US20140221335A1 - Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity - Google Patents

Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity Download PDF

Info

Publication number
US20140221335A1
US20140221335A1 US14/166,146 US201414166146A US2014221335A1 US 20140221335 A1 US20140221335 A1 US 20140221335A1 US 201414166146 A US201414166146 A US 201414166146A US 2014221335 A1 US2014221335 A1 US 2014221335A1
Authority
US
United States
Prior art keywords
independently selected
group
phenyl
alkyl
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/166,146
Other languages
English (en)
Inventor
Christian GNAMM
Thorsten Oost
Stefan Peters
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
Publication of US20140221335A1 publication Critical patent/US20140221335A1/en
Priority to US14/935,915 priority Critical patent/US9290459B1/en
Priority to US14/935,931 priority patent/US20160060231A1/en
Priority to US15/229,160 priority patent/US9670166B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • 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
    • C07D401/06Heterocyclic 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 linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D401/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D405/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D413/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This invention relates to substituted bicyclic dihydropyrimidinones of formula 1
  • compositions containing the same 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 autoimmune 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).
  • PAI-1 and TFPI coagulation and fibrinolytic pathways
  • neutrophil elastase Increased activity of neutrophil elastase is associated with chronic inflammatory and fibrotic diseases of several organs Inhibitors of neutrophil elastase will therefore have an important role for the treatment of different diseases like COPD, idiopathic pulmonary fibrosis and other fibrotic diseases, cancer, acute lung injury, acute respiratory distress syndrome, bronchiectasis, cystic fibrosis, alpha1-antitrypsin deficiency and others.
  • COPD chronic inflammatory and fibrotic diseases of several organs Inhibitors of neutrophil elastase will therefore have an important role for the treatment of different diseases like COPD, idiopathic pulmonary fibrosis and other fibrotic diseases, cancer, acute lung injury, acute respiratory distress syndrome, bronchiectasis, cystic fibrosis, alpha1-antitrypsin deficiency 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 are additionally effective as inhibitors of neutrophil serin protease proteinase 3 and exhibit favourable inhibitory potency, as determined by the half maximal inhibitory concentration (IC 50 ), in an enzymatic inhibition assay.
  • This inhibitory activity on a second neutrophil serin protease may be beneficial for pharmacological efficacy.
  • 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. 2011, 339, 313-320).
  • Some compounds according to the present invention exhibit favourable in vivo potency, as determined, for example, by the half maximal effective dose (EDO, 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).
  • EEO half maximal effective dose
  • Some compounds according to the present invention exhibit favourable in vivo potency, as determined, for example, by the half maximal effective dose (EDO, in a model of LPS/FMLP-induced lung injury in hamster, for instance as described in Mitsuhashi et al. ( Br. J. Pharmacol. 1999, 126, 1147-1152).
  • EEO 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).
  • Some compounds according to the present invention exhibit a favourable, that is low efflux ratio (permeability in the efflux direction divided by the permeability in the influx direction) in an in vitro Caco-2 or MDCK cell layer method 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 27 and references therein.
  • an improved, that is reduced efflux ratio 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).
  • Some compounds according to the present invention exhibit favourable aqueous solubility in a kinetic or thermodynamic solubility method as described in E. Kerns & L. Di ( Drug - like properties: concepts, 15 structure design and methods: from ADME to toxicity optimization , Elsevier, 1 st ed, 2008), chapter 25 and references therein.
  • improved aqueous solubility is expected to translate into a higher fraction of the drug absorbed in the intestinal tract 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, favourable permeability, favourable efflux ratio and favourable aqueous solubility. Accordingly, some compounds of the present invention are expected to exhibit favourable pharmacokinetic (PK) properties after oral dosing, in particular favourable systemic exposure (area under the curve, AUC), thus, leading to favourable efficacy in vivo.
  • PK pharmacokinetic
  • PK properties can be determined in pre-clinical animal species, for example mouse, rat, hamster, 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) and bioavailability after oral administration (F oral ).
  • the compounds of the invention and metabolites thereof are devoid of the hydrazine sub-structure that causes structural alerts for mutagenicity and carcinogenicity as described in Benigni et al. ( Chem. Rev. 2011, 11, 2507-2536).
  • compounds of the invention may bear the advantage of reduced genotoxic potential.
  • Some compounds according to the present invention exhibit favourable inhibition of cytochrome P450 (CYP) isozymes in corresponding in vitro assays for CYP isozyme inhibition 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 32 and references therein.
  • CYP cytochrome P450
  • Reduced inhibition of CYP isozymes is expected to translate into a reduced risk for undesirable drug-drug interactions which is the interference of one drug with the normal metabolic or pharmacokinetic behaviour of a co-administered drug.
  • Some compounds according to the present invention exhibit favourable, i.e. low, inhibition of the hERG channel in a patch clamp assay 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 34 and references cited therein.
  • 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:
  • the asterisk 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.
  • the compounds of the present invention contain at least one asymmetrically substituted carbon atom, and may therefore be isolated as pure enantiomers or as a racemic or non-racemic mixture of both enantiomers. It will be appreciated that some of the compounds of the present invention contain more than one stereogenic center, i.e. more than one asymmetrically substituted carbon or sulfur atom, and may therefore be isolated as pure diastereomers or as diastereomeric mixtures, both in optically active or racemic forms.
  • the invention contemplates all conceivable stereoisomers, particularly the diastereomers and enantiomers mentioned herein, e.g. in substantially pure form, in enriched form (e.g. substantially free of any or all other undesired enantiomers and/or diastereomers and/or in any mixing ratio, including the racemic forms, as well as the salts thereof.
  • substantially pure stereoisomers can be obtained according to synthetic principles known to a person skilled in the field, e.g. by separation of corresponding mixtures, by using stereochemically pure starting materials and/or by stereoselective synthesis. It is known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, e.g. starting from optically active starting materials and/or by using chiral reagents.
  • Enantiomerically pure compounds of this invention or intermediates may be prepared via asymmetric synthesis, for example by preparation and subsequent separation of appropriate diastereomeric compounds or intermediates which can be separated by known methods (e.g. by chromatographic separation or crystallization) and/or by using chiral reagents, such as chiral starting materials, chiral catalysts or chiral auxiliaries.
  • 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)-pyrrolidine, 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 acid
  • 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 n, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n 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 —, H 3 C—CH(CH 3 )—CH 2 —, H 3
  • C 1-n -alkylene wherein n is an integer 2 to n, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to n 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 2 —, —CH 2 —C(CH 3 ) 2 —, —C(CH 3 ) 2 —CH 2 —, —CH(CH 3 )—CH(CH 3 )—, —CH 2 —CH(CH 2 CH 3 )—, —CH(CH 2 CH 3 )—,
  • C 3-n -cycloalkyl wherein n is an integer from 4 to n, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to n C atoms.
  • C 3-7 -cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • halo added to a “alkyl”, “alkylene” or “cycloalkyl” group (saturated or unsaturated) is such a alkyl or cycloalkyl group 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—.
  • 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.
  • heterocyclyl means a saturated or unsaturated mono- or polycyclic-ring system including aromatic ring system containing one or more elements selected from N, O, S, S(O) or S(O) 2 , consisting of 3 to 14 ring atoms wherein none of the heteroatoms is part of the aromatic ring.
  • heterocyclyl is intended to include all the possible isomeric forms; thus, the term “heterocyclyl” includes 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:
  • heteroaryl means a mono- or polycyclic-ring systems containing one or more elements selected from N, O, S, S(O) or S(O) 2 , consisting of 5 to 14 ring atoms wherein at least one of the heteroatoms is part of aromatic ring.
