3-HETARYL-SUBSTITUTED PYRROLO[2,3 B]PYRIDINE DERIVATIVES AS PDK1 INHIBITORS

Abstract
Compounds of the formula I in which Q, R1, R2, R3 and R4 have the meanings indicated in Claim 1, are PDK1 inhibitors and can be employed for the treatment of tumours.
Description

The invention relates to compounds of the formula I




embedded image


  • in which

  • Q denotes Het-diyl,

  • R1 denotes Br, Het1, or phenyl which is monosubstituted by CH2OH,

  • R2, R3 each, independently of one another, denote H, Hal, A, Ar, [C(R5)2]n0H, [C(R5)2]nOA or [C(R5)2]nN(R5)2,

  • R2, R3 together also denote ═O, ═CH2 or an alkylene chain having 2-5 C atoms,

  • R4 denotes H, Ar or Het2,

  • R5 denotes H or A′,

  • Het1 denotes pyrazolyl, which may be monosubstituted by A, CH2OH, (CH2)2OH, COOH, COOA, CH2COHet3 or CONH2,

  • Ar denotes phenyl, naphthyl or biphenyl, each of which is unsubstituted or mono-, di- or trisubstituted by Hal, A, (CH2)nOR5, (CH2)nN(R5)2, SR5, NO2, COOR5, CON(R5)2, NR5COA, NR5SO2A, SO2N(R5)2, COR5, (CH2)nCN and/or S(O)mA,

  • Het denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by Hal, A, [C(R5)2]nOR5, [C(R5)2]nN(R5)2, NO2, CN, COOR5, CON(R5)2, NR5COA, NR5SO2A, COR5, SO2NR5, S(O)mA, ═S, ═NH, ═NA and or ═O (carbonyl oxygen),

  • Het2 denotes a monocyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by Hal, A, OR5, N(R5)2, NO2, CN, COOR5, CON(R5)2, NR5COA, NR5SO2A, COR5, SO2NR5, S(O)mA, ═S, ═NH, ═NA and or ═O (carbonyl oxygen),

  • Het3 denotes an unsubstituted monocyclic saturated heterocycle having 1 to 4 N, and/or O and/or S atoms,

  • A denotes unbranched or branched alkyl having 1-10 C atoms, in which 1-7H atoms may be replaced by F, Cl and/or Br, and/or in which one or two non-adjacent CH and/or CH2 groups may be replaced by NR5, O, S, SO, SO2, CC and/or CH═CH groups,
    • or
    • cyclic alkyl having 3-7 C atoms,

  • A′ denotes unbranched or branched alkyl having 1-6 C atoms,
    • or
    • cyclic alkyl having 3-7 C atoms,

  • Hal denotes F, Cl, Br or I,

  • m denotes 0, 1 or 2,

  • n denotes 0, 1, 2, 3 or 4,


    and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.



The invention was based on the object of finding novel compounds having valuable properties, in particular those which can be used for the preparation of medicaments.


It has been found that the compounds of the formula I and salts, tautomers and stereoisomers thereof have very valuable pharmacological properties while being well tolerated.


In particular, they exhibit a cell proliferation/cell vitality-inhibiting action as antagonists or agonists. The compounds according to the invention can therefore be used for the combating and/or treatment of tumours, tumour growth and/or tumour metastases.


The antiproliferative action can be tested in a proliferation assay/vitality assay.


Pyrimidinyl-2-amine derivatives are described in WO 2008/155000 Other 4-(pyrrolopyridinyl)pyrimidinyl-2-amine derivatives are also described by P. M. Fresneda et al. in Tetrahedron 57 (2001) 2355-2363.


Other 4-(pyrrolopyridinyl)pyrimidinyl-2-amine derivatives are also described by A. Karpov in his dissertation, University of Heidelberg, April 2005.


Other aminopyridine derivatives which carry a 2,2,6,6-tetramethylpiperidin-4-yl radical are described in WO 2004/089913 for the treatment of inflammatory and autoimmune diseases.


Accordingly, the compounds according to the invention or a pharmaceutically acceptable salt thereof are administered for the treatment of cancer, including solid carcinomas, such as, for example, carcinomas (for example of the lungs, pancreas, thyroid, bladder or colon), myeloid diseases (for example myeloid leukaemia) or adenomas (for example villous colon adenoma).


The tumours furthermore include monocytic leukaemia, brain, urogenital, lymphatic system, stomach, laryngeal and lung carcinoma, including lung adenocarcinoma and small-cell lung carcinoma, pancreatic and/or breast carcinoma.


The compounds are furthermore suitable for the treatment of immune deficiency induced by HIV-1 (Human Immunodeficiency Virus Type 1).


Cancer-like hyperproliferative diseases are to be regarded as brain cancer, lung cancer, squamous epithelial cancer, bladder cancer, stomach cancer, pancreatic cancer, liver cancer, renal cancer, colorectal cancer, breast cancer, head cancer, neck cancer, oesophageal cancer, gynaecological cancer, thyroid cancer, lymphomas, chronic leukaemia and acute leukaemia. In particular, cancer-like cell growth is a disease which represents a target of the present invention. The present invention therefore relates to compounds according to the invention as medicaments and/or medicament active compounds in the treatment and/or prophylaxis of the said diseases and to the use of compounds according to the invention for the preparation of a pharmaceutical for the treatment and/or prophylaxis of the said diseases and to a process for the treatment of the said diseases comprising the administration of one or more compounds according to the invention to a patient in need of such an administration.


It can be shown that the compounds according to the invention have an antiproliferative action. The compounds according to the invention are administered to a patient having a hyperproliferative disease, for example to inhibit tumour growth, to reduce inflammation associated with a lymphoproliferative disease, to inhibit transplant rejection or neurological damage due to tissue repair, etc. The present compounds are suitable for prophylactic or therapeutic purposes. As used herein, the term “treatment” is used to refer to both the prevention of diseases and the treatment of pre-existing conditions. The prevention of proliferation/vitality is achieved by administration of the compounds according to the invention prior to the development of overt disease, for example for preventing tumour growth. Alternatively, the compounds are used for the treatment of ongoing diseases by stabilising or improving the clinical symptoms of the patient.


The host or patient can belong to any mammalian species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cows, dogs, cats, etc. Animal models are of interest for experimental investigations, providing a model for treatment of a human disease.


The susceptibility of a particular cell to treatment with the compounds according to the invention can be determined by in vitro testing. Typically, a culture of the cell is incubated with a compound according to the invention at various concentrations for a period of time which is sufficient to allow the active agents to induce cell death or to inhibit cell proliferation, cell vitality or migration, usually between about one hour and one week. In vitro testing can be carried out using cultivated cells from a biopsy sample. The amount of cells remaining after the treatment are then determined.


The dose varies depending on the specific compound used, the specific disease, the patient status, etc. A therapeutic dose is typically sufficient considerably to reduce the undesired cell population in the target tissue, while the viability of the patient is maintained. The treatment is generally continued until a considerable reduction has occurred, for example an at least about 50% reduction in the cell burden, and may be continued until essentially no more undesired cells are detected in the body.


There are many diseases associated with deregulation of cell proliferation and cell death (apoptosis). The conditions of interest include, but are not limited to, the following. The compounds according to the invention are suitable for the treatment of various conditions where there is proliferation and/or migration of smooth muscle cells and/or inflammatory cells into the intimal layer of a vessel, resulting in restricted blood flow through that vessel, for example in the case of neointimal occlusive lesions. Occlusive graft vascular diseases of interest include atherosclerosis, coronary vascular disease after grafting, vein graft stenosis, perianastomatic prosthetic restenosis, restenosis after angioplasty or stent placement, and the like.


The compounds of the formula I, also act as regulators, modulators or inhibitors of protein kinases, in particular of the serine/threonine kinase type, which include, inter alia, phosphoinositide-dependent kinase 1 (PDK 1). The compounds according to the invention exhibit a certain action in the inhibition of serine/threonine kinase PDK1.


PDK1 phosphorylates and activates a sub-group of the AGC protein kinase family, comprising PKB, SGK, S6K and PKC isoforms. These kinases are involved in the PI3K signal transduction pathway and control basic cellular functions, such as survival, growth and differentiation. PDK1 is thus an important regulator of diverse metabolic, proliferative and life-sustaining effects.


Diseases caused by protein kinases are characterised by anomalous activity or hyperactivity of such protein kinases. Anomalous activity relates either to: (1) the expression in cells which do not usually express these protein kinases; (2) increased kinase expression which results in undesired cell proliferation, such as cancer; (3) increased kinase activity which results in undesired cell proliferation, such as cancer, and/or in hyperactivity of the corresponding protein kinases. Hyperactivity relates either to amplification of the gene which encodes a certain protein kinase or the generation of an activity level which can be correlated with a cell proliferation disease (i.e. the severity of one or more symptoms of the cell proliferation disease increases with increasing kinase level) the bioavailability of a protein kinase can also be influenced by the presence or absence of a set of binding proteins of this kinase.


The most important types of cancer that can be treated using a compound according to the invention include colorectal cancer, small-cell lung cancer, nonsmall-cell lung cancer, multiple myeloma as well as renal cell carcinoma and endometrium carcinoma, particularly also types of cancer in which PTEN is mutated, inter alia breast cancer, prostate cancer and glioblastoma.


In addition, the compounds according to the invention can be used to achieve additive or synergistic effects in certain existing cancer chemotherapies and radiotherapies and/or to restore the efficacy of certain existing cancer chemotherapies and radiotherapies.


The invention also relates to the optically active forms (stereoisomers), salts, the enantiomers, the racemates, the diastereomers and the hydrates and solvates of these compounds. Solvates of the compounds are taken to mean adductions of inert solvent molecules onto the compounds which form owing to their mutual attractive force. Solvates are, for example, mono- or dihydrates or alcoholates. Pharmaceutically usable derivatives are taken to mean, for example, the salts of the compounds according to the invention and also so-called prodrug compounds. The invention naturally also encompasses the solvates of the salts of the compounds according to the invention.


Prodrug derivatives are taken to mean compounds of the formula I which have been modified by means of, for example, alkyl or acyl groups, sugars or oligopeptides and which are rapidly cleaved in the organism to form the effective compounds according to the invention.


These also include biodegradable polymer derivatives of the compounds according to the invention, as described, for example, in Int. J. Pharm. 115, 61-67 (1995).


The expression “effective amount” denotes the amount of a medicament or of a pharmaceutical active compound which causes in a tissue, system, animal or human a biological or medical response which is sought or desired, for example, by a researcher or physician.


In addition, the expression “therapeutically effective amount” denotes an amount which, compared with a corresponding subject who has not received this amount, has the following consequence:


improved treatment, healing, prevention or elimination of a disease, syndrome, condition, complaint, disorder or side effects or also the reduction in the advance of a disease, condition or disorder.


The expression “therapeutically effective amount” also encompasses the amounts which are effective for increasing normal physiological function.


The invention also relates to the use of mixtures of the compounds of the formula I, for example mixtures of two diastereomers, for example in the ratio 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:100 or 1:1000.


These are particularly preferably mixtures of stereoisomeric compounds.


Above and below, the radicals R1, R2, R3, R4 and Q have the meanings indicated for the formula I, unless expressly indicated otherwise.


A denotes alkyl, is unbranched (linear) or branched, and has 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 C atoms. A preferably denotes methyl, furthermore ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl or tert-butyl, furthermore also pentyl, 1-, 2- or 3-methylbutyl, 1,1-, 1,2- or 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-, 2-, 3- or 4-methylpentyl, 1,1-, 1,2-, 1,3-, 2,2-, 2,3- or 3,3-dimethylbutyl, 1- or 2-ethylbutyl, 1-ethyl-1-methylpropyl, 1-ethyl-2-methylpropyl, 1,1,2- or 1,2,2-trimethylpropyl, further preferably, for example, trifluoromethyl.


A very particularly preferably denotes alkyl having 1, 2, 3, 4, 5 or 6 C atoms, preferably methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl, trifluoromethyl, pentafluoroethyl or 1,1,1-trifluoroethyl, in which, in addition, one or two CH and/or CH2 groups may preferably be replaced by 0 and/or NR3. A therefore also denotes, for example, CH2OCH3 or CH2OCH2CH2NH2.


A furthermore preferably denotes unbranched or branched alkyl having 1-6 C atoms, in which 1-5H atoms may be replaced by F, or cyclic alkyl having 3-7 C atoms.


A′ preferably denotes alkyl having 1, 2, 3, 4, 5 or 6 C atoms, preferably methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl or hexyl, furthermore cyclopentyl or cyclohexyl.


Cyclic alkyl denotes cyclopropyl, cyclobutyl, cyclopentyl, cyclhexyl or cycloheptyl.


Hal preferably denotes F, Cl or Br, but also I, particularly preferably F or Cl.


R1 preferably denotes Het1.


R2, R3 preferably, in each case independently of one another, denote H, Hal, A, Ar, OH or OA.


R2 particularly preferably denotes H, A or Ar.


R3 particularly preferably denotes H, Hal, OH or OA.


R5 preferably denotes H or methyl.


Het1 preferably denotes pyrazolyl, which may be monosubstituted by A, CH2COHet3 or COOA.


Het1 particularly preferably denotes pyrazolyl which is monosubstituted by A.


Ar denotes, for example, o-, m- or p-tolyl, o-, m- or p-ethylphenyl, o-, m- or p-propylphenyl, o-, m- or p-isopropylphenyl, o-, m- or p-tert-butylphenyl, o-, m- or p-hydroxyphenyl, o-, m- or p-nitrophenyl, o-, m- or p-aminophenyl, o-, m- or p-(nmethylamino)phenyl, o-, m- or p-(n-methylaminocarbonyl)-phenyl, o-, m- or p-acetamidophenyl, o-, m- or p-methoxyphenyl, o-, m- or p-ethoxyphenyl, o-, m- or p-ethoxycarbonylphenyl, o-, m- or p-(N,N-dimethylamino)phenyl, o-, m- or p-(N,N-dimethylaminocarbonyl)phenyl, o-, m- or p-(N-ethylamino)phenyl, o-, m- or p-(N,N-diethylamino)phenyl, o-, m- or p-fluorophenyl, o-, m- or p-bromophenyl, o-, m- or p-chlorophenyl, o-, m- or p-(methylsulfonamido)phenyl, o-, m- or p-(methylsulfonyl)phenyl, o-, m- or p-cyanophenyl, o-, m- or p-carboxyphenyl, o-, m- or p-methoxycarbonylphenyl, o-, m- or p-formylphenyl, o-, m- or p-acetylphenyl, o-, m- or p-aminosulfonylphenyl, furthermore preferably 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-difluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-dichlorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-dibromophenyl, 2,4- or 2,5-dinitrophenyl, 2,5- or 3,4-dimethoxyphenyl, 3-nitro-4-chlorophenyl, 3-amino-4-chloro-, 2-amino-3-chloro-, 2-amino-4-chloro-, 2-amino-5-chloro- or 2-amino-6-chlorophenyl, 2-nitro-4-N,N-dimethylamino- or 3-nitro-4-N,N-dimethylaminophenyl, 2,3-diaminophenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trichlorophenyl, 2,4,6-trimethoxyphenyl, 2-hydroxy-3,5-dichlorphenyl, p-iodophenyl, 3,6-dichloro-4-aminophenyl, 4-fluoro-3-chlorophenyl, 2-fluoro-4-bromophenyl, 2,5-difluoro-4-bromophenyl, 3-bromo-6-methoxyphenyl, 3-chloro-6-methoxyphenyl, 3-chloro-4-acetamidophenyl, 3-fluoro-4-methoxyphenyl, 3-amino-6-methylphenyl, 3-chloro-4-acetamidophenyl or 2,5-dimethyl-4-chlorophenyl.


In a further embodiment, Ar preferably denotes phenyl which is unsubstituted or mono- or disubstituted by Hal, COOR5 and/or CON(R5)2.


Het denotes, irrespective of further substitutions, for example, 2- or 3-furyl, 2- or 3-thienyl, 1-, 2- or 3-pyrrolyl, 1-, 2,4- or 5-imidazolyl, 1-, 3-, 4- or 5-pyrazolyl, 2-, 4- or 5-oxazolyl, 3-, 4- or 5-isoxazolyl, 2-, 4- or 5-thiazolyl, 3-, 4- or 5-isothiazolyl, 2-, 3- or 4-pyridyl, 2-, 4-, 5- or 6-pyrimidinyl, furthermore preferably 1,2,3-triazol-1-, -4- or -5-yl, 1,2,4-triazol-1-, -3- or 5-yl, 1- or 5-tetrazolyl, 1,2,3-oxadiazol-4- or -5-yl, 1,2,4-oxadiazol-3- or -5-yl, 1,3,4-thiadiazol-2- or -5-yl, 1,2,4-thiadiazol-3- or -5-yl, 1,2,3-thiadiazol-4- or -5-yl, 3- or 4-pyridazinyl, pyrazinyl, 1-, 2-, 3-, 4-, 5-, 6- or 7-indolyl, 4- or 5-isoindolyl, indazolyl, 1-, 2-, 4- or 5-benzimidazolyl, 1-, 3-, 4-, 5-, 6- or 7-benzopyrazolyl, 2-, 4-, 5-, 6- or 7-benzoxazolyl, 3-, 4-, 5-, 6- or 7-benzisoxazolyl, 2-, 4-, 5-, 6- or 7-benzothiazolyl, 2-, 4-, 5-, 6- or 7-benzisothiazolyl, 4-, 5-, 6- or 7-benz-2,1,3-oxadiazolyl, 2-, 3-, 4-, 5-, 6-, 7- or 8-quinolyl, 1-, 3-, 4-, 5-, 6-, 7- or 8-isoquinolyl, 3-, 4-, 5-, 6-, 7- or 8-cinnolinyl, 2-, 4-, 5-, 6-, 7- or 8-quinazolinyl, 5- or 6-quinoxalinyl, 2-, 3-, 5-, 6-, 7- or 8-2H-benzo-1,4-oxazinyl, further preferably 1,3-benzodioxol-5-yl, 1,4-benzodioxan-6-yl, 2,1,3-benzothiadiazol-4- or -5-yl, 2,1,3-benzoxadiazol-5-yl or dibenzofuranyl. The heterocyclic radicals may also be partially or fully hydrogenated. Irrespective of further substitutions, Het can thus also denote, for example, 2,3-dihydro 2,3,4 or -5-furyl, 2,5-dihydro 2,3,4 or 5-furyl, tetrahydro-2- or -3-furyl, 1,3-dioxolan-4-yl, tetrahydro-2- or -3-thienyl, 2,3-dihydro 1,2,3,4 or -5-pyrrolyl, 2,5-dihydro 1,2,3,4 or -5-pyrrolyl, 1-, 2- or 3-pyrrolidinyl, tetrahydro-1-, -2- or -4-imidazolyl, 2,3-dihydro 1,2,3,4 or -5-pyrazolyl, tetrahydro-1-, -3- or -4-pyrazolyl, 1,4-dihydro 1,2,3 or -4-pyridyl, 1,2,3,4-tetrahydro 1,2,3,4,5 or -6-pyridyl, 1-, 2-, 3- or 4-piperidinyl, 2-, 3- or 4-morpholinyl, tetrahydro-2-, -3- or -4-pyranyl, 1,4-dioxanyl, 1,3-dioxan-2-, -4- or -5-yl, hexahydro-1-, -3- or -4-pyridazinyl, hexahydro 1,2,4 or -5-pyrimidinyl, 1-, 2- or 3-piperazinyl, 1,2,3,4-tetrahydro 1,2,3,4,5,6,7 or -8-quinolyl, 1,2,3,4-tetrahydro-1-,-2,3,4,5,6,7 or -8-isoquinolyl, 2-, 3-, 5-, 6-, 7- or 8-3,4-dihydro-2H-benzo-1,4-oxazinyl, further preferably 2,3-methylenedioxyphenyl, 3,4-methylenedioxyphenyl, 2,3-ethylenedioxyphenyl, 3,4-ethylenedioxyphenyl, 3,4-(difluoromethylenedioxy)phenyl, 2,3-dihydrobenzofuran-5- or 6-yl, 2,3-(2-oxomethylenedioxy)phenyl or also 3,4-dihydro-2H-1,5-benzodioxepin-6- or -7-yl, furthermore preferably 2,3-dihydrobenzofuranyl, 2,3-dihydro-2-oxofuranyl, 3,4-dihydro-2-oxo-1H-quinazolinyl, 2,3-dihydrobenzoxazolyl, 2-oxo-2,3-dihydrobenzoxazolyl, 2,3-dihydrobenzimidazolyl, 1,3-dihydroindol, 2-oxo-1,3-dihydroindole or 2-oxo-2,3-dihydrobenzimidazolyl.


In a further embodiment, Het preferably denotes a monocyclic aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A and/or [C(R5)2]nOR5.


Het very particularly preferably denotes thiazolyl, thiophenyl, furanyl, pyrrolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazolyl, imidazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, pyridinyl or pyrimidinyl,


where the heterocycles may also be mono- or disubstituted by A and/or [C(R5)2]nOR5.


Het2 denotes, irrespective of further substitutions, for example, 2- or 3-furyl, 2- or 3-thienyl, 1-, 2- or 3-pyrrolyl, 1-, 2,4- or 5-imidazolyl, 1-, 3-, 4- or 5-pyrazolyl, 2-, 4- or 5-oxazolyl, 3-, 4- or 5-isoxazolyl, 2-, 4- or 5-thiazolyl, 3-, 4- or 5-isothiazolyl, 2-, 3- or 4-pyridyl, 2-, 4-, 5- or 6-pyrimidinyl, furthermore preferably 1,2,3-triazol-1-, -4- or -5-yl, 1,2,4-triazol-1-, -3- or 5-yl, 1- or 5-tetrazolyl, 1,2,3-oxadiazol-4- or -5-yl, 1,2,4-oxadiazol-3- or -5-yl, 1,3,4-thiadiazol-2- or -5-yl, 1,2,4-thiadiazol-3- or -5-yl, 1,2,3-thiadiazol-4- or -5-yl, 3- or 4-pyridazinyl, pyrazinyl, 1-, 2-, 3-, 4-, 5-, 6- or 7-indolyl, 4- or 5-isoindolyl, indazolyl, 1-, 2-, 4- or 5-benzimidazolyl, 1-, 3-, 4-, 5-, 6- or 7-benzopyrazolyl, 2-, 4-, 5-, 6- or 7-benzoxazolyl, 3-, 4-, 5-, 6- or 7-benzisoxazolyl, 2-, 4-, 5-, 6- or 7-benzothiazolyl, 2-, 4-, 5-, 6- or 7-benzisothiazolyl, 4-, 5-, 6- or 7-benz-2,1,3-oxadiazolyl, 2-, 3-, 4-, 5-, 6-, 7- or 8-quinolyl, 1-, 3-, 4-, 5-, 6-, 7- or 8-isoquinolyl, 3-, 4-, 5-, 6-, 7- or 8-cinnolinyl, 2-, 4-, 5-, 6-, 7- or 8-quinazolinyl, 5- or 6-quinoxalinyl, 2-, 3-, 5-, 6-, 7- or 8-2H-benzo-1,4-oxazinyl, further preferably 1,3-benzodioxol-5-yl, 1,4-benzodioxan-6-yl, 2,1,3-benzothiadiazol-4- or -5-yl, 2,1,3-benzoxadiazol-5-yl or dibenzofuranyl. The heterocyclic radicals may also be partially or fully hydrogenated. Irrespective of further substitutions, Het2 can thus also denote, for example, 2,3-dihydro 2,3,4 or -5-furyl, 2,5-dihydro 2,3,4 or 5-furyl, tetrahydro-2- or -3-furyl, 1,3-dioxolan-4-yl, tetrahydro-2- or -3-thienyl, 2,3-dihydro 1,2,3,4 or -5-pyrrolyl, 2,5-dihydro 1,2,3,4 or -5-pyrrolyl, 1-, 2- or 3-pyrrolidinyl, tetrahydro-1-, -2- or -4-imidazolyl, 2,3-dihydro 1,2,3,4 or -5-pyrazolyl, tetrahydro-1-, -3- or -4-pyrazolyl, 1,4-dihydro 1,2,3 or -4-pyridyl, 1,2,3,4-tetrahydro 1,2,3,4,5 or -6-pyridyl, 1-, 2-, 3- or 4-piperidinyl, 2-, 3- or 4-morpholinyl, tetrahydro-2-, -3- or -4-pyranyl, 1,4-dioxanyl, 1,3-dioxan-2-, -4- or -5-yl, hexahydro-1-, -3- or -4-pyridazinyl, hexahydro 1,2,4 or -5-pyrimidinyl, 1-, 2- or 3-piperazinyl, 1,2,3,4-tetrahydro 1,2,3,4,5,6,7 or -8-quinolyl, 1,2,3,4-tetrahydro-1-,-2,3,4,5,6,7 or -8-isoquinolyl, 2-, 3-, 5-, 6-, 7- or 8-3,4-dihydro-2H-benzo-1,4-oxazinyl, further preferably 2,3-methylenedioxyphenyl, 3,4-methylenedioxyphenyl, 2,3-ethylenedioxyphenyl, 3,4-ethylenedioxyphenyl, 3,4-(difluoromethylenedioxy)phenyl, 2,3-dihydrobenzofuran-5- or 6-yl, 2,3-(2-oxomethylenedioxy)phenyl or also 3,4-dihydro-2H-1,5-benzodioxepin-6- or -7-yl, furthermore preferably 2,3-dihydrobenzofuranyl, 2,3-dihydro-2-oxofuranyl, 3,4-dihydro-2-oxo-1H-quinazolinyl, 2,3-dihydrobenzoxazolyl, 2-oxo-2,3-dihydrobenzoxazolyl, 2,3-dihydrobenzimidazolyl, 1,3-dihydroindol, 2-oxo-1,3-dihydroindole or 2-oxo-2,3-dihydrobenzimidazolyl.


In a further embodiment, Het2 preferably denotes a monocyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A, OR5 and/or ═O. Het2 very particularly preferably denotes thiazolyl, thiophenyl, furanyl, pyrrolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazolyl, imidazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, pyridinyl, pyrimidinyl, piperidinyl, pyrrolidinyl, morpholinyl, piperazinyl, imidazolidinyl, oxazolidinyl or tetrahydropyranyl, where the heterocycles may also be mono- or disubstituted by A, OR5 and/or ═O.


Het3 preferably denotes piperidinyl, pyrrolidinyl, morpholinyl, piperazinyl, imidazolidinyl, oxazolidinyl or tetrahydropyranyl.


Throughout the invention, all radicals which occur more than once may be identical or different, i.e. are independent of one another.


The compounds of the formula I may have one or more chiral centres and can therefore occur in various stereoisomeric forms. The formula I encompasses all these forms.


Accordingly, the invention relates, in particular, to the compounds of the formula I in which at least one of the said radicals has one of the preferred meanings indicated above. Some preferred groups of compounds may be expressed by the following sub-formulae Ia to Ig, which conform to the formula I and in which the radicals not designated in greater detail have the meaning indicated for the formula I, but in which

  • in Ia R1 denotes Het1;
  • in Ib Het1 denotes pyrazolyl, which may be monosubstituted by A, CH2COHet3 or COOA;
  • in Ic Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, COOR5 and/or CON(R5)2;
  • in Id Het denotes a monocyclic aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A and/or [C(R5)2]nOR5;
  • in Ie Het denotes thiazolyl, thiophenyl, furanyl, pyrrolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazolyl, imidazolyl, triazolyl, thiadiazolyl, pyridazinyl, pyrazinyl, pyridinyl or pyrimidinyl, where the heterocycles may also be mono- or disubstituted by A and/or [C(R5)2]nOR5;
  • in If A denotes unbranched or branched alkyl having 1-6 C atoms, in which 1-5H atoms may be replaced by F,
    • or
    • cyclic alkyl having 3-7 C atoms;
  • in Ig Q denotes Het-diyl,
    • R1 denotes Het1,
    • R2, R3 each, independently of one another, denote H, Hal, A, Ar, [C(R5)2]10H, [C(R5)2]nOA or [C(R5)2]1N(R5)2,
    • R2, R3 together also denote ═O, ═CH2 or an alkylene chain having 2-5 C atoms,
    • R4 denotes H, Ar or Het2,
    • R5 denotes H or A′,
    • Het1 denotes pyrazolyl, which may be monosubstituted by A, CH2COHet3 or COOA,
    • Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, COOR5 and/or CON(R5)2,
    • Het denotes a monocyclic aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A and/or [C(R5)2]nO, R5,
    • Het2 denotes a monocyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A, OR5 and/or ═O,
    • Het3 denotes an unsubstituted monocyclic saturated heterocycle having 1 to 4 N, and/or O and/or S atoms,
    • A denotes unbranched or branched alkyl having 1-6 C atoms, in which 1-5H atoms may be replaced by F,
      • or
      • cyclic alkyl having 3-7 C atoms,
    • A′ denotes unbranched or branched alkyl having 1-6 C atoms,
      • or
      • cyclic alkyl having 3-7 C atoms,
    • Hal denotes F, Cl, Br or I,
    • m denotes 0, 1 or 2,
    • n denotes 0, 1, 2, 3 or 4;


      and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.


The compounds of the formula I and also the starting materials for their preparation are, in addition, prepared by methods known per se, as described in the literature (for example in the standard works, such as Houben-Weyl, Methoden der organischen Chemie [Methods of Organic Chemistry], GeorgThieme-Verlag, Stuttgart), to be precise under reaction conditions which are known and suitable for the said reactions. Use can also be made here of variants known per se which are not mentioned here in greater detail.


Compounds of the formula I can preferably be obtained by reacting compounds of the formula II and with a guanidine salt, such as, for example, guanidinium carbonate.


The compounds of the formula II are generally known. If they are novel, however, they can be prepared by methods known per se.


The reaction is carried out in an inert solvent and is generally carried out in the presence of an acid-binding agent, preferably an organic base, such as DIPEA, triethylamine, dimethylaniline, pyridine or quinoline. The addition of an alkali or alkaline-earth metal hydroxide, carbonate or bicarbonate or another salt of a weak acid of the alkali or alkaline-earth metals, preferably of potassium, sodium, calcium or caesium, may also be favourable.


Depending on the conditions used, the reaction time is between a few minutes and 14 days, the reaction temperature is between about −15° C. and 150° C., normally between 40° C. and 130° C., particularly preferably between 60° C. and 110° C.


Suitable inert solvents are, for example, hydrocarbons, such as hexane, petroleum ether, benzene, toluene or xylene; chlorinated hydrocarbons, such as trichloroethylene, 1,2-dichloroethane, carbon tetrachloride, chloroform or dichloromethane; alcohols, such as methanol, ethanol, isopropanol, n-propanol, n-butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran (THF) or dioxane; glycol ethers, such as ethylene glycol monomethyl or monoethyl ether, ethylene glycol dimethyl ether (diglyme); ketones, such as acetone or butanone; amides, such as acetamide, dimethylacetamide or dimethylformamide (DMF); nitriles, such as acetonitrile; sulfoxides, such as dimethyl sulfoxide (DMSO); carbon disulfide; carboxylic acids, such as formic acid or acetic acid; nitro compounds, such as nitromethane or nitrobenzene; esters, such as ethyl acetate, or mixtures of the said solvents. Particular preference is given to glycol ethers, such as ethylene glycol monomethyl ether, THF, dichloromethane and/or DMF.


The cleavage of an ether is carried out by methods as are known to the person skilled in the art.


A standard method of ether cleavage, for example of a methyl ether, is the use of boron tribromide.


Hydrogenolytically removable groups, for example the cleavage of a benzyl ether, can be cleaved off, for example, by treatment with hydrogen in the presence of a catalyst (for example a noble-metal catalyst, such as palladium, advantageously on a support, such as carbon). Suitable solvents here are those indicated above, in particular, for example, alcohols, such as methanol or ethanol, or amides, such as DMF. The hydrogenolysis is generally carried out at temperatures between about 0 and 100° C. and pressures between about 1 and 200 bar, preferably at 20-30° C. and 1-10 bar.


Esters can be saponified, for example, using acetic acid or using NaOH or KOH in water, water/THF or water/dioxane, at temperatures between 0 and 100° C. Alkylations on the nitrogen are carried out under standard conditions, as are known to the person skilled in the art.


The compounds of the formulae I can furthermore be obtained by liberating them from their functional derivatives by solvolysis, in particular hydrolysis, or by hydrogenolysis.


Preferred starting materials for the solvolysis or hydrogenolysis are those which contain corresponding protected amino and/or hydroxyl groups instead of one or more free amino and/or hydroxyl groups, preferably those which carry an amino-protecting group instead of an H atom bonded to an N atom, for example those which conform to the formula I, but contain an NHR′ group (in which R′ denotes an amino-protecting group, for example BOC or CBZ) instead of an NH2 group.


Preference is furthermore given to starting materials which carry a hydroxyl-protecting group instead of the H atom of a hydroxyl group, for example those which conform to the formula I, but contain an R″O-phenyl group (in which R″ denotes a hydroxyl-protecting group) instead of a hydroxyphenyl group.


It is also possible for a plurality of—identical or different—protected amino and/or hydroxyl groups to be present in the molecule of the starting material. If the protecting groups present are different from one another, they can in many cases be cleaved off selectively.


The expression “amino-protecting group” is known in general terms and relates to groups which are suitable for protecting (blocking) an amino group against chemical reactions, but are easy to remove after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are, in particular, unsubstituted or substituted acyl, aryl, aralkoxymethyl or aralkyl groups. Since the amino-protecting groups are removed after the desired reaction (or reaction sequence), their type and size is furthermore not crucial; however, preference is given to those having 1-20, in particular 1-8, C atoms. The expression “acyl group” is to be understood in the broadest sense in connection with the present process. It includes acyl groups derived from aliphatic, araliphatic, aromatic or heterocyclic carboxylic acids or sulfonic acids, and, in particular, alkoxycarbonyl, aryloxycarbonyl and especially aralkoxycarbonyl groups. Examples of such acyl groups are alkanoyl, such as acetyl, propionyl, butyryl; aralkanoyl, such as phenylacetyl; aroyl, such as benzoyl, tolyl; aryloxyalkanoyl, such as POA; alkoxycarbonyl, such as methoxycarbonyl, ethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, BOC, 2-iodoethoxycarbonyl; aralkoxycarbonyl, such as CBZ (“carbobenzoxy”), 4-methoxybenzyloxycarbonyl, FMOC; arylsulfonyl, such as Mtr, Pbf, Pmc. Preferred amino-protecting groups are BOC and Mtr, furthermore CBZ, Fmoc, benzyl and acetyl.


The expression “hydroxyl-protecting group” is likewise known in general terms and relates to groups which are suitable for protecting a hydroxyl group against chemical reactions, but are easy to remove after the desired chemical reaction has been carried out elsewhere in the molecule. Typical of such groups are the above-mentioned unsubstituted or substituted aryl, aralkyl or acyl groups, furthermore also alkyl groups. The nature and size of the hydroxyl-protecting groups is not crucial since they are removed again after the desired chemical reaction or reaction sequence; preference is given to groups having 1-20, in particular 1-10, C atoms. Examples of hydroxyl-protecting groups are, inter alia, tertbutoxycarbonyl, benzyl, p-nitrobenzoyl, p-toluenesulfonyl, tert-butyl and acetyl, where benzyl and tert-butyl are particularly preferred. The COOH groups in aspartic acid and glutamic acid are preferably protected in the form of their tert-butyl esters (for example Asp(OBut)).


The compounds of the formula I are liberated from their functional derivatives—depending on the protecting group used—for example using strong acids, advantageously using TFA or perchloric acid, but also using other strong inorganic acids, such as hydrochloric acid or sulfuric acid, strong organic carboxylic acids, such as trichloroacetic acid, or sulfonic acids, such as benzene- or p-toluenesulfonic acid. The presence of an additional inert solvent is possible, but is not always necessary. Suitable inert solvents are preferably organic, for example carboxylic acids, such as acetic acid, ethers, such as tetrahydrofuran or dioxane, amides, such as DMF, halogenated hydrocarbons, such as dichloromethane, furthermore also alcohols, such as methanol, ethanol or isopropanol, and water. Mixtures of the above-mentioned solvents are furthermore suitable. TFA is preferably used in excess without addition of a further solvent, perchloric acid is preferably used in the form of a mixture of acetic acid and 70% perchloric acid in the ratio 9:1. The reaction temperatures for the cleavage are advantageously between about 0 and about 50° C., preferably between 15 and 30° C. (room temperature).


The BOC, OBut, Pbf, Pmc and Mtr groups can, for example, preferably be cleaved off using TFA in dichloromethane or using approximately 3 to 5 N HCl in dioxane at 15-30° C., the FMOC group can be cleaved off using an approximately 5 to 50% solution of dimethylamine, diethylamine or piperidine in DMF at 15-30° C.


Hydrogenolytically removable protecting groups (for example CBZ or benzyl) can be cleaved off, for example, by treatment with hydrogen in the presence of a catalyst (for example a noble-metal catalyst, such as palladium, advantageously on a support, such as carbon). Suitable solvents here are those indicated above, in particular, for example, alcohols, such as methanol or ethanol, or amides, such as DMF. The hydrogenolysis is generally carried out at temperatures between about 0 and 100° C. and pressures between about 1 and 200 bar, preferably at 20-30° C. and 1-10 bar. Hydrogenolysis of the CBZ group succeeds well, for example, on 5 to 10% Pd/C in methanol or using ammonium formate (instead of hydrogen) on Pd/C in methanol/DMF at 20-30° C.


The conversion of a radical R6═H into a radical R6═F can be carried out by reaction with Selectfluor® in a solvent, such as, for example, THF.


Pharmaceutical Salts and Other Forms

The said compounds according to the invention can be used in their final non-salt form. On the other hand, the present invention also encompasses the use of these compounds in the form of their pharmaceutically acceptable salts, which can be derived from various organic and inorganic acids and bases by procedures known in the art. Pharmaceutically acceptable salt forms of the compounds of the formula I are for the most part prepared by conventional methods. If the compound of the formula I contains a carboxyl group, one of its suitable salts can be formed by reacting the compound with a suitable base to give the corresponding base-addition salt. Such bases are, for example, alkali metal hydroxides, including potassium hydroxide, sodium hydroxide and lithium hydroxide; alkaline-earth metal hydroxides, such as barium hydroxide and calcium hydroxide; alkali metal alkoxides, for example potassium ethoxide and sodium propoxide; and various organic bases, such as piperidine, diethanolamine and N-methylglutamine. The aluminium salts of the compounds of the formula I are likewise included. In the case of certain compounds of the formula I, acid-addition salts can be formed by treating these compounds with pharmaceutically acceptable organic and inorganic acids, for example hydrogen halides, such as hydrogen chloride, hydrogen bromide or hydrogen iodide, other mineral acids and corresponding salts thereof, such as sulfate, nitrate or phosphate and the like, and alkyl- and monoarylsulfonates, such as ethanesulfonate, toluenesulfonate and benzenesulfonate, and other organic acids and corresponding salts thereof, such as acetate, trifluoroacetate, tartrate, maleate, succinate, citrate, benzoate, salicylate, ascorbate and the like. Accordingly, pharmaceutically acceptable acid-addition salts of the compounds of the formula I include the following: acetate, adipate, alginate, arginate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, citrate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, ethanesulfonate, fumarate, galacterate (from mucic acid), galacturonate, glucoheptanoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, iodide, isethionate, isobutyrate, lactate, lactobionate, malate, maleate, malonate, mandelate, metaphosphate, methanesulfonate, methylbenzoate, monohydrogenphosphate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, oleate, palmoate, pectinate, persulfate, phenylacetate, 3-phenylpropionate, phosphate, phosphonate, phthalate, but this does not represent a restriction.


Furthermore, the base salts of the compounds according to the invention include aluminium, ammonium, calcium, copper, iron(III), iron(II), lithium, magnesium, manganese(III), manganese(II), potassium, sodium and zinc salts, but this is not intended to represent a restriction. Of the above-mentioned salts, preference is given to ammonium; the alkali metal salts sodium and potassium, and the alkaline-earth metal salts calcium and magnesium. Salts of the compounds of the formula I which are derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines, also including naturally occurring substituted amines, cyclic amines, and basic ion exchanger resins, for example arginine, betaine, caffeine, chloroprocaine, choline, N,N′-dibenzylethylenediamine (benzathine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropylamine and tris(hydroxymethyl)methylamine (tromethamine), but this is not intended to represent a restriction.


Compounds of the present invention which contain basic nitrogen-containing groups can be quaternised using agents such as (C1-C4)alkyl halides, for example methyl, ethyl, isopropyl and tert-butyl chloride, bromide and iodide; di(C1-C4)alkyl sulfates, for example dimethyl, diethyl and diamyl sulfate; (C10-C18)alkyl halides, for example decyl, dodecyl, lauryl, myristyl and stearyl chloride, bromide and iodide; and aryl(C1-C4)alkyl halides, for example benzyl chloride and phenethyl bromide. Both water- and oil-soluble compounds according to the invention can be prepared using such salts.


The above-mentioned pharmaceutical salts which are preferred include acetate, trifluoroacetate, besylate, citrate, fumarate, gluconate, hemisuccinate, hippurate, hydrochloride, hydrobromide, isethionate, mandelate, meglumine, nitrate, oleate, phosphonate, pivalate, sodium phosphate, stearate, sulfate, sulfosalicylate, tartrate, thiomalate, tosylate and tromethamine, but this is not intended to represent a restriction.


The acid-addition salts of basic compounds of the formula I are prepared by bringing the free base form into contact with a sufficient amount of the desired acid, causing the formation of the salt in a conventional manner. The free base can be regenerated by bringing the salt form into contact with a base and isolating the free base in a conventional manner. The free base forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free base forms thereof.


As mentioned, the pharmaceutically acceptable base-addition salts of the compounds of the formula I are formed with metals or amines, such as alkali metals and alkaline-earth metals or organic amines. Preferred metals are sodium, potassium, magnesium and calcium. Preferred organic amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methyl-D-glucamine and procaine.


The base-addition salts of acidic compounds according to the invention are prepared by bringing the free acid form into contact with a sufficient amount of the desired base, causing the formation of the salt in a conventional manner. The free acid can be regenerated by bringing the salt form into contact with an acid and isolating the free acid in a conventional manner. The free acid forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free acid forms thereof.


If a compound according to the invention contains more than one group which is capable of forming pharmaceutically acceptable salts of this type, the invention also encompasses multiple salts. Typical multiple salt forms include, for example, bitartrate, diacetate, difumarate, dimeglumine, diphosphate, disodium and trihydrochloride, but this is not intended to represent a restriction.


With regard to that stated above, it can be seen that the expression “pharmaceutically acceptable salt” in the present connection is taken to mean an active compound which comprises a compound of the formula I in the form of one of its salts, in particular if this salt form imparts improved pharmacokinetic properties on the active compound compared with the free form of the active compound or any other salt form of the active compound used earlier. The pharmaceutically acceptable salt form of the active compound can also provide this active compound for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active compound with respect to its therapeutic efficacy in the body.


The invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically usable derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants.


Pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active compound per dosage unit. Such a unit can comprise, for example, 0.5 mg to 1 g, preferably 1 mg to 700 mg, particularly preferably 5 mg to 100 mg, of a compound according to the invention, depending on the condition treated, the method of administration and the age, weight and condition of the patient, or pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active compound per dosage unit. Preferred dosage unit formulations are those which comprise a daily dose or part-dose, as indicated above, or a corresponding fraction thereof of an active compound. Furthermore, pharmaceutical formulations of this type can be prepared using a process which is generally known in the pharmaceutical art.


Pharmaceutical formulations can be adapted for administration via any desired suitable method, for example by oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods. Such formulations can be prepared using all processes known in the pharmaceutical art by, for example, combining the active compound with the excipient(s) or adjuvant(s).


Pharmaceutical formulations adapted for oral administration can be administered as separate units, such as, for example, capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or foam foods; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.


Thus, for example, in the case of oral administration in the form of a tablet or capsule, the active-ingredient component can be combined with an oral, non-toxic and pharmaceutically acceptable inert excipient, such as, for example, ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing it with a pharmaceutical excipient comminuted in a similar manner, such as, for example, an edible carbohydrate, such as, for example, starch or mannitol. A flavour, preservative, dispersant and dye may likewise be present.


Capsules are produced by preparing a powder mixture as described above and filling shaped gelatine shells therewith. Glidants and lubricants, such as, for example, highly disperse silicic acid, talc, magnesium stearate, calcium stearate or polyethylene glycol in solid form, can be added to the powder mixture before the filling operation. A disintegrant or solubiliser, such as, for example, agar-agar, calcium carbonate or sodium carbonate, can likewise be added in order to improve the availability of the medicament after the capsule has been taken.


In addition, if desired or necessary, suitable binders, lubricants and disintegrants as well as dyes can likewise be incorporated into the mixture. Suitable binders include starch, gelatine, natural sugars, such as, for example, glucose or beta-lactose, sweeteners made from maize, natural and synthetic rubber, such as, for example, acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. The lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. The disintegrants include, without being restricted thereto, starch, methylcellulose, agar, bentonite, xanthan gum and the like. The tablets are formulated by, for example, preparing a powder mixture, granulating or dry-pressing the mixture, adding a lubricant and a disintegrant and pressing the entire mixture to give tablets. A powder mixture is prepared by mixing the compound comminuted in a suitable manner with a diluent or a base, as described above, and optionally with a binder, such as, for example, carboxymethylcellulose, an alginate, gelatine or polyvinylpyrrolidone, a dissolution retardant, such as, for example, paraffin, an absorption accelerator, such as, for example, a quaternary salt, and/or an absorbant, such as, for example, bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting it with a binder, such as, for example, syrup, starch paste, acadia mucilage or solutions of cellulose or polymer materials and pressing it through a sieve. As an alternative to granulation, the powder mixture can be run through a tableting machine, giving lumps of non-uniform shape, which are broken up to form granules. The granules can be lubricated by addition of stearic acid, a stearate salt, talc or mineral oil in order to prevent sticking to the tablet casting moulds. The lubricated mixture is then pressed to give tablets. The compounds according to the invention can also be combined with a free-flowing inert excipient and then pressed directly to give tablets without carrying out the granulation or dry-pressing steps. A transparent or opaque protective layer consisting of a shellac sealing layer, a layer of sugar or polymer material and a gloss layer of wax may be present. Dyes can be added to these coatings in order to be able to differentiate between different dosage units.


Oral liquids, such as, for example, solution, syrups and elixirs, can be prepared in the form of dosage units so that a given quantity comprises a pre-specified amount of the compound. Syrups can be prepared by dissolving the compound in an aqueous solution with a suitable flavour, while elixirs are prepared using a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersion of the compound in a non-toxic vehicle. Solubilisers and emulsifiers, such as, for example, ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavour additives, such as, for example, peppermint oil or natural sweeteners or saccharin, or other artificial sweeteners and the like, can likewise be added.


The dosage unit formulations for oral administration can, if desired, be encapsulated in microcapsules. The formulation can also be prepared in such a way that the release is extended or retarded, such as, for example, by coating or embedding of particulate material in polymers, wax and the like.


The compounds of the formula I and salts, solvates and physiologically functional derivatives thereof can also be administered in the form of liposome delivery systems, such as, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from various phospholipids, such as, for example, cholesterol, stearylamine or phosphatidylcholines.


The compounds of the formula I and the salts, solvates and physiologically functional derivatives thereof can also be delivered using monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds can also be coupled to soluble polymers as targeted medicament carriers. Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxyethylaspartamidophenol or polyethylene oxide polylysine, substituted by palmitoyl radicals. The compounds may furthermore be coupled to a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-epsilon-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydroxypyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.


Pharmaceutical formulations adapted for transdermal administration can be administered as independent plasters for extended, close contact with the epidermis of the recipient. Thus, for example, the active compound can be delivered from the plaster by iontophoresis, as described in general terms in Pharmaceutical Research, 3(6), 318 (1986).


Pharmaceutical compounds adapted for topical administration can be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.


For the treatment of the eye or other external tissue, for example mouth and skin, the formulations are preferably applied as topical ointment or cream. In the case of formulation to give an ointment, the active compound can be employed either with a paraffinic or a water-miscible cream base. Alternatively, the active compound can be formulated to give a cream with an oil-in-water cream base or a water-in-oil base.


Pharmaceutical formulations adapted for topical application to the eye include eye drops, in which the active compound is dissolved or suspended in a suitable carrier, in particular an aqueous solvent.


Pharmaceutical formulations adapted for topical application in the mouth encompass lozenges, pastilles and mouthwashes.


Pharmaceutical formulations adapted for rectal administration can be administered in the form of suppositories or enemas.


Pharmaceutical formulations adapted for nasal administration in which the carrier substance is a solid comprise a coarse powder having a particle size, for example, in the range 20-500 microns, which is administered in the manner in which snuff is taken, i.e. by rapid inhalation via the nasal passages from a container containing the powder held close to the nose. Suitable formulations for administration as nasal spray or nose drops with a liquid as carrier substance encompass active-ingredient solutions in water or oil.


Pharmaceutical formulations adapted for administration by inhalation encompass finely particulate dusts or mists, which can be generated by various types of pressurised dispensers with aerosols, nebulisers or insufflators.


Pharmaceutical formulations adapted for vaginal administration can be administered as pessaries, tampons, creams, gels, pastes, foams or spray formulations.


Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions comprising antioxidants, buffers, bacteriostatics and solutes, by means of which the formulation is rendered isotonic with the blood of the recipient to be treated; and aqueous and non-aqueous sterile suspensions, which may comprise suspension media and thickeners. The formulations can be administered in single-dose or multidose containers, for example sealed ampoules and vials, and stored in freeze-dried (lyophilised) state, so that only the addition of the sterile carrier liquid, for example water for injection purposes, immediately before use is necessary. Injection solutions and suspensions prepared in accordance with the recipe can be prepared from sterile powders, granules and tablets.


It goes without saying that, in addition to the above particularly mentioned constituents, the formulations may also comprise other agents usual in the art with respect to the particular type of formulation; thus, for example, formulations which are suitable for oral administration may comprise flavours.


A therapeutically effective amount of a compound of the formula I depends on a number of factors, including, for example, the age and weight of the animal, the precise condition that requires treatment, and its severity, the nature of the formulation and the method of administration, and is ultimately determined by the treating doctor or vet. However, an effective amount of a compound according to the invention for the treatment of neoplastic growth, for example colon or breast carcinoma, is generally in the range from 0.1 to 100 mg/kg of body weight of the recipient (mammal) per day and particularly typically in the range from 1 to 10 mg/kg of body weight per day. Thus, the actual amount per day for an adult mammal weighing 70 kg is usually between 70 and 700 mg, where this amount can be administered as a single dose per day or usually in a series of part-doses (such as, for example, two, three, four, five or six) per day, so that the total daily dose is the same. An effective amount of a salt or solvate or of a physiologically functional derivative thereof can be determined as the fraction of the effective amount of the compound according to the invention per se. It can be assumed that similar doses are suitable for the treatment of other conditions mentioned above.


The invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active compound.


The invention also relates to a set (kit) consisting of separate packs of

  • (a) an effective amount of a compound of the formula I and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios,
    • and
  • (b) an effective amount of a further medicament active compound.


The set comprises suitable containers, such as boxes, individual bottles, bags or ampoules. The set may, for example, comprise separate ampoules, each containing an effective amount of a compound of the formula I and/or pharmaceutically usable derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios,


and an effective amount of a further medicament active compound in dissolved or lyophilised form.


Use

The present compounds are suitable as pharmaceutical active compounds for mammals, especially for humans, in the treatment and control of cancer diseases.


The invention thus relates to the compounds of the formula I and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for use for the treatment of tumours, tumour growth, tumour metastases and/or AIDS.


The present invention encompasses the use of the compounds of the formula I and/or physiologically acceptable salts, tautomers and stereoisomers thereof for the preparation of a medicament for the treatment or prevention of cancer. Preferred carcinomas for the treatment originate from the group cerebral carcinoma, urogenital tract carcinoma, carcinoma of the lymphatic system, stomach carcinoma, laryngeal carcinoma and lung carcinoma bowel cancer. A further group of preferred forms of cancer are monocytic leukaemia, lung adenocarcinoma, small-cell lung carcinomas, pancreatic cancer, glioblastomas and breast carcinoma.


Also encompassed is the use of the compounds of the formula I and/or physiologically acceptable salts, tautomers and stereoisomers thereof for the preparation of a medicament for the treatment and/or control of a tumour-induced disease in a mammal, in which to this method a therapeutically effective amount of a compound according to the invention is administered to a sick mammal in need of such treatment. The therapeutic amount varies according to the particular disease and can be determined by the person skilled in the art without undue effort.


Particular preference is given to the use for the treatment of a disease, where the disease is a solid tumour.


The solid tumour is preferably selected from the group of tumours of the squamous epithelium, of the bladder, of the stomach, of the kidneys, of head and neck, of the oesophagus, of the cervix, of the thyroid, of the intestine, of the liver, of the brain, of the prostate, of the urogenital tract, of the lymphatic system, of the stomach, of the larynx and/or of the lung.


The solid tumour is furthermore preferably selected from the group lung adenocarcinoma, small-cell lung carcinomas, pancreatic cancer, glioblastomas, colon carcinoma and breast carcinoma.


Preference is furthermore given to the use for the treatment of a tumour of the blood and immune system, preferably for the treatment of a tumour selected from the group of acute myeloid leukaemia, chronic myeloid leukaemia, acute lymphatic leukaemia and/or chronic lymphatic leukaemia.


The invention furthermore relates to the use of the compounds according to the invention for the treatment of bone pathologies, where the bone pathology originates from the group osteosarcoma, osteoarthritis and rickets.


The compounds of the formula I may also be administered at the same time as other well-known therapeutic agents that are selected for their particular usefulness against the condition that is being treated.


The present compounds are also suitable for combination with known anti-cancer agents. These known anti-cancer agents include the following: oestrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors and further angiogenesis inhibitors. The present compounds are particularly suitable for administration at the same time as radiotherapy.


“Oestrogen receptor modulators” refers to compounds which interfere with or inhibit the binding of oestrogen to the receptor, regardless of mechanism. Examples of oestrogen receptor modulators include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY 117081, toremifene, fulvestrant, 4-[7-(2,2-dimethyl-1-oxopropoxy-4-methyl-2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-3-yl]phenyl 2,2-dimethylpropanoate, 4,4′-dihydroxybenzophenone-2,4-dinitrophenylhydrazone and SH646.


“Androgen receptor modulators” refers to compounds which interfere with or inhibit the binding of androgens to the receptor, regardless of mechanism. Examples of androgen receptor modulators include finasteride and other 5α-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole and abiraterone acetate.


“Retinoid receptor modulators” refers to compounds which interfere with or inhibit the binding of retinoids to the receptor, regardless of mechanism. Examples of such retinoid receptor modulators include bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, α-difluoromethylornithine, ILX23-7553, trans-N-(4′-hydroxyphenyl)retinamide and N-4-carboxyphenylretinamide.


“Cytotoxic agents” refers to compounds which result in cell death primarily through direct action on the cellular function or inhibit or interfere with cell myosis, including alkylating agents, tumour necrosis factors, intercalators, microtubulin inhibitors and topoisomerase inhibitors.


Examples of cytotoxic agents include, but are not limited to, tirapazimine, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosylate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methylpyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans,trans,trans)bis-mu-(hexane-1,6-diamine)-mu-[diamineplatinum(II)]bis-[diamine(chloro)platinum(II)]tetrachloride, diarisidinylspermine, arsenic trioxide, 1-(11-dodecylamino-10-hydroxyundecyl)-3,7-dimethylxanthine, zorubicin, idarubicin, daunorubicin, bisantrene, mitoxantrone, pirarubicin, pinafide, valrubicin, amrubicin, antineoplaston, 3′-deamino-3′-morpholino-13-deoxo-10-hydroxycaminomycin, annamycin, galarubicin, elinafide, MEN 10755 and 4-demethoxy-3-deamino-3-aziridinyl-4-methyl-sulfonyldaunorubicin (see WO 00/50032).


Examples of microtubulin inhibitors include paclitaxel, vindesine sulfate, 3′,4′-didehydro-4′-deoxy-8′-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS184476, vinflunine, cryptophycin, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzenesulfonamide, anhydrovinblastine, N,N-dimethyl-L-valylL-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t-butylamide, TDX258 and BMS188797.


Topoisomerase inhibitors are, for example, topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3′,4′-O-exobenzylidenechartreusin, 9-methoxy-N,N-dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H)propanamine, 1-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl-1H,12H-benzo[de]pyrano[3′,4′:b,7]indolizino[1,2b]quinoline-10,13(9H,15H)-dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2′-dimethylamino-2′-deoxyetoposide, GL331, N-[2-(dimethylamino)ethyl]-9-hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-1-carboxamide, asulacrine, (5a,5aB,8aa,9b)-9-[2-[N-[2-(dimethylamino)ethyl]-N-methylamino]ethyl]-5-[4-hydroxy-3,5-dimethoxyphenyl]-5,5a,6,8,8a,9-hexohydrofuro(3′,4′:6,7)naphtho(2,3-d)-1,3-dioxol-6-one, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]phenanthridinium, 6,9-bis[(2-aminoethyl)amino]benzo[g]isoquinoline-5,10-dione, 5-(3-aminopropylamino)-7,10-dihydroxy-2-(2-hydroxyethylaminomethyl)-6H-pyrazolo[4,5,1-de]acridin-6-one, N-[1-[2(diethylamino)ethylamino]-7-methoxy-9-oxo-9H-thioxanthen-4-ylmethyl]formamide, N-(2-(dimethylamino)ethyl)acridine-4-carboxamide, 6-[[2-(dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one and dimesna.


“Antiproliferative agents” include antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231 and INX3001 and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2′-deoxy-2′-methylidenecytidine, 2′-fluoromethylene-2′-deoxycytidine, N-[5-(2,3-dihydrobenzofuryl)sulfonyl]-N′-(3,4-dichlorophenyl)urea, N6-[4-deoxy-4-[N2-[2(E),4(E)-tetradecadienoyl]glycylamino]-L-glycero-B-L-mannoheptopyranosyl]adenine, aplidine, ecteinascidin, troxacitabine, 4-[2-amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4-b]-1,4-thiazin-6-yl-(S)-ethyl]-2,5-thienoyl-L-glutamic acid, aminopterin, 5-fluorouracil, alanosine, 11-acetyl-8-(carbamoyloxymethyl)-4-formyl-6-methoxy-14-oxa-1,11-diazatetracyclo(7.4.1.0.0)tetradeca-2,4,6-trien-9-ylacetic acid ester, swainsonine, lometrexol, dexrazoxane, methioninase, 2′-cyano-2′-deoxy-N4-palmitoyl-1-B-D-arabinofuranosyl cytosine and 3-aminopyridine-2-carboxaldehyde thio semicarbazone. “Antiproliferative agents” also include monoclonal antibodies to growth factors other than those listed under “angiogenesis inhibitors”, such as trastuzumab, and tumour suppressor genes, such as p53, which can be delivered via recombinant virus-mediated gene transfer (see U.S. Pat. No. 6,069,134, for example).


Evidence of the Action of Pharmacological Inhibitors on the Proliferation/Vitality of Tumour Cells In Vitro
1. Background

In the present experiment description, the inhibition of tumour cell proliferation/tumour cell vitality by active compounds is described.


The cells are sown in a suitable cell density in microtitre plates (96-well format) and the test substances are added in the form of a concentration series. After four further days of cultivation in serum-containing medium, the tumour cell proliferation/tumour cell vitality can be determined by means of an Alamar Blue test system.


2. Experimental Procedure
2.1 Cell Culture

For example commercially available colon carcinoma cell lines, ovary cell lines, prostate cell lines or breast cell lines, etc.


The cells are cultivated in medium. At intervals of several days, the cells are detached from the culture dishes with the aid of trypsin solution and sown in suitable dilution in fresh medium. The cells are cultivated at 37° C. and 10% CO2.


2.2. Sowing of the Cells

A defined number of cells (for example 2000 cells) per culture/well in a volume of 180 μl of culture medium are sown in microtitre plates (96 well cell-culture plates) using a multichannel pipette. The cells are subsequently cultivated in a CO2 incubator (37° C. and 10% CO2).


2.3. Addition of the Test Substances

The test substances are dissolved, for example, in DMSO and subsequently employed in corresponding concentration (if desired in a dilution series) in the cell culture medium. The dilution steps can be adapted depending on the efficiency of the active compounds and the desired spread of the concentrations. Cell culture medium is added to the test substances in corresponding concentrations. The addition of the test substances to the cells can take place on the same day as the sowing of the cells. To this end, in each case 20 μl of substance solution from the predilution plate are added to the cultures/wells. The cells are cultivated for a further 4 days at 37° C. and 10% CO2.


2.4. Measurement of the Colour Reaction

In each case, 20 μl of Alamar Blue reagent are added per well, and the microtitre plates are incubated, for example, for a further seven hours in a CO2 incubator (at 37° C. and 10% CO2). The plates are measured in a reader with a fluorescence filter at a wavelength of 540 nm. The plates can be shaken gently immediately before the measurement.


3. Evaluation

The absorbance value of the medium control (no cells and test substances used) is subtracted from all other absorbance values. The controls (cells without test substance) are set equal to 100 percent, and all other absorbance values are set in relation thereto (for example in % of control):


Calculation:






100
*

(





value





with





cells





and





test





substance

-






value





of





medium





control




)



(


value





with





cells

-

value





of





medium





control


)





IC50 values (50% inhibition) are determined with the aid of statistics programs, such as, for example, RS1.


IC50 data for compounds according to the invention are shown in Table 1.


4. Test for the Inhibition of PDK1

The experimental batches are carried out in a flashplate system with 384 wells/microtitration plate.


In each case, the PDK1 sample His6-PDK1 (1-50) (3.4 nM), the PDK1 substrate biotin-bA-bA-KTFCGTPEYLAPEVRREPRILSEEEQEMFRDFDYIADWC (400 nM), 4 μM ATP (with 0.2μ Ci of 33P-ATP/well) and the test substance in 50 μl of conventional experimental solution per well are incubated at 30° C. for 60 min. The test substances are employed in corresponding concentrations (if desired in a dilution series). The control is carried out without test substance. The reaction is stopped using standard methods and washed. The activity of the kinase is measured via the incorporated radioactivity in top count. In order to determine the non-specific kinase reaction (blank value), the experimental batches are carried out in the presence of 100 nM staurosporin.


5. Evaluation

The radioactivity (decompositions per minute) of the blank value (no use of test substance in the presence of staurosporin) is subtracted from all other radioactivity values. The controls (kinase activity without test substance) are set equal to 100 percent and all other radioactivity values (after subtracting the blank value) are expressed set in relation thereto (for example in % of the control).


Calculation:







100
*

(





value





of





the





kinase





activity





with





test





substance

-






blank





value




)



(


value





of





the





control

-

blank





value


)


=

%





of





the





control





IC50 values (50% inhibition) are determined with the aid of statistics programmes, such as, for example, RS1. IC50 data of compounds according to the invention are indicated in Table 1.














Material
Order No.
Manufacturer







Microtitre plates for cell culture
167008
Nunc


(Nunclon Surface 96-well plate)


DMEM
P04-03550
Pan Biotech


PBS (10x) Dulbecco
14200-067
Gibco


96-well plates (polypropylene)
267334
Nunc


AlamarBlue ™
BUF012B
Serotec


FCS
1302
Pan Biotech GmbH


Trypsin/EDTA solution 10x
L 2153
Biochrom AG


75 cm2 culture bottles
353136
BD Falcon


A2780
93112519
ECACC


Colo205
CCL222
ATCC


MCF7
HTB22
ATCC


PC3
CRL-1435
ATCC


384-well flash plates
SMP410A001PK
Perkin Elmer





APCI-MS (atmospheric pressure chemical ionisation-mass spectrometry) (M + H)+.






Method for the Cellular Testing of PDK1 Kinase Inhibitors in PC3 Cells.

The cellular assay for the determination of the PDK1 kinase activity is carried out as a Luminex assay in the 96-well format. PC3 cells are sown at 20,000 cells per well in 100 μl of medium (45% of RPMI1460/45% of Ham's F12/10% of FCS) and incubated on the following day for 30 min with a serial dilution of the test substance (7 concentrations) under serum-free conditions. The cells are subsequently lysed using 90 μl of lysis buffer (20 mM tris/HCl pH 8.0, 150 mM NaCl, 1% of NP40, 10% of glycerol, 1% of phosphatase inhibitor I, 1% of phosphatase inhibitor II, 0.1% of protease inhibitor cocktail III, 0.01% of benzonase) per well, and the lysates are separated off from insoluble cell constituents by means of centrifugation through a 96-well filter plate (0.65 μm). The lysates are incubated overnight at 4° C. with shaking with Luminex beads to which an anti-total PKB antibody is coupled. The detection is carried out on the following day by addition of a phospho-T308-PKB antibody and a species-specific peroxidase-labelled secondary antibody. The detection of phospho T308-PKB is carried out by measurement in a Luminex100 instrument by determination of 100 events per cavity in a measurement time of 60 sec. As pharmacological blank, the signals obtained from cells which have been treated with 10 μM staurosporin are subtracted from all other batches. The control value used for maximum phosphorylation of PKB on T308 are the signals from cells which have only been treated with the solvent (0.3% of DMSO). The values of the batches treated with test substance are calculated therefrom as percentage of control, and IC50 values are determined by means of RS 1.


Above and below, all temperatures are indicated in ° C. In the following examples, “conventional work-up” means: water is added if necessary, the pH is adjusted, if necessary, depending on the constitution of the end product, to values between 2 and 10, the mixture is extracted with ethyl acetate or dichloromethane, the phases are separated, the organic phase is dried over sodium sulfate, evaporated and purified by chromatography on silica gel and/or by crystallisation. Rf values on silica gel; eluent: ethyl acetate/methanol 9:1.


Mass spectrometry (MS): EI (electron impact ionisation) M+

    • FAB (fast atom bombardment) (M+H)+
    • ESI (electrospray ionisation) (M+H)+

      APCI-MS (atmospheric pressure chemical ionisation—mass spectrometry) (M+H)+.


      HPLC/SFC conditions:


      N: gradient: 5.5 min; flow rate: 2.75 ml/min from 90:10 to −


0:100 H2O/ACN

water+TFA (0.01% by vol.); acetonitrile+TFA (0.01% by


vol.)


column: Chromolith SpeedROD RP 18e 50-4.6


wavelength: 220 nm


Merck Hitachi La Chrome instrument


P: HPLC method:


gradient: 5.5 min; flow rate: 2.75 ml/min from 99:1 to −0:100


H2O/ACN

water+TFA (0.01% by vol.); acetonitrile+


TFA (0.01% by vol.)

column: Chromolith SpeedROD RP


18e 50-4.6


wavelength: 220 nm


Merck Hitachi La Chrome

instrument


0: HPLC method:


gradient: 5.5 min; flow rate: 2.75 ml/min from 99:1 to


−0:100 H2O/ACN

water+TFA (0.01% by vol.); acetonitrile+


TFA (0.01% by vol.)

column: Chromolith SpeedROD RP 18e 50-4.6


wavelength: 220 nm


Agilent instrument


Q: HPLC method (polar):


gradient: 0 min: 4% of B, 2.8 min: 100% of B; 3.3


min 100% of B; 3.4 min 4% of B

water+HCOOH (0.05% by vol.); acetonitrile+


HCOOH (0.04% by vol.)

column: Chromolith SpeedROD RP 18e 50-4.6


wavelength: 220 nm


Agilent instrument


W: HPLC method:


gradient: 10 min; flow rate: 3 ml/min from 99:1 to


−1:99 H2O/ACN

water+TFA (0.1% by vol.); acetonitrile+


TFA (0.1% by vol.)

column: Chromolith SpeedROD RP


18e 100-4.6


wavelength: 230 nm


Merck Hitachi La Chrom instrument


M: HPLC method:


gradient: 10 min; flow rate: 3 ml/min from 99:1


to −1:99 H2O/ACN

water+TFA (0.1% by vol.); acetonitrile+


TFA (0.1% by vol.)

column: Chromolith SpeedROD RP


18e 100-4.6


wavelength: 220 nm


Merck Hitachi La Chrom instrument


The compounds are accessible via various synthetic routes, some of which are shown here by way of example:




embedded image


embedded image







EXAMPLE 1
Preparation of 1-{3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-isoxazol-5-yl}-1-phenylethanol (“A1”)

1.1 2.96 g of tetrakis(triphenylphosphine)palladium(0) and 76 ml of sodium carbonate solution are added, under nitrogen protection, to a solution of 10.0 g of 5-bromo-1H-pyrrolo[2,3-b]pyridine (5-bromo-7-azaindole) and 18.0 g of 1-methyl-4-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole




embedded image


in 150 ml of DMF. The mixture is stirred at 10° C. for 2 h and at 120° C. for 1.5 h. The mixture is cooled and subjected to conventional work-up, giving 8.87 g of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine




embedded image


1.2 6.08 g of KOH are added to a solution of 8.87 g of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine in 75 ml of DMF. A solution of 75 ml of DMF and 11.01 g of iodine is subsequently added dropwise. The mixture is stirred at room temperature (RT) for a further 1.5 h. The reaction solution is then poured onto a mixture of 600 g of ice and 500 mg of sodium sulfite. The precipitate formed is separated off, washed with water and dried, giving 14.0 g of 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine.


1.3 11.31 ml of triethylamine and 333 mg of 4-(dimethylamino)pyridine are added to a suspension of 14.0 g of 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine in 200 ml of dichloromethane. A solution of 6.40 ml of di-tert-butyl dicarbonate in 100 ml of dichloromethane is then added dropwise, and the mixture is stirred at RT for a further 4 h. The mixture is diluted with dichloromethane and washed 3× with water. The organic phase is dried over sodium sulfate, and the solvent is removed, giving 14.5 g of tert-butyl 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate




embedded image


1.4 200 ml of THF are added to 4.02 g of sodium hydride (in paraffin) under nitrogen, and the mixture is cooled to 0° C. 9.8 g of 2-phenyl-3-butyn-2-ol are then added, and the ice bath is removed. The mixture is stirred for a further 1 h. The mixture is subsequently re-cooled to 0° C., and 8.61 ml of chloromethyl methyl ether are added under nitrogen. The mixture is stirred at RT for a further 14 h. Water is carefully added, and the mixture is subjected to conventional work-up. The crude product is subjected to flash column chromatography.


Conditions:
Apparatus: TELEDYNE-ISCO Combi Flash RF
Column: Silica Sep RF 120 g

Eluent: gradient petroleum ether:ethyl acetate


Flow rate: 85 ml/min


Detection: UV 254 nm

11 g of (1-methoxymethoxy-1-methylprop-2-ynyl)benzene are obtained




embedded image


1.5 The starting materials are combined as follows:


3.45 g of caesium carbonate, 67 mg of copper(I) iodide, 80 mg of palladium(II) acetate, 1.58 g of molybdenum hexacarbonyl, 1.5 g of tert-butyl 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate dissolved in 10 ml of acetonitrile, 1.21 g of (1-methoxymethoxy-1-methylprop-2-ynyl)benzene dissolved in 10 ml of toluene, and finally 0.45 ml of tri-tert-butylphosphine.


The mixture is stirred at 80° C. for 5 min. The reaction mixture is evaporated in a Rotavapor, adsorbed onto silica gel and chromatographed on a Combiflash, giving 1.59 g of tert-butyl 3-(4-methoxymethoxy-4-phenylpent-2-ynoyl)-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate




embedded image


1.6 194 μl of triethylamine and 122 mg of hydroxylammonium chloride are added to a solution of 360 mg of tert-butyl 3-(4-methoxymethoxy-4-phenylpent-2-ynoyl)-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate in 7.5 ml of ethanol. The mixture is stirred at 80° C. for 5 days.


The mixture is cooled and subjected to conventional work-up. The crude product still contains MOM-protected product. The crude product is dissolved in 10 ml of methanol to which 100 μl of 25% HCl have been added, and the mixture is stirred at 55° C. for 30 min.


The mixture is cooled to RT and subjected to conventional work-up. MTB ether is added to the oily residue, and the solvent is then removed, giving 176 mg of 1-{3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]isoxazol-5-yl}-1-phenylethanol (“A1”)




embedded image


HPLC/SFC condition: W; RT/min. 4.65;



1H NMR (400 MHz, DMSO-d6) [ppm] 12.30 (s, 1H), 8.58 (d, J=2.0 Hz, 1H), 8.36 (d, J=2.0 Hz, 1H), 8.27 (s, 1H), 8.13 (d, J=2.9 Hz, 1H), 7.98 (s, 1H), 7.52 (m, 2H), 7.34 (m, 2H), 7.25 (d, J=7.3 Hz, 1H), 6.75 (s, 1H), 6.18 (s, 1H), 3.89 (s, 3H), 1.87 (s, 3H).


An analogous procedure gives

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A6”


embedded image


W
 4.61











1H NMR (400 MHz, DMSO-d6) [ppm] 12.31 (d, J = 2.5 Hz, 1H), 8.59 (d, J = 2.1 Hz, 1H), 8.38 (d, J = 1.9 Hz, 1H), 8.28



(s, 1H), 8.15 (d, J = 2.8 Hz, 1H), 7.99 (d, J = 0.8 Hz, 1H), 7.79 (td, J = 8.0, 1.8 Hz, 1H), 7.41-7.31 (m, 1H), 7.26 (td, J =


7.6, 1.3 Hz, 1H), 7.10 (ddd, J = 11.8, 8.1, 1.1 Hz, 1H), 6.73 (s, 1H), 6.31 (s, 1H), 3.89 (d, J = 3.9 Hz, 3H), 1.89 (d, J =


32.4 Hz, 3H)













“A11”


embedded image


W
 4.98










M + H+ 418













“A12”


embedded image


W
 4.75











1H NMR (400 MHz, DMSO-d6) [ppm] 8.84 (s, 2H), 8.44 (s, 1H), 8.34 (s, 1H), 8.18 (d, J = 0.6 Hz, 1H), 7.86 (td, J = 8.0, 1.8



Hz, 1H), 7.36 (tdd, J = 7.0, 4.9, 1.9 Hz, 1H), 7.27 (td, J = 7.6, 1.3 Hz, 1H), 7.13-7.05 (m, 1H), 6.92 (s, 1H), 5.23 (s,


2H), 3.54 (m, 4H), 2.01 (s, 3H), 1.77-1.43 (m, 6H)













“A18”


embedded image


N
 1.95











1H NMR (400 MHz, DMSO-d6, 90° C.) [ppm] 11.43 (s, 1H), 8.44 (d, J = 2.1 Hz, 1H), 8.35 (s, 1H), 8.02 (s, 1H), 7.78



(s, 1H), 7.74-7.65 (m, 2H), 7.26 (ddd, J = 8.0, 4.9, 1.9 Hz, 1H), 7.16 (td, J = 7.6, 1.3 Hz, 1H), 7.02 (ddd, J = 12.0, 8.1, 1.2


Hz, 1H), 6.39 (s, 1H), 3.88 (s, 3H), 1.95 (d, J = 1.4 Hz, 3H)













“A24”


embedded image


W
 4.10










M + H+ 434













“A25”


embedded image


W
 4.82










M + H+ 418













“A28”


embedded image


N
 2.28











1H NMR (400 MHz, DMSO-d6, 90° C.) [ppm] 12.5 (s, broad, 1H), 11.48 (s, broad, 1H), 8.46 (d, J = 1.8 Hz, 1H), 8.43-



8.34 (m, 1H), 8.05 (s, 1H), 7.82 (s, 1H), 7.75 (s, 1H), 7.46-7.33 (m, 2H), 7.26-7.13 (m, 2H), 6.56 (s, 1H), 6.01 (d, J =


1.1 Hz, 1H), 5.35 (s, 1H), 3.89 (s, 3H)













“A34”


embedded image



 5.54










Column: 3 × 25 cm Chiracel OJ—H, eluent 104 ml of CO2 +25 ml of methanol, 12 min, enantiomer 1; absolute


configuration unknown; M + H+ 386













“A35”


embedded image



 7.07










Column: 3 × 25 cm Chiracel OJ—H, eluent 104 ml of CO2 +25 ml of methanol, 12 min, enantiomer 2; absolute


configuration unknown; M + H+ 386
















“A36”


embedded image


W
 4.63










1H NMR (500 MHz, DMSO-d6) [ppm] 12.34 (d, J = 2.4 Hz, 1H), 8.59 (d, J = 2.0 Hz, 1H), 8.38 (d, J = 2.0 Hz, 1H), 8.28 (s,


1H), 8.15 (d, J = 2.8 Hz, 1H), 8.00 (s, 1H), 7.43-7.25 (m, 3H), 7.14-7.04 (m, 1H), 6.80 (s, 1H), 3.90 (s, 3H), 1.88 (s, 3H)













“A55”


embedded image



 4.81










Column: Chiralcel OJ—H, eluent CO2 + 20% of methanol + 0.5% of DEA; 20 min, enantiomer 1, abs. stereochemistry


unknown; M + H+ 404













“A56”


embedded image



 6.80










Column: Chiralcel OJ—H, eluent CO2 + 20% of methanol + 0.5% of DEA; 20 min, enantiomer 2, abs. stereochemistry


unknown; M + H+ 404













“A61”


embedded image


M
 4.92











1H NMR (500 MHz, DMSO-d6) [ppm] 8.73 (s, 1H), 8.72 (s, 1H), 8.35 (s, 1H), 8.27 (s, 1H), 8.09 (s, 1H), 7.57 (t, J = 7.4 Hz,



1H), 7.38-7.10 (m, 2H), 6.89 (s, 1H), 3.90 (s, 3H), 1.95 (s, 3H)
















“A62”


embedded image



13.42










Column: Chiralpak AS—H, eluent: 20% of isopropOH 0.5% of DEA, 20 min; enantiomer 2; abs. stereochemistry unknown;


M + H+ 404













“A63”


embedded image



 8.47










Column: Chiralpak AS—H, eluent: 20% of isopropOH 0.5% of DEA, 20 min; enantiomer 1; abs. stereochemistry unknown;


M + H+ 404













“A64”


embedded image


M
 4.22











1H NMR (400 MHz, DMSO-d6) [ppm] 8.95 (d, J = 1.8 Hz, 1H), 8.76 (d, J = 1.8 Hz, 1H), 8.31 (s, 1H), 8.23 (s, 1H), 8.09 (t,



J = 9.7 Hz, 1H), 7.61 (t, J = 7.4 Hz, 1H), 7.47-7.12 (m, 2H), 6.93 (s, 1H), 3.96 (s, 3H), 2.08 (d, J = 11.9 Hz, 3H)













“A68”


embedded image


W
 3.78











1H NMR (500 MHz, DMSO-d6) [ppm] 11.73 (d, J = 1.9 Hz, 1H), 8.50 (dd, J = 12.0, 2.1 Hz, 2H), 8.13 (s, 1H), 7.86 (d,



J = 2.2 Hz, 2H), 7.46-7.29 (m, 4H), 7.25 (d, J = 6.6 Hz, 1H), 6.78 (s, 1H), 4.09-3.78 (m, 5H), 3.64 (m, 2H), 1.85 (s, 3H)













“A69”


embedded image


W
411   










M + H+ 411













“A73”


embedded image


W
 4.72











1H NMR (500 MHz, DMSO-d6) [ppm] 9.26 (d, J = 1.8 Hz, 1H), 8.79 (s, 1H), 8.34 (s, 1H), 8.20 (s, 1H), 8.09 (s, 1H), 7.36



(d, J = 4.2 Hz, 5H), 7.05 (s, 1H), 3.96 (s, 3H), 3.54 (s, 3H), 3.26 (s, 3H), 1.89 (s, 3H)













“A74”


embedded image


W












1H NMR (500 MHz, DMSO-d6) [ppm] 8.88 (d, J = 1.9 Hz, 1H), 8.75 (d, J = 1.8 Hz, 1H), 8.33 (s, 1H), 8.24 (s, 1H), 8.08 (d,



J = 0.6 Hz, 1H), 7.55 (d, J = 7.2 Hz, 2H), 7.42 (t, J = 7.5 Hz, 2H), 7.34 (d, J = 7.4 Hz, 1H), 6.96 (s, 1H), 6.01 (s, 1H),


3.96 (s, 3H)













“A75”


embedded image


W
 3.89











1H NMR (500 MHz, DMSO-d6) [ppm] 12.31 (d, J = 2.0 Hz, 1H), 8.59 (d, J = 2.0 Hz, 1H), 8.39 (d, J = 2.0 Hz, 1H), 8.28 (s,



1H), 8.15 (d, J = 2.8 Hz, 1H), 8.00 (s, 1H), 7.51 (d, J = 7.5 Hz, 2H), 7.37 (m, 2H), 7.29 (d, J = 7.3 Hz, 1H), 6.85 (s, 1H),


5.86 (s, 1H), 3.90 (s, 3H)













“A76”


embedded image


W
 3.89










M + H+ 399













“A84”


embedded image


W
 3.39










M + H+ 385













“A88”


embedded image


M
 4.17










M + H+ 419













“A89”


embedded image


M
 4.86










M + H+ 420













“A90”


embedded image


W
 4.06










M + H+ 399













“A92”


embedded image



 4.29










Column: Chiralcel OJ—H, eluent: 20% of isopropanol 0.5% of DEA, enantiomer 1; abs. stereochemistry unknown


M + H+ 418













”A93”


embedded image



 5.87










Column: Chiralcel OJ—H, eluent: 20% of isopropanol 0.5% of DEA, enantiomer 2; abs. stereochemistry unknown


M + H+ 418









EXAMPLE 2
Preparation of 3-(2-benzylthiazol-4-yl)-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine (“A2”)

2.1 100 mg of 5-bromo-1H-pyrrolo[2,3-b]pyridine (7-bromo-7-azaindole) are added to 324.8 mg of aluminium chloride in 10 ml of dichloromethane under argon. After stirring for 30 minutes, 0.31 ml of bromoacetyl chloride is added dropwise, and the mixture is stirred for a further 2 h.


The reaction mixture is cooled in an ice bath and carefully hydrolysed using methanol. The solvents are subsequently removed. The residue is adjusted to pH 7 using NaHCO3 solution and extracted with ethyl acetate. The organic phase is dried, filtered, and the solvent is removed, giving 138 mg of 2-bromo-1-(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)ethanone.


2.2 138 mg of 2-bromo-1-(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)ethanone are added to a solution of 59.5 mg of 2-phenylthioacetamide in 1.2 ml of 2-propanol. The mixture is boiled for 3 minutes.


The mixture is cooled, adjusted to pH 8 using aqueous ammonia solution and diluted with water. The precipitating crystals are separated off, washed with water and dried, giving 134 mg of 3-(2-benzylthiazol-4-yl)-5-bromo-H-pyrrolo[2,3-b]pyridine




embedded image


2.3 19.77 mg of tetrakis(triphenylphosphine)palladium(0) and 511.6 l of sodium carbonate solution are added to a solution of 134 mg of 3-(2-benzylthiazol-4-yl)-5-bromo-1H-pyrrolo[2,3-b]pyridine and 121 mg of 1-methyl-4-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole in 1 mol of DMF under argon. The mixture is stirred at 100° C. for 1 h.


The mixture is subjected to conventional work-up, giving 84 mg of 3-(2-benzyl-thiazol-4-yl)-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A2”)




embedded image


HPLC/SFC condition: N; RT/min. 2.14;



1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.83 (s, 1H), 8.52 (s, 2H), 8.20 (s, 1H), 7.94 (dd, J=5.4, 1.7 Hz, 2H), 7.81 (s, 1H), 7.50-7.21 (m, 5H), 4.42 (s, 2H), 3.90 (s, 3H).


The following compounds are obtained analogously

















HPLC/





SFC
RT/


No.
Name/structure
condition
min.







“A3”


embedded image


N
2.51











1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.85 (s, 1H), 8.50 (d, J = 2.1 Hz,



1H), 8.42 (d, J = 2.1 Hz, 1H), 8.08 (s, 1H), 7.82 (s, 1H), 7.68 (d, J = 2.6


Hz, 1H), 7.35 (ddd, J = 40.2, 21.4, 7.3 Hz, 6H), 4.36 (s, 2H), 3.90 (s, 3H)













“A4”


embedded image


N
2.47











1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.83 (s, 1H), 8.50 (dd, J = 7.5,



2.0 Hz, 2H), 8.17 (s, 1H), 7.92 (d, J = 2.7 Hz, 2H), 7.81 (s, 1H), 7.53 (dd,


J = 8.5, 6.8 Hz, 1H), 7.38 (ddd, J = 7.3, 6.4, 1.7 Hz, 1H), 7.31-7.16 (m,


2H), 4.44 (d, J = 8.9 Hz, 2H), 3.90 (s, 3H)
















“A5”


embedded image


N
2.47











1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.84 (s, 1H), 8.49 (d, J = 2.1 Hz,



1H), 8.37 (d, J = 2.2 Hz, 1H), 8.03 (s, 1H), 7.78 (d, J = 0.6 Hz, 1H),


7.69 (d, J = 2.0 Hz, 1H), 7.45-7.32 (m, 2H), 7.23 (dd, J = 4.5, 3.0 Hz, 1H),


4.46 (s, 2H), 3.91 (s, 3H)













“A7”


embedded image


N
2.45











1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.84 (s, 1H), 8.52 (s, 2H), 8.19



(s, 1H), 7.97-7.93 (m, 2H), 7.82 (s, 1H), 7.49-7.38 (m, 1H), 7.34-7.24


(m, 2H), 7.05-7.19 (m, 1H), 4.45 (s, 2H), 3.90 (s, 3H)
















“A9”


embedded image


N
2.48











1H NMR (400 MHz, DMSO-d6) δ [ppm] 11.83 (s, 1H), 8.50 (dd, J = 7.5,



2.0 Hz, 2H), 8.16 (s, 1H), 7.92 (d, J = 2.8 Hz, 2H), 7.83 (s, 1H), 7.45-


7.31 (m, 2H), 7.24 (dd, J = 5.1, 1.4 Hz, 1H), 4.52 (s, 2H), 3.90 (s, 3H)









EXAMPLE 3
Preparation of 3-[5-(3-fluorobenzyl)-1,2,4-oxadiazol-3-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A8”)

3.1 4.11 g of 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine are suspended in 20 ml of pyridine with stirring and under an argon atmosphere, and 1.81 ml of ethyl chloroformate are then slowly added dropwise. During the dropwise addition, the temperature is kept at 20-25° C. 15 ml of dichloromethane are also added, and the mixture is stirred at RT for a further 1.5 h. The mixture is subjected to conventional work-up, giving 4.8 g of ethyl 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate




embedded image


3.2 The following are weighed out together into a 50 ml round-bottomed flask which has been flushed with acetone and subsequently dried by heating:


2.0 g of ethyl 3-iodo-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate, 355.67 mg of zinc cyanide, 209.01 mg of [Pd2(dba)3]*CHCl3, 231.65 mg of 1,1′-bis(diphenylphosphino)ferrocene and 82.5 mg of zinc (coarsely powdered).


Under argon, 15 ml of N,N-dimethylacetamide are added, the mixture is warmed to 70° C. with stirring and stirred for a further 1 h. The mixture is cooled to RT, the solid material is filtered off through kieselguhr with suction and rinsed with acetone. The filtrate is concentrated to about 3 ml and subsequently chromatographed through a 540 g RP18 silica-gel column of a CombiFlash Companion. Two fraction groups (comprising 2 different products) are isolated. The isolated fractions comprising product are in each case combined and evaporated to an aqueous residue in a rotary evaporator. The residue is rendered basic using saturated NaHCO3 solution. The deposited precipitate is separated off and rinsed with water and diethyl ether. 2 products are obtained:


962 mg of ethyl 3-cyano-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-1-carboxylate,




embedded image


and 157 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonitrile




embedded image


3.3 500 mg of ethyl 3-cyano-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]-pyridine-1-carboxylate, 3.023 g of hydroxylammonium chloride and 2.37 g of KOH are combined, then 40 ml of absolute methanol and 8 ml of DMF are added. The suspension formed is stirred at RT for 60 h. A further 2 ml of DMF are added, and the mixture is stirred at RT for a further 24. The reaction mixture is diluted with water, ethyl acetate is then added, and the mixture is shaken. A precipitate deposits between the phases. This is separated off and rinsed with water and ethyl acetate, subsequently dried at 45° C. in vacuo, giving 222 mg of pale-brown, pulverulent substance (Ns).


The mother liquor (2-phase mixture) is transferred into the separating funnel again, the organic phase is separated off, and the aqueous phase is extracted a further 3× with ethyl acetate. The organic phases are combined, washed 1× with saturated NaCl solution, dried using sodium sulfate, filtered and evaporated. The residue is triturated with diethyl ether, the diethyl ether is removed, and the product is dried, giving 94 mg of yellow-brown, pulverulent substance (Extr. Ns).


Ns. and Extr. Ns. are identical, being N-hydroxy-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboxamidine.




embedded image


3.4


Reaction 1:

31.1 mg of (3-fluorophenyl)acetic acid, 45.7 mg of N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride and 28.6 mg of benzotriazol-1-ol hydrate are weighed out together, 0.7 ml of DMF is added, and the mixture is stirred at RT for 15 min. 50 mg of N-hydroxy-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]-pyridine-3-carboxamidine and 0.3 ml of DMF are then added. The mixture is stirred at RT for 20 min. The mixture is subjected to conventional work-up, giving 59.3 mg of greenish oil which crystallises on standing.


Reaction 2:

1 ml of diethylene glycol dimethyl ether is added to the product from reaction 1, and the mixture is stirred at 130° C. for 15 min. The mixture is subjected to conventional work-up. For purification, the residue is chromatographed over a preparative RP18e silica-gel column.


The isolated fractions are combined and evaporated to an aqueous residue in a rotary evaporator. The aqueous residue is rendered basic using saturated NaHCO3 solution and extracted 3× with ethyl acetate. The organic phases are combined, washed 1× with saturated NaCl solution and dried using sodium sulfate. The mixture is filtered, and the solvent is removed. The residue is dried again at 45° C. for 14 h, giving 26 mg of 3-[5-(3-fluorobenzyl)-1,2,4-oxadiazol-3-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A8”)




embedded image


HPLC/SFC condition: 0; RT/min. 2.94;



1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.41 (s, 1H), 8.61 (d, J=2.1 Hz, 1H), 8.39 (d, J=2.1 Hz, 1H), 8.23 (s, 1H), 8.18 (d, J=1.8 Hz, 1H), 7.91 (d, J=0.7 Hz, 1H), 7.45 (td, J=7.9, 6.3 Hz, 1H), 7.39-7.24 (m, 2H), 7.23-7.11 (m, 1H), 4.49 (s, 2H), 3.90 (s, 3H).


The following compounds are obtained analogously

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A10”


embedded image


P
 3.25











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.38 (s, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.37 (d, J =



2.0 Hz, 1H), 8.21 (s, 1H), 8.15 (d, J = 2.9 Hz, 1H), 7.88 (d, J = 0.7 Hz, 1H), 7.44 (d, J =


8.4 Hz, 1H), 7.34 (d, J = 6.3 Hz, 1H), 7.26 (dd, J = 5.1, 1.4 Hz, 1H), 4.55 (s, 2H), 3.89 (s, 3H)













“A13”


embedded image


O
291   











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.40 (s, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.37 (d, J =



2.1 Hz, 1H), 8.22 (s, 1H), 8.15 (s, 1H), 7.90 (d, J = 0.7 Hz, 1H), 7.53 (dt, J = 9.5, 4.7 Hz, 1H),


7.47-7.36 (m, 1H), 7.32-7.20 (m, 2H), 4.47 (s, 2H), 3.89 (s, 3H)













“A23”


embedded image


P
 2.99











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.38 (s, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.38 (d, J =



2.1 Hz, 1H), 8.21 (s, 1H), 8.17 (d, J = 2.6 Hz, 1H), 7.89 (d, J = 0.7 Hz, 1H), 7.61-7.50 (m, 2H),


7.48-7.30 (m, 3H), 6.82 (d, J = 5.0 Hz, 1H), 6.13 (d, J = 5.0 Hz, 1H), 3.89 (s, 3H)













“A27”


embedded image


P
 3.34










M + H+ 371













“A26”


embedded image


P
 3.24











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.36 (s, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.38 (d, J =



2.1 Hz, 1H), 8.21 (s, 1H), 8.16 (d, J = 2.8 Hz, 1H), 7.89 (d, J = 0.7 Hz, 1H), 7.49-7.25 (m, 5H),


4.43 (s, 2H), 3.89 (s, 3H)













“A31”


embedded image


P
 3.07











1H NMR (400 MHz, DMSO-d6) δ [ppm] 13.2 (s, broad, 1st rotamer), 12.38 (s, broad, 2nd



rotamer), 8.77 (s, 1st rotamer), 8.69 (d, 1st rotamer), 8.60 (d, J = 2.1 Hz, 2nd rotamer),


8.48 (d, 1st rotamer), 8.39 (d, J = 2.1 Hz, 2nd rotamer), 8.27 (s, 1st rotamer), 8.20


(s, 2nd rotamer), 8.18 (d, J = 2.8 Hz, 2nd rotamer), 7.97-7.94 (m, 1st rotamer), 7.89 (d, J =


0.6 Hz, 2nd rotamer), 7.59-7.24 (m, rotamers), 6.76 (s, 2nd rotamer), 5.74 (s, 1st, rotamer),


3.89 (s, rotamer), 2.0 and 1.99 (2s, rotamers)













“A50”


embedded image


N
 2.37











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.38 (d, J = 2.3 Hz, 1H), 8.60 (d, J =2.1 Hz, 1H),



8.39 (d, J = 2.1 Hz, 1H), 8.20 (s, 1H), 8.16 (d, J = 2.9 Hz, 1H), 7.88 (s, 1H), 7.83 (td, J =


8.0, 1.8 Hz, 1H), 7.48-7.39 (m, 1H), 7.33 (td, J = 7.6, 1.1 Hz, 1H), 7.21-7.12 (m, 1H),


6.97 (s, 1H), 3.89 (s, 3H), 2.01 (s, 3H)













“A51”


embedded image


N
 2.42











1H NMR (400 MHz, DMSO-d6) δ [ppm] 13.23 (s, broad, m, 1st rotamer), 12.40 (s, broad, 2nd



rotamer), 8.81 (s, 1st rotamer), 8.70 (d, 1st rotamer), 8.60 (d, J = 2.1 Hz, 2nd rotamer),


8.48 (d, 1st rotamer), 8.39 (d, J = 2.1 Hz, 2nd rotamer), 8.23-8.17 (m, 2nd rotamer), 7.97-


7.94 (m, 1st rotamer), 7.90 (s, 2nd rotamer), 7.52-7.31 (m, rotamers), 7.17 (td, J = 8.2, 2.3


Hz, rotamer), 6.94 (s, rotamer), 3.89 (s, rotamer), 2.01 (s, rotamer)
















“A52”


embedded image


N
 2.45











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.39 (s, broad, 1H), 8.61 (d, J = 2.1 Hz, 1H), 8.39



(d, J = 2.1 Hz, 1H), 8.22-8.17 (m, 2H), 7.89 (d, J = 0.7 Hz, 1H), 7.66-7.58 (m, 1H), 7.52-


7.42 (m, 1H), 7.39-7.31 (m, 1H), 7.17 (s, 1H), 3.89 (s, 3H), 2.03 (s, 3H)









EXAMPLE 4
Preparation of 3-[5-(3-fluorobenzyl)-1H-1,2,4-triazol-3-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A14”)

4.1 33.1 g of aluminium chloride are added to a solution of 10.0 g of 5-bromo-7-azaindole under argon, and the suspension is stirred at RT for 10 min. 8.3 ml of trichloroacetyl chloride are added, and the mixture is stirred for 14 h. The mixture is poured onto ice, stirred until the ice has melted, and the precipitated solid is separated off, washed with dichloromethane and water and dried at 50° C., giving 16.36 g of 1-(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trichloroethanone




embedded image


4.2 9.16 ml of hydrazinium hydroxide are added to a solution of 2.21 g of 1-(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)-2,2,2-trichloroethanone in 40 ml of methanol at RT. The mixture is stirred for 10 min. The solvents are removed to give a residue, which is subjected to conventional work-up, giving 1.25 g of 5-bromo-1H-pyrrolo-[2,3-b]pyridine-3-carbohydrazide




embedded image


4.3HCl gas is passed into a solution of 4.0 ml of 3-fluorophenylacetonitrile in 2.5 ml of ethanol and 25 ml of toluene at 0-5° C. for 30 min. The reaction solution is then warmed to RT, stirred for a further 30 min. The solution is poured into 50 ml of diethyl ether and cooled for 14 h. The deposited crystals are separated off and dried, giving 6.12 g of ethyl 2-(3-fluorophenyl)acetimidate hydrochloride




embedded image


4.4


Reaction 1:

2 ml of saturated NaHCO3 solution are added to 93.86 mg of ethyl 2-(3-fluorophenyl)acetimidate hydrochloride, and the mixture is extracted 3× with 3 ml of dichloromethane each time. The organic phases are combined, dried using sodium sulfate, and the solvent is subsequently removed.


100 mg of 5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide, 2 ml of absolute ethanol are added to the residue, and the mixture is heated to the boil with stirring. The mixture is boiled under reflux for 2 h. The mixture is cooled to RT, and 5 ml of diethyl ether are added. The precipitate is separated off, rinsed with diethyl ether and dried at 45° C., giving 96 mg of “intermediate”.


Reaction 2:

The intermediate is transferred into a microwave vessel, 1 ml of diethylene glycol dimethyl ether is added, and the mixture is warmed to 200° C. in the microwave in 5 min. For purification, the mixture is chromatographed over a preparative RP18e silica-gel column of a preparative HPLC. Conventional work-up gives 48 mg of 5-bromo-3-[5-(3-fluorobenzyl)-1H-1,2,4-triazol-3-yl]-1H-pyrrolo[2,3-b]pyridine




embedded image


4.5 1 ml of DMF and 194 l of sodium carbonate solution (2.0 mol/l) are added to 48 mg of 5-bromo-3-[5-(3-fluorobenzyl)-1H-1,2,4-triazol-3-yl]-1H-pyrrolo-[2,3-b]pyridine, 45.6 mg of 1-methyl-4-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole and 14.9 mg of tetrakis(triphenylphosphine)palladium(0). Argon is subsequently passed through the mixture for 5 min. The suspension is warmed to 120° C. and stirred at 90° C. for 30 min. The mixture is cooled and subjected to conventional work-up. For purification, the mixture is chromatographed over a preparative RP18e silica-gel column of a preparative HPLC. Conventional work-up gives 24.6 mg of 3-[5-(3-fluorobenzyl)-1H-1,2,4-triazol-3-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A14”)




embedded image


HPLC/SFC condition: 0; RT/min. 2.54;



1H NMR (400 MHz, DMSO-d6) δ [ppm] 13.43 (s, broad, 1H), 11.63 (s, 1H), 8.56 (d, J=2.0 Hz, 1H), 8.48 (d, J=1.7 Hz, 1H), 8.02 (s, 1H), 7.91 (s, 1H), 7.78 (s, 1H), 7.43-7.26 (m, 1H), 7.24-7.14 (m, 2H), 7.10-6.97 (m, 1H), 4.13 (s, 2H), 3.89 (s, 3H).


The following compounds are obtained analogously

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A15”


embedded image


P
2.85











1H NMR (400 MHz, DMSO-d6, 90° C.) δ [ppm] 13.50 (s, broad, 1H), 11.66 (s, 1H), 8.54 (d,



J = 2.1 Hz, 1H), 8.48 (d, J = 1.9 Hz, 1H), 8.01 (s, 1H), 7.90 (s, 1H), 7.76 (s, 1H), 7.45-7.34 (m,


1H), 7.32-7.23 (m, 1H), 7.18-7.1 (m, 2H), 4.12 (s, 2H), 3.89 (s, 3H)













“A16”


embedded image


P
2.91











1H NMR (400 MHz, DMSO-d6) δ [ppm] 13.69 (s, broad, 1H), 12.0 (s, broad, 1H), 8.56 (d,



J = 2.1 Hz, 1H), 8.53 (d, J = 2.1 Hz, 1H), 8.13 (s, 1H), 7.96 (s, 1H), 7.85 (s, 1H), 7.44-7.12


(m, 5H), 4.18 (s, 2H), 3.90 (s, 3H)













“A17”


embedded image


P
2.81





“A30”


embedded image


P
2.69





“A39”


embedded image


P
2.95









EXAMPLE 5
Preparation of 3-[5-(3-fluorobenzyl)-1,3,4-oxadiazol-2-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A19”)

5.1 100 mg of 5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide and 116.6 mg of ethyl 2-(3-fluorophenyl)acetimidate hydrochloride are suspended in 1 ml of ethanol, warmed to 85° C. and stirred for 15 h.


The mixture is cooled, the solid is separated off, washed with ethanol and water and dried, giving 99 mg of 5-bromo-3-[5-(3-fluorobenzyl)-1,3,4-oxadiazol-2-yl]-1H-pyrrolo[2,3-b]pyridine




embedded image


5.2 3 ml of DMF are added to 99 mg of 5-bromo-3-[5-(3-fluorobenzyl)-1,3,4-oxadiazol-2-yl]-1H-pyrrolo[2,3-b]pyridine and 80 mg of 1-methyl-4-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole, and the mixture is placed under an argon atmosphere. A solution of 30 mg of tetrakis(triphenylphosphine)-palladium(0) and 80 mg of sodium carbonate in 200 l of water is then added. Argon is passed through the solution for 5 min, the mixture is warmed to 120° C. and stirred for a further 2.5 h. The mixture is cooled and subjected to conventional work-up, giving 35 mg of 3-[5-(3-fluorobenzyl)-1,3,4-oxadiazol-2-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A19”)




embedded image


HPLC/SFC condition: N; RT/min. 2.20;



1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.51 (s, 1H), 8.63 (d, J=2.1 Hz, 1H), 8.42 (d, J=2.1 Hz, 1H), 8.28-8.20 (m, 2H), 7.91 (d, J=0.6 Hz, 1H), 7.51-7.08 (m, 4H), 4.40 (s, 2H), 3.90 (s, 3H).


The following compounds are obtained analogously

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A20”


embedded image


N
2.23











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.58 (s, broad, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.42



(d, J = 2.1 Hz, 1H), 8.26 (s, 1H), 8.24 (s, 1H), 7.93 (d, J = 0.5 Hz, 1H), 7.55-7.18 (m, 3H),


4.49 (s, 2H), 3.91 (s, 3H) “A21”













“A21”


embedded image


N
2.16











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.50 (s, 1H), 8.62 (d, J = 2.1 Hz, 1H), 8.41 (d, J =



2.1 Hz, 1H), 8.19-8.25 (m, 2H), 7.91 (d, J = 0.7 Hz, 1H), 7.49-7.22 (m, 5H), 4.36 (s, 2H),


3.90 (s, 3H)













“A22”


embedded image


N
2.18











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.52 (s, 1H), 8.63 (d, J = 2.0 Hz, 1H), 8.40 (d, J =



2.0 Hz, 1H), 8.22 (s, 2H), 7.91 (s, 1H), 7.59-7.12 (m, 4H), 4.40 (s, 2H), 3.91 (s, 3H)













“A29”


embedded image


N
1.92











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.55 (s, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.41 (d, J =



2.1 Hz, 1H), 8.25 (s, 1H), 8.22 (s, 1H), 7.91 (d, J = 0.7 Hz, 1H), 7.60-7.53 (m, 2H), 7.46-7.29


(m, 3H), 6.74 (d, J = 5.1 Hz, 1H), 6.09 (d, J = 5.0 Hz, 1H), 3.91 (s, 3H)













“A41”


embedded image


N
2.41










M + H+ 371













“A43”


embedded image


N
1.94











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.56 (s, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.39 (d, J = 2.1



Hz, 1H), 8.26 (d, J = 2.8 Hz, 1H), 8.21 (s, 1H), 7.90 (s, 1H), 7.78 (dd, J = 7.6, 6.2 Hz,


1H), 7.49-7.17 (m, 3H), 6.90 (d, J = 5.6 Hz, 1H), 6.27 (d, J = 5.6 Hz, 1H), 3.91 (s, 3H)
















“A45”


embedded image


N
2.11











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.51 (s, broad, 1H), 8.62 (d, J = 2.1 Hz, 1H), 8.38 (d,



J = 2.1 Hz, 1H), 8.20 (s, 1H), 8.19 (s, 1H), 7.88 (s, 1H), 7.54-7.47 (m, 2H), 7.40 (t, J = 7.7 Hz,


2H), 7.35-7.26 (m, 1H), 6.69 (s, 1H), 3.91 (s, 3H), 1.97 (s, 3H)
















“A46”


embedded image


N
2.12











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.52 (s, broad, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.42 (d,



J = 2.1 Hz, 1H), 8.27 (s, 1H), 8.22 (s, 1H), 7.91 (s, 1H), 7.62-7.29 (m, 3H), 7.10 (s, 1H), 6.33


(s, 1H), 3.91 (s, 3H)













“A47”


embedded image


N
2.06











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.57 (s, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.41 (d, J = 2.1



Hz, 1H), 8.28 (s, 1H), 8.23 (s, 1H), 7.91 (s, 1H), 7.50-7.36 (m, 3H), 7.19 (td, J = 8.3, 2.1 Hz,


1H), 6.90 (d, J = 5.1 Hz, 1H), 6.15 (d, J = 4.7 Hz, 1H), 3.91 (s, 3H)













“A82”


embedded image


N
2.47











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.67 (d, J = 2.1 Hz, 1H), 8.65 (d, J = 2.1 Hz, 1H),



8.42 (d, J = 2.0 Hz, 1H), 8.39 (d, J = 3.0 Hz, 1H), 8.24 (s, 1H), 7.92 (s, 1H), 7.62-7.49 (m, 3H),


7.39-7.33 (m, 1H), 7.20 (d, J = 44.9 Hz, 1H), 3.91 (s, 3H)
















“A48”


embedded image


N
2.13











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.53 (d, J = 2.2 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H),



8.29 (d, J = 2.1 Hz, 1H), 8.19 (d, J = 2.9 Hz, 1H), 8.14 (s, 1H), 7.89-7.80 (m, 2H), 7.43 (tdd,


J = 7.2, 5.1, 1.8 Hz, 1H), 7.34 (td, J = 7.6, 1.1 Hz, 1H), 7.20-7.10 (m, 1H), 6.84 (s, 1H), 3.91


(s, 3H), 2.01 (s, 3H)













“A81”


embedded image


N
2.33











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.59 (s, broad, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.40 (d,



J = 2.1 Hz, 1H), 8.29 (s, 1H), 8.23 (s, 1H), 7.91 (d, J = 0.6 Hz, 1H), 7.61-7.53 (m, 2H), 7.48-


7.36 (m, 3H), 5.86 (s, 1H), 3.91 (s, 3H), 3.43 (s, 3H)













“A86”


embedded image


N
2.27











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.60 (s, broad, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.36 (d,



J = 2.0 Hz, 1H), 8.27 (d, J = 2.9 Hz, 1H), 8.22 (s, 1H), 8.13 (d, J = 7.8 Hz, 1H), 7.89 (s, 1H),


7.87-7.74 (m, 2H), 7.62 (t, J = 7.6 Hz, 1H), 7.13 (d, J = 5.3 Hz, 1H), 6.29 (d, J = 5.1 Hz, 1H),


3.91 (s, 3H)













“A96”


embedded image


N
2.2 











1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.59 (s, 1H), 8.64 (s, 1H), 8.37 (s, 1H), 8.28 (s, 1H),



8.21 (s, 1H), 7.94-7.85 (m, 2H), 7.67 (d, J = 7.8 Hz, 1H), 7.55 (t, J = 7.4 Hz, 1H), 7.36 (t, J =


7.3 Hz, 1H), 7.03 (d, J = 5.3 Hz, 1H), 6.24 (d, J = 5.3 Hz, 1H), 3.92 (s, 3H)
















“A97”


embedded image


N
2.44











1H NMR (400 MHz, DMSO-d6) δ [ppm] 12.56 (s, 1H), 8.62 (d, J = 2.1 Hz, 1H), 8.36 (d, J = 2.1



Hz, 1H), 8.24 (s, 1H), 8.19 (s, 1H), 7.88 (s, 1H), 7.53-7.31 (m, 5H), 3.91 (s, 3H), 3.28 (s, 3H),


2.00 (s, 3H)









EXAMPLE 6
Preparation of 3-[3-(2-fluorobenzyl)-1,2,4-oxadiazol-5-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A32”)

6.1 1.27 ml of triethylamine are added to a solution of 684 mg of 2,2,2-tri-chloro-1-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]ethanone




embedded image


in 4 ml of water and 10 ml of THF, and the mixture is stirred at RT for 65 h. The solvents are removed, and the aqueous residue is extracted once with ethyl acetate (the organic phase is discarded). 10% citric acid is added to the aqueous phase, which is then extracted three times with ethyl acetate. The combined organic phases are washed with water, dried over sodium sulfate, and the solvent is subsequently removed, leaving white crystals. Product is still present in the aqueous phase. This is therefore saturated with NaCl and extracted again with ethyl acetate. Removal of the solvent likewise gives white crystals.


Together, 748 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid




embedded image


are obtained.


6.2 10 ml of DMF are added to 748 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid, 620 mg of pentafluorophenol and 694 mg of N,N′-dicyclohexylcarbodiimide under argon. The suspension is stirred for 16 h. 0.54 g of 1,1′-carbonyldiimidazole are then added, and the mixture is stirred for 45 h. 10 ml of dichloromethane are added, and the mixture is subjected to conventional work-up, giving 470 mg of pentafluorophenyl 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylate




embedded image


6.3 1 ml of 2-fluorophenylacetonitrile and 3.3 ml of triethylamine are added to a suspension of 1.05 g of hydroxylammonium chloride in 16 ml of ethanol. The mixture is stirred under reflux for 2 h. The solution is evaporated and subjected to conventional work-up, giving 1.26 g of 2-(2-fluorophenyl)-N-hydroxyacetamidine




embedded image


6.4 1.5 ml of DMF are added to 150 mg of pentafluorophenyl 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylate and 51 mg of 2-(2-fluorophenyl)-N-hydroxyacetamidine under argon, the mixture is warmed to 70° C. and stirred for 14 h. The mixture is heated to 100° C. and stirred for a further 4 h. The mixture is cooled and subjected to conventional work-up. For purification, the crude mixture is purified over a prep-HPLC apparatus, giving 14 mg of 3-[3-(2-fluorobenzyl)-1,2,4-oxadiazol-5-yl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine (“A32”)




embedded image


HPLC/SFC condition: N; RT/min. 2.43;



1H NMR (400 MHz, DMSO-d6) [ppm] 12.76 (s, 1H), 8.64 (d, J=2.1 Hz, 1H), 8.46 (d, J=3.0 Hz, 1H), 8.42 (d, J=2.1 Hz, 1H), 8.24 (s, 1H), 7.92 (s, 1H), 7.54-7.16 (m, 4H), 4.20 (s, 2H), 3.90 (s, 3H).


The following compounds are obtained analogously

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A33”


embedded image


N
2.49











1H NMR (400 MHz, DMSO-d6) [ppm] 8.62 (d, J = 2.1 Hz, 1H), 8.45 (s, 1H), 8.40 (d, J = 2.1 Hz, 1H),



8.23 (s, 1H), 7.91 (d, J = 0.6 Hz, 1H), 7.47-7.16 (m, 3H), 4.26 (s, 2H), 3.90 (s, 3H)













“A38”


embedded image


N
2.42











1H NMR (400 MHz, DMSO-d6) [ppm] 12.79 (s, 1H), 8.65 (d, J = 2.1 Hz, 1H), 8.48 (s, 1H), 8.43 (d, J =



2.1 Hz, 1H), 8.27 (s, 1H), 7.94 (s, 1H), 7.67-7.22 (m, 5H), 4.17 (s, 2H), 3.90 (s, 3H)













“A40”


embedded image


N
2.60











1H NMR (400 MHz, DMSO-d6) [ppm] 12.79 (s, broad, 1H), 8.66 (d, J = 2.1 Hz, 1H), 8.48 (d, J = 2.5



Hz, 1H), 8.45 (d, J = 2.1 Hz, 1H), 8.26 (s, 1H), 7.94 (d, J = 0.7 Hz, 1H), 7.46-7.38 (m, 1H), 7.31-7.23 (m,


2H), 7.17-7.08 (m, 1H), 4.22 (s, 2H), 3.92 (s, 3H)













“A67”


embedded image


N
2.33











1H NMR (500 MHz, DMSO-d6) [ppm] 12.74 (d, J = 2.2 Hz, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.47-8.43 (m,



2H), 8.24 (s, 1H), 7.95-7.92 (m, 1H), 7.56-7.52 (m, 2H), 7.40-7.22 (m, 3H), 6.27 (s, 1H), 3.91 (s, 3H),


1.92 (s, 3H)













“A72”


embedded image


N
2.40











1H NMR (500 MHz, DMSO-d6) [ppm] 12.76 (s, broad, 1H), 8.65 (d, J = 2.1 Hz, 1H), 8.48-8.43 (m, 2H),



8.24 (s, 1H), 7.94 (d, J = 0.6 Hz, 1H), 7.45-7.29 (m, 3H), 7.16-7.06 (m, 1H), 6.45 (s, 1H), 3.91 (s, 3H),


1.93 (s, 3H)













“A77”


embedded image


N
2.34











1H NMR (500 MHz, DMSO-d6) [ppm] 12.73 (s, broad, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.43 (s, 1H), 8.41



(d, J = 2.1 Hz, 1H), 8.21 (s, 1H), 7.89 (d, J = 0.5 Hz, 1H), 7.82 (td, J = 8.0, 1.7 Hz, 1H), 7.42-7.34 (m,


1H), 7.29 (td, J = 7.6, 1.1 Hz, 1H), 7.11 (ddd, J = 11.7, 8.1, 0.9 Hz, 1H), 6.46 (s, 1H), 3.90 (s, 3H), 1.96


(s, 3H)













“A78”


embedded image


N
2.42











1H NMR (500 MHz, DMSO-d6) [ppm] 12.76 (s, broad, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.46 (s, 1H), 8.42



(d, J = 2.1 Hz, 1H), 8.22 (s, 1H), 7.90 (d, J = 0.5 Hz, 1H), 7.62 (t, J = 7.3 Hz, 1H), 7.47-7.35 (m, 1H),


7.33-7.26 (m, 1H), 6.65 (s, 1H), 3.90 (s, 3H), 1.98 (s, 3H)













“A79”


embedded image


N
2.43











1H NMR (500 MHz, DMSO-d6) [ppm] 12.73 (s, broad, 1H), 8.63 (d, J = 1.9 Hz, 1H), 8.44 (s, 1H), 8.39



(d, J = 1.9 Hz, 1H), 8.21 (s, 1H), 8.02 (d, J = 7.5 Hz, 1H), 7.87 (s, 1H), 7.50-7.42 (m, 1H), 7.40-7.31 (m,


2H), 6.50 (s, 1H), 3.90 (s, 3H), 2.03 (s, 3H)













“A80”


embedded image


N
2.51











1H NMR (500 MHz, DMSO-d6) [ppm] 12.75 (s, broad, 1H), 8.64 (d, J= 2.1 Hz, 1H), 8.50-8.41 (m, 2H),



8.23 (s, 1H), 7.94 (d, J = 0.4 Hz, 1H), 7.64 (t, J = 1.8 Hz, 1H), 7.47 (dt, J = 7.7, 1.4 Hz, 1H), 7.42-7.30


(m, 2H), 6.47 (s, 1H), 3.91 (s, 3H), 1.92 (s, 3H)









EXAMPLE 7
Preparation of 1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-2H-1,2,4-triazol-3-yl}-1-phenylethanol (“A42”)

7.1 0.91 ml of triethylamine and 60.48 mg of 4-(dimethylamino)pyridine are added to a suspension of 1.16 g of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]-pyridine-3-carbonitrile in 10 ml of dichloromethane, 0.63 ml of benzenesulfonyl chloride is subsequently added. The mixture is stirred at RT for 30 min and subjected to conventional work-up, giving 673 mg of 1-benzenesulfonyl-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonitrile




embedded image


7.2HCl gas is passed into a solution of 673 mg of 1-benzenesulfonyl-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonitrile in 15 ml of methanol and 20 ml of dichloromethane at 0° C. for 30 min. The suspension is then stirred at RT for 60 h. The solvents are removed, and the residue is subjected to conventional work-up, giving 471 mg of methyl 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboximinate




embedded image


7.3 1.5 ml of ethanol are added to 50.0 mg of methyl 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carboximinate and 33.67 mg of 2-hydroxy-2-phenylpropionohydrazide, and the mixture is warmed to 85° C. (bath temperature) with stirring. The mixture is stirred at this temperature for 18 h. The mixture is cooled, and the solvent is removed. The residue is taken up in 1.5 ml of acetonitrile and, for purification, chromatographed over a preparative RP18 silica-gel column. Work-up gives 24.9 mg of 1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]-2H-1,2,4-triazol-3-yl}-1-phenylethanol (“A42”)




embedded image


HPLC/SFC condition: P; RT/min. 2.77; M+H386.


The following compounds are obtained analogously

















HPLC/





SFC
RT/


No.
Name/structure
condition
min.







“A44”


embedded image


N
2.00











1H NMR (400 MHz, DMSO-d6, 90° C.) [ppm] 8.60 (d, J = 2.1 Hz, 1H),



8.51 (d, J = 2.1 Hz, 1H), 8.04 (s, 1H), 7.93 (s, 1H), 7.81 (s, 1H), 7.52-


7.28 (m, 3H), 7.04 (td, J = 7.9, 1.6 Hz, 1H), 3.93 (s, 3H), 1.97 (s, 3H)













“A49”


embedded image


P
2.89





“A54”


embedded image


N
2.03











1H NMR (400 MHz, DMSO-d6) [ppm] 11.65 (s, broad, 1H), 8.53 (d,



J = 2.1 Hz, 1H), 8.48 (d, J = 2.1 Hz, 1H), 7.97 (s, 1H), 7.89 (s, 1H), 7.74


(d, J = 0.7 Hz, 1H), 7.54 (dd, J = 8.0, 6.8 Hz, 1H), 7.32-7.14 (m, 2H),


3.89 (s, 3H), 2.01 (s, 3H)









EXAMPLE 8
Preparation of (4-fluorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methanol (“A57”)

8.1 7.87 ml of hydrazinium hydroxide are added to a solution of 2.0 g of 2,2,2-trichloro-1-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]ethanone in 40 ml of methanol, and the mixture is stirred at RT for 30 min. The deposited precipitate is separated off, washed with 2-propanol and diethyl ether and dried, giving 1.26 g of grey substance (Ns.). The mother liquor is evaporated to dryness. The residue is taken up in 10 ml of water and, for purification, chromatographed over a 540 g RP18 silica-gel column. Work-up gives 140 mg of substance which is identical to Ns. In this way, 1.4 g of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbohydrazide




embedded image


are obtained.


8.2 A suspension of 100 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbohydrazide and 113.9 mg of ethyl 2-(4-fluorophenyl)-2-hydroxyacetimidate hydrochloride in 1.5 ml of ethanol is stirred at 90° C. for 14 h. The mixture is cooled and subjected to conventional work-up. For purification, the residue is chromatographed over a 12 g Si50 silica-gel column. Work-up gives 61 mg of (4-fluorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methanol (“A57”)




embedded image


HPLC/SFC condition: N; RT/min. 2.12; M+H+391;



1H NMR (500 MHz, DMSO-d6) [ppm] 12.55 (s, broad, 1H), 8.63 (d, J=2.1 Hz, 1H), 8.42 (d, J=2.1 Hz, 1H), 8.25 (s, 1H), 8.22 (s, 1H), 7.92 (s, 1H), 7.64-7.57 (m, 2H), 7.29-7.20 (m, 2H), 6.86 (d, J=5.1 Hz, 1H), 6.11 (d, J=4.9 Hz, 1H), 3.91 (s, 3H).


The following compounds are obtained analogously

















HPLC/SFC



No.
Name/structure
condition
RT/min.







“A58”


embedded image


N
2.17











1H NMR (400 MHz, DMSO-d6) [ppm] 12.57 (s, broad, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.37



(d, J = 2.1 Hz, 1H), 8.26 (d, J= 2.9 Hz, 1H), 8.20 (s, 1H), 7.92-7.87 (m, 2H), 7.56-7.38 (m,


3H), 7.05 (d, J = 5.5 Hz, 1H), 6.29 (d, J = 5.3 Hz, 1H), 3.91 (s, 3H)













“A59”


embedded image


N
2.24











1H NMR (500 MHz, DMSO-d6) [ppm] 12.58 (s, broad, 1H), 8.64 (d, J = 1.9 Hz, 1H), 8.42



(d, J = 1.9 Hz, 1H), 8.28 (d, J = 1.6 Hz, 1H), 8.23 (s, 1H), 7.93 (s, 1H), 7.65 (s, 1H), 7.55-7.40


(m, 3H), 6.93 (d, J = 5.3 Hz, 1H), 6.15 (d, J = 5.3 Hz, 1H), 3.91 (s, 3H)













“A60”


embedded image


N
2.27











1H NMR (400 MHz, DMSO-d6) [ppm] 12.56 (d, J = 2.1 Hz, 1H), 8.63 (d, J = 2.1 Hz, 1H),



8.41 (d, J = 2.1 Hz, 1H), 8.25 (d, J= 2.9 Hz, 1H), 8.22 (s, 1H), 7.91 (d, J = 0.5 Hz, 1H), 7.59


(d, J= 8.5 Hz, 2H), 7.51-7.45 (m, 2H), 6.84 (d, J = 5.2 Hz, 1H), 6.12 (d, J = 5.2 Hz, 1H),


3.91 (s, 3H)













“A65”


embedded image


N
2.35











1H NMR (400 MHz, DMSO-d6) [ppm] 12.58 (s, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.38 (d, J = 2.1



Hz, 1H), 8.27 (d, J = 2.7 Hz, 1H), 8.21 (s, 1H), 7.90 (s, 1H), 7.87 (dd, J = 7.8, 1.1 Hz, 1H), 7.70


(dd, J = 8.0, 1.5 Hz, 1H), 7.54 (t, J = 7.9 Hz, 1H), 7.17 (d, J = 5.7 Hz, 1H), 6.31 (d, J = 5.6 Hz,


1H), 3.91 (s, 3H)













“A70”


embedded image


Q
2.03





“A83”


embedded image


Q
2.09









EXAMPLE 9
Preparation of 1-(3-fluorophenyl)-1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]-2H-pyrazol-3-yl}ethanol (“A37”)

9.1 60 μl of triethylamine and 26.2 μl of hydrazinium hydroxide are added to a solution of 115 mg of tert-butyl 3-[4-(3-fluorophenyl)-4-methoxymethoxy-pent-2-ynoyl]-5-(1-methyl-1H-pyrazol-4-yl)pyrrolo[2,3-b]pyridine-1-carboxylate [preparation analogous to Example 1]




embedded image


in 2.5 ml of ethanol, and the mixture is stirred at RT for 50 h. The mixture is subjected to conventional work-up, giving 98 mg of 3-{5-[1-(3-fluorophenyl)-1-methoxymethoxyethyl]-1H-pyrazol-3-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine




embedded image


9.2 97 mg of 3-{5-[1-(3-fluorophenyl)-1-methoxymethoxyethyl]-1H-pyrazol-3-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine and 3.0 ml of HCl in dioxane (4M) are stirred at RT for 14 h. The solvents are removed, and the product is purified by preparative HPLC, giving 38 mg of 1-(3-fluorophenyl)-1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-2H-pyrazol-3-yl}ethanol (“A37”)




embedded image


HPLC/SFC condition: W; RT/min. 3.89; M+H+403;



1H NMR (500 MHz, DMSO-d6) [ppm] 11.88 (s, 1H), 8.52 (d, J=2.0 Hz, 1H), 8.45 (d, J=1.4 Hz, 1H), 8.17 (s, 1H), 7.91 (s, 1H), 7.86 (d, J=2.6 Hz, 1H), 7.41-7.25 (m, 3H), 7.04 (dd, J=7.8, 1.5 Hz, 1H), 6.61 (s, 1H), 4.5 (s, broad, 1H), 3.90 (s, 3H), 1.87 (s, 3H).


EXAMPLE 10
Preparation of 5-(1-methyl-1H-pyrazol-4-yl)-3-{5-[1-(2-methylpyridin-4-yl)-cyclopropyl]-1,3,4-oxadiazol-2-yl}-1H-pyrrolo[2,3-b]pyridine (“A85”)

10.1 5.31 ml of trimethylaluminium (2M in toluene) and 454.4 mg of tetrakis(triphenylphosphine)palladium(0) are added to a solution of (2-chloro-pyridin-4-yl)acetonitrile in 30 ml of 1,4-dioxane under nitrogen, and the mixture is stirred at 100° for 2 h. The mixture is cooled, the solvent is removed, the residue is subjected to conventional work-up, giving 2.17 g of (2-methyl-pyridin-4-yl)acetonitrile.


10.2 5.0 ml of 50% sodium hydroxide solution are added to a mixture of 670 mg of (2-methylpyridin-4-yl)acetonitrile, 23.1 mg of benzyltriethylammonium chloride and 0.88 ml of 1-bromo-2-chloroethane, and the mixture is stirred at 45° for 3 h. The mixture is cooled, rendered neutral using HCl solution and subjected to conventional work-up. The residue is chromatographed on the Companion, giving 600 mg of 1-(2-methylpyridin-4-yl)cyclopropanecarbonitrile




embedded image


10.3 A mixture of 300 mg of 1-(2-methylpyridin-4-yl)cyclopropane-carbonitrile, 0.95 ml of ethylene glycol and 5.0 ml of 50% sodium hydroxide solution is stirred at 100° for 14 h. The mixture is cooled, rendered slightly acidic using HCl and subjected to conventional work-up. The product is located in the aqueous phase. The water is removed, and the residue is chromatographed on the Companion, giving 300 mg of 1-(2-methylpyridin-4-yl)cyclopropanecarboxylic acid.


10.4 A suspension of 350 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbohydrazide, 300 mg of 1-(2-methylpyridin-4-yl)cyclopropanecarboxylic acid, 306 l of 4-methylmorpholine, 529 mg of N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDCI) and 190.2 mg of 1-hydroxybenzotriazole (HOBt) in 5 ml of DMF is stirred at RT for 5 h. The mixture is poured into 50 ml of water and extracted with 1-butanol. The solvent is removed from the combined organic phases. The residue is taken up in methanol, and the precipitate is separated off. The solvent is removed from the mother liquor, and the residue is chromatographed on the Companion with ethyl acetate/methanol, giving together 250 mg of N′-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl-1-(2-methylpyridin-4-yl)-cyclopropanecarbohydrazide




embedded image


10.5 A mixture of 120 mg of N′-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbonyl-1-(2-methylpyridin-4-yl)cyclopropanecarbohydrazide, 165.2 mg of methyl N-(triethylammoniumsulfonyl)carbamate [Burgess reagent] in 4 ml of tetrahydrofuran is heated in the microwave at 1000 for 35 min. The solvent is removed, and the residue is chromatographed on the preparative HPLC, giving 45.7 mg of 5-(1-methyl-1H-pyrazol-4-yl)-3-{5-[1-(2-methylpyridin-4-yl)-cyclopropyl]-1,3,4-oxadiazol-2-yl}-1H-pyrrolo[2,3-b]pyridine (“A85”)




embedded image


HPLC/SFC condition: G; RT/min. 1.49; M+H+398;


The following compounds are obtained analogously

















HPLC/





SFC
RT/


No.
Name/structure
condition
min.







“A91”


embedded image


Q
1.88





“A94”


embedded image


Q
1.89





“A95”


embedded image


Q
2.02









EXAMPLE 11
Preparation of (2-chlorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]-2H-1,2,4-triazol-3-yl}methanol (“A87”)

11.1 Liberation of ethyl 2-(2-chlorophenyl)-2-hydroxyacetiminate from its HCl salt


1 ml of saturated NaHCO3 solution is added, with stirring, to a suspension of 280 mg of ethyl 2-(2-chlorophenyl)-2-hydroxyacetiminate hydrochloride in 5 ml of ethyl acetate. After stirring for 2 minutes, the aqueous phase is removed, the organic phase is dried using sodium sulfate. The solvent is subsequently removed, giving




embedded image


11.2 A mixture of 279.1 mg of ethyl 2-(2-chlorophenyl)-2-hydroxy-acet-iminate and 260 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide in 5 ml of ethanol is warmed to the boil with stirring and heated under reflux for 16 h. The mixture is cooled, and the solvent is removed. 10 ml of diethyl ether are added, and the mixture is treated in an ultrasound bath. The solid is separated off and dried, giving 410 mg of N′-[2-(2-chlorophenyl)-2-hydroxy-1-iminoethyl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide




embedded image


11.3 A suspension of 297 mg of N′-[2-(2-chlorophenyl)-2-hydroxy-1-imino-ethyl]-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide in 3.5 g of pyridine is heated at 110° for 28 h. The mixture is cooled, and the solvent is removed.


The residue is dissolved in 2.5 ml of DMSO and purified by preparative HPLC (Chromolith prepRod 100-25), giving 27.1 mg of (2-chlorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-2H-1,2,4-triazol-3-yl}-methanol (“A87”)




embedded image


HPLC/SFC condition: P; RT/min. 2.92; M+H+406;


EXAMPLE 11a

The preparation of 1-{3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]isoxazol-5-yl}-1-pyridazin-4-ylethanol (“A98”)




embedded image


is carried out analogously to Example 1 (steps 1-4);


HPLC/SFC condition: M; RT/min (HPLC or SFC): 3.29; [M+H]+ 388;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.34 (d, J=3.0 Hz, 1H), 9.40 (m, 1H), 9.22 (dd, J=5.4, 1.2 Hz, 1H), 8.59 (d, J=2.1 Hz, 1H), 8.38 (d, J=2.2 Hz, 1H), 8.27 (d, J=0.9 Hz, 1H), 8.16 (d, J=2.9 Hz, 1H), 7.99 (d, J=0.9 Hz, 1H), 7.76 (dd, J=5.4, 2.5 Hz, 1H), 6.92 (s, 1H), 6.71 (s, 1H), 3.98 (s, 3H), 1.93 (s, 3H).


The preparation of 6-(1-hydroxy-1-{3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]isoxazol-5-yl}ethyl)-2-methyl-2H-pyridazin-3-one (“A99”)




embedded image


is carried out analogously to Example 1 (steps 1-4); [M+H]+ 418;


HPLC/SFC condition: W; RT/min (HPLC or SFC): 3.36;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.34 (d, J=3.2 Hz, 1H), 8.60 (d, J=2.2 Hz, 1H), 8.40 (d, J=2.2 Hz, 1H), 8.28 (s, 1H), 8.16 (d, J=2.9 Hz, 1H), 8.01 (s, 1H), 7.72 (d, J=9.7 Hz, 1H), 6.97 (m, 2H), 6.49 (s, 1H), 3.90 (s, 3H), 3.63 (s, 3H), 1.88 (s, 3H).


The preparation of 2,2-dimethyl-1-{3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridin-3-yl]isoxazol-5-yl}-1-pyridazin-4-ylpropan-1-ol (“A100”)




embedded image


is carried out analogously to Example 1 (steps 1-4);


HPLC/SFC condition: W; RT/min (HPLC or SFC): 4.16; [M+H]+ 430;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.16 (d, J=2.9 Hz, 1H), 9.46 (m, 1H), 9.25 (dd, J=5.5, 1.2 Hz, 1H), 8.57 (d, J=2.1 Hz, 1H), 8.47 (d, J=2.2 Hz, 1H), 8.26 (d, J=2.8 Hz, 1H), 8.23 (d, J=0.9 Hz, 1H), 7.91 (d, J=0.9 Hz, 1H), 7.87 (dd, J=5.5, 2.5 Hz, 1H), 7.07 (s, 1H), 6.77 (s, 1H), 3.90 (s, 3H), 1.00 (s, 9H).


The preparation of {5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-2H-pyrazol-3-yl}phenyl(tetrahydropyran-4-yl)methanol (“A101”)




embedded image


is carried out analogously to Example 1 (steps 1-4); the cyclisation is carried out using hydrazine;


HPLC/SFC condition: M; RT/min (HPLC or SFC): 3.59; [M+H]+ 455;



1H NMR (500 MHz, DMSO-d6; H/D exchange with TFA-d1) δ [ppm] 9.00 (d, J=1.8 Hz, 1H), 8.80 (d, J=1.7 Hz, 1H), 8.31 (s, 1H), 8.30 (s, 1H), 8.13 (s, 1H), 7.75 (s, 1H), 7.74 (s, 1H), 7.42 (m, 2H), 7.29 (m, 2H), 3.98 (m, 5H), 3.45 (m, 1H), 3.36 (m, 1H), 2.80 (m, 1H), 1.77 (m, 1H), 1.59 (m, 1H), 1.46 (m, 1H), 1.19 (m, 1H).


The following compounds are obtained analogously

















HPLC/SFC
RT/min.;


No.
Name/structure
condition
[M + H]+







“A102”


embedded image


M
4.29; 456 isomeric with “A103”





“A103”


embedded image


M
4.23; 456 isomeric with “A102”





“A104”


embedded image


M
3.01; 351











1H NMR (500 MHz, DMSO-d6) [ppm] 12.49 (d, J = 3.1 Hz, 1H), 9.96 (s, 1H), 8.63 (d, J = 2.0 Hz, 1H),



8.38 (d, J = 2.0 Hz, 1H), 8.26 (m, 2H), 7.98 (d, J = 0.8 Hz, 1H), 7.06 (s, 1H), 4.54 (m, 2H), 3.25 (m, 3H),


2.50 (m, 4H), 1.31 (m, 6H)













“A105”


embedded image


M
2.92; 296











1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.33 (d, J = 2.9 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H), 8.40 (d, J = 2.0



Hz, 1H), 8.31 (s, 1H), 8.16 (d, J = 2.8 Hz, 1H), 8.02 (d, J = 0.8 Hz, 1H), 6.93 (s, 1H), 4.57 (s, 2H), 3.90


(s, 3H)













“A106”


embedded image


isomeric with “A107”






“107”


embedded image


M isomeric with “A106”
3.18; 415









EXAMPLE 12
Preparation of C—((S)—C-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]-pyridin-3-yl]-1,3,4-oxadiazol-2-yl}-C-phenyl)methylamine (“A109”)



embedded image


a) A suspension of 200 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbohydrazide, 296.1 mg of Boc-D-phenylglycine, 175.1 l of 4-methylmorpholine, 302.2 mg of N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDCI), 108.7 mg of 1-hydroxybenzotriazole hydrate (HOBt) in 5 ml of DMF is stirred under an argon atmosphere for 14 hours. The mixture is poured into 50 ml of water, the precipitate is separated, dried, giving 382 mg of tert-butyl (2-{N′-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo-[2,3-b]pyridine-3-carbonyl]hydrazino}-2-oxo-1-phenylethyl)carbamate




embedded image


b) A mixture of 228 mg of the ester obtained under a) and 445.3 mg of Burgess reagent in 5 ml of THF is heated at 100° C. in the microwave for 15 min. After cooling, the mixture is purified on the preparative HPLC. Burgess reagent=methoxycarbonylsulfamoyl)triethylammonium hydroxide, inner salt.


20.3 mg of “A109” are obtained;


HPLC/SFC condition: Q; RT/min (HPLC or SFC): 1.36; [M+H]+ 372.


An analogous procedure gives C—((R)—C-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}-C-phenyl)methylamine (“A108”)




embedded image


HPLC/SFC condition: Q; RT/min (HPLC or SFC): 1.33; [M+H]+ 372;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.53 (s, 1H), 8.64 (s, 1H), 8.42 (s, 1H), 8.28-8.20 (m, 2H), 7.94 (s, 1H), 7.55 (d, 2H), 7.48-7.38 (m, 2H), 7.33 (t, J=7.2 Hz, 1H), 5.47 (s, 1H), 3.92 (s, 3H).


The following compounds are obtained analogously to Example 10


















RT/




HPLC/
min.;




SFC
[M +


No.
Name/structure
condition
H]+







“A110”


embedded image


Q
1.57; 412





“A111”


embedded image


Q
1.54; 400









The following compounds are obtained analogously to “A109”


















RT/




HPLC/
min.;




SFC
[M +


No.
Name/structure
condition
H]+







“A112”


embedded image


Q
1.43; 406





“A113”


embedded image


Q
1.43; 406





“A114”


embedded image


Q
1.42; 400





“A115”


embedded image


Q
1.22; 373





“A116”


embedded image


Q
1.34; 386





“A117”


embedded image


Q
1.44; 408





“A118”


embedded image


Q
1.42; 406









The following compounds are obtained analogously to Example 10

















HPLC/SFC
RT/min.;


No.
Name/structure
condition
[M + H]+







“A119”


embedded image


Q
2.36; 417





“A120”


embedded image


Q
1.67; 384









EXAMPLE 13
Preparation of 5-(1-methyl-1H-pyrazol-4-yl)-3-[5-(1-methyl-1-pyridazin-4-ylethyl)-1,3,4-oxadiazol-2-yl]-1H-pyrrolo[2,3-b]pyridine (“A121”)

13.1 2-(3,6-Dichloropyridazin-4-yl)-2-methylpropionic acid is boiled under reflux with thionyl chloride for 10 min.


The mixture is cooled, toluene is added, and the mixture is evaporated to dryness.


13.2 A solution of the product from 13.1 in 2.5 ml of THF is added dropwise to a suspension of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide, N-ethyldiisopropylamine in 2.5 ml of THF, and the mixture is stirred for a further one hour.


The mixture is subjected to conventional work-up, giving 6-chloro-4-(1-methyl-1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}pyridazin-3-ol




embedded image


13.3 Reaction of the product from 13.2 in acetonitrile with phosphoryl chloride under a protective-gas atmosphere at 900 gives, after conventional work-up, 3-{5-[1-(3,6-dichloropyridazin-4-yl)-1-methylethyl]-1,3,4-oxadiazol-2-yl}-5-(1-methyl-H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine




embedded image


13.4 The product from 13.3 is hydrogenated in ethanol, THF, triethylamine, palladium/aluminium powder (5% of Pd) using hydrogen under pressure. Conventional work-up gives 5-(1-methyl-1H-pyrazol-4-yl)-3-[5-(1-methyl-1-pyridazin-4-ylethyl)-1,3,4-oxadiazol-2-yl]-1H-pyrrolo[2,3-b]pyridine (“A121”)




embedded image


HPLC/SFC condition: P; RT/min (HPLC or SFC): 2.85; [M+H]+ 387;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.55 (s, 1H), 9.41 (dd, J=2.6, 1.2 Hz, 1H), 9.23 (dd, J=5.5, 1.1 Hz, 1H), 8.63 (d, J=2.1 Hz, 1H), 8.37 (d, J=2.1 Hz, 1H), 8.28 (s, 1H), 8.22 (s, 1H), 7.91 (d, J=0.7 Hz, 1H), 7.71 (dd, J=5.5, 2.7 Hz, 1H), 3.91 (s, 3H), 1.89 (s, 6H).


EXAMPLE 14
Preparation of 3-{5-[1-(6-chloropyridin-3-yl)cyclopropyl]-1H-1,2,4-triazol-3-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A122”)



embedded image


and 3-{5-[1-(6-chloropyridin-3-yl)cyclopropyl]-1,3,4-oxadiazol-2-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A123”)




embedded image


A mixture of 194 mg of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbohydrazide and 253 mg of ethyl 1-(6-chloropyridin-3-yl)cyclopropanecarboximinate in 5 ml of ethanol is stirred at 90° C. for 3 days.


The solvent is removed 5 ml of pyrdine are added the mixture is stirred at 150° C. for 2 days. After cooling, the solvent is removed. For purification, the residue is chromatographed over a 40 g Si50 silica-gel column of a CombiFlash Companion. The two products are obtained from the fractions; “A122”:


HPLC/SFC condition: P; RT/min (HPLC or SFC): 3.03; [M+H]+ 417;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 13.62 and 13.44 (2s, 1H), 12.12 and 11.86 (2s, 1H), 8.6-8.45 (m, 3H), 8.13 (s, 1H), 8.05-7.78 (m, 3H), 7.50 (d, J=8.2 Hz, 1H), 3.91 (s, 3H), 1.70-1.28 (m, 4H); “A123”:


HPLC/SFC condition: P; RT/min (HPLC or SFC): 3.14; [M+H]+ 418;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.51 (s, 1H), 8.60 (dd, J=13.1, 2.2 Hz, 2H), 8.32 (d, J=2.1 Hz, 1H), 8.23 (s, 1H), 8.20 (s, 1H), 8.01 (dd, J=8.3, 2.6 Hz, 1H), 7.89 (s, 1H), 7.56 (d, J=8.3 Hz, 1H), 3.91 (s, 3H), 1.80 (q, J=4.8 Hz, 2H), 1.63 (q, J=4.8 Hz, 2H).


Hydrogenation of “A123” gives the compound 5-(1-methyl-1H-pyrazol-4-yl)-3-[5-(1-pyridin-3-ylcyclopropyl)-1,3,4-oxadiazol-2-yl]-1H-pyrrolo[2,3-b]pyridine (“A124”)




embedded image


HPLC/SFC condition: N; RT/min (HPLC or SFC): 1.7; [M+H]+ 384;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.57 (d, J=1.7 Hz, 1H), 8.80 (dd, J=2.3, 0.6 Hz, 1H), 8.67 (d, J=2.1 Hz, 1H), 8.62 (dd, J=4.8, 1.6 Hz, 1H), 8.37 (d, J=2.1 Hz, 1H), 8.29 (d, J=2.8 Hz, 1H), 8.26 (s, 1H), 7.98 (ddd, J=7.9, 2.3, 1.7 Hz, 1H), 7.94 (d, J=0.6 Hz, 1H), 7.50 (ddd, J=7.9, 4.8, 0.7 Hz, 1H), 3.97 (s, 3H), 1.85 (dd, J=7.2, 4.7 Hz, 2H), 1.67 (dd, J=7.3, 4.8 Hz, 2H).


Hydrogenation of “A122” gives the compound 5-(1-methyl-1H-pyrazol-4-yl)-3-[5-(1-pyridin-3-ylcyclopropyl)-1H-1,2,4-triazol-3-yl]-1H-pyrrolo[2,3-b]pyridine (“A125”)




embedded image


HPLC/SFC condition: P; RT/min (HPLC or SFC): 2.63; [M+H]+ 383;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 13.60 (s, 1H), 12.11 and 11.88 (2s, 1H), 8.67 (s, 1H), 8.61-8.42 (m, 3H), 8.14 (s, 1H), 8.07-7.78 (m, 3H), 7.39 (dd, J=7.7, 4.8 Hz, 1H), 3.92 (s, 3H), 1.68-1.25 (m, 4H).


Heating of “A122” in water and conc. HCl (bath temperature 120°; 3 days) and purification by preparative HPLC gives the compound 5-(1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-2H-1,2,4-triazol-3-yl}cyclopropyl)-1H-pyridin-2-one (“A126”)




embedded image


HPLC/SFC condition: P; RT/min (HPLC or SFC): 2.73; [M+H]+ 399;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 13.57 and 13.34 (2s, 1H), 12.12 and 11.86 (2s, 1H), 11.50 (s, 1H), 8.53 (s, 2H), 8.11 (s, 1H), 8.06-7.75 (m, 2H), 7.65-7.24 (m, 2H), 6.35 (d, J=9.4 Hz, 1H), 3.92 (s, 3H), 1.56-1.11 (m, 4H).


An analogous procedure to the preparation of “A42” gives the compound 3-{5-[1-(6-methoxypyridin-3-yl)cyclopropyl]-1H-1,2,4-triazol-3-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (“A 127”)




embedded image


HPLC/SFC condition: P; RT/min (HPLC or SFC): 2.97; [M+H]+ 413;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 13.53 and 13.31 (2s, 1H), 12.09 and 11.82 (2s, 1H), 8.51 (s, 2H), 8.23 (d, J=2.4 Hz, 1H), 8.11 (s, 1H), 8.03-7.68 (m, 3H), 6.80 (d, J=8.4 Hz, 1H), 3.90 (s, 3H), 3.86 (s, 3H), 1.36 (dd, J=83.7, 47.3 Hz, 4H).


An analogous procedure to the preparation of “A57” gives the compound methyl 3-chloro-4-(hydroxy-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]-pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methyl)benzoate (“A128”)




embedded image


HPLC/SFC condition: N; RT/min (HPLC or SFC): 2.26; [M+H]+ 465.


“A128” gives under standard conditions the compound 3-chloro-N-cyclohexyl-4-(hydroxy-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methyl)benzamide (“A129”)




embedded image


HPLC/SFC condition: N; RT/min (HPLC or SFC): 2.41; [M+H]+ 532;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.59 (s, 1H), 8.64 (d, J=2.1 Hz, 1H), 8.40 (d, J=2.1 Hz, 1H), 8.36 (d, J=7.9 Hz, 1H), 8.27 (d, J=1.7 Hz, 1H), 8.22 (s, 1H), 8.0-7.9 (m, 4H), 7.13 (d, J=5.6 Hz, 1H), 6.31 (d, J=5.5 Hz, 1H), 3.91 (s, 3H), 3.82-3.72 (m, 1H), 1.89-1.68 (m, 4H), 1.66-0.77 (m, 6H).


EXAMPLE 15
Preparation of (R)-(2-chlorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methanol (“A131”) and (S)-(2-chlorophenyl)-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}methanol (“A130”) from the racemate “A58”

15 ml of DMF are added to 520 mg of “A58” and 281.9 mg of imidazole. 526.7 1 of tert-butyldiphenylchlorosilane are added to the suspension, and the mixture is stirred at RT for 16 h. Silyl reagent is again added, and the mixture is again stirred for 16. The mixture is subjected to conventional work-up and purified over a 205 g RP18ec silica-gel column of a Combiflash Companion. Work-up gives 3-{5-[(tert-butyldiphenylsilanyloxy)-(2-chlorophenyl)methyl]-1,3,4-oxadiazol-2-yl}-5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine




embedded image


The analytical separation is carried out on Chiralpak AD-H with CO2+20% of methanol.


For preparative separation, in each case 50 mg of the compound obtained are dissolved in 10 ml of boiling methanol and separated over 3×25 cm Chiralpak AD-H column with 80 ml of CO2 and 20 ml of methanol.


2 fractions are collected.


Fraction 1: enantiomer ratio 95:5;


fraction 2: enantiomer ratio 2:98.


The absolute configuration of the two products has not been assigned.


In order to cleave off the silyl group, the products obtained from the two fractions are treated with tetrabutylammonium fluoride in THF. Conventional work-up gives “A131”:


HPLC/SFC condition: N; RT/min (HPLC or SFC): 2.17; [M+H]+ 407; and “A130”:


HPLC/SFC condition: N; RT/min (HPLC or SFC): 2.19; [M+H]+ 407;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.57 (d, J=2.1 Hz, 1H), 8.62 (d, J=2.1 Hz, 1H), 8.36 (d, J=2.1 Hz, 1H), 8.25 (d, J=2.9 Hz, 1H), 8.20 (s, 1H), 7.93-7.85 (m, 2H), 7.55-7.46 (m, 2H), 7.46-7.35 (m, 1H), 6.99 (d, J=5.5 Hz, 1H), 6.28 (d, J=5.5 Hz, 1H), 3.91 (s, 3H).


An analogous procedure to Example 10 gives the compound I-(2-chlorophenyl)-1-{5-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-1,3,4-oxadiazol-2-yl}ethanol (“A132”)




embedded image


HPLC/SFC condition: N; RT/min (HPLC or SFC): 2.26; [M+H]+ 421;



1H NMR (500 MHz, DMSO-d6) δ [ppm] 12.52 (s, 1H), 8.62 (dd, J=8.9, 2.2 Hz, 1H), 8.23 (d, J=2.1 Hz, 1H), 8.18 (d, J=2.5 Hz, 1H), 8.10 (s, 1H), 8.08-8.02 (m, 1H), 7.78 (s, 1H), 7.57-7.49 (m, 1H), 7.47-7.37 (m, 2H), 6.90 (s, 1H), 3.90 (s, 3H), 2.06 (s, 3H).


PDK 1 inhibition


IC50 of compounds according to the invention


















IC50
IC50




[PDK1]
[PDK1]



No.
biochemical
cellular









“A1”
A
A



“A2”

B



“A3”

B



“A4”
B
B



“A6”
A
A



“A7”

B



“A8”
B



“A9”
B
B



“A11”
A
A



“A12”
A
A



“A13”
B



“A14”
A
B



“A15”
A
B



“A16”
A
A



“A17”
A
B



“A18”
A
B



“A19”
A
B



“A20”
A
B



“A21”
A
B



“A22”
A
A



“A23”
A
B



“A24”
A
A



“A25”
A
A



“A26”
B



“A27”

B



“A28”
A
B



“A29”
A
B



“A30”
A



“A31”
A
A



“A34”
A
A



“A35”
A
B



“A36”
A
A



“A37”
A
A



“A39”
B
B



“A42”
A
B



“A43”
A
B



“A44”
A
B



“A45”
A
B



“A46”
A
A



“A47”
A
A



“A48”
A
A



“A49”
A
B



“A50”
A
B



“A51”
A
B



“A52”
A
B



“A54”
A
B



“A55”
A
A



“A56”
A
A



“A57”
A
B



“A58”
A
A



“A59”
A
A



“A60”
B
B



“A61”
A
A



“A62”
A
B



“A63”
A
B



“A64”
A
A



“A65”
A
A



“A67”
B
A



“A68”
A
B



“A69”
B
B



“A70”
A
A



“A72”
A
A



“A73”
B



“A74”
A
A



“A75”
A
A



“A76”
B



“A77”
A
B



“A78”
A
A



“A79”
B
A



“A80”
A
A



“A81”
A
B



“A82”
B
B



“A83”
A
A



“A84”
A
B



“A85”
B
B



“A86”
A
A



“A87”
A
B



“A88”
A
B



“A89”
A
A



“A90”
A



“A91”
B
B



“A92”
A



“A93”
A



“A98”
A



“A99”
A
B



“A100”
A
B



“A101”
A



“A102”
A



“A103”
A



“A104”
A



“A105”
A
B



“A106”
B



“A107”
A
B



“A108”
A



“A109”
A



“A110”
A
B



“A111”
A



“A112”
A
A



“A113”
A
B



“A114”
B



“A115”
B



“A116”
A
B



“A117”
B
B



“A118”
A
A



“A119”

B



“A120”
B
B



“A121”
B



“A122”
B
B



“A123”
B
B



“A124”
A
B



“A125”
A
B



“A126”
A
B



“A127”
B
B



“A128”
B
B



“A129”

B



“A130”
A
A



“A131”
A



“A132”
A
B







IC50: 0.5 nM-1M = A 1M-10 M = B






The following examples relate to medicaments:


EXAMPLE A
Injection Vials

A solution of 100 g of an active compound of the formula I and 5 g of disodium hydrogenphosphate in 3 l of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contains 5 mg of active compound.


EXAMPLE B
Suppositories

A mixture of 20 g of an active compound of the formula I with 100 g of soya lecithin and 1400 g of cocoa butter is melted, poured into moulds and allowed to cool. Each suppository contains 20 mg of active compound.


EXAMPLE C
Solution

A solution is prepared from 1 g of an active compound of the formula I, 9.38 g of NaH2PO4.2H2O, 28.48 g of Na2HPO4.12H2O and 0.1 g of benzalkonium chloride in 940 ml of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 l and sterilised by irradiation. This solution can be used in the form of eye drops.


EXAMPLE D
Ointment

500 mg of an active compound of the formula I are mixed with 99.5 g of Vaseline under aseptic conditions.


EXAMPLE E
Tablets

A mixture of 1 kg of active compound of the formula I, 4 kg of lactose, 1.2 kg of potato starch, 0.2 kg of talc and 0.1 kg of magnesium stearate is pressed in a conventional manner to give tablets in such a way that each tablet contains 10 mg of active compound.


EXAMPLE F
Dragees

Tablets are pressed analogously to Example E and subsequently coated in a conventional manner with a coating of sucrose, potato starch, talc, tragacanth and dye.


EXAMPLE G
Capsules

2 kg of active compound of the formula I are introduced into hard gelatine capsules in a conventional manner in such a way that each capsule contains 20 mg of the active compound.


EXAMPLE H
Ampoules

A solution of 1 kg of active compound of the formula I in 60 l of bidistilled water is sterile filtered, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of active compound.

Claims
  • 1. Compounds of the formula I
  • 2. Compounds according to claim 1in whichR1 denotes Het1,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 3. Compounds according to claim 1in whichHet1 denotes pyrazolyl, which may be monosubstituted by A, CH2COHet3 or COOA,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 4. Compounds according to claim 1in whichAr denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, COOR5 and/or CON(R5)2,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 5. Compounds according to claim 1in whichHet denotes a monocyclic aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A and/or [C(R5)2]nOR5,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 6. Compounds according to claim 1in whichA denotes unbranched or branched alkyl having 1-6 C atoms,in which 1-5H atoms may be replaced by F, orcyclic alkyl having 3-7 C atoms,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 7. Compounds according to claim 1in whichQ denotes Het-diyl,R1 denotes Het1,R2, R3 each, independently of one another, denote H, Hal, A, Ar, [C(R5)2]nOH, [C(R5)2]nOA or [C(R5)2]nN(R5)2,R2, R3 together also denote ═O, ═CH2 or an alkylene chain having 2-5 C atoms,R4 denotes H, Ar or Het2,R5 denotes H or A′,Het1 denotes pyrazolyl, which may be monosubstituted by A, CH2COHet3 or COOA,Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, COOR5 and/or CON(R5)2,Het denotes a monocyclic aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A and/or [C(R5)2]nOR5,Het2 denotes a monocyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by A, OR5 and/or ═O,Het3 denotes an unsubstituted monocyclic saturated heterocycle having 1 to 4 N, and/or O and/or S atoms,A denotes unbranched or branched alkyl having 1-6 C atoms, in which 1-5H atoms may be replaced by F,orcyclic alkyl having 3-7 C atomsA′ denotes unbranched or branched alkyl having 1-6 C atoms, orcyclic alkyl having 3-7 C atoms,Hal denotes F, Cl, Br or I,m denotes 0, 1 or 2,n denotes 0, 1, 2, 3 or 4,and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • 8. Compounds according to claim 1
  • 9. Medicaments comprising at least one compound according to claim 1 and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants.
  • 10. A method for the treatment of tumours, tumour growth, tumour metastases and/or AIDS, comprising administering to a host in need thereof an effective amount of a compound of the formula I and/or pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof.
  • 11. The method according to claim 10, where the tumour originates from the group of tumours of the squamous epithelium, the bladder, the stomach, the kidneys, of head and neck, the oesophagus, the cervix, the thyroid, the intestine, the liver, the brain, the prostate, the urogenital tract, the lymphatic system, the stomach, the larynx and/or the lung.
  • 12. The method according to claim 10, where the tumour originates from the group monocytic leukaemia, lung adenocarcinoma, small-cell lung carcinomas, pancreatic cancer, colon carcinoma, glioblastomas and/or breast carcinoma.
  • 13. The method according to claim 10, where the tumour is a tumour of the blood and immune system.
  • 14. The method according to claim 10, where the tumour originates from the group of acute myeloid leukaemia, chronic myeloid leukaemia, acute lymphatic leukaemia and/or chronic lymphatic leukaemia.
  • 15. A method for the treatment of tumours, comprising administering to a host in need thereof of a therapeutically effective amount of a compound of the formula I according to claim 1 in combination with 1) oestrogen receptor modulator, 2) androgen receptor modulator, 3) retinoid receptor modulator, 4) cytotoxic agent, 5) antiproliferative agent, 6) prenyl-protein transferase inhibitor, 7) HMG-CoA reductase inhibitor, 8) HIV protease inhibitor, 9) reverse transcriptase inhibitor or 10) further angiogenesis inhibitors.
  • 16. A method for the treatment of tumours, comprising administering to a host in need thereof of a therapeutically effective amount of a compound of the formula I according to claim 1 is administered in combination with radiotherapy and 1) oestrogen receptor modulator, 2) androgen receptor modulator, 3) retinoid receptor modulator, 4) cytotoxic agent, 5) antiproliferative agent, 6) prenyl-protein transferase inhibitor, 7) HMG-CoA reductase inhibitor, 8) HIV protease inhibitor, 9) reverse transcriptase inhibitor or 10) further angiogenesis inhibitors.
Priority Claims (1)
Number Date Country Kind
102010053347.5 Dec 2010 DE national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP11/05805 11/17/2011 WO 00 5/30/2013