WO2014029830A1 - Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity - Google Patents

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

Info

Publication number
WO2014029830A1
WO2014029830A1 PCT/EP2013/067425 EP2013067425W WO2014029830A1 WO 2014029830 A1 WO2014029830 A1 WO 2014029830A1 EP 2013067425 W EP2013067425 W EP 2013067425W WO 2014029830 A1 WO2014029830 A1 WO 2014029830A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
methyl
phenyl
preparation
cycloalkyl
Prior art date
Application number
PCT/EP2013/067425
Other languages
French (fr)
Inventor
Thorsten Oost
Dennis Fiegen
Christian GNAMM
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
Priority to JP2015527913A priority Critical patent/JP6319747B2/en
Priority to EP13756358.1A priority patent/EP2888242B1/en
Publication of WO2014029830A1 publication Critical patent/WO2014029830A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • This invention relates to substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of pulmonary, gastrointestinal and genitourinary diseases, inflammatory diseases of the skin and the eye and other 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). 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, fibrosis, cancer and others.
  • the compounds according to the present invention are effective as inhibitors of neutrophil elastase and exhibit favourable inhibitory potency, as determined by the half maximal inhibitory concentration (IC 50 ), in an enzymatic inhibition assay.
  • Some compounds according to the present invention exhibit favourable inhibitory potency, as determined by the half maximal effective concentration (EC 50 ), in a plasma or whole-blood assay, for instance as described in T. Stevens et al, J. Pharm. Exp. Ther. 339, 313-320 (2011).
  • Some compounds according to the present invention exhibit favourable in vivo potency, as determined, for example, by the half maximal effective dose (ED 50 ), in models of human neutrophil elastase-induced lung injury in mice, rat or hamster, for instance as described in Tremblay et al. ⁇ Chest 2002, 121, 582-588) or T. Stevens et al. (J. Pharm. Exp. Ther. 2011, 339, 313-320).
  • 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.
  • An improved metabolic stability in an in vitro test system is expected to translate into a reduced in vivo clearance (CL), because the metabolic conversion in the liver is reduced.
  • Some compounds according to the present invention exhibit favourable permeability in an in vitro Caco-2 cell layer method for permeability as described in E. Kerns & L. Di, Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization, Elsevier, 1 st ed, 2008, chapter 26 and references therein.
  • improved permeability is expected to translate into a higher fraction of the drug absorbed in the intestinal tract, thus, resulting in higher dose-normalized systemic exposure (AUC).
  • Comparatively higher dose-normalized systemic exposure can be advantageous in several ways: (1) If a certain systemic exposure (AUC) needs to be achieved for efficacy, the drug can be dosed in a lower amount. Lower dosages have the advantages of lower drug load (parent drug and metabolites thereof) for the patient causing potentially less side effects, and lower production costs for the drug product. (2) Comparatively higher dose- normalized systemic exposure (AUC) can lead to increased efficacy or prolonged duration of action of the drug when the same dose is applied.
  • Some compounds according to the present invention exhibit favourable metabolic stability and favourable permeability.
  • some compounds of the present invention are expected to exhibit favourable pharmacokinetic (PK) properties, in particular favourable systemic exposure (area under the curve, AUC).
  • PK properties can be determined in pre-clinical animal species, for example mouse, rat, dog, guinea pig, mini pig, cynomolgus monkey, rhesus monkey.
  • the PK properties of a compound can be described, for example, by the following parameters: Mean residence time (MRT), elimination half-live (ti/ 2 ), volume-of-distribution (V D ), area under the curve (AUC), clearance (CL), bioavailability after oral administration (F oral ).
  • A is phenyl or a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected independent from each other from the group consisting ofN, (0 " )-N + , O and S;
  • R la l is Ci_ 4 -alkyl-, C 3 - 6 -cycloalkyl-, Ci_ 4 -haloalkyl-, H 2 N-, C -alkyl- H-, C 3 _ 6 -cycloalkyl-NH-, (Ci_ 4 -alkyl) 2 N-, (C 3 _ 6 -cycloalkyl)(Ci_ 4 -alkyl)N-, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, piperazinyl- Ci_ 4 -alkyl-, Ci_ 4 -alkyl-0-NH-, (Ci_ 4 -alkyl-0)(Ci_ 4 -alkyl)N-;
  • R la 2 is H, NC-, 0 2 N-, Ci_ 4 -alkyl-S0 2 -, aryl-S0 2 -, (aromatic heteroring)-S0 2 -, Ci_ 4 -alkyl-0-(0)C-, Ci_ 4 -alkyl-(0)C-, C 3 _ 6 -cycloalkyl-(0)C-, H 2 N-(0)C-, Ci_ 4 -alkyl-NH-(0)C-, C 3 _ 6 -cycloalkyl-NH-(0)C-, (Ci_ 4 -alkyl) 2 N-(0)C-, (C 3 _6-cycloalkyl)(Ci_ 4 -alkyl)N-(0)C-, azetidinyl-(0)C-, pyrrolidinyl-(0)C- piperidinyl-(0)C-, morpholinyl-(0)C-, piperazinyl-(0)C-, piperazinyl-Ci_ 4 alky
  • R 2 is Ci_6-alkyl or a residue selected from the group consisting of C 3 _6-cycloalkyl-, wherein one element of the ring is optionally replaced by O; phenyl-Ci_4-alkyl-, (aromatic heteroring)-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected
  • each element of one of the rings optionally substituted with one or two groups selected from Ci_4-alkyl-, C 3 _6-cycloalkyl-, halogen, Ci_4-haloalkyl-, Ci_4-alkyl-0-, Ci_4-alkyl-
  • R 3 is H or Ci-4-alkyl-
  • R 4 is phenyl, a five- or six-membered, aromatic heteroring containing one, two or three atoms selected from N, O and S; each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, C 3 _6-cycloalkyl-, halogen or Ci_4-haloalkyl-;
  • R 5 is H, Ci_4-alkyl-, Ci_4-haloalkyl-, halogen;
  • R 6 is H or Ci_ 4 -alkyl-
  • R 7 is H or Ci-4-alkyl-; or R 6 and R 7 are forming together a C 2 -5-alkylene, wherein one element of the formed ring is optionally replaced by O; or a pharmaceutically acceptable salt thereof.
  • A is phenyl or pyridinyl; preferably phenyl;
  • R la l is Ci_4-alkyl-, C 3 _ 6 -cycloalkyl-, Ci_4-alkyl-NH- or (C, _4-alkyl) 2 N-,
  • R la 2 is H, methyl, NC-, Me-S0 2 -;
  • R lb is H, methyl, F, preferably H; and R 2 , R 3 , R 4 , R 5 , R 6 , R 7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
  • A is phenyl or pyridinyl; preferably phenyl;
  • R la l is methyl, ethyl, i-propyl, cyclopropyl
  • R la'2 is NC-
  • R lb is H; and R 2 , R 3 , R 4 , R 5 , R 6 , R 7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
  • R 3 is methyl; and A, R la , R lb , R 2 , R 4 , R 5 , R 6 , R 7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
  • R la , R lb , R 2 , R 4 , R 5 , R 6 , R 7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
  • R 5 is H
  • R 6 is H
  • R 7 is H; and A, R la , R lb , R 2 , R 3 , R 4 , have the above given meaning; or a pharmaceutically
  • A is phenyl or pyridinyl
  • R la is . referably R a'2 N 1 -
  • R la l is Ci_4-alkyl-
  • R la'2 is NC-
  • R lb is H
  • R 2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring containing one or two nitrogen atoms, each of the above rings optionally substituted with
  • R 3 is methyl
  • R 4 is phenyl, a five- or six-membered, aromatic ring containing one or two nitrogen atom, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-;
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • A is phenyl or pyridinyl
  • R la'2 is NC-
  • R I b is H
  • R 2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five-membered, aromatic ring containing two nitrogen atoms; each optionally substituted with methyl or NC-;
  • R 3 is methyl
  • R 4 is phenyl or a six-membered, aromatic heteroring containing one or two nitrogen atom; each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-;
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • A is phenyl or pyridinyl
  • R la l is methyl, ethyl, i-propyl, n-propyl;
  • R 2 is ethyl, i-propyl, 2-butyl, cyclo butyl, 1-methylpyrazolyl, or benzyl or 4-NC- benzyl;
  • R 3 is methyl
  • R 4 is phenyl or pyridinyl, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of methyl, ethyl, propyl, F, Br, CI, F 2 HC- or F 3 C-;
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • A is phenyl
  • R-N* - OR R-N* V a - 1 ; preferably R ⁇ N* -
  • R la l is methyl
  • R la - 2 is NC-; is H; is ethyl, i-propyl, cyclo butyl, 1-methylpyrazolyl, benzyl or 4-NC-benzyl; R 3 is methyl;
  • R 4 is phenyl or pyridinyl, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of F, F 2 HC- or F 3 C-;
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • A is phenyl
  • R la is preferably
  • R la l is methyl
  • R la - 2 is NC-
  • R lb is H
  • R 2 is selected from a roup consisting of
  • R 3 is methyl; is selected from a group consisting of
  • p referably ; preferably
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • R 4 is one of the above mentioned ring carrying the above mentioned optional substituted in meta-position to the element connection R 4 with the compound of formula 1.
  • A is phenyl or pyridinyl; preferably phenyl;
  • R la l is Ci_4-alkyl-, C 3 - 6 -cycloalkyl-, Ci_ 4 -alkyl-NH- or (Ci_ 4 -alkyl) 2 N-,
  • R la 2 is H, methyl, NC-, Me-S0 2 -;
  • R lb is H, methyl, F, preferably H;
  • R 2 is Ci_6-alkyl or a residue selected from the group consisting of C 3 -6-cycloalkyl-, and a five-membered, aromatic heteroring containing one or two nitrogen atoms, each of the above rings optionally substituted with Ci_4-alkyl-;
  • R 3 is H or Ci-4-alkyl-
  • R 4 is phenyl, a six-membered, aromatic heteroring containing one or two N; each optionally substituted with one or two residues selected independently from each other from the group consisting of halogen or Ci_4-haloalkyl-; preferably F, F 2 HC- or F 3 C-;
  • R 5 is H
  • R 6 is H
  • R 7 is H; or a pharmaceutically acceptable salt thereof.
  • A is phenyl
  • R la l is Ci_4-alkyl-
  • R la'2 is NC-
  • R 2 is ethyl, i-propyl, cyclopropyl, eye lo butyl, 1-methyl-pyrazolyl;
  • R 3 is methyl;
  • R 4 is phenyl; substituted with one or two residues selected independently from each other from the group consisting of F, F 2 HC- or F 3 C-;
  • R lb , R 5 , R 6 and R 7 is H
  • Ci_ 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-Ci_ 3 -alkyl-" means an aryl group which is bound to a Ci_ 3 -alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached.
  • aryl-Ci_ 3 -alkyl- means an aryl group which is bound to a Ci_ 3 -alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached.
  • An asterisk, a dashed or a dotted line may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.
  • 3-carboxypropyl-group represents the following substituent:
  • a X with a subscript number or "#”, a dashed or a dotted line may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound.
  • prevention means 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 expression of a disease
  • the purpose of prevention is to combat the development of the disease, condition or disorder, and includes the administration of the active compounds to prevent or delay the onset of the symptoms or complications and to prevent or delay the development of related diseases, conditions or disorders. Success of said preventive treatment is reflected statistically by reduced incidence of said condition within a patient population at risk for this condition in comparison to an equivalent patient population without preventive treatment.
  • treatment means therapeutic treatment of patients having already developed one or more of said conditions in manifest, acute or chronic form, including symptomatic treatment in order to relieve symptoms of the specific indication or causal treatment in order to reverse or partially reverse the condition or to delay the progression of the indication as far as this may be possible, depending on the condition and the severity thereof.
  • treatment of a disease means the management and care of a patient having developed the disease, condition or disorder.
  • the purpose of treatment is to combat the disease, condition or disorder.
  • Treatment includes the administration of the active compounds to eliminate or control the disease, condition or disorder as well as to alleviate the symptoms or complications associated with the disease, condition or disorder.
  • a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc%) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound.
  • halogen generally denotes fluorine, chlorine, bromine and iodine.
  • prodrug refers to (i) an inactive form of a drug that exerts its effects after metabolic processes within the body converting it to a usable or active form, or (ii) a substance that gives rise to a pharmacologically active metabolite, although not itself active (i.e. an inactive precursor).
  • prodrug or “prodrug derivative” mean a covalently-bonded derivative, carrier or precursor of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s).
  • prodrugs either have metabolically cleavable or otherwise convertible groups and are rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood or by activation via oxidation as in case of thioether groups.
  • Most common prodrugs include esters and amide analogs of the parent compounds.
  • prodrug is formulated with the objectives of improved chemical stability, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity).
  • prodrugs themselves have weak or no biological activity and are stable under ordinary conditions.
  • Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: "Design and Applications of Prodrugs”; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug
  • 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, lH-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, l-(2-hydroxyethyl)-pyrrolidinyl, sodium hydroxide, triethanolamine (2, 2', 2"- nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2.2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenes,
  • 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, ) also comprise a part of the invention.
  • Ci_ n -alkyl wherein n is an integer from 2 to 6, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to 6 C atoms.
  • Ci_5-alkyl embraces the radicals H 3 C-,
  • Ci_6-alkylene wherein n is an integer 2 to 6, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to 6 carbon atoms.
  • Ci_4-alkylene
  • C 2 _ n -alkylene wherein n is an integer 3 to 5, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 2 to 5 carbon atoms.
  • C 2 _5-alkylene
  • halo added to a “alkyl”, “alkylene” or “cycloalkyl” group (saturated or unsaturated) is such a alkyl or cycloalkyl group meant wherein one or more hydrogen atoms are replaced by a halogen atom selected from among fluorine, chlorine or bromine, preferably fluorine and chlorine, particularly preferred is fluorine. Examples include:
  • C 3 _6-cycloalkyl wherein n is an integer from 4 to 6, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to 6 C atoms.
  • C 3 _6-cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • aryl denotes a carbocyclic aromatic monocyclic group containing 6 carbon atoms which may be further fused to a second five- or six-membered, carbocyclic group which may be aromatic, saturated or unsaturated.
  • Aryl includes, but is not limited to, phenyl, indanyl, indenyl, naphthyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl and dihydronaphthyl.
  • a phenyl ring contains 6 elements which are all carbon atoms
  • a pyrrol ring contains 5 elements, wherein 4 elements are carbon atoms and the remaining element is a nitrogen atom.
  • the term is intended to include all possible isomeric forms.
  • the term includes (if not otherwise restricted) the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • the compounds according to the present invention and their intermediates may be obtained using methods of synthesis which are known to the one skilled in the art and described in the literature of organic synthesis.
  • the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section.
  • the order in carrying out the reaction steps may be varied. Variants of the reaction methods that are known to the one skilled in the art but not described in detail here may also be used.
  • the general processes for preparing the compounds according to the invention will become apparent to the one skilled in the art studying the following schemes.
  • Starting materials are commercially available or may be prepared by methods that are described in the literature or herein, or may be prepared in an analogous or similar manner. Any functional groups in the starting materials or intermediates may be protected using conventional protecting groups. These protecting groups may be cleaved again at a suitable stage within the reaction sequence using methods familiar to the one skilled in the art.
  • Starting material I can be prepared as described in US2003/87940.
  • Intermediates II can be prepared as described in WO10133973 and US2003/87940 by heating starting material I with amines R-NH 2 in the presence of a strong base, for example sodium tert-butoxide or sodium ethoxide, in an organic solvent, for example ethanol. The reaction usually takes place within 2 to 72 hours. Preferred reaction temperatures are between 50°C and 150°C.
  • a strong base for example sodium tert-butoxide or sodium ethoxide
  • the amide coupling (Step B, intermediates II intermediates III, intermediates IV intermediates V, intermediates VI compounds of the invention) can be achieved by reacting carboxylic acid intermediates II, IV or VI with amines R'-NH 2 in the presence of an amide coupling reagent, for example 0-(Benzotriazol-l-yl)-N, N, ⁇ ', N'- tetramethyluronium hexafluorophosphate (HBTU),0-(Benzotriazol-l-yl)-N, N, ⁇ ', N'- tetramethyluronium tetrafluoroborate (TBTU) or propane phosphonic acid anhydride, and in the presence of a base, for example triethylamine, diisopropylethylamine (DIPEA, Hunig's base) or N-methyl-morpholine, in an organic solvent, for example N, N- dimethylformamide (DMF), N-methyl-2-pyrrolidone (
  • carboxylic acid intermediates can be activated first as described in US2003/87940, for example with 1, l '-carbonyldiimidazole (CDI) in DMF, followed by reaction with the amine R'-NH 2 .
  • CDI 1, l '-carbonyldiimidazole
  • bromination agents for example bromine or N-bromosuccinimide
  • iodination agents for example iodine, iodinechloride (I-Cl) or N-iodosuccinimide
  • organic solvent for example acetic acid, methanol, ethanol, dichloromethane, acetonitrile, N, N-dimethyl- formamide, tetrahydrofuran or mixtures therof.
  • the halogenation reaction usually takes place
  • Step D intermediates IV -7 intermediates VI, intermediates V -7 compounds according to the invention
  • aryl or heteroaryl boronic acids R"-B(OH) 2 or the corresponding boronic esters in the presence of a palladium catalyst, for example tetrakis(triphenylphosphine)palladium(0) or [1, -bis(diphenylphosphino)ferrocene]dichloropalladium(II), and in the presence of a base, for example, potassium carbonate, barium dihydroxide or cesium carbonate, in an organic solvent, for example toluene, benzene, ethanol, ethylene glycol dimethyl ether, acetonitrile, dioxane or mixtures thereof, optionally in the presence of water.
  • a palladium catalyst for example tetrakis(triphenylphosphine)palladium(0) or [1, -bis(diphenylphosphino)ferrocene]dichloro
  • reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 50°C and 150°C.
  • Compounds according to the present invention can also be prepared according to the following scheme starting from 4-hydroxy-6-methyl-nicotinic acid. Halogenation (Step C) as described above, followed by Suzuki coupling (Step D) as described above, followed by amide coupling (Step B) as described above, yields intermediates VII.
  • the alkylation of the pyridone nitrogen can be achieved by reacting intermediate VII with alkylating agents, for example alkyl bromides, alkyl iodides, alkyl tosylates, alkyl mesylates or dialkyl sulfates, in the presence of a base, for example sodium carbonate, potassium carbonate, potassium hydroxide, sodium hydroxide or cesium carbonate, in an organic solvent, N, N-dimethylformamide (DMF), N-methyl-2-pyrrolidone (NMP) or dimethylacetamide (DMA).
  • alkylating agents for example alkyl bromides, alkyl iodides, alkyl tosylates, alkyl mesylates or dialkyl sulfates
