Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THIENOPYRIMIDINES CONTAINING A SUBSTITUTED ALKYL GROUP FOR PHARMACEUTICAL COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2011/104340
Kind Code:
A1
Abstract:
The present invention relates to novel thienopyhmidine compounds of general formula pharmaceutical compositions comprising these compounds and their therapeutic use for the prophylaxis and/or treatment of diseases which can be influenced by the inhibition of the kinase activity of Mnk1 and/or Mnk2 (Mnk2a or Mnk2b) and/or variants thereof.

Inventors:
HECKEL ARMIN (DE)
HIMMELSBACH FRANK (DE)
KLEY JOERG (DE)
LEHMANN-LINTZ THORSTEN (DE)
REDEMANN NORBERT (DE)
SAUER ACHIM (DE)
THOMAS LEO (DE)
WIEDENMAYER DIETER (DE)
BLACK PHILLIP (GB)
BLACKABY WESLEY (GB)
LINNEY IAN (GB)
AUSTEN MATTHIAS (DE)
DANILEWICZ JOHN (GB)
SCHNEIDER MARTIN (DE)
SCHREITER KAY (DE)
Application Number:
PCT/EP2011/052813
Publication Date:
September 01, 2011
Filing Date:
February 25, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
HECKEL ARMIN (DE)
HIMMELSBACH FRANK (DE)
KLEY JOERG (DE)
LEHMANN-LINTZ THORSTEN (DE)
REDEMANN NORBERT (DE)
SAUER ACHIM (DE)
THOMAS LEO (DE)
WIEDENMAYER DIETER (DE)
BLACK PHILLIP (GB)
BLACKABY WESLEY (GB)
LINNEY IAN (GB)
AUSTEN MATTHIAS (DE)
DANILEWICZ JOHN (GB)
SCHNEIDER MARTIN (DE)
SCHREITER KAY (DE)
International Classes:
C07D495/04; A61K31/381; A61P9/00
Domestic Patent References:
WO2006136402A12006-12-28
WO2007059905A22007-05-31
WO2010023181A12010-03-04
WO2003037362A22003-05-08
WO2003003762A22003-01-09
WO2002103361A12002-12-27
WO2005000385A12005-01-06
WO2006066937A22006-06-29
WO2006136402A12006-12-28
WO2007115822A12007-10-18
WO2008006547A22008-01-17
WO2007115822A12007-10-18
Other References:
LADA, DIABETES CARE, vol. 8, 2001, pages 1460 - 1467
DIABETES, vol. 48, 1999, pages 1836 - 1841
JAMA, vol. 288, 2002, pages 2209 - 2716
J. CELL SCI., vol. 110, no. 2, 1997, pages 209 - 219
GENETICS, vol. 156, no. 3, 2000, pages 1219 - 1230
EMBO J., vol. 16, 1997, pages 1909 - 1920
MOL CELL BIOL, vol. 19, 1990, pages 1871 - 1880
MOL CELL BIOL, vol. 21, 2001, pages 743 - 754
MOL CELL BIOL, vol. 24, 2004, pages 6539 - 6549
MOL CELL BIOL, vol. 22, 2001, pages 5500 - 5511
BBA, vol. 1488, 2000, pages 124 - 138
FUKUNAGA; HUNTER, EMBO J, vol. 16, no. 8, 1997, pages 1921 - 1933
UEDA ET AL., MOL CELL BIOL, vol. 24, no. 15, 2004, pages 6539 - 6549
NIKOLCHEVA ET AL., J CLIN INVEST, vol. 110, 2002, pages 119 - 126
CONTI; DIGIOACCHINO, ALLERGY ASTHMA PROC, vol. 22, no. 3, 2001, pages 133 - 7
BUXADE ET AL., IMMUNITY, vol. 23, August 2005 (2005-08-01), pages 177 - 189
KHABAR, J, INTERF CYTOKINE RES, vol. 25, 2005, pages 1 - 10
UEDA ET AL., MOL CELL BIOL., vol. 24, no. 15, 2004, pages 6539 - 49
RUGGERO ET AL., NAT MED., vol. 10, no. 5, May 2004 (2004-05-01), pages 484 - 6
CULJKOVIC ET AL., J CELL BIOL., vol. 175, no. 3, 2006, pages 415 - 26
WENDEL ET AL., GENES DEV., vol. 21, no. 24, 2007, pages 3232 - 7
WENDEL ET AL., NATURE, vol. 428, no. 6980, 2004, pages 332 - 7
GRAFF, CANCER RES., vol. 68, no. 3, 2008, pages 631 - 4
DE BENEDETTI; GRAFF, ONCOGENE, vol. 23, no. 18, 2004, pages 3189 - 99
BARNHART; SIMON, J CLIN INVEST., vol. 117, no. 9, 2007, pages 2385 - 8
GRAFF ET AL., J CLIN INVEST., vol. 117, no. 9, 2007, pages 2638 - 48
TOPISIROVIC ET AL., CANCER RES., vol. 64, no. 23, 2004, pages 8639 - 42
MOL. CELL. BIOL., vol. 21, 2001, pages 5500
MOL CELL BIOL RES COMM, vol. 3, 2000, pages 205
GENOMICS, vol. 69, 2000, pages 63
GORDON; BREACH: "Design and Applications of Prodrugs", 1991
"Desiqn of Prodruqs", 1985, ELSEVIER
"Prodruqs: Topical and Ocular Druq Delivery", 1998, MARCEL DEKKER
"Methods in Enzvmoloqv", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Burqer's Medicinal Chemistry and Druq Discovery", vol. 1, 1995, JOHN WILEY & SONS, pages: 172 - 178,949-
"Pro-Druqs as Novel Delivery Systems", 1975, AM. CHEM. SOC.
"Bioreversible Carriers in Druq Desiqn", 1987, ELSEVIER
"Remington's Pharmaceutical Sciences", 1991, MACK PUBLISHING CO.
KNAUF ET AL., MOL CELL BIOL., vol. 21, no. 16, August 2001 (2001-08-01), pages 5500 - 11
SCHEPER ET AL., MOL CELL BIOL., vol. 21, no. 3, February 2001 (2001-02-01), pages 743 - 54
Attorney, Agent or Firm:
HAMMANN, Heinz et al. (Corporate PatentsBinger Str. 173, Ingelheim Am Rhein, DE)
Download PDF:
Claims:
Claims

1 . A compound of general formula

wherein X is CH or N,

R1 is a hydrogen or halogen atom or CN or an Ch alky! or CONH2 group,

R2 is a straight-chained or branched Ci-6 alkyl group which is independently substituted with one or two fluorine atoms, or one or two trifluoromethyl,

tetrahydropyranyl, cydopropyl, H2N-CO-, R5NHCO- or (R5)2N-CO- groups, wherein the above-mentioned cydopropyl group may be substituted with one or two F or -CH2-CN, and wherein the two R5 groups together with the N atom to which they are attached may form a 4- to 8-membered ring, in which a carbon atom may be replaced by a O, S, SO, SO2 and/or which may be substituted with OH, NH2, N(Ci-3- alkyl)2, NH(Ci-3 alkyl), CF3 orCi-3-alkyl, or a straight-chained or branched C2-6 alkyl group which is independently substituted in position 2 to 6 with one or two hydroxy, Ci-3 alkoxy, amino, CN, R5NH-, (R5)2N-, R5OCONH-, R5CONH-, R5SO2NH-, R5NHCONH- groups, wherein R5 is a Ci-5 alkyl group, preferably a Ci-4 alkyl group, more preferably Me, i-Pr or t-Bu, each optionally substituted with one CF3, NH2, NH(Ci-3 alkyl), N(Ci-3-alkyl)2 or MeO- group,

an wherein the hydrogen atoms of any of the above-mentioned NH moiety may be replaced by methyl,

R3 is a C1-2 alkyl group and

R4 is a carboxy, Ci-3 alkoxy-carbonyl, -CONH2, -CONHR7, -CONH-OR7, -CONH SO2R7 or -CO-NH-L-R6 group,

wherein L is a -(CH2)n-, -CH2-C≡C-CH2-, or

R6 is OH, -NH2, -NHR7, -N(R7)2, -NH-CO2R7 or a 3- to 6-membered cyclic amine such as pyrrolidine or piperidine, n is 2 or 3 and

R7 is C1-4 alkyl, preferably methyl, or a salt thereof.

2. A compound of formula (I) according to claim 1 , wherein

X, R1 , R2 and R4 are defined as in claim 1 and

R is methyl, or a salt thereof.

3. A compound of formula (I) according to claim 1 or 2, wherein R2 to R4 are defined as in claim 1 and X is CH and

R1 is a fluorine atom, or a salt thereof.

4. A compound of formula (I) according to claim 1 or 2, wherein R2 to R4 are defined as in claim 1 and

X is N and

R1 is a hydrogen atom, or a salt thereof.

5. A compound of formula (I) according to any one of claims 1 to 4, wherein

X, R1 , R3 and R4 are defined as in any one of claims 1 to 4 and

R2 is selected from:

(dimethylamino)-carbonyl methyl,

ethyl, 2-amino-ethyl, 1 -(trifluoromethyl)-ethyl;

isopropyl optionally substituted in position 2 with ethoxycarbonyl, amino or tert- butyloxycarbonylamino;

2,2'-diamino-isopropyl, 2,2'-difluoro-isopropyl, 2,2'-di-(ethoxy)-isopropyl, 2,2'-bis- (tert-butyloxycarbonylamino)-isopropyl, 2-[2'-(trifluoromethyl)-ethylamino]-isopropyl, 3-amino-1 -methyl-propyl, 3-(dimethylamino)-1 -methyl-propyl, 3-hydroxy-1 ,3-dimethyl-butyl, or

a fluor-containing residue such as 1 ,3-difluoropropan-2-yl, 1 ,1 ,1 -trifluoropropan-2-yl or 1 ,1 -difluoroethyl, or salt thereof.

6. A compound of formula (I) according to any one of claims 1 to 4, wherein

X, R1, R3 and R4 are defined as in any one of claims 1 to 4 and

R2 is selected from:isopropyl and isobutyl optionally substituted in position 2 or 3 with ethoxycarbonyl, amino, tert-butyloxycarbonylamino or methylsulfonylannino or a salt thereof.

7. A compound of formula (I) according to any one of claims 1 to 6, wherein

X, R1 to R3 are defined as in any one of claims 1 to 6 and

R4 is selected from:

carboxy, Ci-3 alkoxy-carbonyl, aminocarbonyl, N-(Ci-3 alkyl)-aminocarbonyl or N,N- [di-(Ci-3 alkyl)]-aminocarbonyl group, wherein the alkyl moiety of the above-mentioned N-(Ci-3 alkyl)-aminocarbonyl and N,N-[di-(Ci-3 alkyl)]-aminocarbonyl groups may optionally be terminally substituted with a hydroxy, amino, N-(Ci-3 alkyl)-amino or N,N-[di-(Ci_3 alkyl)]- amino group, or a salt thereof.

8. A compound of formula (I) according to any one of claims 1 to 6, wherein

X, R1 to R3 are defined as in any one of claims 1 to 6 and

R4 is selected from:

aminocarbonyl, N-methyl-aminocarbonyl; N-ethyl-aminocarbonyl ternninally substituted in the ethyl moiety with hydroxy or N,N- dimethylamino;

N-(n-propyl)-aminocarbonyl terminally substituted in the n-propyl moiety with N,N- dimethylamino;

carboxy or methoxycarbonyl, or a salt thereof.

9. Compound of formula (I) according to claim 1 selected from: a) Methyl 4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3- d]pyrimidine-6-carboxylate, b) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-N-5-methylthieno[2,3- d]pyrimidine-6-carboxamide, c) Methyl 4-(4-fluoro-(2-(1 -methylsulfonamido)propan-2-yloxy)phenylamino)-5- methyl-thieno[2,3-d]pyrimidine-6-carboxylate, d) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methylthieno[2,3- d) pyrimidine-6-carboxamide, e) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methy-lthieno[2,3- d]pyrimidine-6-carboxylic acid, f) Methyl 4-(2-(4-aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3- d]pyrimidine-6-carboxylate, g) Methyl 4-(4-fluoro-2-(4-(methylsulfonamido)butan-2yloxy)phenylamino)-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylate , h) 4-(2-(4-Aminobutan-2yloxy)pyridin-3ylamino)-5-methylthieno[2,3-d]pyrimidi carboxylate, i) N-(3-(Dimethylannino)propyl)-4-(4-fluoro-2-(4-hydroxy-4-nnethylpentan-2- yloxy)phenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxamide, j) 5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyridin-3-ylannino)thieno[2,3- d]pyrinnidine-6-carboxylic acid, k) 5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyridin-3-ylannino)thieno[2,3- d]pyrimidine-6-carboxamide,

I) N-Methyl- 5-methyl-4-(2-(1 ,1 ,1 tnfluoropropan-2-yloxy)pyridin-3-ylamino)thieno[2,3- d]pyrinnidine-6-N-nnethylcarboxannide, m) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-5-nnethyl-thieno[2,3- d]pyrinnidine-6-carboxannide, n) N-Methyl-4-(2-(1 ,3-difluoropropan-2-yloxy)-4-fluorophenylannino)- 5-methyl-thieno[2,3d]pyrinnidine-6-carboxannide, o) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-N-(3- (dimethylamino)propyl)-5-methyl-thieno[2,3-d]-pyrimidine-6-carboxamide, p) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-

N-(2-(dimethylannino)ethyl)-5-nnethylthieno[2,3-d] pyrinnidine-6-carboxannide, q) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-N-(2-(hydroxyethyl)-5- methylthieno[2,3-d] pyrinnidine-6-carboxannide, r) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-N-(3-(pyrrolidin-1 - yl)propyl)thieno[2,3-d]pyrinnidine-6-carboxannide, s) N-((trans)-2-Aminocyclopropyl)-4-(2-(1 ,3-difluoropropan-2-yloxy)-4- fluorophenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxamide, t) 4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-N-(2-hydroxyethyl)-5-methylthieno[2,3- d]pyrinnidine-6-carboxannide, u) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylam

carboxylic acid, v) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylam

carboxamide, w) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-N-(3-(dimethylamino)propyl)-5- methylthieno[2,3-d]pyrinnidine-6-carboxannide, x) 4-(2-(1 -(Ethylamino)-I -oxopropan-2-yloxy)-4-fluorophenylamino)-N,5- dimethylthieno[2,3-d]pyrinnidine-6-carboxannide and y) 4-(2-(1 -(Ethylamino)-I -oxopropan-2-yloxy)-4-fluorophenylamino)-N,5- dimethylthieno[2,3-d]pyhnnidine-6-carboxannide, or a salt thereof.

10. A pharnnaceutically acceptable salt of a compound according to any one of claims 1 to 9.

1 1 . Pharmaceutical composition comprising a compound according to any one of claims 1 to 9 or a salt according to claim 10 and optionally a pharmaceutically acceptable carrier.

12. Pharmaceutical composition according to claim 1 1 further comprising an additional therapeutic agent.

13. Pharmaceutical composition according to claim 12 wherein the additional therapeutic agent is selected from an antidiabetic agent, a lipid lowering agent, a cardiovascular agent, an antihypertensive agent, a diuretic agent, a thrombocyte aggregation inhibitor, an antineoplastic agent or an anti-obesity agent.

14. Compound as defined in any one of claims 1 to 9 or a salt according to claim 10 for use in inhibiting the activity of the kinase activity of Mnk1 or Mnk2 (Mnk2a, Mnk2b) or variants thereof.

15. Compound as defined in any one of claims 1 to 9 or a salt according to claim 10 for use in the prophylaxis or therapy of metabolic diseases, hematopoietic disorders, neurodegenerative diseases, kidney damage, inflammatory disorders and cancer and their consecutive complications and diseases.

16. Compound according to any one of claims 1 to 9 or a salt according to claim 10 for use in the prophylaxis or therapy of metabolic diseases of the carbohydrate and/or lipid metabolism and their consecutive complications and disorders.

17. Compound according to claim any one of claims 1 to 9 or a salt according to claim 10 for use in the prophylaxis or therapy of diabetes.

18. Compound according to any one of claims 14 to 17 for concomitant or sequential administration to a patient in combination with an additional therapeutic agent.

19. Compound as defined in any of claims 1 to 9 or a salt according to claim 10 for use in treating or preventing cytokine related disorders.

20. Compound according to claim 19 for concomitant or sequential administration to a patient in combination with an additional therapeutic agent.

21 . Compound according to claim 20, wherein the additional therapeutic agent is selected from a histamine antagonist, a bradikinin antagonist, serotonin antagonist, leukotriene, an anti-asthmatic, an NSAID, an antipyretic, a corticosteroid, an antibiotic, an analgetic, a uricosuric agent, chemotherapeutic agent, an anti gout agent, a bronchodilator, a cyclooxygenase-2 inhibitor, a steroid, a 5-lipoxygenase inhibitor, an immunosuppressive agent, a leukotriene antagonist, a cytostatic agent, an antineoplastic agent, a mTor inhibitor, a Tyrosine kinase inhibitor, antibodies or fragments thereof against cytokines and soluble parts (fragments) of cytokine receptors.

Description:
Thienopyrimidines containing a substituted alkyl group for pharmaceutical compositions

The present invention relates to thienopyrimidine compounds and to novel pharmaceutical compositions comprising thienopyrimidine compounds.

Moreover, the present invention relates to the use of the thienopyrimidine compounds of the invention for the production of pharmaceutical compositions for the prophylaxis and/or treatment of diseases which can be influenced by the inhibition of the kinase activity of Mnk1 (Mnkl a or MnKl b) and/or Mnk2 (Mnk2a or Mnk2b) or further variants thereof. Particularly, the present invention relates to the use of the thienopyrimidine compounds of the invention for the production of pharmaceutical compositions for the prophylaxis and/or therapy of metabolic diseases, such as diabetes, hyperlipidemia and obesity, hematopoietic disorders, neurodegenerative diseases, kidney damage, inflammatory disorders, and cancer and their consecutive complications and disorders associated therewith.

Metabolic diseases are diseases caused by an abnormal metabolic process and may either be congenital due to an inherited enzyme abnormality or acquired due to a disease of an endocrine organ or failure of a metabolically important organ such as the liver or the pancreas.

The present invention is more particularly directed to the treatment and/or prophylaxis of in particular metabolic diseases of the lipid and carbohydrate metabolism and the consecutive complications and disorders associated therewith.

Lipid disorders cover a group of conditions which cause abnormalities in the level and metabolism of plasma lipids and lipoproteins. Thus, hyperlipidemias are of particular clinical relevance since they constitute an important risk factor for the development of atherosclerosis and subsequent vascular diseases such as coronary heart disease.

Diabetes mellitus is defined as a chronic hyperglycemia associated with resulting damages to organs and dysfunctions of metabolic processes. Depending on its etiology, one differentiates between several forms of diabetes, which are either due to an absolute (lacking or decreased insulin secretion) or to a relative lack of insulin. Diabetes mellitus Type I (IDDM, insulin-dependent d iabetes mell itus) generally occurs in adolescents under 20 years of age. It is assumed to be of auto-immune etiology, leading to an insulitis with the subsequent destruction of the beta cells of the islets of Langerhans which are responsible for the insulin synthesis. In addition, in latent autoimmune diabetes in adults (LADA; Diabetes Care. 8: 1460-1467, 2001 ) beta cells are being destroyed due to autoimmune attack. The amount of insulin produced by the remaining pancreatic islet cells is too low, resulting in elevated blood glucose levels (hyperglycemia). Diabetes mellitus Type II generally occurs at an older age. It is above all associated with a resistance to insulin in the liver and the skeletal muscles, but also with a defect of the islets of Langerhans. High blood glucose levels (and also high blood l ipid levels) in turn lead to an impairment of beta cell function and to an increase in beta cell apoptosis.

Diabetes is a very disabling disease, because today's common anti-diabetic drugs do not control blood sugar levels well enough to completely prevent the occurrence of high and low blood sugar levels. Out of range blood sugar levels are toxic and cause long-term complications for example retinopathy, Tenopathy, neuropathy and peripheral vascular disease. There is also a host of related conditions, such as obesity, hypertension , heart disease and hyperl ipidemia, for wh ich persons with diabetes are substantially at risk.

Obesity is associated with an increased risk of follow-up diseases such as cardiovascular diseases, hypertension, diabetes, hyperlipidemia and an increased mortality. Diabetes (insulin resistance) and obesity are part of the "metabolic syndrome" which is defined as the linkage between several diseases (also referred to as syndrome X, insulin-resistance syndrome, or deadly quartet). These often occur in the same patients and are major risk factors for development of diabetes type II and cardiovascular disease. It has been suggested that the control of lipid levels and glucose levels is required to treat diabetes type II, heart disease, and other occurrences of metabolic syndrome (see e.g., Diabetes 48: 1836-1841 , 1999; JAMA 288: 2209-2716, 2002). In one embodiment of the present invention the compounds and compositions of the present invention are useful for the treatment and/or prophylaxis of metabolic diseases of the carbohydrate metabolism and their consecutive complications and disorders such as impaired glucose tolerance, diabetes (preferably diabetes type II), diabetic complications such as diabetic gangrene, diabetic arthropathy, diabetic osteopenia, diabetic glomerosclerosis, diabetic nephropathy, diabetic dermopathy, diabetic neuropathy, diabetic cataract and diabetic retinopathy, diabetic maculopathy, d iabetic feet syndrome, d iabetic coma with or without ketoacidosis, d iabetic hyperosmolar coma, hypoglycemic coma, hyperglycemic coma, diabetic acidosis, diabetic ketoacidosis, intracapillary glomerulonephrosis, Kimmelstiel-Wilson synd rome, d iabetic amyotrophy, d iabetic autonom ic neu ropathy, d iabetic mononeuropathy, diabetic polyneuropathy, diabetic angiopathies, diabetic peripheral angiopathy, diabetic ulcer, diabetic arthropathy, or obesity in diabetes.

In a further embodiment the compounds and compositions of the present invention are useful for the treatment and/or prophylaxis of metabolic diseases of the lipid metabolism (i.e. lipid disorders) and their consecutive complications and disorders such as hypercholesterolemia, familial hypercholesterolemia, Fredrickson's hyperlipoproteinemia, hyperbetalipoproteinemia, hyperlipidemia, low-density- lipoprotein-type [LDL] hyperlipoproteinemia, pure hyperglyceridemia, endogenous hyperglyceridemia, isolated hypercholesterolemia, isolated hypertroglyceridemia, cardiovascular diseases such as hypertension, ischemia, varicose veins, retinal vein occlusion, atherosclerosis, angina pectoris, myocardial infarction, stenocardia, pulmonary hypertension, congestive heart failure, glomerulopaty, tubulointestitial disorders, renal failure, angiostenosis, or cerebrovascular disorders, such as cerebral apoplexy.

In a further embodiment of the present invention the compounds and compositions of the present invention are useful for the treatment and/or prophylaxis of hematopoetic disorders and their consecutive complications and disorders such as acute myeloid leukemia (AML), Morbus Hodgkin, Non-Hodgkin's lymphoma; hematopoetic disease, acu te no n-lymphocytic leukemia (ANLL), myeloproliferative disease acute promyelocytic leukemia (APL), acute myelomonocytic leukemia (AMMoL), multiple myeloma, polycythemia vera, lymphoma, acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CCL), Wilm's tumor, or Ewing's Sarcoma.

In a further embodiment of the present invention the compounds and compositions of the present invention are useful for the treatment and/or prophylaxis of cancer and consecutive complications and disorders such as cancer of the upper gastrointestinal tract, pancreatic carcinoma, breast cancer, colon cancer, ovarian carcinoma, cervix carcinoma, endometrial cancer, brain tumor, testicular cancer, laryngeal carcinoma, osteocarcinoma, prostatic cancer, retinoblastoma, liver carcinoma, lung cancer, neuroblastoma, renal carcinoma, thyroid carcinoma, esophageal cancer, soft tissue sarcoma, skin cancer, osteosarcoma, rhabdomyosarcoma, bladder cancer, metastatic cancer, cachexia, or pain.

Certain anti-cancer drugs such as cisplatin are linked to serious side effects such as nephrotoxicity or ototoxicity, which can be dose limiting. Activation of Mnks has been linked to these side effects. In a further embodiment of the present invention, the compounds and compositions of the present invention are useful for the treatment and/or prophylaxis of ear or kidney damage, in particular for the prevention or treatment of ear and kidney drug induced damage

Furthermore, the present invention relates to the use of thienopyrimidine compounds for the production of pharmaceutical compositions for the prophylaxis and/or therapy of cytokine related diseases. Such diseases are i.a. inflammatory diseases, autoimmune diseases, destructive bone disorders, proliferative disorders, infectious diseases, neurodegenerative diseases, allergies, or other conditions associated with proinflammatory cytokines.

Allergic and inflammatory diseases such as acute or chronic inflammation, chronic inflammatory arthritis, rheumatoid arthritis, psoriasis, COPD, inflammatory bowel disease, asthma and septic shock and their consecutive complications and disorders associated therewith. Inflammatory diseases like rheumatoid arthritis, inflammatory lung diseases like COPD, inflammatory bowel disease and psoriasis afflict one in three people in the course of their lives. Not only do those diseases impose immense health care costs, but also they are often crippling and debilitating.

Although inflammation is the unifying pathogenic process of these inflammatory diseases below, the current treatment approach is complex and is generally specific for any one disease. Many of the current therapies available today only treat the symptoms of the disease and not the underlying cause of inflammation.

The compositions of the present invention are useful for the treatment and/or prophylaxis of inflammatory diseases and consecutive complications and disorders, such as chronic or acute inflammation, inflammation of the joints such as chronic inflammatory arthritis, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, juvenile rheumatoid arthritis, Reiter's syndrome, rheumatoid traumatic arthritis, rubella arthritis, acute synovitis and gouty arthritis; inflammatory skin diseases such as sunburn, psoriasis, erythrodermic psoriasis, pustular psoriasis, eczema, dermatitis, acute or chronic graft formation, atopic dermatitis, contact dermatitis, urticaria and scleroderma; inflammation of the gastrointestinal tract such as inflammatory bowel disease, Crohn ' s disease and related conditions, ulcerative colitis, colitis, and diverticulitis; nephritis, urethritis, salpingitis, oophoritis, endomyometritis, spondylitis, systemic lupus erythematosus and related disorders, multiple sclerosis, asthma, meningitis, myelitis, encephalomyelitis, encephalitis, phlebitis, thrombophlebitis, respiratory diseases such as asthma, bronchitis, chronic obstructive pulmonary disease (COPD), inflammatory lung disease and adult respiratory distress syndrome, and allergic rhinitis; endocarditis, osteomyelitis, rheumatic fever, rheumatic pericarditis, rheumatic endocarditis, rheumatic myocarditis, rheumatic mitral valve disease, rheumatic aortic valve disease, prostatitis, prostatocystitis, spondoarthropathies ankylosing spondylitis, synovitis, tenosynovotis, myositis, pharyngitis, polymyalgia rheumatica, shoulder tendonitis or bursitis, gout, pseudo gout, vasculitides, inflammatory diseases of the thyroid selected from granulomatous thyroiditis, lymphocytic thyroiditis, invasive fibrous thyroiditis, acute thyroiditis; Hashimoto ' s thyroiditis, Kawasaki ' s disease, Raynaud ' s phenomenon, Sjogren ' s syndrome, neuroinflammatory disease, sepsis, conjunctivitis, keratitis, iridocyclitis, optic neuritis, otitis, lymphoadenitis, nasopaharingitis, sinusitis, pharyngitis, tonsillitis, laryngitis, epiglottitis, bronchitis, pneumonitis, stomatitis, gingivitis, oesophagitis, gastritis, peritonitis, hepatitis, cholelithiasis, cholecystitis, glomerulonephritis, goodpasture ' s disease, crescentic glomerulonephritis, pancreatitis, endomyometritis, myometritis, metritis, cervicitis, endocervicitis, exocervicitis, parametritis, tuberculosis, vaginitis, vulvitis, silicosis, sarcoidosis, pneumoconiosis, pyresis, inflammatory polyarthropathies, psoriatric arthropathies, intestinal fibrosis, bronchiectasis and enteropathic arthropathies.

Moreover, cytokines are also believed to be implicated in the production and development of various cardiovascular and cerebrovascular disorders such as congestive heart disease, myocardial infarction, the formation of atherosclerotic plaques, hypertension, platelet aggregation, angina, stroke, Alzheimer's disease, reperfusion injury, vascular injury including restenosis and peripheral vascular d isease, and , for example, various d isorders of bone metabol ism such as osteoporosis (including senile and postmenopausal osteoporosis), Paget's disease, bone metastases, hypercalcaemia, hyperparathyroidism, osteosclerosis, osteoporosis and periodontitis, and the abnormal changes in bone metabolism which may accompany rheumatoid arthritis and osteoarthritis.

Excessive cytokine production has also been implicated in mediating certain complications of bacterial, fungal and/or viral infections such as endotoxic shock, septic shock and toxic shock syndrome and in mediating certain complications of CNS surgery or injury such as neurotrauma and ischaemic stroke.

Excessive cytokine production has, moreover, been implicated in mediating or exacerbating the development of diseases involving cartilage or muscle resorption, pulmonary fibrosis, cirrhosis, renal fibrosis, the cachexia found in certain chronic diseases such as malignant disease and acquired immune deficiency syndrome (AIDS), tumour invasiveness and tumour metastasis and multiple sclerosis. The treatment and/or prophylaxis of these diseases are also contemplated by the present invention Additionally, the inventive compositions may be used to treat inflammation associated with autoimmune diseases including, but not limited to, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), glomerulonephritis, rheumatoid arthritis scleroderma, chronic thyroiditis, Graves' disease, autoimmune gastritis, diabetes, autoimmune hemolytic anemia, glomerulonephritis, rheumatoid arthritis autoimmune neutropen ia, th rombocytopen ia, atopic dermatitis, ch ron ic active hepatitis, myasthenia gravis, multiple sclerosis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, psoriasis, and graft vs. host disease.

In a further embodiment the compositions of the present invention may be used for the treatment and prevention of infectious diseases such as sepsis, septic shock, Shigellosis, and Helicobacter pylori and viral diseases including herpes simplex type 1 (HSV-1 ), herpes simplex type 2 (HSV-2), cytomegalovirus, Epstein-Barr, human immunodeficiency virus (HIV), acute hepatitis infection (including hepatitis A, hepatits B, and hepatitis C), HIV infection and CMV retinitis, AIDS or malignancy, malaria, mycobacterial infection and meningitis. These also include viral infections, by influenza virus, varicella-zoster virus (VZV), Epstein-Barr virus, human herpesvirus-6 (HHV-6), human herpesvirus-7 (HHV-7), human herpesvirus-8 (HHV-8), Poxvirus, Vacciniavirus, Monkeypoxvirus, pseudorabies and rhinotracheitis.

The compositions of the present invention may also be used topically in the treatment or prophylaxis of topical disease states mediated by or exacerbated by excessive cytokine prod uction , such as inflamed joints, eczema , psoriasis and other inflammatory skin conditions such as sunburn; inflammatory eye conditions including conjunctivitis; pyresis, pain and other conditions associated with inflammation.

Periodontal disease has also been implemented in cytokine production, both topically and systemically. Hence, use of compositions of the present invention to control the inflammation associated with cytokine production in such peroral diseases such as gingivitis and periodontitis is another aspect of the present invention.

Finally, the compositions of the present invention may also be used to treat or prevent neurodegenerative disease selected from Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, frontotemporal lobar dementia, spinocerebellar ataxia, dementia with Lewy bodies, cerebral ischemia or neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity or hypoxia.

In a preferred embodiment the compositions of the present invention may be used to treat or prevent a disease selected from chronic or acute inflammation, chronic inflammatory arthritis, rheumatoid arthritis, psoriasis, COPD, inflammatory bowel disease, septic shock, Crohn ' s d isease, ulcerative col itis, multiple sclerosis and asthma.

Protein kinases are important enzymes involved in the regulation of many cellular functions. The LK6-serine/threonine-kinase gene of Drosophila melanogaster was described as a short-lived kinase which can associate with microtubules (J. Cell Sci. 1997, 1 10(2): 209-219). Genetic analysis in the development of the compound eye of Drosophila suggested a role in the modulation of the RAS signal pathway (Genetics 2000 156(3): 1219-1230). The closest human homologues of Drosophila LK6-kinase are the MAP-kinase interacting kinase 2 (Mnk2, e.g. the variants Mnk2a and Mnk2b) and MAP-kinase interacting kinase 1 (Mnk1 ) and variants thereof. These kinases are mostly localized in the cytoplasm. Mnks are phosphorylated by the p42 MAP kinases Erk1 and Erk2 and the p38-MAP kinases. This phosphorylation is triggered in a response to growth factors, phorbol esters and oncogenes such as Ras and Mos, and by stress signal ing molecu les and cytokines. The phosphorylation of Mn k proteins stimulates their kinase activity towards eukaryotic initiation factor 4E (elF4E) (EMBO J. 16: 1909-1920, 1997; Mol Cell Biol 19, 1871 -1880, 1990; Mol Cell Biol 21 , 743-754, 2001 ). Simultaneous disruption of both, the Mnk1 and Mnk2 gene in mice diminishes basal and stimulated elF4E phosphorylation (Mol Cell Biol 24, 6539-6549, 2004). Phosphorylation of elF4E results in a regulation of the protein translation (Mol Cell Biol 22: 5500-551 1 , 2001 ).

There are different hypotheses describing the mode of the stimulation of the protein translation by Mnk proteins. Most publications describe a positive stimulatory effect on the cap-dependent protein translation upon activation of MAP kinase-interacting kinases. Thus, the activation of Mnk proteins can lead to an indirect stimulation or regulation of the protein translation, e.g. by the effect on the cytosolic phospholipase 2 alpha (BBA 1488:124-138, 2000).

WO 03/037362 discloses a link between human Mnk genes, particularly the variants of the human Mnk2 genes, and diseases which are associated with the regulation of body weight or thermogenesis. It is postulated that human Mnk genes, particularly the Mnk2 variants are involved in diseases such as e.g. metabolic diseases including obesity, eating disorders, cachexia, diabetes mellitus, hypertension, coronary heart disease, hypercholesterolemia, dyslipidemia, osteoarthritis, biliary stones, cancer of the genitals and sleep apnea, and in diseases connected with the ROS defense, such as e.g. diabetes mellitus and cancer. WO 03/03762 moreover discloses the use of nucleic acid sequences of the MAP kinase-interacting kinase (Mnk) gene family and amino acid sequences encoding these and the use of these sequences or of effectors of Mnk nucleic acids or polypeptides, particularly Mnk inhibitors and activators in the diagnosis, prophylaxis or therapy of diseases associated with the regulation of body weight or thermogenesis. WO 02/103361 describes the use of kinases 2a and 2b (Mnk2a and Mnk2b) i nteracti ng with th e h u m an MAP ki n ase i n assays for th e id entification of pharmacologically active ingredients, particularly useful for the treatment of diabetes mellitus type 2. Moreover, WO 02/103361 discloses also the prophylaxis and/or therapy of d iseases associated with insu l in resistance, by mod u lation of the expression or the activity of Mnk2a or Mnk2b. Apart from peptides, peptidomimetics, am ino acids, amino acid analogues, polynucleotides, polynucleotide analogues, nucleotides and nucleotide analogues, 4-hydroxybenzoic acid methyl ester are described as a substance which binds the human Mnk2 protein. First evidence for a role of M n ks i n i nfl am m ation was provided by stud ies demonstrating activation of Mnk1 by proinflammatory stimuli. The cytokines TNFa and I L-1 β trigger the activation of Mnk1 in vitro (Fu kunaga and Hunter, EMBO J 1 6(8): 1 921 -1933, 1997) and ind uce the phosphorylation of the Mnk-specific substrate elF4E in vivo (Ueda et al ., Mol Cell Biol 24(15): 6539-6549, 2004). In addition, administration of lipopolysaccharide (LPS), a potent stimulant of the inflammatory response, induces activation of Mnk1 and Mnk2 in mice, concomitant with a phosphorylation of their substrate elF4E (Ueda et al., Mol Cell Biol 24(15): 6539-6549, 2004).

Furthermore, Mnk1 has been shown to be involved in regulating the production of proinflammatory cytokines. Mnk1 enhances expression of the chemokine RANTES (Nikolcheva et al ., J Clin Invest 1 10, 1 19-126, 2002). RANTES is a potent chemotractant of monocytes, eosinoph ils, basophiles and , natural killer cells. It activates and induces proliferation of T lymphocytes, mediates degranulation of basophils and induces the respiratory burst in eosinophils (Conti and DiGioacchino, Allergy Asthma Proc 22(3):133-7, 2001 ) WO 2005/00385 and Buxade et al ., Immunity 23: 177-1 89, August 2005 both disclose a link between Mnks and the control of TN Fa biosynthesis. The proposed mechanism is mediated by a regulatory AU-rich element (ARE) in the TNFa mRNA. Buxade et al . demonstrate proteins bind ing and controll ing ARE function to be phosphorylated by Mnk1 and Mnk2. Specifically Mn k-mediated phosphorylation of the ARE-binding protein hnRN P A1 has been suggested to enhance translation of the TNFa mRNA.

TNFa is not the only cytokine regulated by an ARE. Functional AREs are also found in the transcripts of several interleukins, interferones and chemokines (Khabar, J Interf Cytokine Res 25: 1 -1 0, 2005). The Mn k-mediated phosphorylation of ARE- binding proteins has thus the potential to control biosynthesis of cytokines in addition to that of TNFa.

Current evidence demonstrates Mnks as down stream targets of inflammatory signall ing as well as mediators of the inflammatory response. Their involvement in the production of TNFa, RANTES, and potentially additional cytokines suggests inhibition of Mnks as strategy for anti-inflammatory therapeutic intervention. Mnk1 and Mnk2 (including all splice forms) phosphorylate the translation factor elF4E on Serine 209. Mnk1 /2 double knockout mice completely lack phosphorylation on Serine 209, indicating that Mnk kinase are the only kinases able to phosphorylate this site in vivo (Ueda et al ., Mol Cell Biol . 2004; 24(15):6539-49). elF4E is overexpressed in a wide range of human malignancies, and high elF4E expression is frequently associated with more aggressive disease and poor prognosis. Furthermore, elF4E can act as an oncogene when assayed in standard assays for oncogenic activity (e.g. Ruggero et al ., Nat Med. 2004 May;10(5):484-6). elF4E excerts its oncogenic activity by stimulating the translation of oncogenes such as c- myc and cyclinDI (Culjkovic et al ., J Cell Biol . 2006; 175(3):415-26), by increasing the expression of pro-survival factors such as MCP-1 (Wendel et al ., Genes Dev. 2007; 21 (24):3232-7) and by positively regulating pathways of drug resistance (Wendel et al ., Nature 2004; 428(6980):332-7; Graff et el ., Cancer Res. 2008; 68(3):631 -4; De Benedetti and Graff, Oncogene 2004; 23(18):3189-99; Barnhart and Simon, J Clin Invest. 2007; 1 17(9):2385-8). Suppression of elF4E expression by antisense oligonucleotides has shown promise in preclinical experiments with human tumor cells (Graff et al., J Clin Invest. 2007; 1 17(9):2638-48). It has been shown that phosphorylation on Ser209 is strictly required for the oncogenic activity of elF4E in vitro and in vivo (Topisirovic et al ., Cancer Res. 2004; 64(23):8639-42; Wendel et al ., Genes Dev. 2007; 21 (24):3232-7). Thus, inhibition of Mnk1 and Mnk2 is expected to have beneficial effects in human malignancies.

Inhibitors of Mnk (referred to as CGP57380 and CGP052088) have been described (cf. Mol. Cell . Biol . 21 , 5500, 2001 ; Mol Cell Biol Res Comm 3, 205, 2000; Genomics 69, 63, 2000). CGP052088 is a staurosporine derivative having an IC 5 o of 70 nM for inhibition of in vitro kinase activity of Mnk1 . CGP57380 is a low molecular weight selective, non-cytotoxic inhibitor of Mnk2 (Mnk2a or Mnk2b) or of Mnk1 : The addition of CGP57380 to cell culture cells, transfected with Mnk2 (Mnk2a or Mnk2b) or Mnk1 showed a strong reduction of phosphorylated elF4E. Further inhibitors of Mnk have been described. See for example Applicants patent applications WO 06/066937, describing pyrazolopyrimidine compounds, WO 06/136402 describing certain thienopyrimidine compounds, WO 07/1 15822 describing further thienopy midine compounds with modified core ring, and WO 08/006547 describing pyrrolopyrimidines as inhibitors of Mnk kinases.

The problem underlying the present invention is to provide potent and selective Mnk1 and/or Mnk2 inhibitors which may effectively and safely be used for the treatment of metabolic diseases, inflammatory diseases, cancer, neurodegenerative diseases and their consecutive complication and disorders.

It has now been surprisingly found that certain thienopyhmidine compounds are potent inhibitors of the kinase enzymes Mnk1 and/or Mnk2 and/or variants thereof and as such may be useful in the prophylaxis and/or therapy of diseases which can be influenced by the inhibition of the kinase activity of Mnk1 and/or Mnk2 (Mnk2a or Mnk2b) and/or variants thereof. In contrast to the thienopyhmidine compounds known in the art, for example, the compoonds disclosed in the Applicants patent applications WO 06/136402 and WO 2007/1 15822, the thienopyhmidine compounds of the present invention provide several advantages, namely, enhanced solubility, the possibility to form stable salts, improved metabolic stability, improved pharmacokinetic properties, enhanced or retained activity in biochemical or cellular Mnk activity assays and enhanced or retained selectivity against other kinases.

The thienopyhmidine compounds disclosed in WO 06/136402 and WO 07/1 15822 exhibit high activity in Mnk enzyme assays and extremely high selectivity, however they show a very low solubility and are in most cases metabolic unstable resulting in undesired pharmacokinetic properties.

It has been surprisingly found that by the introduction of a polar group at the imposition in the compounds of general formula (I) below leads to surprising substantial metabolic stabilization, rendering the thienopyrimidines of the present invention useful for in vivo pharmacological applications.

Moreover, compounds described in this application also show improved solubility, have strong inhibitory potency in biochemical and cellular assays and are highly selective, resulting in overall greatly improved pharmacological properties.

If not specified otherwise, any alkyl moiety mentioned in this application may be straight-chained or branched.

Thienopyrimidine compounds of the present invention are compounds of the general formula (I):

wherein

X is CH or N,

R 1 is a hydrogen or halogen atom or CN or an C1-3 alkyl or CONH 2 group,

R 2 is a straight-chained or branched Ci-6 alkyl group which is independently

substituted with one or two fluorine atoms, or one or two trifluoromethyl,

tetrahydropyranyl, cyclopropyl, H 2 N-CO-, R 5 NHCO- or (R 5 ) 2 N-CO- groups, wherein the above-mentioned cyclopropyl group may be substituted with one or two F or -CH 2 -CN and wherein the two R5 groups together with the N atom to which they are attached may form a 4- to 8-membered ring, in which a carbon atom may be replaced by a O, S, SO, SO 2 and/or which may be substituted with OH, NH 2 , N(Ci-3-alkyl) 2> NH(Ci -3 alkyl), CF 3 or Ci -3 -alkyl, or a straight-chained or branched C2-6 alkyl group which is independently substituted in position 2 to 6 with one or two hydroxy, Ci-3 alkoxy, amino, CN, R 5 NH-, (R 5 )2N-, R 5 OCONH-, R 5 CONH-, R 5 SO 2 NH-, R 5 NHCONH- groups, wherein R 5 is a Ci -5 alkyl group, preferably a Ci -4 alkyl group, more preferably Me, i-Pr or t-Bu, each optionally substituted with one CF 3 , NH 2 , NH(Ci-3 alkyl), N(Ci-3-alkyl) 2 or MeO- group,

and wherein the hydrogen atoms of any of the above-mentioned NH moiety may be replaced by methyl,

R 3 is a Ci-2 alkyl group and

R 4 is a carboxy, Ci -3 alkoxy-carbonyl, -CONH 2, -CONHR 7 , -CONH-OR 7 , -CONH- SO 2 R 7 or -CO-NH-L-R 6 group,

wherein L is a -(CH 2 ) n -, -CH 2 -C≡C-CH 2 -, or

R 6 is OH, -NH 2 , -NHR 7 , -N(R 7 ) 2 , -NH-CO 2 R 7 or a 3- to 6-membered cyclic amine such as pyrrolidine or piperidine, n is 2 or 3 and

R 7 is Ci-4 alkyl, preferably methyl, or a tautomer, enantiomer or salt thereof.

Preferred compounds of formula (I) are those, wherein X, R 1 , R 2 and R 4 are as defined above and R 3 is methyl, or a tautomer or salt thereof. A preferred subgroup concerns those compound of formula (I), wherein R 2 to R 4 are as defined as above,

X is CH and R 1 is a fluorine atom, or a tautomer or salt thereof.

Another preferred subgroup concerns those compounds of formula (I), wherein R 2 to R 4 are as defined above,

X is N and

R 1 is a hydrogen atom, or a tautomer or salt thereof.

A third preferred subgroup concerns those compounds of formula (I), wherein X, R 1 , R 3 and R 4 are as defined above and R 2 is selected from:

(dimethylamino)-carbonyl methyl,

ethyl, 2-amino-ethyl, 1 -(trifluoromethyl)-ethyl;

isopropyl optionally substituted in position 2 with ethoxycarbonyl, amino or tert- butyloxycarbonylamino;

2,2 ' -diamino-isopropyl, 2,2 ' -difluoro-isopropyl, 2,2 ' -di-(ethoxy)-isopropyl, 2,2 ' -bis- (tert-butyloxycarbonylamino)-isopropyl, 2-[2 ' -(trifluoromethyl)-ethylamino]-isopropyl, 3-amino-1 -methyl-propyl, 3-(dimethylamino)-1 -methyl-propyl,

3-hydroxy-1 ,3-dimethyl-butyl, or a fluor-containing residue such as 1 ,3-difluoropropan-2-yl, 1 ,1 ,1 -trifluoropropan-2-yl or 1 ,1 -difluoroethyl-, or a tautomer or salt thereof, particularly those compounds of formula (I), wherein

X, R 1 , R 3 and R 4 are as defined above and

R 2 is selected from:

isopropyl and isobutyl optionally substituted in position 2 or 3 with ethoxycarbonyl, amino, tert-butyloxycarbonylamino or methylsulfonylamino

or a tautomer or salt thereof.

A fourth preferred subgroup concerns those compounds of formula (I), wherein X, R 1 to R 3 are as defined above and

R 4 is selected from:

carboxy, Ci-3 alkoxy-carbonyl, aminocarbonyl, N-(Ci-3 alkyl)-aminocarbonyl or N,N- [di-(Ci-3 alkyl)]-aminocarbonyl group, wherein the alkyl moiety of the above-mentioned N-(Ci-3 alkyl)-aminocarbonyl and N,N-[di-(Ci-3 alkyl)]-aminocarbonyl groups may optionally be terminally substituted with a hydroxy, amino, N-(Ci-3 alkyl)-amino or N,N-[di-(Ci_3 alkyl)]- amino group, or a tautomer or salt thereof, particularly those compounds of formula (I), wherein

X, R 1 to R 3 are as defined above and

R 4 is selected from:

aminocarbonyl, N-methyl-aminocarbonyl;

N-ethyl-aminocarbonyl terminally substituted in the ethyl moiety with hydroxy or N,N- dimethylamino;

N-(n-propyl)-aminocarbonyl terminally substituted in the n-propyl moiety with N,N- dimethylamino; Carboxy or methoxycarbonyl, or a tautomer or salt thereof. Particularly preferred Compounds of formula (I) are:

a) Methyl 4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- d]pyrimidine-6-carboxylate, b) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-N-5-methylthieno[ 2,3- d]pyrimidine-6-carboxamide, c) Methyl 4-(4-fluoro-(2-(1 -methylsulfonamido)propan-2-yloxy)phenylamino)-5- methyl-thieno[2,3-d]pyrimidine-6-carboxylate, d) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methylthieno[2, 3- d) pyrimidine-6-carboxamide, e) 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methy-lthieno[2 ,3- d]pyrimidine-6-carboxylic acid, f) Methyl 4-(2-(4-aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- d]pyrimidine-6-carboxylate, g) Methyl 4-(4-fluoro-2-(4-(methylsulfonamido)butan-2yloxy)phenylamino )-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylate , h) 4-(2-(4-Aminobutan-2yloxy)pyridin-3ylamino)-5-methylthieno[2 ,3-d]pyrimidine-6- carboxylate, i) N-(3-(Dimethylamino)propyl)-4-(4-fluoro-2-(4-hydroxy-4-methy lpentan-2- yloxy)phenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxa mide, j) 5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyridin-3-ylannino)thieno[2,3- d]pyrimidine-6-carboxylic acid, k) 5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyridin-3-ylannino)thieno[2,3- d]pyrinnidine-6-carboxannide,

I) N-Methyl- 5-methyl-4-(2-(1 ,1 ,1 tnfluoropropan-2-yloxy)pyridin-3-ylamino)thieno[2,3- d]pyrinnidine-6-N-nnethylcarboxannide, m) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-5-nnethyl-t hieno[2,3- d]pyrinnidine-6-carboxannide, n) N-Methyl-4-(2-(1 ,3-difluoropropan-2-yloxy)-4-fluorophenylannino)- 5-methyl-thieno[2,3d]pyrinnidine-6-carboxannide, o) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-N-(3- (dimethylamino)propyl)-5-methyl-thieno[2,3-d]-pyrimidine-6-c arboxamide, p) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)- N-(2-(dimethylannino)ethyl)-5-nnethylthieno[2,3-d] pyrinnidine-6-carboxannide, q) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylannino)-N-(2-(hydro xyethyl)-5- methylthieno[2,3-d] pyrinnidine-6-carboxannide, r) 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-N-( 3-(pyrrolidin-1 - yl)propyl)thieno[2,3-d]pyrinnidine-6-carboxannide, s) N-((trans)-2-Aminocyclopropyl)-4-(2-(1 ,3-difluoropropan-2-yloxy)-4- fluorophenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxa mide, t) 4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-N-(2-hydroxyethyl)- 5-nnethylthieno[2,3- d]pyrimidine-6-carboxamide, u) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-5-methylthieno[2 ,3-d]pyrim carboxylic acid, v) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylam

carboxamide, w) 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-N-(3-(dimethylam ino)propyl)-5- methylthieno[2,3-d]pyrinnidine-6-carboxannide, x) 4-(2-(1 -(Ethylamino)-I -oxopropan-2-yloxy)-4-fluorophenylamino)-N,5- dimethylthieno[2,3-d]pyrinnidine-6-carboxannide and y) 4-(2-(1 -(Ethylamino)-I -oxopropan-2-yloxy)-4-fluorophenylamino)-N,5- dimethylthieno[2,3-d]pyhnnidine-6-carboxannide, or a pharmaceutically acceptable salt thereof.

Typical methods of preparing the compounds of the invention are described below in the experimental section.

The potent inhibitory effect of the compounds of the invention may be determined by in vitro enzyme assays as described in the Examples in more detail.

The compounds of the present invention can be synthesized according to the following synthesis schemes:

D

Compounds of the general formula C can be synthesized by reaction of a compound A with the deprotonated alcohol B in appropriate solvents such as THF or DMF at a temperature between 0 °C and 150 °C. The deprotonated form of B can be obtained by deprotonation with a base such as sodium hydride or lithium hexamethyldisilazane at a preferred temperature of 0 °C. Hydrogenation of compound C in order to obtain a compound of the general formula D can be achieved by reacting C in the presence of hydrogen and a catalyst such as palladium or Raney nickel. The hydrogen can be introduced as a gas or stem from a hydrogen source such as ammonium formate.

Compounds of the general formula C can be also obtained by Mitsunobu reaction of a compound with the general formula E with an alcohol B in the presence of triphenylphosphine and an dialkylazodicarboxylate such as diethylazodicarboxylate, diisopropylazodicarboxylate or di-tert.butylazodiacarboxylate in a solvent such as THF at temperatures between -10 °C and 80 °C, preferrably between 0 °C and 30 °C.

A compound of the formula G can be synthesized by reaction of compound D with F preferably in the presence of an acid such as p-toluene sulfonic acid or hydrochloric acid in solvents such as dioxan at temperatures between 10 °C and 150 °C.

Synthesis of a compound with the general formula H_can be achieved by reaction of compound G_with a base such as sodium hydroxide or lithium hydroxide in solvents such as methanol, ethanol, THF and water or mixtures thereof, preferably in ethanol/THF or THF/water at temperatures between 10 °C and 100 °C. A compound of the general formula J can be obtained by reaction of compound H with amines of the general formula ]_using amide coupling procedures employing reagents such as TBTU, HATU or EDC/ N-Hydroxysuccinimide in the presence or absence of bases such as diisopropylethylamine in solvents such as DMF or THF at temperatures between 0 °C and 120 °C preferably between 0 °C and 30 °C.

Pharmaceutically acceptable salts of the compounds of the invention of formula (1 ) can be formed with numerous organic and inorganic acids and bases. Exemplary acid addition salts including acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, borate, butyrate, citrate, camphorate, camphersulfonate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethane sulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethane sulfonate, lactate, maleate, methane sulfonate, 2-naphthalene sulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenyl sulfonate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, sulfonate, tartrate, thiocyanate, toluene sulfonate such as tosylate, undecanoate, or the like.

Basic nitrogen-containing moieties can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromide and iodide; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long-chain alkyl halides such as decyl, lauryl, myristyl and stearyl chloride, bromide and iodide, or aral kyi hal ides l i ke benzyl and phenethyl brom ides, or others. Water soluble or dispersible products are thereby obtained.

Pharmaceutically acceptable basic addition salts include but are not limited to cations based on the alkaline and alkaline earth metals such as sodium, lithium, potassium, calcium, magnesium, aluminum salts and the like, as well as non toxic ammonium quarternary ammonium, and amine cations, including but not limited to ammonium, tetramethylammon i u m , tetraethyl a m mon i u m , m ethyl a m i n e , d i m ethyl a m i n e , trimethylamine, triethylamine, ethylamine and the like. Other representative amines useful for the formation of base addition salts include benzazethine, dicyclohexyl amine, hydrabine, N-methyl-D-glucamine, N-methyl-D-glucamide, t-butyl am ine, diethylamine, ethylendiamine, ethanolamine, diethanolamine, piperazine and the like and salts with amino acids such as arginine, lysine, or the like.

Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc ..) and racemates thereof as wel l as m ixtures in d ifferent proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates incl ud ing solvates of the free com pounds or solvates of a salt of the compound. As used herein the term "metabolite" refers to (i) a product of metabolism, including intermediate and products, (ii) any substance involved in metabolism (either as a product of metabolism or as necessary for metabolism), or (iii) any substance produced or used during metabolism. In particular it refers to the end product that remains after metabolism.

As used herein the term "prodrug" refers to (i) an inactive form of a drug that exerts its effects after metabolic processes within the body convert it to a usable or active form, or (ii) a substance that gives rise to a pharmacologically active metabolite, although not itself active (i.e. an inactive precursor).

The terms "prodrug" or "prodrug derivative" mean a covalently-bonded derivative or carrier of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s). In general, such prodrugs have metabolically cleavable groups and are rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood, and generally include esters and amide analogs of the parent compounds. The prodrug is formulated with the objectives of improved chemical stabil ity, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g ., increased hydrosolubil ity), and/or decreased side effects (e.g., toxicity). In general, prodrugs themselves have weak or no biological activity and are stable under ordinary conditions. Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textboo k of D ru g Design and Development, Krogsgaard-Larsen and H . Bundgaard (eds.), Gordon & Breach, 1 991 , particularly Chapter 5: "Design and Applications of Prodrugs"; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery, K.B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzvmology, K. Widder et al. (eds.), Vol . 42, Academic Press, 1985, particularly pp. 309-396; Burger's Medicinal Chemistry and Drug Discovery, 5th Ed., M. Wolff (ed.), John Wiley & Sons, 1995, particularly Vol . 1 and pp. 1 72-178 and pp. 949-982; Pro-Drugs as Novel Delivery Systems, T. Higuchi and V. Stella (eds.), Am. Chem. Soc, 1975; Bioreversible Carriers in Drug Design, E.B. Roche (e<±), Elsevier, 1987, each of which is incorporated herein by reference in their entireties.

The term "pharmaceutically acceptable prodrug" as used herein means a prodrug of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible.

As used herein the term "C3-10 cycloalkyl" or "C 3 _ 8 cycloalkyl" refers to mono- or polycyclic carbocyclic alkyl substituent or group having 3 to 10 or 3 to 8 ring atoms respectively, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, cycloheptatrienyl perhydrated naphthalene or indene, adamantyl or norbonanyl and the like.

The term "Ci_s alkyl" as used herein alone or in combination with other terms such as in alkoxy refers to a Ci_s, preferably Ci_4 straight or branched alkyl/alkoxy group such as methyl, ethyl, propyl (iso-, n-), butyl (iso-, n-, sec-, tert-), pentyl, hexyl, methoxy, ethoxy, propoxy (iso-, n-), butoxy (iso-, n-, sec-, tert-), pentoxy, hexoxy; moreover, the term "Ci_ 8 alkyl" also includes an alkyl group which may contain oxygen in the chain and may be substituted with halogen to form an ether or halogenated ether group. Any hydrogen atom, particularly in an alkyl, alkoxy or alkenyl group may be replaced by a fluorine atom.

The term "C 2 -8 alkenyl" by itself or as part of another group refers to a straight or branched alkenyl group of 2 to 8 carbons, preferably 2 to 6 carbons, in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2- propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2- heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl. The term "heterocyclyl" refers to monocyclic saturated or unsaturated heterocyclyl groups with 1 to 4 hetero atoms selected from N, S and O, with the remainder of the ring atoms being carbon atoms and having preferably a total number of ring atoms of 3 to 10, such as morpholino, piperazinyl, piperidinyl, pyridyl, pyrimidinyl, thiazolyl, indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl, thiophenyl or furanyl.

The term "heteroaryl" refers to a mono- or bicyclic aromatic group with 1 to 4 hetero atoms selected from N, S and O, with the remainder of the ring atoms being carbon atoms and having preferably a total number of ring atoms of 5 to 10. Examples without limitation of heteroaryl groups are such as benzofuranyl, furyl, thienyl, benzothienyl, thiazolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, benzothiazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, pyrrolyl, pyranyl, tetrahydropyranyl, pyrazolyl , pyridyl, qu inol inyl , isoqu inol inyl , purinyl , carbazolyl , benzoxazolyl , benzamidazolyl, indolyl, isoindolyl, pyrazinyl, diazinyl, pyrazine, triazinyltriazine, tetrazinyl, tetrazolyl, benzothiophenyl, benzopyridyl and benzimidazolyl.

In a further aspect the present invention provides pharmaceutical compositions comprising a thienopyrimidine compound of the present invention and optionally a pharmaceutically acceptable carrier.

The pharmaceutical composition according to the present invention may further comprise an additional therapeutic agent. Particularly preferred are compositions, wherein the additional therapeutic agent is selected from antidiabetics like insulin, long and short acting insulin analogues, sulfonylureas, biguanides, DPP-IV inhibitors, SGLT2 inhibitors, Ι ΐ β-HSD inhibitors, glucokinase activators, AMPK activators, Glp- 1 receptor agonists, GIP receptor agonists, DGAT inhibitors, PPARgamma agonists, PPARdelta agonists, and other antidiabetics derived from thiazolidinediones, lipid lowering agents such as statines, fibrates, ion exchange resins nicotinic acid derivatives, or HMG-CoA reductase inhibitors, cardiovascular therapeutics such as nitrates, antihypertensiva such as β-blockers, ACE inhibitors, Ca-channel blockers, angiotensin II receptor antagonists, diuretics, thrombocyte aggregation inhibitors, or antineoplastic agents such as al kaloids, al kyl ati ng agents, antibiotics, or antimetabolites, or anti-obesity agents. Further preferred compositions are compositions wherein the additional therapeutic agent is selected from a histamine antagonist, a bradikinin antagonist, serotonin antagonist, leukotriene, an antiasthmatic, an NSAID, an antipyretic, a corticosteroid, an antibiotic, an analgetic, a uricosuric agent, chemotherapeutic agent, an anti gout agent, a bronchodilator, a cyclooxygenase-2 inhibitor, a steroid, a 5-l ipoxyg enase in h ibitor, a n im munosu ppressive agent, a leu kotriene antagon ist, a cytostatic agent, an antineoplastic agent, a mTor inhibitor, a Tyrosine kinase inhibitor, antibodies or fragments thereof against cytokines and soluble parts (fragments) of cytokine receptors.

More particularly preferred are compounds such as human NPH insulin, human lente or ultralente insulin, insulin Lispro, insulin Aspart, insulin Glulisine, insulin detemir or insulin Glargine, metformin, phenformin, acarbose, miglitol, voglibose, pioglitazone, rosiglizatone, rivoglitazone, aleglitazar, alogliptin, saxagliptin, sitagliptin, vildagliptin, exenatide, l iraglutide, albiglutide, praml intide, carbutam ide, chlorpropam ide, glibenclamide (glyburide), gliclazide, glimepiride, glipizide, gliquidone, tolazamide, tolbutamide, atenolol, bisoprolol, metoprolol, esmolol, celiprolol, talinolol, oxprenolol, pindolol, propanolol, bupropanolol, penbutolol, mepindolol, sotalol, certeolol, nadolol, carvedilol, nifedipin, nitrendipin, amlodipin, nicardipin, nisoldipin, diltiazem, enalapril, verapamil, gallopamil, quinapril, captopril, lisinopril, benazepril, ramipril, peridopril, fosinopril, trandolapril, irbesatan, losartan, valsartan, telmisartan, eprosartan, olmesartan, hydrochlorothiazide, piretanid, chlorotalidone, mefruside, furosemide, bendroflumethiazid, triamterene, dehydralazine, acetylsalicylic acid, tirofiban-HCI, dipyramidol, triclopidin, iloprost-trometanol, eptifibatide, clopidogrel, piratecam, abciximab, trapidil, simvastatine, bezafibrate, fenofibrate, gemfibrozil, etofyllin, clofibrate, etofibrate, fluvastatine, lovastatine, pravastatin, colestyramide, colestipol- HCI, xantinol nicotinat, inositol nicotinat, acipimox, nebivolol, glycerolnitrate, isosorbide mononitrate, isosorbide dinitrate, pentaerythrityl tetranitrate, indapamide, cilazepril, urapidil, eprosartan, nilvadipin, metoprolol, doxazosin, molsidormin, moxaverin, acebutolol, prazosine, trapidil, clonidine, vinca alkaloids and analogues such as vinblastin, vincristin, vindesin, vinorelbin, podophyllotoxine derivatives, etoposid, teniposid, alkylating agents, nitroso ureas, N-lost analogues, cycloplonphamid, estamustin, melphalan, ifosfamid, mitoxantron, idarubicin, doxorubicin, bleomycin, mitomycin, dactinomycin, daptomycin, docetaxel, paditaxel, carboplatin, cisplatin, oxaliplatin, BBR3464, satraplatin, busulfan, treosulfan, procarbazine, dacarbazine, temozolomide, chlorambucil, chlormethine, cyclophosphamide, ifosfamide, melphalan, bendamustine, uramustine, ThioTEPA, camptothecin, topotecan, irinotecan, rubitecan, etoposide, teniposide, cetuximab, panitumumab, trastuzumab, rituximab, tositumomab, alemtuzumab, bevacizumab, gemtuzumab, aminolevulinic acid, methyl aminolevulinate, porfimer sodium, verteporfin, axitinib, bosutinib, cediranib, dasatinib, erlotinib, gefitinib, imatinib, lapatinib, lestaurtinib, nilotinib, semaxanib, sorafenib, sunitinib, vandetanib, retinoids (alitretinoin, tretinoin), altretamine, amsacrine, anagrelide, arsenic trioxide, asparaginase (pegaspargase), bexarotene, bortezomib, denileukin diftitox, estramustine, ixabepilone, masoprocol, mitotane, testolactone, tipifarnib, abetimus, deforolimus, everolimus, gusperimus, pimecrolimus, sirolimus, tacrolimus, temsirolimus, antimetabolites such as cytarabin, fluorouracil, fluoroarabin, gemcitabin, tioguanin, capecitabin, combinations such as adriamycin/daunorubicin, cytosine arabinosid/cytarabine, 4-HC, or other phosphamides.

Other particularly preferred compounds are compounds such as clemastine, diphenhydramine, dimenhydrinate, promethazine, cetirizine, astemizole, levocabastine, loratidine, terfenadine, acetylsalicylic acid, sodoum salicylate, salsalate, diflunisal, salicylsalicylic acid, mesalazine, sulfasalazine, osalazine, acetaminophen, indomethacin, sulindac, etodolac, tolmetin, ketorolac, bethamethason, budesonide, chromoglycinic acid, dimeticone, simeticone, domperidone, metoclopramid, acemetacine, oxaceprol, ibuprofen, naproxen, ketoprofen, flubriprofen, fenoprofen, oxaprozin, mefenamic acid, meclofenamic acid, pheylbutazone, oxyphenbutazone, azapropazone, nimesulide, metamizole, leflunamide, eforicoxib, lonazolac, misoprostol, paracetamol, aceclofenac, valdecoxib, parecoxib, celecoxib, propyphenazon, codein, oxapozin, dapson, prednisone, prednisolon, triamcinolone, dexibuprofen, dexamethasone, flunisolide, albuterol, salmeterol, terbutalin, theophylline, caffeine, naproxen, glucosamine sulfate, etanercept, ketoprofen, adalimumab, hyaluronic acid, indometacine, proglumetacine dimaleate, hydroxychloroquine, chloroquine, infliximab, etofenamate, auranofin, gold, [ 224 Ra] radium chloride, tiaprofenic acid, dexketoprofen(trometamol), cloprednol , sod ium auroth iomalate auroth ioglucose, colch icine, allopurinol , probenecid, sulfinpyrazone, benzbromarone, carbamazepine, lornoxicam, fluorcortolon, diclofenac, efalizumab, idarubicin, doxorubicin, bleomycin, mitomycin, dactinomycin, daptomycin, cytarabin, fluorouracil, fluoroarabin, gemcitabin, tioguanin, capecitabin, adriamydin/daunorubicin, cytosine arabinosid/cytarabine, 4-HC, or other phosphamides, penicillamine, a hyaluronic acid preparation, arteparon, glucosamine, MTX, soluble fragments of the TNF-receptor (such as etanercept (Enbrel)) and antibodies against TNF (such as infliximab (Remicade), natalizumab (Tysabri) and adalimumab (Humira)).

It will be appreciated by the person of ordinary skill in the art that the compounds of the invention and the additional therapeutic agent may be formulated in one single dosage form, or may be present in separate dosage forms and may be either administered concomitantly (i.e. at the same time) or sequentially.

The pharmaceutical compositions of the present invention may be in any form suitable for the intended method of administration.

The compounds of the present invention may be administered orally, parenterally, such as bronchopulmonary, subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermally, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable excipients.

Excipients that may be used in the formulation of the pharmaceutical compositions of the present invention comprise carriers, vehicles, diluents, solvents such as monohydric alcohols such as ethanol, isopropanol and polyhydric alcohols such as glycols and edible oils such as soybean oil, coconut oil, olive oil, safflower oil cottonseed oil, oily esters such as ethyl oleate, isopropyl myristate; binders, adjuvants, solubilizers, thicken ing agents, stabilizers, disintergrants, glidants, lubricating agents, buffering agents, emulsifiers, wetting agents, suspending agents, sweetening agents, colorants, flavors, coating agents, preservatives, antioxidants, processing agents, d rug d el ivery mod ifiers and en hancers such as cal ci u m phosphate, magnesium state, talc, monosaccharides, disaccharides, starch, gelatine, cellulose, methylcellulose, sodium carboxymethyl cellulose, dextrose, hydroxypropyl- β-cyclodextrin, polyvinylpyrrolidone, low melting waxes, ion exchange resins.

Other suitable pharmaceutically acceptable excipients are described in Remington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1991 ).

Dosage forms for oral administration include tablets, capsules, lozenges, pills, wafers, granules, oral liquids such as syrups, suspensions, solutions, emulsions, powder for reconstitution.

Dosage forms for parenteral administration include aqueous or olageous solutions or emulsions for infusion, aqueous or olageous solutions, suspensions or emulsions for injection pre-filled syringes, and/or powders for reconstitution.

Dosage forms for local/topical administration comprise insufflations, aerosols, metered aerosols, transdermal therapeutic systems, medicated patches, rectal suppositories, and/or ovula.

The amount of the compound of the present invention that may be combined with the excipients to formulate a single dosage form will vary upon the host treated and the particular mode of administration. The pharmaceutical compositions of the invention can be produced in a manner known per se to the skil led person as described , for example, in Rem ington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1991 ).

In a further aspect of the invention the use of a thienopyrimidine compound of the present invention for the production of a pharmaceutical composition for inhibiting the activity of the kinase activity of Mnk1 or Mnk2 (Mnk2a, Mnk2b) or further variants thereof is provided, in particular for the prophylaxis or therapy of metabolic diseases, hematopoietic disorders, cancer and their consecutive complications and disorders. Whereby the prophylaxis and therapy of metabolic diseases of the carbohydrate and/or lipid metabolism is preferred.

Diseases of the invention that are influenced by the inhibition of the kinase activity of Mnk1 and/or Mnk2 (Mnk2a or Mnk2b) and/or further variants thereof include diseases related to the regulation of metabolic diseases, such as obesity, eating disorders, cachexia, diabetes mellitus, metabolic syndrome, hypertension, coronary heart diseases, hypercholesterolemia, dyslipidemia, osteoarthritis, biliary stones and/or sleep apnea and diseases related to reactive oxygen compounds (ROS defense) such as diabetes mellitus, neurodegenerative diseases and cancer.

The pharmaceutical compositions of the invention are particularly useful for prophylaxis and treatment of obesity, diabetes mellitus and other metabolic diseases of the carbohydrate and lipid metabolism as stated above, in particular diabetes mellitus and obesity.

Thus in a more preferred embodiment of this invention the use of a thienopyrimidine compound for the production of a pharmaceutical composition for the prophylaxis or therapy of metabolic diseases is provided.

In yet a further aspect of the invention the use of a thienopyrimidine compound of the invention for the production of a pharmaceutical composition for treating or preventing a cytokine mediated disorder such as an inflammatory disease is provided.

The pharmaceutical compositions of the invention are thus useful for the prophylaxis or therapy of inflammatory diseases, in particular chronic or acute inflammation, chronic inflammatory arthritis, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, juvenile rheumatoid arthritis, gouty arthritis; psoriasis, erythrodermic psoriasis, pustular psoriasis, inflammatory bowel d isease, Crohn ' s d isease and related conditions, ulcerative colitis, colitis, diverticulitis, nephritis, urethritis, salpingitis, oophoritis, endomyometritis, spondylitis, systemic lupus erythematosus and related disorders, multiple sclerosis, asthma, meningitis, myelitis, encephalomyelitis, encephalitis, phlebitis, thrombophlebitis, chronic obstructive disease (COPD), inflammatory lung disease, allergic rhinitis, endocarditis, osteomyelitis, rheumatic fever, rheumatic pericarditis, rheumatic endocarditis, rheumatic myocarditis, rheumatic mitral valve disease, rheumatic aortic valve disease, prostatitis, prostatocystitis, spondoarthropathies ankylosing spondylitis, synovitis, tenosynovotis, myositis, pharyngitis, polymyalgia rheumatica, shoulder tendonitis or bursitis, gout, pseudo gout, vascul itides, inflammatory diseases of the thyroid selected from granulomatous thyroiditis, lymphocytic thyroiditis, invasive fibrous thyroiditis, acute thyroiditis; Hashimoto ' s thyroiditis, Kawasaki ' s disease, Raynaud ' s phenomenon, Sjogren ' s syndrome, neuroinflammatory disease, sepsis, conjubctivitis, keratitis, iridocyclitis, optic neuritis, otitis, lymphoadenitis, nasopaharingitis, sinusitis, pharyngitis, tonsillitis, laryngitis, epiglottitis, bronchitis, pneumonitis, stomatitis, gingivitis, oesophagitis, gastritis, peritonitis, hepatitis, cholelithiasis, cholecystitis, glomerulonephritis, goodpasture ' s disease, crescentic glomerulonephritis, pancreatitis, dermatitis, endomyometritis, myometritis, metritis, cervicitis, endocervicitis, exocervicitis, parametritis, tuberculosis, vaginitis, vulvitis, silicosis, sarcoidosis, pneumoconiosis, inflammatory polyarthropathies, psoriatric arthropathies, intestinal fibrosis, bronchiectasis and enteropathic arthropathies. As already stated above, the compositions of the present invention are particularly useful for treating or preventing a disease selected from chronic or acute inflammation, chronic inflammatory arthritis, rheumatoid arthritis, psoriasis, COPD, inflammatory bowel d isease, septic shock, Crohn ' s d isease, ulcerative col itis, multiple sclerosis and asthma.

Thus, in a more preferred embodiment of this invention the use of a thienopyrimidine compound for the production of a pharmaceutical composition for the prophylaxis or therapy of inflammatory diseases selected from chronic or acute inflammation, chronic inflammatory arthritis, rheumatoid arthritis, psoriasis, COPD, inflammatory bowel disease, septic shock Crohn ' s disease, ulcerative colitis, multiple sclerosis and asthma is provided. In yet a further aspect of the invention the use of a thienopy midine compound of the invention for the production of a pharmaceutical composition for treating or preventing cancer, viral diseases or neurodegenerative diseases is provided. For the purpose of the present invention, a therapeutically effective dosage will generally be from about 1 to 2000 mg/day, preferably from about 10 to about 1000 mg/day, and most preferably from about 10 to about 500 mg/day, which may be administered in one or multiple doses. It will be appreciated, however, that specific dose level of the compounds of the invention for any particular patient will depend on a variety of factors such as age, sex, body weight, general health condition, diet, individual response of the patient to be treated time of administration, severity of the disease to be treated, the activity of particular compound applied, dosage form, mode of application and concomitant medication. The therapeutically effective amount for a given situation will readily be determined by routine experimentation and is within the skills and judgment of the ordinary clinician or physician.

Abbreviations:

CDI: carbonyldiimidazole

TEA: triethylamine

HATU: (2-(7-aza-1 H-benzotriazole-1 -yl)-1 ,1 ,3,3-tetramethyluronium

hexaflurophosphate)

TBTU: 2-(1 H-Benzotriazol-1 -yl)-1 ,1 ,3,3-tetramethyluroniumtetrafluorborat THF: tetrahydrofuran

EE: ethylacetate

ACN: acetonitrile

EtOH: ethanol

MeOH: methanol

DCM: methylene chloride

DMF: N,N-dimethylformamide

EtOAc: ethyl acetate

HCI: hydrochloric acid t-BuOH: tert.butanol

DTAD: Di-ter-butyl azodicarboxylate

DEAD: diethyl azodicarboxylate

DIAD: diisopropyl azodicarboxylate

□ HMDS: lithium hexymethyldisilazane

DIPEA: diisopropylethyl amine

EDC: 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimid

TFA: trifluoro acetic acid

brine: saturated sodium chloride solution in water rt: room temperature

min: minute

Examples

Intermediate I

tert- Butyl 2-(2-amino-5-fluorophenoxy)propylcarbamate

tert- Butyl 2-(5-fluoro-2-nitrophenoxy)propylcarbamate

2-Nitro-5-fluorophenol (3.0 g) and tert-butyl-N-(2-hydroxypropyl)carbamate were dissolved in THF (20 ml) and triphenylphosphine (7.5 g) and di-tert-butyl- azodicarboxylate were added. The exothermic reaction was cooled in an ice bath. Then the reaction mixture was stirred at room temperature for 2h. The mixture was concentrated in vacuo and the residue was purified by chromatography (silica gel/ dichloromethane: petroleum ether 1 :2). The fractions were combined and

concentrated in vacuo. The residue was dissolved in dichloromethane and washed with 1 M NaOH. The organic layer was separated, dried, filtered and concentrated in vacuo.

Yield: 3.12 g

ESI mass spectrum: m/z = 315 (M+H) +

1.2: tert- Butyl 2-(2-amino-5-fluorophenoxy)propylcarbamate

To a solution of tert-butyl 2-(5-fluoro-2-nitrophenoxy)propylcarbamate (3.1 g) in MeOH (5 ml) was added 10% palladium on carbon (300 mg) and the reaction mixture was hydrogenated at room temperature at 50psi. The catalyst was filtered off and the filtrate was concentrated in vacuo.

Yield: 2.5 g

ESI mass spectrum: m/z = 285 (M+H) +

Intermediate II Tert-butyl 2-hvdrox butylcarbamate

1 -Amino-2-butanol (1 .0 g) was dissolved in dichloromethane (50.0 ml) and di-(tert- butyl) dicarbonate (2.6 g) was added. The reaction mixture was stirred at room temperature for 4h then washed with 1 M NaOH. The organic layer was separated and concentrated in vacuo.

Yield: 1 .8 g

Intermediate III Methyl 4-(4-fluoro-2-hvdroxyphenylamino)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylate

4-Chloro-5-methyl-thieno[2,3-d]pyhmidine-6-carboxylic acid methyl ester (0.5 g), 2- amino-5-fluorophenol (265.0 mg), p-toluenesulfonic acid monohydrate (75.0 mg) and dioxane (5.0 ml) were combined in a microwave tube. The mixture was heated at 140°C for 15min under microwave irradiation. Then the mixture was allowed to cool to room temperature and a precipitate formed. The precipitate was collected by filtration, washed with dioxane, methanol and Et 2 O to yield the title compound.

Yield: 605.0 mg

ESI mass spectrum: m/z = 334 (M+H) + Intermediate IV

Methyl 4-(2-hvdroxypyridin-3-ylamino)-5-methylthieno[2,3-dlpyhmidin e-6-carboxylate

Prepared analogously to example III using 4-chloro-5-methyl-thieno[2,3-d]pyrimidine- 6-carboxylic acid methyl ester and 3-amino-2-hydroxypyridine

Yield: 258.0 mg

ESI mass spectrum: m/z = 317 (M+H) + Intermediate V

tert-Butyl 3-(2-amino-5-fluorophenoxy)butylcarbamate

V.1 tert- Butyl 2-(5-fluoro-2-nitrophenoxy)propylcarbamate

Prepared analogously to example 1-1 using BOC-4-amino-2-butanol and 2-nitro-5- fluorophenol.

Yield: 2.20 g

ESI mass spectrum: m/z = 329 (M+H) + \Λ2 tert-Butyl 3-(2-amino-5-fluorophenoxy)butylcarbamate

Prepared analogously to example 1-2 using tert-butyl 3-(5-fluoro-2- nitrophenoxy)butylcarbamate

Yield: 1 .8 g

Intermediate VI

3-(2-Amino-5-fluoro-phenoxy)-butyric acid ethyl ester Intermediate VI.1 Ethyl 3-(5-fluoro-2-nitrophenoxy)butanoate

Ethyl 3-Hydroxybutyrate (6.5 ml) was dissolved in THF (350.0 ml) at 0°C and NaH (4.8 g) was added. The reaction was stirred at room temperature for 30 min. Then 5,5 g 2,4- difluoronitrobenzene was added and the mixture was heated at reflux overnight. The reaction was concentrated in vacuo and the residue was dissolved in water and dichloromethane. The organic layer was separated, dried and

concentrated in vacuo. The residue was purified by chromatography (silica/ dichloromethane) to yield the title compound.

Yield: 1 .36 g

ESI mass spectrum: m/z = 272 (M+H) +

VI. 2 3-(2-Amino-5-fluoro-phenoxy)-butyric acid ethyl ester

To a solution of Ethyl 3-(5-fluoro-2-nitrophenoxy)butanoate (1 .3 g) in THF (50 ml) was added Raney nickel (150 mg) and the reaction mixture was hydrogenated. The catalyst was filtered off and the filtrate was concentrated in vacuo.

Yield: 2.5 g

ESI mass spectrum: m/z = 285 (M+H) +

Intermediate VII

3[2-(2-Amino-5-fluoro-phenoxy)-3-tert-butoxycarbonylamino-pr opyl1-carbamic acid tert-butyl ester

VII. 1 (3-tert-Butoxycarbonylamino-2-hydroxy-propyl)-carbamic acid tert-butyl ester

Di-tert-butyl dicarbonate (41 .5g) was dissolved in dichloromethane (40.0 ml), a solution of 1 ,3- diamino-propan-2-ol (8.0g) and triethylamine (1 .5 ml) in

dichloromethane/methanol (1 :5; 100 ml) was added and the reaction mixture was stirred at room temperature overnight. The solution was concentrated in vacuo and the residue was dissolved in dichlormethane. The organic layer was washed with water, separated, dried and concentrated in vacuo. The residue was purfied by chromatography (silica / dichloromethane: methanol 25:1 ).

Yield: 17 g

ESI mass spectrum: m/z = 291 (M+H) + VII. 2 [3-tert-Butoxycarbonylamino-2-(5-fluoro-2-nitro-phenoxy)-pro pyl1- carbamic acid tert-butyl ester

2-Nitro-5-fluorophenol (8.4 g) and (3-tert-butoxycarbonylamino-2-hydroxy-propyl)- carbamic acid tert-butyl ester (17.0 g) were dissolved in THF (60 ml) and

triphenylphosphine (21 g) and di-tert-butyl-azodicarboxylate (18.4 g) were added. The exothermic reaction was cooled in an ice bath. Then the mixture was stirred at room temperature overnight. It was concentrated in vacuo and the residue was dissolved in dichloromethane, washed with water and 1 M NaOH. The organic layer was dried and concentrated in vacuo. The residue was purified by chromatography (silica / dichloromethane:methanol 25:1 ).

Yield: 25 g

ESI mass spectrum: m/z = 430 (M+H) +

VII.3 3[2-(2-Amino-5-fluoro-phenoxy)-3-tert-butoxycarbonylamino-pr opyl1-carbamic acid tert-butyl ester

Prepared analogously to example 1-2 using [3-tert-butoxycarbonylamino-2-(5-fluoro-

2-nitro-phenoxy)-propyl]-carbamic acid tert-butyl ester

Yield: 12.7 g

ESI mass spectrum: m/z = 400 (M+H) +

Intermediate VIII

4-(2-amino-5-fluorophenoxy)-2-methylpentan-2-ol VIII.1 4-(5-fluoro-2-nitrophenoxy)-2-methylpentan-2-ol

2-Methylpentane-2,4-diol (2.8 ml) was dissolved in THF (20.0 ml) and NaH (60% in mineral oil; 1 g) was added. At room temperature 2,4-difluoronitrobenzene was added and the reaction mixture was stirred overnight. Then the reaction was quenched with water and concentrated in vacuo. The residue was dissolved in dichloromethane, washed with water and concentrated in vacuo.

Yield: 2.4 g

ESI mass spectrum: m/z = 258 (M+H) +

VIII. 2 4-(2-amino-5-fluorophenoxy)-2-methylpentan-2-ol

Prepared analogously to example 1-2 using 4-(5-fluoro-2-nitrophenoxy)-2- methylpentan-2-ol

Yield: 2.12 g

ESI mass spectrum: m/z = 228 (M+H) +

Intermediate IX

2-(1 ,1 ,1 -Trifluoropropan-2-yloxy)pyridin-3-amine

IX. 1 3-Nitro-2-(1 ,1 ,1 -trifluoropropan-2-yloxy)pyhdine

1 ,1 ,1 - Trifluoro-2-propanol (3.2 g) was dissolved in THF (4.0 ml) and cooled to 0°C. Then LiHMDS (1 M in THF; 28.3 ml) was added dropwise. The reaction mixture was stirred at 0°C for 20 minutes. A solution of 2-fluoro-3-nitro-pyridine (4.0 g) in THF (1 ml) was added and the mixture was stirred overnight. It was quenched by addition of saturated NH CI solution and extracted with dichloromethane. The organic layer was dried and concentrated in vacuo.

Yield: 6.24 g

ESI mass spectrum: m/z = 237 (M+H) + IX. 2 2-(1 ,1 ,1 -Trifluoropropan-2-yloxy)pyridin-3-amine

3-Nitro-2-(1 ,1 ,1 -trifluoropropan-2-yloxy)pyridine (6.2 g) was dissolved in methanol (500 ml) and Raney nickel (1 .0 g) was added. The reaction mixture was

hydrogenated at room temperature and 5 bar. The catalyst was filtered off and the filtrate was concentrated in vacuo.

Yield: 4.83 g

ESI mass spectrum: m/z = 207 (M+H) +

Intermediate X

2-(1 ,3-Difluoropropan-2-yloxy)-4-fluoroaniline

X. 1 2-(1 ,3-Difluoropropa -2-yloxy)-4-fluoro-1 -nitrobenzene

Prepared analogously to example IX.1 using 2,4-difluoronitrobenzene.

Yield: 1 1 . 96g

X.2 2-(1 ,3-Difluoropropan-2-yloxy)-4-fluoroaniline

Prepared analogously to example IX. 2

Yield: 4.42 g

ESI mass spectrum: m/z = 206 (M+H) +

Intermediate XI

2-(2-Fluoropropoxy)pyridin-3-amine

XI.1 2-(2-fluoropropoxy)-3-nitropyhdin

2-Fluoropropan-1 -ol (93.6 mg) was dissolved in THF (10 ml). LiHMDS in THF(1 M; 1 .2 ml) was added and the reaction was stirred for 15 min. Then a solution of 2-fluoro-3- nitro-pyridine (142 mg) in THF was added and the reaction stirred at room

temperature overnight.

An aqueous solution of K2CO3 (2M; 750 μΙ) was added to the reaction mixture and filtered over Alox B. The filtrat was concentrated in vacuo.

Yield: 200 mg

Retention time HPLC: 2.05 min

HPLC method: 003_CC_ZQ6

XI.2 2-(2-Fluoropropoxy)pyridin-3-amine

2-(2-fluoropropoxy)-3-nitropyridine (199.97 mg) was dissolved in a mixture of THF (10 ml) and methanol (5 ml). Pd/C (20 mg) was added and the mixture was hydrogenated at room temperature for 4h and 3 bar.The mixture was concentrated in vacuo.

Yield: 153 mg

Retention time HPLC: 1 .50 min

HPLC method: 002_CC_ZQ4

Intermediate XII

2-(1 -methoxypropan-2-yloxy)pyridin-3-amine

XII.1 2-(1 -methoxypropan-2-yloxy)-3-nitropyhdine

Prepared analogously to example XI.1 using 2-fluoro-3-nitropyridine (142 mg) and 1 - methoxypropan-2-ol (108 mg).

Yield: 212 mg

Retention time HPLC: 2.04 min

HPLC method: 003 CC ZQ6 XII.1 2-(1 -Methoxypropan-2-yloxy)pyridin-3-amine

Prepared analogously to example XI.2 using 2-(1 -methoxypropan-2-yloxy)-3- nitropyridine (212 mg). Yield: 165 mg

Retention time HPLC: 1 .52 min

HPLC method: 002_CC_ZQ4 Intermediate XIII

2-(3-Aminopyridin-2yloxy)ethanol

XIII.1 2-(3-Nitropyridin-2-yloxy)ethanol

Ethylene glycole (88.8 mg) was dissolved in THF (10 ml). LiHMDS in THF(1 M; 1 .2 ml) was added and the reaction was stirred for 15 min. Then a solution of 2-fluoro-3- nitro-pyridine (142 mg) in THF was added and the reaction was stirred at room temperature overnight.

The reaction mixture was concentrated in vacuo and the residue was purified by RP- chromatography (H2O+ 0,1 % TFA/ MeOH= 40%~>99%).

Yield: 176 mg

Retention time HPLC: 1 .68 min

HPLC method: 003_CC_ZQ6

XIII.2 2-(3-Aminopyridin-2yloxy)ethanol

Prepared analogously to example XII.2 using 2-(3-nitropyridin-2-yloxy)ethanol (1 15 mg). Yield: 175 mg

Retention time HPLC: 1 .43 min

HPLC method: 003 CC ZQ7 Intermediate XIV

2-(2,2-Difluoroethoxy)pyridin-3-amine

XIV.1 2-(2,2-Difluoroethoxy)-3-nitropyridine

Prepared analogously to example XII.1 using 2-fluoro-3-nitropyridine (142 mg) and 2,2-difluoroethanol (98 mg).

Yield: 204 mg

Retention time HPLC: 1 .99 min

HPLC method: 003 CC ZQ6 XIV.2 2-(2,2-Difluoroethoxy)pyridin-3-amine

Prepared analogously to example XII.2 using 2-(2,2-difluoroethoxy)-3-nitropyridine (204 mg).

Yield: 171 mg

Retention time HPLC: 1 .58 min

HPLC method: 002_CC_ZQ4

Intermediate XV

2-((3-Aminopyridin-2-yloxy)methyl)propane-1 , 3-diol

Prepared analogously to example XII.1 using 2-fluoro-3-nitropyridine (142 mg) and 2- (hydroxymethyl)-1 ,3-propanediol (148 mg)

Yield: 130 mg

Retention time HPLC: 1 .65 min

HPLC method: 003 CC ZQ6

Prepared analogously to example XII.2 using 2-((3-nitropyridin-2- yloxy)methyl)propane-1 ,3-diol (130 mg).

Yield: 99 mg

Intermediate XVI

2-(1 -((3-Aminopyridin-2-yloxy)methyl)cvclopropyl)acetonithle

XVI .1 2-(1 -((3-Nitropyridin-2-yloxy)methyl)cvclopropyl)acetonitrile

Prepared analogously to example XI.1 using [1 -(hydroxymethyl)cyclopropyl] acetonitrile (267 mg)

Yield: 488 mg

Retention time HPLC: 1 .82 min

HPLC method: 004 CC ZQ6

XVI .2 2-(1 -((3-Aminopyridin-2-yloxy)methyl)cvclopropyl)acetonithle

2-(1 -((3-Nitropyridin-2-yloxy)methyl)cyclopropyl)acetonitrile (466 mg) was dissolved in a mixture of glacial acetic acid (4 ml) and ethanol (8 ml). Ferrum (1 .1 g) was added and the reaction mixture was heated to100°C for 2h. The mixture was concentrated in vacuo and the residue was dissolved in dichloromethane, extracted with an aqueous solution of K2CO3 (2M) and concentrated in vacuo

Yield: 264 mg

Retention time HPLC: 2.07 min

HPLC method: 003 CC ZQ7

Intermediate XVII

2-((2,2-Difluorocvclopropyl)methoxy)pyridine-3-amine

Prepared analogously to example XII.1 using (2,2-difluorocyclopropyl)methanol and

2-fluoro-3-nitro-pyridine

Yield: 469 mg

Retention time HPLC: 2.00 min HPLC method: 004_CC_ZQ6

Prepared analogously to example XII.2 using 2-((2,2-difluorocyclopropyl)methoxy)-3- nitropyridine

Yield: 391 mg

Retention time HPLC: 1 .45 min

HPLC method: 004 CC ZQ6

Intermediate XVIII

4-[2-(1 -Ethylcabamoyl-ethoxy)-4-fluoro-phenylamino1-5-methyl-thieno

[2,3dlpyrimidine-6-carboxylic acid methyl ester

XV 111.1 4-(4-fluoro-2-methoxy-phenylamino)-5-methyl-thieno[2,3-dlpyh midine-6- carboxylic acid methyl ester

4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (15.0 g), 4- fluoro-2-methoxyaniline (9.5 g), 4M hydrochloric acid in dioxane (4.5 ml) and dioxane (100.0 ml) were stirred at 100°C overnight. Then the mixture was filtrated and the solid was dried in vacuo.

Yield: 24.0 g

ESI mass spectrum: m/z = 348 (M+H) +

XVI 11.2 4-(4-Fluoro-2-hvdroxy-phenylamino)-5-methyl-thieno[2,3-dlpyr imidine-6- carboxylic acid methyl ester

The resulting product from XVIII.1 (24.0 g) was dissolved in DCM (500 ml) and was cooled with a dry ice bath.To the mixture was dropped slowly boron tribromide (35 ml) and stirred for 30 min. The mixture was allowed to cool to room temperature overnight. Then the mixture was cooled with an dry ice bath, dropped 100 ml methanol to the mixture, stirred for 30 min and concentrated.

The residue was suspended in methonol and stirred at reflux for 1 hour.

The mixture was filtrated and dried in vacuo.

Yield: 18.6 g

ESI mass spectrum: m/z = 334 (M+H) +

Retention time HPLC: 2.16 min

HPLC method: 007 CC ZQ5

XVIII.3 4-[2-(1 -tert-Butoxycyrbonyl-ethoxy)-4-fluoro-phenylamino1-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

To the resulting product from XVIII.2 (2.0 g) was added 2-bromopropionic acid tert- butyl ester (1 .4 g), cesium carbonate (4.8 g) and ACN (50 ml). The mixture was stirred at 60°C for 2 hours. Then water was added and the mixture was filtrated. Yield: 2.1 g

ESI mass spectrum: m/z = 462 (M+H) +

XVIII.4 4-[2-(1 -Carboxy-ethoxy)-4-fluoro-phenylamino1-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

To the resulting product from XVIII.3 (2.1 g) was added trifluoroacetic acid 50% in

DCM (20 ml) and stirred at rt overnight. The mixture was concentrated and triturated with diethylether.

Yield: 1 .9 g

ESI mass spectrum: m/z = 406 (M+H) +

Retention time HPLC: 1 .96 min

HPLC method: Method A 9

XVIII.5 4-[2-(1 -Ethylcarbamoyl-ethoxy)-4-fluoro-phenylamino1-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

To the resulting product from XVIII.4 (0.5 g) in ACN (10 ml) was added TBTU (0.4 g) and TEA (0.43 ml). The mixture was stirred at rt for 20 min. To the mixture was added ethylamine (2 mol/l (1 .5 ml)) and stirred at rt overnight. Then the mixture was concentrated and triturated with diethylether. The mixture was diluted with methanol and purified by chromatography.

Yield: 0.36 g

ESI mass spectrum: m/z = 433 (M+H) +

Retention time HPLC: 1 .89 min

HPLC method: Method A 9

Intermediate XIX

4-Amino-3-(2-fluoro-1 -fluoromethyl-ethoxy)-benzonithle

XIX.1 3-(2-Fluoro-1 -fluoromethyl-ethoxy)-4-nitro-benzonithle

1 ,3-Difluoro-propan-2-ol (4.2 g) was dissolved under argon in THF (250 ml) and cooled to 0°C. LiHMDS (28 ml) was added to the mixture and stirred at rt for 1 hour. Then the mixture was cooled to 0°C and 3-fluoro-4-nitrobenzonitrile (1 .95 ml) was added in portions and stirred for 2 hours. The mixture was poured in water and extracted with DCM. The organic layer was washed with water, dried and

concentrated.

Yield: 4.9 g

XIX.2 4-Amino-3-(2-fluoro-1 -fluoromethyl-ethoxy)-benzonithle 3-(2-Fluoro-1 -fluoromethyl-ethoxy)-4-nitro-benzonitrile (1 .45 g), tin(ll) chloride dihydrate (4.00 g) and ethanol (60 ml) were stirred at 100°C for 2 hours. The mixture was poured in water and extracted with DCM. The organic layer was washed with water, dried and concentrated.

Yield: 1 .04 g

Intermediate XXIV

4-Chloro-5-methyl-thieno[2,3-dlpyrimidine-6-carboxylic acid amide

XXIV.1 5-Amino-4-cvano- -methyl-thiophene-2-carboxylic acid methyl ester

To a mixture of methylacetoacetate (80.9 ml), malononitrile (49.5 g), sulfur (24 g) in methanol (750 ml) was added morpholin (139.4 ml). The mixture was stirred at rt for

10 min. Then the mixture was stirred at reflux for 3.5 hours. After that time the mixture was cooled with an ice bath and filtrated. The solid was washed with methanol and oven dried at 60°C.

Yield: 88.2 g

ESI mass spectrum: m/z = 197 (M+H) + XXIV.2 5-Methyl-4-oxo-3,4-dihvdro-thienor2,3-dlpyrimidine-6-carboxy lic acid methyl ester

To the product from XXIV.1 (70 g) was added formic acid (875 ml) and the mixture was stirred at reflux overnight. The mixture was cooled down, poured in ice water and filtrated. The solid was washed with water and a small portion of methanol . Then the residue was triturated with diethylether.

Yield: 72.98 g

ESI mass spectrum: m/z = 225 (M+H) +

The product from XXIV.2 (1 g), sodium hydroxide 4M (5 ml) and methanol were stirred at reflux for 1 hour. At rt 5 ml hydrochloric acid 4M were added and the mixture was filtrated. The solid was washed with water and dried at 60°C in an oven overnight.

Yield: 950 mg

ESI mass spectrum: m/z = 21 1 (M+H) +

XXIV.4 4-Chloro-5-methyl-thieno[2,3-c lpyrimidine-6-carbonyl chloride

DMF (0.2 ml) was added to a mixture of the product from XXIV.3 (1 g) and thionylchloride (10 ml). The mixture was stirred at reflux for 1 hour. Then the mixture was concentrated.

Yield: 1 .2 g

XXIV.5 4-Chloro-5-methyl-thieno[2,3-dlpyrimidine-6-carboxylic acid amide

The product from XXIV.4 (1 g) was dissolved warm in ACN (10 ml). This mixture was added dropwise to an ice cooled solution of concentrated ammonia (20 ml) and stirred for 15 min. The mixture was filtrated. The solid was washed with water and oven dried at 50°C.

Yield: 677 mg

ESI mass spectrum: m/z = 226 (M+H) +

Retention time HPLC: 0.766 min H PLC method: M2-SB-C18

Intermediate XXVI

3-(2-Amino-5-fluoro-phenoxy)-2-meth l-butan-2-ol

XXVI .1 2-Methyl-butane-2,3-diol

3-Hydroxy-3-methyl-2-butanon (5 ml) was dissolved in ethanol (20 ml). PtO2 (100 mg) was added and the mixture was hydrogenated at room temperature for 4h and 3 bar.The mixture was concentrated in vacuo.

Yield: 3.87 g

XXVI .2 3-(5-Fluoro-2-nitro- henoxy)-2-methyl-butan-2-ol

Prepared analogously to example XX.2 using the product from XXVI.1 (0.9 ml). Yield: 1 .7 g

ESI mass spectrum: m/z = 261 (M+H) +

Retention time HPLC: 1 .185 min

HPLC method: M2-SB-C18

XXVI .3 3-(2-Amino-5-fluor -phenoxy)-2-methyl-butan-2-ol

Prepared analogously to example XI.2 using the product from XXVI.2 (1 .7 g). Yield: 1 .45 g

ESI mass spectrum: m/z = 214 (M+H) +

Retention time HPLC: 0.681 min

HPLC method: M2-SB-C18

Compound 1

tert-Butyl 2-(5-fluoro-2-(5-methyl-6-(methylcarbamoyl)thieno[2,3-dlpyhm idin-4- ylamino)phenoxy)propyl carbamate 1 .1 Methyl 4-(2-(1 -tert-butoxycarbonylamino)propan-2-yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-dlpyhmidine-6-carboxy late

4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (2.0 g), tert- butyl 2-(2-amino-5-fluorophenoxy)propylcarbamate (2.3 g) and Hunig ' s base (4.2 ml) were dissolved in dioxane (70 ml). The reaction mixture was heated at 100°C for three days. Then the reaction was cooled and the precipitate was filtered, washed with dioxane, water and diethylether.

Yield: 1 .4 g

ESI mass spectrum: m/z = 491 (M+H) +

Retention time HPLC: 3.36 min

HPLC method: A 4

1 .2 4-(2-(1 -(tert-Butoxycarbonylamino)propan-2-yloxy)-4-fluorophenylami no)-5- methyl-thieno[2,3-dlpyhmidine-6-carboxylic acid

Methyl 4-(2-(1 -tert-butoxycarbonylamino)propan-2-yloxy)-4-fluorophenylamin o)-5- methylthieno[2,3-d]pyrinnicline-6-carboxylate (700 mg) and NaOH (1 M; 3.6 ml) was dissolved in a mixture of methanol and THF (1 :1 ; 20 ml). The reaction was stirred at room temperature overnight. Aqueous HCI (1 M; 3.6 ml) was added and the organic solvent was evaporated. The residue was triturated with water and filtered. The residue was washed with water, methanol and Et 2 O and dried at 60°C.

Yield: 0.63 g

ESI mass spectrum: m/z = 477 (M+H) +

Retention time HPLC: 1 .96 min

HPLC method: A 9

1 .3 tert-Butyl 2-(5-fluoro-2-(5-methyl-6-(methylcarbamoyl)thieno[2,3-dlpyhm idin-4- ylamino)phenoxy)propyl carbamate

To a solution of 4-(2-(1 -(tert-Butoxycarbonylamino)propan-2-yloxy)-4- fluorophenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxy lic acid (80 mg) in DMF (1 ml), TBTU (54 mg) and triethylamine (60 μΙ) were added.The mixture was stirred at room temperature for 5 min then methylamine in THF (2M; 420 μΙ) was added and the reaction stirred at room temperature overnight. The mixture was diluted with methanol and purified directly by RP-chromatography (water with 0.2% trifluoroacetic acid/ methanol 72-100%) to give the title compound.

Yield: 38 mg

ESI mass spectrum: m/z = 490 (M+H) +

Retention time HPLC: 1 .86 min

HPLC method: A 9

Further analogues of 1 :

The compounds listed in table 1 were synthesized analogously to example 1 .3 using 4-(2-(1 -(tert-butoxycarbonylamino)propan-2-yloxy)-4-fluorophenylami no)-5- methylthieno[2,3-d]pyrimidine-6-carboxylic acid and the corresponding amine.

Table 1 :

Compound 2 2.1 Methyl 4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylate trifluoroacetate

Methyl 4-(2-(1 -tert-butoxycarbonylamino)propan-2-yloxy)-4-fluorophenylamin o)-5- methyl-thieno[2,3-d]pyrinnidine-6-carboxylate (500 mg) was dissolved in a solution of

25% trifluoracetic acid in dichloromethane (10 ml). The reaction mixture was stirred at room temperature for 2h and then concentrated in vacuo.

Yield: 460 mg

ESI mass spectrum: m/z = 391 (M+H) +

Retention time HPLC: 1 .31 min

HPLC method: A 9

Further analogues of 2:

The compounds listed in table 2 were synthesized analogously to example 2.1 using the corresponding BOC protected derivatives shown in Table 1 .

Table 2:

Compound 3

Methyl 4-(4-fluoro-2-(1 -(methylsulfonamido)propan-2-yloxy)phenylamino)-5-methyl- thieno[2,3-dlpyhmidine-6-carboxylate

3.1 Methyl 4-(4-fluoro-(2-(1 -methylsulfonamido)propan-2-yloxy)phenylamino)-5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carboxylate

To a solution of Methyl 4-(2-(1-aminopropan-2-yloxy)-4-fluorophenylamino)-5 - methyl-thieno[2,3-d]pyhmidine 6-carboxylate trifluoroacetate (80 mg) and

triethylamine (55 μΙ) in dichloromethane (1.5 ml), methansulfonyl chloride (16 μΙ) was added. The reaction mixture was stirred at room temperature overnight. Then the reaction was quenched with water and methanol. The organic layer was separated and concentrated in vacuo. Yield: 68 mg

ESI mass spectrum: m/z = 469 (M+H) +

Retention time HPLC: 1 .99 min

HPLC method: A_9

Further analogues of compound 3 :

The compounds listed in table 3 were synthesized analogously to example 3.1 using the appropriate amine and the corresponding chloride. Table 3:

Compound 4

Methyl 4-(2-(1 -dimethylamino)propan-2yloxy)-4-fluorophenylamino)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylate trifluoroacetate

Methyl 4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- d]pyrimidine-6-carboxylate trifluoroacetate (100 mg) was dissolved in THF (2.5 ml), NaOH (4M; 55 μΙ) and formaldehyde 37% in water (55 μΙ) was added. The reaction mixture was stirred a few minutes. Then sodium triacetoxyborohydride (220 mg) was added. and the reaction was stirred at room temperature for 4h. Then the mixture was concentrated in vacuo. The residue was dissolved in dichloromethane and washed with aqueous NaOH (1 M). The organic layer was separated and concentrated in vacuo. The product was purified by RP-chromatography (water with 0.2%

trifluoroacetic acid/ methanol 72-100%) to afford the title compound.

Yield: 83mg

ESI mass spectrum: m/z = 419 (M+H) +

Retention time HPLC: 1 .32 min

HPLC method: A 9

Compound 5

tert-Butyl 2-(2-(6-carbamoyl-5-methyl-thieno[2,3-dlpyhmidin-4-ylamino)- 5- fluorophenoxy) propylcarbamate

4-(2-(1 -(tert-Butoxycarbonylamino)propan-2-yloxy)-4-fluorophenylami no)-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylic acid (250mg) and TBTU (170 mg) was dissolved in DMF (3 ml) and triethylamine (190 μΙ) was added. The mixture was stirred at room temperature for 10 min, then 0.5 ml cone, ammonia was added. The reaction was stirred for 2h. Then the reaction mixture was concentrated in vacuo and purified by RP-chromatography (water with 0.2% thfluoroacetic acid/ methanol 72-100%) to give the title compound.

Yield: 55 mg

ESI mass spectrum: m/z = 476 (M+H) +

Retention time HPLC: 1 .80 min

HPLC method: A 9

Compound 6

( R) and (S)-4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- dlpyhmidine-6-carboxamide trifluoroacetate

6.1 (R) and (S)- tert-butyl 2-(2-(6-carbamoyl-5-methyl-thienor2,3-dlpyrimidin-4- ylamino)-5-fluorophenoxy)propyl carbamate

The racemate of example 5 was separated by means of HPLC to afford the two enantiomers. The configuration was assigned arbitrarily.

Enantiomer A:

Yield: 10 mg

ESI mass spectrum: m/z = 476 (M+H) +

Retention time HPLC: 1 .80 min HPLC method:

Enantiomer B:

Yield: 14.2 mg

ESI mass spectrum: m/z = 476 (M+H) +

Retention time HPLC 1 .80 min

HPLC method: A 9

6.2 (R) and (S)-4-(2-(1 -aminopropan-2-yloxy)-4-fluorophenylamino)-5- methylthieno[2,3-dlpyhmidine-6-carboxamide trifluoroacetate

iral

Prepared analogously to example 2.1 from (S)- tert-butyl 2-(2-(6-carbamoyl-5- methylthieno[2,3-d]pyrimidin-4-ylamino)-5-fluorophenoxy)prop ylcarbamate and the enantiomer, respectively.

The configuration was assigned arbitrarily.

Enatiomer A

Yield: 7 mg

ESI mass spectrum: m/z = 376 (M+H) +

Retention time HPLC: 1 .16 min

HPLC method: A_9

Enantiomer B

Yield: 1 1 mg

ESI mass spectrum: m/z = 376 (M+H) +

Retention time HPLC: 1 .16 min HPLC method:

Compound 7

4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- dlpyhmidine-6-carboxylic acid thfluoroacetate

Synthesized analogously to example 2-1 from 4-(2-(1 -(tert-butoxycarbonyl- amino)propan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3-d]pyrimidine-6- carboxylic acid

Yield: 89 mg

ESI mass spectrum: m/z = 377 (M+H) +

Retention time HPLC: 1 .26 min

HPLC method: A_9 Compound 8

4-(4-Fluoro-2-(1 -(3,3,3-trifluoropropylamino)propan-2-yloxy)phenylamino-5-me thyl- thieno[2,3-dlpyrimidine-6-carboxamide

8.1 4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- dlpyhmidine-6-carboxamide thfluoroacetate

tert-Butyl 2-(2-(6-carbamoyl-5-methylthieno[2,3-d]pyrimidin-4-ylamino)- 5- fluorophenoxy)propylcarbamate (740mg) was dissolved in dichloromethane (20 and thfluoroacetic acid (4 ml) was added and the reaction was stirred at room temperature overnight. The reaction was concentrated in vacuo to give the title compound.

Yield: 667 mg

ESI mass spectrum: m/z = 376 (M+H) + 8.2 4-(4-Fluoro-2-(1 -(3,3,3-trifluoropropylamino)propan-2-yloxy)phenylamino-5- methyl-thieno[2,3-dlpyhmidine-6-carboxamide

4-(2-(1 -Aminopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2 ,3- d]pyrimidine-6-carboxamide 2,2,2-trifluoroacetate (200 mg) was dissolved in THF (20 ml) and buffer pH 5 (2 ml), 3,3,3,-trifluoropropanal (50.4 mg) and sodium

cyanoborohydride (30.8 mg) were added.

The reacion was stirred at room temperature for 4 days.The residue was diluted with 10 ml water, then the organic solvent was evaporated. The residual aqueous fraction was extracted with ethylacetate. The organic layer was separated, dried and concentrated in vacuo

Yield: 190 mg

ESI mass spectrum: m/z = 472 (M+H) +

Retention time HPLC: 1 .38 min

HPLC method: A 10

Compound 9

Methyl 4-(2-(2-aminoethoxy)-4-fluorophenylamino)-5-methyl-thieno[2, 3-dlpyhmidine- 6-carboxylate hydrochloride

9.1 Methyl 4-(2-(2-(tert-butoxycarbonylamino)ethoxy)-4-fluorophenylamin o)-5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carboxylate

Methyl 4-(4-fluoro-2-hydroxyphenylamino)-5-methyl-thieno[2,3-d]pyhm idine-6- carboxylate (80.0 mg), Boc-amino-ethanol (50.0 mg) and polymer-bound

triphenylphosphine (240.0 mg) was suspended in THF (3.0 ml). The mixture was stirred at room temperature for 5 min. Then di-tert-butyl azodicarboxylate (165.0 mg) was added and the mixture stirred at room temperature overnight. To this reaction celite was added, the resulting mixture was filtered through celite and the filtrate was concentrated in vacuo. The residue was purified by RP-chromatography (water with 0.2% trifluoroacetic acid/ methanol 59-100%)

Yield: 25.0 mg

ESI mass spectrum: m/z = 477 (M+H) +

Retention time HPLC: 2.12 min

HPLC method: A 9 9.2 Methyl 4-(2-(2-aminoethoxy)-4-fluorophenylamino)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylate hydrochloride

Methyl 4-(2-(2-(tert-butoxycarbonylamino)ethoxy)4-fluorophenylamino )-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylate (20 mg) was dissolved in 25% solution of trifluoroacetic acid in dichlormethane (2 ml). The reaction was stirred at room temperature for 2h. The solvent was concentrated in vacuo and the residue was dissolved in methanol and triturated with HCI in methanol.

Yield: 15 mg

ESI mass spectrum: m/z = 377 (M+H) +

Retention time HPLC: 1 .28 min

HPLC method: A_9

Compound 10

Methyl 4-(2-(1 -aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3 - dlpyrimidine-6-carboxylate hydrochloride

10.1 Methyl 4-(2-( 1 -(tert-butoxycarbonylamino)butan-2-yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-dlpyhmidine-6-carboxy late

Synthesized analogously to example 9.1 using methyl 4-(4-fluoro-2- hydroxyphenylamino)-5-methylthieno[2,3-d]pyrimidine-6-carbox ylate and tert-butyl 2- hydroxybutylcarbamate.

Yield: 36 mg

ESI mass spectrum: m/z = 505 (M+H) +

Retention time HPLC: 2.24 min

HPLC method: A 9 10.2 Methyl 4-(2-(1 -aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3 - d]pyrimidine-6-carboxylate hydrochloride

ci

Synthesized analogously to example 9.2 using methyl 4-(2-(1 -(tert- butoxycarbonylamino)butan-2-yloxy)-4-fluorophenylamino)-5-me thylthieno[2,3- d]pyrimidine-6-carboxylate

Yield: 21 mg

ESI mass spectrum: m/z = 405 (M+H) +

Retention time HPLC: 1 .38 min

HPLC method: A 9 Compound 1 1

Methyl 4-(2-(1 -aminopropan-2yloxy)-pyridin-3-ylamino)-5-methyl-thieno[2,3- dlpyhmidine-6-carboxylate hydrochloride

1 1 .1 Methyl 4-(2-(1 -(tert-butoxycarbonylamino)propan-2-yloxy)pyridin-3-ylamino) -5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carbox late

Prepared analogously to example 9.1 using methyl 4-(2-hydroxypyridin-3-ylamir methylthieno[2,3-d]pyrimidine-6-carboxylate and tert-butyl N-(2-hydroxypropyl)- carbamate.

Yield: 65 mg

ESI mass spectrum: m/z = 474 (M+H) +

Retention time HPLC: 1 .97 min

HPLC method: A 9

1 1 .2 Methyl 4-(2-(1 -aminopropan-2yloxy)-pyridin-3-ylamino)-5-methyl-thieno[2,3- dlpyrimidine-6-carboxylate hydrochloride

ci

Prepared analogously to example 9.2 using methyl 4-(2-(1 -(tert-butoxycarbonyl- amino)-propan-2-yloxy)pyridin-3-ylamino)-5-methylthieno[2,3- d]pyrimidine-6- carboxylate Yield: 14 mg

ESI mass spectrum: m/z = 374 (M+H) +

Retention time HPLC: 1 .29 min

HPLC method: A 9

Compound 12

Methyl 4-(2-(4-aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxylate hydrochloride

ci

4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (1 .25 g), tert-butyl 3-(2-amino-5-fluorophenoxy)butylcarbamate (1 .7 g) and p-toloulsulfonic acid (175 mg) was dissolved in dioxane (30ml) and the solution was heated under reflux overnight. Then the reaction mixture was concentrated in vacuo. The residue was treated with trifluoracetic acid 25% in dichloromethane and stirred at room temperature for 1 h. The solution was concentrated in vacuo and the residue was dissolved in dichloromethane and washed with sodium bicarbonate. The organic phase was separated, dried and concentrated in vacuo. The residue was triturated with Et 2 O and the precipitate was filtered.

Yield: 2.14 g

ESI mass spectrum: m/z = 405 (M+H) +

Retention time HPLC: 1 .36 min

HPLC method: A_9

Compound 13

Methyl 4-(4-fluoro-2-(4-(methylsulfonamido)butan-2yloxy)phenylamino )-5-methyl- thieno[2,3-dlpyhmidine-6-carboxylate

Methyl 4-(2-(4-aminobutan-2yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- d]pyrimidine-6-carboxylate hydrochloride (80 mg) and triethylamine (65 μΙ) were dissolved in dichloromethane (1 .5 ml) and methansulfonyl chloride (18 μΙ) was added. The reaction mixture was stirred at room temperature overnight. Then the mixture was diluted with water and some methanol. The organic layer was separated and concentrated in vacuo. The residue was purified by RP-chromatography (water with 0.2% trifluoroacetic acid/ methanol 59-100%)

Yield: 33 mg

ESI mass spectrum: m/z = 483 (M+H) +

Retention time HPLC: 1 .94 min

HPLC method: A_9

Further analogues of compound 13 :

The compounds listed in table 4 were synthesized analogously to example 13 using the appropriate amine and the corresponding chlorides or isocyanate.

Compound 14

Methyl 4-(2-(dimethylamino)butan-2yloxy)-4-fluorophenylamino)-5-met hyl-thieno[2,3- dlpyhmidine-6-carboxylate thfluoroacetate

Prepared analogously to example 4.1 using methyl 4-(2-(4-aminobutan-2yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carbox ylate hydrochloride

Yield: 53 mg

ESI mass spectrum: m/z = 433 (M+H) +

Retention time HPLC: 1 .38 min

HPLC method: A 9

Compound 15 Methyl 4-(2-((4-aminobutan-2yloxy)pyridin-3ylamino)-5-methyl-thien dlpyrimidine-6-carboxylate trifluoracetate

15.1 Methyl 4-(2-((4-aminobutan-2yloxy)pyndin-3ylamino)-5-methylthieno[2 ,3- dlpyrimidine-6-carboxylate

Prepared analogously to example 1 .1 using 4-Chloro-5-methyl-thieno[2,3- d]pyrimidine-6-carboxylic acid methyl ester and tert-butyl 3-(3-aminopyridin- 2yloxy)butyl carbamate

Yield: 1 g

ESI mass spectrum: m/z = 488 (M+H) +

15.2 Methyl 4-(2-((4-aminobutan-2yloxy)pyridin-3ylamino)-5-methyl-thieno [2,3- dlpyrimidine-6-carboxylat trifluoroacetate

Prepared analogously to example 1 .2 from methyl 4-(2-((4aminobutan-2yloxy)pyrid

3ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylate trifluoroacetate

Yield: 100 mg

ESI mass spectrum: m/z = 388 (M+H) +

Retention time HPLC: 1 .26 min

H PLC method: A_10 Compound 16

4-(2-(4-Aminobutan-2-yloxy)pyhdin-3-ylamino)-N-(3-(dimethyla mino)propyl)-5- methylthieno[2,3-dlpyhmidine-6-carboxamide bis-thfluoroacetate

16.1 4-(2-(4-(tert-Butoxycarbonylamino)butan-2yloxy)pyridin-3ylam ino)-5-methyl- thieno[2,3-dlpyhmidine-6-c rboxylic acid

Methyl 4-(2-(4-(tert-butoxycarbonylamino)butan-2yloxy)pyhdin-3ylami no)-5-methyl- thieno[2,3-d]pyhmidine-6-carboxylate (600 mg) was dissolved in methanol (10 ml) and NaOH (1 M; 2.5 ml) was added. The reaction mixture was heated at reflux for 15 minutes. Then it was cooled to room temperature and HCI (1 M; 2.5 ml) was added.

Methanol was evaporated and the residue was dissolved in dichloromethane. The mixture was washed with water, the organic layer was dried and concentrated in vacuo. The residue was triturated with Et 2 O.

Yield: 390 mg

ESI mass spectrum: m/z = 474 (M+H) +

16. 2 4-(2-(4-aminobutan-2yloxy)pyridin-3ylamino)-5-methylthieno[2 ,3-dlpyrimidine-

6-carboxylate trifluoroacetate

Prepared analogously to example 1 .2 from 4-(2-(4-(tert-butoxycarbonylamino

2yloxy)pyridin-3ylamino)-5-methylthieno[2,3-d]pyrimidine-6-c arboxylic acid

Yield: 100 mg

ESI mass spectrum: m/z = 374 (M+H) +

Retention time HPLC: 1 .12 min

HPLC method: A 10

16.3 4-(2-(4-Aminobutan-2-yloxy)pyhdin-3-ylamino)-N-(3-(dimethyla mino)propyl)-5-

4-(2-(4-(t-Butoxycarbonylamino)butan-2yloxy)pyridin-3ylamino )-5-methylthieno[2,3- d]pyrimidine-6-carboxylic acid (200mg) and Hunig's base (160 μΙ) was dissolved in THF (10 ml) and TBTU (148 mg) was added. To this mixture N,N-dimethyl- propandiamine (58 μΙ) was added and the mixture was stirred at room temperature for three days. The reaction mixture was concentrated in vacuo and the residue was dissolved in dichloromethane. The organic layer was washed with water, dried and 3 ml cone. TFA was added. The mixture was stirred at room temperature for 1 h. The mixture was concentrated in vacuo and the product was purified by RP- chromatography (Method amslpolar 1 ).

Yield: 190 mg

ESI mass spectrum: m/z = 458 (M+H) +

Retention time HPLC: 0.86 min

HPLC method: A 10 Compound 16.4: Prepared analogously to example 16.3 using 4-(2-(4-(tert-butoxycarbonylamino) butan-2yloxy)pyridin-3ylamino)-5-methylthieno[2,3-d]pyrimidi ne-6-carboxylic acid and ammonia.

Compound 17

4-(2-(2-(Dimethylamino)-2-oxoethoxy)-4-methylphenylamino)-5- methylthieno[2,3- dlpyhmidine-6-carboxylic acid 17.1 Ethyl 4-(2-(2-dimethylamino)-2-oxoethoxy)-4-methylphenylamino)-5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carboxylate

2-(2-Amino-5-methylphenoxy)-N,N-dimethylacetamide (300 mg) and ethyl 4-chloro-5- methyl-thieno[2,3-d]pyrimidine-6-carboxylate (447 mg) were dissolved in dioxane (5 ml) and p-toluenesulfonic acid monohydrate (44 mg) was added. The mixture was heated at 100°C for 1 .5h. The reaction was allowed to cool to room temperature, the residue was dissolved in dichloromethane and extracted with water. The organic layer was dried and concentrated in vacuo. The residue was purified by

chromatography (silica / dichloromethane: methanol 100-95:5).

Yield: 1 14 mg

ESI mass spectrum: m/z = 377 (M+H) +

17.2 4-(2-(2-(Dimethylamino)-2-oxoethoxy)-4-methylphenylamino)-5- methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid

Ethyl 4-(2-(2-dimethylamino)-2-oxoethoxy)-4-methylphenylamino)-5-m ethyl- thieno[2,3-d]pyrimidine-6-carboxylate (1 14 mg) was dissolved in methanol (5 ml) and aqueous NaOH (2M; 0.66 ml) was added. The reaction was stirred at room

temperature for 4h. Then the reaction mixture was concentrated in vacuo and neutralisied with aqueous HCI (2M; 5 ml). The precipitate was filtered and the residue was dissolved in DMF and purified by RP-chromatography (water with 0.2% trifluoroacetic acid/ methanol 72-100%)

Yield: 26 mg

ESI mass spectrum: m/z = 401 (M+H) +

Retention time HPLC: 1 .69 min

HPLC method: A_10

Compound 18

Methyl 4-(2-(1 ,3-diaminopropan-2-yloxy)-4-fluorophenylamino)-5-methylthien o[2,3- dlpyrimidine-6-carboxylate bis (2,2,2-trifluoroacetate)

18.1 4-{2-[2-tert-Butoxycarbonylamino-1 -(tert-butoxycarbonylamino-methyl)- ethoxy1-4-fluoro-phenylamino)-5-methyl-thieno[2,3-dlpyhmidin e-6-carboxylic acid methyl ester

4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (2 g) and [2- (2-Amino-5-fluoro-phenoxy)-3-tert-butoxycarbonylamiino-propy l]-carbamic acid t-butyl (3.3 g) were dissolved in 20 ml isopropanol and 4.3 ml Hunig ' s base was added. The reaction was heated to 140°C under microwave irradiation for 45 min. The reaction mixture was allowed to stand over the weekend and the resulting precipitate was filtered. The filter cake was purified by chromatography (silica / dichloromethane: methanol 50:1 ).

Yield: 2 g

ESI mass spectrum: m/z = 606 (M+H) +

Retention time HPLC: 2.34 min

HPLC method: A 10

18.2 Methyl 4-(2-(1 ,3-diaminopropan-2-yloxy)-4-fluorophenylamino)-5- methylthieno[2,3-dlpyhmidine-6-carboxylate bis (trifluoroacetate)

Prepared analogously to example 2.1 using 4-{2-[2-tert-Butoxycarbonylamino-1 -(tert- butoxycarbonylamino-methyl)-ethoxy]-4-fluoro-phenylamino}-5- methyl-thieno[2,3- d]pyrimidine-6-carboxylic acid methyl ester

Yield: 2 g ESI mass spectrum: m/z = 406 (M+H) +

Retention time HPLC: 0.87 min

HPLC method: A 10 Compound 19

Methyl 4-(2-(1 -acetamido-3aminopropan-2-yloxy)-4-fluorophenylamino)-5- methylthieno[2,3-dlpyrimidine-6-carboxylate (trifluoroacetate)

19.1 Methyl 4-(2-(1 -acetamido-3-aminopropan-2-yloxy)-4-fluorophenylamino)-5- methylthieno[2,3-dlpyrimidine-6-carboxylate trifluoroacetate

Methyl 4-(2-(1 ,3-diaminopropan-2-yloxy)-4-fluorophenylamino)-5-methylthien o[2,3- d]pyrimidine-6-carboxylate bis (trifluoroacetate) (150 mg) and triethylamine (0.1 ml) were dissolved in dichloromethane, acetylchloride (15μΙ) was added. The reaction mixture was stirred at room temperature overnight. The reaction was diluted with dichloromethane and methanol and then extracted with aqueous HCI (1 M). The organic layer was dried and concentrated in vacuo. The residue was purified by chromatography. The fractions were collected, concentrated in vacuo and triturated with Et 2 O.

Yield: 22 mg

ESI mass spectrum: m/z = 448 (M+H) +

Retention time HPLC: 1 .35 min

HPLC method: A 10

Further analogues of compound 19:

The compounds listed in table 5 were synthesized analogously to example 19.1 using the appropriate amines and the corresponding chlorides. Table 5:

Compound 20

N-(3-(Dimethylamino)propyl)-4-(4-fluoro-2-(4-hvdroxy-4-me thylpentan-2- yloxy)phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-carbox amide trifluoroacetate

20.1 Methyl 4-(4-fluoro-2-(4-hvdroxy-4-methylpentan-2-yloxy)phenylamino) -5- methyl-th ieno[2 ,3-dlpyrim id ine-6-carboxylate

4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (540 mg) and 4-(2-amino-5-fluorophenoxy)-2-methylpentan-2-ol (500 mg) were dissolved in dioxane (20 ml) and p-toluenesulfonic acid monohydrate (70 mg) was added. The reaction was heated under reflux for 2h. Then the reaction mixture was allowed to cool to room temperature and concentrated in vacuo. The residue was dissolved in dichloromethane and washed with water. The organic layer was separated, dried, filtered and evaporated. The residue triturated with methanol and the precipitate was filtered to give the intended product.

Yield: 710 mg

ESI mass spectrum: m/z = 434 (M+H) +

Retention time HPLC: 2.02 min

HPLC method: A 9 20.2 4-(4-fluoro-2-(4-hvdroxy-4-methylpentan-2-yloxy)phenylamino) -5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid

Methyl 4-(4-fluoro-2-(4-hydroxy-4-methylpentan-2-yloxy)phenylamino) -5-methyl- thieno[2,3-d]pyrimidine-6-carboxylate (710 mg) was dissolved in methanol (50 ml) and aqueous NaOH (4M; 5ml) was added. The reaction was stirred at room temperature overnight and then heated at 60°C for 2h. Finally the solution was neutralised and concentrated in vacuo. The residue was triturated with water. The resultant precipitate was filtered and dried. Yield: 600 mg

ESI mass spectrum: m/z = 420 (M+H) +

Retention time HPLC: 1.75 min

HPLC method: A_9

20.3 N-(3-(Dimethylamino)propyl)-4-(4-fluoro-2-(4-hvdroxy-4-methy lpentan-2- yloxy)phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-carbox amide

trifluoroacetate

4-(4-Fluoro-2-(4-hydroxy-4-methylpentan-2-yloxy)phenylami no)-5-methyl-thieno[2,3- d]pyrimidine-6-carboxylic acid (100 mg) was dissolved in DMF (5 ml) and triethylamine (75 μΙ) was added. Then TBTU (80 mg) was added and the reaction was stirred for 5 minutes. Followed by N,N-dimethyl-1,3-propanediamine (35 μΙ) the reaction mixture was stirred at room temperature for 2h. An further aliquot of N,N- dimethyl-1,3-propanediamine and TBTU (40 mg) was added. The mixture was heated at 60°C and stirred for 2h. Then the reaction was concentrated in vacuo. The residue was dissolved in dichloromethane and washed with soda solution. The organic phase was purified by RP-chromatography (water with 0.2% trifluoroacetic acid/ methanol 72-100%) .

Yield: 132 mg

ESI mass spectrum: m/z = 504 (M+H) +

Retention time HPLC: 1.18 min

HPLC method: A 9 Compound 21 5- Methyl-4-(2-(1 ,1 -triflouropropan-2yloxy)pyridin-3-ylamino)^

6- carboxamide

21 .1 Methyl 5-methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyndin-3- ylamino)thieno[2,3-dlpyrinnidine-6-carboxylate

Prepared analogously to example 1 .1 by heating under microwave irradiation at

1 10°C using 2-(1 ,1 ,1 -trifluoropropan-2-yloxy)pyridin-3-amine and 4-Chloro-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester

Yield: 270 mg

ESI mass spectrum: m/z = 413 (M+H) +

Retention time HPLC: 3.37 min

HPLC method: AC 1 21 .2 5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyridin-3-ylamino)thienor2,3- dlpyrimidine-6-carboxylic acid

Prepared analogously to example 1 .2 using Methyl 5-methyl-4-(2-(1 ,1 ,1 - trifluoropropan-2yloxy)pyridin-3-ylamino)thieno[2,3-d]pyrimi dine-6-carboxylate

Yield: 278 mg

ESI mass spectrum: m/z = 399 (M+H) +

Retention time HPLC: 3.25 min

HPLC method: AC 1 21 .3 5-Methyl-4-(2-(1 ,1 ,1 -tnfluoropropan-2yloxy)pyndin-3-ylamino)thieno[2,3- dlpyrinnidine-6-carboxannide

5-Methyl-4-(2-(1 ,1 ,1 -trifluoropropan-2yloxy)pyndin-3-ylamino)thieno[2,3-d]pynmid ine- 6-carboxylic acid (278 mg), HATU (320 mg) and Hunig ' s Base (144 μΙ) were dissolved in DMF (6.5 ml) at 0°C. The reaction was stirred for 30 minutes after this time NH 3 in dioxane (0.5 M; 5.3 ml) was added. The reaction was allowed to warm to room temperature overnight. The reaction mixture was concentrated in vacuo. The residue was extracted with dichloromethane and water. The organic layer was separated, dried and evaporated. The crude product was purified by chromatography (silica / dichloromethane: methanol: NH 3 90:10:1 ).

Yield: 124 mg

ESI mass spectrum: m/z = 398 (M+H) +

Retention time HPLC: 3.93 min

HPLC method: AC 1

Further analogues of example 21 :

Prepared analogously to example 21 .3 using 5-methyl-4-(2-(1 ,1 ,1 -trifluoropropan 2yloxy)pyridin-3-ylamino)thieno[2,3-d]pyrimidine-6-carboxyli c acid and the

corresponding amine

amide

Compound 22 4-(2-(1 ,3-Diethoxypropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxamide

22.1 Methyl 4-(2-(1 ,3-diethoxypropan-2-yloxy)-4-fluorophenylannino)-5-nnethyl- thieno[2,3-dlpyrimidine-6-carboxylate

Prepared analogously to example 18.1 using 4-Chloro-5-methyl-thieno[2,3- d]pyhmidine-6-carboxylic acid methyl ester and 2-(1 ,3-diethoxypropan-2-yloxy)-4- fluoroaniline.

The solvent dioxane was used instead of isopropanol and p-toluonesulfonic acid monohydrate was added.

Yield: 590 mg

ESI mass spectrum: m/z = 464 (M+H) +

Retention time HPLC: 3.75 min

HPLC method: A 10

22.2 4-(2-(1 ,3-Diethoxypropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyhmidine-6-carboxylic acid

Prepared analogously to example 1 .2 using methyl 4-(2-(1 ,3-diethoxypropan-2- yloxy)-4-fluorophenylamino)-5-methylthieno[2,3-d]pyrimidine- 6-carboxylic acid. The reaction was performed at 80°C.

Yield: 390 mg

ESI mass spectrum: m/z = 450 (M+H) +

Retention time HPLC: 1 .91 min

HPLC method: A 10

22.3 4-(2-(1 ,3-Diethoxypropan-2-yloxy)-4-fluorophenylamino)-5-methylthie no[2,3- dlpyhmidine-6-carboxamide

4-(2-(1 ,3-Diethoxypropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- d]pyrimidine-6-carboxylic acid (150 mg), TBTU (107, 2 mg) and Hunig ' s base (122 μΙ) were dissolved in THF (10 ml), ammonia (0,5M; 700μΙ) was added and the mixture was stirred at room temperature overnight. Then it was evaporated and the residue dissolved in dichloromethane. The mixture was extracted with water and aqueous NaOH (0.5M). The organic layer was dried and concentrated in vacuo. The residue was triturated with Et 2 O and a few drops of ethanol .

Yield: 105 mg

ESI mass spectrum: m/z = 449 (M+H) +

Retention time HPLC: 2.02 min

HPLC method: A_10

Compound 23

4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxamide 23.1 Methyl 4-(2-( 1 ,3-difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl- thieno[2,3-dlpyrinnidine- -carboxylate

Prepared analogously to example 21 .1 using 2-(1 ,3-difluoropropan-2-yloxy)-4- fluoroaniline and 4-Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester.

Yield: 3.7 g

ESI mass spectrum: m/z = 412 (M+H) +

Retention time HPLC: 3.37 min

HPLC method: AC 1

23.2 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxylic acid

Prepared analogously to example 1 .2 using methyl 4-(2-(1 ,3-difluoropropan-2-yloxy)-

4-fluorophenylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-c arboxylate

Yield: 1 .93 g

ESI mass spectrum: m/z = 398 (M+H) +

Retention time HPLC: 2.93 min

HPLC method: AC 1

23.3 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thi eno[2,3- dlpyhmidine-6-carboxamide

Prepared analogously to example 21 .3 using 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carbox ylic acid

Yield: 180 mg

ESI mass spectrum: m/z = 397 (M+H) +

Retention time HPLC: 2.70 min

HPLC method: AC 1

Further analogues of compound 23 :

The compounds listed in Table 6 were synthesized analogously to example 23.3 using 4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methylthie no[2,3- d]pyrimidine-6-carboxylic acid and the corresponding amine.

Table 6:

carboxamide Compound 24

4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methyl-N- (methylsulfonv)thieno[2,3-dlpyhmidine-6-carboxamide

4-(2-(1 ,3-Difluoropropan-2-yloxy)-4-fluorophenylamino)-5-methylthie no[2,3- d]pyhmidine-6-carboxylic acid (197 mg), 4-dimethylaminopyhdine (76 mg) and 1 - ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (125 mg) was dissolved in dichloromethane (20 ml). Methanesulfonamide (59.5 mg) was added and the reaction was stirred at room temperature for about 24h. The mixture was washed with a KHSO 4 solution. The organic layer was separated, dried and concentrated in vacuo. The residue was purified by chromatography (dichloromethane: methanol: NH 3 80:20:1 ). The fractions were collected and triturated with diisopropyl ether. Yield: 57 mg

ESI mass spectrum: m/z = 475 (M+H) +

Retention time HPLC: 3.1 1 min

HPLC method: AC 1

Compound 25

N-((trans)-2-Aminocvclopropyl)-4-(2-(1 ,3-difluoropropan-2-yloxy)-4- fluorophenylamino)-5-methylthieno 2,3-dlpyrimidine-6-carboxamide hydrochloride

tert-Butyl (trans)-2-(4-(2-(1 ,3-difluoropropan[2,3-d]pyrimidine-6-carboxamido)cyclo propyl carbamate (130 mg) was dissolved in dioxane (8 ml) and a solution of HCI in dioxane (4M; 1 .34 ml) was added. The reaction was stirred at room temperature overnight. Diethyl ether was added and the precipitate was filtered and washed with Et 2 O to give the title compound.

Yield: 97 mg

ESI mass spectrum: m/z = 452 (M+H) +

Retention time HPLC: 2.10 min

HPLC method: AC 1

Compound 26

N-(3-(Dimethylamino)propyl-4-(4-fluoro-2-((tetrahvdro-2-H -pyran-4- yl)methoxy)phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-c arboxamide

26. 1 Methyl-4-(4-fluoro-2-((tetrahvdro-2-H-pyran-4-yl)methoxy)phe nylamino)-5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carboxylate

Methyl 4-(4-fluoro-2-hydroxyphenylamino)-5-methyl-thieno[2,3-d]pyri midine-6- carboxylate (100 mg) was dissolved in DMF (2 ml), 4-bromomethyltetrahydropyran (107 mg) and potassium carbonate (83 mg) were added. The reaction was stirred at 60°C overnight. Then the reaction mixture was diluted with EtOAc and washed with water and brine. The solvent was removed in vacuo and the residue triturated with Et 2 O.

Yield: 130 mg

ESI mass spectrum: m/z = 432 (M+H) + 26. 2 4-(4-Fluoro-2-((tetrahvdro-2-H-pyran-4-yl)methoxy)phenylamin o)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid

Methyl-4-(4-fluoro-2-((tetrahydro-2-H-pyran-4-yl)methoxy) phenylamino)-5- methylthieno[2,3-d]pyrinnidine-6-carboxylate (82 mg) and NaOH (2M; 0.5 ml) was dissolved in ethanol (2 ml). The reaction was stirred at room temperature overnight. Then the reaction mixture was diluted with dichloromethane and HCI (2M). The suspension was filtered, washed with brine and ether. The residue was dried in vacuo over P2O5.

Yield: 41 mg

26.3 N-(3-(Dimethylamino)propyl-4-(4-fluoro-2-((tetrahvdro-2-H-py ran-4- yl)methoxy)phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-c arboxamide

HATU (36 mg) was added to a solution of 4-(4-fluoro-2-((tetrahydro-2-H-pyran-4- yl)methoxy)phenylamino)-5-methylthieno[2,3-d]pyrimidine-6-ca rboxylic acid (36mg) and DIPEA (17 μΙ) in DMF (1 ml). The reaction mixture was stirred for 40min at room temperature. Then 3-(dimethylamino)propylamine (49 μΙ) was added. The reaction mixture was stirred at room temparture overnight. It was diluted with EtOAc and washed with water and brine. The organic phase was concentrated in vacuo. The residue was suspended in Et 2 O and filtered and dried. Yield: 40 mg

ESI mass spectrum: m/z = 502 (M+H) + Example 27

4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-N-(2-hvdroxyethyl)- 5-methylthieno[2,3- dlpyrimidine-6-carboxamide

27. 1 Methyl-4-(2-(2-fluoropropoxy)pyridin-3-ylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxylate

2-(2-Fluoropropoxy)pyridin-3-amine (153 mg) was dissolved in dioxane (30 ml), 4- Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (174.54 mg) and HCI in dioxane (4M; 25 μΙ) was added. The reactin was stirred at 60°C for three days. The reaction was concentrated in vacuo.

Yield: 333 mg

Retention time HPLC: 2.37 min

HPLC method: 002 CC ZQ4 27. 2 4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-5-methyl-thieno[2,3 -dlpyhmidine-6- carboxylic acid

To a solution of methyl-4-(2-(2-fluoropropoxy)pyridin-3-ylamino)-5-methyl-thi eno[2,3- d]pyhmidine-6-carboxylate (333 mg) in THF (10 ml) was added lithium hydroxide (1 M; 8 ml) and the reaction stirred at room temperature overnight. The mixture was neutralized with HCI (1 M; 8 ml) and concentrated in vacuo. The crude product was purified by chromatography.

Yield: 196 mg

ESI mass spectrum: m/z = 363 (M+H) +

Retention time HPLC: 1 .91 min

HPLC method: 003 CC ZQ7

27.3 4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-N-(2-hvdroxyethyl)- 5- methyl-thieno[2,3-dlpyhmidine-6-carboxamide

4-(2-(2-Fluoropropoxy)pyridin-3-ylamino)-5-methylthieno[2,3- d]pyrimidine-6- carboxylic acid (36.2 mg), HATU (42 mg) and Hunig ' s base (34 μΙ) were dissolved in

DMF (2 ml) and ethanolamine (9 μΙ) was added. The reaction was stirred at room temperature for 2h. The reaction mixture was purified directly by chromatography.

Yield: 31 mg

ESI mass spectrum: m/z = 406 (M+H) +

Retention time HPLC: 2.09 min

HPLC method: 004_CC_ZQ6

Compound 77.4:

Prepared analogously to example 27.3 using 4-(2-(2-Fluoropropoxy)pyridin-3- ylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid and the corresponding amine. tion time method

N-(3-(dimethylamino)- propyl)-4-(2-(2-fluoro- propoxy)pyridin-3-

26 004_C

ylamino)-5-methyl- 447 1 .75

mg C_ZQ6 thieno[2,3-d]pyrimidine-

N— 6-carboxamide

/

hydrochloride

Compound 28

4-(2-(2-Methoxypropan-2-yloxy)pyridin-3-ylamino)-5-methylthi eno[2,3-dlpynmidine-6- carboxamide

28. 1 Methyl-4-(2-(1 -methoxypropan-2-yloxy)pyridin-3-ylamino)-5-methyl-thieno[2, 3- dlpyhmidine-6-carboxylate

Prepared analogously to example 27.1

Yield: 344 mg

Retention time HPLC: 2.40 min

HPLC method: 002_CC_ZQ4

28. 2 4-(2-(1 -Methoxypropan-2-yloxy)pyridin-3-ylamino)-5-methylthieno[2,3 - dlpyhmidine-6-carboxylic acid

Prepared analogously to example 27.2 using methyl-4-(2-(1 -methoxypropan-2- yloxy)pyridin-3-ylamino)-5-methylthieno[2,3-d]pyrimidine-6-c arboxylate

Yield: 1 19 mg

ESI mass spectrum: m/z = 375 (M+H) +

Retention time HPLC: 1 .90 min

HPLC method: 003 CC ZQ7

4-(2-(2-Methoxypropan-2-yloxy)pyridin-3-ylamino)-5-methyl -thieno[2,3- dlpyhmidine-6-carboxamide

Prepared analogously to example 27.3 using 4-(2-(1 -methoxypropan-2-yloxy)pyridin-

3-ylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid (37 mg) and ammonia in dioxane (0.5M; 2 ml)

Yield: 23 mg

ESI mass spectrum: m/z = 374 (M+H) +

Retention time HPLC: 2.13 min

HPLC method: 004 CC ZQ6

Further analogues of 28:

Prepared analogously to example 29.3 using 4-(2-(1 -methoxypropan-2-yloxy)pyridin- 3-ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylic acid and the corresponding amine 28. N-(2-hydroxy-

4 ethyl)-4-(2-(1- methoxy- propan-2- x ; yloxy)pyridin- 16 004_C OH

418 2.1 1

3-ylamino)-5- mg C_ZQ6 H 2 N methylthieno- [2,3-d]- pyrimidine-6- carbox-amide

28. N-(3-(di-

5 methylamino)

propyl )-4-(2- (1-methoxy- rV° propan-2- N yloxy)pyridin- 30 004_C

459 1 .75

3-ylamino)-5- mg C_ZQ6 methlthieno[2,

/

3-d]pyrimi- dine-6- carboxamide

hydrochloride

Compound 29

4-(2-(2-Hvdroxyethoxy)pyridin-3-ylamino)-5-methyl-thieno[2,3 -dlpynmidine-6- carboxamide

29. 1 Methyl-4-(2-(2-hvdroxyethoxy)pyridin-3-ylamino)-5-methyl-thi eno[2,3- dlpyrimidine-6-carboxylate

Prepared analogously to example 28.1 from 2-(3-aminopyridin-2-yloxy)ethanol (175 mg) and chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (121 mg)

Yield: 475 mg

Retention time HPLC: 2.18 min

HPLC method: 002 CC ZQ4

29. 2 4-(2-(2-Hvdroxyethoxy)pyridin-3-ylamino)-5-methyl-thieno[2,3 -dlpyhmidine-6- carboxylic acid

Prepared analogously to example 28.1 from methyl-4-(2-(2-hydroxyethoxy)pyridin-3- ylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylate

Yield: 61 mg

Retention time HPLC: 2.14 min

HPLC method: 003 CC ZQ7

29. 3 4-(2-(2-Hvdroxyethoxy)pyridin-3-ylamino)-5-methylthieno[2,3- dlpyrimidine-6- carboxamide

Prepared analogously to example 28.3 using 4-(2-(2-hydroxyethoxy)pyridin-3- ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylic acid (30.5mg) and ammonia in dioxane (0.5M; 1 .6 ml)

Yield: 13 mg ESI mass spectrum: m/z = 346 (M+H) +

Retention time HPLC: 1 .87 min

HPLC method: 004_CC_ZQ6 Compound 29.4:

Prepared analogously to example 29.3 using 4-(2-(2-hydroxyethoxy)pyridin-3- ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylic acid and the corresponding amine

Compound 30

4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-5-methyl-thieno[ 2,3-dlpyhmidine-6- carboxamide 30. 1 Methyl-4-(2-(2,2-difluoroethoxy)pyridin-3-ylamino)-5-methyl- thienor2,3- dlpyrimidine-6-carboxylate

Prepared analogously to example 28.1 from 2-(2,2-Difluoroethoxy)pyridin-3-amine (171 mg) and Chloro-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (190 mg)

Yield: 376 mg

Retention time HPLC: 2.35min

HPLC method: 002 CC ZQ4

30. 2 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-5-methylthieno[2 ,3-dlpyhmidine-6- carboxylic acid

Prepared analogously to example 28.2 using methyl-4-(2-(2,2-difluoroethoxy)pyrid

3-ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylate (376 mg).

Yield: 220 mg

ESI mass spectrum: m/z = 367 (M+H) +

Retention time HPLC: 1 .91 min

HPLC method: 007 CC ZQ7

30. 3 4-(2-(2,2-Difluoroethoxy)pyridin-3-ylamino)-5-methyl-thieno[ 2,3-dlpyhmidine-6- carboxamide

Prepared analogously to example 28.3 from 4-(2-(2,2-difluoroethoxy)pyridin-3- ylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid (36.6mg) and ammonia in dioxane (0.5M; 2 ml). Yield: 18mg

ESI mass spectrum: m/z = 366 (M+H) +

Retention time HPLC: 2.09min

HPLC method: 004_CC_ZQ6

Further analogues of 30:

Prepared analogously to example 30.3 from 4-(2-(2,2-Difluoroethoxy)pyridin-3- ylamino)-5-methylthieno[2,3-d]pyrimidine-6-carboxylic acid and the corresponding amine

Compound 31

4-(2-(1 -(Ethylamino)-1 -oxopropan-2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3 - dlpyhmidine-6-carboxamide

31 . 1 Methyl-4-(2-(1 -tert-butoxy-1 -oxopropan-2-yloxy)-4-fluorophenylamino)-5- methyl-th ieno[2 ,3-dlpyhm id ine-6-carboxylate

Methyl 4-(4-fluoro-2-hydroxyphenylamino)-5-methyl-thieno[2,3-d]pyri midi^

carboxylate (100 mg) and Caesium carbonate (240 mg) was dissolved in acetonitrile (2 ml) and 2-bromopropionic acid tert-butyl ester (70 mg) was added. The reaction mixture was stirred at 60°C for 2h. Then the reaction was quenched with water and the precipitate was collected by filtration. The residue cake was washed with water and a few drops of methanol .

Yield: 103 mg

ESI mass spectrum: m/z = 462 (M+H) +

31 . 2 2-(5-Fluoro-2-(6-(methoxycarbonyl)-5-methylthieno[2,3-dlpyhm idin-4- ylamino)phenoxy)propanoic acid

Prepared analogously to example 8.1 using methyl-4-(2-(1 -tert-butoxy-1 -oxopropan- 2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3-d]pyrimidi ne-6-carboxylate (2.1 g) and 50% trifluoroacetic acid in dichloromethane (20 ml).

Yield: 1 .9 g

ESI mass spectrum: m/z = 406 (M+H) +

Retention time HPLC: 1 .96 min

HPLC method: A 9 31 . 3 Methyl 4-(2-(1 -(ethylamino)-l -oxopropan-2-yloxy)-4-fluorophenylamino)-5- methylthieno[2,3-dlpyrimidine-6-carboxylate

To a solution of 2-(5-fluoro-2-(6-(methoxycarbonyl)-5-methyl-thieno[2,3-d]pyr imidin-4- ylamino)phenoxy)propanoic acid (500 mg) and TBTU (400mg) in acetonitrile (10 ml) was added triethylamine (430 μΙ). The mixture was stirred for 20 min. Then

ethylamine (2M; 1 .5 ml) was added and the mixture was stirred at room temperature overnight. Then the reaction mixture was diluted with methanol and purified by chromatography.

Yield: 360 mg

ESI mass spectrum: m/z = 433 (M+H) +

Retention time HPLC: 1 .89 min

HPLC method: A 9 31 . 4 4-(2-(1 -(Ethylamino)-1 -oxopropan-2-yloxy)-4-fluorophenylamino)-5-methyl- thieno[2,3-dlpyrimidine-6-carboxylic acid

Prepared analogously to example 1 .2 from methyl 4-(2-(1 -(ethylamino)-1 -oxopropan- 2-yloxy)-4-fluorophenylamino)-5-methyl-thieno[2,3-d]pyrimidi ne-6-carboxylate (360 mg).

The reaction stirred at 50°C overnight. Yield: 299 mg

ESI mass spectrum: m/z = 419 (M+H) +

Retention time HPLC: 2.65 min

HPLC method: A_4

31 . 5 4-(2-(1 -(Ethylamino)-1 -oxopropan-2-yloxy)-4-fluorophenylamino)-5-methyl- tieno[2,3-dlpyrimidine-6-carboxamide

Prepared analogously to example 1 .3 using 4-(2-(1 -(ethylamino)-1 -oxopropan-2- yloxy)-4-fluorophenylamino)-5-methylthieno[2,3-d]pyhmidine-6 -carboxylic acid (60 mg) and ammonia in THF (0.5 M; 600μΙ).

Yield: 25 mg

ESI mass spectrum: m/z = 418 (M+H) +

Retention time HPLC: 1 .41 min

HPLC method: A_9

Further analogues to 31 :

Prepared analogously to example 32.5 from the corresponding amine and 4-(2-(1 - (ethylamino)-1 -oxopropan-2-yloxy)-4-fluorophenylamino)-5-methylthieno[2,3- d]pyrimidine-6-carboxylic acid

31.6 4-(2-(1-

(ethylamino)-

1 -οχο-

'-'Ν propan-2- yloxy)-4- fluorophenyla 30

mino)-N,5- 432 1 .65 A_9

dimethyl- mg

thieno[2,3- d]pyrimidine-

S Ν— 6-carbox- amide

31.7 N-(3-(di- methylamino)

propyl )-4-(2-

(1-(ethyl- amino)-1- N oxo-propan- 32

2-yloxy)-4- 503 1 .97 A_9

fluorophenyla mg

mino)-5- methylthieno[

Ν— 2,3-d]pyrimi-

CI dine-6- carboxamide

hydrochloride

Compound 32

Methyl 4-(2-(1 -(2-(dimethylamino)ethylamino)-1 -oxopropan-2-yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-dlpyhmidine-6-carboxy late

32. 1 Methyl 4-(2-( 1 -(2-(dimethylamino)ethylamino)-1 -oxopropan-2-yloxy)-4- fluorophenylamino)-5-methyl-thieno[2,3-dlpyhmidine-6-carboxy late

Prepared analogously to example 32.3 from 2-(5-fluoro-2-(6-(methoxycarbonyl)-5- methylthieno[2,3-d]pyrimidin-4-ylamino)phenoxy)propanoic acid (200 mg) and 2- dimethylaminoethylamine (70 μΙ) Yield: 136 mg

ESI mass spectrum: m/z = 476 (M+H) +

Retention time HPLC: 2.17 min

HPLC method: A 4

Compounds 32.2- 32.35:

Step 1

Methyl 4-(4-fluoro-2-hydroxyphenylamino)-5-methylthieno[2,3-d]pyhmi dine-6- carboxylate (667 mg) and the corresponding alcohol (e.g ethylen glycol 3eq = 372 mg) were dissolved in THF (30 ml). Under a nitrogen atmosphere triphenylphosphine (1 .1 g) and diisopropyl azodicarboxylate (844 μΙ) were added and the reaction was stirred at room temperature for three days. An additional aliqout of the alcohol, diisopropyl azodicarboxylate and triphenylphosphine was added and the reaction was stirred overnight. Then the reaction mixture was filtered and washed with dichloromethane: methanol 9:1 . The filtrate was concentrated in vacuo and the residue was purified by chromatography to give the intermediate esters

Step 2 :

The intermediate esters (step 1 ) were dissolved in a mixture of methanol (10 ml)/ THF (5 ml) and NaOH (1 M; 5ml) was added. The reaction was stirred at room temperature for three days. The solvent was concentrated in vacuo and the residue was neutralized with HCI (1 M; 5ml) and the precipitate was collected by filtration. The residue cake was dried, suspended in acetonitrile, acidified with HCI and

concentrated in vacuo to give the intermediate acids (a-l)

Step 3

The acid a and Hunig ' s base (25μΙ) was dissolved in DMF (3 ml). After a few minutes HATU (42 mg) was added. To this mixture N,N- dimethyl-1 ,3-propanediamine (19 μΙ) was added. The reaction mixture was stirred at room temperature overnight. The mixture was purified by chromatography to yield compound 35.

The following compounds were synthesized in an analogue manner:

b I I 486 1 .78 002_CC_ZQ4 H 1

/H

c ' N

1 383 2.01 002_CC_ZQ4

H c ' N

1 427 2.00 002_CC_ZQ4

H c 1 I 468 1 .77 002_CC_ZQ4

H 1 d ' N

1 453 2.07 002_CC_ZQ4

H

/H

d ' N

1 409 2.07 002_CC_ZQ4

H d 1 I 494 1 .82 002_CC_ZQ4

H 1 e ' N

1 453 2.02 002_CC_ZQ4

H e 1 I 494 1 .79 002_CC_ZQ4

H 1

/H

e ' N

1 408 2.04 002_CC_ZQ4

H f 1 I 464 1 .80 002_CC_ZQ4 H 1 f ' N

1 422 2.03 002_CC_ZQ4

H

/H

f ' N

1 378 2.05 002_CC_ZQ4

H

' N

g 1 451 1 .87 002_CC_ZQ4

H

/H

' N

g 1 407 1 .89 002_CC_ZQ4

H g 1 I 492 1 .67 002_CC_ZQ4

H 1 32.32 h ' N

I 363 1 .97 002_CC_ZQ4

H

32.33 h ' N

I 407 1 .96 002_CC_ZQ4

H

32.34 h I I 448 1 .73 002_CC_ZQ4

H 1

32.35 i 1 I 476 1 .81 002_CC_ZQ4

H 1

32.36 i ' N

1 435 2.06 002_CC_ZQ4

H

/H

32.37 i ' N

1 391 2.08 002_CC_ZQ4

H

32.38 j 1 I 500 1 .80 002_CC_ZQ4

H 1

32.39 k 1 I 464 1 .8 002_CC_ZQ4

H 1

32.40 I 1 I 514 2.71 003_CC_ZQ7

H 1

32.41 I 526 2.68 003_CC_ZQ7

32.42 I 540 2.81 003_CC_ZQ7

The acid f and Hunig ' s base (15μΙ) was dissolved in DMF (2 ml). After a few minutes TBTU (21 .2 mg) was added. To this mixture was added the amin (0.07 mmol). The reaction mixture was stirred at room temperature overnight. The mixture was purified by chromatography.

The following compounds were synthesized in an analogue manner:

Compound 33

4-(2-((1 -(Cvanomethyl)cvclopropyl)methoxy)pyridin-3-ylamino)-N-(3- (dimethylamino)propyl)-5-methylthieno[2,3-dlpyrimidine-6-car boxamide hydrochloride

33. 1 Methyl 4-(2-((1 -(cvanomethyl)cvclopropyl)methoxy)pyridin-3-ylamino)-5- methylthieno[2,3-dlpyhmidine-6-carboxylate

Prepared analogously to example 32.1 using 2-(1 -((3-aminopyridin-2- yloxy)methyl)cyclopropyl)acetonitrile (264 mg) and chloro-5-methyl-thieno[2,3- d]pyrimidine-6-carboxylic acid methyl ester (152 mg).

Retention time HPLC: 2.13 min

HPLC method: 007_CC_ZQ5

33. 2 4-(2-((1 -(Cvanomethyl)cvclopropyl)methoxy)pyridin-3-ylamino)-5- methylthieno[2,3-dlpyhmidine-6-carboxylic acid

Prepared analogously to example 32 Step 2 using methyl 4-(2-((1 -(cyanomethyl) cyclopropyl)methoxy)pyridin-3-ylamino)-5-methylthieno[2,3-d] pyrimidine-6- carboxylate

Yield: 85mg

Retention time HPLC: 2.23 min

HPLC method: 003_CC_ZQ6

33.3 4-(2-((1 -(Cvanomethyl)cvclopropyl)methoxy)pyridin-3-ylamino)-N-(3-

(dimethylamino)propyl)-5-methyl-thieno[2,3-dlpyrimidine-6 -carboxamide hydrochloride

Prepared analogously to example 32.3 using 4-(2-((1 -(cyanomethyl)cyclopropyl) methoxy)pyridin-3-ylamino)-5-methyl-thie^ acid (42,5 mg) and N,N-dimethyl-1 ,3-propanediamine (17 mg).

Yield: 42.7 mg

ESI mass spectrum: m/z = 480 (M+H) +

Retention time HPLC: 1 .69 min

HPLC method: 004_CC_ZQ6

Compound 33.4:

Prepared analogously to example 32.3 using 4-(2-((1 -(cyanomethyl)cyclopropyl)- methoxy)pyridin-3-ylamino)-5-methylthieno[2,3-d]pyrimidine-6 -carboxylic acid and ammonia.

Compound 34

4-(2-((2,2-Difluorocvclopropyl)methoxy)pyridin-3-ylamino)-N- (3- (dimethylamino)propyl)-5-methylthieno[2,3-dlpyrimidine-6-car boxamide hydrochloride

34. 1 Methyl 4-(2-((2,2-difluorocvclopropyl)methoxy)pyridin-3-ylamino)-5- methylthieno[2,3-dlpyhmidine-6-carboxylate

Prepared analogously to example 32.1 using 2-((2,2- Difluorocyclopropyl)methoxy)pyridin-3-amine (200 mg) and chloro-5-methyl- thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester (194 mg).

Yield: 57 mg

Retention time HPLC: 2.49 min

HPLC method: 004 CC ZQ6

34. 2 4-(2-((2,2-Difluorocvclopropyl)methoxy)pyridin-3-ylamino)-5- methyl-thieno[2,3- dlpyrimidine-6-carboxylic acid

Prepared analogously to example 32 Step 2 using methyl 4-(2-((2,2-difluoro- cyclopropyl)methoxy)pyridin-3-ylamino)-5-methylthieno[2,3-d] pyrimidine-6- carboxylate (56 mg)

Yield: 1 10mg

Retention time HPLC: 2.37 min

HPLC method: 004 CC ZQ6

34. 3 4-(2-((2,2-Difluorocvclopropyl)methoxy)pyridin-3-ylamino)-N- (3- (dimethylamino)propyl)-5-methyl-thieno[2,3-dlpyrimidine-6-ca rboxamide hydrochloride

Prepared analogously to example 32.3 from 4-(2-(2,2)difluorocyclopropyl)- methoxy)pyridin-3-ylamino)-5-methylthieno[2,3-d]pyrimidine-6 -carboxylic acid (27 mg) and N,N-dimethyl-1 ,3-propanediamine (1 1 mg).

Yield: 8.6 mg

ESI mass spectrum: m/z = 477 (M+H) +

Retention time HPLC: 1 .83min

HPLC method: 004_CC_ZQ6

Compound 34.4:

Prepared analogously to example 32.3 using 4-(2-((2,2- difluorocyclopropyl)methoxy)pyridin-3-ylamino)-5-methylthien o[2,3-d]pyhmidine-6- carboxylic acid

Compound 35

4-(2-lsopropoxypyridin-3-ylamino)-5-methylthieno[2,3-dlpyhmi dine-6-carboxyamide 35.1 Methyl 4-(2-isopropoxypyridin-3-ylamino)-5-me

carboxylate

2-lsopropoxypyridin-3-amine (1 .3 g) was added to a solution of 4-Chloro-5-methyl- thieno[2,3-d]pyhmidine-6-carboxylic acid methyl ester (2.3 g), Xantphos (995.2 mg) and Cesium carbonate (3.9 g) in degassed dioxane (40 ml). The mixture was again degassed in a sonicator for 15 min before addition of Pd2(dba)3. The reaction mixture was then heated under a nitrogen atmosphere to 90°C for 3h. The mixture was cooled, filtered and the solid washed with dioxane. The residues were combined and poured into an ammonium hydroxide solution. The solid was filtered off to yield the desired product.

Yield: 2.05 g 35.2 4-(2-lsopropoxypyridin-3-ylamino)-5-methyl-thieno[2,3-dlpyhm idine-6- carboxylic acid

Methyl 4-(2-isopropoxypyridin-3-ylamino)-5-methyl-thieno[2,3-d]pyri midine-6- carboxylate (2.04 g) was dissolved in methanol (24.0 ml), NaOH (2M; 6.0 ml) was added and the mixture was stirred at 60°C for 1 h. Then the reaction was cooled and washed with dichloromethane. The aqueous layer was acidified to pH 5 with cone.

HCI and the precipitate was filtered and washed with water.

Yield: 1 .5 g

ESI mass spectrum: m/z = 345 (M+H) + 35.3 4-(2-lsopropoxypyridin-3-ylamino)-5-methyl-thie^

carboxamide

HATU (182.0 mg) was added to an ice cooled solution of 150 mg 4-(2- lsopropoxypyhdin-3-ylamino)-5-methyl-thieno[2,3-d]pyhmidine- 6-carboxylic acid and DIPEA (83.0 μΙ) in DMF (3.0 ml). The mixture was stirred at 0°C for 20 min then ammonia/ MeOH (7M;1 .5 ml) was added and it was allowed to warm to room temperature overnight. The mixture was diluted with EtOAc and washed with water, 2M NaOH and concentrated in vacuo. The residue was triturated with Et 2 O and filtered to give the title compound.

Yield: 90.0 mg

ESI mass spectrum: m/z = 344 (M+H) + Compound 36

N-(3-(Dimethylamino)propyl)-4-(2-isopropoxypyridin-3-ylamino )-5-methyl-thieno[2,3- dlpyhmidine-6-carboxamide

Prepared analogously to example 1 .3 using 4-(2-isopropoxypyridin-3-ylamino)-5- methylthieno[2,3-d]pyrimidine-6-carboxylic acid (descirbed in I.2) and 3- dimethylaminopropylamine. The resultant product was purified by chromatography.

Yield: 68 mg

ESI mass spectrum: m/z = 427 (M-H) Further analogues of 36

The compounds listed in the following table were synthesized analogously to example 1 .3 using 4-(2-isopropoxypyridin-3-ylamino)-5-methylthieno[2,3- d]pyrimidine-6-carboxylic acid and the corresponding amine

Table 1 :

N d]pyrimidine-6-

Compound 37

4-(4-Fluoro-2-isopropoxyphenylamino)-5-methyl-thieno[2,3-dlp yhmidine-6-carboxylic acid

37.1 Methyl 4-(4-fluoro-2-isopropoxyphenylamino)-5-methyl-thieno[2,3-dlp yhmidine- 6-carboxylate

4-Chloro-5-methyl-thieno[2,3-d]pyhmidine-6-carboxylic acid methyl ester (1 12 mg), 4- fluoro-2-isopropoxyaniline (77.9 mg) and p-toluenesulfonic acid monohydrate (15 mg) were dissolved in dioxane (3.0 ml). The reaction was heated at 1 10°C under microwave irradiation. Then the reaction mixture was concentrated in vacuo and the residue was dissolved in dichloromethane and extracted with water. The organic phase was dried and concentrated in vacuo. The residue was triturated with diisopropyl ether and filtered.

Yield: 106 mg

ESI mass spectrum: m/z = 376 (M+H) +

Retention time HPLC: 4.22 min

HPLC method: AC 1

37.2 4-(4-Fluoro-2-isopropoxyphenylamino)-5-methyl-thieno[2,3-dlp yhmidine-6- carboxylic acid

Methyl 4-(4-fluoro-2-isopropoxyphenylamino)-5-methyl-thieno[2,3-d]p yhmidine-6- carboxylate (451 mg) and lithium hydroxide (865 mg) was dissolved in a mixture of THF: methanol : water (1 :1 :1 ; 100ml) and was stirred at room temperatruer overnight. Then the mixture was acidified with cone. HCI, the resultant precipitate was filtered, dried and triturated with diisopropyl ether to afford the title compound.

Yield: 378 mg

ESI mass spectrum: m/z = 362 (M+H) +

Retention time HPLC: 2.72 min

HPLC method: AC 1 Compound 38

4-(4-Fluoro-2-isopropoxyphenylamino)-5-methyl-thieno[2,3-dlp yhmidine-6-carboxylic acid

Prepared analogously to example 35.3 using 4-(4-Fluoro-2isopropoxyphenylamino) 5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid (descirbed in 37.2) and

N,N-Dimetyhl-1 ,3-propanediamine.

Yield: 355 mg

ESI mass spectrum: m/z = 446 (M+H) +

Retention time HPLC: 2.72 min

HPLC method: W001 001 Compound 39

4-[2-(1 -Ethylcarbamoyl-ethoxy)-4-fluoro-phenylamino1-5-methyl-thien o[2,3-d1 pyrimidine-6-carboxylic acid

To the intermediate XVIII (0.36 g) in THF:methanol 1 :1 (20 ml) was added sodium hydroxide 1 M (2.00 ml) and stirred at 50°C overnight. Then the mixture was acidified by addition of hydrochloric acid and concentrated. The residue was diluted with water, filtrated and the solid was washed with diethylether.

Yield: 0.30 g

ESI mass spectrum: m/z = 419 (M+H) +

Retention time HPLC: 2.65 min

HPLC method: A_ 4

Compound 40 4-[2-(1 -Ethylcarbamoyl-ethoxy)-4-fluoro-phenylamino1-5-methyl-thien

Pyrimidine-6-carboxylic acid (3-dimethylamino-propyl)-amide

Prepared analogously to example 5 using 4-[2-(1 -ethylcarbamoyl-ethoxy)-4-fluoro- phenylamino]-5-methyl-thieno[2,3-d]pyrimidine-6-carboxylic acid (60 mg) and N,N- dimethyl-1 ,3-propanediamine (24 μΙ).

Yield: 0.32 g

ESI mass spectrum: m/z = 503 (M+H) +

Retention time HPLC: 1 .97 min

HPLC method: A 4

Compound 41

The compound listed in the following table was synthesized analogously to example 40 using 4-[2-(1 -ethylcarbamoyl-ethoxy)-4-fluoro-phenylamino]-5-methyl-thien o [2,3-d]pyrimidine-6-carboxylic acid (compound 39) and ethanolamine.

Compound 42

4-[2— (3-Amino-1 -methyl-propoxy)-pyhdin-3-ylamino1-5-methyl- thieno[2,3-d1 pyhmidine-6-carboxylic acid (2-hydroxy-ethyl)-amide

42.1 4- [2-(3-tert-Butoxycarbonylamiino-1 -methyl-propoxy)-pyridin-3-ylamino1

5- methyl-thieno[2,3-d1 pyrinnidine-6-carboxylic acid

To the product from 16.1 (0.20 g) in DMF (5 ml) was added CDI (75 mg) and stirred for 2 hours at 70°C. Then 2-amino-ethanol (28,8 μΙ) was added and stirred at rt overnight. The reaction mixture was concentrated. Purification was achieved by chromatography.

Yield: 0.16 g

ESI mass spectrum: m/z = 417 (M+H) +

Retention time HPLC: 2.18 min

HPLC method: A 10 Compound 43

4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-methyl- thieno[2,3-d1 pyhmidine-6-carboxylic acid (2-hydroxy-ethyl)-amide

Methanesulfonyl chloride (19 μΙ) was added to a mixture of the product from example 42 (0.1 1 g), DCM (5 ml) and DBU (75 μΙ). The mixture was stirred at rt overnight. The reaction mixture was concentrated and then purified by chromatography.

Yield: 9.8 mg

ESI mass spectrum: m/z = 495 (M+H) +

Retention time HPLC: 2.38 min

HPLC method: AC 1 Compound 44

4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-methyl- thieno[2,3-d1 pyhmidine-6-carboxylic acid

44.1 4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5- methyl-thieno[2,3-d1 pyhmidine-6-carboxylic acid methyl ester

Prepared analogously to example 43 using the product from 15.2.

Yield: 330.0 mg

ESI mass spectrum: m/z = 466 (M+H) +

44.2 4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-

Sodium hydroxid 1 M (3 ml) was added to a mixture of the product from example 44.1 (0.27 g) and MeOH (5 ml). The mixture was stirred at 50°C for 1 hour. Then the reaction mixture was neutralized with hydrochloric acid 1 M (3 ml) and concentrated.

The residue was diluted with water and extracted with DCM. The organic layer was washed with a sodium chloride solution, dried and concentrated.

Yield: 210.0 mg

ESI mass spectrum: m/z = 452 (M+H) +

Retention time HPLC: 1 .62 min

HPLC method: A 10

Compound 45

4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-methyl- thieno[2,3-d1 pyrimidine-6-carboxylic acid amide

A reaction mixture of the product from example 44.2 (50 mg), TBTU (36 mg), DIPEA (39 μΙ), ammonia 0.5M in THF (222 μΙ), THF (3 ml) was stirred at rt for 1 hour. Then the mixture was concentrated and then purified by chromatography.

Yield: 18.0 mg

ESI mass spectrum: m/z = 451 (M+H) +

Retention time HPLC: 1 .46 min

HPLC method: A 10 Compound 46

4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-methyl-

Prepared analogously to example 45 using the product from 44.2.

Yield: 62.0 mg

ESI mass spectrum: m/z = 536 (M+H) +

Retention time HPLC: 1 .14 min

HPLC method: A_ 10 Compound 47 4-[2-(3-Methanesulfonylamino-1 -methyl-propoxy)-pyridin-3-ylamino1-5-m

4-Chloro-5-methyl-thieno[2,3-d]pyrinnidine-6-carboxylic acid methyl ester (0.2 g), _4- amino-3-(2-fluoro-1 -fluoromethyl-ethoxy)-benzonitrile (0.17 g), p-toluenesulfonic acid monohydrate (0.16 g) and isopropanol (3.0 ml) were stirred at 90°C for 2 hours.

Then the mixture was poured in water and filtrated.The solid was washed with water and dried in an oven at 70°C.

Yield: 160.0 mg

ESI mass spectrum: m/z = 419 (M+H) +

Retention time HPLC: 2.05 min

HPLC method: AC 1

Compound 48

4-[4-Cvano-2-(2-fluoro-1 -fluoromethyl-ethoxy)-phenylamino1-5-methyl- thieno[2,3-d1 pyrimidine-6-carboxylic acid (3-pyrrolidin-1 -yl-propyl)-amide

48.1 4-[4-Cvano-2-(2-fluoro-1 -fluoromethyl-ethoxy)-phenylamino1-5-methyl- thieno[2,3-d1 pyrimidine-6-carboxylic acid

The product from example 47 (1 .4 g) was dissolved in THF (200 ml). 10 ml water and LiOH (0.48 g) were added. The mixture was stirred at rt for 4 days. Then the mixture was acidified with 10 % citric acid and concentrated. The residue was dried in an oven at 70°C.

Yield: 950.0 mg

48.2 4-[4-Cvano-2-(2-fluoro-1 -fluoromethyl-ethoxy)-phenylamino1-5-methyl- thieno[2,3-d1 pyrimidine-6-carboxylic acid (3-pyrrolidin-1 -yl-propyl)-amide

4-[4-Cyano-2-(2-fluoro-1 -fluoromethyl-ethoxy)-phenylamino]-5-methyl- thieno[2,3-d] pyrimidine-6-carboxylic acid (100 mg), HATU (1 14 mg)

and TEA (52 μΙ) were dissolved in DMF (4 ml). N,N-dimethyl-1 ,3-propanediamine (31 μΙ) was added. The mixture was stirred for 1 hour at rt. Then the mixture was poured in water and filtrated.The solid was washed with water and dried in an oven at 70°C. Yield: 75.0 mg

ESI mass spectrum: m/z = 515 (M+H) +

Retention time HPLC: 1 .95 min

HPLC method: AC 1

The following compounds were prepared analogously to example 48.2 using 4-[4- cyano-2-(2-fluoro-1 -fluoromethyl-ethoxy)-phenylamino]-5-methyl-thieno[2,3-d] pyrimidine-6-carboxylic acid and the corresponding amine.

Compound 54

4-[2-Cvano-methyl-methoxy)-4-fluoro-phenylamino1-5-methyl-th ieno[2,3-dlpyrimidine- 6-carboxylic acid amide

54.1 4-(4-Fluoro-2-hvdroxy-phenylamino)-5-methyl-thieno[2,3-dlpyr imidine-

6-carboxylic acid

Methyl 4-(4-fluoro-2-hydroxyphenylamino)-5-methyl-thieno[2,3-d]pyri midine-6- carboxylate (Intermediate III) (1 .0 g), sodium hydroxide 1 M (10 ml), methanol (20 ml) and THF (20 ml) were stirred at rt overnight. The mixture was concentrated and diluted with EE and water. The aqueous layer was acidified with hydrochloric acid and filtrated.

Yield: 800 mg

ESI mass spectrum: m/z = 320 (M+H) +

Retention time HPLC: 2.22 min

HPLC method: 004 CC ZQ6

54.2 4-(4-Fluoro-2-hvdroxy-phenylamino)-5-methyl-thieno[2,3-dlpyr imidine-

6-carboxylic acid amide

Prepared analogously to example 21 .3 using the product from 66.1

Yield: 30 mg

ESI mass spectrum: m/z = 319 (M+H) +

Retention time HPLC: 1 .9 min

HPLC method: 004 CC ZQ6 54.3 4-[2-Cvano-methyl-methoxy)-4-fluoro-phenylamino1-5-methyl-th ieno[2,3-

The product from 54.2 (30 mg), 2-bromopropionitrile (13 mg), cesium

carbonate (40 mg) inDMF (1 ml) were stirred at rt overnight. The mixture was filtrated and the solid was washed with DMF. Water was added to the filtrate. The solid was isolated by filtration. The combined solids were freeze-dried. Yield: 14 mg

ESI mass spectrum: m/z = 372 (M+H) +

Retention time HPLC: 1 .92 min

HPLC method: 004_CC_ZQ6

Compound 55

4-[2-(1 -Carboxy-ethoxy)-4-fluoro-phenylamino1-5-methyl-thieno[2,3-d 1

Pyrimidine-6-carboxylic acid methyl ester

55.1 4-[2-(1 -tert-Butoxycarbonyl-ethoxy)-4-fluoro-phenylamino1-5-

2-Bromopropionic acid tert-butyl-ester (1 .4 g) was added to methyl 4-(4-fluoro-2- hydroxyphenylamino)-5-methyl-thieno[2,3-d]pyrimidine-6-carbo xylate (Intermediate III) (2.00 g) and cesium carbonate (4.8 g) in ACN (50 ml) and stirred at 60°C for 2 hours. The mixture was diluted with water, filtrated and washed with water.

Yield: 2.1 g

ESI mass spectrum: m/z = 462 (M+H) +

Retention time HPLC: 2.12 min

HPLC method: A 9

55.2 4-[2-(1 -Carboxy-ethoxy)-4-fluoro-phenylamino1-5- methyl-thieno[2,3-dlpyhmidine-6-carboxylic acid methyl ester

A mixture of the product from 55.1 (2.1 g) and 50% trifluoroacetic acid in

DCM was stirred at rt overnight. Then the mixture was concentrated and

the residue was triturated with diethylether.

Yield: 1 .9 g

ESI mass spectrum: m/z = 406 (M+H) +

Retention time HPLC: 1 .96 min

HPLC method: A 9 Comound 56

4-{2-[1 -(3-Amino-propylcarbamoyl)-ethoxy1-4-fluoro-phenylamino)-5-m ethyl- thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

56.1 4-{2-[1 -(3-tert-Butoxycarbonylamino-propylcarbamoyl)-ethoxy1-4-fluo ro- phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

The product from 55 (170 mg) and TBTU (150 mg) was dissolved in ACN

(5 ml). TEA (150 μΙ) was added to the mixture. The mixture was stirred for 15 min. at rt. Then tert-butyl N-(3-Aminopropyl)carbamate (200 mg) was added. The mixture was stirred at 50°C for 2 days. Afterwards the mixture was diluted with water and filtrated. The solid was washed with water and dried in an oven

Yield: 176 mg

ESI mass spectrum: m/z = 562 (M+H) +

Retention time HPLC: 2.21 min

HPLC method: A 9

56.2 4-{2-[1 -(3-Amino-propylcarbamoyl)-ethoxy1-4-fluoro- phenylamino)-5-methyl-thieno[2, -dlpyrimidine-6-carboxylic acid methyl ester

A mixture of the product from 56.1 (30 mg) and 25% trifluoroacetic acid

DCM (2 ml) was stirred at rt for 4 hours. The mixture was concentrated

Yield: 25 mg

ESI mass spectrum: m/z = 462 (M+H) + Retention time HPLC: 1 .96 min

H PLC method: A 9

Comound 57

4-{2-[1 -(3-Amino-propylcarbamoyl)-ethoxy1-4-fluoro-phenylamino)-5-m ethyl-thieno [2,3-dlpyhmidine-6-carboxylic acid (2-hydroxy-ethyl)-amide

57.1 4-{2-[1 -(3-tert-Butoxycarbonylamino-propylcarbamoyl)-ethoxy1-4-fluo ro- phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-carboxylic acid

To the product from 55 (130 mg) in THF:MeOH=1 :1 (5 ml) was added

sodium hydroxide solution 1 M (580 μΙ) and stirred at 40°C overnight. The mixture was cooled down to rt and added Hydrochloric acid 1 M (580 ml). Then the methanol was concentrated and the residue with DCM diluted. The organic layer was seperated by a cartridge and concentrated. The residue was triturated with diethylether.

Yield: 1 13 mg 57.2 4-{2-[1 -(3-Amino-propylcarbamoyl)-ethoxy1-4-fluoro-phenylamino)-5-

The product from 57.1 (1 15 mg) and TBTU (70 mg) were dissolved in DMF (1 .5 ml). TEA (75 μΙ) was added and the mixture was stirred for 5 min. at rt.

Ethanolamine (20 μΙ) was added and the mixture was stirred at rt overnight.

Afterwards the mixture was diluted with methanol . Purification was achieved by chromatography. The combined fractions were concentrated. A mixture of

DCM:TFA=50:50 was added to the residue and the mixture was stirred at rt for 1 hour. Then the mixture was concentrated. A solution of hydrochloric acid in methanol was added and the mixture was concentrated.

Yield: 70 mg

ESI mass spectrum: m/z = 491 (M+H) +

Retention time HPLC: 1 .92 min

HPLC method: A 9

Compound 58

4-{2-[1 -(2-Amino-ethylcarbamoyl)-ethoxy1-4-fluoro- phenylamino)-5-methyl-thieno[2, -dlpyrimidine-6-carboxylic acid methyl ester

58.1 4-{2-[1 -(2-tert-Butoxycarbonylannino-ethylcarbannoyl)-ethoxy1-4-flu oro- phenylamino)-5-methyl-thieno[2,3-dlpyrimidine-6-carboxylic acid methyl ester

Prepared analogously to example 56.1 using the product from 55 (170 mg) and tert- butyl N-(2-aminoethyl)carbamate (90 μΙ).

Yield: 135 mg

ESI mass spectrum: m/z = 548 (M+H) + 58.2 4-{2-[1 -(2-Amino-ethylcarbamoyl)-ethoxy1-4-fluoro-phenylamino)-5-me thyl

Prepared analogously to example 56.2 using the product from 57.3 (170 mg).

Yield: 28 mg

ESI mass spectrum: m/z = 448 (M+H) +

Retention time HPLC: 1 .83 min

HPLC method: A 9

Compound 59 4-{2-[1 -(2-Amino-ethylcarbamoyl)-ethoxy1-4-fluoro-ph

dlpyrinnidine-6-carboxylic acid -hvdroxy-ethyl)annide

59.1 4-{2-[1 -(2-tert-Butoxycarbonylannino-ethylcarbannoyl)-ethoxy1-4-flu oro- phenylamino)-5-meth l-thieno[2,3-dlpyrimidine-6-carboxylic acid

Prepared analogously to example 57.1 using the product from 58 (90 mg)

Yield: 73 mg

ESI mass spectrum: m/z = 534 (M+H) +

59.2 r2-(2-{5-Fluoro-2-i6-(-hvdroxy-etvhlcabamoyl) -5- methyl-thieno[2,3-dlpyrimidin-4-ylamino1-phenoxy)-propionyla mino1- ethyll-carbamic acid tet-butyl ester

The product from 59.1 (73 mg) and TBTU (48 mg) were dissolved in DMF

(1 ml). TEA (48 μΙ) was added and the mixture was stirred for 5 min. at rt before ethanolamine (1 1 μΙ) was added. The mixture was stirred at rt overnight. The mixture was then diluted with methanol. Purification was achieved by chromatography.

Yield: 40 mg

ESI mass spectrum: m/z = 577 (M+H) +

Retention time HPLC: 1 .56 min

HPLC method: A 9

59.3 4-{2-[1 -(2-Amino-ethylcarbamoyl)-ethoxy1-4-fluoro-phenylamino)-5-

A mixture of the product from 59.2 (40 mg) and 25% trifluoroacetic acid in

DCM (5 ml) was stirred at rt for 4 hours. Methanol and a solution of hydrochloric acid in methanol were added. Then the mixture was concentrated.

Yield: 7 mg

ESI mass spectrum: m/z = 462 (M+H) +

Retention time HPLC: 1 .96 min HPLC method: A 9

Compound 60

4-[4-Fluoro-2-(2-hydroxy-1 ,2-dimethyl-propoxy)-phenylamino]-5-methyl-thieno[2,3- d]pyhmidine-6-carboxylic acid amide

4-Chloro-5-methyl-thieno[2,3-d]pyhmidine-6-carboxylic acid amide (intermediate

XXIV) (100 mg), intermediate XXVI (120 mg) and p-toluenesulfonic acid (5 mg) in dioxane (2 ml) were stirred at 100°C for 6 hours. Then the mixture was filtrated. The solid was washed with methanol .

Yield: 145 mg

ESI mass spectrum: m/z = 405 (M+H) +

Retention time HPLC: 1 .245 min

HPLC method: M2-SB-C18

Compound 61

4-[2-(1 -Ethylcarbamoyl-ethoxy)-4-fluoro-phenylamino)-5-methylthieno [2,3dlpyhmidine -6-carboxylic acid methyl ester

The product from 55 (500 mg) and TBTU (400 mg) were dissolved in ACN (10 ml). TEA (430 μΙ) was added and the mixture was stirred for 20 min. at rt before

Ethylamine (1 .5 ml) was added. The mixture was stirred at rt overnight. Then the mixture was diluted with methanol . Purification was achieved by chromatography. Yield: 360 mg

ESI mass spectrum: m/z = 433 (M+H) +

Retention time HPLC: 1 .89 min

H PLC method: A_ 9

The following compound was prepared analogously to example 61 using the product from 55 and the corresponding amine.

Compound 63

4-[2-Cvano-methyl-methoxy)-4-fluoro- phenylamino1-5-methyl-thieno[2,3- dlpyrimidine-6-carboxylic ac -dimethylamino-propy)-amide

63.1 4-(4-Fluoro-2-hvdroxy-phenylamino)-5-methyl-thieno[2,3-dlpyr imidine-6- carboxylic acid

Intermediate III (333 mg), sodium hydroxide solution 1 M (5 ml) in 10 ml methanol and 10 ml THF were stirred at rt overnight. Then the mixture was acidified with

hydrochloric acid and filtrated.

Yield: 256 mg

ESI mass spectrum: m/z = 320 (M+H) +

Retention time HPLC: 2.22 min

HPLC method: 004 CC ZQ6 63.2 4-(4-Fluoro-2-hvdroxy-phenylamino)-5-methyl-thieno[2,3-dlpyr imidine-6- carboxylic acid -dimethylamino-propyl)-amide

Prepared analogously to example 21 .1 using the product from 63.1 (150 mg) and N,N-dimethyl-1 ,3-propanediamine (90 μΙ).

Yield: 157 mg

ESI mass spectrum: m/z = 404 (M+H) +

Retention time HPLC: 1 .62 min

HPLC method: 004 CC ZQ6 63.3 4-[2-Cvano-methyl-methoxy)-4-fluoro- phenylamino1-5-methyl-thieno[2,3- dlpyrimidine-6-carboxylic acid (2-dimethylamino-propy)-amide

To the product from 63.2 (157 mg) in DMF (4 ml) was added cesium carbonate (165 mg). After 5 min. 2-Bromopropionitrile (37 μΙ) was added and the mixture was stirred at rt overnight. Then the mixture was concentrated and diluted with DCM and water. The organic layer was seperated, dried and concentrated.

Yield: 130 mg

ESI mass spectrum: m/z = 457 (M+H) +

MNK 2 -lnhibition

Kinase Fluorescence Polarization Assays

Assay principle: Inhibitory potency of compounds against Mnk1 , Mnk2a and other kinases was assessed with assays based on a format known to those skilled in the art as the indirect (competitive) fluorescence polarization. The assay detection system comprises a small fluorophore-labeled phospho-peptide (termed ligand) bound to a phospho-specific antibody. The product generated by the kinase reaction competes with the ligand for antibody binding. Based on the larger molecular volume of the bound ligand, which results in a lower rotation rate in solution, its emitted light has a higher degree of polarization than the one from the free ligand.

Description of the specific homogenous kinase assay

Example 2a. Mnk1 and Mnk2a in vitro kinase assay

As a source of enzyme, human Mnk1 and human Mnk2a were expressed as GST fusion proteins in E. coli, purified to >80% homogeneity by glutathione affinity chromatography and activated in vitro with pre-activated ERK2. In brief, the open reading frames of human Mnk1 and Mnk2a were amplified from cDNA using the forward/reverse primer pairs SEQ ID NO: 1 5TTTAGGATCCGTATCTTCTCAAAAGTTGG /

SEQ ID NO: 2 5' CTGGGTCGACTCAGAGTGCTGTGGGCGG and

SEQ ID NO: 3 5'ACAGGGATCCGTGCAGAAGAAACCAGCC /

SEQ ID NO: 4 5'GATGGTCGACTCAGGCGTGGTCTCCCACC

(utilized restriction sites underlined), respectively, and cloned into the BamHI and Sail sites of the vector pGEX-4T1 (Amersham, Sweden, cat. no. 27-4580-01 ). These constructs allow prokaryotic expression of Mnk1 or Mnk2a as fusion protein with a N-terminal glutathione S-transferase (GST) tag, referred to as GST-Mnk1 or GST- Mnk2a. The following expression and purification procedure was identical for GST- Mnk1 and GST-Mnk2a, referring in general to GST-Mnk, when not distinguishing between the two isoforms. Expression of GST-Mnk was in E. coli BL21 (Merck Biosciences, Germany, cat. no. 69449). Cells were grown in LB-Bouillon (Merck, Germany, cat. no. 1 .10285) supplemented with 1 00 pg/ml ampicillin (Sigma, Germany, cat. no. A951 8) at 37°C . When the culture had reached a density corresponding to an A 6 oo of 0.8, an equal volume of ice cold LB/ampicillin was added, the cu ltu re transferred to 25°C and i nd uced for 4 h with 1 m M isopropyl thiogalactoside (IPTG, Roth, Germany, cat. no. 2316.4). Cells harvested by centrifugation w e r e r e s u s p e n d e d i n 1 0 m l l y s i s b u f f e r ( 5 0 m M tris(hydroxymethyl)aminomethane hydrochloride (Tris/HCI, Sigma, Germany, cat. no. T5941 ) pH 7.5, 300 mM sodium chloride (NaCI, Sigma, Germany, cat. no. S7653), 5% (w/v) glycerol (Sigma, Germany, cat. no. G5516), 3 mM DTT dithiotreitol (DTT, Sigma, Germany, cat. no. D9779)) per gram wet weight cell pellet. Lysates were prepared by d isru ption of cel ls with a son ifier and subseq uent clearing by centrifugation at 38000 g for 45 min at 4°C.

The lysate was applied to a GSTPrep FF 16/10 column (Amersham, Sweden, cat. no. 17-5234-01 ) equilibrated with lysis buffer. Removal of unbound material was with 3 column volumes (CV) lysis buffer. Elution was with 2 CV of elution buffer (50 mM Tris/HCI pH 7.5, 300 mM NaCI, 5% (w/v) glycerol, 20 mM glutathione (Sigma, Germany, cat. no. G4251 )). Peak fractions were pooled and the protein transferred into storage buffer (50 mM Tris/HCI pH 7.5, 200 mM NaCI, 0.1 mM ethylene glycol- bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA, Aldrich, Germany, cat. no. 23,453-2), 1 mM DTT, 10% (w/v) glycerol, 0.5 M sucrose (Sigma, Germany, cat. no. S0389) by gel filtration on a PD10 desalting column (Amersham, Sweden, cat. no. 17-0851 -01 ). Aliquots were shock frozen in liquid nitrogen and stored at -80°C.

Activation of Mnk1 and Mnk2a was at a concentration of 2.5 μΜ of either purified GST-Mnk1 or GST-Mnk2a by incubation with 150 nM pre-activated NHis-ERK2 (see ERK2 assay for preparation) and 50 μΜ adenosine triphosphate (ATP, Sigma, cat. no . A2699) i n a buffer com pris i ng 20 m M N-(2-hydroxyethyl) piperazine-N'-(2- ethanesulfonic acid) (HEPES, Fluka, Germany, cat. no 54459)/potassium hydroxide (KOH, Roth, Germany, cat. no 6751 .1 ) pH 7.4, 1 0 mM magnesium chloride (MgCI 2 , Sigma, Germany, cat. no. M2670), 0.25 mM DTT, 0.05% (w/v) polyoxyethylene 20 stearylether (Brij 78, Sigma, Germany, cat. no. P401 9) (HMDB buffer) for 45 min at 30°C. After the incubation, the preparation was aliquoted into single-use samples, shock frozen in l iqu id n itrogen, stored at -80°C and util ized for Mn k1 or Mn k2a kinase assays as detailed below. The presence of activating kinase has been tested to not interfere with the Mnk activity assay.

SUBSTRATE: A carboxy-terminal amidated 12mer peptide with the sequence

SEQ ID NO: 5 TATKSGSTTKNR,

derived from the amino acid sequence around serine 209 of the eukaryotic translation initiation factor 4E (elF4E) has been synthesized and purified by high performance liquid chromatography (HPLC) to >95% (Thermo, Germany). The serine residue phosphorylated by Mnk kinases is underlined.

LIGAND: The peptide TATKSG-pS-TTKNR, containing an amidated carboxy- terminus and conjugated at the amino-terminus with the oxazine derived fluorophore depicted below was synthesized and used as ligand.

ANTIBODY: SPF New Zealand White Rabbits have been immunized according to standard protocols with the peptide NH2-CTATKSG-pS-TTKNR-CONH2, coupled to keyhole limpet hemocyanin (KLH). The immune globulin G (IgG) fraction was purified from serum of boosted animals by techniques known in the art. In brief, serum was subjected to protein A affinity chromatography. Eluted material was precipitated at 50% cold saturated ammonium sulfate, pellets dissolved and desalted. The resulting material was appropriate for use in below described assay without further antigen- specific purification.

ASSAY SETUP: Inhibition of kinase activity of Mnk1 and Mnk2a was assessed with the same assay system, using pre-activated GST-Mnk1 or GST-Mnk2a, respectively. The kinase reaction contains 30 μΜ substrate peptide, 20 μΜ ATP, 60 nM ligand and one of either 25 nM pre-activated Mnk1 or 2.5 nM pre-activated Mnk2a. The reaction buffer conditions are 16 mM HEPES/KOH pH 7.4, 8 mM MgCI 2 , 0.4 mM DTT, 0.08 % (w/v) bovine serum albumin (BSA, Sigma, Germany, cat. no. A3059), 0.008% (w/v) Pluronic F127 (Sigma, Germany, cat. no. P2443), 3% (v/v) DMSO (Applichem, Germany, cat. no. A3006). The kinase reaction is at 30°C for 40 min. The kinase reaction is terminated by addition of 0.67 reaction volumes of 1 μΜ antibody in 20 mM HEPES/KOH pH 7.4, 50 mM ethylenediaminetetraacetic acid, disodium salt (EDTA, Sigma, Germany, cat. no. E5134), 0.5 mM DTT, 0.05% (w/v) polyoxyethylene-sorbitan monolaureate (Tween 20, Sigma, Germany, cat. no. P7949). After 1 h equilibration time at room temperature, samples are subjected to fluorescence polarization measurement. The fluorescence polarization readout was generated on an Analyst AD multimode reader (Molecular Devices, Sunnyvale, CA, USA) equipped with a DLRP650 dichroic mirror (Omega Opticals, Brattleboro, VT, USA, cat. no. XF2035), a 630AF50 band pass filter (Omega Opticals, Brattleboro, VT, USA, cat. no. XF1069) on the excitation and a 695AF55 band pass filter on the emission side (Omega Opticals, Brattleboro, VT, USA, cat. no. XF3076).

The activity of Mnk proteins can be assayed also by other in vitro kinase assay formats. For example, suitable kinase assays have been described in the literature in Knauf et al ., Mol Cell Biol . 2001 Aug;21 (16):5500-1 1 or in Scheper et al ., Mol Cell Biol . 2001 Feb;21 (3):743-54. In general, Mnk kinase assays can be performed such that a Mnk substrate such as a protein or a peptide, which may or may not include modifications as further described below, or others are phosphorylated by Mnk proteins having enzymatic activity in vitro. The activity of a candidate agent can then be determined via its ability to decrease the enzymatic activity of the Mnk protein. The kinase activity may be detected by change of the chemical, physical or immunological properties of the substrate due to phosphorylation.

In one example, the kinase substrate may have features, designed or endogenous, to facilitate its binding or detection in order to generate a signal that is suitable for the analysis of the substrates phosphorylation status. These features may be, but are not limited to, a biotin molecule or derivative thereof, a glutathione-S-transferase moiety, a moiety of six or more consecutive histidine residues, an amino acid sequence or hapten to function as an epitope tag, a fluorochrome, an enzyme or enzyme fragment. The kinase substrate may be linked to these or other features with a molecular spacer arm to avoid steric hindrance.

In another example the kinase substrate may be labelled with a fluorophore. The binding of the reagent to the labelled substrate in solution may be followed by the technique of fluorescence polarization as it is described in the literature. In a variation of this example, a fluorescent tracer molecule may compete with the substrate for the analyte to detect kinase activity by a technique which is know to those skilled in the art as indirect fluorescence polarization.

In yet another example, radioactive gamma-ATP is used in the kinase reaction, and the effect of the test agent on the incorporation of radioactive phosphate in the test substrate is determined relative to control conditions. It has been shown that the compounds of the invention exhibit low IC50 values in in vitro biological screening assays as described in example 2a for inhibition of Mnk 1 and/or Mnk 2 kinase activity. The following table contains the test results for exemplary compounds.

Example MNK2 IC50 [nM] Example MNK2 IC50 [nM]

31-6 13 33 47

31-7 20 33-4 16

32-1 267 34 54

32-2 19 34-4 15

32-3 - 35 12

32-4 13 36 29

32-5 8 36-1 35

32-6 23 36-2 42

32.7 5 36-3 26

32-8 - 36-4 44

32-9 13 36-5 25

32-10 18 36-6 55

32-11 14 36-7 27

32-12 12 36-8 19

32-13 - 36-9 63

32-14 65 36-10 32

32-15 78 36-11 45

32-16 66 36-12 31

32-17 16 36-13 54

32-18 30 36-14 64

32-19 23 36-15 24

32-20 19 36-16 22

32-21 8 36-17 46

32-22 36 36-18 26

32-23 26 36-19 14

32-24 25 37 4

32-25 13 38 6

32-26 7 39 19

32-27 7 40 20

32-28 3 41 31

32-29 170 42 70

32-30 56 43 62

32-31 146 44 24

32-32 27 45 20

32-33 65 46 84

32-34 69 47 125

32-35 35 48 12

32-36 25 49 8

32-37 8 50 13

32-38 26 51 10

32-39 35 52 25

32-40 13 53 7

32-41 25 54 11

32-42 12 55 5403

32-43 12 56 314

32-44 6 57 110

32-45 11 58 119 Example MNK2 IC 50 [nM]

59 29

60 37

61 77

62 267

63 17

HPLC methods:

Method A_10

Waters ZQ 2000; Waters 1515 Pumpe; Waters PDA 996 Detektor; Waters 2747 Injektor

DAD 200-420 nm

mobile phases:

A: water with 0.10% formic acid

B: acetonitrile with 0.10% formic acid

time in min %A %B flow rate in ml/min

0.00 95 5 1 .50

2.00 0 100 1 .50

2.50 0 100 1 .50

2.60 95 5 1 .50

Stationary phase:X-terra MS C18; 4.6χ30ηηηη * 2.5μηη Method AC1

Waters ZQ 2000; Waters 1515 Pumpe; Waters PDA 996 Detektor; Waters 2747 Injektor

DAD 210-420 nm

mobile phases:

A: water with 0.10% formic acid

B: acetonitrile with 0.10% formic acid

time in min %A %B flow rate in ml/min

0.00 95 5 1 .00 0.10 95 5 1 .00

3.10 2 98 1 .00

4.50 2 98 1 .00

5.00 95 5 1 .00

Stationary phase: X-terra MS C18; 4.6χ30ηηηη*2.5μηη

Method A_9

(pos/neg switch method)

Waters ZQ 2000; Waters 1515 Pumpe; Waters PDA 996 Detektor; Waters 2747 Injektor

DAD 200-420 nm

mobile phases:

A: water with 0.10% formic acid

B: acetonitrile with 0.10% formic acid

time in min %A %B flow rate in ml/min

0.00 95 5 1 .50

2.00 0 100 1 .50

2.50 0 100 1 .50

2.60 95 5 1 .50

Stationary phase: X-terra MS C18; 4.6χ30ηηηη*2.5μηη

Method C_SF_TFA_MeOH_P30V#004_CC_ZQ1

RP-HPLC MS analyses have been performed on a

Waters ZQ2000 mass spectrometer,

HP1 100 HPLC + DAD (Wavelength range ( nm ): 210

500) and Gilson 215 Autosampler.

Mobile Phases:

A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 80 20 2.00 1 .7 0 100 2.00

2.50 0 100 2.00

2.60 80 20 2.00

Stationary phase: Waters, Sunfire, C18, 3,5 μητι, 4,6 X 50 mm.

Column temp: 60°C.

Diode array detection is at the wavelength range of 210-500 nm.

Method A_4

Waters ZQ 2000; Waters 1515 Pumpe; Waters PDA 996 Detektor; Waters 2747 Injektor

DAD 200-420 nm

mobile phases:

A: water with 0.10% formic acid

B: acetonitrile with 0.10% formic acid time in min %A %B flow rate in ml/min

0.00 95 5 1 .00

0.10 95 5 1 .00

3.10 2 98 1 .00

4.50 2 95 1 .00

5.00 95 5 1 .00

Stationary phase: X-terra MS C18; 4.6χ30ηηηη * 2.5μηη

Method amslstandard:

ZQ 2000MS; Waters 2996 PDA (210-600 nm); Waters 2525 pump; Waters 515 make up pump; Waters 2767 injector/ fraction collector, Waters columns and fluidics organizer (CFO)

mobile phases:

A: water with 0.20% trifluoroacetic acid

B: Methanol

time in min %A %B flow rate in ml/min

0.00 72 18 55.00

2.00 72 18 55.00 2.50 62 38 55.00

9.50 18 72 55.00

10.00 0 100 55.00

12.00 0 100 55.00

12.50 0 100 0

Stationary phase: X-terra MS C18; 30χ100ηηηη*5μηη

Temperature 25°C

Method amslunpolarl :

ZQ 2000MS; Waters 2996 PDA (210-600 nm); Waters 2525 pump; Waters 515 make up pump; Waters 2767 injector/ fraction collector, Waters columns and fluidics organizer (CFO)

mobile phases:

A: water with 0.20% trifluoroacetic acid

B: Methanol

time in min %A %B flow rate in ml/min

0.00 59 41 55.00

2.00 59 41 55.00

2.50 49 51 55.00

9.50 5 95 55.00

10.00 0 100 55.00

12.00 0 100 55.00

12.50 0 100 0

Stationary phase: X-terra MS C18; 30x100mm*5 m

Temperature 25°C

Method 002_CC_ZQ4

RP-HPLC MS analyses have been performed on a Waters

ZQ2000 mass spectrometer,

HP1 100 HPLC + DAD (Wavelength range ( nm ): 210 to 500) and

Gilson 215 Autosampler.

Mobile Phases:

A: Water with 0.1 % TFA B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 95 5 1 .50

1 .3 0 100 1 .50

2.50 0 100 1 .50

2.60 95 5 1 .50

Stationary phase: Waters, Sunfire, C18, 3,5 μιτι, 4,6 X 50 mm. Column temp: constant at 40°C.

Diode array detection is at the wavelength range of 210-500 nm. Method 003_CC_ZQ6

RP-HPLC MS analyses have been performed on a Waters

ZQ2000 mass spectrometer, .Alliance 2695, PDA2996

HP1 100 HPLC + DAD (Wavelength range ( nm ): 210 to 500) and

As 2700

Mobile Phases:

A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 95 5 1 .50

1 .3 0 100 1 .50

3.00 0 100 1 .50

3.40 95 5 1 .50

Stationary phase: Waters, Sunfire, C18, 3,5 μιτι, 4,6 X 50 mm. Column temp: constant at 40°C.

Diode array detection is at the wavelength range of 210-500 nm Method 004_CC_ZQ6

RP-HPLC MS analyses have been performed on a Waters ZQ2000 mass spectrometer,Alliance 2695, PDA2996

DAD (Wavelength range ( nm ): 210 to 500) and 2700 AS

Mobile Phases: A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 80 20 2

1 .7 0 100 2

2.5 0 100 2

2.6 80 20 2

Stationary phase: Waters, Sunfire, C18, 3,5 μιτι, 4,6 X 50 mm. Column temp: 60°C.

Diode array detection is at the wavelength range of 210-500 nm.

Method 004_CC_ZQ7

RP-HPLC MS analyses have been performed on a Waters ZQ2000 mass spectrometer,

DAD (Wavelength range ( nm ): 210 to 500) and 2700 AS Mobile Phases:

A: Water with 0.15% TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 95 5 1 .5

1 .5 95 5 1 .5

2.0 0 100 1 .5

Stationary phase: Xbridge C18, 3,5 μιτι, 4,6 X 50 mm.

Column temp: 40°C.

Diode array detection is at the wavelength range of 210-500 nm.

Method 003_CC_ZQ7

RP-HPLC MS analyses have been performed on a Waters ZQ2000 mass spectrometer,

DAD (Wavelength range ( nm ): 210 to 500) and 2700 AS Mobile Phases:

A: Water with 0.032% NH4OH

B: MeOH Time in min %A %B Flowrate in ml/min

0.00 95 5 1 .5

1 .5 95 5 1 .5

2.0 0 100 1 .5

Stationary phase: Xbridge C18, 3,5 μιτι, 4,6 X 50 mm.

Column temp: 40°C.

Diode array detection is at the wavelength range of 210-500 nm.

Method 007_CC_ZQ5

RP-HPLC MS analyses have been performed on a Waters ZQ2000 mass spectrometer, Alliance 2790

DAD (Wavelength range ( nm ): 210 to 500) and 2700 AS Mobile Phases:

A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 80 20 2

1 .7 0 100 2

2.5 0 100 2

2.6 80 20 2

Stationary phase: Waters, Sunfire C18, 3,5 μιτι, 4,6 X 50 mm. Column temp: 60°C.

Diode array detection is at the wavelength range of 210-500 n

Method 007_CC_ZQ7

RP-HPLC MS analyses have been performed on a Waters

ZQ2000 mass spectrometer, Alliance 2790

DAD (Wavelength range ( nm ): 210 to 500) and 2700 AS

Mobile Phases:

A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min 0.00 80 20 2

1 .7 0 100 2

2.5 0 100 2

2.6 80 20 2

Stationary phase: Waters, Sunfire C18, 3,5 μητι, 4,6 X 50 mm.

Column temp: 60°C.

Diode array detection is at the wavelength range of 210-500 nm. Method M2-SB-C18

RP-HPLC MS analyses have been performed on a

Agilent 1200, MS G6140A, binare Pumpe, DAD

190-400 nm

Mobile Phases:

A: Water with 0.1 % TFA

B: MeOH

Time in min %A %B Flowrate in ml/min

0.00 90 10 3

1 .8 0 100 3

2.0 0 100 3

2.15 90 10 3

2.35 90 10 3

Stationary phase: Agilent, Stable Bond SB-C18, 1 ,8 μπτι, 4,6 X 30 mm.

Diode array detection is at the wavelength range of 190-400 nm.

Method W001 001