HETEROARYL SYK INHIBITORS

The invention relates to new substituted heteroaryls of formula 1 wherein A is selected from the group consisting of N and CH D is selected from the group consisting of CH, N, NH, E is C, T is selected from the group consisting of C and N, G is selected from the group consisting of C and N, and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system, and wherein R1, M and R3 are defined according to claim 1, and to the above compounds for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

The invention relates to new substituted heteroaryls of formula 1

or of formula 1′

wherein

A is selected from the group consisting of N and CH

D is selected from the group consisting of CH, N, NH,

E is C,

T is selected from the group consisting of C and N,

G is selected from the group consisting of C and N,

and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,

and wherein

M, R3 and R1 are defined as in claim 1. Further the invention relates to the above compounds of formula 1 or of formula 1′ for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.

1. BACKGROUND TO THE INVENTION

1.1 SYK-Inhibitors

The present invention describes new compounds that inhibit the protein kinase Syk (spleen tyrosine kinase), the preparation and formulation thereof and their use for preparing a medicament.

Syk is an intracellular tyrosine kinase that has an important mediator function in the signal transduction of different receptors in B-cells, mast cells, monocytes, macrophages, neutrophils, T-cells, dendritic cells and epithelial cells. The receptors in which Syk performs an important function in signal transduction include for example the receptors for IgE (FcεRI) and IgG (FcγR1) on mast cells and B cells, the B-cell receptor (BCR) and the T-cell receptor (TCR) on B- and T-cells, the ICAM1 receptor (ICAM1R) on epithelial cells of the respiratory tract, the DAP12-receptor on natural killer cells, dendritic cells and osteoclasts, the dectin 1-receptor on a subpopulation of T-helper cells (Th-17 cells), as well as the integrin receptors for ⊕1-, β2- and β3-integrins on neutrophils, monocytes and macrophages (Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; Ulanova et al.; Expert Opion. Ther. Target (2005) 9(5); 901-921; Wang et al.; J. Immunol. (2006) 177, 6859-6870; Leib and Gut-Landmann et al.; Nature Immunology (2007) 8, 630-638; Slack et al., European J. Immunol. (2007) 37, 1600-1612). The molecular processes are described best for the signal transduction of the FcεRI. In mast cells the binding of IgE to FcεRI causes the cross-linking of IgE-receptors and the recruiting and activation of Lyn (a tyrosine kinase from the Src family). Active Lyn phoshorylates so-called ITAM motifs, which are present in many of the receptors listed above, and thereby generates binding sites for the SH2-domain of Syk. As a result of the binding to the ITAM motif Syk is activated and then phosphorylates various substrates which are needed for the release of allergic and inflammatory mediators such as e.g. histamine and β-hexosamidase (βHA), as well as for the synthesis of lipid mediators, such as e.g. prostaglandins and leukotrienes.

In view of its central function in different signal transduction pathways Syk has been discussed as a therapeutic target for different diseases such as e.g. allergic rhinitis, asthma, autoimmune diseases, rheumatoid arthritis, osteopenia, osteoporosis, COPD and various leukaemias and lymphomas (Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; Ulanova et al.; Expert Opion. Ther. Target (2005) 9(5); 901-921; Sigh and Masuda. Annual Reports in Medicinal Chemistry (2007) Vol 42; 379-391; Bajpai et al.; Expert Opin. Investig. Drugs (2008) Vol 15 (5); 641-659; Masuda and Schmitz; PPT (2008) Vol 21; 461-467; Riccaboni et al., Drug Discovery Today (2010) Vol 00 (0); 517-530; Efremov and Luarenti, Expert Opin Investig Drugs. (2011) 20(5):623-36).

Allergic rhinitis and asthma are diseases associated with allergic reactions and inflammatory processes and involving different cell types such as e.g. Mast cells, eosinophils, T-cells and dendritic cells. After exposure to allergens has occurred, the high affinity immunoglobulin receptors for IgE (FcεRI) and IgG (FcγR1) are activated and induce the release of pro-inflammatory mediators and bronchoconstrictors. An inhibitor of the Syk kinase activity should thus be able to inhibit these steps.

Rheumatoid arthritis (RA) is an autoimmune disease in which the bones and ligaments structures surrounding the joints are progressively destroyed. In the pathophysiology of RA, B-cells play a significant role, as has been demonstrated for example by the therapeutic use of rituximab, a B cell-depleting antibody. In addition to the function of Syk in the signal transduction of the BCR (which after being stimulated also induces the release of pro-inflammatory mediators), Syk also plays an important part in the maturation and proliferation of B cells (Cheng et al. Nature (1995) 378, 303-306, Cornall et al., PNAS (2000) 97(4), 1713-1718). An inhibitor of the Syk kinase activity may thus offer a therapeutic option for the treatment of autoimmune diseases such as RA and diseases with an increased proliferation of B cells, such as e.g. B-cell lymphomas.

Chronic obstructive pulmonary disease (COPD) is characterised by a successive deterioration in lung function and chronic inflammation of the airways, which is initiated and produced by noxious substances of all kinds and contributes to the maintenance of the course of the disease. At a cellular level, in COPD there is in particular a multiplication of T-lymphocytes, neutrophils, granulocytes and macrophages. In particular, there is an increase in the number of CD8-positive lymphocytes, that is directly connected with the impairment of lung function. Another characteristic of COPD are acute deteriorations in lung function (exacerbations), characterised by viral (e.g. Rhinovirus), or bacterial (e.g. Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis) infections.

In view of the pro-inflammatory function of Syk in macrophages, T-cells and neutrophils as described above (see: Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; and references cited therein) an inhibitor of the Syk kinase activity could be a new therapeutic approach to the treatment of the inflammatory processes that underlie COPD. It has also been shown that Syk in epithelial cells of the respiratory tract is involved in the ICAM1R-mediated uptake and subsequent replication of the Rhinovirus and that a si-RNA against Syk blocks these steps (Wang et al.; J. Immunol. (2006) 177, 6859-6870; Lau et al.; J. Immunol. (2008) 180, 870-880). Thus, an inhibitor of the Syk kinase activity could also be used therapeutically in exacerbations caused by Rhinoviruses.

Various studies suggest that Syk is involved in the malignant transformation of lymphocytes (summarised in Sigh and Masuda, Annual Reports in Medicinal Chemistry (2007) Vol 42; 379-391). A TEL-Syk fusion protein with a constitutive Syk activity transformed B cells of a patient with myelodysplastic syndrome, a constitutively active ITK-Syk fusion protein was isolated from patients with peripheral T-cell lymphomas (PTCL). Moreover, constitutively active Syk was found in B-cell lymphoma cells of patients, especially in B-lineage acute lymphoblastic leukemia (B-ALL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphomas and B cell Non-Hodgkin Lymphomas (NHLs) as well as in acute myeloid leukemia (AML). On the basis of these data it seems that Syk is a proto-oncogene in haematopoietic cells and represents a potential target for the treatment of certain leukaemias and lymphomas.

Idiophathic thrombocytopenic purpura (ITP) is an autoimmune disease in which IgG autoantibodies against antigens present on platelets bind to and destroy platelets. Patients with ITP have an accelerated clearence of circulating IgG-coated platelets via macrophages in the spleen and the liver. In view of the pro-inflammatory FcγR-mediated function of Syk in macrophages an inhibitor of Syk is considered to have a therapeutic benefit in FcγR-mediated cytopenias like ITP. Indeed the Syk inhibitor R788 (R406) improved platelet counts in a single center, oben label study in patients with ITP (Podolanczuk et al; Blood (2009) 113, 3154-3169).

Bullous pemphigoid (Ujiie et al. Journal of Dermatology 2010; 37: 194-204) is a chronic, autoimmune, subepidermal, blistering skin disease that rarely involves mucous membranes. Bullous pemphigoid is characterized by the presence of immunoglobulin G (IgG) autoantibodies specific for the hemidesmosomal bullous pemphigoid antigens BP230 (BPAg1) and BP180 (BPAg2). Pemphigus vulgaris (Venugopal et al. Dermatol. Clin. 2011; 29:373-80) is a chronic blistering skin disease with skin lesions that are rarely pruritic, but which are often painful. Pemphigus vulgaris is an autoimmune disease caused by IgG autoantibodies directed against both desmoglein 1 and desmoglein 3 resulting in the loss of cohesion between keratinocytes in the epidermis. It is characterized by extensive flaccid blisters and mucocutaneous erosions. In both diseases IgG autoantibodies bind to Fc receptor gamma (FcRγ) and activate FcRγ and downstream signaling via Syk kinase. Thus, an inhibitor of the Syk kinase activity which blocks downstream signalling of the FcRg could be used therapeutically to treat patients with bullous pemphigoid and pemphigus vulgaris.

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease which can affect basically any organ of the body. It is characterised by a multisystem inflammation of the microvascular and the presence of autoantibodies. FcγR-deficient mice are protected from several aspects of SLE in disease-related preclinical models, suggesting that an inhibitor of Syk can have a therapeutic benefit in SLE in view of the pro-inflammatory FcγR-mediated function of Syk in various cells.

1.2 Prior Art

1,6-Naphthyridines are known as SYK-inhibitors. For example U.S. Pat. No. 3,928,367, U.S. Pat. No. 4,017,500, U.S. Pat. No. 4,115,395 and U.S. Pat. No. 4,260,759 describe 5-amino-1,6-naphthyridines with an antifungal and antibacterial activity. Further, WO 9918077 describes 5-piperazinyl-1,6-naphthyridines as serotonin antagonists. Additionally, U.S. Pat. No. 7,321,041 describes substituted 1,6-naphthyridines as SYK-inhibitors, however these 1,6-naphthyridines have a completely different substitution pattern from the compounds according to the invention. Also WO 2011092128 discloses 1,6-naphthyridines which are substituted in 5- and in 7-position.

In WO 2012/167733, WO 2012/167423 and in WO 2012/123312 other naphthryidine derivatives such as pyrido[3,4-b]pyrazines which were also substituted in 5- and in 7-position have been disclosed as SYK-inhibitors.

Additionally, WO 01/83485 discloses substituted imidazopyrimidines and triazolopyrimidines as SYK-inhibitors, whereas WO 2008/113469 discloses substituted imidazo- and triazolopyrimidines as GSK 3β-inhibitors.

Also quinolones are known as SYK-inhibitors. For instance, WO 2006038041 and WO 2013014060 both disclose quinoline-compounds which are substituted in the 5- and 7-position, however the substitution pattern—in particular in the 7-position—is completely different from the one of the compounds of formula 1 of the instant invention.

Surprisingly it has now been found that the compounds of formulas 1 and 1′ and in particular the compounds of formulas 1d, 1d′, 1e, 1e′, 1f or 1f′ are particularly suitable for the treatment of respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases, autoimmune diseases, inflammatory diseases and diseases of the peripheral or central nervous system, particularly for the treatment of asthma, allergic rhinitis, rheumatoid arthritis, allergic dermatitis, lupus erythematosus (SLE) and COPD, in particular because all these compounds of the present invention show the following desired capacities:

    • high SYK inhibition (reflected by “low” IC50-values with respect to SYK-inhibition)
    • very low inhibition of the kinase Aurora B (reflected by “high” IC50-values with respect to inhibition of AURB)
    • low inhibition of the kinase FLT-3 (reflected by “high” IC50-values with respect to inhibition of FLT-3)
    • low inhibition of the kinase GSK3β (reflected by “high” IC50-values with respect to inhibition of GSK3β)

This was completely surprising for a person skilled in the art, since the compounds of formula 1 and 1′ of the instant invention have several significant structural differences compared to the previously known prior art compounds. For instance the compounds of formula 1 and 1′ of the instant invention differ from the previously known 1,6-naphthyridines, quinolones, pyrido[3,4-b]pyrazines, imidazopyrimidines and triazolopyrimidines therein that they combine the following features:

    • they all possess diverse core modifications in the central bicyclic heteroaromatic ring system
    • that they all have a methyl-group-substitution attached to position E in formula 1 and/or 1′ and
    • that they all possess a residue of formula T

preferably a residue of formula T′

2. DESCRIPTION OF THE INVENTION

The present invention concerns compounds of formula 1

wherein

A is selected from the group consisting of N and CH

D is selected from the group consisting of CH, N, NH,

E is C,

T is selected from the group consisting of C and N,

G is selected from the group consisting of C and N,

and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
and wherein

M is selected from the group consisting of —CH2—, —O—, —NH— and —N(C1-4-alkyl)-;

R3 is selected from the group consisting of methyl and ethyl;

Het is selected from the group consisting of

a five- to six-membered monocyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O,

and a nine- to eleven-membered bicyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;

Hetaryl is selected from the group consisting of

a five- to six-membered monocyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;

and a nine- to eleven-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;

and wherein

R1 is selected from the group consisting of

Het and Hetaryl;

which is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, —OH, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2, —NH(CH3), —N(CH3)2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, —OH, halogen and C1-3-alkyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

A preferred embodiment of the instant invention relates to the aforementioned compounds of formula 1′

wherein residues A, D, E, T, G, Het, Hetaryl, R1 and R3 are defined as mentioned above, and the pharmaceutically acceptable salts of the aforementioned compounds.

In a further preferred embodiment the instant invention concerns the aforementioned compounds of formula 1 or of formula 1′,

wherein

M is —CH2

and wherein

R3 is methyl,

and wherein

R1 is selected from the group consisting of

Het and Hetaryl;

which is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In a further preferred embodiment the invention concerns compounds of formula 1d

or of formula 1d′,

wherein

A is selected from the group consisting of N and CH,

R1 is selected from the group consisting of

Het and Hetaryl;

which is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—-(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In another preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1 d or formula 1d′, wherein

A is CH

and wherein

R1 is either

    • a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S,
    • or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In another preferred embodiment the instant invention relates to the above compounds of the aformentioned formula 1d or formula 1d′, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl, three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—N HCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O-(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In a particularly preferred embodiment the instant invention relates to the the above compound of the aforementioned formula 1d or formula 1d′, which is

and the pharmaceutically acceptable salts of the aforementioned compound.

In another preferred embodiment the instant invention relates to compounds of formula 1e,

or of formula 1e′,

wherein

A is selected from the group consisting of N and CH

R1 is selected from the group consisting of

Het and Hetaryl;

which is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—N HCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

In a further preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1e or formula 1e′, wherein

A is CH

and wherein

R1 is

    • a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S,
    • or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In another preferred embodiment the instant invention concerns the above compounds of the aforementioned formula 1e or formula 1e′, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl,three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In a particularly preferred embodiment the instant invention relates to the above compound of formula 1e or of formula 1e′, which is

and the pharmaceutically acceptable salts of the aforementioned compound.

In a further preferred embodiment the instant invention refers to compounds of formula 1f or of 1f′

or of formula 1f′ according to claim 2,

wherein

R1 is selected from the group consisting of

Het and Hetaryl;

which is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In a further preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1f or formula 1f′, wherein

R1 is either

    • a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S,
    • or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X,

whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In a further preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1f or formula 1f′, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,

wherein this R1—residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,

whereby each Z is a substituent selected from the group consisting of

—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl,three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,

which is optionally further substituted by one, two or three substituents X, whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,

whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,

and the pharmaceutically acceptable salts of the aforementioned compounds.

In another particularly preferred embodiment the instant invention relates to the above compound of formula 1f or 1f′, which is

and the pharmaceutically acceptable salts thereof.

In a further aspect the instant invention refers to an intermediate compound selected from the group consisting of formula 6

of formula 7

of formula 8

and of formula 11

    • wherein E, D, G, T and R1 are defined as in claim 1 and wherein Hal is Cl or Br and wherein PG is a protecting group selected from the group consisting of benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl.

In a further aspect the instant invention refers to an intermediate compound of formula 10.3

In a further aspect the instant invention refers to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) for the treatment of a disease which can be treated by inhibition of the SYK enzyme.

In another preferred aspect the instant invention relates to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) for the treatment of a disease selected from the group consisting of allergic rhinitis, asthma, COPD, adult respiratory distress syndrome, bronchitis, B-cell lymphoma, dermatitis and contact dermatitis, allergic dermatitis, allergic rhinoconjunctivitis, rheumatoid arthritis, anti-phospholipid syndrome, Berger's disease, Evans's syndrome, ulcerative colitis,allergic antibody-based glomerulonephritis, granulocytopenia, Goodpasture's syndrome, hepatitis, Henoch-Schönlein purpura, hypersensitivity vasculitis, immunohaemolytic anaemia, autoimmune haemolytic anemia, idiopathic thrombocytopenic purpura, Kawasaki syndrome, allergic conjunctivitis, lupus erythematodes, lupus nephritis, capsule cell lymphoma, neutropenia, non-familial lateral sclerosis, artheriosclerosis, Crohn's disease, multiple sclerosis, myasthenia gravis, osteoporosis, osteolytic diseases, osteopenia, psoriasis, Sjogren's syndrome, sclerodermy, T-cell lymphoma, urticaria/angiooedema, Wegener's granulomatosis and coeliac disease.

In another preferred aspect the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, adult respiratory distress syndrome, bronchitis, allergic dermatitis, contact dermatitis, idiopathic thrombocytopenic purpura, rheumatoid arthritis, lupus erythematodes, lupus nephritis and allergic rhinoconjunctivitis.

In another particularly preferred aspect the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.

In another preferred aspect the instant invention concerns pharmaceutical formulations which contain one or more of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) and a pharmaceutically acceptable excipient.

In another preferred aspect the instant invention concerns pharmaceutical formulations which contain one or more compounds of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1d, 1d′, 1e, 1e′, 1f or 1f′) in combination with an active substance selected from the group consisting of anticholinergics, betamimetics, corticosteroids, PDE4-inhibitors, EGFR-inhibitors, LTD4-antagonists, CCR3-inhibitors, iNOS-inhibitors, CRTH2-antagonists, HMG-CoA reductase inhibitors and NSAIDs.

3. TERMS AND DEFINITIONS USED

Unless stated otherwise, all the substituents are independent of one another. If for example a number of C1-6-alkyl groups are possible substituents at a group, in the case of three substituents, for example, C1-6-alkyl could represent, independently of one another, a methyl, an n-propyl and a tert-butyl.

Within the scope of this application, in the definition of possible substituents, these may also be presented in the form of a structural formula. An asterisk (*) in the structural formula of the substituent is to be understood as being the linking point to the rest of the molecule. Mor3eover, the atom of the substituent following the linking point is understood as being the atom in position number 1. Thus for example the groups N-piperidinyl (I), 4-piperidinyl (II), 2-tolyl (III), 3-tolyl (IV) and 4-tolyl (V) are represented as follows:

If there is no asterisk (*) in the structural formula of the substituent, each hydrogen atom may be removed at the substituent and the valency thus freed may serve as a binding site to the rest of a molecule. Thus, for example, VI

may represent 2-tolyl, 3-tolyl, 4-tolyl and benzyl.

Alternatively to the * within the scope of this application X1 is also understood as being the linking point of the group R1 to the structure of formula 1 and X2 as being the linking point of the group R2 to the structure of formula 1.

By the term “C1-6-alkyl” (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms and by the term “C1-3-alkyl” are meant branched and unbranched alkyl groups with 1 to 3 carbon atoms. “C1-4-alkyl” accordingly denotes branched and unbranched alkyl groups with 1 to 4 carbon atoms. Alkyl groups with 1 to 4 carbon atoms are preferred. Examples of these include: methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl or hexyl. The abbreviations Me, Et, n-Pr, i-Pr, n-Bu, i-Bu, t-Bu, etc., may also optionally be used for the above-mentioned groups. Unless stated otherwise, the definitions propyl, butyl, pentyl and hexyl include all the possible isomeric forms of the groups in question. Thus, for example, propyl includes n-propyl and iso-propyl, butyl includes iso-butyl, sec-butyl and tert-butyl etc.

By the term “C1-6-alkylene” (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms and by the term “C1-4-alkylene” are meant branched and unbranched alkylene groups with 1 to 4 carbon atoms. Alkylene groups with 1 to 4 carbon atoms are preferred. Examples of these include: methylene, ethylene, propylene, 1-methylethylene, butylene, 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene, pentylene, 1,1-dimethylpropylene, 2,2-dimethylpropylene, 1,2-dimethylpropylene, 1, 3-dimethylpropylene or hexylene. Unless stated otherwise, the definitions propylene, butylene, pentylene and hexylene include all the possible isomeric forms of the groups in question with the same number of carbons. Thus, for example, propyl includes also 1-methylethylene and butylene includes 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene.

If the carbon chain is substituted by a group which together with one or two carbon atoms of the alkylene chain forms a carbocyclic ring with 3, 5 or 6 carbon atoms, this includes, inter alia, the following examples of the rings:

By the term “C2-6-alkenyl” (including those which are part of other groups) are meant branched and unbranched alkenyl groups with 2 to 6 carbon atoms and by the term “C2-4-alkenyl” are meant branched and unbranched alkenyl groups with 2 to 4 carbon atoms, provided that they have at least one double bond. Alkenyl groups with 2 to 4 carbon atoms are preferred. Examples include: ethenyl or vinyl, propenyl, butenyl, pentenyl or hexenyl. Unless stated otherwise, the definitions propenyl, butenyl, pentenyl and hexenyl include all the possible isomeric forms of the groups in question. Thus, for example, propenyl includes 1-propenyl and 2-propenyl, butenyl includes 1-, 2- and 3-butenyl, 1-methyl-1-propenyl, 1-methyl-2-propenyl etc.

By the term “Cm-alkenylene” (including those which are part of other groups) are meant branched and unbranched alkenylene groups with 2 to 6 carbon atoms and by the term “C2-4-alkenylene” are meant branched and unbranched alkylene groups with 2 to 4 carbon atoms. Alkenylene groups with 2 to 4 carbon atoms are preferred. Examples of these include: ethenylene, propenylene, 1-methylethenylene, butenylene, 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene, pentenylene, 1,1-dimethylpropenylene, 2,2-dimethylpropenylene, 1,2-dimethylpropenylene, 1,3-dimethylpropenylene or hexenylene. Unless stated otherwise, the definitions propenylene, butenylene, pentenylene and hexenylene include all the possible isomeric forms of the groups in question with the same number of carbons. Thus, for example, propenyl also includes 1-methylethenylene and butenylene includes 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene.

By the term “aryl” (including those which are part of other groups) are meant aromatic ring systems with 6 or 10 carbon atoms. Examples include: phenyl or naphthyl, the preferred aryl group being phenyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.

By the term “aryl-C1-6-alkylene” (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by an aromatic ring system with 6 or 10 carbon atoms. Examples include: benzyl, 1- or 2-phenylethyl or 1- or 2-naphthylethyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.

By the term “heteroaryl-C1-6-alkylene” (including those which are part of other groups) are meant—even though they are already included under “aryl-C1-6-alkylene”—branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by a heteroaryl.

If not specifically defined otherwise, a heteroaryl of this kind includes five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed. The following are examples of five- or six-membered heterocyclic aromatic groups or bicyclic heteroaryl rings:

Unless otherwise stated, these heteroaryls may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.

The following are examples of heteroaryl-C1-6-alkylenes:

By the term “C1-6-haloalkyl” (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms, which are substituted by one or more halogen atoms. By the term “C1-4-alkyl” are meant branched and unbranched alkyl groups with 1 to 4 carbon atoms, which are substituted by one or more halogen atoms. Alkyl groups with 1 to 4 carbon atoms are preferred. Examples include: CF3, CHF2, CH2F, CH2CF3.

By the term “C3-7-cycloalkyl” (including those which are part of other groups) are meant cyclic alkyl groups with 3 to 7 carbon atoms, if not specifically defined otherwise. Examples include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Unless otherwise stated, the cyclic alkyl groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.

If not specifically defined otherwise, by the term “C3-10-cycloalkyl” are also meant monocyclic alkyl groups with 3 to 7 carbon atoms and also bicyclic alkyl groups with 7 to 10 carbon atoms, or monocyclic alkyl groups which are bridged by at least one C1-3-carbon bridge.

By the term “heterocyclic rings” or “heterocycle” are meant, unless stated otherwise, five-, six- or seven-membered, saturated, partially saturated or unsaturated heterocyclic rings which may contain one, two or three heteroatoms, selected from among oxygen, sulphur and nitrogen, while the ring may be linked to the molecule through a carbon atom or through a nitrogen atom, if there is one. Although included by the term “heterocyclic rings” or “heterocycles”, the term “saturated heterocyclic ring” refers to five-, six- or seven-membered saturated rings. Examples include:

Although included by the term “heterocyclic rings” or “heterocyclic group”, the term “partially saturated heterocyclic group” refers to five-, six- or seven-membered partially saturated rings which contain one or two double bonds, without so many double bonds being produced that an aromatic system is formed, unless specifically defined otherwise. Examples include:

Although included by the term “heterocyclic rings” or “heterocycles”, the term “heterocyclic aromatic rings” , “unsaturated heterocyclic group” or “heteroaryl” refers to five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed, unless not specifically defined otherwise. Examples of five- or six-membered heterocyclic aromatic groups include:

Unless otherwise mentioned, a heterocyclic ring (or heterocycle) may be provided with a keto group. Examples include:

Although covered by the term “cycloalkyl”, the term “bicyclic cycloalkyls” generally denotes eight-, nine- or ten-membered bicyclic carbon rings. Examples include

Although already included by the term “heterocycle”, the term “bicyclic heterocycles” generally denotes eight-, nine- or ten-membered bicyclic rings which may contain one or more heteroatoms, preferably 1-4, more preferably 1-3, even more preferably 1-2, particularly one heteroatom, selected from among oxygen, sulphur and nitrogen, unless not specifically defined otherwise. The ring may be linked to the molecule through a carbon atom of the ring or through a nitrogen atom of the ring, if there is one. Examples include:

Although already included by the term “aryl”, the term “bicyclic aryl” denotes a 5-10 membered, bicyclic aryl ring which contains sufficient conjugated double bonds to form an aromatic system. One example of a bicyclic aryl is naphthyl.

Although already included under “heteroaryl”, the term “bicyclic heteroaryl” denotes a 5-10 membered, bicyclic heteroaryl ring which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contains sufficient conjugated double bonds to form an aromatic system, unless specifically defined otherwise.

Although included by the term “bicyclic cycloalkyls” or “bicyclic aryl”, the term “fused cycloalkyl” or “fused aryl” denotes bicyclic rings wherein the bridge separating the rings denotes a direct single bond. The following are examples of a fused, bicyclic cycloalkyl:

Although included by the term “bicyclic heterocycles” or “bicyclic heteroaryls”, the term “fused bicyclic heterocycles” of “fused bicyclic heteroaryls” denotes bicyclic 5-10 membered heterorings which contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen and wherein the bridge separating the rings denotes a direct single bond. The “fused bicyclic heteroaryls” moreover contain sufficient conjugated double bonds to form an aromatic system. Examples include pyrrolizine, indole, indolizine, isoindole, indazole, purine, quinoline, isoquinoline, benzimidazole, benzofuran, benzopyran, benzothiazole, benzothiazole, benzoisothiazole, pyridopyrimidine, pteridine, pyrimidopyrimidine,

“Halogen” within the scope of the present invention denotes fluorine, chlorine, bromine or iodine. Unless stated to the contrary, fluorine, chlorine and bromine are regarded as preferred halogens.

Compounds of general formulas 1 or 1′ may have acid groups, mainly carboxyl groups, and/or basic groups such as e.g. amino functions. Compounds of general formulas 1 or 1′ may therefore be present as internal salts, as salts with pharmaceutically usable inorganic acids such as hydrochloric acid, sulphuric acid, phosphoric acid, sulphonic acid or organic acids (such as for example maleic acid, fumaric acid, citric acid, tartaric acid or acetic acid) or as salts with pharmaceutically usable bases such as alkali metal or alkaline earth metal hydroxides or carbonates, zinc or ammonium hydroxides or organic amines such as e.g. diethylamine, triethylamine, triethanolamine, inter alia.

As mentioned previously, the compounds of formulas 1 or 1′ may be converted into the salts thereof, particularly for pharmaceutical use into the physiologically and pharmacologically acceptable salts thereof. These salts may be present on the one hand as physiologically and pharmacologically acceptable acid addition salts of the compounds of formula 1 with inorganic or organic acids. On the other hand, the compound of formulas 1 or 1′ when R is hydrogen may be converted by reaction with inorganic bases into physiologically and pharmacologically acceptable salts with alkali or alkaline earth metal cations as counter-ion. The acid addition salts may be prepared for example using hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid or maleic acid. It is also possible to use mixtures of the above-mentioned acids. To prepare the alkali and alkaline earth metal salts of the compounds of formulas 1 or 1′ wherein R denotes hydrogen, it is preferable to use the alkali and alkaline earth metal hydroxides and hydrides, of which the hydroxides and hydrides of the alkali metals, particularly sodium and potassium, are preferred, while sodium and potassium hydroxide are particularly preferred.

The compounds of general formulas 1 or 1′ may optionally be converted into the salts thereof, particularly for pharmaceutical use into the pharmacologically acceptable acid addition salts with an inorganic or organic acid. Examples of suitable acids for this purpose include succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulphonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulphuric acid, tartaric acid or citric acid. It is also possible to use mixtures of the above-mentioned acids.

The invention relates to the compounds of formula 1 in question, optionally in the form of the individual optical isomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.

The compounds of formula 1, 1d, 1e or 1f according to the invention may optionally be present as racemates, but may also be obtained as pure enantiomers, i.e. in the (R) or (S) form. Preferred are the compounds with the specific stereochemistry of formula 1′, in particular the compounds with the specific stereochemistry of one of formulas 1d′, 1e′ or 1f′.

The invention relates to the compounds in question, optionally in the form of the individual optical isomers, diastereomers, mixtures of diastereomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.

The invention relates to the respective compounds of formulas 1 or 1′ in the form of the pharmacologically acceptable salts thereof. These pharmacologically acceptable salts of the compounds of formulas 1 or 1′ may also be present in the form of their respective hydrates (e.g. Monohydrates, dihydrates, etc.) as well as in the form of their respective solvates.

By a hydrate of the compound according to the formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, containing water of crystallisation.

By a solvate of the compound according to formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, which contains solvent molecules (e.g. Ethanol, methanol etc) in the crystal lattice.

The skilled man will be familiar with the standard methods of obtaining hydrates and solvates (e.g. recrystallisation from the corresponding solvent or from water).

4. METHODS OF PREPARATION

The Examples according to the invention were prepared as shown in Schemes 1, 2 or 3.

D is N, NH or CH,

G is C or N

T is C or N

E is C

Hal is Br or Cl

with X being —B(OH)2, -boronic acid pinacolester, -trifluoroborate or —SnBu3

PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)

and R1 is as herein before defined.

D is CH, N or NH,

G is C or N

T is C or N

E is C

A is CH or N

Hal is Br or Cl

with X being —B(OH)2, -boronic acid pinacolester, -trifluoroborate or —SnBu3

PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)

and R1 is as herein before defined.

4.1. Starting materials of formula 2, 3, 4, 5 and 10

4.1.1. Synthesis of Lactams 2 from Scheme 1, 2 and 3

Synthesis of Synthesis of (R)-4-[(R)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.1 for Example 84 and (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.2 for Examples 6 and 34.

Step 1: Synthesis of (1′R,3R/S)-1-(1′-(4-Methoxyphenylethyl)-5-oxo-3-pyrrolidine carboxylic acid (mixture of diastereoisomers)

A suspension of 100 g of (R)-1-(4-methoxy-phenyl)-ethylamine and 95 g itaconic acid in 0.5 L 1-methyl-2-pyrrolidinone was heated to 80° C. for 1 hour. The solution was stirred for additional 4 hours at 120° C. The reaction mixture was cooled to 25° C. and poured into 1.5 L of demineralized water. The precipitate was filtered, washed with demineralized water and dried at 50° C.

Yield: 195 g (quantitative yield) solid as a mixture of diastereoisomers

Analysis (method G): Rt: 2.6 min and 2.7 min, (M+H)+: 264

Step 2: Synthesis of (R/S)—N-Methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide as a mixture of diastereoisomers

260 g of 1,1′-carbonyldiimidazole (CD) were added to a solution of 285 g (1′R,3R/S)-1-(1′-(4-methoxyphenylethyl)-5-oxo-3-pyrrolidine carboxylic acid (mixture of diastereoisomers) in 1.4 L 2-methyltetrahydrofuran at 20° C. The suspension was stirred at 20° C. for 80 minutes. 235 mL ethyldiisopropylamine (DIPEA) and 130 g of N,O-dimethylhydroxylamine hydrochloride were added. The suspension was stirred for 3 hours at 20° C. Under cooling 850 mL 4M hydrochloric acid was added. The organic phase was separated and washed two times with 500 mL 1 N hydrochloric acid. The aqueous phase was reextracted two times with 500 mL ethyl acetate. The combined organic phases were dried over sodium sulfate. After filtration the solvent was evaporated under reduced pressure.

Yield: 271 g (82% of theory) of (R/S)—N-Methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide (mixture of diastereoisomers) as an oil.

Analysis (method H): Rt: 11.1 min (41 area %) and 13.8 min (59 area %), (M+H)+: 307

Step 3: Synthesis of (R/S)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one as a Mixture of Diastereoisomers

530 mL of a 3M solution of methylmagnesium bromide in diethylether were added slowly to a cooled solution of 271 g of (R/S)—N-methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide (mixture of diastereoisomers) in 1.4 L of 2-methyltetrahydrofuran so that the temperature remained under 0° C. After complete addition the temperature was kept for 75 minutes at 0° C. and then warmed up to 20° C. The suspension was stirred 16 hours at 20° C. Under cooling 650 mL of a 4M hydrochloric acid were added. The organic phase was separated and washed with 500 mL saturated sodium carbonate solution and with 500 mL saturated brine. The organic phase was dried over sodium sulfate. After filtration the solvent was evaporated under reduced pressure.

Yield: 188 g (81% of theory) of (R/S)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one (mixture of diastereoisomers) as an oil.

Analysis (method H): Rt: 7.4 min and 9.6 min, (M+H)+: 262

Step 4: Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one Under Base Induced Epimerization Conditions

103 g of a mixture of diastereoisomers (R/S)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one were dissolved in 155 mL 1-butanol at 25° C. 18 mL benzyltrimethylammonium hydroxide (40% solution in methanol) was added. The solution was stirred for 30 minutes at 25° C. The solution was cooled to 0° C. Precipitation started. The suspension was stirred for 15 minutes at 0° C. 100 mL n-heptane was added slowly and the suspension was stirred for 30 minutes at 0° C. The addition of 100 mL portions of n-heptane was repeated 4 times with subsequent stirring of the suspension at 0° C. for 30 minutes. The precipitate was isolated, washed with n-heptane and dried at 50° C.

Yield: 77.1 g of a beige solid (75% of theory) with a diastereoisomeric purity of ˜95: 5 (method H).

