EP3126358A1 - 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases - Google Patents

2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases

Info

Publication number
EP3126358A1
EP3126358A1 EP15712926.3A EP15712926A EP3126358A1 EP 3126358 A1 EP3126358 A1 EP 3126358A1 EP 15712926 A EP15712926 A EP 15712926A EP 3126358 A1 EP3126358 A1 EP 3126358A1
Authority
EP
European Patent Office
Prior art keywords
compound
mmp
copd
pulmonary
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP15712926.3A
Other languages
German (de)
French (fr)
Inventor
Hartmut Beck
Volkhart Min-Jian Li
Yolanda Cancho Grande
Andreas Timmermann
Dirk Brohm
Hannah JÖRIßEN
Pamela BOGNER
Michael Gerisch
Dieter Lang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Pharma AG
Original Assignee
Bayer Pharma AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Pharma AG filed Critical Bayer Pharma AG
Publication of EP3126358A1 publication Critical patent/EP3126358A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D253/00Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00
    • C07D253/08Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00 condensed with carbocyclic rings or ring systems

Definitions

  • the present application relates to novel 2,5-disubstituted cyclopentanecarboxylic acid derivatives, processes for their preparation, their use alone or in combinations for the treatment and / or prevention of diseases and their use for the preparation of medicaments for the treatment and / or prevention of diseases, in particular for the treatment and / or prevention of respiratory, pulmonary and cardiovascular diseases.
  • Human macrophage elastase belongs to the family of matrix metallo-peptidases (MMPs) and is also called human matrix metallo-peptidase 12 (hMMP-12).
  • MMPs matrix metallo-peptidases
  • hMMP-12 human matrix metallo-peptidase 12
  • the protein is increased i.a. formed by macrophages after contact with "irritating" substances or particles, activated and released.
  • Such substances and particles may, for example, be contained as impurities in suspended particles, as may be mentioned, inter alia. in cigarette smoke or industrial dusts.
  • endogenous and foreign body cell constituents and cellular debris are counted among these irritant particles, as they may be present in some cases in high concentrations in inflammatory processes.
  • the highly active enzyme is capable of degrading a variety of connective tissue proteins, e.g. primarily the protein elastin (hence the name), as well as other proteins and proteoglycans such as collagen, fibronectin, laminin, chondroitin sulfate, heparan sulfate and others.
  • This proteolytic activity of the enzyme enables macrophages to penetrate the basal membrane.
  • Elastin for example, occurs in high concentrations in all tissue types that exhibit high elasticity, e.g. in the lungs and arteries.
  • the HME plays an important role in tissue degradation (tissue remodeling).
  • the HME is an important modulator in inflammatory processes.
  • TGF-ß tumor necrosis factor-alpha
  • TGF-ß transforming growth factor -beta
  • MMP-12 also plays a role in host defense, particularly in the regulation of antiviral immunity, presumably through intervention in the interferon-alpha (IFN- ⁇ ) -mediated signaling pathway [A new transcriptional role-matrix matrix metalloproteinase -12 in antiviral immunity, Marchant et al., Nature Med. 20, 493-502 (2014)].
  • IFN- ⁇ interferon-alpha
  • HME plays an important role in many diseases, injuries and pathological changes, their emergence and / or progression with an infectious or non-infectious inflammatory event and / or a proliferative and hypertrophic tissue and vascular remodeling.
  • diseases and / or damage to the lung, the kidney or the cardiovascular system or these may be cancerous diseases or other inflammatory diseases [Macrophage metalloelasta.se (MMP-12) as a target for inflammatory respiratory diseases, Lagente et al., Expert Opin. Ther.
  • diseases and injuries of the lung are in particular the chronic obstructive pulmonary illness (COPD), the lung emphysema (lung emphysema), interstitial pulmonary diseases (interstitial lung diseases, ILD) such as the pulmonary fibrosis (ideopathic pulmonary fibrosis, IPF) and pulmonary sarcoidosis (pulmonary sareoidosis), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), cystic fibrosis (CF), also called cystic fibrosis).
  • ILD interstitial lung diseases
  • ILD interstitial lung diseases
  • pulmonary fibrosis ideopathic pulmonary fibrosis, IPF
  • pulmonary sarcoidosis pulmonary sarcoidosis
  • CF cystic fibrosis
  • liver fibrosis and systemic sclerosis are mentioned as examples.
  • Atherosclerosis here in particular carotid arteriosclerosis
  • infective endocarditis in particular viral myocarditis
  • cardiomyopathy Cardiac insufficiency in particular viral myocarditis
  • cardiomyopathy Cardiac insufficiency in particular viral myocarditis
  • cardiomyopathy Cardiac insufficiency in particular viral myocarditis
  • cardiomyopathy Cardiac insufficiency CAD
  • cardiogenic shock acute coronary syndrome (ACS)
  • aneurysms myocardial infarct
  • AMDI myocardial infarct
  • CKD chronic kidney disease
  • Alport syndrome also mentioned here are the metabolic syndrome and obesity.
  • SIRS systemic inflammatory response syndrome
  • MODF multi-organ dysfunction
  • intravascular Coagulation dissminated intravascular coagulation, DIC
  • rheumatoid diseases for example rheumatoid arthritis, as well as chronic bowel inflammation (IBD; Crohn's disease, CD, ulcerative colitis, ulcerative colitis , UC).
  • elastase-mediated pathological processes underlie a shifted balance between free elastase (HME) and the body's own elastase inhibitor protein (tissue inhibitor of metalloproteinase, ⁇ ).
  • HME free elastase
  • tissue inhibitor of metalloproteinase
  • MMP-2 oxidative stress, protease-antiprotease imbalance, and inflammation
  • MMP-7 Matrilysins
  • MMP-12 HME (MMP-12) is so far the only representative of metallo-elastase.
  • MMPs are assembled into the group of so-called MT-MMPs (membrane-type MMPs), since they have a characteristic domain that anchors the protein in the membrane (MMP-14, MMP-15, MMP-16, MMP-17 , MMP-24, MMP-25).
  • MMP-14, MMP-15, MMP-16, MMP-17 , MMP-24, MMP-25 common to all MMPs is a conserved zinc-binding region in the active site of the enzyme, which is important for catalytic activity and is also found in other metalloproteins (eg a disintegrin and metalloproteinase, ADAM).
  • ADAM disintegrin and metalloproteinase
  • the complexed zinc is masked by a sulfhydryl group in the N-terminal pro-peptide domain of the protein, resulting in an enzymatically inactive pro-form of the enzyme. Only after cleavage of this pro-peptide domain is the zinc in the active center of the enzyme freed from this coordination and the enzyme thereby activated (so-called activation by cysteine switch) [matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases, Hu et al ., Nature Rev. Drug Discov. 6, 480-498 (2007)].
  • MMPs MMPs and other similar molecules
  • ADAMs eg ADAMs
  • Numerous in vitro and preclinical in vz 'vo experiments have contributed much to a better understanding of MMPs in different disease models (eg transgenic animals, knock-out animals and genetic data from human studies).
  • the validation of a target with regard to a possible drug therapy can ultimately only be carried out in clinical trials on humans or patients.
  • the first generation of MMP inhibitors has been clinically studied in cancer studies.
  • the desired effect on one or more MMP targets has been masked by an undesired effect on one or more MMP anti-targets or by an undesired effect on another target site (off-target) [Validating matrix metalloproteinases as drug targets and anti-targets for Cancer Therapy, Coverall & Kleifeld, Nature Rev. Cancer 6, 227-239 (2006)].
  • MMP-12 inhibitors Newer MMP inhibitors, which are characterized by increased selectivity, have now also been clinically tested, including compounds explicitly referred to as MMP-12 inhibitors, but so far also without any conclusive clinical success. At a closer Look here, too, the previously described as selective inhibitors have been found to be not so selective.
  • test compound MMP408 shows a significantly decreased affinity for the mouse orthologous MMP-12 target: IC 50 2 nM (human MMP-12), IC 50 160 nM (murine MMP-12), IC 50 320 nm (MMP-12 of the Rat) [see above Li et al., 2009; Mukhopadhyay et al., 2010].
  • the potency at the target MMP-12 itself is very important.
  • a highly potent compound will result in a lower therapeutic dose than a less potent compound, and generally a lower dose should be associated with a reduced likelihood of side effects.
  • the free fraction is defined as the available amount of one A compound that is not bound to components of the blood plasma, which are mainly blood protein components such as eg albumin).
  • the specificity is therefore of paramount importance.
  • novel macrophage elastase inhibiting agents should have high selectivity and specificity in order to be able to specifically inhibit HME.
  • a good metabolic stability of the substances is necessary (low clearance).
  • these compounds should be stable under oxidative conditions so as not to lose their inhibitory potency in disease.
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • the first symptoms of the disease usually appear from the fourth to fifth decade of life. In the following years, the shortness of breath is often aggravated and it manifests cough, associated with an extensive and sometimes purulent sputum and a stenosis breathing to a dyspnea.
  • COPD is primarily a disease of smokers: smoking is responsible for 90% of all COPD cases and 80-90% of all COPD deaths. COPD is a major medical problem and is the sixth most common cause of death worldwide. About 4-6% of over 45's are affected.
  • the underlying mechanism involves immune cells that release various chemokines during the inflammatory response of the lungs.
  • neutrophilic cells and subsequently alveolar macrophages are lured to the lung connective tissue and lumen.
  • Neutrophils secrete a protease cocktail containing mainly HNE and proteinase 3.
  • Activated macrophages release the HME.
  • the protease / antiprotease balance is shifted locally in favor of the proteases, which leads inter alia to an uncontrolled elastase activity and, as a consequence thereof, to an excessive degradation of the elastin of the alveolar bodies. This tissue breakdown causes a collapse of the bronchi.
  • HME protein is associated with smoking or COPD status: detectable HME levels are lowest in non-smokers, slightly higher in former smokers and smokers, and in COPD - Patients significantly increased [Elevated MMP-12 protein levels in induced sputum from patients with COPD, Demedts et al., Thorax 61, 196-201 (2006)]. Similar data were collected with human sputum samples and bronchial alveolar washing fluid (BALF).
  • BALF bronchial alveolar washing fluid
  • HME could be detected and quantified on activated macrophages: HME amount COPD patient / smoker> COPD patient / former smoker> former smoker> Non-smoker [Patterns of airway inflammation and MMP-12 expression in smokers and ex-smokers with COPD, Babusyte et al., Respir. Res. 8, 81-90 (2007)].
  • IPD interstitial lung disease
  • IPF idiopathic pulmonary fibrosis
  • MMP-12 expression is also known in ischemic kidney injuries, as is the involvement of MMP-12 in further inflammatory kidney disease [JNK signaling in human and experimental renal ischemia / reperfusion injury, Kanellis et al., Nephrol. Dial. Transplant. 25, 2898-2908 (2010); Macrophage Metalloelastase as a Major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis, Kaneko et al., J. Immun. 170, 3377-3385 (2003); Role for macrophage metallo elastase in Glomerular Basement Membrane Damage Associated with Alport Syndrome, Rao et al., Am. J. Pathol.
  • the object of the present invention was therefore to identify and provide new substances which act as potent, selective and specific inhibitors of human macrophage elastase (HME / MMP-12) and as such for the treatment and / or prevention of, in particular, respiratory diseases, the Lungs and the cardiovascular system are suitable.
  • HME / MMP-12 human macrophage elastase
  • WO 96/15096-A1 is 4-aryl- and 4-biaryl-substituted 4-oxobutanoic acid derivatives with inhibitory activity towards MMP-2, MMP-3, MMP-9 and, to a lesser extent, MMP-1; Because of this profile of action, the compounds have been found to be particularly suitable for the treatment of osteoarthritis, rheumatoid arthritis and tumor diseases.
  • WO 98/09940 A1 and WO 99/18079 A1 other biarylbutanoic acid derivatives have been disclosed as inhibitors of MMP-2, MMP-3 and / or MMP-13, which are suitable for the treatment of various diseases.
  • WO 00/40539 A1 claims the use of 4-biaryl-4-oxobutanoic acids for the treatment of pulmonary and respiratory diseases, based on a different degree of inhibition of MMP-2, MMP-3, MMP-8, MMP-9, MMP-12 and MMP-13 through these compounds.
  • WO 2012/014114-A1 describes 3-hydroxypropionic acid derivatives and WO 2012/038942-A1 describes oxy- or sulfonylacetic acid derivatives as dual MMP-9/12 inhibitors.
  • PTP-1B protein tyrosine phosphatase 1B
  • the compounds of the present invention are also characterized by a significant inhibitory activity and selectivity towards the rodent orthologous MMP-12 peptidases, such as mouse MMP-12 (also referred to as murine macrophage elastase, MME) and rat MMP-12. This allows a more complete preclinical evaluation of the substances in various established animal models of the diseases described above.
  • rodent orthologous MMP-12 peptidases such as mouse MMP-12 (also referred to as murine macrophage elastase, MME) and rat MMP-12.
  • the present invention relates to compounds of the general formula (I)
  • A is -O- or -S-, n is the number 1 or 2 and
  • R 1 is hydrogen, methyl, fluoromethyl, difluoromethyl or trifluoromethyl, and their salts, solvates and solvates of the salts.
  • Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts comprising the compounds of the formulas below and their salts, solvates and solvates of the salts and of the formula (I) encompassed by formula (I), hereinafter referred to as exemplary compounds and their salts, solvates and solvates of the salts, as far as the compounds of formula (I), the compounds mentioned below are not already salts, solvates and solvates of the salts.
  • Salts used in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. Also included are salts which are not suitable for pharmaceutical applications themselves, but can be used, for example, for the isolation, purification or storage of the compounds according to the invention.
  • Physiologically acceptable salts of the compounds according to the invention include, in particular, the salts derived from customary bases, such as, by way of example and by way of preference, alkali metal salts (eg sodium and potassium salts), alkaline earth salts (eg calcium and magnesium salts), zinc salts and ammonium salts derived from ammonia or organic amines having 1 to 16 C atoms, such as, by way of example and by way of preference, ethylamine, diethylamine, triethylamine, / V, / V-diisopropylethylamine, monoethanolamine, diethanolamine, triethanolamine, tromethamine, dimethylaminoethanol, diethylaminoethanol, choline, procaine, dicyclohexylamine, dibenzylamine, / V-methylmor - pholine, / V-methylpiperidine, arginine, lysine and 1,2-ethylenediamine.
  • customary bases such
  • solvates are those forms of the compounds according to the invention which form a complex in the solid or liquid state by coordination with solvent molecules. Hydrates are a special form of solvates that coordinate with water. As solvates, hydrates are preferred in the context of the present invention.
  • the compounds of the invention may exist in different stereoisomeric forms, i. in the form of configurational isomers or optionally also as conformational isomers (enantiomers and / or diastereomers, including those in atropisomers).
  • the present invention therefore includes the enantiomers and diastereomers and their respective mixtures. From such mixtures of enantiomers and / or diastereomers, the stereoisomerically uniform components can be isolated in a known manner; Preferably, chromatographic methods are used for this, in particular HPLC chromatography on achiral or chiral phase.
  • enantiomerically pure in the context of the present invention is understood to mean that the compound in question in terms of the absolute configuration of the chiral centers in an enantiomeric excess of more than 95%, preferably more than 98%.
  • the enantiomeric excess (ene, ee value) is calculated here by evaluating the chromatogram of a HPLC analysis on a chiral phase according to the following formula:
  • Enantiomer 1 (area%) + enantiomer 2 (area%)
  • the present invention encompasses all tautomeric forms.
  • the present invention also includes all suitable isotopic variants of the compounds of the invention.
  • An isotopic variant of a compound according to the invention is understood to mean a compound in which at least one atom within the compound according to the invention is exchanged for another atom of the same atomic number but with a different atomic mass than the atomic mass that usually or predominantly occurs in nature.
  • isotopes which can be incorporated into a compound of the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as ⁇ (deuterium), ⁇ (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 C1, 82 Br, 123 I, 124 I, 129 I and 131 I
  • Certain isotopic variants of a compound of the invention such as, in particular, those in which one or more radioactive isotopes are incorporated, may be useful, for example, for the study of the mechanism of action or drug distribution in the body; Due to the comparatively easy production and detectability, compounds labeled with 3 H or 14 C isotopes in particular are suitable for this purpose.
  • isotopes such as deuterium may result in certain therapeutic benefits as a result of greater metabolic stability of the compound, such as prolonging the body's half-life or reducing the required effective dose;
  • modifications of the compounds of the invention may therefore optionally also constitute a preferred embodiment of the present invention.
  • Isotopic variants of the compounds according to the invention can be prepared by generally customary processes known to the person skilled in the art, for example by the methods described below and the rules reproduced in the exemplary embodiments by using corresponding isotopic modifications of the respective reagents and / or starting compounds.
  • the present invention also includes prodrugs of the compounds of the invention.
  • prodrugs here denotes compounds which may themselves be biologically active or inactive, but are converted during their residence time in the body by, for example, metabolic or hydrolytic routes to compounds of the invention.
  • the present invention comprises, as prodrugs, hydrolyzable ester derivatives of the carboxylic acids of the formula (I) according to the invention.
  • esters which can be hydrolyzed in physiological media, under the conditions of the biological assays described below, and in particular in vivo enzymatically or chemically to the free carboxylic acids, as the main biologically active compounds.
  • esters preference is given to (C 1 -C -alkyl esters in which the alkyl group may be straight-chain or branched.) Particular preference is given to methyl, ethyl or ethyl-butyl esters.
  • radicals are substituted in the compounds according to the invention, the radicals can, unless otherwise specified, be monosubstituted or polysubstituted. Substitution with one or two identical or different substituents is preferred. Particularly preferred is the substitution with a substituent.
  • A is -O-, n is the number 1 or 2 and
  • R 1 is hydrogen, methyl or trifluoromethyl, and their salts, solvates and solvates of the salts.
  • A is -O-, n is the number 2 and
  • R 1 is hydrogen, methyl or trifluoromethyl, and their salts, solvates and solvates of the salts.
  • A, n and R 1 have the meanings defined above and the groups bound to the central cyclopentane ring have a relative irans arrangement to each other, and mixtures of these compounds, wherein A, n or R 1 in such a mixture of (IA ) and (IB) are each identical, and the salts, solvates and solvates of the salts of these compounds and their mixtures.
  • Preferred in the context of the present invention are the compounds of the formula (IA)
  • A, n and R 1 have the meanings defined above, in enantiomerically pure form with a, as designated, (lS, 2R, 5S) configuration on the central cyclopentane ring and the salts, solvates and solvates of the salts of these compounds.
  • the invention further provides a process for the preparation of the compounds according to the invention, which comprises reacting a compound of the formula (II)
  • X is a leaving group such as, for example, chlorine, bromine, iodine, mesylate, triflate or tosylate, to give a compound of the formula (IV)
  • n, A and R 1 have the meanings given above, alkylated and then the 2- (trimethylsilyl) ethyl ester group with the aid of an acid or a fluoride reagent to the carboxylic acid of formula (I)
  • n, A and R 1 have the abovementioned meanings, and if appropriate separates the compounds of the formula (I) thus obtained into their enantiomers and / or diastereomers and / or with the corresponding (i) solvents and / or (ii) Bases are converted into their solvates, salts and / or solvates of the salts.
  • Suitable bases for the alkylation reaction (II) + ( ⁇ ) - (IV) are in particular suitable alkali metal carbonates such as lithium, sodium, potassium or cesium carbonate, alkali alcoholates such as sodium or potassium methoxide, sodium or potassium ethanolate or sodium or potassium ieri-butoxide, alkali metal hydrides such as sodium or potassium hydride, amide bases such as lithium diisopropylamide or lithium, sodium or potassium bis (trimethylsilyl) amide, or conventional organometallic bases such as phenyllithium or n-, sec- or ieri. butyllithium. Preference is given to using potassium carbonate or potassium ieri-butoxide.
  • alkali metal carbonates such as lithium, sodium, potassium or cesium carbonate
  • alkali alcoholates such as sodium or potassium methoxide, sodium or potassium ethanolate or sodium or potassium ieri-butoxide
  • alkali metal hydrides such as sodium or potassium hydride
  • amide bases such as lithium diiso
  • Suitable inert solvents for this reaction are, for example, ethers, such as diethyl ether, diisopropyl ether, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis (2-methoxyethyl) ether, hydrocarbons, such as benzene, toluene, Xylene, pentane, hexane or cyclohexane, or dipolar aprotic solvents such as acetonitrile, butyronitrile, N, N-dimethylformamide (DMF), ⁇ , ⁇ -Dimefhylacetamid (DMA), ⁇ , ⁇ '-Dimefhylpropylenharnstoff (DMPU), / V Methyl pyrrolidinone (NMP) or dimethyl sulfoxide (DMSO). It is also possible to use mixtures of such solvents.
  • the reaction (II) + ( ⁇ ) - (IV) is generally carried out in a temperature range from 0 ° C to + 120 ° C, depending on the reactivity of the components involved.
  • the cleavage of the 2- (trimethylsilyl) ethyl ester grouping in process step (IV) - (I) is carried out by customary methods with the aid of a strong acid, in particular trifluoroacetic acid, in an inert solvent such as dichloromethane or with the aid of a fluoride, in particular tetrabutylammonium fluoride ( TBAF), in an ethereal solvent such as tetrahydrofuran.
  • the ester cleavage is usually carried out in a temperature range from -20 ° C to + 30 ° C.
  • the compounds of the formula ( ⁇ ), in the case where A is -O-, can be prepared by reacting a compound of the formula (V)
  • PG is a temporary protecting group such as benzyl, in the presence of an alkyl or aryl phosphine and an azodicarboxylate with a triazine-4 (3 //) -one derivative of the formula (VI)
  • R 1 has the meaning given above, splits off.
  • reaction (V) + (VI) - (VII) is carried out under the usual conditions of a "Mitsunobu reaction" in the presence of a phosphine and an azodicarboxylate [see, e.g. D.L. Hughes, Org. Reactions 42, 335 (1992); D.L. Hughes, Org. Prep. Proced. Int. 28 (2), 127 (1996)].
  • Suitable phosphine components are triphenylphosphine, tri-n-butylphosphine, 1,2-bis (diphenylphosphino) ethane (DPPE), diphenyl (2-pyridyl) phosphine, (4-dimethylaminophenyl) diphenylphosphine or tris (4-dimethylaminophenyl ) phosphine.
  • DPPE 1,2-bis (diphenylphosphino) ethane
  • diphenyl (2-pyridyl) phosphine diphenyl (2-pyridyl) phosphine
  • (4-dimethylaminophenyl) diphenylphosphine or tris (4-dimethylaminophenyl ) phosphine.
  • azodicarboxylate for example, diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), Oi tert-butylazodicarboxylat, / V, / V, / VW tetramethylazodicarboxamide (TMAD), l, l '- (azodicarbonyl) di-piperidine (ADDP) or 4,7-dimethyl-3,5,7-hexahydro-l, 2,4,7-tetrazocine-3,8-dione (DHTD) can be used.
  • DEAD diethyl azodicarboxylate
  • DIAD diisopropyl azodicarboxylate
  • TMAD Oi tert-butylazodicarboxylat, / V, / V, / VW tetramethylazodicarboxamide
  • ADDP 4,7-dimethyl-3,5,7-hexahydro-l, 2,4,
  • Inert solvents for this reaction are, for example, ethers, such as diethyl ether, diisopropyl ether, methyl tert. -b tyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis (2-meth- oxyethyl) ether, hydrocarbons such as benzene, toluene, xylene, pentane, hexane or cyclohexane, or polar aprotic solvents such as acetonitrile , Butyronitrile, dimethylsulfoxide (DMSO), N, N-dimethylformamide (DMF), N, N-dimethylacetamide (DMA), N, N'-dimethylpropylurea (DMPU) or / V-methylpyrrolidinone (NMP).
  • ethers such as diethyl ether, diisopropyl ether,
  • reaction (V) + (VI) -> (VII) is generally carried out in a temperature range from -20 ° C to + 60 ° C, preferably at 0 ° C to + 40 ° C.
  • the use of a microwave apparatus in this reaction may be advantageous.
  • the removal of benzyl as a temporary protective group PG in process step (VII) - (II-A) is carried out in the usual manner by hydrogenation with gaseous hydrogen or, in the sense of a transfer hydrogenation, with the aid of a hydrogen donor such as ammonium formate, cyclohexene or cyclohexadiene, in each case in the presence of a suitable hydrogenation catalyst such as in particular palladium on activated carbon.
  • the reaction is preferably carried out in an alcoholic solvent such as methanol or ethanol, in ethyl acetate or tetrahydrofuran or in a mixture of such solvents, optionally with the addition of water, in a temperature range from + 20 ° C to + 80 ° C [to possible alternative protecting groups and for introduction and removal of such protecting groups see also: TW Greene and P.G.M. Wuts, Protective Croups in Organic Synthesis, Wiley, New York, 1999].
  • an alcoholic solvent such as methanol or ethanol
  • ethyl acetate or tetrahydrofuran or in a mixture of such solvents
  • water optionally with the addition of water
  • the preparation of the trifluoromethanesulfonate (VIII) starting from the phenol (II-A) takes place in a conventional manner by reaction with trifluoromethanesulfonic anhydride in the presence of an amine base such as / V-diisopropylethylamine or pyridine.
  • an amine base such as / V-diisopropylethylamine or pyridine.
  • chlorinated hydrocarbons such as dichloromethane or chloroform are generally used, and the reaction is usually carried out in a temperature range of -20 ° C to + 25 ° C.
  • Suitable catalysts are, for example, palladium (II) acetate, palladium (II) chloride, bis (triphenylphosphine) palladium (II) chloride, bis (acetonitrile) palladium (II) chloride, tetrakis (triphenylphosphine) palladium (O), Bis (dibenzylideneacetone) palladium (0), tris (dibenzylideneacetone) dipalladium (O) or [1,1'-bis (diphenylphosphino) ferrocene] palladium (II) chloride, each in combination with a phosphine ligand such as 2-dicyclohexylphosphino 2 ', 4', 6'-triisopropylbiphenyl (X-Phos), 2-dicyclohexylphosphino-2 ', 6'-dimethoxybiphenyl (S-phos), 1,2,3,
  • the reaction is usually carried out in the presence of a base.
  • a base such are alkali metal carbonates such as sodium, potassium or cesium carbonate, alkali metal phosphates such as sodium or potassium phosphate, alkali fluorides such as potassium or cesium fluoride, alkali ieri.-butylates such as sodium or potassium ieri.-butoxide, tertiary amine bases such as triethylamine, -Mefhylmorpholin, / V-methylpiperidine, -Diisopropylefhylamin, pyridine or 4-A i, / V-dimethylaminopyridine, or Amide bases such as lithium, sodium or potassium bis (trimethylsilyl) amide.
  • the reaction is carried out in an inert solvent such as toluene, xylene, 1,2-dimethoxyethane, tetrahydrofuran, 1,4-dioxane, acetonitrile, dimethyl sulfoxide (DMSO), N, N-dimethylformamide (DMF) or / V, / V Dimethylacetamide (DMA) or mixtures thereof in a temperature range of + 50 ° C to + 150 ° C, the use of a microwave apparatus may be advantageous.
  • an inert solvent such as toluene, xylene, 1,2-dimethoxyethane, tetrahydrofuran, 1,4-dioxane, acetonitrile, dimethyl sulfoxide (DMSO), N, N-dimethylformamide (DMF) or / V, / V Dimethylacetamide (DMA) or mixtures thereof in a temperature range of + 50 ° C to + 150 ° C, the
  • Preferred for the transformation (VIII) - (II-B) is a catalyst / ligand / base system consisting of tris (dibenzylideneacetone) dipalladium (0), 4,5-bis (diphenylphosphino) -9,9-dimethyl-xanthene ( Xantphos) and -Diisopropylefhylamin used and 1,4-dioxane used as a solvent.
  • the trialkylsilylsulfide initially formed in this reaction is split again under the conditions used here for aqueous reaction work-up and chromatographic product purification, so that the free thiophenol ( ⁇ - ⁇ ) is obtained directly [cf. also M. Kreis and S.
  • the compounds of the formula (V), in turn, can be prepared in various ways, starting from compounds of the formula (IX) or (X), based on published synthesis processes.
  • Hai is a halogen atom [see, e.g. the general preparative methods described in WO 96/15096-A1, pages 26-44, in particular the methods A, G, H and K].
  • VA (VB)
  • PG in which PG has the abovementioned meaning
  • PG can be prepared in analogy to published synthesis methods, for example by reacting e o-2- (trimethylsilyl) efhyl 2-oxobicyclo [2.2.1] heptane-7 carboxylate of the formula (XI)
  • R 1 has the abovementioned meaning, accessible with sodium nitrite in aqueous hydrochloric acid [see, eg, D. Fernandez-Forner et al., Tetrahedron 47 (42), 8917-8930 (1991)].
  • stereoisomers enantiomers and / or diastereomers
  • the separation of stereoisomers (enantiomers and / or diastereomers) of the compounds of the formula (I) according to the invention can be achieved by customary methods known to the person skilled in the art.
  • chromatographic methods for achiral or chiral separation phases are used for this purpose.
  • a separation of the compounds according to the invention into the corresponding enantiomers and / or diastereomers may, if appropriate, also be carried out at the stage of intermediates (II), (IV), (V), (VII), (II-A), (II) ⁇ - ⁇ ) or (VA) / (VB), which are then further reacted in separated form according to the reaction sequence described above.
  • intermediates II), (IV), (V), (VII), (II-A), (II) ⁇ - ⁇ ) or (VA) / (VB)
  • the compounds according to the invention have valuable pharmacological properties and can be used for the prevention and treatment of diseases in humans and animals.
  • the compounds of the invention are potent, non-reactive and selective inhibitors of human macrophage elastase (HME / hMMP-12), which have a significantly improved profile of potency and selectivity compared to the compounds known in the art.
  • the compounds of the invention show good solubility in aqueous systems and low non-specific binding to blood plasma components such as albumin.
  • the compounds of the invention also have low in vivo clearance and good metabolic stability. Overall, this property profile makes it possible to expect low dosability for the compounds according to the invention and, as a result of the more targeted mode of action, a reduced risk of the occurrence of undesired side effects in the therapy.
  • the compounds according to the invention are therefore particularly suitable for the treatment and / or prevention of diseases and pathological processes, in particular those in which, in the course of an infectious or non-infectious inflammatory event and / or a tissue or vascular remodeling, the macrophage elastase (HME / hMMP-12).
  • these include, in particular, diseases of the respiratory tract and the lungs, such as chronic obstructive pulmonary disease (COPD), asthma and the group of interstitial lung diseases (ILD), and diseases of the cardiovascular system, such as arteriosclerosis and aneurysms ,
  • COPD chronic obstructive pulmonary disease
  • pulmonary emphysema e.g. Cigarette smoke-induced pulmonary emphysema, chronic bronchitis (CB), pulmonary hypertension in COPD (PH-COPD), bronchiectasis (BE) and combinations thereof, especially in acute exacerbating stages of the disease (AE-COPD).
  • CB chronic bronchitis
  • PH-COPD pulmonary hypertension in COPD
  • BE bronchiectasis
  • AE-COPD acute exacerbating stages of the disease
  • Types of asthma include asthmatic diseases of varying severity with intermittent or persistent events, such as refractory asthma, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, and medication-induced or dust-induced asthma.
  • interstitial lung diseases includes idiopathic pulmonary fibrosis (IPF), pulmonary sarcoidosis and acute interstitial pneumonia, non-specific interstitial pneumonia, lymphoid interstitial pneumonia, respiratory bronchiolitis with interstitial lung disease, cryptogenic organizing pneumonia, desquamative interstitial pneumonia and non-classifiable idiopathic interstitial pneumonia, granulomatous maternal interstitial lung disease, interstitial lung disease of known cause and other interstitial lung diseases of unknown cause.
  • IPF idiopathic pulmonary fibrosis
  • pulmonary sarcoidosis and acute interstitial pneumonia
  • non-specific interstitial pneumonia non-specific interstitial pneumonia
  • lymphoid interstitial pneumonia lymphoid interstitial pneumonia
  • respiratory bronchiolitis with interstitial lung disease cryptogenic organizing pneumonia
  • desquamative interstitial pneumonia and non-classifiable idiopathic interstitial pneumonia granulomatous maternal interstitial lung disease
  • the compounds of the invention may also be used for the treatment and / or prevention of other respiratory and pulmonary diseases, e.g. Pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH), bronchiolitis obliterans syndrome (BOS), acute respiratory tract syndrome (ARDS), acute lung injury (ALI), alpha-1-antitrypsin deficiency (AATD ) and cystic fibrosis (CF), various forms of bronchitis (chronic bronchitis, infectious bronchitis, eosinophilic bronchitis), bronchiectasis, pneumonia, farmer's lung and related diseases, infectious and non-infectious cough and cold diseases (chronic inflammatory cough, iatrogenic cough), nasal mucosal inflammation (including medicinal rhinitis, vasomotor rhinitis and seasonal, allergic rhinitis, eg hay fever) and polyps.
  • PH Pulmonary arterial hypertension
  • PH bronchiolitis
  • the group of diseases of the cardiovascular system includes, in particular, arteriosclerosis and its secondary diseases, such as, for example, Stroke in arteriosclerosis of the cervical arteries (carotid arteriosclerosis), myocardial infarction in arteriosclerosis of the coronary arteries, peripheral arterial occlusive disease (PAOD) due to arteriosclerosis of the leg arteries, as well as aneurysms, in particular aneurysms of the aorta, e.g.
  • arteriosclerosis and its secondary diseases such as, for example, Stroke in arteriosclerosis of the cervical arteries (carotid arteriosclerosis), myocardial infarction in arteriosclerosis of the coronary arteries, peripheral arterial occlusive disease (PAOD) due to arteriosclerosis of the leg arteries, as well as aneurysms, in particular aneurysms of the aorta, e.g.
  • arteriosclerosis hypertension, injuries and inflammations, infections (eg rheumatic fever, syphilis, Lyme disease), congenital connective tissue weaknesses (eg in Marfan syndrome and Ehlers-Danlos syndrome) or as a result of a volume burden of the aorta in congenital heart defects with right-left shunt or shunt-dependent perfusion of the lungs, as well as aneurysms on coronary vessels in the course of a disease in Kawasaki syndrome and in brain areas in patients with a congenital malformation of the aortic valve.
  • arteriosclerosis hypertension, injuries and inflammations, infections (eg rheumatic fever, syphilis, Lyme disease), congenital connective tissue weaknesses (eg in Marfan syndrome and Ehlers-Danlos syndrome) or as a result of a volume burden of the aorta in congenital heart defects with right-left shunt or shunt-dependent perfusion of the lungs, as well as
  • the compounds of the invention may also be used for the treatment and / or prevention of other cardiovascular diseases such as hypertension, heart failure, coronary heart disease, stable and unstable angina pectoris, renal hypertension, peripheral and cardial vascular diseases, arrhythmias, atrial arrhythmias and of the ventricles as well as conduction disorders such as atrio-ventricular blockades of grade I-III, supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, torsades de pointes tachycardia, extrasystoles of the atrium and ventricle, atrioventricular extrasystoles, Sick sinus syndrome, syncope, AV nodal reentry tachycardia, Wolff-Parkinson-White syndrome, acute Coronary syndrome (ACS), autoimmune heart disease (pericarditis, endocarditis, valvolitis, aortitis,
  • cardiac failure encompasses both acute and chronic manifestations of cardiac insufficiency as well as specific or related forms of disease thereof, such as acute decompensated heart failure, right heart failure, left heart failure, global insufficiency, ischemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart disease, heart valve defects, cardiac insufficiency in valvular heart failure, mitral stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspid stenosis, tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined valvular heart failure, myocarditis, chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic toxicity Cardiomyopathy, cardiac storage disorders as well as diastolic
  • kidney diseases in particular renal insufficiency and kidney failure.
  • renal insufficiency and renal failure include both acute and chronic manifestations thereof as well as underlying or related renal diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial disorders, nephropathic disorders such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as renal transplant rejection and Alport syndrome, immune complex-induced kidney disease, toxic-induced nephropathy, contrast-induced nephropathy, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive Nephrosclerosis and nephrotic syndrome, which are diagnosed by, for
  • the present invention also encompasses the use of the compounds of the invention for the treatment and / or prevention of sequelae of renal insufficiency, such as hypertension, pulmonary edema, heart failure, uremia, anemia, electrolyte imbalances (eg, hyperkalemia, hyponatremia) and disorders in bone and carbohydrate metabolism.
  • sequelae of renal insufficiency such as hypertension, pulmonary edema, heart failure, uremia, anemia, electrolyte imbalances (eg, hyperkalemia, hyponatremia) and disorders in bone and carbohydrate metabolism.
  • the compounds according to the invention are suitable for the treatment and / or prevention of diseases of the genitourinary system, such as benign prostatic syndrome (BPS), benign prostatic hyperplasia (BPH), benign prostatic hyperplasia (BPE), bladder emptying disorders (BOO), lower urinary tract syndromes (LUTS) , neurogenic overactive bladder (OAB), incontinence such as mixed, urgency, stress or overflow incontinence (MUI, UUI, SUI, OUI), pelvic pain, as well as erectile dysfunction and female sexual dysfunction.
  • BPS benign prostatic syndrome
  • BPH benign prostatic hyperplasia
  • BPE benign prostatic hyperplasia
  • BOO bladder emptying disorders
  • LUTS lower urinary tract syndromes
  • OAB neurogenic overactive bladder
  • incontinence such as mixed, urgency, stress or overflow incontinence (MUI, UUI, SUI, OUI), pelvic pain, as well as erectile dysfunction and female sexual dysfunction.
  • the compounds of the invention have anti-inflammatory activity and can therefore be used as anti-inflammatory agents for the treatment and / or prevention of sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory diseases of the kidney, chronic intestinal inflammation (IBD, Crohn's disease, ulcerative colitis ), Pancreatitis, peritonitis, cystitis, urethritis, prostatitis, epidymitis, oophoritis, salpingitis, vulvovaginitis, rheumatoid diseases, inflammatory diseases of the central nervous system, multiple sclerosis, inflammatory skin diseases and inflammatory ocular diseases.
  • SIRS sepsis
  • MODS multiple organ failure
  • IBD chronic intestinal inflammation
  • Crohn's disease chronic intestinal inflammation
  • ulcerative colitis ulcerative colitis
  • Pancreatitis peritonitis
  • cystitis cystitis
  • urethritis prostatitis
  • epidymitis oophoritis
  • the compounds according to the invention are furthermore suitable for the treatment and / or prevention of fibrous diseases of the internal organs, such as, for example, the lung, the heart, the kidney, the bone marrow and in particular the liver, as well as dermatological fibroses and fibroid diseases of the eye .
  • the term fibrotic disorders includes in particular such diseases as liver fibrosis, liver cirrhosis, pulmonary fibrosis, endomyocardial fibrosis, nephropathy, glomerulonephritis, interstitial kidney fibrosis, fibrotic damage as a result of diabetes, bone marrow fibrosis, peritoneal fibrosis and similar fibrotic disorders, scleroderma, morphea, Keloids, hypertrophic scarring, nevi, diabetic retinopathy, proliferative vitroretinopathy and connective tissue disorders (eg sarcoidosis).
  • the compounds of the invention may also be used to promote wound healing, to combat postoperative scarring, eg, after glaucoma surgery, and for cosmetic purposes on aging or keratinizing skin.
  • the compounds of the invention may be used for the treatment and / or prevention of anemias, such as hemolytic anemias, especially hemoglobinopathies such as sickle cell anemia and thalassemias, megaloblastic anemias, iron deficiency anemias, acute blood loss anemia, crowding anaemias and aplastic anemias.
  • the compounds of the invention are also useful in the treatment of cancers such as skin cancer, brain tumors, breast cancer, bone marrow tumors, leukemias, liposarcomas, carcinomas of the gastrointestinal tract, liver, pancreas, lung, kidney, ureter, prostate and genital tract, and of malignant tumors of the lymphoproliferative system, such as Hodgkin's and Non-Hodgkin's Lymphoma.
  • cancers such as skin cancer, brain tumors, breast cancer, bone marrow tumors, leukemias, liposarcomas, carcinomas of the gastrointestinal tract, liver, pancreas, lung, kidney, ureter, prostate and genital tract
  • malignant tumors of the lymphoproliferative system such as Hodgkin's and Non-Hodgkin's Lymphoma.
  • the compounds according to the invention can be used for the treatment and / or prevention of lipid metabolism disorders and dyslipidemias (hypolipoproteinemia, hypertriglyceridemia, hyperlipidemia, combined hyperlipidemias, hypercholesterolemia, abetalipoproteinemia, sitosterolemia), xanthomatosis, Tangier's disease, obesity, obesity , metabolic disorders (metabolic syndrome, hyperglycemia, insulin-dependent diabetes, non-insulin-dependent diabetes, gestational diabetes, hyperinsulinemia, insulin resistance, glucose intolerance and diabetic sequelae such as retinopathy, nephropathy and neuropathy), diseases of the gastrointestinal tract and the abdomen (Glositis, gingivitis, periodontitis, esophagitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, colitis, proctitis, pruritis ani, diarrhea, celiac disease, hepatitis, liver fibrosis, liver cirr
  • the compounds according to the invention are particularly suitable for the treatment and / or prevention of respiratory and pulmonary diseases, especially chronic obstructive pulmonary disease (COPD), in particular pulmonary emphysema, chronic bronchitis (CB), pulmonary Hypertension in COPD (PH-COPD) and bronchiectasis (BE) as well as combinations of these diseases, especially in acute exacerbating stages of COPD disease (AE-COPD), asthma and interstitial lung diseases, in particular idiopathic pulmonary fibrosis ( IPF) and pulmonary sarcoidosis, diseases of the cardiovascular system, in particular atherosclerosis, especially carotid arteriosclerosis, as well as viral myocarditis, cardiomyopathy and aneurysms, including their sequelae such as stroke, myocardial infarction and peripheral artery disease (PAOD), as well as chronic kidneys - diseases and the Alport syndrome.
  • COPD chronic obstructive pulmonary disease
  • CB chronic bronchit
  • treatment includes inhibiting, delaying, arresting, alleviating, attenuating, restraining, reducing, suppressing, restraining or curing a disease, a disease, a disease, an injury or a medical condition , the unfolding, the course or progression of such conditions and / or the symptoms of such conditions.
  • therapy is understood to be synonymous with the term “treatment”.
  • prevention means the avoidance or reduction of the risk, a disease, a disease, a disease, an injury or a health disorder, a development or a Progression of such conditions and / or to get, experience, suffer or have the symptoms of such conditions.
  • the treatment or the prevention of a disease, a disease, a disease, an injury or a health disorder can be partial or complete.
  • Another object of the present invention is the use of the compounds of the invention for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
  • Another object of the present invention is the use of the compounds of the invention for the manufacture of a medicament for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
  • Another object of the present invention is a pharmaceutical composition containing at least one of the compounds of the invention, for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
  • Another object of the present invention is the use of the compounds of the invention in a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
  • Another object of the present invention is a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases, using an effective amount of at least one of the compounds of the invention.
  • the compounds according to the invention can be used alone or as needed in combination with one or more other pharmacologically active substances, as long as this combination does not lead to undesired and unacceptable side effects.
  • a further subject of the present invention are therefore medicaments containing at least one of the compounds according to the invention and one or more further active compounds, in particular for the treatment and / or prevention of the aforementioned diseases.
  • Suitable combination active ingredients for this purpose are by way of example and preferably mentioned:
  • Anti-obstructive / bronchodilatory agents e.g. for the treatment of chronic obstructive pulmonary disease (COPD) or bronchial asthma, by way of example and preferably from the group of inhaled or systemically administered beta-adrenergic receptor agonists (beta-mimetics), the inhaled anti-muscarinic substances and the PDE 4 inhibitors;
  • COPD chronic obstructive pulmonary disease
  • bronchial asthma by way of example and preferably from the group of inhaled or systemically administered beta-adrenergic receptor agonists (beta-mimetics), the inhaled anti-muscarinic substances and the PDE 4 inhibitors;
  • organic nitrates and NO donors such as sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
  • Compounds which inhibit the degradation of cyclic guanosine monophosphate (cGMP) and / or cyclic adenosine monophosphate (cAMP) for example inhibitors of phosphodiesterases (PDE) 1, 2, 3, 4 and / or 5, in particular PDE 4 inhibitors such as roflumi last and PDE 5 inhibitors such as sildenafil, vardenafil, tadalafil, uddenafil, dasantafil, avanafil, mirodenafil or lodenafil;
  • PDE phosphodiesterases
  • roflumi last and PDE 5 inhibitors such as sildenafil, vardenafil, tadalafil, uddenafil,
  • sGC soluble guanylate cyclase
  • HNE human neutrophil elastase
  • Sivelastat Sivelastat
  • DX-890 Reltran
  • Prostacyclin analogs and IP receptor agonists such as by way of example and preferably iloprost, beraprost, treprostinil, epoprostenol or NS-304; Endothelin receptor antagonists, such as by way of example and preferably bosentan, darusentan, ambrisentan or sitaxsentan; anti-inflammatory, immunomodulatory, immunosuppressant and / or cytotoxic agents, by way of example and preferably from the group of systemic or inhaled corticosteroids, and acetylcysteine, montelukast, azathioprine, cyclophosphamide, hydroxycarbamide, azithromycin, IFN- ⁇ , pirfenidone or etanercept; antifibrotic agents, such as, by way of example and by way of preference, lysophosphatidic acid receptor 1 (LPA-1) antagonists, lysyl oxidase (LOX) inhibitors, ly
  • Compounds which block the binding of serotonin to its receptor by way of example and preferably antagonists of the 5-HT2B receptor such as PRX-08066; Antagonists of growth factors, cytokines and chemokines, by way of example and preferably antagonists of TGF- ⁇ , CTGF, IL-1, IL-4, IL-5, IL-6, IL-8, IL-13 and integrins;
  • the Rho kinase inhibiting compounds such as by way of example and preferably Fasudil, Y-27632, SLx-2119, BF-66851, BF-66852, BF-66853, KI-23095 or BA-1049;
  • the energy metabolism of the heart affecting compounds such as by way of example and preferably etomoxir, dichloroacetate, ranolazine or trimetazidine;
  • Antithrombotic agents by way of example and preferably from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances;
  • chemotherapeutic agents as e.g. used for the treatment of neoplasms of the lungs or other organs; and or
  • Antibiotics in particular from the group of fluoroquinolonecarboxylic acids, such as by way of example and preferably ciprofloxacin or moxifloxacin.
  • the compounds according to the invention are administered in combination with a beta-adrenergic receptor agonist such as, for example and preferably, albuterol, isoproterenol, metaproterenol, terbutaline, fenoterol, formoterol, repro sterol, salbutamol or salmeterol.
  • a beta-adrenergic receptor agonist such as, for example and preferably, albuterol, isoproterenol, metaproterenol, terbutaline, fenoterol, formoterol, repro sterol, salbutamol or salmeterol.
  • the compounds according to the invention are administered in combination with an anti-muscarinergic substance, such as by way of example and preferably ipratropium bromide, tiotropium bromide or oxitropium bromide.
  • an anti-muscarinergic substance such as by way of example and preferably ipratropium bromide, tiotropium bromide or oxitropium bromide.
  • the compounds according to the invention are administered in combination with a corticosteroid, such as by way of example and preferably prednisone, prednisolone, methylprednisolone, triamcinolone, dexamethasone, beclomethasone, betamethasone, flunisolide, budesonide or fluticasone.
  • a corticosteroid such as by way of example and preferably prednisone, prednisolone, methylprednisolone, triamcinolone, dexamethasone, beclomethasone, betamethasone, flunisolide, budesonide or fluticasone.
  • Antithrombotic agents are preferably understood as meaning compounds from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances.
  • the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, such as, by way of example and by way of preference, aspirin, clopidogrel, ticlopidine or dipyridamole.
  • a platelet aggregation inhibitor such as, by way of example and by way of preference, aspirin, clopidogrel, ticlopidine or dipyridamole.
  • the compounds according to the invention are administered in combination with a thrombin inhibitor, such as, by way of example and by way of preference, ximelagatran, melagatran, dabigatran, bivalirudin or Clexane.
  • the compounds according to the invention are administered in combination with a GPIIb / nia antagonist, such as, by way of example and by way of preference, tirofiban or abciximab.
  • a GPIIb / nia antagonist such as, by way of example and by way of preference, tirofiban or abciximab.
  • the compounds according to the invention are used in combination with a factor Xa inhibitor, such as by way of example and preferably rivaraban, apixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA -1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • a factor Xa inhibitor such as by way of example and preferably rivaraban, apixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA -1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative.
  • LMW low molecular weight
  • the compounds according to the invention are administered in combination with a vitamin K antagonist, such as by way of example and preferably coumarin.
  • antihypertensive agents are preferably compounds from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blocker, beta-receptor blocker, mineralocorticoid receptor Antagonists and diuretics understood.
  • the compounds according to the invention are administered in combination with a calcium antagonist, such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem.
  • a calcium antagonist such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem.
  • the compounds according to the invention are administered in combination with an alpha-1-receptor blocker, such as by way of example and preferably prazosin.
  • the compounds according to the invention are used in combination with a beta-receptor blocker, such as by way of example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipropanol, nadolol, mepindolol, carazalol, Sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, Carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucine dolol administered.
  • a beta-receptor blocker such as by way of example and preferably propranolol, atenolol, timolol
  • the compounds according to the invention are administered in combination with an angiotensin AII antagonist, such as by way of example and preferably losartan, candesartan, valsartan, telmisartan or embursatan.
  • an angiotensin AII antagonist such as by way of example and preferably losartan, candesartan, valsartan, telmisartan or embursatan.
  • the compounds according to the invention are administered in combination with an ACE inhibitor such as, by way of example and by way of preference, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • an ACE inhibitor such as, by way of example and by way of preference, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • an endothelin antagonist such as, by way of example and by way of preference, bosentan, darusentan, ambrisentan or sitaxsentan.
  • the compounds of the invention are administered in combination with a renin inhibitor, such as by way of example and preferably aliskiren, SPP-600 or SPP-800.
  • a renin inhibitor such as by way of example and preferably aliskiren, SPP-600 or SPP-800.
  • the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist, such as by way of example and preferably spironolactone, eplerenone or finerenone.
  • a mineralocorticoid receptor antagonist such as by way of example and preferably spironolactone, eplerenone or finerenone.
  • the compounds according to the invention are administered in combination with a diuretic, such as by way of example and preferably furosemide, bumetanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazide, polythiazide, trichloromethiazide, chlorthalidone, indapamide, metolazone, quineth- azon, acetazolamide, dichlorophenamide, methazolamide, glycerol, isosorbide, mannitol, amiloride or triamterene.
  • a diuretic such as by way of example and preferably furosemide, bumetanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazide, polythiazide, trich
  • the lipid metabolizing agents are preferably compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase or squalene synthesis inhibitors, the ACAT inhibitors, MTP inhibitors, PPAR alpha- , PPAR gamma and / or PPAR delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, lipase inhibitors and the lipoprotein (a) antagonists understood.
  • the compounds according to the invention are administered in combination with a CETP inhibitor, such as by way of example and preferably torcetrapib (CP-529 414), JJT-705 or CETP vaccine (Avant).
  • a CETP inhibitor such as by way of example and preferably torcetrapib (CP-529 414), JJT-705 or CETP vaccine (Avant).
  • the compounds according to the invention are administered in combination with a thyroid receptor agonist such as, by way of example and by way of preference, D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214) ,
  • a thyroid receptor agonist such as, by way of example and by way of preference, D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214) ,
  • the compounds according to the invention are administered in combination with an HMG-CoA reductase inhibitor from the class of statins, such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
  • statins such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
  • the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, such as by way of example and preferably BMS-188494 or TAK-475.
  • a squalene synthesis inhibitor such as by way of example and preferably BMS-188494 or TAK-475.
  • the compounds according to the invention are administered in combination with an ACAT inhibitor, such as, for example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • an ACAT inhibitor such as, for example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • the compounds according to the invention are administered in combination with an MTP inhibitor such as, for example and preferably, implitapide, BMS-201038, R-103757 or JTT-130.
  • an MTP inhibitor such as, for example and preferably, implitapide, BMS-201038, R-103757 or JTT-130.
  • the compounds of the invention are administered in combination with a PPAR-gamma agonist such as, by way of example and by way of preference, pioglitazone or rosiglitazone.
  • the compounds according to the invention are administered in combination with a PPAR delta agonist, such as by way of example and preferably GW 501516 or BAY 68-5042.
  • the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
  • a cholesterol absorption inhibitor such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
  • the compounds according to the invention are administered in combination with a lipase inhibitor, such as, for example and preferably, orlistat.
  • a lipase inhibitor such as, for example and preferably, orlistat.
  • the compounds of the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and by way of preference, cholestyramine, colestipol, colesolvam, cholesta gel or colestimide.
  • ASBT IBAT
  • AZD-7806 S-8921
  • AK-105 AK-105
  • BARI-1741 AK-105
  • SC-435 SC-635.
  • the compounds according to the invention are administered in combination with a lipoprotein (a) antagonist, such as by way of example and preferably gemcabene calcium (CI-1027) or nicotinic acid.
  • a lipoprotein (a) antagonist such as by way of example and preferably gemcabene calcium (CI-1027) or nicotinic acid.
  • compositions according to the invention with one or more further active compounds selected from the group consisting of corticosteroids, beta-adrenergic receptor agonists, anti-muscarcinogenic substances, PDE 4 inhibitors, PDE 5 inhibitors, sGC activators, sGC Stimulants, HNE inhibitors, prostacyclin analogs, endothelin antagonists, statins, antifibrotic agents, antiinflammatory agents, immunomodulating agents, immunosuppressive agents and cytotoxic agents.
  • further active compounds selected from the group consisting of corticosteroids, beta-adrenergic receptor agonists, anti-muscarcinogenic substances, PDE 4 inhibitors, PDE 5 inhibitors, sGC activators, sGC Stimulants, HNE inhibitors, prostacyclin analogs, endothelin antagonists, statins, antifibrotic agents, antiinflammatory agents, immunomodulating agents, immunosuppressive agents and cytotoxic agents.
  • compositions containing at least one compound of the invention usually together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the purposes mentioned above.
  • the compounds according to the invention can act systemically and / or locally.
  • they may be applied in a suitable manner, e.g. oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic, or as an implant or stent.
  • the compounds according to the invention can be administered in suitable administration forms.
  • the compounds of the invention rapidly and / or modified donating application forms containing the compounds of the invention in crystalline and / or amorphized and / or dissolved form, such as tablets (uncoated or coated Tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention), rapidly disintegrating tablets or films / wafers, films / lyophilisates, capsules (for example hard or soft gelatin capsules), dragees, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
  • tablets uncoated or coated Tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention
  • Parenteral administration can be done bypassing a resorption step (eg intravenously, intraarterially, intracardially, intraspinally or intralumbarly) or using absorption (for example by inhalation, intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally).
  • a resorption step eg intravenously, intraarterially, intracardially, intraspinally or intralumbarly
  • absorption for example by inhalation, intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally.
  • suitable application forms include injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
  • Inhalation medicaments including powder inhalers, nebulizers, metered dose aerosols
  • nasal drops solutions or sprays
  • lingual, sublingual or buccal tablets films / wafers or capsules
  • suppositories ear or ophthalmic preparations
  • vaginal capsules aqueous suspensions (lotions, shake mixtures)
  • lipophilic suspensions ointments
  • creams transdermal therapeutic systems (for example patches)
  • milk pastes, foams, scattering powders, implants or stents.
  • the compounds according to the invention can be converted into the stated administration forms. This can be done in a conventional manner by mixing with inert, non-toxic, pharmaceutically suitable excipients.
  • excipients for example microcrystalline cellulose, lactose, mannitol
  • solvents for example liquid polyethylene glycols
  • emulsifiers and dispersants or wetting agents for example sodium dodecyl sulfate, polyoxysorbitanoleate
  • binders for example polyvinylpyrrolidone
  • synthetic and natural polymers for example albumin
  • Stabilizers eg antioxidants such as ascorbic acid
  • dyes eg inorganic pigments such as iron oxides
  • flavor and / or odoriferous include, among others.
  • Excipients for example microcrystalline cellulose, lactose, mannitol
  • solvents for example liquid polyethylene glycols
  • emulsifiers and dispersants or wetting agents for example sodium dodecyl
  • the dosage is about 0.01 to 100 mg kg, preferably about 0.01 to 20 mg / kg and most preferably 0.1 to 10 mg / kg body weight.
  • the amount is generally about 0.1 to 50 mg per inhalation.
  • a designation "IRS, 2RS, 5SR" in the IUP AC name of the relevant example, in conjunction with the term “racemate”, means that this is a racemic mixture of IR, 2R, 5S - Enantiomers (- each 1st letter after the position number in "IRS, 2RS, 5SR”) with the appropriate lS, 2S, 5R-enantiomer (-> each 2nd letter after the position number) acts.
  • Example 8A 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- (4-hydroxybenzoyl) -5- ⁇ [4-oxo-6- (trifluoromethyl) -l, 2,3-benzotriazine-3 (4 /) - yl] methyl ⁇ cyclopentanecarboxylate (racemate)
  • the mixture was combined with the reaction mixtures of two similar preliminary experiments (batch size in each case 47 mg (0.08 mmol) of the compound from Example 8A). After removal of the DMF, 60 ml of water and 60 ml of ethyl acetate were added to this combined mixture. After separating the phases, the aqueous phase was extracted once with 30 ml of ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and concentrated.
  • the aqueous phase was extracted once with 30 ml of ieri.-butyl-methyl ether and twice with 50 ml of ethyl acetate.
  • the combined organic phases were dried over sodium sulfate, filtered and concentrated.
  • the residue was taken up in dichloromethane and purified by column chromatography (25 g silica gel, eluent cyclohexane / ethyl acetate 7: 3). There were obtained 138 mg (46% of theory, purity 100%) of the title compound.
  • the pharmacological activity of the compounds according to the invention can be detected by in vitro and in vzvo studies, as are known to those skilled in the art.
  • the following application examples describe the biological activity of the compounds according to the invention without restricting the invention to these examples.
  • the potency of the compounds of the invention compared to HME is determined in a in vi tro-inhibited st.
  • the HME-mediated amidolytic cleavage of a suitable peptide substrate results here in a fluorescence light increase.
  • the signal intensity of the fluorescence light is directly proportional to the enzyme activity.
  • the effective concentration of a test compound in which half of the enzyme is inhibited is given as IC 5 o value.
  • This test corresponds to the standard HME inhibition test described above, but using a modified reaction buffer.
  • This reaction buffer additionally contains bovine serum albumin (BSA, fatty acid free, A6003, Sigma-Aldrich) of a final concentration of 2% (w / w), which corresponds approximately to half of the physiological serum albumin content.
  • BSA bovine serum albumin
  • the enzyme concentration in this modified assay is slightly elevated (e.g., 0.75 nM) as is the incubation time (e.g., three hours).
  • Table 1A Inhibition of human macrophage elastase (HME / hMMP-12)
  • Table 1B Inhibition of human macrophage elastase (HME / hMMP-12) in the absence (-) or presence (+) of serum albumin (BSA)
  • the potency of the compounds of the invention over other MMPs is also determined in vz 'iro inhibition assays.
  • the MMP-mediated amidolytic cleavage of a suitable peptide substrate also leads here to a fluorescence light increase.
  • the signal intensity of the fluorescent light is directly proportional to the enzyme activity.
  • the effective concentration of a test compound in which half of the enzyme is inhibited (50% signal intensity of the fluorescent light) is given as IC 50 value.
  • Recombinant MMP-1 (R & D Systems, 901-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-1 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Recombinant MMP-2 (R & D Systems, 902-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentra- tion, for example, 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl, 150 mM NaCl, 0.05% Brij ® -35
  • concentrations for example 1 nM to 30 ⁇
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Recombinant MMP-3 (R & D Systems, 513-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is carried out by adding the intramolecularly quenched substrate McA-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-002 ) so that a total test volume of 50 ⁇ results.
  • the course of the MMP-3 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant MMP-7 (R & D Systems, 907-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-7 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • In vitro MMP-8 inhibition test :
  • Recombinant MMP-8 (R & D Systems, 908-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant MMP-9 (R & D Systems, 911-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • In vitro ⁇ -10 inhibition test :
  • Recombinant MMP-10 (R & D Systems, 910-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is carried out by adding the intramolecularly quenched substrate McA-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-002 ) so that a total test volume of 50 ⁇ results.
  • the course of the MMP-10 reaction is determined by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C).
  • Recombinant MMP-13 (R & D Systems, 511-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-13 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • In vitro ⁇ -14 inhibition test :
  • Recombinant MMP-14 (R & D Systems, 918-MP) is enzymatically activated according to the manufacturer's instructions by using recombinant furin (R & D Systems, 1503-SE).
  • activated enzyme final concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as Solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 ⁇ ; R & D Systems, ES-010) a total test volume of 50 ⁇ results.
  • the course of the MMP-14 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Recombinant MMP-16 (R & D Systems, 1785-MP) is enzymatically activated according to the manufacturer's instructions by using recombinant furin (R & D Systems, 1503-SE).
  • activated enzyme final concentration eg 1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is achieved by addition of the intramolecular quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 ⁇ , R & D Systems, ES-010) is started so that a total test volume of 50 ⁇ results.
  • the course of the MMP-16 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • the IC 50 values from these assays for inhibiting human MMPs are reported (in part as averages of several independent determinations and rounded to two significant digits):
  • Recombinant mouse MMP-2 (R & D Systems, 924-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) that a total test volume of 50 ⁇ results.
  • the course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • In vitro MMP-3 inhibition test of the mouse In vitro MMP-3 inhibition test of the mouse:
  • Recombinant mouse MMP-3 (R & D Systems, 548-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • the examined test compound as a solution in DMSO, suitable concentrations pipetted example 1 nM to 30 ⁇
  • the enzymatic reaction is carried out by addition of the intramolecularly quenched substrate Mca-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH2 (final concentration eg 5 ⁇ ; R & D Systems, ES-002) so that a total test volume of 50 ⁇ results.
  • the course of the MMP-3 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant mouse MMP-7 (R & D Systems, 2967-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.5 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 ⁇ ; R & D Systems, ES-010) a total test volume of 50 ⁇ results.
  • the course of the MMP-7 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Recombinant mouse MMP-8 (R & D Systems, 2904-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 ⁇ ; R & D Systems, ES-010) a total test volume of 50 ⁇ results.
  • the course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • In vitro MMP-9 inhibition test of the mouse In vitro MMP-9 inhibition test of the mouse:
  • Recombinant mouse MMP-9 (R & D Systems, 909-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant mouse MMP-12 (R & D Systems, 3467-MP) is autocatalytically activated according to the manufacturer's instructions.
  • activated enzyme final concentration eg 1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 ⁇ ; R & D Systems, ES-010) that a total test volume of 50 ⁇ results.
  • the course of the MMP-12 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Recombinant rat MMP-2 (R & D Systems, 924-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • the examined test compound as a solution in DMSO, suitable concentrations pipetted example 1 nM to 30 ⁇
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant rat MMP-8 (R & D Systems, 3245-MP) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 2 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 ⁇ ; R & D Systems, ES-010) a total test volume of 50 ⁇ results.
  • the course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Recombinant mouse MMP-9 (R & D Systems, 5427-MM) is chemically activated according to the manufacturer's instructions by using APMA.
  • activated enzyme final concentration eg 0.1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35
  • 1 ⁇ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 ⁇ ) in a white 384-well microtiter plate (MTP) pipetted.
  • MTP white 384-well microtiter plate
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
  • Rat in vitro MMP-12 inhibition test :
  • Rat MMP-12 (Uniprot NP_446415.1; construct L96-V277) is expressed with an additional N-terminal His tag and a TEV consecutive cleavage sequence using a pDEco7 vector in E. coli (BL21).
  • the recombinantly expressed protein forms an intracellular insoluble protein compo- sition (so-called inclusion body). This is solubilized after separation and intensive washing under denaturing conditions.
  • the inclusion body pellet fraction from a 250 ml E. coli culture in a volume of 120 ml of buffer A (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl 2 , 10 mM CaCl 2 , 8 M urea).
  • the soluble protein is renatured by dialysing each 60 ml of the sample several times at 4-8 ° C against buffer B (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl 2 , 10 mM CaCl 2 ). After dialysis, the sample is centrifuged (25,000 xg). The refolded protein is in the supernatant with a yield of 3.7 mg per 250 ml-E. coli culture. The protein thus obtained is enzymatically active without further purification operations or protease-mediated cleavage processes.
  • MMP-12 protein final concentration, for example 1 nM
  • reaction buffer 50 mM Tris / HCl pH 7.5, 10 mM CaCl 2, 150 mM NaCl, 0.05% Brij ® -35
  • concentrations eg 1 nM to 30 ⁇ in a white 384-well microtiter plate (MTP) pipetted.
  • the enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 ⁇ ; R & D Systems, ES-001) Total test volume of 50 ⁇ results.
  • the course of the MMP-12 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
  • Table 3 shows representative IC 50 values from mouse MMP inhibition assays (in part as averages of several independent determinations and rounded to two significant digits) for representative embodiments of the invention:
  • Elastase-induced pulmonary emphysema in mouse, rat or hamster is a widely used animal model of pulmonary emphysema [The Fas / Fas-ligand pathway does not mediate the apoptosis in elastase-induced emphysema in mice, Sawada et al., Exp. Lung Res. 33, 277-288 (2007)].
  • the animals receive orotracheal instillation of porcine pancreatic elastase.
  • the treatment of the animals with the test substance starts on the day of the instillation of the porcine pancreatic elastase and extends over a period of 3 weeks.
  • lung compliance is determined and alveolar morphometry performed.
  • B-4 Animal model of silica-induced lung inflammation
  • Orotracheal administration of silica to mouse, rat or hamster leads to lung inflammation [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 ( 2010)].
  • the animals are treated with the test substance on the day of the instillation of the silica. After 24 hours bronchioalveolar lavage is performed to determine cell content and biomarkers.
  • Silica-induced lung fibrosis in mouse, rat or hamster is a widely used animal model of pulmonary fibrosis [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 (2010 )].
  • the animals receive oro-tracheal instillation of silica.
  • the treatment of the animals with the test substance starts during the day the instillation of the silica or therapeutically a week later and extends over a period of 6 weeks.
  • a bronchioalveolar lavage is performed to determine cell content and biomarkers, and a histological assessment of pulmonary fibrosis is performed.
  • Intratracheal administration of ATP (adenosine triphosphate) to the mouse causes lung inflammation [ATP via the P2Y receptors: An experimental study, Matsuyama et al., Respir. Res. 9:79 (2008)].
  • the animals are treated with the test substance for 24 h on the day of instillation of ATP (by gavage, by addition in feed or drinking water, by osmotic minipump, by subcutaneous or intraperitoneal injection or by inhalation).
  • a bronchio-alveolar lavage is performed to determine the cell content and the pro-inflammatory markers.
  • the ability of substances to inhibit the human CYP enzymes CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in human is examined using pooled human liver microsomes as enzyme source in the presence of standard substrates (see above) which form CYP-specific metabolites.
  • the inhibition effects are investigated at six different concentrations of the test compounds [2.8, 5.6, 8.3, 16.7, 20 (or 25) and 50 ⁇ ], compared with the extent of CYP-specific metabolite formation of the standard substrates in the absence of the test compounds and the corresponding IC 50 values calculated.
  • a standard inhibitor that specifically inhibits a single CYP isoform is always incubated to make comparisons between different series comparable.
  • test compounds are preferably dissolved in acetonitrile.
  • 96-well plates are incubated for a defined time at 37 ° C with pooled human liver microsomes. The reactions are stopped by addition of 100 ⁇ L acetonitrile, which is a suitable internal standard. Precipitated proteins are separated by centrifugation, the supernatants are pooled and analyzed by LC-MS / MS. B-eighth Hepatocyte assay for determination of metabolic stability
  • the metabolic stability of test compounds to hepatocytes is determined by incubating the compounds at low concentrations (preferably below or around 1 ⁇ ) and at low cell counts (preferably at 1 ⁇ 10 6 cells / ml) to ensure the best possible linear kinetic conditions in the experiment , Seven samples from the incubation solution are taken at a fixed time interval for LC-MS analysis to determine the half-life (ie, degradation) of each compound. From this half-life different "clearance” parameters (CL) and "Fmax” values are calculated (see below).
  • the CL and Fmax values represent a measure of the phase 1 and phase 2 metabolism of the compounds in the hepatocytes.
  • hepatocyte cell count in the liver 1.1 * 10 8 cells / g liver is expected.
  • CL parameters calculated using half-lives that are significantly longer than the incubation time can only be considered as rough guidelines.
  • liver microsomes or with primary fresh hepatocytes of various animal species eg rat, dog
  • primary fresh hepatocytes of various animal species eg rat, dog
  • the compounds of the invention are incubated at a concentration of about 1-10 ⁇ .
  • stock solutions of the compounds are prepared at a concentration of 0.1-1 mM in acetonitrile and then pipetted with a 1: 100 dilution in the incubation mixture.
  • the liver microsomes are incubated in 50 mM potassium phosphate buffer pH 7.4 with and without NADPH-generating system consisting of 1 mM NADP + , 10 mM glucose-6-phosphate and 1 unit glucose-6-phosphate dehydrogenase at 37 ° C.
  • Primary hepatocytes are also incubated in suspension in William's E medium at 37 ° C.
  • the incubation mixtures are stopped with acetonitrile (final concentration about 30%) and the protein is centrifuged off at about 15,000 ⁇ g.
  • the thus stopped samples are either analyzed directly or stored at -20 ° C until analysis.
  • the analysis is carried out by means of high performance liquid chromatography with ultraviolet and mass spectrometric detection (HPLC-UV-MS / MS).
  • HPLC-UV-MS / MS ultraviolet and mass spectrometric detection
  • the supernatants of the incubation samples are chromatographed with suitable C18 reverse phase columns and variable eluent mixtures of acetonitrile and 10 mM aqueous ammonium formate solution or 0.05% aqueous formic acid.
  • the UV chromatograms in combination with the mass spectrometric data serve to identify, structure elucidate and quantitatively estimate the metabolites and to quantitatively determine the metabolic decrease of the compounds according to the invention in the incubation mixtures.
  • the substance to be tested is administered intravenously to rats or mice as a solution (eg in appropriate plasma with a small amount of DMSO or in a PEG / ethanol / water mixture), the oral administration is carried out as a solution (eg in Solutol / Ethanol / water or PEG / ethanol / water mixtures) or as a suspension (eg in Tylose) in each case via a gavage.
  • a solution eg in Solutol / Ethanol / water or PEG / ethanol / water mixtures
  • a suspension eg in Tylose
  • the pharmacokinetic parameters are calculated using an internal standard and with the aid of a validated computer program, such as AUC (area under the concentration-time curve), Cmax (maximum plasma concentration), t (half-life), Vss (distribution volume) and CL (clearance) and the absolute and relative bioavailability F and F re i (iv / po comparison or comparison of suspension to solution after po administration).
  • test substance is dissolved in DMSO. An aliquot is taken from this solution and introduced into PBS buffer pH 6.5 (DMSO content: 1%). This solution / suspension is shaken for 24 h at room temperature. After ultra-centrifugation at 114,000 g for 30 min, the supernatant is removed, diluted with acetonitrile / water 8: 2 and analyzed by LC-MSMS. Quantification is via a five-point calibration curve of the test compound in DMSO.
  • Eluent A 0.5 ml of formic acid / liter of water
  • eluent B 0.5 ml of formic acid / liter of acetonitrile
  • Gradient 0 min 90% A -> 0.5 min 5% A -> 0.84 min 5% A -> 0.85 min 90% A ⁇ 1.22 min 90% A
  • Flow 2.5 ml / min
  • Injection volume 15 ⁇
  • Column Waters OASIS HLB, 2.1 x 20 mm, 25 ⁇ ; Column temperature: 30 ° C; Splitter (before MS): 1:20.
  • FIA Flow Injection Analysis
  • MRM Multiple Reaction Monitoring
  • the compounds according to the invention can be converted into pharmaceutical preparations as follows:
  • composition
  • the mixture of compound of the invention, lactose and starch is granulated with a 5% solution (m / m) of the PVP in water.
  • the granules are mixed after drying with the magnesium stearate for 5 minutes.
  • This mixture is compressed with a conventional tablet press (for the tablet format see above).
  • a pressing force of 15 kN is used as a guideline for the compression.
  • the rhodigel is suspended in ethanol, the compound according to the invention is added to the suspension. While stirring, the addition of water. Until the completion of the swelling of Rhodigels is stirred for about 6 h.
  • the compound of the invention is suspended in the mixture of polyethylene glycol and polysorbate with stirring. The stirring is continued until complete dissolution of the compound according to the invention.
  • i.v. solution The compound of the invention is dissolved at a concentration below the saturation solubility in a physiologically acceptable solvent (e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%). The solution is sterile filtered and filled into sterile and pyrogen-free injection containers.
  • a physiologically acceptable solvent e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%.

Abstract

The invention relates to novel 2,5-disubstituted cyclopentane carboxylic acid derivatives, to methods for the preparation thereof, to the use thereof alone or in combination for the treatment and/or prevention of disorders, and to the use thereof for producing medicaments for the treatment and/or prevention of disorders, especially for treatment and/or prevention of diseases of the respiratory tract, lung and of the cardiovascular system.

Description

2,5-DISUBSTITUIERTE CYCLOPENTANCARBONSÄUREN ZUR BEHANDLUNG VON  2,5-DISUBSTITUTED CYCLOPENTANCARBOXYLIC ACIDS FOR THE TREATMENT OF
ATEMWEGSERKRANKUNGEN RESPIRATORY DISEASES
Die vorliegende Anmeldung betrifft neue 2,5-disubstituierte Cyclopentancarbonsäure-Derivate, Verfahren zu ihrer Herstellung, ihre Verwendung allein oder in Kombinationen zur Behandlung und/oder Prävention von Krankheiten sowie ihre Verwendung zur Herstellung von Arzneimitteln zur Behandlung und/oder Prävention von Krankheiten, insbesondere zur Behandlung und/oder Prävention von Erkrankungen der Atemwege, der Lunge und des Herz-Kreislauf-Systems.  The present application relates to novel 2,5-disubstituted cyclopentanecarboxylic acid derivatives, processes for their preparation, their use alone or in combinations for the treatment and / or prevention of diseases and their use for the preparation of medicaments for the treatment and / or prevention of diseases, in particular for the treatment and / or prevention of respiratory, pulmonary and cardiovascular diseases.
Die humane Makrophagen-Elastase (HME, EC 3.4.24.65) gehört zur Familie der Matrix-Metallo- Peptidasen (MMPs) und wird auch humane Matrix-Metallo-Peptidase 12 (hMMP-12) genannt. Das Protein wird vermehrt u.a. von Makrophagen nach Kontakt mit "reizenden" Stoffen oder Partikeln gebildet, aktiviert und freigesetzt. Solche Stoffe und Partikel können beispielsweise als Fremdstoffe in Schwebeteilchen enthalten sein, wie sie u.a. in Zigarettenrauch oder Industriestäuben vorkommen. Im weiteren Sinne werden auch körpereigene und körperfremde Zellbestandteile und Zelltrümmer zu diesen Reizpartikeln gezählt, wie sie in zum Teil hoher Konzentration bei entzündlichen Prozessen vorliegen können. Das hochaktive Enzym ist in der Lage, eine Vielzahl von Bindegewebs-Proteinen abzubauen, z.B. vornehmlich das Protein Elastin (daher der Name), sowie weitere Proteine und Proteoglykane wie Kollagen, Fibronektin, Laminin, Chondroitinsulfat, Hepa- ransulfat und andere mehr. Durch diese proteolytische Aktivität des Enzyms werden Makrophagen in die Lage versetzt, die basale Membran zu penetrieren. Elastin zum Beispiel kommt in hohen Konzentrationen in allen Gewebetypen vor, die eine hohe Elastizität zeigen, z.B. in der Lunge und in Arterien. Bei einer Vielzahl von pathologischen Prozessen, wie Gewebeverletzungen, spielt die HME eine wichtige Rolle beim Gewebeabbau und -umbau (engl, tissue remodeling). Darüber hinaus ist die HME ein wichtiger Modulator bei entzündlichen Prozessen. Es ist ein Schlüsselmolekül bei der Rekrutierung von Entzündungszellen, indem es zum Beispiel den zentralen Entzündungsmediator Tumor-Nekrosefaktor-alpha (TNF-α) freisetzt und in den durch transformieren- den Wachstumsfaktor -beta (TGF-ß) vermittelten Signalweg eingreift [Hydrolysis of a Broad Spectrum of Extracellular Matrix Proteins by Human Macrophage Elastase, Gronski et al., J. Biol. Chem. 272, 12189-12194 (1997)] . MMP-12 spielt auch eine Rolle in der körpereigenen Abwehr (engl, host defense), insbesondere bei der Regulation der antiviralen Immunität, vermutlich durch einen Eingriff in den Interferon-alpha (IFN-a)-vermittelten Signalweg [A new transcriptional roleor matrix metalloproteinase-12 in antiviral immunity, Marchant et al., Nature Med. 20, 493-502 (2014)] . Human macrophage elastase (HME, EC 3.4.24.65) belongs to the family of matrix metallo-peptidases (MMPs) and is also called human matrix metallo-peptidase 12 (hMMP-12). The protein is increased i.a. formed by macrophages after contact with "irritating" substances or particles, activated and released. Such substances and particles may, for example, be contained as impurities in suspended particles, as may be mentioned, inter alia. in cigarette smoke or industrial dusts. In a broader sense, endogenous and foreign body cell constituents and cellular debris are counted among these irritant particles, as they may be present in some cases in high concentrations in inflammatory processes. The highly active enzyme is capable of degrading a variety of connective tissue proteins, e.g. primarily the protein elastin (hence the name), as well as other proteins and proteoglycans such as collagen, fibronectin, laminin, chondroitin sulfate, heparan sulfate and others. This proteolytic activity of the enzyme enables macrophages to penetrate the basal membrane. Elastin, for example, occurs in high concentrations in all tissue types that exhibit high elasticity, e.g. in the lungs and arteries. In a variety of pathological processes, such as tissue injury, the HME plays an important role in tissue degradation (tissue remodeling). In addition, the HME is an important modulator in inflammatory processes. It is a key molecule in the recruitment of inflammatory cells, for example by releasing the central inflammatory mediator tumor necrosis factor-alpha (TNF-α) and interfering with the transforming growth factor -beta (TGF-ß) mediated signaling pathway [Hydrolysis of a Broad Spectrum of Extracellular Matrix Proteins by Human Macrophage Elastase, Gronski et al., J. Biol. Chem. 272, 12189-12194 (1997)]. MMP-12 also plays a role in host defense, particularly in the regulation of antiviral immunity, presumably through intervention in the interferon-alpha (IFN-α) -mediated signaling pathway [A new transcriptional role-matrix matrix metalloproteinase -12 in antiviral immunity, Marchant et al., Nature Med. 20, 493-502 (2014)].
Es wird daher angenommen, dass die HME bei vielen Erkrankungen, Verletzungen und pathologischen Veränderungen eine wichtige Rolle spielt, deren Entstehung und/oder Progression mit einem infektiösen oder nicht-infektiösen entzündlichen Geschehen und/oder einem proliferativen und hypertrophen Gewebe- und Gefäßumbau in Zusammenhang steht. Dies können insbesondere Erkrankungen und/oder Schädigungen der Lunge, der Niere oder des Herz-Kreislauf-Systems sein, oder es kann sich hierbei um Krebs-Erkrankungen oder um andere entzündliche Erkrankungen handeln [Macrophage metalloelasta.se (MMP-12) as a target for inflammatory respiratory dis- eases, Lagente et al., Expert Opin. Ther. Targets 13, 287-295 (2009); Macrophage Metalloelastase as a major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis, Kaneko et al., J. Immunol. 170, 3377-3385 (2003); A Selective Matrix Metalloelastase-12 Inhibitor Retards Atherosclerotic Plaque Development in Apolipoprotein E Knock-out Mice, Johnson et al., Arterioscler. Thromb. Vase. Biol. 31, 528-535 (2011); Impaired Coronary Collateral Growth in the Metabolie Syndrome Is in Part Mediated by Matrix Metalloelastase 12-dependent Production of Endo statin and Angiostatin, Dodd et al., Arterioscler. Thromb. Vase. Biol. 33, 1339- 1349 (2013); Matrix metalloproteinase pharmacogenomics in non-small-cell lung Carcinoma, Chetty et al., Pharmacogenomics 12, 535-546 (2011)] . It is therefore assumed that the HME plays an important role in many diseases, injuries and pathological changes, their emergence and / or progression with an infectious or non-infectious inflammatory event and / or a proliferative and hypertrophic tissue and vascular remodeling. These may be, in particular, diseases and / or damage to the lung, the kidney or the cardiovascular system, or these may be cancerous diseases or other inflammatory diseases [Macrophage metalloelasta.se (MMP-12) as a target for inflammatory respiratory diseases, Lagente et al., Expert Opin. Ther. Targets 13, 287-295 (2009); Macrophage Metalloelastase as a Major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis, Kaneko et al., J. Immunol. 170, 3377-3385 (2003); A Selective Matrix Metalloelastase-12 Inhibitor Retards Atherosclerotic Plaque Development in Apolipoprotein E Knock-out Mice, Johnson et al., Arterioscler. Thromb. Vase. Biol. 31, 528-535 (2011); Impaired Coronary Collateral Growth in the Metabolic Syndrome is Mediated by Matrix Metalloelastase 12-dependent Production of Endodontin and Angiostatin, Dodd et al., Arterioscler. Thromb. Vase. Biol. 33, 1339-1349 (2013); Carcinoma, Chetty et al., Pharmacogenomics 12, 535-546 (2011)]. Matrix metalloproteinase pharmacogenomics in non-small-cell lung carcinoma.
In diesem Kontext zu nennende Erkrankungen und Schädigungen der Lunge sind insbesondere die chronisch-obstruktive Lungenerkrankung (engl, chronic obstructive pulmonary disease, COPD), das Lungenemphysem (lung emphysema), interstitielle Lungenerkrankungen (interstitial lung diseases, ILD) wie z.B. die Lungenfibrose (ideopathic pulmonary fibrosis, IPF) und die Lungen- sarkoidose (pulmonary sareoidosis), die akute Lungenschädigung (acute lung injury, ALI), das akute Atemwegssyndrom (acute respiratory distress Syndrome, ARDS), zystische Fibrose (cystic fibrosis, CF; auch Mukoviszidose genannt), Asthma sowie infektiös, insbesondere viral bedingte Atemwegserkrankungen. Als andere fibrotische Erkrankungen seien hier beispielhaft die Leber - fibrose und die systemische Sklerose erwähnt. Erkrankungen und Schädigungen des Herz-Kreislauf-Systems, in denen die HME involviert ist, sind zum Beispiel Gewebe- und Gefäßveränderungen bei einer Arteriosklerose, hier insbesondere die karotide Arteriosklerose, die infektive Endo- karditis, hier insbesondere die virale Myokarditis, die Kardiomyopathie, die Herzinsuffizienz, der kardiogene Schock, das akute Koronarsyndrom (acute coronary Syndrome, ACS), Aneurysmen, Reperfusionsschäden nach einem Myokardinfarkt (acute myocardial infaret, AMI), ischämische Schädigungen der Niere oder der Retina sowie deren chronische Verläufe, wie zum Beispiel die chronische Nierenerkrankung (chronic kidney disease, CKD) und das Alport-Syndrom. Genannt seien hier auch das metabolische Syndrom und Adipositas. Erkrankungen in Zusammenhang mit einer Sepsis sind beispielsweise eine systemische entzündliche Reaktion (systemic inflammatory response Syndrome, SIRS), die schwere Sepsis, der septische Schock und das multiple Organversagen (multi-organ failure, MOF; multi-organ dysfunetion, MODS) sowie die intravaskuläre Gerinnung (disseminated intravascular coagulation, DIC). Beispiele für einen Gewebeab- und -umbau bei Krebsprozessen sind das Einwandern von Krebszellen in das gesunde Gewebe (Metastasenbildung) und die Neuausbildung von versorgenden Blutgefäßen (Neo-Angiogenese). Andere entzündliche Krankheiten, bei denen die HME eine Rolle spielt, sind rheumatoide Erkrankungen, zum Beispiel die rheumatoide Arthritis, sowie chronische Darmentzündungen (inflammatory bowel disease, IBD; Morbus Crohn, engl. Crohn 's disease, CD; Colitis ulcerosa, engl, ulcerative Colitis, UC). Im Allgemeinen geht man davon aus, dass Elastase-vermittelten pathologischen Prozessen ein verschobenes Gleichgewicht zwischen der freien Elastase (HME) und dem körpereigenen Elastase- Inhibitorprotein (tissue Inhibitor of metalloproteinase, ΤΓΜΡ) zugrunde liegt. In verschiedenen pathologischen, insbesondere entzündlichen Prozessen ist die Konzentration an freier Elastase (HME) erhöht, so dass lokal die Balance zwischen Protease und Anti-Protease zu Gunsten der Pro- tease verschoben ist. Ein ähnliches (Un-)Gleichgewicht besteht zwischen der Elastase der neutro- philen Zellen (human neutrophil elastase, HNE, ein Mitglied der Serinprotease-Familie) und der körpereigenen Anti-Protease AAT (alpha- 1 anti-Trypsin, ein Mitglied der Serinprotease-Inhibi- toren, SERPINs). Beide Gleichgewichte sind miteinander gekoppelt, da HME den Inhibitor der HNE spaltet und inaktiviert und umgekehrt HNE den HME-Inhibitor spaltet und inaktiviert, wo- durch sich die jeweiligen Protease/ Anti-Protease-Ungleichgewichte zusätzlich verschieben können. Außerdem herrschen im Umfeld von lokalen Entzündungen stark oxidierende Bedingungen (engl. oxidative burst), wodurch das Protease/ Anti-Protease-Ungleichgewicht weiter verstärkt wird [Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation, Fischer et al., Int. J. COPD 6, 413-421 (2011)]. Es sind derzeit mehr als 20 MMPs bekannt, die historisch grob in verschiedene Klassen hinsichtlich ihrer prominentesten Substrate eingeteilt werden, z.B. Gelatinasen (MMP-2, MMP-9), Kollagenasen (MMP-1, MMP-8, MMP-13), Stromelysine (MMP-3, MMP-10, MMP-11) und Matri- lysine (MMP-7, MMP-26). HME (MMP-12) ist bisher einziger Vertreter der Metallo-Elastase. Darüber hinaus werden weitere MMPs zur Gruppe der sogenannten MT-MMPs (membrane-type MMPs) zusammengefügt, da diese eine charakteristische Domäne besitzen, die das Protein in der Membran verankert (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24, MMP-25). Allen MMPs ist eine konservierte Zink-bindende Region im aktiven Zentrum des Enzyms gemeinsam, die für die kataly tische Aktivität wichtig ist und die auch in anderen Metallo-Proteinen zu finden ist (z.B. a disintegrin and metalloproteinase, ADAM). Das komplexierte Zink wird durch eine Sulfhydryl- Gruppe in der N-terminalen Pro-Peptid-Domäne des Proteins maskiert, was zu einer enzymatisch inaktiven Pro-Form des Enzyms führt. Erst durch eine Abspaltung dieser Pro-Peptid-Domäne wird das Zink im aktiven Zentrum des Enzyms von dieser Koordinierung befreit und das Enzym dadurch aktiviert (sog. Aktivierung durch cysteine switch) [Matrix metalloproteinase Inhibitors as therapy for inflammatory and vascular diseases, Hu et al., Nature Rev. Drug Discov. 6, 480-498 (2007)]. Die meisten bekannten synthetischen MMP-Inhibitoren verfügen über eine Zink-komplexierende funktionelle Gruppe, sehr häufig zum Beispiel ein Hydroxamat, ein Carboxylat oder ein Thiol [Recent Developments in the Design of Specific Matrix Metalloproteina.se Inhibitors aided by Structural and Computational Studies, B.G. Rao, Curr. Pharm. Des. 11, 295-322 (2005)]. Die Gerüststruktur (scaffold) dieser Inhibitoren ähnelt häufig noch Peptiden, man spricht dann von sogenannten Peptidomimetika (in der Regel mit einer schlechten oralen Bioverfügbarkeit), oder sie weist keine Ähnlichkeit zu Peptiden auf, man spricht dann allgemeiner von kleinen Molekülen (small molecules, SMOLs). Die physikochemischen und pharmakokinetischen Eigenschaften dieser Inhibitoren haben, ganz allgemein gesagt, einen großen Einfluss darauf, welche Zielmoleküle (targets) und welche unerwünschten Moleküle (anti-targets, off-targets) in welchem Gewebe und in welchem Zeitraum in welchem Ausmaß "getroffen" werden. In this context to be named diseases and injuries of the lung are in particular the chronic obstructive pulmonary illness (COPD), the lung emphysema (lung emphysema), interstitial pulmonary diseases (interstitial lung diseases, ILD) such as the pulmonary fibrosis (ideopathic pulmonary fibrosis, IPF) and pulmonary sarcoidosis (pulmonary sareoidosis), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), cystic fibrosis (CF), also called cystic fibrosis). , Asthma and infectious, especially viral respiratory diseases. As other fibrotic diseases, liver fibrosis and systemic sclerosis are mentioned as examples. Diseases and injuries of the cardiovascular system in which the HME is involved are, for example, tissue and vascular changes in atherosclerosis, here in particular carotid arteriosclerosis, infective endocarditis, in particular viral myocarditis, cardiomyopathy Cardiac insufficiency, cardiogenic shock, acute coronary syndrome (ACS), aneurysms, myocardial infarct (AMI) reperfusion injury, ischemic damage to the kidney or retina, and chronic disease such as chronic kidney disease. chronic kidney disease, CKD) and Alport syndrome. Also mentioned here are the metabolic syndrome and obesity. Diseases associated with sepsis include systemic inflammatory response syndrome (SIRS), severe sepsis, septic shock and multi-organ dysfunction (MODF), as well as intravascular Coagulation (disseminated intravascular coagulation, DIC). Examples of tissue degradation and remodeling in cancer processes are the invasion of cancer cells into the healthy tissue (metastasis) and the reformation of supplying blood vessels (neo-angiogenesis). Other Inflammatory diseases in which the HME plays a role are rheumatoid diseases, for example rheumatoid arthritis, as well as chronic bowel inflammation (IBD; Crohn's disease, CD, ulcerative colitis, ulcerative colitis , UC). In general, it is believed that elastase-mediated pathological processes underlie a shifted balance between free elastase (HME) and the body's own elastase inhibitor protein (tissue inhibitor of metalloproteinase, ΤΓΜΡ). In various pathological, especially inflammatory processes, the concentration of free elastase (HME) is increased, so that locally the balance between protease and anti-protease is shifted in favor of the protease. A similar (in) equilibrium exists between the elastase of the neutrophil cells (human neutrophil elastase, HNE, a member of the serine protease family) and the endogenous anti-protease AAT (alpha-1 anti-trypsin, a member of the serine protease family). Inhibitors, SERPINs). Both equilibria are coupled with each other since HME cleaves and inactivates the HNE inhibitor and, conversely, HNE cleaves and inactivates the HME inhibitor, thereby additionally shifting the respective protease / anti-protease imbalances. In addition, in the environment of local inflammation, oxidative bursts prevail, further enhancing protease / anti-protease imbalance [Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation, Fischer et al. , Int. J. COPD 6, 413-421 (2011)]. There are currently more than 20 known MMPs that are historically roughly classified into different classes in terms of their most prominent substrates, eg gelatinases (MMP-2, MMP-9), collagenases (MMP-1, MMP-8, MMP-13), stromelysins (MMP-3, MMP-10, MMP-11) and Matrilysins (MMP-7, MMP-26). HME (MMP-12) is so far the only representative of metallo-elastase. In addition, further MMPs are assembled into the group of so-called MT-MMPs (membrane-type MMPs), since they have a characteristic domain that anchors the protein in the membrane (MMP-14, MMP-15, MMP-16, MMP-17 , MMP-24, MMP-25). Common to all MMPs is a conserved zinc-binding region in the active site of the enzyme, which is important for catalytic activity and is also found in other metalloproteins (eg a disintegrin and metalloproteinase, ADAM). The complexed zinc is masked by a sulfhydryl group in the N-terminal pro-peptide domain of the protein, resulting in an enzymatically inactive pro-form of the enzyme. Only after cleavage of this pro-peptide domain is the zinc in the active center of the enzyme freed from this coordination and the enzyme thereby activated (so-called activation by cysteine switch) [matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases, Hu et al ., Nature Rev. Drug Discov. 6, 480-498 (2007)]. Most known synthetic MMP inhibitors have a zinc-complexing functional group, very often for example a hydroxamate, a carboxylate or a thiol [Recent Developments in the Design of Specific Matrix Metalloproteinase Inhibitors aided by Structural and Computational Studies, BG Rao , Curr. Pharm. Des. 11, 295-322 (2005)]. The scaffold of these inhibitors is often similar to peptides, one speaks of so-called peptidomimetics (usually with a poor oral bioavailability), or it has no similarity to peptides, one speaks more generally of small molecules (small molecules, SMOLs ). The physicochemical and pharmacokinetic properties of these inhibitors have, in general terms, a great influence on which targets and which undesired molecules (anti-targets, off-targets) are "hit" in which tissue and for what period to what extent ,
Es ist hierbei eine große Herausforderung, die spezifische Rolle einer bestimmten MMP in einem Krankheitsgeschehen zu bestimmen. Erschwert wird dies insbesondere durch den Umstand, dass es eine Vielzahl von MMPs und weiterer ähnlicher Moleküle (z.B. ADAMs) gibt, verbunden mit einer Vielzahl an jeweils möglichen physiologischen Substraten und damit unter Umständen auch einhergehenden inhibitorischen oder aktivatorischen Effekten in vielfältigen Signaltransduktions- wegen. Zahlreiche in vitro- und präklinische in vz'vo-Experimente haben viel zu einem besseren Verständnis der MMPs in verschiedenen Krankheitsmodellen beigetragen (z.B. transgene Tiere, knock-out-Tiere sowie genetische Daten aus Humanstudien). Die Validierung eines Targets hin- sichtlich einer möglichen medikamentösen Therapie kann letztendlich nur in klinischen Testreihen am Menschen bzw. Patienten erfolgen. Die erste Generation von MMP-Inhibitoren wurde hierbei in Krebsstudien klinisch untersucht. Zu dieser Zeit waren erst wenige Vertreter der MMP-Protein- familie bekannt. Keiner der untersuchten Inhibitoren konnte klinisch überzeugen, da bei wirksamen Dosierungen die aufgetretenen Nebenwirkungen nicht tolerierbar waren. Wie sich im Zuge der Kenntnis weiterer MMPs herausstellte, handelte es sich bei den Vertretern der ersten Inhibitor- Generation um nicht-selektive Inhibitoren, d.h. eine Vielzahl verschiedener MMPs wurde gleichermaßen inhibiert (pan-MMP-Inhibitoren, pan-MMPIs). Vermutlich wurde die erwünschte Wirkung an einem oder mehreren MMP-Targets überdeckt von einer unerwünschten Wirkung an einem oder mehreren MMP-anti-Targets oder durch eine unerwünschte Wirkung an einem sonstigen Ziel- ort (off-target) [Validating matrix metallo proteinases as drug targets and anti-targets for Cancer therapy, Overall & Kleifeld, Nature Rev. Cancer 6, 227-239 (2006)]. It is a major challenge to determine the specific role of a particular MMP in a disease process. This is compounded in particular by the fact that there are a large number of MMPs and other similar molecules (eg ADAMs), associated with a large number of physiological substrates that are possible in each case and thus possibly also associated inhibitory or activatory effects in a variety of signal transduction pathways. Numerous in vitro and preclinical in vz 'vo experiments have contributed much to a better understanding of MMPs in different disease models (eg transgenic animals, knock-out animals and genetic data from human studies). The validation of a target with regard to a possible drug therapy can ultimately only be carried out in clinical trials on humans or patients. The first generation of MMP inhibitors has been clinically studied in cancer studies. At that time, only a few representatives of the MMP protein family were known. None of the tested inhibitors could clinically convince, since at effective dosages, the side effects were not tolerated. As it became clear in the course of the knowledge of further MMPs, the representatives of the first inhibitor generation were non-selective inhibitors, ie a large number of different MMPs were equally inhibited (pan-MMP inhibitors, pan-MMPIs). Presumably, the desired effect on one or more MMP targets has been masked by an undesired effect on one or more MMP anti-targets or by an undesired effect on another target site (off-target) [Validating matrix metalloproteinases as drug targets and anti-targets for Cancer Therapy, Coverall & Kleifeld, Nature Rev. Cancer 6, 227-239 (2006)].
Neuere MMP-Inhibitoren, die sich durch eine erhöhte Selektivität auszeichnen, sind nun ebenfalls klinisch getestet worden, darunter auch explizit als MMP-12-Inhibitoren bezeichnete Verbindungen, bislang allerdings ebenso ohne durchschlagenden klinischen Erfolg. Bei einem genaueren Hinsehen haben sich auch hier die zuvor als selektiv beschriebenen Inhibitoren als nicht ganz so selektiv herausgestellt. Newer MMP inhibitors, which are characterized by increased selectivity, have now also been clinically tested, including compounds explicitly referred to as MMP-12 inhibitors, but so far also without any conclusive clinical success. At a closer Look here, too, the previously described as selective inhibitors have been found to be not so selective.
So wird für die klinische Testverbindung "MMP408" als ΜΜΡ-12-Inhibitor eine gewisse bis deutliche Selektivität in vitro gegenüber MMP-13, MMP-3, MMP-14, MMP-9, Agg-1, MMP-1, Agg-2, MMP-7 und TACE beschrieben [A Selective Matrix Metalloprotease 12 Inhibitor for Potential Treatment of Chronic Obstructive Pulmonary Disease (COPD): Discovery of (S)-2-(8-(Methoxy- carbonylamino)dibenzo[b,d]furan-3-sulfonamido)-3-methylbutanoic acid (MMP408), Li et al., J. Med. Chem. 52, 1799-1802 (2009)] . In vitro -Wirkdaten zu MMP-2 und MMP-8 deuten auf eine weniger vorteilhafte Selektivität gegenüber diesen beiden MMP-Vertretern hin [Matrix metallo- proteinase-12 is a therapeutic targetfor asthma in children and young adults, Mukhopadhyay et al., J. Allergy Clin. Immunol. 126, 70-76 (2010)] . Thus, for the clinical test compound "MMP408" as a ΜΜΡ-12 inhibitor, a certain to marked selectivity in vitro over MMP-13, MMP-3, MMP-14, MMP-9, Agg-1, MMP-1, Agg-2 , MMP-7 and TACE [A Selective Matrix Metalloprotease 12 Inhibitor for Potential Treatment of Chronic Obstructive Pulmonary Disease (COPD): Discovery of (S) -2- (8- (methoxycarbonylamino) dibenzo [b, d] furan- 3-sulfonamido) -3-methylbutanoic acid (MMP408), Li et al., J. Med. Chem. 52, 1799-1802 (2009)]. In vitro micrographs of MMP-2 and MMP-8 indicate less favorable selectivity towards these two MMPs [matrix metalloproteinase-12 is a therapeutic target for asthma in children and young adults, Mukhopadhyay et al., J. Biol. Allergy Clin. Immunol. 126, 70-76 (2010)].
Ähnlich verhält es sich mit der klinischen Testsubstanz AZD1236 zur Behandlung von COPD, die als dualer MMP-9/12-Inhibitor beschrieben wird [Effects of an oral MMP-9 and -12 Inhibitor, AZD1236, on biomarkers in moderate/ 'severe COPD: A randomised controlled trial, Dahl et al., Pulm. Pharmacol. Therap. 25, 169-177 (2012)] . Die Entwicklung dieser Verbindung ist im Jahr 2012 eingestellt worden; auch hier wird eine merkliche Inhibition von MMP-2 und MMP-13 angeführt [http://www.wipo.int/research/en/details.jsp?id=2301]. The same is true for the clinical test substance AZD1236 for the treatment of COPD, which is described as a dual MMP-9/12 inhibitor. [Effects of an oral MMP-9 and -12 inhibitor, AZD1236, on biomarkers in moderate / severe COPD: A randomized controlled trial, Dahl et al., Pulm. Pharmacol. Therap. 25, 169-177 (2012)]. The development of this compound was discontinued in 2012; here, too, a marked inhibition of MMP-2 and MMP-13 is cited [http://www.wipo.int/research/en/details.jsp?id=2301].
Bei der Bewertung der MMP-Selektivität ist zudem eine vorsichtige Einschätzung der Aussagekraft von Tiermodellen angezeigt. Die Testverbindung MMP408 beispielsweise zeigt eine wesent- lieh verringerte Affinität zum orthologen MMP-12-Target der Maus: IC50 2 nM (humanes MMP- 12), IC50 160 nM (murines MMP-12), IC50 320 nm (MMP-12 der Ratte) [siehe oben Li et al., 2009; Mukhopadhyay et al., 2010]. Angaben zur Wirkstärke gegenüber anderen MMPs der Maus sind nicht publiziert. Ähnlich scheint es sich bei der Testsubstanz AZD1236 darzustellen [siehe die unter http://www.wipo. int/research/en/details.jsp?id=2301 angegebenen Informationen zur Kreuz- reaktivität bei verschiedenen Tierspezies] . When assessing MMP selectivity, a cautious assessment of the predictive power of animal models is also indicated. For example, the test compound MMP408 shows a significantly decreased affinity for the mouse orthologous MMP-12 target: IC 50 2 nM (human MMP-12), IC 50 160 nM (murine MMP-12), IC 50 320 nm (MMP-12 of the Rat) [see above Li et al., 2009; Mukhopadhyay et al., 2010]. Information on the potency against other mouse MMPs has not been published. It seems similar with the test substance AZD1236 [see http: //www.wipo. int / research / en / details.jsp? id = 2301 for cross-reactivity in various animal species].
Neben dem Selektivitätsprofil über Speziesgrenzen hinweg ist auch die Wirkstärke am Target MMP-12 selbst sehr wichtig. Bei einem vergleichsweise ähnlichen pharmakokinetischen Profil wird eine hochpotente Verbindung zu einer geringeren therapeutischen Dosis führen als eine weniger potente Verbindung, und im Allgemeinen sollte eine geringere Dosis mit einer vermin- derten Wahrscheinlichkeit von Nebenwirkungen einhergehen. Dies gilt insbesondere unter Einbeziehung der sogenannten "freien Fraktion" (fraction unbound, fu) einer Verbindung, die mit dem gewünschten Target bzw. unerwünschten anti- und off-Targets wechselwirken kann (die "freie Fraktion" ist definiert als die verfügbare Menge einer Verbindung, die nicht an Bestandteile des Blutplasmas gebunden ist; hierbei handelt es sich hauptsächlich um Bluteiweiß-Bestandteile wie z.B. Albumin). Neben der MMP-Selektivität ist also auch die Spezifität von herausragender Bedeutung. In addition to the selectivity profile across species boundaries, the potency at the target MMP-12 itself is very important. With a comparably similar pharmacokinetic profile, a highly potent compound will result in a lower therapeutic dose than a less potent compound, and generally a lower dose should be associated with a reduced likelihood of side effects. This is particularly true with the inclusion of the so-called "free fraction" (fraction unbound, f u ) of a compound which can interact with the desired target or unwanted anti and off targets (the "free fraction" is defined as the available amount of one A compound that is not bound to components of the blood plasma, which are mainly blood protein components such as eg albumin). In addition to the MMP selectivity, the specificity is therefore of paramount importance.
Neue die Makrophagen-Elastase inhibierende Wirkstoffe sollten demnach eine hohe Selektivität und Spezifität aufweisen, um in der Lage zu sein, gezielt die HME zu inhibieren. Hierzu ist auch eine gute metabolische Stabilität der Substanzen notwendig (geringe Clearance). Außerdem sollten diese Verbindungen stabil sein unter oxidativen Bedingungen, um im Krankheitsgeschehen nicht an inhibitorischer Potenz zu verlieren. Accordingly, novel macrophage elastase inhibiting agents should have high selectivity and specificity in order to be able to specifically inhibit HME. For this purpose, a good metabolic stability of the substances is necessary (low clearance). In addition, these compounds should be stable under oxidative conditions so as not to lose their inhibitory potency in disease.
Die chronisch-obstruktive Lungenerkrankung (COPD) ist eine langsam fortschreitende Lungenerkrankung, die durch eine Behinderung der Atemströmung charakterisiert ist, welche durch ein Lungenemphysem und/oder eine chronische Bronchitis hervorgerufen wird. Die ersten Symptome der Erkrankung zeigen sich in der Regel ab dem vierten bis fünften Lebensjahrzehnt. In den darauf folgenden Lebensjahren verschlimmert sich häufig die Kurzatmigkeit und es manifestiert sich Husten, verbunden mit einem ausgiebigen und stellenweise eitrigen Auswurf und einer Stenose- Atmung bis hin zu einer Atemnot (Dyspnoe). COPD ist in erster Linie eine Krankheit von Rauchern: Rauchen ist verantwortlich für 90% aller COPD-Fälle und 80-90% aller COPD-Todes- fälle. COPD ist ein großes medizinisches Problem und stellt weltweit die sechsthäufigste Todesursache dar. Von den über 45-jährigen Menschen sind ca. 4-6% betroffen. Chronic obstructive pulmonary disease (COPD) is a slowly progressive lung disease characterized by obstruction of respiratory flow, which is caused by pulmonary emphysema and / or chronic bronchitis. The first symptoms of the disease usually appear from the fourth to fifth decade of life. In the following years, the shortness of breath is often aggravated and it manifests cough, associated with an extensive and sometimes purulent sputum and a stenosis breathing to a dyspnea. COPD is primarily a disease of smokers: smoking is responsible for 90% of all COPD cases and 80-90% of all COPD deaths. COPD is a major medical problem and is the sixth most common cause of death worldwide. About 4-6% of over 45's are affected.
Obwohl die Behinderung der Atemströmung nur partiell und zeitlich befristet sein kann, ist COPD nicht heilbar. Behandlungsziel ist folglich eine Verbesserung der Lebensqualität, die Linderung der Symptome, die Verhinderung akuter Verschlechterungen und die Verlangsamung der fortschreitenden Beeinträchtigung der Lungenfunktion. Bestehende Pharmakotherapien, die sich seit den letzten zwei bis drei Jahrzehnten kaum geändert haben, sind das Verwenden von Broncho- dilatoren, um blockierte Atemwege zu öffnen, und in bestimmten Situationen Kortikosteroide, um die Entzündung der Lunge einzudämmen [Chronic Obstructive Pulmonary Disease, PJ. Barnes, N. Engl. J. Med. 343, 269-280 (2000)] . Die chronische Entzündung der Lunge, hervorgerufen durch Zigarettenrauch oder andere Reizstoffe, ist die treibende Kraft der Krankheitsentwicklung. Der zugrunde liegende Mechanismus beinhaltet Immunzellen, die im Zuge der inflammatorischen Reaktion der Lunge verschiedene Chemokine ausschütten. Hierdurch werden neutrophile Zellen und im weiteren Verlauf alveolare Makrophagen zum Lungenbindegewebe und Lumen gelockt. Neutrophile Zellen sezernieren einen Protease-Cocktail, der hauptsächlich HNE und Proteinase 3 enthält. Aktivierte Makrophagen setzen die HME frei. Hierdurch wird lokal die Protease/ Antipro- tease-Balance zu Gunsten der Proteasen verschoben, was u.a. zu einer unkontrollierten Elastase- Aktivität und in Folge hiervon zu einem überschießenden Abbau des Elastins der Alveolaren führt. Dieser Gewebeabbau verursacht einen Kollaps der Bronchien. Dies geht einher mit einer vermin- derten Elastizität der Lunge, was zu einer Behinderung der Atemströmung und beeinträchtigter Atmung führt. Darüber hinaus kann eine häufige und andauernde Entzündung der Lunge zu einem Remodeling der Bronchien und in der Folge zu einer Ausbildung von Läsionen führen. Solche Läsionen tragen zum Auftreten des chronischen Hustens bei, der eine chronische Bronchitis kenn- zeichnet. Although the obstruction of the respiratory flow can be only partially and temporally limited, COPD is not curable. The aim of the treatment is thus to improve the quality of life, alleviate the symptoms, prevent acute worsening and slow down the progressive impairment of lung function. Existing pharmacotherapies, which have barely changed over the last two to three decades, include the use of bronchodilators to open blocked airways and, in certain situations, corticosteroids to control lung inflammation [Chronic Obstructive Pulmonary Disease, PJ. Barnes, N. Engl. J. Med. 343, 269-280 (2000)]. The chronic inflammation of the lungs, caused by cigarette smoke or other irritants, is the driving force of disease development. The underlying mechanism involves immune cells that release various chemokines during the inflammatory response of the lungs. As a result, neutrophilic cells and subsequently alveolar macrophages are lured to the lung connective tissue and lumen. Neutrophils secrete a protease cocktail containing mainly HNE and proteinase 3. Activated macrophages release the HME. As a result, the protease / antiprotease balance is shifted locally in favor of the proteases, which leads inter alia to an uncontrolled elastase activity and, as a consequence thereof, to an excessive degradation of the elastin of the alveolar bodies. This tissue breakdown causes a collapse of the bronchi. This goes hand in hand with a reduction in dermal elasticity of the lung, resulting in obstruction of the respiratory flow and impaired breathing. In addition, frequent and prolonged inflammation of the lungs may lead to bronchial remodeling and, as a consequence, to the formation of lesions. Such lesions contribute to the onset of chronic cough that marks chronic bronchitis.
Aus Untersuchungen mit humanen Sputum-Proben ist bekannt, dass die Menge an HME-Protein mit dem Rauch- bzw. COPD-Status einhergeht: Die nachweisbaren HME-Mengen sind bei Nichtrauchern am niedrigsten, bei ehemaligen Rauchern und Rauchern etwas erhöht, sowie bei COPD- Patienten deutlich erhöht [Elevated MMP-12 protein levels in induced Sputum from patients with COPD, Demedts et al., Thorax 61, 196-201 (2006)] . Ähnliche Daten wurden mit humanen Sputumproben und der bronchial-alveolaren Waschflüssigkeit (bronchial alveolar washing fluid, BALF) erhoben. Hier konnte HME auf aktivierten Makrophagen nachgewiesen und quantifiziert werden: HME-Menge COPD-Patient / Raucher > COPD-Patient / ehemaliger Raucher > ehemaliger Raucher > Nichtraucher [Patterns of airway inflammation and MMP-12 expression in smokers and ex-smokers with COPD, Babusyte et al., Respir. Res. 8, 81-90 (2007)] . Studies with human sputum samples have shown that the amount of HME protein is associated with smoking or COPD status: detectable HME levels are lowest in non-smokers, slightly higher in former smokers and smokers, and in COPD - Patients significantly increased [Elevated MMP-12 protein levels in induced sputum from patients with COPD, Demedts et al., Thorax 61, 196-201 (2006)]. Similar data were collected with human sputum samples and bronchial alveolar washing fluid (BALF). Here, HME could be detected and quantified on activated macrophages: HME amount COPD patient / smoker> COPD patient / former smoker> former smoker> Non-smoker [Patterns of airway inflammation and MMP-12 expression in smokers and ex-smokers with COPD, Babusyte et al., Respir. Res. 8, 81-90 (2007)].
Eine der COPD in gewisser Weise ähnliche entzündliche Lungenerkrankung ist die interstitielle Lungenerkrankung (ILD), insbesondere hier die Ausprägung als idiopathische Lungenfibrose (idiopathic pulmonary fibrosis, IPF) und Sarkoidose [Commonalities between the pro-fibrotic mechanisms in COPD and IPF, L.A. Murray, Pulm. Pharmacol. Therap. 25, 276-280 (2012); The pathogenesis of COPD and IPF: distinct horns of the same devil?, Chilosi et al., Respir. Res. 13:3 (2012)] . Auch hier ist die Homöostase der extrazellulären Matrix gestört. Daten aus Genom- weiten Assoziationsstudien lassen eine besondere Rolle der HME im Krankheitsgeschehen solcher fibro- tischen Erkrankungen vermuten [Gene Expression Profiling Identifies MMP-12 and ADAMDEC1 as Potential Pathogenic Mediators of Pulmonary Sarcoidosis, Crouser et al., Am. J. Respir. Crit. Care Med. 179, 929-938 (2009); Association of a Functional Polymorphism in the Matrix Metallo- proteinase-12 Promoter Region with Systemic Sclerosis in an Italian Population, Manetti et al., J. Rheumatol. 37, 1852-1857 (2010); Increased serum levels and tissue expression of matrix metallo- proteinase-12 in patients with systemic sclerosis: correlation with severity of skin and pulmonary fibrosis and vascular damage, Manetti et al., Ann. Rheum. Dis. 7J_, 1064-1070 (2012)]. Darüber hinaus gibt es weitere präklinische Evidenz für eine massgebliche Rolle der HME in ischämisch-entzündlichen Krankheitsprozessen [Macrophage Metalloelastase (MMP-12) Defici- ency Mitigates Retinal Inflammation and Pathological Angiogenesis in Ischemic Retinopathy, Li et al., PLoS ONE 7 (12), e52699 (2012)] . Eine deutlich höhere MMP-12-Expression ist auch bei ischämischen Nierenverletzungen bekannt, ebenso die Beteiligung von MMP-12 bei weiteren ent- zündlichen Nierenerkrankungen [JNK signalling in human and experimental renal ischaemia/ reperfusion injury, Kanellis et al., Nephrol. Dial. Transplant. 25, 2898-2908 (2010); Macrophage Metalloelastase as a Major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis, Kaneko et al., J. Immun. 170, 3377-3385 (2003); Role for Macrophage Metallo- elastase in Glomerular Basement Membrane Damage Associated with Alport Syndrome, Rao et al., Am. J. Pathol. 169, 32-46 (2006); Differential regulation ofmetzincins in experimental chronic renal allograft rejection: Potential markers and novel therapeutic targets, Berthier et al., Kidney Int. 69, 358-368 (2006); Macrophage Infiltration and renal damage are independent of Matrix Metalloproteinase 12 (MMP-12) in the obstructed kidney, Abraham et al., Nephrology 17, 322-329 (2012)] . One of the COPD-related inflammatory lung diseases is interstitial lung disease (ILD), in particular idiopathic pulmonary fibrosis (IPF) and sarcoidosis [Commonalities between the pro-fibrotic mechanisms in COPD and IPF, LA Murray, Pulm , Pharmacol. Therap. 25, 276-280 (2012); The pathogenesis of COPD and IPF: distinct horns of the same devil ?, Chilosi et al., Respir. Res. 13: 3 (2012)]. Again, the homeostasis of the extracellular matrix is disturbed. Data from genome-wide association studies suggest a special role of HME in the disease process of such fibro- genetic diseases [Gene Expression Profiling Identifies MMP-12 and ADAMDEC1 as Potential Pathogenic Mediators of Pulmonary Sarcoidosis, Crouser et al., Am. J. Respir. Crit. Care Med. 179, 929-938 (2009); Association of a Functional Polymorphism in the Matrix Metalloproteinase-12 Promoter Region with Systemic Sclerosis in an Italian Population, Manetti et al., J. Rheumatol. 37, 1852-1857 (2010); Increased serum levels and tissue expression of matrix metalloproteinase-12 in patients with systemic sclerosis: correlation with severity of skin and pulmonary fibrosis and vascular damage, Manetti et al., Ann. Rheum. Dis. 7J, 1064-1070 (2012)]. In addition, there is further preclinical evidence for a significant role of HME in ischemic-inflammatory disease processes [Macrophage Metalloelastase (MMP-12) Deficiency Mitigates Retinal Inflammation and Pathological Angiogenesis in Ischemic Retinopathy, Li et al., PLoS ONE 7 (12)]. , e52699 (2012)]. Significantly higher MMP-12 expression is also known in ischemic kidney injuries, as is the involvement of MMP-12 in further inflammatory kidney disease [JNK signaling in human and experimental renal ischemia / reperfusion injury, Kanellis et al., Nephrol. Dial. Transplant. 25, 2898-2908 (2010); Macrophage Metalloelastase as a Major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis, Kaneko et al., J. Immun. 170, 3377-3385 (2003); Role for macrophage metallo elastase in Glomerular Basement Membrane Damage Associated with Alport Syndrome, Rao et al., Am. J. Pathol. 169, 32-46 (2006); Differential regulation of metabolites in experimental chronic renal allograft rejection: potential markers and novel therapeutic targets, Berthier et al., Kidney Int. 69, 358-368 (2006); Macrophage infiltration and renal damage are independent of matrix metalloproteinase 12 (MMP-12) in the obstructed kidney, Abraham et al., Nephrology 17, 322-329 (2012)].
Aufgabe der vorliegenden Erfindung war somit die Identifizierung und Bereitstellung neuer Substanzen, welche als potente, selektive und spezifische Inhibitoren der humanen Makrophagen- Elastase (HME / MMP-12) agieren und als solche zur Behandlung und/oder Prävention insbesondere von Erkrankungen der Atemwege, der Lunge und des Herz-Kreislauf-Systems geeignet sind. Aus den Patentanmeldungen WO 96/15096-A1, WO 97/43237-A1, WO 97/43238-A1, WO 97/ 43239-A1, WO 97/43240-A1, WO 97/43245-A1 und WO 97/43247-A1 sind 4-Aryl- und 4-Biaryl- substituierte 4-Oxobutansäure-Derivate mit inhibitorischer Aktivität gegenüber MMP-2, MMP-3, MMP-9 und, in geringerem Ausmaß, MMP-1 bekannt; aufgrund dieses Wirkprofils wurden die Verbindungen als insbesondere für die Behandlung von Osteoarthritis, rheumatoider Arthritis und Tumorerkrankungen geeignet betrachtet. In WO 98/09940-A1 und WO 99/18079-A1 wurden weitere Biarylbutansäure-Derivate als Inhibitoren von MMP-2, MMP-3 und/oder MMP-13 offenbart, die zur Behandlung verschiedenartiger Erkrankungen geeignet sind. In WO 00/40539- AI wird die Verwendung von 4-Biaryl-4-oxobutansäuren zur Behandlung von Lungen- und Atemwegserkrankungen beansprucht, basierend auf einer unterschiedlich ausgeprägten Inhibition von MMP-2, MMP-3, MMP-8, MMP-9, MMP-12 und MMP-13 durch diese Verbindungen. Ferner werden in WO 2012/014114-A1 3-Hydroxypropionsäure-Derivate und in WO 2012/038942- AI Oxy- oder Sulfonylessigsäure -Derivate als duale MMP-9/12-Inhibitoren beschrieben. The object of the present invention was therefore to identify and provide new substances which act as potent, selective and specific inhibitors of human macrophage elastase (HME / MMP-12) and as such for the treatment and / or prevention of, in particular, respiratory diseases, the Lungs and the cardiovascular system are suitable. From the patent applications WO 96/15096-A1, WO 97/43237-A1, WO 97/43238-A1, WO 97/43239-A1, WO 97/43240-A1, WO 97/43245-A1 and WO 97/43247 A1 is 4-aryl- and 4-biaryl-substituted 4-oxobutanoic acid derivatives with inhibitory activity towards MMP-2, MMP-3, MMP-9 and, to a lesser extent, MMP-1; Because of this profile of action, the compounds have been found to be particularly suitable for the treatment of osteoarthritis, rheumatoid arthritis and tumor diseases. In WO 98/09940 A1 and WO 99/18079 A1 other biarylbutanoic acid derivatives have been disclosed as inhibitors of MMP-2, MMP-3 and / or MMP-13, which are suitable for the treatment of various diseases. WO 00/40539 A1 claims the use of 4-biaryl-4-oxobutanoic acids for the treatment of pulmonary and respiratory diseases, based on a different degree of inhibition of MMP-2, MMP-3, MMP-8, MMP-9, MMP-12 and MMP-13 through these compounds. Furthermore, WO 2012/014114-A1 describes 3-hydroxypropionic acid derivatives and WO 2012/038942-A1 describes oxy- or sulfonylacetic acid derivatives as dual MMP-9/12 inhibitors.
Vor dem Hintergrund der oben beschriebenen Aufgabe zeigte es sich allerdings, dass diese MMP- Inhibitoren aus dem Stand der Technik oftmals Nachteile aufweisen, wie insbesondere eine unzu- reichende inhibitorische Potenz gegenüber MMP-12, eine ungenügende Selektivität für MMP-12 im Vergleich zu anderen MMPs und/oder eine eingeschränkte metabolische Stabilität. In view of the above-described object, however, it has been found that these prior art MMP inhibitors often have disadvantages, in particular an inadequate inhibitory potency for MMP-12, an insufficient selectivity for MMP-12 compared to others MMPs and / or limited metabolic stability.
Weitere Arylalkancarbonsäure-Derivate werden in WO 2004/092146-A2, WO 2004/099168-A2, WO 2004/099170- A2, WO 2004/099171-A2, WO 2006/050097-A1 und WO 2006/055625- A2 als Inhibitoren der Protein-Tyrosin-Phosphatase 1B (PTP-1B) zur Behandlung von Diabetes, Krebserkrankungen und neurodegenerativen Erkrankungen beschrieben. Further arylalkanecarboxylic acid derivatives are described in WO 2004/092146-A2, WO 2004/099168-A2, WO 2004/099170-A2, WO 2004/099171-A2, WO 2006/050097-A1 and WO 2006/055625-A2 Inhibitors of protein tyrosine phosphatase 1B (PTP-1B) are described for the treatment of diabetes, cancers and neurodegenerative diseases.
Es wurde nun überraschenderweise gefunden, dass bestimmte 2,5-disubstituierte Cyclopentan- carbonsäure-Derivate ein signifikant verbessertes Profil bezüglich ihrer Wirkstärke und Selektivi- tät gegenüber der humanen Makrophagen-Elastase (HME / hMMP-12) im Vergleich zu den aus dem Stand der Technik bekannten Verbindungen besitzen. Darüber hinaus zeigen die erfindungsgemäßen Verbindungen eine gute Löslichkeit in wässrigen Systemen und eine geringe unspezifische Bindung an Blutplasma-Bestandteile wie Albumin. Die erfindungsgemäßen Verbindungen weisen zudem eine niedrige in vziro-Clearance und eine gute metabolische Stabilität auf. Dieses Eigenschaftsprofil insgesamt lässt für die erfindungsgemäßen Verbindungen eine niedrige Dosier - barkeit und - als Folge der gezielteren Wirkungsweise - ein vermindertes Risiko des Auftretens von unerwünschten Nebenwirkungen in der Therapie erwarten. It has now surprisingly been found that certain 2,5-disubstituted cyclopentanecarboxylic acid derivatives have a significantly improved profile with regard to their potency and selectivity with respect to the human macrophage elastase (HME / hMMP-12) in comparison to those of the prior art Technology known compounds. In addition, the compounds of the invention show good solubility in aqueous systems and low non-specific binding to blood plasma components such as albumin. The compounds of the invention also have low in vivo clearance and good metabolic stability. This property profile as a whole makes it possible for the compounds according to the invention to have low dosability and, as a consequence of the more targeted mode of action, a reduced risk of the occurrence of undesired side effects in the therapy.
Die erfindungsgemäßen Verbindungen zeichnen sich außerdem durch eine signifikante inhibitorische Aktivität und Selektivität gegenüber den orthologen MMP-12-Peptidasen der Nagetiere aus, wie MMP-12 der Maus (auch als murine Makrophagen-Elastase, MME, bezeichnet) und MMP-12 der Ratte. Dies ermöglicht eine umfassendere präklinische Evaluierung der Substanzen in verschiedenen etablierten Tiermodellen der oben beschriebenen Erkrankungen. The compounds of the present invention are also characterized by a significant inhibitory activity and selectivity towards the rodent orthologous MMP-12 peptidases, such as mouse MMP-12 (also referred to as murine macrophage elastase, MME) and rat MMP-12. This allows a more complete preclinical evaluation of the substances in various established animal models of the diseases described above.
Gegenstand der vorliegenden Erfindung sind Verbindungen der allgemeinen Formel (I) The present invention relates to compounds of the general formula (I)
in welcher in which
A für -O- oder -S- steht, n für die Zahl 1 oder 2 steht und A is -O- or -S-, n is the number 1 or 2 and
R1 für Wasserstoff, Methyl, Fluormethyl, Difluormethyl oder Trifluormethyl steht, sowie ihre Salze, Solvate und Solvate der Salze. Erfindungsgemäße Verbindungen sind die Verbindungen der Formel (I) und deren Salze, Solvate und Solvate der Salze, die von Formel (I) umfassten Verbindungen der nachfolgend genannten Formeln und deren Salze, Solvate und Solvate der Salze sowie die von Formel (I) umfassten, nachfolgend als Ausführungsbeispiele genannten Verbindungen und deren Salze, Solvate und Solvate der Salze, soweit es sich bei den von Formel (I) umfassten, nachfolgend genannten Verbindungen nicht bereits um Salze, Solvate und Solvate der Salze handelt. R 1 is hydrogen, methyl, fluoromethyl, difluoromethyl or trifluoromethyl, and their salts, solvates and solvates of the salts. Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts comprising the compounds of the formulas below and their salts, solvates and solvates of the salts and of the formula (I) encompassed by formula (I), hereinafter referred to as exemplary compounds and their salts, solvates and solvates of the salts, as far as the compounds of formula (I), the compounds mentioned below are not already salts, solvates and solvates of the salts.
Als Salze sind im Rahmen der vorliegenden Erfindung physiologisch unbedenkliche Salze der erfindungsgemäßen Verbindungen bevorzugt. Umfasst sind auch Salze, die für pharmazeutische Anwendungen selbst nicht geeignet sind, jedoch beispielsweise für die Isolierung, Reinigung oder Lagerung der erfindungsgemäßen Verbindungen verwendet werden können. Salts used in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. Also included are salts which are not suitable for pharmaceutical applications themselves, but can be used, for example, for the isolation, purification or storage of the compounds according to the invention.
Physiologisch unbedenkliche Salze der erfindungsgemäßen Verbindungen umfassen insbesondere die von üblichen Basen abgeleiteten Salze, wie beispielhaft und vorzugsweise Alkalimetallsalze (z.B. Natrium- und Kaliumsalze), Erdalkalisalze (z.B. Calcium- und Magnesiumsalze), Zinksalze sowie Ammoniumsalze abgeleitet von Ammoniak oder organischen Aminen mit 1 bis 16 C- Atomen, wie beispielhaft und vorzugsweise Ethylamin, Diethylamin, Triethylamin, /V,/V-Diisopro- pylethylamin, Monoethanolamin, Diethanolamin, Triethanolamin, Tromethamin, Dimethylamino- ethanol, Diethylaminoethanol, Cholin, Procain, Dicyclohexylamin, Dibenzylamin, /V-Mefhylmor- pholin, /V-Mefhylpiperidin, Arginin, Lysin und 1,2-Ethylendiamin. Physiologically acceptable salts of the compounds according to the invention include, in particular, the salts derived from customary bases, such as, by way of example and by way of preference, alkali metal salts (eg sodium and potassium salts), alkaline earth salts (eg calcium and magnesium salts), zinc salts and ammonium salts derived from ammonia or organic amines having 1 to 16 C atoms, such as, by way of example and by way of preference, ethylamine, diethylamine, triethylamine, / V, / V-diisopropylethylamine, monoethanolamine, diethanolamine, triethanolamine, tromethamine, dimethylaminoethanol, diethylaminoethanol, choline, procaine, dicyclohexylamine, dibenzylamine, / V-methylmor - pholine, / V-methylpiperidine, arginine, lysine and 1,2-ethylenediamine.
Als Solvate werden im Rahmen der Erfindung solche Formen der erfindungsgemäßen Verbindun- gen bezeichnet, welche in festem oder flüssigem Zustand durch Koordination mit Lösungsmittelmolekülen einen Komplex bilden. Hydrate sind eine spezielle Form der Solvate, bei denen die Koordination mit Wasser erfolgt. Als Solvate sind im Rahmen der vorliegenden Erfindung Hydrate bevorzugt. In the context of the invention, solvates are those forms of the compounds according to the invention which form a complex in the solid or liquid state by coordination with solvent molecules. Hydrates are a special form of solvates that coordinate with water. As solvates, hydrates are preferred in the context of the present invention.
Die erfindungsgemäßen Verbindungen können in Abhängigkeit von ihrer Struktur in unterschied- liehen stereoisomeren Formen existieren, d.h. in Gestalt von Konfigurationsisomeren oder gegebenenfalls auch als Konformationsisomere (Enantiomere und/oder Diastereomere, einschließlich solcher bei Atropisomeren). Die vorliegende Erfindung umfasst deshalb die Enantiomere und Diastereomere und ihre jeweiligen Mischungen. Aus solchen Mischungen von Enantiomeren und/ oder Diastereomeren lassen sich die stereoisomer einheitlichen Bestandteile in bekannter Weise isolieren; vorzugsweise werden hierfür chromatographische Verfahren verwendet, insbesondere die HPLC-Chromatographie an achiraler bzw. chiraler Phase. Depending on their structure, the compounds of the invention may exist in different stereoisomeric forms, i. in the form of configurational isomers or optionally also as conformational isomers (enantiomers and / or diastereomers, including those in atropisomers). The present invention therefore includes the enantiomers and diastereomers and their respective mixtures. From such mixtures of enantiomers and / or diastereomers, the stereoisomerically uniform components can be isolated in a known manner; Preferably, chromatographic methods are used for this, in particular HPLC chromatography on achiral or chiral phase.
Der Begriff "enantiomerenrein" wird im Rahmen der vorliegenden Erfindung dahingehend verstanden, dass die betreffende Verbindung hinsichtlich der Absolutkonfiguration der chiralen Zentren in einem Enantiomerenüberschuss von mehr als 95%, bevorzugt von mehr als 98% vorliegt. Der Enantiomerenüberschuss (engl, enantiomeric excess, ee-Wert) wird hierbei durch Auswertung des Chromatogramms einer HPLC- Analyse an chiraler Phase nach der folgenden Formel berechnet: The term "enantiomerically pure" in the context of the present invention is understood to mean that the compound in question in terms of the absolute configuration of the chiral centers in an enantiomeric excess of more than 95%, preferably more than 98%. The enantiomeric excess (ene, ee value) is calculated here by evaluating the chromatogram of a HPLC analysis on a chiral phase according to the following formula:
Enantiomer 1 (Flächenprozent) — Enantiomer 2 (Flächenprozent) Enantiomer 1 (area%) - enantiomer 2 (area%)
ee = x 100%  ee = x 100%
Enantiomer 1 (Flächenprozent) + Enantiomer 2 (Flächenprozent) Sofern die erfindungsgemäßen Verbindungen in tautomeren Formen vorkommen können, umfasst die vorliegende Erfindung sämtliche tautomere Formen.  Enantiomer 1 (area%) + enantiomer 2 (area%) As long as the compounds of this invention can exist in tautomeric forms, the present invention encompasses all tautomeric forms.
Die vorliegende Erfindung umfasst auch alle geeigneten isotopischen Varianten der erfindungsgemäßen Verbindungen. Unter einer isotopischen Variante einer erfindungsgemäßen Verbindung wird hierbei eine Verbindung verstanden, in welcher mindestens ein Atom innerhalb der erfin- dungsgemäßen Verbindung gegen ein anderes Atom der gleichen Ordnungszahl, jedoch mit einer anderen Atommasse als der gewöhnlich oder überwiegend in der Natur vorkommenden Atommasse ausgetauscht ist. Beispiele für Isotope, die in eine erfindungsgemäße Verbindung inkorporiert werden können, sind solche von Wasserstoff, Kohlenstoff, Stickstoff, Sauerstoff, Phosphor, Schwefel, Fluor, Chlor, Brom und Iod, wie Ή (Deuterium), Ή (Tritium), 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36C1, 82Br, 123I, 124I, 129I und 131I. Bestimmte isotopische Varianten einer erfindungsgemäßen Verbindung, wie insbesondere solche, bei denen ein oder mehrere radioaktive Isotope inkorporiert sind, können von Nutzen sein beispielsweise für die Untersuchung des Wirkmechanismus oder der Wirkstoff -Verteilung im Körper; aufgrund der vergleichsweise leichten Herstell- und Detektierbarkeit sind hierfür insbesondere mit 3H- oder 14C -Isotopen markierte Verbindungen geeignet. Darüber hinaus kann der Einbau von Isotopen, wie beispielsweise von Deuterium, zu bestimmten therapeutischen Vorteilen als Folge einer größeren metabolischen Stabilität der Verbindung führen, wie beispielsweise zu einer Verlängerung der Halbwertszeit im Körper oder zu einer Reduktion der erforderlichen Wirkdosis; solche Modifikationen der erfindungsgemäßen Verbindungen können daher gegebenenfalls auch eine bevorzugte Ausführungsform der vorliegenden Erfindung darstellen. Isotopische Varianten der erfindungsgemäßen Verbindungen können nach allgemein gebräuchlichen, dem Fachmann bekannten Verfahren hergestellt werden, so beispielsweise nach den weiter unten beschriebenen Methoden und den bei den Ausführungsbeispielen wiedergegebenen Vorschriften, indem hierbei entsprechende isotopische Modifikationen der jeweiligen Reagentien und/oder Ausgangsverbindungen eingesetzt werden. Außerdem umfasst die vorliegende Erfindung auch Prodrugs der erfindungsgemäßen Verbindungen. Der Begriff "Prodrugs" bezeichnet hierbei Verbindungen, welche selbst biologisch aktiv oder inaktiv sein können, jedoch während ihrer Verweilzeit im Körper auf beispielsweise metabolischem oder hydrolytischem Wege zu erfindungsgemäßen Verbindungen umgesetzt werden. Insbesondere umfasst die vorliegende Erfindung als Prodrugs hydrolysierbare Ester-Derivate der erfindungsgemäßen Carbonsäuren der Formel (I). Hierunter werden Ester verstanden, die in physiologischen Medien, unter den Bedingungen der im weiteren beschriebenen biologischen Tests und insbesondere in vivo auf enzymatischem oder chemischem Wege zu den freien Carbonsäuren, als den biologisch hauptsächlich aktiven Verbindungen, hydrolysiert werden können. Als solche Ester werden (Ci-C -Alkylester, in welchen die Alkylgruppe geradkettig oder verzweigt sein kann, bevorzugt. Besonders bevorzugt sind Methyl-, Ethyl- oder ieri.-Butylester. The present invention also includes all suitable isotopic variants of the compounds of the invention. An isotopic variant of a compound according to the invention is understood to mean a compound in which at least one atom within the compound according to the invention is exchanged for another atom of the same atomic number but with a different atomic mass than the atomic mass that usually or predominantly occurs in nature. Examples of isotopes which can be incorporated into a compound of the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as Ή (deuterium), Ή (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 C1, 82 Br, 123 I, 124 I, 129 I and 131 I Certain isotopic variants of a compound of the invention, such as, in particular, those in which one or more radioactive isotopes are incorporated, may be useful, for example, for the study of the mechanism of action or drug distribution in the body; Due to the comparatively easy production and detectability, compounds labeled with 3 H or 14 C isotopes in particular are suitable for this purpose. In addition, the incorporation of isotopes such as deuterium may result in certain therapeutic benefits as a result of greater metabolic stability of the compound, such as prolonging the body's half-life or reducing the required effective dose; Such modifications of the compounds of the invention may therefore optionally also constitute a preferred embodiment of the present invention. Isotopic variants of the compounds according to the invention can be prepared by generally customary processes known to the person skilled in the art, for example by the methods described below and the rules reproduced in the exemplary embodiments by using corresponding isotopic modifications of the respective reagents and / or starting compounds. In addition, the present invention also includes prodrugs of the compounds of the invention. The term "prodrugs" here denotes compounds which may themselves be biologically active or inactive, but are converted during their residence time in the body by, for example, metabolic or hydrolytic routes to compounds of the invention. In particular, the present invention comprises, as prodrugs, hydrolyzable ester derivatives of the carboxylic acids of the formula (I) according to the invention. These are understood to mean esters which can be hydrolyzed in physiological media, under the conditions of the biological assays described below, and in particular in vivo enzymatically or chemically to the free carboxylic acids, as the main biologically active compounds. As such esters, preference is given to (C 1 -C -alkyl esters in which the alkyl group may be straight-chain or branched.) Particular preference is given to methyl, ethyl or ethyl-butyl esters.
Im Rahmen der vorliegenden Erfindung gilt, dass für alle Reste, die mehrfach auftreten, deren Bedeutung unabhängig voneinander ist. Wenn Reste in den erfindungsgemäßen Verbindungen sub- stituiert sind, können die Reste, soweit nicht anders spezifiziert, ein- oder mehrfach substituiert sein. Eine Substitution mit einem oder mit zwei gleichen oder verschiedenen Substituenten ist bevorzugt. Besonders bevorzugt ist die Substitution mit einem Substituenten. In the context of the present invention, the meaning is independent of each other for all radicals which occur repeatedly. If radicals are substituted in the compounds according to the invention, the radicals can, unless otherwise specified, be monosubstituted or polysubstituted. Substitution with one or two identical or different substituents is preferred. Particularly preferred is the substitution with a substituent.
Bevorzugt im Rahmen der vorliegenden Erfindung sind Verbindungen der Formel (I), in welcher Preferred in the context of the present invention are compounds of the formula (I) in which
A für -O- steht, n für die Zahl 1 oder 2 steht und A is -O-, n is the number 1 or 2 and
R1 für Wasserstoff, Methyl oder Trifluormethyl steht, sowie ihre Salze, Solvate und Solvate der Salze. R 1 is hydrogen, methyl or trifluoromethyl, and their salts, solvates and solvates of the salts.
Besonders bevorzugt im Rahmen der vorliegenden Erfindung sind Verbindungen der Formel (I), in welcher Particularly preferred in the context of the present invention are compounds of the formula (I) in which
A für -O- steht, n für die Zahl 2 steht und A is -O-, n is the number 2 and
R1 für Wasserstoff, Methyl oder Trifluormethyl steht, sowie ihre Salze, Solvate und Solvate der Salze. R 1 is hydrogen, methyl or trifluoromethyl, and their salts, solvates and solvates of the salts.
Von besonderer Bedeutung im Rahmen der vorliegenden Erfindung sind Verbindungen der Formeln (I-A) und (I-B) Of particular importance in the context of the present invention are compounds of the formulas (IA) and (IB)
worin A, n und R1 die oben definierten Bedeutungen haben und die an den zentralen Cyclopentan- Ring gebundenen Gruppen eine relative irans-Anordnung zueinander aufweisen, sowie Gemische dieser Verbindungen, wobei A, n bzw. R1 in einem solchen Gemisch von (I-A) und (I-B) jeweils identisch sind, und die Salze, Solvate und Solvate der Salze dieser Verbindungen und ihrer Gemische. Bevorzugt im Rahmen der vorliegenden Erfindung sind die Verbindungen der Formel (I-A) wherein A, n and R 1 have the meanings defined above and the groups bound to the central cyclopentane ring have a relative irans arrangement to each other, and mixtures of these compounds, wherein A, n or R 1 in such a mixture of (IA ) and (IB) are each identical, and the salts, solvates and solvates of the salts of these compounds and their mixtures. Preferred in the context of the present invention are the compounds of the formula (IA)
worin A, n und R1 die oben definierten Bedeutungen haben, in enantiomerenreiner Form mit einer, wie bezeichnet, (lS,2R,5S)-Konfiguration am zentralen Cyclopentan-Ring sowie die Salze, Solvate und Solvate der Salze dieser Verbindungen. wherein A, n and R 1 have the meanings defined above, in enantiomerically pure form with a, as designated, (lS, 2R, 5S) configuration on the central cyclopentane ring and the salts, solvates and solvates of the salts of these compounds.
Die in den jeweiligen Kombinationen bzw. bevorzugten Kombinationen von Resten im einzelnen angegebenen Reste-Definitionen werden unabhängig von den jeweiligen angegebenen Kombinationen der Reste beliebig auch durch Reste-Definitionen anderer Kombinationen ersetzt. The residue definitions given in detail in the respective combinations or preferred combinations of residues are also replaced by residue definitions of other combinations, regardless of the particular combinations of the residues indicated.
Ganz besonders bevorzugt sind Kombinationen von zwei oder mehreren der oben genannten Vorzugsbereiche. Weiterer Gegenstand der Erfindung ist ein Verfahren zur Herstellung der erfindungsgemäßen Verbindungen, dadurch gekennzeichnet, dass man eine Verbindung der Formel (II) Very particular preference is given to combinations of two or more of the abovementioned preferred ranges. The invention further provides a process for the preparation of the compounds according to the invention, which comprises reacting a compound of the formula (II)
in welcher A und R1 die oben angegebenen Bedeutungen haben, in which A and R 1 have the meanings given above,
Gegenwart einer Base mit einer Verbindung der Formel (ΠΙ) in welcher n die oben angegebene Bedeutung hat und Presence of a base with a compound of the formula (ΠΙ) in which n has the meaning given above and
X für eine Abgangsgruppe wie beispielsweise Chlor, Brom, lod, Mesylat, Triflat oder Tosylat steht, zu einer Verbindung der Formel (IV) X is a leaving group such as, for example, chlorine, bromine, iodine, mesylate, triflate or tosylate, to give a compound of the formula (IV)
in welcher n, A und R1 die oben angegebenen Bedeutungen haben, alkyliert und anschließend die 2-(Trimethylsilyl)ethyl-Estergruppe mit Hilfe einer Säure oder eines Fluorid-Reagenzes zur Carbonsäure der Formel (I) in which n, A and R 1 have the meanings given above, alkylated and then the 2- (trimethylsilyl) ethyl ester group with the aid of an acid or a fluoride reagent to the carboxylic acid of formula (I)
in welcher n, A und R1 die oben angegebenen Bedeutungen haben, abspaltet und gegebenenfalls die so erhaltenen Verbindungen der Formel (I) in ihre Enantiomere und/oder Diastereomere trennt und/oder mit den entsprechenden (i) Lösungsmitteln und/oder (ii) Basen in ihre Solvate, Salze und/oder Solvate der Salze überführt. in which n, A and R 1 have the abovementioned meanings, and if appropriate separates the compounds of the formula (I) thus obtained into their enantiomers and / or diastereomers and / or with the corresponding (i) solvents and / or (ii) Bases are converted into their solvates, salts and / or solvates of the salts.
Als Base für die Alkylierungsreaktion (II) + (ΠΙ)— (IV) sind insbesondere geeignet Alkalicarbo- nate wie Lithium-, Natrium-, Kalium- oder Cäsiumcarbonat, Alkali-Alkoholate wie Natrium- oder Kaliummethanolat, Natrium- oder Kaliumethanolat oder Natrium- oder Kalium-ieri.-butylat, Alkalihydride wie Natrium- oder Kaliumhydrid, Amid-Basen wie Lithiumdiisopropylamid oder Lithium-, Natrium- oder Kalium-bis(trimethylsilyl)amid, oder übliche metallorganische Basen wie Phenyllithium oder n-, sec- oder ieri.-Butyllithium. Bevorzugt wird Kaliumcarbonat oder Kalium- ieri.-butylat eingesetzt. Als inertes Lösungsmittel für diese Reaktion eignen sich beispielsweise Ether wie Diethylether, Diisopropylether, Methyl-ieri.-butylether, Tetrahydrofuran, 1,4-Dioxan, 1,2-Dimethoxyethan oder Bis(2-methoxyethyl)ether, Kohlenwasserstoffe wie Benzol, Toluol, Xylol, Pentan, Hexan oder Cyclohexan, oder dipolar-aprotische Lösungsmittel wie Acetonitril, Butyronitril, N,N-Dimethyl- formamid (DMF), Ν,Ν-Dimefhylacetamid (DMA), Ν,Ν'-Dimefhylpropylenharnstoff (DMPU), /V-Methylpyrrolidinon (NMP) oder Dimethylsulfoxid (DMSO). Auch Gemische solcher Lösungsmittel können eingesetzt werden. Bevorzugt wird Acetonitril oder -Dimefhylformamid (DMF) verwendet. Suitable bases for the alkylation reaction (II) + (ΠΙ) - (IV) are in particular suitable alkali metal carbonates such as lithium, sodium, potassium or cesium carbonate, alkali alcoholates such as sodium or potassium methoxide, sodium or potassium ethanolate or sodium or potassium ieri-butoxide, alkali metal hydrides such as sodium or potassium hydride, amide bases such as lithium diisopropylamide or lithium, sodium or potassium bis (trimethylsilyl) amide, or conventional organometallic bases such as phenyllithium or n-, sec- or ieri. butyllithium. Preference is given to using potassium carbonate or potassium ieri-butoxide. Suitable inert solvents for this reaction are, for example, ethers, such as diethyl ether, diisopropyl ether, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis (2-methoxyethyl) ether, hydrocarbons, such as benzene, toluene, Xylene, pentane, hexane or cyclohexane, or dipolar aprotic solvents such as acetonitrile, butyronitrile, N, N-dimethylformamide (DMF), Ν, Ν-Dimefhylacetamid (DMA), Ν, Ν'-Dimefhylpropylenharnstoff (DMPU), / V Methyl pyrrolidinone (NMP) or dimethyl sulfoxide (DMSO). It is also possible to use mixtures of such solvents. Preference is given to using acetonitrile or dimethylformamide (DMF).
Die Umsetzung (II) + (ΠΙ)— (IV) wird im Allgemeinen, je nach Reaktivität der beteiligten Komponenten, in einem Temperaturbereich von 0°C bis +120°C durchgeführt. Die Abspaltung der 2-(Trimethylsilyl)ethyl-Estergruppierung im Verfahrensschritt (IV)— (I) erfolgt nach üblichen Methoden mit Hilfe einer starken Säure, wie insbesondere Trifluoressigsäure, in einem inerten Lösungsmittel wie Dichlormethan oder mit Hilfe eines Fluorids, wie insbesondere Tetrabutylammoniumfluorid (TBAF), in einem etherischen Lösungsmittel wie Tetrahydrofuran. Die Esterspaltung wird in der Regel in einem Temperaturbereich von -20°C bis +30°C durchgeführt. The reaction (II) + (ΠΙ) - (IV) is generally carried out in a temperature range from 0 ° C to + 120 ° C, depending on the reactivity of the components involved. The cleavage of the 2- (trimethylsilyl) ethyl ester grouping in process step (IV) - (I) is carried out by customary methods with the aid of a strong acid, in particular trifluoroacetic acid, in an inert solvent such as dichloromethane or with the aid of a fluoride, in particular tetrabutylammonium fluoride ( TBAF), in an ethereal solvent such as tetrahydrofuran. The ester cleavage is usually carried out in a temperature range from -20 ° C to + 30 ° C.
Die Verbindungen der Formel (Π) können im Fall, dass A für -O- steht, dadurch hergestellt werden, dass man eine Verbindung der Formel (V) The compounds of the formula (Π), in the case where A is -O-, can be prepared by reacting a compound of the formula (V)
in welcher in which
PG für eine temporäre Schutzgruppe wie beispielsweise Benzyl steht, in Gegenwart eines Alkyl- oder Arylphosphans und eines Azodicarboxylats mit einem Triazin- 4(3//)-on-Derivat der Formel (VI) PG is a temporary protecting group such as benzyl, in the presence of an alkyl or aryl phosphine and an azodicarboxylate with a triazine-4 (3 //) -one derivative of the formula (VI)
in welcher R1 die oben angegebene Bedeutung hat, zu einer Verbindung der Formel (VII) in which R 1 has the abovementioned meaning, to give a compound of the formula (VII)
(VII), in welcher PG und R1 die oben angegebenen Bedeutungen haben, umsetzt und anschließend die Schutzgruppe PG unter Erhalt der Verbindung der Formel (II-A) (VII) in which PG and R 1 have the abovementioned meanings, and then the protective group PG to give the compound of the formula (II-A)
in welcher R1 die oben angegebene Bedeutung hat, abspaltet. in which R 1 has the meaning given above, splits off.
Die Umsetzung (V) + (VI)— (VII) wird unter den üblichen Bedingungen einer "Mitsunobu-Reak- tion" in Gegenwart eines Phosphins und eines Azodicarboxylats durchgeführt [siehe z.B. D. L. Hughes, Org. Reactions 42, 335 (1992); D. L. Hughes, Org. Prep. Proced. Int. 28 (2), 127 (1996)] . Als Phosphin-Komponente eignet sich beispielsweise Triphenylphosphin, Tri-n-butylphosphin, l,2-Bis(diphenylphosphino)ethan (DPPE), Diphenyl(2-pyridyl)phosphin, (4-Dimethylamino- phenyl)diphenylphosphin oder Tris(4-dimethylaminophenyl)phosphin. Als Azodicarboxylat kann beispielsweise Diethylazodicarboxylat (DEAD), Diisopropylazodicarboxylat (DIAD), Oi-tert.- butylazodicarboxylat, /V,/V,/VW-Tetramethylazodicarboxamid (TMAD), l,l'-(Azodicarbonyl)di- piperidin (ADDP) oder 4,7-Dimethyl-3,5,7-hexahydro-l,2,4,7-tetrazocin-3,8-dion (DHTD) eingesetzt werden. Bevorzugt wird hier Tri-n-butylphosphin in Verbindung mit Diethylazodicarboxylat (DEAD) verwendet. The reaction (V) + (VI) - (VII) is carried out under the usual conditions of a "Mitsunobu reaction" in the presence of a phosphine and an azodicarboxylate [see, e.g. D.L. Hughes, Org. Reactions 42, 335 (1992); D.L. Hughes, Org. Prep. Proced. Int. 28 (2), 127 (1996)]. Examples of suitable phosphine components are triphenylphosphine, tri-n-butylphosphine, 1,2-bis (diphenylphosphino) ethane (DPPE), diphenyl (2-pyridyl) phosphine, (4-dimethylaminophenyl) diphenylphosphine or tris (4-dimethylaminophenyl ) phosphine. As azodicarboxylate, for example, diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), Oi tert-butylazodicarboxylat, / V, / V, / VW tetramethylazodicarboxamide (TMAD), l, l '- (azodicarbonyl) di-piperidine (ADDP) or 4,7-dimethyl-3,5,7-hexahydro-l, 2,4,7-tetrazocine-3,8-dione (DHTD) can be used. Preferably here tri-n-butylphosphine is used in conjunction with diethyl azodicarboxylate (DEAD).
Inerte Lösungsmittel für diese Reaktion sind beispielsweise Ether wie Diethylether, Diisopropyl- ether, Methyl- tert. -b ty lether, Tetrahydrofuran, 1,4-Dioxan, 1,2-Dimethoxyethan oder Bis(2-meth- oxyethyl)ether, Kohlenwasserstoffe wie Benzol, Toluol, Xylol, Pentan, Hexan oder Cyclohexan, oder polar-aprotische Lösungsmittel wie Acetonitril, Butyronitril, Dimethylsulfoxid (DMSO), N,N-Dimethylformamid (DMF), N,N-Dimefhylacetamid (DMA), N,N'-Dimefhylpropylenharnstoff (DMPU) oder /V-Methylpyrrolidinon (NMP). Auch können Gemische solcher Lösungsmittel verwendet werden. Bevorzugt wird Tetrahydrofuran, Toluol oder ein Gemisch dieser beiden eingesetzt. Die Umsetzung (V) + (VI)— > (VII) erfolgt in der Regel in einem Temperaturbereich von -20°C bis +60°C, bevorzugt bei 0°C bis +40°C. Gegebenenfalls kann die Verwendung einer Mikrowellenapparatur bei dieser Reaktion von Vorteil sein. Inert solvents for this reaction are, for example, ethers, such as diethyl ether, diisopropyl ether, methyl tert. -b tyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis (2-meth- oxyethyl) ether, hydrocarbons such as benzene, toluene, xylene, pentane, hexane or cyclohexane, or polar aprotic solvents such as acetonitrile , Butyronitrile, dimethylsulfoxide (DMSO), N, N-dimethylformamide (DMF), N, N-dimethylacetamide (DMA), N, N'-dimethylpropylurea (DMPU) or / V-methylpyrrolidinone (NMP). Also, mixtures of such solvents can be used. Preference is given to using tetrahydrofuran, toluene or a mixture of these two. The reaction (V) + (VI) -> (VII) is generally carried out in a temperature range from -20 ° C to + 60 ° C, preferably at 0 ° C to + 40 ° C. Optionally, the use of a microwave apparatus in this reaction may be advantageous.
Die Abspaltung von Benzyl als temporärer Schutzgruppe PG im Verfahrensschritt (VII)— (II-A) erfolgt auf übliche Weise durch Hydrierung mit gasförmigem Wasserstoff oder, im Sinne einer Transfer-Hydrierung, mit Hilfe eines Wasserstoff-Donators wie Ammoniumformiat, Cyclohexen oder Cyclohexadien, jeweils in Gegenwart eines geeigneten Hydrierkatalysators wie insbesondere Palladium auf Aktivkohle. Die Reaktion wird vorzugsweise in einem alkoholischen Lösungsmittel wie Methanol oder Ethanol, in Ethylacetat oder Tetrahydrofuran oder in einem Gemisch solcher Lösungsmittel, gegebenenfalls unter Zusatz von Wasser, in einem Temperaturbereich von +20°C bis +80°C durchgeführt [zu möglichen alternativen Schutzgruppen sowie zur Einführung und Entfernung solcher Schutzgruppen siehe auch: T.W. Greene und P.G.M. Wuts, Protective Croups in Organic Synthesis, Wiley, New York, 1999]. The removal of benzyl as a temporary protective group PG in process step (VII) - (II-A) is carried out in the usual manner by hydrogenation with gaseous hydrogen or, in the sense of a transfer hydrogenation, with the aid of a hydrogen donor such as ammonium formate, cyclohexene or cyclohexadiene, in each case in the presence of a suitable hydrogenation catalyst such as in particular palladium on activated carbon. The reaction is preferably carried out in an alcoholic solvent such as methanol or ethanol, in ethyl acetate or tetrahydrofuran or in a mixture of such solvents, optionally with the addition of water, in a temperature range from + 20 ° C to + 80 ° C [to possible alternative protecting groups and for introduction and removal of such protecting groups see also: TW Greene and P.G.M. Wuts, Protective Croups in Organic Synthesis, Wiley, New York, 1999].
Verbindungen der Formel (Π), worin A für -S- steht, können hergestellt werden, indem man die oben beschriebene Verbindung der Formel (Π-Α) in das korrespondierende Trifluormethansulfonat der Formel (VIII) Compounds of the formula (Π) in which A is -S- can be prepared by reacting the above-described compound of the formula (Π-Α) in the corresponding trifluoromethanesulfonate of the formula (VIII)
in welcher R1 die oben angegebene Bedeutung hat, überführt und dieses dann in Gegenwart eines geeigneten Palladium-Katalysators mit einem Tri- alkylsilanthiol, beispielsweise Triisopropylsilanthiol, zur Verbindung der Formel (Π-Β) in welcher R1 die oben angegebene Bedeutung hat, umsetzt. in which R 1 has the abovementioned meaning, and then converting this in the presence of a suitable palladium catalyst with a trialkylsilanethiol, for example triisopropylsilanethiol, to give the compound of the formula (Π-Β) in which R 1 has the meaning given above, converts.
Die Herstellung des Trifluormethansulfonats (VIII) ausgehend vom Phenol (II-A) erfolgt auf üb- liehe Weise durch Umsetzung mit Trifluormethansulfonsäureanhydrid in Gegenwart einer Amin- Base wie beispielsweise /V-Diisopropylethylamin oder Pyridin. Als inertes Lösungsmittel werden im Allgemeinen chlorierte Kohlenwasserstoffe wie Dichlormethan oder Chloroform verwendet, und die Reaktion wird in der Regel in einem Temperaturbereich von -20°C bis +25 °C durchgeführt. Die weitere Überführung des Trifluormethansulfonats (VIII) in das Thiophenol (Π-Β) erfolgt durch Palladium-katalysierte Umsetzung mit einem Trialkylsilanthiol wie beispielsweise Triisopropyl- silanthiol. Als Katalysator geeignet sind zum Beispiel Palladium(II)acetat, Palladium(II)chlorid, Bis(triphenylphosphin)palladium(II)chlorid, Bis(acetonitril)palladium(II)chlorid, Tetrakis(tri- phenylphosphin)palladium(O), Bis(dibenzylidenaceton)palladium(0), Tris(dibenzylidenaceton)- dipalladium(O) oder [l,l'-Bis(diphenylphosphino)ferrocen]palladium(II)chlorid, jeweils in Kombination mit einem Phosphin-Liganden wie beispielsweise 2-Dicyclohexylphosphino-2',4',6'-triiso- propylbiphenyl (X-Phos), 2-Dicyclohexylphosphino-2',6'-dimethoxybiphenyl (S-Phos), 1,2,3,4,5- Pentaphenyl- 1 '-(di-tert. -butylphosphino)ferrocen (Q-Phos), 4,5-Bis(diphenylphosphino)-9,9-di- methylxanthen (Xantphos), 2,2'-Bis(diphenylphosphino)-l,l'-binaphthyl (BINAP), 2-Dicyclo- hexylphosphino-2'-(A',/V-dimethylamino)biphenyl oder 2-Di-ieri.-butylphosphino-2'-(/V,/V-dimethyl- amino)biphenyl. The preparation of the trifluoromethanesulfonate (VIII) starting from the phenol (II-A) takes place in a conventional manner by reaction with trifluoromethanesulfonic anhydride in the presence of an amine base such as / V-diisopropylethylamine or pyridine. As the inert solvent, chlorinated hydrocarbons such as dichloromethane or chloroform are generally used, and the reaction is usually carried out in a temperature range of -20 ° C to + 25 ° C. Further conversion of the trifluoromethanesulfonate (VIII) into the thiophenol (Π-Β) takes place by palladium-catalyzed reaction with a trialkylsilanethiol, for example triisopropylsilanethiol. Suitable catalysts are, for example, palladium (II) acetate, palladium (II) chloride, bis (triphenylphosphine) palladium (II) chloride, bis (acetonitrile) palladium (II) chloride, tetrakis (triphenylphosphine) palladium (O), Bis (dibenzylideneacetone) palladium (0), tris (dibenzylideneacetone) dipalladium (O) or [1,1'-bis (diphenylphosphino) ferrocene] palladium (II) chloride, each in combination with a phosphine ligand such as 2-dicyclohexylphosphino 2 ', 4', 6'-triisopropylbiphenyl (X-Phos), 2-dicyclohexylphosphino-2 ', 6'-dimethoxybiphenyl (S-phos), 1,2,3,4,5-pentaphenyl-1' - (di-tert-butylphosphino) ferrocene (Q-phos), 4,5-bis (diphenylphosphino) -9,9-dimethylxanthene (xantphos), 2,2'-bis (diphenylphosphino) -l, l- binaphthyl (BINAP), 2-dicyclohexylphosphino-2 '- (A', / V-dimethylamino) biphenyl or 2-di-ieri-butylphosphino-2 '- (/ V, / V-dimethylamino) biphenyl.
Die Reaktion wird in der Regel in Gegenwart einer Base durchgeführt. Als solche eignen sich Alkalicarbonate wie Natrium-, Kalium- oder Cäsiumcarbonat, Alkaliphosphate wie Natrium- oder Kaliumphosphat, Alkalifluoride wie Kalium- oder Cäsiumfluorid, Alkali-ieri.-butylate wie Natrium- oder Kalium-ieri.-butylat, tertiäre Amin-Basen wie Triethylamin, -Mefhylmorpholin, /V-Methylpiperidin, -Diisopropylefhylamin, Pyridin oder 4-Ai,/V-Dimethylaminopyridin, oder Amid-Basen wie Lithium-, Natrium- oder Kalium-bis(trimethylsilyl)amid. Die Umsetzung erfolgt in einem inerten Lösungsmittel wie beispielsweise Toluol, Xylol, 1,2-Dimethoxyethan, Tetra- hydrofuran, 1,4-Dioxan, Acetonitril, Dimethylsulfoxid (DMSO), N,N-Dimethylformamid (DMF) oder /V,/V-Dimethylacetamid (DMA) oder Mischungen hiervon in einem Temperaturbereich von +50°C bis +150°C, wobei die Verwendung einer Mikrowellenapparatur von Vorteil sein kann. The reaction is usually carried out in the presence of a base. As such are alkali metal carbonates such as sodium, potassium or cesium carbonate, alkali metal phosphates such as sodium or potassium phosphate, alkali fluorides such as potassium or cesium fluoride, alkali ieri.-butylates such as sodium or potassium ieri.-butoxide, tertiary amine bases such as triethylamine, -Mefhylmorpholin, / V-methylpiperidine, -Diisopropylefhylamin, pyridine or 4-A i, / V-dimethylaminopyridine, or Amide bases such as lithium, sodium or potassium bis (trimethylsilyl) amide. The reaction is carried out in an inert solvent such as toluene, xylene, 1,2-dimethoxyethane, tetrahydrofuran, 1,4-dioxane, acetonitrile, dimethyl sulfoxide (DMSO), N, N-dimethylformamide (DMF) or / V, / V Dimethylacetamide (DMA) or mixtures thereof in a temperature range of + 50 ° C to + 150 ° C, the use of a microwave apparatus may be advantageous.
Bevorzugt wird für die Transformation (VIII)— (II-B) ein Katalysator/Ligand/Base-System bestehend aus Tris(dibenzylidenaceton)dipalladium(0), 4,5-Bis(diphenylphosphino)-9,9-dimethyl- xanthen (Xantphos) und -Diisopropylefhylamin eingesetzt sowie 1,4-Dioxan als Lösungsmittel verwendet. Das bei dieser Umsetzung zunächst entstehende Trialkylsilylsulfid wird unter den hier verwendeten Bedingungen einer wässrigen Reaktionsaufarbeitung und chromatographischen Produktreinigung wieder gespalten, so dass direkt das freie Thiophenol (Π-Β) erhalten wird [vgl. auch M. Kreis und S. Brase, Adv. Synth. Catal. 347 (2-3), 313-319 (2005); M. S. Chambers et al, Int. Pat. Appl. WO 2006/059149-A1, Seite 9; C.-K. Pei und M. Shi, Tetrahedron: Asymmetry 22 (11), 1239-1248 (2011)]. Preferred for the transformation (VIII) - (II-B) is a catalyst / ligand / base system consisting of tris (dibenzylideneacetone) dipalladium (0), 4,5-bis (diphenylphosphino) -9,9-dimethyl-xanthene ( Xantphos) and -Diisopropylefhylamin used and 1,4-dioxane used as a solvent. The trialkylsilylsulfide initially formed in this reaction is split again under the conditions used here for aqueous reaction work-up and chromatographic product purification, so that the free thiophenol (Π-Β) is obtained directly [cf. also M. Kreis and S. Brase, Adv. Synth. Catal. 347 (2-3), 313-319 (2005); M.S. Chambers et al, Int. Pat. Appl. WO 2006/059149-A1, page 9; C.-K. Pei and M. Shi, Tetrahedron: Asymmetry 22 (11), 1239-1248 (2011)].
Die zuvor beschriebenen einzelnen Verfahrensschritte können bei normalem, bei erhöhtem oder bei erniedrigtem Druck durchgeführt werden (z.B. im Bereich von 0.5 bis 5 bar); im Allgemeinen arbeitet man jeweils bei Normaldruck. The individual process steps described above can be carried out at normal, at elevated or at reduced pressure (for example in the range from 0.5 to 5 bar); In general, one works at normal pressure.
Die Verbindungen der Formel (V) ihrerseits können in Anlehnung an publizierte Syntheseverfah- ren auf verschiedenen Wegen ausgehend von Verbindungen der Formel (IX) oder (X) The compounds of the formula (V), in turn, can be prepared in various ways, starting from compounds of the formula (IX) or (X), based on published synthesis processes.
(IX) (X),  (IX) (X),
in welchen PG die oben angegebene Bedeutung hat und Hai für ein Halogenatom steht, erhalten werden [siehe z.B. die in WO 96/15096-A1, Seite 26-44, beschriebenen allgemeinen prä- parativen Methoden, insbesondere die Methoden A, G, H und K] . wherein PG is as defined above and Hai is a halogen atom [see, e.g. the general preparative methods described in WO 96/15096-A1, pages 26-44, in particular the methods A, G, H and K].
Verbindungen der Formel (V) im Besonderen, welche eine relative trans -Anordnung der an den zentralen Cyclopentan-Ring gebundenen Gruppen aufweisen, d.h. Verbindungen der FormelnIn particular, compounds of formula (V) which have a relative trans arrangement of the groups attached to the central cyclopentane ring, i. Compounds of the formulas
(V-A) und (V-B) (VA) and (VB)
(V-A) (V-B), in welchen PG die oben angegebene Bedeutung hat, können in Analogie zu publizierten Syntheseverfahren beispielsweise dadurch hergestellt werden, dass man e o-2-(Trimefhylsilyl)efhyl 2-oxobicyclo[2.2.1]heptan-7-carboxylat der Formel (XI)  (VA) (VB), in which PG has the abovementioned meaning, can be prepared in analogy to published synthesis methods, for example by reacting e o-2- (trimethylsilyl) efhyl 2-oxobicyclo [2.2.1] heptane-7 carboxylate of the formula (XI)
mit einer Phenyl-Grignard- Verbindung der Formel (XII) with a phenyl Grignard compound of the formula (XII)
in welcher PG und Hai die oben angegebenen Bedeutungen haben, zum Addukt der Formel (XIII) in which PG and Hai have the meanings given above, to the adduct of the formula (XIII)
in welcher PG die oben angegebene Bedeutung hat, umsetzt, nachfolgend die tertiäre Hydroxygruppe über das korrespondierende Mesylat zum Olefin der Formel (XIV) in which PG has the abovementioned meaning, then reacting the tertiary hydroxy group via the corresponding mesylate to give the olefin of the formula (XIV)
in welcher PG die oben angegebene Bedeutung hat, eliminiert, dieses dann mit -Methylmorpholin- -oxid zusammen mit Osmiumtetroxid als Katalysator zum 1,2-Diol der Formel (XV) in which PG has the abovementioned meaning, it is then eliminated with methylmorpholine oxide together with osmium tetroxide as catalyst to give the 1,2-diol of the formula (XV)
pQ (XV), in welcher PG die oben angegebene Bedeutung hat, oxidiert, anschließend dieses bicyclische Diol mit Hilfe von Bleitetraacetat oder Natriumperiodat zum 2-Formyl-5-Keto-Cyclopentancarbonsäureester der Formel (XVI) pQ (XV), in which PG has the abovementioned meaning, then oxidizing this bicyclic diol with the aid of lead tetraacetate or sodium periodate to give the 2-formyl-5-keto-cyclopentanecarboxylic acid ester of the formula (XVI)
in welcher PG die oben angegebene Bedeutung hat, spaltet und schließlich mit Natriumborhydrid zur Hydroxymethyl- Verbindung der Formel (V-A) in welcher PG die oben angegebene Bedeutung hat, reduziert [vgl. WO 96/15096-A1, präparative Methode K (Seite 42-44)] . in which PG has the meaning given above, and finally with sodium borohydride to the hydroxymethyl compound of the formula (VA) in which PG has the meaning given above, reduced [cf. WO 96/15096-A1, preparative method K (pages 42-44)].
Bei der zuvor beschriebenen Synthesesequenz (XI) + (XII) -> (ΧΙΠ) -> (XIV) -> (XV) -> (XVI) — (V-A) wurde zur vereinfachten Darstellung der relativen Konfiguration der chiralen Zentren jeweils nur die Strukturformel eines Enantiomers wiedergegeben, auch wenn die betreffenden Verbindungen in racemischer Form eingesetzt bzw. erhalten wurden; tatsächliches Endprodukt eines solcherart in racemischer Form durchgeführten Herstellverfahrens ist das racemische Gemisch der Verbindungen (V-A) und (V-B). Die l,2,3-Triazin-4(3//)-on-Derivate der Formel (VI) sind auf einfache Weise durch Behandlung von oriÄo-Aminobenzamiden der Formel (XVII) In the above-described synthetic sequence (XI) + (XII) -> (ΧΙΠ) -> (XIV) → (XV) → (XVI) - (VA), in order to simplify the relative configuration of the chiral centers, only the structural formula was used an enantiomer, even if the compounds in question were used or obtained in racemic form; The actual end product of such a racemic preparation process is the racemic mixture of compounds (V-A) and (V-B). The 1,2,3-triazine-4 (3 //) -one derivatives of the formula (VI) can be prepared in a simple manner by treatment of aminoaminobenzamides of the formula (XVII)
in welcher R1 die oben angegebene Bedeutung hat, mit Natriumnitrit in wässriger Salzsäure zugänglich [siehe z.B. D. Fernandez-Forner et ah , Tetra- hedron 47 (42), 8917-8930 (1991)] . in which R 1 has the abovementioned meaning, accessible with sodium nitrite in aqueous hydrochloric acid [see, eg, D. Fernandez-Forner et al., Tetrahedron 47 (42), 8917-8930 (1991)].
Die Auftrennung von Stereoisomeren (Enantio- und/oder Diastereomeren) der erfindungsgemäßen Verbindungen der Formel (I) läßt sich nach üblichen, dem Fachmann geläufigen Methoden erreichen. Vorzugsweise werden hierfür chromatographische Verfahren an achiralen bzw. chiralen Trennphasen angewandt. Alternativ kann auch eine Trennung über diastereomere Salze der Car- bonsäuren der Formel (I) mit chiralen Amin-Basen erfolgen. Eine Trennung der erfindungsgemäßen Verbindungen in die entsprechenden Enantiomere und/ oder Diastereomere kann gegebenenfalls, je nach Zweckmäßigkeit, auch bereits auf der Stufe der Intermediate (Π), (IV), (V), (VII), (II-A), (Π-Β) oder (V-A)/(V-B) erfolgen, welche dann in separierter Form gemäß der zuvor beschriebenen Reaktionssequenz weiter umgesetzt werden. Für eine solche Auftrennung der Stereoisomere von Intermediaten werden gleichfalls bevorzugt chromatographische Verfahren an achiralen bzw. chiralen Trennphasen angewandt. The separation of stereoisomers (enantiomers and / or diastereomers) of the compounds of the formula (I) according to the invention can be achieved by customary methods known to the person skilled in the art. Preferably, chromatographic methods for achiral or chiral separation phases are used for this purpose. Alternatively, it is also possible to carry out a separation via diastereomeric salts of the carboxylic acids of the formula (I) with chiral amine bases. A separation of the compounds according to the invention into the corresponding enantiomers and / or diastereomers may, if appropriate, also be carried out at the stage of intermediates (II), (IV), (V), (VII), (II-A), (II) Π-Β) or (VA) / (VB), which are then further reacted in separated form according to the reaction sequence described above. For such a separation of the stereoisomers of intermediates, it is likewise preferred to use chromatographic methods on achiral or chiral separation phases.
Die Verbindungen der Formeln (ΙΠ), (IX), (X), (XI), (ΧΠ) und (XVII) sind entweder kommerziell erhältlich oder als solche in der Literatur beschrieben, oder sie können, ausgehend von anderen kommerziell erhältlichen Verbindungen, nach dem Fachmann geläufigen, literaturbekannten Me- thoden hergestellt werden. Zahlreiche detaillierte Vorschriften und weitere Literaturangaben befinden sich auch im Experimentellen Teil im Abschnitt zur Herstellung der Ausgangsverbindungen und Intermediate. The compounds of formulas (ΙΠ), (IX), (X), (XI), (ΧΠ) and (XVII) are either commercially available or described as such in the literature or, starting from other commercially available compounds, be prepared by the skilled worker, literature known methods. Numerous detailed regulations and further references are also contained in the Experimental Section in the section on the Preparation of Starting Compounds and Intermediates.
Die Herstellung der erfindungsgemäßen Verbindungen kann durch die folgenden Reaktionsschemata beispielhaft veranschaulicht werden: The preparation of the compounds of the invention can be exemplified by the following reaction schemes:
Schema 1 Scheme 1
(als racemisches Gemisch) Schema 2 (as a racemic mixture) Scheme 2
Die erfindungsgemäßen Verbindungen besitzen wertvolle pharmakologische Eigenschaften und können zur Vorbeugung und Behandlung von Erkrankungen bei Menschen und Tieren verwendet werden. Die erfindungsgemäßen Verbindungen stellen potente, nicht-reaktive und selektive Inhibitoren der humanen Makrophagen-Elastase (HME / hMMP-12) dar, die ein signifikant verbessertes Profil bezüglich der Wirkstärke und Selektivität im Vergleich zu den aus dem Stand der Technik bekannten Verbindungen besitzen. Darüber hinaus zeigen die erfindungsgemäßen Verbindungen eine gute Löslichkeit in wässrigen Systemen und eine geringe unspezifische Bindung an Blutplasma- Bestandteile wie Albumin. Die erfindungsgemäßen Verbindungen weisen zudem eine niedrige in vziro-Clearance und eine gute metabolische Stabilität auf. Dieses Eigenschaftsprofil insgesamt lässt für die erfindungsgemäßen Verbindungen eine niedrige Dosierbarkeit und - als Folge der gezielteren Wirkungsweise - ein vermindertes Risiko des Auftretens von unerwünschten Neben- Wirkungen in der Therapie erwarten. The compounds according to the invention have valuable pharmacological properties and can be used for the prevention and treatment of diseases in humans and animals. The compounds of the invention are potent, non-reactive and selective inhibitors of human macrophage elastase (HME / hMMP-12), which have a significantly improved profile of potency and selectivity compared to the compounds known in the art. In addition, the compounds of the invention show good solubility in aqueous systems and low non-specific binding to blood plasma components such as albumin. The compounds of the invention also have low in vivo clearance and good metabolic stability. Overall, this property profile makes it possible to expect low dosability for the compounds according to the invention and, as a result of the more targeted mode of action, a reduced risk of the occurrence of undesired side effects in the therapy.
Die erfindungsgemäßen Verbindungen eignen sich daher in besonderem Maße zur Behandlung und/oder Prävention von Erkrankungen und pathologischen Prozessen, insbesondere solcher, bei denen im Zuge eines infektiösen oder nicht-infektiösen Entzündungsgeschehens und/oder eines Gewebe- oder Gefäßumbaus die Makrophagen-Elastase (HME / hMMP-12) involviert ist. Dazu zählen im Sinne der vorliegenden Erfindung insbesondere Erkrankungen der Atemwege und der Lunge, wie die chronisch-obstruktive Lungenerkrankung (COPD), Asthma und die Gruppe der interstitiellen Lungenerkrankungen (ILD), sowie Erkrankungen des Herz-Kreislauf-Systems, wie die Arteriosklerose und Aneurysmen. The compounds according to the invention are therefore particularly suitable for the treatment and / or prevention of diseases and pathological processes, in particular those in which, in the course of an infectious or non-infectious inflammatory event and / or a tissue or vascular remodeling, the macrophage elastase (HME / hMMP-12). For the purposes of the present invention, these include, in particular, diseases of the respiratory tract and the lungs, such as chronic obstructive pulmonary disease (COPD), asthma and the group of interstitial lung diseases (ILD), and diseases of the cardiovascular system, such as arteriosclerosis and aneurysms ,
Zu den Ausprägungen der chronisch-obstruktiven Lungenerkrankung (COPD) gehören insbeson- dere das Lungenemphysem, z.B. das durch Zigarettenrauch induzierte Lungenemphysem, die chronische Bronchitis (CB), die pulmonale Hypertension in der COPD (PH-COPD), Bronchiektasie (BE) und Kombinationen hiervon, insbesondere in akut exazerbierenden Stadien der Erkrankung (AE-COPD). The manifestations of chronic obstructive pulmonary disease (COPD) include, in particular, pulmonary emphysema, e.g. Cigarette smoke-induced pulmonary emphysema, chronic bronchitis (CB), pulmonary hypertension in COPD (PH-COPD), bronchiectasis (BE) and combinations thereof, especially in acute exacerbating stages of the disease (AE-COPD).
Zu den Ausprägungen von Asthma gehören asthmatische Erkrankungen unterschiedlicher Schwe- regrade mit intermittierendem oder persistierendem Verlauf, wie refraktäres Asthma, bronchiales Asthma, allergisches Asthma, intrinsisches Asthma, extrinsisches Asthma und durch Medikamente oder Staub induziertes Asthma. Types of asthma include asthmatic diseases of varying severity with intermittent or persistent events, such as refractory asthma, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, and medication-induced or dust-induced asthma.
Zu der Gruppe der interstitiellen Lungenerkrankungen (ILD) gehören die idiopathische pulmonale Fibrose (IPF), die Lungensarkoidose und die akute interstitielle Pneumonie, nicht-spezifische interstitielle Pneumonien, lymphoide interstitielle Pneumonien, respiratorische Bronchiolitis mit interstitieller Lungenerkrankung, kryptogene organisierende Pneumonien, desquamative interstitielle Pneumonien und nicht-klassifizierbare idiopathische interstitielle Pneumonien, ferner granulo- matöse interstitielle Lungenerkrankungen, interstitielle Lungenerkrankungen bekannter Ursache und andere interstitielle Lungenerkrankungen unbekannter Ursache. The group of interstitial lung diseases (ILD) includes idiopathic pulmonary fibrosis (IPF), pulmonary sarcoidosis and acute interstitial pneumonia, non-specific interstitial pneumonia, lymphoid interstitial pneumonia, respiratory bronchiolitis with interstitial lung disease, cryptogenic organizing pneumonia, desquamative interstitial pneumonia and non-classifiable idiopathic interstitial pneumonia, granulomatous maternal interstitial lung disease, interstitial lung disease of known cause and other interstitial lung diseases of unknown cause.
Die erfindungsgemäßen Verbindungen können auch zur Behandlung und/oder Prävention von weiteren Erkrankungen der Atemwege und der Lunge verwendet werden, wie z.B. der pulmonalen arteriellen Hypertonie (PAH) und anderer Formen der pulmonalen Hypertonie (PH), des Bronchiolitis obliterans-Syndroms (BOS), des akuten Atemwegssyndroms (ARDS), der akuten Lungenschädigung (ALI), der alpha- 1-Antitrypsin-Defizienz (AATD) und der zystischen Fibrose (CF), von verschiedenen Formen der Bronchitis (chronische Bronchitis, infektiöse Bronchitis, eosinophile Bronchitis), von Bronchiektasie, Pneumonie, Farmerlunge und verwandten Krankheiten, infektiös und nicht-infektiös bedingten Husten- und Erkältungskrankheiten (chronischer entzündlicher Husten, iatrogener Husten), Nasenschleimhautentzündungen (einschließlich medikamentöse Rhinitis, vasomotorische Rhinitis und jahreszeitabhängige, allergische Rhinitis, z.B. Heuschnupfen) und von Polypen. The compounds of the invention may also be used for the treatment and / or prevention of other respiratory and pulmonary diseases, e.g. Pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH), bronchiolitis obliterans syndrome (BOS), acute respiratory tract syndrome (ARDS), acute lung injury (ALI), alpha-1-antitrypsin deficiency (AATD ) and cystic fibrosis (CF), various forms of bronchitis (chronic bronchitis, infectious bronchitis, eosinophilic bronchitis), bronchiectasis, pneumonia, farmer's lung and related diseases, infectious and non-infectious cough and cold diseases (chronic inflammatory cough, iatrogenic cough), nasal mucosal inflammation (including medicinal rhinitis, vasomotor rhinitis and seasonal, allergic rhinitis, eg hay fever) and polyps.
Zu der Gruppe der Erkrankungen des Herz-Kreislauf-Systems gehören im Sinne der vorliegenden Erfindung insbesondere die Arteriosklerose und deren Folgeerkrankungen, wie z.B. Schlaganfall bei einer Arteriosklerose der Halsarterien (karotide Arteriosklerose), Herzinfarkt bei einer Arteriosklerose der Herzkranzgefäße, periphere arterielle Verschlusskrankheit (pAVK) in Folge einer Arteriosklerose der Beinarterien, sowie Aneurysmen, insbesondere Aneurysmen der Aorta, z.B. in Folge von Arteriosklerose, Bluthochdruck, Verletzungen und Entzündungen, Infektionen (z.B. bei rheumatischem Fieber, Syphilis, Lyme-Borreliose), angeborenen Bindegewebsschwächen (z.B. beim Marfan-Syndrom und Ehlers-Danlos-Syndrom) oder als Folge einer Volumenbelastung der Aorta bei angeborenen Herzfehlern mit Rechts-Links-Shunt oder einer Shunt-abhängigen Perfusion der Lungen, sowie Aneurysmen an Herzkranzgefäßen im Zuge einer Erkrankung am Kawa- saki-Syndrom und in Hirnarealen bei Patienten mit einer angeborenen Fehlbildung der Aorten- klappe. For the purposes of the present invention, the group of diseases of the cardiovascular system includes, in particular, arteriosclerosis and its secondary diseases, such as, for example, Stroke in arteriosclerosis of the cervical arteries (carotid arteriosclerosis), myocardial infarction in arteriosclerosis of the coronary arteries, peripheral arterial occlusive disease (PAOD) due to arteriosclerosis of the leg arteries, as well as aneurysms, in particular aneurysms of the aorta, e.g. as a result of arteriosclerosis, hypertension, injuries and inflammations, infections (eg rheumatic fever, syphilis, Lyme disease), congenital connective tissue weaknesses (eg in Marfan syndrome and Ehlers-Danlos syndrome) or as a result of a volume burden of the aorta in congenital heart defects with right-left shunt or shunt-dependent perfusion of the lungs, as well as aneurysms on coronary vessels in the course of a disease in Kawasaki syndrome and in brain areas in patients with a congenital malformation of the aortic valve.
Die erfindungsgemäßen Verbindungen können darüber hinaus zur Behandlung und/oder Prävention weiterer kardiovaskulärer Erkrankungen eingesetzt werden, wie beispielsweise Bluthochdruck (Hypertonie), Herzinsuffizienz, koronare Herzerkrankung, stabile und instabile Angina pectoris, renale Hypertonie, periphere und kardiale Gefäßerkrankungen, Arrhythmien, Rhythmusstörungen der Vorhöfe und der Kammern sowie Überleitungsstörungen wie beispielsweise atrio-ventrikuläre Blockaden des Grades I-III, supraventrikuläre Tachyarrhythmie, Vorhofflimmern, Vorhofflattern, Kammerflimmern, Kammerflattern, ventrikuläre Tachyarrhythmie, Torsade de pointes-Tachykar- die, Extrasystolen des Vorhofs und des Ventrikels, AV-junktionale Extrasystolen, Sick-Sinus- Syndrom, Synkopen, AV-Knoten-Reentry-Tachykardie, Wolff-Parkinson-White-Syndrom, akutes Koronarsyndrom (ACS), autoimmune Herzerkrankungen (Perikarditis, Endokarditis, Valvolitis, Aortitis, Kardiomyopathien), Boxerkardiomyopathie, Schock wie kardiogener Schock, septischer Schock und anaphylaktischer Schock, ferner zur Behandlung und/oder Prävention von thrombo- embolischen Erkrankungen und Ischämien, wie myokardiale Ischämie, Herzhypertrophie, transito- rische und ischämische Attacken, Präeklampsie, entzündliche kardiovaskuläre Erkrankungen, Spasmen der Koronararterien und peripherer Arterien, Ödembildung wie beispielsweise pulmonales Ödem, Hirnödem, renales Ödem oder Herzinsuffizienz-bedingtes Ödem, periphere Durchblutungsstörungen, Reperfusionsschäden, arterielle und venöse Thrombosen, Mikroalbuminurie, Herzmuskelschwäche, endotheliale Dysfunktion, mikro- und makrovaskuläre Schädigungen (Vas- kulitis), sowie zur Verhinderung von Restenosen beispielsweise nach Thrombolyse-Therapien, percutan-transluminalen Angioplastien (PTA), percutan-transluminalen Koronarangioplastien (PTCA), Herztransplantationen und Bypass-Operationen. The compounds of the invention may also be used for the treatment and / or prevention of other cardiovascular diseases such as hypertension, heart failure, coronary heart disease, stable and unstable angina pectoris, renal hypertension, peripheral and cardial vascular diseases, arrhythmias, atrial arrhythmias and of the ventricles as well as conduction disorders such as atrio-ventricular blockades of grade I-III, supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, torsades de pointes tachycardia, extrasystoles of the atrium and ventricle, atrioventricular extrasystoles, Sick sinus syndrome, syncope, AV nodal reentry tachycardia, Wolff-Parkinson-White syndrome, acute Coronary syndrome (ACS), autoimmune heart disease (pericarditis, endocarditis, valvolitis, aortitis, cardiomyopathy), boxer cardiomyopathy, shock such as cardiogenic shock, septic shock and anaphylactic shock, as well as for the treatment and / or prevention of thromboembolic disorders and ischemia, such as myocardial ischemia , Cardiac hypertrophy, transitory and ischemic attacks, preeclampsia, inflammatory cardiovascular diseases, coronary and peripheral arterial spasm, edema formation such as pulmonary edema, cerebral edema, renal edema or congestive heart failure, peripheral circulatory disorders, reperfusion injury, arterial and venous thrombosis, microalbuminuria , Myocardial insufficiency, endothelial dysfunction, micro- and macrovascular damage (vasculitis), as well as for the prevention of restenosis, for example after thrombolytic therapies, percutaneous transluminal angioplasties (PTA), percutaneous transluminal cor onarangioplasties (PTCA), heart transplants and bypass surgery.
Im Sinne der vorliegenden Erfindung umfasst der Begriff Herzinsuffizienz sowohl akute als auch chronische Erscheinungsformen der Herzinsuffizienz wie auch spezifische oder verwandte Krank- heitsformen hiervon, wie akute dekompensierte Herzinsuffizienz, Rechtsherzinsuffizienz, Linksherzinsuffizienz, Globalinsuffizienz, ischämische Kardiomyopathie, dilatative Kardiomyopathie, hypertrophe Kardiomyopathie, idiopathische Kardiomyopathie, angeborene Herzfehler, Herzklappenfehler, Herzinsuffizienz bei Herzklappenfehlern, Mitralklappenstenose, Mitralklappen- insuffizienz, Aortenklappenstenose, Aortenklappeninsuffizienz, Trikuspidalstenose, Trikuspidal- Insuffizienz, Pulmonalklappenstenose, Pulmonalklappeninsuffizienz, kombinierte Herzklappenfehler, Herzmuskelentzündung (Myokarditis), chronische Myokarditis, akute Myokarditis, virale Myokarditis, diabetische Herzinsuffizienz, alkoholtoxische Kardiomyopathie, kardiale Speichererkrankungen sowie diastolische und systolische Herzinsuffizienz. For the purposes of the present invention, the term cardiac failure encompasses both acute and chronic manifestations of cardiac insufficiency as well as specific or related forms of disease thereof, such as acute decompensated heart failure, right heart failure, left heart failure, global insufficiency, ischemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart disease, heart valve defects, cardiac insufficiency in valvular heart failure, mitral stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspid stenosis, tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined valvular heart failure, myocarditis, chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic toxicity Cardiomyopathy, cardiac storage disorders as well as diastolic and systolic heart failure.
Die erfindungsgemäßen Verbindungen eignen sich außerdem zur Behandlung und/oder Prävention von Nierenerkrankungen, insbesondere von Niereninsuffizienz und Nierenversagen. Im Sinne der vorliegenden Erfindung umfassen die Begriffe Niereninsuffizienz und Nierenversagen sowohl akute als auch chronische Erscheinungsformen hiervon wie auch diesen zugrundeliegende oder verwandte Nierenerkrankungen, wie renale Hypoperfusion, intradialytische Hypotonie, obstruktive Uropathie, Glomerulopathien, Glomerulonephritis, akute Glomerulonephritis, Glomerulosklerose, tubulointerstitielle Erkrankungen, nephropathische Erkrankungen wie primäre und angeborene Nierenerkrankung, Nierenentzündung, immunologische Nierenerkrankungen wie Nierentransplantat-Abstoßung und das Alport-Syndrom, Immunkomplex-induzierte Nierenerkrankungen, durch toxische Substanzen induzierte Nephropathie, Kontrastmittel-induzierte Nephropathie, diabetische und nicht-diabetische Nephropathie, Pyelonephritis, Nierenzysten, Nephrosklerose, hypertensive Nephrosklerose und nephrotisches Syndrom, welche diagnostisch beispielsweise durch abnorm verminderte Kreatinin- und/oder Wasser-Ausscheidung, abnorm erhöhte Blutkonzentrationen von Harnstoff, Stickstoff, Kalium und/oder Kreatinin, veränderte Aktivität von Nierenenzymen wie z.B. Glutamylsynthetase, veränderte Urinosmolarität oder Urinmenge, erhöhte Mikroalbuminurie, Makroalbuminurie, Läsionen an Glomerula und Arteriolen, tubuläre Dilatation, Hyperphosphat- ämie und/oder die Notwendigkeit zur Dialyse charakterisiert werden können. Die vorliegende Erfindung umfasst auch die Verwendung der erfindungsgemäßen Verbindungen zur Behandlung und/oder Prävention von Folgeerscheinungen einer Niereninsuffizienz, wie beispielsweise Hypertonie, Lungenödem, Herzinsuffizienz, Urämie, Anämie, Elektrolytstörungen (z.B. Hyperkalämie, Hyponaträmie) und Störungen im Knochen- und Kohlenhydrat-Metabolismus. Darüber hinaus sind die erfindungsgemäßen Verbindungen zur Behandlung und/oder Prävention von Erkrankungen des Urogenitalsystems geeignet, wie beispielsweise benignes Prostata-Syndrom (BPS), benigne Prostatahyperplasie (BPH), benigne Prostatavergrößerung (BPE), Blasenentleerungsstörungen (BOO), untere Harnwegssyndrome (LUTS), neurogene überaktive Blase (OAB), Inkontinenz wie beispielsweise Misch-, Drang-, Stress- oder Überlauf-Inkontinenz (MUI, UUI, SUI, OUI), Beckenschmerzen sowie erektile Dysfunktion und weibliche sexuelle Dysfunktion. The compounds according to the invention are also suitable for the treatment and / or prevention of kidney diseases, in particular renal insufficiency and kidney failure. For the purposes of the present invention, the terms renal insufficiency and renal failure include both acute and chronic manifestations thereof as well as underlying or related renal diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial disorders, nephropathic disorders such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as renal transplant rejection and Alport syndrome, immune complex-induced kidney disease, toxic-induced nephropathy, contrast-induced nephropathy, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive Nephrosclerosis and nephrotic syndrome, which are diagnosed by, for example, abnormal decreased creatinine and / or water excretion, abnormally elevated blood levels of urea, nitrogen, potassium and / or creatinine, altered activity of renal enzymes such as glutamylsynthetase, altered urinosmolarity or urine levels, increased microalbuminuria, macroalbuminuria, glomerular and arteriolar lesions, tubular dilatation , Hyperphosphatemia and / or the need for dialysis can be characterized. The present invention also encompasses the use of the compounds of the invention for the treatment and / or prevention of sequelae of renal insufficiency, such as hypertension, pulmonary edema, heart failure, uremia, anemia, electrolyte imbalances (eg, hyperkalemia, hyponatremia) and disorders in bone and carbohydrate metabolism. Moreover, the compounds according to the invention are suitable for the treatment and / or prevention of diseases of the genitourinary system, such as benign prostatic syndrome (BPS), benign prostatic hyperplasia (BPH), benign prostatic hyperplasia (BPE), bladder emptying disorders (BOO), lower urinary tract syndromes (LUTS) , neurogenic overactive bladder (OAB), incontinence such as mixed, urgency, stress or overflow incontinence (MUI, UUI, SUI, OUI), pelvic pain, as well as erectile dysfunction and female sexual dysfunction.
Zudem besitzen die erfindungsgemäßen Verbindungen anti-inflammatorische Wirkung und können daher als entzündungshemmende Mittel zur Behandlung und/oder Prävention von Sepsis (SIRS), multiplem Organversagen (MODS, MOF), entzündlichen Erkrankungen der Niere, chronischen Darmentzündungen (IBD, Morbus Crohn, Colitis ulcerosa), Pankreatitis, Peritonitis, Cystitis, Ure- thritis, Prostatitis, Epidimytitis, Oophoritis, Salpingitis, Vulvovaginitis, rheumatoiden Erkrankungen, entzündlichen Erkrankungen des Zentralnervensystems, multipler Sklerose, entzündlichen Hauterkrankungen und entzündlichen Augenerkrankungen eingesetzt werden. In addition, the compounds of the invention have anti-inflammatory activity and can therefore be used as anti-inflammatory agents for the treatment and / or prevention of sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory diseases of the kidney, chronic intestinal inflammation (IBD, Crohn's disease, ulcerative colitis ), Pancreatitis, peritonitis, cystitis, urethritis, prostatitis, epidymitis, oophoritis, salpingitis, vulvovaginitis, rheumatoid diseases, inflammatory diseases of the central nervous system, multiple sclerosis, inflammatory skin diseases and inflammatory ocular diseases.
Die erfindungsgemäßen Verbindungen sind ferner zur Behandlung und/oder Prävention von fibro- tischen Erkrankungen der inneren Organe, wie beispielsweise der Lunge, des Herzens, der Niere, des Knochenmarks und insbesondere der Leber, sowie von dermatologischen Fibrosen und fibro- tischen Erkrankungen des Auges geeignet. Im Sinne der vorliegenden Erfindung umfasst der Begriff fibrotische Erkrankungen insbesondere solche Erkrankungen wie Leberfibrose, Leberzirrhose, Lungenfibrose, Endomyokardfibrose, Nephropathie, Glomerulonephritis, interstitielle Nieren- fibrose, fibrotische Schäden in Folge von Diabetes, Knochenmarksfibrose, Peritonealfibrose und ähnliche fibrotische Erkrankungen, Sklerodermie, Morphaea, Keloide, hypertrophe Narbenbildung, Naevi, diabetische Retinopathie, proliferative Vitroretinopathie und Erkrankungen des Bindegewebes (z.B. Sarkoidose). Die erfindungsgemäßen Verbindungen können ebenso verwendet werden zur Förderung der Wundheilung, zur Bekämpfung postoperativer Narbenbildung, z.B. nach Glaukom-Operationen, und zu kosmetischen Zwecken bei alternder oder verhornender Haut. Auch können die erfindungsgemäßen Verbindungen zur Behandlung und/oder Prävention von Anämien verwendet werden, wie hämolytischen Anämien, insbesondere Hämoglobinopathien wie Sichelzellanämie und Thalassämien, megaloblastären Anämien, Eisenmangel-Anämien, Anämien durch akuten Blutverlust, Verdrängungsanämien und aplastischen Anämien. Die erfindungsgemäßen Verbindungen sind zudem zur Behandlung von Krebserkrankungen geeignet, wie beispielsweise von Hautkrebs, Hirntumoren, Brustkrebs, Knochenmarktumoren, Leukämien, Liposarcomen, Karzinomen des Magen-Darm- Traktes, der Leber, Bauchspeicheldrüse, Lunge, Niere, Harnleiter, Prostata und des Genitaltraktes sowie von bösartigen Tumoren des lymphoproliferativen Systems, wie z.B. Hodgkin's und Non-Hodgkin's Lymphom. Darüber hinaus können die erfindungsgemäßen Verbindungen eingesetzt werden zur Behandlung und/oder Prävention von Lipidstoffwechselstörungen und Dyslipidämien (Hypolipoproteinämie, Hypertriglyceridämie, Hyperlipidämie, kombinierte Hyperlipidämien, Hypercholesterolämie, Abetalipoproteinämie, Sitosterolämie), Xanthomatose, Tangier-Krankheit, Fettsucht (Adipositas), Fettleibigkeit (Obesitas), metabolischen Erkrankungen (Metabolisches Syndrom, Hyperglykämie, Insulin-abhängiger Diabetes, nicht-Insulin-abhängiger Diabetes, Gestationsdiabetes, Hyperinsulin- ämie, Insulinresistenz, Glukose-Intoleranz und diabetische Spätfolgen wie Retinopathie, Nephropathie und Neuropathie), von Erkrankungen des Gastrointestinaltrakts und des Abdomen (Glos- sitis, Gingivitis, Periodontitis, Oesophagitis, eosinophile Gastroenteritis, Mastocytose, Morbus Crohn, Colitis, Proctitis, Pruritis ani, Diarrhöe, Zöliakie, Hepatitis, Leberfibrose, Leberzirrhose, Pankreatitis und Cholecystitis), von Erkrankungen des Zentralen Nervensystems und von neuro- degenerativen Störungen (Schlaganfall, Alzheimer'sche Krankheit, Parkinson'sche Krankheit, Demenz, Epilepsie, Depressionen, Multiple Sklerose), Immunerkrankungen, Schilddrüsenerkrankungen (Hyperthyreose), Hauterkrankungen (Psoriasis, Akne, Ekzeme, Neurodermitis, vielfältige Formen der Dermatitis wie z.B. Dermatitis abacribus, Dermatitis actinica, Dermatitis allergica, Dermatitis ammoniacalis, Dermatitis artefacta, Dermatitis autogenica, Dermatitis atrophicans, Dermatitis calorica, Dermatitis combustionis, Dermatitis congelationis, Dermatitis cosmetica, Dermatitis escharotica, Dermatitis exfoliativa, Dermatitis gangraenose, Dermatitis haemostatica, Dermatitis herpetiformis, Dermatitis lichenoides, Dermatitis linearis, Dermatitis maligna, Dermatitis medimencatosa, Dermatitis palmaris et plantaris, Dermatitis parasitaria, Dermatitis photoallergica, Dermatitis phototoxica, Dermatitis pustularis, Dermatitis seborrhoica, Dermatitis solaris, Dermatitis toxica, Dermatitis ulcerosa, Dermatitis veneata, infektiöse Dermatitis, pyogene Dermatitis und Rosazea-artige Dermatitis, sowie Keratitis, Bullosis, Vasculitis, Cellulitis, Panniculitis, Lupus erythematodes, Erythema, Lymphome, Hautkrebs, Sweet-Syndrom, Weber-Christian-Syndrom, Narbenbildung, Warzenbildung, Frostbeulen), von entzündlichen Augenerkrankungen (Saccoidosis, Blepharitis, Conjunctivitis, Iritis, Uveitis, Chorioiditis, Ophthalmitis), viralen Erkrankungen (durch Influenza-, Adeno- und Coronaviren, wie z.B. HPV, HCMV, HIV, SARS), von Erkrankungen des Skelettknochens und der Gelenke sowie der Skelettmuskel (vielfältige Formen der Arthritis wie z.B. Arthritis alcaptonurica, Arthritis ankylosans, Arthritis dysenterica, Arthritis exsudativa, Arthritis fungosa, Arthritis gonorrhoica, Arthritis mutilans, Arthritis psoriatica, Arthritis purulenta, Arthritis rheumatica, Arthritis serosa, Arthritis syphilitica, Arthritis tuberculosa, Arthritis urica, Arthritis villonodularis pigmentosa, atypische Arthritis, hämophile Arthritis, juvenile chronische Arthritis, rheumatoide Arthritis und metastatische Arthritis, des weiteren das Still-Syndrom, Felty- Syndrom, Sjörgen-Syndrom, Clutton-Syndrom, Poncet-Syndrom, Pott-Syndrom und Reiter-Syn- drom, vielfältige Formen der Arthropathien wie z.B. Arthropathie deformans, Arthropathie neuro- pathica, Arthropathie ovaripriva, Arthropathie psoriatica und Arthropathie tabica, systemische Sklerosen, vielfältige Formen der entzündlichen Myopathien wie z.B. Myopathie epidemica, Myopathie fibrosa, Myopathie myoglobinurica, Myopathie ossificans, Myopathie ossificans neurotica, Myopathie ossificans progressiva multiplex, Myopathie purulenta, Myopathie rheumatica, Myopathie trichinosa, Myopathie tropica und Myopathie typhosa, sowie das Günther-Syndrom und das Münchmeyer-Syndrom), von entzündlichen Arterienveränderungen (vielfältige Formen der Arteri- tis wie z.B. Endarteritis, Mesarteritis, Periarteritis, Panarteritis, Arteritis rheumatica, Arteritis deformans, Arteritis temporalis, Arteritis cranialis, Arteritis gigantocellularis und Arteritis granulomatosa, sowie das Horton-Syndrom, Churg-Strauss-Syndrom und die Takayasu-Arteritis), des Mückle -Well-Syndroms, der Kikuchi-Krankheit, von Polychondritis, Sklerodermia sowie von wei- teren Erkrankungen mit einer entzündlichen oder immunologischen Komponente, wie beispielsweise Katarakt, Kachexie, Osteoporose, Gicht, Inkontinenz, Lepra, Sezary-Syndrom und paraneoplastisches Syndrom, bei Abstossungsreaktionen nach Organtransplantationen und zur Wundheilung und Angiogenese insbesondere bei chronischen Wunden. The compounds according to the invention are furthermore suitable for the treatment and / or prevention of fibrous diseases of the internal organs, such as, for example, the lung, the heart, the kidney, the bone marrow and in particular the liver, as well as dermatological fibroses and fibroid diseases of the eye , For the purposes of the present invention, the term fibrotic disorders includes in particular such diseases as liver fibrosis, liver cirrhosis, pulmonary fibrosis, endomyocardial fibrosis, nephropathy, glomerulonephritis, interstitial kidney fibrosis, fibrotic damage as a result of diabetes, bone marrow fibrosis, peritoneal fibrosis and similar fibrotic disorders, scleroderma, morphea, Keloids, hypertrophic scarring, nevi, diabetic retinopathy, proliferative vitroretinopathy and connective tissue disorders (eg sarcoidosis). The compounds of the invention may also be used to promote wound healing, to combat postoperative scarring, eg, after glaucoma surgery, and for cosmetic purposes on aging or keratinizing skin. Also, the compounds of the invention may be used for the treatment and / or prevention of anemias, such as hemolytic anemias, especially hemoglobinopathies such as sickle cell anemia and thalassemias, megaloblastic anemias, iron deficiency anemias, acute blood loss anemia, crowding anaemias and aplastic anemias. The compounds of the invention are also useful in the treatment of cancers such as skin cancer, brain tumors, breast cancer, bone marrow tumors, leukemias, liposarcomas, carcinomas of the gastrointestinal tract, liver, pancreas, lung, kidney, ureter, prostate and genital tract, and of malignant tumors of the lymphoproliferative system, such as Hodgkin's and Non-Hodgkin's Lymphoma. In addition, the compounds according to the invention can be used for the treatment and / or prevention of lipid metabolism disorders and dyslipidemias (hypolipoproteinemia, hypertriglyceridemia, hyperlipidemia, combined hyperlipidemias, hypercholesterolemia, abetalipoproteinemia, sitosterolemia), xanthomatosis, Tangier's disease, obesity, obesity , metabolic disorders (metabolic syndrome, hyperglycemia, insulin-dependent diabetes, non-insulin-dependent diabetes, gestational diabetes, hyperinsulinemia, insulin resistance, glucose intolerance and diabetic sequelae such as retinopathy, nephropathy and neuropathy), diseases of the gastrointestinal tract and the abdomen (Glositis, gingivitis, periodontitis, esophagitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, colitis, proctitis, pruritis ani, diarrhea, celiac disease, hepatitis, liver fibrosis, liver cirrhosis, pancreatitis and cholecystitis), from E diseases of the central nervous system and of neurodegenerative disorders (stroke, Alzheimer's disease, Parkinson's disease, dementia, epilepsy, depression, multiple sclerosis), immune diseases, thyroid diseases (hyperthyroidism), skin diseases (psoriasis, acne, eczema, neurodermatitis, various forms of dermatitis such as dermatitis abacribus, dermatitis actinica, dermatitis allergica, ammoniacal dermatitis, dermatitis artefacta, autogenica dermatitis, dermatitis atrophicans, dermatitis calorica, dermatitis, dermatitis congelationis, dermatitis cosmetica, dermatitis escharotica, exfoliative dermatitis, gangrenous dermatitis, dermatitis haemostatica , Dermatitis herpetiformis, dermatitis lichenoides, dermatitis linearis, malignant dermatitis, median catosis, dermatitis palmaris et plantaris, dermatitis parasitaria, dermatitis photoallergica, dermatitis phototoxica, dermatitis pustularis, dermatitis seborrhoica, dermatitis sola Risks, dermatitis toxica, ulcerative colitis, dermatitis veneata, infectious dermatitis, pyogenic dermatitis and rosacea-like dermatitis, as well as keratitis, bullosis, vasculitis, cellulitis, panniculitis, lupus erythematosus, erythema, lymphoma, skin cancer, sweet syndrome, Weber-Christian Syndrome, scarring, warting, frostbite), inflammatory eye diseases (saccoidosis, blepharitis, conjunctivitis, iritis, uveitis, choroiditis, ophthalmitis), viral diseases (by Influenza, adeno and coronaviruses such as HPV, HCMV, HIV, SARS), diseases of the skeletal bone and joints, and skeletal muscle (various forms of arthritis such as arthritis alcaptonurica, arthritis ankylosans, arthritis dysenterica, arthritis exsudativa, arthritis fungosa , Arthritis gonorrhoica, arthritis mutilans, psoriatic arthritis, purulenta arthritis, arthritis rheumatica, arthritis serosa, arthritis syphilitica, arthritis tuberculosa, arthritis urica, arthritis villonodularis pigmentosa, atypical arthritis, hemophilic arthritis, juvenile chronic arthritis, rheumatoid arthritis, and metastatic arthritis Still's syndrome, Felty's syndrome, Sjörgen's syndrome, Clutton's syndrome, Poncet's syndrome, Pott syndrome and Reiter's syndrome, multiple forms of arthropathies such as arthropathy deformans, neuropathic arthropathy, ovaripriva arthropathy, psoriatic arthropathy and arthropathy tabica, systemic sclerosis, multiple forms of inflammatory myopathies such as myopathy epidemica, myopathy fibrosa, myopathy myoglobinurica, myopathy ossificans, myopathy ossificans neurotica, myopathy ossificans progressiva multiplex, myopathy purulenta, myopathy rheumatica, myopathy trichinosa, myopathy tropica and myopathy typhosa, as well as the Günther syndrome and the Münchmeyer syndrome ), inflammatory arterial changes (various forms of arteritis such as endarteritis, mesarteritis, periarteritis, panarteritis, rheumatoid arthritis, arteritis deformans, temporal arteritis, cranial arteritis, gigantocellular arteritis and granulomatous arteritis, and Horton syndrome, Churg-Strauss disease). Syndrome and Takayasu's arteritis), Mückle -Well's syndrome, Kikuchi's disease, polychondritis, sclerodermia and other diseases with an inflammatory or immunological component such as cataract, cachexia, osteoporosis, gout, incontinence, leprosy , Sezary syndrome and par Anophilic syndrome, rejection after organ transplantation and wound healing and angiogenesis especially in chronic wounds.
Aufgrund ihres Eigenschaftsprofils eignen sich die erfindungsgemäßen Verbindungen insbeson- dere zur Behandlung und/oder Prävention von Erkrankungen der Atemwege und der Lunge, vor allem der chronisch-obstruktiven Lungenerkrankung (COPD), hier insbesondere des Lungenemphysems, der chronischen Bronchitis (CB), der pulmonalen Hypertension in der COPD (PH- COPD) und von Bronchiektasie (BE) sowie von Kombinationen dieser Krankheitsformen insbesondere in akut exazerbierenden Stadien der COPD-Erkrankung (AE-COPD), des weiteren von Asthma und von interstitiellen Lungenerkrankungen, hier insbesondere der idiopathischen Lungenfibrose (IPF) und der Lungensarkoidose, von Erkrankungen des Herz-Kreislauf-Systems, insbesondere von Arteriosklerose, speziell der karotiden Arteriosklerose, sowie viraler Myokarditis, Kardiomyopathie und Aneurysmen, einschließlich deren Folgeerkrankungen wie Schlaganfall, Myokardinfarkt und periphere arterielle Verschlusskrankheit (pAVK), sowie von chronischen Nieren- erkrankungen und dem Alport-Syndrom. Die zuvor genannten, gut charakterisierten Krankheiten des Menschen können mit vergleichbarer Ätiologie auch in anderen Säugetieren vorkommen und dort ebenfalls mit den Verbindungen der vorliegenden Erfindung behandelt werden. Because of their property profile, the compounds according to the invention are particularly suitable for the treatment and / or prevention of respiratory and pulmonary diseases, especially chronic obstructive pulmonary disease (COPD), in particular pulmonary emphysema, chronic bronchitis (CB), pulmonary Hypertension in COPD (PH-COPD) and bronchiectasis (BE) as well as combinations of these diseases, especially in acute exacerbating stages of COPD disease (AE-COPD), asthma and interstitial lung diseases, in particular idiopathic pulmonary fibrosis ( IPF) and pulmonary sarcoidosis, diseases of the cardiovascular system, in particular atherosclerosis, especially carotid arteriosclerosis, as well as viral myocarditis, cardiomyopathy and aneurysms, including their sequelae such as stroke, myocardial infarction and peripheral artery disease (PAOD), as well as chronic kidneys - diseases and the Alport syndrome. The aforementioned well-characterized human diseases of similar etiology may also be present in other mammals and also be treated there with the compounds of the present invention.
Im Sinne der vorliegenden Erfindung umfasst der Begriff "Behandlung" oder "behandeln" ein Hemmen, Verzögern, Aufhalten, Lindern, Abschwächen, Einschränken, Verringern, Unterdrücken, Zurückdrängen oder Heilen einer Krankheit, eines Leidens, einer Erkrankung, einer Verletzung oder einer gesundheitlichen Störung, der Entfaltung, des Verlaufs oder des Fortschreitens solcher Zustände und/oder der Symptome solcher Zustände. Der Begriff "Therapie" wird hierbei als synonym mit dem Begriff "Behandlung" verstanden. Die Begriffe "Prävention", "Prophylaxe" oder "Vorbeugung" werden im Rahmen der vorliegenden Erfindung synonym verwendet und bezeichnen das Vermeiden oder Vermindern des Risikos, eine Krankheit, ein Leiden, eine Erkrankung, eine Verletzung oder eine gesundheitliche Störung, eine Entfaltung oder ein Fortschreiten solcher Zustände und/oder die Symptome solcher Zustände zu bekommen, zu erfahren, zu erleiden oder zu haben. Die Behandlung oder die Prävention einer Krankheit, eines Leidens, einer Erkrankung, einer Verletzung oder einer gesundheitlichen Störung können teilweise oder vollständig erfolgen. For the purposes of the present invention, the term "treatment" or "treating" includes inhibiting, delaying, arresting, alleviating, attenuating, restraining, reducing, suppressing, restraining or curing a disease, a disease, a disease, an injury or a medical condition , the unfolding, the course or progression of such conditions and / or the symptoms of such conditions. The term "therapy" is understood to be synonymous with the term "treatment". The terms "prevention", "prophylaxis" or "prevention" are used interchangeably in the context of the present invention and denote the avoidance or reduction of the risk, a disease, a disease, a disease, an injury or a health disorder, a development or a Progression of such conditions and / or to get, experience, suffer or have the symptoms of such conditions. The treatment or the prevention of a disease, a disease, a disease, an injury or a health disorder can be partial or complete.
Weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung der erfindungsgemäßen Verbindungen zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen. Weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung der erfindungsgemäßen Verbindungen zur Herstellung eines Arzneimittels zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen. Another object of the present invention is the use of the compounds of the invention for the treatment and / or prevention of diseases, in particular the aforementioned diseases. Another object of the present invention is the use of the compounds of the invention for the manufacture of a medicament for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
Weiterer Gegenstand der vorliegenden Erfindung ist ein Arzneimittel, enthaltend mindestens eine der erfindungsgemäßen Verbindungen, zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen. Another object of the present invention is a pharmaceutical composition containing at least one of the compounds of the invention, for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
Weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung der erfindungsgemäßen Verbindungen in einem Verfahren zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen. Another object of the present invention is the use of the compounds of the invention in a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
Weiterer Gegenstand der vorliegenden Erfindung ist ein Verfahren zur Behandlung und/oder Prä- vention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen, unter Verwendung einer wirksamen Menge von mindestens einer der erfindungsgemäßen Verbindungen. Die erfindungsgemäßen Verbindungen können allein oder bei Bedarf in Kombination mit einer oder mehreren anderen pharmakologisch wirksamen Substanzen eingesetzt werden, solange diese Kombination nicht zu unerwünschten und inakzeptablen Nebenwirkungen führt. Weiterer Gegenstand der vorliegenden Erfindung sind daher Arzneimittel, enthaltend mindestens eine der erfin- dungsgemäßen Verbindungen und einen oder mehrere weitere Wirkstoffe, insbesondere zur Behandlung und/oder Prävention der zuvor genannten Erkrankungen. Als hierfür geeignete Kombinationswirkstoffe seien beispielhaft und vorzugsweise genannt: Another object of the present invention is a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases, using an effective amount of at least one of the compounds of the invention. The compounds according to the invention can be used alone or as needed in combination with one or more other pharmacologically active substances, as long as this combination does not lead to undesired and unacceptable side effects. A further subject of the present invention are therefore medicaments containing at least one of the compounds according to the invention and one or more further active compounds, in particular for the treatment and / or prevention of the aforementioned diseases. Suitable combination active ingredients for this purpose are by way of example and preferably mentioned:
• anti-obstruktiv / bronchodilatorisch wirkende Mittel, wie sie z.B. zur Therapie der chronischobstruktiven Lungenerkrankung (COPD) oder eines Asthma bronchiale eingesetzt werden, bei- spielhaft und vorzugsweise aus der Gruppe der inhalativ oder systemisch angewendeten beta- adrenergen Rezeptor-Agonisten (beta-Mimetika), der inhalativ angewendeten anti-muscariner- gen Substanzen und der PDE 4-Inhibitoren; Anti-obstructive / bronchodilatory agents, e.g. for the treatment of chronic obstructive pulmonary disease (COPD) or bronchial asthma, by way of example and preferably from the group of inhaled or systemically administered beta-adrenergic receptor agonists (beta-mimetics), the inhaled anti-muscarinic substances and the PDE 4 inhibitors;
• organische Nitrate und NO-Donatoren, wie beispielsweise Natriumnitroprussid, Nitroglycerin, Isosorbidmononitrat, Isosorbiddinitrat, Molsidomin oder SIN-1, sowie inhalatives NO; · Verbindungen, die den Abbau von cyclischem Guanosinmonophosphat (cGMP) und/oder cyclischem Adenosinmonophosphat (cAMP) inhibieren, wie beispielsweise Inhibitoren der Phosphodiesterasen (PDE) 1, 2, 3, 4 und/oder 5, insbesondere PDE 4-Inhibitoren wie Roflumi- last und PDE 5-Inhibitoren wie Sildenafil, Vardenafil, Tadalafil, Udenafil, Dasantafil, Avanafil, Mirodenafil oder Lodenafil; · NO- und Häm-unabhängige Aktivatoren der löslichen Guanylatcyclase (sGC), wie insbesondere die in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 und WO 02/070510 beschriebenen Verbindungen; • organic nitrates and NO donors, such as sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO; Compounds which inhibit the degradation of cyclic guanosine monophosphate (cGMP) and / or cyclic adenosine monophosphate (cAMP), for example inhibitors of phosphodiesterases (PDE) 1, 2, 3, 4 and / or 5, in particular PDE 4 inhibitors such as roflumi last and PDE 5 inhibitors such as sildenafil, vardenafil, tadalafil, uddenafil, dasantafil, avanafil, mirodenafil or lodenafil; · NO and heme-independent activators of soluble guanylate cyclase (sGC), in particular the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510 ;
• NO-unabhängige, jedoch Häm-abhängige Stimulatoren der löslichen Guanylatcyclase (sGC), wie insbesondere Riociguat sowie die in WO 00/06568, WO 00/06569, WO 02/42301, WO 03/095451, WO 2011/147809, WO 2012/004258, WO 2012/028647 und WO 2012/059549 beschriebenen Verbindungen; NO-independent, but heme-dependent stimulators of soluble guanylate cyclase (sGC), in particular riociguat, as well as those 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;
• Verbindungen, die die humane neutrophile Elastase (HNE) inhibieren, wie insbesondere Sivele- stat, DX-890 (Reltran) sowie die in WO 2004/020410, WO 2004/020412, WO 2004/024700, WO 2004/024701, WO 2005/080372, WO 2005/082863, WO 2005/082864, WO 2009/080199, WO 2009/135599, WO 2010/078953 und WO 2010/115548 beschriebenen Verbindungen; Compounds which inhibit human neutrophil elastase (HNE), in particular Sivelastat, DX-890 (Reltran) and those described in WO 2004/020410, WO 2004/020412, WO 2004/024700, WO 2004/024701, WO 2005 / 080372, WO 2005/082863, WO 2005/082864, WO 2009/080199, WO 2009/135599, WO 2010/078953 and WO 2010/115548;
• Prostacyclin-Analoga und IP-Rezeptor-Agonisten, wie beispielhaft und vorzugsweise Iloprost, Beraprost, Treprostinil, Epoprostenol oder NS-304; Endothelin-Rezeptor-Antagonisten, wie beispielhaft und vorzugsweise Bosentan, Darusentan, Ambrisentan oder Sitaxsentan; entzündungshemmende, immunmodulierende, immunsuppressive und/oder zytotoxische Mittel, beispielhaft und vorzugsweise aus der Gruppe der systemisch oder inhalativ angewendeten Corticosteroide sowie Acetylcystein, Montelukast, Azathioprin, Cyclophosphamid, Hydroxy- carbamid, Azithromycin, IFN-γ, Pirfenidon oder Etanercept; antifibrotisch wirkende Mittel, wie beispielhaft und vorzugsweise Lysophosphatidsäure -Rezeptor 1 (LPA-l)-Antagonisten, Lysyloxidase (LOX) -Inhibitoren, Lysyloxidase-like-2-Inhibitoren, Vasoaktives intestinales Peptid (VIP), VIP-Analoga, avß6-Integrin-Antagonisten, Cholchicin, IFN-ß, D-Penicillamin, Inhibitoren des WNT-Signalwegs oder CCR2 -Antagonisten; den Fettstoffwechsel verändernde Wirkstoffe, beispielhaft und vorzugsweise aus der Gruppe der Thyroidrezeptor-Agonisten, Cholesterinsynthese-Inhibitoren wie beispielhaft und vorzugsweise HMG-CoA-Reduktase- oder Squalensynthese -Inhibitoren, der ACAT-Inhibitoren, CETP- Inhibitoren, MTP-Inhibitoren, PPAR-alpha-, PPAR-gamma- und/oder PPAR-delta-Agonisten, Cholesterin-Absorptionshemmer, Lipase-Inhibitoren, polymeren Gallensäureadsorber, Gallensäure -Reabsorptionshemmer und Lipoprotein(a)-Antagonisten; den Blutdruck senkende Wirkstoffe, beispielhaft und vorzugsweise aus der Gruppe der Calcium-Antagonisten, Angiotensin AII-Antagonisten, ACE-Hemmer, Vasopeptidase-Inhibitoren, Endothelin-Antagonisten, Renin-Inhibitoren, alpha-Rezeptoren-Blocker, beta-Rezeptoren- Blocker, Mineralocorticoid-Rezeptor- Antagonisten sowie der Diuretika; die Signaltransduktionskaskade inhibierende Verbindungen, beispielhaft und vorzugsweise aus der Gruppe der Kinase-Inhibitoren, insbesondere aus der Gruppe der Tyrosinkinase- und/oder Serin/Threoninkinase-Inhibitoren, wie beispielhaft und vorzugsweise Nintedanib, Dasatinib, Nilotinib, Bosutinib, Regorafenib, Sorafenib, Sunitinib, Cediranib, Axitinib, Telatinib, Imati- nib, Brivanib, Pazopanib, Vatalanib, Gefitinib, Erlotinib, Lapatinib, Canertinib, Lestaurtinib, Pelitinib, Semaxanib oder Tandutinib; Prostacyclin analogs and IP receptor agonists, such as by way of example and preferably iloprost, beraprost, treprostinil, epoprostenol or NS-304; Endothelin receptor antagonists, such as by way of example and preferably bosentan, darusentan, ambrisentan or sitaxsentan; anti-inflammatory, immunomodulatory, immunosuppressant and / or cytotoxic agents, by way of example and preferably from the group of systemic or inhaled corticosteroids, and acetylcysteine, montelukast, azathioprine, cyclophosphamide, hydroxycarbamide, azithromycin, IFN-γ, pirfenidone or etanercept; antifibrotic agents, such as, by way of example and by way of preference, lysophosphatidic acid receptor 1 (LPA-1) antagonists, lysyl oxidase (LOX) inhibitors, lysyl oxidase like 2 inhibitors, vasoactive intestinal peptide (VIP), VIP analogs, α v β6- Integrin antagonists, cholchicine, IFN-β, D-penicillamine, inhibitors of the WNT signaling pathway or CCR2 antagonists; lipid metabolism-altering agents, by way of example and preferably from the group of thyroid receptor agonists, cholesterol synthesis inhibitors such as by way of example and preferably HMG-CoA reductase or squalene synthesis inhibitors, ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha , PPAR gamma and / or PPAR delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors and lipoprotein (a) antagonists; hypotensive agents, by way of example and preferably from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid Receptor antagonists and diuretics; the signal transduction cascade inhibiting compounds, for example and preferably from the group of kinase inhibitors, in particular from the group of tyrosine kinase and / or serine / threonine kinase inhibitors, such as by way of example and preferably nintedanib, dasatinib, nilotinib, bosutinib, regorafenib, sorafenib, sunitinib, Cediranib, axitinib, telatinib, imatinib, brivanib, pazopanib, vatalanib, gefitinib, erlotinib, lapatinib, canertinib, lestaurtinib, pelitinib, semaxanib or tandutinib;
Verbindungen, die die Bindung von Serotonin an dessen Rezeptor blockieren, beispielhaft und vorzugsweise Antagonisten des 5 -HT2B -Rezeptors wie PRX -08066; Antagonisten von Wachstumsfaktoren, Zytokinen und Chemokinen, beispielhaft und vorzugs- weise Antagonisten von TGF-ß, CTGF, IL-1, IL-4, IL-5, IL-6, IL-8, IL-13 und Integrinen; • die Rho-Kinase inhibierende Verbindungen, wie beispielhaft und vorzugsweise Fasudil, Y-27632, SLx-2119, BF-66851, BF-66852, BF-66853, KI-23095 oder BA-1049; Compounds which block the binding of serotonin to its receptor, by way of example and preferably antagonists of the 5-HT2B receptor such as PRX-08066; Antagonists of growth factors, cytokines and chemokines, by way of example and preferably antagonists of TGF-β, CTGF, IL-1, IL-4, IL-5, IL-6, IL-8, IL-13 and integrins; The Rho kinase inhibiting compounds, such as by way of example and preferably Fasudil, Y-27632, SLx-2119, BF-66851, BF-66852, BF-66853, KI-23095 or BA-1049;
• Verbindungen, die die lösliche Epoxidhydrolase (sEH) inhibieren, wie beispielsweise N,N'-Oi- cyclohexylharnstoff, 12-(3-Adamantan-l-yl-ureido)-dodecansäure oder l-Adamantan-l-yl-3-{5- [2-(2-ethoxyethoxy)ethoxy]pentyl } -harnstoff ; Compounds which inhibit the soluble epoxide hydrolase (sEH), such as N, N'-cycloalkyloxyurea, 12- (3-adamantan-1-yl-ureido) -dodecanoic acid or 1-adamantan-1-yl-3 { 5- [2- (2-ethoxyethoxy) ethoxy] pentyl} urea;
• den Energiestoffwechsel des Herzens beeinflussende Verbindungen, wie beispielhaft und vorzugsweise Etomoxir, Dichloracetat, Ranolazin oder Trimetazidin; • the energy metabolism of the heart affecting compounds, such as by way of example and preferably etomoxir, dichloroacetate, ranolazine or trimetazidine;
• antithrombotisch wirkende Mittel, beispielhaft und vorzugsweise aus der Gruppe der Thrombozytenaggregationshemmer, der Antikoagulantien und der profibrinolytischen Substanzen; Antithrombotic agents, by way of example and preferably from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances;
• Chemotherapeutika, wie sie z.B. zur Therapie von Neubildungen (Neoplasien) der Lunge oder anderer Organe eingesetzt werden; und/oder • chemotherapeutic agents, as e.g. used for the treatment of neoplasms of the lungs or other organs; and or
• Antibiotika, insbesondere aus der Gruppe der Fluorchinoloncarbonsäuren, wie beispielhaft und vorzugsweise Ciprofloxacin oder Moxifloxacin. Antibiotics, in particular from the group of fluoroquinolonecarboxylic acids, such as by way of example and preferably ciprofloxacin or moxifloxacin.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem beta-adrenergen Rezeptor-Agonisten, wie beispielhaft und vorzugsweise Albuterol, Isoproterenol, Metaproterenol, Terbutalin, Fenoterol, Formoterol, Repro- terol, Salbutamol oder Salmeterol, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a beta-adrenergic receptor agonist such as, for example and preferably, albuterol, isoproterenol, metaproterenol, terbutaline, fenoterol, formoterol, repro sterol, salbutamol or salmeterol.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einer anti-muscarinergen Substanz, wie beispielhaft und vorzugsweise Ipratropiumbromid, Tiotropiumbromid oder Oxitropiumbromid, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an anti-muscarinergic substance, such as by way of example and preferably ipratropium bromide, tiotropium bromide or oxitropium bromide.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Corticosteroid, wie beispielhaft und vorzugsweise Prednison, Prednisolon, Methylprednisolon, Triamcinolon, Dexamethason, Beclomethason, Betamethason, Flunisolid, Budesonid oder Fluticason, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a corticosteroid, such as by way of example and preferably prednisone, prednisolone, methylprednisolone, triamcinolone, dexamethasone, beclomethasone, betamethasone, flunisolide, budesonide or fluticasone.
Unter antithrombotisch wirkenden Mittel werden vorzugsweise Verbindungen aus der Gruppe der Thrombozytenaggregationshemmer, der Antikoagulantien und der profibrinolytischen Substanzen verstanden. Antithrombotic agents are preferably understood as meaning compounds from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Thrombozytenaggregationshemmer, wie beispielhaft und vorzugsweise Aspirin, Clopidogrel, Ticlopidin oder Dipyridamol, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Thrombin-Inhibitor, wie beispielhaft und vorzugsweise Ximela- gatran, Melagatran, Dabigatran, Bivalirudin oder Clexane, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, such as, by way of example and by way of preference, aspirin, clopidogrel, ticlopidine or dipyridamole. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thrombin inhibitor, such as, by way of example and by way of preference, ximelagatran, melagatran, dabigatran, bivalirudin or Clexane.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem GPIIb/nia-Antagonisten, wie beispielhaft und vorzugsweise Tirofiban oder Abciximab, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a GPIIb / nia antagonist, such as, by way of example and by way of preference, tirofiban or abciximab.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Faktor Xa-Inhibitor, wie beispielhaft und vorzugsweise Riva- roxaban, Apixaban, Fidexaban, Razaxaban, Fondaparinux, Idraparinux, DU-176b, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR- 126512 oder SSR-128428, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are used in combination with a factor Xa inhibitor, such as by way of example and preferably rivaraban, apixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA -1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit Heparin oder einem low molecular weight (LMW)-Heparin-Derivat verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Vitamin K-Antagonisten, wie beispielhaft und vorzugsweise Coumarin, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vitamin K antagonist, such as by way of example and preferably coumarin.
Unter den Blutdruck senkenden Mitteln werden vorzugsweise Verbindungen aus der Gruppe der Calcium-Antagonisten, Angiotensin AII-Antagonisten, ACE-Hemmer, Endothelin-Antagonisten, Renin-Inhibitoren, alpha-Rezeptoren-Blocker, beta-Rezeptoren-Blocker, Mineralocorticoid-Rezep- tor-Antagonisten sowie der Diuretika verstanden. Among the antihypertensive agents are preferably compounds from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blocker, beta-receptor blocker, mineralocorticoid receptor Antagonists and diuretics understood.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Calcium- Antagonisten, wie beispielhaft und vorzugsweise Nifedipin, Amlodipin, Verapamil oder Diltiazem, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem alpha- 1 -Rezeptoren-Blocker, wie beispielhaft und vorzugsweise Prazosin, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcium antagonist, such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alpha-1-receptor blocker, such as by way of example and preferably prazosin.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem beta-Rezeptoren-Blocker, wie beispielhaft und vorzugsweise Propranolol, Atenolol, Timolol, Pindolol, Alprenolol, Oxprenolol, Penbutolol, Bupranolol, Meti- pranolol, Nadolol, Mepindolol, Carazalol, Sotalol, Metoprolol, Betaxolol, Celiprolol, Bisoprolol, Carteolol, Esmolol, Labetalol, Carvedilol, Adaprolol, Landiolol, Nebivolol, Epanolol oder Bucin- dolol, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are used in combination with a beta-receptor blocker, such as by way of example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipropanol, nadolol, mepindolol, carazalol, Sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, Carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucine dolol administered.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Angiotensin AII-Antagonisten, wie beispielhaft und vorzugs- weise Losartan, Candesartan, Valsartan, Telmisartan oder Embursatan, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an angiotensin AII antagonist, such as by way of example and preferably losartan, candesartan, valsartan, telmisartan or embursatan.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem ACE-Hemmer, wie beispielhaft und vorzugsweise Enalapril, Captopril, Lisinopril, Ramipril, Delapril, Fosinopril, Quinopril, Perindopril oder Trandopril, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Endothelin-Antagonisten, wie beispielhaft und vorzugsweise Bosentan, Darusentan, Ambrisentan oder Sitaxsentan, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACE inhibitor such as, by way of example and by way of preference, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an endothelin antagonist such as, by way of example and by way of preference, bosentan, darusentan, ambrisentan or sitaxsentan.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Renin-Inhibitor, wie beispielhaft und vorzugsweise Aliskiren, SPP-600 oder SPP-800, verabreicht. In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a renin inhibitor, such as by way of example and preferably aliskiren, SPP-600 or SPP-800.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Mineralocorticoid-Rezeptor-Antagonisten, wie beispielhaft und vorzugsweise Spironolacton, Eplerenon oder Finerenon, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist, such as by way of example and preferably spironolactone, eplerenone or finerenone.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem Diuretikum, wie beispielhaft und vorzugsweise Furosemid, Bumetanid, Torsemid, Bendroflumethiazid, Chlorthiazid, Hydrochlorthiazid, Hydroflumethiazid, Methyclothiazid, Polythiazid, Trichlormethiazid, Chlorthalidon, Indapamid, Metolazon, Quineth- azon, Acetazolamid, Dichlorphenamid, Methazolamid, Glycerin, Isosorbid, Mannitol, Amilorid oder Triamteren, verabreicht. Unter den Fettstoffwechsel verändernden Mitteln werden vorzugsweise Verbindungen aus der Gruppe der CETP-Inhibitoren, Thyroidrezeptor-Agonisten, Cholesterinsynthese-Inhibitoren wie HMG-CoA-Reduktase- oder Squalensynthese-Inhibitoren, der ACAT-Inhibitoren, MTP-Inhibi- toren, PPAR-alpha-, PPAR-gamma- und/oder PPAR-delta-Agonisten, Cholesterin-Absorptions- hemmer, polymeren Gallensäureadsorber, Gallensäure-Reabsorptionshemmer, Lipase -Inhibitoren sowie der Lipoprotein(a) -Antagonisten verstanden. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem CETP-Inhibitor, wie beispielhaft und vorzugsweise Torcetrapib (CP-529 414), JJT-705 oder CETP-vaccine (Avant), verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a diuretic, such as by way of example and preferably furosemide, bumetanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazide, polythiazide, trichloromethiazide, chlorthalidone, indapamide, metolazone, quineth- azon, acetazolamide, dichlorophenamide, methazolamide, glycerol, isosorbide, mannitol, amiloride or triamterene. Among the lipid metabolizing agents are preferably compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase or squalene synthesis inhibitors, the ACAT inhibitors, MTP inhibitors, PPAR alpha- , PPAR gamma and / or PPAR delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, lipase inhibitors and the lipoprotein (a) antagonists understood. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CETP inhibitor, such as by way of example and preferably torcetrapib (CP-529 414), JJT-705 or CETP vaccine (Avant).
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem Thyroidrezeptor-Agonisten, wie beispielhaft und vorzugsweise D-Thyroxin, 3,5,3'-Triiodothyronin (T3), CGS 23425 oder Axitirome (CGS 26214), verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thyroid receptor agonist such as, by way of example and by way of preference, D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214) ,
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem HMG-CoA-Reduktase-Inhibitor aus der Klasse der Statine, wie beispielhaft und vorzugsweise Lovastatin, Simvastatin, Pravastatin, Fluvastatin, Atorvastatin, Rosuvastatin oder Pitavastatin, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an HMG-CoA reductase inhibitor from the class of statins, such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Squalensynthese-Inhibitor, wie beispielhaft und vorzugsweise BMS-188494 oder TAK-475, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, such as by way of example and preferably BMS-188494 or TAK-475.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem ACAT-Inhibitor, wie beispielhaft und vorzugsweise Avasimibe, Melinamide, Pactimibe, Eflucimibe oder SMP-797, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACAT inhibitor, such as, for example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem MTP-Inhibitor, wie beispielhaft und vorzugsweise Implitapide, BMS-201038, R-103757 oder JTT-130, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem PPAR-gamma-Agonisten, wie beispielhaft und vorzugsweise Pioglitazone oder Rosiglitazone, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an MTP inhibitor such as, for example and preferably, implitapide, BMS-201038, R-103757 or JTT-130. In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a PPAR-gamma agonist such as, by way of example and by way of preference, pioglitazone or rosiglitazone.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem PPAR-delta-Agonisten, wie beispielhaft und vorzugsweise GW 501516 oder BAY 68-5042, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR delta agonist, such as by way of example and preferably GW 501516 or BAY 68-5042.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Cholesterin- Absorptionshemmer, wie beispielhaft und vorzugsweise Ezetimibe, Tiqueside oder Pamaqueside, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem Lipase-Inhibitor, wie beispielhaft und vorzugsweise Orlistat, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem polymeren Gallensäureadsorber, wie beispielhaft und vorzugsweise Cholestyramin, Colestipol, Colesolvam, CholestaGel oder Colestimid, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipase inhibitor, such as, for example and preferably, orlistat. In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and by way of preference, cholestyramine, colestipol, colesolvam, cholesta gel or colestimide.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbin- düngen in Kombination mit einem Gallensäure -Reabsorptionshemmer, wie beispielhaft und vorzugsweise ASBT (= IBAT)-Inhibitoren wie z.B. AZD-7806, S-8921, AK-105, BARI- 1741, SC-435 oder SC-635, verabreicht. In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a bile acid reabsorption inhibitor such as, by way of example and preferably, ASBT (= IBAT) inhibitors such as e.g. AZD-7806, S-8921, AK-105, BARI-1741, SC-435 or SC-635.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Lipoprotein(a)-Antagonisten, wie beispielhaft und vorzugs- weise Gemcabene calcium (CI-1027) oder Nicotinsäure, verabreicht. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipoprotein (a) antagonist, such as by way of example and preferably gemcabene calcium (CI-1027) or nicotinic acid.
Besonders bevorzugt sind Kombinationen der erfindungsgemäßen Verbindungen mit einem oder mehreren weiteren Wirkstoffen ausgewählt aus der Gruppe bestehend aus Corticosteroiden, beta- adrenergen Rezeptor- Agonisten, anti-muscarinergen Substanzen, PDE 4-Inhibitoren, PDE 5-Inhibi- toren, sGC-Aktivatoren, sGC-Stimulatoren, HNE-Inhibitoren, Prostacyclin-Analoga, Endothelin- Antagonisten, Statinen, antifibrotisch wirkenden Mitteln, entzündungshemmend wirkenden Mitteln, immunmodulierend wirkenden Mitteln, immunsuppressiv wirkenden Mitteln und zytotoxisch wirkenden Mitteln. Particularly preferred are combinations of the compounds according to the invention with one or more further active compounds selected from the group consisting of corticosteroids, beta-adrenergic receptor agonists, anti-muscarcinogenic substances, PDE 4 inhibitors, PDE 5 inhibitors, sGC activators, sGC Stimulants, HNE inhibitors, prostacyclin analogs, endothelin antagonists, statins, antifibrotic agents, antiinflammatory agents, immunomodulating agents, immunosuppressive agents and cytotoxic agents.
Weiterer Gegenstand der vorliegenden Erfindung sind Arzneimittel, die mindestens eine erfindungsgemäße Verbindung, üblicherweise zusammen mit einem oder mehreren inerten, nicht- toxischen, pharmazeutisch geeigneten Hilfsstoffen enthalten, sowie deren Verwendung zu den zuvor genannten Zwecken. Another object of the present invention are pharmaceutical compositions containing at least one compound of the invention, usually together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the purposes mentioned above.
Die erfindungsgemäßen Verbindungen können systemisch und/oder lokal wirken. Zu diesem Zweck können sie auf geeignete Weise appliziert werden, wie z.B. oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otisch oder als Implan- tat oder Stent. The compounds according to the invention can act systemically and / or locally. For this purpose, they may be applied in a suitable manner, e.g. oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic, or as an implant or stent.
Für diese Applikationswege können die erfindungsgemäßen Verbindungen in geeigneten Applikationsformen verabreicht werden. For these administration routes, the compounds according to the invention can be administered in suitable administration forms.
Für die orale Applikation eignen sich nach dem Stand der Technik funktionierende, die erfindungsgemäßen Verbindungen schnell und/oder modifiziert abgebende Applikationsformen, die die erfindungsgemäßen Verbindungen in kristalliner und/oder amorphisierter und/oder gelöster Form enthalten, wie z.B. Tabletten (nicht-überzogene oder überzogene Tabletten, beispielsweise mit magensaftresistenten oder sich verzögert auflösenden oder unlöslichen Überzügen, die die Frei- setzung der erfindungsgemäßen Verbindung kontrollieren), in der Mundhöhle schnell zerfallende Tabletten oder Filme/Oblaten, Filme/Lyophilisate, Kapseln (beispielsweise Hart- oder Weichgelatinekapseln), Dragees, Granulate, Pellets, Pulver, Emulsionen, Suspensionen, Aerosole oder Lösungen. Die parenterale Applikation kann unter Umgehung eines Resorptionsschrittes geschehen (z.B. intravenös, intraarteriell, intrakardial, intraspinal oder intralumbal) oder unter Einschaltung einer Resorption (z.B. inhalativ, intramuskulär, subcutan, intracutan, percutan oder intraperitoneal). Für die parenterale Applikation eignen sich als Applikationsformen u.a. Injektions- und Infusionszubereitungen in Form von Lösungen, Suspensionen, Emulsionen, Lyophilisaten oder sterilen Pul- vern. For oral administration are according to the prior art functioning, the compounds of the invention rapidly and / or modified donating application forms containing the compounds of the invention in crystalline and / or amorphized and / or dissolved form, such as tablets (uncoated or coated Tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention), rapidly disintegrating tablets or films / wafers, films / lyophilisates, capsules (for example hard or soft gelatin capsules), dragees, granules, pellets, powders, emulsions, suspensions, aerosols or solutions. Parenteral administration can be done bypassing a resorption step (eg intravenously, intraarterially, intracardially, intraspinally or intralumbarly) or using absorption (for example by inhalation, intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). For parenteral administration, suitable application forms include injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
Für die sonstigen Applikationswege eignen sich z.B. Inhalationsarzneiformen (u.a. Pulverinhalatoren, Nebulizer, Dosieraerosole), Nasentropfen, -lösungen oder -sprays, lingual, sublingual oder buccal zu applizierende Tabletten, Filme/Oblaten oder Kapseln, Suppositorien, Ohren- oder Augenpräparationen, Vaginalkapseln, wäßrige Suspensionen (Lotionen, Schüttelmixturen), lipo- phile Suspensionen, Salben, Cremes, transdermale therapeutische Systeme (z.B. Pflaster), Milch, Pasten, Schäume, Streupuder, Implantate oder Stents. For the other routes of administration are suitable, for example Inhalation medicaments (including powder inhalers, nebulizers, metered dose aerosols), nasal drops, solutions or sprays, lingual, sublingual or buccal tablets, films / wafers or capsules, suppositories, ear or ophthalmic preparations, vaginal capsules, aqueous suspensions (lotions, shake mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (for example patches), milk, pastes, foams, scattering powders, implants or stents.
Bevorzugt sind die orale, die intrapulmonale (inhalative) und die intravenöse Applikation. Preferred are oral, intrapulmonary (inhalative) and intravenous administration.
Die erfindungsgemäßen Verbindungen können in die angeführten Applikationsformen überführt werden. Dies kann in an sich bekannter Weise durch Mischen mit inerten, nichttoxischen, pharma- zeutisch geeigneten Hilfsstoffen geschehen. Zu diesen Hilfsstoffen zählen u.a. Trägerstoffe (beispielsweise mikrokristalline Cellulose, Lactose, Mannitol), Lösungsmittel (z.B. flüssige Poly- ethylenglycole), Emulgatoren und Dispergier- oder Netzmittel (beispielsweise Natriumdodecyl- sulfat, Polyoxysorbitanoleat), Bindemittel (beispielsweise Polyvinylpyrrolidon), synthetische und natürliche Polymere (beispielsweise Albumin), Stabilisatoren (z.B. Antioxidantien wie beispiels- weise Ascorbinsäure), Farbstoffe (z.B. anorganische Pigmente wie beispielsweise Eisenoxide) und Geschmacks- und/oder Geruchskorrigentien. The compounds according to the invention can be converted into the stated administration forms. This can be done in a conventional manner by mixing with inert, non-toxic, pharmaceutically suitable excipients. These adjuvants include, among others. Excipients (for example microcrystalline cellulose, lactose, mannitol), solvents (for example liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecyl sulfate, polyoxysorbitanoleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), Stabilizers (eg antioxidants such as ascorbic acid), dyes (eg inorganic pigments such as iron oxides) and flavor and / or odoriferous.
Im Allgemeinen hat es sich als vorteilhaft erwiesen, bei parenteraler Applikation Mengen von etwa 0.001 bis 1 mg kg, vorzugsweise etwa 0.01 bis 0.5 mg kg Körpergewicht zur Erzielung wirksamer Ergebnisse zu verabreichen. Bei oraler Applikation beträgt die Dosierung etwa 0.01 bis 100 mg kg, vorzugsweise etwa 0.01 bis 20 mg/kg und ganz besonders bevorzugt 0.1 bis 10 mg/kg Körpergewicht. Bei intrapulmonaler Applikation beträgt die Menge im Allgemeinen etwa 0.1 bis 50 mg je Inhalation. Trotzdem kann es gegebenenfalls erforderlich sein, von den genannten Mengen abzuweichen, und zwar in Abhängigkeit von Körpergewicht, Applikationsweg, individuellem Verhalten gegenüber dem Wirkstoff, Art der Zubereitung und Zeitpunkt bzw. Intervall, zu welchem die Applikation erfolgt. So kann es in einigen Fällen ausreichend sein, mit weniger als der vorgenannten Mindestmenge auszukommen, während in anderen Fällen die genannte obere Grenze überschritten werden muss. Im Falle der Applikation größerer Mengen kann es empfehlenswert sein, diese in mehreren Einzelgaben über den Tag zu verteilen. In general, it has proven to be advantageous, when administered parenterally, to administer amounts of about 0.001 to 1 mg kg, preferably about 0.01 to 0.5 mg kg body weight, in order to achieve effective results. When administered orally, the dosage is about 0.01 to 100 mg kg, preferably about 0.01 to 20 mg / kg and most preferably 0.1 to 10 mg / kg body weight. For intrapulmonary administration, the amount is generally about 0.1 to 50 mg per inhalation. Nevertheless, it may be necessary to deviate from the stated amounts, depending on body weight, route of administration, individual behavior towards the active ingredient, type of preparation and time or interval at which the application is carried out. Thus, in some cases it may be sufficient to manage with less than the aforementioned minimum amount, while in other cases, the said upper limit must be exceeded. In the case of the application of larger quantities, it may be advisable to distribute these in several single doses throughout the day.
Die nachfolgenden Ausführungsbeispiele erläutern die Erfindung. Die Erfindung ist nicht auf die Beispiele beschränkt. The following embodiments illustrate the invention. The invention is not limited to the examples.
Beispiele Examples
Abkürzungen und Akronyme: abs. absolut  Abbreviations and acronyms: abs. absolutely
Ac Acetyl  Ac acetyl
aq. wässrig, wässrige Lösung aq. aqueous, aqueous solution
br. breit (bei NMR-Signal) br. wide (at NMR signal)
Bsp. Beispiel  Example. Example
Bu Butyl  Bu Butyl
c Konzentration c concentration
ca. circa, ungefähr about circa, about
cat. katalytisch cat. catalytic
CI chemische Ionisation (bei MS)  CI chemical ionization (in MS)
d Dublett (bei NMR) d doublet (by NMR)
d Tag(e) d day (s)
(dba)3Pd2 Tris(dibenzylidenaceton)dipalladium(0) (dba) 3 Pd 2 tris (dibenzylideneacetone) dipalladium (0)
DC Dünnschichtchromatographie TLC thin layer chromatography
DCI direkte chemische Ionisation (bei MS) DCI direct chemical ionization (in MS)
dd Dublett von Dublett (bei NMR) dd doublet of doublet (by NMR)
DEAD Diethylazodicarboxylat  DEAD Diethyl azodicarboxylate
DMF N, -Dimefhylf ormamid DMF N, -dimethylamine
DMSO Dimethylsulfoxid DMSO dimethyl sulfoxide
dt Dublett von Triplett (bei NMR) dt doublet of triplet (by NMR)
d. Th. der Theorie (bei chemischer Ausbeute) ee Enantiomerenüberschuss d. Th. Of theory (in chemical yield) ee enantiomeric excess
EI Elektronenstoß-Ionisation (bei MS)  EI electron impact ionization (in MS)
ent enantiomerenrein, Enantiomer ent enantiomerically pure, enantiomer
eq. Äquivalent(e) eq. Equivalent (s)
ESI Elektrospray-Ionisation (bei MS)  ESI electrospray ionization (in MS)
Et Ethyl  Et ethyl
h Stunde(n) h hour (s)
HPLC Hochdruck-, Hochleistungsflüssigchromatographie  HPLC high pressure, high performance liquid chromatography
iPr Isopropyl iPr isopropyl
konz konzentriert (bei Lösung) concentrate concentrated (at solution)
LC Flüssigchromatographie  LC liquid chromatography
LC/MS Flüssigchromatographie-gekoppelte Massenspektrometrie LC / MS liquid chromatography-coupled mass spectrometry
Lit. Literatur(stelle) Literature (position)
m Multiplett (bei NMR) m multiplet (by NMR)
Me Methyl  Me methyl
min Minute(n) min minute (s)
MPLC Mitteldruckflüssigchromatographie (über Kieselgel; auch "flash- MPLC medium pressure liquid chromatography (over silica gel; also "flash"
Chromatographie" genannt) Called chromatography ")
Ms Methansulfonyl (Mesyl)  Ms methanesulfonyl (mesyl)
MS Massenspektrometrie  MS mass spectrometry
NMO N-Methylmorpholin-N-oxid  NMO N-methylmorpholine N-oxide
NMR Kernresonanzspektrometrie  NMR nuclear magnetic resonance spectrometry
Pd/C Palladium auf Aktivkohle  Pd / C palladium on charcoal
Pr Propyl  Pr Propyl
q (oder quart) Quartett (bei NMR) q (or quart) quartet (by NMR)
qd Quartett von Dublett (bei NMR) qd Quartet by Dublett (by NMR)
quant. quantitativ (bei chemischer Ausbeute) quant. quantitative (at chemical yield)
quint Quintett (bei NMR) quint quintet (by NMR)
rac racemisch, Racemat rac racemic, racemate
Rf Retentionsindex (bei DC)  Rf Retention Index (at DC)
RP reverse phase (Umkehrphase, bei HPLC)  RP reverse phase (reversed phase, for HPLC)
RT Raumtemperatur  RT room temperature
Rt Retentionszeit (bei HPLC, LC/MS) R t retention time (for HPLC, LC / MS)
s Singulett (bei NMR) s singlet (by NMR)
sept Septett (bei NMR) sept septet (by NMR)
SFC superkritische Flüssigchromatographie t Triplett (bei NMR) SFC supercritical liquid chromatography t triplet (by NMR)
tBu teri.-Butyl tBu teri-butyl
td Triplett von Dublett (bei NMR) td triplet of doublet (by NMR)
TFA Trifluoressigsäure  TFA trifluoroacetic acid
THF Tetrahydrofuran  THF tetrahydrofuran
UV Ultraviolett-Spektrometrie  UV ultraviolet spectrometry
v/v Volumen zu Volumen- Verhältnis (einer Lösung) v / v volume to volume ratio (of a solution)
Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthen  Xantphos 4,5-bis (diphenylphosphino) -9,9-dimethylxanthene
zus. zusammen together
HPLC- und LC/MS-Methoden: HPLC and LC / MS methods:
Methode 1 (LC/MS): Method 1 (LC / MS):
Instrument: Waters ACQUITY SQD UPLC System; Säule: Waters Acquity UPLC HSS T3 1.8 μ, 50 x 1 mm; Eluent A: 1 1 Wasser + 0.25 ml 99%-ige Ameisensäure, Eluent B: 1 1 Acetonitril + 0.25 ml 99%-ige Ameisensäure; Gradient: 0.0 min 90% A -> 1.2 min 5% A -> 2.0 min 5% A; Ofen: 50°C; Fluss: 0.40 ml/min; UV-Detektion: 208-400 nm.  Instrument: Waters ACQUITY SQD UPLC System; Column: Waters Acquity UPLC HSS T3 1.8 μ, 50 x 1 mm; Eluent A: 1 l of water + 0.25 ml of 99% formic acid, eluent B: 1 l of acetonitrile + 0.25 ml of 99% formic acid; Gradient: 0.0 min 90% A -> 1.2 min 5% A -> 2.0 min 5% A; Oven: 50 ° C; Flow: 0.40 ml / min; UV detection: 208-400 nm.
Methode 2 (LC/MS): Method 2 (LC / MS):
Instrument: Micromass Quattro Premier mit Waters UPLC Acquity; Säule: Thermo Hypersil GOLD 1.9 μ, 50 x 1 mm; Eluent A: 1 1 Wasser + 0.5 ml 50%-ige Ameisensäure, Eluent B: 1 1 Acetonitril + 0.5 ml 50%-ige Ameisensäure; Gradient: 0.0 min 97% A— > 0.5 min 97% A— > 3.2 min 5% A -> 4.0 min 5% A; Ofen: 50°C; Fluss: 0.3 ml/min; UV-Detektion: 210 nm.  Instrument: Micromass Quattro Premier with Waters UPLC Acquity; Column: Thermo Hypersil GOLD 1.9 μ, 50 x 1 mm; Eluent A: 1 l of water + 0.5 ml of 50% formic acid, eluent B: 1 l of acetonitrile + 0.5 ml of 50% formic acid; Gradient: 0.0 min 97% A-> 0.5 min 97% A-> 3.2 min 5% A -> 4.0 min 5% A; Oven: 50 ° C; Flow: 0.3 ml / min; UV detection: 210 nm.
Methode 3 (LC/MS): Method 3 (LC / MS):
Instrument MS: Waters Micromass QM; Instrument HPLC: Agilent 1100 Serie; Säule: Agilent ZORBAX Extend-C18 3.5 μ, 3.0 x 50 mm; Eluent A: 1 1 Wasser + 0.01 mol Ammoniumcarbonat, Eluent B: 1 1 Acetonitril; Gradient: 0.0 min 98% A -> 0.2 min 98% A -> 3.0 min 5% A -> 4.5 min 5% A; Ofen: 40°C; Fluss: 1.75 ml/min; UV-Detektion: 210 nm.  Instrument MS: Waters Micromass QM; Instrument HPLC: Agilent 1100 series; Column: Agilent ZORBAX Extend-C18 3.5 μ, 3.0 x 50 mm; Eluent A: 1 l of water + 0.01 mol of ammonium carbonate, eluent B: 1 l of acetonitrile; Gradient: 0.0 min 98% A -> 0.2 min 98% A -> 3.0 min 5% A -> 4.5 min 5% A; Oven: 40 ° C; Flow: 1.75 ml / min; UV detection: 210 nm.
Methode 4 (präparative HPLC): Method 4 (preparative HPLC):
Säule: Reprosil C18, 10 μιη, 250 x 30 mm; Eluent: Acetonitril/Wasser mit 0.1% TFA; Gradient: 0-5.00 min 10:90, Probeninjektion bei 3.00 min; 5.00-23.00 min bis 95:5; 23.00-30.00 min 95:5; 30.00-30.50 min bis 10:90; 30.50-31.20 min 10:90. Methode 5 (präparative HPLC): Column: Reprosil C18, 10 μm, 250 × 30 mm; Eluent: acetonitrile / water with 0.1% TFA; Gradient: 0-5.00 min 10:90, sample injection at 3.00 min; 5.00-23.00 min to 95: 5; 23.00-30.00 min 95: 5; 30.00-30.50 min to 10:90; 30.50-31.20 min 10:90. Method 5 (preparative HPLC):
Säule: Reprosil C18, 10 μιη, 250 x 30 mm; Eluent: AcetonitrilAVasser mit 0.1% TFA; Gradient: 0-5.00 min 10:90, Probeninjektion bei 3.00 min; 5.00-20.00 min bis 95:5; 20.00-30.00 min 95:5; 30.00-30.50 min bis 10:90; 30.50-31.20 min 10:90. Methode 6 (präparative HPLC):  Column: Reprosil C18, 10 μm, 250 × 30 mm; Eluent: acetonitrile / water with 0.1% TFA; Gradient: 0-5.00 min 10:90, sample injection at 3.00 min; 5.00-20.00 min to 95: 5; 20.00-30.00 min 95: 5; 30.00-30.50 min to 10:90; 30.50-31.20 min 10:90. Method 6 (preparative HPLC):
Säule: Reprosil C18, 10 μιη, 125 x 30 mm; Eluent: AcetonitrilA asser mit 0.1% TFA; Gradient: 0-6.00 min 35:65, Probeninjektion bei 3.00 min; 6.00-27.00 min bis 80:20; 27.00-30.00 min 95:5; 30.00-33.00 min bis 35:65.  Column: Reprosil C18, 10 μm, 125 × 30 mm; Eluent: acetonitrile / water with 0.1% TFA; Gradient: 0-6.00 min 35:65, sample injection at 3.00 min; 6.00-27.00 min to 80:20; 27.00-30.00 min 95: 5; 30.00-33.00 min to 35:65.
Methode 7 (präparative HPLC): Method 7 (preparative HPLC):
Säule: Reprosil-Pur C18, 10 μιη; Eluent: Wasser/Methanol; Gradient: 70:30 -> 50:50 (bis 6 min) -> 20:80 (bis 22 min), bis 75 min 20:80. Column: Reprosil-Pur C18, 10 μιη; Eluent: water / methanol; Gradient: 70:30 -> 50:50 (to 6 min) -> 20:80 (to 22 min), to 75 min 20:80.
Methode 8 (präparative HPLC): Method 8 (preparative HPLC):
Säule: Reprosil-Pur C18, 10 μιη; Eluent: Wasser/Methanol; Gradient: 70:30 -> 50:50 (bis 6 min) -> 20:80 (bis 20 min), bis 115 min 20:80. Methode 9 (präparative HPLC):  Column: Reprosil-Pur C18, 10 μιη; Eluent: water / methanol; Gradient: 70:30 -> 50:50 (to 6 min) -> 20:80 (to 20 min), to 115 min 20:80. Method 9 (preparative HPLC):
Säule: Reprosil-Pur C18, 10 μιη; Eluent: Wasser/Methanol; Gradient: 70:30 -> 50:50 (bis 6 min) -> 20:80 (bis 21 min), bis 75 min 20:80.  Column: Reprosil-Pur C18, 10 μιη; Eluent: water / methanol; Gradient: 70:30 -> 50:50 (to 6 min) -> 20:80 (to 21 min), to 75 min 20:80.
Methode 10 (präparative HPLC): Method 10 (preparative HPLC):
Säule: Reprosil-Pur C18, 10 μιη; Eluent: Wasser/Methanol; Gradient: 70:30 -> 50:50 (bis 6 min) -> 20:80 (bis 25 min), bis 75 min 20:80.  Column: Reprosil-Pur C18, 10 μιη; Eluent: water / methanol; Gradient: 70:30 -> 50:50 (to 6 min) -> 20:80 (to 25 min), to 75 min 20:80.
Methode 11 (präparative HPLC): Method 11 (preparative HPLC):
Säule: Reprosil-Pur C18, 10 μιη; Eluent: Wasser/Methanol; Gradient: 70:30 -> 50:50 (bis 6 min) -> 20:80 (bis 20 min), bis 75 min 20:80.  Column: Reprosil-Pur C18, 10 μιη; Eluent: water / methanol; Gradient: 70:30 -> 50:50 (to 6 min) -> 20:80 (to 20 min), to 75 min 20:80.
Weitere Angaben: More information:
Die Prozentangaben in den folgenden Beispiel- und Testbeschreibungen sind, sofern nicht anders angegeben, Gewichtsprozente; Teile sind Gewichtsteile. Lösungsmittelverhältnisse, Verdünnungsverhältnisse und Konzentrationsangaben von flüssig/flüssig-Lösungen beziehen sich jeweils auf das Volumen. Reinheitsangaben beziehen sich in der Regel auf entsprechende Peak-Integrationen im LC/MS- Chromatogramm, können aber zusätzlich auch unter Zuhilfenahme des ^-NMR-Spektrums ermittelt worden sein. Wenn keine Reinheit angegeben ist, handelt es sich in der Regel um eine 100%-Reinheit laut automatischer Peak-Integration im LC/MS-Chromatogramm oder die Reinheit wurde nicht explizit ermittelt. The percentages in the following example and test descriptions are by weight unless otherwise indicated; Parts are parts by weight. Solvent ratios, dilution ratios and concentration data of liquid / liquid solutions are based on volume. Purity specifications usually refer to corresponding peak integrations in the LC / MS chromatogram, but may additionally have been determined with the help of the--NMR spectrum. If no purity is specified, it is usually a 100% purity according to automatic peak integration in the LC / MS chromatogram, or purity was not explicitly determined.
Angaben zu Ausbeuten in % d. Th. sind in der Regel reinheitskorrigiert, sofern eine Reinheit <100% angegeben ist. Bei lösungsmittelhaltigen oder verunreinigten Chargen kann die Ausbeute formal ">100%" betragen; in diesen Fällen ist die Ausbeute nicht lösungsmittel- bzw. reinheitskorrigiert. Die nachfolgenden Beschreibungen der Kopplungsmuster von ^-NMR-Signalen wurden teilweise direkt den Vorschlägen des ACD SpecManagers (ACD/Labs Release 12.00, Product version 12.5) entnommen und nicht notwendigerweise streng hinterfragt. Teilweise wurden die Vorschläge des SpecManagers manuell angepasst. Manuell angepasste bzw. zugewiesene Beschreibungen orientieren sich in der Regel an dem optischen Erscheinungsbild der betreffenden Signale und entsprechen nicht notwendigerweise einer strengen, physikalisch korrekten Interpretation. In der Regel bezieht sich die Angabe zur chemischen Verschiebung auf das Zentrum des betreffenden Signals. Bei breiten Multipletts erfolgt die Angabe eines Intervalls. Durch Lösungsmittel oder Wasser verdeckte Signale wurden entweder tentativ zugeordnet oder sind nicht aufgeführt. Data on yields in% d. Th. Are purity-corrected as a rule, provided that a purity of <100% is specified. For solvent-containing or contaminated batches, the yield may be formally "> 100%"; in these cases, the yield is not solvent or purity corrected. The following descriptions of the coupling patterns of ^ -NMR signals were partly taken directly from the suggestions of the ACD SpecManager (ACD / Labs Release 12.00, Product version 12.5) and not necessarily rigorously questioned. In part, the suggestions of the SpecManager were adapted manually. Manually customized descriptions are usually based on the visual appearance of the signals in question and do not necessarily correspond to a rigorous, physically correct interpretation. As a rule, the indication of the chemical shift refers to the center of the relevant signal. For wide multiplets, an interval is specified. Solvent or water concealed signals were either tentatively assigned or are not listed.
Schmelzpunkte und Schmelzbereiche, soweit angegeben, sind nicht korrigiert. Für alle Reaktanden oder Reagenzien, deren Herstellung im Folgenden nicht explizit beschrieben ist, gilt, dass sie von allgemein zugänglichen Quellen kommerziell bezogen wurden. Für alle übrigen Reaktanden oder Reagenzien, deren Herstellung im Folgenden ebenfalls nicht beschrieben ist und die nicht kommerziell erhältlich waren oder von Quellen bezogen wurden, die nicht allgemein zugänglich sind, ist ein Verweis auf die veröffentlichte Literatur angegeben, in der ihre Her- Stellung beschrieben ist. Melting points and melting ranges, where indicated, are not corrected. For all reactants or reagents, the preparation of which is not explicitly described below, it is true that they were obtained commercially from generally available sources. For all other reactants or reagents, the preparation of which is also not described below and which were not commercially available or obtained from sources that are not generally available, reference is made to the published literature describing their preparation ,
Bei den im Folgenden beschriebenen Intermediaten und Ausführungsbeispielen bedeutet eine im IUP AC -Namen des betreffenden Beispiels aufgeführte Bezeichnung " IRS,2RS,5SR" in Verbindung mit der Angabe "Racemat", dass es sich hierbei um ein racemisches Gemisch des IR,2R,5S- Enantiomeren (— jeweils 1. Buchstabe nach der Positionsziffer in " IRS,2RS,5SR") mit dem ent- sprechenden lS,2S,5R-Enantiomeren (— > jeweils 2. Buchstabe nach der Positionsziffer) handelt. Die Bezeichnung " IRS,2RS,5SR" in Verbindung mit den Angaben "Enantiomer 1" und "Enantio- mer 2" bedeutet, dass es sich hierbei um die beiden Enantiomere in separierter, isolierter Form handelt, wobei eine Zuordnung der Absolutkonfiguration (IR,2R,5S oder IS,2S,5R) zu diesen Enantiomeren nicht vorgenommen wurde. Ähnliche Bezeichnungen wie " IRS,2SR,5RS", die sich aus der veränderten Priorität und/oder Reihenfolge von Namensbestandteilen aufgrund der IUPAC -Nomenklatur-Regeln ergeben, sind nach dieser Anleitung auf analoge Weise zu interpretieren. Zur vereinfachten Darstellung der relativen stereochemischen Konfiguration chiraler Zentren wird im Folgenden bei den Strukturformeln racemischer Beispielverbindungen nur die Strukturformel eines der beteiligten Enantiomere wiedergegeben; wie aus der Angabe "Racemat" beim zugehörigen IUPAC -Namen ersichtlich, ist in diesen Fällen das zweite Enantiomer mit der jeweils entgegengesetzten Absolutkonfiguration immer mit eingeschlossen. In the intermediates and exemplary embodiments described below, a designation "IRS, 2RS, 5SR" in the IUP AC name of the relevant example, in conjunction with the term "racemate", means that this is a racemic mixture of IR, 2R, 5S - Enantiomers (- each 1st letter after the position number in "IRS, 2RS, 5SR") with the appropriate lS, 2S, 5R-enantiomer (-> each 2nd letter after the position number) acts. The term "IRS, 2RS, 5SR" in connection with the statements "enantiomer 1" and "enantiomer 2" means that these are the two enantiomers in separated, isolated form, wherein an assignment of the absolute configuration (IR, 2R, 5S or IS, 2S, 5R) to these Enantiomers was not made. Similar terms such as "IRS, 2SR, 5RS", which result from the changed priority and / or order of parts of names due to the IUPAC nomenclature rules, shall be interpreted in an analogous manner according to this manual. In order to simplify the description of the relative stereochemical configuration of chiral centers, only the structural formula of one of the participating enantiomers is reproduced below in the structural formulas of racemic example compounds; As can be seen from the term "racemate" in the associated IUPAC name, in these cases the second enantiomer with the respectively opposite absolute configuration is always included.
Ausgangsverbindungen und Intermediate: Starting compounds and intermediates:
Beispiel 1A Example 1A
6-(Trifluormethyl)-l,2,3-benzotriazin-4(3//)-on 6- (trifluoromethyl) -l, 2,3-benzotriazin-4 (3 //) - one
Zu einer Suspension von 24.4 g (119.51 mmol) 2-Amino-5-(trifluormethyl)benzamid in 174 ml eines 2: 1 -Gemisches von Wasser und konz. Salzsäure bei 0°C wurde eine Lösung von 9.08 g (131.47 mmol) Natriumnitrit in 74 ml Wasser langsam hinzugegeben, wobei die Innentemperatur unter 5°C gehalten wurde. Nach 30 min Rühren bei 0°C Badtemperatur wurden unter weiterer Eisbad-Kühlung langsam 74 ml (0.74 mol) 10 M Natronlauge hinzugegeben, wobei die Innentempera- tur auf ca. 20°C anstieg. Es bildete sich zunächst eine Lösung, aus welcher dann eine Suspension entstand, die zur besseren Rührbarkeit mit 100 ml Wasser verdünnt wurde. Nach 1.5 h Rühren bei RT wurde das Gemisch vorsichtig mit konz. Salzsäure sauer gestellt (pH = 2). Der gebildete Niederschlag wurde abfiltriert und dreimal mit Wasser nachgewaschen. Nach Trocknen an der Luft und danach im Vakuum wurden 24.74 g (96% d. Th.) der Titelverbindung erhalten. Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 15.31 (br. s, 1H), 8.46 (s, 1H), 8.40 (d, 2H). To a suspension of 24.4 g (119.51 mmol) of 2-amino-5- (trifluoromethyl) benzamide in 174 ml of a 2: 1 mixture of water and conc. Hydrochloric acid at 0 ° C, a solution of 9.08 g (131.47 mmol) of sodium nitrite in 74 ml of water was added slowly, keeping the internal temperature below 5 ° C. After stirring for 30 minutes at a bath temperature of 0 ° C., 74 ml (0.74 mol) of 10 M sodium hydroxide solution were slowly added with further ice-bath cooling, the internal temperature rising to about 20 ° C. It first formed a solution from which then a suspension was formed, which was diluted for better stirrability with 100 ml of water. After stirring at RT for 1.5 h, the mixture was carefully poured with conc. Hydrochloric acid acidified (pH = 2). The precipitate formed was filtered off and washed three times with water. After drying in air and then in vacuo, 24.74 g (96% of theory) of the title compound were obtained. Ή NMR (400 MHz, DMSO-de): δ [ppm] = 15.31 (br.s, 1H), 8.46 (s, 1H), 8.40 (d, 2H).
LC/MS (Methode 1, ESIpos): Rt = 0.78 min, m/z = 216 [M+H] Beispiel 2A LC / MS (Method 1, ESIpos): R t = 0.78 min, m / z = 216 [M + H] Example 2A
6-Mefhyl-l,2,3-benzotriazin-4(3//)-on 6-Mefhyl-l, 2,3-benzotriazin-4 (3 //) - one
Zu einer Suspension von 32.0 g (213.08 mmol) 2-Amino-5-methylbenzamid in 300 ml eines 2: 1- Gemisches von Wasser und konz. Salzsäure bei 0°C wurde eine Lösung von 16.17 g (234.38 mmol) Natriumnitrit in 120 ml Wasser langsam hinzugegeben, wobei die Innentemperatur unter 5°C gehalten wurde. Nach 30 min Rühren bei 0°C Badtemperatur wurden unter weiterer Eisbad- Kühlung langsam 120 ml (1.2 mol) 10 M Natronlauge hinzugegeben, wobei die Innentemperatur auf ca. 20°C anstieg und vorhandener Feststoff in Lösung ging. Nach 1 h Rühren bei RT wurde das Gemisch vorsichtig mit konz. Salzsäure sauer gestellt (pH = 2). Der gebildete Niederschlag wurde abfiltriert und dreimal mit Wasser nachgewaschen. Nach Trocknen an der Luft und im Vakuum wurden 33.80 g (98% d. Th.) der Titelverbindung erhalten. To a suspension of 32.0 g (213.08 mmol) of 2-amino-5-methylbenzamide in 300 ml of a 2: 1 mixture of water and conc. Hydrochloric acid at 0 ° C, a solution of 16.17 g (234.38 mmol) of sodium nitrite in 120 ml of water was added slowly, keeping the internal temperature below 5 ° C. After stirring for 30 minutes at 0 ° C. bath temperature, 120 ml (1.2 mol) of 10 M sodium hydroxide solution were slowly added with further cooling of the ice bath, during which the internal temperature rose to about 20 ° C. and the solid present went into solution. After stirring for 1 h at RT, the mixture was carefully poured with conc. Hydrochloric acid acidified (pH = 2). The precipitate formed was filtered off and washed three times with water. After drying in air and in vacuo, 33.80 g (98% of theory) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 14.85 (br. s, 1H), 8.08 (d, 1H), 8.02 (s, 1H), 7.90 (d, 1H). LC/MS (Methode 3, ESIpos): Rt = 1.40 min, m/z = 162 [M+H]+. Beispiel 3A Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 14.85 (br.s, 1H), 8.08 (d, 1H), 8.02 (s, 1H), 7.90 (d, 1H). LC / MS (Method 3, ESIpos): R t = 1.40 min, m / z = 162 [M + H] + . Example 3A
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-[4-(benzyloxy)benzoyl]-5-[(4-oxo-l,2,3-benzotriazin- 3(4 /)-yl)methyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- [4- (benzyloxy) benzoyl] -5 - [(4-oxo-l, 2,3-benzotriazine - 3 (4 /) - yl) methyl] cyclopentanecarboxylate (racemate)
Schritt 1: Step 1:
2-(Trimethylsilyl)ethyl 2- [4-(benzyloxy)phenyl] -2-hydroxybicyclo [2.2.1 ]heptan-7-carboxylat  2- (trimethylsilyl) ethyl 2- [4- (benzyloxy) phenyl] -2-hydroxybicyclo [2.2.1] heptane-7-carboxylate
Eine Lösung von 24.30 g (95.52 mmol) e o-2-(Trimemylsilyl)ethyl 2-oxobicyclo[2.2.1]heptan-7- carboxylat [WO 96/15096, Beispiel 360 / Stufe 1] in 60 ml THF wurde bei ca. -5°C Innentemperatur unter Argon langsam mit 114.62 ml (114.62 mmol) einer 1 M Lösung von 4-(Benzyloxy)- phenylmagnesiumbromid in THF versetzt, wobei die Innentemperatur auf maximal 0°C anstieg. Anschließend wurde das Kältebad entfernt und das Gemisch 1 h nachgerührt. Das Gemisch wurde dann mit 200 ml 5%-iger Zitronensäure-Lösung versetzt und zweimal mit Dichlormethan extra- hiert. Die vereinigten organischen Phasen wurden über Magnesiumsulfat getrocknet und eingeengt. Der Rückstand wurde mittels Flash-Chromatographie an 1 kg Kieselgel gereinigt (Laufmittel Cyclohexan/Ethylacetat 9: 1). Es wurden 28.70 g (66% d. Th., Reinheit 97%) der Titelverbindung erhalten. A solution of 24.30 g (95.52 mmol) of e o-2- (trimemylsilyl) ethyl 2-oxobicyclo [2.2.1] heptane-7-carboxylate [WO 96/15096, Example 360 / step 1] in 60 ml of THF was added at ca. .1 ° C internal temperature under argon slowly added to 114.62 ml (114.62 mmol) of a 1 M solution of 4- (benzyloxy) - phenylmagnesium bromide in THF, wherein the internal temperature rose to a maximum of 0 ° C. Subsequently, the cooling bath was removed and the mixture stirred for 1 h. The mixture was then treated with 200 ml of 5% citric acid solution and extracted twice with dichloromethane. The combined organic phases were dried over magnesium sulfate and concentrated. The residue was purified by flash chromatography on 1 kg of silica gel (mobile phase cyclohexane / ethyl acetate 9: 1). There were obtained 28.70 g (66% of theory, purity 97%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 7.49-7.27 (m, 7H), 6.95 (d, 2H), 5.09 (s, 2H), 5.05 (s IH), 4.10-4.00 (m, 2H), 2.44-2.37 (m, IH), 2.33-2.24 (m, IH), 2.23-2.11 (m, IH), 1.78-1.60 (m IH), 1.52-1.26 (m, 4H), 0.95-0.80 (m, 2H), 0.00 (s, 9H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 7.49-7.27 (m, 7H), 6.95 (d, 2H), 5.09 (s, 2H), 5.05 (s IH), 4.10-4.00 (m, 2H), 2.44-2.37 (m, IH), 2.33-2.24 (m, IH), 2.23-2.11 (m, IH), 1.78-1.60 (m IH), 1.52-1.26 (m, 4H), 0.95-0.80 (m, 2H), 0.00 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 3.15 min, m/z = 421 [M+H-H20]+. LC / MS (Method 1, ESIpos): R t = 3.15 min, m / z = 421 [M + HH 2 0] + .
2-(Trimethylsilyl)ethyl 2-[4-(benzyloxy)phenyl]bicyclo[2.2.1]hept-2-en-7-carboxylat 2- (trimethylsilyl) ethyl 2- [4- (benzyloxy) phenyl] bicyclo [2.2.1] hept-2-ene-7-carboxylate
Zu einer Lösung von 28.70 g (63.466 mmol) der Verbindung aus Beispiel 3A / Schritt 1 in 150 ml Dichlormethan unter Argon wurden bei ca. 0°C zunächst 26.50 ml (190.40 mmol) Triethylamin und dann langsam 9.82 ml (126.93 mmol) Methansulfonsäurechlorid gegeben, wobei die Innentemperatur 5°C nicht überschritt. Anschließend wurde 1.5 h bei 0°C nachgerührt. Danach wurde das Gemisch mit Dichlormethan verdünnt und mit Wasser extrahiert. Die organische Phase wurde über Magnesiumsulfat getrocknet und eingeengt und der Rückstand mittels Flash-Chromatographie an 1 kg Kieselgel gereinigt (Laufmittel Cyclohexan/Ethylacetat 95:5). Es wurden 20.06 g (75% d. Th.) der Titelverbindung erhalten. To a solution of 28.70 g (63.466 mmol) of the compound from Example 3A / step 1 in 150 ml of dichloromethane under argon was added at about 0 ° C, first 26.50 ml (190.40 mmol) of triethylamine and then slowly 9.82 ml (126.93 mmol) of methanesulfonyl chloride , wherein the internal temperature did not exceed 5 ° C. The mixture was then stirred at 0 ° C for 1.5 h. Thereafter, the mixture was diluted with dichloromethane and extracted with water. The organic phase was dried over magnesium sulfate and concentrated, and the residue was purified by flash chromatography on 1 kg of silica gel (mobile phase cyclohexane / ethyl acetate 95: 5). 20.06 g (75% of theory) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 7.48-7.28 (m, 7H), 6.97 (d, 2H), 6.30 (d, 1H), 5.11 (s, 2H), 4.15-4.06 (m, 2H), 3.43 (br. s, 1H), 3.06 (br. s, 1H), 1.85-1.71 (m, 2H), 1.17-1.06 (m, 1H), 1.04-0.87 (m, 3H), 0.04 (s, 9H). Ή NMR (400 MHz, DMSO-de): δ [ppm] = 7.48-7.28 (m, 7H), 6.97 (d, 2H), 6.30 (d, 1H), 5.11 (s, 2H), 4.15-4.06 (m, 2H), 3.43 (brs s, 1H), 3.06 (brs s, 1H), 1.85-1.71 (m, 2H), 1.17-1.06 (m, 1H), 1.04-0.87 (m, 3H) , 0.04 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.61 min, m/z = 421 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.61 min, m / z = 421 [M + H] + .
Schritt 3: Step 3:
2-(Trimethylsilyl)ethyl 2-[4-(benzyloxy)phenyl]-2,3-dihydroxybicyclo[2.2.1]heptan-7-carboxylat  2- (trimethylsilyl) ethyl 2- [4- (benzyloxy) phenyl] -2,3-dihydroxybicyclo [2.2.1] heptane-7-carboxylate
Zu einer entgasten Lösung von 25.37 g (60.314 mmol, nicht reinheitskorrigiert) der Verbindung aus Beispiel 3A / Schritt 2 in 150 ml THF unter Argon wurde bei 0°C eine entgaste Lösung von 15.90 g (135.71 mmol) /V-Methylmorpholin-/V-oxid (NMO) in 42 ml Wasser unter Argon gegeben. Zu diesem Gemisch wurden dann langsam unter Rühren 116 ml (9.05 mmol) einer 2.5%-igen Lösung von Osmiumtetroxid in tert. -Butanol gegeben. Anschließend wurde 1 h bei 0°C nachgerührt. Nach weiteren 16 h Rühren bei RT wurde das Gemisch mit 150 ml Ethylacetat verdünnt und zweimal mit jeweils 250 ml 10%-iger Zitronensäure-Lösung, zweimal mit jeweils 300 ml gesättigter Natriumhydrogencarbonat-Lösung und zweimal mit jeweils 300 ml gesättigter Natriumchlorid- Lösung extrahiert. Die organische Phase wurde anschließend über Natriumsulfat getrocknet und eingeengt. Es wurden 27.51 g (75% d. Th., Reinheit 75%) der Titelverbindung erhalten. LC/MS (Methode 1, ESIpos): Rt = 1.40 min, m/z = 437 [M+H-H20]+. Schritt 4: To a degassed solution of 25.37 g (60.314 mmol, not purity-corrected) of the compound from Example 3A / step 2 in 150 ml of THF under argon at 0 ° C was a degassed solution of 15.90 g (135.71 mmol) / V-Methylmorpholin- / V oxide (NMO) in 42 ml of water under argon. 116 ml (9.05 mmol) of a 2.5% solution of osmium tetroxide in tert. Given -butanol. The mixture was then stirred at 0 ° C for 1 h. After a further 16 h stirring at RT, the mixture was diluted with 150 ml of ethyl acetate and extracted twice with 250 ml of 10% citric acid solution, twice with 300 ml of saturated sodium bicarbonate solution and twice with 300 ml of saturated sodium chloride solution. The organic phase was then dried over sodium sulfate and concentrated. There were obtained 27.51 g (75% of theory, purity 75%) of the title compound. LC / MS (Method 1, ESIpos): R t = 1.40 min, m / z = 437 [M + HH 2 O] + . Step 4:
2-(Trimethylsilyl)ethyl (lRS,2RS,5SR)-2-[4-(benzyloxy)benzoyl]-5-formylcyclopentancarboxylat 2- (trimethylsilyl) ethyl (1RS, 2RS, 5SR) -2- [4- (benzyloxy) benzoyl] -5-formylcyclopentanecarboxylate
(Racemat) (Racemate)
Methode A: Method A:
Zu einer Lösung von 27.42 g (60.31 mmol, nicht reinheitskorrigiert) der Verbindung aus Beispiel 3A / Schritt 3 in 170 ml Methanol unter Argon wurden bei -15°C Badtemperatur langsam 30.96 g (66.34 mmol, Reinheit 95%) Bleitetraacetat gegeben. Das Gemisch wurde 1 h bei -15°C gerührt. Nach Erwärmen auf RT wurde das Gemisch über Celite filtriert und der Filtrationsrückstand dreimal mit jeweils 50 ml Methanol nachgewaschen. Das Filtrat wurde eingeengt und der Rückstand in 500 ml Dichlormethan und 500 ml Wasser aufgenommen, ohne dass sich eine Phasentrennung einstellte. Daraufhin wurde das Gemisch über Kieselgel filtriert und das Kieselgel mit Dichlormethan nachgewaschen. Nach Phasentrennung wurde die wässrige Phase noch einmal mit 150 ml Dichlor - methan extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet und eingeengt. Es wurden 27.1 g (86% d. Th., Reinheit 87%) der Titelverbindung erhalten.  To a solution of 27.42 g (60.31 mmol, not purity-corrected) of the compound from Example 3A / step 3 in 170 ml of methanol under argon was added slowly at -15 ° C bath temperature 30.96 g (66.34 mmol, purity 95%) of lead tetraacetate. The mixture was stirred at -15 ° C for 1 h. After warming to RT, the mixture was filtered through Celite and the filtration residue washed three times with 50 ml of methanol. The filtrate was concentrated and the residue was taken up in 500 ml of dichloromethane and 500 ml of water, without any phase separation. The mixture was then filtered through silica gel and the silica gel washed with dichloromethane. After phase separation, the aqueous phase was extracted once more with 150 ml of dichloromethane. The combined organic phases were dried over sodium sulfate and concentrated. There were obtained 27.1 g (86% of theory, purity 87%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 9.72 (d, 1H), 8.02 (d, 2H), 7.53-7.34 (m, 5H), 7.18 (d, 2H), 5.25 (s, 2H), 4.17 (q, 1H), 4.09 (dd, 2H), 3.74 (t, 1H), 3.23-3.14 (m, 1H), 2.24-2.13 (m, 1H), 2.08-1.88 (m, 2H), 1.61-1.49 (m, 1H), 0.87-0.79 (m, 2H), 0.00 (s, 9H). LC/MS (Methode 1, ESIpos): Rt = 1.45 min, m/z = 425 [M+H-28] Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 9.72 (d, 1H), 8.02 (d, 2H), 7.53-7.34 (m, 5H), 7.18 (d, 2H), 5.25 ( s, 2H), 4.17 (q, 1H), 4.09 (dd, 2H), 3.74 (t, 1H), 3.23-3.14 (m, 1H), 2.24-2.13 (m, 1H), 2.08-1.88 (m, 2H), 1.61-1.49 (m, 1H), 0.87-0.79 (m, 2H), 0.00 (s, 9H). LC / MS (Method 1, ESIpos): R t = 1.45 min, m / z = 425 [M + H-28]
Methode B: Method B:
Zu einer Lösung von 69.0 g (131 mmol, ca. 80% Reinheit) der Verbindung aus Beispiel 3A / Schritt 2 in einem Gemisch aus Aceton/Wasser/THF (3: 1 : 1) wurden bei 0°C unter Argon zunächst 76.87 g (656 mmol) N-Mefhylmorpholin-N-oxid (NMO) und dann 2.09 g (8.20 mmol) einer 4%- igen Lösung von Osmiumtetroxid in Wasser gegeben. Das Gemisch wurde 3 Tage bei RT gerührt. Anschließend wurden 105.26 g (492 mmol) Natriumperiodat hinzugefügt und das Gemisch weiter über Nacht bei RT gerührt. Nach Versetzen mit Ethylacetat und 10%-iger wässriger Zitronensäure wurde die wässrige Phase abgetrennt und einmal mit Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumhydrogencarbonat-Lösung gewaschen und dann mit Magnesiumsilikat (Fluorisil) verrührt. Nach Filtration wurde der Filterrückstand mit Ethylacetat nachgewaschen. Nach Einengen des Filtrats wurde der so erhaltene Rückstand mit den Rückständen aus zwei ähnlich durchgeführten Vorversuchen [eingesetzte Mengen der Verbindung aus Beispiel 3A / Schritt 2: 3.0 g (7.13 mmol) bzw. 3.2 g (7.61 mmol)] vereinigt und gemeinsam mittels Flash-Chromatographie gereinigt (Kieselgel, Laufmittel Petrolether/Ethylacetat 8:2). Es wurden auf diese Weise 53 g (58% d. Th. unter Berücksichtigung der Vorversuche, Reinheit 89%) der Titelverbindung erhalten. To a solution of 69.0 g (131 mmol, about 80% purity) of the compound from Example 3A / step 2 in a mixture of acetone / water / THF (3: 1: 1) were added at 0 ° C under argon first Add 76.87 g (656 mmol) of N-methylmorpholine N-oxide (NMO) and then 2.09 g (8.20 mmol) of a 4% solution of osmium tetroxide in water. The mixture was stirred at RT for 3 days. Subsequently, 105.26 g (492 mmol) of sodium periodate were added and the mixture was further stirred overnight at RT. After addition of ethyl acetate and 10% aqueous citric acid, the aqueous phase was separated and extracted once with ethyl acetate. The combined organic phases were washed once with saturated sodium bicarbonate solution and then stirred with magnesium silicate (Fluorisil). After filtration, the filter residue was washed with ethyl acetate. After concentration of the filtrate, the residue thus obtained was combined with the residues from two similar preliminary experiments [amounts used of the compound from Example 3A / step 2: 3.0 g (7.13 mmol) or 3.2 g (7.61 mmol)] and mixed together by flash chromatography. Purified by chromatography (silica gel, eluent petroleum ether / ethyl acetate 8: 2). This gave 53 g (58% of theory, taking into account preliminary tests, purity 89%) of the title compound.
Schritt 5: Step 5:
2-(Trimethylsilyl)ethyl (lRS,2RS,5SR)-2-[4-(benzyloxy)benzoyl]-5-(hydroxymefhyl)cyclopentan- carboxylat (Racemat) 2- (trimethylsilyl) ethyl (1RS, 2RS, 5SR) -2- [4- (benzyloxy) benzoyl] -5- (hydroxyethyl) cyclopentane carboxylate (racemate)
Zu einer Lösung von 27.0 g (59.65 mmol, nicht reinheitskorrigiert) der Verbindung aus Beispiel 3A / Schritt 4 in 135 ml Ethanol wurden bei RT langsam 677 mg (17.895 mmol) Natriumborhydrid gegeben und das Gemisch 30 min bei RT gerührt. Anschließend wurde das Gemisch mit jeweils 400 ml Ammoniumchlorid-Lösung und Wasser versetzt und zweimal mit jeweils 300 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet und eingeengt. Es wurden 21.90 g (70% d. Th., Reinheit 87%) der Titelverbindung erhalten. Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 7.95 (d, 2H), 7.48-7.31 (m, 5H), 7.12 (d, 2H), 5.20 (s, 2H), 4.64 (t, 1H), 4.07-3.98 (m, 3H), 3.53-3.45 (m, 1H), 3.40-3.34 (m, 1H), 2.94 (t, 1H), 2.34-2.23 (m, 1H), 2.12-2.01 (m, 1H), 1.90-1.78 (m, 1H), 1.67-1.47 (m, 2H), 0.82-0.75 (m, 2H), 0.00 (s, 9H). To a solution of 27.0 g (59.65 mmol, not purity-corrected) of the compound from Example 3A / step 4 in 135 ml of ethanol was added slowly at RT 677 mg (17.895 mmol) of sodium borohydride and the mixture was stirred at RT for 30 min. The mixture was then mixed with 400 ml of ammonium chloride solution and water and extracted twice with 300 ml of ethyl acetate. The combined organic phases were dried over sodium sulfate and concentrated. This gave 21.90 g (70% of theory, purity 87%) of the title compound. Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 7.95 (d, 2H), 7.48-7.31 (m, 5H), 7.12 (d, 2H), 5.20 (s, 2H), 4.64 ( t, 1H), 4.07-3.98 (m, 3H), 3.53-3.45 (m, 1H), 3.40-3.34 (m, 1H), 2.94 (t, 1H), 2.34-2.23 (m, 1H), 2.12- 2.01 (m, 1H), 1.90-1.78 (m, 1H), 1.67-1.47 (m, 2H), 0.82-0.75 (m, 2H), 0.00 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.34 min, m/z = 455 [M+H]+. Schritt 6: LC / MS (Method 1, ESIpos): R t = 1.34 min, m / z = 455 [M + H] + . Step 6:
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-[4-(benzyloxy)benzoyl]-5-[(4-oxo-l,2,3-benzotriazin- 3(4//)-yl)mefhyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- [4- (benzyloxy) benzoyl] -5 - [(4-oxo-l, 2,3-benzotriazine - 3 (4 //) - yl) methyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 500 mg (1.10 mmol, nicht reinheitskorrigiert) der Verbindung aus Beispiel 3A / Schritt 5 in 6 ml THF unter Argon wurden 243 mg (1.65 mmol) l,2,3-Benzotriazin-4(3 /)-on und 1.11 g (5.50 mmol) Tributylphosphan gegeben. Anschließend wurden 1.50 ml (3.30 mmol) einer 40% -igen Lösung von Diethylazodicarboxylat (DEAD) in Toluol bei 0°C hinzugetropft. Das Gemisch wurde ca. 1 h bei RT gerührt, dann mit Ethylacetat verdünnt und zweimal mit jeweils 5 ml Wasser und zweimal mit gesättigter Natriumchlorid-Lösung extrahiert. Die organische Phase wurde über Magnesiumsulfat getrocknet und eingeengt. Der Rückstand wurde mittels präparativer HPLC (Methode 6) gereinigt. Es wurden 334 mg (52% d. Th.) der Titelverbindung erhalten. To a solution of 500 mg (1.10 mmol, not purity corrected) of the compound of Example 3A / Step 5 in 6 mL of THF under argon was added 243 mg (1.65 mmol) of l, 2,3-benzotriazine-4 (3 /) -one and Add 1.11 g (5.50 mmol) of tributylphosphine. Subsequently, 1.50 ml (3.30 mmol) of a 40% solution of diethyl azodicarboxylate (DEAD) in toluene were added dropwise at 0 ° C. The mixture was stirred at RT for about 1 h, then diluted with ethyl acetate and extracted twice with 5 ml each of water and twice with saturated sodium chloride solution. The organic phase was dried over magnesium sulfate and concentrated. The residue was purified by preparative HPLC (Method 6). There were obtained 334 mg (52% of theory) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.44 (dd, 1H), 8.38 (d, 1H), 8.27 (td, 1H), 8.15-8.08 (m, 3H), 7.65-7.48 (m, 5H), 7.29 (d, 2H), 5.37 (s, 2H), 4.74-4.62 (m, 2H), 4.26 (q, 1H), 3.40 (t, 1H), 3.13-3.01 (m, 1H), 2.36-2.25 (m, 1H), 2.21-2.10 (m, 1H), 1.96-1.84 (m, 1H), 1.77-1.65 (m, 1H), 0.53-0.46 (m, 2H), 0.17 (s, 9H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.44 (dd, 1H), 8.38 (d, 1H), 8.27 (td, 1H), 8.15-8.08 (m, 3H), 7.65- 7.48 (m, 5H), 7.29 (d, 2H), 5.37 (s, 2H), 4.74-4.62 (m, 2H), 4.26 (q, 1H), 3.40 (t, 1H), 3.13-3.01 (m, 1H), 2.36-2.25 (m, 1H), 2.21-2.10 (m, 1H), 1.96-1.84 (m, 1H), 1.77-1.65 (m, 1H), 0.53-0.46 (m, 2H), 0.17 ( s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.51 min, m/z = 584 [M+H]+. Beispiel 4A LC / MS (Method 1, ESIpos): R t = 1.51 min, m / z = 584 [M + H] + . Example 4A
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-(4-hydroxybenzoyl)-5-[(4-oxo-l,2,3-benzotriazin-3(4 /)- yl)methyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- (4-hydroxybenzoyl) -5 - [(4-oxo-l, 2,3-benzotriazin-3 ( 4 /) - yl) methyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 270 mg (0.46 mmol) der Verbindung aus Beispiel 3A in 12 ml Ethylacetat unter Argon wurden 25 mg (0.024 mmol) Palladium auf Aktivkohle (10% Pd) gegeben. Anschließend wurde 42 h unter Normaldruck hydriert. Das Gemisch wurde danach über Kieselgur filtriert, der Filter-Rückstand mit Ethylacetat nachgewaschen und das Filtrat eingeengt. Der so erhaltene Rückstand wurde in wenig Dichlormethan aufgenommen und mittels Säulenchromatographie ge- reinigt (25 g Kieselgel, Laufmittel Cyclohexan/Ethylacetat 7:3). Es wurden 165 mg (72% d. Th., Reinheit 100%) der Titelverbindung erhalten. To a solution of 270 mg (0.46 mmol) of the compound of Example 3A in 12 mL of ethyl acetate under argon was added 25 mg (0.024 mmol) of palladium on charcoal (10% Pd). The mixture was then hydrogenated under normal pressure for 42 h. The mixture was then filtered through kieselguhr, the filter residue washed with ethyl acetate and the filtrate was concentrated. The residue thus obtained was taken up in a little dichloromethane and purified by column chromatography (25 g of silica gel, mobile phase cyclohexane / ethyl acetate 7: 3). 165 mg (72% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, CDC13): δ [ppm] = 8.37 (d, 1H), 8.16 (d, 1H), 7.98-7.88 (m, 3H), 7.84-7.77 (m, 1H), 6.89 (d, 2H), 6.67 (br. s, 1H), 4.77-4.70 (m, 1H), 4.68-4.60 (m, 1H), 4.20-4.10 (m, 1H), 3.88-3.81 (m, 2H), 3.46 (t, 1H), 3.08-2.94 (m, 1H), 2.19-2.04 (m, 1H), 2.01-1.86 (m, 2H), 1.72- 1.64 (m, teilweise verdeckt, 1H), 0.63-0.55 (m, 2H), -0.09 (s, 9H). Ή NMR (400 MHz, CDCl 3 ): δ [ppm] = 8.37 (d, 1H), 8.16 (d, 1H), 7.98-7.88 (m, 3H), 7.84-7.77 (m, 1H), 6.89 ( d, 2H), 6.67 (br s, 1H), 4.77-4.70 (m, 1H), 4.68-4.60 (m, 1H), 4.20-4.10 (m, 1H), 3.88-3.81 (m, 2H), 3.46 (t, 1H), 3.08-2.94 (m, 1H), 2.19-2.04 (m, 1H), 2.01-1.86 (m, 2H), 1.72- 1.64 (m, partially occluded, 1H), 0.63-0.55 ( m, 2H), -0.09 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.23 min, m/z = 494 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.23 min, m / z = 494 [M + H] + .
Beispiel 5A Example 5A
2-(Trimethylsilyl)ethyl (lR5,25R,5R5)-2-[(4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetra- hydro-2/f-pyran-4-ylmefhoxy)benzoyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (1R5,25R, 5R5) -2 - [(4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) methyl] -5- [4- (tetrahydro -2 / f-pyran-4-ylmefhoxy) benzoyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 164 mg (0.33 mmol) der Verbindung aus Beispiel 4A in 3.7 ml Acetonitril unter Argon wurden 92 mg (0.66 mmol) Kaliumcarbonat und 71 mg (0.40 mmol) 4-(Brommefhyl)- tetrahydropyran gegeben und das Gemisch 20 h unter Rückfluss gerührt. Anschließend wurden weitere 36 mg (0.20 mmol) 4-(Brommethyl)tetrahydropyran hinzugefügt und das Gemisch erneut 7 h unter Rückfluss gerührt. Danach wurden nochmals 71 mg (0.40 mmol) 4-(Brommethyl)tetra- hydropyran sowie 46 mg (0.33 mmol) Kaliumcarbonat hinzugegeben und das Gemisch weitere 17 h unter Rückfluss gerührt. Nach Abkühlen auf RT wurde das Gemisch mit 30 ml Wasser und 30 ml Ethylacetat verdünnt, und nach Trennen der Phasen wurde die wässrige Phase einmal mit 30 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde mittels präparativer HPLC (Methode 4) gereinigt. Die vereinigten produkthaltigen Fraktionen wurden mit gesättigter wässriger Natrium- hydrogencarbonat-Lösung auf pH 7-8 gestellt, dann bis auf einen Rest an wässriger Phase eingeengt und diese zweimal mit Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet, eingeengt und der Rückstand im Vakuum getrocknet. Es wurden 85 mg (42% d. Th., Reinheit 97%) der Titelverbindung erhalten. To a solution of 164 mg (0.33 mmol) of the compound from Example 4A in 3.7 ml of acetonitrile under argon was added 92 mg (0.66 mmol) of potassium carbonate and 71 mg (0.40 mmol) of 4- (bromomethyl) -tetrahydropyran and the mixture under 20 h under Reflux stirred. Subsequently, another 36 mg (0.20 mmol) of 4- (bromomethyl) tetrahydropyran were added and the mixture stirred again under reflux for 7 h. Then another 71 mg (0.40 mmol) of 4- (bromomethyl) tetrahydropyran and 46 mg (0.33 mmol) of potassium carbonate were added and the mixture was stirred under reflux for a further 17 h. After cooling to RT, the mixture was diluted with 30 ml of water and 30 ml of ethyl acetate, and after separating the phases, the aqueous phase was extracted once with 30 ml of ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and concentrated. The residue was purified by preparative HPLC (Method 4). The combined product-containing fractions were adjusted to pH 7-8 with saturated aqueous sodium bicarbonate solution, then concentrated to a residual aqueous phase and extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, concentrated and the residue was dried in vacuo. There were obtained 85 mg (42% of theory, purity 97%) of the title compound.
Ή-NMR (400 MHz, CDCL): δ [ppm] = 8.37 (dd, 1H), 8.15 (d, 1H), 7.99-7.91 (m, 3H), 7.83-7.76 (m, 1H), 6.91 (d, 2H), 4.77-4.68 (m, 1H), 4.67-4.59 (m, 1H), 4.23-4.13 (m, 1H), 4.03 (dd, 2H), 3.89-3.80 (m, 4H), 3.50-3.39 (m, teilweise verdeckt, 3H), 3.09-2.93 (m, 1H), 2.19-2.03 (m, 2H), 2.01-1.86 (m, 2H), 1.76 (dd, 2H), 1.71-1.62 (m, teilweise verdeckt, 1H), 1.47 (qd, 2H), 0.65-0.53 (m, 2H), -0.09 (s, 9H). Ή NMR (400 MHz, CDCL): δ [ppm] = 8.37 (dd, 1H), 8.15 (d, 1H), 7.99-7.91 (m, 3H), 7.83-7.76 (m, 1H), 6.91 (i.e. , 2H), 4.77-4.68 (m, 1H), 4.67-4.59 (m, 1H), 4.23-4.13 (m, 1H), 4.03 (dd, 2H), 3.89-3.80 (m, 4H), 3.50-3.39 (m, partially occluded, 3H), 3.09-2.93 (m, 1H), 2.19-2.03 (m, 2H), 2.01-1.86 (m, 2H), 1.76 (dd, 2H), 1.71-1.62 (m, partial occluded, 1H), 1.47 (qd, 2H), 0.65-0.53 (m, 2H), -0.09 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.43 min, m/z = 592 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.43 min, m / z = 592 [M + H] + .
Beispiel 6A Example 6A
2-(Trimethylsilyl)ethyl (lR5,25R,5R5)-2-[(4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2- (tetrahydro-2i7-pyran-4-yl)ethoxy]benzoyl } cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (1R5,25R, 5R5) -2 - [(4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) -methyl] -5- {4- [2- (2H) tetrahydro-2α-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylate (racemate)
Zu einer Lösung von 3.88 g (7.47 mmol, Reinheit 95%) der Verbindung aus Beispiel 4A in 41 ml DMF unter Argon wurden 1.01 g (8.96 mmol) Kalium-feri.-butylat gegeben. Nach 5 min Rühren bei RT wurden 1.73 g (8.96 mmol) 4-(2-Bromefhyl)tetrahydro-2i7-pyran hinzugegeben, und das Gemisch wurde 2 h bei 100°C Badtemperatur gerührt. Nach Abkühlen auf RT wurden dem Gemisch Wasser und Ethylacetat zugesetzt. Nach Trennen der Phasen wurde die wässrige Phase einmal mit Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumchlorid-Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde mittels Säulenchromatographie gereinigt (300 g Kieselgel, Laufmittel Cyclo- hexan/Ethylacetat 7:3). Es wurden 2.91 g (64% d. Th., Reinheit 99%) der Titelverbindung erhalten. To a solution of 3.88 g (7.47 mmol, purity 95%) of the compound from Example 4A in 41 ml of DMF under argon was added 1.01 g (8.96 mmol) of potassium feri-butoxide. After stirring at RT for 5 min, 1.73 g (8.96 mmol) of 4- (2-bromoethyl) tetrahydro-2i-7-pyran were added, and the mixture was stirred for 2 h at a bath temperature of 100.degree. After cooling to RT, water and ethyl acetate were added to the mixture. After separating the phases, the aqueous phase was extracted once with ethyl acetate. The combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. The residue was purified by column chromatography (300 g of silica gel, eluent cyclohexane / ethyl acetate 7: 3). There were obtained 2.91 g (64% of theory, purity 99%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.27 (d, 1H), 8.20 (d, 1H), 8.10 (t, 1H), 7.97-7.89 (m, 3H), 7.03 (d, 2H), 4.57-4.44 (m, 2H), 4.14-4.03 (m, 3H), 3.82 (dd, 2H), 3.63-3.46 (m, 2H), 3.31- 3.17 (m, teilweise verdeckt, 3H), 2.97-2.84 (m, 1H), 2.18-2.05 (m, 1H), 2.04-1.92 (m, 1H), 1.80- 1.48 (m, 6H), 1.29-1.13 (m, 3H), 0.37-0.26 (m, 2H), -0.17 (s, 9H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.27 (d, 1H), 8.20 (d, 1H), 8.10 (t, 1H), 7.97-7.89 (m, 3H), 7.03 ( d, 2H), 4.57-4.44 (m, 2H), 4.14-4.03 (m, 3H), 3.82 (dd, 2H), 3.63-3.46 (m, 2H), 3.31- 3.17 (m, partially concealed, 3H) , 2.97-2.84 (m, 1H), 2.18-2.05 (m, 1H), 2.04-1.92 (m, 1H), 1.80- 1.48 (m, 6H), 1.29-1.13 (m, 3H), 0.37-0.26 ( m, 2H), -0.17 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.46 min, m/z = 606 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.46 min, m / z = 606 [M + H] + .
Beispiel 7A Example 7A
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-[4-(benzyloxy)benzoyl]-5-{ [4-oxo-6-(trifluormethyl)- l,2,3-benzotriazin-3(4 /)-yl]methyl}cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- [4- (benzyloxy) benzoyl] -5- {[4-oxo-6- (trifluoromethyl) - l , 2,3-benzotriazine-3 (4 /) -yl] methyl} cyclopentanecarboxylate (racemate)
Zu einer Lösung von 13.88 g (30.53 mmol, nicht reinheitskorrigiert) der Verbindung aus Beispiel 3A / Schritt 5 in 200 ml Toluol unter Argon wurden 7.88 g (36.64 mmol) der Verbindung aus Beispiel 1A und 9.88 g (48.85 mmol) Tributylphosphan gegeben. Anschließend wurden 13.90 ml (30.53 mmol) einer 40%-igen Lösung von Diethylazodicarboxylat in Toluol bei 0°C hinzugetropft. Das Gemisch wurde 1 Tag bei RT gerührt und dann eingeengt. Der Rückstand wurde mittels Flash-Chromatographie gereinigt (1 kg Kieselgel, Laufmittel Cyclohexan/Ethylacetat 9: 1). Es wurden 9.06 g (44% d. Th., Reinheit 98%) der Titelverbindung erhalten. To a solution of 13.88 g (30.53 mmol, not purity corrected) of the compound of Example 3A / Step 5 in 200 mL of toluene under argon was added 7.88 g (36.64 mmol) of the compound from Example 1A and 9.88 g (48.85 mmol) of tributylphosphine. Subsequently, 13.90 ml (30.53 mmol) of a 40% solution of diethyl azodicarboxylate in toluene were added dropwise at 0 ° C. The mixture was stirred at RT for 1 day and then concentrated. The residue was purified by flash chromatography (1 kg silica gel, eluent cyclohexane / ethyl acetate 9: 1). 9.06 g (44% of theory, purity 98%) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.52 (s, 1H), 8.47-8.43 (m, 2H), 7.95 (d, 2H), 7.48- 7.30 (m, 5H), 7.12 (d, 2H), 5.20 (s, 2H), 4.60-4.50 (m, 2H), 4.10 (q, 1H), 3.65-3.49 (m, 2H), 3.25 (t, 1H), 2.96-2.83 (m, 1H), 2.19-2.07 (m, 1H), 2.07-1.95 (m, 1H), 1.80-1.68 (m, 1H), 1.63-1.50 (m, 1H), 0.39-0.22 (m, 2H), -0.18 (s, 9H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.52 (s, 1H), 8.47-8.43 (m, 2H), 7.95 (d, 2H), 7.48-7.30 (m, 5H), 7.12 (d, 2H), 5.20 (s, 2H), 4.60-4.50 (m, 2H), 4.10 (q, 1H), 3.65-3.49 (m, 2H), 3.25 (t, 1H), 2.96-2.83 ( m, 1H), 2.19-2.07 (m, 1H), 2.07-1.95 (m, 1H), 1.80-1.68 (m, 1H), 1.63-1.50 (m, 1H), 0.39-0.22 (m, 2H), -0.18 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.57 min, m/z = 652 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.57 min, m / z = 652 [M + H] + .
Beispiel 8A 2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-(4-hydroxybenzoyl)-5-{ [4-oxo-6-(trifluormethyl)-l,2,3- benzotriazin-3(4 /)-yl]methyl}cyclopentancarboxylat (Racemat) Example 8A 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- (4-hydroxybenzoyl) -5- {[4-oxo-6- (trifluoromethyl) -l, 2,3-benzotriazine-3 (4 /) - yl] methyl} cyclopentanecarboxylate (racemate)
Zu einer Lösung von 9.05 g (13.89 mmol) der Verbindung aus Beispiel 7A in einem Gemisch aus 100 ml Ethylacetat und 100 ml Ethanol unter Argon wurden 1.05 g (16.66 mmol) Ammonium- formiat und 369 mg (0.35 mmol) Palladium auf Aktivkohle (10% Pd) gegeben. Anschließend wur- de das Gemisch 1 h bei 75°C gerührt. Danach wurden weitere 105 mg (1.67 mmol) Ammonium- formiat hinzugegeben und das Gemisch nochmals 30 min bei 75°C gerührt. Nach Abkühlen auf RT wurde das Gemisch über Kieselgur filtriert, der Filter-Rückstand mit Ethylacetat und Ethanol nachgewaschen und das Filtrat eingeengt. Es wurden 7.86 g (100% d. Th.) der Titelverbindung erhalten. Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 10.40 (br. s, 1H), 8.52 (s, 1H), 8.47-8.42 (m, 2H), 7.85 (d, 2H), 6.84 (d, 2H), 4.60-4.51 (m, 2H), 4.05 (q, 1H), 3.64-3.48 (m, 2H), 3.23 (t, 1H), 2.96-2.82 (m, 1H), 2.17-2.05 (m, 1H), 2.05-1.94 (m, 1H), 1.79-1.67 (m, 1H), 1.62-1.49 (m, 1H), 0.38-0.22 (m, 2H), -0.18 (s, 9H). 1.05 g (16.66 mmol) of ammonium formate and 369 mg (0.35 mmol) of palladium on activated carbon (10 % Pd). The mixture was then stirred for 1 h at 75.degree. Thereafter, a further 105 mg (1.67 mmol) of ammonium formate were added and the mixture was stirred for a further 30 min at 75.degree. After cooling to RT, the mixture was filtered through kieselguhr, the filter residue washed with ethyl acetate and ethanol and the filtrate was concentrated. There were obtained 7.86 g (100% of theory) of the title compound. Ή NMR (400 MHz, DMSO-de): δ [ppm] = 10.40 (br.s, 1H), 8.52 (s, 1H), 8.47-8.42 (m, 2H), 7.85 (d, 2H), 6.84 (d, 2H), 4.60-4.51 (m, 2H), 4.05 (q, 1H), 3.64-3.48 (m, 2H), 3.23 (t, 1H), 2.96-2.82 (m, 1H), 2.17-2.05 (m, 1H), 2.05-1.94 (m, 1H), 1.79-1.67 (m, 1H), 1.62-1.49 (m, 1H), 0.38-0.22 (m, 2H), -0.18 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.31 min, m/z = 562 [M+H]+. Beispiel 9A LC / MS (Method 1, ESIpos): R t = 1.31 min, m / z = 562 [M + H] + . Example 9A
2-(Trimethylsilyl)ethyl (lRS,2SR^^ 2- (trimethylsilyl) ethyl (IRS, 2SR ^^
methyl } -5-[4-(tetrahydro-2 i-pyran-4-ylmethoxy)benzoyl]cyclopentancarboxylat (Racemat) methyl} -5- [4- (tetrahydro-2-pyran-4-ylmethoxy) benzoyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 1.07 g (1.90 mmol) der Verbindung aus Beispiel 8A in 20 ml DMF unter Argon wurden 256 mg (2.28 mmol) Kalium-ieri.-butylat gegeben. Nach 5 min Rühren bei RT wurden 408 mg (2.28 mmol) 4-(Brommethyl)tetrahydropyran hinzugegeben, und das Gemisch wurde 2 h bei 100°C Badtemperatur gerührt. Anschließend wurden weitere 136 mg (0.76 mmol) 4-(Brom- methyl)tetrahydropyran hinzugegeben und das Gemisch nochmals 2 h bei 100°C Badtemperatur gerührt. Nach Abkühlen auf RT wurde das Gemisch mit den Reaktionsgemischen aus zwei ähnlich durchgeführten Vorversuchen (Ansatzgröße jeweils 47 mg (0.08 mmol) der Verbindung aus Beispiel 8A) vereinigt. Nach Entfernen des DMF wurde diesem vereinten Gemisch 60 ml Wasser und 60 ml Ethylacetat zugesetzt. Nach Trennen der Phasen wurde die wässrige Phase einmal mit 30 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde in einem Gemisch aus Cyclohexan und Ethylacetat (9: 1) aufgenommen und mittels Säulenchromatographie gereinigt (120 g Kieselgel, Laufmittel Cyclohexan/Ethylacetat 9: 1). Es wurden 590 mg (47% d. Th., Reinheit 100%) der Titelverbindung erhalten. To a solution of 1.07 g (1.90 mmol) of the compound from Example 8A in 20 ml of DMF under argon was added 256 mg (2.28 mmol) of potassium ieri-butoxide. After stirring at RT for 5 min, 408 mg (2.28 mmol) of 4- (bromomethyl) tetrahydropyran were added, and the mixture was stirred for 2 h at a bath temperature of 100.degree. Subsequently, a further 136 mg (0.76 mmol) of 4- (bromomethyl) tetrahydropyran were added and the mixture was stirred for a further 2 hours at 100 ° C. bath temperature. After cooling to RT, the mixture was combined with the reaction mixtures of two similar preliminary experiments (batch size in each case 47 mg (0.08 mmol) of the compound from Example 8A). After removal of the DMF, 60 ml of water and 60 ml of ethyl acetate were added to this combined mixture. After separating the phases, the aqueous phase was extracted once with 30 ml of ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and concentrated. The residue was taken up in a mixture of cyclohexane and ethyl acetate (9: 1) and purified by column chromatography (120 g silica gel, eluent cyclohexane / ethyl acetate 9: 1). 590 mg (47% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, CDC13): δ [ppm] = 8.66 (s, 1H), 8.28 (d, 1H), 8.14 (dd, 1H), 7.95 (d, 2H), 6.92 (d, 2H), 4.78-4.62 (m, 2H), 4.21-4.13 (m, 1H), 4.03 (dd, 2H), 3.89-3.81 (m, 4H), 3.50-3.40 (m, 3H), 3.07-2.93 (m, 1H), 2.19-2.03 (m, 2H), 2.03-1.87 (m, 2H), 1.76 (dd, 2H), 1.72-1.61 (m, 1H), 1.47 (qd, 2H), 0.63-0.53 (m, 2H), -0.09 (s, 9H). LC/MS (Methode 1, ESIpos): Rt = 1.51 min, m/z = 560 [M+H]+. Ή NMR (400 MHz, CDC1 3 ): δ [ppm] = 8.66 (s, 1H), 8.28 (d, 1H), 8.14 (dd, 1H), 7.95 (d, 2H), 6.92 (d, 2H) , 4.78-4.62 (m, 2H), 4.21-4.13 (m, 1H), 4.03 (dd, 2H), 3.89-3.81 (m, 4H), 3.50-3.40 (m, 3H), 3.07-2.93 (m, 1H), 2.19-2.03 (m, 2H), 2.03-1.87 (m, 2H), 1.76 (dd, 2H), 1.72-1.61 (m, 1H), 1.47 (qd, 2H), 0.63-0.53 (m, 2H), -0.09 (s, 9H). LC / MS (Method 1, ESIpos): R t = 1.51 min, m / z = 560 [M + H] + .
Beispiel 10A Example 10A
2-(Trimethylsilyl)ethyl (lRS,2SR^^ 2- (trimethylsilyl) ethyl (IRS, 2SR ^^
methyl}-5-{4-[2-(tetrahydro-2ii-pyran-4-yl)ethoxy]benzoyl}cyclopentancarboxylat (Racemat) methyl} -5- {4- [2- (tetrahydro-2ii-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylate (racemate)
Zu einer Lösung von 250 mg (0.45 mmol) der Verbindung aus Beispiel 8A in 4.5 ml DMF unter Argon wurden 60 mg (0.53 mmol) Kalium-feri.-butylat gegeben. Nach 5 min Rühren bei RT wurden 103 mg (0.53 mmol) 4-(2-Bromethyl)tetrahydro-2i7-pyran hinzugegeben, und das Gemisch wurde 1 h bei 100°C Badtemperatur gerührt. Nach Abkühlen auf RT wurden dem Gemisch 60 ml Wasser und 60 ml ieri.-Butyl-methylether zugesetzt. Nach Trennen der Phasen wurde die wässrige Phase einmal mit 30 ml ieri.-Butyl-methy lether und zweimal mit jeweils 50 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden über Natriumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde in Dichlormethan aufgenommen und mittels Säulenchromatographie gereinigt (25 g Kieselgel, Laufmittel Cyclohexan/Ethylacetat 7:3). Es wurden 138 mg (46% d. Th., Reinheit 100%) der Titelverbindung erhalten. To a solution of 250 mg (0.45 mmol) of the compound from Example 8A in 4.5 ml of DMF under argon was added 60 mg (0.53 mmol) of potassium feri-butoxide. After stirring at RT for 5 min, 103 mg (0.53 mmol) of 4- (2-bromoethyl) tetrahydro-2i-7-pyran were added, and the mixture was stirred for 1 h at a bath temperature of 100.degree. After cooling to RT, 60 ml of water and 60 ml of tert-butyl methyl ether were added to the mixture. After separating the phases, the aqueous phase was extracted once with 30 ml of ieri.-butyl-methyl ether and twice with 50 ml of ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and concentrated. The residue was taken up in dichloromethane and purified by column chromatography (25 g silica gel, eluent cyclohexane / ethyl acetate 7: 3). There were obtained 138 mg (46% of theory, purity 100%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.52 (s, 1H), 8.45 (s, 2H), 7.93 (d, 2H), 7.04 (d, 2H), 4.60-4.50 (m, 2H), 4.15-4.07 (m, 3H), 3.82 (dd, 2H), 3.64-3.47 (m, 2H), 3.31-3.18 (m, 3H), 2.97- 2.83 (m, 1H), 2.19-2.06 (m, 1H), 2.06-1.94 (m, 1H), 1.81-1.49 (m, 6H), 1.29-1.14 (m, 3H), 0.36- 0.22 (m, 2H), -0.18 (s, 9H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.52 (s, 1H), 8.45 (s, 2H), 7.93 (d, 2H), 7.04 (d, 2H), 4.60-4.50 ( m, 2H), 4.15-4.07 (m, 3H), 3.82 (dd, 2H), 3.64-3.47 (m, 2H), 3.31-3.18 (m, 3H), 2.97- 2.83 (m, 1H), 2.19- 2.06 (m, 1H), 2.06-1.94 (m, 1H), 1.81-1.49 (m, 6H), 1.29-1.14 (m, 3H), 0.36-0.22 (m, 2H), -0.18 (s, 9H) ,
LC/MS (Methode 1, ESIpos): Rt = 1.53 min, m/z = 674 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.53 min, m / z = 674 [M + H] + .
Beispiel IIA Example IIA
2-(Trimethylsilyl)ethyl (lRS,2SR^^ 2- (trimethylsilyl) ethyl (IRS, 2SR ^^
methyl}-5-(4-{ [(trifluormethyl)sulfonyl]oxy}benzoyl)cyclopentancarboxylat (Racemat) methyl} -5- (4- {[(trifluoromethyl) sulfonyl] oxy} benzoyl) cyclopentanecarboxylate (racemate)
Zu einer Lösung von 1.00 g (1.78 mmol) der Verbindung aus Beispiel 8A in 5.0 ml Dichlormethan unter Argon wurden bei 0°C zunächst 0.25 ml (3.12 mmol) Pyridin und anschließend langsam 0.45 ml (2.67 mmol) Trifluormethansulfonsäureanhydrid hinzugegeben. Das Gemisch wurde 1 h bei 0°C gerührt, anschließend mit Dichlormethan versetzt und jeweils einmal mit Wasser und gesättigter Natriumhydrogencarbonat-Lösung gewaschen. Die organische Phase wurde über Magnesiumsulfat getrocknet, filtriert und eingeengt. Es wurden 1.21 g (98% d. Th., Reinheit 100%) der Titelverbindung erhalten. 0.25 ml (3.12 mmol) of pyridine and then, slowly, 0.45 ml (2.67 mmol) of trifluoromethanesulfonic anhydride were added at 0 ° C. to a solution of 1.00 g (1.78 mmol) of the compound from Example 8A in 5.0 ml of dichloromethane under argon. The mixture was stirred for 1 h at 0 ° C, then treated with dichloromethane and washed once each with water and saturated sodium bicarbonate solution. The organic phase was dried over magnesium sulfate, filtered and concentrated. 1.21 g (98% of theory, purity 100%) of the title compound were obtained.
LC/MS (Methode 2, ESIpos): Rt = 3.41 min, m/z = 694 [M+H]+. Beispiel 12A LC / MS (Method 2, ESIpos): R t = 3.41 min, m / z = 694 [M + H] + . Example 12A
2-(Trimethylsilyl)ethyl (lRS,2SR^^ 2- (trimethylsilyl) ethyl (IRS, 2SR ^^
methyl } -5-(4-sulfanylbenzoyl)cyclopentancarboxylat (Racemat) methyl} -5- (4-sulfanylbenzoyl) cyclopentanecarboxylate (racemate)
Zu einer Lösung von 800 mg (1.15 mmol) der Verbindung aus Beispiel I IA in 10 ml Dioxan wur- den nacheinander 264 mg (1.38 mmol) Triisopropylsilanthiol, 298 mg (2.31 mmol) NN-Diiso- propylethylamin, 26 mg (0.03 mmol) Tris(dibenzylidenaceton)dipalladium und 33 mg (0.06 mmol) 4,5-Bis(diphenylphosphin)-9,9-dimethylxanthen (Xantphos) gegeben. Anschließend wurde das Ge- misch entgast, mit Argon gespült und 2.5 h unter Rückfluss gerührt. Nach Abkühlen auf RT wurde das Gemisch mit Ethylacetat versetzt und einmal mit Wasser gewaschen. Nach einmaliger Extraktion der wässrigen Phase mit Ethylacetat wurden die vereinigten organischen Phasen einmal mit gesättigter Natriumchlorid-Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und ein- geengt. Der Rückstand wurde mittels präparativer HPLC (Methode 4) gereinigt. Die produkt- haltigen Fraktionen wurden vereinigt, mit gesättigter wässriger Natriumhydrogencarbonat-Lösung neutralisiert und bis auf ein kleines Restvolumen an Wasser eingeengt. Nach zweimaliger Extraktion dieser wässrigen Phase mit Dichlormethan wurden die vereinigten organischen Phasen über Magnesiumsulfat getrocknet, filtriert, eingeengt und der Rückstand im Vakuum getrocknet. Es wurden 350 mg (35% d. Th., Reinheit 67%) der Titelverbindung erhalten. Laut LC/MS war darin das korrespondierende Disulfid (dimerisiertes Produkt, (+/-)-Bis[2-(trimethylsilyl)ethyl] 2,2'-[di- sulf andiylbis(benzol-4, 1 -diylcarbonyl)]bis(5- { [4-oxo-6-(trifluormethyl)- 1 ,2,3-benzotriazin-3(4 /)- yl]methyl}cyclopentancarboxylat) zu 25% enthalten. To a solution of 800 mg (1.15 mmol) of the compound of Example IA in 10 ml of dioxane was successively 264 mg (1.38 mmol) of triisopropylsilanethiol, 298 mg (2.31 mmol) of N, N-diisopropylethylamine, 26 mg (0.03 mmol). Tris (dibenzylideneacetone) dipalladium and 33 mg (0.06 mmol) of 4,5-bis (diphenylphosphine) -9,9-dimethylxanthene (xanthphos). Subsequently, the degassed, purged with argon and stirred under reflux for 2.5 h. After cooling to RT, the mixture was treated with ethyl acetate and washed once with water. After a single extraction of the aqueous phase with ethyl acetate, the combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. The residue was purified by preparative HPLC (Method 4). The product-containing fractions were combined, neutralized with saturated aqueous sodium bicarbonate solution and concentrated to a small residual volume of water. After extracting this aqueous phase twice with dichloromethane, the combined organic phases were dried over magnesium sulfate, filtered, concentrated and the residue was dried in vacuo. 350 mg (35% of theory, purity 67%) of the title compound were obtained. According to LC / MS, it contained the corresponding disulfide (dimerized product, (+/-) - bis [2- (trimethylsilyl) ethyl] 2,2 '- [disulfandiylbis (benzene-4,1-diylcarbonyl)] bis ( 5- {[4-oxo-6- (trifluoromethyl) -1,2,3-benzotriazine-3 (4 /) -yl] methyl} cyclopentanecarboxylate) at 25%.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.52 (s, 1H), 8.44 (s, 2H), 7.83 (d, 2H), 7.42 (d, 2H), 6.03 (br. s, 1H), 4.60-4.48 (m, 2H), 4.08 (q, 1H), 3.64-3.48 (m, 2H), 3.23 (t, 1H), 2.97-2.81 (m, 1H), 2.19-2.06 (m, 1H), 2.06-1.94 (m, 1H), 1.79-1.67 (m, 1H), 1.62-1.48 (m, 1H), 0.37-0.22 (m, 2H), -0.18 (s, 9H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.52 (s, 1H), 8.44 (s, 2H), 7.83 (d, 2H), 7.42 (d, 2H), 6.03 (br. s, 1H), 4.60-4.48 (m, 2H), 4.08 (q, 1H), 3.64-3.48 (m, 2H), 3.23 (t, 1H), 2.97-2.81 (m, 1H), 2.19-2.06 ( m, 1H), 2.06-1.94 (m, 1H), 1.79-1.67 (m, 1H), 1.62-1.48 (m, 1H), 0.37-0.22 (m, 2H), -0.18 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.44 min, m/z = 578 [M+H]+. Beispiel 13A 2-(Trimethylsilyl)ethyl (lRlS,,2lS,R,5R.S,)-2-{ [4-oxo-6-(trifluormethyl)-l,2,3-benzotriazin-3(4 /)-yl]- methyl}-5-{4-[(tetrahydro-2ii-pyran-4-ylmethyl)sulfanyl]benzoyl}cyclopentancarboxylatLC / MS (Method 1, ESIpos): R t = 1.44 min, m / z = 578 [M + H] + . Example 13A 2- (trimethylsilyl) ethyl (lR l S, l 2 S, R, 5R.S,) -2- {[4-oxo-6- (trifluoromethyl) -l, 2,3-benzotriazin-3 ( 4 /) - yl] - methyl} -5- {4 - [(tetrahydro-2ii-pyran-4-ylmethyl) sulfanyl] benzoyl} cyclopentanecarboxylate
(Racemat) (Racemate)
Zu einer Lösung von 200 mg der Verbindung aus Beispiel 12A (0.35 mmol, nicht reinheits- korrigiert, ca. 25% korrespondierendes Disulfid enthalten) in 14 ml DMF wurden 96 mg (0.69 mmol) Kaliumcarbonat gegeben und das Gemisch 2 min bei RT gerührt. Anschließend wurden 136 mg (0.76 mmol) 4-(Brommethyl)tetrahydropyran sowie 123 mg (1.04 mmol) Natriumhydroxy- methansulfinat hinzugegeben und das Gemisch weitere 30 min bei RT gerührt. Das Gemisch wurde danach eingeengt, und der Rückstand wurde mit Wasser versetzt und zweimal mit Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumchlorid- Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und eingeengt. Es wurden 248 mg (100% d. Th., Reinheit 95%) der Titelverbindung erhalten. LC/MS (Methode 1, ESIpos): Rt = 1.50 min, m/z = 676 [M+H]+. To a solution of 200 mg of the compound from Example 12A (0.35 mmol, not purity-corrected, containing about 25% corresponding disulfide) in 14 ml of DMF, 96 mg (0.69 mmol) of potassium carbonate were added and the mixture was stirred for 2 min at RT. Subsequently, 136 mg (0.76 mmol) of 4- (bromomethyl) tetrahydropyran and 123 mg (1.04 mmol) of sodium hydroxy methanesulfinate were added and the mixture was stirred at RT for a further 30 min. The mixture was then concentrated, and the residue was added with water and extracted twice with ethyl acetate. The combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. 248 mg (100% of theory, purity 95%) of the title compound were obtained. LC / MS (Method 1, ESIpos): R t = 1.50 min, m / z = 676 [M + H] + .
Beispiel 14A Example 14A
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-[4-(benzyloxy)benzoyl]-5-[(6-methyl-4-oxo-l,2,3-benzo- triazin-3(4 /)-yl)methyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- [4- (benzyloxy) benzoyl] -5 - [(6-methyl-4-oxo-l, 2 , 3-benzotriazine-3 (4 /) -yl) methyl] cyclopentanecarboxylate (racemate)
Zu einer Suspension von 9.60 g (20.06 mmol, Reinheit 95%) der Verbindung aus Beispiel 3A / Schritt 5 in 110 ml Toluol unter Argon wurden 3.88 g (24.07 mmol) der Verbindung aus Beispiel 2A gegeben. Anschließend wurden 25.1 ml (100.30 mmol) Tributylphosphan und 27.4 ml (60.18 mmol) einer 40%-igen Lösung von Diethylazodicarboxylat in Toluol bei 0°C hinzugetropft. Nach 2 h Rühren bei RT wurde das Gemisch mit Ethylacetat verdünnt und einmal mit Wasser gewa- sehen. Die wässrige Phase wurde einmal mit Ethylacetat rückextrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumchlorid-Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde mittels Flash-Chromatographie gereinigt (Kieselgel, Laufmittel Cyclohexan/Ethylacetat 85: 15 -> 80:20). Es wurden 6.28 g (51 % d. Th., Reinheit 98%) der Titel Verbindung erhalten. Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.12-8.05 (m, 2H), 7.97-7.88 (m, 3H), 7.48-7.27 (m, 5H), 7.12 (d, 2H), 5.20 (s, 2H), 4.56-4.42 (m, 2H), 4.08 (q, 1H), 3.61-3.46 (m, 2H), 3.22 (t, 1H), 2.96-2.83 (m, 1H), 2.55 (s, 3H), 2.17-2.05 (m, 1H), 2.03-1.92 (m, 1H), 1.78-1.67 (m, 1H), 1.59- 1.47 (m, 1H), 0.38-0.23 (m, 2H), -0.17 (s, 9H). LC/MS (Methode 1, ESIpos): Rt = 1.49 min, m/z = 598 [M+H]+. 3.88 g (24.07 mmol) of the compound from Example 2A were added to a suspension of 9.60 g (20.06 mmol, purity 95%) of the compound from Example 3A / Step 5 in 110 ml of toluene under argon. Subsequently, 25.1 ml (100.30 mmol) of tributylphosphane and 27.4 ml (60.18 mmol) of a 40% solution of diethyl azodicarboxylate in toluene were added dropwise at 0 ° C. After stirring at RT for 2 h, the mixture was diluted with ethyl acetate and washed once with water. The aqueous phase was back-extracted once with ethyl acetate. The combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. The residue was purified by flash chromatography (silica gel, eluent cyclohexane / ethyl acetate 85:15 → 80:20). There were obtained 6.28 g (51% of theory, purity 98%) of the title compound. Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.12-8.05 (m, 2H), 7.97-7.88 (m, 3H), 7.48-7.27 (m, 5H), 7.12 (d, 2H ), 5.20 (s, 2H), 4.56-4.42 (m, 2H), 4.08 (q, 1H), 3.61-3.46 (m, 2H), 3.22 (t, 1H), 2.96-2.83 (m, 1H), 2.55 (s, 3H), 2.17-2.05 (m, 1H), 2.03-1.92 (m, 1H), 1.78-1.67 (m, 1H), 1.59- 1.47 (m, 1H), 0.38-0.23 (m, 2H ), -0.17 (s, 9H). LC / MS (Method 1, ESIpos): R t = 1.49 min, m / z = 598 [M + H] + .
Beispiel 15A Example 15A
2-(Trimethylsilyl)ethyl (lRlS,,2RlS,,5.S,R)-2-(4-hydroxybenzoyl)-5-[(6-methyl-4-oxo-l,2,3-benzo- triazin-3(4//)-yl)mefhyl]cyclopentancarboxylat (Racemat) 2- (trimethylsilyl) ethyl (lR l S, 2R S l, 5.S, R) -2- (4-hydroxybenzoyl) -5 - [(6-methyl-4-oxo-l, 2,3- benzotriazine-3 (4 //) - yl) -methyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 6.25 g (10.25 mmol, Reinheit 98%) der Verbindung aus Beispiel 14A in einem Gemisch aus 50 ml Ethylacetat und 50 ml Ethanol unter Argon wurden 273 mg (0.26 mmol) Palladium auf Aktivkohle (10% Pd) und 969 mg (15.37 mmol) Ammoniumformiat gegeben. Anschließend wurde das Gemisch 2 h bei 70°C gerührt. Nach Abkühlen auf RT wurde das Gemisch über Kieselgur filtriert, der Filter-Rückstand mit Ethylacetat nachgewaschen, das Filtrat eingeengt und der Rückstand im Vakuum getrocknet. Es wurden 5.20 g (97% d. Th., Reinheit 97%) der Titelverbindung erhalten. To a solution of 6.25 g (10.25 mmol, purity 98%) of the compound of Example 14A in a mixture of 50 ml of ethyl acetate and 50 ml of ethanol under argon was added 273 mg (0.26 mmol) of palladium on charcoal (10% Pd) and 969 mg (15.37 mmol) of ammonium formate. Subsequently, the mixture was stirred at 70 ° C for 2 h. After cooling to RT, the mixture was filtered through kieselguhr, the filter residue was washed with ethyl acetate, the filtrate was concentrated and the residue was dried in vacuo. There were obtained 5.20 g (97% of theory, purity 97%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 10.40 (br. s, 1H), 8.12-8.05 (m, 2H), 7.92 (dd, 1H), 7.84 (d, 2H), 6.84 (d, 2H), 4.57-4.42 (m, 2H), 4.03 (q, 1H), 3.62-3.46 (m, 2H), 3.21 (t, 1H), 2.96- 2.83 (m, 1H), 2.55 (s, 3H), 2.17-2.04 (m, 1H), 2.03-1.91 (m, 1H), 1.78-1.66 (m, 1H), 1.60-1.48 (m, 1H), 0.39-0.25 (m, 2H), -0.17 (s, 9H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 10.40 (br.s, 1H), 8.12-8.05 (m, 2H), 7.92 (dd, 1H), 7.84 (d, 2H), 6.84 (d, 2H), 4.57-4.42 (m, 2H), 4.03 (q, 1H), 3.62-3.46 (m, 2H), 3.21 (t, 1H), 2.96-2.83 (m, 1H), 2.55 ( s, 3H), 2.17-2.04 (m, 1H), 2.03-1.91 (m, 1H), 1.78-1.66 (m, 1H), 1.60-1.48 (m, 1H), 0.39-0.25 (m, 2H), -0.17 (s, 9H).
LC/MS (Methode 1, ESIpos): Rt = 1.28 min, m/z = 508 [M+H] Beispiel 16A LC / MS (Method 1, ESIpos): R t = 1.28 min, m / z = 508 [M + H] Example 16A
2-(Trimethylsilyl)ethyl (lRS,2SR,5^ 2- (trimethylsilyl) ethyl (IRS, 2SR, 5 ^
5-[4-(tetrahydro-2/f-pyran-4-ylmefhoxy)benzoyl]cyclopentancarboxylat (Racemat)  5- [4- (tetrahydro-2 / f-pyran-4-ylmefloxy) benzoyl] cyclopentanecarboxylate (racemate)
Zu einer Lösung von 500 mg (0.96 mmol, Reinheit 97%) der Verbindung aus Beispiel 15A in 5.3 ml DMF unter Argon wurden 129 mg (1.15 mmol) Kalium-ieri.-butylat gegeben. Nach 5 min Rühren bei RT wurden 205 mg (1.15 mmol) 4-(Brommethyl)tetrahydro-2i7-pyran hinzugegeben, und das Gemisch wurde 1 h bei 100°C Badtemperatur gerührt. Nach Abkühlen und Stehenlassen über Nacht bei RT wurden dem Gemisch 60 ml Wasser und 60 ml Ethylacetat zugesetzt. Nach Trennen der Phasen wurde die wässrige Phase einmal mit 30 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumchlorid-Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde mittels Säulenchromatographie gereinigt (90 g Kieselgel, Laufmittel Cyclohexan/Ethylacetat 7:3). Es wurden 290 mg (41 % d. Th., Reinheit 82%) der Titelverbindung erhalten. LC/MS (Methode 1, ESIpos): Rt = 1.43 min, m/z = 606 [M+H]+. To a solution of 500 mg (0.96 mmol, purity 97%) of the compound from Example 15A in 5.3 ml of DMF under argon was added 129 mg (1.15 mmol) of potassium ieri-butoxide. After stirring at RT for 5 min, 205 mg (1.15 mmol) of 4- (bromomethyl) tetrahydro-2i-7-pyran were added and the mixture was stirred for 1 h at a bath temperature of 100.degree. After cooling and standing at RT overnight, 60 ml of water and 60 ml of ethyl acetate were added to the mixture. After separating the phases, the aqueous phase was extracted once with 30 ml of ethyl acetate. The combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. The residue was purified by column chromatography (90 g silica gel, eluent cyclohexane / ethyl acetate 7: 3). 290 mg (41% of theory, purity 82%) of the title compound were obtained. LC / MS (Method 1, ESIpos): R t = 1.43 min, m / z = 606 [M + H] + .
Beispiel 17A Example 17A
2-(Trimethylsilyl)ethyl (lRS,2SR,5^ 2- (trimethylsilyl) ethyl (IRS, 2SR, 5 ^
5-{4-[2-(tetrahydro-2 i-pyran-4-yl)ethoxy]benzoyl}cyclopentancarboxylat (Racemat) 5- {4- [2- (tetrahydro-2-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylate (racemate)
Zu einer Lösung von 200 mg (0.38 mmol, Reinheit 97%) der Verbindung aus Beispiel 15A in 2.1 ml DMF unter Argon wurden 51 mg (0.46 mmol) Kalium-ieri. -butylat gegeben. Nach 5 min Rühren bei RT wurden 89 mg (0.46 mmol) 4-(2-Bromethyl)tetrahydro-2i7-pyran hinzugegeben, und das Gemisch wurde 2 h bei 100°C Badtemperatur gerührt. Nach Abkühlen auf RT wurden dem Gemisch 60 ml Wasser und 60 ml Ethylacetat zugesetzt. Nach Trennen der Phasen wurde die wässrige Phase einmal mit 30 ml Ethylacetat extrahiert. Die vereinigten organischen Phasen wurden einmal mit gesättigter Natriumchlorid-Lösung gewaschen, über Magnesiumsulfat getrocknet, filtriert und eingeengt. Der Rückstand wurde mittels Säulenchromatographie gereinigt (40 g Kie- selgel, Laufmittel Cyclohexan/Ethylacetat 7:3). Es wurden 142 mg (60% d. Th., Reinheit 100%) der Titelverbindung erhalten. To a solution of 200 mg (0.38 mmol, purity 97%) of the compound from Example 15A in 2.1 ml of DMF under argon, 51 mg (0.46 mmol) of potassium ieri. given butylate. After stirring at RT for 5 min, 89 mg (0.46 mmol) of 4- (2-bromoethyl) tetrahydro-2i-7-pyran were added and the mixture was stirred for 2 h at a bath temperature of 100.degree. After cooling to RT, 60 ml of water and 60 ml of ethyl acetate were added to the mixture. After separating the phases, the aqueous phase was extracted once with 30 ml of ethyl acetate. The combined organic phases were washed once with saturated sodium chloride solution, dried over magnesium sulfate, filtered and concentrated. The residue was purified by column chromatography (40 g of silica gel, eluent cyclohexane / ethyl acetate 7: 3). There were obtained 142 mg (60% of theory, purity 100%) of the title compound.
LC/MS (Methode 1, ESIpos): Rt = 1.46 min, m/z = 620 [M+H] LC / MS (Method 1, ESIpos): R t = 1.46 min, m / z = 620 [M + H]
Ausf ührungsbeispiele : Embodiments:
Beispiel 1 example 1
(+/-)-(lRS,2SR,5RS)-2-[(4-Oxo-l,2,3-benzotria^^ (+/-) - (lRS, 2SR, 5RS) -2 - [(4-oxo-l, 2,3-benzotria ^^
ylmethoxy)benzoyl] cyclopentancarbonsäure (Racemat) ylmethoxy) benzoyl] cyclopentane carboxylic acid (racemate)
Eine Lösung von 83 mg (0.14 mmol) der Verbindung aus Beispiel 5A in 0.5 ml Dichlormethan wurde bei 0°C mit 0.25 ml (3.24 mmol) Trifluoressigsäure versetzt. Das Gemisch wurde 2.5 h bei 0°C gerührt und danach eingeengt. Der Rückstand wurde in Acetonitril aufgenommen und mittels präparativer HPLC (Methode 4) gereinigt. Es wurden 60 mg (85% d. Th., Reinheit 98%) der Titel- Verbindung erhalten. A solution of 83 mg (0.14 mmol) of the compound from Example 5A in 0.5 ml of dichloromethane was treated at 0 ° C. with 0.25 ml (3.24 mmol) of trifluoroacetic acid. The mixture was stirred for 2.5 h at 0 ° C and then concentrated. The residue was taken up in acetonitrile and purified by preparative HPLC (Method 4). There were obtained 60 mg (85% of theory, purity 98%) of the title compound.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.12 (s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.99-7.90 (m, 3H), 7.04 (d, 2H), 4.59-4.46 (m, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.38-3.32 (teilweise verdeckt, 2H), 3.23 (t, 1H), 2.94-2.81 (m, 1H), 2.17-1.95 (m, 2H), 1.95- 1.83 (m, 1H), 1.72-1.61 (m, 3H), 1.57-1.45 (m, 1H), 1.33 (qd, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.06 min, m/z = 492 [M+H]+. Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.12 (s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.99-7.90 ( m, 3H), 7.04 (d, 2H), 4.59-4.46 (m, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.38-3.32 (partially obscured , 2H), 3.23 (t, 1H), 2.94-2.81 (m, 1H), 2.17-1.95 (m, 2H), 1.95-1.83 (m, 1H), 1.72-1.61 (m, 3H), 1.57-1.45 (m, 1H), 1.33 (qd, 2H). LC / MS (Method 1, ESIpos): R t = 1.06 min, m / z = 492 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
30 mg der racemischen Verbindung aus Beispiel 1 wurden in 12 ml Methanol/ Acetonitril in der Wärme gelöst und mittels präparativer SFC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 2 und 3) [Säule: Daicel Chiralpak AZ-H, 5 μιη, 250 mm x 20 mm; Fluss: 80 ml/min; Detektion: 210 nm; Injektionsvolumen: 1.0 ml; Temperatur: 40°C; Eluent: 60% Kohlendioxid / 40% Ethanol].  30 mg of the racemic compound from Example 1 were dissolved in 12 ml of methanol / acetonitrile under heat and separated by means of preparative SFC on chiral phase into the enantiomers (see Examples 2 and 3) [column: Daicel Chiralpak AZ-H, 5 μιη, 250 mm x 20 mm; Flow: 80 ml / min; Detection: 210 nm; Injection volume: 1.0 ml; Temperature: 40 ° C; Eluent: 60% carbon dioxide / 40% ethanol].
Beispiel 2 Example 2
(+)-(lR5,25R,5R5)-2-[(4-Oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetrahydro-2ii-pyran-4-yl- methoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 1) Ausbeute: 14 mg; ehem. Reinheit = 100%; ee-Wert = 99% [a]D 20 = +66.9°, 589 nm, c = 0.27 g/100 ml, Chloroform (+) - (1R5,25R, 5R5) -2 - [(4-Oxo-1,2,3-benzotriazine-3 (4 /) -yl) -methyl] -5- [4- (tetrahydro-2ii-pyran 4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 1) Yield: 14 mg; former purity = 100%; ee value = 99% [α] D 20 = + 66.9 °, 589 nm, c = 0.27 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.10 (br. s, 1H), 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99-7.89 (m, 3H), 7.04 (d, 2H), 4.59-4.46 (m, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.32-3.19 (m, teilweise verdeckt, 3H), 2.94-2.80 (m, 1H), 2.18-1.96 (m, 2H), 1.95-1.84 (m, 1H), 1.72-1.61 (m, 3H), 1.58-1.44 (m, 1H), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.10 (br.s, 1H), 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99 -7.89 (m, 3H), 7.04 (d, 2H), 4.59-4.46 (m, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.32-3.19 (m, partially occluded, 3H), 2.94-2.80 (m, 1H), 2.18-1.96 (m, 2H), 1.95-1.84 (m, 1H), 1.72-1.61 (m, 3H), 1.58-1.44 (m , 1H), 1.33 (qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.04 min, m/z = 492 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.04 min, m / z = 492 [M + H] + .
Beispiel 3 Example 3
(-)-(lR5,25R,5R^-2-[(4-Oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetrahydro-2ii-pyran-4-yl- methoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 2) (-) - (1R5,25R, 5R ^ -2 - [(4-Oxo-l, 2,3-benzotriazine-3 (4 /) - yl) methyl] -5- [4- (tetrahydro-2ii-pyran -4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 2)
Ausbeute: 17 mg; ehem. Reinheit = 100%; ee-Wert = 99% Yield: 17 mg; former purity = 100%; ee value = 99%
[a]D 20 = -56.4°, 589 nm, c = 0.28 g/100 ml, Chloroform [a] D 20 = -56.4 °, 589 nm, c = 0.28 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.07 (br. s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.99-7.89 (m, 3H), 7.04 (d, 2H), 4.53 (dd, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.34-3.28 (m, teilweise verdeckt, 2H), 3.24 (t, 1H), 2.98-2.81 (m, 1H), 2.17-1.96 (m, 2H), 1.95- 1.83 (m, 1H), 1.73-1.60 (m, 3H), 1.57-1.45 (m, 1H), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.07 (br.s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.99- 7.89 (m, 3H), 7.04 (d, 2H), 4.53 (dd, 2H), 4.14-4.05 (m, 1H), 3.93 (d, 2H), 3.87 (dd, 2H), 3.34-3.28 (m, partially obscured, 2H), 3.24 (t, 1H), 2.98-2.81 (m, 1H), 2.17-1.96 (m, 2H), 1.95- 1.83 (m, 1H), 1.73-1.60 (m, 3H), 1.57 -1.45 (m, 1H), 1.33 (qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.04 min, m/z = 492 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.04 min, m / z = 492 [M + H] + .
Beispiel 4 Example 4
(+/-)-(lR5,25R,5R5)-2-[(4-Oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2-(tetrahydro-2ii-pyran- 4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Racemat) (+/-) - (1R5,25R, 5R5) -2 - [(4-Oxo-1,2,3-benzotriazine-3 (4 /) - yl) methyl] -5- {4- [2- (2H) tetrahydro-2ii-pyran-4-yl) ethoxy] benzoyl} cyclopentane carboxylic acid (racemate)
Eine Lösung von 2.91 g (4.75 mmol, Reinheit 99%) der Verbindung aus Beispiel 6A in 16 ml Dichlormethan wurde bei 0°C mit 8.0 ml (104 mmol) Trifluoressigsäure versetzt und 2 h bei 0°C gerührt. Anschließend wurde das Gemisch eingeengt und der Rückstand im Vakuum getrocknet. Nach Zugabe von wenig Ethylacetat wurde ein Feststoff erhalten, welcher abfiltriert, einmal mit wenig Ethylacetat und Pentan nachgewaschen und im Vakuum getrocknet wurde. Es wurden so 2.11 g (88% d. Th., Reinheit 100%) einer ersten Charge der Titelverbindung erhalten. Die verbliebene Mutterlauge wurde eingeengt und der Rückstand mittels präparativer HPLC gereinigt [Säule: Kinetix C18, 5 μιη, 100 mm x 21.2 mm; Fluss: 25 ml/min; Detektion: 210 nm; Injektionsvolumen: 0.5 ml; Temperatur: 40°C; Eluent: 44% Wasser / 45% Acetonitril / 11 % Ameisensäure in Wasser, isokratisch über 8 min] . Es wurden auf diese Weise 52 mg (2% d. Th., Reinheit 100%) einer zweiten Charge der Titelverbindung erhalten. A solution of 2.91 g (4.75 mmol, purity 99%) of the compound from Example 6A in 16 ml of dichloromethane was admixed at 0 ° C. with 8.0 ml (104 mmol) of trifluoroacetic acid and stirred at 0 ° C. for 2 h. The mixture was then concentrated and the residue was dried in vacuo. After adding a little ethyl acetate, a solid was obtained which was filtered off, rinsed once with a little ethyl acetate and pentane and dried in vacuo. This gave 2.11 g (88% of theory, purity 100%) of a first batch of the title compound. The remaining mother liquor was concentrated and the residue was purified by preparative HPLC [column: Kinetix C18, 5 μm, 100 mm × 21.2 mm; Flow: 25 ml / min; Detection: 210 nm; Injection volume: 0.5 ml; Temperature: 40 ° C; Eluent: 44% water / 45% acetonitrile / 11% formic acid in water, isocratic over 8 min]. 52 mg (2% of theory, purity 100%) of a second batch of the title compound were obtained in this way.
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.14 (br. s, 1H), 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99-7.90 (m, 3H), 7.04 (d, 2H), 4.58-4.47 (m, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.34-3.19 (m, 3H), 2.94-2.81 (m, 1H), 2.16-2.04 (m, 1H), 1.95-1.83 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.45 (m, 1H), 1.29-1.14 (m, 2H). Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.14 (br.s, 1H), 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99 -7.90 (m, 3H), 7.04 (d, 2H), 4.58-4.47 (m, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.34-3.19 (m, 3H), 2.94 -2.81 (m, 1H), 2.16-2.04 (m, 1H), 1.95-1.83 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.45 (m, 1H), 1.29-1.14 (m, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.05 min, m/z = 506 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.05 min, m / z = 506 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
2.00 g der racemischen Verbindung aus Beispiel 4 wurden in 20 ml Dioxan angelöst, mit 180 ml eines Methanol/ Acetonitril-Gemisches versetzt, in der Wärme in Lösung gebracht und anschließend mittels präparativer SFC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 5 und 6) [Säule: Daicel Chiralpak AY-H, 5 μιη, 250 mm x 20 mm; Fluss: 80 ml/min; Detektion: 210 nm; Injektionsvolumen: 1.2 ml; Temperatur: 40°C; Eluent: 70% Kohlendioxid / 30% Ethanol, Laufzeit 16 min] . 2.00 g of the racemic compound from Example 4 were dissolved in 20 ml of dioxane, mixed with 180 ml of a methanol / acetonitrile mixture, brought into solution in the heat and then separated by means of preparative SFC on a chiral phase into the enantiomers (see Examples 5 and 6 ) [Column: Daicel Chiralpak AY-H, 5 μm, 250 mm × 20 mm; Flow: 80 ml / min; Detection: 210 nm; Injection volume: 1.2 ml; Temperature: 40 ° C; Eluent: 70% carbon dioxide / 30% ethanol, running time 16 min].
Beispiel 5 Example 5
(+)-(lR5,25R,5R5)-2-[(4-Oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2-(tetrahydro-2ii-pyran- 4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Enantiomer 1) (+) - (lR5,25R, 5R5) -2 - [(4-Oxo-l, 2,3-benzotriazine-3 (4 /) - yl) methyl] -5- {4- [2- (tetrahydrofuran) 2ii-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylic acid (enantiomer 1)
Es wurden 910 mg (ehem. Reinheit = 97%, ee-Wert = 100%) der Titelverbindung erhalten, welche in 20 ml Acetonitril aufgenommen und nochmals chromatographisch nachgereinigt wurden [Säule: Kinetix C18, 5 μιη, 100 mm x 30 mm; Fluss: 60 ml/min; Detektion: 210 nm; Injektionsvolumen: 1.0 ml; Temperatur: 30°C; Eluent: 45% Wasser / 50% Acetonitril / 5% Ameisensäure in Wasser, isokratisch über 4 min] . Es wurden so 850 mg der Titelverbindung in einer ehem. Reinheit von 100% erhalten. [a]D 20 = +71.0°, 589 nm, c = 0.37 g/100 ml, Chloroform Ή-NMR (500 MHz, DMSO-d6): δ [ppm] = 12.13 (br. s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.98-7.90 (m, 3H), 7.04 (d, 2H), 4.58-4.47 (m, 2H), 4.14-4.05 (m, 3H), 3.83 (dd, 2H), 3.32- 3.20 (m, teilweise verdeckt, 3H), 2.88 (sext, 1H), 2.15-2.05 (m, 1H), 1.93-1.84 (m, 1H), 1.76-1.58 (m, 6H), 1.56-1.47 (m, 1H), 1.27-1.16 (m, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.07 min, m/z = 506 [M+H]+. 910 mg (former purity = 97%, ee value = 100%) of the title compound were obtained, which were taken up in 20 ml of acetonitrile and further purified by chromatography [column: Kinetix C18, 5 μm, 100 mm × 30 mm; Flow: 60 ml / min; Detection: 210 nm; Injection volume: 1.0 ml; Temperature: 30 ° C; Eluent: 45% water / 50% acetonitrile / 5% formic acid in water, isocratic over 4 min]. There were thus obtained 850 mg of the title compound in a former purity of 100%. [a] D 20 = + 71.0 °, 589 nm, c = 0.37 g / 100 ml, chloroform Ή-NMR (500 MHz, DMSO-d 6 ): δ [ppm] = 12.13 (br.s, 1H), 8.26 (dd, 1H), 8.20 (d, 1H), 8.08 (td, 1H), 7.98- 7.90 (m, 3H), 7.04 (d, 2H), 4.58-4.47 (m, 2H), 4.14-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, partially obscured, 3H) , 2.88 (sext, 1H), 2.15-2.05 (m, 1H), 1.93-1.84 (m, 1H), 1.76-1.58 (m, 6H), 1.56-1.47 (m, 1H), 1.27-1.16 (m, 2H). LC / MS (Method 1, ESIpos): R t = 1.07 min, m / z = 506 [M + H] + .
Beispiel 6 Example 6
(-)-(lR5,25R,5R^-2-[(4-Oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2-(tetrahydro-2ii-pyran-4- yl)ethoxy]benzoyl}cyclopentancarbonsäure (Enantiomer 2) (-) - (1R5,25R, 5R ^ -2 - [(4-Oxo-l, 2,3-benzotriazine-3 (4 /) - yl) methyl] -5- {4- [2- (tetrahydro-) 2ii-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylic acid (enantiomer 2)
Ausbeute: 903 mg; ehem. Reinheit = 100%; ee-Wert = 100% [a]D 20 = -70.1°, 589 nm, c = 0.35 g/100 ml, Chloroform Yield: 903 mg; former purity = 100%; ee value = 100% [α] D 20 = -70.1 °, 589 nm, c = 0.35 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99- 7.90 (m, 3H), 7.04 (d, 2H), 4.59-4.47 (m, 2H), 4.14-4.06 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.87 (sext, 1H), 2.17-2.04 (m, 1H), 1.95-1.83 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.45 (m, 1H), 1.29-1.15 (m, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.05 min, m/z = 506 [M+H]+. Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 8.26 (d, 1H), 8.20 (d, 1H), 8.11-8.05 (m, 1H), 7.99-7.90 (m, 3H), 7.04 (d, 2H), 4.59-4.47 (m, 2H), 4.14-4.06 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.87 (sec, 1H), 2.17- 2.04 (m, 1H), 1.95-1.83 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.45 (m, 1H), 1.29-1.15 (m, 2H). LC / MS (Method 1, ESIpos): R t = 1.05 min, m / z = 506 [M + H] + .
Beispiel 7 Example 7
(+/-)-(lRS,2SR,5RS)-2-{ [4-Oxo-6-(trifl^^ (+/-) - (lRS, 2SR, 5RS) -2- {[4-oxo-6- (trifl ^^
hydro-2 i-pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Racemat) hydro-2-pyran-4-ylmethoxy) benzoyl] cyclopentane carboxylic acid (racemate)
Eine Lösung von 585 mg (0.89 mmol) der Verbindung aus Beispiel 9A in 3 ml Dichlormethan wurde bei 0°C mit 1.5 ml (19.47 mmol) Trifluoressigsäure versetzt. Das Gemisch wurde 5.5 h bei 0°C gerührt und danach eingeengt. Der Rückstand wurde in 5 ml Acetonitril aufgenommen. Dabei fiel ein Feststoff aus, welcher abfiltriert und im Vakuum getrocknet wurde. Es wurden 468 mg (95% d. Th., Reinheit 100%) der Titelverbindung erhalten. Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.13 (s, IH), 8.51 (s, IH), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.38-3.28 (verdeckt, 2H), 3.24 (t, IH), 2.94-2.79 (m, IH), 2.18-1.87 (m, 3H), 1.73-1.61 (m, 3H), 1.59-1.46 (m, IH), 1.33 (qd, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.17 min, m/z = 660 [M+H]+. A solution of 585 mg (0.89 mmol) of the compound from Example 9A in 3 ml of dichloromethane was admixed at 0 ° C. with 1.5 ml (19.47 mmol) of trifluoroacetic acid. The mixture was stirred for 5.5 h at 0 ° C and then concentrated. The residue was taken up in 5 ml of acetonitrile. This precipitated a solid, which was filtered off and dried in vacuo. There were obtained 468 mg (95% of theory, purity 100%) of the title compound. Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.13 (s, IH), 8.51 (s, IH), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 ( d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.38-3.28 (covert, 2H), 3.24 (t, IH) , 2.94-2.79 (m, IH), 2.18-1.87 (m, 3H), 1.73-1.61 (m, 3H), 1.59-1.46 (m, IH), 1.33 (qd, 2H). LC / MS (Method 1, ESIpos): R t = 1.17 min, m / z = 660 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
465 mg der racemischen Verbindung aus Beispiel 7 wurden in 15 ml DMSO und 30 ml Ethanol gelöst und mittels präparativer SFC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 8 und 9) [Säule: Daicel Chiralpak AY, 20 μιη, 250 mm x 30 mm; Fluss: 175 ml/min; Detektion: 210 nm; Injektionsvolumen: 1.3 ml; Temperatur: 38°C; Eluent: 75% Kohlendioxid / 25% Ethanol, Laufzeit 16.5 min].  465 mg of the racemic compound from Example 7 were dissolved in 15 ml of DMSO and 30 ml of ethanol and separated into the enantiomers by means of preparative SFC on a chiral phase (see Examples 8 and 9) [column: Daicel Chiralpak AY, 20 μm, 250 mm × 30 mm; Flow: 175 ml / min; Detection: 210 nm; Injection volume: 1.3 ml; Temperature: 38 ° C; Eluent: 75% carbon dioxide / 25% ethanol, running time 16.5 min].
Beispiel 8 Example 8
(+)-(lRS,2SR,5RS)-2-{ [4-Oxo-6-(trifluom^ (+) - (IRS, 2SR, 5RS) -2- {[4-oxo-6- (trifluoro ^
hydro-2 i-pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 1) Ausbeute: 239 mg; ehem. Reinheit = 100%; ee-Wert = 100% [ab20 = +80.2°, 589 nm, c = 0.31 g/100 ml, Chloroform hydro-2-pyran-4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 1) Yield: 239 mg; former purity = 100%; ee value = 100% [from 20 = + 80.2 °, 589 nm, c = 0.31 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.13 (br. s, IH), 8.51 (s, IH), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (verdeckt, 2H), 3.24 (t, IH), 2.94-2.80 (m, IH), 2.17-1.88 (m, 3H), 1.72-1.61 (m, 3H), 1.58-1.46 (m, IH), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.13 (br.s, IH), 8.51 (s, IH), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (obscured, 2H), 3.24 (t, IH), 2.94-2.80 (m, IH), 2.17-1.88 (m, 3H), 1.72-1.61 (m, 3H), 1.58-1.46 (m, IH), 1.33 (qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.17 min, m/z = 660 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.17 min, m / z = 660 [M + H] + .
Beispiel 9 Example 9
(-)-(lRS,2SR,5R^-2-{ [4-Oxo-6-(trifluorme (-) - (IRS, 2SR, 5R ^ -2- {[4-oxo-6- (trifluoromethylene)
hydro-2 i-pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 2) Ausbeute: 228 mg; ee-Wert = 100% hydro-2-pyran-4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 2) Yield: 228 mg; ee value = 100%
[a]D 20 = -88.9°, 589 nm, c = 0.31 g/100 ml, Chloroform [a] D 20 = -88.9 °, 589 nm, c = 0.31 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.13 (br. s, IH), 8.51 (s, IH), 8.46-8.37 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.14-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (ver- deckt, 2H), 3.23 (t, 1H), 2.94-2.80 (m, 1H), 2.17-1.87 (m, 3H), 1.73-1.61 (m, 3H), 1.58-1.46 (m, 1H), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.13 (br.s, IH), 8.51 (s, IH), 8.46-8.37 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.14-4.05 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 ( covers, 2H), 3.23 (t, 1H), 2.94-2.80 (m, 1H), 2.17-1.87 (m, 3H), 1.73-1.61 (m, 3H), 1.58-1.46 (m, 1H), 1.33 ( qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.17 min, m/z = 660 [M+H]+. Beispiel 10 (+/-)-(lRS,2SR,5RS)-2- { [4-Oxo-6-(trifluormethyl)- 1 ,2,3-benzotriazin-3(4//)-yl] methyl } -5- { 4- [2- (tetrahydro-2/i-pyran-4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Racemat) LC / MS (Method 1, ESIpos): R t = 1.17 min, m / z = 660 [M + H] + . Example 10 (+/-) - (IRS, 2SR, 5RS) -2- {[4-Oxo-6- (trifluoromethyl) -1,2,3-benzotriazine-3 (4 //) - yl] methyl} - 5- {4- [2- (tetrahydro-2-i-pyran-4-yl) ethoxy] benzoyl} cyclopentane carboxylic acid (racemate)
Eine Lösung von 135 mg (0.20 mmol) der Verbindung aus Beispiel 10A in 0.7 ml Dichlormethan wurde bei 0°C mit 0.35 ml (4.54 mmol) Trifluoressigsäure versetzt. Das Gemisch wurde 2.5 h bei 0°C gerührt und danach eingeengt. Der Rückstand wurde in 2 ml Acetonitril aufgenommen und mittels präparativer HPLC (Methode 5) gereinigt. Es wurden 91 mg (80% d. Th., Reinheit 100%) der Titelverbindung erhalten. A solution of 135 mg (0.20 mmol) of the compound from Example 10A in 0.7 ml of dichloromethane was treated at 0 ° C with 0.35 ml (4.54 mmol) of trifluoroacetic acid. The mixture was stirred for 2.5 h at 0 ° C and then concentrated. The residue was taken up in 2 ml of acetonitrile and purified by preparative HPLC (Method 5). 91 mg (80% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.14-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93-2.81 (m, 1H), 2.17-2.03 (m, 1H), 2.00-1.88 (m, 1H), 1.76-1.45 (m, 7H), 1.29-1.14 (m, 2H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 ( d, 2H), 4.57 (d, 2H), 4.14-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93-2.81 (m, 1H), 2.17-2.03 ( m, 1H), 2.00-1.88 (m, 1H), 1.76-1.45 (m, 7H), 1.29-1.14 (m, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.21 min, m/z = 574 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.21 min, m / z = 574 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
83 mg der racemischen Verbindung aus Beispiel 10 wurden in 2 ml Ethanol gelöst und mittels präparativer HPLC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 11 und 12) [Säule: Daicel Chiralpak AY-H, 5 μιη, 250 mm x 20 mm; Fluss: 15 ml/min; Detektion: 220 nm; Injektionsvolumen: 1 ml; Temperatur: 45°C; Eluent: t = 0-15 min 25% Isohexan / 75% Ethanol + 0.2% Essigsäure].  83 mg of the racemic compound from Example 10 were dissolved in 2 ml of ethanol and separated into the enantiomers by preparative HPLC on a chiral phase (see Examples 11 and 12) [column: Daicel Chiralpak AY-H, 5 μm, 250 mm × 20 mm; Flow: 15 ml / min; Detection: 220 nm; Injection volume: 1 ml; Temperature: 45 ° C; Eluent: t = 0-15 min 25% isohexane / 75% ethanol + 0.2% acetic acid].
Beispiel 11 Example 11
(+)-(lRS,2SR,5RS)-2- { [4-Oxo-6-(trifluormefhyl)- 1 ,2,3-benzotriazin-3(4//)-yl] methyl } -5- { 4- [2- (tetrahydro-2 i-pyran-4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Enantiomer 1) Ausbeute: 38 mg; ee-Wert = 100% (+) - (IRS, 2SR, 5RS) -2- {[4-Oxo-6- (trifluoromethyl) -1,3,3-benzotriazine-3 (4 //) - yl] methyl} -5- {4 - [2- (tetrahydro-2-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylic acid (enantiomer 1) Yield: 38 mg; ee value = 100%
[a]D 20 = +70.7°, 589 nm, c = 0.10 g/100 ml, Chloroform [a] D 20 = + 70.7 °, 589 nm, c = 0.10 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.14-4.06 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.91-2.83 (m, 1H), 2.16-2.04 (m, 1H), 1.99-1.88 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.47 (m, 1H), 1.29-1.15 (m, 2H). Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (i.e. , 2H), 4.57 (d, 2H), 4.14-4.06 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.91-2.83 (m, 1H), 2.16-2.04 (m , 1H), 1.99-1.88 (m, 1H), 1.75-1.58 (m, 6H), 1.57-1.47 (m, 1H), 1.29-1.15 (m, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.22 min, m/z = 574 [M+H]+. Beispiel 12 LC / MS (Method 1, ESIpos): R t = 1.22 min, m / z = 574 [M + H] + . Example 12
(-)-(lR5,25R,5R^-2-{ [4-Oxo-6-(trifluormethyl)-l,2,3-benzotriazin-3(4 ^-yl]methyl }-5-{4-[2- (tetrahydro-2/i-pyran-4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Enantiomer 2) (-) - (1R5,25R, 5R ^ -2- {[4-Oxo-6- (trifluoromethyl) -l, 2,3-benzotriazine-3 (4 ^ -yl] methyl} -5- {4- [ 2- (tetrahydro-2-i-pyran-4-yl) ethoxy] benzoyl} cyclopentane carboxylic acid (enantiomer 2)
Ausbeute: 45 mg; ee-Wert = 100% Yield: 45 mg; ee value = 100%
[a]D 20 = -77.1°, 589 nm, c = 0.37 g/100 ml, Chloroform [α] D 20 = -77.1 °, 589 nm, c = 0.37 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 (d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93-2.81 (m, 1H), 2.17-2.04 (m, 1H), 1.99-1.87 (m, 1H), 1.74-1.58 (m, 6H), 1.58-1.47 (m, 1H), 1.29-1.15 (m, 2H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.13 (s, 1H), 8.51 (s, 1H), 8.46-8.38 (m, 2H), 7.96 (d, 2H), 7.04 ( d, 2H), 4.57 (d, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93-2.81 (m, 1H), 2.17-2.04 ( m, 1H), 1.99-1.87 (m, 1H), 1.74-1.58 (m, 6H), 1.58-1.47 (m, 1H), 1.29-1.15 (m, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.22 min, m/z = 574 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.22 min, m / z = 574 [M + H] + .
Beispiel 13 Example 13
(+/-)-(lR5,25R,5R5)-2-{ [4-Oxo-6-(trifluormethyl)-l,2,3-benzotriazin-3(4 /)-yl]methyl}-5-{4- [(tetrahydro-2i7-pyran-4-ylmefhyl)sulfanyl]benzoyl Jcyclopentancarbonsäure (Racemat) (+/-) - (1R5,25R, 5R5) -2- {[4-oxo-6- (trifluoromethyl) -1,3,3-benzotriazine-3 (4 /) -yl] methyl} -5- { 4- [(tetrahydro-2-yl-pyran-4-ylmethyl) sulfanyl] benzoyl-1-cyclopentanecarboxylic acid (racemate)
Eine Lösung von 249 mg (0.35 mmol, Reinheit 95%) der Verbindung aus Beispiel 13A in 3.5 ml Dichlormethan wurde bei 0°C mit 1.75 ml Trifluoressigsäure versetzt. Das Gemisch wurde zu- nächst 15 min bei 0°C und dann 1 h bei RT gerührt und anschließend eingeengt. Der Rückstand wurde mittels präparativer HPLC (Methode 4) gereinigt. Es wurden 163 mg (81 % d. Th., Reinheit 100%) der Titelverbindung erhalten. A solution of 249 mg (0.35 mmol, purity 95%) of the compound from Example 13A in 3.5 ml of dichloromethane was treated at 0 ° C with 1.75 ml of trifluoroacetic acid. The mixture was added stirred for 15 min at 0 ° C and then for 1 h at RT and then concentrated. The residue was purified by preparative HPLC (Method 4). 163 mg (81% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.16 (br. s, 1H), 8.51 (s, 1H), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.62-4.52 (m, 2H), 4.15-4.06 (m, 1H), 3.83 (dd, 2H), 3.30-3.19 (m, 3H), 3.02 (d, 2H), 2.94-2.81 (m, 1H), 2.20-2.04 (m, 1H), 2.01-1.87 (m, 1H), 1.84-1.61 (m, 4H), 1.60-1.45 (m, 1H), 1.35-1.17 (m, 2H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.16 (br.s, 1H), 8.51 (s, 1H), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.62-4.52 (m, 2H), 4.15-4.06 (m, 1H), 3.83 (dd, 2H), 3.30-3.19 (m, 3H), 3.02 (d, 2H), 2.94- 2.81 (m, 1H), 2.20-2.04 (m, 1H), 2.01-1.87 (m, 1H), 1.84-1.61 (m, 4H), 1.60-1.45 (m, 1H), 1.35-1.17 (m, 2H ).
LC/MS (Methode 1, ESIpos): Rt = 1.20 min, m/z = 576 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.20 min, m / z = 576 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
150 mg der racemischen Verbindung aus Beispiel 13 wurden in 3 ml Acetonitril/Ethanol gelöst und mittels präparativer HPLC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 14 und 15) [Säule: Daicel Chiralpak AS-H, 5 μιη, 250 mm x 4.6 mm; Fluss: 20 ml/min; Detektion: 230 nm; Injektionsvolumen: 0.06 ml; Temperatur: 25°C; Eluent: t = 0-16 min 20% Ethanol / 76% Acetonitril / 4% 5%-ige Essigsäure in Acetonitril] . Beispiel 14 150 mg of the racemic compound from Example 13 were dissolved in 3 ml of acetonitrile / ethanol and separated into the enantiomers by preparative HPLC on a chiral phase (see Examples 14 and 15) [column: Daicel Chiralpak AS-H, 5 μm, 250 mm × 4.6 mm; Flow: 20 ml / min; Detection: 230 nm; Injection volume: 0.06 ml; Temperature: 25 ° C; Eluent: t = 0-16 min 20% ethanol / 76% acetonitrile / 4% 5% acetic acid in acetonitrile]. Example 14
(-)-(lRS,2SR,5R^-2-{ [4-Oxo-6-(trifluorme (-) - (IRS, 2SR, 5R ^ -2- {[4-oxo-6- (trifluoromethylene)
hydro-2 i-pyran-4-ylmethyl)sulfanyl]benzoyl}cyclopentancarbonsäure (Enantiomer 1) hydro-2-pyran-4-ylmethyl) sulfanyl] benzoyl} cyclopentanecarboxylic acid (enantiomer 1)
Ausbeute: 59 mg; ehem. Reinheit = 100%; ee-Wert = 100% Yield: 59 mg; former purity = 100%; ee value = 100%
[a]D 20 = -85.6°, 589 nm, c = 0.39 g/100 ml, Chloroform Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.15 (br. s, 1H), 8.51 (s, 1H), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.63-4.51 (m, 2H), 4.16-4.03 (m, 1H), 3.83 (dd, 2H), 3.29-3.19 (m, 3H), 3.02 (d, 2H), 2.94-2.81 (m, 1H), 2.18-2.05 (m, 1H), 2.02-1.86 (m, 1H), 1.83-1.61 (m, 3H), 1.59-1.44 (m, 1H), 1.36-1.19 (m, 2H). [a] D 20 = -85.6 °, 589 nm, c = 0.39 g / 100 ml, chloroform Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.15 (br. s, 1H), 8.51 (s, 1H), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.63-4.51 (m, 2H), 4.16-4.03 (m, 1H), 3.83 (dd , 2H), 3.29-3.19 (m, 3H), 3.02 (d, 2H), 2.94-2.81 (m, 1H), 2.18-2.05 (m, 1H), 2.02-1.86 (m, 1H), 1.83-1.61 (m, 3H), 1.59-1.44 (m, 1H), 1.36-1.19 (m, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.21 min, m/z = 576 [M+H]+. Beispiel 15 LC / MS (Method 1, ESIpos): R t = 1.21 min, m / z = 576 [M + H] + . Example 15
(+)-(lRS,2SR,5RS)-2- { [4-Oxo-6-(trifluormethyl)- 1 ,2,3-benzotriazin-3(4 /)-yl] methyl } -5- { 4- [(tetra- hydro-2 i-pyran-4-ylmethyl)sulfanyl]benzoyl}cyclopentancarbonsäure (Enantiomer 2) (+) - (IRS, 2SR, 5RS) -2- {[4-Oxo-6- (trifluoromethyl) -1,2,3-benzotriazine-3 (4 /) -yl] methyl} -5- {4- [(tetrahydro-2-pyran-4-ylmethyl) sulfanyl] benzoyl} cyclopentanecarboxylic acid (enantiomer 2)
Ausbeute: 61 mg; ehem. Reinheit = 100%; ee-Wert [a]D 20 = +53.1°, 589 nm, c = 0.16 g/100 ml, Chloroform Yield: 61 mg; former purity = 100%; ee [a] D 20 = + 53.1 °, 589 nm, c = 0.16 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.14 (br. s, IH), 8.51 (s, IH), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.63-4.50 (m, 2H), 4.16-4.05 (m, IH), 3.83 (dd, 2H), 3.29-3.17 (m, 3H), 3.02 (d, 2H), 2.94-2.77 (m, IH), 2.18-2.04 (m, IH), 2.02-1.86 (m, IH), 1.83-1.62 (m, 3H), 1.60-1.45 (m, IH), 1.35-1.20 (m, 2H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.14 (br.s, IH), 8.51 (s, IH), 8.46-8.37 (m, 2H), 7.90 (d, 2H), 7.41 (d, 2H), 4.63-4.50 (m, 2H), 4.16-4.05 (m, IH), 3.83 (dd, 2H), 3.29-3.17 (m, 3H), 3.02 (d, 2H), 2.94- 2.77 (m, IH), 2.18-2.04 (m, IH), 2.02-1.86 (m, IH), 1.83-1.62 (m, 3H), 1.60-1.45 (m, IH), 1.35-1.20 (m, 2H ).
LC/MS (Methode 1, ESIpos): Rt = 1.21 min, m/z = 576 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.21 min, m / z = 576 [M + H] + .
Beispiel 16 Example 16
(+/-)-(lR5,25R,5R5)-2-[(6-Methyl-4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetrahydro-2ii- pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Racemat) (+/-) - (1R5,25R, 5R5) -2 - [(6-methyl-4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) -methyl] -5- [4- (tetrahydro-2-pyran-4-ylmethoxy) benzoyl] cyclopentane carboxylic acid (racemate)
Eine Lösung von 290 mg (0.39 mmol, Reinheit 82%) der Verbindung aus Beispiel 16A in 1.3 ml Dichlormethan wurde bei 0°C mit 0.7 ml (8.65 mmol) Trifluoressigsäure versetzt. Das Gemisch wurde 1 h bei RT gerührt und anschließend eingeengt. Der Rückstand wurde mittels präparativer HPLC (Methode 4) gereinigt. Es wurden 153 mg (77% d. Th., Reinheit 100%) der Titelverbindung erhalten. A solution of 290 mg (0.39 mmol, purity 82%) of the compound from Example 16A in 1.3 ml of dichloromethane was treated at 0 ° C with 0.7 ml (8.65 mmol) of trifluoroacetic acid. The mixture was stirred at RT for 1 h and then concentrated. The residue was purified by preparative HPLC (Method 4). 153 mg (77% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.12 (br. s, IH), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.13-4.04 (m, IH), 3.93 (d, 2H), 3.88 (dd, 2H), 3.37-3.28 (m, 2H), 3.23 (t, IH), 2.93-2.80 (m, IH), 2.55 (s, 3H), 2.16-1.95 (m, 2H), 1.93-1.82 (m, IH), 1.71-1.61 (m, 3H), 1.50 (dd, IH), 1.33 (qd, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.06 min, m/z = 506 [M+H]+. Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.12 (br.s, IH), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.13-4.04 (m, IH), 3.93 (d, 2H), 3.88 (dd, 2H), 3.37-3.28 (m, 2H), 3.23 (t, IH), 2.93-2.80 (m, IH), 2.55 (s, 3H), 2.16-1.95 (m, 2H), 1.93-1.82 (m, IH), 1.71-1.61 (m, 3H), 1.50 (dd, IH), 1.33 (qd, 2H). LC / MS (Method 1, ESIpos): R t = 1.06 min, m / z = 506 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
144 mg der racemischen Verbindung aus Beispiel 16 wurden in 11 ml Ethanol gelöst und mittels präparativer SFC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 17 und 18) [Säule: Phenomenex Amylose Π, 5 μιη, 250 mm x 20 mm; Fluss: 100 ml/min; Detektion: 210 nm; Injek- tionsvolumen: 0.40 ml; Temperatur: 40°C; Eluent: 65% Kohlendioxid / 35% Ethanol, Laufzeit 15 min] . 144 mg of the racemic compound from Example 16 were dissolved in 11 ml of ethanol and separated into the enantiomers by means of preparative SFC on a chiral phase (see Examples 17 and 18) [column: Phenomenex amylose®, 5 μm, 250 mm × 20 mm; Flow: 100 ml / min; Detection: 210 nm; injections volume: 0.40 ml; Temperature: 40 ° C; Eluent: 65% carbon dioxide / 35% ethanol, running time 15 min].
Beispiel 17 Example 17
(+)-(lR5,25R,5R5)-2-[(6-Methyl-4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetrahydro-2ii- pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 1) (+) - (IR 5,25 R, 5 R 5) -2 - [(6-Methyl-4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) methyl] -5- [4- (tetrahydro 2-pyran-4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 1)
Ausbeute: 63 mg; ehem. Reinheit = 94%; ee-Wert = 100% Yield: 63 mg; former purity = 94%; ee value = 100%
[ab20 = +68.4°, 589 nm, c = 0.39 g/100 ml, Chloroform [from 20 = + 68.4 °, 589 nm, c = 0.39 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.12 (br. s, IH), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.14-4.04 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (teil- weise verdeckt, 2H), 3.23 (t, IH), 2.92-2.80 (m, IH), 2.55 (s, 3H), 2.16-1.94 (m, 2H), 1.93-1.82 (m, IH), 1.67 (d, 3H), 1.56-1.44 (m, IH), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.12 (br.s, IH), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.14-4.04 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (partially obscured, 2H), 3.23 (t, IH), 2.92-2.80 (m, IH), 2.55 (s, 3H), 2.16-1.94 (m, 2H), 1.93-1.82 (m, IH), 1.67 (d, 3H), 1.56-1.44 (m, IH), 1.33 (qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.07 min, m/z = 506 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.07 min, m / z = 506 [M + H] + .
Beispiel 18 Example 18
(-)-(lR5,25R,5R^-2-[(6-Methyl-4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-[4-(tetrahydro-2ii- pyran-4-ylmethoxy)benzoyl]cyclopentancarbonsäure (Enantiomer 2) (-) - (1R5,25R, 5R ^ -2 - [(6-Methyl-4-oxo-l, 2,3-benzotriazine-3 (4 /) -yl) methyl] -5- [4- (tetrahydro 2-pyran-4-ylmethoxy) benzoyl] cyclopentanecarboxylic acid (enantiomer 2)
Ausbeute: 63 mg; ehem. Reinheit = 100%; ee-Wert = 100% Yield: 63 mg; former purity = 100%; ee value = 100%
[a]D 20 = -63.7°, 589 nm, c = 0.37 g/100 ml, Chloroform [a] D 20 = -63.7 °, 589 nm, c = 0.37 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.12 (br. s, IH), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.14-4.04 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (teil- weise verdeckt, 2H), 3.22 (t, IH), 2.93-2.79 (m, IH), 2.55 (s, 3H), 2.16-1.96 (m, 2H), 1.94-1.81 (m, IH), 1.72-1.60 (m, 3H), 1.56-1.43 (m, IH), 1.33 (qd, 2H). Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.12 (br.s, IH), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.14-4.04 (m, IH), 3.93 (d, 2H), 3.87 (dd, 2H), 3.37-3.28 (partially obscured, 2H), 3.22 (t, IH), 2.93-2.79 (m, IH), 2.55 (s, 3H), 2.16-1.96 (m, 2H), 1.94-1.81 (m, IH), 1.72-1.60 (m, 3H), 1.56 -1.43 (m, IH), 1.33 (qd, 2H).
LC/MS (Methode 1, ESIpos): Rt = 1.07 min, m/z = 506 [M+H]+. LC / MS (Method 1, ESIpos): R t = 1.07 min, m / z = 506 [M + H] + .
Beispiel 19 Example 19
(+/-)-(lR5,25R,5R5)-2-[(6-Methyl-4-oxo-l,2,3-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2-(tetrahydro- 2i7-pyran-4-yl)efhoxy]benzoyl}cyclopentancarbonsäure (Racemat) (+/-) - (IR 5,25R, 5R 5) -2 - [(6-methyl-4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) -methyl] -5- [2- (tetrahydro-2i-pyran-4-yl) efhoxy] benzoyl} cyclopentane carboxylic acid (racemate)
Eine Lösung von 142 mg (0.23 mmol) der Verbindung aus Beispiel 17A in 0.8 ml Dichlormethan wurde bei 0°C mit 0.4 ml (5.03 mmol) Trifluoressigsäure versetzt. Das Gemisch wurde 1 h bei RT gerührt und anschließend eingeengt. Der Rückstand wurde mittels präparativer HPLC (Methode 6) gereinigt. Es wurden 84 mg (71 % d. Th., Reinheit 100%) der Titelverbindung erhalten. A solution of 142 mg (0.23 mmol) of the compound from Example 17A in 0.8 ml of dichloromethane was treated at 0 ° C with 0.4 ml (5.03 mmol) of trifluoroacetic acid. The mixture was stirred at RT for 1 h and then concentrated. The residue was purified by preparative HPLC (Method 6). 84 mg (71% of theory, purity 100%) of the title compound were obtained.
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.12 (br. s, 1H), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, 1H), 7.04 (d, 2H), 4.51 (d, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93- 2.80 (m, 1H), 2.55 (s, 3H), 2.16-2.04 (m, 1H), 1.93-1.82 (m, 1H), 1.76-1.57 (m, 6H), 1.57-1.44 (m, 1H), 1.30-1.12 (m, 2H). LC/MS (Methode 1, ESIpos): Rt = 1.14 min, m/z = 520 [M+H]+. Ή-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.12 (br.s, 1H), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, 1H), 7.04 (d, 2H), 4.51 (d, 2H), 4.15-4.05 (m, 3H), 3.83 (dd, 2H), 3.32-3.20 (m, 3H), 2.93-2.80 (m, 1H), 2.55 ( s, 3H), 2.16-2.04 (m, 1H), 1.93-1.82 (m, 1H), 1.76-1.57 (m, 6H), 1.57-1.44 (m, 1H), 1.30-1.12 (m, 2H). LC / MS (Method 1, ESIpos): R t = 1.14 min, m / z = 520 [M + H] + .
Trennung der Enantiomere: Separation of the enantiomers:
69 mg der racemischen Verbindung aus Beispiel 19 wurden in 10 ml Ethanol/Acetonitril gelöst und mittels präparativer SFC an chiraler Phase in die Enantiomere getrennt (siehe Beispiele 20 und 21) [Säule: Phenomenex Amylose II, 5 μιη, 250 mm x 20 mm; Fluss: 100 ml/min; Detektion: 210 nm; Injektionsvolumen: 0.40 ml; Temperatur: 40°C; Eluent: 70% Kohlendioxid / 30% Ethanol, Laufzeit 18 min] .  69 mg of the racemic compound from Example 19 were dissolved in 10 ml of ethanol / acetonitrile and separated into the enantiomers by means of preparative SFC on a chiral phase (see Examples 20 and 21) [column: Phenomenex amylose II, 5 μm, 250 mm × 20 mm; Flow: 100 ml / min; Detection: 210 nm; Injection volume: 0.40 ml; Temperature: 40 ° C; Eluent: 70% carbon dioxide / 30% ethanol, running time 18 min].
Beispiel 20 Example 20
(+)-( lRS,2SR,5RS)-2- [(6-Methyl-4-oxo- 1 ,2,3-benzotriazin-3(4 /)-yl)methyl] -5- { 4- [2-(tetrahydro- 2i7-pyran-4-yl)ethoxy]benzoyl}cyclopentancarbonsäure (Enantiomer 1) Ausbeute: 22 mg; ehem. Reinheit = 100%; ee-Wert = 100% (+) - (IRS, 2SR, 5RS) -2- [(6-methyl-4-oxo-1,2,3-benzotriazine-3 (4 /) -yl) methyl] -5- {4- [2 - (tetrahydro-2-i-pyran-4-yl) ethoxy] benzoyl} cyclopentanecarboxylic acid (enantiomer 1) Yield: 22 mg; former purity = 100%; ee value = 100%
[a]D 20 = +50.6°, 589 nm, c = 0.32 g/100 ml, Chloroform [a] D 20 = + 50.6 °, 589 nm, c = 0.32 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.10 (br. s, 1H), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (d, 1H), 7.04 (d, 2H), 4.51 (d, 2H), 4.16-4.04 (m, 3H), 3.83 (dd, 2H), 3.32-3.19 (m, teilweise verdeckt, 3H), 2.93-2.79 (m, 1H), 2.55 (s, 3H), 2.17-2.04 (m, 1H), 1.94-1.82 (m, 1H), 1.76-1.58 (m, 6H), 1.56-1.44 (m, 1H), 1.29-1.11 (m, 2H). Ή-NMR (400 MHz, DMSO-de): δ [ppm] = 12.10 (br.s, 1H), 8.12-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (d, 1H), 7.04 (d, 2H), 4.51 (d, 2H), 4.16-4.04 (m, 3H), 3.83 (dd, 2H), 3.32-3.19 (m, partially occluded, 3H), 2.93-2.79 (m, 1H), 2.55 (s, 3H), 2.17-2.04 (m, 1H), 1.94-1.82 (m, 1H), 1.76-1.58 (m, 6H), 1.56-1.44 (m, 1H), 1.29-1.11 (m, 2H ).
LC/MS (Methode 1, ESIpos): Rt = 1.10 min, m/z = 520 [M+H]+. Beispiel 21 LC / MS (Method 1, ESIpos): R t = 1.10 min, m / z = 520 [M + H] + . Example 21
(-)-(lR5,25R,5R^-2-[(6-Methyl-4-oxo-l,23-benzotriazin-3(4 /)-yl)methyl]-5-{4-[2-(tetrahydro- 2/f-pyran-4-yl)ethoxy]berizoyl}cyclopentaricarborisäure (Enantiomer 2) (-) - (1R5,25R, 5R ^ -2 - [(6-Methyl-4-oxo-l, 23-benzotriazine-3 (4 /) -yl) methyl] -5- {4- [2- ( tetrahydro-2 / f-pyran-4-yl) ethoxy] berizoyl} cyclopentaricarboxylic acid (enantiomer 2)
Ausbeute: 20 mg; ehem. Reinheit = 95%; ee-Wert = 100% [a]D 20 = -50.6°, 589 nm, c = 0.31 g/100 ml, Chloroform Yield: 20 mg; former purity = 95%; ee value = 100% [a] D 20 = -50.6 °, 589 nm, c = 0.31 g / 100 ml, chloroform
Ή-NMR (400 MHz, DMSO-d6): δ [ppm] = 12.10 (br. s, IH), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.15-4.04 (m, 3H), 3.83 (dd, 2H), 3.32-3.19 (m, teilweise verdeckt, 3H), 2.93-2.79 (m, IH), 2.55 (s, 3H), 2.16-2.04 (m, IH), 1.94-1.81 (m, IH), 1.74-1.57 (m, 6H), 1.56-1.44 (m, IH), 1.29-1.14 (m, 2H). Ή NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 12.10 (br.s, IH), 8.11-8.04 (m, 2H), 7.96 (d, 2H), 7.90 (dd, IH), 7.04 (d, 2H), 4.51 (d, 2H), 4.15-4.04 (m, 3H), 3.83 (dd, 2H), 3.32-3.19 (m, partially occluded, 3H), 2.93-2.79 (m, IH) , 2.55 (s, 3H), 2.16-2.04 (m, IH), 1.94-1.81 (m, IH), 1.74-1.57 (m, 6H), 1.56-1.44 (m, IH), 1.29-1.14 (m, 2H).
B. Bewertung der pharmakologischen Wirksamkeit B. Evaluation of Pharmacological Activity
Die pharmakologische Aktivität der erfindungsgemäßen Verbindungen kann durch in vitro- und in vzvo-Untersuchungen, wie sie dem Fachmann bekannt sind, nachgewiesen werden. Die nachfolgenden Anwendungsbeispiele beschreiben die biologische Wirkung der erfindungsgemäßen Verbindungen, ohne die Erfindung auf diese Beispiele zu beschränken. The pharmacological activity of the compounds according to the invention can be detected by in vitro and in vzvo studies, as are known to those skilled in the art. The following application examples describe the biological activity of the compounds according to the invention without restricting the invention to these examples.
Abkürzungen und Akronyme: Abbreviations and acronyms:
APMA 4-Aminophenylquecksilberacetat APMA 4-aminophenyl mercuric acetate
Brij®-35 Polyoxyethylenlaurylether Brij®- 35 polyoxyethylene lauryl ether
BSA bovines Serumalbumin  BSA bovine serum albumin
CYP Cytochrom P450  CYP cytochrome P450
Dap (oder Dpa) L-2,3-Diaminopropionsäure (ß-Amino-L-alanin)  Dap (or Dpa) L-2,3-diaminopropionic acid (β-amino-L-alanine)
DMSO Dimethylsulfoxid  DMSO dimethyl sulfoxide
Dnp 2,4-Dinitrophenyl  Dnp 2,4-dinitrophenyl
EDTA Ethylendiamintetraessigsäure  EDTA ethylenediaminetetraacetic acid
HEPES 2-[4-(2-Hydroxyethyl)piperazin-l-yl]ethansulfonsäure  HEPES 2- [4- (2-hydroxyethyl) piperazin-1-yl] ethanesulfonic acid
HME humane Makrophagen-Elastase  HME human macrophage elastase
IC Inhibitionskonzentration  IC inhibitory concentration
Mca (7 -Methoxycumarin-4-yl) acetyl  Mca (7 -methoxycumarin-4-yl) acetyl
MMP Matrixmetallopeptidase  MMP matrix metallopeptidase
MTP Mikrotiterplatte  MTP microtiter plate
NADP+ Nikotinsäureamid-Adenin-Dinukleotid-Phosphat (oxidierte Form) NADP + nicotinamide adenine dinucleotide phosphate (oxidized form)
NADPH Nikotinsäureamid-Adenin-Dinukleotid-Phosphat (reduzierte Form) NADPH nicotinic acid amide adenine dinucleotide phosphate (reduced form)
Nval Norvalin Nval Norvaline
PBS Phosphat-gepufferte Salzlösung  PBS phosphate buffered saline
PEG Polyethylenglykol  PEG polyethylene glycol
Tris Tris(hydroxymethyl)aminomethan  Tris tris (hydroxymethyl) aminomethane
v/v Volumen zu Volumen- Verhältnis (einer Lösung) v / v volume to volume ratio (of a solution)
w/w Gewicht zu Gewicht- Verhältnis (einer Lösung) w / w weight to weight ratio (of a solution)
B-l. In vitro HME-Inhibitionstest B-l. In vitro HME inhibition test
Die Wirkstärke der erfindungsgemäßen Verbindungen gegenüber HME (MMP-12) wird in einem in vi tro -Hemmte st ermittelt. Die HME-vermittelte amidolytische Spaltung eines geeigneten Peptid- substrats führt hierin zu einer Fluoreszenzlichtzunahme. Die Signalintensität des Fluoreszenz- lichtes ist direkt proportional zur Enzymaktivität. Die Wirkkonzentration einer Testverbindung, bei der die Hälfte des Enzyms inhibiert ist (50% Signalintensität des Fluoreszenzlichtes), wird als IC5o-Wert angegeben. The potency of the compounds of the invention compared to HME (MMP-12) is determined in a in vi tro-inhibited st. The HME-mediated amidolytic cleavage of a suitable peptide substrate results here in a fluorescence light increase. The signal intensity of the fluorescence light is directly proportional to the enzyme activity. The effective concentration of a test compound in which half of the enzyme is inhibited (50% signal intensity of the fluorescent light), is given as IC 5 o value.
Standard-m vitro HME-Inhibitionstest: Standard in vitro HME inhibition test:
In einer 384 Loch-Mikrotiterplatte werden in einem Testvolumen von insgesamt 41 μΐ der Testpuffer (0.1 M HEPES pH 7.4, 0.15 M NaCl, 0.03 M CaCl2, 0.004 mM ZnCl2, 0.02 M EDTA, 0.005% Brij®), das Enzym (0.5 nM HME; Fa. R&D Systems, 917-MP, autokatalytische Aktivierung nach Angaben des Herstellers) und das intramolekular gequenchte Substrat [5 μΜ Mca-Pro- Leu-Gly-Leu-Glu-Glu-Ala-Dap(Dnp)-NH2; Fa. Bachem, M-2670] bei An- und Abwesenheit der Testsubstanz (als Lösung in DMSO) zwei Stunden lang bei 37°C inkubiert. Die Fluoreszenzlichtintensität der Testansätze wird gemessen (Excitation 323 nm, Emission 393 nm). Die ICso-Werte werden durch eine Auftragung der Fluoreszenzlichtintensität gegenüber der Wirkstoffkonzentration ermittelt. In a 384 well microtiter plate are used in a test volume of 41 μΐ of assay buffer (0.1 M HEPES pH 7.4, 0.15M NaCl, 0.03M CaCl 2, 0.004 mM ZnCl 2, 0.02M EDTA, 0.005% Brij ®), the enzyme ( 0.5 nM HME, R & D Systems, 917-MP, autocatalytic activation according to the manufacturer's instructions) and the intramolecularly quenched substrate [5 μM Mca-Pro-Leu-Gly-Leu-Glu-Glu-Ala-Dap (Dnp) -NH 2 ; Bachem, M-2670] in the presence and absence of the test substance (as a solution in DMSO) for two hours at 37 ° C incubated. The fluorescent light intensity of the test batches is measured (excitation 323 nm, emission 393 nm). The IC 50 values are determined by plotting the fluorescent light intensity versus the drug concentration.
Hochsensitiver in vitro HME-Inhibitionstest: Highly sensitive in vitro HME inhibition test:
Ergeben sich bei hochpotenten Testsubstanzen im oben beschriebenen Standard-HME-Inhibitions- test subnanomolare IC-Werte, so wird zu deren genauerer Ermittlung ein modifizierter Test verwendet. Hierbei wird eine zehnfach niedrigere Enzymkonzentration eingesetzt (finale Konzentration z.B. 0.05 nM), um eine erhöhte Sensitivität des Tests zu erreichen. Die Inkubationszeit des Tests wird entsprechend länger gewählt (z.B. 16 Stunden). In vitro HME-Inhibitionstest in Anwesenheit von Serumalbumin im Reaktionspuffer: If subnanomolar IC values are obtained in the case of highly potent test substances in the standard HME inhibition test described above, a modified test is used for their more precise determination. In this case, a ten-fold lower enzyme concentration is used (final concentration, for example, 0.05 nM) in order to achieve an increased sensitivity of the test. The incubation time of the test is chosen to be longer (e.g., 16 hours). In vitro HME inhibition test in the presence of serum albumin in the reaction buffer:
Dieser Test entspricht dem oben beschriebenen Standard-HME-Inhibitionstest, jedoch unter Verwendung eines modifizierten Reaktionspuffers. Dieser Reaktionspuffer enthält zusätzlich Rinderserumalbumin (BSA, fettsäurefrei, A6003, Fa. Sigma-Aldrich) einer finalen Konzentration von 2% (w/w), was in etwa der Hälfte des physiologischen Serumalbumingehaltes entspricht. Die Enzym- konzentration in diesem modifizierten Test ist leicht erhöht (z.B. 0.75 nM), ebenso die Inkubationsdauer (z.B. drei Stunden).  This test corresponds to the standard HME inhibition test described above, but using a modified reaction buffer. This reaction buffer additionally contains bovine serum albumin (BSA, fatty acid free, A6003, Sigma-Aldrich) of a final concentration of 2% (w / w), which corresponds approximately to half of the physiological serum albumin content. The enzyme concentration in this modified assay is slightly elevated (e.g., 0.75 nM) as is the incubation time (e.g., three hours).
In der folgenden Tabelle 1A sind für individuelle Ausführungsbeispiele der Erfindung die ICso- Werte aus dem Standard- bzw. hochsensitiven HME-Inhibitionstest wiedergegeben (zum Teil als Mittelwerte aus mehreren unabhängigen Einzelbestimmungen und gerundet auf zwei signifikante Stellen): Tabelle 1A: Hemmung der humanen Makrophagen-Elastase (HME / hMMP-12) In the following Table 1A, for individual embodiments of the invention, the IC 50 values from the standard or highly sensitive HME inhibition test are reproduced (partly as averages from several independent individual determinations and rounded to two significant digits): Table 1A: Inhibition of human macrophage elastase (HME / hMMP-12)
In der folgenden Tabelle 1B sind für repräsentative Ausführungsbeispiele der Erfindung die IC50- Werte aus dem HME-Inhibitionstest in Abwesenheit (vgl. Daten in Tabelle 1A) bzw. Anwesenheit von Serumalbumin gegenübergestellt (zum Teil als Mittelwerte aus mehreren unabhängigen Einzelbestimmungen und gerundet auf zwei signifikante Stellen): Representative embodiments of the invention are compared in the following Table 1B with the IC 50 values from the HME inhibition test in the absence (see data in Table 1A) and presence of serum albumin (partly as averages from several independent determinations and rounded to two significant digits):
Tabelle 1B: Hemmung der humanen Makrophagen-Elastase (HME / hMMP-12) in Abwesenheit (-) bzw. Anwesenheit (+) von Serumalbumin (BSA) Table 1B: Inhibition of human macrophage elastase (HME / hMMP-12) in the absence (-) or presence (+) of serum albumin (BSA)
Beispiel HME ICso [nM] HME ICso [nM] Example HME ICso [nM] HME ICso [nM]
Nr. (- BSA) (+ BSA)  No. (- BSA) (+ BSA)
1 0.040 6.45  1 0.040 6.45
2 0.071 6.71  2 0.071 6.71
4 0.37 37.3  4 0.37 37.3
5 0.085 4.06  5 0.085 4.06
8 0.016 2.45  8 0.016 2.45
11 0.19 39.2  11 0.19 39.2
15 0.026 16.0  15 0.026 16.0
19 0.12 32.0 Beispiel HME ICso [nM] HME ICso [nM] 19 0.12 32.0 Example HME ICso [nM] HME ICso [nM]
Nr. (- BSA) (+ BSA)  No. (- BSA) (+ BSA)
20 0.053 12.2  20 0.053 12.2
Beim Vergleich der in Tabelle 1B wiedergegebenen Daten zeigt sich, dass die erfindungsgemäßen Verbindungen auch in Gegenwart von Serumalbumin noch eine hohe inhibitorische Potenz (häufig im nanomolaren Bereich) gegenüber HME aufweisen. Dies deutet auf eine weniger stark aus- geprägte unspezifische Wechselwirkung der erfindungsgemäßen Verbindungen mit Blutplasma- Bestandteilen hin und lässt so eine erhöhte "freie Fraktion" dieser Verbindungen im Blut erwarten, was sich günstig auf die in vzVo-Wirksamkeit auswirken sollte. Comparison of the data presented in Table 1B shows that the compounds according to the invention still have a high inhibitory potency (frequently in the nanomolar range) compared to HME, even in the presence of serum albumin. This indicates a less pronounced nonspecific interaction of the compounds according to the invention with blood plasma components and thus allows an increased "free fraction" of these compounds in the blood, which should have a favorable effect on the vzVo activity.
B-2. In vitro MMP-Inhibitionstests B-2. In vitro MMP inhibition tests
Die Wirkstärke der erfindungsgemäßen Verbindungen gegenüber anderen MMPs (und damit ihre Selekivität) wird ebenfalls in in vz'iro-Hemmtests ermittelt. Die MMP-vermittelte amidolytische Spaltung eines geeigneten Peptidsubstrats führt auch hier zu einer Fluoreszenzlichtzunahme. Die Signalintensität des Fluoreszenzlichtes ist direkt proportional zur Enzymaktivität. Die Wirkkonzentration einer Testverbindung, bei der die Hälfte des Enzyms inhibiert ist (50% Signalintensität des Fluoreszenzlichtes), wird als ICso-Wert angegeben. a) Humane MMPs: The potency of the compounds of the invention over other MMPs (and thus their Selekivität) is also determined in vz 'iro inhibition assays. The MMP-mediated amidolytic cleavage of a suitable peptide substrate also leads here to a fluorescence light increase. The signal intensity of the fluorescent light is directly proportional to the enzyme activity. The effective concentration of a test compound in which half of the enzyme is inhibited (50% signal intensity of the fluorescent light) is given as IC 50 value. a) Human MMPs:
In vitro MMP-1 -Inhibitionstest: In vitro MMP-1 inhibition test:
Rekombinantes MMP-1 (Fa. R&D Systems, 901-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca- Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES- 001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-l-Reak- tion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-1 (R & D Systems, 901-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-1 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-2 -Inhibitionstest: In vitro MMP-2 inhibition test:
Rekombinantes MMP-2 (Fa. R&D Systems, 902-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentra- tion z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca- Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES- 001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-2-Reak- tion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-2 (R & D Systems, 902-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentra- tion, for example, 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl, 150 mM NaCl, 0.05% Brij ® -35) is 1 μΐ of the examined test compound (as a solution in DMSO, suitable concentrations, for example 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP). The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-3-Inhibitionstest: In vitro MMP-3 inhibition test:
Rekombinantes MMP-3 (Fa. R&D Systems, 513-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca- Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys(Dnp)-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-002) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-3 -Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-3 (R & D Systems, 513-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is carried out by adding the intramolecularly quenched substrate McA-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-002 ) so that a total test volume of 50 μΐ results. The course of the MMP-3 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-7 -Inhibitionstest: In vitro MMP-7 inhibition test:
Rekombinantes MMP-7 (Fa. R&D Systems, 907-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.5 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-7 - Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro MMP-8-Inhibitionstest: Recombinant MMP-7 (R & D Systems, 907-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.5 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-7 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.). In vitro MMP-8 inhibition test:
Rekombinantes MMP-8 (Fa. R&D Systems, 908-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.5 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-8- Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-8 (R & D Systems, 908-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.5 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-9-Inhibitionstest: In vitro MMP-9 inhibition test:
Rekombinantes MMP-9 (Fa. R&D Systems, 911-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-9- Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro ΜΜΡ-10-Inhibitionstest: Recombinant MMP-9 (R & D Systems, 911-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.). In vitro ΜΜΡ-10 inhibition test:
Rekombinantes MMP-10 (Fa. R&D Systems, 910-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Kon- zentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca- Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys(Dnp)-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-002) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-10-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-10 (R & D Systems, 910-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is carried out by adding the intramolecularly quenched substrate McA-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-002 ) so that a total test volume of 50 μΐ results. The course of the MMP-10 reaction is determined by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C).
In vitro ΜΜΡ-13-Inhibitionstest: In vitro ΜΜΡ-13 inhibition test:
Rekombinantes MMP-13 (Fa. R&D Systems, 511-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-13- Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro ΜΜΡ-14-Inhibitionstest: Recombinant MMP-13 (R & D Systems, 511-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-13 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.). In vitro ΜΜΡ-14 inhibition test:
Rekombinantes MMP-14 (Fa. R&D Systems, 918-MP) wird den Herstellerangaben entsprechend durch Verwenden von rekombinantem Furin (Fa. R&D Systems, 1503-SE) enzymatisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.5 nM) in Reaktionspuffer (50 mM Tris/ HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Test- Verbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-14-Reaktion wird durch Messung der Fluoreszenz- Intensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant MMP-14 (R & D Systems, 918-MP) is enzymatically activated according to the manufacturer's instructions by using recombinant furin (R & D Systems, 1503-SE). To 24 μΐ activated enzyme (final concentration eg 0.5 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as Solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 μΜ; R & D Systems, ES-010) a total test volume of 50 μΐ results. The course of the MMP-14 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
In vitro ΜΜΡ-16-Inhibitionstest: In vitro ΜΜΡ-16 inhibition test:
Rekombinantes MMP-16 (Fa. R&D Systems, 1785-MP) wird den Herstellerangaben entsprechend durch Verwenden von rekombinantem Furin (Fa. R&D Systems, 1503-SE) enzymatisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 1 nM) in Reaktionspuffer (50 mM Tris/ HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-16-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In den folgenden Tabellen 2A und 2B sind für repräsentative Ausführungsbeispiele der Erfindung die ICso-Werte aus diesen Tests zur Inhibition humaner MMPs wiedergegeben (zum Teil als Mittelwerte aus mehreren unabhängigen Einzelbestimmungen und gerundet auf zwei signifikante Stellen): Recombinant MMP-16 (R & D Systems, 1785-MP) is enzymatically activated according to the manufacturer's instructions by using recombinant furin (R & D Systems, 1503-SE). To 24 μΐ activated enzyme (final concentration eg 1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is achieved by addition of the intramolecular quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 μΜ, R & D Systems, ES-010) is started so that a total test volume of 50 μΐ results. The course of the MMP-16 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.). In the following Tables 2A and 2B, for representative embodiments of the invention, the IC 50 values from these assays for inhibiting human MMPs are reported (in part as averages of several independent determinations and rounded to two significant digits):
Tabelle 2A: Hemmung humaner MMPs Table 2A: Inhibition of human MMPs
Tabelle 2B: Hemmung humaner MMPs Table 2B: Inhibition of human MMPs
Beispiel MMP-9 MMP-10 MMP-13 MMP-14 MMP- 16 Example MMP-9 MMP-10 MMP-13 MMP-14 MMP-16
Nr. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]  Nos. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]
1 450 120 110 160 1500  1 450 120 110 160 1500
2 360 80 49 99 550  2 360 80 49 99 550
4 5000 170 460 2700 7100  4 5000 170 460 2700 7100
5 460 13 67 250 940 Beispiel MMP-9 MMP-10 MMP-13 MMP-14 MMP-16 5 460 13 67 250 940 Example MMP-9 MMP-10 MMP-13 MMP-14 MMP-16
Nr. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]  Nos. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]
7 120 11000 140 170 830  7 120 11000 140 170 830
8 55 12 51 100 280  8 55 12 51 100 280
10 310 5100 230 600 1800  10 310 5100 230 600 1800
11 660 18 470 1200 3800  11 660 18 470 1200 3800
15 150 9 220 150 760  15 150 9 220 150 760
19 960 58 590 2700 2500  19 960 58 590 2700 2500
20 370 31 170 1000 3100  20 370 31 170 1000 3100
Beim Vergleich der in den Tabellen 1A und 2A/2B wiedergegebenen Inhibitionsdaten zeigt sich, dass die erfindungsgemäßen Verbindungen im Allgemeinen und deren aktivere Stereoisomere im Besonderen eine sehr hohe inhibitorische Potenz (häufig im sub-nanomolaren Bereich) gegenüber HME und zugleich eine hohe bis sehr hohe Selektivität (in der Regel ein bis drei Größenordnungen oder noch darüber) gegenüber verwandten humanen MMPs aufweisen. b ) MMPs der Nager: Comparing the inhibition data presented in Tables 1A and 2A / 2B shows that the compounds according to the invention in general and their more active stereoisomers in particular have a very high inhibitory potency (frequently in the sub-nanomolar range) compared to HME and at the same time a high to very high Have selectivity (usually one to three orders of magnitude or more) against related human MMPs. b) MMPs of rodents:
In vitro MMP-2 -Inhibitionstest der Maus: In vitro MMP-2 inhibition test of the mouse:
Rekombinantes MMP-2 der Maus (Fa. R&D Systems, 924-MP) wird den Herstellerangaben ent- sprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Sub- strats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-2 -Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro MMP-3 -Inhibitionstest der Maus: Recombinant mouse MMP-2 (R & D Systems, 924-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) that a total test volume of 50 μΐ results. The course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.). In vitro MMP-3 inhibition test of the mouse:
Rekombinantes MMP-3 der Maus (Fa. R&D Systems, 548-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.5 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys(Dnp)-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-002) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-3-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant mouse MMP-3 (R & D Systems, 548-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.5 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) is 1 μΐ of the examined test compound (as a solution in DMSO, suitable concentrations pipetted example 1 nM to 30 μΜ) (in a white 384 well microtitre plate MTP). The enzymatic reaction is carried out by addition of the intramolecularly quenched substrate Mca-Arg-Pro-Lys-Pro-Val-Glu-Nval-Trp-Arg-Lys (Dnp) -NH2 (final concentration eg 5 μΜ; R & D Systems, ES-002) so that a total test volume of 50 μΐ results. The course of the MMP-3 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-7 -Inhibitionstest der Maus: In vitro MMP-7 inhibition test of the mouse:
Rekombinantes MMP-7 der Maus (Fa. R&D Systems, 2967-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.5 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-7-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Tempera- tur von 32°C). Recombinant mouse MMP-7 (R & D Systems, 2967-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.5 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 μΜ; R & D Systems, ES-010) a total test volume of 50 μΐ results. The course of the MMP-7 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-8-Inhibitionstest der Maus: In vitro MMP-8 inhibition test of the mouse:
Rekombinantes MMP-8 der Maus (Fa. R&D Systems, 2904-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-8-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro MMP-9-Inhibitionstest der Maus: Recombinant mouse MMP-8 (R & D Systems, 2904-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 μΜ; R & D Systems, ES-010) a total test volume of 50 μΐ results. The course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.). In vitro MMP-9 inhibition test of the mouse:
Rekombinantes MMP-9 der Maus (Fa. R&D Systems, 909-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-9-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant mouse MMP-9 (R & D Systems, 909-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro ΜΜΡ-12-Inhibitionstest der Maus: In vitro ΜΜΡ-12 inhibition test of the mouse:
Rekombinantes MMP-12 der Maus (Fa. R&D Systems, 3467-MP) wird den Herstellerangaben ent- sprechend autokatalytisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly- Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-12-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant mouse MMP-12 (R & D Systems, 3467-MP) is autocatalytically activated according to the manufacturer's instructions. To 24 μΐ activated enzyme (final concentration eg 1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 μΜ; R & D Systems, ES-010) that a total test volume of 50 μΐ results. The course of the MMP-12 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
Hochsensitiver in vitro MMP-12-Inhibitionstest der Maus: Highly sensitive in vitro MMP-12 inhibition test of the mouse:
Ergeben sich bei hochpotenten Testsubstanzen im oben beschriebenen MMP-12-Inhibitionstest der Maus subnanomolare IC -Werte, so wird zu deren genauerer Ermittlung ein modifizierter Test verwendet. Hierbei wird eine zehnfach niedrigere Enzymkonzentration eingesetzt (finale Konzentration z.B. 0.1 nM), um eine erhöhte Sensitivität des Tests zu erreichen. Die Inkubationszeit des Tests wird entsprechend länger gewählt (z.B. 16 Stunden). In vitro MMP-2 -Inhibitionstest der Ratte: If subnanomolar IC values are obtained in the case of highly potent test substances in the mouse MMP-12 inhibition test described above, a modified test is used for more precise determination. Here, a tenfold lower enzyme concentration is used (final concentration, e.g., 0.1 nM) to achieve increased sensitivity of the assay. The incubation time of the test is chosen to be longer (e.g., 16 hours). Rat in vitro MMP-2 inhibition test:
Rekombinantes MMP-2 der Ratte (Fa. R&D Systems, 924-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 10 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-2 -Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). Recombinant rat MMP-2 (R & D Systems, 924-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) is 1 μΐ of the examined test compound (as a solution in DMSO, suitable concentrations pipetted example 1 nM to 30 μΜ) (in a white 384 well microtitre plate MTP). The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 10 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-2 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-8-Inhibitionstest der Ratte: Rat in vitro MMP-8 inhibition test:
Rekombinantes MMP-8 der Ratte (Fa. R&D Systems, 3245-MP) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 2 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Lys-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-010) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-8-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Tempera- tur von 32°C). Recombinant rat MMP-8 (R & D Systems, 3245-MP) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 2 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCh, 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO , suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by addition of the intramolecularly quenched substrate Mca-Lys-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH2 (final concentration eg 5 μΜ; R & D Systems, ES-010) a total test volume of 50 μΐ results. The course of the MMP-8 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.).
In vitro MMP-9-Inhibitionstest der Ratte: Rat in vitro MMP-9 inhibition test:
Rekombinantes MMP-9 der Maus (Fa. R&D Systems, 5427 -MM) wird den Herstellerangaben entsprechend durch Verwenden von APMA chemisch aktiviert. Zu 24 μΐ aktivierten Enzyms (finale Konzentration z.B. 0.1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-9-Reaktion wird durch Messung der Fluoreszenzintensität (Excitation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). In vitro MMP-12-Inhibitionstest der Ratte: Recombinant mouse MMP-9 (R & D Systems, 5427-MM) is chemically activated according to the manufacturer's instructions by using APMA. To 24 μΐ activated enzyme (final concentration eg 0.1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2 , 150 mM NaCl, 0.05% Brij ® -35) 1 μΐ of the test compound to be tested (as a solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-9 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (eg over 120 minutes at a temperature of 32 ° C.). Rat in vitro MMP-12 inhibition test:
MMP-12 der Ratte (Uniprot NP_446415.1 ; Konstrukt L96-V277) wird mit einem zusätzlichen N-terminalen His-Tag und einer konsekutiven TEV-Spaltsequenz mittels eines pDEco7-Vektors in E. coli (BL21) exprimiert. Das so rekombinant exprimierte Protein bildet ein intrazelluläres un- lösliches Proteinkomp artiment (sog. inclusion body). Dieses wird nach Trennen und intensivem Waschen unter denaturierenden Bedingungen solubilisiert. Hierzu wird die inclusion body-Pellet- Fraktion aus einer 250 ml-E. coli-Kultur in einem Volumen von 120 ml Puffer A (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl2, 10 mM CaCl2, 8 M Harnstoff) aufgenommen. Das lösliche Protein wird renaturiert, indem je 60 ml der Probe mehrmals bei 4-8°C gegen Puffer B (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl2, 10 mM CaCl2) dialysiert werden. Nach der Dialyse wird die Probe zentrifugiert (25.000 x g). Das rückgefaltete Protein wird im Überstand mit einer Ausbeute von 3.7 mg pro 250 ml-E. coli-Kultur erhalten. Das so gewonnene Protein ist ohne weitere Reinigungsoperationen oder Protease-vermittelte Spaltprozesse enzymatisch aktiv. Rat MMP-12 (Uniprot NP_446415.1; construct L96-V277) is expressed with an additional N-terminal His tag and a TEV consecutive cleavage sequence using a pDEco7 vector in E. coli (BL21). The recombinantly expressed protein forms an intracellular insoluble protein compo- sition (so-called inclusion body). This is solubilized after separation and intensive washing under denaturing conditions. For this purpose, the inclusion body pellet fraction from a 250 ml E. coli culture in a volume of 120 ml of buffer A (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl 2 , 10 mM CaCl 2 , 8 M urea). The soluble protein is renatured by dialysing each 60 ml of the sample several times at 4-8 ° C against buffer B (50 mM Tris pH 7.4, 100 mM NaCl, 0.03 mM ZnCl 2 , 10 mM CaCl 2 ). After dialysis, the sample is centrifuged (25,000 xg). The refolded protein is in the supernatant with a yield of 3.7 mg per 250 ml-E. coli culture. The protein thus obtained is enzymatically active without further purification operations or protease-mediated cleavage processes.
Zu 24 μΐ MMP-12-Protein (finale Konzentration z.B. 1 nM) in Reaktionspuffer (50 mM Tris/HCl pH 7.5, 10 mM CaCl2, 150 mM NaCl, 0.05% Brij®-35) wird 1 μΐ der zu untersuchenden Testverbindung (als Lösung in DMSO, geeignete Konzentrationen z.B. 1 nM bis 30 μΜ) in einer weißen 384 Loch-Mikrotiterplatte (MTP) pipettiert. Die enzymatische Reaktion wird durch Zugabe des intramolekular gequenchten Substrats Mca-Pro-Leu-Gly-Leu-Dpa(Dnp)-Ala-Arg-NH2 (finale Konzentration z.B. 5 μΜ; R&D Systems, ES-001) gestartet, so dass ein Gesamttestvolumen von 50 μΐ resultiert. Der Verlauf der MMP-12-Reaktion wird durch Messung der Fluoreszenzintensität (Ex- citation 320 nm, Emission 410 nm) über einen geeigneten Zeitraum hinweg gemessen (z.B. über 120 min bei einer Temperatur von 32°C). To 24 μΐ MMP-12 protein (final concentration, for example 1 nM) in reaction buffer (50 mM Tris / HCl pH 7.5, 10 mM CaCl 2, 150 mM NaCl, 0.05% Brij ® -35) is 1 μΐ of the examined test compound ( as solution in DMSO, suitable concentrations eg 1 nM to 30 μΜ) in a white 384-well microtiter plate (MTP) pipetted. The enzymatic reaction is started by adding the intramolecularly quenched substrate Mca-Pro-Leu-Gly-Leu-Dpa (Dnp) -Ala-Arg-NH 2 (final concentration eg 5 μΜ; R & D Systems, ES-001) Total test volume of 50 μΐ results. The course of the MMP-12 reaction is measured by measuring the fluorescence intensity (excitation 320 nm, emission 410 nm) over a suitable period of time (for example over 120 minutes at a temperature of 32 ° C.).
In der folgenden Tabelle 3 sind für repräsentative Ausführungsbeispiele der Erfindung die IC50- Werte aus den Tests zur Inhibition von Maus-MMPs wiedergegeben (zum Teil als Mittelwerte aus mehreren unabhängigen Einzelbestimmungen und gerundet auf zwei signifikante Stellen): Table 3 below shows representative IC 50 values from mouse MMP inhibition assays (in part as averages of several independent determinations and rounded to two significant digits) for representative embodiments of the invention:
Tabelle 3: Hemmung von MMPs der Maus Table 3: Inhibition of mouse MMPs
Beispiel MMP-2 MMP-3 MMP-7 MMP-8 MMP-9 MMP-12Example MMP-2 MMP-3 MMP-7 MMP-8 MMP-9 MMP-12
Nr. IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM]No. IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM] IC50 [nM]
1 170 570 85 40 560 3.31 170 570 85 40 560 3.3
2 58 710 52 21 280 0.852 58 710 52 21 280 0.85
4 490 4000 1400 370 1500 7.74 490 4000 1400 370 1500 7.7
5 45 270 130 54 210 0.67 Beispiel MMP-2 MMP-3 MMP-7 MMP-8 MMP-9 MMP-12 Nr. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]5 45 270 130 54 210 0.67 Example MMP-2 MMP-3 MMP-7 MMP-8 MMP-9 MMP-12 No. ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM] ICso [nM]
10 300 120 130 14 930 2.810 300 120 130 14 930 2.8
11 640 620 210 34 1900 9.411 640 620 210 34 1900 9.4
19 840 3100 630 150 2200 7.619 840 3100 630 150 2200 7.6
20 340 670 200 46 800 1.7 20 340 670 200 46 800 1.7
Beim Vergleich der in Tabelle 3 wiedergegebenen Inhibitionsdaten zeigt sich, dass die erfindungsgemäßen Verbindungen im Allgemeinen und deren aktivere Stereoisomere im Besonderen eine sehr hohe inhibitorische Potenz (häufig im nanomolaren oder sogar sub-nanomolaren Bereich) ge- genüber MMP-12 der Maus und zugleich eine hohe Selektivität (in der Regel ein bis zwei Größenordnungen) gegenüber verwandten murinen MMPs aufweisen. Comparing the inhibition data presented in Table 3 shows that the compounds according to the invention in general and their more active stereoisomers in particular have a very high inhibitory potency (frequently in the nanomolar or even sub-nanomolar range) in relation to mouse MMP-12 and at the same time have high selectivity (usually one to two orders of magnitude) over related murine MMPs.
B-3. Tiermodell des Lungenemphysems B-3. Animal model of pulmonary emphysema
Elastase-induziertes Lungenemphysem bei Maus, Ratte oder Hamster ist ein weit verbreitetes Tiermodell für Lungenemphysem [The Fas/Fas-ligand pathway does not mediate the apoptosis in elastase-induced emphysema in mice, Sawada et al., Exp. Lung Res. 33, 277-288 (2007)] . Die Tiere erhalten eine orotracheale Instillation porciner Pankreas-Elastase. Die Behandlung der Tiere mit der Testsubstanz beginnt am Tag der Instillation der porcinen Pankreas-Elastase und erstreckt sich über einen Zeitraum von 3 Wochen. Am Studienende wird die Lungen-Compliance bestimmt und eine Alveolarmorphometrie durchgeführt. B-4. Tiermodell der Silica-induzierten Lungeninflammation Elastase-induced pulmonary emphysema in mouse, rat or hamster is a widely used animal model of pulmonary emphysema [The Fas / Fas-ligand pathway does not mediate the apoptosis in elastase-induced emphysema in mice, Sawada et al., Exp. Lung Res. 33, 277-288 (2007)]. The animals receive orotracheal instillation of porcine pancreatic elastase. The treatment of the animals with the test substance starts on the day of the instillation of the porcine pancreatic elastase and extends over a period of 3 weeks. At the end of the study, lung compliance is determined and alveolar morphometry performed. B-4. Animal model of silica-induced lung inflammation
Eine orotracheale Gabe von Silica bei Maus, Ratte oder Hamster führt zu einer Inflammation in der Lunge [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 (2010)] . Die Tiere werden am Tag der Instillation des Silica mit der Testsubstanz behandelt. Nach 24 Stunden wird eine bronchio-alveoläre Lavage zur Bestimmung des Zellgehaltes und der Biomarker durchgeführt. Orotracheal administration of silica to mouse, rat or hamster leads to lung inflammation [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 ( 2010)]. The animals are treated with the test substance on the day of the instillation of the silica. After 24 hours bronchioalveolar lavage is performed to determine cell content and biomarkers.
B-5. Tiermodell der Silica-induzierten Lungenfibrose B-fifth Animal model of silica-induced pulmonary fibrosis
Silica-induzierte Lungenfibrose bei Maus, Ratte oder Hamster ist ein weit verbreitetes Tiermodell für Lungenfibrose [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 (2010)] . Die Tiere erhalten eine oro- tracheale Instillation von Silica. Die Behandlung der Tiere mit der Testsubstanz beginnt am Tag der Instillation des Silica oder therapeutisch eine Woche später und erstreckt sich über einen Zeitraum von 6 Wochen. Am Studienende werden eine bronchio-alveoläre Lavage zur Bestimmung des Zellgehaltes und der Biomarker sowie eine histologische Beurteilung der Lungenfibrose durchgeführt. B-6. Tiermodell der ATP-induzierten pulmonalen Inflammation Silica-induced lung fibrosis in mouse, rat or hamster is a widely used animal model of pulmonary fibrosis [Involvement of leukotrienes in the pathogenesis of silica-induced pulmonary fibrosis in mice, Shimbori et al., Exp. Lung Res. 36, 292-301 (2010 )]. The animals receive oro-tracheal instillation of silica. The treatment of the animals with the test substance starts during the day the instillation of the silica or therapeutically a week later and extends over a period of 6 weeks. At the end of the study, a bronchioalveolar lavage is performed to determine cell content and biomarkers, and a histological assessment of pulmonary fibrosis is performed. B-sixth Animal model of ATP-induced pulmonary inflammation
Eine intratracheale Gabe von ATP (Adenosintriphosphat) an der Maus führt zu einer Inflammation in der Lunge [Acute lung inflammation and ventilator-induced lung injury caused by ATP via the P2Y receptors: An experimental study, Matsuyama et al., Respir. Res. 9:79 (2008)]. Die Tiere werden am Tag der Instillation von ATP für eine Dauer von 24 h mit der Testsubstanz behandelt (per gavage, durch Zusatz im Futter oder Trinkwasser, per osmotischer Minipumpe, per subkutaner oder intraperitonealer Injektion oder per Inhalation). Am Versuchsende wird eine bronchio-alveoläre Lavage zur Bestimmung des Zellgehalts und der pro-inflammatorischen Marker durchgeführt. Intratracheal administration of ATP (adenosine triphosphate) to the mouse causes lung inflammation [ATP via the P2Y receptors: An experimental study, Matsuyama et al., Respir. Res. 9:79 (2008)]. The animals are treated with the test substance for 24 h on the day of instillation of ATP (by gavage, by addition in feed or drinking water, by osmotic minipump, by subcutaneous or intraperitoneal injection or by inhalation). At the end of the experiment a bronchio-alveolar lavage is performed to determine the cell content and the pro-inflammatory markers.
B-7. CYP-Inhibitionstest B. 7 CYP Inhibition Assay
Die Fähigkeit von Substanzen, die CYP-Enzyme CYP1A2, CYP2C9, CYP2D6 und CYP3A4 im Menschen zu inhibieren, wird untersucht mit gepoolten Human-Lebermikrosomen als Enzymquelle in Gegenwart von Standardsubstraten (s.u.), die CYP-spezifische Metaboliten bilden. Die Inhibitionseffekte werden bei sechs verschiedenen Konzentrationen der Testverbindungen untersucht [2.8, 5.6, 8.3, 16.7, 20 (oder 25) sowie 50 μΜ], mit dem Ausmaß der CYP-spezifischen Metabolitenbildung der Standardsubstrate in Abwesenheit der Testverbindungen verglichen und die entsprechenden ICso-Werte berechnet. Ein Standard-Inhibitor, der eine einzelne CYP-Isoform spezifisch inhibiert, wird immer mitinkubiert, um Ergebnisse zwischen verschiedenen Serien vergleichbar zu machen. The ability of substances to inhibit the human CYP enzymes CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in human is examined using pooled human liver microsomes as enzyme source in the presence of standard substrates (see above) which form CYP-specific metabolites. The inhibition effects are investigated at six different concentrations of the test compounds [2.8, 5.6, 8.3, 16.7, 20 (or 25) and 50 μΜ], compared with the extent of CYP-specific metabolite formation of the standard substrates in the absence of the test compounds and the corresponding IC 50 values calculated. A standard inhibitor that specifically inhibits a single CYP isoform is always incubated to make comparisons between different series comparable.
Die Inkubation von Phenacetin, Diclofenac, Tolbutamid, Dextromethorphan oder Midazolam mit Human-Lebermikrosomen in Gegenwart von jeweils sechs verschiedenen Konzentrationen einer Testverbindung (als potentiellem Inhibitor) wird auf einer Workstation durchgeführt (Tecan, Genesis, Crailsheim, Deutschland). Standard-Inkubationsgemische enthalten 1.3 mM NADP+, 3.3 mM MgC x 6 H2O, 3.3 mM Glukose -6-phosphat, Glukose-6-phosphat-Dehydrogenase (0.4 U/ml) und 100 mM Phosphat-Puffer (pH 7.4) in einem Gesamtvolumen von 200 μΐ. Testverbindungen werden bevorzugt in Acetonitril gelöst. 96-Lochplatten werden eine definierte Zeit bei 37°C mit gepoolten Human-Lebermikrosomen inkubiert. Die Reaktionen werden durch Zugabe von 100 μΐ Acetonitril, worin sich ein geeigneter interner Standard befindet, abgestoppt. Gefällte Proteine werden durch Zentrifugation abgetrennt, die Überstände werden vereinigt und mittels LC- MS/MS analysiert. B-8. Hepatozytenassav zur Bestimmung der metabolischen Stabilität Incubation of phenacetin, diclofenac, tolbutamide, dextromethorphan or midazolam with human liver microsomes in the presence of six different concentrations of each test compound (as a potential inhibitor) is performed on a workstation (Tecan, Genesis, Crailsheim, Germany). Standard incubation mixtures contain 1.3 mM NADP + , 3.3 mM MgC x 6 H 2 O, 3.3 mM glucose-6-phosphate, glucose-6-phosphate dehydrogenase (0.4 U / ml) and 100 mM phosphate buffer (pH 7.4) in a total volume of 200 μΐ. Test compounds are preferably dissolved in acetonitrile. 96-well plates are incubated for a defined time at 37 ° C with pooled human liver microsomes. The reactions are stopped by addition of 100 μL acetonitrile, which is a suitable internal standard. Precipitated proteins are separated by centrifugation, the supernatants are pooled and analyzed by LC-MS / MS. B-eighth Hepatocyte assay for determination of metabolic stability
Die metabolische Stabilität von Testverbindungen gegenüber Hepatozyten wird bestimmt, indem die Verbindungen bei niedrigen Konzentrationen (bevorzugt unter oder um 1 μΜ) und bei niedrigen Zellzahlen (bevorzugt bei 1 * 106 Zellen/ml) inkubiert werden, um möglichst lineare kinetische Bedingungen im Versuch sicherzustellen. Sieben Proben aus der Inkubationslösung werden in einem festgelegten Zeitraster für die LC-MS-Analytik entnommen, um die Halbwertszeit (d.h. den Abbau) der jeweiligen Verbindung zu bestimmen. Aus dieser Halbwertszeit werden unterschiedliche "Clearance"-Parameter (CL) und "Fmax" - Werte berechnet (s.u.). The metabolic stability of test compounds to hepatocytes is determined by incubating the compounds at low concentrations (preferably below or around 1 μΜ) and at low cell counts (preferably at 1 × 10 6 cells / ml) to ensure the best possible linear kinetic conditions in the experiment , Seven samples from the incubation solution are taken at a fixed time interval for LC-MS analysis to determine the half-life (ie, degradation) of each compound. From this half-life different "clearance" parameters (CL) and "Fmax" values are calculated (see below).
Die CL- und Fmax-Werte stellen ein Maß für den Phase 1- und Phase 2-Metabolismus der Ver- bindungen in den Hepatozyten dar. Um den Einfluss des organischen Lösungsmittels auf die Enzyme in den Inkubationsansätzen möglichst klein zu halten, wird dessen Konzentration im Allgemeinen auf 1% (Acetonitril) bzw. 0.1 % (DMSO) begrenzt. The CL and Fmax values represent a measure of the phase 1 and phase 2 metabolism of the compounds in the hepatocytes. In order to minimize the influence of the organic solvent on the enzymes in the incubation mixtures, its concentration in the Generally limited to 1% (acetonitrile) and 0.1% (DMSO).
Für alle Spezies und Rassen wird mit einer Hepatozyten-Zellzahl in der Leber von 1.1 * 108 Zellen/g Leber gerechnet. CL-Parameter, deren Berechnung auf Halbwertszeiten beruhen, die we- sentlich über die Inkubationszeit hinausgehen (üblicherweise 90 Minuten), können nur als grobe Richtwerte angesehen werden. For all species and breeds, a hepatocyte cell count in the liver of 1.1 * 10 8 cells / g liver is expected. CL parameters calculated using half-lives that are significantly longer than the incubation time (usually 90 minutes) can only be considered as rough guidelines.
Die berechneten Parameter und deren Bedeutung sind: The calculated parameters and their meaning are:
Fmax well-stirred [ ] maximal mögliche Bioverfügbarkeit nach oraler ApplikationFmax well-stirred [] maximum bioavailability after oral administration
Berechnung: (l-CLbi00d well-stirred/QH) * 100 Calculation: (l-CLbi 00 d well-stirred / QH) * 100
CLbiood well-stirred [L/(h*kg)] berechnete Blut-Clearance (well stirred-Modell)  CLbiood well-stirred [L / (h * kg)] calculated blood clearance (well-stirred model)
Berechnung: (QH * CL';„trinsic) / (QH + CL';„trinsic)  Calculation: (QH * CL '; "trinsic) / (QH + CL';" trinsic)
CL'intrinsic [ml/(min*kg)] maximale Fähigkeit der Leber (der Hepatozyten), eine Verbindung zu metabolisieren (unter der Annahme, dass der Leber - blutfluss nicht geschwindigkeitslimitierend ist)  CL'intrinsic [ml / (min * kg)] maximum ability of liver (hepatocytes) to metabolize a compound (assuming that liver blood flow is not rate limiting)
Berechnung: CL'mtrinsic, apparent * speziesspezifische Hepatozytenzahl [1.1 * 108/ g Leber] * speziesspezifisches Lebergewicht [g/kg] Calculation: CL'mtrinsic, apparent * species-specific hepatocyte count [1.1 * 10 8 / g liver] * species-specific liver weight [g / kg]
CL'mtrinsic, apparent [ml/(min*mg)] normiert die Eliminationskonstante, indem diese durch die eingesetzte Hepatozyten-Zellzahl x (x * 106/ml) dividiert wird Berechnung: kei [1/min] / (Zellzahl [x * 106] / Inkubationsvolumen [ml])CL'mtrinsic, apparent [ml / (min * mg)] normalizes the elimination constant by dividing it by the hepatocyte cell number x (x * 10 6 / ml) used. Calculation: k e i [1 / min] / (cell number [x * 10 6 ] / incubation volume [ml])
(QH = speziesspezifischer Leberb lutfluss). Γη der folgenden Tabelle 4 sind für repräsentative Ausführungsbeispiele der Erfindung die CL- und Fmax-Werte aus diesem Assay nach Inkubation der Verbindungen mit Ratten-Hepatozyten wiedergegeben (zum Teil als Mittelwerte aus mehreren unabhängigen Einzelbestimmungen): (QH = species-specific liver blood flow). In the following Table 4, for representative embodiments of the invention, the CL and Fmax values from this assay are shown after incubation of the compounds with rat hepatocytes (partly as averages of several independent individual determinations):
Tabelle 4: berechnete Blut-Clearance und Bioverfügbarkeit nach Inkubation mit Ratten- Hepatozyten Table 4: Calculated blood clearance and bioavailability after incubation with rat hepatocytes
B-9. Metabolismus-Untersuchung B. 9 Metabolism investigation
Zur Bestimmung des Metabolismus-Profils der erfindungsgemäßen Verbindungen werden diese mit Lebermikrosomen oder mit primären frischen Hepatozyten verschiedener Tierspezies (z.B. Ratte, Hund) als auch humanen Ursprungs inkubiert, um Informationen über einen möglichst kompletten hepatischen Phase I- und Phase II-Metabolismus sowie über die am Metabolismus beteiligten Enzyme zu erhalten und zu vergleichen. To determine the metabolism profile of the compounds according to the invention, they are incubated with liver microsomes or with primary fresh hepatocytes of various animal species (eg rat, dog) as well as of human origin in order to obtain information on the most complete hepatic phase I and phase II metabolism and on the Obtain and compare enzymes involved in metabolism.
Die erfindungsgemäßen Verbindungen werden mit einer Konzentration von etwa 1-10 μΜ inkubiert. Dazu werden Stammlösungen der Verbindungen mit einer Konzentration von 0.1-1 mM in Acetonitril hergestellt und dann mit einer l: 100-Verdünnung in den Inkubationsansatz pipettiert. Die Lebermikrosomen werden in 50 mM Kaliumphosphatpuffer pH 7.4 mit und ohne NADPH- generierendem System, bestehend aus 1 mM NADP+, 10 mM Glukose-6-phosphat und 1 Unit Glu- kose-6-phosphat-Dehydrogenase, bei 37°C inkubiert. Primäre Hepatozyten werden in Suspension in William's E-Medium ebenfalls bei 37°C inkubiert. Nach einer Inkubationszeit von 0-4 h werden die Inkubationsansätze mit Acetonitril abgestoppt (Endkonzentration ca. 30%) und das Protein bei ca. 15000 x g abzentrifugiert. Die so abgestoppten Proben werden entweder direkt analysiert oder bis zur Analyse bei -20°C gelagert. Die Analyse erfolgt mittels Hochleistungsflüssigkeitschromatographie mit Ultraviolett- und massenspektrometrischer Detektion (HPLC-UV-MS/MS). Dazu werden die Überstände der Inkubationsproben mit geeigneten C18-reverse phase-Säulen und variablen Eluenten-Gemischen aus Acetonitril und 10 mM wässriger Ammoniumformiat-Lösung oder 0.05% wässriger Ameisensäure chromatographiert. Die UV-Chromatogramme in Verbindung mit den massenspektrometrischen Daten dienen zur Identifizierung, Strukturaufklärung und quantitativen Abschätzung der Metabolite und zur quantitativen Bestimmung der metabolischen Abnahme der erfindungsgemäßen Verbindungen in den Inkubationsansätzen. The compounds of the invention are incubated at a concentration of about 1-10 μΜ. For this purpose, stock solutions of the compounds are prepared at a concentration of 0.1-1 mM in acetonitrile and then pipetted with a 1: 100 dilution in the incubation mixture. The liver microsomes are incubated in 50 mM potassium phosphate buffer pH 7.4 with and without NADPH-generating system consisting of 1 mM NADP + , 10 mM glucose-6-phosphate and 1 unit glucose-6-phosphate dehydrogenase at 37 ° C. Primary hepatocytes are also incubated in suspension in William's E medium at 37 ° C. After an incubation period of 0-4 h, the incubation mixtures are stopped with acetonitrile (final concentration about 30%) and the protein is centrifuged off at about 15,000 × g. The thus stopped samples are either analyzed directly or stored at -20 ° C until analysis. The analysis is carried out by means of high performance liquid chromatography with ultraviolet and mass spectrometric detection (HPLC-UV-MS / MS). For this, the supernatants of the incubation samples are chromatographed with suitable C18 reverse phase columns and variable eluent mixtures of acetonitrile and 10 mM aqueous ammonium formate solution or 0.05% aqueous formic acid. The UV chromatograms in combination with the mass spectrometric data serve to identify, structure elucidate and quantitatively estimate the metabolites and to quantitatively determine the metabolic decrease of the compounds according to the invention in the incubation mixtures.
B-10. Pharmakokinetische Untersuchungen in vivo Die zu untersuchende Substanz wird Ratten oder Mäusen intravenös als Lösung appliziert (z.B. in entsprechendem Plasma mit geringem DMSO-Zusatz oder in einem PEG/Ethanol/W asser- Gemisch), die perorale Applikation erfolgt als Lösung (z.B. in Solutol/Ethanol/W asser- oder PEG/ Ethanol/Wasser-Gemischen) oder als Suspension (z.B. in Tylose) jeweils über eine Schlundsonde. Nach Substanzgabe wird den Tieren zu festgelegten Zeitpunkten Blut entnommen. Dieses wird heparinisiert, anschließend wird daraus durch Zentrifugation Plasma gewonnen. Die Testsubstanz wird im Plasma über LC-MSMS analytisch quantifiziert. Aus den so ermittelten Plasmakonzentration-Zeit-Verläufen werden unter Verwendung eines internen Standards und mit Hilfe eines validierten Rechenprogramms die pharmakokinetischen Kenngrößen berechnet, wie AUC (Fläche unter der Konzentration-Zeit-Kurve), Cmax (maximale Plasmakonzentration), t (Halbwertszeit), Vss (Verteilungsvolumen) und CL (Clearance) sowie die absolute und die relative Bioverfügbarkeit F und Frei (i.v./p.o. -Vergleich bzw. Vergleich von Suspension zu Lösung nach p.o.-Gabe). B-tenth Pharmacokinetic studies in vivo The substance to be tested is administered intravenously to rats or mice as a solution (eg in appropriate plasma with a small amount of DMSO or in a PEG / ethanol / water mixture), the oral administration is carried out as a solution (eg in Solutol / Ethanol / water or PEG / ethanol / water mixtures) or as a suspension (eg in Tylose) in each case via a gavage. After substance administration, the animals are bled at fixed times. This is heparinized, then plasma is recovered therefrom by centrifugation. The test substance is analytically quantified in the plasma via LC-MSMS. From the plasma concentration-time curves determined in this way, the pharmacokinetic parameters are calculated using an internal standard and with the aid of a validated computer program, such as AUC (area under the concentration-time curve), Cmax (maximum plasma concentration), t (half-life), Vss (distribution volume) and CL (clearance) and the absolute and relative bioavailability F and F re i (iv / po comparison or comparison of suspension to solution after po administration).
B-ll. Bestimmung der Löslichkeit B-ll. Determination of solubility
Versuchsdurchführung : Experimental procedure:
Die Prüfsubstanz wird in DMSO gelöst. Aus dieser Lösung wird ein Aliquot entnommen und in PBS-Puffer pH 6.5 eingebracht (DMSO- Anteil: 1%). Diese Lösung/Suspension wird für 24 h bei Raumtemperatur geschüttelt. Nach Ultra-Zentrifugation bei 114000 g für 30 min wird der Überstand abgenommen, mit Acetonitril/Wasser 8:2 verdünnt und per LC-MSMS analysiert. Quantifiziert wird über eine Fünf-Punkt-Kalibrationskurve der Testverbindung in DMSO.  The test substance is dissolved in DMSO. An aliquot is taken from this solution and introduced into PBS buffer pH 6.5 (DMSO content: 1%). This solution / suspension is shaken for 24 h at room temperature. After ultra-centrifugation at 114,000 g for 30 min, the supernatant is removed, diluted with acetonitrile / water 8: 2 and analyzed by LC-MSMS. Quantification is via a five-point calibration curve of the test compound in DMSO.
Geräte für die LC-MSMS-Quantifizierung: Devices for LC-MSMS quantification:
AB Sciex TRIPLE QUAD 4500; Agilent 1260 mit primärer Pumpe (G1312B Minity), Degasser (G4225A Minity), Säulenthermostat (G1316C Minity); CTC Analytics PAL Injektionssystem THC-xt. HPLC-Methode: AB Sciex TRIPLE QUAD 4500; Agilent 1260 with Primary Pump (G1312B Minity), Degasser (G4225A Minity), Column Thermostat (G1316C Minity); CTC Analytics PAL injection system THC-xt. HPLC method:
Eluent A: 0.5 ml Ameisensäure / Liter Wasser, Eluent B: 0.5 ml Ameisensäure / Liter Acetonitril; Gradient: 0 min 90% A -> 0.5 min 5% A -> 0.84 min 5% A -> 0.85 min 90% A ^ 1.22 min 90% A; Fluss: 2.5 ml/min; Injektionsvolumen: 15 μΐ; Säule: Waters OASIS HLB, 2.1 x 20 mm, 25 μ; Säulentemperatur: 30°C; Splitter (vor MS): 1:20.  Eluent A: 0.5 ml of formic acid / liter of water, eluent B: 0.5 ml of formic acid / liter of acetonitrile; Gradient: 0 min 90% A -> 0.5 min 5% A -> 0.84 min 5% A -> 0.85 min 90% A ^ 1.22 min 90% A; Flow: 2.5 ml / min; Injection volume: 15 μΐ; Column: Waters OASIS HLB, 2.1 x 20 mm, 25 μ; Column temperature: 30 ° C; Splitter (before MS): 1:20.
MS -Methoden: MS methods:
Flow Injection Analysis (FIA) für die Optimierung, Multiple Reaction Monitoring (MRM) für die Quantifizierung; Eluent A: 0.5 ml Ameisensäure / Liter Wasser, Eluent B: 0.5 ml Ameisensäure / Liter Acetonitril; Fluss: 0.25 ml/min; Injektionsvolumen: 15 μΐ; Säule: Edelstahlkapillare; Kapil- lartemperatur: 25°C.  Flow Injection Analysis (FIA) for optimization, Multiple Reaction Monitoring (MRM) for quantification; Eluent A: 0.5 ml of formic acid / liter of water, eluent B: 0.5 ml of formic acid / liter of acetonitrile; Flow: 0.25 ml / min; Injection volume: 15 μΐ; Column: stainless steel capillary; Capillary temperature: 25 ° C.
In der folgenden Tabelle 5 sind die so bestimmten Löslichkeitswerte repräsentativer Ausführungsbeispiele in PBS-Puffer pH 6.5 dargestellt: Table 5 below shows the solubility values of representative embodiments thus determined in PBS buffer pH 6.5:
Tabelle 5: Löslichkeit in PBS-Puffer pH 6.5 TABLE 5 Solubility in PBS buffer pH 6.5
Beispiel-Nr. Löslichkeit [mg/Liter] Example no. Solubility [mg / liter]
1 52.2  1 52.2
4 51.4  4 51.4
5 338.1  5 338.1
6 354.9  6 354.9
8 4.5  4.5
10 33.4  10 33.4
11 210  11 210
12 240  12 240
14 80  14 80
15 100  15 100
17 250  17 250
18 330  18,330
19 17  19 17
20 425.2 C. Ausführungsbeispiele für pharmazeutische Zusammensetzungen 20 425.2 C. Embodiments of Pharmaceutical Compositions
Die erfindungsgemäßen Verbindungen können folgendermaßen in pharmazeutische Zubereitungen überführt werden: The compounds according to the invention can be converted into pharmaceutical preparations as follows:
Tablette: Zusammensetzung: Tablet: composition:
100 mg der erfindungsgemäßen Verbindung, 50 mg Lactose (Monohydrat), 50 mg Maisstärke (nativ), 10 mg Polyvinylpyrrolidon (PVP 25) (Fa. BASF, Ludwigshafen, Deutschland) und 2 mg Magnesiumstearat.  100 mg of the compound according to the invention, 50 mg of lactose (monohydrate), 50 mg of corn starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (BASF, Ludwigshafen, Germany) and 2 mg of magnesium stearate.
Tablettengewicht 212 mg. Durchmesser 8 mm, Wölbungsradius 12 mm. Herstellung: Tablet weight 212 mg. Diameter 8 mm, radius of curvature 12 mm. production:
Die Mischung aus erfindungsgemäßer Verbindung, Lactose und Stärke wird mit einer 5%-igen Lösung (m/m) des PVPs in Wasser granuliert. Das Granulat wird nach dem Trocknen mit dem Magnesiumstearat 5 Minuten gemischt. Diese Mischung wird mit einer üblichen Tablettenpresse verpresst (Format der Tablette siehe oben). Als Richtwert für die Verpressung wird eine Presskraft von 15 kN verwendet.  The mixture of compound of the invention, lactose and starch is granulated with a 5% solution (m / m) of the PVP in water. The granules are mixed after drying with the magnesium stearate for 5 minutes. This mixture is compressed with a conventional tablet press (for the tablet format see above). As a guideline for the compression, a pressing force of 15 kN is used.
Oral applizierbare Suspension: Orally administrable suspension:
Zusammensetzung: Composition:
1000 mg der erfindungsgemäßen Verbindung, 1000 mg Ethanol (96%), 400 mg Rhodigel® (Xanthan gum der Firma FMC, Pennsylvania, USA) und 99 g Wasser. Einer Einzeldosis von 100 mg der erfindungsgemäßen Verbindung entsprechen 10 ml orale Suspension. 1000 mg of the compound of the invention, 1000 mg of ethanol (96%), 400 mg of Rhodigel ® (xanthan gum of the firm FMC, Pennsylvania, USA) and 99 g of water. A single dose of 100 mg of the compound of the invention corresponds to 10 ml of oral suspension.
Herstellung: production:
Das Rhodigel wird in Ethanol suspendiert, die erfindungsgemäße Verbindung wird der Suspension zugefügt. Unter Rühren erfolgt die Zugabe des Wassers. Bis zum Abschluß der Quellung des Rhodigels wird ca. 6 h gerührt. Oral applizierbare Lösung: The rhodigel is suspended in ethanol, the compound according to the invention is added to the suspension. While stirring, the addition of water. Until the completion of the swelling of Rhodigels is stirred for about 6 h. Orally administrable solution:
Zusammensetzung: Composition:
500 mg der erfindungsgemäßen Verbindung, 2.5 g Polysorbat und 97 g Polyethylenglycol 400. Einer Einzeldosis von 100 mg der erfindungsgemäßen Verbindung entsprechen 20 g orale Lösung. Herstellung:  500 mg of the compound according to the invention, 2.5 g of polysorbate and 97 g of polyethylene glycol 400. A single dose of 100 mg of the compound according to the invention correspond to 20 g of oral solution. production:
Die erfindungsgemäße Verbindung wird in der Mischung aus Polyethylenglycol und Polysorbat unter Rühren suspendiert. Der Rührvorgang wird bis zur vollständigen Auflösung der erfindungsgemäßen Verbindung fortgesetzt. i.v.-Lösung: Die erfindungsgemäße Verbindung wird in einer Konzentration unterhalb der Sättigungslöslichkeit in einem physiologisch verträglichen Lösungsmittel (z.B. isotonische Kochsalzlösung, Glucose- lösung 5% und/oder PEG 400-Lösung 30%) gelöst. Die Lösung wird steril filtriert und in sterile und pyrogenfreie Injektionsbehältnisse abgefüllt.  The compound of the invention is suspended in the mixture of polyethylene glycol and polysorbate with stirring. The stirring is continued until complete dissolution of the compound according to the invention. i.v. solution: The compound of the invention is dissolved at a concentration below the saturation solubility in a physiologically acceptable solvent (e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%). The solution is sterile filtered and filled into sterile and pyrogen-free injection containers.

Claims

Patentansprüche Patent claims
1. Verbindung der Formel (I) 1. Compound of formula (I)
in welcher in which
A für -O- oder -S- steht, A stands for -O- or -S-,
für die Zahl 1 oder 2 steht stands for the number 1 or 2
und and
R1 für Wasserstoff, Methyl, Fluormethyl, Difluormethyl oder Trifluormethyl steht, oder ein Salz, Solvat oder Solvat eines Salzes dieser Verbindung. R 1 represents hydrogen, methyl, fluoromethyl, difluoromethyl or trifluoromethyl, or a salt, solvate or solvate of a salt of this compound.
2. Verbindung der Formel (I) nach Anspruch 1, in welcher 2. A compound of formula (I) according to claim 1, in which
A für -O- steht, A stands for -O-,
für die Zahl 1 oder 2 steht stands for the number 1 or 2
und and
für Wasserstoff, Methyl oder Trifluormethyl steht, represents hydrogen, methyl or trifluoromethyl,
oder ein Salz, Solvat oder Solvat eines Salzes dieser Verbindung. or a salt, solvate or solvate of a salt of this compound.
3. Verbindung der Formel (I) nach Anspruch 1 oder 2, in welcher 3. A compound of formula (I) according to claim 1 or 2, in which
A für -O- steht, A stands for -O-,
für die Zahl 2 steht stands for the number 2
und and
R1 für Wasserstoff, Methyl oder Trifluormethyl steht, oder ein Salz, Solvat oder Solvat eines Salzes dieser Verbindung. R 1 represents hydrogen, methyl or trifluoromethyl, or a salt, solvate or solvate of a salt of this compound.
4. Verbindung gemäß Anspruch 1, 2 oder 3 mit der Formel (I-A) oder (I-B) 4. A compound according to claim 1, 2 or 3 with the formula (I-A) or (I-B)
worin A, n und R1 die in Anspruch 1, 2 oder 3 definierten Bedeutungen haben und die an den zentralen Cyclopentan-Ring gebundenen Gruppen eine relative irans-Anordnung zueinander aufweisen, oder ein Gemisch dieser Verbindungen, wobei A, n bzw. R1 in einem solchen Gemisch von (I-A) und (I-B) jeweils identisch sind, oder ein Salz, Solvat oder Solvat eines Salzes dieser Verbindungen oder ihres Gemisches. 5. Verbindung gemäß Anspruch 1, 2, 3 oder 4 mit der Formel (I-A) wherein A, n and R 1 have the meanings defined in claim 1, 2 or 3 and the groups bonded to the central cyclopentane ring have a relative irans arrangement to one another, or a mixture of these compounds, where A, n and R 1 , respectively in such a mixture of (IA) and (IB) are each identical, or a salt, solvate or solvate of a salt of these compounds or their mixture. 5. Compound according to claim 1, 2, 3 or 4 with the formula (IA)
worin A, n und R1 die in Anspruch 1, 2 oder 3 definierten Bedeutungen haben, in enantio- merenreiner Form mit einer, wie bezeichnet, (lS,2R,5S)-Konfiguration am zentralen Cyclopentan-Ring oder ein Salz, Solvat oder Solvat eines Salzes dieser Verbindung. wherein A, n and R 1 have the meanings defined in claim 1, 2 or 3, in enantiomerically pure form with an, as designated, (IS,2R,5S) configuration on the central cyclopentane ring or a salt, solvate or Solvate of a salt of this compound.
Verfahren zur Herstellung einer Verbindung, wie in den Ansprüchen 1 bis 5 definiert, dadurch gekennzeichnet, dass man eine Verbindung der Formel (Π) Process for the preparation of a compound as defined in claims 1 to 5, characterized in that a compound of formula (Π)
in welcher A und R1 die in den Ansprüchen 1 bis 5 angegebenen Bedeutungen haben, in Gegenwart einer Base mit einer Verbindung der Formel (III) in welcher n die in den Ansprüchen 1 bis 5 angegebene Bedeutung hat und in which A and R 1 have the meanings given in claims 1 to 5, in the presence of a base with a compound of the formula (III) in which n has the meaning given in claims 1 to 5 and
X für eine Abgangsgruppe wie beispielsweise Chlor, Brom, Iod, Mesylat, Triflat oder Tosylat steht, zu einer Verbindung der Formel (IV) X represents a leaving group such as chlorine, bromine, iodine, mesylate, triflate or tosylate to give a compound of the formula (IV)
in welcher n, A und R1 die in den Ansprüchen 1 bis 5 angegebenen Bedeutungen haben, alkyliert und anschließend die 2-(Trimethylsilyl)ethyl-Estergruppe mit Hilfe einer Säure oder eines Fluorid-Reagenzes zur Carbonsäure der Formel (I) in which n, A and R 1 have the meanings given in claims 1 to 5, alkylated and then the 2-(trimethylsilyl)ethyl ester group using an acid or a fluoride reagent to give the carboxylic acid of the formula (I)
in welcher n, A und R1 die in den Ansprüchen 1 bis 5 angegebenen Bedeutungen haben, abspaltet und gegebenenfalls die so erhaltenen Verbindungen der Formel (I) in ihre Enantiomere und/oder Diastereomere trennt und/oder mit den entsprechenden (i) Lösungsmitteln und/oder (ii) Basen in ihre Solvate, Salze und/oder Solvate der Salze überführt. in which n, A and R 1 have the meanings given in claims 1 to 5, splits off and optionally separates the compounds of formula (I) thus obtained into their enantiomers and/or diastereomers and/or with the corresponding (i) solvents and /or (ii) bases converted into their solvates, salts and/or solvates of the salts.
Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, zur Behandlung und/oder Prävention von Krankheiten. A compound as defined in any one of claims 1 to 5 for the treatment and/or prevention of diseases.
Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, zur Verwendung in einem Verfahren zur Behandlung und/oder Prävention von chronisch-obstruktiver Lungenerkrankung (COPD), Lungenemphysem, chronischer Bronchitis, pulmonaler Hypertension in der COPD (PH-COPD), Bronchiektasie, Asthma, interstitiellen Lungenerkrankungen, idiopathischer Lungenfibrose (IPF) und Lungensarkoidose, von Arteriosklerose, karotider Arteriosklerose, viraler Myokarditis, Kardiomyopathie und Aneurysmen, einschließlich deren Folgeerkrankungen wie Schlaganfall, Myokardinfarkt und periphere arterielle Verschlusskrankheit, sowie von chronischen Nierenerkrankungen und dem Alport-Syndrom. A compound as defined in any one of claims 1 to 5 for use in a method for the treatment and/or prevention of chronic obstructive pulmonary disease (COPD), pulmonary emphysema, chronic bronchitis, pulmonary hypertension in COPD (PH-COPD), bronchiectasis, Asthma, interstitial lung disease, idiopathic pulmonary fibrosis (IPF) and pulmonary sarcoidosis, arteriosclerosis, carotid arteriosclerosis, viral myocarditis, cardiomyopathy and aneurysms, including their complications such as stroke, myocardial infarction and peripheral arterial disease, as well as chronic kidney disease and Alport syndrome.
9. Verwendung einer Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, zur Herstellung eines Arzneimittels zur Behandlung und/oder Prävention von chronisch-obstruktiver Lungenerkrankung (COPD), Lungenemphysem, chronischer Bronchitis, pulmonaler Hypertension in der COPD (PH-COPD), Bronchiektasie, Asthma, interstitiellen Lungenerkrankungen, idiopathischer Lungenfibrose (IPF) und Lungensarkoidose, von Arteriosklerose, karotider Arteriosklerose, viraler Myokarditis, Kardiomyopathie und Aneurysmen, einschließlich deren Folgeerkrankungen wie Schlaganfall, Myokardinfarkt und periphere arterielle Verschlusskrankheit, sowie von chronischen Nierenerkrankungen und dem Alport-Syndrom. Arzneimittel enthaltend eine Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, in Kombination mit einem oder mehreren inerten, nicht-toxischen, pharmazeutisch geeigneten Hilfsstoffen. 9. Use of a compound as defined in any one of claims 1 to 5 for the production of a medicament for the treatment and/or prevention of chronic obstructive pulmonary disease (COPD), pulmonary emphysema, chronic bronchitis, pulmonary hypertension in COPD (PH-COPD) , bronchiectasis, asthma, interstitial lung diseases, idiopathic pulmonary fibrosis (IPF) and pulmonary sarcoidosis, arteriosclerosis, carotid arteriosclerosis, viral myocarditis, cardiomyopathy and aneurysms, including their complications such as stroke, myocardial infarction and peripheral arterial disease, as well as chronic kidney disease and Alport syndrome . Medicaments containing a compound as defined in any one of claims 1 to 5, in combination with one or more inert, non-toxic, pharmaceutically suitable excipients.
Arzneimittel enthaltend eine Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, in Kombination mit einem oder mehreren weiteren Wirkstoffen ausgewählt aus der Gruppe bestehend aus Corticosteroiden, beta-adrenergen Rezeptor-Agonisten, anti-muscarinergen Substanzen, PDE 4-Inhibitoren, PDE 5-Inhibitoren, sGC -Aktivatoren, sGC-Stimulatoren, HNE-Inhibitoren, Prostacyclin-Analoga, Endothelin-Antagonisten, Statinen, antifibrotisch wirkenden Mitteln, entzündungshemmend wirkenden Mitteln, immunmodulierend wirkenden Mitteln, immunsuppressiv wirkenden Mitteln und zytotoxisch wirkenden Mitteln. Medicament containing a compound as defined in one of claims 1 to 5, in combination with one or more further active ingredients selected from the group consisting of corticosteroids, beta-adrenergic receptor agonists, anti-muscarinic substances, PDE 4 inhibitors, PDE 5 inhibitors, sGC activators, sGC stimulators, HNE inhibitors, prostacyclin analogues, endothelin antagonists, statins, antifibrotic agents, anti-inflammatory agents, immunomodulatory agents, immunosuppressive agents and cytotoxic agents.
Arzneimittel nach Anspruch 10 oder 11 zur Behandlung und/oder Prävention von chronisch-obstruktiver Lungenerkrankung (COPD), Lungenemphysem, chronischer Bronchitis, pulmonaler Hypertension in der COPD (PH-COPD), Bronchiektasie, Asthma, interstitiellen Lungenerkrankungen, idiopathischer Lungenfibrose (IPF) und Lungensarkoidose, von Arteriosklerose, karotider Arteriosklerose, viraler Myokarditis, Kardiomyopathie und Aneurysmen, einschließlich deren Folgeerkrankungen wie Schlaganfall, Myokardinfarkt und periphere arterielle Verschlusskrankheit, sowie von chronischen Nierenerkrankungen und dem Alport-Syndrom. Medicament according to claim 10 or 11 for the treatment and/or prevention of chronic obstructive pulmonary disease (COPD), pulmonary emphysema, chronic bronchitis, pulmonary hypertension in COPD (PH-COPD), bronchiectasis, asthma, interstitial lung diseases, idiopathic pulmonary fibrosis (IPF) and Pulmonary sarcoidosis, arteriosclerosis, carotid arteriosclerosis, viral myocarditis, cardiomyopathy and aneurysms, including their complications such as stroke, myocardial infarction and peripheral arterial disease, as well as chronic kidney disease and Alport syndrome.
Verfahren zur Behandlung und/oder Prävention von chronisch-obstruktiver Lungenerkrankung (COPD), Lungenemphysem, chronischer Bronchitis, pulmonaler Hypertension in der COPD (PH-COPD), Bronchiektasie, Asthma, interstitiellen Lungenerkrankungen, idiopathischer Lungenfibrose (IPF) und Lungensarkoidose, von Arteriosklerose, karotider Arteriosklerose, viraler Myokarditis, Kardiomyopathie und Aneurysmen, einschließlich deren Folgeerkrankungen wie Schlaganfall, Myokardinfarkt und periphere arterielle Verschlusskrankheit, sowie von chronischen Nierenerkrankungen und dem Alport-Syndrom bei Menschen und Tieren durch Verabreichung einer wirksamen Menge mindestens einer Verbindung, wie in einem der Ansprüche 1 bis 5 definiert, oder eines Arzneimittels, wie in einem der Ansprüche 10 bis 12 definiert. Methods for the treatment and/or prevention of chronic obstructive pulmonary disease (COPD), pulmonary emphysema, chronic bronchitis, pulmonary hypertension in COPD (PH-COPD), bronchiectasis, asthma, interstitial lung diseases, idiopathic pulmonary fibrosis (IPF) and pulmonary sarcoidosis, arteriosclerosis, carotid arteriosclerosis, viral myocarditis, cardiomyopathy and aneurysms, including their sequelae such as stroke, myocardial infarction and peripheral arterial disease, as well as chronic kidney disease and Alport syndrome in humans and animals by administering an effective amount of at least one compound as in one of claims 1 to 5, or a medicament as defined in one of claims 10 to 12.
EP15712926.3A 2014-04-03 2015-03-31 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases Withdrawn EP3126358A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP14163306 2014-04-03
PCT/EP2015/056943 WO2015150350A1 (en) 2014-04-03 2015-03-31 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases

Publications (1)

Publication Number Publication Date
EP3126358A1 true EP3126358A1 (en) 2017-02-08

Family

ID=50397042

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15712926.3A Withdrawn EP3126358A1 (en) 2014-04-03 2015-03-31 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases

Country Status (6)

Country Link
US (1) US20170114049A1 (en)
EP (1) EP3126358A1 (en)
JP (1) JP2017509665A (en)
CN (1) CN106661008A (en)
CA (1) CA2944614A1 (en)
WO (1) WO2015150350A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2018007152A (en) 2015-12-14 2018-08-15 Ironwood Pharmaceuticals Inc USE OF sGC STIMULATORS FOR THE TREATMENT OF GASTROINTESTINAL SPHINCTER DYSFUNCTION.
CN105891375B (en) * 2016-06-17 2017-12-15 安徽瑞思威尔科技有限公司 A kind of method that chiral ethyl lactate in phase chromatogram quick detection white wine is closed using ultra high efficiency
WO2018111795A2 (en) 2016-12-13 2018-06-21 Ironwood Pharmaceuticals, Inc. Use of sgc stimulators for the treatment of esophageal motility disorders
CN108084104A (en) * 2017-12-27 2018-05-29 温州大学 1,2,3- phentriazine -4(3H)The synthetic method of -one compound
EP3820476A1 (en) 2018-07-11 2021-05-19 Cyclerion Therapeutics, Inc. Use of sgc stimulators for the treatment of mitochonrial disorders

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5789434A (en) 1994-11-15 1998-08-04 Bayer Corporation Derivatives of substituted 4-biarylbutyric acid as matrix metalloprotease inhibitors
MY132470A (en) 1996-05-15 2007-10-31 Bayer Corp Substituted oxobutyric acids as matrix metalloprotease inhibitors
MY116903A (en) * 1996-05-15 2004-04-30 Bayer Corp Substituted 4-arylbutyric acid derivatives as matrix metalloprotease inhibitors
TNSN97083A1 (en) * 1996-05-15 2005-03-15 Bayer Corp INHIBITION OF METALLOPROTEASE MATRICES BY SUBSTITUTION OF OXOBUTYRIC BIARYL ACID
TNSN97082A1 (en) 1996-05-15 2005-03-15 Bayer Corp INHIBITION OF METALLOPROTEASE MATRICES BY SUBSTITUTION OF PHENETYLE COMPOUNDS
ZA974031B (en) 1996-05-15 1998-02-19 Bayer Ag Biaryl acetylenes as inhibitors of matrix metalloproteases.
TNSN97081A1 (en) 1996-05-15 2005-03-15 Bayer Corp INHIBITION OF METALLOPROTEINASE MATRICES BY 2- (W-AROYLKYL) -4-BIARYL-4-OXUBUTYRIC ACIDS
AU4159197A (en) 1996-09-04 1998-03-26 Warner-Lambert Company Biphenyl butyric acids and their derivatives as inhibitors of matrix metalloproteinases
US6399612B1 (en) 1997-10-06 2002-06-04 Warner-Lambert Company Heteroaryl butyric acids and their derivatives as inhibitors of matrix metalloproteinases
DE19834047A1 (en) 1998-07-29 2000-02-03 Bayer Ag Substituted pyrazole derivatives
DE19834044A1 (en) 1998-07-29 2000-02-03 Bayer Ag New substituted pyrazole derivatives
BR9916669A (en) 1998-12-30 2001-10-16 Bayer Ag Use of derivatives of substituted 4-biarylbutyric and 5-biarylpentanoic acids as matrix metalloprotease inhibitors for the treatment of respiratory diseases
DE19943636A1 (en) 1999-09-13 2001-03-15 Bayer Ag Novel dicarboxylic acid derivatives with pharmaceutical properties
DE19943639A1 (en) 1999-09-13 2001-03-15 Bayer Ag Dicarboxylic acid derivatives with novel pharmaceutical properties
DE19943635A1 (en) 1999-09-13 2001-03-15 Bayer Ag Novel aminodicarboxylic acid derivatives with pharmaceutical properties
DE19943634A1 (en) 1999-09-13 2001-04-12 Bayer Ag Novel dicarboxylic acid derivatives with pharmaceutical properties
AR031176A1 (en) 2000-11-22 2003-09-10 Bayer Ag NEW DERIVATIVES OF PIRAZOLPIRIDINA SUBSTITUTED WITH PIRIDINE
DE10110749A1 (en) 2001-03-07 2002-09-12 Bayer Ag Substituted aminodicarboxylic acid derivatives
DE10110750A1 (en) 2001-03-07 2002-09-12 Bayer Ag Novel aminodicarboxylic acid derivatives with pharmaceutical properties
DE10220570A1 (en) 2002-05-08 2003-11-20 Bayer Ag Carbamate-substituted pyrazolopyridines
AU2003293356A1 (en) 2002-08-27 2004-03-19 Bayer Healthcare Ag Dihydropyridinone derivatives as hne inhibitors
GB0219896D0 (en) 2002-08-27 2002-10-02 Bayer Ag Dihydropyridine derivatives
EP1539710B1 (en) 2002-09-10 2010-10-27 Bayer Schering Pharma Aktiengesellschaft Heterocyclic derivatives
US7687510B2 (en) 2002-09-10 2010-03-30 Bayer Healthcare Ag Pyrimidinone derivatives as therapeutic agents against acute and chronic inflammatory, ischaemic and remodelling processes
JP2006524248A (en) 2003-04-14 2006-10-26 ジ インスチチュート フォー ファーマシューティカル ディスカバリー、エルエルシー N-((((((1,3-thiazol-2-yl) amino) carbonyl) phenyl) sulfonyl) phenylalanine derivatives and related compounds for the treatment of diabetes
AU2004236247B2 (en) 2003-04-30 2008-11-06 The Institutes For Pharmaceutical Discovery, Llc Substituted amino carboxylic acids as inhibitors of protein tyrosine phosphatase-1 B
WO2004099170A2 (en) 2003-04-30 2004-11-18 The Institutes For Pharmaceutical Discovery, Llc Phenyl substituted carboxylic acids as inhibitors of protein tyrosine phosphatase-1b
KR20060006953A (en) 2003-04-30 2006-01-20 디 인스티튜트스 포 파마슈티컬 디스커버리, 엘엘씨 Substituted carboxylic acids
ATE516285T1 (en) 2004-02-19 2011-07-15 Bayer Schering Pharma Ag DIHYDROPYRIDINONE DERIVATIVES
US7893073B2 (en) 2004-02-26 2011-02-22 Bayer Schering Pharma Aktiengesellschaft Heterocyclic derivatives
JP4825194B2 (en) 2004-02-26 2011-11-30 バイエル・シェーリング・ファルマ・アクチェンゲゼルシャフト 1,4-Diaryl-dihydropyrimidin-2-one compounds and their use as human neutrophil elastase inhibitors
TW200630327A (en) 2004-10-28 2006-09-01 Inst For Pharm Discovery Inc Substituted phenylalkanoic acids
JP2008520683A (en) 2004-11-18 2008-06-19 ジ インスチチュート フォー ファーマシューティカル ディスカバリー、エルエルシー Heterocyclylbiphenyl derivatives as protein tyrosine phosphatase inhibitors
GB0426313D0 (en) 2004-12-01 2005-01-05 Merck Sharp & Dohme Therapeutic agents
DE102008022521A1 (en) 2008-05-07 2009-11-12 Bayer Schering Pharma Aktiengesellschaft 1,4-Diaryl-pyrimidopyridazine-2,5-diones and their use
SG10201604819TA (en) 2007-12-20 2016-07-28 Bayer Ip Gmbh 4-(4-cyano-2-thioaryl)dihydropyrimidinones and use thereof
DE102009004197A1 (en) 2009-01-09 2010-07-15 Bayer Schering Pharma Aktiengesellschaft Heterocyclic fused diaryldihydropyrimidine derivatives and their use
DE102009016553A1 (en) 2009-04-06 2010-10-07 Bayer Schering Pharma Aktiengesellschaft Sulfonamide- and sulfoximine-substituted diaryldihydropyrimidinones and their use
DE102010021637A1 (en) 2010-05-26 2011-12-01 Bayer Schering Pharma Aktiengesellschaft Substituted 5-fluoro-1H-pyrazolopyridines and their use
MX2013000198A (en) 2010-07-09 2013-01-28 Bayer Ip Gmbh Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases.
CA2807097A1 (en) 2010-07-30 2012-02-02 Ranbaxy Laboratories Limited Matrix metalloproteinase inhibitors
DE102010040233A1 (en) 2010-09-03 2012-03-08 Bayer Schering Pharma Aktiengesellschaft Bicyclic aza heterocycles and their use
CN103228634A (en) 2010-09-24 2013-07-31 兰贝克赛实验室有限公司 Matrix metalloproteinase inhibitors
DE102010043379A1 (en) 2010-11-04 2012-05-10 Bayer Schering Pharma Aktiengesellschaft Substituted 6-fluoro-1H-pyrazolo [4,3-b] pyridines and their use

Also Published As

Publication number Publication date
CN106661008A (en) 2017-05-10
CA2944614A1 (en) 2015-10-08
US20170114049A1 (en) 2017-04-27
WO2015150350A1 (en) 2015-10-08
JP2017509665A (en) 2017-04-06

Similar Documents

Publication Publication Date Title
EP2234985B1 (en) 4-(4-cyano-2-thioaryl)dihydropyrimidinones and use thereof
WO2015189117A1 (en) Heterobicyclically substituted 4-oxobutane acid derivatives and use thereof
EP2417112B1 (en) Sulfonic amide and sulfoximine-substituted diaryl-dihydropyrimidinones and usage thereof
EP2285809B1 (en) 1,4-diaryl-pyrimidopyridazine-2,5-diones and their use
WO2015150350A1 (en) 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases
DE102007061766A1 (en) New 4-(4-cyano-2-thioaryl)-dihydro-pyrimidinone compounds are human neutrophil elastase inhibitor, useful for the treatment or prevention of e.g. pulmonary arterial hypertonia, acute lung injury and diseases of the cardiovascular system
AU2020257075B2 (en) Methods and agents for treating disease
WO2015044174A1 (en) Substituted phenylalanine derivatives as modulators of factor xia
WO2015150364A1 (en) Substituted benzotriazinone butane acids and use thereof
EP3126340A2 (en) Chiral 2,5-disubstituted cyclopentanecarboxylic acid derivatives and use thereof
WO2015150363A1 (en) 2,5-disubstituted cyclopentane carboxylic acids and use thereof
EP1966218B1 (en) Novel, cyclic substituted furopyrimidine derivatives and use thereof for treating cardiovascular diseases
CN114981257A (en) Substituted pyrazolopiperidinecarboxylic acids
WO2015150366A1 (en) Cyclically substituted phenol ether derivatives and use thereof
EP3707141B1 (en) Substituted 2,4-dihydro-3h-1,2,4-triazol-3-ones and use of same
EP2158206A1 (en) Substituted furopyrimidines and use thereof
CA3204494A1 (en) Substituted pyrazolo piperidine carboxylic acids
CN116829545A (en) Substituted pyrazolylpiperidine carboxylic acids
DE102008052013A1 (en) New 4-(4-cyano-2-thioaryl)dihydropyrimidinone compounds are neutrophil elastase inhibitors useful to treat or prevent e.g. pulmonary arterial hypertension, chronic obstructive pulmonary disease, acute lung injury, or cystic fibrosis
EP3455224B1 (en) Substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine-3(2h)-ones and 2,5,6,7-tetrahydro-3h-pyrrolo[2,1-c][1,2,4]triazol-3-ones, and use thereof
DE102010030187A1 (en) New 4-cyano-2-sulfonylphenyl-pyrazolyl-substituted pyridinones and pyrazinones compounds are human neutrophil elastase inhibitors, useful to treat and prevent e.g. pulmonary arterial hypertension and chronic obstructive pulmonary disease

Legal Events

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

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

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

Free format text: ORIGINAL CODE: 0009012

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20161103

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Extension state: BA ME

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

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

18D Application deemed to be withdrawn

Effective date: 20181002