  • heteroaryl is intended to include all the possible isomeric forms;
  • heteroaryl includes 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:
  • R 1 is R 1.a and R 1.a is phenyl or pyridinyl; each ring optionally substituted by one, two or three residues independently selected from the group consisting of halogen, O 2 N—, NC—, H 2 N—, HO—, R 1.1 , R 1.1 O—, R 1.2 R 1.3 S—, R 1.3 (O)S— and R 1.3 (O) 2 S—.
  • R 1 is R 1.b and R 1.b is phenyl or pyridinyl; each ring optionally substituted by one, two or three residues independently selected from the group consisting of halogen, NC—, R 1.1 , R 1.3 (O)S— and, R 1.3 (O) 2 S—.
  • R 1 is R 1.c and R 1.c is phenyl or pyridinyl; each ring optionally substituted by one, two or three residues independently selected from the group consisting of F, Cl, Br—, NC—, R 1.1 , R 1.3 (O)S— and R 1.3 (O) 2 S—, and
  • R 1 is R 1.a and R 1.a is phenyl or pyridinyl; each ring optionally substituted by one, two or three residues independently selected from the group consisting of F, Cl, Br—, NC—, Me, Et, i-Pr, t-Bu, cyclopropyl, Me(O)S—, Me(O) 2 S—, Et(O) 2 S—, i-Pr(O) 2 S—, t-Bu(O) 2 S— and cyclopropyl(O) 2 S—.
  • R 1 is R 1.a and R 1.a is phenyl or pyridinyl; each ring optionally substituted by one, two or three residues independently selected from the group consisting of F, Cl, Br—, NC—, Me, Me(O)S—, Me(O) 2 S— and Et(O) 2 S—.
  • R 1 is R 1.e and R 1.e is phenyl or pyridinyl; each ring optionally substituted by one or two residues independently selected from the group consisting of NC—, Me(O)S—, Me(O) 2 S and Et(O) 2 S.
  • R 2 is R 2.a and R 2.a is phenyl or a six-membered heteroaryl; wherein one or two elements are replaced by an element independently selected from the group consisting of N, O and S; each ring optionally substituted with a substituent independently selected from the group consisting of halogen, C 1-4 -alkyl-, C 1-4 -haloalkyl- and C 1-4 -alkyl-O—.
  • R 2 is R 2.b and R 2.b is phenyl or a six-membered heteroaryl; wherein one or two elements are replaced by N; each ring optionally substituted with a substituent independently selected from the group consisting of halogen, C 1-4 -alkyl- and C 1-4 -haloalkyl-.
  • R 2 is R 2.c and R 2.c is phenyl or pyridinyl; each optionally substituted with a substituent independently selected from the group consisting of halogen, C 1-4 -alkyl- and C 1-4 -haloalkyl-.
  • R 2 is R 2.d and R 2.d is phenyl or pyridinyl; each optionally substituted with a substituent independently selected from among F 3 C—, F 2 HC— and FH 2 C—.
  • R 2 is R 2.d and R 2.d is phenyl or pyridinyl; each optionally substituted with a substituent independently selected from among F 3 C— and F 2 HC—.
  • R 2 is R 2.e and R 2.e is phenyl, optionally substituted with a substituent independently selected from the group consisting of F 3 C— and F 2 HC—.
  • R 2 is R 2.f and R 2.f is pyridinyl, optionally substituted with a substituent independently selected from the group consisting of F 3 C— and F 2 HC—.
  • R 2 is one of the above mentioned rings carrying the above mentioned substituent in meta-position to the connection of R 2 with the compound of formula 1.
  • R 3 is R 3.a and R 3.a is selected from the group consisting of
  • R 3 is R 3.b and R 3.b is selected from the group consisting of
  • R 3 is independently selected from among HO(O)C—H 2 C—, MeO(O)C—H 2 C—, H 2 N(O)C—H 2 C—, MeHN(O)C—H 2 C—, Me 2 N(O)C—H 2 C—, morpholinyl-(O)C—H 2 C—, azetidinyl-(O)C—H 2 C—, pyrrolidinyl-(O)C—H 2 C—, MeHN(O)C—, EtHN(O)C—, HO(CH 2 ) 2 HN(O)C—, HO(CMe 2 )(CH 2 )HN(O)C—, HO(CH 2 ) 3 HN(O)C—, Me(O)S(CH 2 ) 2 HN(O)C—, Me(O) 2 S(CH 2 ) 2 HN(O)C—, Et(O) 2 S— and Me(O)C— and Me(O)C—
  • R 3 is independently selected from among HO(O)C—H 2 C—, MeO(O)C—H 2 C—, H 2 N(O)C—H 2 C—, MeHN(O)C—H 2 C—, Me 2 N(O)C—H 2 C—, morpholinyl-(O)C—H 2 C—, azetidinyl-(O)C—H 2 C— and pyrrolidinyl-(O)C—H 2 C—.
  • R 3 is independently selected from among MeHN(O)C—, EtHN(O)C—, HO(CH 2 ) 2 HN(O)C—, HO(CMe 2 )(CH 2 )HN(O)C—, HO(CH 2 ) 3 HN(O)C—, Me(O)S(CH 2 ) 2 HN(O)C— and Me(O) 2 S(CH 2 ) 2 HN(O)C—.
  • R 3 is selected from among the examples (E#) 1 to 59 of Table 1 R 3 — Embodiments of the invention for R 3 , R 3.2 , R 3.3 , R 3.4 , R 3.5 , R 3.6 , R 3.7 , R 3.8 (if present):
  • R 3 R 3.2 R 3.3 R 3.4 R 3.5 R 3.6 R 3.7 R 3.8 1.
  • R 3.1 is R 3.1.a and R 3.1.a is H, R 3.3 , R 3.4 , C 1-6 -alkyl-C 3-6 -cycloalkyl-, C 3-6 -cycloalkyl-C 1-6 -alkyl-, each optionally substituted with one or two substituents independently selected from R 3.1.1 —; and R 3.1.1 is selected from among HO—, halogen, NC—, R 3.3 O—, R 3.5 , R 3.6 and R 3.7 .
  • R 3.1 is R 3.1.b and R 3.1.b is selected from among H, R 3.3 , R 3.4 , C 1-6 -alkyl-C 3-6 -cycloalkyl- and C 3-6 -cycloalkyl-C 1-6 -alkyl-.
  • R 3.1 is R 3.1.c and R 3.1.c is selected from among H, R 3.4 and C 1-6 -alkyl-, optionally substituted with one or two substituents independently selected from R 3.1.1 —; and R 3.1.1 is a ring independently selected from among phenyl and a four-membered heterocyclic ring containing one element independently selected from among N, O, S, S(O) and S(O) 2 ; or
  • R 3.1 is R 3.1.d and R 3.1.d is independently selected from among H, R 3.4 and C 1-6 -alkyl-, optionally substituted with one or two substituents independently selected from among R 3.1.1 —; and
  • R 3.2 is R 3.2.a and R 3.2.a is R 3.1a .
  • R 3.2 is R 3.2.b and R 3.2.b is R 3.1b .
  • R 3.2 is R 3.2.c and R 3.2.c is phenyl.
  • R 3.2 is R 3.2.d and R 3.2.d is a five- or six-membered heterocyclic or heteroaryl ring containing one, two or three elements independently selected from among N, O, S, S(O) and S(O) 2 ; each ring optionally substituted with one or two substituents independently selected from among HO—, O ⁇ , NC—, halogen, R 3.3 , R 3.3 O—, R 3.3 —(O)C—, R 3.4 , R 3.5 , R 3.6 and R 3.7 or two substituents are together R 3.8 .
  • R 3.2 is R 3.2.e and two R 3.2.e are together a three-, four-, five- or six-membered monocyclic or a six-, seven-, eight-, nine- or ten-membered bicyclic heterocyclic or heterocyclic ring optionally containing additional to the nitrogen one or two elements independently selected from among N, O, S, S(O) and S(O) 2 ; optionally substituted with one or two substituents, independently selected from among HO—, F, O ⁇ , NC—, R 3.3 , R 3.3 O—, R 3.3 —(O)C—, R 3.4 , R 3.5 , R 3.7 and R 3.6 or two substituents are together R 3.8 .
  • R 3.2 is R 3.2.f and two R 3.2.f are together a three-, four-, five- or six-membered heterocyclic or heteroaryl ring optionally containing additional to the nitrogen one or two elements independently selected from among N, O, S, S(O) and S(O) 2 ; optionally substituted with one or two substituents, independently selected from the group consisting of HO—, F, O ⁇ , NC—, R 3.3 , R 3.3 O—, R 3.3 —(O)C—, R 3.4 , R 3.5 , R 3.7 , R 3.6 or two substituents are together R 3.8 .
  • R 3.2 is R 3.2.g and two R 3.2.g are together a six-, seven-, eight-, nine- or ten-membered bicyclic heterocyclic or heteroaryl ring optionally containing additional to the nitrogen one or two elements independently selected from the group consisting of N, O, S, S(O) and S(O) 2 ; optionally substituted with one or two substituents, independently selected from the group consisting of HO—, F, O ⁇ , NC—, R 3.3 , R 3.3 O—, R 3.3 —(O)C—, R 3.4 , R 3.5 , R 3.7 and R 3.6 or two substituents are together R 3.8 .
  • R 3.2 is R 3.2.h and R 3.2.h is selected from the group consisting of H, Me, Et, n-Pr, i-Pr and cyclopropyl.
  • R 3.3 is R 3.3.a and R 3.3.a is selected from the group consisting of Me, Et, n-Pr, i-Pr, n-Bu, t-Bu, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, F 3 C—, F 2 HC—, F 3 C—CH 2 —, F 2 HC—CH 2 — and FH 2 C—CH 2 —.
  • R 3.4 is R 3.4.a and R 3.4.a is selected from the group consisting of HO—CH 2 —, HO—CH 2 —CH 2 —, HO—CH 2 —CH 2 —CH 2 —, R 3.3.a O—CH 2 —, R 3.3.a O—CH 2 —CH 2 — and R 3.3.a O—CH 2 —CH 2 —CH 2 —.
  • R 3.4 is R 3.4.b and R 3.4.b is selected from the group consisting of HO—CH 2 —, HO—CH 2 —CH 2 —, HO—CH 2 —CH 2 —CH 2 —, MeO—CH 2 —, MeO—CH 2 —CH 2 —, MeO—CH 2 —CH 2 —CH 2 —, EtO—CH 2 -EtO—CH 2 —CH 2 — and EtO—CH 2 —CH 2 —CH 2 —.
  • R 3.5 is R 3.5.a and R 3.5.a is selected from the group consisting of H 2 N—, R 3.3.a HN—, (R 3.3.a ) 2 N—, R 3.3.a (O)C—HN— and R 3.3.a —(O)C—(R 3.3.a )N—.
  • R 3.5 is R 3.5.b and R 3.5.b is selected from the group consisting of H 2 N—, MeHN—, (Me) 2 N—, EtHN—, (Et) 2 N—, i-PrHN—, (i-Pr)(Me)N—, t-BuHN—, (t-Bu)(Me)N—, Me(O)C—HN—, Et(O)C—HN—, n-Pr(O)C—HN—, i-Pr(O)C—HN— and t-Bu(O)C—HN—.
  • R 3.6 is R 3.6.a and R 3.6.a is selected from the group consisting of R 3.3.a (O)S—, R 3.3.a (O) 2 S—, R 3.3.a (HN)S—, R 3.3.a (HN)(O)S—, R 3.3.a (R 3.3.a N)S—, R 3.3.a (R 3.3.a N)(O)S—, R 3.3.a (R 3.4.a N)S—, R 3.3.a (R 3.4.a N)(O)S—, R 3.3.a (NC—N)S— and R 3.3.a (NC—N)(O)S—.
  • R 3.6 is R 3.6.b and R 3.6.b is selected from the group consisting of Me(O)S—, Et(O)S—, i-Pr(O)S—, Me(O) 2 S—, Et(O) 2 S—, i-Pr(O) 2 S—, Me(HN)S—, Et(HN)S—, i-Pr(HN)S—, Me(HN)(O)S—, Et(HN)(O)S—, i-Pr(HN)(O)S—, Me(MeN)S—, Et(MeN)S—, i-Pr(MeN)S—, Me(MeN)(O)S—, Et(MeN)(O)S—, i-Pr(MeN)(O)S—, Me(MeN)(O)S—, Et(MeN)(O)S—, i-Pr(MeN)(O)S—, Me(
  • R 3.7 is R 3.7.a and R 3.7.a is selected from the group consisting of HO(O)C—, H 2 N(O)C—, R 3.3.a O(O)C—, R 3.3.a NH(O)C— and (R 3.3.a ) 2 N(O)C—.
  • R 3.7 is R 3.7.b and R 3.7.b is selected from the group consisting of HO(O)C—, H 2 N(O)C—, MeO(O)C—, EtO(O)C—, i-PrO(O)C—, t-BuO(O)C—, MeNH(O)C—, EtNH(O)C—, i-PrNH(O)C—, t-BuNH(O)C—, (Me) 2 N(O)C—, (Et) 2 N(O)C—, (i-Pr)(Me)N(O)C—, (t-Bu)(Me)N(O)C—, Et(Me)N(O)C—, i-Pr(Me)N(O)C— and t-Bu(Me)N(O)C—.
  • R 3.8 is R 3.8.a and R 3.8.a is independently selected from the group consisting of —CH 2 —, —CH 2 CH 2 —, —CH 2 CH 2 CH 2 —, —CH 2 CH 2 CH 2 CH 2 — and —CH 2 CH 2 CH 2 CH 2 CH 2 —, wherein optionally one or two CH 2 -groups are independently replaced by a group selected from among —HN—, -MeN—, -EtN—, -(Me(O)C—)N—, -(Et(O)C—)N—, -(MeO(O)C—)N—, -(EtO(O)C—)N—, —O—, —S—, —S(O)— and —S(O) 2 —.
  • R 3.8 is R 3.8.b and R 3.8.b is selected from the group consisting of —CH 2 —, —CH 2 CH 2 —, —CH 2 CH 2 CH 2 —, —CH 2 CH 2 CH 2 CH 2 — and —CH 2 CH 2 CH 2 CH 2 CH 2 —, wherein optionally one or two CH 2 -groups are independently replaced by a group selected from among —HN—, -MeN—, -EtN—, —O—, —S—, —S(O)— and —S(O) 2 —.
  • A is A a and A a is —CH 2 —, optionally substituted with one or two substituents independently selected from the group consisting of halogen, R 3.3 , R 3.3 O— and R 3.4 or two substituents together are —CH 2 CH 2 —.
  • A is A b and A b is —CH 2 —, optionally substituted with one or two substituents independently selected from the group consisting of F, Me, Et, i-Pr, MeO, EtO, HOCH 2 O— and MeOCH 2 —.
  • R 4 is R 4.a and R 4.a is selected from the group consisting of halogen, C 1-6 -alkyl-, C 3-6 -cycloalkyl-, C 1-6 -haloalkyl- and C 3-6 -halocycloalkyl.
  • R 4 is R 4.c and R 4.c is C 1-6 -alkyl-.
  • Particularly preferred are the above compounds of formula 1, wherein R 4 is R 4.c and R 4.c is Me.
  • R 3 is a residue independently selected from the group consisting of
  • 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.
  • Step A intermediate I ⁇ intermediate II
  • a carbamate for example methyl carbamate, ethyl carbamate (urethane) or benzyl carbamate in the presence of a strong Br ⁇ nsted or a Lewis acid, for example sulfuric acid, hydrogen chloride, p-toluenesulfonic acid, Amberlyst 15, tetrafluoroboric acid, trifluoroacetic acid or boron trifluoride, either without solvent as a melt or in a suitable solvent, such as benzene, toluene, acetonitrile, diethyl ether, chloroform, acetic anhydride or mixtures thereof.
  • a Lewis acid for example sulfuric acid, hydrogen chloride, p-toluenesulfonic acid, Amberlyst 15, tetrafluoroboric acid, trifluoroacetic acid or boron trifluoride, either without solvent as a melt or in a suitable solvent, such as benzene
  • reaction takes place within 1 to 24 hours.
  • Preferred reaction temperatures are between room temperature and 160° C., or the boiling point of the solvent, respectively.
  • the reaction is done with molten ethyl carbamate as reactant and a catalytic amount of concentrated sulfuric acid at temperatures of 140-160° C. without any additional solvent.
  • Step B intermediate II ⁇ intermediate III
  • a chlorinating agent for example phosphorous pentachloride, phosphoryl chloride or sulfuryl chloride in an organic solvent, for example benzene or toluene.
  • the reaction takes place within 1 to 24 hours.
  • Preferred reaction temperatures are between 50° C. and 150° C.
  • intermediates III can be prepared as described in Jochims et al. ( Chem. Ber. 1982, 115, 860-870) by ⁇ -halogenation of aliphatic isocyanates, for example benzyl isocyanate, using for example a bromination agent, for example N-bromosuccinimide Isocyanates can be synthesized as described in U.S. Pat. No. 6,207,665 and in Charalambides et al. ( Synth. Commun. 2007, 37, 1037-1044), by reacting an amine precursor with phosgene.
  • Step C intermediate IV intermediates V
  • a catalyst for example Ytterbium triflate [Yb(OTf) 3 ] or an acid, for example hydrogen chloride or p-toluenesulfonic acid, optionally in a solvent, for example water, acetic acid, acetonitrile, benzene, toluene.
  • a solvent for example water, acetic acid, acetonitrile, benzene, toluene.
  • Preferred reaction temperatures are between room temperature and 120° C., most preferred room temperature.
  • intermediates V can be prepared as described in Scott et al. ( J. Med. Chem. 1993, 36, 1947-1955) by direct condensation of the 1,3-dicarbonyl compound with an amine under reflux in a suitable solvent, for example benzene or toluene with azeotropic removal of water.
  • intermediates V can be prepared as described in Mariano et al. ( J. Org. Chem. 1984, 49, 220-228) by reacting an amine with 3-chloro-2-cyclopenten-1-one, which can be prepared from cyclopentane-1,3-dione.
  • Step D intermediates III ⁇ compounds of the invention VI
  • Step D intermediates III ⁇ compounds of the invention VI
  • Step D intermediates III ⁇ compounds of the invention VI
  • Step D intermediates III ⁇ compounds of the invention VI
  • Step D intermediates III ⁇ compounds of the invention VI
  • an organic solvent for example dichloromethane, chloroform, benzene or toluene.
  • the reaction takes place within 1-24 hours.
  • Preferred reaction temperatures are between 0° C. and 100° C.
  • Step F compounds of the invention VI ⁇ compounds of the invention VIII
  • Step E compounds of the invention VI ⁇ compounds of the invention VII
  • an appropriate alkyl haloacetate as alkylating agent, for example methyl bromoacetate.
  • Step G compounds of the invention VIII ⁇ compounds of the invention IX
  • a suitable base for example sodium hydroxide, potassium hydroxide, caesium hydroxide, lithium hydroxide, sodium carbonate, potassium carbonate, sodium methoxide or sodium ethoxide in a suitable solvent, for example water, methanol, ethanol, propanol, N,N-dimethylformamide, tetrahydrofuran, 1,4-dioxane, acetonitrile or mixtures thereof.
  • a suitable solvent for example water, methanol, ethanol, propanol, N,N-dimethylformamide, tetrahydrofuran, 1,4-dioxane, acetonitrile or mixtures thereof.
  • Preferred reaction temperatures are between 0° C. and 100° C.
  • the amide coupling (Step H, compounds of the invention IX ⁇ compounds of the invention X) can be achieved by reacting the carboxylic acid intermediate IX with amines R III NH 2 or R III R IV NH in the m presence of an amide coupling reagent, for example N,N,N′,N′-tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate (TBTU) or N,N,N′,N′-tetramethyl-O-(benzotriazol-1-yl)uronium hexafluorophosphate (HBTU), in the presence of a base, for example triethylamine, N,N-diisopropylethylamine or N-methylmorpholine in an organic solvent, for example N-methyl-2-pyrrolidone N,N-dimethylformamide, N,N-dimethylacetamide or mixtures thereof.
  • the reaction takes place within 1-72 hours. P
  • Step K compounds of the invention VI ⁇ intermediates XII
  • a base for example triethylamine, N,N-diisopropylethylamine or N-methylmorpholine
  • a catalyst for example 4-dimethylaminopyridine
  • organic solvent for example dichloromethane, tetrahydrofuran, acetonitrile or N,N-dimethylformamide.
  • Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • Step L intermediates XII ⁇ compounds of the invention XIII
  • an organic solvent for example dichloromethane, acetonitrile, tetrahydrofuran, 1,4-dioxane, toluene or N,N-dimethylformamide.
  • the reaction takes place within 1-72 hours.
  • Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • Compounds of the invention XIV (Step M, compounds of the invention VI ⁇ compounds of the invention XIV) can be prepared as described in WO07046513 or JP2000273087, by reacting compounds of the invention VI with a suitable chloroformate ClCO 2 R VI , for example methyl chloroformate or benzyl chloroformate, in the presence of a suitable base, for example potassium carbonate, sodium hydride, sodium hydroxide, cesium carbonate, lithium diisopropylamide, potassium hexamethyldisilazide, lithium hexamethyldisilazide, an organolithium reagent, for example tert-butyllithium or a Grignard reagent, for example isopropylmagnesiumchloride, in an organic solvent, for example tetrahydrofuran, N,N-dimethylformamide, acetonitrile, 1,4-dioxane, dichloromethane or toluene
  • compounds of the invention XIV can be prepared as described in WO03101917 or WO11085211, by reacting intermediates XII with a suitable alcohol, for example methanol, iso-propanol, 2-methoxyethanol or benzyl alcohol, in the presence of a suitable base, for example potassium carbonate, potassium tert-butoxide or sodium hexamethyldisilazide in an organic solvent, for example tetrahydrofuran, N,N-dimethylformamide, acetonitrile, dichloromethane or dimethylsulfoxide.
  • a suitable alcohol for example methanol, iso-propanol, 2-methoxyethanol or benzyl alcohol
  • a suitable base for example potassium carbonate, potassium tert-butoxide or sodium hexamethyldisilazide
  • organic solvent for example tetrahydrofuran, N,N-dimethylformamide, acetonitrile, dichloromethane or dimethyls
  • R E.1 , R E.2 have the meanings as defined hereinbefore and hereinafter.
  • Step O intermediate I ⁇ intermediate XV
  • Step O intermediate I ⁇ intermediate XV
  • a suitable lewis acid for example trimethylsilyl chloride
  • acetonitrile or toluene can be used as solvent.
  • the reaction takes place within 1-6 days. Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • Intermediates XVI (Step P, intermediate XV ⁇ intermediate XVI) can be prepared in analogy to the method described for the preparation of compounds of the invention VI (Scheme 1, Step D, intermediate III ⁇ compound of the invention VI), by reacting intermediates XV with intermediates V in the presence of a suitable base, for example sodium hydride or sodium tert-butoxide, in a suitable organic solvent, for example tetrahydrofuran or 2-methyltetrahydrofuran. The reaction takes place within 1-24 h. Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • Intermediates XVII (Step Q, intermediate XVI ⁇ intermediate XVII) can be prepared by reacting intermediates XVI with a suitable acid, for example hydrogen chloride, in a suitable solvent, for example 1,4-dioxane. The reaction takes place between 1-72 hours. Preferred reaction temperatures are between 0° C. and room temperature, most preferred room temperature.
  • Step R intermediate XVII ⁇ compound of the invention VI
  • a suitable reagent for example phosgene, triphosgene or carbonyl diimidazole
  • a suitable base for example triethylamine, N,N-diisopropylethylamine, pyridine or sodium carbonate
  • a suitable solvent for example acetonitrile, dichloromethane or toluene.
  • Preferred reaction temperatures are between 0° C. and 50° C., most preferred room temperature.
  • room temperature denotes a temperature of about 20° C.
  • 1 H NMR spectra and/or mass spectra have been obtained of the compounds prepared. Compounds given with a specific configuration at a stereocenter are isolated as pure isomers.
  • example 1A has been assigned unambigously by X-ray structure analysis to be (R).
  • This (R)-enantiomer (example 1A) is significantly more potent with respect to the inhibition of neutrophil elastase than the (S)-enantiomer (example 1B), as can be seen from the measured IC 50 values of 11.5 nM (example 1A) and 8040 nM (example 1B), respectively.
  • the absolute configuration of all other pure enantiomers described has been assigned in analogy to example 1A, that is, the more potent enantiomer (the eutomer) with respect to the inhibition of neutrophil elastase, i.e. the enantiomer with the lower IC 50 value has been assigned to have the same absolute configuration as example 1A.
  • 4-Nitrophenyl chloroformate (1.11 g, 5.52 mmol) is added to a solution of 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)benzonitrite (example 1, 1.33 g, 3.35 mmol), N,N-diisopropylethylamine (2.28 mL, 13.4 mmol) and 4-(dimethylamino)pyridine (409 mg, 3.35 mmol) in dichloromethane (24 mL). After 1 h the mixture is washed with water and concentrated.
  • Phosphorous pentachloride (5.47 g, 26.2 mmol) is added to a suspension of diethyl (4-bromo-2-methylsulfonyl)phenyl)methylenedicarbamate (intermediate 7, 5.05 g, 11.9 mmol) in toluene (30 mL) and the mixture is heated at reflux for 3 h. The toluene is evaporated and the mixture is then purified by distillation under reduced pressure (ca. 160° C., 0.1 mbar). Yield: 945 mg.
  • Formic acid (3.9 mL, 104 mmol) is added to a solution of tert-butyl carbamate (1.90 g, 16.2 mmol), 2-bromo-4-cyanobenzaldehyde (3.41 g, 16.2 mmol) and sodium benzenesulfinate (2.67 g, 16.2 mmol) in a mixture of tetrahydrofuran (7.0 mL) and water (60 mL), and the mixture is stirred at room temperature for 6 days. Water (180 mL) is added, and the precipitate is filtered and washed with water. The precipitate is treated with tert-butyl methyl ether (30 mL), and the mixture is stirred for 30 min.
  • Sodium hydride (60% in mineral oil, 360 mg, 9.00 mmol) is added in portions to a mixture of 3-(3-(trifluoromethyl)phenylamino)cyclopent-2-enone (2.16 g, 8.96 mmol) and 2-methyltetrahydrofuran (30 mL). After 30 min tert-butyl (2-bromo-4-cyanophenyl)(phenylsulfonyl)methylcarbamate (Step 1, 3.35 g, 7.43 mmol) is added and the mixture is stirred at room temperature for 2 h. Water is added and the phases are separated.
  • Sodium hydride (60% in mineral oil, 6 mg, 150 ⁇ mol) is added to a mixture of 5-methyl-3-(3-(trifluoromethyl)phenylamino)cyclopent-2-enone (intermediate 23, 38 mg, 150 ⁇ mol) and 2-methyltetrahydrofuran (2 mL). After 20 min tert-butyl (4-cyanophenyl)(phenylsulfonyl)methylcarbamate (60 mg, 150 ⁇ mol based on 90% purity) is added, and the mixture is stirred at room temperature over night.
  • Triethylamine 250 ⁇ L, 1.81 mmol is added to a mixture of 2-(amino(4-bromo-2-(methylthio)phenyl)methyl)-3-(3-(trifluoromethyl)phenyl amino)cyclopent-2-enone hydrochloride (intermediate 20.5, 4.08 g, 7.23 mmol based on 90% purity) and 1,1′-carbonyldiimidazole (1.46 g, 9.04 mmol) in acetonitrile (54 mL), and the mixture is stirred at room temperature for 1 h. All volatiles are removed under reduced pressure, and the residue is treated with water.
  • example 1A can be prepared as follows:
  • Aqueous sodium hydroxide solution (1 M, 15.0 mL, 15.0 mmol) is added to a solution of methyl 2-(4-(4-cyanophenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-6,7-dihydro-1H-cyclopenta[d]pyrimidin-3(2H,4H,5H)-yl)acetate (example 3, 2.64 g, 5.62 mmol) in tetrahydrofuran (40 mL) and the mixture is stirred at room temperature for 4 h. Water is added and the mixture is extracted three times with ethyl acetate.
  • N,N,N′,N′-Tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate 43 mg, 0.13 mmol
  • 2-(4-(4-cyanophenyl)-2,5-dioxo-1-(3-(trifluoro-methyl)phenyl)-6,7-dihydro-1H-cyclopenta[d]pyrimidin-3(2H,4H,5H)-yl)acetic acid (example 4, 60 mg, 0.13 mmol) and N,N-diisopropylethylamine (50 ⁇ L, 0.29 mmol) in N,N-dimethylformamide (0.5 mL).
  • Triethylamine (0.43 mL, 3.0 mmol) is added to a mixture of 4-(amino(5-oxo-2-(3-(trifluoromethyl)phenylamino)cyclopent-1-enyl)methyl)-3-bromobenzonitrile hydrochloride (intermediate 20, 5.90 g, 12.1 mmol) and 1,1′-carbonyldiimidazole (2.46 g, 15.2 mmol) in acetonitrile (60 mL), and the mixture is stirred at room temperature over night. Water (700 mL) is added and the precipitate is filtered, washed with water and dried. Yield: 5.45 g.
  • Triethylamine (0.38 mL, 2.70 mmol) is added to a mixture of 4-(amino(5-oxo-2-(3-(trifluoromethyl)phenylamino)cyclopent-1-enyl)methyl)-3-chlorobenzonitrile hydrochloride (intermediate 20.1, 660 mg, 1.34 mmol based on 90% purity) and 1,1′-carbonyldiimidazole (270 mg, 1.68 mmol) in acetonitrile (5 mL), and the mixture is stirred at room temperature over night. Water and dichloromethane are added, and the phases are separated.
  • Table 7 The following examples of Table 7 are prepared in analogy to 3-chloro-4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)benzonitrile (example 15), using the appropriate starting material and the purification method as indicated in the table (Method A: Waters XbridgeTM-C 18 , gradient of acetonitrile in water, 0.1% TFA; Method B: Waters SunFireTM-C 18 , gradient of acetonitrile in water, 0.1% TFA; Method C: Waters SunFireTM-C 18 , gradient of acetonitrile in water, 0.1% formic acid).
  • meta-Chloroperoxybenzoic acid (77%, 390 mg, 1.74 mmol) is added at room temperature to a solution of 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylthio)benzonitrile (example 16, 776 mg, 1.75 mmol) in dichloromethane, and the mixture is stirred for 30 min Saturated aqueous NaHCO 3 solution is added, and the mixture is extracted with dichloromethane.
  • Phosphorous pentachloride (9.63 g, 46.2 mmol) is added to a mixture of diethyl (4-cyano-2-fluorophenyl)methylenedicarbamate (intermediate 26, 6.50 g, 21.0 mmol) in toluene (25.0 mL), and the mixture is heated at reflux for 3 h. The toluene is evaporated, and the mixture is then purified by distillation under reduced pressure. The first fraction (ca. 35° C., ca. 0.2 mbar) is discarded. The second fraction (ca. 112° C., ca. 0.1 mbar) is collected. Yield: 1.90 g.
  • Aqueous sodium hydroxide solution (1.0 M, 10.0 mL, 10.0 mmol) is added to a solution of ethyl 2-(4-(4-cyano-2-(methylsulfonyl)phenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-6,7-dihydro-1H-cyclopenta[d]pyrimidin-3(2H,4H,5H)-yl)acetate (intermediate 29, 1.80 g, 3.20 mmol) in tetrahydrofuran (40 mL), and the mixture is stirred at room temperature over night.
  • Table 8 The following examples of Table 8 are prepared in analogy to 2-(4-(4-cyano-2-(methylsulfonyl)phenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-6,7-dihydro-1H-cyclopenta[d]-pyrimidin-3(2H,4H,5H)-yl)-N-(2-hydroxyethyl)-N-methylacetamid (example 21), using the appropriate amine as reagent.
  • Bromoethane (20 ⁇ L, 0.27 mmol) is added to a solution of 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 10, 60 mg, 0.11 mmol based on 90% purity) and cesium carbonate (74 mg, 0.23 mmol) in N,N-dimethylformamide (2.0 mL).
  • Table 9 The following examples of Table 9 are prepared in analogy to 4-(3-ethyl-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 24), substituting bromoethane with the appropriate alkylating reagent and using the purification method indicated in the table (Method A: Waters SunFireTM-C 18 , gradient of acetonitrile in water, 0.1% TFA; Method B: Waters XbridgeTM-C 18 , gradient of acetonitrile in water, 0.1% NH 3 ; Method C: Waters XbridgeTM-Phenyl, gradient of methanol in water, 0.1% TFA).
  • Table 10 The following examples of Table 10 are prepared in analog to 4-(1-(3-(difluoromethyl)-phenyl)-3-methyl-2,5-dioxo-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-benzonitrile (example 26), using the appropriate starting material as indicated in the table and substituting tetrahydrofuran with acetonitrile as solvent.
  • Methyl iodide (15 ⁇ L, 0.24 mmol) is added to a solution of 4-(1-(3-(difluoromethyl)-phenyl)-2,5-dioxo-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 15.4, 69 mg, 0.15 mmol) and cesium carbonate (98 mg, 0.30 mmol) in N,N-dimethylformamide (1.0 mL).
  • Table 11 The following examples of Table 11 are prepared in analog to 4-(1-(3-(difluoromethyl)-phenyl)-3-methyl-2,5-dioxo-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 27), using the appropriate starting material as indicated in the table.
  • Methyl iodide (2 M in tert-butyl methyl ether, 63 ⁇ L, 0.13 mmol) is added to a solution of 4-(2,5-dioxo-1-(2-(trifluoromethyl)pyridin-4-yl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]-pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 15.6, 50 mg, 0.11 mmol) and cesium carbonate (68 mg, 0.21 mmol) in N,N-dimethylformamide (2.0 mL), and the mixture is stirred at room temperature over night.
  • Table 14 The following examples of Table 14 are prepared in analogy to 3-(methylsulfonyl)-4-(3-(methylsulfonyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)benzonitrile (example 37), using the appropriate starting material as indicated in the table.