  • a base for example sodium carbonate, potassium carbonate, potassium hydroxide, sodium hydroxide or cesium carbonate
  • NMF N-dimethylformamide
  • NMP N-methyl-2-pyrrolidone
  • Preferred reaction temperatures are between 50°C and 150°C.
  • Intermediates VIII can be prepared as described in Bolm et al, Org. Lett. 9, 3809-3811 (2007) via direct imination of a thioether (intermediates VII) with cyanamide (Step F, intermediates VII intermediates VIII) by reaction with an halogenating agent, for example N-bromo- or N-chlorosuccinimide, tert-butyl hypochlorite or iodine in the presence of cyanamide and a base, for example potassium or sodium tert-butoxide, in an organic solvent, for example methanol, tetrahydrofuran (THF) or acetonitrile.
  • an organic solvent for example methanol, tetrahydrofuran (THF) or acetonitrile.
  • THF tetrahydrofuran
  • Preferred reaction temperatures are between 0 °C and 50°C, most preferred is room temperature.
  • the imination (Step F, intermediates VII intermediates VIII) can be achieved as described in Bolm et al., Org. Lett. 9, 2951-2954 (2007), by using phenyliodo diacetate [PhI(OAc) 2 ] and cyanamide in an organic solvent, for example acetonitrile or tetrahydrofuran (THF).
  • the reaction usually takes place within 1 to 72 hours.
  • Preferred reaction temperatures are between 0°C and room temperature.
  • Step G intermediates VIII intermediates IX, intermediates XI intermediates XII
  • a peroxycarboxylic acid for example meto-chloroperoxybenzoic acid (mCPBA)
  • mCPBA meto-chloroperoxybenzoic acid
  • a base for example potassium carbonate
  • organic solvent for example ethanol, methanol, dichloromethane or chloroform.
  • Preferred reaction temperatures are between 0°C and room temperature.
  • oxidizing agents for example hydrogen peroxide, tert-butyl hydroperoxide, sodium hypochlorite, sodium iodate, sodium periodate, potassium permanganate, ruthenium tetroxide, potassium peroxymonosulfate (Oxone) or dimethyldioxirane.
  • Intermediates X can be obtained by imination of intermediates VII (Step H, intermediates VII -» intermediates X) as described in Bolm. et al, Org. Lett. 6, 1305-1307 (2004), by reacting a thioether (intermediates VII) with trifluoroacetamide, phenyliodo diacetate [PhI(OAc) 2 ], rhodium acetate dimer [Rh 2 (OAc) 4 ] and MgO in an organic solvent, for example dichloromethane. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and room temperature.
  • Step K intermediates X intermediates XI
  • a base for example potassium carbonate
  • organic solvent for example methanol
  • intermediates XI can be prepared by the imination of intermediates VII using other electrophilic nitrogen sources, for example tert-butyl 3-(4-cyano-phenyl)- oxaziridine-2-carboxylate (followed by cleavage of the Boc-protecting group), O-mesityl sulfonyl hydroxylamine (MSH), or hydrazoic azid.
  • other electrophilic nitrogen sources for example tert-butyl 3-(4-cyano-phenyl)- oxaziridine-2-carboxylate (followed by cleavage of the Boc-protecting group), O-mesityl sulfonyl hydroxylamine (MSH), or hydrazoic azid.
  • Step K intermediates XI intermediates VIII, intermediates XII intermediates IX
  • Step K intermediates XI intermediates VIII, intermediates XII intermediates IX
  • cyanogen bromide optionally 4-dimethylaminopyridine (DMAP) and optionally a base, for example triethylamine
  • DMAP 4-dimethylaminopyridine
  • a base for example triethylamine
  • organic solvent for example dichloromethane.
  • Preferred reaction temperatures are between 0°C and 50°C, most preferred room temperature.
  • the synthesis of sulfonimidamides has been described in Johnson et al, J. Org.
  • Sulfmamides (starting material XIII) can be prepared as described in Johnson et al., J. Org Chem. 44, 2055-2061 (1975) by reacting sulfmyl chlorides with a primary or secondary amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example dichloromethane, tetrahydrofuran, diethylether, acetonitrile, toluene, N,N-dimethylformamide, ethanol or ethyl acetate.
  • the reaction usually takes place within 1-72 hours.
  • Preferred reaction temperatures are between 0 °C and room temperature.
  • Sulfonimidoyl chlorides can be prepared as described in Johnson et al, J. Org. Chem. 44, 2055-2061 (1975), by reacting sulfinamides (starting material XIII) with a chlorinating agent, for example chlorine, N-chlorosuccinimide (NCS), N-chloro- benzotriazole or tert-butyl hypochlorite in an organic solvent, for example dichloromethane, diethylether, benzene, toluene, carbon tetrachloride, acetonitrile or tetrahydrofuran (Step L). The reaction usually takes place within less than 1 hour.
  • a chlorinating agent for example chlorine, N-chlorosuccinimide (NCS), N-chloro- benzotriazole or tert-butyl hypochlorite
  • an organic solvent for example dichloromethane, diethylether, benzene, tol
  • reaction temperatures are between -78 °C and room temperature, mostly preferred between 0 °C and room temperature.
  • Sulfonimidoyl fluorides can be prepared as described in Johnson et al, J. Org. Chem. 48, 1-3 (1983) and Gnamm et al, Bioorg. Med. Chem. Lett.
  • Step M by reacting sulfonimidoyl chlorides (intermediates XIV) with a fluoride salt, for example potassium fluoride, sodium fluoride or tetrabutylammonium fluoride, optionally in the presence of a crown ether, for example 18-crown-6, in an organic solvent, for example acetonitrile or tetrahydrofuran (Step M).
  • a fluoride salt for example potassium fluoride, sodium fluoride or tetrabutylammonium fluoride
  • a crown ether for example 18-crown-6
  • organic solvent for example acetonitrile or tetrahydrofuran
  • Sulfonimidamides can be prepared as described in Johnson et al, J. Org. Chem. 44, 2055-2061 (1975), Bolm et al, J. Org. Chem. 75, 3301-3310 (2010), and WO09156336 by reacting sulfonimidoyl chlorides (intermediates XIV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step N).
  • the reaction takes place within 1 to 72 hours.
  • Preferred reaction temperatures are between 0 °C and room temperature.
  • sulfonimidamides can be prepared as described in Gnamm et al, Bioorg. Med. Chem. Lett. 22, 3800-3806 (2012) by reacting sulfonimidoyl fluorides (intermediates XV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, for example l,8-diazabicyclo[5.4.0]undec-7-ene (DBU), in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step O).
  • the reaction takes place within 1 to 72 hours.
  • Preferred reaction temperatures are between 0°C and the boiling point of the solvent, mostly preferred between room temperature and 110°C.
  • Preparation 2a is prepared following the procedure described for preparation lb, substituting isopropylamine with ethylamine.
  • ESI mass spectrum: [M+H] + 182; r.t. HPLC: 0.62 min (Z002 006).
  • 2b 5-Bromo- 1 -ethyl-6-methyl-4-o - 1 ,4-dihydro-pyridine-3-carboxylic acid
  • Preparation 2b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 2a as starting material.
  • ESI mass spectrum: 5 [M+H] + 260 (bromine isotope pattern); r.t. HPLC: 0.89 min (Z002 007).
  • Preparation 3a is prepared following the procedure described for preparation lb, substituting isopropylamine with cyclopropylamine.
  • ESI mass spectrum: [M+H] + 208; r.t. HPLC: 0.62 min (Z002 002).
  • Preparation 3b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 3a as starting material.
  • ESI mass spectrum: [M+H] + 286 (bromine isotope pattern); r.t. HPLC: 0.82 min (Z002 002).
  • 3c 5-Bromo-l-cyclobutyl-6-methyl-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid 4-(N- cyano-S-methylsulfonimidoy ⁇ -benzylamide
  • Preparation 3c is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 3b as starting material and substituting TBTU with HBTU as coupling reagent.
  • ESI mass spectrum: [M+H] + 477 (bromine isotope pattern); r.t. HPLC: 0.90 min (Z018 S04).
  • Preparation 4a is prepared following the procedure described for preparation lb, substituting isopropylamine with l-methyl-lH-pyrazol-5-ylamine.
  • ESI mass spectrum: [M+H] + 234; r.t. HPLC: 0.63 min (Z018 S04).
  • Preparation 4b is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 4a as starting material and substituting TBTU with HBTU as coupling reagent.
  • ESI mass spectrum: [M+H] + 425; r.t. HPLC: 0.79 min (Z018 S04).
  • Preparation 5a is prepared following the procedure described for preparation lb, substituting isopropylamine with 4-cyanobenzylamine.
  • ESI mass spectrum: [M+H] + 269; r.t. HPLC: 0.86 min (Z002 005).
  • Preparation 5b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 5a as starting material.
  • ESI mass spectrum: [M+H] + 347 (bromine isotope pattern); r.t. HPLC: 1.07 min (Z002 005).
  • Example 1 5-(3-Difluoromethyl-phenyl)-l-isopropyl-6-methyl-4-oxo-l,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
  • Example 1A and Example IB Enantiomers of Example 1
  • racemic example 1 63 mg are separated by chiral HPLC (Daicel IB, 250mm x 20mm, 15% MeOH + 0.2% diethylamine in supercritical C0 2 , 40°C).
  • Example 2 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
  • Example 2 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid.
  • Example 2 A and Example 2B Enantiomers of Example 2
  • racemic example 2 100 mg are separated by chiral HPLC (Daicel OZ-H, 250mm x 20mm, 40% MeOH + 0.2% diethylamine in supercritical C0 2 , 40°C).
  • Example 3 l-Isopropyl-2-methyl-4-oxo-2 '-trifluor omethyl- 1 ,4-dihydro- [3,4 '] - bipyridinyl-5-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
  • Example 3 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid.
  • Example 3 A and Example 3B Enantiomers of Example 3
  • racemic example 3 50 mg are separated by chiral HPLC (Daicel IB, 250mm x 20mm, 15% MeOH + 0.2% diethylamine in supercritical C0 2 , 40°C).
  • Example 4 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid and substituting preparation 1 with preparation 2 as starting material.
  • ESI mass spectrum :
  • Example 4 A and Example 4B Enantiomers of Example 4
  • racemic example 4 are separated by chiral HPLC (Daicel OZH, 250mm x 20mm, 40% MeOH + 0.2% diethylamine in supercritical C0 2 , 40°C).
  • Example 5 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 2 as starting material.
  • ESI mass spectrum: [M+H] + 499; r.t. HPLC: 0.79 min (Z01 1 S03).
  • Example 6 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 3 as starting material.
  • ESI mass spectrum: [M+H] + 525; r.t. HPLC: 0.84 min (Z01 1 S03).
  • Example 7 5-(3-Difluoromethyl-phenyl)-6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4- oxo-1, 4-dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)- benzylamide
  • Example 7 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 4 as starting material.
  • ESI mass spectrum: [M+H] + 551; r.t. HPLC: 0.96 min (Z018 S04).
  • Example 8 6-Methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-5-(3-trifluoromethyl- phenyl)- 1, 4-dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)- benzylamide
  • Example 8 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid and substituting preparation 1 with preparation 4 as starting material.
  • ESI mass spectrum: [M+H] + 569; r.t. HPLC: 1.00 min (Z018 S04).
  • Example 9 l-(4-Cyano-benzyl)-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4- dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
  • Example 9 is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 5 as starting material.
  • ESI mass spectrum: [M+H] + 604; r.t. HPLC: 0.63 min (X012 S01).
  • Example 9 A and Example 9B Enantiomers of Example 9
  • racemic example 9 58 mg are separated by chiral HPLC (Daicel ASH, 250mm x 20mm, 20% MeOH + 0.2% diethylamine in supercritical C0 2 , 40°C).
  • bromo intermediates are prepared as described for preparation lc, employing the appropriate des-bromo intermediates as starting material.
  • Racemic preparation 6c (2.0 g) is separated by chiral HPLC (Daicel Chiralcel OZ-H, 250 mm x 30 mm, 32% MeOH in supercritical C0 2 ).
  • racemic preparation 6d The early eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-5'-methyl- sulfonimidoyl)benzyl)isoindoline-l ,3-dione with preparation 1 1 A.
  • the late eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-S'-methylsulfonimidoyl)- benzyl)isoindoline-l ,3-dione with preparation 1 IB.
  • Phthalic anhydride (4.42 g, 30 mmol) is added to a solution of 4-(ethylthio)benzylamine (5.00 g, 30 mmol) in acetic acid. The mixture is heated at reflux for 5 h and cooled at room temperature. Water ist added, and the precipitate is filtered and dried.
  • WO05026124, 4.28 g, 28.5 mmol) in THF is cooled at -78 °C in a dry ice / ethanol bath.
  • Lithium aluminium hydride (2.4 M in THF, 12 mL, 28.8 mmol) is added dropwise. After 10 min water (1 mL) and THF (3 mL) are added. After 5 min, aqueous sodium hydroxide solution (4 M, 1 mL) and water (3 mL) is added.
  • Example 60 5-(3-Trifluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V VyV-trimethylsulfamimidoyl)-benzylamide
  • 60a Thioacetic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2-ylmethyl)-phenyl ester
  • a solution of acetic anhydride (4.34 mL, 45.9 mmol) in dichloromethane (50 mL) is added dropwise to a precooled solution of thioacetic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2- ylmethyl)-phenyl ester (preparation 60a, 13.0 g, 41.8 mmol) in dichloromethane (500 mL) at -20 °C.
  • a solution of sulfuryl chloride (7.4 mL, 91.9 mmol) in dichloromethane (50 mL) is added at this temperature, and the mixture is stirred for 1 h. The mixture is warmed to room temperature, and all volatiles are removed under reduced pressure.
  • Example 60 5-(3-Trifluoromethyl-phenyl)-l-isopropyl-6-methyl-4-oxo-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(N,N,N'-trimethylsulfamimidoyl)-benzylamide
  • Example 61 5-(3-Difluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V ⁇ yV-trimethylsulfamimidoyl)-benzylamide
  • Example 61 is prepared as described for Example 60, substituting preparation 9.2 with preparation 9.12.
  • ESI mass spectrum: [M+H] + 531 ; r.t. HPLC: 0.86 min (Z01 1 S03).
  • Example 62 5-(3-Trifluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid (5-( V-cyano-S-methylsulfonimidoyl)-l-oxy-pyridin-2-yl)- methylamide (Pyridine N-oxide of Exam le 16)
  • eto-chloroperoxybenzoic acid (77%, 8.0 mg, 36 ⁇ ) is added to a solution of 5-(3-trifiuoromethyl-phenyl)- 1 -isopropyl-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3- carboxylic acid (5-(N-cyano-5'-methylsulfonimidoyl)pyridin-2-yl)methylamide (example 16, 17.0 mg, 32 ⁇ ) in dichloromethane (0.5 mL), and the mixture is stirred at room temperature for 2 h. The mixture is heated at reflux and stirred for 2 h. The mixture is cooled at room temperature and stirred over night.
  • Example 63 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-(S-cyclopropyl- V-cyanosulfonimidoyl)-benzylamide
  • Example 64 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-(N-(cyanomethyl)-S-methylsulfonimidoyl)-benzylamide
  • Example 64 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(N-(cyanomethyl)-5'-methylsulfonimidoyl)-benzylamide
  • Example 65 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid [l-(4-( V-cyano-S-methylsulfonimidoyl)-phenyl)-ethyl]- amide
  • Example 65 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro- pyridine-3-carboxylic acid 1 -(4-(N-cyano-5'-methylsulfonimidoyl)-phenyl)-ethyl]-amide
  • Example 66 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 3-methyl-4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
  • N-Bromosuccinimide (63 mg, 0.35 mmol) is added to a mixture of l-isopropyl-6-methyl-4- oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3-carboxylic acid 3-methyl-4- methylsulfanyl-benzylamide (preparation 66c, 115 mg, 0.24 mmol), cyanamide (14 mg, 0.33 mmol) and potassium tert-butoxide (32 mg, 0.29 mmol) in methanol (1.1 mL). After 20 min saturated aqueous sodium thiosulfate solution is added, and the mixture is extracted twice with dichloromethane.
  • Example 67 l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-( V,S-dimethylsulfonimidoyl)-benzylamide
  • 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: lOOmM Tris, 500mM NaCl, adjusted to pH 7.5; Assay buffer: lOOmM Tris, 500mM NaCl, adjusted to pH 7.5, containing 0.01%BSA.
  • Assay conditions Test compounds were prediluted in DMSO and subsequently in compound buffer (5% DMSO final).
  • IC50 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%. IC50 values of selected compound in the Neutrophil Elastase assay:
  • Example IC50 [nM] Example IC50 [nM]
  • the compounds of general formula I may be used on their own or combined with other active substances of formula I according to the invention.
  • the compounds of general formula I may optionally also be combined with other pharmacologically active substances. These include, B2-adrenoceptor-agonists (short and long-acting),
  • Hydrolase inhibitors or FLAP inhibitors Hydrolase inhibitors or FLAP inhibitors, MMP9-inhibitors, MMP12-inhibitors
  • Non-steroidale anti-inflammatory agents NSAIDs
  • Cathepsin C or DPPI / Dipeptidyl- aminopeptidase I
  • CRTH2 antagonists DPI -receptor modulators
  • Thromboxane receptor antagonists CCR3 antagonists, CCR 4 antagonists, CCR 1 antagonists, CCR5 antagonists, CCR6 antagonists, CCR7 antagonists, CCR8 antagonists, CCR9 antagonists, CCR30 antagonists, , CXCR 3 antagonists, CXCR 4 antagonists, CXCR 2 antagonists,
  • CXCR antagonists CXCR5 antagonists, CXCR6 antagonists, CX3CR antagonists, Neurokinin (NK1, NK2) antagonists, Sphingosine 1 -Phosphate receptor modulators, Sphingosine 1 phosphate lyase inhibitors, Adenosine receptor modulators as for example A2a-agonists, modulators of purinergic receptors as for example P2X7 inhibitors, Histone Deacetylase (HDAC) activators, Bradykinin (BK1, BK2) antagonists, TACE inhibitors, PPAR gamma modulators, Rho-kinase inhibitors, interleukin 1-beta converting enzyme (ICE) inhibitors, Toll-Like receptor (TLR) modulators, HMG-CoA reductase inhibitors, VLA-4 antagonists, ICAM-1 inhibitors, SHIP agonists, GABAa receptor antagonist, ENaC-inhibitors, Prostasin-inhibitors, Melanocortin receptor (MC1
  • betamimetics are anticholinergics, corticosteroids, PDE4-inhibitors,
  • LTD4-antagonists LTD4-antagonists, EGFR-inhibitors, Cathepsin C inhibitors, CRTH2 inhibitors,
  • 5-LO-inhibitors Histamine receptor antagonists and SYK- inhibitors, especially Cathepsin C inhibitors, but also combinations of two or three active substances, i.e.:
  • Betamimetics with corticosteroids PDE4-inhibitors, CRTH2-inhibitors or
  • 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:
  • 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; alpha 1 -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; vasculi
  • skin psoriasis, atopic dermatitis, contact dermatitis or other eczematous dermatoses, and delayed-type hypersensitivity reactions; phyto- and photodermatitis; seborrhoeic dermatitis, dermatitis herpetiformis, lichen planus, lichen sclerosus et atrophica, pyoderma gangrenosum, skin sarcoid, discoid lupus erythematosus, pemphigus, pemphigoid, epidermolysis bullosa, urticaria, angioedema, vasculitides, toxic erythemas, cutaneous eosinophilias, alopecia areata, male-pattern baldness, Sweet's syndrome, Weber-Christian syndrome, erythema multiforme; cellulitis, both infective and non-infective; panniculitis; cutaneous lymphomas, non-melanom
  • drug-induced disorders including fixed drug eruptions;
  • 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 vulvo -vaginitis
  • Peyronie's disease erectile dysfunction (both male and female); 5. allograft rejection: acute and chronic following, for example, transplantation of kidney, heart, liver, lung, bone marrow, skin or cornea or following blood transfusion; or chronic graft versus host disease;
  • 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,
  • 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,
  • 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,
  • bacterial diseases such as tuberculosis and mycobacterium avium, leprosy
  • infectious diseases such as fungal diseases, chlamydia, Candida, aspergillus, cryptococcal meningitis, Pneumocystis carnii, cryptosporidiosis, histoplasmosis, toxoplasmosis, trypanosome infection and leishmaniasis.
  • 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. In any case the active ingredient will be