For further purification the crude product was dissolved in 310 mL 2-methyl-2-butanol at 40° C. (temperature <50° C.). The solution was slowly cooled to 0° C. Precipitation started. At 0° C. 385 mL of n-heptane were added and the suspension was stirred for 1 hour. The precipitate was filtrated, washed with n-heptane and dried at 50° C.

Yield: 68.7 g (67% of theory) of a colorless solid with a diastereoisomeric purity of >99:1.

Analysis (method H): Rt: 6.8 min, (M+H)+: 262

Step 4: Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one Under Base Induced Epimerization Conditions

13.2 g of a mixture of diastereoisomers (R/S)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one were dissolved in 18 mL of 1-butanol at 25° C. The solution was cooled to 3° C. and treated with 100mg of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one. The resulting mixture was agitated for 15 min at 3° C.; at which point, 2.3 mL benzyltrimethylammonium hydroxide (40% solution in methanol) were added. The solution was stirred for 30 minutes at 3° C. 64 mL n-heptane was added slowly over 1 h at 0 to 3° C. and the suspension was stirred for 60 minutes at 0° C. The precipitate was isolated, washed with n-heptane and dried at 30° C.

Yield: 10.6 g of a beige solid (80% of theory) with a diastereoisomeric purity of ˜98: 2 (method H).

Analysis (method H): Rt: 6.8 min, (M+H)+: 262

Step 5: Synthesis of (R)-4-[(R)-1-Hydroxyethyl]-1-[(S)-11-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.1

94.6 mg of dichloro (pentamethylcyclopentadienyl)-iridium(III) dimer and 105 mg of (S,S)—N-(p-toluenesulfonyl)-1,2-diphenylethylendiamine [(R,R)-TsDPEN] were dissolved in 20 mL of acetonitrile and subsequently charged to a slurry of 50 g of (R)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one and 65 g of sodium formate in 500 mL of water at 25° C. The slurry was heated to 60° C. and agitated at this temperature while sparging with nitrogen for 3 h. The reaction was diluted at 60° C. with 500 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed twice with 300 mL of water. The organic portion was concentrated to an oily solid. The residual material was crystallized three times from ethyl acetate and hexanes followed by drying in a vacuum oven with a nitrogen stream at 30° C.

25.4 g of a beige solid with a diastereomeric purity of >99:1

Step 5: Synthesis of (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.2

9.46 mg of dichloro (pentamethylcyclopentadienyl)-iridium(III) dimer and 10.52 mg of (R,R)—N-(p-toluenesulfonyl)-1,2-diphenylethylendiamine [(R,R)-TsDPEN]were dissolved in 1 mL of acetonitrile and subsequently charged to a slurry of 5 g of (R)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one and 6.5 g of sodium formate in 50 mL of water at 25° C. The slurry was heated to 60° C. and agitated at this temperature while sparging with nitrogen for 3 h. The reaction was diluted at 60° C. with 50 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed with 20 mL of water. The organic portion was concentrated to an oil. The oil was dissolved in 8 mL of isopropyl acetate at reflux. The solution was cooled to ambient temperature wherein crystallization occurred. The mixture was diluted dropwise with 10 mL of heptane at ambient temperature. The mixture was agitated for 30 minutes. The solids were collected by filtration, washed with a solution of 20 vol % isopropyl acetate in heptane and dried in a vacuum oven with a nitrogen stream at 55° C. 3.82 g of a beige solid with a diastereomeric purity of 99:1

Analysis (method I): Rt: 12.9 min, (M+H)+: 264

Synthesis of [(1S)-1-[(3R)-1-[(1S)-1-(4-Methoxyphenyl)ethyl]-5-oxo-pyrrolidin-3-yl]ethyl]4-methylbenzenesulfonate 2.3 for Example 6

To a mixture of 20.0 g of (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.2, 21.67 g p-toluenesulfonyl chloride and 0.92 g N,N-dimethylpyridin-4-amine was added 42 mL pyridine and 42 mL dichloromethane (DCM). The resulting mixture was stirred at 34° C. for 18 h under argon atmosphere. The reaction mixture was diluted with isopropyl acetate and washed with water and 2M hydrochloric acid. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was taken up in isopropyl acetate and n-heptane. The precipitate was filtered off, washed with n-heptane/isopropyl acetate and dried to yield 19.83 g of [(1S)-1-[(3R)-1-[(1S)-1-(4-methoxyphenyl)ethyl]-5-oxo-pyrrolidin-3-yl]ethyl] 4-methylbenzenesulfonate 2.3 as solid.

Analysis: HPLC-MS: Rt=0.680 min (method J), M+H=418

4.1.2. Synthesis of Boronic Acids, Boronic Esters, BF3 Borates and Stannanes with Formula 4

X.1.2.1. Synthesis of R1-Hal 3

Synthesis of 4-Bromo-1-tert-butyl-pyrazole 3.1 for Examples 6, 34 and 84 Step 1: Synthesis of 1-tert-Butyl-pyrazole

To a stirred mixture of 34.48 g of 1,1,3,3-tetramethoxy-propane and 26.20 g tert.-butylhydrazine hydrochloride in 230 mL ethanol was added 40.0 mL conc. hydrochloric acid dropwise below 50° C., then the mixture was stirred under reflux for 2 h. The reaction mixture was diluted with water. The solvent was almost removed by destillation and the aqueous residue extracted with diethylether. The combined aqueous phases were basified with 10N sodium hydroxide solution and extracted with diethylether. The combined organic phases were washed with saturated brine, dried over sodium sulfate, filtered and concentrated in vacuo to yield 21.90 g of 1-tert-butyl-pyrazole as oil.

Analysis: HPLC-MS: Rt=0.412 min (method A), M+H=125

Step 2: Synthesis of 4-Bromo-1-tert-butyl-pyrazole

To a mixture of 21.9 g of 1-tert-butyl-pyrazole in 150 mL DCM was added 31.5 g N-bromosuccinimide in portions between 0 and 10° C. The resulting mixture was stirred for 30 min. The reaction mixture was allowed to reach ambient temperature. The precipitate was filtered off and washed with DCM. The combined organic extracts were washed with water and saturated brine, dried over magnesium sulfate, filtered and concentrated in vacuo to yield 34.0 g of 4-bromo-1-tert-butyl-pyrazole as oil.

Analysis: HPLC-MS: Rt=1.35 min (method B), M+H=203/205

4.1.3. Synthesis of Compounds of Formula 4 (R1—X) (Scheme 1 and 2)

Synthesis of 1-tert-Butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.19 for Examples, 6, 34 and 84

To a stirred mixture of 50 g of 4-bromo-1-tert-butyl-pyrazole 3.1 in 230 mL THF was added dropwise 100 mL 2.5M N-butyllithium solution in hexane under argon atmosphere below −60° C., then the mixture was stirred at this temperature for 5 min, before 52 mL 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane were added dropwise below −60° C. The reaction mixture was allowed to reach ambient temperature. The mixture was cooled with an ice bath and diluted with aqueous phosphate buffer solution and water and neutralized with 2M aqueous hydrochloric acid. The organic solvent was removed by destillation and the residue was extracted with DCM. The combined organic extracts were washed with saturated brine, dried over sodium sulfate, filtered and concentrated in vacuo to yield 44.26 g of 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole as solid.

Analysis: HPLC-MS: Rt=0.904 min (method F), M+H=251

4.1.4. Synthesis of Heterocyclic 5 and 10 from Scheme 1 and 2

Synthesis of 5,7-Dichloro-2-methyl[1,2,4]triazolo[1,5-c]pyrimidine (5.2) for Example 84

5.2 was synthesized according to WO 2008113469.

Step 1: Synthesis of (6-Chloro-2-methylsulfanyl-pyrimidin-4-yl)-hydrazine

A solution of 10.0 g 4,6-dichloro-2-methylsulfanyl-pyrimidine and 10.1 mL hydrazine monohydrate in 40 mL DCM was stirred at 0° C. overnight and at room temperature for 2 days. The precipitate was filtered off, washed with DCM and dried to afford 10.0 g of (6-chloro-2-methylsulfanyl-pyrimidin-4-yl)-hydrazine as solid.

Analysis: HPLC-MS: Rt=0.30 min (method J), M+H=191

Step 2: Synthesis of 7-Chloro-2-methyl-5-methylsulfanyl41,2,4]triazolo[1,5-c]pyrimidine

A solution of 2.35 g (6-chloro-2-methylsulfanyl-pyrimidin-4-yl)-hydrazine in 12 mL triethyl orthoacetate and 10.7 mL glacial acid was refluxed for 1 h. The solvent was removed by destillation and the residue was purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 870 mg 7-chloro-2-methyl-5-methylsulfanyl-[1,2,4]triazolo[1,5-c]pyrimidine as solid.

Analysis: HPLC-MS: Rt=0.40 min (method C), M+H=215

Step 3: Synthesis of 7-Chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-ol

To a solution of 1.67 g of 7-chloro-2-methyl-5-methylsulfanyl-[1,2,4]triazolo[1,5-c]pyrimidine in 35 mL methanol was added 3.13 mL aqueous potassium hydroxide solution (40%), then the mixture was stirred at room temperature for 10 min. The solvent was removed by destillation. The residue was taken up in water and a neutral pH was adjusted with 4M aqueous hydrochloric acid. The precipitate was filtered off, washed with water and dried to yield 1.25 g of 7-chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-ol as solid.

Analysis: HPLC-MS: Rt=0.23 min (method J), M+H=185

Step 4: Synthesis of 5,7-Dichloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidine (8.2)

A mixture of 600 mg 7-chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-ol and 103.5 μL N,N-diethylaniline in 19.15 mL phosphorus oxychloride was stirred at 100° C. for 2 h. The solvent was removed by destillation and the residue taken up in water. A neutral pH was adjusted with saturated aqueous sodium bicarbonate solution and aqueous phosphate buffer solution. The precipitate was filtered off, washed with water and triturated with diethyl ether and petroleum to yield 504 mg of 5,7-dichloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidine 5.2 as solid.

Analysis: HPLC-MS: Rt=0.403 min (method J), M+H=203

Synthesis of 6-Bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-ol 10.3 for Example 34 Step 1: Synthesis of 2,4-dinitro-phenolatel-amino-3-bromo-5-methoxy-pyridinium

A mixture of 3.0 g of 3-bromo-5-methoxy-pyridine and 3.34 g of O-(2,4-dinitro-phenyl)-hydroxylamine in 40 mL acetonitrile was stirred at 50° C. for 2 days, then at 80° C. for 4 h. The reaction mixture was allowed to reach ambient temperature and diluted with TBME. The precipitate was filtered off, washed with TBME and dried to yield 4.24 g of 2,4-dinitro-phenolatel-amino-3-bromo-5-methoxy-pyridinium as solid.

Analysis: 1H NMR (DMSO-d6, 500 MHz): δ=8.7 (t, J=1.3 Hz, 1H), 8.63-8.55 (m, 2H), 8.44-7.99 (m, 3H), 7.76 (dd, J=9.8, 3.2 Hz, 1H), 6.30 (d, J=9.8 Hz, 1H), 3.97 (s, 3H).

Step 2: Synthesis of 6-Bromo-4-methoxy-2-methyl-pyrazolo[1,5-a]pyridine-3-carboxylic acid methyl ester

To a mixture of 4.24 g of 2,4-dinitro-phenolatel-amino-3-bromo-5-methoxy-pyridinium and 3.02 g potassium carbonate in 42.4 mL DMF was added 1.49 mL but-2-ynoic acid methyl ester. The reaction mixture was stirred at 45° C. for 18 h. The reaction mixture was allowed to reach ambient temperature, then diluted with water and extracted with ethyl acetate/heptane (1:1). The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/TBME/ethyl acetate) to give 1.95 g of 6-bromo-4-methoxy-2-methyl-pyrazolo[1,5-a]pyridine-3-carboxylic acid methyl ester as solid.

Analysis: HPLC-MS: Rt=2.94 min (method R), M+H=299/301

Step 3: Synthesis of 6-Bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-ol 10.3

A mixture of 2.34 g of 6-bromo-4-methoxy-2-methyl-pyrazolo[1,5-a]pyridine-3-carboxylic acid methyl ester in 3.51 mL hydrogen bromide (48% aqueous) was stirred in a sealed tube at 150° C. for 8 h. The reaction mixture was allowed to reach ambient temperature then diluted with water. The mixture was worked up by adding saturated aqueous sodium bicarbonate solution, followed by extraction with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/ethyl acetate/methanol) to give 1.54 g (73% per HPLC) of 6-bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-ol 10.3 as solid.

Analysis: HPLC-MS: Rt=1.63 min (method K), M+H=227/229

4.2. Synthesis of Intermediates 6, 7, 8 and 11 from Scheme 1 and 2

Synthesis of (4R)-4-[(1R)-1-[(7-Chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl)oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 6.2 for Example 84

To a mixture of 537 mg (R)-4-[(R)-1-hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.1 in 7.5 mL DMA was added 49 mg NaH at room temperature. The resulting mixture was stirred at room temperature for 10 min, before 414 mg 5,7-dichloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidine 5.2 were added. The resulting mixtures was stirred at room temperature for 2 h and at 40° C. for 2 h. Additional 49 mg NaH were added and the reaction mixture was stirred at 40° C. for 2 h. The reaction mixture was quenched with saturated ammonium chloride solution and extracted with DCM. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo. The crude material was purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 251 mg of (4R)-4-[(1R)-1-[(7-chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl)oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 6.2 as solid.

Analysis: HPLC-MS: Rt=0.55 min (method C), M+H=430

Synthesis of (4R)-4-[(1R)-1-(6-bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-yl)oxyethyl] pyrrolidin-2-one (7.6) for Example 34

This intermediate was prepared from 6-bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-ol 10.3 in two steps according to the preparation of (4R)-4-[(1 R)-1-(6-bromo-2-methyl-indazol-4-yl)poxyethyl]pyrrolidin-2-one 7.5.

Analysis: HPLC-MS: Rt=1.67 min (method K), M+H=338/340

4.3 Synthesis of the patent Examples of Formula 1

Synthesis of (4R)-4-[(1R)-1-[[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-1H-benzimidazol-4-yl]oxy]ethyl]pyrrolidin-2-one (Example 6) Step 1: Synthesis of 1-Benzyl-4-benzyloxy-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazole

To a mixture of 730 mg 1-benzyl-4-benzyloxy-6-bromo-2-methyl-benzimidazole, 538 mg 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) and 146 mg 1,1′-bis(diphenylphospino)ferrocenedichloropalladium(II) (complex with DCM (1:1)) in dioxane was added 2.69 mL 2M aqueous sodium carbonat solution. The reaction mixture was stirred at 120° C. for 30 min under microwave irradiation, then filtered through Agilent PL-Thiol MP-SPE resin and washed with methanol. The solvent was removed by destillation and the residue was taken up in DCM and washed with water. The combined organic phases were concentrated in vacuo. The resulting residue was dissolved in acetonitrile/methanol, filtered through rp-SiO2, washed with MeOH. The solvent was removed by destillation and the residue was purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 217 mg of 1-benzyl-4-benzyloxy-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazole as solid.

Analysis: HPLC-MS: Rt=0.76 min (method A), M+H=451

Step 2: Synthesis of 1-Benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-ol

A mixture of 200 mg 1-benzyl-4-benzyloxy-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazole and 200 mg palladium on carbon in 5 mL ethyl acetate was hydrogenated at room temperature for 7 h at 1.01 bar. The catalyst was removed by filtration and the solvent was evaporated in vacuo to yield 160 mg (90% per HPLC) 1-benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-ol as solid.

Analysis: HPLC-MS: Rt=0.99 min (method L), M+H=361

Step 3: Synthesis of (4R)-4-[(1R)-141-Benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-yl] oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one

A mixture of 130 mg 1-benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-ol, 166 mg [(1S)-1-[(3R)-1-[(1S)-1-(4-methoxyphenyl)ethyl]-5-oxo-pyrrolidin-3-yl]ethyl] 4-methyl-benzenesulfonate 2.3 and 150 mg potassium carbonate in 2 mL DMF was stirred at 60° C. for 16 h. The reaction mixture was allowed to reach ambient temperature, diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with saturated brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 42 mg of (4R)-4-[(1R)-1-[1-benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one as solid.

Analysis: HPLC-MS: Rt=1.30 min (method L), M+H=606

Step 4: Synthesis of (4R)-4-[(1R)-1-[[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-1H-benzimidazol-4-yl] oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one

A mixture of 42 mg (4R)-4-[(1R)-1-[1-benzyl-6-(1-tert-butylpyrazol-4-yl)-2-methyl-benzimidazol-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one, 42 mg palladium on carbon and 140 μL glacial acetic acid in 3 mL ethanol was hydrogenated at room temperature for 5.5 h at 1 bar. The catalyst was removed by filtration and the solvent was evaporated in vacuo to yield 14 mg (4R)-4-[(1R)-1-[[6-(1-tert-butylpyrazol-4-yl)-2-methyl-1H-benzimidazol-4-yl]oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one.

Analysis: HPLC-MS: Rt=0.86 min (method F), M+H=516

Step 5: Synthesis of (4R)-4-[(1R)-1-[[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-1H-benzimidazol-4-yl]oxy]ethyl]pyrrolidin-2-one (Example 6)

A mixture of 14 mg (4R)-4-[(1R)-1-[[6-(1-tert-butylpyrazol-4-yl)-2-methyl-1H-benzimidazol-4-yl]oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one in 200 μL TFA was stirred at 70° C. for 5 h. The reaction mixture was diluted with methanol, filtered and purified by rpHPLC (SunfireC18, acetonitrile/water, TFA) to yield after lyophilisation 7 mg of Example 6 as solid.

Analysis: HPLC-MS: Rt=0.425 min (method S), M+H=382

Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[1,5-a]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 34)

To a mixture of 100 mg (4R)-4-[(1R)-1-(6-bromo-2-methyl-pyrazolo[1,5-a]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.6, 111 mg 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.19 and 10.4 mg 1,1′-bis(triphenylphosphine)palladium(II) chloride in 1.50 mL ethanol (80% with toluene) was added 443 μL 2M aqueous sodium carbonat solution. The resulting mixture was stirred at 95° C. for 1 h. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol) and by rpHPLC to yield 73 mg (yield: 65%) of Example 34.

Analysis: HPLC-MS: Rt=2.63 min (method R), M+H=382

1H NMR (DMSO, 500 MHz) □=1.29 (3H, d, J=6.1 Hz), 1.55 (9H, s), 2.16-2.33 (2H, m), 2.34 (3H, s), 2.75 (1H, h, J=8.0 Hz), 3.10 (1H, dd, J=9.7, 6.5 Hz), 3.38 (1H, t, J=9.1 Hz), 4.77 (1H, p, J=5.9 Hz), 6.31 (1H, s), 6.91 (1H, s), 7.58 (1H, s), 7.96 (1H, s), 8.35 (1H, s), 8.50 (1H, s)

Synthesis of (4R)-4-[(1R)-1-[[7-(1-tert-Butylpyrazol-4-yl)-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl]oxy]ethyl]pyrrolidin-2-one (Example 84) Step 1: Synthesis of (4R)-4-[(1R)-1-[[7-(1-tert-butylpyrazol-4-yl)-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl]oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one

A mixture of 80 mg (4R)-4-[(1R)-1-[(7-chloro-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl)oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 7.2, 56 mg 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) 4.19, 15.2 mg 1,1′-bis(diphenyl-phospino) ferrocenedichloropalladium(II) (complex with DCM (1:1)) and 280 μL 2M aqueous sodium carbonat solution in 1 mL dioxane was stirred at 120° C. for 25 min under microwave irradiation. Additional 14 mg 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) were added and the reaction mixture was stirred at 120° C. for 10 min under microwave irradiation. The reaction mixture was filtered through Agilent PL-Thiol MP-SPE, washed with methanol and purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 82 mg of (4R)-4-[(1R)-1-[[7-(1-tert-butylpyrazol-4-yl)-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl]oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 8.2 as oil.

Analysis: HPLC-MS: Rt=0.60 min (method C), M+H=518

Step 2: Synthesis of (4R)-4-[(1R)-1-[[7-(1-tert-Butylpyrazol-4-yl)-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl]oxy]ethyl]pyrrolidin-2-one (Example 84)

A mixture of 89 mg of (4R)-4-[(1R)-1-[[7-(1-tert-butylpyrazol-4-yl)-2-methyl-[1,2,4]triazolo[1,5-c]pyrimidin-5-yl]oxy]ethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 8.2 and 319 μL anisole in 1.13 mL TFA was stirred at 80° C. for 2.25 h. The reaction mixture was filtered and purified by rpHPLC to yield after lyophilisation 31 mg of Example 84 as solid.

Analysis: HPLC-MS: Rt=0.42 min (method X), M+H=384

4.5 Analytical Methods

The Example compounds prepared according to the foregoing synthesis schemes were characterised by the following chromatographic methods and/or NMR spectroscopy.

4.5.1 Chromatographic Methods (HPLC-MS Methods)

Method A

Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm Column supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% NH ] [Acetonitril] [ml/min] [° C.] 0.00 99 1 1.3 60 0.02 99 1 1.3 60 1.00 0 100 1.3 60 1.10 0 100 1.3 60

Method B:

Column: Sunfire C18, 3 × 30 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% TFA] [Methanol] [ml/min] [° C.] 0.0 95 5 1.8 60 0.25 95 5 1.8 60 1.70 0 100 1.8 60 1.75 0 100 2.5 60 1.90 0 100 2.5 60

Method C:

Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm Column supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.] 0.00 95 5 1.3 60 0.02 95 5 1.3 60 1.00 0 100 1.3 60 1.10 0 100 1.3 60

Method D:

Column: XBridge C18, 2.1 × 20 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.10% TFA] [Methanol] [ml/min] [° C.] 0.0 95 5 1.4 60 0.05 95 5 1.4 60 1.00 0 100 1.4 60 1.1 0 100 1.4 60

Method E:

Column: Sunfire C18, 2.1 × 20 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.10% TFA] [Methanol] [ml/min] [° C.] 0.00 99 1 1.3 60 0.15 99 1 1.3 60 1.10 0 100 1.3 60 1.25 0 100 1.3 60

Method F:

Column: XBridge C18, 3 × 30 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.] 0.00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60

Method G:

Eluent A: Water/0.2% KH2PO4 pH = 3 Eluent B: Acetonitrile Time [min] % A % B Flow rate [mL/min] 0.00 80 20 1.50 5.00 20 80 1.50 8.00 20 80 1.50

The stationary phase used was a Inertsil C8-3 (GL Sciences), 5 μm; dimension: 100×4.0 mm,

(column temperature: constant at 30° C.). Detection UV 220 nm.

Method H:

Eluent A: Hexane Eluent B: 2-Propanol Time [min] % A % B Flow rate [mL/min] 00.00 90 10 1.0 20.00 90 10 1.0

The stationary phase used was a Chiralpak AD-H (Daicel), 5 μm; dimension: 150×4.6 mm,

(column temperature: constant at 10° C.). Detection DAD 225 nm.

Method I:

Eluent A: Hexane Eluent B: 2-Propanol Time [min] % A % B Flow rate [mL/min] 00.00 90 10 1.0 25.00 90 10 1.0

The stationary phase used was a Chiralpak AD-H (Daicel), 5 μm; dimension: 150×4.6 mm,

(column temperature: constant at 10° C.).

Detection DAD 225 nm.

Method J:

Column: Sunfire C18, 2.1 × 30 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.] 0.0 99 1 1.5 60 0.02 99 1 1.5 60 1.00 0 100 1.5 60 1.10 0 100 1.5 60

Method K:

Column: Waters Atlantis dC18 (2.1×50 mm, 3 μm column)

Flow rate: 1 mL/min

Solvent A: 0.1% Formic acid/water

Solvent B: 0.1% Formic acid/acetonitrile

Injection volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 2.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 2.5 to 2.7 minutes, 100% solvent B; 2.71 to 3.0 minutes, 95% solvent A+5% solvent B.

MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV

UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array, Waters 2487 UV or Shimadzu SPD-M20A PDA

Method L:

Column: XBridge C18, 4.6 × 30 mm, 3.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% NH3] [ACN] [ml/min] [° C.] 0.0 97 3 5 60 0.2 97 3 5 60 1.6 0 100 5 60 1.7 0 100 5 60

Method M:

Column: Waters SymmetryShield RP8 (2.1×50 mm, 3.5 μm column)

Flow rate: 1 mL/min

Solvent A: 0.1% Formic acid/water

Solvent B: 0.1% Formic acid/acetonitrile

Injection volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 2.2 minutes, constant gradient from 95% solvent A +5% solvent B to 100% solvent B; 2.2 to 2.7 minutes, 100% solvent B; 2.71 to 3.0 minutes, 95% solvent A+5% solvent B.

MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV

UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array, Waters 2487 UV or Shimadzu SPD-M20A PDA

Method N:

Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.] 0.0 99 1 1.6 60 0.02 99 1 1.6 60 1.00 0 100 1.6 60 1.10 0 100 1.6 60

Method O:

Column: Xbridge BEH Phenyl, 2.1 × 30 mm, 1.7 μm Column supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.] 0.00 95 5 1.3 60 0.02 95 5 1.3 60 1.00 0 100 1.3 60 1.10 0 100 1.3 60

Method P:

Column: Supelco Ascentis Express (2.1×30 mm, 2.7 μm column)

Flow rate: 1 ml/min

Solvent A: 0.1% Formic acid/water

Solvent B: 0.1% Formic acid/acetonitrile

Injection volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 1.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 1.5 to 1.6 minutes, 100% solvent B; 1.60 to 1.61 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 1.61 to 2.00 minutes, 95% solvent A+5% solvent B.

MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV

UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array, Waters 2487 UV or Shimadzu SPD-M20A PDA

Method Q

Column: Atlantis d C18; 50×3 mm; 3 μ

Flow rate: 0.6 ml/min

Solvent A: 0.1% Formic acid in water

Solvent B: 0.1% Formic acid in acetonitrile

Injection Volume: 5 μL

Column temperature: 35° C.

UV Detection wavelength: Spectra A max (with scan in the region of 200-400nm) Eluent: 0 to 3.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 3.5 to 3.8 minutes, 100% solvent B; 3.8 to 3.9 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 3.9 to 4.5 minutes, 95% solvent A+5% solvent B.

MS detection using Waters 3100, SQ detector, ES +ve and −ve modes (Cone voltage: 30V, Capillary voltage 3.0 KV)

UV detection using Waters 2996 photodiode array

Method R:

Column: Phenomenex Kinetex-XB C18 (2.1×100 mm, 1.7 μm column)

Flow rate: 0.6 mL/min

Solvent A: 0.1% Formic acid/water

Solvent B: 0.1% Formic acid/acetonitrile

Injection volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 5.3 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 5.3 to 5.8 minutes, 100% solvent B; 5.80 to 5.82 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.82 to 7mins, 95% solvent A+5% solvent B

MS detection using Waters SQD

UV detection using Waters Acquity photodiode array

Method S:

Column: Sunfire C18, 2.1 × 30 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.] 0.0 99 1 1.3 60 0.02 99 1 1.3 60 1.00 0 100 1.3 60 1.10 0 100 1.3 60

Method T:

Column: Phenomenex Gemini C18 (2.0 mm×100 mm, 3 μm column)

Flow rate: 0.5 mL/min

Solvent A: 2 mM Ammonium bicarbonate modified to pH 10 with Ammonium Hydroxide/water

Solvent B: Acetonitrile

Injection volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 5.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 5.5 to 5.9 minutes, 100% solvent B; 5.90 to 5.92 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.92 to 9.00 minutes, 95% solvent A+5% solvent B.

Method U:

Column: XBridge C18_3.0 × 30 mm, 2.5 μm Column producer: Waters Description: Gradient/Solvent % Sol [H2O % Sol Flow Temp Time [min] 0.1% NH4OH] [Acetonitrile] [ml/min] [° C.] 0.0 98.0 2.0 2.0 60.0 1.2 0.0 100.0 2.0 60.0 1.4 0.0 100.0 2.0 60.0

Method V:

Column: XBridge C18_3.0 × 30 mm, 2.5 μm Column producer: Waters Gradient/Solvent % Sol [H2O % Sol Flow Temp Time [min] 0.1% NH4OH] [Acetonitrile] [ml/min] [° C.] 0.0 98.0 2.0 2.0 60.0 1.2 0.0 100.0 2.0 60.0 1.4 0.0 100.0 2.0 60.0

Method W:

Column: Sunfire C18_3.0 × 30 mm, 2.5 μm Column producer: Waters Description: Gradient/Solvent % Sol [H2O % Sol Flow Temp Time [min] 0.1% TFA] [Acetonitrile] [ml/min] [° C.] 0.0 98.0 2.0 2.0 60.0 1.2 0.0 100.0 2.0 60.0 1.4 0.0 100.0 2.0 60.0

Method X:

Column: Sunfire C18_2.1 × 50 mm, 2.5 μm Column producer: Waters Description: Gradient/Solvent % Sol [H2O % Sol [Acetonitrile Flow Temp Time [min] 0.1% TFA] 0.08% TFA] [ml/min] [° C.] 0.0 95.0 5.0 1.5 60.0 0.75 0.0 100.0 1.5 60.0 0.85 0.0 100.0 1.5 60.0

Method Y:

Device description: Waters Acquity with 3100 MS Column: XBridge BEH C18_3.0 × 30 mm, 1.7 μm Column producer: Waters Description: Gradient/Solvent % Sol [H2O % Sol Flow Temp Time [min] 0.1% NH4OH] [Acetonitrile] [ml/min] [° C.] 0.0 95.0 5.0 1.5 60.0 0.7 0.1 99.9 1.5 60.0 0.8 0.1 99.9 1.5 60.0 0.81 95.0 5.0 1.5 60.0 1.1 95.0 5.0 1.5 60.0

Method Z:

Column: Waters Atlantis dC18 (2.1×100 mm, 3 μm column)

Flow rate: 0.6 mL/min

Solvent A: 0.1% Formic acid/water

Solvent B: 0.1% Formic acid/acetonitrile

Injection Volume: 3 μL

Column temperature: 40° C.

UV Detection wavelength: 215 nm

Eluent: 0 to 5 minutes, constant gradient from 95% solvent A +5% solvent B to 100% solvent B; 5 to 5.4 minutes, 100% solvent B; 5.4 to 5.42 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.42 to 7.00 minutes, 95% solvent A+5% solvent B.

MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV

UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array, Waters 2487 UV or Shimadzu SPD-M20A PDA

Method Z1:

Method Name: Method Name: Column: Sunfire, 3 × 30 mm, 2.5 μm Column Supplier: Waters Gradient/Solvent % Sol % Sol Flow Temp Time [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.] 0.00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60

4.5.2 NMR Spectroscopy

Configuration of the Bruker DRX 500 MHz NMR

High performance digital NMR spectrometer, 2-channel microbay console and Windows XP host workstation running Topspin version 1.3.

Equipped with:

    • Oxford instruments magnet 11.74 Tesla (500 MHz proton resonance frequency)
    • B-VT 3000 temperature controller
    • GRASP II gradient spectroscopy accessory for fast acquisition of 2D pulse sequences
    • Deuterium lock switch for gradient shimming
    • 5 mm Broad Band Inverse geometry double resonance probe with automated tuning and matching (BBI ATMA). Allows 1H observation with pulsing/decoupling of nuclei in the frequency range 15N and 31P with 2H lock and shielded z-gradient coils.

Configuration of the Bruker DPX 400 MHz NMR

High performance one bay Bruker 400 MHz digital two channel NMR spectrometer console and Windows XP host workstation running XwinNMR version 3.5.

Equipped with:

    • Oxford instruments magnet 9.39 Tesla (400 MHz proton resonance frequency)
    • B-VT 3300 variable temperature controller unit
    • Four nucleus (QNP) switchable probe for observation of 1H, 13C, 19F and 31P with 2H lock

Configuration of the Bruker 500 MHz NMR

High performance digital NMR spectrometer, 2-channel one bay console and Linux host workstation running Topspin version 2.1 PL6.

Equipped with:

    • Bruker-Biospin AVANCE III 500A magnet 11.75 Tesla (500 MHz proton resonance frequency)
    • B-VT 3000 temperature controller
    • 5 mm Multinuclear Broad Band fluorine observe (BBFO) probe with digital tuning covering the range from 15N and 31P as well as 19F with 1H decoupling.

Configuration of the Bruker DPX 400 MHz NMR

High performance digital NMR spectrometer, 2-channel microbay console and Linux host workstation running Topspin version 2.1 PL6

Equipped with:

    • Bruker-Biospin AVANCE Ill DPX400C magnet 9.40 Tesla (400 MHz proton resonance frequency)
    • B-VT 3200 variable temperature controller unit

5 mm Multinuclear Broad Band fluorine observe (BBFO) probe with digital tuning covering the range from 15N and 31P as well as 19F with 1H decoupling.

5. EXAMPLES

The following Examples were prepared analogously to the methods of synthesis described above. These compounds are suitable as SYK inhibitors and have IC50-values with regard to SYK-inhibition of less than or equal to 1 pmol. Additionally these compounds exhibit a very good SYK-selectivity which means that—whereas SYK is inhibited effectively—other kinases such as Aurora B (AURB), FLT-3 and GSK 3β are not or almost not inhibited at all. Consequently undesired side effects of these effective SYK-inhibitors of the invention are minimized.

AURB phosphorylates Ser10 and Ser28 on histone H3, a key event in mitosis and cellular proliferation. Inhibition of AURB therefore has the potential to block cellular proliferation, and could compromise tissues that exhibit a high cellular turnover, such as the intestine or the bone marrow. It is therefore desired to avoid parallel AURB inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds show IC50-values with regard to Aurora B inhibition of more than 1 μM, preferably more than 6 μM, more preferably more than 10 μM, more preferably more than 30 μM, particularly preferably more than 50 μM. The AURB-IC50/SYK-IC50-ratios of all example compounds are preferably more than 30, more preferably more than 100.

FLT-3 is a tyrosine kinase receptor. When an FLT-3 ligand binds to the receptor, the intrinsic tyrosine kinase activity of the receptor is activated, which in turn phosphorylates and activates signal transduction molecules (such as SHC) which in turn propagates the signal in the cell. Signaling through FLT-3 plays a role in cell survival, proliferation, and differentiation and is important for lymphocyte (B cell and T cell) development. It is therefore desired to avoid parallel FLT-3 inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds of the instant invention show IC50-values with regard to FLT-3 inhibition of more than 0.30 μM, preferably more than 1 μM, more preferably more than 10 μM, particularly preferably more than 30 μM. The FLT-3-IC50/SYK-IC50-ratios of all example compounds are preferably more than 10, more preferably more than 30.

Glycogen synthase kinase 3 beta (GSK 3β) is a proline-directed serine-threonine kinase that is prominent in the TGF-β and Wnt intracellular signalling pathways. GSK 3β facilitates a number of intracellular signalling pathways including the activation of β-catenin complex. In adults, GSK 3β is involved in cellular proliferation and energy metabolism, whilst in neonates is involved in neuronal cell development and body pattern formation. It is therefore desired to avoid parallel GSK3β inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds of the invention show IC50-values with regard to GSK 3β inhibition of more than 1 μM, preferably of more than 10 μM.

Further it is desirable for an SYK-inhibitor to have certain human liver microsomal stability (corresponding to CI<60% Qh; % Qh=percentage of liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated.

The IC50-values with respect to SYK-inhibition,with respect to Aurora B inhibition,with respect to FLT3-inhibition and with respect to GSKbeta-inhibition as well as the human liver microsomal stablities (CI [% Qh]) for each of the individual example substances are shown in the following Table 1 and were experimentally determined as follows:

5.1 Syk Kinase Test

Recombinant human Syk (amino acids 342-635) was expressed as a fusion protein with an N-terminal GST tag, affinity-purified and deep-frozen at a concentration of approx. 50-100 μM in storage buffer (25 mM HEPES pH7.5; 25 mM MgCl2; 5 mM MnCl2; 50 mM KCl; 0.2% BSA; 0.01% CHAPS; 100 μM Na3VO4; 0.5 mM DTT, 10% glycerol) at −80° C. until use.

The catalytic activity of the GST-Syk kinase fusion protein was determined using the Kinase Glo® Luminescence Kinase test (Promega; V6712). In this homogeneous test the amount of ATP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ATP still present and thus correlates inversely with the activity of the kinase.

Method

The test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 1 mM. Serial Dilution is done in 100% DMSO. All further dilutions of the substances were carried out with test buffer (25 mM HEPES pH7.5; 25 mM MgCl2; 5 mM MnCl2; 50 mM KCl; 0.2% HSA; 0.01% CHAPS; 100 μM Na3VO4; 0.5 mM DTT). Dilution steps and concentration range were adapted according to need. 7 pl aliquots of these dilutions were transferred into a 384-well Optiplate (Perkin Elmer, #6007290). GST-Syk was diluted to 12 nM in the test buffer and 5 μl of this dilution were used in the kinase test (final concentration of Syk=4 nM in a total volume of 15 μl). After 15 minutes incubation at room temperature 3 μl of a mixture of 750 nM ATP and 100 μg/ml poly (L-Glutamic acid L-Tyrosine 4:1), Fluka #81357) in test buffer were added to each well and the incubation was continued for a further 60 minutes at room temperature.

Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.

After 60 minutes, 10 μl Kinase-Glo® solution (Promega, Cat. #V6712) (heated to room temperature) were added to each well and incubation was continued for a further 15 minutes. The plates were read in Envision Luminescence Reader (Perkin-Elmer).

Data Evaluation and Calculation:

The output file of the reader is a csv file that contains the well number and measured relative light units (RLU). For data evaluation and calculation, the measurement of the negative control was set as 100% ctrl and the measurement of the positive control was set as 0% ctrl. Based on this values the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics. The IC50 values were calculated from the % ctrl values using Assay Explorer software. Calculation:[y=(a−d)/(1+(x/c)̂b)+d]a=low value, d=high value; x=conc M; c=IC50 M; b=hill; y=% ctrl.

5.2 Aurora B Kinase Test

Recombinant human Aurora B (amino acids 1-344, clone number DU1773, Molecular weight 40.2 kDa, University of Dundee) was expressed as a fusion protein with an N-terminal His tag, affinity-purified and deep-frozen at a concentration of approx. 0.25-0.5 mg/ml in storage buffer (50 mM Tris-HCl pH 8; 25 mM Na-β-glycerophosphat; 0.1 mM EGTA; 150 mM NaCl; 0.03% Brij-35; 1 mM DTT and 10% glycerol) at −80° C. until use.

The activity of the Aurora B kinase protein was determined using the ADP Glo® Luminescence Kinase test (Promega; V9103X). In this homogeneous test the amount of ADP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ADP still present and thus correlates with the activity of the protein kinase.

Method

The test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 5 mM. Serial Dilution is done in 1:10 steps in 100% DMSO. All further dilutions of the substances were carried out with test buffer (50 mM Hepes, pH 7.5, 10 mM MgCl2, 1 mM EGTA, 60 μM Ultra Pure ATP, 0.01% Brij35, 0.1% BSA, 5 mM β-Glycerophosphate) until a concentration was reached which was 2.5 times above the final test concentration (final concentration of the compounds: 50 μM to 0.005 nM). 4 μl aliquots of these dilutions were transferred into a 384-well Optiplate (Perkin Elmer, #6007290). His-Aurora B was diluted to 125 nM in the test buffer and 4 μl of this dilution were used in the kinase test (final concentration of Aurora B=50 nM in a total volume of 10 μl). After 15 minutes incubation at room temperature 2 μl of 250 μM substrate ([LRRLSLGLRRLSLGLRRLSLGLRRLSLG]; University of Dundee) in test buffer were added to each well and the incubation was continued for a further 60 minutes at room temperature.

Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.

After 60 minutes, 10 μl ADP-Glo® solution (ADP-Glo Reagent #V912B Promega) (heated to room temperature) were added to each well and incubation was continued for a further 40. minutes. Then 20 μl Kinase detection mix (Detection Buffer #V913B Promega; Kinase Detection Substrate #V914B Promega) were added and incubated for 40 minutes at room temperature. The plates were read in Envision Luminescence Reader (Perkin-Elmer).

Data Evaluation and Calculation:

The output file of the reader is a csv file that contains the well number and measured RLU. For data evaluation and calculation, the measurement of the negative control was set as 0% ctrl and the measurement of the positive control was set as 100% ctrl. Based on this values the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics. The IC50 values were calculated from the % ctrl values using Assay Explorer software.Calculation: [y=(a−d)/(1+(x/c)̂b)+d], a=low value, d=high value; x=conc M; c=IC50 M; b=hill; y=% ctrl.

5.3 FLT3 Kinase Test

FLT3 is obtained from Invitrogen in 50 mM Tris (pH7.5); 100 mM NaCl; 0.05 mM EDTA, 0.05% NP-40, 2 mM DTT; 50% Glycerol #PV3182; Lot 286671; sequence see below). The enzyme is diluted to 720 nM (35 μg/ml) in enzyme dilution buffer and 10 μl aliquots are stored at −80° C.

The activity of FLT3 is measured using the Z′-LYTETM assay technology from Invitrogen (#PV3191)

Method

The assay is performed in 384 black plates from Corning (#3676) in a final volume of 10 μl by adding 5 μl of kinase peptide mix and 2.5 μl of compound dilution. The reaction is started by addition of 2.5 μl of the 4×]ATP solution.

Final concentration in assay: FLT3 2 nM, Tyr2 peptide 4 μM, ATP 470 μM (ATP Km for FLT3)

Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no kinase. As a further control, the phosphopeptide solution is added to wells without kinase(=100% phosphorylation control). The non inhibited kinase reaction will result in a phosphorylation corresponding to 20%-30% of the phosphorylation control.

The reaction is performed for 1 h at room temperature before 5 μl of the development solution is added. After a further incubation for 1 h at room temperature 5 μl of the stop reagent is added. The plates are read on a Flex Station II 384 (Molecular Devices).

To control for any potential inhibition of the protease present in the development solution, the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 μM or 10 μM).

Data Evaluation and Calculation:

The output text file is evaluated in an “MS-Excel-VB-Makro” and “GraphPadPrism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of FLT3 are reported in M. data for the inhibition of the protease are reported in % CTL.

5.4 GSK 3β Kinase-Test

Human GSK3beta (expressed and purified from SF21 cells)is obtained from the University Dundee/Scotland (Dr. James Hastie—Dept. of Biochemistry) in 50 mM Tris (pH7.5); 150 mM NaCl; 0.1 mM EGTA, 270 mM Succrose, 0.1% R-mercaptoethanol, 1 mM benzamidine, 0.2 mM PMSF; sequence see below). The enzyme is diluted to 3.56 μM (168 μg/ml) in enzyme dilution buffer and 6 μl aliquots are stored at −80° C.

The activity of GSK3β kinase protein is measured using the Z′-LYTETM assay technology from Invitrogen (#PV3324).

Method:

The assay is performed in 384 black plates from Corning (#3676) in a final volume of 10 μl by adding 5 μl of kinase peptide mix and 2.5 μl of compound dilution. The reaction is started by addition of 2.5 μl of the 4× ATP solution.

Final concentration in assay: GSK3β 5 nM, Ser/Thr9 peptide 2 μM, ATP 7 μM (ATP Km for GSK3β)

Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no ATP. As a further control, the phosphopeptide solution is added to wells without kinase and without ATP(=100% phosphorylation control). The non inhibited kinase reaction will result in a phosphorylation corresponding to 20%-30% of the phosphorylation control.

The reaction is performed 1 h at room temperature. After 1 h 5 μl of the development solution is added. After a further incubation for 1 h at room temperature 5 μl of the stop reagent is added. Finally the plates are read on a Flex Station II 384 (Molecular Devices).

To control for any potential inhibition of the protease present in the development solution, the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 μM).

Data Evaluation and Calculation:

The output text file is evaluated in an “MS-Excel-VB-Makro” and “Graph Pad Prism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of GSK3beta are reported in M. data for the inhibition of the protease are reported in % CTL.

5.5 Human Liver Microsomal Stability Test

Further it is desirable for an SYK-inhibitor that is sufficiently SYK-specific as described above to have certain human liver microsomal stability (corresponding to CI<60% Qh; % Qh=percentage of liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated.

Method:

The metabolic degradation for a specific SYK-inhibitor is performed at 37° C. with pooled human liver microsomes (human liver microsomes are commercially available as “BD UltraPool™” by Corning Life Sciences, Fogostraat 12, 1060 LJ Amsterdam, The Netherlands). The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 μM.

Following a short preincubation period at 37° C., the reaction is initiated by addition of beta-nicotinamide adenine dinucleotide phosphate in its reduced form (NADPH, 1 mM) and terminated by transfering an aliquot into solvent after different time points. Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point.

The quenched (terminated) incubations are then pelleted by centrifugation (10000 g, 5 min).

An aliquot of the supernatant is assayed by LC-MS/MS for the remaining amount of parent compound. The half-life (t1/2 INVITRO) is determined by the slope of the semilogarithmic plot of the concentration-time profile.

Data Evaluation and Calculation:

The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation:


CL_INTRINSIC [μl/min/mg protein]=(Ln 2/(t1/2 INVITRO [min] * protein content [mg/ml]))*1000

The protein content [mg/ml] was determined with the “Bicinchoninic Acid Kit” of Sigma Aldrich (commercially available).

The upscaled intrinsic Clearance (CL_UP_INT) is calculated by considering the liver weight [g liver/kg body weight] and the microsomal recovery [mg protein/g liver]:


CL_UP_INT [ml/min/kg]=0.001*CL_INTRINSIC*liver weight*microsomal recovery

    • with microsomal recovery=45 mg protein/g liver
    • with liver weight=25.7 g liver/kg body weight

The percent hepatic blood flow (% Qh) is finally calculated by considering the human liver blood flow Q [ml/min/kg]:


% Qh [%]=((Q*CL_UP_INT)/(Q+CL_UP_INT)/Q)*100

    • with liver blood flow (Q)=20.7 ml/min/kg.

TABLE 1 SYK- AURB- FLT3- inhibition inhibition inhibition IC50- IC50- IC50- Example value value value No. Structure [μM] [μM] [μM]  6 0.1508 >50 35.80 34 0.0122 >50 7.93 84 0.6366 >50 >50

6. COMPARISON OF SYK-INHIBITORY CAPACITY AND OF SYK-SELECTIVITY OF THE COMPOUNDS OF THE INVENTION COMPARED TO SELECTED COMPOUNDS OF WO 2013/014060 AND OF WO 2011/092128

To have an efficient SYK-inhibitory capacity is not the only important aspect which a SYK-inhibitor to be used as a medicament to treat SYK-related diseases must show. Similarly important like the low IC50-value with regard to SYK-inhibition (IV50 (SYK)≦1 μM) is that the candidate compound does not show undesired inhibitory effects on other kinases which could lead to unwanted or even dangerous side effects. Examples of such other kinases that should not be inhibited by the candidate SYK-inhibitor are AURB, FLT3 and GSKbeta.

Consequently the IC50-values with regard to SYK, AURB, FLT3 and GSKbeta for structurally close compounds disclosed in WO 2013/014060 and of WO 2011/092128 have been experimentally determined according to the same assays as described in chapter 5. The measured IC50-values with regard to SYK, AURB, FLT3 and GSKbeta of these structurally closest prior art compounds are in the following tables 2a to 6c compared to the respective previously determined IC50-values of a representative selection of compounds of the invention (same assay conditions).

Further it is desirable for an SYK-inhibitor that is sufficiently SYK-specific as described above to have certain human liver microsomal stability (corresponding to CI<60% Qh; Qh=liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated. Consequently also the CI-values for structurally close compounds disclosed in WO 2013/014060 and in WO 2011/092128 have been experimentally determined according to the same human liver microsomal-test as described in chapter 5. An experimentally determined CI-value of more than 60% Qh is regarded to be inacceptable in order to reach an adequate plasma level of the respective SYK-inhibitor in the patient to be treated.

6.1 Comparisons with Structurally Similar Prior Art Compounds

Whereas all compounds of the invention (see Table 2a), of WO 2013/014060 (see Table 2b) and of WO 2011/092128 (see Table 2c) have suitable IC50 (SYK)-values of smaller than 1 μM, only the compounds of the invention (see Table 2a) have IC50-values with regard to AURB of more than 50 μM (compared to IC50-values (AURB) of below 3 μM for the compounds of WO 2013/014060 in Table 2b and for the compounds of W02011/092128 in Table 2c). Also the IC50-values with respect to FLT3 are larger for the compounds of the invention (Table 2a) compared to the compounds of WO 2013/014060 (see Table 2b) and to the compounds of WO2011/092128 (see Table 2c). Consequently the compounds of the invention are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 2b) and of WO2011/092128 (see Table 2c)), but also do not have unwanted inhibitory effects on other kinases such as AURB, FLT3 and GSK3beta (unlike the compounds of WO 2013/014060 (see Table 2b) and of WO2011/092128 (see Table 2c)). The compounds of the invention therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060 and in WO2011/092128.

TABLE 2a compounds of the invention GSK3- SYK- AURB- FLT3- beta inhibition inhibition inhibition inhib. micosomal IC50- IC50- IC50- IC50- stability Ex. value value value value Cl No. Structure [μM] [μM] [μM] [μM] [% Qh]  6 0.1508 >50 35.80 >10 <23 34 0.0122 >50 7.93 >10 <23 84 0.6366 >50 >50 >10 <23

TABLE 2b Compounds of WO 2013/014060; IC50 IC50 IC50 IC50 microsomal in μM in μM in μM in μM stability Ex. (SYK- (AURB- (FLT3- (GSK3beta- Cl No. Structure Inhibition) Inhibition) Inhibition) Inhibition) [% Qh] 112 0.006  2.96 1.25  >10 <23 114 0.0002 2.54 0.049 >10  26 115 0.0002  0.021 0.041 >10 <23

TABLE 2c: Compounds of WO 2011/092128: IC50 IC50 IC50 IC50 microsomal in μM in μM in μM in μM stability Ex. (SYK- (AURB- (FLT3- (GSK3beta- Cl No. Structure Inhibition) Inhibition) Inhibition) Inhibition) [% Qh]  3 0.0122 1.04  1.82  >10 24 45 0.0002 0.086 0.100 >10 53

6. INDICATIONS

As has been found, the compounds of formula 1 are characterised by their range of applications in the therapeutic field. Particular mention should be made of those applications for which the compounds of formula 1 according to the invention are preferably used on the basis of their pharmaceutical activity as Syk-inhibitors. Examples include respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases or complaints, immune or autoimmune diseases, allergic diseases, inflammatory diseases, e.g. inflammatory diseases of the joints, skin and eyes and diseases of the peripheral or central nervous system.

Particular mention should be made of the prevention and treatment of respiratory tract and pulmonary diseases which are accompanied by increased mucus production, inflammation and/or obstructive diseases of the airways. Examples of these include asthma, paediatric asthma, ARDS (Adult Respiratory Distress Syndrome), acute, allergic or chronic bronchitis, autoimmune haemolytic anemia, chronic obstructive bronchitis (COPD) (including the treatment of Rhinovirus-induced exacerbations), coughs, allergic rhinitis or sinusitis, allergic rhinoconjunctivitis, chronic rhinitis or sinusitis, alveolitis, farmers' lung, hyperreactive airways, infectious bronchitis or pneumonitis, bronchiectasis, pulmonary arterial hypertension, pulmonary fibrosis, bronchial oedema, pulmonary oedema, pneumonia or interstitial pneumonia triggered by various causes such as aspiration, inhalation of toxic gases or bronchitis, pneumonia or interstitial pneumonia triggered by cardiac insufficiency, radiation, chemotherapy, cystic fibrosis or mucoviscidosis, alpha 1-antitrypsin deficiency.

The compounds according to the invention are preferably also suitable for the treatment of allergic diseases such as for example allergic rhinitis, allergic rhinoconjunctivitis, allergic conjunctivitis, and contact dermatitis, urticaria/angiooedema and allergic dermatitis.

Mention should also preferably be made of the treatment of inflammatory diseases of the gastrointestinal tract. Examples of these are Crohn's disease and ulcerative colitis.

The compounds according to the invention are preferably also suitable for the treatment of inflammatory diseases of the joints, of the blood vessels and of the kidney or inflammatory diseases of the skin and eyes. Examples of these are rheumatoid arthritis, antibody-based glomerulonephritis, psoriasis, Kawasaki syndrome, coeliac disease (sprue), arteriosclerosis and Wegener's granulomatosis, osteoarthritis, systemic scleroderma, ankylosing spondylitis.

The compounds according to the invention are preferably also suitable for the treatment of autoimmune diseases. Examples of these are hepatitis (autoimmune-based), lupus erythematodes, lupus nephritis, systemic lupus, Systemic lupus erythematosus,discoid lupus, cutaneous lupus erythematosus (acute, subacute, chronic), anti-phospholipid syndrome, Berger's disease, Evans's syndrome, immunohaemolytic anaemia, ITP (idiopathic thrombocytopenic purpura; adult, neonatal and paediatric), myasthenia gravis, Sjögren's syndrome, sclerodermy, Bullous pemphigoid and Pemphigus vulgaris.

The compounds according to the invention are preferably also suitable for the treatment of B-cell lymphomas, like chronic lymphocytic leukaemia and non-Hodgkin's lymphomas or T cell lymphomas.

The compounds according to the invention are preferably also suitable for the treatment of Graft-versus-host disease.

Mention may preferably also be made of the prevention and treatment of diseases of the peripheral or central nervous system. Examples of these are acute and chronic multiple sclerosis or non-familial lateral sclerosis.

Mention may preferably also be made of the prevention and treatment of osteoporotic diseases such as for example disease-associated osteopenia, osteoporosis and osteolytic diseases.

The present invention relates particularly preferably to the use of compounds of formula 1 for preparing a pharmaceutical composition for the treatment of diseases selected from among asthma, COPD, allergic rhinitis, Adult Respiratory Distress Syndrome, bronchitis, allergic dermatitis, contact dermatitis, ITP, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, and allergic rhinoconjunctivitis.

Most preferably, the compounds of formula 1 may be used for the treatment of a disease selected from among asthma, allergic rhinitis, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, allergic dermatitis and COPD.

7. COMBINATIONS

The compounds of formula 1 may be used on their own or in conjunction with other active substances of formula 1 according to the invention. The compounds of formula 1 may optionally also be used in conjunction with other pharmacologically active substances. Preferably the active substances used here may be selected for example from among the betamimetics, anticholinergics, corticosteroids, PDE4-inhibitors, LTD4-antagonists, EGFR-inhibitors, MRP4-inhibitors, dopamine agonists, H1-antihistamines, PAF-antagonists, iNos-inhibitos, HMG-CoA reductase inhibitors (statins), PI3-kinase-inhibitors, CCR3-antagonists, CCR2-antagonists, CCR1-antagonists, IKK2-inhibitors, A2a agonists, alpha-4-integrin-inhibitors, CRTH2-antagonists, histamine 1, combined H1/H3-antagonists, p38 kinase inhibitors, methylxanthines, ENaC-inhibitors, CXCR1-antagonists, CXCR2-antagonists, ICE-inhibitors, LTB4-antagonists, 5-LO antagonists, FLAP-antagonists. LTB4-antagonists; cromoglycine, dissociated glucocorticoid mimetics, immunesuppressive agents, cytostatica, non-steroidal anti-inflammatory drugs (NSAIDs), chloroquine, hydroxychloroquine, 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-IL6 antibodies, anti-IL6 receptor antibodies, anti-IL-13-antibodies, anti-IL_18 antibodies, anti-CD30 L antibodies, anti-Ox40L-antibodies, anti-IL-4/IL-13-antibodies, anti-IL-23 (p19) antibodies, anti-IL-12/IL-23 (p40) antibodies, anti-CD3 antibodies, anti-CD4 antibodies, anti-CD154 antibodies, CD89 antibodies, anti-IL-2 receptor/CD25 antibodies, anti-CD22 antibodies, anti-interferon antibodies, anti-ICOS antibodies, anti-ICOS antibodies, anti-CD20 antibodies, anti-CD40 antibodies, anti-BAFF/BLyS antibodies, anti-CD18 antibodies, anti-CD62L antibodies, anti-CD147 antibodies, anti-integrin antibodies, agents interfering with LFA-1, IL-36 pathway modulators, M-CSF/c-fms antagonists, CTLA-4 fusions, mTor modulators, Toll like receptors 7 inhibitors (TLR7 inhibitor), Toll like receptor 9 inhibitors (TLR9 inhibitors), T cell-costimulatory modulators such as CTLA-4 fusions, JAK inhibitors, IRF modulators, CX3 chemokine receptor antagonists (CX3CR1 antagonists), IRAK inhibitors (in particular IRAK1- and IRAK4-inhibitors), Sphingosine-1-phosphate modulators (S1P pathway modulators),

or double or triple combinations thereof, such as for example combinations of one, two or three compounds selected from among the

    • Syk-inhibitors of formula 1, betamimetics, corticosteroids, EGFR-inhibitors and PDE4-antagonists,
    • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids, EGFR-inhibitors and PDE4-antagonists,
    • Syk-inhibitors of formula 1, PDE4-inhibitors, corticosteroids and EGFR-inhibitors,
    • Syk-inhibitors of formula 1, EGFR-inhibitors and PDE4-inhibitors,
    • Syk-inhibitors of formula 1 and EGFR-inhibitors,
    • Syk-inhibitors of formula 1, betamimetics and anticholinergics
    • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids and PDE4-inhibitors,
    • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids, iNOS inhibitors, HMG-CoA reductase inhibitors.

Combinations of three active substances each taken from one of the above-mentioned categories of compounds are also an object of the invention.

Suitable betamimetics used are preferably compounds selected from among arformoterol, carmoterol, formoterol, indacaterol, sal meterol, albuterole, bambuterol, bitolterol, broxaterol, carbuterol, clenbuterol, fenoterol, hexoprenalin, ibuterol, isoetharin, isoprenalin, levosalbutamol, mabuterol, meluadrin, metaproterenol, milveterol, orciprenalin, pirbuterol, procaterol, reproterol, rimiterol, ritodrin, salmefamol, soterenol, sulphonterol, terbutalin, tiaramide, tolubuterol, zinterol, 6-Hydroxy-8-{1-hydroxy-2-[2-(4-methoxy-phenyl)-1,1-dimethyl-ethylamino]-ethyl}-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(2,4-Difluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(3,5-Difluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(4-Ethoxy-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo [1,4]oxazine-3-one; 8-{2-[2-(4-Fluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; N-(5-{2-[3-(4,4-Diethyl-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenylymethansulfonamide; N-(5-{2-[3-(4,4-Diethyl-6-fluoro-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenylymethansulfonamide; N-(5-{2-[3-(4,4-Diethyl-6-methoxy-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; N-(5-{2-[1,1-Dimethyl-3-(2-oxo-4,4-dipropyl-4H-benzo[d][1,3]oxazine-1-yl)-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; 8-{2-[1,1-Dimethyl-3-(2-oxo-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(6-methyl-2-oxo-2,3-dihydro-benzoimidazole-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(2-oxo-5-trifluormethyl-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(3-methyl-2-oxo-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; N-[2-Hydroxy-5-((1R)-1-hydroxy-2-{2-[4-(2-hydroxy-2-phenyl-ethylamino)-phenyl]-ethylamino}-ethyly)-phenyl]-formamide; 8-Hydroxy-5-((1R)-1-hydroxy-2-{2-[4-(6-methoxy-biphenyl-3-ylamino)-phenyl]-ethylamino}-ethyl)-1H-quinoline-2-one; 8-Hydroxy-5-[(1R)-1-hydroxy-2-(6-phenethylamino-hexylamino)-ethyl]-1H-quinoline-2-one; 5-[(1R)-2-(2-{4-[4-(2-Amino-2-methyl-propoxy)-phenylamino]-phenyl}-ethylamino)-1-hydroxy-ethyl]-8-hydroxy-1H-quinoline-2-one; [3-(4-{6-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-hexyloxy}-butyl)-5-methyl-phenyl]-urea; 4-((1R)-2-{6-[2-(2,6-Dichlor-benzyloxy)-ethoxy]-hexylamino}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol; 3-(4-{6-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-hexyloxy}-butyl)-benzenesulfonamide; 3-(3-{7-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-heptyloxy}-propyl)-benzenesulfonamide; 4-((1R)-2-{6-[4-(3-Cyclopentanesulfonyl-phenyl)-butoxy]-hexylaminoyl}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol, 4-(2-{6-[2-(2,6-dichloro-benzyloxy)-ethoxy]-hexylamino}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol; Vilanterol; N-1-Adamantanyl-2-{3-[(2R)-2-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl}amino)propyl]phenyl}acetamide; 2-(3-{2-[2-hydroxy-3-methanesulfonylamino-phenyl)-ethylamino]-propyl}-phenyl)-N-[4-(4-hydroxy-phenyl)-2-vinyl-penta-2,4-dienyl]-acetamide; (1R)-5-{2-[6-(2,2-Difluor-2-phenyl-ethoxy)-hexylamino]-1-hydroxy-ethyl}-8-hydroxy-1H-quinoline-2-one; (R,S)-4-(2-{[6-(2,2-Difluor-4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[4,4-Difluor-6-(4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(4,4-Difluor-4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-5-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxy-ethyl)-8-hydroxyquinoline-2(1H)-one; (R,S)-[2-({6-[2,2-Difluor-2-(3-methylphenyl)ethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; 4-(1R)-2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxyethyl)-2-(hydroxymethyl)phenol; (R,S)-2-(Hydroxymethyl)-4-(1-hydroxy-2-{[4,4,5I5-tetrafluor-6-(3-phenylpropoxy)-hexyl]amino}ethyl)phenol; (R,S)-[5-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxy-ethyl)-2-hydroxyphenyl]formamide; (R,S)-4-[2-({6-[2-(3-Bromophenyl)-2,2-difluoroethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; (R,S)—N-[3-(1,1-Difluor-2-{[6-({2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]-ethyl}amino)hexyl]oxy}ethyl)phenyl]-urea; 3-[3-(1,1-Difluor-2-{[6-({2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl) phenyl]ethyl}amino)hexyl]oxy}ethyl)phenyl]imidazolidine-2,4-dione; (R,S)-4-[2-({6-[2,2-Difluor-2-(3-methoxyphenyl)ethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; 5-((1R)-2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxyethyl)-8-hydroxyquinoline-2(1H)-one; 4-((1R)-2-{[4,4-Difluor-6-(4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(3,3-Difluor-3-phenylpropoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-(2-{[6-(2,2-Difluor-2-phenylethoxy)-4,4-difluorohexyl]amino}-1-hydroxyethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(2,2-Difluor-3-phenylpropoxy)hexyl]amino}-1-hydroxy ethyl)-2-(hydroxymethyl)phenol; 3-[2-(3-Chlor-phenyl)-ethoxy]-N-(2-diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethylypropionamide; N-(2-Diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-3-(2-naphthalen-1-yl-ethoxy)-propionamide; 7-[2-(2-{3-[2-(2-Chlor-phenyl)-ethylamino]-propylsulfanyl}-ethylamino)-1-hydroxy-ethyl]-4-hydroxy-3H-benzothiazol-2-one, optionally in the form of the racemates, enantiomers, diastereomers and optionally in the form of the pharmacologically acceptable acid addition salts, solvates or hydrates thereof.

According to the invention the acid addition salts of the betamimetics are preferably selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably the hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate. Of the above-mentioned acid addition salts the salts of hydrochloric acid, methanesulphonic acid, benzoic acid and acetic acid are particularly preferred according to the invention.

The anticholinergics used are preferably compounds selected from among tiotropium salts, particularly the bromide salt, oxitropium salts, particularly the bromide salt, flutropium salts, particularly the bromide salt, ipratropium salts, particularly the bromide salt, Aclidinium salts, particularly the bromide salt, glycopyrronium salts, particularly the bromide salt, trospium salts, particularly the chloride salt, tolterodin, (3R)-1-Phenethyl-3-(9H-xanthene-9-carbonyloxy)-1-azoniabicyclo[2.2.2]octan-salts; 2,2-Diphenyl propionic acid tropenole ester-methobromide; 2,2-Diphenyl propionic acid scopine ester-methobromide; 2-Fluor-2,2-Diphenyl acetic acid scopine ester-methobromide; 2-Fluor-2,2-Diphenyl acetic acid tropenole ester-methobromide; 3,3′,4,4′-Tetrafluor benzilic acid tropenole ester-methobromide; 3,3′,4,4′-Tetrafluor benzilic acid scopine ester-methobromide; 4,4′-Difluor benzilic acid tropenole ester-methobromide; 4,4′-Difluor benzilic acid scopine ester-methobromide; 3,3′-Difluor benzilic acid tropenole ester-methobromide; 3,3′-Difluor benzilic acid scopine ester-methobromide; 9-Hydroxy-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Fluor-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxy-fluorene-9-carboxylic acid scopine ester-methobromide; 9-Fluor-fluorene-9-carboxylic acid scopine ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid scopine ester-methobromide; Benzilic acid cyclopropyl tropine ester-methobromide; 2,2-Diphenyl propionic acid cyclopropyltropine ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Hydroxy-fluorene-9-carboxilic acid cyclopropyltropine ester-methobromide; 4,4′-Difluor benzilic acid methyl ester cyclopropyltropine ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid scopine ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid scopine ester-methobromide; 9-Ethyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Difluormethyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxymethyl-xanthene-9-carboxylic acid scopine ester-methobromide;

3-[2-(3-Chloro-phenyl)-ethoxy]-N-(2-diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-propionamide;

N-(2-Diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-3-(2-naphthalen-1-yl-ethoxy)-propionamide;

7-[2-(2-{3-[2-(2-Chloro-phenyl)-ethylamino]-propylsulfanyl}-ethylamino)-1-hydroxy-ethyl]-4-hydroxy-3H-benzothiazol-2-one and Darotropium;

optionally in the form of the solvates or hydrates thereof.

In the above-mentioned salts the cations tiotropium, oxitropium, flutropium, ipratropium, glycopyrronium, aclidinium and trospium are the pharmacologically active ingredients. As anions, the above-mentioned salts may preferably contain chloride, bromide, iodide, sulphate, phosphate, methanesulphonate, nitrate, maleate, acetate, citrate, fumarate, tartrate, oxalate, succinate, benzoate or p-toluenesulphonate, while chloride, bromide, iodide, sulphate, methanesulphonate or p-toluenesulphonate are preferred as counter-ions. Of all the salts, the chlorides, bromides, iodides and methanesulphonate are particularly preferred.

Of particular importance is tiotropium bromide. In the case of tiotropium bromide the pharmaceutical combinations according to the invention preferably contain it in the form of the crystalline tiotropium bromide monohydrate, which is known from WO 02/30928. If the tiotropium bromide is used in anhydrous form in the pharmaceutical combinations according to the invention, it is preferable to use anhydrous crystalline tiotropium bromide, which is known from WO 03/000265.

Corticosteroids used here are preferably compounds selected from among beclomethasone, betamethasone, budesonide, butixocort, ciclesonide, deflazacort, dexamethasone, etiprednole, flunisolide, fluticasone, loteprednole, mometasone, prednisolone, prednisone, rofleponide, triamcinolone, tipredane; Pregna-1,4-diene-3,20-dione, 6-fluoro-11-hydroxy-16,17-[(1-methylethylidene) bis(oxy)]-21-[[4-[(nitrooxy)methyl]benzoyl]oxy]-, (6-alpha,11-beta,16-alpha)-(9Cl); 16,17-butylidenedioxy-6,9-difluoro-11-hydroxy-17-(methylthio)androst-4-en-3-one; 6,9-Difluor-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-dien-17-carbothione acid (S)-fluoromethylester; (S)-fluoromethyl 6,9-difluoro-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-diene-17-carbothionate; 6-alpha,9-alpha-difluoro-11-beta-hydroxy-16alpha-methyl-3-oxo-17alpha-(2,2,3,3-tetramethylcyclopropylcarbonyl)oxy-and rosta-1,4-diene-17beta-carboxylic acid cyanomethyl ester, each optionally in the form of the racemates, enantiomers or diastereomers thereof and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.

Particularly preferably the steroid is selected from among budesonide, fluticasone, mometasone, ciclesonide and (S)-fluoromethyl 6,9-difluoro-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-diene-17-carbothionate, optionally in the form of the racemates, enantiomers or diastereomers thereof and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.

Any reference to steroids includes a reference to any salts or derivatives, hydrates or solvates thereof which may exist. Examples of possible salts and derivatives of the steroids may be: alkali metal salts, such as for example sodium or potassium salts, sulfobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates thereof.

PDE4 inhibitors which may be used are preferably compounds selected from among enprofyllin, theophyllin, roflumilast, ariflo (cilomilast), tofimilast, pumafentrin, lirimilast, apremilast, arofyllin, atizoram, oglemilast, tetomilast; 5-[(N-(2,5-dichloro-3-pyridinyl)-carboxamide]-8-methoxy-Quinoline (D-4418); 5-[N-(3,5-dichloro-1-oxido-4-pyridinyl)-carboxamide]-8-methoxy-2-(trifluoromethyl)-Quinoline (D-4396 (Sch-351591)); N-(3,5-dichloropyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indol-3-yl]glyoxylic acid amide (AWD-12-281 (GW-842470)); 9-[(2-fluorophenyl)methyl]-N-methyl-2-(trifluoromethyl)-9H-Purin-6-amine (NCS-613); 4-[(2R)-2-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-phenylethyl]-Pyridine (CDP-840); N-[(3R)-3,4,6,7-tetrahydro-9-methyl-4-oxo-1-phenylpyrrolo[3,2,1-jk][1,4]benzodiazepin-3-yl]-4-Pyridinecarboxamide (PD-168787); 4-[6,7-diethoxy-2,3-bis(hydroxymethyl)-1-naphthalenyl]-1-(2-methoxyethyl)-2(1H)-Pyridinone (T-440); 2-[4-[6,7-diethoxy-2,3-bis(hydroxymethyl)-1-naphthalenyl]-2-pyridinyl]-4-(3-pyridinyl)-1(2H)-Phthalazinone (T-2585); (3-(3-cyclopenyloxy-4-methoxybenzyl)-6-ethylamino-8-isopropyl-3H-purine (V-11294A); beta-[3-(cyclopentyloxy)-4-methoxyphenyl]-1,3-dihydro-1,3-dioxo-2H-lsoindole-2-propanamide (CDC-801); Imidazo[1,5-a]pyrido[3,2-e]pyrazine-6(5H)-one, 9-ethyl-2-methoxy-7-methyl-5-propyl-(D-22888); 5-[3-(cyclopentyloxy)-4-methoxypheny]-3-[(3-methylphenyl)methyl]-, (3S,5S)-2-Piperidinon (HT-0712); 4-[1-[3,4-bis(difluoromethoxy)phenyl]-2-(3-methyl-1-oxido-4-pyridinyl)ethyl]-alpha,alpha-bis(trifluoromethyl)-Benzenemethanol (L-826141); N-(3,5-Dichloro-1-oxo-pyridin-4-yl)-4-difluormethoxy-3-cyclopropylmethoxybenzamide; (−)p-[(4aR*,10bS*)-9-Ethoxy-1,2,3,4,4a,10b-hexahydro-8-methoxy-2-methylbenzo[s][1,6]naphthyridin-6-yl]-N,N-diisopropylbenzamide; (R)-(+)-1-(4-Brombenzyl)-4-[(3-cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidon; 3-(Cyclopentyloxy-4-methoxyphenyl)-1-(4-N′-[N-2-cyano-S-methyl-isothioureido]benzyl)-2-pyrrolidon; cis[4-Cyano-4-(3-cyclopentyloxy-4-methoxyphenyl)cyclohexan-1-carboxylic acid]; 2-carbomethoxy-4-cyano-4-(3-cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-1-one; cis[4-Cyano-4-(3-cyclopropylmethoxy-4-difluormethoxyphenyl)cyclohexan-1-ol]; (R)-(+)-Ethyl[4-(3-cyclopentyloxy-4-methoxyphenyl)pyrrolidin-2-yliden]acetat; (S)-(−)-Ethyl[4-(3-cyclopentyloxy-4-methoxyphenyl)pyrrolidin-2-yliden]acetat; 9-Cyclopentyl-5,6-dihydro-7-ethyl-3-(2-thienyl)-9H-pyrazolo[3,4-c]-1,2,4-triazolo[4,3-a]pyridin; 9-Cyclopentyl-5,6-dihydro-7-ethyl-3-(tert-butyl)-9H-pyrazolo[3,4-c]-1,2,4-triazolo[4,3-a]pyridin,

optionally in the form of the racemates, enantiomers or diastereomers and optionally in the form of the pharmacologically acceptable acid addition salts, solvates and/or hydrates thereof.

By acid addition salts with pharmacologically acceptable acids which the above-mentioned PDE4-inhibitors might be in a position to form are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.

LTD4-antagonists which may be used are preferably compounds selected from among montelukast, pranlukast, zafirlukast; (E)-8-[2-[4-[4-(4-Fluorophenyl)butoxy]phenyl]ethenyl]-2-(1H-tetrazol-5-yl)-4H-1-benzopyran-4-one (MEN-91507); 4-[6-Acetyl-3-[3-(4-acetyl-3-hydroxy-2-propylphenylthio)propoxy]-2-propylphenoxy]-butyric acid (MN-001); 1-(((R)-(3-(2-(6,7-Difluor-2-quinolinyl)ethenyl)phenyl)-3-(2-(2-hydroxy-2-propyl)phenyl)thio)methylcyclopropane-acetic acid; 1-(((1(R)-3(3-(2-(2,3-Dichlorthieno[3,2-b]pyridin-5-yl)-(E)-ethenyl)phenyl)-3-(2-(1-hydroxy-1-methylethyl)phenyl) propyl)thio)methyl)cyclopropane acetic acid; [2-[[2-(4-tert-Butyl-2-thiazolyl)-5-benzofuranyl]oxymethyl]phenyl] acetic acid,

optionally in the form of the racemates, enantiomers or diastereomers, optionally in the form of the pharmacologically acceptable acid addition salts and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.

By acid addition salts with pharmacologically acceptable acids which the LTD4-antagonists may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate. By salts or derivatives which the LTD4-antagonists may be capable of forming are meant, for example: alkali metal salts, such as, for example, sodium or potassium salts, alkaline earth metal salts, sulphobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates.

The EGFR-inhibitors used are preferably compounds selected from among 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-diethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-2-methoxymethyl-6-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-(N,N-bis-(2-methoxy-ethyl)-amino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4N-(2-methoxy-ethyl)-N-ethyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4-[N-(tetrahydropyran-4-yl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-Phenyl-ethyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-Phenyl-ethyl)amino]-6-({4-[N-(tetrahydropyran-4-yl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-((R)-tetrahydrofuran-3-yloxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-((S)-tetrahydrofuran-3-yloxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N-cyclopropyl-N-methyl-amino)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-[(R)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6.7-bis-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-7-[3-(morpholine-4-yl)-propyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(R)-(1-phenyl-ethypamino]-6-(4-hydroxy-phenyl)-7H-pyrrolo[2,3-d]pyrimidine, 3-cyano-4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-ethoxy-quinoline, 4-{[3-chloro-4-(3-fluoro-benzyloxy)-phenyl]amino}-6-(5-{[(2-methanesulphonyl-ethyl)amino]methyl}-furan-2-yl)quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]-amino}-7-[(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N,N-bis-(2-methoxy-ethyl)-amino]-1-oxo-2-butene-1-yl}amino)-7-[(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-{[4-(5.5-dimethyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-[(R)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-6-[(S)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{2-[4-(2-oxo-morpholine-4-yl)-piperidine-1-yl]-ethoxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(tert.-butyloxycarbonyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-amino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-methanesulphonylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-3-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(methoxymethyl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(piperidine-3-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(2-acetylamino-ethyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-ethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-((S)-tetrahydrofuran-3-yloxy)-7-hydroxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[(dimethylamino)sulphonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[(morpholine-4-yl)carbonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[(morpholine-4-yl)sulphonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-acetylamino-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-methanesulphonylamino-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(piperidine-1-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-aminocarbonylmethyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(tetrahydropyran-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(morpholine-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazolin; 4-{2-[4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yloxy]-ethyl}-6-methyl-morpholine-2-one, 4-{4-[4-(3-chloro-2-fluoro-phenylamino)-7-methoxy-quinazolin-6-yloxy]-cyclohexyl}-1-methyl-piperazine-2-one, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(morpholine-4-yl)sulphonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-ethansulphonylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-ethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(2-methoxy-acetyl)-piperidine-4-yloxy]-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-acetylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-[1-(tert.-butyloxycarbonyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(tetrahydropyran-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(piperidine-1-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(4-methyl-piperazine-1-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{cis-4-[(morpholine-4-yl)carbonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[2-(2-oxopyrrolidin-1-yl)ethyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-acetyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-isopropyloxycarbonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-methylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{cis-4-[N-(2-methoxy-acetyl)-N-methyl-amino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-[1-(2-methoxy-acetyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(cis-2,6-dimethyl-morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(2-methyl-morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(S,S)-(2-oxa-5-aza-bicyclo[2,2,1]hept-5-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(N-methyl-N-2-methoxyethyl-amino)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-ethyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(2-methoxyethyl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(3-methoxypropyl-amino)-carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[cis-4-(N-methanesulphonyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[cis-4-(N-acetyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-methylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[trans-4-(N-methanesulphonyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-dimethylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-{N-[(morpholine-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-[(S)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-cyano-piperidine-4-yloxy)-7-methoxy-quinazoline, 3-Cyano-4-[(3-chlor-4-fluorphenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-ethoxy-quinoline, [4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-(homomorpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-(2-{4-[(S)-(2-oxo-tetrahydrofuran-5-yl)carbonyl]-piperazine-1-yl}-ethoxy)-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((S)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-(2-{4-[(S)-(2-oxo-tetrahydrofuran-5-yl)carbonyl]-piperazine-1-yl}-ethoxy)-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((S)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, cetuximab, trastuzumab, panitumumab(=ABX-EGF), Mab ICR-62, gefitinib, pelitinib, canertinib and erlotinib, optionally in the form of the racemates, enantiomers or diastereomers thereof, optionally in the form of the pharmacologically acceptable acid addition salts thereof, the solvates and/or hydrates thereof.

By acid addition salts with pharmacologically acceptable acids which the EGFR-inhibitors may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.

Examples of dopamine agonists which may be used preferably include compounds selected from among bromocriptine, cabergoline, alpha-dihydroergocryptine, lisuride, pergolide, pramipexol, roxindol, ropinirol, talipexol, terguride and viozan. Any reference to the above-mentioned dopamine agonists within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts and optionally hydrates thereof which may exist. By the physiologically acceptable acid addition salts which may be formed by the above-mentioned dopamine agonists are meant, for example, pharmaceutically acceptable salts which are selected from the salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid and maleic acid.

Examples of H1-antihistamines preferably include compounds selected from among epinastine, cetirizine, azelastine, fexofenadine, levocabastine, loratadine, mizolastine, ketotifen, emedastine, dimetinden, clemastine, bamipin, cexchlorpheniramine, pheniramine, doxylamine, chlorophenoxamine, dimenhydrinate, diphenhydramine, promethazine, ebastine, olopatadine, desloratidine and meclozine. Any reference to the above-mentioned H1-antihistamines within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts which may exist.

Examples of PAF-antagonists preferably include compounds selected from among lexipafant, 4-(2-chlorophenyl)-9-methyl-2-[3(4-morpholinyl)-3-propanon-1-yl]-6H-thieno-[3,2-f]-[1,2,4]triazolo[4,3-a][1,4]diazepines, 6-(2-chlorophenyl)-8,9-dihydro-1-methyl-8-[(4-morpho-linyl)carbonyl]-4H,7H-cyclo-penta-[4,5]thieno-[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepines. Any reference to the above-mentioned above-mentioned PAF-antagonists includes within the scope of the present invention a reference to any pharmacologically acceptable acid addition salts thereof which may exist.

Examples of non-steroidal anti-inflammatory drugs (NSAIDs) preferably include compounds selected from among Aceclofenac, Acemetacin, Acetylsalicylsäure, Alclofenac, Alminoprofen, Amfenac, Ampiroxicam, Antolmetinguacil, Anirolac, Antrafenin, Azapropazon, Benorilat, Bermoprofen, Bindarit, Bromfenac, Bucloxinsäure, Bucolom, Bufexamac, Bumadizon, Butibufen, Butixirat, Carbasalatcalcium, Carprofen, Cholin Magnesium Trisalicylat, Celecoxib, Cinmetacin, Cinnoxicam, Clidanac, Clobuzarit, Deboxamet, Dexibuprofen, Dexketoprofen, Diclofenac, Diflunisal, Droxicam, Eltenac, Enfenaminsäure, Etersalat, Etodolac, Etofenamat, Etoricoxib, Feclobuzon, Felbinac, Fenbufen, Fenclofenac, Fenoprofen, Fentiazac, Fepradinol, Feprazon, Flobufen, Floctafenin, Flufenaminsäure, Flufenisal, Flunoxaprofen, Flurbiprofen, Flurbiprofenaxetil, Furofenac, Furprofen, Glucametacin, Ibufenac, Ibuprofen, Indobufen, Indometacin, Indometacinfarnesil, Indoprofen, Isoxepac, Isoxicam, Ketoprofen, Ketorolac, Lobenzarit, Lonazolac, Lornoxicam, Loxoprofen, Lumiracoxib, Meclofenaminsäure, Meclofen, Mefenaminsäure, Meloxicam, Mesalazin, Miroprofen, Mofezolac, Nabumeton, Naproxen, Nifluminsäure, Olsalazin, Oxaprozin, Oxipinac, Oxyphenbutazon, Parecoxib, Phenylbutazon, Pelubiprofen, Pimeprofen, Pirazolac, Priroxicam, Pirprofen, Pranoprofen, Prifelon, Prinomod, Proglumetacin, Proquazon, Protizininsäure, Rofecoxib, Romazarit, Salicylamid, Salicylsäure, Salmistein, Salnacedin, Salsalat, Sulindac, Sudoxicam, Suprofen, Talniflumat, Tenidap, Tenosal, Tenoxicam, Tepoxalin, Tiaprofensäure, Taramid, Tilnoprofenarbamel, Timegadin, Tinoridin, Tiopinac, Tolfenaminsäure, Tolmetin, Ufenamat, Valdecoxib, Ximoprofen, Zaltoprofen and Zoliprofen.

MRP4-inhibitors used are preferably compounds selected from among N-acetyl-dinitrophenyl-cysteine, cGMP, cholate, diclofenac, dehydroepiandrosterone 3-glucuronide, dehydroepiandrosterone 3-sulphate, dilazep, dinitrophenyl-s-glutathione, estradiol 17-beta-glucuronide, estradiol 3,17-disulphate, estradiol 3-glucuronide, estradiol 3-sulphate, estrone 3-sulphate, flurbiprofen, folate, N5-formyl-tetrahydrofolate, glycocholate, glycolithocholic acid sulphate, ibuprofen, indomethacin, indoprofen, ketoprofen, lithocholic acid sulphate, methotrexate,((E)-3-[[[3-[2-(7-chloro-2-q uinol inyl)ethenyl]phenyl]-[[3-dimethylamino)-3-oxopropyl]thio]methyl]thio]-propanoic acid), alpha-naphthyl-beta-D-glucuronide, nitrobenzyl mercaptopurine riboside, probenecid, sildenafil, sulfinpyrazone, taurochenodeoxycholate, taurocholate, taurodeoxycholate, taurolithocholate, taurolithocholic acid sulphate, topotecan, trequinsin and zaprinast, dipyridamole, optionally in the form of the racemates, enantiomers, diastereomers and the pharmacologically acceptable acid addition salts and hydrates thereof.

Examples of JAK inhibitors preferably include compounds selected from among Tofacitinib and Ruxolitinib.

Examples of immunesuppressive agents preferably include compounds selected from among mycophenolate mofetil, mycophenolic acid, azathioprine, cyclosporine, tacrolimus, pimecrolimus, abetimus, gusperimus and leflunomide.

An example of a cytostaticum is cyclophosphamide.

The invention relates more preferably to the use of MRP4-inhibitors for preparing a pharmaceutical composition for treating respiratory complaints, containing the Syk-inhibitors of formula 1 and MRP4-inhibitors according to the invention, the MRP4-inhibitors preferably being selected from among dehydroepiandrosterone 3-sulphate, estradiol 3,17-disulphate, flurbiprofen, indomethacin, indoprofen, taurocholate, optionally in the form of the racemates, enantiomers, diastereomers and the pharmacologically acceptable acid addition salts and hydrates thereof. The separation of enantiomers from the racemates can be carried out using methods known from the art (e.g. chromatography on chiral phases, etc.).

By acid addition salts with pharmacologically acceptable acids are meant, for example, salts selected from among the hydrochlorides, hydrobromides, hydroiodides, hydrosulphates, hydrophosphates, hydromethanesulphonates, hydronitrates, hydromaleates, hydroacetates, hydrobenzoates, hydronitrates, hydrofumarates, hydrotartrates, hydrooxalates, hydrosuccinates, hydrobenzoates and hydro-p-toluenesulphonates, preferably the hydrochlorides, hydrobromides, hydrosulphates, hydrophosphates, hydrofumarates and hydromethanesulphonates.

The invention further relates to pharmaceutical preparations which contain a triple combination of the Syk-inhibitors of formula 1, MRP4-inhibitors and another active substance according to the invention, such as, for example, an anticholinergic, a PDE4 inhibitor, a steroid, an LTD4-antagonist or a betamimetic, and the preparation thereof and the use thereof for treating respiratory complaints.

Compounds which may be used as iNOS inhibitors are compounds selected from among: S-(2-aminoethyl)isothiourea, aminoguanidine, 2-aminomethylpyridine, 5,6-dihydro-6-methyl-4H-1,3-Thiazine-2-amine(=AMT), L-canavanine, 2-iminopiperidine, S-isopropylisothiourea, S-methylisothiourea, S-ethylisothiourea, S-methyltiocitrullin, S-ethylthiocitrulline, L-NA (Nω-nitro-L-arginine), L-NAME (Nω-nitro-L-argininemethylester), L-NMMA (NG-monomethyl-L-arginine), L-NIO (Nω-iminoethyl-L-ornithine), L-NIL (Nω-iminoethyl-lysine), (S)-6-acetimidoylamino-2-amino-hexanoic acid (1H-tetrazol-5-yl)-amide (SC-51) (J. Med. Chem. 2002, 45, 1686-1689), N-[[3-(aminomethyl)phenyl]methyl]-Ethanimidamide(=1400W), (S)-4-(2-acetimidoylamino-ethylsulphanyl)-2-amino-butyric acid (GW274150) (Bioorg. Med. Chem. Lett. 2000, 10, 597-600), 2-[2-(4-methoxy-pyridin-2-yl)-ethyl]-3H-imidazo[4,5-b]pyridine (BYK191023) (Mol. Pharmacol. 2006, 69, 328-337), 2-((R)-3-amino-1-phenyl-propoxy)-4-chloro-5-fluorobenzonitrile (WO 01/62704), 2-((1R,3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-6-trifluoromethyl-nicotinonitrile (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-4-chloro-benzonitrile (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-5-chloro-benzonitrile (WO 2004/041794), (2S.4R)-2-amino-4-(2-chloro-5-trifluoromethyl-phenylsulphanyl)-4-thiazol-5-yl-butan-1-ol (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-5-chloro-nicotinonitrile (WO 2004/041794), 4-((S)-3-amino-4-hydroxy-1-phenyl-butylsulphanyl)-6-methoxy-nicotinonitrile (WO 02/090332), substituted 3-phenyl-3,4-dihydro-1-isoquinolinamine such as e.g. (1S.5S.6R)-7-chloro-5-methyl-2-aza-bicyclo[4.1.0]hept-2-en-3-ylamine (ONO-1714) (Biochem. Biophys. Res. Commun. 2000, 270, 663-667), (4R,5R)-5-ethyl-4-methyl-thiazolidin-2-ylideneamine (Bioorg. Med. Chem. 2004, 12, 4101), (4R,5R)-5-ethyl-4-methyl-selenazolidin-2-ylideneamine (Bioorg. Med. Chem. Lett. 2005, 15, 1361), 4-aminotetrahydrobiopterine (Curr. Drug Metabol. 2002, 3, 119-121), (E)-3-(4-chloro-phenyl)-N-(1-{2-oxo-2-[4-(6-trifluoromethyl-pyrimidin-4-yloxy)-piperidine-1-yl]ethylcarbamoyl}-2-pyridin-2-yl-ethylyacrylamide (FR260330) (Eur. J. Pharmacol. 2005, 509, 71-76), 3-(2,4-difluoro-phenyl)-6-[2-(4-imidazol-1-ylmethyl-phenoxy)-ethoxy]-2-phenyl-pyridine (PPA250) (J. Pharmaco Exp. Ther. 2002, 303, 52-57), 3-{[(benzo[1,3]dioxol-5-ylmethyl)-carbamoyl]-methyl}-4-(2-imidazol-1-yl-pyrimidin-4-yl)-piperazine-1-carboxylate (BBS-1) (Drugs Future 2004, 29, 45-52), (R)-1-(2-imidazol-1-yl-6-methyl-pyrimidin-4-yl)-pyrrolidine-2-carboxylic acid (2-benzo[1,3]dioxol-5-yl-ethyl)amide (BBS-2) (Drugs Future 2004, 29, 45-52) and the pharmaceutical salts, prodrugs or solvates thereof.

Examples of iNOS-inhibitors within the scope of the present invention may also include antisense oligonucleotides, particularly those antisense oligonucleotides which bind iNOS-coding nucleic acids. For example, WO 01/52902 describes antisense oligonucleotides, particularly antisense oligonucleotides, which bind iNOS coding nucleic acids, for modulating the expression of iNOS. iNOS-antisense oligonucleotides as described particularly in WO 01/52902 may therefore also be combined with the PDE4-inhibitors of the present invention on account of their similar effect to the iNOS-inhibitors.

Suitable HMG-CoA reductase inhibitors (also called statins) which may be preferably used in double or triple combinations with the compounds of formula 1 are selected from among Atorvastatin, Cerivastatin, Flurvastatin, Lovastatin, Pitavastatin, Pravastatin, Rosuvastatin, Simvastatin, optionally in form of their pharmaceutically available acid addition salts, prodrugs, solvates or hydrates thereof.

8. FORMULATIONS

Suitable forms for administration are for example tablets, capsules, solutions, syrups, emulsions or inhalable powders or aerosols. The content of the pharmaceutically effective compound(s) in each case should be in the range from 0.1 to 90 wt. %, preferably 0.5 to 50 wt. % of the total composition, i.e. in amounts which are sufficient to achieve the dosage range specified hereinafter.

The preparations may be administered orally in the form of a tablet, as a powder, as a powder in a capsule (e.g. a hard gelatine capsule), as a solution or suspension. When administered by inhalation the active substance combination may be given as a powder, as an aqueous or aqueous-ethanolic solution or using a propellant gas formulation.

Preferably, therefore, pharmaceutical formulations are characterised by the content of one or more compounds of formula 1 according to the preferred embodiments above.

It is particularly preferable if the compounds of formula 1 are administered orally, and it is also particularly preferable if they are administered once or twice a day. Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate. The tablets may also comprise several layers.

Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arabic, talc, titanium dioxide or sugar. To achieve delayed release or prevent incompatibilities the core may also consist of a number of layers. Similarly the tablet coating may consist of a number of layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.

Syrups containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavour enhancer, e.g. a flavouring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.

Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gelatine capsules. Suitable suppositories may be made for example by mixing with carriers provided for this purpose, such as neutral fats or polyethyleneglycol or the derivatives thereof.

Excipients which may be used include, for example, water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose), emulsifiers (e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone) and lubricants (e.g. magnesium stearate, talc, stearic acid and sodium lauryl sulphate).

For oral administration the tablets may, of course, contain, apart from the abovementioned carriers, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additives such as starch, preferably potato starch, gelatine and the like. Moreover, lubricants such as magnesium stearate, sodium lauryl sulphate and talc may be used at the same time for the tabletting process. In the case of aqueous suspensions the active substances may be combined with various flavour enhancers or colourings in addition to the excipients mentioned above.

It is also preferred if the compounds of formula 1 are administered by inhalation, particularly preferably if they are administered once or twice a day. For this purpose, the compounds of formula 1 have to be made available in forms suitable for inhalation. Inhalable preparations include inhalable powders, propellant-containing metered-dose aerosols or propellant-free inhalable solutions, which are optionally present in admixture with conventional physiologically acceptable excipients.

Within the scope of the present invention, the term propellant-free inhalable solutions also includes concentrates or sterile ready-to-use inhalable solutions. The preparations which may be used according to the invention are described in more detail in the next part of the specification.

Inhalable Powders

If the active substances of formula 1 are present in admixture with physiologically acceptable excipients, the following physiologically acceptable excipients may be used to prepare the inhalable powders according to the invention: monosaccharides (e.g. glucose or arabinose), disaccharides (e.g. lactose, saccharose, maltose), oligo- and polysaccharides (e.g. dextran), polyalcohols (e.g. sorbitol, mannitol, xylitol), salts (e.g. sodium chloride, calcium carbonate) or mixtures of these excipients with one another. Preferably, mono- or disaccharides are used, while the use of lactose or glucose is preferred, particularly, but not exclusively, in the form of their hydrates. For the purposes of the invention, lactose is the particularly preferred excipient, while lactose monohydrate is most particularly preferred. Methods of preparing the inhalable powders according to the invention by grinding and micronising and by finally mixing the components together are known from the prior art.

Propellant-Containing Inhalable Aerosols

The propellant-containing inhalable aerosols which may be used according to the invention may contain the compounds of formula 1 dissolved in the propellant gas or in dispersed form. The propellant gases which may be used to prepare the inhalation aerosols according to the invention are known from the prior art. Suitable propellant gases are selected from among hydrocarbons such as n-propane, n-butane or isobutane and halohydrocarbons such as preferably fluorinated derivatives of methane, ethane, propane, butane, cyclopropane or cyclobutane. The propellant gases mentioned above may be used on their own or in mixtures thereof. Particularly preferred propellant gases are fluorinated alkane derivatives selected from TG134a (1,1,1,2-tetrafluoroethane), TG227 (1,1,1,2,3,3,3-heptafluoropropane) and mixtures thereof. The propellant-driven inhalation aerosols used within the scope of the use according to the invention may also contain other ingredients such as co-solvents, stabilisers, surfactants, antioxidants, lubricants and pH adjusters. All these ingredients are known in the art.

Propellant-Free Inhalable Solutions

The compounds of formula 1 according to the invention are preferably used to prepare propellant-free inhalable solutions and inhalable suspensions. Solvents used for this purpose include aqueous or alcoholic, preferably ethanolic solutions. The solvent may be water on its own or a mixture of water and ethanol. The solutions or suspensions are adjusted to a pH of 2 to 7, preferably 2 to 5, using suitable acids. The pH may be adjusted using acids selected from inorganic or organic acids. Examples of particularly suitable inorganic acids include hydrochloric acid, hydrobromic acid, nitric acid, sulphuric acid and/or phosphoric acid. Examples of particularly suitable organic acids include ascorbic acid, citric acid, malic acid, tartaric acid, maleic acid, succinic acid, fumaric acid, acetic acid, formic acid and/or propionic acid etc. Preferred inorganic acids are hydrochloric and sulphuric acids. It is also possible to use the acids which have already formed an acid addition salt with one of the active substances. Of the organic acids, ascorbic acid, fumaric acid and citric acid are preferred. If desired, mixtures of the above acids may also be used, particularly in the case of acids which have other properties in addition to their acidifying qualities, e.g. as flavourings, antioxidants or complexing agents, such as citric acid or ascorbic acid, for example. According to the invention, it is particularly preferred to use hydrochloric acid to adjust the pH.

Co-solvents and/or other excipients may be added to the propellant-free inhalable solutions used for the purpose according to the invention. Preferred co-solvents are those which contain hydroxyl groups or other polar groups, e.g. alcohols—particularly isopropyl alcohol, glycols—particularly propyleneglycol, polyethyleneglycol, polypropyleneglycol, glycolether, glycerol, polyoxyethylene alcohols and polyoxyethylene fatty acid esters. The terms excipients and additives in this context denote any pharmacologically acceptable substance which is not an active substance but which can be formulated with the active substance or substances in the pharmacologically suitable solvent in order to improve the qualitative properties of the active substance formulation. Preferably, these substances have no pharmacological effect or, in connection with the desired therapy, no appreciable or at least no undesirable pharmacological effect. The excipients and additives include, for example, surfactants such as soya lecithin, oleic acid, sorbitan esters, such as polysorbates, polyvinylpyrrolidone, other stabilisers, complexing agents, antioxidants and/or preservatives which guarantee or prolong the shelf life of the finished pharmaceutical formulation, flavourings, vitamins and/or other additives known in the art. The additives also include pharmacologically acceptable salts such as sodium chloride as isotonic agents. The preferred excipients include antioxidants such as ascorbic acid, for example, provided that it has not already been used to adjust the pH, vitamin A, vitamin E, tocopherols and similar vitamins or provitamins occurring in the human body. Preservatives may be used to protect the formulation from contamination with pathogens. Suitable preservatives are those which are known in the art, particularly cetyl pyridinium chloride, benzalkonium chloride or benzoic acid or benzoates such as sodium benzoate in the concentration known from the prior art.

For the treatment forms described above, ready-to-use packs of a medicament for the treatment of respiratory complaints are provided, containing an enclosed description including for example the words respiratory disease, COPD or asthma, together with a imidazolyl-pyrimidine according to formula 1 and one or more combination partners selected from those described above.

Claims

1. Compound of formula 1 or 1′

wherein
A is selected from the group consisting of N and CH
D is selected from the group consisting of CH, N, NH,
E is C
T is selected from the group consisting of C and N,
G is selected from the group consisting of C and N,
and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system, and wherein
M is selected from the group consisting of —CH2—, —O—, —NH— and —N(C1-4-alkyl)-;
R3 is selected from the group consisting of methyl and ethyl;
Het is selected from the group consisting of
a five- to six-membered monocyclic heterocycle with 1, 2, 3 or 4 heteroatoms each
independently from one another selected from N, S and O,
and a nine- to eleven-membered bicyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;
Hetaryl is selected from the group consisting of
a five- to six-membered monocyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;
and a nine- to eleven-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and O;
and wherein
R1 is selected from the group consisting of
Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, —OH, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2, —NH(CH3), —N(CH3)2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, —OH, halogen and C1-3-alkyl,
and the pharmaceutically acceptable salts of the aforementioned compounds.

2. (canceled)

3. The compound of formula 1 according to claim 1 or of formula 1′ of claim 1, wherein

M is —CH2—
and wherein
R3 is methyl,
and wherein
R1 is selected from the group consisting of
Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

4. The compound of formula 1d according to claim 1

or of formula 1d′ according to claim 1,
wherein
A is selected from the group consisting of N and CH,
R1 is selected from the group consisting of
Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

5. The compound of formula 1d or of formula 1d′ according to claim 4,

wherein
A is CH and wherein
R1 is either a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S, or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, 1'(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

6. The compound of formula 1d or of formula 1d′ according to claim 4, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl, three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

7. The compound of formula 1d or of formula 1d′ according to claim 4, which is

and the pharmaceutically acceptable salts of the aforementioned compounds.

8. The compound of formula 1e according to claim 1,

or of formula 1e′ according to claim 1,
wherein
A is selected from the group consisting of N and CH
R1 is selected from the group consisting of
Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.)

9. The compound of formula 1e or of formula 1e′ according to claim 8,

wherein
A is CH and wherein
R1 is a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S, or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

10. The compound of formula 1e or of formula 1e′ according to claim 8, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl, three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

11. The compound of formula 1e or of formula 1e′ according to claim 8, which is

and the pharmaceutically acceptable salts of the aforementioned compound.

12. The compound of formula 1f according to claim 1,

or of formula 1f′0 according to claim 1,
wherein
R1 is selected from the group consisting of
Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

13. The compound of formula 1f or of formula 1f′ according to claim 12, wherein

R1 is either a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S, or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

14. The compound of formula 1f or of formula 1f′ according to claim 12, wherein

R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O-methyl, —O-ethyl, O-propyl, O-butyl, —C1-3-haloalkyl, three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl, and the pharmaceutically acceptable salts of the aforementioned compounds.

15. The compound of formula 1 according to formula 1f or of 1f′ of claim 12 which is

and the pharmaceutically acceptable salts thereof.

16. An intermediate compound selected from the group consisting of formula 6

of formula 7
of formula 8
and of formula 11
wherein E, D, G, T and R1 are defined as in claim 1 and wherein Hal is Cl or Br and wherein PG is a protecting group selected from the group consisting of benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl.

17. An intermediate compound selected from the group consisting of formula 10.3

18. A method for the treatment of a disease which can be treated by inhibition of the SYK enzyme comprising administering a therapeutically effective amount of a compound of claim 1 to a patient in need thereof.

19. The method of claim 18 for the treatment of a disease selected from the group consisting of allergic rhinitis, asthma, COPD, adult respiratory distress syndrome, bronchitis, B-cell lymphoma, dermatitis and contact dermatitis, allergic dermatitis, allergic rhinoconjunctivitis, rheumatoid arthritis, anti-phospholipid syndrome, Berger's disease, Evans's syndrome, ulcerative colitis, allergic antibody-based glomerulonephritis, granulocytopenia, Goodpasture's syndrome, hepatitis, Henoch-Schönlein purpura, hypersensitivity vasculitis, immunohaemolytic anaemia, autoimmune haemolytic anemia, idiopathic thrombocytopenic purpura, Kawasaki syndrome, allergic conjunctivitis, lupus erythematodes, lupus nephritis, capsule cell lymphoma, neutropenia, non-familial lateral sclerosis, artheriosclerosis, Crohn's disease, multiple sclerosis, myasthenia gravis, osteoporosis, osteolytic diseases, osteopenia, psoriasis, Sjögren's syndrome, sclerodermy, T-cell lymphoma, urticaria/angiooedema, Wegener's granulomatosis and coeliac disease.

20. The method according to claim 19 wherein the disease is selected from the group consisting of asthma, COPD, allergic rhinitis, adult respiratory distress syndrome, bronchitis, allergic dermatitis, contact dermatitis, idiopathic thrombocytopenic purpura, rheumatoid arthritis, lupus erythematodes, lupus nephritis and allergic rhinoconjunctivitis.

21. The method according to claim 20 wherein the disease is selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.

22. Pharmaceutical formulations, characterised in that they contain one or more compounds according to claim 1 and a pharmaceutically acceptable excipient.

23. Pharmaceutical formulations, characterised in that they contain one or more compounds according to claim 1 in combination with an active substance selected from the group consisting of anticholinergics, betamimetics, corticosteroids, PDE4-inhibitors, EGFR-inhibitors, LTD4-antagonists, CCR3-inhibitors, iNOS-inhibitors, CRTH2-antagonists, HMG-CoA reductase inhibitors and NSAIDs.

Patent History
Publication number: 20170008896
Type: Application
Filed: Mar 12, 2015
Publication Date: Jan 12, 2017
Applicant: Boehringer Ingelheim International GmbH (Ingelheim am Rhein)
Inventors: Georg DAHMANN (Biberach an der Riss), Matthias HOFFMANN (Mittelbiberach), Jasna KLICIC (Geneva), David James LAMB (Mittelbiberach), Clive McCARTHY (Wantage), Spencer NAPIER (Abingdon), Karen PARRISH (Abingdon), John SCOTT (Abingdon), Jennifer L. Swantek FITZGERALD (Ridgefield, CT), Edward WALKER (Didcot)
Application Number: 15/125,599
Classifications
International Classification: C07D 487/04 (20060101); C07D 471/04 (20060101); A61K 31/437 (20060101); A61K 31/4184 (20060101); A61K 31/519 (20060101); C07D 403/14 (20060101); A61K 45/06 (20060101);