  • Table 15 is prepared in analogy to 3-(methylsulfonyl)-4-(3-(methylsulfonyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)benzonitrile (example 37), using the appropriate starting material as indicated in the table and replacing tetrahydrofuran with acetonitrile as solvent.
  • Table 16 The following examples of Table 16 are prepared in analog to 4-(4-cyanophenyl)-N-(2-hydroxy-2-methylpropyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 41), using the appropriate amine as reagent.
  • N,N-Diisopropylethylamine (170 ⁇ L, 1.00 mmol), 4-dimethylaminopyridine (34 mg, 0.28 mmol) and 4-nitrophenyl chloroformate (56 mg, 0.28 mmol) is added to a solution of 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]-pyrimidin-4-yl)benzonitrile (example 1, 100 mg, 0.25 mmol) in acetonitrile (2.0 mL), and the mixture is stirred at room temperature over night.
  • 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]-pyrimidin-4-yl)benzonitrile (example 1, 100 mg, 0.25 mmol) in
  • Table 17 The following examples of Table 17 are prepared in analogy to 4-(4-cyanophenyl)-2,5-dioxo-N-(1,1-dioxo-1 ⁇ 6 -thietan-3-yl)-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 42), using the appropriate amine as reagent.
  • Methyltrioxorhenium(VII) (1 mg, 4 ⁇ mol) is added, and the mixture is stirred at ⁇ 78° C. for 30 min Another portion of methyltrioxorhenium(VII) (1 mg, 4 ⁇ mol) is added, and the mixture is stirred at ⁇ 78° C. for 1 h.
  • Aqueous potassium hydrogen sulfate solution (10%, 0.5 mL) and water is (10 mL) is added, and the mixture is filtered.
  • Table 18 The following examples of Table 18 are prepared in analogy to 4-(4-cyanophenyl)-N-(2-hydroxy-2-methylpropyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 41), using (R)-4-nitrophenyl 4-(4-cyanophenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxylate (intermediate 30.1) as starting material and the appropriate amine as reagent.
  • N,N-Diisopropylethylamine (137 ⁇ L, 0.81 mmol), 4-dimethylaminopyridine (27 mg, 0.22 mmol) and 4-nitrophenyl chloroformate (45 mg, 0.22 mmol) is added to a solution of (R)-4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]-pyrimidin-4-yl)benzonitrile (example 1A, 80 mg, 0.20 mmol) in acetonitrile (2.0 mL), and the mixture is stirred at room temperature over night.
  • Table 19 are prepared in analog to 4-(4-cyanophenyl)-N-(2-hydroxy-2-methylpropyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 41.0), using (S)-4-nitrophenyl 4-(4-cyano-2-(methylsulfonyl)phenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxylate (intermediate 20.2) as starting material and the appropriate amine as reagent.
  • Table 20 The following examples of Table 20 are prepared in analog to 4-(4-cyanophenyl)-2,5-dioxo-N-(1,1-dioxo-1 ⁇ 6 -thietan-3-yl)-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 42), using (S)-4-nitrophenyl 4-(4-cyano-2-(methylsulfonyl)phenyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxylate (intermediate 30.2) as starting material and the appropriate amine as reagent.
  • 4-Nitrophenyl chloroformate (23 mg, 0.11 mmol) is added to a solution of 4-(2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-fluorobenzonitrile (example 18, 43 mg, 0.10 mmol), N,N-diisopropylethylamine (70 ⁇ L, 0.41 mmol) and 4-dimethylaminopyridine (14 mg, 0.11 mmol) in acetonitrile (3.0 mL), and the mixture is stirred at room temperature over night.
  • Table 21 The following examples of Table 21 are prepared in analogy 4-(4-cyano-2-fluorophenyl)-N-methyl-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]-pyrimidine-3(2H)-carboxamide (example 49), substituting methylamine with the appropriate amine as reagent.
  • Table 22 The following examples of Table 22 are prepared in analogy to 4-(4-cyano-2-fluorophenyl)-N-methyl-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 49), using the appropriate starting material as indicated in the table.
  • Table 24 The following examples of Table 24 are prepared in analogy to 4-(4-cyanophenyl)-1-(3-(difluoromethyl)phenyl)-N-(3-hydroxypropyl)-2,5-dioxo-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 52), replacing 3-aminopropanol with the appropriate amine as reagent.
  • Table 25 The following examples of Table 25 are prepared in analogy to 4-(4-cyano-2-fluorophenyl)-N-methyl-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 49), using 4-(1-(3-(difluoromethyl)-phenyl)-2,5-dioxo-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 15.4) as starting material and employing the appropriate amine as reagent.
  • Table 26 The following examples of Table 26 are prepared in analogy to 4-(4-cyanophenyl)-N-(2-hydroxy-2-methylpropyl)-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 41), using 4-nitrophenyl 4-(4-cyanophenyl)-2,5-dioxo-1-(2-(trifluoromethyl)pyridin-4-yl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxylate (intermediate 30.3) as starting material and employing the appropriate amine as reagent.
  • Table 27 The following examples of Table 27 are prepared in analogy to 4-(4-cyano-2-fluorophenyl)-N-methyl-2,5-dioxo-1-(3-(trifluoromethyl)phenyl)-4,5,6,7-tetrahydro-1H-cyclopenta[d]pyrimidine-3(2H)-carboxamide (example 49), using 4-(2,5-dioxo-1-(2-(trifluoro-methyl)pyridin-4-yl)-2,3,4,5,6,7-hexahydro-1H-cyclopenta[d]pyrimidin-4-yl)-3-(methylsulfonyl)benzonitrile (example 15.6) as starting material and employing the appropriate amine as reagent.
  • 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.
  • the following buffers were used: 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 nM 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).
  • IC 50 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%.
  • the IC 50 values of selected compound in the Neutrophil Elastase assay are listed in Table 28.
  • Citrated blood from human healthy donors is mixed with zymosan suspension and incubated at room temperature. This leads to the stimulation of neutrophils and the release of neutrophil elastase into the plasma. The stimulated blood is centrifuged to generate the neutrophil elastase enriched plasma.
  • Zymosan (100 mg) is mixed with saline (0.9%, 10 mL) and stored at 4° C. for up to one week (note: zymosan does not dissolve in the saline and is used as a suspension).
  • the analysis of the data is performed by the calculation of the percentage of fluorescence in the presence of the test compound compared to the fluorescence of the vehicle control after subtracting the background fluorescence:
  • An inhibitor of the neutrophil elastase enzyme will give values between 100% control (no inhibition) and 0% control (complete inhibition).
  • the human plasma shift of selected compounds can be calculated using the following equation:
  • Human plasma shift (EC 50 in human plasma assay)/(IC 50 in human neutrophil elastase assay)
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A disclosed in WO 2005/0828683), the cyclic ketone example 1A exhibits a significantly lower EC 50 value, i.e. significantly improved potency, in the human plasma assay described above. Furthermore, example 1A exhibits a human plasma shift of less than 2 which is significantly lower than the human plasma shift for example 8A in WO 2005/0828683 and is likely attributable to reduced binding to human plasma proteins. This observation is surprising, since example 1A differs from example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the metabolic degradation of the test compound is assayed at 37° C. with pooled human liver microsomes.
  • the final incubation volume of 100 ⁇ l per time point contains TRIS buffer pH 7.6 (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 ⁇ M.
  • the reactions are initiated by addition of beta-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM) and terminated by transferring an aliquot into acetonitrile after different time points. Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point.
  • test compound after NADPH independent incubation is reflected by the parameter c(control) (metabolic stability).
  • the quenched incubations are pelleted by centrifugation (10′000 g, 5 min) An aliquot of the supernatant is assayed by LC-MS/MS for the amount of parent compound.
  • the half-life (t 1/2 INVITRO) is determined by the slope of the semilogarithmic plot of the concentration-time profile.
  • the intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation:
  • CL_INTRINSIC[ ⁇ l/min/mg protein] (ln 2/(half-life [min]*protein content [mg/ml]))*1′000.
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A disclosed in WO 2005/0828683), the cyclic ketone example 1A exhibits improved half life, i.e. improved stability, in the metabolic stability assay described above. This observation is surprising, since example 1A differs from example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the metabolic degradation of the test compound is assayed in a human hepatocyte suspension.
  • Human hepatocytes typically cryopreserved are incubated in an appropriate buffer system (e.g. Dulbecco's modified eagle medium plus 3.5 ⁇ g glucagon/500 mL, 2.5 mg insulin/500 mL and 3.75 mg/500 mL hydrocortison) containing 5% species serum.
  • an appropriate buffer system e.g. Dulbecco's modified eagle medium plus 3.5 ⁇ g glucagon/500 mL, 2.5 mg insulin/500 mL and 3.75 mg/500 mL hydrocortison
  • test compound solution 80 ⁇ M; from 2 mM stock solution in DMSO diluted 1:25 with medium
  • 395 ⁇ l hepatocyte suspension cell density in the range 0.25-5*10 6 cells/mL, typically 1*10 6 cells/mL; final concentration of test compound 1 ⁇ M, final DMSO concentration 0.05%).
  • the cells are incubated for six hours (incubator, orbital shaker) and samples (25 ⁇ l) are taken at 0, 0.5, 1, 2, 4 and 6 hours.
  • the intrinsic clearance CL_INTRINSIC is calculated as follows:
  • the calculated in vitro hepatic intrinsic clearance can be scaled up to the intrinsic in vivo hepatic clearance and used to predict hepatic in vivo blood clearance (CL) by the use of a liver model (well stirred model):
  • CL_INTRINSIC_INVIVO[ml/min/kg] (CL_INTRINSIC[ ⁇ L/min/10 6 cells]*hepatocellularity[10 6 cells/g liver]*liver factor[g/kg bodyweight])/1′000
  • CL[ml/min/kg] CL_INTRINSIC_INVIVO[ml/min/kg]*hepatic blood flow[ml/min/kg]/(CL_INTRINSIC_INVIVO[ml/min/kg]+hepatic blood flow[ml/min/kg])
  • liver factor human: 25.7 g/kg bodyweight
  • blood flow human: 21 ml/(min*kg)
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A disclosed in WO 2005/0828683), the cyclic ketone example 1A exhibits reduced clearance, i.e. improved stability, in the metabolic stability assay described above. This observation is surprising, since example 1A differs from example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the assay provides information on the potential of a compound to pass the cell membrane, on the extent of oral absorption as well as on whether the compound is actively transported by uptake and/or efflux transporters.
  • confluent human cancer colon carcinoma cells 2 (Caco-2) cell monolayers grown on permeable filter supports are used as the in vitro absorption model.
  • Apparent permeability coefficients (PE) of the compounds across the Caco-2 monolayers are measured (pH 7.2, 37° C.) in apical-to-basal (AB) (absorptive) and basal-to-apical (BA) (secretory) transport direction.
  • AB permeability (PEAB) represents drug absorption from the intestine into the blood
  • BA permeability (PEBA) drug secretion from the blood back into the intestine via both passive permeability as well as active transport mechanisms mediated by efflux and uptake transporters that are expressed on the Caco-2 cells.
  • the compounds are assigned to permeability/absorption classes by comparison of the AB permeabilities with the AB permeabilities of reference compounds with known in vitro permeability and oral absorption in the human. Identical or similar permeabilities in both transport directions indicate passive permeation, vectorial permeability points to additional active transport mechanisms.
  • Higher PEBA than PEAB suggests the involvement of an apical efflux transporter (like P-gp) and/or basolateral uptake transporter; higher PEAB than PEBA permeability suggests involvement of an apical uptake transporter (like PepT1) and/or basolateral efflux transporter (like MRP3).
  • Active transport is concentration-dependently saturable.
  • Caco-2 cells (1-2*10 5 cells/cm 2 area) are seeded on filter inserts (Costar transwell polycarbonate or PET filters, 0.4 nm pore size) and cultured (DMEM) for 10 to 25 days. Compounds are dissolved in appropriate solvent (like DMSO, 1-20 mM stock solutions).
  • HTP-4 buffer (128.13 mM NaCl, 5.36 mM KCl, 1 mM MgSO 4 , 1.8 mM CaCl 2 , 4.17 mM NaHCO 3 , 1.19 mM Na 2 HPO 4 x7H 2 O, 0.41 mM NaH 2 PO 4 xH 2 O, 15 mM HEPES, 20 mM glucose, pH 7.2
  • the transport solution (TL) is applied to the apical or basolateral donor side for measuring A-B or B-A permeability (3 filter replicates), respectively.
  • the receiver side contains HTP-4 buffer supplemented with 2% BSA. Samples are collected at the start and end of experiment from the donor and at various time intervals for up to 2 hours also from the receiver side for concentration measurement by LC-MS/MS or scintillation counting. Sampled receiver volumes are replaced with fresh receiver solution.
  • the cyclic ketone example 1A Compared to the cyclic amide derivative (example 4 disclosed in WO 2007/129060), the cyclic ketone example 1A exhibits improved AB permeability and a reduced efflux ratio.
  • the AB permeability and efflux ratio of example 1A are in the favorable range for an orally administered drug.
  • the cyclic ketone example 10A exhibits improved AB permeability.
  • the aqueous solubility of a compound is determined by comparing the amount dissolved in aqueous buffer (containing 2.5% DMSO) to the amount dissolved in an acetonitrile/water (1/1) solution. Starting from a 10 mM DMSO stock solution, aliquots are diluted with acetonitrile/water (1/1) and McIlvaine buffer pH 6.8, respectively. After 24 h of shaking, the solutions or suspensions are filtered and analyzed by LC-UV. The amount dissolved in buffer is compared to the amount dissolved in the acetonitrile/water (1/1) solution. Solubility is measured from 0.001 to 0.125 mg/ml at a DMSO concentration of 2.5%. If more than 90% of the compound is dissolved in buffer, the value is marked with “>”.
  • the inhibition of cytochrome P450 2C9-isoenzyme catalysed hydroxylation of Diclofenac by the test compound is assayed at 37° C. with human liver microsomes. All assays are carried out on a robotic system in 96 well plates. The final incubation volume contains TRIS buffer (0.1 M), MgCl 2 (5 mM), human liver microsomes (0.1 mg/ml), Diclofenac (10 ⁇ M) and the test compound at five different concentrations or no compound (high control) in duplicate (e.g. highest concentration 10-50 ⁇ M with subsequent serial 1:4 dilutions).
  • % control activity (100% control activity/(1+( I/IC 50 )* S )) ⁇ B
  • the IC 50 is assigned “ ⁇ lowest concentration tested” (usually ⁇ 0.4 ⁇ M). If the inhibition of the reaction is still ⁇ 50% at the highest concentration of the test compound, the IC 50 is assigned “>highest concentration tested” (usually >50 ⁇ M).
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A disclosed in WO 2005/0828683), the cyclic ketone example 1A exhibits reduced CYP2C9 inhibition in the assay described above. This observation is surprising, since example 1A differs from Example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the inhibition of cytochrome P450 2C19-isoenzyme catalysed hydroxylation of Mephenytoin by the test compound is assayed at 37° C. with human liver microsomes. All assays are carried out on a robotic system in 96 well plates. The final incubation volume contains TRIS buffer (0.1 M), MgCl 2 (5 mM), human liver microsomes (0.5 mg/ml), (S)-Mephenytoin (70 ⁇ M) and the test compound at five different concentrations or no compound (high control) in duplicate (e.g. highest concentration 10-50 ⁇ M with subsequent serial 1:4 dilutions).
  • % control activity (100% control activity/(1+( I/IC 50 )* S )) ⁇ B
  • the IC 50 is assigned “ ⁇ lowest concentration tested” (usually ⁇ 0.4 ⁇ M). If the inhibition of the reaction is still ⁇ 50% at the highest concentration of the test compound, the IC 50 is assigned “>highest concentration tested” (usually >50 ⁇ M).
  • IC 50 values of selected compounds in the CYP2C19 inhibition assay described above are listed in Table 35.
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A in WO 2005/0828683), the cyclic ketone example 1A exhibits reduced CYP2C19 inhibition in the assay described above. This observation is surprising, since example 1A differs from example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the inhibition of cytochrome P450 2C8-isoenzyme catalysed deethylation of Amodiaquine by the test compound is assayed at 37° C. with human liver microsomes. All assays are carried out on a robotic system in 96 well plates. The final incubation volume contains TRIS buffer (0.1 M), MgCl 2 (5 mM), human liver microsomes (0.05 mg/ml), Amodiaquine (1 ⁇ M) and the test compound at five different concentrations or no compound (high control) in duplicate (e.g. highest concentration 10-50 ⁇ M with subsequent serial 1:4 dilutions).
  • % control activity (100% control activity/(1+( I/IC 50 )* S )) ⁇ B
  • the IC 50 is assigned “ ⁇ lowest concentration tested” (usually ⁇ 0.4 ⁇ M). If the inhibition of the reaction is still ⁇ 50% at the highest concentration of the test compound, the IC 50 is assigned “>highest concentration tested” (usually >50 ⁇ M).
  • IC 50 values of selected compounds in the CYP2C8 inhibition assay described above are listed in Table 36.
  • example 1A Compared to the acyclic methyl ketone derivative (example 8A disclosed in WO 2005/0828683), the cyclic ketone example 1A exhibits reduced CYP2C8 inhibition in the assay described above. This observation is surprising, since example 1A differs from example 8A in WO 2005/0828683 by only a single carbon-carbon bond.
  • the compounds of general formula 1 may be used on their own or combined with other active substances of formula 1 according to the invention.
  • the compounds of general formula 1 may optionally also be combined with other pharmacologically active substances. These include, ⁇ 2-adrenoceptor-agonists (short and long-acting), anticholinergics (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-melanoma
  • 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;
  • 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);
  • 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;
  • 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,
  • 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)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Pain & Pain Management (AREA)
  • Oncology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US14/166,146 2013-02-06 2014-01-28 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity Abandoned US20140221335A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/935,915 US9290459B1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US14/935,931 US20160060231A1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US15/229,160 US9670166B2 (en) 2013-02-06 2016-08-05 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13154256.5 2013-02-06
EP13154256 2013-02-06

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/935,931 Continuation US20160060231A1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US14/935,915 Continuation US9290459B1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Publications (1)

Publication Number Publication Date
US20140221335A1 true US20140221335A1 (en) 2014-08-07

Family

ID=47681764

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/166,146 Abandoned US20140221335A1 (en) 2013-02-06 2014-01-28 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US14/935,931 Abandoned US20160060231A1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US14/935,915 Active US9290459B1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US15/229,160 Active US9670166B2 (en) 2013-02-06 2016-08-05 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/935,931 Abandoned US20160060231A1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US14/935,915 Active US9290459B1 (en) 2013-02-06 2015-11-09 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US15/229,160 Active US9670166B2 (en) 2013-02-06 2016-08-05 Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Country Status (39)

Country Link
US (4) US20140221335A1 (zh)
EP (1) EP2953943B1 (zh)
JP (1) JP6130519B2 (zh)
KR (1) KR102221201B1 (zh)
CN (4) CN106045920B (zh)
AP (1) AP2015008535A0 (zh)
AR (2) AR095164A1 (zh)
AU (1) AU2014214031B2 (zh)
BR (1) BR112015017929B1 (zh)
CA (1) CA2900308C (zh)
CL (1) CL2015002030A1 (zh)
CY (1) CY1119063T1 (zh)
DK (1) DK2953943T3 (zh)
EA (1) EA030569B1 (zh)
EC (1) ECSP15036282A (zh)
ES (1) ES2629023T3 (zh)
GE (1) GEP201706741B (zh)
HK (1) HK1211573A1 (zh)
HR (1) HRP20171040T1 (zh)
HU (1) HUE14705723T2 (zh)
IL (1) IL239472B (zh)
LT (1) LT2953943T (zh)
MA (1) MA38327B1 (zh)
ME (1) ME02697B (zh)
MX (1) MX361166B (zh)
NZ (1) NZ709041A (zh)
PE (1) PE20151429A1 (zh)
PH (1) PH12015501697B1 (zh)
PL (1) PL2953943T3 (zh)
PT (1) PT2953943T (zh)
RS (1) RS56063B1 (zh)
SG (1) SG11201505959QA (zh)
SI (1) SI2953943T1 (zh)
TN (1) TN2015000328A1 (zh)
TW (2) TWI634109B (zh)
UA (1) UA117007C2 (zh)
UY (1) UY35311A (zh)
WO (1) WO2014122160A1 (zh)
ZA (1) ZA201504257B (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9458113B2 (en) 2014-07-31 2016-10-04 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9475779B2 (en) 2014-07-31 2016-10-25 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US10189776B2 (en) 2015-09-18 2019-01-29 Boehringer Ingelheim International Gmbh Stereoselective process

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140221335A1 (en) * 2013-02-06 2014-08-07 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
KR20200020210A (ko) 2018-08-16 2020-02-26 서울대학교산학협력단 식물 추출물을 함유하는 호중구 엘라스타아제 억제용 조성물
GB201820450D0 (en) * 2018-12-14 2019-01-30 Z Factor Ltd Compound and its use for the treatment of alpha1-antitryspin deficiency
CN109331009A (zh) * 2018-12-24 2019-02-15 王萌萌 一种京尼平眼用制剂
JP2023500182A (ja) 2019-09-17 2023-01-05 メレオ バイオファーマ 4 リミテッド 移植片拒絶反応、閉塞性細気管支炎症候群、及び移植片対宿主病の治療に使用するためのアルベレスタット
WO2021209739A1 (en) 2020-04-16 2021-10-21 Mereo Biopharma 4 Limited Methods involving neutrophil elastase inhibitor alvelestat for treating respiratory disease mediated by alpha-1 antitrypsin deficiency
GB202009074D0 (en) * 2020-06-15 2020-07-29 Z Factor Ltd Compound
GB202009069D0 (en) * 2020-06-15 2020-07-29 Z Factor Ltd Process
CA3234399A1 (en) 2021-10-20 2023-04-27 Mereo Biopharma 4 Limited Neutrophil elastase inhibitors for use in the treatment of fibrosis

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6228861B1 (en) 1995-11-16 2001-05-08 Synaptic Pharmaceutical Corporation Dihydropyrimidines and uses thereof
US6207665B1 (en) 1997-06-12 2001-03-27 Schering Aktiengesellschaft Piperazine derivatives and their use as anti-inflammatory agents
JP2001139556A (ja) * 1999-03-18 2001-05-22 Nippon Soda Co Ltd インデノピリジンおよびインデノピリミジン化合物並びにその製造法
JP2000273087A (ja) 1999-03-24 2000-10-03 Mitsui Chemicals Inc 非対称2−イミダゾリジノン類の製造方法
CA2470813A1 (en) 2001-12-20 2003-07-03 Bayer Healthcare Ag 1,4-dihydro-1,4-diphenylpyridine derivatives
AU2003263574A1 (en) 2002-05-31 2003-12-19 Patel, Hetalkumar, Virendrabhai A process for the preparation of phenylcarbamates
WO2004020410A2 (en) 2002-08-27 2004-03-11 Bayer Healthcare Ag Dihydropyridinone derivatives as hne inhibitors
GB0219896D0 (en) 2002-08-27 2002-10-02 Bayer Ag Dihydropyridine derivatives
US7566723B2 (en) 2002-09-10 2009-07-28 Bayer Healthcare Ag 1-phenyl1-3,4-dihydropyrimidin-2(1H)-one derivatives and their use
PL375647A1 (en) 2002-09-10 2005-12-12 Bayer Healthcare Ag Pyrimidinone derivatives as therapeutic agents against acute and chronic inflammatory, ischaemic and remodelling processes
GB2392910A (en) 2002-09-10 2004-03-17 Bayer Ag 2-Oxopyrimidine derivatives and their use as human leukocyte elastase inhibitors
JP4825194B2 (ja) 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−ジアリール−ジヒドロピリミジン−2−オン化合物およびヒト好中球エラスターゼ阻害剤としてのそれらの使用
EP1568696A1 (en) 2004-02-26 2005-08-31 4Sc Ag Compounds as inhibitors of cell proliferation and viral infections
WO2005082863A2 (en) * 2004-02-26 2005-09-09 Bayer Healthcare Ag 1,4 diaryl-dihydropyrimidin-2 ones and their use as a human neutrophil elastase inhibitors
GB0502258D0 (en) 2005-02-03 2005-03-09 Argenta Discovery Ltd Compounds and their use
DE102005028845A1 (de) 2005-06-22 2006-12-28 Sanofi-Aventis Deutschland Gmbh Substituierte Heterocyclen, ihre Verwendung als Medikament sowie enthaltende pharmazeutische Zubereitungen
GB0512940D0 (en) 2005-06-24 2005-08-03 Argenta Discovery Ltd Compounds and their use
EP1937629A2 (en) 2005-10-20 2008-07-02 Sumitomo Chemical Company, Limited Benzoylurea compounds and use thereof
WO2007129060A1 (en) * 2006-05-04 2007-11-15 Argenta Discovery Limited Tetrahydropyrrolopyrimidinediones and their use as human neutrophil elastase inhibitors
GB0608844D0 (en) * 2006-05-04 2006-06-14 Argenta Discovery Ltd Enzyme inhibitors
WO2009013444A1 (en) 2007-07-25 2009-01-29 Argenta Discovery Limited Tetrahydropyrrolopyrimidinediones and their use as human neutrophil elastase inhibitors
US8198288B2 (en) 2006-05-04 2012-06-12 Pulmagen Therapeutics (Inflammation) Limited Tetrahydropyrrolopyrimidinediones and their use in therapy
GB0610765D0 (en) 2006-05-31 2006-07-12 Proskelia Sas Imidazolidine derivatives, uses therefor, preparation thereof and compositions comprising such
US20100010024A1 (en) 2006-07-01 2010-01-14 Bayer Healthcare Ag Use of1,4-diaryl-dihydropyrimidine-2-on derivatives for treating pulmonary arterial hypertension
DE102006031314A1 (de) 2006-07-01 2008-01-03 Bayer Healthcare Aktiengesellschaft Verwendung von 1,4-Diaryl-dihydropyrimidin-2-on-Derivaten zur Behandlung der pulmonalen arteriellen Hyptertonie
JP2008273852A (ja) * 2007-04-26 2008-11-13 Kowa Co 3環性ピリミジノン誘導体及びこれを含有するstat6活性化阻害剤
WO2009037413A1 (en) 2007-09-19 2009-03-26 Argenta Discovery Limited Dimers of 5- [ (4-cyanophenyl) sulfinyl] -6-methyl-2-oxo-1- [3- (trifluoromethyl)phenyl] -1,2-dihydropyridine-3-carboxamide as inhibitors of human neutrophil elastase for treating respiratory diseases
WO2009060158A1 (en) 2007-11-07 2009-05-14 Argenta Discovery Limited 4- (4-cyanophenyl) -1- (3-trifluoromethylphenyl) -3,4, 6, 7-tetrahydro-1h-pyrrolo [3, 4- d] pyrimidine-2, 5-dione derivatives and their use as human neutrophil elastase inhibitors
GB0721866D0 (en) 2007-11-07 2007-12-19 Argenta Discovery Ltd Pharmaceutical compounds having dual activities
WO2009060206A1 (en) 2007-11-07 2009-05-14 Argenta Discovery Limited 3,4,6,7-tetrahydro-1h-pyrrolo[3,4-d]pyrimidine-2,5-diones and their therapeutic use
DE102007061766A1 (de) 2007-12-20 2009-06-25 Bayer Healthcare Ag 4-(4-Cyano-2-thioaryl)-dihydropyrimidinone und ihre Verwendung
DE102008022521A1 (de) 2008-05-07 2009-11-12 Bayer Schering Pharma Aktiengesellschaft 1,4-Diaryl-pyrimidopyridazin-2,5-dione und ihre Verwendung
MX2010006033A (es) * 2007-12-20 2010-07-16 Bayer Schering Pharma Ag 4-(4-ciano-2-tioaril)dihidropirimidinonas y su uso.
ES2409262T3 (es) 2008-06-02 2013-06-26 Janssen Pharmaceutica, N.V. Antagonistas de TRPA1 de 3,4-dihidropirimidina
UY32138A (es) * 2008-09-25 2010-04-30 Boehringer Ingelheim Int Amidas sustituidas del ácido 2-(2,6-dicloro-fenilamino)-6-fluoro-1-metil-1h-bencimidazol-5-carboxílico y sus sales farmacéuticamente aceptables
DE102009004197A1 (de) 2009-01-09 2010-07-15 Bayer Schering Pharma Aktiengesellschaft Heterocyclisch anellierte Diaryldihydropyrimidin-Derivate und ihre Verwendung
US8530487B1 (en) * 2009-01-29 2013-09-10 Hydra Biosciences, Inc. Compounds useful for treating disorders related to TRPA1
DE102009016553A1 (de) 2009-04-06 2010-10-07 Bayer Schering Pharma Aktiengesellschaft Sulfonamid- und Sulfoximin-substituierte Diaryldihydropyrimidinone und ihre Verwendung
WO2011042145A1 (en) 2009-10-07 2011-04-14 Siena Biotech S.P.A. Wnt pathway antagonists
TWI508725B (zh) 2010-01-08 2015-11-21 Catabasis Pharmaceuticals Inc 脂肪酸富馬酸酯衍生物及其用途
GB201004178D0 (en) 2010-03-12 2010-04-28 Pulmagen Therapeutics Inflamma Enzyme inhibitors
GB201004179D0 (en) 2010-03-12 2010-04-28 Pulmagen Therapeutics Inflamma Enzyme inhibitors
JP2013177318A (ja) 2010-06-30 2013-09-09 Dainippon Sumitomo Pharma Co Ltd ジヒドロピリミジノン誘導体およびその医薬用途
CN104024241B (zh) 2011-08-01 2016-01-20 大日本住友制药株式会社 尿嘧啶衍生物和其用于医学目的的用途
BR112014005669A2 (pt) 2011-09-14 2017-04-04 Chiesi Farm Spa composto, composição farmacêutica e uso de um composto
US20140221335A1 (en) * 2013-02-06 2014-08-07 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9115093B2 (en) 2013-03-04 2015-08-25 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Groutas et al. Expert Opin Ther Pat. 2011 March ; 21(3): 339-354. *
Snyder et al., J. Med. Liban 48(4): 208-214, 2000. PubMed Abstract. *
Sugar et al., Diagno Microbiol. Infect. Dis. 21" 129-133, 1995. *
Turner et al., Current Pharmaceutical Design. 2, 209-224, 1996. *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9458113B2 (en) 2014-07-31 2016-10-04 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9475779B2 (en) 2014-07-31 2016-10-25 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US10189776B2 (en) 2015-09-18 2019-01-29 Boehringer Ingelheim International Gmbh Stereoselective process

Also Published As

Publication number Publication date
AU2014214031B2 (en) 2018-06-28
UY35311A (es) 2014-08-29
MX361166B (es) 2018-11-29
MA38327A2 (fr) 2016-12-30
AR128259A2 (es) 2024-04-10
KR102221201B1 (ko) 2021-03-03
CN106008369A (zh) 2016-10-12
HRP20171040T1 (hr) 2017-10-06
EA201500816A1 (ru) 2016-01-29
KR20150114575A (ko) 2015-10-12
PT2953943T (pt) 2017-07-18
CY1119063T1 (el) 2018-01-10
HK1211573A1 (zh) 2016-05-27
JP2016511240A (ja) 2016-04-14
PE20151429A1 (es) 2015-10-14
US20160340319A1 (en) 2016-11-24
PL2953943T3 (pl) 2017-10-31
CL2015002030A1 (es) 2015-12-11
CA2900308A1 (en) 2014-08-14
SI2953943T1 (sl) 2017-07-31
PH12015501697A1 (en) 2015-10-12
JP6130519B2 (ja) 2017-05-17
CN106045920B (zh) 2018-10-19
CN106045921B (zh) 2018-09-25
BR112015017929B1 (pt) 2023-01-24
NZ709041A (en) 2020-01-31
LT2953943T (lt) 2017-06-12
ME02697B (me) 2017-10-20
TWI663156B (zh) 2019-06-21
TW201831455A (zh) 2018-09-01
ES2629023T3 (es) 2017-08-07
HUE14705723T2 (hu) 2018-02-28
MX2015009512A (es) 2015-10-30
RS56063B1 (sr) 2017-10-31
EA030569B1 (ru) 2018-08-31
CN104995186B (zh) 2017-06-20
US20160060231A1 (en) 2016-03-03
AU2014214031A1 (en) 2015-07-02
MA38327B1 (fr) 2019-03-29
BR112015017929A2 (pt) 2017-07-11
AP2015008535A0 (en) 2015-06-30
PH12015501697B1 (en) 2015-10-12
CN104995186A (zh) 2015-10-21
ECSP15036282A (es) 2016-01-29
EP2953943B1 (en) 2017-04-12
CA2900308C (en) 2021-01-12
BR112015017929A8 (pt) 2019-11-05
TN2015000328A1 (en) 2017-01-03
US9670166B2 (en) 2017-06-06
MA38327A3 (fr) 2019-01-31
AR095164A1 (es) 2015-09-30
DK2953943T3 (en) 2017-07-03
TW201444804A (zh) 2014-12-01
GEP201706741B (en) 2017-09-25
US9290459B1 (en) 2016-03-22
UA117007C2 (uk) 2018-06-11
CN106008369B (zh) 2019-06-21
WO2014122160A1 (en) 2014-08-14
IL239472A0 (en) 2015-07-30
CN106045921A (zh) 2016-10-26
ZA201504257B (en) 2017-11-29
US20160060230A1 (en) 2016-03-03
SG11201505959QA (en) 2015-08-28
EP2953943A1 (en) 2015-12-16
TWI634109B (zh) 2018-09-01
CN106045920A (zh) 2016-10-26
IL239472B (en) 2018-06-28

Similar Documents

Publication Publication Date Title
US9670166B2 (en) Substituted bicyclic dihydropyrimidinones 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
US9440930B2 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US20140057926A1 (en) Substituted 4-pyridones 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
US9221807B2 (en) Substituted pyridones and pyrazinones 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
STCB Information on status: application discontinuation

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