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This invention relates to substituted 4-pyridones of formula (1) and their use as inhibitors of neutrophil elastase activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of pulmonary, gastrointestinal and genitourinary diseases, inflammatory diseases of the skin and the eye and other auto-immune and allergic disorders, allograft rejection, and oncological diseases.

Description

SUBSTITUTED 4-PYRIDONES AND THEIR USE AS INHIBITORS OF
NEUTROPHIL ELASTASE ACTIVITY
FIELD OF THE INVENTION
This invention relates to substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of pulmonary, gastrointestinal and genitourinary diseases, inflammatory diseases of the skin and the eye and other autoimmune and allergic disorders, allograft rejection, and oncological diseases.
BACKGROUND INFORMATION
• The following references describe neutrophil elastase inhibitors with a 2-pyridone central core: WO04043924, WO05026123, WO05026124, WO06098683,
WO06098684, WO07129962, WO10094964, WOl 1039528.
• The following references describe neutrophil elastase inhibitors with a 2-pyrazinone central core: WO07129963, WO09061271, WO09058076, WOl 1110852.
• For a review on various inhibitors of neutrophil elastase see: P. Sjo {Future Med.
Chem. 2012, 4, 651-660).
BRIEF SUMMARY OF THE INVENTION
Neutrophil elastase is a 29 kDa serine protease. It is expressed in bone marrow precursor cells, stored in the granula of peripheral blood granulocytes at high concentrations and it is released upon cellular activation. To the substrates of NE belong major elements of the extracellular matrix: elastin, fibronectin, laminin, collagen and proteoglycans. Neutrophil elastase activity leads to ECM degradation, increases migration and chemotaxis of monocytes and vascular smooth muscle cells and directly affects components of the coagulation and fibrinolytic pathways (PAI-1 and TFPI). Increased activity of neutrophil elastase is associated with chronic inflammatory and fibrotic diseases of several organs. Inhibitors of neutrophil elastase will therefore have an important role for the treatment of different diseases like COPD, fibrosis, cancer and others.
The compounds according to the present invention, including the physiologically acceptable salts, are effective as inhibitors of neutrophil elastase and exhibit favourable inhibitory potency, as determined by the half maximal inhibitory concentration (IC50), in an enzymatic inhibition assay.
Some compounds according to the present invention, including the physiologically acceptable salts, exhibit favourable inhibitory potency, as determined by the half maximal effective concentration (EC50), in a plasma or whole-blood assay, for instance as described in T. Stevens et al, J. Pharm. Exp. Ther. 339, 313-320 (2011).
Some compounds according to the present invention, including the physiologically acceptable salts, exhibit favourable in vivo potency, as determined, for example, by the half maximal effective dose (ED50), 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).
Some compounds according to the present invention, including the physiologically acceptable salts, 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, 1st ed, 2008, chapter 29 and references therein.
Some compounds according to the present invention, including the physiologically acceptable salts, 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, 1st ed, 2008, chapter 29 and references therein. An improved metabolic stability in an in vitro test system is expected to translate into a reduced in vivo clearance (CL), because the metabolic conversion in the liver is reduced. Based on the pharmacokinetic equation CL/Forai = Dose / AUC (Foral: oral bioavailability, AUC: area under the curve), a reduced in vivo clearance is expected lead to higher dose- normalized systemic exposure (AUC) of the drug.
Some compounds according to the present invention, including the physiologically acceptable salts, 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, 1st ed, 2008, chapter 26 and references therein. For an oral drug, improved permeability is expected to translate into a higher fraction of the drug absorbed in the intestinal tract, thus, resulting in higher dose-normalized systemic exposure (AUC).
Comparatively higher dose-normalized systemic exposure (AUC) 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, including the physiologically acceptable salts, exhibit favourable metabolic stability and favourable permeability.
Accordingly, some compounds of the present invention are expected to exhibit favourable pharmacokinetic (PK) properties, in particular favourable systemic exposure (area under the curve, AUC).
Some compounds according to the present invention, including the physiologically acceptable salts, exhibit favourable pharmacokinetic (PK) properties. The PK properties can be determined in pre-clinical animal species, for example mouse, rat, dog, guinea pig, mini pig, cynomolgus monkey, rhesus monkey. The PK properties of a compound can be described, for example, by the following parameters: Mean residence time (MRT), elimination half-live (ti/2), volume-of-distribution (VD), area under the curve (AUC), clearance (CL), bioavailability after oral administration (Foral).
DETAILED DESCRIPTION OF THE INVENTION
Compounds of formula 1
Figure imgf000005_0001
wherein
A is phenyl or a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected independent from each other from the group consisting ofN, (0")-N+, O and S;
R la
IS
Figure imgf000005_0002
R-N* - or R-N* a-1 ; preferably R-N* -
Rla l is Ci_4-alkyl-, C3-6-cycloalkyl-, Ci_4-haloalkyl-, H2N-, C -alkyl- H-, C3_6-cycloalkyl-NH-, (Ci_4-alkyl)2N-, (C3_6-cycloalkyl)(Ci_4-alkyl)N-, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, piperazinyl- Ci_4-alkyl-, Ci_4-alkyl-0-NH-, (Ci_4-alkyl-0)(Ci_4-alkyl)N-;
Rla 2 is H, NC-, 02N-, Ci_4-alkyl-S02-, aryl-S02-, (aromatic heteroring)-S02-, Ci_4-alkyl-0-(0)C-, Ci_4-alkyl-(0)C-, C3_6-cycloalkyl-(0)C-, H2N-(0)C-, Ci_4-alkyl-NH-(0)C-, C3_6-cycloalkyl-NH-(0)C-, (Ci_4-alkyl)2N-(0)C-, (C3_6-cycloalkyl)(Ci_4-alkyl)N-(0)C-, azetidinyl-(0)C-, pyrrolidinyl-(0)C- piperidinyl-(0)C-, morpholinyl-(0)C-, piperazinyl-(0)C-, piperazinyl-Ci_4 alkyl-(0)C-, Ci_4-alkyl-, C3_6-cycloalkyl-, Ci_4-haloalkyl-, aryl, heteroaryl; R is H, Ci_4-alkyl-, C3_6-cycloalkyl-, Ci_4-haloalkyl- or halogen; or Rla and Rlb are together C2-4-alkylene forming a carbocyclic heteroring;
R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, wherein one element of the ring is optionally replaced by O; phenyl-Ci_4-alkyl-, (aromatic heteroring)-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected
independent from each other from the group consisting of N, O and S; each element of one of the rings optionally substituted with one or two groups selected from Ci_4-alkyl-, C3_6-cycloalkyl-, halogen, Ci_4-haloalkyl-, Ci_4-alkyl-0-, Ci_4-alkyl-
Figure imgf000006_0001
R3 is H or Ci-4-alkyl-;
R4 is phenyl, a five- or six-membered, aromatic heteroring containing one, two or three atoms selected from N, O and S; each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, C3_6-cycloalkyl-, halogen or Ci_4-haloalkyl-;
R5 is H, Ci_4-alkyl-, Ci_4-haloalkyl-, halogen;
R6 is H or Ci_4-alkyl-;
R7 is H or Ci-4-alkyl-; or R6 and R7 are forming together a C2-5-alkylene, wherein one element of the formed ring is optionally replaced by O; or a pharmaceutically acceptable salt thereof.
PREFERRED EMBODIMENTS
Preferred are the above compounds of formula 1, wherein A is phenyl or pyridinyl; preferably phenyl;
Rla is
°VXl 3Λ °VXl
Rla l is Ci_4-alkyl-, C3_6-cycloalkyl-, Ci_4-alkyl-NH- or (C, _4-alkyl)2N-,
preferably Ci-4-alkyl- or C3_6-cycloalkyl-,;
Rla 2 is H, methyl, NC-, Me-S02-;
Rlb is H, methyl, F, preferably H; and R2, R3, R4, R5, R6, R7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl or pyridinyl; preferably phenyl;
Rla is
°V NXr l 3Λ °VXl
R N R or ; preferably
Rla l is methyl, ethyl, i-propyl, cyclopropyl;
Rla'2 is NC-;
Rlb is H; and R2, R3, R4, R5, R6, R7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
R3 is methyl; and A, Rla, Rlb, R2, R4, R5, R6, R7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein R4 is selected from a group consisting of
Figure imgf000008_0001
and A, Rla, Rlb, R2, R4, R5, R6, R7 have the above given meaning; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
R5 is H;
R6 is H;
R7 is H; and A, Rla, Rlb, R2, R3, R4, have the above given meaning; or a pharmaceutically
acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl or pyridinyl;
Rla is
Figure imgf000009_0001
. referably R a'2N 1-
Rla l is Ci_4-alkyl-;
Rla'2 is NC-;
Rlb is H;
R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring containing one or two nitrogen atoms, each of the above rings optionally substituted with
Ci_4-alkyl-, halogen or NC-;
R3 is methyl;
R4 is phenyl, a five- or six-membered, aromatic ring containing one or two nitrogen atom, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl or pyridinyl;
Rla is
°VXl 8Λ °VXl
R-N* - or R-N* Va-1 ; preferably R-N* - Rla l is Ci_4-alkyl-;
Rla'2 is NC-;
RI b is H;
R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five-membered, aromatic ring containing two nitrogen atoms; each optionally substituted with methyl or NC-;
R3 is methyl;
R4 is phenyl or a six-membered, aromatic heteroring containing one or two nitrogen atom; each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl or pyridinyl;
Rla is
°VXl 8Λ °VXl
R-N* - or R-N* Va-1 ; preferably R-N* -
Rla l is methyl, ethyl, i-propyl, n-propyl;
Rla'2 is NC-; RIb is H;
R2 is ethyl, i-propyl, 2-butyl, cyclo butyl, 1-methylpyrazolyl, or benzyl or 4-NC- benzyl;
R3 is methyl;
R4 is phenyl or pyridinyl, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of methyl, ethyl, propyl, F, Br, CI, F2HC- or F3C-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl;
Rla is
°VXl 3Λ °VXl
R-N* - OR R-N* Va-1 ; preferably R^N* -
Rla l is methyl;
Rla-2 is NC-; is H; is ethyl, i-propyl, cyclo butyl, 1-methylpyrazolyl, benzyl or 4-NC-benzyl; R3 is methyl;
R4 is phenyl or pyridinyl, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of F, F2HC- or F3C-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
Preferred are the above compounds of formula 1, wherein
A is phenyl;
Rla is
Figure imgf000012_0001
preferably
Rla l is methyl;
Rla-2 is NC-;
Rlb is H;
R2 is selected from a roup consisting of
Figure imgf000012_0002
R3 is methyl; is selected from a group consisting of
p
Figure imgf000013_0001
referably ; preferably
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
In a preferred embodiment of the invention R4 is one of the above mentioned ring carrying the above mentioned optional substituted in meta-position to the element connection R4 with the compound of formula 1.
From the above mentioned compounds those are preferred wherein R4 is
Figure imgf000013_0002
From the above mentioned compounds those are referred wherein R4 is
Figure imgf000013_0003
From the above mentioned compounds those are preferred wherein R4 is
Figure imgf000014_0001
From the above mentioned compounds those are referred wherein R4 is
Figure imgf000014_0002
From the above mentioned compounds those are referred wherein R4 is
Figure imgf000014_0003
In another embodiment of the invention preferred are the above compounds of formula 1, wherein
A is phenyl or pyridinyl; preferably phenyl;
R la
IS
Figure imgf000014_0004
Rla l is Ci_4-alkyl-, C3-6-cycloalkyl-, Ci_4-alkyl-NH- or (Ci_4-alkyl)2N-,
preferably Ci_4-alkyl- or C3_6-cycloalkyl-,;
Rla 2 is H, methyl, NC-, Me-S02-;
Rlb is H, methyl, F, preferably H; R2 is Ci_6-alkyl or a residue selected from the group consisting of C3-6-cycloalkyl-, and a five-membered, aromatic heteroring containing one or two nitrogen atoms, each of the above rings optionally substituted with Ci_4-alkyl-;
R3 is H or Ci-4-alkyl-;
R4 is phenyl, a six-membered, aromatic heteroring containing one or two N; each optionally substituted with one or two residues selected independently from each other from the group consisting of halogen or Ci_4-haloalkyl-; preferably F, F2HC- or F3C-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention preferred are the above compounds of formula 1, wherein
A is phenyl;
Rla is
°vXi
2N* V
Rla l is Ci_4-alkyl-;
Rla'2 is NC-;
R2 is ethyl, i-propyl, cyclopropyl, eye lo butyl, 1-methyl-pyrazolyl;
R3 is methyl; R4 is phenyl; substituted with one or two residues selected independently from each other from the group consisting of F, F2HC- or F3C-;
Rlb, R5, R6 and R7 is H;
or a pharmaceutically acceptable salt thereof.
Preferred are
Figure imgf000016_0001
Figure imgf000017_0001

Figure imgf000018_0001

Figure imgf000019_0001

Figure imgf000020_0001
20
Figure imgf000021_0001
Figure imgf000022_0001
# example numbers are according to the numbering in the Preparation / experimental section
The residue Rla is chiral, preferred is the Eutomer. USED TERMS AND DEFINITIONS
Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to.
In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example, Ci_6-alkyl means an alkyl group or radical having 1 to 6 carbon atoms.
In general in single groups like HO, H2N, OS, 02S, NC (cyano), HOOC, F3C or the like, the skilled artisan can see the radical attachment point(s) to the molecule from the free valences of the group itself. For combined groups comprising two or more subgroups, the last named subgroup is the radical attachment point, for example, the substituent "aryl-Ci_ 3-alkyl-" means an aryl group which is bound to a Ci_3-alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached. In case a compound of the present invention is depicted in form of a chemical name and as a formula in case of any discrepancy the formula shall prevail. An asterisk, a dashed or a dotted line may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.
For example, the term "3-carboxypropyl-group" represents the following substituent:
Figure imgf000023_0001
wherein the carboxy group is attached to the third carbon atom of the propyl group. The terms "1-methylpropyl-", "2, 2-dimethylpropyl-" or "cyclopropylmethyl-" group represent the following groups:
Figure imgf000023_0002
A X with a subscript number or "#", a dashed or a dotted line may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.
Many of the followings terms may be used repeatedly in the definition of a formula or group and in each case have one of the meanings given above, independently of one another.
The term "substituted" as used herein, 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. The expressions "prevention", "prophylaxis", "prophylactic treatment" or "preventive treatment" used herein 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. Thus the expression "prevention of a disease" as used herein 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.
The expression "treatment" or "therapy" 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. Thus the expression "treatment of a disease" as used herein 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.
Unless specifically indicated, throughout the specification and the appended claims, 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.
When in the claimed ring system A (e.g. SA) the two residues Rla and Rlb are "together a C2-4-alkylene forming a carbocyclic heteroring; rings similar to example SB are meant in this case Rla and Rlb are in ortho position to each other.
Figure imgf000025_0001
The term halogen generally denotes fluorine, chlorine, bromine and iodine.
As used herein the term "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).
The terms "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). Such 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. The 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). In general, 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. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug
Delivery, K.B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzymology, K. Widder et al. (eds.), Vol. 42, Academic Press, 1985, particularly pp. 309-396; Burger's Medicinal Chemistry and Drug Discovery, 5th Ed., M. Wolff (ed.), John Wiley & Sons, 1995, particularly Vol. 1 and pp. 172-178 and pp. 949-982; Pro-Drugs as Novel Delivery
Systems, T. Higuchi and V. Stella (eds.), Am. Chem. Soc, 1975; Bioreversible Carriers in Drug Design, E.B. Roche (ed.), Elsevier, 1987, each of which is incorporated herein by reference in their entireties.
The term "pharmaceutically acceptable prodrug" as used herein 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.
The phrase "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.
As used herein, "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. For example, 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, lH-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, l-(2-hydroxyethyl)-pyrrolidinyl, sodium hydroxide, triethanolamine (2, 2', 2"- nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2.2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 2, 5-dihydroxybenzoic acid, 4-acetamido-benzoic acid, (+)-camphoric acid, (+)-camphor- 10-sulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, decanoic acid, dodecylsulfuric acid, ethane- 1, 2-disulfonic acid, ethanesulfonic acid, 2-hydroxy- ethanesulfonic acid, ethylenediaminetetraacetic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, D-glucoheptonic acid, D-gluconic acid, D-glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, hexanoic acid, hippuric acid, hydrobromic acid, hydrochloric acid, isobutyric acid, DL- lactic acid, lactobionic acid, lauric acid, lysine, maleic acid, (-)-L-malic acid, malonic acid, DL-mandelic acid, methanesulfonic acid, galactaric acid, naphthalene- 1, 5-disulfonic acid, naphthalene-2-sulfonic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, octanoic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid (embonic acid), phosphoric acid, propionic acid, (-)-L-pyroglutamic acid, salicylic acid, 4-amino- salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L- tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid. Further pharmaceutically acceptable salts can be formed with cations from metals like aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like, (also see
Pharmaceutical salts, Berge, S.M. et al, J. Pharm. Sci., (1977), 66, 1-19).
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, ) also comprise a part of the invention.
The term "Ci_n-alkyl", wherein n is an integer from 2 to 6, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to 6 C atoms. For example the term Ci_5-alkyl embraces the radicals H3C-,
H3C-CH2-, H3C-CH2-CH2-, H3C-CH(CH3)-, H3C-CH2-CH2-CH2-, H3C-CH2-CH(CH3)-, H3C-CH(CH3)-CH2-, H3C-C(CH3)2-, H3C-CH2-CH2-CH2-CH2-, H3C-CH2-CH2-CH(CH3)-, H3C-CH2-CH(CH3)-CH2-, H3C-CH(CH3)-CH2-CH2-, H3C-CH2-C(CH3)2-,
H3C-C(CH3)2-CH2-, H3C-CH(CH3)-CH(CH3)- and H3C-CH2-CH(CH2CH3)-.
The term "Ci_6-alkylene" wherein n is an integer 2 to 6, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to 6 carbon atoms. For example the term Ci_4-alkylene
includes -CH2-, -CH2-CH2-, -CH(CH3)-, -CH2-CH2-CH2-, -C(CH3)2-, -CH(CH2CH3)-, -CH (CH3)-CH2-, -CH2-CH(CH3)-, -CH2-CH2-CH2-CH2-, -CH2-CH2-CH(CH3)-, -CH(CH3)-CH2 -CH2-, -CH2-CH(CH3)-CH2-, -CH2-C(CH3)2-, -C(CH3)2-CH2-, -CH(CH3)-CH(CH3)-, -CH2- CH(CH2CH3)-, -CH(CH2CH3)-CH2-, -CH(CH2CH2CH3)- , -CH(CH(CH3))2- and -C(CH3)( CH2CH3)-.
The term "C2_n-alkylene" wherein n is an integer 3 to 5, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 2 to 5 carbon atoms. For example the term C2_5-alkylene
includes -CH2-CH2-, -CH(CH3)-, -CH2-CH2-CH2-, -C(CH3)2-, -CH(CH2CH3)-, -CH(CH3)- CH2-, -CH2-CH(CH3)-, -CH2-CH2-CH2-CH2-, -CH2-CH2-CH(CH3)-, -CH(CH3)-CH2-CH2-, -CH2-CH(CH3)-CH2-, -CH2-C(CH3)2-, -C(CH3)2-CH2-, -CH(CH3)-CH(CH3)-, -CH2-CH(C H2CH3)-, -CH(CH2CH3)-CH2-, -CH(CH2CH2CH3)- , -CH(CH(CH3))2- and -C(CH3)(CH2C ¾)-.
By the term "halo" added to a "alkyl", "alkylene" or "cycloalkyl" group (saturated or unsaturated) is such a alkyl or cycloalkyl group meant wherein one or more hydrogen atoms are replaced by a halogen atom selected from among fluorine, chlorine or bromine, preferably fluorine and chlorine, particularly preferred is fluorine. Examples include:
H2FC-, HF2C-, F3C-.
The term "C3_6-cycloalkyl", wherein n is an integer from 4 to 6, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to 6 C atoms. For example the term C3_6-cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "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.
With the elements of a ring the atoms forming this ring are meant. So, a phenyl ring contains 6 elements which are all carbon atoms, a pyrrol ring contains 5 elements, wherein 4 elements are carbon atoms and the remaining element is a nitrogen atom.
The term "aromatic heteroring" means a unsaturated monocyclic-ring system containing one, two, three or four heteroatoms selected from N, (0")N+, O or (0)rS, wherein r = 0, 1 or 2, consisting of four, five or six ring atoms. If the term is connected with a more detailed definition of the amount or kind of heteroatoms and the possible size of the aromatic heteroring, the detailed definition is restricting the above mentioned definition.
Furthermore the term is intended to include all possible isomeric forms. Thus, the term includes (if not otherwise restricted) the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
Figure imgf000030_0001
PREPARATION
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. Preferably, the compounds are obtained in analogous fashion to the methods of preparation explained more fully hereinafter, in particular as described in the experimental section. In some cases, 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.
Figure imgf000031_0001
Compounds according
VI
to the invention
Starting material I can be prepared as described in US2003/87940.
Intermediates II can be prepared as described in WO10133973 and US2003/87940 by heating starting material I with amines R-NH2 in the presence of a strong base, for example sodium tert-butoxide or sodium ethoxide, in an organic solvent, for example ethanol. The reaction usually takes place within 2 to 72 hours. Preferred reaction temperatures are between 50°C and 150°C.
The amide coupling (Step B, intermediates II intermediates III, intermediates IV intermediates V, intermediates VI compounds of the invention) can be achieved by reacting carboxylic acid intermediates II, IV or VI with amines R'-NH2 in the presence of an amide coupling reagent, for example 0-(Benzotriazol-l-yl)-N, N, Ν', N'- tetramethyluronium hexafluorophosphate (HBTU),0-(Benzotriazol-l-yl)-N, N, Ν', N'- tetramethyluronium tetrafluoroborate (TBTU) or propane phosphonic acid anhydride, and in the presence of a base, for example triethylamine, diisopropylethylamine (DIPEA, Hunig's base) or N-methyl-morpholine, in an organic solvent, for example N, N- dimethylformamide (DMF), N-methyl-2-pyrrolidone (NMP) or dimethylacetamide (DMA) or mixtures thereof. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and 50°C, most preferred room temperature.
Alternatively, the carboxylic acid intermediates can be activated first as described in US2003/87940, for example with 1, l '-carbonyldiimidazole (CDI) in DMF, followed by reaction with the amine R'-NH2.
The bromination (Step C, X=Br, intermediates II -7 intermediates IV, intermediates III - intermediates V) can be achieved by reacting intermediates II or III with bromination agents, for example bromine or N-bromosuccinimide, in an organic solvent, for example acetic acid, dichloromethane, methanol, acetonitrile, tetrahydrofuran or mixtures thereof. The iodination (Step C, X=I, intermediates II -7 intermediates IV, intermediates III -7 intermediates V) can be achieved by reacting intermediates II or III with iodination agents, for example iodine, iodinechloride (I-Cl) or N-iodosuccinimide, in an organic solvent, for example acetic acid, methanol, ethanol, dichloromethane, acetonitrile, N, N-dimethyl- formamide, tetrahydrofuran or mixtures therof. The halogenation reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and 50°C, most preferred room temperature.
The Suzuki coupling (Step D, intermediates IV -7 intermediates VI, intermediates V -7 compounds according to the invention) can be achieved by reacting intermediates IV or V with aryl or heteroaryl boronic acids R"-B(OH)2 or the corresponding boronic esters in the presence of a palladium catalyst, for example tetrakis(triphenylphosphine)palladium(0) or [1, -bis(diphenylphosphino)ferrocene]dichloropalladium(II), and in the presence of a base, for example, potassium carbonate, barium dihydroxide or cesium carbonate, in an organic solvent, for example toluene, benzene, ethanol, ethylene glycol dimethyl ether, acetonitrile, dioxane or mixtures thereof, optionally in the presence of water. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 50°C and 150°C. Compounds according to the present invention can also be prepared according to the following scheme starting from 4-hydroxy-6-methyl-nicotinic acid. Halogenation (Step C) as described above, followed by Suzuki coupling (Step D) as described above, followed by amide coupling (Step B) as described above, yields intermediates VII. The alkylation of the pyridone nitrogen (Step E) can be achieved by reacting intermediate VII with alkylating agents, for example alkyl bromides, alkyl iodides, alkyl tosylates, alkyl mesylates or dialkyl sulfates, in the presence of a base, for example sodium carbonate, potassium carbonate, potassium hydroxide, sodium hydroxide or cesium carbonate, in an organic solvent, N, N-dimethylformamide (DMF), N-methyl-2-pyrrolidone (NMP) or dimethylacetamide (DMA). The reaction usually takes place within 1 to 72 hours.
Preferred reaction temperatures are between 50°C and 150°C.
Figure imgf000033_0001
The intermediates for compounds according to the present invention with 2-alkyl substituents can be prepared according to Venkatramani et al., J. Het. Chem. 30, 723-738 (1993).
The synthesis of sulfinimides (sulfilimines) and sulfonimides (sulfoximines) has been extensively described in Reggelin et al, Synthesis, 1-64 (2000) and Bolm et al, Chem. Lett. 33, 482-487 (2004). The general processes for the preparation of N-cyano-sulfon- imides (N-cyano-sulfoximines) are illustrated in the following scheme. As indicated, the order of imination and oxidation can usually be interchanged, and the syntheses can likewise start from sulfoxides instead of sulfides.
K
Figure imgf000034_0001
Intermediates VIII can be prepared as described in Bolm et al, Org. Lett. 9, 3809-3811 (2007) via direct imination of a thioether (intermediates VII) with cyanamide (Step F, intermediates VII intermediates VIII) by reaction with an halogenating agent, for example N-bromo- or N-chlorosuccinimide, tert-butyl hypochlorite or iodine in the presence of cyanamide and a base, for example potassium or sodium tert-butoxide, in an organic solvent, for example methanol, tetrahydrofuran (THF) or acetonitrile. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0 °C and 50°C, most preferred is room temperature. Alternatively, the imination (Step F, intermediates VII intermediates VIII) can be achieved as described in Bolm et al., Org. Lett. 9, 2951-2954 (2007), by using phenyliodo diacetate [PhI(OAc)2] and cyanamide in an organic solvent, for example acetonitrile or tetrahydrofuran (THF). The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and room temperature.
The oxidation (Step G, intermediates VIII intermediates IX, intermediates XI intermediates XII) can be achieved using a peroxycarboxylic acid as oxidizing agent, for example meto-chloroperoxybenzoic acid (mCPBA), optionally in the presence of a base, for example potassium carbonate, in an organic solvent, for example ethanol, methanol, dichloromethane or chloroform. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and room temperature. Alternatively, other common oxidizing agents may be used to achieve this transformation, for example hydrogen peroxide, tert-butyl hydroperoxide, sodium hypochlorite, sodium iodate, sodium periodate, potassium permanganate, ruthenium tetroxide, potassium peroxymonosulfate (Oxone) or dimethyldioxirane.
Intermediates X can be obtained by imination of intermediates VII (Step H, intermediates VII -» intermediates X) as described in Bolm. et al, Org. Lett. 6, 1305-1307 (2004), by reacting a thioether (intermediates VII) with trifluoroacetamide, phenyliodo diacetate [PhI(OAc)2], rhodium acetate dimer [Rh2(OAc)4] and MgO in an organic solvent, for example dichloromethane. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and room temperature.
The cleavage of the trifluoroacetamide (Step K, intermediates X intermediates XI) can be achieved as described in Bolm. et al, Org. Lett. 6, 1305-1307 (2004), using a base, for example potassium carbonate, in an organic solvent, for example methanol. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and 50°C, most preferred room temperature.
Alternatively, intermediates XI can be prepared by the imination of intermediates VII using other electrophilic nitrogen sources, for example tert-butyl 3-(4-cyano-phenyl)- oxaziridine-2-carboxylate (followed by cleavage of the Boc-protecting group), O-mesityl sulfonyl hydroxylamine (MSH), or hydrazoic azid.
The cyanation of sulfinimides or sulfonimides (Step K, intermediates XI intermediates VIII, intermediates XII intermediates IX) can be achieved as described in Bolm et al., Org. Lett. 9, 2951-2954 (2007), by reacting the starting material with cyanogen bromide, optionally 4-dimethylaminopyridine (DMAP) and optionally a base, for example triethylamine, in an organic solvent, for example dichloromethane. The reaction usually takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and 50°C, most preferred room temperature. The synthesis of sulfonimidamides has been described in Johnson et al, J. Org. Chem. 44, 2055-2061 (1975), Bolm et al, J. Org. Chem. 75, 3301-3310 (2010) and in WO09156336. The general synthetic scheme for the preparation of sulfonimidamides from sulfmamides is depicted in the following scheme.
Figure imgf000036_0001
XV XVI
Sulfmamides (starting material XIII) can be prepared as described in Johnson et al., J. Org Chem. 44, 2055-2061 (1975) by reacting sulfmyl chlorides with a primary or secondary amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example dichloromethane, tetrahydrofuran, diethylether, acetonitrile, toluene, N,N-dimethylformamide, ethanol or ethyl acetate. The reaction usually takes place within 1-72 hours. Preferred reaction temperatures are between 0 °C and room temperature.
Sulfonimidoyl chlorides (intermediates XIV) can be prepared as described in Johnson et al, J. Org. Chem. 44, 2055-2061 (1975), by reacting sulfinamides (starting material XIII) with a chlorinating agent, for example chlorine, N-chlorosuccinimide (NCS), N-chloro- benzotriazole or tert-butyl hypochlorite in an organic solvent, for example dichloromethane, diethylether, benzene, toluene, carbon tetrachloride, acetonitrile or tetrahydrofuran (Step L). The reaction usually takes place within less than 1 hour. Preferred reaction temperatures are between -78 °C and room temperature, mostly preferred between 0 °C and room temperature. Sulfonimidoyl fluorides (intermediates XV) can be prepared as described in Johnson et al, J. Org. Chem. 48, 1-3 (1983) and Gnamm et al, Bioorg. Med. Chem. Lett. 12, 3800-3806 (2012), by reacting sulfonimidoyl chlorides (intermediates XIV) with a fluoride salt, for example potassium fluoride, sodium fluoride or tetrabutylammonium fluoride, optionally in the presence of a crown ether, for example 18-crown-6, in an organic solvent, for example acetonitrile or tetrahydrofuran (Step M). The reaction takes place within 1 to 24 hours. Preferred reaction temperatures are between 0 °C and room temperature, mostly preferred room temperature.
Sulfonimidamides (intermediates XVI) can be prepared as described in Johnson et al, J. Org. Chem. 44, 2055-2061 (1975), Bolm et al, J. Org. Chem. 75, 3301-3310 (2010), and WO09156336 by reacting sulfonimidoyl chlorides (intermediates XIV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step N). The reaction takes place within 1 to 72 hours. Preferred reaction temperatures are between 0 °C and room temperature.
Alternatively, sulfonimidamides (intermediates XVI) can be prepared as described in Gnamm et al, Bioorg. Med. Chem. Lett. 22, 3800-3806 (2012) by reacting sulfonimidoyl fluorides (intermediates XV) with a primary or secondary, aliphatic or aromatic amine, optionally in the presence of an additional base, for example pyridine or a tertiary amine, for example l,8-diazabicyclo[5.4.0]undec-7-ene (DBU), in an organic solvent, for example acetonitrile, tetrahydrofuran, N,N-dimethylformamide or benzene (Step O). The reaction takes place within 1 to 72 hours. Preferred reaction temperatures are between 0°C and the boiling point of the solvent, mostly preferred between room temperature and 110°C.
Preliminary remarks: Ή-NMR have been obtained for the examples of the invention. The HPLC data given are measured under the following conditions (r.t. = retention time):
Figure imgf000038_0001
Figure imgf000038_0003
Figure imgf000038_0002
Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000039_0003
Figure imgf000039_0004
Method Name: Z002 002
Column: Sunfire CI 8, 3 x 30 mm, 2.5 μιη
Column Supplier: Waters
Figure imgf000040_0001
Figure imgf000040_0003
Figure imgf000040_0002
Figure imgf000041_0001
Figure imgf000041_0002
Figure imgf000041_0003
Figure imgf000041_0004
Method Name: K011 S03
Figure imgf000042_0001
Figure imgf000042_0002
Figure imgf000042_0003
Method Name: LCMS-FA-8
Figure imgf000043_0001
Preparation 1 : 5-Bromo- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3- carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000044_0001
1 a: 3-Dimethylaminomethylene-6-methyl-pyran-2,4-dione
Figure imgf000044_0002
To a solution of 4-hydroxy-6-methyl-2-pyrone (1 1.50 g, 91.2 mmol) in toluene (30 mL) is added N, N-dimethylformamide dimethyl acetal (13.00 mL, 97.9 mmol). After stirring for 2 h at room temperature, the reaction mixture is evaporated under reduced pressure and co- evaporated with toluene several times. Yield: 18.5 g; ESI mass spectrum: [M+H]+ = 182; r.t. HPLC: 0.72 min (Z002 007). lb: l-Isopropyl-6-methyl-4-oxo-l ,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000044_0003
A solution of 3-dimethylaminomethylene-6-methyl-pyran-2,4-dione (preparation la, 10.00 g, 38.6 mmol based on 70% purity), isopropylamine (5.00 mL, 58.4 mmol) and sodium tert-butoxide (5.50 g, 57.2 mmol) in ethanol (20 mL) is heated for 18 h at 90°C. The reaction mixture is evaporated under reduced pressure, treated with water and extracted with dichloromethane. The aqueous layer is acidified with 4 N aqueous HCl and extracted with dichloromethane. The combined organic layer is washed with water, dried over Na2S04 and evaporated under reduced pressure. Yield: 7.16 g; ESI mass spectrum:
[M+H]+ = 196; r.t. HPLC: 0.71 min (Z002 006).
1 c: 5-Bromo- 1 -isopropyl-6-methyl- -oxo- 1 ,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000045_0001
To a solution of l-isopropyl-6-methyl-4-oxo-l ,4-dihydro-pyridine-3-carboxylic acid (preparation lb, 1.50 g, 6.92 mmol based on 90% purity) in glacial acetic acid (10 ml) is added at room temperature bromine (0.60 mL, 1 1.7 mmol). After stirring for 3 d at room temperature, additional bromine (1.00 mL, 19.5 mmol) is added to the reaction mixture and stirring is continued for 2 h at room temperature. The reaction mixture is diluted with water. Upon addition of dichloromethane a precipitate forms which is filtered off and dried. Yield: 2.55 g; ESI mass spectrum: [M+H]+ = 274 (bromine isotope pattern); r.t. HPLC: 0.76 min (Z002 002).
Id: 5-Bromo-l-isopropyl-6-methyl-4-oxo-l ,4-dihydro-pyridine-3-carboxylic acid 4-(N- cyano-S-methylsulfonimidoy^-benzylamide
0 HN A solution of 5-bromo-l-isopropyl-6-methyl-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid (preparation lc, 270 mg, 0.99 mmol), TBTU (316 mg, 0.99 mmol) and DIPEA (513 μί, 2.97 mmol) in DMF (3.00 mL) is stirred for 10 min at room temperature. Then, a solution of (4-(N-cyano-5'-methylsulfonimidoyl)phenyl)methanamine (preparation 6, 247 mg, 1.18 mmol) in DMF (1.00 mL) is added and the reaction mixture is stirred for 18 h at room temperature. The reaction mixture is purified by preparative reversed phase HPLC
(Sunfire, gradient of acetonitrile in water, 0.1% TFA, 60°C). Yield: 70 mg (15% of theory); ESI mass spectrum: [M+H]+ = 465 (bromine isotope pattern); r.t. HPLC: 0.86 min (Z017 S04).
Preparation 2 : 5-Bromo- l-ethyl-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
2a: 1 -Ethyl-6-methyl-4-oxo- 1 ,4- xylic acid
Figure imgf000046_0001
Preparation 2a is prepared following the procedure described for preparation lb, substituting isopropylamine with ethylamine. ESI mass spectrum: [M+H]+ = 182; r.t. HPLC: 0.62 min (Z002 006). 2b: 5-Bromo- 1 -ethyl-6-methyl-4-o - 1 ,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000047_0001
Preparation 2b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 2a as starting material. ESI mass spectrum: 5 [M+H]+ = 260 (bromine isotope pattern); r.t. HPLC: 0.89 min (Z002 007).
2c: 5-Bromo- l-ethyl-6-methyl-4-oxo-l ,4-dihydro-pyridine-3-carboxylic acid 4-(N-cyano- S-methylsulfonimidoy^-benzylamide
Figure imgf000047_0002
10 Preparation 2c is prepared following the procedure described for preparation Id,
substituting preparation lc with preparation 2b as starting material and substituting TBTU with HBTU as coupling reagent. ESI mass spectrum: [M+H] = 451 (bromine isotope pattern); r.t. HPLC: 0.81 min (Z012 S04). i s Preparation 3: 5-Bromo-l-cyclobutyl-6-methyl-4-oxo-l,4-dihydro-pyridine-3- carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000048_0001
3a: 1 -Cyclobutyl-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000048_0002
Preparation 3a is prepared following the procedure described for preparation lb, substituting isopropylamine with cyclopropylamine. ESI mass spectrum: [M+H]+ = 208; r.t. HPLC: 0.62 min (Z002 002).
3b: 5-Bromo-l-cyclobutyl-6-methy -4-oxo-l,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000048_0003
Preparation 3b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 3a as starting material. ESI mass spectrum: [M+H]+ = 286 (bromine isotope pattern); r.t. HPLC: 0.82 min (Z002 002). 3c: 5-Bromo-l-cyclobutyl-6-methyl-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid 4-(N- cyano-S-methylsulfonimidoy^-benzylamide
Figure imgf000049_0001
Preparation 3c is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 3b as starting material and substituting TBTU with HBTU as coupling reagent. ESI mass spectrum: [M+H]+ = 477 (bromine isotope pattern); r.t. HPLC: 0.90 min (Z018 S04).
Preparation 4 : 5-Bromo-6-methyl- l-(2-methyl-2H-pyrazol-3-yl)-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000049_0002
4a: 6-Methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000050_0001
Preparation 4a is prepared following the procedure described for preparation lb, substituting isopropylamine with l-methyl-lH-pyrazol-5-ylamine. ESI mass spectrum: [M+H]+ = 234; r.t. HPLC: 0.63 min (Z018 S04).
4b: 6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-l ,4-dihydro-pyridine-3-carboxylic acid 4-(N-cyano-5'-methylsulfonimidoyl -benzylamide
Figure imgf000050_0002
Preparation 4b is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 4a as starting material and substituting TBTU with HBTU as coupling reagent. ESI mass spectrum: [M+H]+ = 425; r.t. HPLC: 0.79 min (Z018 S04).
4c: 5-Bromo-6-methyl- 1 -(2-methyl-2H-pyrazol-3-yl)-4-oxo- 1 ,4-dihydro-pyridine-3- carboxylic acid 4-(N-cyano-5'-methylsulfonimidoyl)-benzylamide
Figure imgf000051_0001
To a solution of 6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-l,4-dihydro-pyridine-3- carboxylic acid 4-(N-cyano-5'-methylsulfonimidoyl)-benzylamide (preparation 4b, 150 mg, 0.1 18 mmol) in dichloromethane (2 mL) is added N-bromosuccinimide (21 mg, 0.35 mmol) and five drops of glacial acetic acid. After stirring for 18 h at room temperature the reaction mixture is concentrated under reduced pressure and purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1% TFA, 60°C). Yield: 40 mg (23% of theory); ESI mass spectrum: [M+H]+ = 503 (bromine isotope pattern); r.t. HPLC: 0.82 min (Z012 S04).
Preparation 5: l-(4-Cyano-benzyl)-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4- dihydro-pyridine-3-carboxylic acid
Figure imgf000051_0002
5a: 1 -(4-Cyano-benzyl)-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000052_0001
Preparation 5a is prepared following the procedure described for preparation lb, substituting isopropylamine with 4-cyanobenzylamine. ESI mass spectrum: [M+H]+ = 269; r.t. HPLC: 0.86 min (Z002 005).
5b: 5-Bromo-l-(4-cyano-benzyl -6-methyl-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid
Figure imgf000052_0002
Preparation 5b is prepared following the procedure described for preparation lc, substituting preparation lb with preparation 5a as starting material. ESI mass spectrum: [M+H]+ = 347 (bromine isotope pattern); r.t. HPLC: 1.07 min (Z002 005).
5c: 1 -(4-Cyano-benzyl)-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro- pyridine-3-carboxylic acid
Figure imgf000053_0001
A mixture of 5-bromo-l-(4-cyano-benzyl)-6-methyl-4-oxo-l,4-dihydro-pyridine-3- carboxylic acid (preparation 5b, 2.55 g, 7.34 mmol), 3-(trifluoromethyl)phenylboronic acid (1.65 g, 8.69 mmol), l, -[bis(diphenylphosphino)ferrocene]dichloropalladium(II) (340 mg, 0.47 mmol) and CS2CO3 (4.20 g, 12.9 mmol) in dioxane (20.0 mL) is heated for 72 h at 80°C. The reaction mixture is filtered over silica and the filtrate is concentrated under reduced pressure and purified by preparative reversed phase HPLC (first purification: Sunfire, gradient of methanol in water, 0.1% TFA; second purification: Xbridge, gradient of methanol in water, 0.1% NH4OH, 60°C). Yield: 514 mg (17 % of theory); ESI mass spectrum: [M+H]+ = 413; r.t. HPLC: 1.37 min (Z002 005).
Preparation 6: (4-( V-cyano-S-methylsulfonimidoyl)phenyl)methanamine
Figure imgf000053_0002
A mixture of 4-(methylthio)benzylamine (2.00 mL, 2.20 g, 14.38 mmol) and phthalic anhydride (2.39 g, 15.79 mmol) in acetic acid (40 mL, 42 g, 700 mmol) is heated at reflux for 3 h. The mixture is concentrated under reduced pressure, and the residue is partitioned between water and dichloromethane. The phases are separated, and the aqueous phase is extracted twice with dichloromethane. The combined organic layers are concentrated under reduced pressure and the residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 9: 1 to 7:3). Yield: 3.60 g (88% of theory); ESI mass spectrum: [M+H]+ = 284; r.t. HPLC: 0.75 min (X001 004).
6b: 2-(4-(N-cyano-5,-methylsulfinimidoyl)benzyl)isoindoline-l ,3-dione
Figure imgf000054_0001
A solution of 2-(4-(methylthio)benzyl)isoindoline- l ,3-dione (preparation 6a, 3.60 g, 12.71 mmol), cyanamide (694 mg, 16.5 mmol) and potassium tert-butoxide (1.71 g, 15.25 mmol) in methanol (30 mL) is treated with N-bromosuccinimide (3.39 g,
19.06 mmol). The mixture is stirred for 1 h and concentrated under reduced pressure. Saturated aqueous Na2S2C"3 solution is added and the mixture is extracted twice with dichloromethane. The combined organic layers are dried over Na2S04 and concentrated under reduced pressure. The residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 1 : 1 to ethyl acetate to ethyl acetate/methanol 4: 1). Yield: 2.43 g (59 % of theory); ESI mass spectrum: [M+H]+ = 324; r.t. HPLC: 0.52 min (X001 004).
6c: 2-(4-(N-cyano-5'-methylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000054_0002
A solution of 2-(4-(N-cyano-5,-methylsulfinimidoyl)benzyl)isoindoline-l ,3-dione
(preparation 6b, 5.75 g, 17.78 mmol) and potassium carbonate (7.37 g, 53.34 mmol) in ethanol (130 mL) is cooled at 0 °C and treated with meto-chloroperoxybenzoic acid (mCPBA, 5.98 g, 26.67 mmol). The reaction mixture is warmed to room temperature and stirred overnight before more mCPBA (2.39 g, 10.66 mmol) is added and the mixture is stirred for 4 h. Saturated aqueous Na2S2C"3 solution is added, and the mixture is extracted twice with dichloromethane. The combined organic layers are dried over Na2S04 and concentrated under reduced pressure. The residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 1 : 1 to ethyl acetate). Yield: 2.47 g (41 % of theory); ESI mass spectrum: [M+H]+ = 334; r.t. HPLC: 1.02 min (V003 003).
6d: (4-(N-cyano-5'-methylsulfonimidoyl)phenyl)methanamine
Figure imgf000055_0001
A solution of 2-(4-(N-cyano-5'-methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione
(preparation 6c, 684 mg, 2.02 mmol) and 1 ,2-diaminoethane (243 μί, 219 mg, 3.64 mmol) in a mixture of acetonitrile (8 mL), tetrahydrofuran (4 mL) and ethanol (4 mL) is heated at 60 °C for 5 h. All volatiles are removed under reduced pressure and the residue is suspended in dichloromethane. The mixture is filtered and the filtrate is concentrated under reduced pressure. Yield: 326 mg (77% of theory); ESI mass spectrum: [M+H]+ = 210; r.t. HPLC: 0.223 min (V001 007).
Example 1: 5-(3-Difluoromethyl-phenyl)-l-isopropyl-6-methyl-4-oxo-l,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000055_0002
To a solution of preparation 1 (70 mg, 0.15 mmol), 3-(difluoromethyl)phenylboronic acid (36 mg, 0.21 mmol) and 1, -[bis(diphenylphosphino)ferrocene]dichloropalladium(II) (11 mg, 0.015 mmol) in acetonitrile (2.00 mL) is added aqueous K2CO3 solution (2 M, 0.155 mL, 0.31 mmol). After stirring for 18 h at 75°C, the reaction mixture is filtered and the filtrate is purified by preparative reversed phase HPLC (XBridge, gradient of methanol in water, 0.1 % NH4OH, 60°C). Yield: 32 mg (42 % of theory); ESI mass spectrum: [M+H]+ = 513; r.t. HPLC: 0.90 min (Z003 001).
Example 1A and Example IB: Enantiomers of Example 1
63 mg of racemic example 1 are separated by chiral HPLC (Daicel IB, 250mm x 20mm, 15% MeOH + 0.2% diethylamine in supercritical C02, 40°C).
Early eluting enantiomer (Example 1A): r.t. chiral HPLC = 5.10 min (Daicel
Chiralcel®OZ-H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2%
diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum:
[M+H]+ = 513; Yield: 23 mg
Late eluting enantiomer (Example IB): r.t. chiral HPLC = 7.80 min (Daicel Chiralcel®OZ- H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum: [M+H]+ = 513; Yield: 20 mg
Example 2: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000057_0001
Example 2 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid.
ESI mass spectrum: [M+H]+ = 531; r.t. HPLC: 0.99 min (Z003 001).
Example 2 A and Example 2B: Enantiomers of Example 2
100 mg of racemic example 2 are separated by chiral HPLC (Daicel OZ-H, 250mm x 20mm, 40% MeOH + 0.2% diethylamine in supercritical C02, 40°C).
Early eluting enantiomer (Example 2A): r.t. chiral HPLC = 3.62 min (Daicel
Chiralcel®OZ-H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2%
diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum:
[M+H]+ = 531; Yield: 41 mg
Late eluting enantiomer (Example 2B): r.t. chiral HPLC = 5.23 min (Daicel Chiralcel®OZ- H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum: [M+H]+ = 531; Yield: 43 mg.
Example 3 : l-Isopropyl-2-methyl-4-oxo-2 '-trifluor omethyl- 1 ,4-dihydro- [3,4 '] - bipyridinyl-5-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000058_0001
Example 3 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid.
ESI mass spectrum: [M+H]+ = 532; r.t. HPLC: 0.86 min (Z003 001).
Example 3 A and Example 3B: Enantiomers of Example 3
50 mg of racemic example 3 are separated by chiral HPLC (Daicel IB, 250mm x 20mm, 15% MeOH + 0.2% diethylamine in supercritical C02, 40°C).
Early eluting enantiomer (Example 3 A): r.t. chiral HPLC = 2.81 min (Daicel
Chiralcel®OZ-H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2%
diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum:
[M+H]+ = 532; Yield: 23 mg.
Late eluting enantiomer (Example 3B): r.t. chiral HPLC = 4.25 min (Daicel Chiralcel®OZ- H 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 40% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 100 bar back pressure); ESI mass spectrum: [M+H]+ = 532; Yield: 20 mg.
Example 4: l-Ethyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000059_0001
Example 4 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 2-(trifluoromethyl)pyridine-4-boronic acid and substituting preparation 1 with preparation 2 as starting material. ESI mass spectrum:
[M+H]+ = 517; r.t. HPLC: 0.85 min (Z011 S03).
Example 4 A and Example 4B: Enantiomers of Example 4
30 mg of racemic example 4 are separated by chiral HPLC (Daicel OZH, 250mm x 20mm, 40% MeOH + 0.2% diethylamine in supercritical C02, 40°C).
Early eluting enantiomer (Example 4A): r.t. chiral HPLC = 5.08 min (Daicel Chiralpak®IB, 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 20% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 150 bar back pressure); ESI mass spectrum: [M+H]+ = 517; Yield: 11 mg.
Late eluting enantiomer (Example 4B): r.t. chiral HPLC = 5.47 min (Daicel Chiralpak®IB, 4.6 mm x 250mm, 5μιη, 4ml/min, lOmin, 20% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 150 bar back pressure); ESI mass spectrum: [M+H]+ = 517; Yield: 9 mg.
Example 5 : 5-(3-Difluoromethyl-phenyl)- l-ethyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000060_0001
Example 5 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 2 as starting material. ESI mass spectrum: [M+H]+ = 499; r.t. HPLC: 0.79 min (Z01 1 S03).
Example 6 : 5-(3-Difluoromethyl-phenyl)- l-cyclobutyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( -cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000060_0002
Example 6 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 3 as starting material. ESI mass spectrum: [M+H]+ = 525; r.t. HPLC: 0.84 min (Z01 1 S03). Example 7: 5-(3-Difluoromethyl-phenyl)-6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4- oxo-1, 4-dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)- benzylamide
Figure imgf000061_0001
Example 7 is prepared following the procedure described for example 1 , substituting preparation 1 with preparation 4 as starting material. ESI mass spectrum: [M+H]+ = 551; r.t. HPLC: 0.96 min (Z018 S04).
Example 8: 6-Methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-5-(3-trifluoromethyl- phenyl)- 1, 4-dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)- benzylamide
Figure imgf000061_0002
Example 8 is prepared following the procedure described for example 1, substituting 3- (difluoromethyl)phenylboronic acid with 3-(trifluoromethyl)phenylboronic acid and substituting preparation 1 with preparation 4 as starting material. ESI mass spectrum: [M+H]+ = 569; r.t. HPLC: 1.00 min (Z018 S04).
Example 9: l-(4-Cyano-benzyl)-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4- dihydro-pyridine-3-carboxylic acid 4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000062_0001
Example 9 is prepared following the procedure described for preparation Id, substituting preparation lc with preparation 5 as starting material. ESI mass spectrum: [M+H]+ = 604; r.t. HPLC: 0.63 min (X012 S01).
Example 9 A and Example 9B: Enantiomers of Example 9
58 mg of racemic example 9 are separated by chiral HPLC (Daicel ASH, 250mm x 20mm, 20% MeOH + 0.2% diethylamine in supercritical C02, 40°C).
Early eluting enantiomer (Example 9A): r.t. chiral HPLC = 5.76 min (Daicel
Chiralpak®ASH, 250mm x 4.6mm, 5 μιη, 4ml/min, lOmin, 20% MeOH + 0.2%
diethylamine in supercritical C02, 40°C, 150 bar back pressure); ESI mass spectrum: [M+H]+ = 604; Yield: 8 mg Late eluting enantiomer (Example 9B): r.t. chiral HPLC = 6.86 min (Daicel
Chiralpak®ASH, 250mm x 4.6mm, 5 μιη, 4ml/min, lOmin, 20% MeOH + 0.2% diethylamine in supercritical C02, 40°C, 150 bar back pressure); ESI mass spectrum: [M+H]+ = 604; Yield: 7 mg
The following intermediates are prepared as described for preparation lb, substituting isopropylamine with the appropriate amine.
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
The following bromo intermediates are prepared as described for preparation lc, employing the appropriate des-bromo intermediates as starting material.
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Preparation 8.20: 5-Bromo-6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4- dihydro-pyridine-3-carboxylic acid methyl ester
Figure imgf000072_0002
8.20a 6-Methyl- l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-l,4-dihydro-pyridine-3-carboxylic acid methyl ester
Figure imgf000073_0001
To a solution of preparation 4a (1.8 g, 90% purity, 6.95 mmol) in methanol (5 mL) is added slowly thionyl chloride (1.64 g, 13.8 mmol) at 0°C. The reaction mixture is stirred at 70°C for 1.5 h, concentrated under reduced pressure and purified by reversed-phase HPLC (Gilson: Sunfire, 30°C, gradient of acetonitrile in water + 0.1% formic acid). Yield: 199 mg (12% of theory). ESI mass spectrum: [M+H]+ = 248; r.t. HPLC: 0.56 min (Z018 S04).
8.20b 5-Bromo-6-methyl-l-(2-methyl-2H-pyrazol-3-yl)-4-oxo-l,4-dihydro-pyridine-3- carboxylic acid methyl ester
Figure imgf000073_0002
The title compound is prepared as described for preparation 4c, substituting preparation 4b with preparation 8.20a as starting material. ESI mass spectrum: [M+H]+ = 326 (bromine pattern); r.t. HPLC: 0.69 min (Z012 S04). The following intermediates are prepared as described for Example 2, substituting preparation 1 with the appropriate bromo intermediate as starting material.
Starting MS r.t.
Preparation Structure
Material [M+H]+ HPLC Method
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
The following intermediates are prepared as described for Example 1, substituting preparation 1 with the appropriate bromo intermediate as starting material.
Figure imgf000076_0002
Figure imgf000077_0001
The following intermediates are prepared as described for Example 1, substituting preparation 1 with preparation lc and employing the appropriate boronic acid or ester in the Suzuki reaction.
Figure imgf000078_0001
The following intermediates are prepared as described for Example 3, substituting preparation 1 with the appropriate bromo intermediate as starting material.
Starting MS r.t.
Preparation Structure
Material [M+H]+ HPLC Method
Figure imgf000079_0001
The following intermediates are prepared as described for preparation Id, substituting preparation lc with the appropriate starting material acid and substituting racemic preparation 6 with enantiomerically pure preparation 12B. The asterisk in the structures indicates that the compound is prepared from the enantiomer preparation 12B.
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Preparation 11A and Preparation 11B: Enantiomers of preparation 6c
Racemic preparation 6c (2.0 g) is separated by chiral HPLC (Daicel Chiralcel OZ-H, 250 mm x 30 mm, 32% MeOH in supercritical C02).
Early eluting enantiomer (preparation 1 1A): r.t. chiral HPLC = 10.19 min (Daicel
Chiralpak® IA 4.6 mm x 250 mm x 5 μιη, 1 mL/min, 20 min, 70% hexane + 15% isopropanol + 15% dichloromethane + 0.1% TFA, 20 °C); ESI mass spectrum: [M+H]+ = 334; Yield: 600 mg.
Late eluting enantiomer (preparation 1 IB): r.t. chiral HPLC = 10.70 min (Daicel
Chiralpak® IA 4.6 mm x 250 mm x 5 μιη, 1 mL/min, 20 min, 70% hexane + 15% isopropanol + 15% dichloromethane + 0.1% TFA, 20 °C); ESI mass spectrum: [M+H]+ = 334; Yield: 700 mg.
Preparation 12A: Early eluting enantiomer of preparation 6d
The early eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-5'-methyl- sulfonimidoyl)benzyl)isoindoline-l ,3-dione with preparation 1 1 A.
r.t. chiral HPLC = 2.92 min (Daicel Chiralpak® IC 4.6 mm x 250 mm 5μιη, 4 ml min, 10 min, 30%) MeOH + 0.2%> diethylamine in supercritical C02, 40 °C, 100 bar back pressure); ESI mass spectrum: [M+H]+ = 210. Preparation 12B: Late eluting enantiomer of preparation 6d
The late eluting enantiomer of racemic preparation 6d is prepared following the procedure described for preparation 6d, substituting racemic 2-(4-(N-cyano-S'-methylsulfonimidoyl)- benzyl)isoindoline-l ,3-dione with preparation 1 IB.
r.t. chiral HPLC = 3.59 min (Daicel Chiralpak® IC 4.6 mm x 250 mm 5μιη, 4 ml/min, 10 min, 30% MeOH + 0.2% diethylamine in supercritical C02, 40 °C, 100 bar back pressure); ESI mass spectrum: [M+H]+ = 210.
Preparation 13: (4-(N-Cyano-S-eth lsulfonimidoyl)phenyl)methanamine
Figure imgf000084_0001
13a: 2-(4-(N-cyano-5'-ethylsulfinimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000084_0002
Phthalic anhydride (4.42 g, 30 mmol) is added to a solution of 4-(ethylthio)benzylamine (5.00 g, 30 mmol) in acetic acid. The mixture is heated at reflux for 5 h and cooled at room temperature. Water ist added, and the precipitate is filtered and dried.
The residue is dissolved in methanol (50 mL), and the mixture is treated with potassium tert-butoxide (2.26 g, 20 mmol) and cyanamide (920 mg, 22 mmol). N-Bromosuccinimide (4.49 g, 25 mmol) is added, and the mixture is stirred at room temperature for 2 h. The mixture is concentrated under reduced pressure and treated with saturated aqueous Na2S203 solution. The mixture is extracted four times with dichloromethane, and the combined organic layers are dried over Na2S04 and concentrated under reduced pressure. The residue is purified by flash chromatography on silica (dichloromethane/methanol 95 :5). Yield: 3.0g (30% of theory); ESI mass spectrum: [M+H]+ = 338; r.t. HPLC: 3.14 min (LCMS-FA-8). 13b: 2-(4-(N-cyano-5'-ethylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000085_0001
The title compound is prepared in analogy to preparation 6c, substituting 2-(4-(N-cyano-lS- methylsulfinimidoyl)benzyl)isoindoline- 1 ,3-dione with 2-(4-(N-cyano-5'- ethylsulfinimidoyl)benzyl)isoindoline-l ,3-dione (preparation 13a). ESI mass spectrum: [M+H]+ = 354; r.t. HPLC: 3.33 min (LCMS-FA-8).
13c: (4-(N-Cyano-5'-ethylsulfonimido l)phenyl)methanamine
Figure imgf000085_0002
The title compound is prepared in analogy to preparation 6d, substituting 2-(4-(N-cyano-lS- methylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione with 2-(4-(N-cyano-5'- ethylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (preparation 13b). ESI mass spectrum: [M+H]+ = 224; r.t. HPLC: 0.52 min (Z01 1 S03).
Preparation 14: (4-( V-Cyano-S-iso ropylsulfonimidoyl)phenyl)methanamine
Figure imgf000085_0003
14a: 2-(4-(N-cyano-5'-isopropylsulfinimidoyl)benzyl)isoindoline-l ,3-dione
Figure imgf000085_0004
The title compound is prepared in analogy to preparation 13 a, substituting 4-(ethylthio)- benzylamine with 4-(isopropylthio)benzylamine. ESI mass spectrum: [M+H]+ = 352; r.t. HPLC: 0.50 min (X012 S01). 14b: 2-(4-(N-cyano-5'-isopropylsulfonimidoyl)benzyl)isoindoline-l ,3-dione
Figure imgf000086_0001
The title compound is prepared in analogy to preparation 6c, substituting 2-(4-(N-cyano-lS- methylsulfinimidoyl)benzyl)isoindoline- 1 ,3-dione with 2-(4-(N-cyano-5'- isopropylsulfinimidoyl)benzyl)isoindoline-l ,3-dione (preparation 14a). ESI mass spectrum: [M+H]+ = 368; r.t. HPLC: 3.42 min (LCMS-FA-8).
14c: (4-(N-Cyano-5'-isopropylsulfonimido l)phenyl)methanamine
Figure imgf000086_0002
The title compound is prepared in analogy to preparation 6d, substituting
2-(4-(N-cyano-5'-methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione with 2-(4-(N-cyano-5'- isopropylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (preparation 14b). ESI mass spectrum: [M+H]+ = 238; r.t. HPLC: 0.59 min (Z01 1 S03).
Preparation 15: 3-Fluoro-4-( -cyano-S-methylsulfonimidoyl)benzylamine
Figure imgf000086_0003
15a: 2-(3-Fluoro-4-methylsulfanyl-benzyl)-isoindole- 1 ,3-dione
Figure imgf000087_0001
A mixture of 3-fluoro-4-methylsulfanyl-benzonitrile (6.00 g, 34.1 mmol), Raney-Nickel (100 mg) and concentrated aqueous ammonia (30 mL) in methanol (300 mL) is treated with hydrogen (3.4 bar) at room temperature for 3 h. The mixture is filtered through a pad of silica gel, and the filtrate is concentrated under reduced pressure.
The residue is dissolved in toluene (250 mL), treated with phthalic anhydride (4.0 g, 26.3 mmol) and triethylamine (0.98 g, 9.50 mmol), and the mixture is heated at reflux over night. All volatiles are removed, and the residue is recrystallized from ethanol. Yield: 4.8 g (46 % of theory). ESI mass spectrum: [M+H]+ = 302; r.t. HPLC: 0.67 min (X012 S01).
15b: 2-(3-Fluoro-4-(N-cyano-5'-methylsulfinimidoyl)-benzyl)-isoindole-l ,3-dione
Figure imgf000087_0002
Cyanamide (350 mg, 8.20 mmol) and potassium tert-butoxide (860 mg, 7.60 mmol) are added to a solution of 2-(3-fluoro-4-methylsulfanyl-benzyl)-isoindole-l ,3-dione
(preparation 15a, 2.00g, 6.31 mmol) in methanol (50 mL). N-Bromosuccinimide (1.70 g, 9.50 mmol) is added and the mixture is stirred at room temperature over night. All volatiles are removed under reduced pressure, and the residue is treated with dichloromethane (20 mL). The mixture is washed with saturated aqueous sodium chloride solution, dried over Na2S04 and concentrated under reduced pressure. The residue is directly used in the next step without further purification. Yield: 1.61 g (60% of theory); ESI mass spectrum: [M+H]+ = 342; r.t. HPLC: 0.76 min (5-95 AB).
15c: 2-(3-Fluoro-4-(N-cyano-5'-methylsulfonimidoyl)-benzyl)-iso indole- 1 ,3-dione
Figure imgf000088_0001
2-(3-Fluoro-4-(N-cyano-5'-methylsulfinimidoyl)-benzyl)-isoindole-l ,3-dione
(preparationl5b, 1.60 g, 4.22 mmol) and acetic acid (0.25 g, 4.22 mmol) are added to a solution of potassium permanganate (670 mg, 4.22 mmol) in a mixture of water (8 mL) and acetonitrile (9.6 ml). The mixture is stirred over night, and saturated aqueous Na2S203 solution is added. The mixture is extracted with ethyl acetate and the organic layer is dried over Na2S04. The residue is directly used in the next step without further purification. Yield: 1.34 g (80% of theory); ESI mass spectrum: [M+H]+ = 358; r.t. HPLC: 0.73 min (5- 95AB).
15d: 3-Fluoro-4-(N-cyano-5'-methylsulfonimidoyl)benzylamine
Figure imgf000088_0002
Hydrazine (85% in water, 2.0 g, 53 mmol) is added to a solution of 2-(3-fluoro-4-(N- cyano-^-methylsulfonimidoy^-benzy^-isoindole-l ^-dione (preparation 15c, 3.5 g, 8.8 mmol) in a 1 : 1 mixture of dichloromethane and methanol (140 mL). The mixture is stirred over night and concentrated under reduced pressure. The residue is purified by preparative reversed phase HPLC (Luna C I 8(2), gradient of acetonitrile in water, 0.01 M N¾). Yield: 1.2 g (58% of theory); ESI mass spectrum: [M+H]+ = 228; r.t. HPLC: 0.26 min
(X01 1 S03).
Preparation 16: (5-( V-Cyano-S-methylsulfonimidoyl)pyridin-2-yl)methanamine
Figure imgf000088_0003
16a: 5-(Methylthio)pyridin-2-yl)methylamine
Figure imgf000089_0001
A solution of 5-methylsulfanyl-pyridine-2-carbonitrile (preparation described in
WO05026124, 4.28 g, 28.5 mmol) in THF is cooled at -78 °C in a dry ice / ethanol bath. Lithium aluminium hydride (2.4 M in THF, 12 mL, 28.8 mmol) is added dropwise. After 10 min water (1 mL) and THF (3 mL) are added. After 5 min, aqueous sodium hydroxide solution (4 M, 1 mL) and water (3 mL) is added. After 10 min the mixture is concentrated under reduced pressure, and the residue is dissolved in a mixture of water, methanol and acetic acid and purified by preparative reversed phase HPLC (Sunfire, gradient of acetonitrile in water, 0.1 % formic acid). Yield: 1.36 g (31% of theory); ESI mass spectrum: [M+H]+ = 155; r.t. HPLC: 0.44 min (Z018 S04).
16b: 2-(5-Methylsulfanyl-pyridin-2-ylmethyl)-isoindole- 1 ,3-dione
Figure imgf000089_0002
The title compound is prepared in analogy to preparation 6a, substituting 4-(methylthio)- benzylamine with (5-methylsulfanyl-pyridin-2-yl)-methylamine (preparation 16a). ESI mass spectrum: [M+H]+ = 285; r.t. HPLC: 0.51 min (X012 S01).
16c: 2-(5-(N-Cyano-5,-methylsulfinimidoyl)pyridin-2-yl)-iso indole- 1 ,3-dione
Figure imgf000089_0003
The title compound is prepared in analogy to preparation 6b, substituting 2-(4- (methylthio)benzyl)isoindoline- 1 ,3-dione with 2-(5-methylsulfanyl-pyridin-2-ylmethyl)- isoindole-l ,3-dione (preparation 16b). ESI mass spectrum: [M+H]+ = 325; r.t. HPLC: 0.63 min (Z018_S04). 16d: 2-(5-(N-Cyano-5'-methylsulfonimidoyl)pyridin-2-yl)-iso indole- 1 ,3-dione
Figure imgf000090_0001
The title compound is prepared in analogy to preparation 6c, substituting 2-(4-(N-cyano-lS- methylsulfinimidoyl)benzyl)isoindoline- 1 ,3-dione with 2-(5-(N-cyano-lS- methylsulfinimidoyl)pyridin-2-yl)-isoindole-l ,3-dione (preparation 16c). ESI mass spectrum: [M+H]+ = 341 ; r.t. HPLC: 0.88 min (Z018 S04).
16e: (5-(N-Cyano-5'-methylsulfonimidoyl)pyridin-2-yl)methanamine
Figure imgf000090_0002
The title compound is prepared in analogy to preparation 6d, substituting 2-(4-(N-cyano-lS- methylsulfonimidoyl)benzyl)isoindoline-l , 3-dione with 2-(5-(N-cyano-lS- methylsulfonimidoyl)pyridin-2-yl)-isoindole-l ,3-dione (preparation 16d). ESI mass spectrum: [M+H]+ = 21 1 ; r.t. HPLC: 0.23 min (Z01 1 S03). Preparation 17: (4-(S-Methyl- V- methylsulfonyl))phenyl)methanamine
Figure imgf000090_0003
17a: 2-(4-(N-Trifluoracetyl-5'-methylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000091_0001
A solution of 2-(4-(N-cyano-5'-methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione
(preparation 6c, 2.00 g, 6.00 mmol) in dichloromethane (20 mL) is cooled at 0 °C, and trifluoroacetic anhydride (2.48 mL, 3.72g, 17.7 mmol) is added. The mixture is allowed to warm to room temperature, stirred for 2 h and poured into water. The aqueous layer is extracted three times with dichloromethane, and the combined organic layers are dried over Na2S04 and concentrated under reduced pressure. The residue is purified by flash chromatography on silica (ethyl acetate/dichloro methane 9: 1). Yield: 2.10 g (86% of theory); ESI mass spectrum: [M+H]+ = 41 1 , r.t. HPLC: 4.50 min (LCMS-FA-2).
17b: 2-(4-(5'-Methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione
Figure imgf000091_0002
Potassium carbonate (3.37 g, 24.4 mmol) is added to a solution of 2-(4-(N-trifluoracetyl-5'- methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (preparation 17a, 2.00 g, 4.88 mmol) in methanol (20 mL). The mixture is stirred at room temperature for 16 h and filtered. The filtrate is concentrated under reduced pressure, and the residue is purified by flash chromatography on silica (methanol/dichloromethane 7:3). Yield: 0.90 g (58% of theory); ESI mass spectrum: [M+H]+ = 315, r.t. HPLC: 1.08 min (V003 003).
17c: 2-(4-(5'-Methyl-N-(methylsulfonyl)sulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000092_0001
A solution of 2-(4-(5'-methylsulfonimidoyl)benzyl)isoindoline-l,3-dione (preparation 17b, 2.50 g, 7.95 mmol) and triethylamine (2.2 mL, 16 mmol) in dichloromethane (20 mL) is cooled at 0 °C and treated with methanesulfonyl chloride (620 μί, 8.0 mmol). The mixture is stirred at this temperature for 1.5 h and then washed with water. The organic layer is concentrated under reduced pressure, and the residue is suspended in a mixture of methanol and water. The precipitate is filtered, washed with water, and dried. Yield: 273 mg (8% of theory); ESI mass spectrum: [M+H]+ = 393, r.t. HPLC: 0.44 min (X012 S01).
17d: (4-(5'-Methyl-N-(methylsulfon l))phenyl)methanamine
Figure imgf000092_0002
A solution of 2-(4-(5'-methyl-N-(methylsulfonyl)sulfonimidoyl)benzyl)isoindo line- 1,3- dione (preparation 17c, 270 mg, 0.69 mmol) and 1,2-diamino ethane (83 μΐ,, 74 mg, 1.24 mmol) in a 2: 1 : 1 mixture of acetonitrile, tetrahydrofuran and ethanol (3 mL) is heated at 60 °C for 4 h. The mixture is concentrated under reduced pressure, and the residue is suspended in dichloromethane. The mixture is filtered and the filtrate is concentrated under reduced pressure. Yield: 178 mg (99% of theory); ESI mass spectrum: [M+H]+ = 263, r.t. HPLC: 0.17 min (X012 S01).
Preparation 18: 2-Fluoro-4-( V-cyano-S-methylsulfonimidoyl)benzylamine
Figure imgf000093_0001
18a: 2-Fluoro-4-methylsulfanyl-benzylamine
Figure imgf000093_0002
A mixture of 2-fluoro-4-methylsulfanyl-benzonitrile (8.0 g, 45.5 mmol), Raney-Nickel (20.0 g) and concentrated aqueous ammonia (300 mL) in methanol (3.0 L) is treated with hydrogen (3.4 bar) at room temperature and stirred over night. The mixture is filtered through a pad of silica gel, and the filtrate is concentrated under reduced pressure. Yield: 6.3 g (81% of theory); ESI mass spectrum: [M+H-NH3]+ = 155; r.t. HPLC: 1.38 min (0- 30AB).
18b: (2-Fluoro-4-methylsulfan l-benzyl)-carbamic acid tert-butyl ester
Figure imgf000093_0003
Triethylamine (3.40 g, 33.3 mmol) and di-tert-butyl dicarbonate (7.4 g, 33.3 mmol) are added to a solution of 2-fluoro-4-methylsulfanyl-benzylamine (preparation 18a, 4.0 g, 22.2 mmol) in dichloro methane (100 mL). After 30 min water is added and the mixture is extracted with dichloromethane. The organic layer is washed with brine, dried over Na2S04 and concentrated under reduced pressure. Yield: 5.0 g (79% of theory); ESI mass spectrum: [M+Na]+ = 294; r.t. HPLC: 0.86 min (5-95 AB).
18c: (2-Fluoro-4-(N-cyano-5'-methylsulfinimidoyl)-benzyl)-carbamic acid tert-butyl ester
Figure imgf000094_0001
The title compound is prepared following the procedure described for preparation 15b, using (2-fluoro-4-methylsulfanyl-benzyl)-carbamic acid tert-butyl ester (preparation 18b) as starting material. ESI mass spectrum: [M+H]+ = 312; r.t. HPLC: 0.68 min (5-95 AB).
18d: (2-Fluoro-4-(N-cyan -5'-methylsulfonimidoyl)-benzyl)-carbamic acid tert-butyl ester
Figure imgf000094_0002
The title compound is prepared following the procedure described for preparation 15c, using (2-fluoro-4-(N-cyano-5'-methylsulfinimidoyl)-benzyl)-carbamic acid tert-butyl ester (preparation 18c) as starting material. ESI mass spectrum: [M+H]+ = 312; r.t. HPLC: 0.68 min (5-95AB).
18e: 2-Fluoro-4-(N-cyano-5'-methylsulfonimidoyl)benzylamine
Figure imgf000094_0003
A solution of (2-fluoro-4-(N-cyano-5'-methylsulfonimidoyl)-benzyl)-carbamic acid tert- butyl ester (preparation 18c, 2.20 g, 6.38 mmol) in dichloromethane (20 mL) is cooled at 0 °C and treated with a solution of hydrogen chloride in ethyl acetate (4 M, 16 mL, 64 mmol). After 2 h all volatiles are removed under reduced pressure and the residue is purifed by preparative reversed phase HPLC (Gemini, gradient of acetonitrile in water, 0.04% NH3). ESI mass spectrum: [M+H]+ = 228; r.t. HPLC: 2.16 min (0-30AB). Preparation 19: 2-Methyl-4-( V-cyano-S-methylsulfonimidoyl)benzylamine
Figure imgf000095_0001
The title compound is prepared following the procedure described for preparation 15, substituting 3-fluoro-4-methylsulfanyl-benzonitrile with 2-methyl-4-methylsulfanyl- benzonitrile as starting material. ESI mass spectrum: [M+H]+ = 224; r.t. HPLC: 2.18 min (0-30AB).
The following examples are prepared as described for preparation Id, substituting preparation lc with the appropriate starting material acid and substituting preparation 6 with the appropriate starting material amine. The asterisk in the structures indicates that the compound is prepared from the enantiomer preparation 12B (prep. = preparation).
Figure imgf000095_0002
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
The following examples are prepared as described for Example 2, substituting preparation 1 with the appropriate bromo starting material (prep. = preparation).
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Several racemic examples are separated into the respective enantiomers by preparative chiral HPLC as described for the separation of racemic Example 1 into the enantiomers Example 1 A and Example IB, employing HPLC conditions as indicated in the following table. Preparative
Enantio¬
Racemic Chiral Analytical Chiral HPLC r.t. MS meric
Example HPLC Method [M+H]+
Example
Conditions
Daicel Chiralpak®OJ-H,
Daicel OJ-H 10A 250mmx4.6mm, 4ml/min, 2.33 min 531
10 10% MeOH lOmin, 15% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
10B 2.53 min 531
C02, 40°C
Daicel Chiralpak®OJ-H,
Daicel OJ-H 21A 250mmx4.6mm, 4ml/min, 2.24 min 543
21 10% MeOH lOmin, 15% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
21B 2.41 min 543
C02, 40°C
Daicel Chiralpak®AS-H,
22A 2.05 min 557
Daicel AS-H 250mmx4.6mm, 4ml/min,
22 20% MeOH lOmin, 15% MeOH + 0.2%
+0.2% DEA 22B diethylamine in supercritical 2.35 min 557
C02, 40°C
Daicel Chiralpak®OJ-H,
Daicel OJ-H 29A 250mmx4.6mm, 4ml/min, 3.48 min 513
29 10% MeOH lOmin, 15% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
29B 3.91 min 513
C02, 40°C
Daicel Chiralpak®OZ-H,
Daicel OZ-H 30A 250mmx4.6mm, 4ml/min, 6.50 min 527
30 35% MeOH lOmin, 40% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
30B 7.55 min 527
C02, 40°C Preparative
Enantio¬
Racemic Chiral Analytical Chiral HPLC r.t. MS meric
Example HPLC Method [M+H]+
Example
Conditions
Daicel Chiralpak®OJ-H,
Daicel OJ-H 31A 250mmx4.6mm, 4ml/min, 2.44 min 527
31 10% MeOH lOmin, 15% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
31B 2.71 min 527
C02, 40°C
Daicel Chiralpak®OJ-H,
Daicel OJ-H 5.5A 250mmx4.6mm, 4ml/min, 2.36 min 499
5.5 10% MeOH lOmin, 20% MeOH + 0.2%
+0.2% DEA diethylamine in supercritical
5.5B 2.65 min 499
C02, 40°C
Example 60 : 5-(3-Trifluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V VyV-trimethylsulfamimidoyl)-benzylamide
Figure imgf000115_0001
60a: Thioacetic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2-ylmethyl)-phenyl ester
Figure imgf000115_0002
A solution of 2-benzyl-isoindole-l,3-dione (prepared as described in WO07093452, 22.0 g, 92.7 mmol) in dichloromethane (150 mL) is cooled in an ice bath at 0°C. Chlorosulfonic acid (12.3 mL, 185 mmol) is added dropwise and the mixture is allowed to warm to room temperature. After 1.5 h the mixture is cooled at 0 °C, and thionyl chloride (10.1 mL, 139 mmol) is added. The mixture is heated at reflux for 3.5 h, and chlorosulfonic acid (3.1 mL, 46 mmol) and thionyl chloride (2.0 mL, 28 mmol) is added. The mixture is stirred at room temperature over night and poured into ice water. The aqueous phase is extracted twice with dichloromethane, and the combined organic phases are concentrated under reduced pressure.
The residue is dissolved in toluene (250 mL), and the mixture is cooled at 0 °C. A solution of triphenylphosphine (70.9 g, 270 mmol) in toluene (250 mL) is added dropwise, and the mixture is warmed at room temperature. After 2.5 h water is added and the phases are separated.
The organic layer is concentrated under reduced pressure and the residue is dissolved in DMF (200 mL). Acetic anhydride (29 mL) is added and the mixture is stirred at room temperature for 2 h. The mixture is concentrated under reduced pressure and purified by flash chromatography on silica (gradient cyclohexane to cyclohexane/ethyl acetate 8:2). Yield: 14 g (15 % of theory). ESI mass spectrum: [M+H]+ = 312; r.t. HPLC: 1.36 min (V001 007).
60b: 4-(l,3-Dioxo-l,3-dihyd -isoindol-2-ylmethyl)-benzenesulfinic acid methylamide
Figure imgf000116_0001
A solution of acetic anhydride (4.34 mL, 45.9 mmol) in dichloromethane (50 mL) is added dropwise to a precooled solution of thioacetic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2- ylmethyl)-phenyl ester (preparation 60a, 13.0 g, 41.8 mmol) in dichloromethane (500 mL) at -20 °C. A solution of sulfuryl chloride (7.4 mL, 91.9 mmol) in dichloromethane (50 mL) is added at this temperature, and the mixture is stirred for 1 h. The mixture is warmed to room temperature, and all volatiles are removed under reduced pressure.
The residue is dissolved in dichloromethane (400 mL), and the solution is cooled in an ice bath at 0 °C. Methylamine (2 M in tetrahydrofuran, 84 mL, 168 mmmol) is added dropwise at 0 °C, and the mixture is stirred for 2 and then warmed to room temperature. Saturated sodium chloride solution is added, and the mixture is extracted with dichloromethane. The combined organic layers are dried over Na2S04 and concentrated under reduced pressure. Yield: 3.44 g (23% of theory). ESI mass spectrum: [M+H]+ = 315; r.t. HPLC: 0.89 min (X012 S01).
60c: 4-((l,3-Dioxoisoindolin-2-yl)methyl)-N,N,N'-trimethyl-benzene-sulfonimidamide
Figure imgf000117_0001
A mixture of 4-(l,3-dioxo-l,3-dihydro-isoindol-2-ylmethyl)-benzenesulfinic acid methylamide (preparation 60b, 250 mg, 0.795 mmol) in acetonitrile is cooled at 0 °C in an ice bath and treated with tert-butyl hypochlorite (100 μί, 0.88 mmol). After 15 min dimethylamine (2 M in tetrahydrofuran, 600 μί, 1.2 mmol) is added and the mixture is warmed to room temperature. Water is added, and the mixture is extracted twice with dichloromethane. The combined organic layers are dried over Na2S04 and concentrated under reduced pressure. The residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 7:3 to ethyl acetate). Yield: 284 mg (99% of theory). ESI mass spectrum: [M+H]+ = 358; r.t. HPLC: 0.43 min (X012 S01).
60d: 4-(Aminomethyl)-N,N,N'-trimeth lbenzenesulfonimidamide
Figure imgf000117_0002
A solution of 4-((l,3-dioxoisoindolin-2-yl)methyl)-N,N,N'-trimethyl-benzene-sulfonimid- amide (preparation 60c, 280 mg, 0.78 mmol) in a 2: 1 : 1 mixture of acetonitrile,
tetrahydrofuran and ethanol (8 mL) is treated with 1 ,2-diaminoethane (116 μί, 1.74 mmol). The mixture is heated at 60 °C for 5 h, and another portion of 1 ,2-diaminoethane (26 μΐ,, 0.39 mmol) is added. After 1.5 h all volatiles are removed under reduced pressure. The residue is treated with dichloromethane, and the mixture is filtered. The filtrate is concentrated under reduced pressure. Yield: 172 mg (97% of theory). ESI mass spectrum: [M+H]+ = 228; r.t. HPLC: 0.66 min (V01 1 S01).
Example 60: 5-(3-Trifluoromethyl-phenyl)-l-isopropyl-6-methyl-4-oxo-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(N,N,N'-trimethylsulfamimidoyl)-benzylamide
Figure imgf000118_0001
A solution of l-isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3 -carboxylic acid (preparation 9.2, 186 mg, 0.55 mmol) in DMF (5 mL) is treated with DIPEA (300 μί, 1.72 mmol) and stirred at room temperature for 10 min. HBTU (312 mg, 0.82 mmol) is added, and the mixture is stirred for 20 min. A solution of 4-(aminomethyl)-N,N,N'-trimethylbenzenesulfonimidamide (preparation 60d, 156 mg, 0.69 mmol) in DMF (0.5 mL) is added and the mixture is stirred for 2 h. Water is added and the mixture is extracted with dichloromethane. The organic layer is dried over Na2S04 and concentrated under reduced pressure. The residue is purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1 % TFA). Yield: 41 mg (1 1% of theory); ESI mass spectrum: [M+H]+ = 549; r.t. HPLC: 1.17 min (Z001 005).
Example 61 : 5-(3-Difluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid 4-( V \yV-trimethylsulfamimidoyl)-benzylamide
Figure imgf000118_0002
Example 61 is prepared as described for Example 60, substituting preparation 9.2 with preparation 9.12. ESI mass spectrum: [M+H]+ = 531 ; r.t. HPLC: 0.86 min (Z01 1 S03).
Example 62 : 5-(3-Trifluoromethyl-phenyl)- l-isopropyl-6-methyl-4-oxo- 1 ,4-dihydro- pyridine-3-carboxylic acid (5-( V-cyano-S-methylsulfonimidoyl)-l-oxy-pyridin-2-yl)- methylamide (Pyridine N-oxide of Exam le 16)
Figure imgf000119_0001
eto-chloroperoxybenzoic acid (77%, 8.0 mg, 36 μιηοΐ) is added to a solution of 5-(3-trifiuoromethyl-phenyl)- 1 -isopropyl-6-methyl-4-oxo- 1 ,4-dihydro-pyridine-3- carboxylic acid (5-(N-cyano-5'-methylsulfonimidoyl)pyridin-2-yl)methylamide (example 16, 17.0 mg, 32 μιηοΐ) in dichloromethane (0.5 mL), and the mixture is stirred at room temperature for 2 h. The mixture is heated at reflux and stirred for 2 h. The mixture is cooled at room temperature and stirred over night. A second portion of m-CPBA (77%, 4.3 mg, 19 μιηοΐ) is added and the mixture is heated at reflux for 2.5 h. A third portion of m-CPBA (77%o, 4.3 mg, 19 μιηοΐ) is added and the mixture is stirred at room temperature over night. Saturated aqueous sodium thio sulfate solution is added and the mixture is extracted twice with dichloromethane. The organic layer is dried and concentrated under reduced pressure. The residue is purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1 % NH3). Yield: 3.3 mg (19%> of theory); ESI mass spectrum: [M+H]+ = 548; r.t. HPLC: 0.54 min (X01 1 S03).
Example 63: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-(S-cyclopropyl- V-cyanosulfonimidoyl)-benzylamide
Figure imgf000120_0001
63a: 4-Cyclopropylsulfanyl-benzamide
Figure imgf000120_0002
Thionyl chloride (340 μί, 4.7 mmol) is added to a solution of 4-cyclopropylsulfanyl- benzoic acid (prepared as described in WO07003960, 190 mg, 0.98 mmol) in
dichloromethane (1.9 mL), and the mixture is heated at reflux for 30 min. All volatiles are removed under reduced pressure, and the residue is dissolved in chloroform (1.9 mL). Concentrated aqueous ammonia (28%, 770 μί, 9.8 mmol) is added and the mixture is stirred at room temperature for 1 h. Water is added and the phases are separated. The organic layer is concentrated under reduced pressure. Yield: 137 mg (72% of theory); ESI mass spectrum: [M+H]+ = 194; r.t. HPLC: 0.45 min (X011 S03).
64b : 4-Cy clopropylsulfanyl-benz lamine
Figure imgf000120_0003
A solution of 4-cyclopropylsulfanyl-benzamide (preparation 63a, 137 mg, 0.71 mmol) in THF is added to lithium aluminium hydride (1 M in THF, 2.1 mL, 2.1 mmol) precooled at 0 °C. The mixture is stirred at 0 °C for 1 h and then heated at reflux for 1 h. The mixture is cooled to room temperature and stirred for 1.5 h. Water is added, and the mixture is extracted twice with dichloromethane. The combined organic layers are dried over Na2SC>4 and concentrated under reduced pressure. Yield: 75 mg (60%> of theory); ESI mass spectrum: [M+H]+ = 180; r.t. HPLC: 0.51 min (X011 S03). 63c: 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3- carboxylic acid 4-cyclopropylsulfanyl-benzylamide
Figure imgf000121_0001
The title compound is prepared following the procedure described for example 60, substituting 4-(aminomethyl)-N,N,N'-trimethylbenzenesulfonimidamide with 4- cyclopropylsulfanyl-benzylamine (preparation 63b), and replacing HBTU with TBTU. ESI mass spectrum: [M+H]+ = 501; r.t. HPLC: 0.77 min (X012 S01).
63d: 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3- carboxylic acid 4-(5,-cyclopropyl-N-cyanosulfinimidoyl)-benzylamide
Figure imgf000121_0002
A mixture of l-isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-cyclopropylsulfanyl-benzylamide (preparation 63 c, 75 mg, 0.15 mmol), cyanamide (8 mg, 0.19 mmol), N-bromosuccinimide (40 mg, 0.22 mmol) and potassium tert-butoxide (20 mg, 0.18 mmol) in methanol (1 mL) is stirred at room temperature for 1 h. Saturated aqueous sodium thiosulfate solution is added, and the mixture is extracted twice with dichloromethane. The organic layer is dried over Na2S04 and concentrated under reduced pressure. The residue is purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1 % NH3). Yield: 7 mg (8% of theory); ESI mass spectrum: [M+H]+ = 541; r.t. HPLC: 0.64 min (X012 S02). Example 63: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-(S-cyclopropyl- V-cyanosulfonimidoyl)-benzylamide
Figure imgf000122_0001
A mixture of l-isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(5,-cyclopropyl-N-cyanosulfinimidoyl)-benzylamide
(preparation 63d, 40 mg, 74 μηιοΐ), meto-chloroperoxybenzoic acid (77%, 25 mg, 0.1 1 mmol) and potassium carbonate (31 mg, 0.22 mmol) in ethanol is stirred at room temperature for 1 h. Another portion of meto-chloroperoxybenzoic acid (77%, 5 mg, 0.02 mmol) is added and stirring is continued for 1.5 h. Saturated aqueous sodium thiosulfate solution is added, and the mixture is extracted with dichloromethane. The organic layer is dried over Na2S04 and concentrated under reduced pressure. The residue i purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1 % TFA). Yield: 13 mg (31% of theory); ESI mass spectrum: [M+H]+ = 557; r.t. HPLC 0.65 min (X01 1_S03).
Example 64: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-(N-(cyanomethyl)-S-methylsulfonimidoyl)-benzylamide
Figure imgf000122_0002
64a: 2-(4-(N-Cyanomethyl-5'-methylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000123_0001
A mixture of 2-(4-(5'-methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (preparation 17b, 500 mg, 1.59 mmol) and 1 ,2-dimethoxy ethane (2 mL) is added to a mixture of potassium hydride (30%, 260 mg, 1.95 mmol) in 1 ,2-dimethoxyethane (3 mL). The mixture is stirred for 15 min at room temperature and treated with tetraethylammonium bromide (10 mg, 0.05 mmol) and bromoacetonitrile (220 μί, 3.16 mmol). The mixture is heated at 1 10 °C over night and cooled to room temperature. Water is added, and the mixture is extracted twice with dichloromethane. The organic layer is concentrated under reduced pressure, and the residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 1 :0 to 0: 1). Yield: 155 mg (27% of theory). ESI mass spectrum: [M+H]+ = 354; r.t. HPLC: 0.47 min (X011 S03).
64b : 4-(N-cyanomethyl-5'-meth lsulfonimidoyl)benzylamine
Figure imgf000123_0002
The title compounds is prepared in analogy to preparation 60d, using 2-(4-(N-cyano- methyl-^-methylsulfonimidoy^benzy^isoindoline- l ^-dione (preparation 64a) as starting material. ESI mass spectrum: [M+H]+ = 224; r.t. HPLC: 0.22 min (X01 1_S03).
Example 64: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(N-(cyanomethyl)-5'-methylsulfonimidoyl)-benzylamide
Figure imgf000124_0001
The title compound is prepared following the procedure described for example 60, substituting 4-(aminomethyl)-N,N,N'-trimethylbenzenesulfonimidamide with 4-(N-cyano- methyl-S-methylsulfonimidoyl)benzylamine (preparation 64b), and replacing HBTU with TBTU. ESI mass spectrum: [M+H]+ = 545; r.t. HPLC: 0.59 min (X012 S01).
Example 65: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid [l-(4-( V-cyano-S-methylsulfonimidoyl)-phenyl)-ethyl]- amide
Figure imgf000124_0002
65a: 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3- carboxylic acid [l-(4-methylsulfanyl-phenyl)-ethyl] -amide
Figure imgf000124_0003
The title compound is prepared following the procedure described for example 60, substituting 4-(aminomethyl)-N,N,N'-trimethylbenzenesulfonimidamide with l-(4- methylsulfanyl-phenyl)-ethylamine, and replacing HBTU with TBTU. ESI mass spectrum: [M+H]+ = 489; r.t. HPLC: 0.76 min (X011 S03).
Example 65 : 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro- pyridine-3-carboxylic acid 1 -(4-(N-cyano-5'-methylsulfonimidoyl)-phenyl)-ethyl]-amide
Figure imgf000125_0001
A mixture of l-isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid [l-(4-methylsulfanyl-phenyl)-ethyl] -amide (preparation 65 a, 50 mg, 0.10 mmol), cyanamide (6 mg, 0.14 mmol) and potassium tert-butoxide (14 mg, 0.13 mmol) in methanol (0.5 mL) is treated with N-bromosuccinimide (27 mg, 0.15 mmol) and stirred at room temperature for 15 min. Saturated aqueous Na2S2C"3 solution is added, and the mixture is extracted with dichloromethane. The organic layer is concentrated under reduced pressure, and the residue is dissolved in ethanol (0.5 mL).
Potassium carbonate (16 mg, 120 μιηοΐ) and meto-chloroperoxybenzoic acid (77%, 13 mg, 60 μιηοΐ) is added, and the mixture is stirred at room temperature for 1.5 h. Another portion of potassium carbonate (10 mg, 70 μιηοΐ) and meto-chloroperoxybenzoic acid (77%, 13 mg, 60 μιηοΐ) is added and stirring is continued for 1 h. Saturated aqueous Na2S2C"3 solution is added, and the mixture is extracted twice with dichloromethane. The residue is purified by flash chromatography on silica (gradient cyclohexane/ethyl acetate 8:2 to to ethyl acetate). Yield: 5 mg (25 %> of theory); ESI mass spectrum: [M+H]+ = 545; r.t. HPLC: 0.72 min (Y001 U01).
Example 66: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 3-methyl-4-( V-cyano-S-methylsulfonimidoyl)-benzylamide
Figure imgf000126_0001
66a: 3-Methyl-4-methylsulfanyl-benzonitrile
Figure imgf000126_0002
Sodium methanethiolate (1.60 g, 22.8 mmol) is added to a solution of 4-fluoro-3-methyl- benzonitrile (2.50 g, 18.5 mmol) in DMF, and the mixture is heated at 80 °C for 15 min. All volatiles are evaporated, and the residue is treated with water. The mixture is extracted with ethyl acetate twice, and the organic layer is dried over Na2SC"4 and concentrated. Yield: 3.0 g (99 % of theory); ESI mass spectrum: [M+H]+ = 164; r.t. HPLC: 0.61 min (X011 S03).
66b : 3 -Methyl-4-methylsulfanyl-benzylamine
Figure imgf000126_0003
A solution of lithium aluminium hydride (1.0 M in THF, 18.4 mL, 18.4 mmol) is cooled in an ice bath and treated slowly with a solution of 3-methyl-4-methylsulfanyl-benzonitrile (preparation 66a, 1.0 g, 6.1 mmol) in THF (10 mL) while the temperature is kept below 5 °C. The mixture is allowed to warm to room temperature and stirred over night. The mixture is cooled again at 0 °C, and water (2 mL) and aqueous sodium hydroxide (4 M, 2 mL) is added. The mixture is filtered and the filtrate is extracted twice with
dichloromethane. The combined organic layers are concentrated under reduced pressure. Yield: 923 mg (90 % of theory); ESI mass spectrum: [M+H-NH3]+ = 151; r.t. HPLC: 0.61 min (X011_S03). 66c: 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3- carboxylic acid 3-methyl-4-methylsulfanyl-benzylamide
Figure imgf000127_0001
The title compound is prepared following the procedure described for example 60, using 3-methyl-4-methylsulfanyl-benzylamine (preparation 66b) as starting material, and replacing HBTU with TBTU. ESI mass spectrum: [M+H]+ = 489; r.t. HPLC: 0.77 min (X011 S03).
66d: 1 -Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3- carboxylic acid 3-methyl- -(N-cyano-5'-methylsulfinimidoyl)-benzylamide
Figure imgf000127_0002
N-Bromosuccinimide (63 mg, 0.35 mmol) is added to a mixture of l-isopropyl-6-methyl-4- oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro-pyridine-3-carboxylic acid 3-methyl-4- methylsulfanyl-benzylamide (preparation 66c, 115 mg, 0.24 mmol), cyanamide (14 mg, 0.33 mmol) and potassium tert-butoxide (32 mg, 0.29 mmol) in methanol (1.1 mL). After 20 min saturated aqueous sodium thiosulfate solution is added, and the mixture is extracted twice with dichloromethane. The combined organic layers is dried over Na2S04 and concentrated under reduced pressure. The residue is directly used in the next step without further purification. Yield: 99 mg (80% of theory); ESI mass spectrum: [M+H]+ = 529; r.t. HPLC: 0.58 min (X012 S01). Example 66: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3-carboxylic acid 3-methyl-4-(N-cyano-5'-methylsulfonimidoyl)-benzylamide
Figure imgf000128_0001
A solution of 1 -isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)- 1 ,4-dihydro- pyridine-3-carboxylic acid 3-methyl-4-(N-cyano-5'-methylsulfinimidoyl)-benzylamide (preparation 66d, 25 mg, 47 μιηοΐ) and acetic acid (3 μί, 50 μmol) in acetonitrile (0.2 mL) is added to a solution of potassium permanganate (8 mg, 50 μιηοΐ) in acetonitrile
(0.15 mL) and water (0.15 mL). After 3 h another portion of potassium permanganate (8 mg, 50 μιηοΐ) is added and the mixture is heated at reflux. After 2 h saturated aqueous sodium thiosulfate solution is added, and the mixture is extracted twice with dichloro- methane. The organic layer is dried over Na2S04 and concentrated under reduced pressure. The residue is purified by preparative reversed phase HPLC (Xbridge, gradient of acetonitrile in water, 0.1 % TFA). Yield: 2 mg (8% of theory); ESI mass spectrum:
[M+H]+ = 545; r.t. HPLC: 0.61 min (X012 S01).
Example 67: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l,4-dihydro- pyridine-3-carboxylic acid 4-( V,S-dimethylsulfonimidoyl)-benzylamide
Figure imgf000129_0001
67a: 2-(4-(N,5'-Dimethylsulfonimidoyl)benzyl)isoindoline- 1 ,3-dione
Figure imgf000129_0002
A mixture of 2-(4-(5'-methylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (preparation 17b, 5 200 mg, 63.6 mmol) and paraformaldehyde (1 15 mg, 1.27 mmol) in acetic acid (2 mL) is heated in a microwave at 100 °C for 4 h. The mixture is concentrated under reduced pressure, and the residue is purified by flash chromatography on silica (gradient dichloromethane to dichloromethane/methanol 95 :5). Yield: 189 mg (90 % of theory); ESI mass spectrum: [M+H]+ = 329; r.t. HPLC: 0.45 min (X01 1 S03).
10
67b : 4-(N,5'-Dimethylsulfonimido l)benzylamine
Figure imgf000129_0003
A mixture of 2-(4-(N,5'-dimethylsulfonimidoyl)benzyl)isoindoline-l ,3-dione (189 mg, 0.58 mmol) and 1 ,2-diamino ethane (132 μΐ^, 3.44 mmol) in a 2: 1 : 1 mixture of acetonitrile, i s tetrahydroiuran and ethanol (2 mL) is heated at reflux for 1 h. All volatiles are removed under reduced pressure, and the residue is treated with dichloromethane. The mixture is filtered, and the filtrate is concentrated under reduced pressure. Yield: 1 14 mg (85 % of theory); ESI mass spectrum: [M+H]+ = 199; r.t. HPLC: 0.22 min (X01 1 S03). Example 67: l-Isopropyl-6-methyl-4-oxo-5-(3-trifluoromethyl-phenyl)-l ,4-dihydro- pyridine-3 -carboxylic acid 4-(N, '-dimethylsulfonimidoyl)-benzylamide
Figure imgf000130_0001
The title compound is prepared following the procedure described for example 60, substituting 4-(aminomethyl)-N,N,N'-trimethylbenzenesulfonimidamide with 4-(N,S- Dimethylsulfonimidoyl)benzylamine (preparation 67b), and replacing HBTU with TBTU. ESI mass spectrum: [M+H]+ = 520; r.t. HPLC: 0.58 min (X01 1 S03).
EXAMPLES
Other features and advantages of the present invention will become apparent from the following more detailed examples which illustrate, by way of example, the principles of the invention.
Human neutrophil elastase assay
Materials: 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: lOOmM Tris, 500mM NaCl, adjusted to pH 7.5; Assay buffer: lOOmM Tris, 500mM NaCl, adjusted to pH 7.5, containing 0.01%BSA. Assay conditions: Test compounds were prediluted in DMSO and subsequently in compound buffer (5% DMSO final). 5 μΐ, of these compound dilutions were mixed with 10 μΐ 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 substrate solution in assay buffer were added (250 μΜ 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). IC50 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%. IC50 values of selected compound in the Neutrophil Elastase assay:
Example IC50 [nM] Example IC50 [nM]
1 1,3 6 2,4
1A 3,1 7 2,1
IB 1,3 8 2,4
2 1,7 9 <1
2A 2,3 9A <1
2B 1,1 9B <1
3 4,7 10 1,2
3A 12,0 10A 2,7
3B 5,1 10B 1,2
4 3,0 5.5A 5,0
4A 4,1 5.5B 2,7
4B 1,6 11 <1
5 2,5 12 <1 Example IC50 [nM] Example IC50 [nM]
13 2,4 36 1,0
14 1,5 37 <1
15 2,2 38 3,7
16 1,9 39 3,7
17 7,3 40 4,0
18 1,5 41 1,8
19 1,0 42 4,7
20 5,6 43 13,4
21A 5,2 44 14,9
21B 2,1 45 14,4
22A 4,5 46 11,7
22B 1,2 47 1,2
23 2,6 48 9,4
24 5,5 49 7,3
25 4,3 50 8,6
26 3,3 53 2,3
27 2,5 54 2,3
28 3,3 55 1,8
29 1,5 56 3,1
29A 2,2 57 1,7
29B <1 58 4,7
30 <1 59 7,9
30A 1,8 60 22,0
30B <1 61 10,2
31 <1 62 2,7
31A <1 63 1,4
31B <1 64 24,5
32 1,1 65 57,1
33 2,5 66 3,4
34 2,4 67 49,3
35 8,0 COMBINATIONS
The compounds of general formula I may be used on their own or combined with other active substances of formula I according to the invention. The compounds of general formula I may optionally also be combined with other pharmacologically active substances. These include, B2-adrenoceptor-agonists (short and long-acting),
anti-cholinergics (short and long-acting), anti-inflammatory steroids (oral and topical corticosteroids), cromoglycate, methylxanthine, dissociated-glucocorticoidmimetics, PDE3 inhibitors, PDE4- inhibitors, PDE7- inhibitors, LTD4 antagonists, EGFR- inhibitors, Dopamine agonists, PAF antagonists, Lipoxin A4 derivatives, FPRL1 modulators, LTB4-receptor (BLT1, BLT2) antagonists, Histamine HI receptor antagonists, Histamine H4 receptor antagonists, dual Histamine H1/H3 -receptor antagonists, PI3-kinase inhibitors, inhibitors of non-receptor tyrosine kinases as for example LYN, LCK, SYK, ZAP-70, FYN, BTK or ITK, inhibitors of MAP kinases as for example p38, ERK1, ERK2, JNK1, JNK2, JNK3 or SAP, inhibitors of the NF-κΒ signalling pathway as for example IKK2 kinase inhibitors, iNOS inhibitors, MRP4 inhibitors, leukotriene biosynthese inhibitors as for example 5 -Lipoxygenase (5-LO) inhibitors, cPLA2 inhibitors, Leukotriene A4
Hydrolase inhibitors or FLAP inhibitors, MMP9-inhibitors, MMP12-inhibitors
Non-steroidale anti-inflammatory agents (NSAIDs), Cathepsin C (or DPPI / Dipeptidyl- aminopeptidase I) inhibitors, CRTH2 antagonists, DPI -receptor modulators, Thromboxane receptor antagonists, CCR3 antagonists, CCR4 antagonists, CCR1 antagonists, CCR5 antagonists, CCR6 antagonists, CCR7 antagonists, CCR8 antagonists, CCR9 antagonists, CCR30 antagonists, , CXCR3 antagonists, CXCR4 antagonists, CXCR2 antagonists,
1 3
CXCR antagonists, CXCR5 antagonists, CXCR6 antagonists, CX3CR antagonists, Neurokinin (NK1, NK2) antagonists, Sphingosine 1 -Phosphate receptor modulators, Sphingosine 1 phosphate lyase inhibitors, Adenosine receptor modulators as for example A2a-agonists, modulators of purinergic receptors as for example P2X7 inhibitors, Histone Deacetylase (HDAC) activators, Bradykinin (BK1, BK2) antagonists, TACE inhibitors, PPAR gamma modulators, Rho-kinase inhibitors, interleukin 1-beta converting enzyme (ICE) inhibitors, Toll-Like receptor (TLR) modulators, HMG-CoA reductase inhibitors, VLA-4 antagonists, ICAM-1 inhibitors, SHIP agonists, GABAa receptor antagonist, ENaC-inhibitors, Prostasin-inhibitors, Melanocortin receptor (MC1R, MC2R, MC3R, MC4R, MC5R) modulators, CGRP antagonists, Endothelin antagonists, TNFa antagonists, anti-TNF antibodies, anti-GM-CSF antibodies, anti-CD46 antibodies, anti-IL-1 antibodies, anti-IL-2 antibodies, anti-IL-4 antibodies, anti-IL-5 antibodies, anti-IL-13 antibodies, anti-IL-4/IL-13 antibodies, anti-TSLP antibodies, anti-OX40 antibodies, mucoregulators, immunotherapeutic agents, compounds against swelling of the airways, compounds against cough, VEGF inhibitors, but also combinations of two or three active substances.
Preferred are betamimetics, anticholinergics, corticosteroids, PDE4-inhibitors,
LTD4-antagonists, EGFR-inhibitors, Cathepsin C inhibitors, CRTH2 inhibitors,
5-LO-inhibitors, Histamine receptor antagonists and SYK- inhibitors, especially Cathepsin C inhibitors, but also combinations of two or three active substances, i.e.:
• Betamimetics with corticosteroids, PDE4-inhibitors, CRTH2-inhibitors or
LTD4-antagonists,
• Anticholinergics with betamimetics, corticosteroids, PDE4-inhibitors,
CRTH2-inhibitors or LTD4-antagonists,
• Corticosteroids with PDE4-inhibitors, CRTH2-inhibitors or LTD4-antagonists
• PDE4-inhibitors with CRTH2-inhibitors or LTD4-antagonists
• CRTH2-inhibitors with LTD4-antagonists.
INDICATIONS
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:
1. 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; alpha 1 -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 and thrombotic disorders of the lung vasculature, and pulmonary hypertension; antitussive activity including treatment of chronic cough associated with inflammatory and secretory conditions of the airways, and iatrogenic cough; acute and chronic rhinitis including rhinitis medicamentosa, and vasomotor rhinitis; perennial and seasonal allergic rhinitis including rhinitis nervosa (hay fever); nasal polyposis; acute viral infection including the common cold, and infection due to respiratory syncytial virus, influenza, coronavirus (including SARS) and adenovirus; acute lung injury (ALI) and adult respiratory diseases syndrome (ARDS).
2. 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 skin cancer and other dysplastic lesions;
drug-induced disorders including fixed drug eruptions;
3. eyes: blepharitis; conjunctivitis, including perennial and vernal allergic conjunctivitis; iritis; anterior and posterior uveitis; choroiditis; autoimmune, degenerative or inflammatory disorders affecting the retina; ophthalmitis including sympathetic ophthalmitis; sarcoidosis; infections including viral , fungal, and bacterial;
4. genitourinary: nephritis including interstitial and glomerulonephritis; nephrotic syndrome; cystitis including acute and chronic (interstitial) cystitis and Hunner's ulcer; acute and chronic urethritis, prostatitis, epididymitis, oophoritis and salpingitis;
vulvo -vaginitis; Peyronie's disease; erectile dysfunction (both male and female); 5. allograft rejection: acute and chronic following, for example, transplantation of kidney, heart, liver, lung, bone marrow, skin or cornea or following blood transfusion; or chronic graft versus host disease;
6. other auto-immune and allergic disorders including rheumatoid arthritis, irritable bowel syndrome, systemic lupus erythematosus, multiple sclerosis, Hashimoto's thyroiditis, Graves' disease, Addison's disease, diabetes mellitus, idiopathic thrombocytopaenic purpura, eosinophilic fasciitis, hyper-IgE syndrome, antiphospholipid syndrome and Sazary syndrome;
7. oncology: treatment of common cancers including prostate, breast, lung, ovarian, pancreatic, bowel and colon, stomach, skin and brain tumors and malignancies affecting the bone marrow (including the leukaemias) and lymphoproliferative systems, such as Hodgkin's and non-Hodgkin's lymphoma; including the prevention and treatment of metastatic disease and tumour recurrences, and paraneoplastic syndromes; and,
8. 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.
For treatment of the above-described diseases and conditions, 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. For Example, for administration to a 70 kg person, 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. In any case 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.

Claims

WHAT WE CLAIM
1. Compounds of formula 1
Figure imgf000138_0001
wherein
A is phenyl or a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected independent from each other from the group consisting ofN, (0")-N+, O and S;
R is
Figure imgf000138_0002
Rla l is Ci_4-alkyl-, C3-6-cycloalkyl-, Ci_4-haloalkyl-, H2N-, Ci_4-alkyl-NH-, C3_6-cycloalkyl-NH-, (Ci_4-alkyl)2N-, (C3_6-cycloalkyl)(Ci_4-alkyl)N-, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, piperazinyl- Ci_4-alkyl-, Ci_4-alkyl-0-NH-, (Ci_4-alkyl-0)(Ci_4-alkyl)N-;
Rla 2 is H, NC-, 02N-, Ci_4-alkyl-S02-, aryl-S02-, (aromatic heteroring)-S02-, Ci_4-alkyl-0-(0)C-, Ci_4-alkyl-(0)C-, C3_6-cycloalkyl-(0)C-, H2N-(0)C-, Ci_4-alkyl-NH-(0)C-, C3_6-cycloalkyl-NH-(0)C-, (Ci_4-alkyl)2N-(0)C-, (C3_6-cycloalkyl)(Ci_4-alkyl)N-(0)C-, azetidinyl-(0)C-, pyrrolidinyl-(0)C- piperidinyl-(0)C-, morpholinyl-(0)C-, piperazinyl-(0)C-, piperazinyl-Ci_4 alkyl-(0)C-, Ci_4-alkyl-, C3_6-cycloalkyl-, Ci_4-haloalkyl-, aryl, heteroaryl;
R is H, Ci_4-alkyl-, C3_6-cycloalkyl-, Ci_4-haloalkyl- or halogen; and R are together C2_4-alkylene forming a carbocyclic heteroring; R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, wherein one element of the ring is optionally replaced by O; phenyl-Ci_4-alkyl-, (aromatic heteroring)-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring, wherein one, two or three elements are replaced by an element selected
independent from each other from the group consisting of N, O and S; each element of one of the rings optionally substituted with one or two groups selected from Ci_4-alkyl-, C3_6-cycloalkyl-, halogen, Ci_4-haloalkyl-, Ci_4-alkyl-0-, Ci_4-alkyl-
Figure imgf000139_0001
R3 is H or Ci-4-alkyl-;
R4 is phenyl, a five- or six-membered, aromatic heteroring containing one, two or three atoms selected from N, O and S; each optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, C3_6-cycloalkyl-, halogen or Ci_4-haloalkyl-;
R5 is H, Ci_4-alkyl-, Ci_4-haloalkyl-, C3_6-cycloalkyl-;
R6 is H or Ci_4-alkyl-;
R7 is H or Ci-4-alkyl-; or R6 and R7 are forming together a C2_5-alkylene, wherein one element of the formed ring is optionally replaced by O; or a pharmaceutically acceptable salt thereof.
2. Compounds of formula 1 , according to claim 2, wherein is phenyl or pyridinyl;
is
°VXl s Xl
R1aV V 1 or R1a"2N* a-1 Rla l is Ci_4-alkyl-;
Rla'2 is NC-;
RI b is H;
R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five- or six-membered, aromatic heteroring containing one or two nitrogen atoms, each of the above rings optionally substituted with
Ci_4-alkyl-, halogen or NC-;
R3 is methyl;
R4 is phenyl, a five- or six-membered, aromatic ring containing one or two nitrogen atom, each ring optionally substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-; is H;
is H;
is H; or a pharmaceutically acceptable salt thereof.
3. Compounds of formula 1 , according to one of the claims 1 or 2, wherein
A is phenyl or pyridinyl;
Rla is
R1 2N .A_ R
Rla l is Ci_4-alkyl-; Rla'2 is NC-;
RIb is H;
R2 is Ci_6-alkyl or a residue selected from the group consisting of C3_6-cycloalkyl-, phenyl-Ci_4-alkyl- and a five-membered, aromatic ring containing two nitrogen atoms; each optionally substituted with methyl or NC-;
R3 is methyl;
R4 is phenyl or a six-membered, aromatic heteroring containing one or two nitrogen atom, each ring substituted with one or two residues selected independently from each other from the group consisting of Ci_4-alkyl-, halogen or Ci_4-haloalkyl-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
4. Compounds of formula 1, according to one of the claims 1 to 3, wherein
A is phenyl or pyridinyl;
Rla is
Figure imgf000141_0001
Rla l is methyl, ethyl, i-propyl, n-propyl;
Rla'2 is NC-;
RIb is H; is ethyl, i-propyl, 2-butyl, cyclo butyl, 1-methylpyrazolyl, or benzyl or 4-NC- benzyl; is methyl;
is phenyl or pyridinyl, each ring substituted with one or two residues selected independently from each other from the group consisting of methyl, ethyl, propyl, F, Br, CI, F2HC- or F3C-;
R5 is H;
R6 is H;
R7 is H; or a pharmaceutically acceptable salt thereof.
5. Compounds of formula 1, according to one of the claims 1 to 4, wherein
A is phenyl;
R la
IS
Figure imgf000142_0001
R la.l is methyl;
R la.2
is NC-;
R lb is H; is ethyl, i-propyl, cyclo butyl, 1-methylpyrazolyl, benzyl or 4-NC-benzyl;
R3 is methyl;
R4 is phenyl or pyridinyl, each ring substituted with one or two residues selected independently from each other from the group consisting of F, F2HC- or F3C-; R5 isH;
R6 isH;
R7 isH; or a pharmaceutically acceptable salt thereof.
6. Compounds of formula 1, according to one of the claims 1 to5, wherein A is phenyl;
R la
IS
Figure imgf000143_0001
la.l
R is methyl;
la.2
R is NC-;
R lb isH; is selected from a roup consisting of
Figure imgf000143_0002
is methyl;
is selected from a roup consisting of
Figure imgf000143_0003
R5 isH;
R6 isH; R7 is H; or a pharmaceutically acceptable salt thereof.
7. A compound of formula 1 according to any one claims 1 to 6 for use as a medicament.
8. A compound of formula 1 according to any one claims 1 to 6 for use as a medicament for the treatment of asthma and allergic diseases, gastrointestinal inflammatory diseases, eosinophilic diseases, chronic obstructive pulmonary disease, infection by pathogenic microbes and rheumatoid arthritis.
9. Pharmaceutical composition, characterised in that it contains one or more compounds of formula 1 according to any one of claims 1 to 6 or a pharmaceutically active salt thereof.
10. Method of treatment or prevention of diseases in which neutrophil elastase inhibitors have a therapeutic benefit, which method comprises administration of a therapeutically or preventively effective amount of a compounds of formula 1 according to one of claims 1 to 6 to a patient in need thereof.
11. A pharmaceutical composition comprising additionally to a compound of formula 1, according to any one of claims 1 to 6, a pharmaceutically active compound selected from the group consisting of betamimetics, anticholinergics, corticosteroids, PDE4-inhibitors, LTD4-antagonists, EGFR-inhibitors, Cathepsin C inhibitors, CRTH2 inhibitors,
5-LO-inhibitors, Histamine receptor antagonists and SYK- inhibitors, but also combinations of two or three active substances.
PCT/EP2013/067425 2012-08-23 2013-08-22 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity WO2014029830A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2015527913A JP6319747B2 (en) 2012-08-23 2013-08-22 Substituted 4-pyridones and their use as neutrophil elastase activity inhibitors
EP13756358.1A EP2888242B1 (en) 2012-08-23 2013-08-22 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Applications Claiming Priority (2)

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

Publications (1)

Publication Number Publication Date
WO2014029830A1 true WO2014029830A1 (en) 2014-02-27

Family

ID=46963420

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/067425 WO2014029830A1 (en) 2012-08-23 2013-08-22 Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity

Country Status (7)

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

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9290457B2 (en) 2014-07-31 2016-03-22 Boehringer Ingelheim International Gmbh Substituted dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
WO2016124557A1 (en) 2015-02-05 2016-08-11 Bayer Cropscience Aktiengesellschaft 2-(het)aryl-substituted condensed bicyclic heterocycle derivatives as pest control agents
US9440930B2 (en) 2014-07-31 2016-09-13 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9458113B2 (en) 2014-07-31 2016-10-04 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9475779B2 (en) 2014-07-31 2016-10-25 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9657015B2 (en) 2014-07-31 2017-05-23 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
WO2021053058A1 (en) 2019-09-17 2021-03-25 Mereo Biopharma 4 Limited Alvelestat for use in the treatment of graft rejection, bronchiolitis obliterans syndrome and graft versus host disease
EP3896065A1 (en) 2015-08-07 2021-10-20 Bayer CropScience Aktiengesellschaft 2-(het)aryl-substituted condensed heterocycle derivatives as pesticides
WO2021209740A1 (en) 2020-04-16 2021-10-21 Mereo Biopharma 4 Limited Methods involving neutrophil elastase inhibitor alvelestat for treating coronavirus infection
WO2023067103A1 (en) 2021-10-20 2023-04-27 Mereo Biopharma 4 Limited Neutrophil elastase inhibitors for use in the treatment of fibrosis

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201900604A (en) 2017-05-18 2019-01-01 印度商Pi工業公司 Novel bismuth compound

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030087940A1 (en) 2000-11-30 2003-05-08 Claiborne Akiyo K. Farnesyltransferase inhibitors
WO2004043924A1 (en) 2002-11-12 2004-05-27 Astrazeneca Ab 2-pyridone derivatives as inhibitors of neutrophile elastase
WO2005026124A1 (en) 2003-09-18 2005-03-24 Astrazeneca Ab 2-pyridone derivatives as netrophil elastase inhibitors and their use
WO2005026123A1 (en) 2003-09-18 2005-03-24 Astrazeneca Ab 2-pyridone derivatives as neutrophil elastase inhibitors and their use
WO2006098684A1 (en) 2005-03-16 2006-09-21 Astrazeneca Ab Novel compounds ii 2-pyridine derivatives as inhibitors of neutrophile elastase.
WO2007003960A1 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007093452A1 (en) 2006-02-16 2007-08-23 Polimeri Europa S.P.A. Improved process for the preparation of stable nitroxyl radicals
WO2007129963A1 (en) 2006-05-08 2007-11-15 Astrazeneca Ab 2-pyrazinone derivatives for the treatment of disease or condition in which inhibition of neutrophil elastase activity is beneficial.
WO2007129962A1 (en) 2006-05-08 2007-11-15 Astrazeneca Ab 2-pyridone derivatives for the treatment of disease or condition in which inhibition of neutrophil elastase activity is beneficial.
WO2009058076A1 (en) 2007-11-02 2009-05-07 Astrazeneca Ab 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
WO2009061271A1 (en) 2007-11-06 2009-05-14 Astrazeneca Ab Some 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
WO2009094417A1 (en) * 2008-01-23 2009-07-30 Bristol-Myers Squibb Company 4-pyridinone compounds and their use for cancer
WO2009156336A1 (en) 2008-06-23 2009-12-30 Basf Se Sulfoximinamide compounds for combating animal pests
WO2010094964A1 (en) 2009-02-20 2010-08-26 Astrazeneca Ab Tosylate salt of a 5-pyrazolyl-2-pyridone derivative, useful in the treatment of copd
WO2010133973A1 (en) 2009-05-20 2010-11-25 Universite Lille 2 Droit Et Sante 1,4 dihydropyridine derivatives and their uses
WO2011039528A1 (en) 2009-10-02 2011-04-07 Astrazeneca Ab 2-pyridone compounds used as inhibitors of neutrophil elastase
WO2011110852A1 (en) 2010-03-10 2011-09-15 Astrazeneca Ab Polymorphic forms of 6- [2- (4 -cyanophenyl) - 2h - pyrazol - 3 - yl] - 5 -methyl - 3 - oxo - 4 - (trifluoromethyl - phenyl) 3,4-dihydropyrazine-2-carboxylic acid ethylamide

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4844732A (en) * 1985-10-24 1989-07-04 Daicel Chemical Industries Ltd. Pyridine-3-carboxamide derivatives

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030087940A1 (en) 2000-11-30 2003-05-08 Claiborne Akiyo K. Farnesyltransferase inhibitors
WO2004043924A1 (en) 2002-11-12 2004-05-27 Astrazeneca Ab 2-pyridone derivatives as inhibitors of neutrophile elastase
EP1562902A1 (en) * 2002-11-12 2005-08-17 AstraZeneca AB 2-pyridone derivatives as inhibitors of neutrophile elastase
WO2005026124A1 (en) 2003-09-18 2005-03-24 Astrazeneca Ab 2-pyridone derivatives as netrophil elastase inhibitors and their use
WO2005026123A1 (en) 2003-09-18 2005-03-24 Astrazeneca Ab 2-pyridone derivatives as neutrophil elastase inhibitors and their use
WO2006098684A1 (en) 2005-03-16 2006-09-21 Astrazeneca Ab Novel compounds ii 2-pyridine derivatives as inhibitors of neutrophile elastase.
WO2006098683A1 (en) 2005-03-16 2006-09-21 Astrazeneca Ab 2-pyridine derivatives as inhibitors of neutrophile elastase
WO2007003960A1 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007093452A1 (en) 2006-02-16 2007-08-23 Polimeri Europa S.P.A. Improved process for the preparation of stable nitroxyl radicals
WO2007129962A1 (en) 2006-05-08 2007-11-15 Astrazeneca Ab 2-pyridone derivatives for the treatment of disease or condition in which inhibition of neutrophil elastase activity is beneficial.
WO2007129963A1 (en) 2006-05-08 2007-11-15 Astrazeneca Ab 2-pyrazinone derivatives for the treatment of disease or condition in which inhibition of neutrophil elastase activity is beneficial.
WO2009058076A1 (en) 2007-11-02 2009-05-07 Astrazeneca Ab 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
WO2009061271A1 (en) 2007-11-06 2009-05-14 Astrazeneca Ab Some 2-pyrazinone derivatives and their use as inhibitors of neutrophile elastase
WO2009094417A1 (en) * 2008-01-23 2009-07-30 Bristol-Myers Squibb Company 4-pyridinone compounds and their use for cancer
WO2009156336A1 (en) 2008-06-23 2009-12-30 Basf Se Sulfoximinamide compounds for combating animal pests
WO2010094964A1 (en) 2009-02-20 2010-08-26 Astrazeneca Ab Tosylate salt of a 5-pyrazolyl-2-pyridone derivative, useful in the treatment of copd
WO2010133973A1 (en) 2009-05-20 2010-11-25 Universite Lille 2 Droit Et Sante 1,4 dihydropyridine derivatives and their uses
EP2261211A1 (en) * 2009-05-20 2010-12-15 Université de Lille 2 Droit et Santé 1,4-dihydropyridine derivatives and their uses
WO2011039528A1 (en) 2009-10-02 2011-04-07 Astrazeneca Ab 2-pyridone compounds used as inhibitors of neutrophil elastase
WO2011110852A1 (en) 2010-03-10 2011-09-15 Astrazeneca Ab Polymorphic forms of 6- [2- (4 -cyanophenyl) - 2h - pyrazol - 3 - yl] - 5 -methyl - 3 - oxo - 4 - (trifluoromethyl - phenyl) 3,4-dihydropyrazine-2-carboxylic acid ethylamide

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"A Textbook of Drug Design and Development", 1991, GORDON & BREACH, article "Design and Applications of Prodrugs"
"Bioreversible Carriers in Drug Design", 1987, ELSEVIER
"Burger's Medicinal Chemistry and Drug Discovery", vol. 1, 1995, JOHN WILEY & SONS, pages: 172 - 178,949-
"Design of Prodrugs", 1985, ELSEVIER
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Pro-Drugs as Novel Delivery Systems", 1975, AM. CHEM. SOC.
"Prodrugs: Topical and Ocular Drug Delivery", 1998, MARCEL DEKKER
BERGE, S.M. ET AL.: "Pharmaceutical salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
BOLM ET AL., CHEM. LETT., vol. 33, 2004, pages 482 - 487
BOLM ET AL., J. ORG. CHEM., vol. 75, 2010, pages 3301 - 3310
BOLM ET AL., ORG. LETT., vol. 9, 2007, pages 2951 - 2954
BOLM ET AL., ORG. LETT., vol. 9, 2007, pages 3809 - 3811
BOLM. ET AL., ORG. LETT., vol. 6, 2004, pages 1305 - 1307
E. KERNS; L. DI: "Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization", 2008, ELSEVIER
GNAMM ET AL., BIOORG. MED. CHEM. LETT., vol. 12, 2012, pages 3800 - 3806
GNAMM ET AL., BIOORG. MED. CHEM. LETT., vol. 22, 2012, pages 3800 - 3806
JOHNSON ET AL., J. ORG. CHEM., vol. 44, 1975, pages 2055 - 2061
JOHNSON ET AL., J. ORG. CHEM., vol. 48, 1983, pages 1 - 3
P. SJÖ, FUTURE MED. CHEM., vol. 4, 2012, pages 651 - 660
REGGELIN ET AL., SYNTHESIS, 2000, pages 1 - 64
T. STEVENS ET AL., J. PHARM. EXP. THER., vol. 339, 2011, pages 313 - 320
TREMBLAY ET AL., CHEST, vol. 121, 2002, pages 582 - 588
VENKATRAMANI ET AL., J. HET. CHEM., vol. 30, 1993, pages 723 - 738

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3539952A1 (en) 2014-07-31 2019-09-18 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
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
US9657015B2 (en) 2014-07-31 2017-05-23 Boehringer Ingelheim International Gmbh Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
US9290457B2 (en) 2014-07-31 2016-03-22 Boehringer Ingelheim International Gmbh Substituted dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
EP3604308A1 (en) 2014-07-31 2020-02-05 Boehringer Ingelheim International GmbH Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
WO2016124557A1 (en) 2015-02-05 2016-08-11 Bayer Cropscience Aktiengesellschaft 2-(het)aryl-substituted condensed bicyclic heterocycle derivatives as pest control agents
EP3896065A1 (en) 2015-08-07 2021-10-20 Bayer CropScience Aktiengesellschaft 2-(het)aryl-substituted condensed heterocycle derivatives as pesticides
EP3896066A2 (en) 2015-08-07 2021-10-20 Bayer CropScience Aktiengesellschaft 2-(het)aryl-substituted condensed heterocycle derivatives as pesticides
WO2021053058A1 (en) 2019-09-17 2021-03-25 Mereo Biopharma 4 Limited Alvelestat for use in the treatment of graft rejection, bronchiolitis obliterans syndrome and graft versus host disease
WO2021209740A1 (en) 2020-04-16 2021-10-21 Mereo Biopharma 4 Limited Methods involving neutrophil elastase inhibitor alvelestat for treating coronavirus infection
WO2023067103A1 (en) 2021-10-20 2023-04-27 Mereo Biopharma 4 Limited Neutrophil elastase inhibitors for use in the treatment of fibrosis

Also Published As

Publication number Publication date
US20160130265A1 (en) 2016-05-12
TW201422586A (en) 2014-06-16
AR092205A1 (en) 2015-04-08
US9346794B1 (en) 2016-05-24
EP2888242B1 (en) 2016-07-13
US20140057926A1 (en) 2014-02-27
EP2888242A1 (en) 2015-07-01
JP2015526453A (en) 2015-09-10
JP6319747B2 (en) 2018-05-09
UY34989A (en) 2014-02-28

Similar Documents

Publication Publication Date Title
EP2888242B1 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
EP2888249B1 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
US9290459B1 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
EP2888248B1 (en) Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity
EP2964617B1 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
EP3107911B1 (en) Substituted pyridones and pyrazinones and their use as inhibitors of neutrophil elastase activity
EP3174860A1 (en) Substituted dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
WO2016016368A1 (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
CA2956745C (en) Substituted bicyclic dihydropyrimidinones and their use as inhibitors of neutrophil elastase activity
EP3174876A1 (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
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13756358

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2013756358

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2013756358

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015527913

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE