US20140057926A1 - Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity - Google Patents

Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity Download PDF

Info

Publication number
US20140057926A1
US20140057926A1 US13/971,378 US201313971378A US2014057926A1 US 20140057926 A1 US20140057926 A1 US 20140057926A1 US 201313971378 A US201313971378 A US 201313971378A US 2014057926 A1 US2014057926 A1 US 2014057926A1
Authority
US
United States
Prior art keywords
alkyl
methyl
phenyl
preparation
cycloalkyl
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.)
Abandoned
Application number
US13/971,378
Other languages
English (en)
Inventor
Thorsten Oost
Dennis Fiegen
Christian GNAMM
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.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Assigned to BOEHRINGER INGELHEIM INTERNATIONAL GMBH reassignment BOEHRINGER INGELHEIM INTERNATIONAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FIEGEN, DENNIS, OOST, THORSTEN, GNAMM, Christian
Publication of US20140057926A1 publication Critical patent/US20140057926A1/en
Priority to US14/981,999 priority Critical patent/US9346794B1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D405/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • This invention relates to substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of pulmonary, gastrointestinal and genitourinary diseases, inflammatory diseases of the skin and the eye and other auto-immune and allergic disorders, allograft rejection, and oncological diseases.
  • Neutrophil elastase is a 29 kDa serine protease. It is expressed in bone marrow precursor cells, stored in the granula of peripheral blood granulocytes at high concentrations and it is released upon cellular activation. To the substrates of NE belong major elements of the extracellular matrix: elastin, fibronectin, laminin, collagen and proteoglycans. Neutrophil elastase activity leads to ECM degradation, increases migration and chemotaxis of monocytes and vascular smooth muscle cells and directly affects components of the coagulation and fibrinolytic pathways (PAI-1 and TFPI). Increased activity of neutrophil elastase is associated with chronic inflammatory and fibrotic diseases of several organs.
  • PAI-1 and TFPI coagulation and fibrinolytic pathways
  • Inhibitors of neutrophil elastase will therefore have an important role for the treatment of different diseases like COPD, fibrosis, cancer and others.
  • the compounds according to the present invention are effective as inhibitors of neutrophil elastase and exhibit favourable inhibitory potency, as determined by the half maximal inhibitory concentration (IC 50 ), in an enzymatic inhibition assay.
  • Some compounds according to the present invention exhibit favourable inhibitory potency, as determined by the half maximal effective concentration (EC 50 ), in a plasma or whole-blood assay, for instance as described in T. Stevens et al., J. Pharm. Exp. Ther. 339, 313-320 (2011).
  • Some compounds according to the present invention exhibit favourable in vivo potency, as determined, for example, by the half maximal effective dose (ED 50 ), in models of human neutrophil elastase-induced lung injury in mice, rat or hamster, for instance as described in Tremblay et al. ( Chest 2002, 121, 582-588) or T. Stevens et al. ( J. Pharm. Exp. Ther. 2011, 339, 313-320).
  • ED 50 half maximal effective dose
  • Some compounds according to the present invention exhibit favourable metabolic stability in an in vitro microsomal assay for metabolic stability as described in E. Kerns & L. Di, Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization, Elsevier, 1 st ed, 2008, chapter 29 and references therein.
  • Some compounds according to the present invention exhibit favourable metabolic stability in an in vitro hepatocytes assay for metabolic stability as described in E. Kerns & L. Di, Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization, Elsevier, 1 st ed, 2008, chapter 29 and references therein.
  • Some compounds according to the present invention exhibit favourable permeability in an in vitro Caco-2 cell layer method for permeability as described in E. Kerns & L. Di, Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization, Elsevier, 1 st ed, 2008, chapter 26 and references therein.
  • improved permeability is expected to translate into a higher fraction of the drug absorbed in the intestinal tract, thus, resulting in higher dose-normalized systemic exposure (AUC).
  • Comparatively higher dose-normalized systemic exposure can be advantageous in several ways: (1) If a certain systemic exposure (AUC) needs to be achieved for efficacy, the drug can be dosed in a lower amount. Lower dosages have the advantages of lower drug load (parent drug and metabolites thereof) for the patient causing potentially less side effects, and lower production costs for the drug product. (2) Comparatively higher dose-normalized systemic exposure (AUC) can lead to increased efficacy or prolonged duration of action of the drug when the same dose is applied.
  • Some compounds according to the present invention exhibit favourable metabolic stability and favourable permeability. Accordingly, some compounds of the present invention are expected to exhibit favourable pharmacokinetic (PK) properties, in particular favourable systemic exposure (area under the curve, AUC).
  • PK pharmacokinetic
  • PK properties can be determined in pre-clinical animal species, for example mouse, rat, dog, guinea pig, mini pig, cynomolgus monkey, rhesus monkey.
  • the PK properties of a compound can be described, for example, by the following parameters: Mean residence time (MRT), elimination half-live (t 1/2 ), volume-of-distribution (V D ), area under the curve (AUC), clearance (CL), bioavailability after oral administration (L oral ).
  • R 4 is one of the above mentioned ring carrying the above mentioned optional substituted in meta-position to the element connection R 4 with the compound of formula 1.
  • Example # Structure 1 2 4 5 6 8 10 11 12 18 19 21 29 30 31 41 42 47 # example numbers are according to the numbering in the Preparation/experimental section
  • the residue R 1a is chiral, preferred is the Eutomer.
  • C 1-6 -alkyl means an alkyl group or radical having 1 to 6 carbon atoms.
  • radical attachment point(s) to the molecule from the free valences of the group itself.
  • the last named subgroup is the radical attachment point, for example, the substituent “aryl-C 1-3 -alkyl-” means an aryl group which is bound to a C 1-3 -alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached.
  • 3-carboxypropyl-group represents the following substituent:
  • a X with a subscript number or “#”, a dashed or a dotted line may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound.
  • prevention should be understood synonymous and in the sense that the risk to develop a condition mentioned hereinbefore is reduced, especially in a patient having elevated risk for said conditions or a corresponding anamnesis, e.g. elevated risk of developing metabolic disorder such as diabetes or obesity or another disorder mentioned herein.
  • prevention of a disease means the management and care of an individual at risk of developing the disease prior to the clinical onset of the disease.
  • the purpose of prevention is to combat the development of the disease, condition or disorder, and includes the administration of the active compounds to prevent or delay the onset of the symptoms or complications and to prevent or delay the development of related diseases, conditions or disorders. Success of said preventive treatment is reflected statistically by reduced incidence of said condition within a patient population at risk for this condition in comparison to an equivalent patient population without preventive treatment.
  • treatment means therapeutic treatment of patients having already developed one or more of said conditions in manifest, acute or chronic form, including symptomatic treatment in order to relieve symptoms of the specific indication or causal treatment in order to reverse or partially reverse the condition or to delay the progression of the indication as far as this may be possible, depending on the condition and the severity thereof.
  • treatment of a disease means the management and care of a patient having developed the disease, condition or disorder.
  • the purpose of treatment is to combat the disease, condition or disorder.
  • Treatment includes the administration of the active compounds to eliminate or control the disease, condition or disorder as well as to alleviate the symptoms or complications associated with the disease, condition or disorder.
  • a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc. . . . ) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound.
  • halogen generally denotes fluorine, chlorine, bromine and iodine.
  • prodrug refers to (i) an inactive form of a drug that exerts its effects after metabolic processes within the body converting it to a usable or active form, or (ii) a substance that gives rise to a pharmacologically active metabolite, although not itself active (i.e. an inactive precursor).
  • prodrug or “prodrug derivative” mean a covalently-bonded derivative, carrier or precursor of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s).
  • prodrugs either have metabolically cleavable or otherwise convertible groups and are rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood or by activation via oxidation as in case of thioether groups.
  • Most common prodrugs include esters and amide analogs of the parent compounds.
  • prodrug is formulated with the objectives of improved chemical stability, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity).
  • prodrugs themselves have weak or no biological activity and are stable under ordinary conditions.
  • Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: “Design and Applications of Prodrugs”; Design of Prodrugs, H.
  • pharmaceutically acceptable prodrug means a prodrug of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • such salts include salts from ammonia, L-arginine, betaine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine (2,2′-iminobis(ethanol)), diethylamine, 2-(diethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-glucamine, hydrabamine, 1H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, 1-(2-hydroxyethyl)-pyrrolidinyl, sodium hydroxide, triethanolamine (2,2′,2′′-nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2,2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof.
  • Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention e.g. trifluoro acetate salts
  • Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention also comprise a part of the invention.
  • C 1-n -alkyl wherein n is an integer from 2 to 6, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to 6 C atoms.
  • C 1-5 -alkyl embraces the radicals H 3 C—, H 3 C—CH 2 —, H 3 C—CH 2 —CH 2 —, H 3 C—CH(CH 3 )—, H 3 C—CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH(CH 3 )—, H 3 C—CH(CH 3 )—CH 2 —, H 3 C—C(CH 3 ) 2 —, H 3 C—CH 2 —CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH 2 —CH(CH 3 )—, H 3 C—CH 2 —CH(CH 3 )—CH 2 —CH 2 —, H 3 C—CH 2 —CH(CH 3 )—, H
  • C 1-6 -alkylene wherein n is an integer 2 to 6, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to 6 carbon atoms.
  • C 1-4 -alkylene includes —CH 2 —, —CH 2 —CH 2 —, —CH(CH 3 )—, —CH 2 —CH 2 —CH 2 —, —C(CH 3 ) 2 —, —CH(CH 2 CH 3 )—, —CH(CH 3 )—CH 2 —, —CH 2 —CH(CH 3 )—, —CH 2 —CH 2 —CH 2 —CH 2 —, —CH 2 —CH 2 —CH(CH 3 )—, —CH(CH 3 )—CH 2 —CH 2 —, —CH 2 —CH(CH 3 )—, —CH(CH 3 )—CH 2 —CH 2 —, —
  • C 2-n -alkylene wherein n is an integer 3 to 5, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 2 to 5 carbon atoms.
  • C 2-5 -alkylene includes —CH 2 —CH 2 —, —CH(CH 3 )—, CH 2 —CH 2 —CH 2 —, —C(CH 3 ) 2-9 —CH(CH 2 CH 3 )—, CH(CH 3 )—CH 2 —, —CH 2 —CH(CH 3 )—, CH 2 —CH 2 —CH 2 —CH 2-9 —CH 2 —CH 2 —CH(CH 3 )—, CH(CH 3 )—CH 2 —CH 2 —, —CH 2 —CH(CH 3 )—CH 2-9 —CH 2 —C(CH 3 ) 2-9 —C(CH 3 ) 2 —CH 2-9 —CH(CH(CH 3
  • halo added to a “alkyl”, “alkylene” or “cycloalkyl” group (saturated or unsaturated) is such a alkyl or cycloalkyl group meant wherein one or more hydrogen atoms are replaced by a halogen atom selected from among fluorine, chlorine or bromine, preferably fluorine and chlorine, particularly preferred is fluorine. Examples include: H 2 FC—, HF 2 C—, F 3 C—.
  • C 3-6 -cycloalkyl wherein n is an integer from 4 to 6, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to 6 C atoms.
  • C 3-6 -cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • aryl as used herein, either alone or in combination with another radical, denotes a carbocyclic aromatic monocyclic group containing 6 carbon atoms which may be further fused to a second five- or six-membered, carbocyclic group which may be aromatic, saturated or unsaturated.
  • Aryl includes, but is not limited to, phenyl, indanyl, indenyl, naphthyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl and dihydronaphthyl.
  • a phenyl ring contains 6 elements which are all carbon atoms
  • a pyrrol ring contains 5 elements, wherein 4 elements are carbon atoms and the remaining element is a nitrogen atom.
  • the term is intended to include all possible isomeric forms.
  • the term includes (if not otherwise restricted) the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • the compounds according to the present invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis.
  • the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section.
  • the order in carrying out the reaction steps may be varied. Variants of the reaction methods that are known to the one skilled in the art but not described in detail here may also be used.
  • the general processes for preparing the compounds according to the invention will become apparent to the one skilled in the art studying the following schemes.
  • Starting materials are commercially available or may be prepared by methods that are described in the literature or herein, or may be prepared in an analogous or similar manner. Any functional groups in the starting materials or intermediates may be protected using conventional protecting groups. These protecting groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the one skilled in the art.
  • Starting material I can be prepared as described in US2003/87940.
  • Intermediates II can be prepared as described in WO10133973 and US2003/87940 by heating starting material I with amines R—NH 2 in the presence of a strong base, for example sodium tert-butoxide or sodium ethoxide, in an organic solvent, for example ethanol. The reaction usually takes place within 2 to 72 hours. Preferred reaction temperatures are between 50° C. and 150° C.
  • the amide coupling (Step B, intermediates II ⁇ intermediates III, intermediates IV ⁇ intermediates V, intermediates VI ⁇ compounds of the invention) can be achieved by reacting carboxylic acid intermediates II, IV or VI with amines R′—NH 2 in the presence of an amide coupling reagent, for example O-(Benzotriazol-1-yl)-N,N,N′,N-tetramethyluronium hexafluorophosphate (HBTU), O-(Benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) or propane phosphonic acid anhydride, and in the presence of a base, for example triethylamine, diisopropylethylamine (DIPEA, Hünig's base) or N-methyl-morpholine, in an organic solvent, for example N,N-dimethylformamide (DMF), N-methyl-2-pyrroli
  • reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • carboxylic acid intermediates can be activated first as described in US2003/87940, for example with 1,1′-carbonyldiimidazole (CDI) in DMF, followed by reaction with the amine R′—NH 2 .
  • CDI 1,1′-carbonyldiimidazole
  • the bromination (Step C, X ⁇ Br, intermediates II ⁇ intermediates IV, intermediates III ⁇ intermediates V) can be achieved by reacting intermediates II or III with bromination agents, for example bromine or N-bromosuccinimide, in an organic solvent, for example acetic acid, dichloromethane, methanol, acetonitrile, tetrahydrofuran or mixtures thereof.
  • bromination agents for example bromine or N-bromosuccinimide
  • the iodination (Step C, X ⁇ I, intermediates II ⁇ intermediates IV, intermediates III ⁇ intermediates V) can be achieved by reacting intermediates II or III with iodination agents, for example iodine, iodinechloride (I—Cl) or N-iodosuccinimide, in an organic solvent, for example acetic acid, methanol, ethanol, dichloromethane, acetonitrile, N,N-dimethyl-formamide, tetrahydrofuran or mixtures thereof.
  • iodination agents for example iodine, iodinechloride (I—Cl) or N-iodosuccinimide
  • organic solvent for example acetic acid, methanol, ethanol, dichloromethane, acetonitrile, N,N-dimethyl-formamide, tetrahydrofuran or mixtures thereof.
  • the halogenation reaction usually takes place within 1 to 72
  • Step D intermediates IV ⁇ intermediates VI, intermediates V ⁇ compounds according to the invention
  • aryl or heteroaryl boronic acids R′′—B(OH) 2 or the corresponding boronic esters in the presence of a palladium catalyst, for example tetrakis(triphenylphosphine)palladium(0) or [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II)
  • a base for example, potassium carbonate, barium dihydroxide or cesium carbonate
  • organic solvent for example toluene, benzene, ethanol, ethylene glycol dimethyl ether, acetonitrile, dioxane or mixtures thereof, optionally in the presence of water.
  • Preferred reaction temperatures are between 50° C. and 150° C.
  • the alkylation of the pyridone nitrogen can be achieved by reacting intermediate VII with alkylating agents, for example alkyl bromides, alkyl iodides, alkyl tosylates, alkyl mesylates or dialkyl sulfates, in the presence of a base, for example sodium carbonate, potassium carbonate, potassium hydroxide, sodium hydroxide or cesium carbonate, in an organic solvent, N,N-dimethylformamide (DMF), N-methyl-2-pyrrolidone (NMP) or dimethylacetamide (DMA).
  • alkylating agents for example alkyl bromides, alkyl iodides, alkyl tosylates, alkyl mesylates or dialkyl sulfates
  • a base for example sodium carbonate, potassium carbonate, potassium hydroxide, sodium hydroxide or cesium carbonate
  • DMF N,N-dimethylformamide
  • NMP N-methyl-2-pyrrolidone
  • Intermediates VIII can be prepared as described in Bolm et al., Org. Lett. 9, 3809-3811 (2007) via direct imination of a thioether (intermediates VII) with cyanamide (Step F, intermediates VII ⁇ intermediates VIII) by reaction with an halogenating agent, for example N-bromo- or N-chlorosuccinimide, tert-butyl hypochlorite or iodine in the presence of cyanamide and a base, for example potassium or sodium tert-butoxide, in an organic solvent, for example methanol, tetrahydrofuran (THF) or acetonitrile.
  • an organic solvent for example methanol, tetrahydrofuran (THF) or acetonitrile.
  • Preferred reaction temperatures are between 0° C. and 50° C., most preferred is room temperature.
  • the imination (Step F, intermediates VII ⁇ intermediates VIII) can be achieved as described in Bolm et al., Org. Lett. 9, 2951-2954 (2007), by using phenyliodo diacetate [PhI(OAc) 2 ] and cyanamide in an organic solvent, for example acetonitrile or tetrahydrofuran (THF).
  • the reaction usually takes place within 1 to 72 hours.
  • Preferred reaction temperatures are between 0° C. and room temperature.
  • Step G intermediates VIII ⁇ intermediates IX, intermediates XI ⁇ intermediates XII
  • a peroxycarboxylic acid for example meta-chloroperoxybenzoic acid (mCPBA)
  • mCPBA meta-chloroperoxybenzoic acid
  • a base for example potassium carbonate
  • organic solvent for example ethanol, methanol, dichloromethane or chloroform.
  • Preferred reaction temperatures are between 0° C. and room temperature.
  • other common oxidizing agents may be used to achieve this transformation, for example hydrogen peroxide, tert-butyl hydroperoxide, sodium hypochlorite, sodium iodate, sodium periodate, potassium permanganate, ruthenium tetroxide, potassium peroxymonosulfate (Oxone) or dimethyldioxirane.
  • Intermediates X can be obtained by imination of intermediates VII (Step H, intermediates VII ⁇ intermediates X) as described in Bolm. et al., Org. Lett. 6, 1305-1307 (2004), by reacting a thioether (intermediates VII) with trifluoroacetamide, phenyliodo diacetate [PhI(OAc) 2 ], rhodium acetate dimer [Rh 2 (OAc) 4 ] and MgO in an organic solvent, for example dichloromethane. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0° C. and room temperature.
  • Step K intermediates X ⁇ intermediates XI
  • a base for example potassium carbonate
  • organic solvent for example methanol
  • intermediates XI can be prepared by the imination of intermediates VII using other electrophilic nitrogen sources, for example tert-butyl 3-(4-cyano-phenyl)-oxaziridine-2-carboxylate (followed by cleavage of the Boc-protecting group), O-mesityl sulfonyl hydroxylamine (MSH), or hydrazoic azid.
  • other electrophilic nitrogen sources for example tert-butyl 3-(4-cyano-phenyl)-oxaziridine-2-carboxylate (followed by cleavage of the Boc-protecting group), O-mesityl sulfonyl hydroxylamine (MSH), or hydrazoic azid.
  • Step K intermediates XI ⁇ intermediates VIII, intermediates XII ⁇ intermediates IX
  • Step K intermediates XI ⁇ intermediates VIII, intermediates XII ⁇ intermediates IX
  • cyanogen bromide optionally 4-dimethylaminopyridine (DMAP) and optionally a base, for example triethylamine
  • DMAP 4-dimethylaminopyridine
  • a base for example triethylamine
  • Sulfinamides (starting material XIII) can be prepared as described in Johnson et al., J. Org. Chem. 44, 2055-2061 (1975) by reacting sulfinyl chlorides with a primary or secondary amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example dichloromethane, tetrahydrofuran, diethylether, acetonitrile, toluene, N,N-dimethylformamide, ethanol or ethyl acetate.
  • the reaction usually takes place within 1-72 hours.
  • Preferred reaction temperatures are between 0° C. and room temperature.
  • Sulfonimidoyl chlorides can be prepared as described in Johnson et al., J. Org. Chem. 44, 2055-2061 (1975), by reacting sulfinamides (starting material XIII) with a chlorinating agent, for example chlorine, N-chlorosuccinimide (NCS), N-chloro-benzotriazole or tert-butyl hypochlorite in an organic solvent, for example dichloromethane, diethylether, benzene, toluene, carbon tetrachloride, acetonitrile or tetrahydrofuran (Step L).
  • a chlorinating agent for example chlorine, N-chlorosuccinimide (NCS), N-chloro-benzotriazole or tert-butyl hypochlorite
  • an organic solvent for example dichloromethane, diethylether, benzene, toluene, carbon tetrach
  • Sulfonimidoyl fluorides can be prepared as described in Johnson et al., J. Org. Chem. 48, 1-3 (1983) and Gnamm et al., Bioorg. Med. Chem. Lett. 12, 3800-3806 (2012), by reacting sulfonimidoyl chlorides (intermediates XIV) with a fluoride salt, for example potassium fluoride, sodium fluoride or tetrabutylammonium fluoride, optionally in the presence of a crown ether, for example 18-crown-6, in an organic solvent, for example acetonitrile or tetrahydrofuran (Step M). The reaction takes place within 1 to 24 hours. Preferred reaction temperatures are between 0° C. and room temperature, mostly preferred room temperature.
  • Sulfonimidamides can be prepared as described in Johnson et al., J. Org. Chem. 44, 2055-2061 (1975), Bolm et al., J. Org. Chem. 75, 3301-3310 (2010), and WO09156336 by reacting sulfonimidoyl chlorides (intermediates XIV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step N). The reaction takes place within 1 to 72 hours. Preferred reaction temperatures are between 0° C. and room temperature.
  • sulfonimidamides can be prepared as described in Gnamm et al., Bioorg. Med. Chem. Lett. 22, 3800-3806 (2012) by reacting sulfonimidoyl fluorides (intermediates XV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, for example 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU), in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step O).
  • the reaction takes place within 1 to 72 hours.
  • Preferred reaction temperatures are between 0° C. and the boiling point of the solvent, mostly preferred between room temperature and 110° C.
  • Preparation 2a is prepared following the procedure described for preparation 1b, substituting isopropylamine with ethylamine.
  • ESI mass spectrum: [M+H] + 182; r.t.
  • Preparation 2b is prepared following the procedure described for preparation 1c, substituting preparation 1b with preparation 2a as starting material.
  • ESI mass spectrum: [M+H] + 260 (bromine isotope pattern); r.t. HPLC: 0.89 min (Z002 — 007).
  • Preparation 2c is prepared following the procedure described for preparation 1d, substituting preparation 1c with preparation 2b as starting material and substituting TBTU with HBTU as coupling reagent.
  • ESI mass spectrum: [M+H] + 451 (bromine isotope pattern); r.t. HPLC: 0.81 min (Z012_SO 4 ).
  • Preparation 3a is prepared following the procedure described for preparation 1b, substituting isopropylamine with cyclopropylamine.
  • ESI mass spectrum: [M+H] + 208; r.t. HPLC: 0.62 min (Z002 — 002).
  • Preparation 3b is prepared following the procedure described for preparation 1c, substituting preparation 1b with preparation 3a as starting material.
  • ESI mass spectrum: [M+H] + 286 (bromine isotope pattern); r.t. HPLC: 0.82 min (Z002 — 002).
  • Preparation 3c is prepared following the procedure described for preparation 1d, substituting preparation 1c with preparation 3b as starting material and substituting TBTU with HBTU as coupling reagent.
  • ESI mass spectrum: [M+H] + 477 (bromine isotope pattern); r.t. HPLC: 0.90 min (Z018_S04).
  • Preparation 4a is prepared following the procedure described for preparation 1b, substituting isopropylamine with 1-methyl-1H-pyrazol-5-ylamine.
  • ESI mass spectrum: [M+H] + 234; r.t. HPLC: 0.63 min (Z018_S04).
  • Preparation 4b is prepared following the procedure described for preparation 1d, substituting preparation 1c with preparation 4a as starting material and substituting TBTU with HBTU as coupling reagent.
  • ESI mass spectrum: [M+H] + 425; r.t. HPLC: 0.79 min (Z018_S04).
  • Preparation 5a is prepared following the procedure described for preparation 1b, substituting isopropylamine with 4-cyanobenzylamine.
  • ESI mass spectrum: [M+H] + 269; r.t. HPLC: 0.86 min (Z002 — 005).
  • Preparation 5b is prepared following the procedure described for preparation 1c, substituting preparation 1b with preparation 5a as starting material.
  • ESI mass spectrum: [M+H] + 347 (bromine isotope pattern); r.t. HPLC: 1.07 min (Z002 — 005).
  • racemic example 1 63 mg are separated by chiral HPLC (Daicel IB, 250 mm ⁇ 20 mm, 15% MeOH+0.2% diethylamine in supercritical CO 2 , 40° C.).
  • Example 2 is prepared following the procedure described for example 1, substituting 3-(difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid.
  • ESI mass spectrum: [M+H] + 531; r.t. HPLC: 0.99 min (Z003 — 001).
  • racemic example 2 100 mg are separated by chiral HPLC (Daicel OZ-H, 250 mm ⁇ 20 mm, 40% MeOH+0.2% diethylamine in supercritical CO 2 , 40° C.).
  • Example 3 is prepared following the procedure described for example 1, substituting 3-(difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid.
  • ESI mass spectrum: [M+H] + 532; r.t. HPLC: 0.86 min (Z003 — 001).
  • racemic example 3 50 mg are separated by chiral HPLC (Daicel IB, 250 mm ⁇ 20 mm, 15% MeOH+0.2% diethylamine in supercritical CO 2 , 40° C.).
  • Example 4 is prepared following the procedure described for example 1, substituting 3-(difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid and substituting preparation 1 with preparation 2 as starting material.
  • ESI mass spectrum: [M+H] + 517; r.t. HPLC: 0.85 min (Z011_S03).
  • racemic example 4 are separated by chiral HPLC (Daicel OZH, 250 mm ⁇ 20 mm, 40% MeOH+0.2% diethylamine in supercritical CO 2 , 40° C.).
  • Example 5 is prepared following the procedure described for example 1, substituting preparation 1 with preparation 2 as starting material.
  • ESI mass spectrum: [M+H] + 499; r.t. HPLC: 0.79 min (Z011_S03).
  • Example 6 is prepared following the procedure described for example 1, substituting preparation 1 with preparation 3 as starting material.
  • ESI mass spectrum: [M+H] + 525; r.t. HPLC: 0.84 min (Z011_S03).
  • Example 7 is prepared following the procedure described for example 1, substituting preparation 1 with preparation 4 as starting material.
  • ESI mass spectrum: [M+H] + 551; r.t. HPLC: 0.96 min (Z018_S04).
  • Example 8 is prepared following the procedure described for example 1, substituting 3-(difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid and substituting preparation 1 with preparation 4 as starting material.
  • ESI mass spectrum: [M+H] + 569; r.t. HPLC: 1.00 min (Z018_S04).
  • Example 9 is prepared following the procedure described for preparation 1d, substituting preparation 1c with preparation 5 as starting material.
  • ESI mass spectrum: [M+H] + 604; r.t. HPLC: 0.63 min (X012_S01).
  • racemic example 9 58 mg are separated by chiral HPLC (Daicel ASH, 250 mm ⁇ 20 mm, 20% MeOH+0.2% diethylamine in supercritical CO 2 , 40° C.).
  • bromo intermediates are prepared as described for preparation 1c, employing the appropriate des-bromo intermediates as starting material.
  • Racemic preparation 6c (2.0 g) is separated by chiral HPLC (Daicel Chiralcel OZ-H, 250 mm ⁇ 30 mm, 32% MeOH in supercritical CO 2 ).
  • racemic preparation 6d The early eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-S-methyl-sulfonimidoyl)benzyl)isoindoline-1,3-dione with preparation 11A.
  • the late eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-S-methylsulfonimidoyl)-benzyl)isoindoline-1,3-dione with preparation 11B.
  • Phthalic anhydride (4.42 g, 30 mmol) is added to a solution of 4-(ethylthio)benzylamine (5.00 g, 30 mmol) in acetic acid. The mixture is heated at reflux for 5 h and cooled at room temperature. Water ist added, and the precipitate is filtered and dried. The residue is dissolved in methanol (50 mL), and the mixture is treated with potassium tert-butoxide (2.26 g, 20 mmol) and cyanamide (920 mg, 22 mmol). N-Bromosuccinimide (4.49 g, 25 mmol) is added, and the mixture is stirred at room temperature for 2 h.
  • a solution of 5-methylsulfanyl-pyridine-2-carbonitrile (preparation described in WO05026124, 4.28 g, 28.5 mmol) in THF is cooled at ⁇ 78° C. in a dry ice/ethanol bath.
  • Lithium aluminium hydride (2.4 M in THF, 12 mL, 28.8 mmol) is added dropwise. After 10 min water (1 mL) and THF (3 mL) are added. After 5 min, aqueous sodium hydroxide solution (4 M, 1 mL) and water (3 mL) is added.
  • 60a Thioacetic acid 4-(1,3-dioxo-1,3-dihydro-isoindol-2-ylmethyl)-phenyl ester
  • Example 61 is prepared as described for Example 60, substituting preparation 9.2 with preparation 9.12.
  • ESI mass spectrum: [M+H] + 531; r.t. HPLC: 0.86 min (Z011 — 503).
  • Meta-chloroperoxybenzoic acid (77%, 8.0 mg, 36 ⁇ mol) is added to a solution of 5-(3-trifluoromethyl-phenyl)-1-isopropyl-6-methyl-4-oxo-1,4-dihydro-pyridine-3-carboxylic acid (5-(N-cyano-S-methylsulfonimidoyl)pyridin-2-yl)methylamide (example 16, 17.0 mg, 32 ⁇ mol) in dichloromethane (0.5 mL), and the mixture is stirred at room temperature for 2 h. The mixture is heated at reflux and stirred for 2 h. The mixture is cooled at room temperature and stirred over night.
  • a solution of lithium aluminium hydride (1.0 M in THF, 18.4 mL, 18.4 mmol) is cooled in an ice bath and treated slowly with a solution of 3-methyl-4-methylsulfanyl-benzonitrile (preparation 66a, 1.0 g, 6.1 mmol) in THF (10 mL) while the temperature is kept below 5° C.
  • the mixture is allowed to warm to room temperature and stirred over night.
  • the mixture is cooled again at 0° C., and water (2 mL) and aqueous sodium hydroxide (4 M, 2 mL) is added.
  • the mixture is filtered and the filtrate is extracted twice with dichloromethane.
  • the combined organic layers are concentrated under reduced pressure.
  • Human neutrophil elastase was purchased from Calbiochem (Cat.No.: 324681) and the elastase substrate MeOSuc-Ala-Ala-Pro-Val-AMC from Bachem (Cat.No.: I-1270). All other materials were of the highest grade commercially available.
  • Compound buffer 100 mM Tris, 500 mM NaCl, adjusted to pH 7.5
  • Assay buffer 100 mM Tris, 500 mM NaCl, adjusted to pH 7.5, containing 0.01% BSA.
  • Test compounds were prediluted in DMSO and subsequently in compound buffer (5% DMSO final). 5 ⁇ L of these compound dilutions were mixed with 10 ⁇ l Neutrophil elastase (9 ng/ml in assay buffer) in a black 384 well OptiPlate (Perkin Elmer, Cat No.: 6007270) and incubated for 15 min at room temperature. Subsequently 10 ⁇ L substrate solution in assay buffer were added (250 ⁇ M final concentration) and the plates were incubated for 60 min at room temperature. After inactivation of the enzyme, fluorescence intensities were measured at 380 nm excitation and 460 nm emission wavelengths.
  • Each plate contains wells with a high value control (DMSO+enzyme+substrate) and wells with a low value control (DMSO+inactivated enzyme+substrate).
  • 1050 values were estimated using a sigmoidal concentration response curve with variable slope. Means of low values were taken as 0%, means of high values as 100%. 1050 values of selected compound in the Neutrophil Elastase assay:
  • Example IC50 [nM] Example IC50 [nM] 1 1.3 6 2.4 1A 3.1 7 2.1 1B 1.3 8 2.4 2 1.7 9 ⁇ 1 2A 2.3 9A ⁇ 1 2B 1.1 9B ⁇ 1 3 4.7 10 1.2 3A 12.0 10A 2.7 3B 5.1 10B 1.2 4 3.0 5.5A 5.0 4A 4.1 5.5B 2.7 4B 1.6 11 ⁇ 1 5 2.5 12 ⁇ 1 13 2.4 36 1.0 14 1.5 37 ⁇ 1 15 2.2 38 3.7 16 1.9 39 3.7 17 7.3 40 4.0 18 1.5 41 1.8 19 1.0 42 4.7 20 5.6 43 13.4 21A 5.2 44 14.9 21B 2.1 45 14.4 22A 4.5 46 11.7 22B 1.2 47 1.2 23 2.6 48 9.4 24 5.5 49 7.3 25 4.3 50 8.6 26 3.3 53 2.3 27 2.5 54 2.3 28 3.3 55 1.8 29 1.5 56 3.1 29A 2.2 57 1.7 29B ⁇ 1 58 4.7 30 ⁇ 1 59 7.9
  • the compounds of general formula I may be used on their own or combined with other active substances of formula I according to the invention.
  • the compounds of general formula I may optionally also be combined with other pharmacologically active substances. These include, ⁇ 2-adrenoceptor-agonists (short and long-acting), anti-cholinergics (short and long-acting), anti-inflammatory steroids (oral and topical corticosteroids), cromoglycate, methylxanthine, dissociated-glucocorticoidmimetics, PDE3 inhibitors, PDE4-inhibitors, PDE7-inhibitors, LTD4 antagonists, EGFR-inhibitors, Dopamine agonists, PAF antagonists, Lipoxin A4 derivatives, FPRL1 modulators, LTB4-receptor (BLT1, BLT2) antagonists, Histamine H1 receptor antagonists, Histamine H4 receptor antagonists, dual Histamine H1/H3-receptor antagonists, PI3-kinase inhibitors
  • the compounds of the invention and their pharmaceutically acceptable salts have activity as pharmaceuticals, in particular as inhibitors of neutrophil elastase, and thus may be used in the treatment of:
  • respiratory tract obstructive diseases of the airways including: asthma, including bronchial, allergic, intrinsic, extrinsic, exercise-induced, drug-induced (including aspirin and NSAID-induced) and dust-induced asthma, both intermittent and persistent and of all severities, and other causes of airway hyper-responsiveness; chronic obstructive pulmonary disease (COPD); bronchitis, including infectious and eosinophilic bronchitis; emphysema; alpha1-antitrypsin deficiency; bronchiectasis; cystic fibrosis; sarcoidosis; farmer's lung and related diseases; hypersensitivity pneumonitis; lung fibrosis, including cryptogenic fibrosing alveolitis, idiopathic interstitial pneumonias, fibrosis complicating anti-neoplastic therapy and chronic infection, including tuberculosis and aspergillosis and other fungal infections; complications of lung transplantation; vasculitic
  • skin psoriasis, atopic dermatitis, contact dermatitis or other eczematous dermatoses, and delayed-type hypersensitivity reactions; phyto- and photodermatitis; seborrhoeic dermatitis, dermatitis herpetiformis, lichen planus, lichen sclerosus et atrophica, pyoderma gangrenosum, skin sarcoid, discoid lupus erythematosus, pemphigus, pemphigoid, epidermolysis bullosa, urticaria, angioedema, vasculitides, toxic erythemas, cutaneous eosinophilias, alopecia areata, male-pattern baldness, Sweet's syndrome, Weber-Christian syndrome, erythema multiforme; cellulitis, both infective and non-infective; panniculitis; cutaneous lymphomas, non-melanom
  • eyes blepharitis; conjunctivitis, including perennial and vernal allergic conjunctivitis; ulceris; anterior and posterior uveitis; choroiditis; autoimmune, degenerative or inflammatory disorders affecting the retina; ophthalmitis including sympathetic ophthalmitis; sarcoidosis; infections including viral, fungal, and bacterial; 4.
  • nephritis including interstitial and glomerulonephritis; nephrotic syndrome; cystitis including acute and chronic (interstitial) cystitis and Hunner's ulcer; acute and chronic urethritis, prostatitis, epididymitis, oophoritis and salpingitis; vulvo-vaginitis; Peyronie's disease; erectile dysfunction (both male and female); 5. allograft rejection: acute and chronic following, for example, transplantation of kidney, heart, liver, lung, bone marrow, skin or cornea or following blood transfusion; or chronic graft versus host disease; 6.
  • oncology treatment of common cancers including prostate, breast, lung, ovarian, pancreatic, bowel and colon, stomach, skin and brain tumors and malignancies affecting the bone marrow (including the leukaemias) and lymphoproliferative systems, such as Hodgkin's and non-Hodgkin's lymphoma; including the prevention and treatment of metastatic disease and tumour recurrences, and paraneoplastic syndromes; and, 8.
  • common cancers including prostate, breast, lung, ovarian, pancreatic, bowel and colon, stomach, skin and brain tumors and malignancies affecting the bone marrow (including the leukaemias) and lymphoproliferative systems, such as Hodgkin's and non-Hodgkin's lymphoma; including the prevention and treatment of metastatic disease and tumour recurrences, and paraneoplastic syndromes; and, 8.
  • infectious diseases virus diseases such as genital warts, common warts, plantar warts, hepatitis B, hepatitis C, herpes simplex virus, molluscum contagiosum, variola, human immunodeficiency virus (HIV), human papilloma virus (HPV), cytomegalovirus (CMV), varicella zoster virus (VZV), rhinovirus, adenovirus, coronavirus, influenza, para-influenza; bacterial diseases such as tuberculosis and mycobacterium avium , leprosy; other infectious diseases, such as fungal diseases, chlamydia, Candida, aspergillus , cryptococcal meningitis, Pneumocystis carnii , cryptosporidiosis, histoplasmosis, toxoplasmosis, trypanosome infection and leishmaniasis.
  • virus diseases such as genital warts, common warts,
  • a therapeutically effective dose will generally be in the range from about 0.01 mg to about 100 mg/kg of body weight per dosage of a compound of the invention; preferably, from about 0.1 mg to about 20 mg/kg of body weight per dosage.
  • the dosage range would be from about 0.7 mg to about 7000 mg per dosage of a compound of the invention, preferably from about 7.0 mg to about 1400 mg per dosage.
  • Some degree of routine dose optimization may be required to determine an optimal dosing level and pattern.
  • the active ingredient may be administered from 1 to 6 times a day.
  • the actual pharmaceutically effective amount or therapeutic dosage will of course depend on factors known by those skilled in the art such as age and weight of the patient, route of administration and severity of disease.
  • the active ingredient will be administered at dosages and in a manner which allows a pharmaceutically effective amount to be delivered based upon patient's unique condition.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US13/971,378 2012-08-23 2013-08-20 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity Abandoned US20140057926A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/981,999 US9346794B1 (en) 2012-08-23 2015-12-29 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12181542 2012-08-23
EP12181542.7 2012-08-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/981,999 Continuation US9346794B1 (en) 2012-08-23 2015-12-29 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Publications (1)

Publication Number Publication Date
US20140057926A1 true US20140057926A1 (en) 2014-02-27

Family

ID=46963420

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/971,378 Abandoned US20140057926A1 (en) 2012-08-23 2013-08-20 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
US14/981,999 Active US9346794B1 (en) 2012-08-23 2015-12-29 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/981,999 Active US9346794B1 (en) 2012-08-23 2015-12-29 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Country Status (7)

Country Link
US (2) US20140057926A1 (es)
EP (1) EP2888242B1 (es)
JP (1) JP6319747B2 (es)
AR (1) AR092205A1 (es)
TW (1) TW201422586A (es)
UY (1) UY34989A (es)
WO (1) WO2014029830A1 (es)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018211442A1 (en) 2017-05-18 2018-11-22 Pi Industries Ltd. Formimidamidine compounds useful against phytopathogenic microorganisms

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9475779B2 (en) 2014-07-31 2016-10-25 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9458113B2 (en) 2014-07-31 2016-10-04 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9440930B2 (en) 2014-07-31 2016-09-13 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9657015B2 (en) 2014-07-31 2017-05-23 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9290457B2 (en) 2014-07-31 2016-03-22 Boehringer Ingelheim International Gmbh Substituted dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
UY36547A (es) 2015-02-05 2016-06-01 Bayer Cropscience Ag Derivados heterocíclicos condensados bicíclicos sustituidos por 2-(het)arilo como pesticidas
EP3896065A1 (de) 2015-08-07 2021-10-20 Bayer CropScience Aktiengesellschaft 2-(het)aryl-substituierte kondensierte heterocyclen-derivate als schädlingsbekämpfungsmittel
WO2021053058A1 (en) 2019-09-17 2021-03-25 Mereo Biopharma 4 Limited Alvelestat for use in the treatment of graft rejection, bronchiolitis obliterans syndrome and graft versus host disease
MX2022012942A (es) 2020-04-16 2023-01-11 Mereo Biopharma 4 Ltd Metodos que involucran el inhibidor de la elastasa de neutrofilo alvelestat para el tratamiento de enfermedades respiratorias mediadas por deficiencia de alfa-1 antitripsina.
AU2022373971A1 (en) 2021-10-20 2024-04-04 Mereo Biopharma 4 Limited Neutrophil elastase inhibitors for use in the treatment of fibrosis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4844732A (en) * 1985-10-24 1989-07-04 Daicel Chemical Industries Ltd. Pyridine-3-carboxamide derivatives
WO2006098683A1 (en) * 2005-03-16 2006-09-21 Astrazeneca Ab 2-pyridine derivatives as inhibitors of neutrophile elastase

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7211595B2 (en) 2000-11-30 2007-05-01 Abbott Laboratories Farnesyltransferase inhibitors
TW200500341A (en) 2002-11-12 2005-01-01 Astrazeneca Ab Novel compounds
SE0302487D0 (sv) 2003-09-18 2003-09-18 Astrazeneca Ab Novel compounds
SE0302486D0 (sv) 2003-09-18 2003-09-18 Astrazeneca Ab Novel compounds
US20090325924A1 (en) 2005-06-30 2009-12-31 Stuart Edward GPCR Agonists
ITMI20060285A1 (it) 2006-02-16 2007-08-17 Polimeri Europa Spa Procdedimento perfezionato per la preparazione di radicali nitrosilici stabili
TW200808763A (en) 2006-05-08 2008-02-16 Astrazeneca Ab Novel compounds I
TW200808771A (en) 2006-05-08 2008-02-16 Astrazeneca Ab Novel compounds II
WO2009058076A1 (en) 2007-11-02 2009-05-07 Astrazeneca Ab 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
AU2008325288B2 (en) 2007-11-06 2011-12-22 Astrazeneca Ab Some 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
KR101596527B1 (ko) * 2008-01-23 2016-02-22 브리스톨-마이어스 스큅 컴퍼니 4-피리디논 화합물 및 암에 대한 그의 용도
KR20110036586A (ko) 2008-06-23 2011-04-07 바스프 에스이 동물 해충의 퇴치를 위한 술폭시민아미드 화합물
TW201036957A (en) 2009-02-20 2010-10-16 Astrazeneca Ab Novel salt 628
EP2261211A1 (en) 2009-05-20 2010-12-15 Université de Lille 2 Droit et Santé 1,4-dihydropyridine derivatives and their uses
EP2483244A1 (en) 2009-10-02 2012-08-08 AstraZeneca AB 2-pyridone compounds used as inhibitors of neutrophil elastase
WO2011110852A1 (en) 2010-03-10 2011-09-15 Astrazeneca Ab Polymorphic forms of 6- [2- (4 -cyanophenyl) - 2h - pyrazol - 3 - yl] - 5 -methyl - 3 - oxo - 4 - (trifluoromethyl - phenyl) 3,4-dihydropyrazine-2-carboxylic acid ethylamide

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4844732A (en) * 1985-10-24 1989-07-04 Daicel Chemical Industries Ltd. Pyridine-3-carboxamide derivatives
WO2006098683A1 (en) * 2005-03-16 2006-09-21 Astrazeneca Ab 2-pyridine derivatives as inhibitors of neutrophile elastase

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018211442A1 (en) 2017-05-18 2018-11-22 Pi Industries Ltd. Formimidamidine compounds useful against phytopathogenic microorganisms

Also Published As

Publication number Publication date
WO2014029830A1 (en) 2014-02-27
EP2888242A1 (en) 2015-07-01
UY34989A (es) 2014-02-28
AR092205A1 (es) 2015-04-08
JP2015526453A (ja) 2015-09-10
US9346794B1 (en) 2016-05-24
EP2888242B1 (en) 2016-07-13
JP6319747B2 (ja) 2018-05-09
US20160130265A1 (en) 2016-05-12
TW201422586A (zh) 2014-06-16

Similar Documents

Publication Publication Date Title
US9346794B1 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
US9340507B2 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
US9670166B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
USRE46630E1 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
US9115093B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9290457B2 (en) Substituted dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9221807B2 (en) Substituted pyridones and pyrazinones and their use as inhibitors of neutrophil elastase activity
US20160031831A1 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9475779B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9458113B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9657015B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
USRE47493E1 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
OA17445A (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Legal Events

Date Code Title Description
AS Assignment

Owner name: BOEHRINGER INGELHEIM INTERNATIONAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OOST, THORSTEN;FIEGEN, DENNIS;GNAMM, CHRISTIAN;SIGNING DATES FROM 20131122 TO 20131204;REEL/FRAME:031895/0883

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE