US20180072744A1 - Heteroaryl syk inhibitors - Google Patents

Heteroaryl syk inhibitors Download PDF

Info

Publication number
US20180072744A1
US20180072744A1 US15/817,435 US201715817435A US2018072744A1 US 20180072744 A1 US20180072744 A1 US 20180072744A1 US 201715817435 A US201715817435 A US 201715817435A US 2018072744 A1 US2018072744 A1 US 2018072744A1
Authority
US
United States
Prior art keywords
methyl
group
hplc
alkyl
het
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/817,435
Inventor
Georg Dahmann
Matthias Hoffmann
Jasna Klicic
David James LAMB
Clive McCarthy
Spencer Charles R. Napier
Karen PARRISH
John Scott
Jennifer L. Swantek FITZGERALD
Edward Walker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Priority to US15/817,435 priority Critical patent/US20180072744A1/en
Publication of US20180072744A1 publication Critical patent/US20180072744A1/en
Priority to US16/535,337 priority patent/US10947243B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41621,2-Diazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention relates to new substituted heteroaryls of formula 1
  • A is selected from the group consisting of N and CH
  • E is selected from the group consisting of C and N,
  • G is selected from the group consisting of C and N,
  • each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
  • M, R 3 and R 1 are defined as in claim 1 .
  • the invention relates to the above compounds of formula 1 or of formula 1′ for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.
  • the present invention describes new compounds that inhibit the protein kinase Syk (spleen tyrosine kinase), the preparation and formulation thereof and their use for preparing a medicament.
  • Syk is an intracellular tyrosine kinase that has an important mediator function in the signal transduction of different receptors in B-cells, mast cells, monocytes, macrophages, neutrophils, T-cells, dendritic cells and epithelial cells.
  • the receptors in which Syk performs an important function in signal transduction include for example the receptors for IgE (Fc ⁇ RI) and IgG (Fc ⁇ R1) on mast cells and B cells, the B-cell receptor (BCR) and the T-cell receptor (TCR) on B- and T-cells, the ICAM1 receptor (ICAM1 R) on epithelial cells of the respiratory tract, the DAP12-receptor on natural killer cells, dendritic cells and osteoclasts, the dectin 1-receptor on a subpopulation of T-helper cells (Th-17 cells), as well as the integrin receptors for ⁇ 1-, ⁇ 2- and ⁇ 3-integrins on neutrophils, monocytes and macrophages (Wong et al.; Expert Opin.
  • IgE In mast cells the binding of IgE to Fc ⁇ RI causes the cross-linking of IgE-receptors and the recruiting and activation of Lyn (a tyrosine kinase from the Src family). Active Lyn phoshorylates so-called ITAM motifs, which are present in many of the receptors listed above, and thereby generates binding sites for the SH2-domain of Syk. As a result of the binding to the ITAM motif Syk is activated and then phosphorylates various substrates which are needed for the release of allergic and inflammatory mediators such as e.g. histamine and ⁇ -hexosamidase (BHA), as well as for the synthesis of lipid mediators, such as e.g. prostaglandins and leukotrienes.
  • BHA ⁇ -hexosamidase
  • Syk has been discussed as a therapeutic target for different diseases such as e.g. allergic rhinitis, asthma, autoimmune diseases, rheumatoid arthritis, osteopenia, osteoporosis, COPD and various leukaemias and lymphomas (Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; Ulanova et al.; Expert Opion. Ther. Target (2005) 9(5); 901-921; Sigh and Masuda. Annual Reports in Medicinal Chemistry (2007) Vol 42; 379-391; Bajpai et al.; Expert Opin. Investig.
  • Allergic rhinitis and asthma are diseases associated with allergic reactions and inflammatory processes and involving different cell types such as e.g. Mast cells, eosinophils, T-cells and dendritic cells.
  • the high affinity immunoglobulin receptors for IgE (FcERI) and IgG (Fc ⁇ R1) are activated and induce the release of pro-inflammatory mediators and bronchoconstrictors.
  • An inhibitor of the Syk kinase activity should thus be able to inhibit these steps.
  • RA Rheumatoid arthritis
  • B-cells play a significant role, as has been demonstrated for example by the therapeutic use of rituximab, a B cell-depleting antibody.
  • Syk In addition to the function of Syk in the signal transduction of the BCR (which after being stimulated also induces the release of pro-inflammatory mediators), Syk also plays an important part in the maturation and proliferation of B cells (Cheng et al. Nature (1995) 378, 303-306, Cornall et al., PNAS (2000) 97(4), 1713-1718).
  • An inhibitor of the Syk kinase activity may thus offer a therapeutic option for the treatment of autoimmune diseases such as RA and diseases with an increased proliferation of B cells, such as e.g. B-cell lymphomas.
  • COPD chronic obstructive pulmonary disease
  • a cellular level in COPD there is in particular a multiplication of T-lymphocytes, neutrophils, granulocytes and macrophages.
  • a multiplication of T-lymphocytes, neutrophils, granulocytes and macrophages in particular, there is an increase in the number of CD8-positive lymphocytes, that is directly connected with the impairment of lung function.
  • Another characteristic of COPD are acute deteriorations in lung function (exacerbations), characterised by viral (e.g. Rhinovirus), or bacterial (e.g. Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis) infections.
  • an inhibitor of the Syk kinase activity could be a new therapeutic approach to the treatment of the inflammatory processes that underlie COPD. It has also been shown that Syk in epithelial cells of the respiratory tract is involved in the ICAM1R-mediated uptake and subsequent replication of the Rhinovirus and that a si-RNA against Syk blocks these steps (Wang et al.; J. Immunol. (2006) 177, 6859-6870; Lau et al.; J. Immunol. (2008) 180, 870-880). Thus, an inhibitor of the Syk kinase activity could also be used therapeutically in exacerbations caused by Rhinoviruses.
  • B-ALL B-lineage acute lymphoblastic leukemia
  • FL follicular lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • NHS mantle cell lymphomas
  • AML acute myeloid leukemia
  • Idiophathic thrombocytopenic purpura is an autoimmune disease in which IgG autoantibodies against antigens present on platelets bind to and destroy platelets.
  • Patients with ITP have an accelerated clearence of circulating IgG-coated platelets via macrophages in the spleen and the liver.
  • an inhibitor of Syk is considered to have a therapeutic benefit in Fc ⁇ R-mediated cytopenias like ITP.
  • the Syk inhibitor R788 (R406) improved platelet counts in a single center, Being label study in patients with ITP (Podolanczuk et al; Blood (2009) 113, 3154-3169).
  • Bullous pemphigoid (Ujiie et al. Journal of Dermatology 2010; 37: 194-204) is a chronic, autoimmune, subepidermal, blistering skin disease that rarely involves mucous membranes. Bullous pemphigoid is characterized by the presence of immunoglobulin G (IgG) autoantibodies specific for the hemidesmosomal bullous pemphigoid antigens BP230 (BPAg1) and BP180 (BPAg2).
  • IgG immunoglobulin G
  • Pemphigus vulgaris (Venugopal et al. Dermatol. Clin. 2011;29:373-80) is a chronic blistering skin disease with skin lesions that are rarely pruritic, but which are often painful.
  • Pemphigus vulgaris is an autoimmune disease caused by IgG autoantibodies directed against both desmoglein 1 and desmoglein 3 resulting in the loss of cohesion between keratinocytes in the epidermis. It is characterized by extensive flaccid blisters and mucocutaneous erosions. In both diseases IgG autoantibodies bind to Fc receptor gamma (FcR ⁇ ) and activate FcR ⁇ and downstream signaling via Syk kinase. Thus, an inhibitor of the Syk kinase activity which blocks downstream signalling of the FcRg could be used therapeutically to treat patients with bullous pemphigoid and pemphigus vulgaris.
  • FcR ⁇ Fc receptor gamma
  • SLE Systemic lupus erythematosus
  • 1,6-Naphthyridines are known as SYK-inhibitors.
  • U.S. Pat. Nos. 3,928,367, 4,017,500, 4,115,395 and 4,260,759 describe 5-amino-1,6-naphthyridines with an antifungal and antibacterial activity.
  • WO 9918077 describes 5-piperazinyl-1,6-naphthyridines as serotonin antagonists.
  • U.S. Pat. No. 7,321,041 describes substituted 1,6-naphthyridines as SYK-inhibitors, however these 1,6-naphthyridines have a completely different substitution pattern from the compounds according to the invention.
  • WO 2011092128 discloses 1,6-naphthyridines which are substituted in 5- and in 7-position.
  • WO 01/83485 discloses substituted imidazopyrimidines and triazolopyrimidines as SYK-inhibitors
  • WO 2008/113469 discloses substituted imidazo- and triazolopyrimidines as GSK 3 ⁇ -inhibitors.
  • quinolones are known as SYK-inhibitors.
  • SYK-inhibitors For instance, WO 2006038041 and WO 2013014060 both disclose quinoline-compounds which are substituted in the 5- and 7-position, however the substitution pattern—in particular in the 7-position—is completely different from the one of the compounds of formula 1 of the instant invention.
  • the compounds of formulas 1 and 1′ and in particular the compounds of formulas 1a, 1a′, 1c, 1c′ are particularly suitable for the treatment of respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases, autoimmune diseases, inflammatory diseases and diseases of the peripheral or central nervous system, particularly for the treatment of asthma, allergic rhinitis, rheumatoid arthritis, allergic dermatitis, lupus erythematosus (SLE) and COPD, in particular because all these compounds of the present invention show the following desired capacities:
  • the compounds of formula 1 and 1′ of the instant invention have several significant structural differences compared to the previously known prior art compounds.
  • the compounds of formula 1 and 1′ of the instant invention differ from the previously known 1,6-naphthyridines,quinolones, pyrido[3,4-b]pyrazines, imidazopyrimidines and triazolopyrimidines therein that they combine the following features:
  • the present invention concerns compounds of formula 1
  • A is selected from the group consisting of N and CH
  • E is selected from the group consisting of C and N,
  • G is selected from the group consisting of C and N,
  • each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
  • M is selected from the group consisting of —CH 2 —, —O—, —NH— and —N(C 1-4 -alkyl)-;
  • R 3 is selected from the group consisting of methyl and ethyl
  • Het is selected from the group consisting of
  • Hetaryl is selected from the group consisting of
  • R 1 is selected from the group consisting of
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, —OH, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,—NH(CH 3 ), —N(CH 3 ) 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, —OH, halogen and C 1-3 -alkyl,
  • a preferred embodiment of the instant invention relates to the aforementioned compounds of formula 1′
  • the instant invention concerns the aforementioned compounds of formula 1 or of formula 1′, wherein
  • M is —CH 2 —
  • R 3 is methyl
  • G is C (carbon)
  • R 1 is selected from the group consisting of
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the instant invention relates to the aforementioned compounds of formula 1a
  • R 1 is selected from the group consisting of
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the invention relates to compounds of the above-mentioned formula 1a or of formula 1a′, wherein R 1 is
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl,
  • each X is selected from the group consisting of halogen, oxo and —C 1-4 -alkyl,
  • the invention relates to compounds of the above-mentioned formula 1a or of formula 1a′, wherein
  • R 1 is either
  • R 1 -residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the invention relates to the above compounds of the aforementioned formula 1a or formula 1a′, wherein
  • R 1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
  • R 1 -residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the invention concerns the above compounds of the aformementioned formula 1a or of formula 1a′, wherein
  • R 1 is phenyl
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, Hetaryl, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the instant invention relates to the above compounds of the aforementioned formula 1a or formula 1a′, which is selected from the group consisting of
  • the instant invention concerns compounds of formula 1c
  • R 1 is selected from the group consisting of
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein R 1 is
  • the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R 1 is either
  • R 1 -residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R 1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
  • R 1 -residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the instant invention concerns the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R 1 is phenyl
  • each Z is a substituent selected from the group consisting of
  • each X is selected from the group consisting of halogen, oxo, —C 1-4 -alkyl, —O—C 1-4 -alkyl, —C 1-4 -haloalkyl, —O—(C 1-4 -alkylene)-Het, Het, —NH 2 ,
  • substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • the instant invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, which is selected from the group consisting of
  • the instant invention refers to an intermediate compound selected from the group consisting of
  • the instant invention refers to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease which can be treated by inhibition of the SYK enzyme.
  • the instant invention relates to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of allergic rhinitis, asthma, COPD, adult respiratory distress syndrome, bronchitis, B-cell lymphoma, dermatitis and contact dermatitis, allergic dermatitis, allergic rhinoconjunctivitis, rheumatoid arthritis, anti-phospholipid syndrome, Berger's disease, Evans's syndrome, ulcerative colitis,allergic antibody-based glomerulonephritis, granulocytopenia, Goodpasture's syndrome, hepatitis, Henoch-Schönlein purpura, hypersensitivity vasculitis, immunohaemolytic anaemia, autoimmune haemolytic anemia, idiopathic thrombocytopenic purpura, Kawasaki syndrome, allergic conjunctivitis, l
  • the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, adult respiratory distress syndrome, bronchitis, allergic dermatitis, contact dermatitis, idiopathic thrombocytopenic purpura, rheumatoid arthritis, lupus erythematodes, lupus nephritis and allergic rhinoconjunctivitis.
  • a disease selected from the group consisting of asthma, COPD, allergic rhinitis, adult respiratory distress syndrome, bronchitis, allergic dermatitis, contact dermatitis, idiopathic thrombocytopenic purpura, rheumatoid arthritis, lupus erythematodes, lupus nephritis and allergic rhinoconjunctivitis.
  • the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.
  • a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.
  • the instant invention concerns pharmaceutical formulations which contain one or more of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) and a pharmaceutically acceptable excipient.
  • the instant invention concerns pharmaceutical formulations which contain one or more compounds of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) in combination with an active substance selected from the group consisting of anticholinergics, betamimetics, corticosteroids, PDE4-inhibitors, EGFR-inhibitors, LTD4-antagonists, CCR3-inhibitors, iNOS-inhibitors, CRTH2-antagonists,HMG-CoA reductase inhibitors and NSAIDs.
  • an active substance selected from the group consisting of anticholinergics, betamimetics, corticosteroids, PDE4-inhibitors, EGFR-inhibitors, LTD4-antagonists, CCR3-inhibitors, iNOS-inhibitors, CRTH2-antagonists,HMG-CoA reductase inhibitors and NSAIDs.
  • C 1-6 -alkyl groups are possible substituents at a group, in the case of three substituents, for example, C 1-6 -alkyl could represent, independently of one another, a methyl, an n-propyl and a tert-butyl.
  • each hydrogen atom may be removed at the substituent and the valency thus freed may serve as a binding site to the rest of a molecule.
  • X 1 is also understood as being the linking point of the group R 1 to the structure of formula 1 and X 2 as being the linking point of the group R 2 to the structure of formula 1.
  • C 1-6 -alkyl (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms and by the term “C 1-3 -alkyl” are meant branched and unbranched alkyl groups with 1 to 3 carbon atoms. “C 1-4 -alkyl” accordingly denotes branched and unbranched alkyl groups with 1 to 4 carbon atoms. Alkyl groups with 1 to 4 carbon atoms are preferred.
  • Examples of these include: methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl or hexyl.
  • the abbreviations Me, Et, n-Pr, i-Pr, n—Bu, i—Bu, t—Bu, etc. may also optionally be used for the above-mentioned groups.
  • the definitions propyl, butyl, pentyl and hexyl include all the possible isomeric forms of the groups in question.
  • propyl includes n-propyl and iso-propyl
  • butyl includes iso-butyl, sec-butyl and tert-butyl etc.
  • C 1-6 -alkylene (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms and by the term “C 1-4 -alkylene” are meant branched and unbranched alkylene groups with 1 to 4 carbon atoms.
  • Alkylene groups with 1 to 4 carbon atoms are preferred. Examples of these include: methylene, ethylene, propylene, 1-methylethylene, butylene, 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene, pentylene, 1,1-dimethylpropylene, 2,2-dimethylpropylene, 1,2-dimethylpropylene, 1,3-dimethylpropylene or hexylene.
  • propylene, butylene, pentylene and hexylene include all the possible isomeric forms of the groups in question with the same number of carbons.
  • propyl includes also 1-methylethylene and butylene includes 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene.
  • carbon chain is substituted by a group which together with one or two carbon atoms of the alkylene chain forms a carbocyclic ring with 3, 5 or 6 carbon atoms, this includes, inter alia, the following examples of the rings:
  • C 2-6 -alkenyl (including those which are part of other groups) are meant branched and unbranched alkenyl groups with 2 to 6 carbon atoms and by the term “C 2-4 -alkenyl” are meant branched and unbranched alkenyl groups with 2 to 4 carbon atoms, provided that they have at least one double bond.
  • Alkenyl groups with 2 to 4 carbon atoms are preferred. Examples include: ethenyl or vinyl, propenyl, butenyl, pentenyl or hexenyl. Unless stated otherwise, the definitions propenyl, butenyl, pentenyl and hexenyl include all the possible isomeric forms of the groups in question. Thus, for example, propenyl includes 1-propenyl and 2-propenyl, butenyl includes 1-, 2- and 3-butenyl, 1-methyl-1-propenyl, 1-methyl-2-propenyl etc.
  • C 2-6 -alkenylene (including those which are part of other groups) are meant branched and unbranched alkenylene groups with 2 to 6 carbon atoms and by the term “C 2-4 -alkenylene” are meant branched and unbranched alkylene groups with 2 to 4 carbon atoms. Alkenylene groups with 2 to 4 carbon atoms are preferred.
  • propenylene examples include: ethenylene, propenylene, 1-methylethenylene, butenylene, 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene, pentenylene, 1,1-dimethylpropenylene, 2,2-dimethylpropenylene, 1,2-dimethylpropenylene, 1,3-dimethylpropenylene or hexenylene.
  • propenylene, butenylene, pentenylene and hexenylene include all the possible isomeric forms of the groups in question with the same number of carbons.
  • propenyl also includes 1-methylethenylene and butenylene includes 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene.
  • aryl aromatic ring systems with 6 or 10 carbon atoms. Examples include: phenyl or naphthyl, the preferred aryl group being phenyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • aryl-C 1-6 -alkylene (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by an aromatic ring system with 6 or 10 carbon atoms. Examples include: benzyl, 1- or 2-phenylethyl or 1- or 2-naphthylethyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • heteroaryl-C 1-6 -alkylene (including those which are part of other groups) are meant—even though they are already included under “aryl-C 1-6 -alkylene”—branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by a heteroaryl.
  • a heteroaryl of this kind includes five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed.
  • heteroaryls may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • heteroaryl-C 1-6 -alkylenes The following are examples of heteroaryl-C 1-6 -alkylenes:
  • C 1-6 -haloalkyl (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms, which are substituted by one or more halogen atoms.
  • C 1-4 -alkyl are meant branched and unbranched alkyl groups with 1 to 4 carbon atoms, which are substituted by one or more halogen atoms.
  • Alkyl groups with 1 to 4 carbon atoms are preferred. Examples include: CF 3 , CHF 2 , CH 2 F, CH 2 CF 3 .
  • C 3-7 -cycloalkyl (including those which are part of other groups) are meant cyclic alkyl groups with 3 to 7 carbon atoms, if not specifically defined otherwise. Examples include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Unless otherwise stated, the cyclic alkyl groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • C 3-10 -cycloalkyl are also meant monocyclic alkyl groups with 3 to 7 carbon atoms and also bicyclic alkyl groups with 7 to 10 carbon atoms, or monocyclic alkyl groups which are bridged by at least one C 1-3 -carbon bridge.
  • heterocyclic rings or “heterocycle” are meant, unless stated otherwise, five-, six- or seven-membered, saturated, partially saturated or unsaturated heterocyclic rings which may contain one, two or three heteroatoms, selected from among oxygen, sulphur and nitrogen, while the ring may be linked to the molecule through a carbon atom or through a nitrogen atom, if there is one.
  • heterocyclic rings or “heterocycles”
  • saturated heterocyclic ring refers to five-, six- or seven-membered saturated rings. Examples include:
  • heterocyclic rings or “heterocyclic group”
  • partially saturated heterocyclic group refers to five-, six- or seven-membered partially saturated rings which contain one or two double bonds, without so many double bonds being produced that an aromatic system is formed, unless specifically defined otherwise. Examples include:
  • heterocyclic aromatic rings or “heterocycles”
  • heterocyclic aromatic rings unsaturated heterocyclic group” or “heteroaryl” refers to five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed, unless not specifically defined otherwise.
  • heterocyclic aromatic groups include:
  • heterocyclic ring may be provided with a keto group.
  • keto group examples include:
  • bicyclic cycloalkyls generally denotes eight-, nine- or ten-membered bicyclic carbon rings. Examples include
  • bicyclic heterocycles generally denotes eight-, nine- or ten-membered bicyclic rings which may contain one or more heteroatoms, preferably 1-4, more preferably 1-3, even more preferably 1-2, particularly one heteroatom, selected from among oxygen, sulphur and nitrogen, unless not specifically defined otherwise.
  • the ring may be linked to the molecule through a carbon atom of the ring or through a nitrogen atom of the ring, if there is one. Examples include:
  • bicyclic aryl denotes a 5-10 membered, bicyclic aryl ring which contains sufficient conjugated double bonds to form an aromatic system.
  • aryl is a 5-10 membered, bicyclic aryl ring which contains sufficient conjugated double bonds to form an aromatic system.
  • aryl is naphthyl.
  • bicyclic heteroaryl denotes a 5-10 membered, bicyclic heteroaryl ring which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contains sufficient conjugated double bonds to form an aromatic system, unless specifically defined otherwise.
  • bicyclic cycloalkyls or “bicyclic aryl”
  • fused cycloalkyl or “fused aryl” denotes bicyclic rings wherein the bridge separating the rings denotes a direct single bond.
  • fused, bicyclic cycloalkyl
  • bicyclic heterocycles or “bicyclic heteroaryls”
  • fused bicyclic heterocycles of “fused bicyclic heteroaryls” denotes bicyclic 5-10 membered heterorings which contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen and wherein the bridge separating the rings denotes a direct single bond.
  • the “fused bicyclic heteroaryls” moreover contain sufficient conjugated double bonds to form an aromatic system.
  • Examples include pyrrolizine, indole, indolizine, isoindole, indazole, purine, quinoline, isoquinoline, benzimidazole, benzofuran, benzopyran, benzothiazole, benzothiazole, benzoisothiazole, pyridopyrimidine, pteridine, pyrimidopyrimidine,
  • Halogen within the scope of the present invention denotes fluorine, chlorine, bromine or iodine. Unless stated to the contrary, fluorine, chlorine and bromine are regarded as preferred halogens.
  • Compounds of general formulas 1 or 1′ may have acid groups, mainly carboxyl groups, and/or basic groups such as e.g. amino functions. Compounds of general formulas 1 or 1′ may therefore be present as internal salts, as salts with pharmaceutically usable inorganic acids such as hydrochloric acid, sulphuric acid, phosphoric acid, sulphonic acid or organic acids (such as for example maleic acid, fumaric acid, citric acid, tartaric acid or acetic acid) or as salts with pharmaceutically usable bases such as alkali metal or alkaline earth metal hydroxides or carbonates, zinc or ammonium hydroxides or organic amines such as e.g. diethylamine, triethylamine, triethanolamine, inter alia.
  • pharmaceutically usable inorganic acids such as hydrochloric acid, sulphuric acid, phosphoric acid, sulphonic acid or organic acids (such as for example maleic acid, fumaric acid, citric acid, tarta
  • the compounds of formulas 1 or 1′ may be converted into the salts thereof, particularly for pharmaceutical use into the physiologically and pharmacologically acceptable salts thereof.
  • These salts may be present on the one hand as physiologically and pharmacologically acceptable acid addition salts of the compounds of formula 1 with inorganic or organic acids.
  • the compound of formulas 1 or 1′ when R is hydrogen may be converted by reaction with inorganic bases into physiologically and pharmacologically acceptable salts with alkali or alkaline earth metal cations as counter-ion.
  • the acid addition salts may be prepared for example using hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid or maleic acid. It is also possible to use mixtures of the above-mentioned acids.
  • R denotes hydrogen it is preferable to use the alkali and alkaline earth metal hydroxides and hydrides, of which the hydroxides and hydrides of the alkali metals, particularly sodium and potassium, are preferred, while sodium and potassium hydroxide are particularly preferred.
  • the compounds of general formulas 1 or 1′ may optionally be converted into the salts thereof, particularly for pharmaceutical use into the pharmacologically acceptable acid addition salts with an inorganic or organic acid.
  • suitable acids for this purpose include succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulphonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulphuric acid, tartaric acid or citric acid. It is also possible to use mixtures of the above-mentioned acids.
  • the invention relates to the compounds of formula 1 in question, optionally in the form of the individual optical isomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • pharmacologically acceptable acids such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • the compounds of formula 1, 1a and 1c according to the invention may optionally be present as racemates, but may also be obtained as pure enantiomers, i.e. in the (R) or (S) form.
  • the invention relates to the compounds in question, optionally in the form of the individual optical isomers, diastereomers, mixtures of diastereomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • pharmacologically acceptable acids such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • the invention relates to the respective compounds of formulas 1 or 1′ in the form of the pharmacologically acceptable salts thereof.
  • These pharmacologically acceptable salts of the compounds of formulas 1 or 1′ may also be present in the form of their respective hydrates (e.g. Monohydrates, dihydrates, etc.) as well as in the form of their respective solvates.
  • a hydrate of the compound according to the formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, containing water of crystallisation.
  • a solvate of the compound according to formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, which contains solvent molecules (e.g. Ethanol, methanol etc) in the crystal lattice.
  • solvent molecules e.g. Ethanol, methanol etc
  • G is C or N
  • E is C or N, preferably N
  • PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)
  • R 1 is as herein before defined.
  • G is C or N
  • E is C or N, preferably N
  • A is CH or N
  • PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)
  • R 1 is as herein before defined.
  • Step 1 Synthesis of (1′R,3R,/S)-1-(1′′-(4-Methoxyphenylethyl)-5-oxo-3-pyrrolidine carboxylic acid (mixture of diastereoisomers)
  • Step 2 Synthesis of (R/S)-N-Methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide as a mixture of diastereoisomers
  • Step 3 Synthesis of (R/S)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidine-2-one as a mixture of diastereoisomers
  • Step 4 Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one under base induced epimerization conditions
  • the crude product was dissolved in 310 mL 2-methyl-2-butanol at 40° C. (temperature ⁇ 50° C). The solution was slowly cooled to 0° C. Precipitation started. At 0° C. 385 mL of n-heptane were added and the suspension was stirred for 1 hour. The precipitate was filtrated, washed with n-heptane and dried at 50° C.
  • Step 4 Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one under base induced epimerization conditions
  • Step 5 Synthesis of (R)-4-[(R)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.1
  • the reaction was diluted at 60° C. with 500 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed twice with 300 mL of water. The organic portion was concentrated to an oily solid. The residual material was crystallized three times from ethyl acetate and hexanes followed by drying in a vacuum oven with a nitrogen stream at 30° C.
  • Step 5 Synthesis of (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.2
  • the reaction was diluted at 60° C. with 50 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed with 20 mL of water. The organic portion was concentrated to an oil. The oil was dissolved in 8 mL of isopropyl acetate at reflux. The solution was cooled to ambient temperature wherein crystallization occurred. The mixture was diluted dropwise with 10 mL of heptane at ambient temperature. The mixture was agitated for 30 minutes.
  • HATU 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluroniumhexafluorophosphat
  • NMP N-methyl-2-pyrrolidinon
  • THF tetra
  • Step 2 Synthesis of 4-Bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6
  • This intermediate was prepared from (S)-tetrahydro-furan-3-ol in two steps according to the preparation of 4-bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6.
  • Step 2 Synthesis of 4-Bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8
  • Step 1 Synthesis of N′-(4,4-difluoro-cyclohexylidene)-hydrazinecarboxylic acid tert-butyl ester
  • Step 2 Synthesis of N′-(4,4-difluoro-cyclohexyl)-hydrazinecarboxylic acid tert-butyl ester
  • This intermediate was prepared from 4-bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8 according to the preparation of 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.19.
  • Step 1 Synthesis of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • 6.1 can be synthesized as following:
  • the reaction mixture was diluted with 200 mL of isopropyl acetate and water (100 mL). The layers were separated, and the aqueous portion was extracted with twice with 100 mL of isopropyl acetate. The organic portions were assayed to show 40.78g of of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one in a solution mass of 796.3g for a 99% yield.
  • Purification of 6.1 can be conducted by concentration of a crude isopropyl acetate solution of 50 g 6.1 in vacuo to 200 mL wherein solids crystallized. 500 mL of heptane was slowly charged to the slurry at 20° C. and the mixture was agitated for 2 h. The solids were collected by filtration, washed with heptane, and dried at 30° C. 46.9 g of 6.1 was isolated as a beige solid in 92% recovery.
  • Step 2 Synthesis of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1
  • Step 1 Synthesis of (4R)-4-[(1R)-1-(6-Bromo-2-methyl-indazol-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • Step 2 Synthesis of (4R)-4-[(1R)-1-(6-Bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one 7.5
  • Step 1 Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • Step 2 Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 2)
  • 8.1 can be synthesized as following:
  • the crude paste was suspended in hot isopropyl acetate (75 mL) and filtered hot.
  • the filtrate was concentrated in vacuo to approximately 15 mL wherein a solid crystallized upon cooling to ambient temperature.
  • the mixture was diluted by the dropwise addition of heptane (30 mL).
  • the mixture was agitated for 1h at ambient temperature.
  • Ethyl 3-amino-1-methyl-1H-pyrazole-4-carboxylate was isolated by filtration, washed with heptane and dried under vacuum to provide 3.7g as a yellow-orange solid in 74% yield.
  • Isopropyl magnesium chloride lithium chloride complex (1.3 M in THF, 28.4 mL, 37 mmol) was charged to a solution of 4-bromo-1-(tert-butyl)-1H-pyrazole (5.0 g, 25 mmol) in anhydrous THF (25 mL) under argon at ambient temperature.
  • Anhydrous dioxane (3.3 g, 37 mmol) was charged to the reaction, and the reaction was agitated at 45° C. for 4 h.
  • the resulting mixture was cooled to ambient temperature and charged to an anhydrous solution of acetic anhydride (7.5 g, 73 mmol) in THF (25 mL) at ⁇ 20° C.
  • Phosphorous (V) oxychloride (848 mg, 5.53 mmol) was charged to a mixture of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2,5-dihydro-4H-pyrazolo[4,3-c]pyridin-4-one (500mg, 1.84 mmol) in anhydrous acetonitrile (5 mL) under argon. The reaction was agitated at 75-80° C. for 3 h; at which point, the reaction was cooled to ambient temperature. The reaction slowly poured into a saturated sodium bicarbonate aqueous solution (45 mL).
  • the reaction mixture was cooled to ambient temperature and quenched by the slow addition of a hydrogen chloride solution (4 M in 1,4 dioxane, 0.245 mL, 0.98 mmol).
  • the reaction was diluted with isopropyl acetate (20 mL) and water (20 mL). The layers were separated, and the aqueous portion was extracted twice with isopropyl acetate (20 ml). The combined organic layers were concentrated to an oil.
  • Example 17 was synthesized in analogous manner to Example 3 using 1-[(4-bromophenyl)methyl]-2-methyl-1H-imidazole 3.27.
  • Example 8 as solid.
  • Step 1 Synthesis of tert-Butyl 4-[4-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]pyrazol-1-yl]piperidine-1-carboxylate
  • reaction mixture was stirred at 140° C. for 15 min under microwave irradiation.
  • the reaction mixture was filtered through rpSiO 2 , washed with methanol and purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 90 mg (80% per NMR) of tert-butyl 4-[4-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo-[4,3-c]pyridin-6-yl]pyrazol-1-yl]piperidine-1-carboxylate as solid.
  • Step 2 Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(4-piperidyl)pyrazol-4-yl]pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 9)
  • Example was synthesized in analogous manner to Example 29, but with modified reaction time.
  • Example 45 Example 45 as solid.
  • the reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo.
  • the crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol). The residue was dissolved in 2 mL DCM, 500 ⁇ L TFA were added and the resulting mixture was stirred at room temperature for 1 h.
  • the reaction mixture was purified by flash chromatography (DCM/methanol/ammonia) and by elution through a SCX (Biotage SCX-3) column and subsequent rpHPLC to yield 92 mg (yield: 61%) of Example 45 as solid.
  • Example 8 A mixture of 68 mg of Example 8, 59 mg of tetrahydropyran-4-yl 4-methylbenzenesulfonate and 39 mg potassium carbonate in 2 mL DMF was stirred at 80° C. for 3 h and at 100° C. for 10 h. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The resulting residue was purified by rpHPLC to yield after lyophilisation 37 mg of Example 73 as solid.
  • Example 9 To a mixture of 68 mg of Example 9 and 31 mg sodium acetate in 3 mL DCM and 0.5 mL methanol was added 17 ⁇ L formaldehyde (aqueous 37%). The resulting mixture was stirred at room temperature for 10 min, before 46 mg sodium triacetoxyborohydride was added. The reaction mixture was stirred for 1.75 h before quenched with water. The organic solvent was removed by destillation. The resulting residue was purified by rpHPLC to yield after lyophilisation 64 mg of Example 82 as solid.
  • Example compounds prepared according to the foregoing synthesis schemes were characterised by the following chromatographic methods and/or NMR spectroscopy.
  • the stationary phase used was a Inertsil C8-3 (GL Sciences), 5 ⁇ m; dimension: 100 ⁇ 4.0 mm,
  • the stationary phase used was a Chiralpak AD-H (Daicel), 5 ⁇ m; dimension: 150 ⁇ 4.6 mm, (column temperature: constant at 10° C.). Detection DAD 225 nm.
  • the stationary phase used was a Chiralpak AD-H (Daicel), 5 ⁇ m; dimension: 150 ⁇ 4.6 mm,
  • UV Detection wavelength 215 nm
  • UV Detection wavelength 215 nm
  • UV Detection wavelength 215 nm
  • Solvent B 0.1% Formic acid in acetonitrile
  • UV Detection wavelength Spectra A max (with scan in the region of 200-400 nm)
  • UV Detection wavelength 215 nm
  • Solvent A 2 mM Ammonium bicarbonate modified to pH 10 with Ammonium Hydroxide/ water
  • UV Detection wavelength 215 nm
  • UV Detection wavelength 215 nm
  • Method Name Method Name: Column: Sunfire, 3 ⁇ 30 mm, 2.5 ⁇ m Column Supplier: Waters Gradient/ Solvent Time % Sol % Sol Flow Temp [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.] 0.00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60
  • BBFO Multinuclear Broad Band fluorine observe
  • AURB phosphorylates Ser10 and Ser28 on histone H3, a key event in mitosis and cellular proliferation. Inhibition of AURB therefore has the potential to block cellular proliferation, and could compromise tissues that exhibit a high cellular turnover, such as the intestine or the bone marrow. It is therefore desired to avoid parallel AURB inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds show IC 50 -values with regard to Aurora B inhibition of more than 1 ⁇ M, preferably more than 6 ⁇ M, more preferably more than 10 ⁇ M, more preferably more than 30 ⁇ M, more preferably of more than 45 ⁇ M, particularly preferably more than 50 ⁇ M.
  • the AURB-IC 50 /SYK-IC 50 -ratios of all example compounds are preferably more than 30, more preferably more than 100.
  • FLT-3 is a tyrosine kinase receptor.
  • an FLT-3 ligand binds to the receptor, the intrinsic tyrosine kinase activity of the receptor is activated, which in turn phosphorylates and activates signal transduction molecules (such as SHC) which in turn propagates the signal in the cell.
  • Signaling through FLT-3 plays a role in cell survival, proliferation, and differentiation and is important for lymphocyte (B cell and T cell) development. It is therefore desired to avoid parallel FLT-3 inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound.
  • all example compounds of the instant invention show IC 50 -values with regard to FLT-3 inhibition of more than 0,30 ⁇ M, preferably more than 1 ⁇ M, more preferably more than 10 ⁇ M, particularly preferably more than 30 ⁇ M.
  • the FLT-3-IC 50 /SYK-IC 50 -ratios of all example compounds are preferably more than 10, more preferably more than 30.
  • Glycogen synthase kinase 3 beta (GSK 3 ⁇ ) is a proline-directed serine-threonine kinase that is prominent in the TGF- ⁇ and Wnt intracellular signalling pathways. GSK 3 ⁇ facilitates a number of intracellular signalling pathways including the activation of ⁇ -catenin complex. In adults, GSK 3 ⁇ is involved in cellular proliferation and energy metabolism, whilst in neonates is involved in neuronal cell development and body pattern formation. It is therefore desired to avoid parallel GSK3 ⁇ inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds of the invention show IC 50 -values with regard to GSK 3 ⁇ inhibition of more than 1 ⁇ M, preferably of more than 10 ⁇ M.
  • Recombinant human Syk (amino acids 342-635) was expressed as a fusion protein with an N-terminal GST tag, affinity-purified and deep-frozen at a concentration of approx. 50-100 ⁇ M in storage buffer (25 mM HEPES pH7.5; 25 mM MgCl 2 ; 5 mM MnCl 2 ; 50 mM KCl; 0.2% BSA; 0.01% CHAPS; 100 ⁇ M Na 3 VO 4 ; 0.5 mM DTT, 10% glycerol) at ⁇ 80° C. until use.
  • storage buffer 25 mM HEPES pH7.5; 25 mM MgCl 2 ; 5 mM MnCl 2 ; 50 mM KCl; 0.2% BSA; 0.01% CHAPS; 100 ⁇ M Na 3 VO 4 ; 0.5 mM DTT, 10% glycerol
  • the catalytic activity of the GST-Syk kinase fusion protein was determined using the Kinase Glo® Luminescence Kinase test (Promega; V6712). In this homogeneous test the amount of ATP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ATP still present and thus correlates inversely with the activity of the kinase.
  • test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 1 mM. Serial Dilution is done in 100% DMSO. All further dilutions of the substances were carried out with test buffer (25 mM HEPES pH7.5; 25 mM MgCl 2 ; 5 mM MnCl 2 ; 50 mM KCl; 0.2% HSA; 0.01% CHAPS; 100 ⁇ M Na 3 VO 4 ; 0.5 mM DTT). Dilution steps and concentration range were adapted according to need. 7 ⁇ l aliquots of these dilutions were transferred into a 384-well Optiplate (Perkin Elmer, # 6007290).
  • Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.
  • the output file of the reader is a csv file that contains the well number and measured relative light units (RLU).
  • RLU measured relative light units
  • the measurement of the negative control was set as 100% ctrl and the measurement of the positive control was set as 0% ctrl.
  • the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics.
  • the IC 50 values were calculated from the % ctrl values using Assay Explorer software.
  • Recombinant human Aurora B (amino acids 1-344, clone number DU1773, Molecular weight 40,2kDa, University of Dundee) was expressed as a fusion protein with an N-terminal His tag, affinity-purified and deep-frozen at a concentration of approx. 0.25-0.5 mg/ml in storage buffer (50 mM Tris-HCl pH 8; 25 mM Na- ⁇ -glycerophosphat; 0.1 mM EGTA; 150 mM NaCl; 0.03% Brij-35; 1 mM DTT and 10% glycerol) at ⁇ 80° C. until use.
  • storage buffer 50 mM Tris-HCl pH 8; 25 mM Na- ⁇ -glycerophosphat; 0.1 mM EGTA; 150 mM NaCl; 0.03% Brij-35; 1 mM DTT and 10% glycerol
  • the activity of the Aurora B kinase protein was determined using the ADP Glo® Luminescence Kinase test (Promega; V9103X). In this homogeneous test the amount of ADP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ADP still present and thus correlates with the activity of the protein kinase.
  • test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 5 mM. Serial Dilution is done in 1:10 steps in 100% DMSO. All further dilutions of the substances were carried out with test buffer (50 mM Hepes, pH 7.5, 10 mM MgCl2, 1 mM EGTA, 60 ⁇ M Ultra Pure ATP, 0.01% Brij35, 0.1% BSA, 5 mM ⁇ -Glycerophosphate) until a concentration was reached which was 2.5 times above the final test concentration (final concentration of the compounds: 50 ⁇ M to 0.005 nM).
  • test buffer 50 mM Hepes, pH 7.5, 10 mM MgCl2, 1 mM EGTA, 60 ⁇ M Ultra Pure ATP, 0.01% Brij35, 0.1% BSA, 5 mM ⁇ -Glycerophosphate
  • Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.
  • ADP-Glo® solution ADP-Glo Reagent #V912B Promega (heated to room temperature) were added to each well and incubation was continued for a further 40. minutes. Then 20 ⁇ l Kinase detection mix (Detection Buffer #V913B Promega; Kinase Detection Substrate #V914B Promega) were added and incubated for 40 minutes at room temperature. The plates were read in Envision Luminescence Reader (Perkin-Elmer).
  • the output file of the reader is a csv file that contains the well number and measured RLU.
  • the measurement of the negative control was set as 0% ctrl and the measurement of the positive control was set as 100% ctrl.
  • the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics.
  • FLT3 is obtained from Invitrogen in 50 mM Tris (pH7.5); 100 mM NaCl; 0.05 mM EDTA, 0.05% NP-40, 2 mM DTT; 50%Glycerol # PV3182; Lot 286671; sequence see below).
  • the enzyme is diluted to 720 nM (35 ⁇ g/ml) in enzyme dilution buffer and 10 ⁇ l aliquots are stored at ⁇ 80° C.
  • the activity of FLT3 is measured using the Z′-LYTETM assay technology from Invitrogen (# PV3191)
  • the assay is performed in 384 black plates from Corning (# 3676) in a final volume of 10 ⁇ l by adding 5 ⁇ l of kinase peptide mix and 2.5 ⁇ l of compound dilution. The reaction is started by addition of 2.5 ⁇ l of the 4 ⁇ ATP solution.
  • Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no kinase.
  • the reaction is performed for 1 h at room temperature before 5 ⁇ l of the development solution is added. After a further incubation for 1 h at room temperature 5 ⁇ l of the stop reagent is added. The plates are read on a Flex Station II 384 (Molecular Devices).
  • the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 ⁇ M or 10 ⁇ M).
  • the output text file is evaluated in an “MS-Excel - VB -Makro” and “GraphPadPrism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of FLT3 are reported in M. data for the inhibition of the protease are reported in % CTL.
  • Human GSK3beta (expressed and prified from SF21 cells)is obtained from the University Dundee/Scotland (Dr. James Hastie—Dept. of Biochemistry) in 50 mM Tris (pH7.5); 150 mM NaCl; 0.1 mM EGTA, 270 mM Succrose, 0,1% ⁇ -mercaptoethanol, 1 mM benzamidine, 0,2 mM PMSF ; sequence see below).
  • the enzyme is diluted to 3,56 ⁇ M (168 ⁇ g/ml) in enzyme dilution buffer and 6 ⁇ l aliquots are stored at ⁇ 80° C.
  • GSK3 ⁇ kinase protein The activity of GSK3 ⁇ kinase protein is measured using the Z′-LYTETM assay technology from Invitrogen (# PV3324).
  • the assay is performed in 384 black plates from Corning (# 3676) in a final volume of 10 ⁇ l by adding 5 ⁇ l of kinase peptide mix and 2.5 ⁇ l of compound dilution. The reaction is started by addition of 2.5 ⁇ l of the 4 ⁇ ATP solution.
  • Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no ATP.
  • the reaction is performed 1 h at room temperature. After 1 h 5 ⁇ l of the development solution is added. After a further incubation for 1 h at room temperature 5 ⁇ l of the stop reagent is added. Finally the plates are read on a Flex Station II 384 (Molecular Devices).
  • the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 ⁇ M).
  • the output text file is evaluated in an “MS-Excel-VB-Makro” and “GraphPadPrism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of GSK3beta are reported in M. data for the inhibition of the protease are reported in % CTL.
  • the metabolic degradation for a specific SYK-inhibitor is performed at 37° C. with pooled human liver microsomes (human liver microsomes are commercially available as “BD UltraPoolTM” by Corning Life Sciences, Fogostraat 12, 1060 LJ Amsterdam, The Netherlands).
  • the final incubation volume of 100 ⁇ l per time point contains TRIS buffer pH 7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 ⁇ M.
  • the reaction is initiated by addition of beta-nicotinamide adenine dinucleotide phosphate in its reduced form (NADPH, 1 mM) and terminated by transfering an aliquot into solvent after different time points. Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point.
  • beta-nicotinamide adenine dinucleotide phosphate in its reduced form (NADPH, 1 mM) and terminated by transfering an aliquot into solvent after different time points. Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point.
  • the quenched (terminated) incubations are then pelleted by centrifugation (10000 g, 5 min).
  • CL_INTRINSIC The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation:
  • the protein content [mg/ml] was determined with the “Bicinchoninic Acid Kit” of Sigma Aldrich (commercially available).
  • the upscaled intrinsic Clearance (CL_UP_INT) is calcuated by considering the liver weight [g liver/kg body weight] and the microsomal recovery [mg protein/g liver]:
  • the percent hepatic blood flow (% Q h ) is finally calculated by considering the human liver blood flow Q [ml/min/kg]:
  • SYK-inhibitory capacity is not the only important aspect which a SYK-inhibitor to be used as a medicament to treat SYK-related diseases must show.
  • IC 50 (SYK) ⁇ 1 ⁇ M the candidate compound does not show undesired inhibitory effects on other kinases which could lead to unwanted or even dangerous side effects.
  • Examples of such other kinases that should not be inhibited by the candidate SYK-inhibitor are AURB, FLT3 and GSKbeta.
  • the compounds of the invention with alkyl-substituted pyrazole structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 2b)), but also do not have unwanted inhibitory effects on other kinases such as AURB, FLT3 and GSK3beta (unlike the compounds of WO 2013/014060 (see Table 2b)).
  • the compounds of the invention with alkyl-substituted pyrazole structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • the compounds of the invention with optionally substituted bicyclic heteroaryl structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 3b)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2013/014060 (see Table 3b)).
  • the compounds of the invention with optionally substituted bicyclic heteroaryl structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • the compounds of the invention have IC 50 -values with regard to AURB of more than 45 ⁇ M, often even of more than 50 ⁇ M
  • the IC 50 -values (AURB) of WO 2011/092128 (see Table 4b) and of WO 2013/014060 (see Table 4c) are mostly ⁇ 5 ⁇ M, often even below 1 ⁇ M and consequently the compounds of the invention with alkoxy-substituted phenyl structures have less unwanted inhibitory effects on other kinases such as AURB compared to most of the compounds of WO 2011/092128 (Table 4b) and of WO 2013/014060 (see Table 4c).
  • the compounds of the invention with alkoxy-substituted phenyl structures therefore show a significantly improved SYK-selectivity and additionally an acceptable human liver microsomal stability compared to all of the structurally closest compounds disclosed in WO2013/014060 and in WO2011/092128.
  • the compounds of the invention with heterocycle-substituted or heterocycle-annulated phenyl structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 5b)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2013/014060 (see Table 5b)).
  • the compounds of the invention with heterocycle-substituted or heterocycle-annulated phenyl structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • the compounds of the invention with optionally substituted pyridine structures are not only efficient SYK-inhibitors (like the compounds of WO 2011/092128 (Table 6b) and of WO 2013/014060 (see Table 6c)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2011/092128 (Table 6b) and of WO 2013/014060 (see Table 6c)).
  • the compounds of the invention with optionally substituted pyridine structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO 2011/092128 or in WO2013/014060.
  • the compounds of formula 1 are characterised by their range of applications in the therapeutic field. Particular mention should be made of those applications for which the compounds of formula 1 according to the invention are preferably used on the basis of their pharmaceutical activity as Syk-inhibitors. Examples include respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases or complaints, immune or autoimmune diseases, allergic diseases, inflammatory diseases, e.g. inflammatory diseases of the joints, skin and eyes and diseases of the peripheral or central nervous system.
  • pulmonary diseases which are accompanied by increased mucus production, inflammation and/or obstructive diseases of the airways.
  • these include asthma, paediatric asthma, ARDS (Adult Respiratory Distress Syndrome), acute, allergic or chronic bronchitis, autoimmune haemolytic anemia, chronic obstructive bronchitis (COPD) (including the treatment of Rhinovirus-induced exacerbations), coughs, allergic rhinitis or sinusitis, allergic rhinoconjunctivitis, chronic rhinitis or sinusitis, alveolitis, farmers' lung, hyperreactive airways, infectious bronchitis or pneumonitis, bronchiectasis, pulmonary arterial hypertension, pulmonary fibrosis, bronchial oedema, pulmonary oedema, pneumonia or interstitial pneumonia triggered by various causes such as aspiration, inhalation of toxic gases or bronchitis, pneumonia or
  • the compounds according to the invention are preferably also suitable for the treatment of allergic diseases such as for example allergic rhinitis, allergic rhinoconjunctivitis, allergic conjunctivitis, and contact dermatitis, urticaria/angiooedema and allergic dermatitis.
  • allergic diseases such as for example allergic rhinitis, allergic rhinoconjunctivitis, allergic conjunctivitis, and contact dermatitis, urticaria/angiooedema and allergic dermatitis.
  • the compounds according to the invention are preferably also suitable for the treatment of inflammatory diseases of the joints, of the blood vessels and of the kidney or inflammatory diseases of the skin and eyes.
  • inflammatory diseases of the joints of the blood vessels and of the kidney or inflammatory diseases of the skin and eyes.
  • examples of these are rheumatoid arthritis, antibody-based glomerulonephritis, psoriasis, Kawasaki syndrome, coeliac disease (sprue), arteriosclerosis and Wegener's granulomatosis, osteoarthritis, systemic scleroderma, ankylosing spondylitis.
  • the compounds according to the invention are preferably also suitable for the treatment of autoimmune diseases.
  • autoimmune diseases examples include hepatitis (autoimmune-based), lupus erythematodes, lupus nephritis, systemic lupus, Systemic lupus erythematosus,discoid lupus, cutaneous lupus erythematosus (acute, subacute, chronic), anti-phospholipid syndrome, Berger's disease, Evans's syndrome, immunohaemolytic anaemia, ITP (idiopathic thrombocytopenic purpura; adult, neonatal and paediatric), myasthenia gravis, Sjogren's syndrome, sclerodermy, Bullous pemphigoid and Pemphigus vulgaris.
  • ITP idiopathic thrombocytopenic purpura
  • sclerodermy Bullous pemphigoid and Pemphigus vulgaris.
  • the compounds according to the invention are preferably also suitable for the treatment of B-cell lymphomas, like chronic lymphocytic leukaemia and non-Hodgkin's lymphomas or T cell lymphomas.
  • the compounds according to the invention are preferably also suitable for the treatment of Graft-versus -host disease.
  • Mention may preferably also be made of the prevention and treatment of diseases of the peripheral or central nervous system. Examples of these are acute and chronic multiple sclerosis or non-familial lateral sclerosis.
  • Mention may preferably also be made of the prevention and treatment of osteoporotic diseases such as for example disease-associated osteopenia, osteoporosis and osteolytic diseases.
  • osteoporotic diseases such as for example disease-associated osteopenia, osteoporosis and osteolytic diseases.
  • the present invention relates particularly preferably to the use of compounds of formula 1 for preparing a pharmaceutical composition for the treatment of diseases selected from among asthma, COPD, allergic rhinitis, Adult Respiratory Distress Syndrome, bronchitis, allergic dermatitis, contact dermatitis, ITP, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, and allergic rhinoconjunctivitis.
  • diseases selected from among asthma, COPD, allergic rhinitis, Adult Respiratory Distress Syndrome, bronchitis, allergic dermatitis, contact dermatitis, ITP, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, and allergic rhinoconjunctivitis.
  • the compounds of formula 1 may be used for the treatment of a disease selected from among asthma, allergic rhinitis, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, allergic dermatitis and COPD.
  • a disease selected from among asthma, allergic rhinitis, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, allergic dermatitis and COPD.
  • the compounds of formula 1 may be used on their own or in conjunction with other active substances of formula 1 according to the invention.
  • the compounds of formula 1 may optionally also be used in conjunction with other pharmacologically active substances.
  • the active substances used here may be selected for example from among the betamimetics, anticholinergics, corticosteroids, PDE4-inhibitors, LTD4-antagonists, EGFR-inhibitors, MRP4-inhibitors, dopamine agonists, H1-antihistamines, PAF-antagonists, iNos-inhibitos, HMG-CoA reductase inhibitors (statins), PI3-kinase-inhibitors, CCR3-antagonists, CCR2-antagonists, CCR1-antagonists, IKK2-inhibitors, A2a agonists, alpha-4-integrin-inhibitors, CRTH2-antagonists, histamine 1, combined H1/H3-antagonists, p38 kinas
  • LTB4-antagonists cromoglycine, dissociated glucocorticoid mimetics, immunesuppressive agents, cytostatica, non-steroidal anti-inflammatory drugs (NSAIDs), chloroquine, hydroxychloroquine, anti-TNF-antibodies, anti-GM-CSF antibodies, anti-CD46-antibodies, anti-IL-1- antibodies, anti-IL-2-antibodies, anti-IL-4- antibodies, anti-IL-5-antibodies, anti-IL6 antibodies, anti-IL6 receptor antibodies, anti-IL-13- antibodies, anti-IL_18 antibodies, anti-CD30 L antibodies, anti-Ox40L-antibodies, anti-IL-4/IL-13-antibodies, anti-IL-23 (p19) antibodies, anti-IL-12/IL-23 (p40) antibodies, anti-CD3 antibodies, anti-CD4 antibodies, anti-CD154 antibodies, CD89 antibodies, anti-IL-2 receptor/CD25 antibodies, anti-CD22 antibodies, anti-interferon antibodies, anti-ICOS
  • Suitable betamimetics used are preferably compounds selected from among arformoterol, carmoterol, formoterol, indacaterol, salmeterol, albuterole, bambuterol, bitolterol, broxaterol, carbuterol, clenbuterol, fenoterol, hexoprenalin, ibuterol, isoetharin, isoprenalin, levosalbutamol, mabuterol, meluadrin, metaproterenol, milveterol, orciprenalin, pirbuterol, procaterol, reproterol, rimiterol, ritodrin, salmefamol, soterenol, sulphonterol, terbutalin, tiaramide, tolubuterol, zintero1,6-Hydroxy-8- ⁇ 1-hydroxy-2-[2-(4-methoxy-phenyl)-1,1-dimethyl-e
  • the acid addition salts of the betamimetics are preferably selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably the hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • the salts of hydrochloric acid, methanesulphonic acid, benzoic acid and acetic acid are particularly preferred according to the invention.
  • the anticholinergics used are preferably compounds selected from among tiotropium salts, particularly the bromide salt, oxitropium salts, particularly the bromide salt, flutropium salts, particularly the bromide salt, ipratropium salts, particularly the bromide salt, Aclidinium salts, particularly the bromide salt, glycopyrronium salts, particularly the bromide salt, trospium salts, particularly the chloride salt, tolterodin, (3R)-1-Phenethyl-3-(9H-xanthene-9-carbonyloxy)-1-azoniabicyclo[2.2.2]octan-salts; 2,2-Diphenyl propionic acid tropenole ester-methobromide; 2,2-Diphenyl propionic acid scopine ester-methobromide; 2-Fluor-2,2-Diphenyl acetic acid scopine ester-methobromide; 2-Fluor-2,2-Di
  • the cations tiotropium, oxitropium, flutropium, ipratropium, glycopyrronium, aclidinium and trospium are the pharmacologically active ingredients.
  • the above-mentioned salts may preferably contain chloride, bromide, iodide, sulphate, phosphate, methanesulphonate, nitrate, maleate, acetate, citrate, fumarate, tartrate, oxalate, succinate, benzoate or p-toluenesulphonate, while chloride, bromide, iodide, sulphate, methanesulphonate or p-toluenesulphonate are preferred as counter-ions.
  • the chlorides, bromides, iodides and methanesulphonate are particularly preferred.
  • tiotropium bromide Of particular importance is tiotropium bromide.
  • the pharmaceutical combinations according to the invention preferably contain it in the form of the crystalline tiotropium bromide monohydrate, which is known from WO 02/30928. If the tiotropium bromide is used in anhydrous form in the pharmaceutical combinations according to the invention, it is preferable to use anhydrous crystalline tiotropium bromide, which is known from WO 03/000265.
  • Corticosteroids used here are preferably compounds selected from among beclomethasone, betamethasone, budesonide, butixocort, ciclesonide, deflazacort, dexamethasone, etiprednole, flunisolide, fluticasone, loteprednole, mometasone, prednisolone, prednisone, rofleponide, triamcinolone, tipredane; Pregna-1,4-diene-3,20-dione, 6-fluoro-11-hydroxy-16,17-[(1-methylethylidene) bis(oxy)]-21-[[4-[(nitrooxy)methyl]benzoyl]oxy]-, (6-alpha,11-beta,16-alpha)-(9Cl); 16,17-butylidenedioxy-6,9-difluoro-11-hydroxy-17-(methylthio)androst-4-en-3-one; 6,9-Difluor
  • the steroid is selected from among budesonide, fluticasone, mometasone, ciclesonide and (S)-fluoromethyl 6,9-difluoro-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-diene-17-carbothionate, optionally in the form of the racemates, enantiomers or diastereomers thereof and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.
  • any reference to steroids includes a reference to any salts or derivatives, hydrates or solvates thereof which may exist.
  • Examples of possible salts and derivatives of the steroids may be: alkali metal salts, such as for example sodium or potassium salts, sulfobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates thereof.
  • PDE4 inhibitors which may be used are preferably compounds selected from among enprofyllin, theophyllin, roflumilast, ariflo (cilomilast), tofimilast, pumafentrin, lirimilast, apremilast, arofyllin, atizoram, oglemilast, tetomilast; 5-[N-(2,5-dichloro-3-pyridinyl)-carboxamide]-8-methoxy-Quinoline (D-4418); 5-[N-(3,5-dichloro-1-oxido-4-pyridinyl)-carboxamide]-8-methoxy-2-(trifluoromethyl)-Quinoline (D-4396 (Sch-351591)); N-(3,5-dichloropyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indol-3-yl]glyoxylic acid amide (AW
  • acid addition salts with pharmacologically acceptable acids which the above-mentioned PDE4-inhibitors might be in a position to form are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • LTD4-antagonists which may be used are preferably compounds selected from among montelukast, pranlukast, zafirlukast; (E)-8-[2-[4-[4-(4-Fluorophenyl)butoxy]phenyl]ethenyl]-2-(1H-tetrazol-5-yl)-4H-1-benzopyran-4-one (MEN-91507); 4-[6-Acetyl-3-[3-(4-acetyl-3-hydroxy-2-propylphenylthio)propoxy]-2-propylphenoxy]-butyric acid (MN-001); 1-(((R)-(3-(2-(6,7-Difluor-2-quinolinyl)ethenyl)phenyl)-3-(2-(2-hydroxy-2-propyl)phenyl)thio)methylcyclopropane-acetic acid; 1-(((1(R)-3(3-(2-(2,3-
  • acid addition salts with pharmacologically acceptable acids which the LTD4-antagonists may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • salts or derivatives which the LTD4-antagonists may be capable of forming are meant, for example: alkali metal salts, such as, for example, sodium or potassium salts, alkaline earth metal salts, sulphobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates.
  • alkali metal salts such as, for example, sodium or potassium salts, alkaline earth metal salts, sulphobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates.
  • the EGFR-inhibitors used are preferably compounds selected from among 4-[(3-chloro-4-fluorophenyl)amino]-6- ⁇ [4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]amino ⁇ -7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6- ⁇ [4-(N,N-diethylamino)-1-oxo-2-butene-1-yl]amino ⁇ -7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6- ⁇ [4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino ⁇ -7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)a
  • acid addition salts with pharmacologically acceptable acids which the EGFR-inhibitors may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • dopamine agonists which may be used preferably include compounds selected from among bromocriptine, cabergoline, alpha-dihydroergocryptine, lisuride, pergolide, pramipexol, roxindol, ropinirol, talipexol, terguride and viozan.
  • Any reference to the above-mentioned dopamine agonists within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts and optionally hydrates thereof which may exist.
  • physiologically acceptable acid addition salts which may be formed by the above-mentioned dopamine agonists are meant, for example, pharmaceutically acceptable salts which are selected from the salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid and maleic acid.
  • H1-antihistamines preferably include compounds selected from among epinastine, cetirizine, azelastine, fexofenadine, levocabastine, loratadine, mizolastine, ketotifen, emedastine, dimetinden, clemastine, bamipin, cexchlorpheniramine, pheniramine, doxylamine, chlorophenoxamine, dimenhydrinate, diphenhydramine, promethazine, ebastine, olopatadine, desloratidine and meclozine.
  • Any reference to the above-mentioned H1-antihistamines within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts which may exist.
  • Examples of PAF-antagonists preferably include compounds selected from among lexipafant, 4-(2-chlorophenyl)-9-methyl-2-[3(4-morpholinyl)-3-propanon-1-yl]-6H-thieno-[3,2-f]-[1,2,4]triazolo[4,3-a][1,4]diazepines, 6-(2-chlorophenyl)-8,9-dihydro-1-methyl-8-[(4-morpho-linyl)carbonyl]-4H,7H-cyclo-penta-[4,5]thieno-[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepines.
  • An reference to the above-mentioned above-mentioned PAF-antagonists includes within the scope of the present invention a reference to any pharmacologically acceptable acid addition salts thereof which may exist.
  • non-steroidal anti-inflammatory drugs preferably include compounds selected from among Aceclofenac, Acemetacin, Acetylsalicylklare, Alclofenac, Alminoprofen, Amfenac, Ampiroxicam, Antolmetinguacil, Anirolac, Antrafenin, Azapropazon, Benorilat, Bermoprofen, Bindarit, Bromfenac, Bucloxin Textre, Bucolom, Bufexamac, Bumadizon, Butibufen, Butixirat, Carbasalatcalcium, Carprofen, Cholin Magnesium Trisalicylat, Celecoxib, Cinmetacin, Cinnoxicam, Clidanac, Clobuzarit, Deboxamet, Dexibuprofen, Dexketoprofen, Diclofenac, Diflunisal, Droxicam, Eltenac, Enfenaminklare, Etersalat, Etod
  • MRP4-inhibitors used are preferably compounds selected from among N-acetyl-dinitrophenyl-cysteine, cGMP, cholate, diclofenac, dehydroepiandrosterone 3-glucuronide, dehydroepiandrosterone 3-sulphate, dilazep, dinitrophenyl-s-glutathione, estradiol 17-beta-glucuronide, estradiol 3,17-disulphate, estradiol 3-glucuronide, estradiol 3-sulphate, estrone 3-sulphate, flurbiprofen, folate, N5-formyl-tetrahydrofolate, glycocholate, glycolithocholic acid sulphate, ibuprofen, indomethacin, indoprofen, ketoprofen, lithocholic acid sulphate, methotrexate, ((E)-3-[[[3-[2-(7-chloro-2-quino
  • JAK inhibitors preferably include compounds selected from among Tofacitinib and Ruxolitinib.
  • immunesuppressive agents preferably include compounds selected from among mycophenolate mofetil, mycophenolic acid, azathioprine, cyclosporine, tacrolimus, pimecrolimus, abetimus, gusperimus and leflunomide.
  • cytostaticum is cyclophosphamide.
  • the invention relates more preferably to the use of MRP4-inhibitors for preparing a pharmaceutical composition for treating respiratory complaints, containing the Syk-inhibitors of formula 1 and MRP4-inhibitors according to the invention, the MRP4-inhibitors preferably being selected from among dehydroepiandrosterone 3-sulphate, estradiol 3,17-disulphate, flurbiprofen, indomethacin, indoprofen, taurocholate, optionally in the form of the racemates, enantiomers, diastereomers and the pharmacologically acceptable acid addition salts and hydrates thereof.
  • the separation of enantiomers from the racemates can be carried out using methods known from the art (e.g. chromatography on chiral phases, etc.).
  • acid addition salts with pharmacologically acceptable acids are meant, for example, salts selected from among the hydrochlorides, hydrobromides, hydroiodides, hydrosulphates, hydrophosphates, hydromethanesulphonates, hydronitrates, hydromaleates, hydroacetates, hydrobenzoates, hydrocitrates, hydrofumarates, hydrotartrates, hydrooxalates, hydrosuccinates, hydrobenzoates and hydro-p-toluenesulphonates, preferably the hydrochlorides, hydrobromides, hydrosulphates, hydrophosphates, hydrofumarates and hydromethanesulphonates.
  • the invention further relates to pharmaceutical preparations which contain a triple combination of the Syk-inhibitors of formula 1, MRP4-inhibitors and another active substance according to the invention, such as, for example, an anticholinergic, a PDE4 inhibitor, a steroid, an LTD4-antagonist or a betamimetic, and the preparation thereof and the use thereof for treating respiratory complaints.
  • a triple combination of the Syk-inhibitors of formula 1, MRP4-inhibitors and another active substance according to the invention such as, for example, an anticholinergic, a PDE4 inhibitor, a steroid, an LTD4-antagonist or a betamimetic, and the preparation thereof and the use thereof for treating respiratory complaints.
  • Examples of iNOS-inhibitors within the scope of the present invention may also include antisense oligonucleotides, particularly those antisense oligonucleotides which bind iNOS-coding nucleic acids.
  • antisense oligonucleotides particularly antisense oligonucleotides, which bind iNOS coding nucleic acids, for modulating the expression of iNOS.
  • iNOS-antisense oligonucleotides as described particularly in WO 01/52902 may therefore also be combined with the PDE4-inhibitors of the present invention on account of their similar effect to the iNOS-inhibitors.
  • Suitable HMG-CoA reductase inhibitors which may be preferably used in double or triple combinations with the compounds of formula 1 are selected from among Atorvastatin, Cerivastatin, Flurvastatin, Lovastatin, Pitavastatin, Pravastatin, Rosuvastatin, Simvastatin, optionally in form of their pharmaceutically available acid addition salts, prodrugs, solvates or hydrates thereof.
  • Suitable forms for administration are for example tablets, capsules, solutions, syrups, emulsions or inhalable powders or aerosols.
  • the content of the pharmaceutically effective compound(s) in each case should be in the range from 0.1 to 90 wt. %, preferably 0.5 to 50 wt. % of the total composition, i.e. in amounts which are sufficient to achieve the dosage range specified hereinafter.
  • the preparations may be administered orally in the form of a tablet, as a powder, as a powder in a capsule (e.g. a hard gelatine capsule), as a solution or suspension.
  • a tablet e.g. a powder
  • a capsule e.g. a hard gelatine capsule
  • the active substance combination may be given as a powder, as an aqueous or aqueous-ethanolic solution or using a propellant gas formulation.
  • pharmaceutical formulations are characterised by the content of one or more compounds of formula 1 according to the preferred embodiments above.
  • Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate.
  • excipients for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate.
  • the tablets may also comprise several layers.
  • Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arabic, talc, titanium dioxide or sugar.
  • the core may also consist of a number of layers.
  • the tablet coating may consist of a number of layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.
  • Syrups containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavour enhancer, e.g. a flavouring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.
  • a sweetener such as saccharine, cyclamate, glycerol or sugar
  • a flavour enhancer e.g. a flavouring such as vanillin or orange extract.
  • suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.
  • Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gelatine capsules.
  • Suitable suppositories may be made for example by mixing with carriers provided for this purpose, such as neutral fats or polyethyleneglycol or the derivatives thereof.
  • Excipients which may be used include, for example, water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose), emulsifiers (e.g.
  • pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly disper
  • lignin e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone
  • lubricants e.g. magnesium stearate, talc, stearic acid and sodium lauryl sulphate.
  • the tablets may, of course, contain, apart from the abovementioned carriers, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additives such as starch, preferably potato starch, gelatine and the like.
  • additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additives such as starch, preferably potato starch, gelatine and the like.
  • lubricants such as magnesium stearate, sodium lauryl sulphate and talc may be used at the same time for the tabletting process.
  • the active substances may be combined with various flavour enhancers or colourings in addition to the excipients mentioned above.
  • the compounds of formula 1 are administered by inhalation, particularly preferably if they are administered once or twice a day.
  • the compounds of formula 1 have to be made available in forms suitable for inhalation.
  • Inhalable preparations include inhalable powders, propellant-containing metered-dose aerosols or propellant-free inhalable solutions, which are optionally present in admixture with conventional physiologically acceptable excipients.
  • propellant-free inhalable solutions also includes concentrates or sterile ready-to-use inhalable solutions.
  • the preparations which may be used according to the invention are described in more detail in the next part of the specification.
  • physiologically acceptable excipients may be used to prepare the inhalable powders according to the invention: monosaccharides (e.g. glucose or arabinose), disaccharides (e.g. lactose, saccharose, maltose), oligo- and polysaccharides (e.g. dextran), polyalcohols (e.g. sorbitol, mannitol, xylitol), salts (e.g. sodium chloride, calcium carbonate) or mixtures of these excipients with one another.
  • monosaccharides e.g. glucose or arabinose
  • disaccharides e.g. lactose, saccharose, maltose
  • oligo- and polysaccharides e.g. dextran
  • polyalcohols e.g. sorbitol, mannitol, xylitol
  • salts e.g. sodium chloride, calcium carbonate
  • lactose is the particularly preferred excipient, while lactose monohydrate is most particularly preferred.
  • the propellant-containing inhalable aerosols which may be used according to the invention may contain the compounds of formula 1 dissolved in the propellant gas or in dispersed form.
  • the propellant gases which may be used to prepare the inhalation aerosols according to the invention are known from the prior art. Suitable propellant gases are selected from among hydrocarbons such as n-propane, n-butane or isobutane and halohydrocarbons such as preferably fluorinated derivatives of methane, ethane, propane, butane, cyclopropane or cyclobutane.
  • the propellant gases mentioned above may be used on their own or in mixtures thereof.
  • propellant gases are fluorinated alkane derivatives selected from TG134a (1,1,1,2-tetrafluoroethane), TG227 (1,1,1,2,3,3,3-heptafluoropropane) and mixtures thereof.
  • the propellant-driven inhalation aerosols used within the scope of the use according to the invention may also contain other ingredients such as co-solvents, stabilisers, surfactants, antioxidants, lubricants and pH adjusters. All these ingredients are known in the art.
  • the compounds of formula 1 according to the invention are preferably used to prepare propellant-free inhalable solutions and inhalable suspensions.
  • Solvents used for this purpose include aqueous or alcoholic, preferably ethanolic solutions.
  • the solvent may be water on its own or a mixture of water and ethanol.
  • the solutions or suspensions are adjusted to a pH of 2 to 7, preferably 2 to 5, using suitable acids.
  • the pH may be adjusted using acids selected from inorganic or organic acids. Examples of particularly suitable inorganic acids include hydrochloric acid, hydrobromic acid, nitric acid, sulphuric acid and/or phosphoric acid.
  • organic acids examples include ascorbic acid, citric acid, malic acid, tartaric acid, maleic acid, succinic acid, fumaric acid, acetic acid, formic acid and/or propionic acid etc.
  • Preferred inorganic acids are hydrochloric and sulphuric acids. It is also possible to use the acids which have already formed an acid addition salt with one of the active substances.
  • ascorbic acid, fumaric acid and citric acid are preferred.
  • mixtures of the above acids may also be used, particularly in the case of acids which have other properties in addition to their acidifying qualities, e.g. as flavourings, antioxidants or complexing agents, such as citric acid or ascorbic acid, for example.
  • hydrochloric acid it is particularly preferred to use hydrochloric acid to adjust the pH.
  • Co-solvents and/or other excipients may be added to the propellant-free inhalable solutions used for the purpose according to the invention.
  • Preferred co-solvents are those which contain hydroxyl groups or other polar groups, e.g. alcohols—particularly isopropyl alcohol, glycols—particularly propyleneglycol, polyethyleneglycol, polypropyleneglycol, glycolether, glycerol, polyoxyethylene alcohols and polyoxyethylene fatty acid esters.
  • excipients and additives in this context denote any pharmacologically acceptable substance which is not an active substance but which can be formulated with the active substance or substances in the pharmacologically suitable solvent in order to improve the qualitative properties of the active substance formulation.
  • these substances have no pharmacological effect or, in connection with the desired therapy, no appreciable or at least no undesirable pharmacological effect.
  • the excipients and additives include, for example, surfactants such as soya lecithin, oleic acid, sorbitan esters, such as polysorbates, polyvinylpyrrolidone, other stabilisers, complexing agents, antioxidants and/or preservatives which guarantee or prolong the shelf life of the finished pharmaceutical formulation, flavourings, vitamins and/or other additives known in the art.
  • the additives also include pharmacologically acceptable salts such as sodium chloride as isotonic agents.
  • the preferred excipients include antioxidants such as ascorbic acid, for example, provided that it has not already been used to adjust the pH, vitamin A, vitamin E, tocopherols and similar vitamins or provitamins occurring in the human body.
  • Preservatives may be used to protect the formulation from contamination with pathogens. Suitable preservatives are those which are known in the art, particularly cetyl pyridinium chloride, benzalkonium chloride or benzoic acid or benzoates such as sodium benzoate in the concentration known from the prior art.
  • ready-to-use packs of a medicament for the treatment of respiratory complaints are provided, containing an enclosed description including for example the words respiratory disease, COPD or asthma, together with a imidazolyl-pyrimidine according to formula 1 and one or more combination partners selected from those described above.

Abstract

The invention relates to new substituted heteroaryls of formula 1
Figure US20180072744A1-20180315-C00001
    • wherein
    • A is selected from the group consisting of N and CH
    • D is selected from the group consisting of CH, N, NH, S and O,
    • E is selected from the group consisting of C and N,
    • T is selected from the group consisting of C and N,
    • G is selected from the group consisting of C and N,
    • and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
    • and wherein
    • R1, M and R3 are defined according to claim 1, and to the above compounds for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.

Description

  • The invention relates to new substituted heteroaryls of formula 1
  • Figure US20180072744A1-20180315-C00002
  • or of formula 1′
  • Figure US20180072744A1-20180315-C00003
  • wherein
  • A is selected from the group consisting of N and CH
  • D is CH,
  • E is selected from the group consisting of C and N,
  • T is C,
  • G is selected from the group consisting of C and N,
  • and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
  • and wherein
  • M, R3 and R1 are defined as in claim 1. Further the invention relates to the above compounds of formula 1 or of formula 1′ for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.
  • 1. BACKGROUND TO THE INVENTION 1.1 SYK-inhibitors
  • The present invention describes new compounds that inhibit the protein kinase Syk (spleen tyrosine kinase), the preparation and formulation thereof and their use for preparing a medicament.
  • Syk is an intracellular tyrosine kinase that has an important mediator function in the signal transduction of different receptors in B-cells, mast cells, monocytes, macrophages, neutrophils, T-cells, dendritic cells and epithelial cells. The receptors in which Syk performs an important function in signal transduction include for example the receptors for IgE (FcεRI) and IgG (FcγR1) on mast cells and B cells, the B-cell receptor (BCR) and the T-cell receptor (TCR) on B- and T-cells, the ICAM1 receptor (ICAM1 R) on epithelial cells of the respiratory tract, the DAP12-receptor on natural killer cells, dendritic cells and osteoclasts, the dectin 1-receptor on a subpopulation of T-helper cells (Th-17 cells), as well as the integrin receptors for β1-, β2- and β3-integrins on neutrophils, monocytes and macrophages (Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; Ulanova et al.; Expert Opion. Ther. Target (2005) 9(5); 901-921; Wang et al.; J. Immunol. (2006) 177, 6859-6870; Leib and Gut-Landmann et al.; Nature Immunology (2007) 8, 630-638; Slack et al., European J. Immunol. (2007) 37, 1600-1612). The molecular processes are described best for the signal transduction of the FcεRI. In mast cells the binding of IgE to FcεRI causes the cross-linking of IgE-receptors and the recruiting and activation of Lyn (a tyrosine kinase from the Src family). Active Lyn phoshorylates so-called ITAM motifs, which are present in many of the receptors listed above, and thereby generates binding sites for the SH2-domain of Syk. As a result of the binding to the ITAM motif Syk is activated and then phosphorylates various substrates which are needed for the release of allergic and inflammatory mediators such as e.g. histamine and β-hexosamidase (BHA), as well as for the synthesis of lipid mediators, such as e.g. prostaglandins and leukotrienes.
  • In view of its central function in different signal transduction pathways Syk has been discussed as a therapeutic target for different diseases such as e.g. allergic rhinitis, asthma, autoimmune diseases, rheumatoid arthritis, osteopenia, osteoporosis, COPD and various leukaemias and lymphomas (Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; Ulanova et al.; Expert Opion. Ther. Target (2005) 9(5); 901-921; Sigh and Masuda. Annual Reports in Medicinal Chemistry (2007) Vol 42; 379-391; Bajpai et al.; Expert Opin. Investig. Drugs (2008) Vol 15 (5); 641-659; Masuda and Schmitz; PPT (2008) Vol 21; 461-467; Riccaboni et al., Drug Discovery Today (2010) Vol 00 (0); 517-530; Efremov and Luarenti, Expert Opin Investig Drugs. (2011) 20(5):623-36).
  • Allergic rhinitis and asthma are diseases associated with allergic reactions and inflammatory processes and involving different cell types such as e.g. Mast cells, eosinophils, T-cells and dendritic cells. After exposure to allergens has occurred, the high affinity immunoglobulin receptors for IgE (FcERI) and IgG (FcγR1) are activated and induce the release of pro-inflammatory mediators and bronchoconstrictors. An inhibitor of the Syk kinase activity should thus be able to inhibit these steps.
  • Rheumatoid arthritis (RA) is an autoimmune disease in which the bones and ligaments structures surrounding the joints are progressively destroyed. In the pathophysiology of RA, B-cells play a significant role, as has been demonstrated for example by the therapeutic use of rituximab, a B cell-depleting antibody. In addition to the function of Syk in the signal transduction of the BCR (which after being stimulated also induces the release of pro-inflammatory mediators), Syk also plays an important part in the maturation and proliferation of B cells (Cheng et al. Nature (1995) 378, 303-306, Cornall et al., PNAS (2000) 97(4), 1713-1718). An inhibitor of the Syk kinase activity may thus offer a therapeutic option for the treatment of autoimmune diseases such as RA and diseases with an increased proliferation of B cells, such as e.g. B-cell lymphomas.
  • Chronic obstructive pulmonary disease (COPD) is characterised by a successive deterioration in lung function and chronic inflammation of the airways, which is initiated and produced by noxious substances of all kinds and contributes to the maintenance of the course of the disease. At a cellular level, in COPD there is in particular a multiplication of T-lymphocytes, neutrophils, granulocytes and macrophages. In particular, there is an increase in the number of CD8-positive lymphocytes, that is directly connected with the impairment of lung function. Another characteristic of COPD are acute deteriorations in lung function (exacerbations), characterised by viral (e.g. Rhinovirus), or bacterial (e.g. Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis) infections.
  • In view of the pro-inflammatory function of Syk in macrophages, T-cells and neutrophils as described above (see: Wong et al.; Expert Opin. Investig. Drugs (2004) 13(7), 743-762; and references cited therein) an inhibitor of the Syk kinase activity could be a new therapeutic approach to the treatment of the inflammatory processes that underlie COPD. It has also been shown that Syk in epithelial cells of the respiratory tract is involved in the ICAM1R-mediated uptake and subsequent replication of the Rhinovirus and that a si-RNA against Syk blocks these steps (Wang et al.; J. Immunol. (2006) 177, 6859-6870; Lau et al.; J. Immunol. (2008) 180, 870-880). Thus, an inhibitor of the Syk kinase activity could also be used therapeutically in exacerbations caused by Rhinoviruses.
  • Various studies suggest that Syk is involved in the malignant transformation of lymphocytes (summarised in Sigh and Masuda, Annual Reports in Medicinal Chemistry (2007) Vol 42; 379-391). A TEL-Syk fusion protein with a constitutive Syk activity transformed B cells of a patient with myelodysplastic syndrome, a constitutively active ITK-Syk fusion protein was isolated from patients with peripheral T-cell lymphomas (PTCL). Moreover, constitutively active Syk was found in B-cell lymphoma cells of patients, especially in B-lineage acute lymphoblastic leukemia (B-ALL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphomas and B cell Non-Hodgkin Lymphomas (NHLs) as well as in acute myeloid leukemia (AML). On the basis of these data it seems that Syk is a proto-oncogene in haematopoietic cells and represents a potential target for the treatment of certain leukaemias and lymphomas.
  • Idiophathic thrombocytopenic purpura (ITP) is an autoimmune disease in which IgG autoantibodies against antigens present on platelets bind to and destroy platelets. Patients with ITP have an accelerated clearence of circulating IgG-coated platelets via macrophages in the spleen and the liver. In view of the pro-inflammatory FcγR-mediated function of Syk in macrophages an inhibitor of Syk is considered to have a therapeutic benefit in FcγR-mediated cytopenias like ITP. Indeed the Syk inhibitor R788 (R406) improved platelet counts in a single center, oben label study in patients with ITP (Podolanczuk et al; Blood (2009) 113, 3154-3169).
  • Bullous pemphigoid (Ujiie et al. Journal of Dermatology 2010; 37: 194-204) is a chronic, autoimmune, subepidermal, blistering skin disease that rarely involves mucous membranes. Bullous pemphigoid is characterized by the presence of immunoglobulin G (IgG) autoantibodies specific for the hemidesmosomal bullous pemphigoid antigens BP230 (BPAg1) and BP180 (BPAg2). Pemphigus vulgaris (Venugopal et al. Dermatol. Clin. 2011;29:373-80) is a chronic blistering skin disease with skin lesions that are rarely pruritic, but which are often painful. Pemphigus vulgaris is an autoimmune disease caused by IgG autoantibodies directed against both desmoglein 1 and desmoglein 3 resulting in the loss of cohesion between keratinocytes in the epidermis. It is characterized by extensive flaccid blisters and mucocutaneous erosions. In both diseases IgG autoantibodies bind to Fc receptor gamma (FcRγ) and activate FcRγ and downstream signaling via Syk kinase. Thus, an inhibitor of the Syk kinase activity which blocks downstream signalling of the FcRg could be used therapeutically to treat patients with bullous pemphigoid and pemphigus vulgaris.
  • Systemic lupus erythematosus (SLE) is a chronic autoimmune disease which can affect basically any organ of the body. It is characterised by a multisystem inflammation of the microvascular and the presence of autoantibodies. FcγR-deficient mice are protected from several aspects of SLE in disease-related preclinical models, suggesting that an inhibitor of Syk can have a therapeutic benefit in SLE in view of the pro-inflammatory FcγR-mediated function of Syk in various cells.
  • 1.2 Prior art
  • 1,6-Naphthyridines are known as SYK-inhibitors. For example U.S. Pat. Nos. 3,928,367, 4,017,500, 4,115,395 and 4,260,759 describe 5-amino-1,6-naphthyridines with an antifungal and antibacterial activity. Further, WO 9918077 describes 5-piperazinyl-1,6-naphthyridines as serotonin antagonists. Additionally, U.S. Pat. No. 7,321,041 describes substituted 1,6-naphthyridines as SYK-inhibitors, however these 1,6-naphthyridines have a completely different substitution pattern from the compounds according to the invention. Also WO 2011092128 discloses 1,6-naphthyridines which are substituted in 5- and in 7-position.
  • In WO 2012/167733, WO 2012/167423 and in WO 201 2/1 2331 2 other naphthryidine derivatives such as pyrido[3,4-b]pyrazines which were also substituted in 5- and in 7-position have been disclosed as SYK-inhibitors.
  • Additionally, WO 01/83485 discloses substituted imidazopyrimidines and triazolopyrimidines as SYK-inhibitors, whereas WO 2008/113469 discloses substituted imidazo- and triazolopyrimidines as GSK 3β-inhibitors.
  • Also quinolones are known as SYK-inhibitors. For instance, WO 2006038041 and WO 2013014060 both disclose quinoline-compounds which are substituted in the 5- and 7-position, however the substitution pattern—in particular in the 7-position—is completely different from the one of the compounds of formula 1 of the instant invention.
  • Surprisingly it has now been found that the compounds of formulas 1 and 1′ and in particular the compounds of formulas 1a, 1a′, 1c, 1c′, are particularly suitable for the treatment of respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases, autoimmune diseases, inflammatory diseases and diseases of the peripheral or central nervous system, particularly for the treatment of asthma, allergic rhinitis, rheumatoid arthritis, allergic dermatitis, lupus erythematosus (SLE) and COPD, in particular because all these compounds of the present invention show the following desired capacities:
      • high SYK inhibition (reflected by “low” IC50-values with respect to SYK-inhibition)
      • very low inhibition of the kinase Aurora B (reflected by “high” IC50-values with respect to inhibition of AURB)
      • low inhibition of the kinase FLT-3 (reflected by “high” IC50-values with respect to inhibition of FLT-3)
      • low inhibition of the kinase GSK3β (reflected by “high” IC50-values with respect to inhibition of GSK3β)
  • This was completely surprising for a person skilled in the art, since the compounds of formula 1 and 1′ of the instant invention have several significant structural differences compared to the previously known prior art compounds.. For instance the compounds of formula 1 and 1′ of the instant invention differ from the previously known 1,6-naphthyridines,quinolones, pyrido[3,4-b]pyrazines, imidazopyrimidines and triazolopyrimidines therein that they combine the following features:
      • they all possess diverse core modifications in the central bicyclic heteroaromatic ring system (for instance core modifications resulting in benzopyrazoles etc.)
      • that they all have a methyl-group-substitution attached to position E in formula 1 and/or 1′ and
      • that they all possess a residue of formula T
  • Figure US20180072744A1-20180315-C00004
  • preferably a residue of formula T′
  • Figure US20180072744A1-20180315-C00005
  • 2. DESCRIPTION OF THE INVENTION
  • The present invention concerns compounds of formula 1
  • Figure US20180072744A1-20180315-C00006
  • wherein
  • A is selected from the group consisting of N and CH
  • D is CH
  • E is selected from the group consisting of C and N,
  • T is C
  • G is selected from the group consisting of C and N,
  • and wherein each of the broken (dotted) double bonds in ring 1 are selected from either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
  • and wherein
  • M is selected from the group consisting of —CH2—, —O—, —NH— and —N(C1-4-alkyl)-;
  • R3 is selected from the group consisting of methyl and ethyl;
  • Het is selected from the group consisting of
  • a five- to six-membered monocyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and 0,
  • and a nine- to eleven-membered bicyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and 0;
  • Hetaryl is selected from the group consisting of
  • a five- to six-membered monocyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and 0;
  • and a nine- to eleven-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from N, S and 0;
  • and wherein
  • R1 is selected from the group consisting of
  • C6-10-aryl, Het and Hetaryl;
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, —OH, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,—NH(CH3), —N(CH3)2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, —OH, halogen and C1-3-alkyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • A preferred embodiment of the instant invention relates to the aforementioned compounds of formula 1′
  • Figure US20180072744A1-20180315-C00007
  • wherein residues A, D, E, T, G, Het, Hetaryl, R1 and R3 are defined as mentioned above,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the instant invention concerns the aforementioned compounds of formula 1 or of formula 1′, wherein
  • D is CH,
  • and wherein E is N
  • and wherein
  • M is —CH2
  • and wherein
  • R3 is methyl,
  • and wherein
  • G is C (carbon)
  • R1 is selected from the group consisting of
  • phenyl, Het and Hetaryl;
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In another preferred embodiment the instant invention relates to the aforementioned compounds of formula 1a
  • Figure US20180072744A1-20180315-C00008
  • or of formula 1a′,
  • Figure US20180072744A1-20180315-C00009
  • wherein
  • R1 is selected from the group consisting of
  • phenyl, Het and Hetaryl;
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the invention relates to compounds of the above-mentioned formula 1a or of formula 1a′, wherein R1 is
      • a) either selected from the group consisting of Het and Hetaryl;
      • which is optionally further substituted by one, two or three substituents Z,
      • whereby each Z is a substituent selected from the group consisting of —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl,
  • —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
      • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • or wherein R1 is
  • b) phenyl,
  • which is optionally further substituted by one, two or three substituents Z,
      • whereby each Z is a substituent selected from the group consisting of, —CN, halogen, —C1-6—alkyl, —(C1-3-alkylene)-Hetaryl, —(C1-3-alkylene)-Het, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo and —C1-4-alkyl,
      • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In another preferred embodiment the invention relates to compounds of the above-mentioned formula 1a or of formula 1a′, wherein
  • R1 is either
      • a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S,
      • or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,
  • wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the invention relates to the above compounds of the aforementioned formula 1a or formula 1a′, wherein
  • R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
  • wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—methyl, —O—ethyl, O-propyl, O-butyl, —C1-3-haloalkyl,three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In another preferred embodiment the invention concerns the above compounds of the aformementioned formula 1a or of formula 1a′, wherein
  • R1 is phenyl,
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a particularly preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1a or formula 1a′, which is selected from the group consisting of
  • Figure US20180072744A1-20180315-C00010
    Figure US20180072744A1-20180315-C00011
    Figure US20180072744A1-20180315-C00012
    Figure US20180072744A1-20180315-C00013
    Figure US20180072744A1-20180315-C00014
    Figure US20180072744A1-20180315-C00015
    Figure US20180072744A1-20180315-C00016
    Figure US20180072744A1-20180315-C00017
    Figure US20180072744A1-20180315-C00018
    Figure US20180072744A1-20180315-C00019
    Figure US20180072744A1-20180315-C00020
    Figure US20180072744A1-20180315-C00021
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In another preferred embodiment the instant invention concerns compounds of formula 1c
  • Figure US20180072744A1-20180315-C00022
  • or of formula 1c′
  • Figure US20180072744A1-20180315-C00023
  • wherein
  • R1 is selected from the group consisting of
  • phenyl, Het and Hetaryl;
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein R1 is
  • a) either selected from the group consisting of Het and Hetaryl;
      • which is optionally further substituted by one, two or three substituents Z,
      • whereby each Z is a substituent selected from the group consisting of —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
      • which is optionally further substituted by one, two or three substituents X,
      • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl,
      • —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,
      • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • or wherein R1 is
  • b) phenyl,
      • which is optionally further substituted by one, two or three substituents Z,
      • whereby each Z is a substituent selected from the group consisting of, —CN, halogen, —C1-6-alkyl, —(C1-3-alkylene)-Hetaryl, —(C1-3-alkylene)-Het, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl,
      • which is optionally further substituted by one, two or three substituents X,
      • whereby each X is selected from the group consisting of halogen, oxo and —C1-4-alkyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In another preferred embodiment the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R1 is either
      • a monocyclic five- to six-membered heteroaromate with 1, 2 or 3 heteroatoms each independently from one another selected from the group consisting of N, O and S,
      • or a 9- to 11-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, O and S,
  • wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R1 is selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, thiophenyl, furanyl, pyrazolopyridinyl, indazolyl, thiazolyl, imidazo-pyridinyl and indolyl,
  • wherein this R1-residue is attached to the rest of the molecule either via a C-atom or via an N-atom and is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, -methyl, -ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, —O—methyl, —O—ethyl, O-propyl, O-butyl, —C1-3-haloalkyl,three-, four, five- or six-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further preferred embodiment the instant invention concerns the above compounds of the aforementioned formula 1c or formula 1c′, wherein
  • R1 is phenyl,
  • which is optionally further substituted by one, two or three substituents Z,
  • whereby each Z is a substituent selected from the group consisting of
  • —OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), —O-Het,
  • which is optionally further substituted by one, two or three substituents X,
  • whereby each X is selected from the group consisting of halogen, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, —NH2,
  • whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl,
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a particularly preferred embodiment the instant invention relates to the above compounds of the aforementioned formula 1c or formula 1c′, which is selected from the group consisting of
  • Figure US20180072744A1-20180315-C00024
    Figure US20180072744A1-20180315-C00025
    Figure US20180072744A1-20180315-C00026
    Figure US20180072744A1-20180315-C00027
    Figure US20180072744A1-20180315-C00028
    Figure US20180072744A1-20180315-C00029
  • and the pharmaceutically acceptable salts of the aforementioned compounds.
  • In a further aspect the instant invention refers to an intermediate compound selected from the group consisting of formula 6
  • Figure US20180072744A1-20180315-C00030
      • of formula 7
  • Figure US20180072744A1-20180315-C00031
      • of formula 8
  • Figure US20180072744A1-20180315-C00032
      • and of formula 11
  • Figure US20180072744A1-20180315-C00033
      • wherein E, D, G, T and R1 are defined above or as defined in claim 1 and wherein Hal is Cl or Br
      • and wherein PG is a protecting group selected from the group consisting of benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl.
  • In a further aspect the instant invention refers to an intermediate compound selected from the group consisting of
      • formula 5.1
  • Figure US20180072744A1-20180315-C00034
  • and
      • of formula 10.2
  • Figure US20180072744A1-20180315-C00035
  • In a further aspect the instant invention refers to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease which can be treated by inhibition of the SYK enzyme.
  • In another preferred aspect the instant invention relates to one of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of allergic rhinitis, asthma, COPD, adult respiratory distress syndrome, bronchitis, B-cell lymphoma, dermatitis and contact dermatitis, allergic dermatitis, allergic rhinoconjunctivitis, rheumatoid arthritis, anti-phospholipid syndrome, Berger's disease, Evans's syndrome, ulcerative colitis,allergic antibody-based glomerulonephritis, granulocytopenia, Goodpasture's syndrome, hepatitis, Henoch-Schönlein purpura, hypersensitivity vasculitis, immunohaemolytic anaemia, autoimmune haemolytic anemia, idiopathic thrombocytopenic purpura, Kawasaki syndrome, allergic conjunctivitis, lupus erythematodes, lupus nephritis, capsule cell lymphoma, neutropenia, non-familial lateral sclerosis, artheriosclerosis, Crohn's disease, multiple sclerosis, myasthenia gravis, osteoporosis, osteolytic diseases, osteopenia, psoriasis, Sjogren's syndrome, sclerodermy, T-cell lymphoma, urticaria/angiooedema, Wegener's granulomatosis and coeliac disease.
  • In another preferred aspect the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, adult respiratory distress syndrome, bronchitis, allergic dermatitis, contact dermatitis, idiopathic thrombocytopenic purpura, rheumatoid arthritis, lupus erythematodes, lupus nephritis and allergic rhinoconjunctivitis.
  • In another particularly preferred aspect the instant invention concerns the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) for the treatment of a disease selected from the group consisting of asthma, COPD, allergic rhinitis, allergic dermatitis, lupus erythematodes, lupus nephritis and rheumatoid arthritis.
  • In another preferred aspect the instant invention concerns pharmaceutical formulations which contain one or more of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) and a pharmaceutically acceptable excipient.
  • In another preferred aspect the instant invention concerns pharmaceutical formulations which contain one or more compounds of the aforementioned compounds of formula 1 or 1′ (or of any of the sub-formulas 1a, 1a′, 1c, 1c′) in combination with an active substance selected from the group consisting of anticholinergics, betamimetics, corticosteroids, PDE4-inhibitors, EGFR-inhibitors, LTD4-antagonists, CCR3-inhibitors, iNOS-inhibitors, CRTH2-antagonists,HMG-CoA reductase inhibitors and NSAIDs.
  • 3. TERMS AND DEFINITIONS USED
  • Unless stated otherwise, all the substituents are independent of one another. If for example a number of C1-6-alkyl groups are possible substituents at a group, in the case of three substituents, for example, C1-6-alkyl could represent, independently of one another, a methyl, an n-propyl and a tert-butyl.
  • Within the scope of this application, in the definition of possible substituents, these may also be presented in the form of a structural formula. An asterisk (*) in the structural formula of the substituent is to be understood as being the linking point to the rest of the molecule. Mor3eover, the atom of the substituent following the linking point is understood as being the atom in position number 1. Thus for example the groups N-piperidinyl (I), 4-piperidinyl (II), 2-tolyl (III), 3-tolyl (IV) and 4-tolyl (V) are represented as follows:
  • Figure US20180072744A1-20180315-C00036
  • If there is no asterisk (*) in the structural formula of the substituent, each hydrogen atom may be removed at the substituent and the valency thus freed may serve as a binding site to the rest of a molecule. Thus, for example, VI
  • Figure US20180072744A1-20180315-C00037
  • may represent 2-tolyl, 3-tolyl, 4-tolyl and benzyl.
  • Alternatively to the * within the scope of this application X1 is also understood as being the linking point of the group R1 to the structure of formula 1 and X2 as being the linking point of the group R2 to the structure of formula 1.
  • By the term “C1-6-alkyl” (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms and by the term “C1-3-alkyl” are meant branched and unbranched alkyl groups with 1 to 3 carbon atoms. “C1-4-alkyl” accordingly denotes branched and unbranched alkyl groups with 1 to 4 carbon atoms. Alkyl groups with 1 to 4 carbon atoms are preferred. Examples of these include: methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl or hexyl. The abbreviations Me, Et, n-Pr, i-Pr, n—Bu, i—Bu, t—Bu, etc., may also optionally be used for the above-mentioned groups. Unless stated otherwise, the definitions propyl, butyl, pentyl and hexyl include all the possible isomeric forms of the groups in question. Thus, for example, propyl includes n-propyl and iso-propyl, butyl includes iso-butyl, sec-butyl and tert-butyl etc.
  • By the term “C1-6-alkylene” (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms and by the term “C1-4-alkylene” are meant branched and unbranched alkylene groups with 1 to 4 carbon atoms. Alkylene groups with 1 to 4 carbon atoms are preferred. Examples of these include: methylene, ethylene, propylene, 1-methylethylene, butylene, 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene, pentylene, 1,1-dimethylpropylene, 2,2-dimethylpropylene, 1,2-dimethylpropylene, 1,3-dimethylpropylene or hexylene. Unless stated otherwise, the definitions propylene, butylene, pentylene and hexylene include all the possible isomeric forms of the groups in question with the same number of carbons. Thus, for example, propyl includes also 1-methylethylene and butylene includes 1-methylpropylene, 1,1-dimethylethylene, 1,2-dimethylethylene.
  • If the carbon chain is substituted by a group which together with one or two carbon atoms of the alkylene chain forms a carbocyclic ring with 3, 5 or 6 carbon atoms, this includes, inter alia, the following examples of the rings:
  • Figure US20180072744A1-20180315-C00038
  • By the term “C2-6-alkenyl” (including those which are part of other groups) are meant branched and unbranched alkenyl groups with 2 to 6 carbon atoms and by the term “C2-4-alkenyl” are meant branched and unbranched alkenyl groups with 2 to 4 carbon atoms, provided that they have at least one double bond. Alkenyl groups with 2 to 4 carbon atoms are preferred. Examples include: ethenyl or vinyl, propenyl, butenyl, pentenyl or hexenyl. Unless stated otherwise, the definitions propenyl, butenyl, pentenyl and hexenyl include all the possible isomeric forms of the groups in question. Thus, for example, propenyl includes 1-propenyl and 2-propenyl, butenyl includes 1-, 2- and 3-butenyl, 1-methyl-1-propenyl, 1-methyl-2-propenyl etc.
  • By the term “C2-6-alkenylene” (including those which are part of other groups) are meant branched and unbranched alkenylene groups with 2 to 6 carbon atoms and by the term “C2-4-alkenylene” are meant branched and unbranched alkylene groups with 2 to 4 carbon atoms. Alkenylene groups with 2 to 4 carbon atoms are preferred. Examples of these include: ethenylene, propenylene, 1-methylethenylene, butenylene, 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene, pentenylene, 1,1-dimethylpropenylene, 2,2-dimethylpropenylene, 1,2-dimethylpropenylene, 1,3-dimethylpropenylene or hexenylene. Unless stated otherwise, the definitions propenylene, butenylene, pentenylene and hexenylene include all the possible isomeric forms of the groups in question with the same number of carbons. Thus, for example, propenyl also includes 1-methylethenylene and butenylene includes 1-methylpropenylene, 1,1-dimethylethenylene, 1,2-dimethylethenylene.
  • By the term “aryl” (including those which are part of other groups) are meant aromatic ring systems with 6 or 10 carbon atoms. Examples include: phenyl or naphthyl, the preferred aryl group being phenyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • By the term “aryl-C1-6-alkylene” (including those which are part of other groups) are meant branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by an aromatic ring system with 6 or 10 carbon atoms. Examples include: benzyl, 1- or 2-phenylethyl or 1- or 2-naphthylethyl. Unless otherwise stated, the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • By the term “heteroaryl-C1-6-alkylene” (including those which are part of other groups) are meant—even though they are already included under “aryl-C1-6-alkylene”—branched and unbranched alkylene groups with 1 to 6 carbon atoms, which are substituted by a heteroaryl.
  • If not specifically defined otherwise, a heteroaryl of this kind includes five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed. The following are examples of five- or six-membered heterocyclic aromatic groups or bicyclic heteroaryl rings:
  • Figure US20180072744A1-20180315-C00039
  • Unless otherwise stated, these heteroaryls may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • The following are examples of heteroaryl-C1-6-alkylenes:
  • Figure US20180072744A1-20180315-C00040
  • By the term “C1-6-haloalkyl” (including those which are part of other groups) are meant branched and unbranched alkyl groups with 1 to 6 carbon atoms, which are substituted by one or more halogen atoms. By the term “C1-4-alkyl” are meant branched and unbranched alkyl groups with 1 to 4 carbon atoms, which are substituted by one or more halogen atoms. Alkyl groups with 1 to 4 carbon atoms are preferred. Examples include: CF3, CHF2, CH2F, CH2CF3.
  • By the term “C3-7-cycloalkyl” (including those which are part of other groups) are meant cyclic alkyl groups with 3 to 7 carbon atoms, if not specifically defined otherwise. Examples include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Unless otherwise stated, the cyclic alkyl groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • If not specifically defined otherwise, by the term “C3-10-cycloalkyl” are also meant monocyclic alkyl groups with 3 to 7 carbon atoms and also bicyclic alkyl groups with 7 to 10 carbon atoms, or monocyclic alkyl groups which are bridged by at least one C1-3-carbon bridge.
  • By the term “heterocyclic rings” or “heterocycle” are meant, unless stated otherwise, five-, six- or seven-membered, saturated, partially saturated or unsaturated heterocyclic rings which may contain one, two or three heteroatoms, selected from among oxygen, sulphur and nitrogen, while the ring may be linked to the molecule through a carbon atom or through a nitrogen atom, if there is one. Although included by the term “heterocyclic rings” or “heterocycles”, the term “saturated heterocyclic ring” refers to five-, six- or seven-membered saturated rings. Examples include:
  • Figure US20180072744A1-20180315-C00041
  • Although included by the term “heterocyclic rings” or “heterocyclic group”, the term “partially saturated heterocyclic group” refers to five-, six- or seven-membered partially saturated rings which contain one or two double bonds, without so many double bonds being produced that an aromatic system is formed, unless specifically defined otherwise. Examples include:
  • Figure US20180072744A1-20180315-C00042
  • Although included by the term “heterocyclic rings” or “heterocycles”, the term “heterocyclic aromatic rings”, “unsaturated heterocyclic group” or “heteroaryl” refers to five- or six-membered heterocyclic aromatic groups or 5-10-membered, bicyclic heteroaryl rings which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contain so many conjugated double bonds that an aromatic system is formed, unless not specifically defined otherwise. Examples of five- or six-membered heterocyclic aromatic groups include:
  • Figure US20180072744A1-20180315-C00043
  • Unless otherwise mentioned, a heterocyclic ring (or heterocycle) may be provided with a keto group. Examples include:
  • Figure US20180072744A1-20180315-C00044
  • Although covered by the term “cycloalkyl”, the term “bicyclic cycloalkyls” generally denotes eight-, nine- or ten-membered bicyclic carbon rings. Examples include
  • Figure US20180072744A1-20180315-C00045
  • Although already included by the term “heterocycle”, the term “bicyclic heterocycles” generally denotes eight-, nine- or ten-membered bicyclic rings which may contain one or more heteroatoms, preferably 1-4, more preferably 1-3, even more preferably 1-2, particularly one heteroatom, selected from among oxygen, sulphur and nitrogen, unless not specifically defined otherwise. The ring may be linked to the molecule through a carbon atom of the ring or through a nitrogen atom of the ring, if there is one. Examples include:
  • Figure US20180072744A1-20180315-C00046
  • Although already included by the term “aryl”, the term “bicyclic aryl” denotes a 5-10 membered, bicyclic aryl ring which contains sufficient conjugated double bonds to form an aromatic system. One example of a bicyclic aryl is naphthyl.
  • Although already included under “heteroaryl”, the term “bicyclic heteroaryl” denotes a 5-10 membered, bicyclic heteroaryl ring which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and contains sufficient conjugated double bonds to form an aromatic system, unless specifically defined otherwise.
  • Although included by the term “bicyclic cycloalkyls” or “bicyclic aryl”, the term “fused cycloalkyl” or “fused aryl” denotes bicyclic rings wherein the bridge separating the rings denotes a direct single bond. The following are examples of a fused, bicyclic cycloalkyl:
  • Figure US20180072744A1-20180315-C00047
  • Although included by the term “bicyclic heterocycles” or “bicyclic heteroaryls”, the term “fused bicyclic heterocycles” of “fused bicyclic heteroaryls” denotes bicyclic 5-10 membered heterorings which contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen and wherein the bridge separating the rings denotes a direct single bond. The “fused bicyclic heteroaryls” moreover contain sufficient conjugated double bonds to form an aromatic system. Examples include pyrrolizine, indole, indolizine, isoindole, indazole, purine, quinoline, isoquinoline, benzimidazole, benzofuran, benzopyran, benzothiazole, benzothiazole, benzoisothiazole, pyridopyrimidine, pteridine, pyrimidopyrimidine,
  • Figure US20180072744A1-20180315-C00048
  • “Halogen” within the scope of the present invention denotes fluorine, chlorine, bromine or iodine. Unless stated to the contrary, fluorine, chlorine and bromine are regarded as preferred halogens.
  • Compounds of general formulas 1 or 1′ may have acid groups, mainly carboxyl groups, and/or basic groups such as e.g. amino functions. Compounds of general formulas 1 or 1′ may therefore be present as internal salts, as salts with pharmaceutically usable inorganic acids such as hydrochloric acid, sulphuric acid, phosphoric acid, sulphonic acid or organic acids (such as for example maleic acid, fumaric acid, citric acid, tartaric acid or acetic acid) or as salts with pharmaceutically usable bases such as alkali metal or alkaline earth metal hydroxides or carbonates, zinc or ammonium hydroxides or organic amines such as e.g. diethylamine, triethylamine, triethanolamine, inter alia.
  • As mentioned previously, the compounds of formulas 1 or 1′ may be converted into the salts thereof, particularly for pharmaceutical use into the physiologically and pharmacologically acceptable salts thereof. These salts may be present on the one hand as physiologically and pharmacologically acceptable acid addition salts of the compounds of formula 1 with inorganic or organic acids. On the other hand, the compound of formulas 1 or 1′ when R is hydrogen may be converted by reaction with inorganic bases into physiologically and pharmacologically acceptable salts with alkali or alkaline earth metal cations as counter-ion. The acid addition salts may be prepared for example using hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid or maleic acid. It is also possible to use mixtures of the above-mentioned acids. To prepare the alkali and alkaline earth metal salts of the compounds of formulas 1 or 1′ wherein R denotes hydrogen, it is preferable to use the alkali and alkaline earth metal hydroxides and hydrides, of which the hydroxides and hydrides of the alkali metals, particularly sodium and potassium, are preferred, while sodium and potassium hydroxide are particularly preferred.
  • The compounds of general formulas 1 or 1′ may optionally be converted into the salts thereof, particularly for pharmaceutical use into the pharmacologically acceptable acid addition salts with an inorganic or organic acid. Examples of suitable acids for this purpose include succinic acid, hydrobromic acid, acetic acid, fumaric acid, maleic acid, methanesulphonic acid, lactic acid, phosphoric acid, hydrochloric acid, sulphuric acid, tartaric acid or citric acid. It is also possible to use mixtures of the above-mentioned acids.
  • The invention relates to the compounds of formula 1 in question, optionally in the form of the individual optical isomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • The compounds of formula 1, 1a and 1c according to the invention may optionally be present as racemates, but may also be obtained as pure enantiomers, i.e. in the (R) or (S) form. Preferred are the compounds with the specific stereochemistry of formula 1′, in particular the compounds with the specific stereochemistry of one of formulas 1a′ and 1c′.
  • The invention relates to the compounds in question, optionally in the form of the individual optical isomers, diastereomers, mixtures of diastereomers, mixtures of the individual enantiomers or racemates, in the form of the tautomers as well as in the form of the free bases or the corresponding acid addition salts with pharmacologically acceptable acids—such as for example acid addition salts with hydrohalic acids—for example hydrochloric or hydrobromic acid—or organic acids—such as for example oxalic, fumaric, diglycolic or methanesulphonic acid.
  • The invention relates to the respective compounds of formulas 1 or 1′ in the form of the pharmacologically acceptable salts thereof. These pharmacologically acceptable salts of the compounds of formulas 1 or 1′ may also be present in the form of their respective hydrates (e.g. Monohydrates, dihydrates, etc.) as well as in the form of their respective solvates.
  • By a hydrate of the compound according to the formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, containing water of crystallisation.
  • By a solvate of the compound according to formulas 1 or 1′ is meant, for the purposes of the invention, a crystalline salt of the compound according to formulas 1 or 1′, which contains solvent molecules (e.g. Ethanol, methanol etc) in the crystal lattice.
  • The skilled man will be familiar with the standard methods of obtaining hydrates and solvates (e.g. recrystallisation from the corresponding solvent or from water).
  • 4. METHODS OF PREPARATION
  • The Examples according to the invention were prepared as shown in Schemes 1, 2 or 3.
  • Figure US20180072744A1-20180315-C00049
  • D is CH,
  • G is C or N
  • T is C
  • E is C or N, preferably N
  • Hal is Br or Cl
  • with X being —B(OH)2, -boronic acid pinacolester, -trifluoroborate or —SnBu3
  • PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)
  • and R1 is as herein before defined.
  • Figure US20180072744A1-20180315-C00050
  • D is CH,
  • G is C or N
  • T is C
  • E is C or N, preferably N
  • A is CH or N
  • Hal is Br or Cl
  • with X being —B(OH)2, -boronic acid pinacolester, -trifluoroborate or —SnBu3
  • PG is protecting group (e.g. benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl)
  • and R1 is as herein before defined.
  • 4.1. Starting Materials of Formula 2, 3, 4, 5 and 10 4.1.1. Synthesis of Lactams 2 from Scheme 1 and 2
  • Synthesis of Synthesis of (R)-4-[(R)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.1 for Examples 1-3, 7-13, 17, 50-84 and (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.2 for Examples 4-6, 14-16, 18-49.
  • Step 1: Synthesis of (1′R,3R,/S)-1-(1″-(4-Methoxyphenylethyl)-5-oxo-3-pyrrolidine carboxylic acid (mixture of diastereoisomers)
  • Figure US20180072744A1-20180315-C00051
  • A suspension of 100 g of (R)-1-(4-methoxy-phenyl)-ethylamine and 95 g itaconic acid in 0.5 L 1-methyl-2-pyrrolidinone was heated to 80° C. for 1 hour. The solution was stirred for additional 4 hours at 120° C. The reaction mixture was cooled to 25° C. and poured into 1.5 L of demineralized water. The precipitate was filtered, washed with demineralized water and dried at 50° C.
  • Yield: 195 g (quantitative yield) solid as a mixture of diastereoisomers
  • Analysis (method G): Rt: 2.6 min and 2.7 min, (M+H)+: 264
  • Step 2: Synthesis of (R/S)-N-Methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide as a mixture of diastereoisomers
  • Figure US20180072744A1-20180315-C00052
  • 260 g of 1,1′-carbonyldiimidazole (CDI) were added to a solution of 285 g (1′R,3R/S)-1-(1′-(4-methoxyphenylethyl)-5-oxo-3-pyrrolidine carboxylic acid (mixture of diastereoisomers) in 1.4 L 2-methyltetrahydrofuran at 20° C. The suspension was stirred at 20° C. for 80 minutes. 235 mL ethyldiisopropylamine (DIPEA) and 130 g of N,O-dimethylhydroxylamine hydrochloride were added. The suspension was stirred for 3 hours at 20° C. Under cooling 850 mL 4M hydrochloric acid was added. The organic phase was separated and washed two times with 500 mL 1 N hydrochloric acid. The aqueous phase was reextracted two times with 500 mL ethyl acetate. The combined organic phases were dried over sodium sulfate. After filtration the solvent was evaporated under reduced pressure.
  • Yield: 271 g (82% of theory) of (R/S)-N-Methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide (mixture of diastereoisomers) as an oil.
  • Analysis (method H): Rt: 11.1 min (41 area %) and 13.8 min (59 area %), (M+H)+: 307
  • Step 3: Synthesis of (R/S)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidine-2-one as a mixture of diastereoisomers
  • Figure US20180072744A1-20180315-C00053
  • 530 mL of a 3M solution of methylmagnesium bromide in diethylether were added slowly to a cooled solution of 271 g of (R/S)-N-methoxy-5-oxo-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-3-carboxamide (mixture of diastereoisomers) in 1.4 L of 2-methyltetrahydrofuran so that the temperature remained under 0° C. After complete addition the temperature was kept for 75 minutes at 0° C. and then warmed up to 20° C. The suspension was stirred 16 hours at 20° C. Under cooling 650 mL of a 4M hydrochloric acid were added. The organic phase was separated and washed with 500 mL saturated sodium carbonate solution and with 500 mL saturated brine. The organic phase was dried over sodium sulfate. After filtration the solvent was evaporated under reduced pressure.
  • Yield: 188 g (81% of theory) of (R/S)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidine-2-one (mixture of diastereoisomers) as an oil.
  • Analysis (method H): Rt: 7.4 min and 9.6 min, (M+H)+: 262
  • Step 4: Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one under base induced epimerization conditions
  • Figure US20180072744A1-20180315-C00054
  • 103 g of a mixture of diastereoisomers (R/S)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one were dissolved in 155 mL 1-butanol at 25° C. 18 mL benzyltrimethylammonium hydroxide (40% solution in methanol) was added. The solution was stirred for 30 minutes at 25° C. The solution was cooled to 0° C. Precipitation started. The suspension was stirred for 15 minutes at 0° C. 100 mL n-heptane was added slowly and the suspension was stirred for 30 minutes at 0° C. The addition of 100 mL portions of n-heptane was repeated 4 times with subsequent stirring of the suspension at 0° C. for 30 minutes. The precipitate was isolated, washed with n-heptane and dried at 50° C.
  • Yield: 77.1 g of a beige solid (75% of theory) with a diastereoisomeric purity of ˜95: 5 (method H).
  • For further purification the crude product was dissolved in 310 mL 2-methyl-2-butanol at 40° C. (temperature<50° C). The solution was slowly cooled to 0° C. Precipitation started. At 0° C. 385 mL of n-heptane were added and the suspension was stirred for 1 hour. The precipitate was filtrated, washed with n-heptane and dried at 50° C.
  • Yield: 68.7 g (67% of theory) of a colorless solid with a diastereoisomeric purity of >99: 1.
  • Analysis (method H): Rt: 6.8 min, (M+H)+: 262
  • Step 4: Crystallization of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one under base induced epimerization conditions
  • Figure US20180072744A1-20180315-C00055
  • 13.2 g of a mixture of diastereoisomers (R/S)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one were dissolved in 18 mL of 1-butanol at 25° C. The solution was cooled to 3 ° C. and treated with 100mg of (R)-4-Acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl] pyrrolidine-2-one. The resulting mixture was agitated for 15 min at 3° C.; at which point, 2.3 mL benzyltrimethylammonium hydroxide (40% solution in methanol) were added. The solution was stirred for 30 minutes at 3° C. 64 mL n-heptane was added slowly over 1 h at 0 to 3° C. and the suspension was stirred for 60 minutes at 0° C. The precipitate was isolated, washed with n-heptane and dried at 30° C.
  • Yield: 10.6 g of a beige solid (80% of theory) with a diastereoisomeric purity of ˜98: 2 (method H).
  • Analysis (method H): Rt: 6.8 min, (M+H)+: 262
  • Step 5: Synthesis of (R)-4-[(R)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.1
  • Figure US20180072744A1-20180315-C00056
  • 94.6 mg of dichloro (pentamethylcyclopentadienyl)-iridium(III) dimer and 105 mg of (S,S)-N-(p-toluenesulfonyl)-1,2-diphenylethylendiamine [(R,R)-TsDPEN] were dissolved in 20 mL of acetonitrile and subsequently charged to a slurry of 50 g of (R)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one and 65 g of sodium formate in 500 mL of water at 25° C. The slurry was heated to 60° C. and agitated at this temperature while sparging with nitrogen for 3 h. The reaction was diluted at 60° C. with 500 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed twice with 300 mL of water. The organic portion was concentrated to an oily solid. The residual material was crystallized three times from ethyl acetate and hexanes followed by drying in a vacuum oven with a nitrogen stream at 30° C.
  • 25.4 g of a beige solid with a diastereomeric purity of >99:1
  • Step 5: Synthesis of (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.2
  • Figure US20180072744A1-20180315-C00057
  • 9.46 mg of dichloro (pentamethylcyclopentadienyl)-iridium(III) dimer and 10.52 mg of (R,R)-N-(p-toluenesulfonyl)-1,2-diphenylethylendiamine [(R,R)-TsDPEN]were dissolved in 1 mL of acetonitrile and subsequently charged to a slurry of 5 g of (R)-4-acetyl-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidine-2-one and 6.5 g of sodium formate in 50 mL of water at 25° C. The slurry was heated to 60° C. and agitated at this temperature while sparging with nitrogen for 3 h. The reaction was diluted at 60° C. with 50 mL of isopropyl acetate and subsequently cooled to ambient temperature. The layers were separated, and the organic portion was washed with 20 mL of water. The organic portion was concentrated to an oil. The oil was dissolved in 8 mL of isopropyl acetate at reflux. The solution was cooled to ambient temperature wherein crystallization occurred. The mixture was diluted dropwise with 10 mL of heptane at ambient temperature. The mixture was agitated for 30 minutes. The solids were collected by filtration, washed with a solution of 20 vol % isopropyl acetate in heptane and dried in a vacuum oven with a nitrogen stream at 55° C. 3.82 g of a beige solid with a diastereomeric purity of 99:1
  • Analysis (method I): Rt: 12.9 min, (M+H)+: 264
  • Synthesis of [(1S)-1-[(3R)-1-[(1S)-1-(4-Methoxyphenyl)ethyl]-5-oxo-pyrrolidin-3-yl]ethyl]4-methylbenzenesulfonate 2.3
  • Figure US20180072744A1-20180315-C00058
  • To a mixture of 20.0 g of (R)-4-[(S)-1-Hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.2, 21.67 g p-toluenesulfonyl chloride and 0.92 g N,N-dimethylpyridin-4-amine was added 42 mL pyridine and 42 mL dichloromethane (DCM). The resulting mixture was stirred at 34° C. for 18 h under argon atmosphere. The reaction mixture was diluted with isopropyl acetate and washed with water and 2M hydrochloric acid. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was taken up in isopropyl acetate and n-heptane. The precipitate was filtered off, washed with n-heptane/isopropyl acetate and dried to yield 19.83 g of [(1S)-1-[(3R)-1-[(1S)-1-(4-methoxyphenyl)ethyl]-5-oxo-pyrrolidin-3-yl]ethyl] 4-methylbenzenesulfonate 2.3 as solid.
  • Analysis: HPLC-MS: Rt=0.680 min (method J), M+H=418
  • 4.1.2. Synthesis of Boronic Acids, Boronic Esters, BF3 Borates and Stannanes with Formula 4 X.1.2.1. Synthesis of R1-Hal 3 Synthesis of 4-Bromo-1-tert-butyl-pyrazole 3.1 for Examples 2, 18,
  • Step 1: Synthesis of 1-tert-Butyl-pyrazole
  • Figure US20180072744A1-20180315-C00059
  • To a stirred mixture of 34.48 g of 1,1,3,3-tetramethoxy-propane and 26.20 g tert.-butylhydrazine hydrochloride in 230 mL ethanol was added 40.0 mL conc. hydrochloric acid dropwise below 50° C., then the mixture was stirred under reflux for 2 h. The reaction mixture was diluted with water. The solvent was almost removed by destillation and the aqueous residue extracted with diethylether. The combined aqueous phases were basified with 10N sodium hydroxide solution and extracted with diethylether. The combined organic phases were washed with saturated brine, dried over sodium sulfate, filtered and concentrated in vacuo to yield 21.90 g of 1-tert-butyl-pyrazole as oil.
  • Analysis: HPLC-MS: Rt=0.412 min (method A), M+H=125
  • Step 2: Synthesis of 4-Bromo-1-tert-butyl-pyrazole
  • Figure US20180072744A1-20180315-C00060
  • To a mixture of 21.9 g of 1-tert-butyl-pyrazole in 150 mL DCM was added 31.5 g N-bromosuccinimide in portions between 0 and 10° C. The resulting mixture was stirred for 30 min. The reaction mixture was allowed to reach ambient temperature. The precipitate was filtered off and washed with DCM. The combined organic extracts were washed with water and saturated brine, dried over magnesium sulfate, filtered and concentrated in vacuo to yield 34.0 g of 4-bromo-1-tert-butyl-pyrazole as oil.
  • Analysis: HPLC-MS: Rt=1.35 min (method B), M+H=203/205
  • Synthesis of trans 4-[4-(4-Bromo-pyrazol-1-yl)-cyclohexyl]-1-methyl-piperazin-2-one 3.2 for Example 12
  • The starting material 1-spiro[7-azoniabicyclo[2,2,1]heptane-7,4′-[1′-methyl-2′-oxo-4′-piperazinium]methane-sulphonate] was obtained as described in WO2011092128.
  • Figure US20180072744A1-20180315-C00061
  • To a solution of 506 mg of 4-bromopyrazole in 7.5 mL dimethylacetamide (DMA) was added 91 mg sodium hydride (NaH). The resulting mixture was stirred at room temperature for 10 min, before 1.0 g of 1-spiro[7-azoniabicyclo[2,2,1]heptane-7,4′-[1′-methyl-2′-oxo-4′-piperazinium]methane-sulphonate] was added and the mixture was stirred at 100° C. for 40 min. Additional 70 mg NaH were added and the reaction mixture was stirred at 120° C. for 40 min. The solvent was removed by destillation and the residue taken up in MeOH and purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 850 mg of trans 4-[4-(4-bromo-pyrazol-1-yl)-cyclohexyl]-1-methyl-piperazin-2-one as solid.
  • Analysis: HPLC-MS: Rt=0.45 min (method C), M+H=341/343
  • Synthesis of 5-Bromo-2-(difluoromethyl)pyridine 3.3 for Example 51
  • Figure US20180072744A1-20180315-C00062
  • A solution of 1 g of 5-bromopyridine-2-carbaldehyde in 50 mL DCM was cooled to −70° C., then 1.55 mL diethylaminosulfurtrifluoride were added dropwise over 20 minutes. The suspension was stirred for 30 minutes at room temperature, then 10 mL water were added at 0° C. followed by slow addition of 20 mL saturated NaHCO3 (gas formation). The phases were separated and 2 mL of 4M HCl in dioxane were added to the organic phase which was concentrated in vacuo to provide 1.06 g product as yellow solid.
  • Analysis: HPLC-MS: Rt=0.72 min (method D), M+H=208/210.
  • Synthesis of 7-Bromo-3-methyl-1,2,4,5-tetrahydro-3-benzazepine 3.4 for Example 55
  • 7-Bromo-3-methyl-1,2,4,5-tetrahydro-3-benzazepine can be obtained as described in Shah, Unmesh; Lankin, Claire M.; Boyle, Craig D.; Chackalamannil, Samuel; Greenlee, William J.; Neustadt, Bernard R.; Cohen-Williams, Mary E.; Higgins, Guy A.; Ng, Kwokei; Varty, Geoffrey B.; Zhang, Hongtao; Lachowicz, Jean E. Bioorganic and Medicinal Chemistry Letters, 2008, 18, 4204-4209.
  • Figure US20180072744A1-20180315-C00063
  • Synthesis of 6-Bromo-N,N,1-trimethyl-indole-2-carboxamide 3.5 for Example 56
  • Figure US20180072744A1-20180315-C00064
  • A mixture of 0.68 g of 6-bromo-1-methyl-indole-2-carboxylic acid, 1.1 g of 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluroniumhexafluorophosphat (HATU) and 0.55 mL triethylamine in 2 mL N-methyl-2-pyrrolidinon (NMP) and 3 mL tetrahydrofuran (THF) was stirred at room temperature for 2 h followed by addition of 4.0 mL 2M dimetylamine solution in THF. The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate solution. The combined organic phases were concentrated in vacuo. The crude material was purified by flash chromatography (DCM→DCM: methanol 90:10) to yield 0.66 g of 6-bromo-N,N,1-trimethyl-indole-2-carboxamide 3.5 as oil.
  • Analysis: HPLC-MS: Rt=0.85 min (method E), M+H=281/283
  • Synthesis of 4-Bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6 for Example 70
  • Step 1: Synthesis of [(3R)-Tetrahydrofuran-3-yl] 4-methylbenzenesulfonate
  • Figure US20180072744A1-20180315-C00065
  • To a solution of 25.43 g (R)-tetrahydro-furan-3-ol in 60 mL pyridine and 250 mL DCM was added 73.0 g of 4-methyl-benzenesulfonyl chloride followed by 1.0 g N,N-dimethylpyridin-4-amin. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM and washed with 2M hydrochloric acid and water. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (DCM→DCM: methanol 95:5) to yield 59.46 g [(3R)-tetrahydrofuran-3-yl] 4-methylbenzenesulfonate as oil.
  • Analysis: MS: M+H=243
  • Step 2: Synthesis of 4-Bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6
  • Figure US20180072744A1-20180315-C00066
  • A mixture of 650 mg [(3R)-tetrahydrofuran-3-yl] 4-methylbenzenesulfonate, 400 mg 4-bromo-1H-pyrazole and 1.40 g cesium carbonate in 10 mL N,N-dimethylformamide (DMF) was stirred at 65° C. for 6 h. Additional 20 mg 4-bromo-1H-pyrazole were added and the reaction mixture was stirred at 65° C. overnight. The reaction mixture was diluted with ethyl acetate and washed with brine. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (cyclohexane/ethyl acetate 9:1→1:1) to yield 476 mg of 4-bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6 as solid.
  • Analysis: MS: M+H=217/219
  • Synthesis of 4-Bromo-1-[(3R)-tetrahydrofuran-3-yl]pyrazole 3.7 for Example 74
  • Figure US20180072744A1-20180315-C00067
  • This intermediate was prepared from (S)-tetrahydro-furan-3-ol in two steps according to the preparation of 4-bromo-1-[(3S)-tetrahydrofuran-3-yl]pyrazole 3.6.
  • Analysis: MS: M+H=217/219
  • Synthesis of 4-Bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8 for Example 79
  • Step 1: Synthesis of 5-Methyl-1-tetrahydropyran-4-yl-pyrazole
  • Figure US20180072744A1-20180315-C00068
  • To a stirred mixture of 2.26 g of 1-tetrahydropyran-4-ylpyrazole in 20 mL THF was added 11.14 mL of 1.6M N-butyllithium solution in hexane dropwise at −50° C. under argon atmosphere. The mixture was stirred between −20° to −15° C. for 1.5 h, before 1.11 mL methyl iodide were added dropwise.The resulting mixture was stirred between −20° to −15° C. for 1.5 h. 10 mL water were added dropwise and the mixture was allowed to reach ambient temperature. The reaction mixture was diluted with water, followed by extraction with ethyl acetate. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by rpHPLC (basic) to yield after lyophilisation 1.51 g of 5-methyl-1-tetrahydropyran-4-yl-pyrazole as solid.
  • Analysis: HPLC-MS: Rt=0.61 min (method F), M+H=167
  • Step 2: Synthesis of 4-Bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8
  • Figure US20180072744A1-20180315-C00069
  • To a mixture of 1.0 g of 5-methyl-1-tetrahydropyran-4-yl-pyrazole in 20 mL THF and 20 mL ethyl acetate was added 1.09 g bromosuccinimide in portions between 10 and 20° C. The reaction mixture was stirred at room temperature for 30 min and then quenched with saturated aqueous potassium carbonate solution. The solvent was evaporated and the residue was purified by rpHPLC (basic) to yield after lyophilisation 1.20 g of 4-bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8 as solid.
  • Analysis: HPLC-MS: Rt=0.65 min (method F), M+H=245/247
  • Synthesis of 4-(4-Bromophenyl)-1-methyl-piperidine 3.9 for Example 80
  • Figure US20180072744A1-20180315-C00070
  • To a mixture of 100 mg of 4-(4-bromophenyl)-piperidine hydrochlorid and 100 mg sodium acetate in 3 mL DCM and 0.5 mL methanol was added 50 μL formaldehyde (aqueous 37%). The resulting mixture was stirred at room temperature for 10 min, before 155 mg sodium triacetoxyborohydride were added. The reaction mixture was stirred for 2 h and quenched with saturated aqueous sodium bicarbonate solution, followed by extraction with DCM. The combined organic phases were concentrated in vacuo to yield 88 mg of 4-(4-bromophenyl)-1-methyl-piperidine 3.9 as solid.
  • Analysis: HPLC-MS: Rt=0.38 min (method J), M+H=254/256
  • Synthesis of 4-(4-Bromo-3-trifluoromethyl-phenyl)-1-methyl-piperidine 3.10 for Example 32
  • Figure US20180072744A1-20180315-C00071
  • To a solution of 640 mg of 4-(4-bromo-3-trifluoromethyl-phenyl)-piperidine in 16.3 mL DCM and 2.9 mL methanol was added 6.40 mL formaldehyde (aqueous 37%). The resulting mixture was stirred at ambient temperature for 1 h, before cooled to 0° C. and 1.02 g sodium triacetoxyborohydride were added in portions. Then the reaction was allowed to warm to ambient temperature and was stirred for 1 h. The reaction mixture was quenched with saturated aqueous potassium carbonate solution, followed by extraction with DCM. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 650 mg of 4-(4-bromo-3-trifluoromethyl-phenyl)-1-methyl-piperidine 3.10 as oil.
  • Analysis: HPLC-MS: Rt=1.37 min (method K), M+H=323/325
  • Synthesis of 4-(4-Bromo-2-trifluoromethyl-phenyl)-morpholine 3.11 for Example 38
  • Figure US20180072744A1-20180315-C00072
  • A mixture of 1.0 g of 4-bromo-2-(trifluoromethyl)aniline, 786 μL bis(2-bromoethyl)ether and 1.45 mL diisopropylamine in 3 mL DMA was stirred in a sealed tube at 140° C. for 2 days. The reaction mixture was poured into water and extracted with TBME. The combined organic extracts were washed with saturated aqueous sodium bicarbonate solution, dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 833 mg (84% per HPLC) of 4-(4-bromo-2-trifluoromethyl-phenyl)-morpholine 3.11 as oil.
  • Analysis: HPLC-MS: Rt=2.37 min (method K), M+H=310/312
  • Synthesis of 4-Bromo-1-(4,4-difluorocyclohexyl)pyrazole 3.12 for Example 10
  • Step 1: Synthesis of N′-(4,4-difluoro-cyclohexylidene)-hydrazinecarboxylic acid tert-butyl ester
  • Figure US20180072744A1-20180315-C00073
  • A solution of 0.99 g hydrazinecarboxylic acid tert-butyl ester in 5 mL methanol was added dropwise to a solution of 1.0 g of 4,4-difluoro-cyclohexanone in 5 mL methanol. The resulting mixture was stirred at room temperature for 1 h. The solvent was removed by destillation to yield 1.76 g N′-(4,4-difluoro-cyclohexylidene)-hydrazinecarboxylic acid tert-butyl ester as solid.
  • Analysis: MS: M+H=249
  • Step 2: Synthesis of N′-(4,4-difluoro-cyclohexyl)-hydrazinecarboxylic acid tert-butyl ester
  • Figure US20180072744A1-20180315-C00074
  • A mixture of 8.0 g N′-(4,4-difluoro-cyclohexylidene)-hydrazinecarboxylic acid tert-butyl ester and 800 mg palladium on carbon in 48 mL methanol was hydrogenated at 40° C. for 16 h at 10 bar. The catalyst was removed by filtration and the solvent was evaporated in vacuo to yield 7.82 g of N′-(4,4-difluoro-cyclohexyl)-hydrazinecarboxylic acid tert-butyl ester.
  • Analysis: MS: M−H=249
  • Step 3: Synthesis of (4,4-Difluoro-cyclohexyl)-hydrazine hydrochlorid
  • Figure US20180072744A1-20180315-C00075
  • To a mixture of 5.0 g of N′-(4,4-difluoro-cyclohexyl)-hydrazinecarboxylic acid tert-butyl ester in 20 mL DCM was added 40 mL 6M hydrochloric acid in isopropanol and the resulting mixture was stirred at room temperature for 12 h. The solvent was evaporated and the residue triturated with toluene. The precipitate was filtered off and dried to yield 3.72 g of (4,4-difluoro-cyclohexyl)-hydrazine hydrochlorid as solid.
  • Analysis: ESI-MS: M+H=151
  • Step 4: Synthesis of 1-(4,4-difluorocyclohexyl)pyrazole
  • Figure US20180072744A1-20180315-C00076
  • To a mixture of 2.04 g of (4,4-difluoro-cyclohexyl)-hydrazine hydrochlorid in 15 mL ethanol was added 3.50 mL conc. hydrochloric acid followed by 1.80 g 1,1,3,3-tetramethoxypropane, then the mixture was refluxed for 1h. The reaction mixture was diluted with water, ethanol was removed by destillation. The residue was alkalized with aqueous sodium hydroxid solution (30%) and extracted with diethylether. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo to yield 2.02 g of 1-(4,4-difluorocyclohexyl)pyrazole as oil.
  • Analysis: HPLC-MS: Rt=0.46 min (method C), M+H=187
  • Step 5: Synthesis of 4-Bromo-1-(4,4-difluorocyclohexyl)pyrazole
  • Figure US20180072744A1-20180315-C00077
  • To a solution of 2.0 g of 1-(4,4-difluorocyclohexyl)pyrazole in 5 mL DCM was added 0.55 mL bromine at 0° C. and the mixture was stirred at room temperature for 15 min. The solvent was removed by destillation and the residue taken up in DCM and washed with semi saturated brine. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo to yield 2.81 g of 4-bromo-1-(4,4-difluorocyclohexyl)pyrazole 3.12 as solid.
  • Analysis: HPLC-MS: Rt=0.60 min (method C), M+H=265/267
  • Synthesis of 4-Bromo-1-(difluoromethyl) imidazole 3.13 for Example 83
  • Figure US20180072744A1-20180315-C00078
  • Into a mixture of 10.0 g of 4-bromo-1H-imidazole in 60 mL DMF was passed 30 g chlorodifluoromethane under dry ice cooling, then 15.0 g potassium carbonate were added and the reaction mixture was heated to 110° C. overnight in a sealed tube (19 bar). Additional 30 g chlorodifluoromethane and 5 g potassium carbonate were added and the reaction mixture was heated to 110° C. overnight (9 bar). The reaction mixture was allowed to reach ambient temperature, then diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with saturated brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by rpHPLC (SunfireC18, acetonitrile/water trifluoroacetic acid) to yield 3.07 g 4-bromo-1-(difluoromethyl) imidazole.
  • Analysis: HPLC-MS: Rt=0.371 min (method J), M+H=197/199
  • The following bromides were commercially available:
      • 3-Bromoimidazo[1,2-a]pyridine-6-carbonitrile 3.14 for Example 43,
      • 4-Bromo-1-(3,3,3-trifluoro-propyl)pyrazole 3.15 for Example 15, 76
      • 3-Bromo-1H-indazole-5-carbonitrile 3.16 for Example 16
      • 2-Bromo-5-fluoro-pyridine 3.17 for Example 41
      • 4-(4-Bromophenyl)-1-methyl-piperidine 3.18 for Example 42
      • 2-(4-Bromo-2-methyl-phenyl)-1,2-thiazolidine 1,1-dioxide 3.19 for Example 71
      • 7-Bromo-2-methyl-3,4-dihydro-1H-isoquinoline 3.20 for Example 75
      • 4-Bromo-1-isopropoxy-2-methoxy-benzene 3.22 for Example 81
      • 5-Bromo-1-methyl-indazol-3-amine 3.23 for Example 36,
      • 2-(4-Bromophenyl)-1,2-thiazolidine 1,1-dioxide 3.24 for Example 33
      • 4-Bromo-1-(difluoromethyl)pyrazole 3.25 for Example 40
      • 4-Bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole 3.26 for Example 3
      • 1-[(4-Bromophenyl)methyl]-2-methyl-1H-imidazole 3.27 for Example 17
    4.1.3. Synthesis of Compounds of Formula 4 (R1-X) (Scheme 1 and 2) Synthesis of Tributyl-[2-(difluoromethyl)thiazol-4-yl]stannane 4.1 for Example 35
  • Figure US20180072744A1-20180315-C00079
  • To a mixture of 500 mg of 4-tributylstannylthiazole-2-carbaldehyde in 5 mL DCM was slowly added 1.01 mL 2.7M [bis(2-methoxyethyl)amino]sulfur trifluoride solution (in toluene) at 0° C., then the mixture was allowed to warm to ambient temperature and stirred for 2 h. The reaction mixture was diluted with DCM and washed with water. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 440 mg (82% per HPLC) tributyl-[2-(difluoromethyl)thiazol-4-yl]stannane 4.1 as oil.
  • Analysis: HPLC-MS: Rt=2.72 min (method M), M+H=425
  • The following stannane was commercially available:
      • Tributyl(thiazol-4-yl)stannane 4.2 for Example 29
    Synthesis of 3-Methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,4,5-tetrahydro-3-benzazepine 4.3 for Example 55
  • Figure US20180072744A1-20180315-C00080
  • 100 mg of 7-bromo-3-methyl-1,2,4,5-tetrahydro-3-benzazepine, 127 mg bis-(pinacolato)-diboron, 20 mg 1,1′-bis(diphenylphospino)ferrocenedichloropalladium(II) and 123 mg potassium acetate were suspended in 2 mL dioxane and the mixture stirred at 100° C. for 1.25 h. The mixture was diluted after cooling with dioxane, filtered through Celite, washed with dioxane and the solvent was evaporated in vacuo to yield 220 mg (92%, content 50%) 3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,4,5-tetrahydro-3-benzazepine 4.3 as solid, which was used in the next step without further purification.
  • Analysis: HPLC-MS: Rt=0.45 min (method N), M+H=288
  • The following boronic esters were synthesized in analogy and were used without further purification:
      • 1-Methyl-4-{4-[4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)-pyrazol-1-yl]-cyclohexyl}-piperazin-2-one 4.4 starting from 3.2 (for Example 12). Reaction conditions: 4 h, 100° C. Yield: 81% (content 36%). Analysis: HPLC-MS: Rt=0.41 min (method 0), M+H=389
      • 2-Difluoromethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-pyridine 4.5 starting from 3.3 (for Example 51). Reaction conditions: 45 min, 100° C. Yield: 82% (content 40%). Analysis: HPLC: Rt=0.25 min (method N); MS: M+H=256
      • N,N,1-Trimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indole-2-carboxamide 4.6 starting from 3.5 (for Example 56). Reaction conditions: 3 h, 100° C. Yield: 84% (content 50%). Analysis: HPLC-MS: Rt=0.69 min (method N), M+H=329
      • 1-[(3S)-Tetrahydrofuran-3-yl]-4-(4,4,5, 5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.7 starting from 3.6 (for Example 70). Reaction conditions: 3 h, 100° C. Yield: 59% (content 50%). Analysis: HPLC-MS: Rt=0.49 min (method N), M+H=265
      • 2-[2-Methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1,2-thiazolidine 1,1-dioxide 4.8 starting from 3.19 (for Example 71). Reaction conditions: 3 h, 100° C. Yield: 86% (content 50%). Analysis: HPLC-MS: Rt=0.64 min (method N), M+H=338
      • 1-[(3R)-Tetrahydrofuran-3-yl]-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.9 starting from 3.7 (for Example 74). Reaction conditions: 5 h, 100° C. Yield: 43% (content 38%). Analysis: HPLC-MS: Rt=0.54 min (method J), M+H=265
      • 2-Methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydro-1H-isoquinoline 4.10 starting from 3.20 (for Example 75). Reaction conditions: 2 h, 100° C. Yield: 99% (content 43%). Analysis: HPLC-MS: Rt=0.41 min (method J), M+H=274
      • 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-1-(3,3,3-trifluoropropyl)pyrazole 4.11 starting from 3.15 (for Example 76). Reaction conditions: 4 h, 100° C. Yield: 29% (content 25%). Analysis: HPLC-MS: Rt=0.61 min (method J), M+H=291
      • 1-Methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]piperidine 4.12 starting from 3.9 (for Example 80). Reaction conditions: 4 h, 100° C. Yield: 84% (content 50%). Analysis: HPLC-MS: Rt=0.47 min (method J), M+H=302
      • 2-(4-lsopropoxy-3-methoxy-phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane 4.13 starting from 3.22 (for Example 81). Reaction conditions: 3 h, 100° C. Yield: 87% (content 40%). Analysis: HPLC-MS: Rt=0.75 min (method J), M+H=293
      • 2-[4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-1,2-thiazolidine 1,1-dioxide 4.14 starting from 3.24 (for Example 33). Reaction conditions: 2.5 h, 80° C., in DMF. Yield: 68% (content 95%). Analysis: HPLC-MS: Rt=2.04 min (method K), M+H=324
      • The following examples were synthesized in analogy to the described example but using bis(triphenylphosphine)palladium(II) chloride (0.05 eq.) instead of 1,1′-bis(diphenylphospino)ferrocenedichloropalladium(II) as catalyst (see description above):
      • 1-Methyl-5-(4,4,5, 5-tetramethyl-1,3,2-dioxaborolan-2-yl) indazol-3-amine 4.15 starting from 3.23 (for Example 36). Reaction conditions: 1 h, 95° C. Yield: 89% (content 85%). Analysis: HPLC-MS: Rt=1.76 min (method K), M+H=274
      • 1-Methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(trifluoromethyl)phenyl]piperazine 4.16 starting from 3.10 (for Example 32). Reaction conditions: 1.5 h, 95° C. Yield: 61% (content 95%). Analysis: HPLC-MS: Rt=1.66 min (method K), M+H=371
      • 1-(Difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.17 starting from 3.25 (for Example 40). Reaction conditions: 2.5 h, 95° C. Yield: 70% (content 95%). Analysis: HPLC-MS: Rt=1.97 min (method K), M+H=245
      • 4-[4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(trifluoromethyl)phenyl]morpholine 4.18 starting from 3.11 (for Example 38). Reaction conditions: 1 h, 95° C. Yield: 99% (content 78%). Analysis: HPLC-MS: Rt=2.67 min (method K), M+H=358
    Synthesis of 1-tert-Butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.19 for Examples 2,18,
  • Figure US20180072744A1-20180315-C00081
  • To a stirred mixture of 50 g of 4-bromo-1-tert-butyl-pyrazole 3.1 in 230 mL THF was added dropwise 100 mL 2.5M N-butyllithium solution in hexane under argon atmosphere below -60° C., then the mixture was stirred at this temperature for 5 min, before 52 mL 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane were added dropwise below −60° C. The reaction mixture was allowed to reach ambient temperature. The mixture was cooled with an ice bath and diluted with aqueous phosphate buffer solution and water and neutralized with 2M aqueous hydrochloric acid. The organic solvent was removed by destillation and the residue was extracted with DCM. The combined organic extracts were washed with saturated brine, dried over sodium sulfate, filtered and concentrated in vacuo to yield 44.26 g of 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole as solid.
  • Analysis: HPLC-MS: Rt=0.904 min (method F), M+H=251
  • Synthesis of 5-Methyl-1-tetrahydropyran-4-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.20 for Example 79
  • Figure US20180072744A1-20180315-C00082
  • This intermediate was prepared from 4-bromo-5-methyl-1-tetrahydropyran-4-yl-pyrazole 3.8 according to the preparation of 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.19.
  • Yield: 94% of 4.20
  • Analysis: HPLC-MS: Rt=0.58 min (method J), M+H=293
  • Synthesis of 1-Methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine 4.21 for Example 49
  • Figure US20180072744A1-20180315-C00083
  • A mixture of 250 mg 4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-pyrazol-1-yl]piperidine-1-carboxylic acid tert-butyl ester 4.51 and 0.5 mL trifluoroacetic acid in 2 mL DCM was stirred at room temperature for 1 h. The solvent was removed by destillation and the residue taken up in 10 mL DCM, followed by addition of 494 μL formaldehyde (aqueous 37%). The reaction mixture was stirred at room temperature for 1 h, before 421 mg sodium triacetoxyborhydride were added. The resulting mixture was stirred at room temperature for 30 min, diluted with saturated aqueous sodium bicarbonate solution and extracted with DCM. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo to yield 99 mg of 1-methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine 4.21 as solid.
  • Analysis: HPLC-MS: Rt=1.25 min (method K), M+H=292
  • The Following Boronic Acids, Boronic Esters and BF3 Borates were Commercially Available:
      • (3,4-Dimethoxyphenyl)boronic acid 4.22 for Example 1
      • 2-Cyclopropyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine 4.23 for Example 50
      • 2-Methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol 4.24 for Examples 8, 73
      • (3,4,5-Trimethoxyphenyl)boronic acid 4.25 for Example 52
      • tert-Butyl-5-methoxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indole-1-carboxylate 4.26 for Example 53
      • 1-(2-Methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.27 for Example 57
      • 1-Ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.28 for Example 58
      • [6-(Trifluoromethyl)-3-pyridyl]boronic acid 4.29 for Examples 4, 61
      • (1-Methylindazol-5-yl)boronic acid 4.30 for Example 62
      • 1-Methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.31 for Example 63
      • 1-Tetrahydropyran-4-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.32 for Examples 47, 64
      • 1-Isopropyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.33 for Example 65
      • 4-[5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-2-pyridyl]morpholine 4.34 for Example 66
      • 1-Methyl-4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]piperazine 4.35 for Examples 26, 67
      • 1,3-Dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.36 for Example 68
      • 1,5-Dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.37 for Example 69
      • (1-Methylindazol-6-yl)boronic acid 4.38 for Example 72
      • 7-Chloro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole 4.39 for Examples 48, 77
      • 1-Cyclopropyl-4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyrazole 4.40 for Example 78
      • tert-Butyl 4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine-1-carboxylate 4.41 for Examples 9, 45, 82
      • Potassium 5-methyl-2-thiophenetrifluoroborate 4.42 for Example 54
      • Potassium 6-methoxy-3-pyridyltrifluoroborate 4.43 for Example 59
      • Potassium 4-(trifluoromethyl)phenyltrifluoroborate 4.44 for Example 60
      • 5-Fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine 4.45
      • (4-Morpholinophenyl)boronic acid 4.46 for Example 22
      • (1-Methylindazol-5-yl)boronic acid 4.47 for Examples 28
      • 2-lsopropoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine 4.48 for Example 27
      • 1-Methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole 4.49 for Example 14
      • 1-Cyclohexyl-4-(4,4, 5, 5-tetramethyl-1,3,2-dioxoborolan-2-yl)-1H-pyrazole 4.50 for Example 7
      • 4-[4-(4,4,5, 5-tetramethyl-1,3,2-dioxaborolan-2-yl)-pyrazol-1-yl]piperidine-1-carboxylic acid tert-butyl ester 4.51 for Example 49
    4.1.4. Synthesis of Heterocyclic 5 and 10 from Scheme 1 and 2 Synthesis of 4,6-Dichloro-2-methyl-pyrazolo[4,3-c]pyridine (5.1) for Examples 1-3, 7-13, 17, 50-83
  • Figure US20180072744A1-20180315-C00084
  • To a mixture of 9.5 g trimethyloxonium tetrafluoroborate in 300 mL DCM was added 10.0 g of 4,6-dichloro-3aH-pyrazolo[4,3-c]pyridine (commercially available from Sphinx Scientific Laboratory Corporation) under argon atmosphere. The reaction mixture was stirred at room temperature overnight. Additional 2.7 g trimethyl-oxonium tetrafluoroborate and 2.0 mL ethyldiisopropylamine (DIPEA) were added and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water, filtered and the organic phase was concentrated in vacuo. The crude material was taken up in semi saturated aqueous sodium bicarbonate solution. The precipitate was filtered off, washed with water and dried to yield 8.2 g of 4,6-dichloro-2-methyl-pyrazolo[4,3-c]pyridine 5.1 as solid.
  • Analysis: HPLC-MS: Rt=0.45 min (method C), M+H=202/204
  • Synthesis of 6-Bromo-2-methyl-indazol-4-ol 10.2 for Examples 14-16, 18, 22, 26-29, 32, 33, 35, 36, 38, 40-43, 45, 47-49
  • Step 1: Synthesis of 6-Bromo-4-methoxy-2-methyl-indazole
  • Figure US20180072744A1-20180315-C00085
  • To a solution of 5.0 g of 6-bromo-4-methoxy-1H-indazole in 50 mL 1,4-dioxane was added 4.23 g trimethyloxonium tetrafluoroborate at room temperature. The reaction mixture was stirred at 40° C. for 3 h and left standing overnight. The reaction mixture was poured into water. The precipitate was filtered off, washed with water and dried to yield 4.26 g of 6-bromo-4-methoxy-2-methyl-indazole as solid.
  • Analysis: HPLC-MS: Rt=1.78 min (method K), M+H=241/243
  • Step 2: Synthesis of 6-Bromo-2-methyl-indazol-4-ol 10.2
  • Figure US20180072744A1-20180315-C00086
  • To a suspension of 4.26 g of 6-bromo-4-methoxy-2-methyl-indazole in 42.6 mL DCM was added 53.06 mL boron tribromide solution (1M in DCM) at 0° C. The reaction mixture was allowed to reach ambient temperature and stirred for 3 days. Additional 10 mL boron tribromide solution (1M in DCM) was added and the reaction mixture was stirred at room temperature for 8 h. The mixture was poured into water. The precipitate was filtered off and triturated with acetonitrile to yield 2.8 g of 6-bromo-2-methyl-indazol-4-ol 10.2 as solid. The acetonitrile filtrate was combined with the DCM layer and concentrated in vacuo. The residue was triturated with acetonitrile to yield 1.08 g of 6-bromo-2-methyl-indazol-4-ol 10.2 as solid. The two solids were combined to yield 3.88g of 6-bromo-2-methyl-indazol-4-ol 10.2 as solid.
  • Analysis: HPLC-MS: Rt=1.49 min (method K), M+H=227/229
  • 4.2. Synthesis of Intermediates 6, 7, 8 and 11 from Scheme 1 and 2 Synthesis of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one (7.1) for Examples 1-3, 7-13, 17, 50-83
  • Step 1: Synthesis of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • Figure US20180072744A1-20180315-C00087
  • To a mixture of 20.0 g of 4,6-dichloro-2-methyl-pyrazolo[4,3-c]pyridine 5.1 and 30.1g (R)-4-[(R)-1-hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]pyrrolidin-2-one 2.1 in 1 L dioxane was added 4.51 g NaH in mineral oil (60%). The resulting mixture was stirred at 100° C. for 15 h. The solvent was removed by destillation to ⅓. The residue was taken up in DCM and washed with saturated ammonium chloride solution and water. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo to yield 44.8 g (88% per HPLC) of (R)-4-[(R)-1-(6-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yloxy)-ethyl]-1-[(S)-1-(4-methoxy-phenyl)-ethyl]-pyrrolidin-2-one as solid.
  • Analysis: HPLC-MS: Rt=0.65 min (method J), M+H=429
  • Alternatively, 6.1 can be synthesized as following:
  • A solution of 20.0 g of 4,6-dichloro-2-methyl-pyrazolo[4,3-c]pyridine 5.1 and 25.4 g (R)-4-[(R)-1-hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.1 in 250 mL dioxane was added to a slurry of 9.6 g NaH in mineral oil (60%) in 50 mL of dioxane at 20° C. The resulting mixture was stirred at 40° C. for 5.5 h. The mixture was cooled to ambient temperature and quenched by the slow addition of 36 mL of 4M HCl in dioxane. The reaction mixture was diluted with 200 mL of isopropyl acetate and water (100 mL). The layers were separated, and the aqueous portion was extracted with twice with 100 mL of isopropyl acetate. The organic portions were assayed to show 40.78g of of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one in a solution mass of 796.3g for a 99% yield. Purification of 6.1 can be conducted by concentration of a crude isopropyl acetate solution of 50 g 6.1 in vacuo to 200 mL wherein solids crystallized. 500 mL of heptane was slowly charged to the slurry at 20° C. and the mixture was agitated for 2 h. The solids were collected by filtration, washed with heptane, and dried at 30° C. 46.9 g of 6.1 was isolated as a beige solid in 92% recovery.
  • Step 2: Synthesis of (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1
  • Figure US20180072744A1-20180315-C00088
  • A mixture of 1.0 g of (R)-4-[(R)-1-(6-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yloxy)-ethyl]-1-[(S)-1-(4-methoxy-phenyl)-ethyl]-pyrrolidin-2-one 6.1 and 1 mL anisole in 5 mL trifluoroacetic acid (TFA) was stirred at 80° C. for 17 h. The solvent was removed by destillation. The residue was taken up in DCM and washed with saturated aqueous sodium bicarbonate solution. The combined organic phases were concentrated in vacuo and the residue was triturated with diethyl ether. The precipitate was filtered off and dried to yield 0.37 g of (R)-4-[(R)-1-(6-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yloxy)-ethyl]pyrrolidin-2-one 7.1 as solid.
  • Analysis: HPLC-MS: Rt=0.47 min (method J), M+H=295
  • Synthesis of (4R)-4-[(1R)-1-(6-Bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one (7.5) for Examples 14-16, 18, 22, 26-29, 32, 33, 35, 36, 38, 40-43, 45, 47-49
  • Step 1: Synthesis of (4R)-4-[(1R)-1-(6-Bromo-2-methyl-indazol-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • Figure US20180072744A1-20180315-C00089
  • To a mixture of 1.46 g 6-bromo-2-methyl-indazol-4-ol 10.2, 1.86 g (R)-4-[(S)-1-hydroxyethyl]-1-[(S)-1-(4-methoxyphenyl)-ethyl]-pyrrolidin-2-one 2.2 and 5.06 g triphenylphosphine in 36.5 mL THF was added 4.44 g di-tert-butyl azodicarboxylate (DBAD) over 30 min. The resulting mixture was stirred at room temperature for 18 h.The solvent was evaporated and the residue triturated with TBME. The precipitate was filtered off and washed with TBME. The filtrate was concentrated in vacuo and the resulting residue was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 2.36 g (54% per HPLC) of (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 6.5 as solid.
  • Analysis: HPLC-MS: Rt=2.00 min (method K), M+H=472/474
  • Step 2: Synthesis of (4R)-4-[(1R)-1-(6-Bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one 7.5
  • Figure US20180072744A1-20180315-C00090
  • A mixture of 583 mg (63% per HPLC) of (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl) oxyethyl]-1-[(1S)-1-(4-methoxyphenyl) ethyl] pyrrolidin-2-one 6.5 in 10 mL TFA was stirred at 70° C. for 18 h. The solvent was removed by destillation. The residue was taken up in DCM, poured into saturated aqueous sodium bicarbonate solution and extracted. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 321 mg (84% per HPLC) of (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl) oxyethyl]pyrrolidin-2-one 7.5.
  • Analysis: HPLC-MS: Rt=1.63 min (method K), M+H=338/340
  • 4.1.6. Synthesis of Boronic Acids and Boronic Esters 11 from Scheme 1 and 2 Synthesis of [2-Methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]boronic acid 11.1 for Examples 3, 10, 13, 17, 83
  • Figure US20180072744A1-20180315-C00091
  • 400 mg of (R)-4-[(R)-1-(6-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yloxy)-ethyl]pyrrolidin-2-one 7.1, 620 mg bis-(pinacolato)-diboron, 122 mg 1,1′-bis(diphenylphospino) ferrocenedichloropalladium(II) and 360 mg potassium acetate were suspended in 4 mL dioxane and the mixture stirred at 100° C. for 1 h. The mixture was diluted with dioxane, filtered through Celite, washed with dioxane and the solvent was evaporated in vacuo to yield 1.09 g (crude) [2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]boronic acid 11.1 as oil, which was used in the next step without further purification.
  • Analysis: HPLC-MS: Rt=0.26 min (method S), M+H=305
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indazol-4-yl]oxyethyl]pyrrolidin-2-one 11.3 for Examples 15, 16, 41, 42, 43
  • Figure US20180072744A1-20180315-C00092
  • 500 mg of (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one 7.5, 563 mg bis-(pinacolato)-diboron, 52 mg bis(triphenylphosphine)palladium(II) chloride and 435 mg potassium acetate were suspended in 5 mL dioxane and the resulting mixture was stirred at 95° C. for 1 h. The reaction mixture was allowed to reach ambient temperature, diluted with water and extracted with DCM. The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (heptane/ethyl acetate/methanol) to yield 505 mg (78% per HPLC) of (4R)-4-[(1R)-1-[2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indazol-4-yl]oxyethyl]pyrrolidin-2-one 11.3.
  • Analysis: HPLC-MS: Rt=1.76 min (method K), M+H=387
  • 4.3 Synthesis of the Patent Examples of Formula 1 Synthesis of (4R)-4-[(1R)-1-[6-(3,4-Dimethoxyphenyl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 1)
  • Figure US20180072744A1-20180315-C00093
  • A mixture of 150 mg (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1, 139 mg (3,4-dimethoxyphenyl)boronic acid 4.22, 17.9 mg bis(triphenylphosphine)palladium(II) chloride and 764 μL 2M aqueous sodium carbonat solution in 1.7 mL DMF was stirred at 90° C. for 20 h. The reaction mixture was allowed to reach ambient temperature and purified by rpHPLC to yield after lyophilisation 42 mg of Example 1.
  • Analysis: HPLC-MS: Rt=3.72 min (method T), M+H=397
  • Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 2)
  • Step 1: Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one
  • Figure US20180072744A1-20180315-C00094
  • To a mixture of 1.0 g (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 6.1, 790 mg 1-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) 4.19 and 170 mg 1,1′-bis(diphenylphospino) ferrocenedichloropalladium(II) (complex with DCM (1:1)) in 6 mL dioxane was added 5.0 mL 2M aqueous sodium carbonat solution. The resulting mixture was stirred in a sealed tube at 140° C. for 1h. The reaction mixture was poured into DCM. The precipitate was filtered off. The filtrate was concentrated in vacuo and the resulting residue was purified by flash chromatography (DCM/methanol=1/0→9/1) to yield 1.0 g (50% per NMR) of (4R)-4-[(1R)-1-[6-(1-tert-butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 8.1 as oil.
  • Analysis: HPLC-MS: Rt=0.63 min (method C), M+H=517
  • Step 2: Synthesis of (4R)-4-[(1R)-1-[6-(1-tert-Butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 2)
  • Figure US20180072744A1-20180315-C00095
  • A mixture of 3.90 g (36% per HPLC) of (4R)-4-[(1R)-1-[6-(1-tert-butylpyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]-1-[(1S)-1-(4-methoxyphenyl)ethyl]pyrrolidin-2-one 8.1 in 25 mL TFA was stirred at 80° C. for 3h. The reaction mixture was purified by rpHPLC (SunfireC18, acetonitrile/water, TFA and XbridgeC18, acetonitrile/water, ammonia) and the desired fractions were lyophilized. The residue was triturated with TBME and a small amount of acetone. The solvent was removed by destillation and the residue was dried to yield 410 mg of Example 2 as solid.
  • Analysis: HPLC-MS: Rt=0.52 min (method C), M+H=383
  • Alternatively, 8.1 can be synthesized as following:
  • Synthesis of Ethyl 3-amino-1-methyl-1H-pyrazole-4-carboxylate
  • Figure US20180072744A1-20180315-C00096
  • A solution of ethyl 2-cyano-3-ethoxyacrylate (5.0g, 30 mmol) in anhydrous THF (10 mL) was charged dropwise to a mixture of sodium ethoxide (4.02g, 59 mmol) and methylhydrazine (1.36 g, 30 mmol) in absolute ethanol (10 mL) at 0° C. under nitrogen. After aging the agitated mixture for 90 min, a solution of anhydrous hydrochloric acid (3.22 M in CPME, 28 mL, 90 mmol) was charged to the reaction dropwise. The reaction was then concentrated in vacuo to a solid, diluted with isopropyl acetate (20 mL) and concentrated to a solid. The crude paste was suspended in hot isopropyl acetate (75 mL) and filtered hot. The filtrate was concentrated in vacuo to approximately 15 mL wherein a solid crystallized upon cooling to ambient temperature. The mixture was diluted by the dropwise addition of heptane (30 mL). The mixture was agitated for 1h at ambient temperature. Ethyl 3-amino-1-methyl-1H-pyrazole-4-carboxylate was isolated by filtration, washed with heptane and dried under vacuum to provide 3.7g as a yellow-orange solid in 74% yield. 1H NMR (500 MHz, DMSO-d6) δ=7.87 (s, 1H), 5.3 (bs, 2H), 4.15 (q, J=7.2 Hz, 2H), 3.60 (s, 3H), 1.23 (t, J=7.2 Hz, 3H).
  • Synthesis of Ethyl 3-bromo-1-methyl-1H-pyrazole-4-carboxylate
  • Figure US20180072744A1-20180315-C00097
  • Isoamyl nitrite (420 mg, 3.6 mmol) was charged dropwise to an agitated mixture of ethyl 3-amino-1-methyl-1H-pyrazole-4-carboxylate (400 mg, 2.4 mmol) and copper (II) bromide (660 mg, 3.0 mmol) in anhydrous acetonitrile (10 mL) at ambient temperature. The reaction was agitated for one hour; at which point, the reaction was diluted with isopropyl acetate (100 mL). The mixture was washed with water (100 mL) and concentrated to an oily residue. Purification by silica gel chromatography with methyl t-butyl ether and hexanes provided ethyl 3-bromo-1-methyl-1H-pyrazole-4-carboxylate (350 mg) as a white solid in 64% yield. 1H NMR (500 MHz, CDCl3) δ=7.83 (s, 1H), 4.30 (q, J=7.1Hz, 2H), 3.90 (s, 3H), 1.35 (t, J=7.2 Hz, 3H).
  • Synthesis of 3-Bromo-1-methyl-1H-pyrazole-4-carboxylic acid
  • Figure US20180072744A1-20180315-C00098
  • An aqueous solution of sodium hydroxide (2 M, 10 mL, 20 mmol) was charged to a mixture of ethyl 3-bromo-1-methyl-1H-pyrazole-4-carboxylate (2.0g, 8.6 mmol) in ethanol (20 mL) at ambient temperature. The reaction was agitated at 50° C. for 1h; at which point, the reaction was cooled to ambient temperature. An aqueous solution of hydrochloric acid (3 M, 6.7 mL, 20 mmol) was charged dropwise to the reaction to induce crystallization. The solids of 3-bromo-1-methyl-1H-pyrazole-4-carboxylic acid were collected by filtration, washed with water followed by heptane, and dried to afford 1.4 g as a white solid in 79% yield. 1H NMR (400 MHz, DMSO-d6) δ=12.59 (s, 1H), 8.27 (s, 1H), 3.85 (s, 3H).
  • Synthesis of 1-(1-(tert-Butyl)-1H-pyrazol-4-yl)ethan-1-one
  • Figure US20180072744A1-20180315-C00099
  • Isopropyl magnesium chloride lithium chloride complex (1.3 M in THF, 28.4 mL, 37 mmol) was charged to a solution of 4-bromo-1-(tert-butyl)-1H-pyrazole (5.0 g, 25 mmol) in anhydrous THF (25 mL) under argon at ambient temperature. Anhydrous dioxane (3.3 g, 37 mmol) was charged to the reaction, and the reaction was agitated at 45° C. for 4 h. The resulting mixture was cooled to ambient temperature and charged to an anhydrous solution of acetic anhydride (7.5 g, 73 mmol) in THF (25 mL) at −20° C. The resulting mixture was warmed to ambient temperature and concentrated to a residue. The mixture was dissolved in methyl t-butyl ether (50 mL) and washed with water (25 mL). The organic portion was concentrated to provide crude 1-(1-(tert-butyl)-1H-pyrazol-4-yl)ethan-1-one as an oil (7.6 g, 36 wt %) and 67% yield. Crystallization in a mixture of methyl t-butyl ether and heptane provided analytically pure material. 1H NMR (500 MHz, CDCl3) δ=7.96 (s, 1H), 7.86 (s, 1H), 2.37 (s, 3H), 1.55 (s, 9H).
  • Synthesis of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methylpyrano[4,3-c]pyrazol-4(2H)-one
  • Figure US20180072744A1-20180315-C00100
  • A mixture of lithium tert-butoxide (0.970 g, 12 mmol), palladium (II) acetate (27 mg, 0.12 mmol), and di-tert-butyl(2,4,6-trisopropylbiphenyl-2-yl)phosphine (77 mg, 18 mmol) in anhydrous and degassed 1,4 dioxane (4 mL) under argon was agitated at ambient temperature for 15 minutes. The mixture was heated to 90° C., and agitated at this temperature for 5 minutes. A solution of 3-bromo-1-methyl-1H-pyrazole-4-carboxylic acid (500 mg, 2.4 mmol) and 1-(1-(tert-Butyl)-1H-pyrazol-4-yl)ethan-1-one (490 mg, 2.9 mmol) in anhydrous and degassed 1,4-dioxane (7 mL) under argon was charged to the catalysts base slurry at 90° C. dropwise over 50 minutes. The reaction was agitated at 90° C. for 30 minutes. The reaction was cooled to ambient temperature and quenched by the addition of trifluoroacetic acid (5 mL). The reaction was concentrated to an oily solid. The mixture was suspended in a mixture of acetonitrile (20 mL) and trifluoroacetic acid (20 mL). The mixture was agitated at 75° C. for 14 h then concentrated to an oily solid. The mixture was suspended in isopropyl acetate (70 mL) and washed twice with water (40 mL). The organic portion was concentrated to a solid. Purification by silica gel chromatography with ethyl acetate and hexanes provided 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methylpyrano[4,3-c]pyrazol-4(2H)-one as an orange solid (650 mg) in 98% yield. 1H NMR (500 MHz, CLCl3) δ=8.06 (s, 1H), 7.94 (s, 1H), 7.84 (s, 1H), 6.68 (s, 1H), 4.04 (s, 3H), 1.60 (s, 9H).
  • Synthesis of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2,5-dihydro-4H-pyrazolo[4,3-c]pyridin-4-one
  • Figure US20180072744A1-20180315-C00101
  • A mixture of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methylpyrano[4,3-c]pyrazol-4(2H)-one (980 mg, 3.6 mmol) and ammonium acetate (1.11 g, 14 mmol) in anhydrous DMSO (4 mL) was agitated at 110° C. for 4 h; at which point, additional ammonium acetate (1.11 g, 3.6 mmol) was charged to the reaction. After agitation for another 4 h at 110° C., the reaction was cooled to ambient temperature and diluted with water (20 mL). The solids of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2,5-dihydro-4H-pyrazolo[4,3-c]pyridin-4-one were isolated by filtration, washed with water, and dried to provide 880 mg for an 90% yield. 1H NMR (500 MHz, DMSO-d6) δ=10.59 (s, 1H), 8.54 (s, 1H), 8.39 (s, 1H), 8.06 (s, 1H), 6.73 (s, 1H), 3.99 (s, 3H), 1.53 (s, 9H).
  • Synthesis of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-4-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridine
  • Figure US20180072744A1-20180315-C00102
  • Phosphorous (V) oxychloride (848 mg, 5.53 mmol) was charged to a mixture of 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2,5-dihydro-4H-pyrazolo[4,3-c]pyridin-4-one (500mg, 1.84 mmol) in anhydrous acetonitrile (5 mL) under argon. The reaction was agitated at 75-80° C. for 3 h; at which point, the reaction was cooled to ambient temperature. The reaction slowly poured into a saturated sodium bicarbonate aqueous solution (45 mL). The mixture was agitated for 20 min and concentrated in vacuo to remove the acetonitrile solvent. The resulting aqueous slurry was diluted with isopropyl acetate (50 mL) and washed with water (2×20 mL). The organic portion was concentrated to an oil which solidified upon standing to provide 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-4-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridine as a yellow solid (690 mg, 73 wt %) in 92% yield. Analytically pure material was obtained by crystallization in n-propanol and heptane. 1H NMR (500 MHz, CLCl3) δ=8.08 (s, 1H), 8.00 (s, 1H), 7.95 (s, 1H), 7.52 (s, 1H), 4.20 (s, 3H), 1.62 (s, 9H).
  • Synthesis of (R)-4-((R)-1-((6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yl)oxy)ethyl)-1-((S)-1-(4-methoxyphenyl)ethyl)pyrrolidin-2-one (8.1)
  • Figure US20180072744A1-20180315-C00103
  • A mixture of (R)-4-((R)-1-hydroxyethyl)-1-((S)-1-(4-methoxyphenyl)ethyl)pyrrolidin-2-one (173 mg, 0.66 mmol), 6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-4-chloro-2-methyl-2H-pyrazolo[4,3-c]pyridine (190 mg, 66 mmol) and sodium hydride (60 wt %, 66 mg, 1.6 mmol) in anhydrous 1,4 dioxane (3 mL) was agitated under argon at 50-70° C. for 18 h. The reaction mixture was cooled to ambient temperature and quenched by the slow addition of a hydrogen chloride solution (4 M in 1,4 dioxane, 0.245 mL, 0.98 mmol). The reaction was diluted with isopropyl acetate (20 mL) and water (20 mL). The layers were separated, and the aqueous portion was extracted twice with isopropyl acetate (20 ml). The combined organic layers were concentrated to an oil. Purification by silica gel chromatography with methanol and methyl tert-butyl ether provided (R)-4-((R)-1-((6-(1-(tert-Butyl)-1H-pyrazol-4-yl)-2-methyl-2H-pyrazolo[4,3-c]pyridin-4-yl)oxy)ethyl)-1-((S)-1-(4-methoxyphenyl)ethyl)pyrrolidin-2-one (185 mg) as a white foam in 55% yield. 1H NMR (500 MHz, CDCl3) δ=7.94 (s, 1H), 7.89 (s, 1H), 7.76 (s, 1H), 7.20 (s, 1H), 7.10 (d, J=8.8 Hz, 2H), 6.57 (d, J=8.7 Hz, 2H), 5.48 (dddd, J=4.8, 6.1, 6.1, 6.1Hz, 1H), 5.43 (q, J=7.0 Hz, 1H), 4.13 (s, 3H), 3.66 (s, 3H), 3.47 (t, J=9.0 Hz, 1H), 2.96 (dd, J=5.1, 9.8 Hz, 1H), 2.74-2.66 (m, 1H), 2.66-2.57 (m, 2H), 1.64 (s, 9H), 1.48 (d, J=7.1Hz, 3H), 1.33 (d, J=6.2 Hz, 3H).
  • Synthesis of (4R)-4-[(1R)-1-[6-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 3)
  • Figure US20180072744A1-20180315-C00104
  • To a mixture of 50 mg [2-Methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]boronic acid 11.1, 46.2 mg 4-Bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole 3.26 and 20 mg palladium-X-phos was added to 3 mL dioxane and 65 μL 5M aqueous sodium carbonat solution. The resulting mixture was heated at 110° C. for 14h. The mixture was cooled to ambient temperature aund purified via rpHPLC. The combined organic phases were lyophilized to provide example 3 in 21 mg.
  • Analysis: HPLC-MS: Rt=0.86 min (method Z1), M+H=411
  • Example 17 was synthesized in analogous manner to Example 3 using 1-[(4-bromophenyl)methyl]-2-methyl-1H-imidazole 3.27.
  • Analysis: HPLC-MS: Rt=0.73 min (method Z1), M+H=431
  • Synthesis of (4R)-4-[(1R)-1-[6-(4-Hydroxy-3-methoxy-phenyl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 8)
  • Figure US20180072744A1-20180315-C00105
  • To a mixture of 100 mg (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1, 110 mg 2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol 4.24 and 37 mg 1,1′-bis(diphenylphospino)ferrocene-dichloropalladium(II) (complex with DCM (1:1)) in 1 mL dioxane and 0.5 mL methanol was added 400 μL 2M aqueous sodium carbonat solution. The reaction mixture was stirred at 140° C. for 15 min under microwave irradiation. The reaction mixture was filtered through rpSiO2, washed with methanol and purified by rpHPLC to yield after lyophilisation 80 mg (yield: 68%) of Example 8 as solid.
  • Analysis: HPLC-MS: Rt=0.47 min (method S), M+H=383
  • The following Examples were synthesized in analogous manner to Example 8.
  • Boronic acid/ester
    or BF3 borates
    (corresponding to
    Example formula 4) Yield Analysis
    Example 7 1-Cyclohexyl-4- 50 mg HPLC-MS:
    (R)-4-((R)-1-(6-(1-cyclohexyl- (4,4,5,5-tetramethyl- (47%) Rt = 0.72 min
    1H-pyrazol-4-yl)-2-methyl-2H- 1,3,2-dioxoborolan- (method U),
    pyrazolo[4,3-c]pyridin-4- 2-yl)-1H-pyrazole M + H = 409
    yloxy)ethyl)pyrrolidin-2-one 4.50
    Example 53 tert-butyl 5-meth- 51 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(5-methoxy- oxy-3-(4,4,5,5- (51%) Rt = 0.64 min
    1H-indol-3-yl)-2-methyl- tetramethyl-1,3,2- (method U),
    pyrazolo[4,3-c]pyridin-4- dioxaborolan-2- M + H = 406
    yl]oxyethyl]pyrrolidin-2-one yl)indole-1- 1H NMR (DMSO,
    carboxylate 4.26 400 MHz) δ = 1.42
    (3H, d), 2.20-2.36
    (2H, m), 2.75-
    2.89 (1H, m), 3.12
    (dd, 1H), 3.40(1H,
    t), 3.82 (3H, s), 4.12
    (3H, s), 5.58-5.68
    (1H, m), 6.79
    (1H, d), 7.32 (1H,
    d), 7.41 (1H, s),
    7.53 (1H, s), 7.80
    (1H, s), 7.96 (1H,
    s), 8.42 (s, 1H),
    11.2 (1H, s).
    Example 54 potassium 5-methyl- 36 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(5- 2-thiophene-tri- (67%) Rt = 0.68 min
    methyl-2-thienyl)pyrazolo-[4,3- fluoroborate 4.42 (method V),
    c]pyridin-4-yl]oxyethyl]- M + H = 357
    pyrrolidin-2-one
    Example 55 3-methyl-7-(4,4,5,5- 29 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(3- tetramethyl-1,3,2- (46%) Rt = 0.62 min
    methyl-1,2,4,5-tetrahydro-3- dioxaborolan-2-yl)- (method V),
    benzazepin-7-yl)pyrazolo-[4,3- 1,2,4,5-tetrahydro-3- M + H = 420
    c]pyridin-4-yl]oxyethyl]- benzazepine 4.3
    pyrrolidin-2-one
    Example 56 N,N,1-trimethyl-6- 34 mg HPLC-MS:
    N,N,1-trimethyl-6-[2-methyl-4- (4,4,5,5-tetramethyl- (49%) Rt = 0.59 min
    [(1R)-1-[(3R)-5-oxopyrrol-idin- 1,3,2-dioxaborolan- (method V),
    3-yl]ethoxy]pyrazolo[4,3- 2-yl)indole-2- M + H = 461
    c]pyridin-6-yl]indole-2- carboxamide 4.6
    carboxamide
    Example 59 potassium 6- 37 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(6-methoxy- methoxy-3-pyridyl- (66%) Rt = 0.57 min
    3-pyridyl)-2-methyl-pyrazolo- trifluoroborate (method V),
    [4,3-c]pyridin-4-yl]oxyethyl]- 4.44 M + H = 368
    pyrrolidin-2-one
    Example 60 potassium 4- 19 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[4- (trifluoromethyl)phenyl- (31%) Rt = 0.75 min
    (trifluoromethyl)phenyl]- trifluoroborate 4.44 (method V),
    pyrazolo[4,3-c]pyridin-4-yl]- M + H = 405
    oxyethyl]pyrrolidin-2-one
    Example 64 1-tetrahydropyran-4- 48 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(1- yl-4-(4,4,5,5- (76%) Rt = 0.57 min
    tetrahydropyran-4-yl-pyrazol- tetramethyl-1,3,2- (method W),
    4-yl)pyrazolo[4,3-c]pyridin-4- dioxaborolan-2- M + H = 411
    yl]oxyethyl]-pyrrolidin-2-one yl)pyrazole 4.32
    Example 65 1-isopropyl-4- 31 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1-iso- (4,4,5,5-tetramethyl- (55%) Rt = 0.61 min
    propylpyrazol-4-yl)-2-methyl- 1,3,2-dioxaborolan- (method W),
    pyrazolo[4,3-c]pyridin-4-yl]- 2-yl)pyrazole 4.33 M + H = 369
    oxyethyl]pyrrolidin-2-one 1H NMR (DMSO,
    400 MHz) δ = 1.35
    (3H, d), 1.46
    (6H, d), 2.20-2.32
    (2H, m), 2.72-
    2.83 (1H, m), 3.12
    (m, 1H), 3.38(1H,
    t), 4.10 (3H, s),
    4.48-4.59 (1H, m),
    5.50-5.60 (1H, m),
    7.30 (1H, s), 7.51
    (1H, s), 7.99 (1H,
    s), 8.21 (1H, s),
    8.41 (s, 1H).
    Example 66 4-[5-(4,4,5,5- 49 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(6- tetramethyl-1,3,2- (76%) Rt = 0.46 min
    morpholino-3-pyridyl)- dioxaborolan-2-yl)-2- (method W),
    pyrazolo[4,3-c]pyridin-4- pyridyl]-morpholine M + H = 423
    yl]oxyethyl]pyrrolidin-2-one 4.34
    Example 67 1-methyl-4-[4- 20 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[4- (4,4,5,5-tetramethyl- (24%) Rt = 0.46 min
    (4-methylpiperazin-1- 1,3,2-dioxaborolan- (method W),
    yl)phenyl]pyrazolo[4,3- 2-yl)phenyl]- M + H = 435
    c]pyridin-4-yl]oxyethyl]- piperazine 4.35
    pyrrolidin-2-one
    Example 68 1,3-dimethyl-4- 36 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1,3- (4,4,5,5-tetramethyl- (67%) Rt = 0.52 min
    dimethylpyrazol-4-yl)-2- 1,3,2-dioxaborolan- (method W),
    methyl-pyrazolo[4,3-c]pyridin- 2-yl)pyrazole 4.36 M + H = 355
    4-yl]oxyethyl]pyrrolidin-2-one
    Example 69 1,5-dimethyl-4- 39 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1,5-dimeth- (4,4,5,5-tetramethyl- (72%) Rt = 0.52 min
    ylpyrazol-4-yl)-2-methyl- 1,3,2-dioxaborolan- (method W),
    pyrazolo[4,3-c]pyridin-4- 2-yl)pyrazole 4.37 M + H = 355
    yl]oxyethyl]pyrrolidin-2-one
    Example 70 1-[(3S)- 14 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[1- tetrahydrofuran-3- (23%) Rt = 0.55 min
    [(3S)-tetrahydrofuran-3- yl]-4-(4,4,5,5- (method W),
    yl]pyrazol-4-yl]pyrazolo[4,3- tetramethyl-1,3,2- M + H = 397
    c]pyridin-4-yl]oxyethyl]- dioxaborolan-2-
    pyrrolidin-2-one yl)pyrazole 4.7
    Example 71 2-[2-methyl-4- 42 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-[4-(1,1- (4,4,5,5-tetramethyl- (58%) Rt = 0.66 min
    dioxo-1,2-thiazolidin-2-yl)-3- 1,3,2-dioxaborolan- (method W),
    methyl-phenyl]-2-methyl- 2-yl)phenyl]-1,2- M + H = 470
    pyrazolo[4,3-c]pyridin-4- thiazolidine 1,1-
    yl]oxyethyl]pyrrolidin-2-one dioxide 4.8
    Example 72 (1-methylindazol-6- 34 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(1- yl)boronic acid 4.38 (57%) Rt = 0.67 min
    methylindazol-6-yl)pyraz- (method W),
    olo[4,3-c]pyridin-4- M + H = 391
    yl]oxyethyl]pyrrolidin-2-one
    Example 74 1-[(3R)-tetrahydro- 13 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[1- furan-3-yl]-4- (25%) Rt = 0.47 min
    [(3R)-tetrahydrofuran-3- (4,4,5,5-tetramethyl- (method V),
    yl]pyrazol-4-yl]pyrazolo[4,3- 1,3,2-dioxaborolan- M + H = 397
    c]pyridin-4-yl]oxyethyl]- 2-yl)pyrazole 4.9
    pyrrolidin-2-one
    Example 75 2-methyl-7-(4,4,5,5- 18 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(2- tetramethyl-1,3,2- (23%) Rt = 0.61 min
    methyl-3,4-dihydro-1H- dioxaborolan-2-yl)- (method V),
    isoquinolin-7-yl)pyrazolo[4,3- 3,4-dihydro-1H- M + H = 406
    c]pyridin-4-yl]oxyethyl]- isoquinoline 4.10
    pyrrolidin-2-one
    Example 76 4-(4,4,5,5- 17 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[1- tetramethyl-1,3,2- (25%) Rt = 0.44 min
    (3,3,3-trifluoropropyl)-pyrazol- dioxaborolan-2-yl)-1- (method X),
    4-yl]pyrazolo[4,3-c]pyridin-4- (3,3,3-trifluoro- M + H = 423
    yl]oxyethyl]-pyrrolidin-2-one propyl)pyrazole 4.11
    Example 77 7-chloro-2-(4,4,5,5- 27 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(7-chloro-1H- tetramethyl-1,3,2- (32%) Rt = 0.59 min
    indol-2-yl)-2-methyl- dioxaborolan-2-yl)- (method X),
    pyrazolo[4,3-c]pyridin-4- 1H-indole 4.39 M + H = 410
    yl]oxyethyl]pyrrolidin-2-on
    Example 78 1-cyclopropyl-4- 40 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1- (4,4,5,5-tetramethyl- (51%) Rt = 0.40 min
    cyclopropylpyrazol-4-yl)-2- 1,3,2-dioxaborolan- (method X),
    methyl-pyrazolo[4,3-c]pyridin- 2-yl)pyrazole 4.40 M + H = 367
    4-yl]oxyethyl]pyrrolidin-2-one
    Example 79 5-methyl-1- 26 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(3- tetrahydropyran-4-yl- (28%) Rt = 0.39 min
    methyl-1-tetrahydropyran-4-yl- 4-(4,4,5,5- (method Y),
    pyrazol-4-yl)pyrazolo-[4,3- tetramethyl-1,3,2- M + H = 425
    c]pyridin-4-yl]oxyethyl]- dioxaborolan-2-
    pyrrolidin-2-one yl)pyrazole 4.20
    Example 80 1-methyl-4-[4- 24 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[4- (4,4,5,5-tetramethyl- (32%) Rt = 0.50 min
    (1-methyl-4-piperidyl)- 1,3,2-dioxaborolan- (method Y),
    phenyl]pyrazolo[4,3-c]pyridin- 2-yl)phenyl]- M + H = 434
    4-yl]oxyethyl]pyrrolidin-2-one piperidine 4.12
    Example 81 2-(4-isopropoxy-3- 35 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(4- methoxy-phenyl)- (35%) Rt = 0.51 min
    isopropoxy-3-methoxy- 4,4,5,5-tetramethyl- (method Y),
    phenyl)-2-methyl-pyrazolo- 1,3,2-dioxaborolane M + H = 425
    [4,3-c]pyridin-4-yl]oxyethyl]- 4.13
    pyrrolidin-2-one
  • The following examples were synthesized in analogy to Example 8 but using different reaction solvents:
  • Boronic acid/ester
    (corresponding to Yield
    Example formula 4) Solvent Analysis
    Example 11 11 mg HPLC-MS:
    (4S)-4-[(1R)-1-[2-methyl-6-[2- (9%) Rt = 0.463 min
    methyl-4-[(1R)-1-[(3R)-5- dioxane (method C)
    oxopyrrolidin-3-yl]ethoxy]- M + H = 519
    pyrazolo[4,3-c]pyridin-6-
    yl]pyrazolo[4,3-c]pyridin-4-
    yl]oxyethyl]pyrrolidin-2-one
    Example 12 trans 1-methyl-4-{4- 13 mg HPLC-MS:
    1-methyl-4-[4-[4-[2-methyl-4- [4-(4,4,5,5- (10%) Rt = 0.423 min
    [(1R)-1-[(3R)-5-oxopyrrolidin- tetramethyl-1,3,2- dioxane (method C)
    3-yl]ethoxy]pyrazolo[4,3- dioxaborolan-2-yl)- M + H = 521
    c]pyridin-6-yl]pyrazol-1-yl]- pyrazol-1-yl]-
    cyclohexyl]piperazin-2-one cyclohexyl}-piper-
    azin-2-one 4.4
    Example 62 (1-methylindazol-5- 33 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(1- yl)boronic acid 4.30 (50%) Rt = 0.55 min
    methylindazol-5- DMA (method V),
    yl)pyrazolo[4,3-c]pyridin-4- M + H = 391
    yl]oxyethyl]pyrrolidin-2-one 1H NMR (DMSO,
    400 MHz) δ =
    1.42 (3H, d), 2.20-
    2.38 (2H, m),
    2.75-2.88 (1H,
    m), 3.18 (dd, 1H),
    3.40(1H, t), 4.08
    (3H, s), 4.13
    (3H, s), 5.59-5.68
    (1H, m), 7.53 (1H,
    s), 7.65 (1H, s),
    7.70 (1H, d), 7.80
    (1H, s), 8.12 (1H,
    s), 8.16 (1H, d).
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(4-piperidyl)pyrazol-4-yl]pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 9)
  • Step 1: Synthesis of tert-Butyl 4-[4-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]pyrazol-1-yl]piperidine-1-carboxylate
  • Figure US20180072744A1-20180315-C00106
  • To a mixture of 100 mg (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1, 170 mg tert-butyl 4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine-1-carboxylate 4.41 and 37 mg 1,1′-bis(diphenylphospino)ferrocenedichloropalladium(II) (complex with DCM (1:1)) in 1 mL dioxane and 0.5 mL methanol was added 400 μL 2M aqueous sodium carbonat solution. The reaction mixture was stirred at 140° C. for 15 min under microwave irradiation. The reaction mixture was filtered through rpSiO2, washed with methanol and purified by rpHPLC (XbridgeC18, acetonitrile/water, ammonia) to yield after lyophilisation 90 mg (80% per NMR) of tert-butyl 4-[4-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo-[4,3-c]pyridin-6-yl]pyrazol-1-yl]piperidine-1-carboxylate as solid.
  • Analysis: HPLC-MS: Rt=0.45 min (method J), M+H=510
  • Step 2: Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(4-piperidyl)pyrazol-4-yl]pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 9)
  • Figure US20180072744A1-20180315-C00107
  • A solution of 90 mg (80% per NMR) of tert-butyl 4-[4-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]pyrazol-1-yl]piperidine-1-carboxylate in 2 mL TFA was stirred at room temperature for 15 min. The reaction mixture was purified by rpHPLC (SunfireC18, acetonitrile/water, TFA) to yield after lyophilisation 83 mg of Example 9 as solid.
  • Analysis: HPLC-MS: Rt=0.32 min (method J), M+H=410
  • Synthesis of (4R)-4-[(1R)-1-[6-[1-(4,4-Difluorocyclohexyl)pyrazol-4-yl]-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 10)
  • Figure US20180072744A1-20180315-C00108
  • A mixture of 100 mg [2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]boronic acid 11.1, 87 mg 4-bromo-1-(4,4-difluoro-cyclohexyl)pyrazole 3.12, 27 mg 1,1′-bis(diphenylphospino)ferrocenedichloropalladium(II) (complex with DCM (1:1)) and 493 μL 2M aqueous sodium carbonat solution in 2 mL dioxane was stirred at 120° C. for 15 min under microwave irradiation. The reaction mixture was filtered and purified by rpHPLC to yield after lyophilisation 23 mg of Example 10 as solid.
  • Analysis: HPLC-MS: Rt=0.535 min (method C), M+H=445
  • 1H NMR (DMSO, 400 MHz) δ=1.38 (3H, d), 1.58 (9H, s), 2.00-2.20 (10H, m), 2.73-2.86 (1H, m), 3.11-3.16 (m, 1H), 3.38(1H, t), 4.09 (3H,$), 4.39-4.49 (1H,m), 5.50-5.61 (1H,m), 7.31 (1H, s), 7.50 (1H, s), 8.00 (1H, s), 8.26 (1H, s), 8.40 (s, 1H).
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(3,3,3-trifluoropropyl)pyrazol-4-yl]indazol-4-yl]oxyethyl]pyrrolidin-2-one (Example 15)
  • Figure US20180072744A1-20180315-C00109
  • To a mixture of 100 mg (4R)-4-[(1R)-1-[2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indazol-4-yl]oxyethyl]pyrrolidin-2-one 11.3, 95 mg 4-bromo-1-(3,3,3-trifluoro-propyl)pyrazole 3.15 and 9.1 mg 1,1′-bis(triphenylphosphine)palladium(II) chloride in 1.5 mL ethanol (80% with toluene) was added 389 μL 2M aqueous sodium carbonat solution. The resulting mixture was stirred at 100° C. for 1 h under microwave irradiation. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol) and by rpHPLC to yield 27 mg (yield: 25%) of Example 15 as solid.
  • Analysis: HPLC-MS: Rt=2.33 min (method R), M+H=422
  • The following Examples were synthesized in analogous manner to Example 15.
  • Bromide
    (corresponding to
    Example formula 3) Yield Analysis
    Example 16 3-bromo-1H- 12 mg HPLC-MS:
    3-[2-methyl-4-[(1R)-1-[(3R)-5- indazole-5-carbo- (12%) Rt = 2.20 min
    oxopyrrolidin-3-yl]- nitrile 3.16 (method R),
    ethoxy]indazol-6-yl]-1H- M + H = 401
    indazole-5-carbonitrile
    Example 41 2-bromo-5-fluoro- 19 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(5-fluoro-2- pyridine 3.17 (27%) Rt = 2.23 min
    pyridyl)-2-methyl-indazol-4- (Method R),
    yl]oxyethyl]pyrrolidin-2-one M + H = 355
    Example 42 4-(4-bromo- 34 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[4- phenyl)-1-methyl- (40%) Rt = 1.53 min
    (1-methyl-4-piperidyl)- piperidine 3.18 (method R),
    phenyl]indazol-4-yl]oxyethyl]- M + H = 433
    pyrrolidin-2-one
    Example 43 3-bromoimidazo- 21 mg HPLC-MS:
    3-[2-methyl-4-[(1R)-1-[(3R)-5- [1,2-a]pyridine-6- (26%) Rt = 1.65 min
    oxopyrrolidin-3-yl]eth- carbonitrile 3.14 (method R),
    oxy]indazol-6-yl]imidazo[1,2- M + H = 401
    a]pyridine-6-carbonitrile
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-(4-morpholinophenyl)indazol-4-yl]oxyethyl]-pyrrolidin-2-one (Example 22)
  • Figure US20180072744A1-20180315-C00110
  • To a mixture of 105 mg (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one 7.4, 96 mg (4-morpholinophenyl)boronic acid 4.46 and 10.9 mg 1,1′-bis(triphenylphosphine)palladium(II) chloride in 1.58 mL ethanol (80% with toluene) was added 466 μL 2M aqueous sodium carbonat solution. The resulting mixture was stirred at 95° C. for 1 h. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol) and by rpHPLC to yield 74 mg (yield: 56%) of Example 22 as solid.
  • Analysis: HPLC-MS: Rt=2.35 min (method R), M+H=421
  • The following Examples were synthesized in analogous manner to Example 22, but with modified reaction time.
  • Boronic acid/ester Yield
    (corresponding to Reaction
    Example formula 4) time Analysis
    Example 14 1-methyl-3-(4,4,5,5- 49 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- tetramethyl-1,3,2- (66%) Rt = 1.86 min
    (1-methylpyrazol-3- dioxaborolan-2- 2 h (method R),
    yl)indazol-4-yl]oxyethyl]- yl)pyrazole 4.49 M + H = 340
    pyrrolidin-2-one
    Example 18 1-tert-butyl-4- 95 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1-tert- (4,4,5,5-tetramethyl- (80%) Rt = 2.35 min
    butylpyrazol-4-yl)-2-methyl- 1,3,2-dioxaborolan- 1 h (method R),
    indazol-4- 2-yl)pyrazole 4.19 M + H = 382
    yl]oxyethyl]pyrrolidin-2-one 1H NMR (DMSO,
    500 MHz) δ = 1.28
    (3H, d, J = 6.1 Hz),
    1.56 (9H, s), 2.19-
    2.35 (2H, m), 2.74
    (1H, h, J = 8.2 Hz),
    3.12 (1H, dd, J = 9.6,
    6.8 Hz), 3.37 (1H, t,
    J = 9.0 Hz), 4.08 (3H,
    s), 4.76 (1H, d,
    J = 6.0 Hz), 6.71 (1H,
    s), 7.34 (1H, s), 7.57
    (1H, s), 7.91 (1H, s),
    8.22 (1H, s), 8.29
    (1H, s)
    Example 26 1-methyl-4-[4- 97 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- (4,4,5,5-tetramethyl- (72%) Rt = 1.43 min
    [4-(4-methylpiperazin-1- 1,3,2-dioxaborolan- 1.5 h (method R),
    yl)phenyl]indazol-4- 2-yl)phenyl]- M + H = 434
    yl]oxyethyl]pyrrolidin-2-one piperazine 4.35
    Example 27 2-isopropoxy-5- 78 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(6- (4,4,5,5-tetramethyl- (90%) Rt = 2.75 min
    isopropoxy-3-pyridyl)-2- 1,3,2-dioxaborolan- 1.5 h (method R),
    methyl-indazol-4- 2-yl)pyridine 4.48 M + H = 395
    yl]oxyethyl]pyrrolidin-2-one
    Example 28 (1-methylindazol-5- 67 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- yl)boronic acid 4.47 (78%) Rt = 2.30 min
    (1-methylindazol-5-yl)indaz- 1.5 h (method R),
    ol-4-yl]oxyethyl]pyrrolidin-2- M + H = 390
    one
    Example 32 1-methyl-4-[4- 75 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- (4,4,5,5-tetramethyl- (67%) Rt = 1.75 min
    [4-(4-methylpiperazin-1-yl)-3- 1,3,2-dioxaborolan- 1 h (method R),
    (trifluoromethyl)phenyl]- 2-yl)-2-(trifluoro- M + H = 502
    indazol-4-yl]oxyethyl]pyrrolidin- methyl)phenyl]piperazine
    2-one 4.16
    Example 33 2-[4-(4,4,5,5- 45 mg HPLC-MS:
    2-[4-(4,4,5,5-tetramethyl- tetramethyl-1,3,2- (45%) Rt = 2.25 min
    1,3,2-dioxaborolan-2- dioxaborolan-2- 1 h (method R),
    yl)phenyl]-1,2-thiazolidine yl)phenyl]-1,2- M + H = 455
    1,1-dioxide thiazolidine 1,1-
    dioxide 4.14
    Example 36 1-methyl-5-(4,4,5,5- 66 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(3-amino-1- tetramethyl-1,3,2- (73%) Rt = 1.97 min
    methyl-indazol-5-yl)-2- dioxaborolan-2- 1 h (method R),
    methyl-indazol-4- yl)indazol-3-amine M + H = 405
    yl]oxyethyl]pyrrolidin-2-one 4.15
    Example 38 4-[4-(4,4,5,5- 43 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- tetramethyl-1,3,2- (40%) Rt = 3.11 min
    [4-morpholino-3- dioxaborolan-2-yl)-2- 1 h (method R),
    (trifluoromethyl)phenyl]indazol- (trifluoromethyl)- M + H = 489
    4-yl]oxyethyl]pyrrolidin-2- phenyl]morpholine
    one 4.18
    Example 40 1-(difluoromethyl)-4- 53 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-[1- (4,4,5,5-tetramethyl- (63%) Rt = 2.16 min
    (difluoromethyl)pyrazol-4-yl]- 1,3,2-dioxaborolan- 1 h (method R),
    2-methyl-indazol-4- 2-yl)pyrazole 4.17 M + H = 376
    yl]oxyethyl]pyrrolidin-2-one
    Example 47 1-tetrahydropyran-4- 68 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- yl-4-(4,4,5,5- (75%) Rt = 1.95 min
    (1-tetrahydropyran-4- tetramethyl-1,3,2- 1.5 h (method R),
    ylpyrazol-4-yl)indazol-4- dioxaborolan-2- M + H = 410
    yl]oxyethyl]pyrrolidin-2-one yl)pyrazole 4.32 1H NMR (DMSO,
    500 MHz) δ = 1.29
    (3H, d, J = 6.1 Hz),
    1.92-2.08 (4H, m),
    2.18-2.35 (2H, m),
    2.75 (1H, dt, J = 14.9,
    7.6 Hz), 3.12 (1H,
    dd, J = 9.5, 6.8 Hz),
    3.38 (1H, t, J = 9.1
    Hz), 3.48 (2H, td,
    J = 11.4, 3.1 Hz), 3.98
    (2H, d, J = 11.2 Hz),
    4.08 (3H, s), 4.40
    (1H, tt, J = 10.1, 5.1
    Hz), 4.74 (1H, p,
    J = 6.0 Hz), 6.69 (1H,
    s), 7.33 (1H, s), 7.58
    (1H, s), 7.94 (1H, s),
    8.23 (1H, s), 8.31
    (1H, s)
    Example 48 7-chloro-2-(4,4,5,5- 65 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(7-chloro- tetramethyl-1,3,2- (71%) Rt = 3.05 min
    1H-indol-2-yl)-2-methyl- dioxaborolan-2-yl)- 1 h (method R),
    indazol-4-yl]oxyethyl]- 1H-indole 4.39 under M + H = 409/411
    pyrrolidin-2-one microwave
    irradiation
    Example 49 1-methyl-4-[4- 64 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- (4,4,5,5-tetramethyl- (68%) Rt = 1.25 min
    [1-(1-methyl-4-piperidyl)- 1,3,2-dioxaborolan- 1 h (method R),
    pyrazol-4-yl]indazol-4-yl]- 2-yl)pyrazol-1- M + H = 423
    oxyethyl]pyrrolidin-2-one yl]piperidine 4.21
  • Synthesis of (4R)-4-[(1R)-1-(2-methyl-6-thiazol-4-yl-indazol-4-yl)oxyethyl]pyrrolidin-2-one (Example 29)
  • Figure US20180072744A1-20180315-C00111
  • A mixture of 75 mg (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one (9.2), 102 μL tributyl(thiazol-4-yl)stannane and 7.8 mg 1,1′-bis(triphenylphosphine) palladium(II) chloride in 1.5 mL dioxane was stirred at 95° C. for 18 h. The reaction mixture was concentrated in vacuo. The crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol) and by rpHPLC to yield 13 mg (yield: 19%) of Example 29 as solid.
  • Analysis: HPLC-MS: Rt=1.98 min (method R), M+H=343
  • The following Example was synthesized in analogous manner to Example 29, but with modified reaction time.
  • Stannane Yield
    (corresponding to Reaction
    Example formula 4) time Analysis
    Example 35 2-difluoromethyl-4- 49 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-[2- tributylstannanyl- (42%) Rt = 2.50 min
    (difluoromethyl)thiazol-4-yl]- thiazole 4.1 1 h (method R),
    2-methyl-indazol-4-yl]- M + H = 393
    oxyethyl]pyrrolidin-2-one
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(4-piperidyl)pyrazol-4-yl]indazol-4-yl]oxyethyl]pyrrolidin-2-one (Example 45)
  • Figure US20180072744A1-20180315-C00112
  • To a mixture of 125 mg (4R)-4-[(1R)-1-(6-bromo-2-methyl-indazol-4-yl)oxyethyl]pyrrolidin-2-one 7.5, 167 mg tert-butyl 4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine-1-carboxylate 4.41 and 13 mg 1,1′-bis(triphenylphosphine)palladium(II) chloride in 1.88 mL ethanol (80% with toluene) was added 554 μL 2M aqueous sodium carbonat solution. The resulting mixture was stirred at 95° C. for 1 h. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The crude residue was purified by flash chromatography (heptane/ethyl acetate/methanol). The residue was dissolved in 2 mL DCM, 500 μL TFA were added and the resulting mixture was stirred at room temperature for 1 h. The reaction mixture was purified by flash chromatography (DCM/methanol/ammonia) and by elution through a SCX (Biotage SCX-3) column and subsequent rpHPLC to yield 92 mg (yield: 61%) of Example 45 as solid.
  • Analysis: HPLC-MS: Rt=1.25 min (method R), M+H=409
  • Synthesis of (4R)-4-[(1R)-1-[6-(6-Cyclopropyl-3-pyridyl)-2-methyl-pyrazolo[4,3-c]pyridine-4-yl]oxyethyl]pyrrolidin-2-one (Example 50)
  • Figure US20180072744A1-20180315-C00113
  • To a mixture of 50 mg (4R)-4-[(1R)-1-(6-chloro-2-methyl-pyrazolo[4,3-c]pyridin-4-yl)oxyethyl]pyrrolidin-2-one 7.1, 94 mg 2-cyclopropyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine 4.23 and 20 mg 1,1′-bis(triphenylphosphine)palladium(II) chloride in 1 mL DMA was added 400 μL 2M aqueous sodium carbonat solution. The reaction mixture was stirred at 130° C. for 25 min under microwave irradiation. The reaction mixture was filtered through rpSiO2, washed with methanol and purified by rpHPLC to yield after lyophilisation 22 mg (yield: 37%) of Example 50 as solid.
  • Analysis: HPLC-MS: Rt=0.69 min (method U), M+H=378
  • The following Examples were synthesized in analogous manner to Example 50.
  • Boronic acid/ester
    (corresponding to
    Example formula 4) Yield Analysis
    Example 2 1-tert-butyl-4- 1.11 g HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1-tert- (4,4,5,5- (54%) Rt = 0.50 min
    butylpyrazol-4-yl)-2-methyl- tetramethyl-1,3,2- (method C),
    pyrazolo[4,3-c]pyridin-4- dioxaborolan-2- M + H = 383
    yl]oxyethyl]pyrrolidin-2-one yl)pyrazole 4.19 1H NMR (DMSO,
    400 MHz) δ = 1.38
    (3H, d), 1.58 (9H,
    s), 2.20-2.34 (2H,
    m), 2.73-2.85 (1H,
    m), 3.11-3.19 (m,
    1H), 3.38(1H, t),
    4.10 (3H, s), 5.50-
    5.60 (1H, m), 7.33
    (1H, s), 7.52 (1H,
    s), 7.98 (1H, s),
    8.26 (1H, s), 8.41
    (s, 1H).
    Example 51 2-difluoromethyl-5- 24 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-[6- (4,4,5,5- (40%) Rt = 0.65 min
    (difluoromethyl)-3-pyridyl]-2- tetramethyl-1,3,2- (method U),
    methyl-pyrazolo[4,3-c]pyridin- dioxaborolan-2-yl)- M + H = 388
    4-yl]oxyethyl]pyrrolidin-2-one pyridine 4.5
    Example 52 (3,4,5-trimethoxy- 35 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6- phenyl)boronic acid (54%) Rt = 0.67 min
    (3,4,5-trimethoxyphenyl)- 4.25 (method W),
    pyrazolo[4,3-c]pyridin-4- M + H = 427
    yl]oxyethyl]pyrrolidin-2-one
    Example 57 1-(2-methoxy- 36 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-[1-(2- ethyl)-4-(4,4,5,5- (62%) Rt = 0.53 min
    methoxyethyl)pyrazol-4-yl]-2- tetramethyl-1,3,2- (method W),
    methyl-pyrazolo[4,3-c]pyridin- dioxaborolan-2- M + H = 385
    4-yl]oxyethyl]pyrrolidin-2-one yl)pyrazole 4.27
    Example 58 1-ethyl-4-(4,4,5,5- 31 mg HPLC-MS:
    (4R)-4-[(1R)-1-[6-(1- tetramethyl-1,3,2- (45%) Rt = 0.48 min
    ethylpyrazol-4-yl)-2-methyl- dioxaborolan-2- (method V),
    pyrazolo[4,3-c]pyridin-4- yl)pyrazole 4.28 M + H = 355
    yl]oxyethyl]pyrrolidin-2-one
    Example 61 [6-(trifluoro- 15 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-[6- methyl)-3-pyridyl] (23%) Rt = 0.65 min
    (trifluoromethyl)-3- boronic acid 4.29 (method V),
    pyridyl]pyrazolo[4,3-c]pyri-din- M + H = 406
    4-yl]oxyethyl]pyrrolidin-2-one
    Example 63 1-methyl-4- 26 mg HPLC-MS:
    (4R)-4-[(1R)-1-[2-methyl-6-(1- (4,4,5,5- (50%) Rt = 0.44 min
    methylpyrazol-4- tetramethyl-1,3,2- (method V),
    yl)pyrazolo[4,3-c]pyridin-4- dioxaborolan-2- M + H = 341
    yl]oxyethyl]pyrrolidin-2-one yl)pyrazole 4.31
  • Synthesis of (4R)-4-[(1R)-1-[6-(3-Methoxy-4-tetrahydropyran-4-yloxy-phenyl)-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 73)
  • Figure US20180072744A1-20180315-C00114
  • A mixture of 68 mg of Example 8, 59 mg of tetrahydropyran-4-yl 4-methylbenzenesulfonate and 39 mg potassium carbonate in 2 mL DMF was stirred at 80° C. for 3 h and at 100° C. for 10 h. The reaction mixture was diluted with water and extracted with DCM. The combined organic phases were concentrated in vacuo. The resulting residue was purified by rpHPLC to yield after lyophilisation 37 mg of Example 73 as solid.
  • Analysis: HPLC-MS: Rt=0.70 min (method W), M+H=467
  • Synthesis of (4R)-4-[(1R)-1-[2-Methyl-6-[1-(1-methyl-4-piperidyl)pyrazol-4-yl]pyrazolo [4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 82)
  • Figure US20180072744A1-20180315-C00115
  • To a mixture of 68 mg of Example 9 and 31 mg sodium acetate in 3 mL DCM and 0.5 mL methanol was added 17 μL formaldehyde (aqueous 37%). The resulting mixture was stirred at room temperature for 10 min, before 46 mg sodium triacetoxyborohydride was added. The reaction mixture was stirred for 1.75 h before quenched with water. The organic solvent was removed by destillation. The resulting residue was purified by rpHPLC to yield after lyophilisation 64 mg of Example 82 as solid.
  • Analysis: HPLC-MS: Rt=0.28 min (method X), M+H=424
  • Synthesis of (4R)-4-[(1R)-1-[6-[1-(4,4-(Difluoromethyl)imidazol-4-yl]-2-methyl-pyrazolo[4,3-c]pyridin-4-yl]oxyethyl]pyrrolidin-2-one (Example 83) and 2-Methyl-6-[2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]-5H-pyrazolo[4,3-c]pyridin-4-one (Example 13)
  • Figure US20180072744A1-20180315-C00116
  • To a mixture of 300 mg (crude) [2-methyl-4-[(1R)-1-[(3R)-5-oxopyrrolidin-3-yl]ethoxy]pyrazolo[4,3-c]pyridin-6-yl]boronic acid 11.1, 60 mg 4-bromo-1-(difluoromethyl)imidazole and 41 mg 1,1′-bis(diphenylphospino)ferrocenedichloro palladium(II) (complex with DCM (1:1)) in 2 mL dioxane and 0.5 mL methanol was added 670 μL 2M aqueous sodium carbonat solution. The reaction mixture was stirred at 140° C. for 15 min under microwave irradiation. The reaction mixture was filtered through Agilent PL-Thiol MP-SPE, washed with methanol and purified by rpHPLC to yield after lyophilisation 9 mg of Example 83 and 4 mg of Example 13 as solids.
  • Analysis (Example 83): HPLC-MS: Rt=0.43 min (method C), M+H=377
  • Analysis (Example 13): HPLC-MS: Rt=0.38 min (method C), M+H=408
  • 4.5 Analytical Methods
  • The Example compounds prepared according to the foregoing synthesis schemes were characterised by the following chromatographic methods and/or NMR spectroscopy.
  • 4.5.1 Chromatographic Methods (HPLC-MS Methods)
  • Method A
  • Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm
    Column supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.]
    0.00 99 1 1.3 60
    0.02 99 1 1.3 60
    1.00 0 100 1.3 60
    1.10 0 100 1.3 60
  • Method B:
  • Column: Sunfire C18, 3 × 30 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% TFA] [Methanol] [ml/min] [° C.]
    0.0 95 5 1.8 60
    0.25 95 5 1.8 60
    1.70 0 100 1.8 60
    1.75 0 100 2.5 60
    1.90 0 100 2.5 60
  • Method C:
  • Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm
    Column supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.]
    0.00 95 5 1.3 60
    0.02 95 5 1.3 60
    1.00 0 100 1.3 60
    1.10 0 100 1.3 60
  • Method D:
  • Column: XBridge C18, 2.1 × 20 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.10% TFA] [Methanol] [ml/min] [° C.]
    0.0 95 5 1.4 60
    0.05 95 5 1.4 60
    1.00 0 100 1.4 60
    1.1 0 100 1.4 60
  • Method E:
  • Column: Sunfire C18, 2.1 × 20 mm, 2,5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.10% TFA] [Methanol] [ml/min] [° C.]
    0.00 99 1 1.3 60
    0.15 99 1 1.3 60
    1.10 0 100 1.3 60
    1.25 0 100 1.3 60
  • Method F:
  • Column: XBridge C18, 3 × 30 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.]
    0.00 97 3 2.2 60
    0.20 97 3 2.2 60
    1.20 0 100 2.2 60
    1.25 0 100 3 60
    1.40 0 100 3 60
  • Method G:
  • Eluent A: Water/0.2% KH2PO4 pH=3
  • Eluent B: Acetonitrile
  • Time Flow rate
    [min] % A % B [mL/min]
    0.00 80 20 1.50
    5.00 20 80 1.50
    8.00 20 80 1.50
  • The stationary phase used was a Inertsil C8-3 (GL Sciences), 5 μm; dimension: 100×4.0 mm,
  • (column temperature: constant at 30° C.). Detection UV 220 nm.
  • Method H:
  • Eluent A: Hexane
  • Eluent B: 2-Propanol
  • Time Flow rate
    [min] % A % B [mL/min]
    00.00 90 10 1.0
    20.00 90 10 1.0
  • The stationary phase used was a Chiralpak AD-H (Daicel), 5 μm; dimension: 150×4.6 mm, (column temperature: constant at 10° C.). Detection DAD 225 nm.
  • Method I:
  • Eluent A: Hexane
  • Eluent B: 2-Propanol
  • Time Flow rate
    [min] % A % B [mL/min]
    00.00 90 10 1.0
    25.00 90 10 1.0
  • The stationary phase used was a Chiralpak AD-H (Daicel), 5 μm; dimension: 150×4.6 mm,
  • (column temperature: constant at 10° C.).
  • Detection DAD 225 nm.
  • Method J:
  • Column: Sunfire C18, 2.1 × 30 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.]
    0.0 99 1 1.5 60
    0.02 99 1 1.5 60
    1.00 0 100 1.5 60
    1.10 0 100 1.5 60
  • Method K:
  • Column: Waters Atlantis dC18 (2.1×50 mm, 3 μm column)
  • Flow rate: 1 mL/min
  • Solvent A: 0.1% Formic acid/water
  • Solvent B: 0.1% Formic acid/acetonitrile
  • Injection volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 2.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 2.5 to 2.7 minutes, 100% solvent B; 2.71 to 3.0 minutes, 95% solvent A+5% solvent B.
  • MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV
  • UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array,Waters 2487 UV or Shimadzu SPD-M20A PDA
  • Method L:
  • Column: XBridge C18, 4.6 × 30 mm, 3.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% NH3] [ACN] [ml/min] [° C.]
    0.0 97 3 5 60
    0.2 97 3 5 60
    1.6 0 100 5 60
    1.7 0 100 5 60
  • Method M:
  • Column: Waters SymmetryShield RP8 (2.1×50 mm, 3.5 μm column)
  • Flow rate: 1 mUmin
  • Solvent A: 0.1% Formic acid/water
  • Solvent B: 0.1% Formic acid/acetonitrile
  • Injection volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 2.2 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 2.2 to 2.7 minutes, 100% solvent B; 2.71 to 3.0 minutes, 95% solvent A+5% solvent B.
  • MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV
  • UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array,Waters 2487 UV or Shimadzu SPD-M20A PDA
  • Method N:
  • Column: Xbridge BEH C18, 2.1 × 30 mm, 1.7 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.]
    0.0 99 1 1.6 60
    0.02 99 1 1.6 60
    1.00 0 100 1.6 60
    1.10 0 100 1.6 60
  • Method O:
  • Column: Xbridge BEH Phenyl, 2.1 × 30 mm, 1.7 μm
    Column supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% NH3] [Acetonitril] [ml/min] [° C.]
    0.00 95 5 1.3 60
    0.02 95 5 1.3 60
    1.00 0 100 1.3 60
    1.10 0 100 1.3 60
  • Method P:
  • Column: Supelco Ascentis Express (2.1×30 mm, 2.7 μm column)
  • Flow rate: 1 ml/min
  • Solvent A: 0.1% Formic acid/water
  • Solvent B: 0.1% Formic acid/acetonitrile
  • Injection volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 1.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 1.5 to 1.6 minutes, 100% solvent B; 1.60 to 1.61 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 1.61 to 2.00 minutes, 95% solvent A+5% solvent B.
  • MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV
  • UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array,Waters 2487 UV or Shimadzu SPD-M20A PDA
  • Method Q
  • Column: Atlantis d C18; 50×3 mm; 3 μ
  • Flow rate: 0.6m1/min
  • Solvent A: 0.1% Formic acid in water
  • Solvent B: 0.1% Formic acid in acetonitrile
  • Injection Volume: 5 μL
  • Column temperature: 35° C.
  • UV Detection wavelength: Spectra A max (with scan in the region of 200-400 nm)
  • Eluent: 0 to 3.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 3.5 to 3.8 minutes, 100% solvent B; 3.8 to 3.9 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 3.9 to 4.5 minutes, 95% solvent A+5% solvent B.
  • MS detection using Waters 3100, SQ detector, ES+ve and −ve modes (Cone voltage: 30V, Capillary voltage 3.0 KV)
  • UV detection using Waters 2996 photodiode array
  • Method R:
  • Column: Phenomenex Kinetex-XB C18 (2.1×100 mm, 1.7 μm column)
  • Flow rate: 0.6 mL/min
  • Solvent A: 0.1% Formic acid/water
  • Solvent B: 0.1% Formic acid/acetonitrile
  • Injection volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 5.3 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 5.3 to 5.8 minutes, 100% solvent B; 5.80 to 5.82 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.82 to 7mins, 95% solvent A+5% solvent B
  • MS detection using Waters SQD
  • UV detection using Waters Acquity photodiode array
  • Method S:
  • Column: Sunfire C18, 2.1 × 30 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.]
    0.0 99 1 1.3 60
    0.02 99 1 1.3 60
    1.00 0 100 1.3 60
    1.10 0 100 1.3 60
  • Method T:
  • Column: Phenomenex Gemini C18 (2.0 mm×100 mm, 3 μm column)
  • Flow rate: 0.5 mL/min
  • Solvent A: 2 mM Ammonium bicarbonate modified to pH 10 with Ammonium Hydroxide/ water
  • Solvent B: Acetonitrile
  • Injection volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 5.5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 5.5 to 5.9 minutes, 100% solvent B; 5.90 to 5.92 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.92 to 9.00 minutes, 95% solvent A+5% solvent B.
  • Method U:
  • Column: XBridge C18_3.0 × 30 mm, 2.5 μm
    Column producer: Waters
    Description:
    Gradient/ % Sol
    Solvent Time [H2O 0.1% % Sol Flow Temp
    [min] NH4OH] [Acetonitrile] [ml/min] [° C.]
    0.0 98.0 2.0 2.0 60.0
    1.2 0.0 100.0 2.0 60.0
    1.4 0.0 100.0 2.0 60.0
  • Method V:
  • Column: XBridge C18_3.0 × 30 mm, 2.5 μm
    Column producer: Waters
    Gradient/ % Sol
    Solvent Time [H2O 0.1% % Sol Flow Temp
    [min] NH4OH] [Acetonitrile] [ml/min] [° C.]
    0.0 98.0 2.0 2.0 60.0
    1.2 0.0 100.0 2.0 60.0
    1.4 0.0 100.0 2.0 60.0
  • Method W:
  • Column: Sunfire C18_3.0 × 30 mm, 2.5 μm
    Column producer: Waters
    Description:
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O 0.1% TFA] [Acetonitrile] [ml/min] [° C.]
    0.0 98.0 2.0 2.0 60.0
    1.2 0.0 100.0 2.0 60.0
    1.4 0.0 100.0 2.0 60.0
  • Method X:
  • Column: Sunfire C18_2.1 × 50 mm, 2.5 μm
    Column producer: Waters
    Description:
    Gradient/ % Sol
    Solvent Time % Sol [Acetonitrile Flow Temp
    [min] [H2O 0.1% TFA] 0.08% TFA] [ml/min] [° C.]
    0.0 95.0 5.0 1.5 60.0
    0.75 0.0 100.0 1.5 60.0
    0.85 0.0 100.0 1.5 60.0
  • Method Y:
  • Device description: Waters Acquity with 3100 MS
    Column: XBridge BEH C18_3.0 × 30 mm, 1.7 μm
    Column producer: Waters
    Description:
    Gradient/ % Sol
    Solvent Time [H2O 0.1% % Sol Flow Temp
    [min] NH4OH] [Acetonitrile] [ml/min] [° C.]
    0.0 95.0 5.0 1.5 60.0
    0.7 0.1 99.9 1.5 60.0
    0.8 0.1 99.9 1.5 60.0
    0.81 95.0 5.0 1.5 60.0
    1.1 95.0 5.0 1.5 60.0
  • Method Z:
  • Column: Waters Atlantis dC18 (2.1×100 mm, 3 μm column)
  • Flow rate: 0.6 mL/min
  • Solvent A: 0.1% Formic acid/water
  • Solvent B: 0.1% Formic acid/acetonitrile
  • Injection Volume: 3 μL
  • Column temperature: 40° C.
  • UV Detection wavelength: 215 nm
  • Eluent: 0 to 5 minutes, constant gradient from 95% solvent A+5% solvent B to 100% solvent B; 5 to 5.4 minutes, 100% solvent B; 5.4 to 5.42 minutes, constant gradient from 100% solvent B to 95% solvent A+5% solvent B; 5.42 to 7.00 minutes, 95% solvent A+5% solvent B.
  • MS detection using Waters LCT Premier, QTof micro, ZQ or Shimadzu LCMS2010EV
  • UV detection using Waters 2996 photodiode array, Waters 2998 photodiode array, Waters 2487 UV or Shimadzu SPD-M20A PDA
  • Method Z1:
  • Method Name:
    Method Name:
    Column: Sunfire, 3 × 30 mm, 2.5 μm
    Column Supplier: Waters
    Gradient/
    Solvent Time % Sol % Sol Flow Temp
    [min] [H2O, 0.1% TFA] [Acetonitril] [ml/min] [° C.]
    0.00 97 3 2.2 60
    0.20 97 3 2.2 60
    1.20 0 100 2.2 60
    1.25 0 100 3 60
    1.40 0 100 3 60
  • 4.5.2 NMR Spectroscopy
  • Configuration of the Bruker DRX 500 MHz NMR
  • High performance digital NMR spectrometer, 2-channel microbay console and Windows XP host workstation running Topspin version 1.3.
  • Equipped with:
      • Oxford instruments magnet 11.74 Tesla (500 MHz proton resonance frequency)
      • B-VT 3000 temperature controller
      • GRASP II gradient spectroscopy accessory for fast acquisition of 2D pulse sequences
      • Deuterium lock switch for gradient shimming
      • 5 mm Broad Band Inverse geometry double resonance probe with automated tuning and matching (BBI ATMA). Allows 1H observation with pulsing/decoupling of nuclei in the frequency range 15N and 31P with 2H lock and shielded z-gradient coils.
  • Configuration of the Bruker DPX 400MHz NMR
  • High performance one bay Bruker 400 MHz digital two channel NMR spectrometer console and Windows XP host workstation running XwinNMR version 3.5.
      • Equipped with:
      • Oxford instruments magnet 9.39 Tesla (400 MHz proton resonance frequency)
      • B-VT 3300 variable temperature controller unit
      • Four nucleus (QNP) switchable probe for observation of 1H, 13C, 19F and 31P with 2H lock
  • Configuration of the Bruker 500 MHz NMR
  • High performance digital NMR spectrometer, 2-channel one bay console and Linux host workstation running Topspin version 2.1 PL6.
  • Equipped with:
      • Bruker-Biospin AVANCE III 500A magnet 11.75 Tesla (500 MHz proton resonance frequency)
      • B-VT 3000 temperature controller
      • 5 mm Multinuclear Broad Band fluorine observe (BBFO) probe with digital tuning covering the range from 15N and 31P as well as 19F with 1H decoupling.
  • Configuration of the Bruker DPX 400 MHz NMR
  • High performance digital NMR spectrometer, 2-channel microbay console and Linux host workstation running Topspin version 2.1 PL6
  • Equipped with:
      • Bruker—Biospin AVANCE III DPX400C magnet 9.40 Tesla (400 MHz proton resonance frequency)
      • B-VT 3200 variable temperature controller unit
  • 5 mm Multinuclear Broad Band fluorine observe (BBFO) probe with digital tuning covering the range from 15N and 31P as well as 19F with 1H decoupling.
  • 5. EXAMPLES
  • The following Examples were prepared analogously to the methods of synthesis described above. These compounds are suitable as SYK inhibitors and have IC50-values with regard to SYK-inhibition of less than or equal to 1 μmol. Additionally these compounds exhibit a very good SYK-selectivity which means that—whereas SYK is inhibited effectively—other kinases such as Aurora B (AURB), FLT-3 and GSK 3β are not or almost not inhibited at all. Consequently undesired side effects of these effective SYK-inhibitors of the invention are minimized.
  • AURB phosphorylates Ser10 and Ser28 on histone H3, a key event in mitosis and cellular proliferation. Inhibition of AURB therefore has the potential to block cellular proliferation, and could compromise tissues that exhibit a high cellular turnover, such as the intestine or the bone marrow. It is therefore desired to avoid parallel AURB inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds show IC50-values with regard to Aurora B inhibition of more than 1 μM, preferably more than 6 μM, more preferably more than 10 μM, more preferably more than 30 μM, more preferably of more than 45 μM, particularly preferably more than 50 μM. The AURB-IC50/SYK-IC50-ratios of all example compounds are preferably more than 30, more preferably more than 100.
  • FLT-3 is a tyrosine kinase receptor. When an FLT-3 ligand binds to the receptor, the intrinsic tyrosine kinase activity of the receptor is activated, which in turn phosphorylates and activates signal transduction molecules (such as SHC) which in turn propagates the signal in the cell. Signaling through FLT-3 plays a role in cell survival, proliferation, and differentiation and is important for lymphocyte (B cell and T cell) development. It is therefore desired to avoid parallel FLT-3 inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds of the instant invention show IC50-values with regard to FLT-3 inhibition of more than 0,30 μM, preferably more than 1 μM, more preferably more than 10 μM, particularly preferably more than 30 μM. The FLT-3-IC50/SYK-IC50-ratios of all example compounds are preferably more than 10, more preferably more than 30.
  • Glycogen synthase kinase 3 beta (GSK 3β) is a proline-directed serine-threonine kinase that is prominent in the TGF-β and Wnt intracellular signalling pathways. GSK 3β facilitates a number of intracellular signalling pathways including the activation of β-catenin complex. In adults, GSK 3β is involved in cellular proliferation and energy metabolism, whilst in neonates is involved in neuronal cell development and body pattern formation. It is therefore desired to avoid parallel GSK3β inhibition of an effective SYK inhibitor to improve the overall clinical safety profile of the compound. Consequently all example compounds of the invention show IC50-values with regard to GSK 3β inhibition of more than 1 μM, preferably of more than 10 μM.
  • Further it is desirable for an SYK-inhibitor to have certain human liver microsomal stability (corresponding to Cl <60% Qh; % Qh=percentage of liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated.
  • The IC50-values with respect to SYK-inhibition,with respect to Aurora B inhibition,with respect to FLT3-inhibition and with respect to GSKbeta-inhibition as well as the human liver microsomal stablities (CI [% Qh]) for each of the individual example substances are shown in the following Table 1 and were experimentally determined as follows:
  • 5.1 Syk Kinase Test
  • Recombinant human Syk (amino acids 342-635) was expressed as a fusion protein with an N-terminal GST tag, affinity-purified and deep-frozen at a concentration of approx. 50-100 μM in storage buffer (25 mM HEPES pH7.5; 25 mM MgCl2; 5 mM MnCl2; 50 mM KCl; 0.2% BSA; 0.01% CHAPS; 100 μM Na3VO4; 0.5 mM DTT, 10% glycerol) at −80° C. until use.
  • The catalytic activity of the GST-Syk kinase fusion protein was determined using the Kinase Glo® Luminescence Kinase test (Promega; V6712). In this homogeneous test the amount of ATP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ATP still present and thus correlates inversely with the activity of the kinase.
  • Method
  • The test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 1 mM. Serial Dilution is done in 100% DMSO. All further dilutions of the substances were carried out with test buffer (25 mM HEPES pH7.5; 25 mM MgCl2; 5 mM MnCl2; 50 mM KCl; 0.2% HSA; 0.01% CHAPS; 100 μM Na3VO4; 0.5 mM DTT). Dilution steps and concentration range were adapted according to need. 7 μl aliquots of these dilutions were transferred into a 384-well Optiplate (Perkin Elmer, # 6007290). GST-Syk was diluted to 12 nM in the test buffer and 5 μl of this dilution were used in the kinase test (final concentration of Syk=4 nM in a total volume of 15 μl). After 15 minutes incubation at room temperature 3 μl of a mixture of 750 nM ATP and 100 μg/ml poly (L-Glutamic acid L-Tyrosine 4:1), Fluka # 81357) in test buffer were added to each well and the incubation was continued for a further 60 minutes at room temperature.
  • Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.
  • After 60 minutes, 10 μl Kinase-Glo® solution (Promega, Cat. # V6712) (heated to room temperature) were added to each well and incubation was continued for a further 15 minutes. The plates were read in Envision Luminescence Reader (Perkin-Elmer).
  • Data evaluation and Calculation:
  • The output file of the reader is a csv file that contains the well number and measured relative light units (RLU). For data evaluation and calculation, the measurement of the negative control was set as 100% ctrl and the measurement of the positive control was set as 0% ctrl. Based on this values the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics. The IC50 values were calculated from the % ctrl values using Assay Explorer software. Calculation: [y=(a−d)/(1+(x/c)̂b)+d] a=low value, d=high value; x=conc M; c=IC50 M; b=hill; y=% ctrl.
  • 5.2 Aurora B Kinase Test
  • Recombinant human Aurora B (amino acids 1-344, clone number DU1773, Molecular weight 40,2kDa, University of Dundee) was expressed as a fusion protein with an N-terminal His tag, affinity-purified and deep-frozen at a concentration of approx. 0.25-0.5 mg/ml in storage buffer (50 mM Tris-HCl pH 8; 25 mM Na-β-glycerophosphat; 0.1 mM EGTA; 150 mM NaCl; 0.03% Brij-35; 1 mM DTT and 10% glycerol) at −80° C. until use.
  • The activity of the Aurora B kinase protein was determined using the ADP Glo® Luminescence Kinase test (Promega; V9103X). In this homogeneous test the amount of ADP remaining after the kinase reaction is quantified by a luciferin-luciferase reaction using luminescence. The luminescence signal obtained correlates with the amount of ADP still present and thus correlates with the activity of the protein kinase.
  • Method
  • The test compounds were dissolved in 100% DMSO at a concentration of 10 mM and diluted in DMSO to a concentration of 5 mM. Serial Dilution is done in 1:10 steps in 100% DMSO. All further dilutions of the substances were carried out with test buffer (50 mM Hepes, pH 7.5, 10 mM MgCl2, 1 mM EGTA, 60 μM Ultra Pure ATP, 0.01% Brij35, 0.1% BSA, 5 mM β-Glycerophosphate) until a concentration was reached which was 2.5 times above the final test concentration (final concentration of the compounds: 50 μM to 0.005 nM). 4 μl aliquots of these dilutions were transferred into a 384-well Optiplate (Perkin Elmer, # 6007290). His-Aurora B was diluted to 125 nM in the test buffer and 4 μl of this dilution were used in the kinase test (final concentration of Aurora B=50 nM in a total volume of 10 μl). After 15 minutes incubation at room temperature 2 μl of 250 μM substrate ([LRRLSLGLRRLSLGLRRLSLGLRRLSLG]; University of Dundee) in test buffer were added to each well and the incubation was continued for a further 60 minutes at room temperature.
  • Positive controls are the reaction mixtures that contain no test substance; negative controls (blanks) are reaction mixtures that contain no kinase.
  • After 60 minutes, 10 μl ADP-Glo® solution (ADP-Glo Reagent #V912B Promega) (heated to room temperature) were added to each well and incubation was continued for a further 40. minutes. Then 20 μl Kinase detection mix (Detection Buffer #V913B Promega; Kinase Detection Substrate #V914B Promega) were added and incubated for 40 minutes at room temperature. The plates were read in Envision Luminescence Reader (Perkin-Elmer).
  • Data evaluation and calculation:
  • The output file of the reader is a csv file that contains the well number and measured RLU. For data evaluation and calculation, the measurement of the negative control was set as 0% ctrl and the measurement of the positive control was set as 100% ctrl. Based on this values the % value for the measurement of each substance concentration was calculated using an Assay Explorer software (Accelrys). Normally, the % ctrl values calculated are between 0% and 100% values but may also occur outside these limits in individual cases based on variability or compound characteristics. The IC50 values were calculated from the % ctrl values using Assay Explorer software.Calculation: [y=(a−d)/(1+(x/c)̂b)+d], a=low value, d=high value; x=conc M; c=IC50 M; b=hill; y=% ctrl.
  • 5.3 FLT3 Kinase Test
  • FLT3 is obtained from Invitrogen in 50 mM Tris (pH7.5); 100 mM NaCl; 0.05 mM EDTA, 0.05% NP-40, 2 mM DTT; 50%Glycerol # PV3182; Lot 286671; sequence see below). The enzyme is diluted to 720 nM (35 μg/ml) in enzyme dilution buffer and 10 μl aliquots are stored at −80° C.
  • The activity of FLT3 is measured using the Z′-LYTETM assay technology from Invitrogen (# PV3191)
  • Method
  • The assay is performed in 384 black plates from Corning (# 3676) in a final volume of 10 μl by adding 5 μl of kinase peptide mix and 2.5 μl of compound dilution. The reaction is started by addition of 2.5 μl of the 4× ATP solution.
  • Final concentration in assay: FLT3 2 nM, Tyr2 peptide 4 μM, ATP 470 μM (ATP Km for FLT3)
  • Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no kinase. As a further control, the phosphopeptide solution is added to wells without kinase (=100% phosphorylation control). The non inhibited kinase reaction will result in a phosphorylation corresponding to 20%-30% of the phosphorylation control.
  • The reaction is performed for 1 h at room temperature before 5 μl of the development solution is added. After a further incubation for 1 h at room temperature 5 μl of the stop reagent is added. The plates are read on a Flex Station II 384 (Molecular Devices).
  • To control for any potential inhibition of the protease present in the development solution, the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 μM or 10 μM).
  • Data Evaluation and Calculation:
  • The output text file is evaluated in an “MS-Excel - VB -Makro” and “GraphPadPrism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of FLT3 are reported in M. data for the inhibition of the protease are reported in % CTL.
  • 5.4 GSK 3β Kinase-Test
  • Human GSK3beta (expressed and prified from SF21 cells)is obtained from the University Dundee/Scotland (Dr. James Hastie—Dept. of Biochemistry) in 50 mM Tris (pH7.5); 150 mM NaCl; 0.1 mM EGTA, 270 mM Succrose, 0,1% β-mercaptoethanol, 1 mM benzamidine, 0,2 mM PMSF ; sequence see below). The enzyme is diluted to 3,56 μM (168 μg/ml) in enzyme dilution buffer and 6 μl aliquots are stored at −80° C.
  • The activity of GSK3β kinase protein is measured using the Z′-LYTETM assay technology from Invitrogen (# PV3324).
  • Method:
  • The assay is performed in 384 black plates from Corning (# 3676) in a final volume of 10 μl by adding 5 μl of kinase peptide mix and 2.5 μl of compound dilution. The reaction is started by addition of 2.5 μl of the 4× ATP solution.
  • Final concentration in assay: GSK3β 5 nM, Ser/Thr9 peptide 2 μM, ATP 7 μM (ATP Km for GSK3β)
  • Positive controls are reaction mixtures containing no test compound; negative controls (blanks) are reaction mixtures containing no ATP. As a further control, the phosphopeptide solution is added to wells without kinase and without ATP (=100% phosphorylation control). The non inhibited kinase reaction will result in a phosphorylation corresponding to 20%-30% of the phosphorylation control.
  • The reaction is performed 1 h at room temperature. After 1 h 5 μl of the development solution is added. After a further incubation for 1 h at room temperature 5 μl of the stop reagent is added. Finally the plates are read on a Flex Station II 384 (Molecular Devices).
  • To control for any potential inhibition of the protease present in the development solution, the phosphopeptide is incubated with the development solution in the presence of the highest concentration of the test compound (usually 100 μM).
  • Data Evaluation and Calculation:
  • The output text file is evaluated in an “MS-Excel-VB-Makro” and “GraphPadPrism”(Version 5) (GraphPad Software Inc.) is used to calculate the results. Data for the inhibition of GSK3beta are reported in M. data for the inhibition of the protease are reported in % CTL.
  • 5.5 Human Liver Microsomal Stability Test
  • Further it is desirable for an SYK-inhibitor that is sufficiently SYK-specific as described above to have certain human liver microsomal stability (corresponding to Cl<60% Oh; % Oh=percentage of liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated.
  • Method:
  • The metabolic degradation for a specific SYK-inhibitor is performed at 37° C. with pooled human liver microsomes (human liver microsomes are commercially available as “BD UltraPool™” by Corning Life Sciences, Fogostraat 12, 1060 LJ Amsterdam, The Netherlands). The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and the test compound at a final concentration of 1 μM.
  • Following a short preincubation period at 37° C., the reaction is initiated by addition of beta-nicotinamide adenine dinucleotide phosphate in its reduced form (NADPH, 1 mM) and terminated by transfering an aliquot into solvent after different time points. Additionally, the NADPH-independent degradation is monitored in incubations without NADPH, terminated at the last time point.
  • The quenched (terminated) incubations are then pelleted by centrifugation (10000 g, 5 min).
  • An aliquot of the supernatant is assayed by LC-MS/MS for the remaining amount of parent compound. The half-life (t½INVITRO) is determined by the slope of the semilogarithmic plot of the concentration-time profile.
  • Data Evaluation and Calculation:
  • The intrinsic clearance (CL_INTRINSIC) is calculated by considering the amount of protein in the incubation:
  • CL_INTRINSIC [μl/min/mg protein]=(Ln 2/(t½INVITRO [min]*protein content [mg/ml])) *1000
  • The protein content [mg/ml] was determined with the “Bicinchoninic Acid Kit” of Sigma Aldrich (commercially available).
  • The upscaled intrinsic Clearance (CL_UP_INT) is calcuated by considering the liver weight [g liver/kg body weight] and the microsomal recovery [mg protein/g liver]:
  • CL_UP_INT [ml/min/kg] =0.001*CL_INTRINSIC*liver weight*microsomal recovery
      • with microsomal recovery=45 mg protein/g liver
      • with liver weight=25.7 g liver/kg body weight
  • The percent hepatic blood flow (% Qh) is finally calculated by considering the human liver blood flow Q [ml/min/kg]:
  • % Qh [%]=((Q*CL_UP_INT)/(Q+CL_UP_INT)/Q)*100
      • with liver blood flow (Q)=20.7 ml/min/kg.
  • TABLE 1
    SYK- AURB- FLT3-
    inhibiton inhibition inhibition
    Example IC50-value IC50-value IC50-value
    No. Structure [μM] [μM] [μM]
     1
    Figure US20180072744A1-20180315-C00117
    0.0160 >50 3.99
     2
    Figure US20180072744A1-20180315-C00118
    0.0144 >50 14.84
     3
    Figure US20180072744A1-20180315-C00119
    0.0466 >50 17.45
     4 not defined not defined not defined not defined
     5 not defined not defined not defined not defined
     6 not defined not defined not defined not defined
     7
    Figure US20180072744A1-20180315-C00120
    0.0147 37.30 5.77
     8
    Figure US20180072744A1-20180315-C00121
    0.0274 >50 13.50
     9
    Figure US20180072744A1-20180315-C00122
    0.0424 >50 3.67
    10
    Figure US20180072744A1-20180315-C00123
    0.0218 36.00 8.03
    11
    Figure US20180072744A1-20180315-C00124
    0.9352 >50 37.38
    12
    Figure US20180072744A1-20180315-C00125
    0.0123 34.09 8.33
    13
    Figure US20180072744A1-20180315-C00126
    0.8310 >50 >50
    14
    Figure US20180072744A1-20180315-C00127
    0.0421 >50 40.80
    15
    Figure US20180072744A1-20180315-C00128
    0.0187 33.40 10.08
    16
    Figure US20180072744A1-20180315-C00129
    0.0065 >50 10.60
    17
    Figure US20180072744A1-20180315-C00130
    0.0316 >50 1.61
    18
    Figure US20180072744A1-20180315-C00131
    0.0046 >50 13.40
    19 not defined not defined not defined not defined
    20 not defined not defined not defined not defined
    21 not defined not defined not defined not defined
    22
    Figure US20180072744A1-20180315-C00132
    0.0128 46.90 1.17
    23 not defined not defined not defined not defined
    24 not defined not defined not defined not defined
    25 not defined not defined not defined not defined
    26
    Figure US20180072744A1-20180315-C00133
    0.0258 >50 0.95
    27
    Figure US20180072744A1-20180315-C00134
    0.1382 >50 26.64
    28
    Figure US20180072744A1-20180315-C00135
    0.0335 >50 3.69
    29
    Figure US20180072744A1-20180315-C00136
    0.0492 >50 20.56
    30 not defined not defined not defined not defined
    31 not defined not defined not defined not defined
    32
    Figure US20180072744A1-20180315-C00137
    0.0272 >50 2.35
    33
    Figure US20180072744A1-20180315-C00138
    0.0245 >50 3.88
    34 not defined not defined not defined not defined
    35
    Figure US20180072744A1-20180315-C00139
    0.0345 >50 19.66
    36
    Figure US20180072744A1-20180315-C00140
    0.0172 35.22 2.39
    37 not defined not defined not defined not defined
    38
    Figure US20180072744A1-20180315-C00141
    0.0609 47.10 11.35
    39 not defined not defined not defined not defined
    40
    Figure US20180072744A1-20180315-C00142
    0.0343 31.43 8.95
    41
    Figure US20180072744A1-20180315-C00143
    0.1638 >50 42.82
    42
    Figure US20180072744A1-20180315-C00144
    0.0167 >50 0.55
    43
    Figure US20180072744A1-20180315-C00145
    0.0649 >50 15.05
    44 not defined not defined not defined not defined
    45
    Figure US20180072744A1-20180315-C00146
    0.0147 45.21 2.04
    46 not defined not defined not defined not defined
    47
    Figure US20180072744A1-20180315-C00147
    0.0067 25.97 4.77
    48
    Figure US20180072744A1-20180315-C00148
    0.0556 >50 16.86
    49
    Figure US20180072744A1-20180315-C00149
    0.0148 >50 2.78
    50
    Figure US20180072744A1-20180315-C00150
    0.0554 47.95 9.52
    51
    Figure US20180072744A1-20180315-C00151
    0.3660 49.83 20.87
    52
    Figure US20180072744A1-20180315-C00152
    0.0413 >50 >50
    53
    Figure US20180072744A1-20180315-C00153
    0.0139 >50 34.12
    54
    Figure US20180072744A1-20180315-C00154
    0.0892 >50 16.00
    55
    Figure US20180072744A1-20180315-C00155
    0.0436 >50 3.07
    56
    Figure US20180072744A1-20180315-C00156
    0.0939 >50 8.94
    57
    Figure US20180072744A1-20180315-C00157
    0.0901 >50 31.97
    58
    Figure US20180072744A1-20180315-C00158
    0.0366 >50 17.10
    59
    Figure US20180072744A1-20180315-C00159
    0.1437 >50 46.20
    60
    Figure US20180072744A1-20180315-C00160
    0.1448 34.70 9.75
    61
    Figure US20180072744A1-20180315-C00161
    0.4749 >50 48.10
    62
    Figure US20180072744A1-20180315-C00162
    0.0193 >50 4.78
    63
    Figure US20180072744A1-20180315-C00163
    0.0473 >50 13.24
    64
    Figure US20180072744A1-20180315-C00164
    0.0258 >50 21.62
    65
    Figure US20180072744A1-20180315-C00165
    0.0211 >50 14.85
    66
    Figure US20180072744A1-20180315-C00166
    0.0385 >50 4.44
    67
    Figure US20180072744A1-20180315-C00167
    0.0152 >50 1.68
    68
    Figure US20180072744A1-20180315-C00168
    0.0652 >50 22.43
    69
    Figure US20180072744A1-20180315-C00169
    0.1973 >50 45.15
    70
    Figure US20180072744A1-20180315-C00170
    0.0345 >50 16.99
    71
    Figure US20180072744A1-20180315-C00171
    0.0150 >50 7.65
    72
    Figure US20180072744A1-20180315-C00172
    0.0526 >50 19.59
    73
    Figure US20180072744A1-20180315-C00173
    0.0224 49.10 4.17
    74
    Figure US20180072744A1-20180315-C00174
    0.0322 >50 11.15
    75
    Figure US20180072744A1-20180315-C00175
    0.0998 8.54 1.59
    76
    Figure US20180072744A1-20180315-C00176
    0.0583 46.20 14.80
    77
    Figure US20180072744A1-20180315-C00177
    0.0498 43.38 3.75
    78
    Figure US20180072744A1-20180315-C00178
    0.0359 >50 22.04
    79
    Figure US20180072744A1-20180315-C00179
    0.0364 >50 26.74
    80
    Figure US20180072744A1-20180315-C00180
    0.0224 39.74 1.11
    81
    Figure US20180072744A1-20180315-C00181
    0.0240 48.40 5.24
    82
    Figure US20180072744A1-20180315-C00182
    0.0483 >50 7.78
    83
    Figure US20180072744A1-20180315-C00183
    0.7324 >50 25.60
    84 not defined not defined not defined not defined
  • 6. Comparison of SYK-inhibitory Capacity and of SYK-selectivity of the Compounds of the Invention Compared to Selected Compounds of WO 2013/014060 and of WO 2011/092128
  • To have an efficient SYK-inhibitory capacity is not the only important aspect which a SYK-inhibitor to be used as a medicament to treat SYK-related diseases must show. Similarly important like the low IC50-value with regard to SYK-inhibition (IC50 (SYK)≦1 μM) is that the candidate compound does not show undesired inhibitory effects on other kinases which could lead to unwanted or even dangerous side effects. Examples of such other kinases that should not be inhibited by the candidate SYK-inhibitor are AURB, FLT3 and GSKbeta.
  • Consequently the IC50-values with regard to SYK, AURB, FLT3 and GSKbeta for structurally close compounds disclosed in WO 2013/014060 and of WO 2011/092128 have been experimentally determined according to the same assays as described in chapter 5. The measured IC50-values with regard to SYK, AURB, FLT3 and GSKbeta of these structurally closest prior art compounds are in the following tables 2a to 6c compared to the respective previously determined IC50-values of a representative selection of compounds of the invention (same assay conditions).
  • Further it is desirable for an SYK-inhibitor that is sufficiently SYK-specific as described above to have certain human liver microsomal stability (corresponding to Cl <60% Qh; Qh=liver blood flow). Otherwise it will be difficult to reach an adequate plasma level of the SYK-inhibitor in the patient to be treated. Consequently also the Cl-values for structurally close compounds disclosed in WO 2013/014060 and in WO 2011/092128 have been experimentally determined according to the same human liver microsomal-test as described in chapter 5. An experimentally determined Cl-value of more than 60% Oh is regarded to be inacceptable in order to reach an adequate plasma level of the respective SYK-inhibitor in the patient to be treated.
  • 6.1 Comparisons for Compounds with Alkyl-substituted Pyrazole Structures
  • Whereas all compounds of the invention (see Table 2a) and of WO 2013/014060 (see Table 2b) with alkyl-substituted pyrazole structures have suitable IC50 (SYK)-values of smaller than 1 μM, only the compounds of the invention (see Table 2a) have IC50-values with regard to AURB of more than 50 μM (compared to IC50-values (AURB) of significantly below 5 μM for the compounds of WO 2013/014060 in Table 2b). Also the IC50-values with respect to FLT3 are larger for the compounds of the invention (Table 2a) compared to the compounds of WO 2013/014060 (see Table 2b). Consequently the compounds of the invention with alkyl-substituted pyrazole structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 2b)), but also do not have unwanted inhibitory effects on other kinases such as AURB, FLT3 and GSK3beta (unlike the compounds of WO 2013/014060 (see Table 2b)). The compounds of the invention with alkyl-substituted pyrazole structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • TABLE 2a
    compounds of the invention with alkyl-substituted pyrazole structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
     2
    Figure US20180072744A1-20180315-C00184
    0.0144 >50 14.8 >10 <23
    18
    Figure US20180072744A1-20180315-C00185
    0.0046 >50 13.4 >10 <23
    n.d. = not detected
  • TABLE 2b
    Compounds of WO 2013/014060 with alkyl-substituted pyrazole structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    112
    Figure US20180072744A1-20180315-C00186
    0.006  2.96 1.25  >10 <23
    114
    Figure US20180072744A1-20180315-C00187
    0.0002 2.54 0.049 >10 26
    115
    Figure US20180072744A1-20180315-C00188
    0.0002 0.021 0.041 >10 <23
  • 6.2 Comparisons for Compounds with Optionally Substituted Bicyclic Heteroaryl Structures
  • Whereas all compounds of the invention (see Table 3a) and of WO 2013/014060 (see Table 3b) with optionally substituted bicyclic heteroaryl structures have suitable IC50 (SYK)-values of smaller than 1 μM, only the compounds of the invention (see Table 3a) have IC50-values with regard to AURB of more than 30 μM, most of them even of more than 50 μM (compared to IC50-values (AURB) of mostly below 1μM for the compounds of WO 2013/014060 in Table 3b). Consequently the compounds of the invention with optionally substituted bicyclic heteroaryl structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 3b)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2013/014060 (see Table 3b)).
  • The compounds of the invention with optionally substituted bicyclic heteroaryl structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • TABLE 3a
    Compounds of the invention with optionally substituted bicyclic heteroaryl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    62
    Figure US20180072744A1-20180315-C00189
    0.0193 >50 4.78 >10 <23
    72
    Figure US20180072744A1-20180315-C00190
    0.0526 >50 19.6 1.45 <23
    36
    Figure US20180072744A1-20180315-C00191
    0.0172 35.22 2.39 >10 <23
    28
    Figure US20180072744A1-20180315-C00192
    0.0335 >50 3.69 >10 <23
    n.d. = not detected
  • TABLE 3b
    Compounds of WO 2013/014060 with optionally substituted bicyclic heteroaryl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
     29
    Figure US20180072744A1-20180315-C00193
    0.002 0.293 0.315 >10 44
     41
    Figure US20180072744A1-20180315-C00194
    0.001 0.308 0.505 >10 37
     42
    Figure US20180072744A1-20180315-C00195
    0.002 0.630 0.618 >10 <23
     61
    Figure US20180072744A1-20180315-C00196
    0.002 0.250 0.411 >10 <23
     81
    Figure US20180072744A1-20180315-C00197
    0.003 0.949 0.514 >10 <23
     85
    Figure US20180072744A1-20180315-C00198
    0.002 0.460 0.321 6.82 49
     94
    Figure US20180072744A1-20180315-C00199
    0.001 0.298 0.267 n.d. n.d.
    105
    Figure US20180072744A1-20180315-C00200
    0.001 0.479 0.309 >10 64
    107
    Figure US20180072744A1-20180315-C00201
    0.009 1.82  1.76  >10 38
    109
    Figure US20180072744A1-20180315-C00202
    0.001 0.260 0.330 >10 41
  • 6.3 Comparisons for Compounds with Alkoxy-substituted Phenyl Structures
  • All compounds of the invention (see Table 4a), of WO 2011/092128 (see Table 4b) and of WO 2013/014060 (see Table 4c) with alkoxy-substituted phenyl structures have suitable IC50 (SYK)-values of smaller than 1 μM. However, whereas the compounds of the invention (see Table 4a) have IC50-values with regard to AURB of more than 45 μM, often even of more than 50 μM, the IC50-values (AURB) of WO 2011/092128 (see Table 4b) and of WO 2013/014060 (see Table 4c) are mostly ≦5μM, often even below 1 μM and consequently the compounds of the invention with alkoxy-substituted phenyl structures have less unwanted inhibitory effects on other kinases such as AURB compared to most of the compounds of WO 2011/092128 (Table 4b) and of WO 2013/014060 (see Table 4c).
  • Only example 8 of WO 2011/092128 (see Table 4b) also shows an IC50-value (with regard to AURB) of more than 50 μM which seems comparable to the compounds of the instant invention, however example 8 of WO 2011/092128 (see Table 4b) shows with Cl=77% Qh a human liver microsomal stability of significantly lower than 60% Qh that would lead to an inadequately low plasma level of the SYK-inhibitor in a patient to be treated. Also example 2 of WO2013/014060 (see Table 4c) shows with IC50 (AURB)=16.9 μM a slightly larger IC50-value with respect to AURB than the other prior art compounds, however also here the human liver microsomal stability is with Cl=81% Qh inadequate. In contrast to that the compounds of the invention with alkoxy-substituted phenyl structures (see Table 4a) show acceptable Cl-values of lower than 60% Qh (all Cl=<23)% Oh).
  • The compounds of the invention with alkoxy-substituted phenyl structures therefore show a significantly improved SYK-selectivity and additionally an acceptable human liver microsomal stability compared to all of the structurally closest compounds disclosed in WO2013/014060 and in WO2011/092128.
  • TABLE 4a
    Compounds of the invention with alkoxy-substituted phenyl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl[% Qh]
     1
    Figure US20180072744A1-20180315-C00203
    0.0160 >50 3.99 >10 <23
     8
    Figure US20180072744A1-20180315-C00204
    0.0274 >50 13.50 >10 <23
    52
    Figure US20180072744A1-20180315-C00205
    0.0413 >50 >50 >10 <23
    81
    Figure US20180072744A1-20180315-C00206
    0.0240 48.40 5.24 >10 <23
    n.d. = not determined
  • TABLE 4b
    Compounds of WO 2011/092128 with alkoxy-substituted phenyl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    35
    Figure US20180072744A1-20180315-C00207
    0.001 5.21 1.18 >10 62
     7
    Figure US20180072744A1-20180315-C00208
    0.002 2.33 0.452 >10 79
     8
    Figure US20180072744A1-20180315-C00209
    0.012 >50 >50 >10 77
     4
    Figure US20180072744A1-20180315-C00210
    0.003 1.85 0.633 >10 33
     1
    Figure US20180072744A1-20180315-C00211
    0.009 5.09 1.53 >10 n.d.
    n.d. = not determined
  • TABLE 4c
    Compounds of 2013/014060 with alkoxy-substituted phenyl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
     2
    Figure US20180072744A1-20180315-C00212
    0.007 16.9 7.39  >10 81
     8
    Figure US20180072744A1-20180315-C00213
    0.001 0.643 0.369 >10 58
     10
    Figure US20180072744A1-20180315-C00214
    0.001 0.271 0.312 >10 57
     11
    Figure US20180072744A1-20180315-C00215
    0.002 0.752 0.738 >10 24
    102
    Figure US20180072744A1-20180315-C00216
    0.013 2.63 2.27  >10 54
  • 6.4 Comparisons for Compounds with Heterocycle-substituted or Heterocycle-annulated Phenyl Structures
  • Whereas all compounds of the invention (see Table 5a), of WO 2013/014060 (see Table 5b) with heterocycle-substituted or heterocycle-annulated phenyl structures have suitable IC50 (SYK)-values of smaller than 1 μM, only the compounds of the invention (see Table 5a) have IC50-values with regard to AURB of mostly more than 45 μM, mostly of more than 50 μM (compared to IC50-values (AURB) of below 1 μM for the compounds of WO 2013/014060 in Table 5b). Consequently the compounds of the invention with heterocycle-substituted or heterocycle-annulated phenyl structures are not only efficient SYK-inhibitors (like the compounds of WO 2013/014060 (see Table 5b)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2013/014060 (see Table 5b)).
  • The compounds of the invention with heterocycle-substituted or heterocycle-annulated phenyl structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO2013/014060.
  • TABLE 5a
    Compounds of the invention with heterocycle-substituted or heterocycle-annulated phenyl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    22
    Figure US20180072744A1-20180315-C00217
    0.0128 46.90 1.17 >10 25
    26
    Figure US20180072744A1-20180315-C00218
    0.0258 >50 0.95 >10 <23
    42
    Figure US20180072744A1-20180315-C00219
    0.0167 >50 0.55 >10 <23
    67
    Figure US20180072744A1-20180315-C00220
    0.0152 >50 1.68 >10 34
  • TABLE 5b
    Compounds of WO 2013/014060 with heterocycle-substituted or heterocycle-annulated phenyl structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    43
    Figure US20180072744A1-20180315-C00221
    0.001 0.144 0.157 n.d. <23
    76
    Figure US20180072744A1-20180315-C00222
    0.002 0.075 0.169 >10 <23
    101
    Figure US20180072744A1-20180315-C00223
    0.002 1.05  0.140 >10 <23
  • 6.5 Comparisons for Compounds with Optionally Substituted Pyridine Structures
  • Whereas all compounds of the invention (see Table 6a), of WO 2011/092128 (see Table 6b) and of WO 2013/014060 (see Table 6c) with optionally substituted pyridine structures have suitable IC50 (SYK)-values of smaller than 1 μM, only the compounds of the invention (see Table 6a) have IC50-values with regard to AURB of more than 49 μM, mostly even of more than 50 μM (compared to IC50-values (AURB) of around 1 μM for the compounds of WO 2011/092128 in Table 6b and of WO 2013/014060 in Table 6c). Consequently the compounds of the invention with optionally substituted pyridine structures are not only efficient SYK-inhibitors (like the compounds of WO 2011/092128 (Table 6b) and of WO 2013/014060 (see Table 6c)), but also do not have unwanted inhibitory effects on other kinases such as AURB (unlike the compounds of WO 2011/092128 (Table 6b) and of WO 2013/014060 (see Table 6c)).
  • The compounds of the invention with optionally substituted pyridine structures therefore show a significantly improved SYK-selectivity compared to the structurally closest compounds disclosed in WO 2011/092128 or in WO2013/014060.
  • TABLE 6a
    Compounds of the invention with optionally substituted pyridine structures
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    59
    Figure US20180072744A1-20180315-C00224
    0.1437 >50 46.20 >10 <23
    61
    Figure US20180072744A1-20180315-C00225
    0.4749 >50 48.10 >10 <23
    51
    Figure US20180072744A1-20180315-C00226
    0.3660 49.83 20.87 >10 <23
    41
    Figure US20180072744A1-20180315-C00227
    0.1638 >50 42.82 >10 <23
    27
    Figure US20180072744A1-20180315-C00228
    0.1382 >50 26.64 >10 <23
    n.d. = not determined
  • TABLE 6b
    Compound of WO 2011/092128 with optionally substituted pyridine structure
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
     3
    Figure US20180072744A1-20180315-C00229
    0.0122 1.04  1.82  >10 24
    45
    Figure US20180072744A1-20180315-C00230
    0.0002 0.086 0.100 >10 53
  • TABLE 6c
    Compound of WO 2013/014060 with optionally substituted pyridine structure
    IC50 in μM IC50 in μM IC50 in μM IC50 in μM microsomal
    (SYK- (AURB- (FLT3- (GSK3beta- stability
    Ex. No. Structure Inhibition) Inhibition) Inhibition) Inhibition) Cl [% Qh]
    6
    Figure US20180072744A1-20180315-C00231
    0.004 1.03 0.640 >1 24
  • 6. INDICATIONS
  • As has been found, the compounds of formula 1 are characterised by their range of applications in the therapeutic field. Particular mention should be made of those applications for which the compounds of formula 1 according to the invention are preferably used on the basis of their pharmaceutical activity as Syk-inhibitors. Examples include respiratory complaints, allergic diseases, osteoporosis, gastrointestinal diseases or complaints, immune or autoimmune diseases, allergic diseases, inflammatory diseases, e.g. inflammatory diseases of the joints, skin and eyes and diseases of the peripheral or central nervous system.
  • Particular mention should be made of the prevention and treatment of respiratory tract and pulmonary diseases which are accompanied by increased mucus production, inflammation and/or obstructive diseases of the airways. Examples of these include asthma, paediatric asthma, ARDS (Adult Respiratory Distress Syndrome), acute, allergic or chronic bronchitis, autoimmune haemolytic anemia, chronic obstructive bronchitis (COPD) (including the treatment of Rhinovirus-induced exacerbations), coughs, allergic rhinitis or sinusitis, allergic rhinoconjunctivitis, chronic rhinitis or sinusitis, alveolitis, farmers' lung, hyperreactive airways, infectious bronchitis or pneumonitis, bronchiectasis, pulmonary arterial hypertension, pulmonary fibrosis, bronchial oedema, pulmonary oedema, pneumonia or interstitial pneumonia triggered by various causes such as aspiration, inhalation of toxic gases or bronchitis, pneumonia or interstitial pneumonia triggered by cardiac insufficiency, radiation, chemotherapy, cystic fibrosis or mucoviscidosis, alpha 1-antitrypsin deficiency.
  • The compounds according to the invention are preferably also suitable for the treatment of allergic diseases such as for example allergic rhinitis, allergic rhinoconjunctivitis, allergic conjunctivitis, and contact dermatitis, urticaria/angiooedema and allergic dermatitis.
  • Mention should also preferably be made of the treatment of inflammatory diseases of the gastrointestinal tract. Examples of these are Crohn's disease and ulcerative colitis.
  • The compounds according to the invention are preferably also suitable for the treatment of inflammatory diseases of the joints, of the blood vessels and of the kidney or inflammatory diseases of the skin and eyes. Examples of these are rheumatoid arthritis, antibody-based glomerulonephritis, psoriasis, Kawasaki syndrome, coeliac disease (sprue), arteriosclerosis and Wegener's granulomatosis, osteoarthritis, systemic scleroderma, ankylosing spondylitis.
  • The compounds according to the invention are preferably also suitable for the treatment of autoimmune diseases. Examples of these are hepatitis (autoimmune-based), lupus erythematodes, lupus nephritis, systemic lupus, Systemic lupus erythematosus,discoid lupus, cutaneous lupus erythematosus (acute, subacute, chronic), anti-phospholipid syndrome, Berger's disease, Evans's syndrome, immunohaemolytic anaemia, ITP (idiopathic thrombocytopenic purpura; adult, neonatal and paediatric), myasthenia gravis, Sjogren's syndrome, sclerodermy, Bullous pemphigoid and Pemphigus vulgaris.
  • The compounds according to the invention are preferably also suitable for the treatment of B-cell lymphomas, like chronic lymphocytic leukaemia and non-Hodgkin's lymphomas or T cell lymphomas.
  • The compounds according to the invention are preferably also suitable for the treatment of Graft-versus -host disease.
  • Mention may preferably also be made of the prevention and treatment of diseases of the peripheral or central nervous system. Examples of these are acute and chronic multiple sclerosis or non-familial lateral sclerosis.
  • Mention may preferably also be made of the prevention and treatment of osteoporotic diseases such as for example disease-associated osteopenia, osteoporosis and osteolytic diseases.
  • The present invention relates particularly preferably to the use of compounds of formula 1 for preparing a pharmaceutical composition for the treatment of diseases selected from among asthma, COPD, allergic rhinitis, Adult Respiratory Distress Syndrome, bronchitis, allergic dermatitis, contact dermatitis, ITP, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, and allergic rhinoconjunctivitis.
  • Most preferably, the compounds of formula 1 may be used for the treatment of a disease selected from among asthma, allergic rhinitis, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, allergic dermatitis and COPD.
  • 7. COMBINATIONS
  • The compounds of formula 1 may be used on their own or in conjunction with other active substances of formula 1 according to the invention. The compounds of formula 1 may optionally also be used in conjunction with other pharmacologically active substances. Preferably the active substances used here may be selected for example from among the betamimetics, anticholinergics, corticosteroids, PDE4-inhibitors, LTD4-antagonists, EGFR-inhibitors, MRP4-inhibitors, dopamine agonists, H1-antihistamines, PAF-antagonists, iNos-inhibitos, HMG-CoA reductase inhibitors (statins), PI3-kinase-inhibitors, CCR3-antagonists, CCR2-antagonists, CCR1-antagonists, IKK2-inhibitors, A2a agonists, alpha-4-integrin-inhibitors, CRTH2-antagonists, histamine 1, combined H1/H3-antagonists, p38 kinase inhibitors, methylxanthines, ENaC-inhibitors, CXCR1-antagonists, CXCR2-antagonists, ICE-inhibitors, LTB4-antagonists, 5-LO antagonists, FLAP-antagonists. LTB4-antagonists; cromoglycine, dissociated glucocorticoid mimetics, immunesuppressive agents, cytostatica, non-steroidal anti-inflammatory drugs (NSAIDs), chloroquine, hydroxychloroquine, anti-TNF-antibodies, anti-GM-CSF antibodies, anti-CD46-antibodies, anti-IL-1- antibodies, anti-IL-2-antibodies, anti-IL-4- antibodies, anti-IL-5-antibodies, anti-IL6 antibodies, anti-IL6 receptor antibodies, anti-IL-13- antibodies, anti-IL_18 antibodies, anti-CD30 L antibodies, anti-Ox40L-antibodies, anti-IL-4/IL-13-antibodies, anti-IL-23 (p19) antibodies, anti-IL-12/IL-23 (p40) antibodies, anti-CD3 antibodies, anti-CD4 antibodies, anti-CD154 antibodies, CD89 antibodies, anti-IL-2 receptor/CD25 antibodies, anti-CD22 antibodies, anti-interferon antibodies, anti-ICOS antibodies, anti-ICOS antibodies, anti-CD20 antibodies, anti-CD40 antibodies, anti-BAFF/BLyS antibodies, anti-CD18 antibodies, anti-CD62L antibodies, anti-CD147 antibodies, anti-integrin antibodies, agents interfering with LFA-1, IL-36 pathway modulators, M-CSF/c-fms antagonists, CTLA-4 fusions, mTor modulators,Toll like receptors 7 inhibitors (TLR7 inhibitor),Toll like receptor 9 inhibitors (TLR9 inhibitors), T cell-costimulatory modulators such as CTLA-4 fusions,JAK inhibitors, IRF modulators,CX3 chemokine receptor antagonists (CX3CR1 antagonists), IRAK inhibitors (in particular IRAK1- and IRAK4-inhibitors), Sphingosine-1-phosphate modulators (S1 P pathway modulators),
  • or double or triple combinations thereof, such as for example combinations of one, two or three compounds selected from among the
      • Syk-inhibitors of formula 1, betamimetics, corticosteroids, EGFR-inhibitors and PDE4-antagonists,
      • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids, EGFR-inhibitors and PDE4-antagonists,
      • Syk-inhibitors of formula 1, PDE4-inhibitors, corticosteroids and EGFR-inhibitors,
      • Syk-inhibitors of formula 1, EGFR-inhibitors and PDE4-inhibitors,
      • Syk-inhibitors of formula 1 and EGFR-inhibitors,
      • Syk-inhibitors of formula 1, betamimetics and anticholinergics
      • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids and PDE4-inhibitors,
      • Syk-inhibitors of formula 1, anticholinergics, betamimetics, corticosteroids, iNOS inhibitors, HMG-CoA reductase inhibitors.
  • Combinations of three active substances each taken from one of the above-mentioned categories of compounds are also an object of the invention.
  • Suitable betamimetics used are preferably compounds selected from among arformoterol, carmoterol, formoterol, indacaterol, salmeterol, albuterole, bambuterol, bitolterol, broxaterol, carbuterol, clenbuterol, fenoterol, hexoprenalin, ibuterol, isoetharin, isoprenalin, levosalbutamol, mabuterol, meluadrin, metaproterenol, milveterol, orciprenalin, pirbuterol, procaterol, reproterol, rimiterol, ritodrin, salmefamol, soterenol, sulphonterol, terbutalin, tiaramide, tolubuterol, zintero1,6-Hydroxy-8-{1-hydroxy-2-[2-(4-methoxy-phenyl)-1,1-dimethyl-ethylamino]-ethyl}-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(2,4-Difluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(3,5-Difluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[2-(4-Ethoxy-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo [1,4]oxazine-3-one; 8-{2-[2-(4-Fluor-phenyl)-1,1-dimethyl-ethylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; N-(5-{2-[3-(4,4-Diethyl-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; N-(5-{2-[3-(4,4-Diethyl-6-fluoro-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; N-(5-{2-[3-(4,4-Diethyl-6-methoxy-2-oxo-4H-benzo[d][1,3]oxazine-1-yl)-1,1-dimethyl-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; N-(5-{2-[1,1-Dimethyl-3-(2-oxo-4,4-dipropyl-4H-benzo[d][1,3]oxazine-1-yl)-propylamino]-1-hydroxy-ethyl}-2-hydroxy-phenyl)-methansulfonamide; 8-{2-[1,1-Dimethyl-3-(2-oxo-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(6-methyl-2-oxo-2,3-dihydro-benzoimidazole-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(2-oxo-5-trifluormethyl-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; 8-{2-[1,1-Dimethyl-3-(3-methyl-2-oxo-2,3-dihydro-benzoimidazol-1-yl)-propylamino]-1-hydroxy-ethyl}-6-hydroxy-4H-benzo[1,4]oxazine-3-one; N-[2-Hydroxy-5-((1R)-1-hydroxy-2-{2-[4-(2-hydroxy-2-phenyl-ethylamino)-phenyl]-ethylamino}-ethyl)-phenyl]-formamide; 8-Hydroxy-5-((1R)-1-hydroxy-2-{2-[4-(6-methoxy-biphenyl-3-ylamino)-phenyl]-ethylamino}-ethyl)-1H-quinoline-2-one; 8-Hydroxy-5-[(1R)-1-hydroxy-2-(6-phenethylamino-hexylamino)-ethyl]-1H-quinoline-2-one; 5-[(1R)-2-(2-{4-[4-(2-Amino-2-methyl-propoxy)-phenylamino]-phenyl}-ethylamino)-1-hydroxy-ethyl]-8-hydroxy-1H-quinoline-2-one; [3-(4-{6-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-hexyloxy}-butyl)-5-methyl-phenyl]-urea; 4-((1R)-2-{6-[2-(2,6-Dichlor-benzyloxy)-ethoxy]-hexylamino}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol; 3-(4-{6-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-hexyloxy}-butyl)-benzenesulfonamide; 3-(3-{7-[(2R)-2-Hydroxy-2-(4-hydroxy-3-hydroxymethyl-phenyl)-ethylamino]-heptyloxy}-propyl)-benzenesulfonamide; 4-((1R)-2-{6-[4-(3-Cyclopentanesulfonyl-phenyl)-butoxyFhexylamino}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol, 4-(2-{6-[2-(2,6-dichloro-benzyloxy)-ethoxy]-hexylamino}-1-hydroxy-ethyl)-2-hydroxymethyl-phenol; Vilanterol; N-1-Adamantanyl-2-{3-[(2R)-2-({(2R)-2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]ethyl}amino)propyl]phenyl}acetamide; 2-(3-{2-[2-hydroxy-3-methanesulfonylamino-phenyl)-ethylamino]-propyl}-phenyl)-N-[4-(4-hydroxy-phenyl)-2-vinyl-penta-2,4-dienyl]-acetamide; (1R)-5-{2-[6-(2,2-Difluor-2-phenyl-ethoxy)-hexylamino]-1-hydroxy-ethyl}-8-hydroxy-1H-quinoline-2-one; (R,S)-4-(2-{[6-(2,2-Difluor-4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[4,4-Difluor-6-(4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2- (hydroxymethyl)phenol; (R,S)-4-(2-{[6-(4,4-Difluor-4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-5-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxy-ethyl)-8-hydroxyquinoline-2(1H)-one; (R,S)[2-({6-[2,2-Difluor-2-(3-methylphenyl)ethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; 4-(1R)-2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxyethyl)-2-(hydroxymethyl)phenol; (R,S)-2-(Hydroxymethyl)-4-(1-hydroxy-2-{[4,4,5]5-tetrafluor-6-(3-phenylpropoxy)-hexyl]amino}ethyl)phenol; (R,S)-[5-(2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxy-ethyl)-2-hydroxyphenyl]formamide; (R,S)-4-[2-({6-[2-(3-Bromophenyl)-2,2-difluoroethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; (R,S)-N-[3-(1,1-Difluor-2-{[6-({2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl)phenyl]-ethyl}amino)hexyl]oxy}ethyl)phenyl]-urea; 3-[3-(1,1-Difluor-2-{[6-({2-hydroxy-2-[4-hydroxy-3-(hydroxymethyl) phenyl]ethyl}amino)hexyl]oxy}ethyl)phenyl]imidazolidine-2,4-dione; (R,S)-4-[2-({6-[2,2-Difluor-2-(3-methoxyphenyl)ethoxy]hexyl}amino)-1-hydroxyethyl]-2-(hydroxymethyl)phenol; 5-((1R)-2-{[6-(2,2-Difluor-2-phenylethoxy)hexyl]amino}-1-hydroxyethyl)-8-hydroxyquinoline-2(1H)-one; 4-((1R)-2-{[4,4-Difluor-6-(4-phenylbutoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(3,3-Difluor-3-phenylpropoxy)hexyl]amino}-1-hydroxy-ethyl)-2-(hydroxymethyl)phenol; (R,S)-(2-{[6-(2,2-Difluor-2-phenylethoxy)-4,4-difluorohexyl]amino}-1-hydroxyethyl)-2-(hydroxymethyl)phenol; (R,S)-4-(2-{[6-(2,2-Difluor-3-phenylpropoxy)hexyl]amino}-1-hydroxy ethyl)-2-(hydroxymethyl)phenol; 3-[2-(3-Chlor-phenyl)-ethoxy]-N-(2-diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-propionamide; N-(2-Diethylamino-ethyl-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-3-(2-naphthalen-1-yl-ethoxy)-propionamide; 7-[2-(2-{3-[2-(2-Chlor-phenyl)-ethylamino]-propylsulfanyl}-ethylamino)-1-hydroxy-ethyl]-4-hydroxy-3H-benzothiazol-2-one, optionally in the form of the racemates, enantiomers, diastereomers and optionally in the form of the pharmacologically acceptable acid addition salts, solvates or hydrates thereof.
  • According to the invention the acid addition salts of the betamimetics are preferably selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably the hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate. Of the above-mentioned acid addition salts the salts of hydrochloric acid, methanesulphonic acid, benzoic acid and acetic acid are particularly preferred according to the invention.
  • The anticholinergics used are preferably compounds selected from among tiotropium salts, particularly the bromide salt, oxitropium salts, particularly the bromide salt, flutropium salts, particularly the bromide salt, ipratropium salts, particularly the bromide salt, Aclidinium salts, particularly the bromide salt, glycopyrronium salts, particularly the bromide salt, trospium salts, particularly the chloride salt, tolterodin, (3R)-1-Phenethyl-3-(9H-xanthene-9-carbonyloxy)-1-azoniabicyclo[2.2.2]octan-salts; 2,2-Diphenyl propionic acid tropenole ester-methobromide; 2,2-Diphenyl propionic acid scopine ester-methobromide; 2-Fluor-2,2-Diphenyl acetic acid scopine ester-methobromide; 2-Fluor-2,2-Diphenyl acetic acid tropenole ester-methobromide; 3,3′,4,4′-Tetrafluor benzilic acid tropenole ester-methobromide; 3,3′,4,4′-Tetrafluor benzilic acid scopine ester-methobromide; 4,4′-Difluor benzilic acid tropenole ester-methobromide; 4,4′-Difluor benzilic acid scopine ester-methobromide; 3,3′-Difluor benzilic acid tropenole ester-methobromide; 3,3′-Difluor benzilic acid scopine ester-methobromide; 9-Hydroxy-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Fluor-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxy-fluorene-9-carboxylic acid scopine ester-methobromide; 9-Fluor-fluorene-9-carboxylic acid scopine ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid tropenole ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid scopine ester-methobromide; Benzilic acid cyclopropyl tropine ester-methobromide; 2,2-Diphenyl propionic acid cyclopropyltropine ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Methyl-fluorene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid cyclopropyltropine ester-methobromide; 9-Hydroxy-fluorene-9-carboxilic acid cyclopropyltropine ester-methobromide; 4,4′-Difluor benzilic acid methyl ester cyclopropyltropine ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxy-xanthene-9-carboxylic acid scopine ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Methyl-xanthene-9-carboxylic acid scopine ester-methobromide; 9-Ethyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Difluormethyl-xanthene-9-carboxylic acid tropenole ester-methobromide; 9-Hydroxymethyl-xanthene-9-carboxylic acid scopine ester-methobromide;
  • 3-[2-(3-Chloro-phenyl)-ethoxy]-N-(2-diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-propionamide;
  • N-(2-Diethylamino-ethyl)-N-{2-[2-(4-hydroxy-2-oxo-2,3-dihydro-benzothiazol-7-yl)-ethylamino]-ethyl}-3-(2-naphthalen-1-yl-ethoxy)-propionamide;
  • 7-[2-(2-{3-[2-(2-Chloro-phenyl)-ethylamino]-propylsulfanyl}-ethylamino)-1-hydroxy-ethyl]-4-hydroxy-3H-benzothiazol-2-one and Darotropium;
  • optionally in the form of the solvates or hydrates thereof.
  • In the above-mentioned salts the cations tiotropium, oxitropium, flutropium, ipratropium, glycopyrronium, aclidinium and trospium are the pharmacologically active ingredients. As anions, the above-mentioned salts may preferably contain chloride, bromide, iodide, sulphate, phosphate, methanesulphonate, nitrate, maleate, acetate, citrate, fumarate, tartrate, oxalate, succinate, benzoate or p-toluenesulphonate, while chloride, bromide, iodide, sulphate, methanesulphonate or p-toluenesulphonate are preferred as counter-ions. Of all the salts, the chlorides, bromides, iodides and methanesulphonate are particularly preferred.
  • Of particular importance is tiotropium bromide. In the case of tiotropium bromide the pharmaceutical combinations according to the invention preferably contain it in the form of the crystalline tiotropium bromide monohydrate, which is known from WO 02/30928. If the tiotropium bromide is used in anhydrous form in the pharmaceutical combinations according to the invention, it is preferable to use anhydrous crystalline tiotropium bromide, which is known from WO 03/000265.
  • Corticosteroids used here are preferably compounds selected from among beclomethasone, betamethasone, budesonide, butixocort, ciclesonide, deflazacort, dexamethasone, etiprednole, flunisolide, fluticasone, loteprednole, mometasone, prednisolone, prednisone, rofleponide, triamcinolone, tipredane; Pregna-1,4-diene-3,20-dione, 6-fluoro-11-hydroxy-16,17-[(1-methylethylidene) bis(oxy)]-21-[[4-[(nitrooxy)methyl]benzoyl]oxy]-, (6-alpha,11-beta,16-alpha)-(9Cl); 16,17-butylidenedioxy-6,9-difluoro-11-hydroxy-17-(methylthio)androst-4-en-3-one; 6,9-Difluor-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-dien-17-carbothione acid (S)-fluoromethylester; (S)-fluoromethyl 6,9-difluoro-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-diene-17-carbothionate;6-alpha,9-alpha-difluoro-11-beta-hydroxy-16alpha-methyl-3-oxo-17alpha-(2,2,3,3-tetramethylcyclopropylcarbonyl)oxy-androsta-1,4-diene-17beta-carboxylic acid cyanomethyl ester, each optionally in the form of the racemates, enantiomers or diastereomers thereof and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.
  • Particularly preferably the steroid is selected from among budesonide, fluticasone, mometasone, ciclesonide and (S)-fluoromethyl 6,9-difluoro-17-[(2-furanylcarbonyl)oxy]-11-hydroxy-16-methyl-3-oxo-androsta-1,4-diene-17-carbothionate, optionally in the form of the racemates, enantiomers or diastereomers thereof and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.
  • Any reference to steroids includes a reference to any salts or derivatives, hydrates or solvates thereof which may exist. Examples of possible salts and derivatives of the steroids may be: alkali metal salts, such as for example sodium or potassium salts, sulfobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates thereof.
  • PDE4 inhibitors which may be used are preferably compounds selected from among enprofyllin, theophyllin, roflumilast, ariflo (cilomilast), tofimilast, pumafentrin, lirimilast, apremilast, arofyllin, atizoram, oglemilast, tetomilast; 5-[N-(2,5-dichloro-3-pyridinyl)-carboxamide]-8-methoxy-Quinoline (D-4418); 5-[N-(3,5-dichloro-1-oxido-4-pyridinyl)-carboxamide]-8-methoxy-2-(trifluoromethyl)-Quinoline (D-4396 (Sch-351591)); N-(3,5-dichloropyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indol-3-yl]glyoxylic acid amide (AWD-12-281 (GW-842470)); 9-[(2-fluorophenyl)methyl]-N-methyl-2-(trifluoromethyl)-9H-Purin-6-amine (NCS-613); 4-[(2R)-2-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-phenylethyl]-Pyridine (CDP-840); N-[(3R)-3,4,6,7-tetrahydro-9-methyl-4-oxo-1-phenylpyrrolo[3,2,1-jk][1,4]benzodiazepin-3-yl]-4-Pyridinecarboxamide (PD-168787); 4-[6,7-diethoxy-2,3-bis(hydroxymethyl)-1-naphthalenyl]-1-(2-methoxyethyl)-2(1H)-Pyridinone (T-440); 2-[4-[6,7-diethoxy-2,3-bis(hydroxymethyl)-1-naphthalenyl]-2-pyridinyl]-4-(3-pyridinyl)-1(2H)-Phthalazinone (T-2585); (3-(3-cyclopenyloxy-4-methoxybenzyl)-6-ethylamino-8-isopropyl-3H-purine (V-11294A); beta-[3-(cyclopentyloxy)-4-methoxyphenyl]-1,3-dihydro-1,3-dioxo-2H-lsoindole-2-propanamide (CDC-801); Imidazo[1,5-a]pyrido[3,2-e]pyrazine-6(5H)-one, 9-ethyl-2-methoxy-7-methyl-5-propyl-(D-22888); 5-[3-(cyclopentyloxy)-4-methoxyphenyl]-3-[(3-methylphenyl)methyl]-,(3S,5S)-2-Piperidinon (HT-0712); 4-[1-[3,4-bis(difluoromethoxy)phenyl]-2-(3-methyl-1-oxido-4-pyridinyl)ethyl]-alpha, alpha-bis(trifluoromethyl)-Benzenemethanol (L-826141); N-(3,5-Dichloro-1-oxo-pyridin-4-yl)-4-dif luormethoxy-3-cyclopropylmethoxybenzamide; (−)p-[(4aR*,10bS*)-9-Ethoxy-1,2,3,4,4a,10b-hexahydro-8-methoxy-2-methylbenzo[s][1,6]naphthyridin-6-yl]-N,N-diisopropylbenzamide; (R)-(+)-1-(4-Brombenzyl)-4-[(3-cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidon; 3-(Cyclopentyloxy-4-methoxyphenyl)-1-(4-N′-[N-2-cyano-S-methyl-isothioureido]benzyl)-2-pyrrolidon; cis[4-Cyano-4-(3-cyclopentyloxy-4-methoxyphenyl)cyclohexan-1-carboxylic acid]; 2-carbomethoxy-4-cyano-4-(3-cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-1-one; cis[4-Cyano-4-(3-cyclopropylmethoxy-4-difluormethoxyphenyl)cyclohexan-1-ol]; (R)-(+)-Ethyl[4-(3-cyclopentyloxy-4-methoxyphenyl)pyrrolidin-2-yliden]acetat; (S)-(−)-Ethyl[4-(3-cyclopentyloxy-4-methoxyphenyl)pyrrolidin-2-yliden]acetat; 9-Cyclopentyl-5,6-dihydro-7-ethyl-3-(2-thienyl)-9H-pyrazolo[3,4-c]-1,2,4-triazolo[4,3-a]pyridin; 9-Cyclopentyl-5,6-dihydro-7-ethyl-3-(tert-butyl)-9 H-pyrazolo[3,4-c]-1, 2,4-triazolo[4,3-a]pyridin,
  • optionally in the form of the racemates, enantiomers or diastereomers and optionally in the form of the pharmacologically acceptable acid addition salts, solvates and/or hydrates thereof.
  • By acid addition salts with pharmacologically acceptable acids which the above-mentioned PDE4-inhibitors might be in a position to form are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • LTD4-antagonists which may be used are preferably compounds selected from among montelukast, pranlukast, zafirlukast; (E)-8-[2-[4-[4-(4-Fluorophenyl)butoxy]phenyl]ethenyl]-2-(1H-tetrazol-5-yl)-4H-1-benzopyran-4-one (MEN-91507); 4-[6-Acetyl-3-[3-(4-acetyl-3-hydroxy-2-propylphenylthio)propoxy]-2-propylphenoxy]-butyric acid (MN-001); 1-(((R)-(3-(2-(6,7-Difluor-2-quinolinyl)ethenyl)phenyl)-3-(2-(2-hydroxy-2-propyl)phenyl)thio)methylcyclopropane-acetic acid; 1-(((1(R)-3(3-(2-(2,3-Dichlorthieno[3,2-b]pyridin-5-yl)-(E)-ethenyl)phenyl)-3-(2-(1-hydroxy-1-methylethyl)phenyl) propyl)thio)methyl)cyclopropane acetic acid; [2-[[2-(4-tert-Butyl-2-thiazolyl)-5-benzofuranyl]oxymethyl]phenyl] acetic acid,
  • optionally in the form of the racemates, enantiomers or diastereomers, optionally in the form of the pharmacologically acceptable acid addition salts and optionally in the form of the salts and derivatives, solvates and/or hydrates thereof.
  • By acid addition salts with pharmacologically acceptable acids which the LTD4-antagonists may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate. By salts or derivatives which the LTD4-antagonists may be capable of forming are meant, for example: alkali metal salts, such as, for example, sodium or potassium salts, alkaline earth metal salts, sulphobenzoates, phosphates, isonicotinates, acetates, propionates, dihydrogen phosphates, palmitates, pivalates or furoates.
  • The EGFR-inhibitors used are preferably compounds selected from among 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-diethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-((R)-2-methoxymethyl-6-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-(N,N-bis-(2-methoxy-ethyl)-amino)-1-oxo-2-butene-1-yl]amino}-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-ethyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-({4-[N-(tetrahydropyran-4-yl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-Phenyl-ethyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(R)-(1-Phenyl-ethyl)amino]-6-({4-[N-(tetrahydropyran-4-yl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopropylmethoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-((R)-tetrahydrofuran-3-yloxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-((S)-tetrahydrofuran-3-yloxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N-(2-methoxy-ethyl)-N-methyl-amino]-1-oxo-2-butene-1-yl}amino)-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N-cyclopropyl-N-methyl-amino)-1-oxo-2-butene-1-yl]amino}-7-cyclopentyloxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-[(R)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-21-yl)methoxy]-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6.7-bis-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-7-[3-(morpholine-4-yl)-propyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-(4-hydroxy-phenyl)-7H-pyrrolo[2,3-d]pyrimidine, 3-cyano-4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-ethoxy-quinoline, 4-{[3-chloro-4-(3-fluoro-benzyloxy)-phenyl]amino}-6-(5-{[(2-methanesulphonyl-ethyl)amino]methyl}-furan-2-yl)quinazoline, 4-[(R)-(1-phenyl-ethyl)amino]-6-{[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(morpholine-4-yl)-1-oxo-2-butene-1-yl]-amino}-7-[(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluorophenyl)amino]-6-({4-[N,N-bis-(2-methoxy-ethyl)-amino]-1-oxo-2-butene-1-yl}amino)-7-[(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-{[4-(5.5-dimethyl-2-oxo-morpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-[(R)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-6-[(S)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{2-[4-(2-oxo-morpholine-4-yl)-piperidine-1-yl]-ethoxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(tert.-butyloxycarbonyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-amino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-methanesulphonylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-3-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(methoxymethyl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(piperidine-3-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(2-acetylamino-ethyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-ethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-((S)-tetrahydrofuran-3-yloxy)-7-hydroxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[dimethylamino)sulphonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[(morpholine-4-yl)carbonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{trans-4-[(morpholine-4-yl)sulphonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-acetylamino-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(tetrahydropyran-4-yloxy)-7-(2-methanesulphonylamino-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(piperidine-1-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-aminocarbonylmethyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(tetrahydropyran-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(morpholine-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazolin; 4-{2-[4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yloxy]-ethyl}-6-methyl-morpholine-2-one, 4-{4-[4-(3-chloro-2-fluoro-phenylamino)-7-methoxy-quinazolin-6-yloxy]-cyclohexyl}-1-methyl-piperazine-2-one, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(morpholine-4-yl)sulphonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-ethansulphonylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-ethoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[1-(2-methoxy-acetyl)-piperidine-4-yloxy]-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-acetylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-[1-(tert.-butyloxycarbonyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(tetrahydropyran-4-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(piperidine-1-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-{N-[(4-methyl-piperazine-1-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{cis-4-[(morpholine-4-yl)carbonylamino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[2-(2-oxopyrrolidin-1-yl)ethyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-(2-methoxy-ethoxy)-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-acetyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(1-methanesulphonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-methyl-piperidine-4-yloxy)-7(2-methoxy-ethoxy)-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-isopropyloxycarbonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(cis-4-methylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{cis-4[N-(2-methoxy-acetyl)-N-methyl-amino]-cyclohexan-1-yloxy}-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-(piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-[1-(2-methoxy-acetyl)-piperidine-4-yloxy]-7-methoxy-quinazoline, 4-[(3-ethynyl-phenyl)amino]-6-{1-[(morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(cis-2,6-dimethyl-morpholine-4-yl)carbony]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(2-methyl-morpholine-4-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(S,S)-(2-oxa-5-aza-bicyclo[2,2,1]hept-5-yl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(N-methyl-N-2-methoxyethyl-amino)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-ethyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(2-methoxyethyl)carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{1-[(3-methoxypropyl-amino)-carbonyl]-piperidine-4-yloxy}-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[cis-4-(N-methanesulphonyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[cis-4-(N-acetyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-methylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[trans-4-(N-methanesulphonyl-N-methyl-amino)-cyclohexan-1-yloxy]-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-dimethylamino-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(trans-4-{N-[(morpholine-4-yl)carbonyl]-N-methyl-amino}-cyclohexan-1-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-[2-(2.2-dimethyl-6-oxo-morpholine-4-yl)-ethoxy]-7-[(S)-(tetrahydrofuran-2-yl)methoxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1- methanesulphonyl-piperidine-4-yloxy)-7-methoxy-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-6-(1-cyano-piperidine-4-yloxy)-7-methoxy-quinazoline, 3-Cyano-4-[(3-chlor-4-fluorphenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-butene-1-yl]amino}-7-ethoxy-quinoline, [4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-(homomorpholine-4-yl)-1-oxo-2-butene-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-(2-{4-[(S)-(2-oxo-tetrahydrofuran-5-yl)carbonyl]-piperazine-1-yl}-ethoxy)-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((S)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-(2-{4-[(S)-(2-oxo-tetrahydrofuran-5-yl)carbonyl]-piperazine-1-yl}-ethoxy)-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[2-((S)-6-methyl-2-oxo-morpholine-4-yl)-ethoxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((R)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, 4-[(3-chloro-4-fluoro-phenyl)amino]-7-[4-((S)-6-methyl-2-oxo-morpholine-4-yl)-butyloxy]-6-[(vinylcarbonyl)amino]-quinazoline, cetuximab, trastuzumab, panitumumab (=ABX-EGF), Mab ICR-62, gefitinib, pelitinib, canertinib and erlotinib, optionally in the form of the racemates, enantiomers or diastereomers thereof, optionally in the form of the pharmacologically acceptable acid addition salts thereof, the solvates and/or hydrates thereof.
  • By acid addition salts with pharmacologically acceptable acids which the EGFR-inhibitors may be capable of forming are meant, for example, salts selected from among the hydrochloride, hydrobromide, hydroiodide, hydrosulphate, hydrophosphate, hydromethanesulphonate, hydronitrate, hydromaleate, hydroacetate, hydrobenzoate, hydrocitrate, hydrofumarate, hydrotartrate, hydrooxalate, hydrosuccinate, hydrobenzoate and hydro-p-toluenesulphonate, preferably hydrochloride, hydrobromide, hydrosulphate, hydrophosphate, hydrofumarate and hydromethanesulphonate.
  • Examples of dopamine agonists which may be used preferably include compounds selected from among bromocriptine, cabergoline, alpha-dihydroergocryptine, lisuride, pergolide, pramipexol, roxindol, ropinirol, talipexol, terguride and viozan. Any reference to the above-mentioned dopamine agonists within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts and optionally hydrates thereof which may exist. By the physiologically acceptable acid addition salts which may be formed by the above-mentioned dopamine agonists are meant, for example, pharmaceutically acceptable salts which are selected from the salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, citric acid, tartaric acid and maleic acid.
  • Examples of H1-antihistamines preferably include compounds selected from among epinastine, cetirizine, azelastine, fexofenadine, levocabastine, loratadine, mizolastine, ketotifen, emedastine, dimetinden, clemastine, bamipin, cexchlorpheniramine, pheniramine, doxylamine, chlorophenoxamine, dimenhydrinate, diphenhydramine, promethazine, ebastine, olopatadine, desloratidine and meclozine. Any reference to the above-mentioned H1-antihistamines within the scope of the present invention includes a reference to any pharmacologically acceptable acid addition salts which may exist.
  • Examples of PAF-antagonists preferably include compounds selected from among lexipafant, 4-(2-chlorophenyl)-9-methyl-2-[3(4-morpholinyl)-3-propanon-1-yl]-6H-thieno-[3,2-f]-[1,2,4]triazolo[4,3-a][1,4]diazepines, 6-(2-chlorophenyl)-8,9-dihydro-1-methyl-8-[(4-morpho-linyl)carbonyl]-4H,7H-cyclo-penta-[4,5]thieno-[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepines. An reference to the above-mentioned above-mentioned PAF-antagonists includes within the scope of the present invention a reference to any pharmacologically acceptable acid addition salts thereof which may exist.
  • Examples of non-steroidal anti-inflammatory drugs (NSAIDs) preferably include compounds selected from among Aceclofenac, Acemetacin, Acetylsalicylsäure, Alclofenac, Alminoprofen, Amfenac, Ampiroxicam, Antolmetinguacil, Anirolac, Antrafenin, Azapropazon, Benorilat, Bermoprofen, Bindarit, Bromfenac, Bucloxinsäure, Bucolom, Bufexamac, Bumadizon, Butibufen, Butixirat, Carbasalatcalcium, Carprofen, Cholin Magnesium Trisalicylat, Celecoxib, Cinmetacin, Cinnoxicam, Clidanac, Clobuzarit, Deboxamet, Dexibuprofen, Dexketoprofen, Diclofenac, Diflunisal, Droxicam, Eltenac, Enfenaminsäure, Etersalat, Etodolac, Etofenamat, Etoricoxib, Feclobuzon, Felbinac, Fenbufen, Fenclofenac, Fenoprofen, Fentiazac, Fepradinol, Feprazon, Flobufen, Floctafenin, Flufenaminsäure, Flufenisal, Flunoxaprofen, Flurbiprofen, Flurbiprofenaxetil, Furofenac, Furprofen, Glucametacin, Ibufenac, Ibuprofen, Indobufen, Indometacin, Indometacinfarnesil, Indoprofen, Isoxepac, Isoxicam, Ketoprofen, Ketorolac, Lobenzarit, Lonazolac, Lornoxicam, Loxoprofen, Lumiracoxib, Meclofenaminsäure, Meclofen, Mefenaminsäure, Meloxicam, Mesalazin, Miroprofen, Mofezolac, Nabumeton, Naproxen, Nifluminsäure, Olsalazin, Oxaprozin, Oxipinac, Oxyphenbutazon, Parecoxib, Phenylbutazon, Pelubiprofen, Pimeprofen, Pirazolac, Priroxicam, Pirprofen, Pranoprofen, Prifelon, Prinomod, Proglumetacin, Proquazon, Protizininsäure, Rofecoxib, Romazarit, Salicylam id, Salicylsäure, Salmistein, Salnacedin, Salsalat, Sulindac, Sudoxicam, Suprofen, Talniflumat, Tenidap, Tenosal, Tenoxicam, Tepoxalin, Tiaprofensäure, Taramid, Tilnoprofenarbamel, Timegadin, Tinoridin, Tiopinac, Tolfenaminsäure, Tolmetin, Ufenamat, Valdecoxib, Ximoprofen, Zaltoprofen and Zoliprofen.
  • MRP4-inhibitors used are preferably compounds selected from among N-acetyl-dinitrophenyl-cysteine, cGMP, cholate, diclofenac, dehydroepiandrosterone 3-glucuronide, dehydroepiandrosterone 3-sulphate, dilazep, dinitrophenyl-s-glutathione, estradiol 17-beta-glucuronide, estradiol 3,17-disulphate, estradiol 3-glucuronide, estradiol 3-sulphate, estrone 3-sulphate, flurbiprofen, folate, N5-formyl-tetrahydrofolate, glycocholate, glycolithocholic acid sulphate, ibuprofen, indomethacin, indoprofen, ketoprofen, lithocholic acid sulphate, methotrexate, ((E)-3-[[[3-[2-(7-chloro-2-quinolinyl)ethenyl]phenyl]-[[3-dimethylamino)-3-oxopropyl]thio]methyl]thio]-propanoic acid), alpha-naphthyl-beta-D-glucuronide, nitrobenzyl mercaptopurine riboside, probenecid , sildenafil, sulfinpyrazone, taurochenodeoxycholate, taurocholate, taurodeoxycholate, taurolithocholate, taurolithocholic acid sulphate, topotecan, trequinsin and zaprinast, dipyridamole, optionally in the form of the racemates, enantiomers, diastereomers and the pharmacologically acceptable acid addition salts and hydrates thereof.
  • Examples of JAK inhibitors preferably include compounds selected from among Tofacitinib and Ruxolitinib.
  • Examples of immunesuppressive agents preferably include compounds selected from among mycophenolate mofetil, mycophenolic acid, azathioprine, cyclosporine, tacrolimus, pimecrolimus, abetimus, gusperimus and leflunomide.
  • An example of a cytostaticum is cyclophosphamide.
  • The invention relates more preferably to the use of MRP4-inhibitors for preparing a pharmaceutical composition for treating respiratory complaints, containing the Syk-inhibitors of formula 1 and MRP4-inhibitors according to the invention, the MRP4-inhibitors preferably being selected from among dehydroepiandrosterone 3-sulphate, estradiol 3,17-disulphate, flurbiprofen, indomethacin, indoprofen, taurocholate, optionally in the form of the racemates, enantiomers, diastereomers and the pharmacologically acceptable acid addition salts and hydrates thereof. The separation of enantiomers from the racemates can be carried out using methods known from the art (e.g. chromatography on chiral phases, etc.).
  • By acid addition salts with pharmacologically acceptable acids are meant, for example, salts selected from among the hydrochlorides, hydrobromides, hydroiodides, hydrosulphates, hydrophosphates, hydromethanesulphonates, hydronitrates, hydromaleates, hydroacetates, hydrobenzoates, hydrocitrates, hydrofumarates, hydrotartrates, hydrooxalates, hydrosuccinates, hydrobenzoates and hydro-p-toluenesulphonates, preferably the hydrochlorides, hydrobromides, hydrosulphates, hydrophosphates, hydrofumarates and hydromethanesulphonates.
  • The invention further relates to pharmaceutical preparations which contain a triple combination of the Syk-inhibitors of formula 1, MRP4-inhibitors and another active substance according to the invention, such as, for example, an anticholinergic, a PDE4 inhibitor, a steroid, an LTD4-antagonist or a betamimetic, and the preparation thereof and the use thereof for treating respiratory complaints.
  • Compounds which may be used as iNOS inhibitors are compounds selected from among: S-(2-aminoethyl)isothiourea, aminoguanidine, 2-aminomethylpyridine, 5,6-dihydro-6-methyl-4H-1,3-Thiazine-2-amine (=AMT), L-canavanine, 2-iminopiperidine, S-isopropylisothiourea, S-methylisothiourea, S-ethylisothiourea, S-methyltiocitrullin, S-ethylthiocitrulline, L-NA (Nw -nitro-L-arginine), L-NAME (Nw-nitro-L-argininemethylester), L-NMMA (NG-monomethyl-L-arginine), L-NIO (Nw-iminoethyl-L-ornithine), L-NIL (Nw-iminoethyl-lysine), (S)-6-acetimidoylamino-2-amino-hexanoic acid (1H-tetrazol-5-yl)-amide (SC-51) (J. Med. Chem. 2002, 45, 1686-1689), N-[[3-(aminomethyl)phenyl]methyl]-Ethanimidamide (=1400W), (S)-4-(2-acetimidoylamino-ethylsulphanyl)-2-amino-butyric acid (GW274150) (Bioorg. Med. Chem. Lett. 2000, 10, 597-600), 2-[2-(4-methoxy-pyridin-2-yl)-ethyl]-3H-imidazo[4,5-b]pyridine (BYK191023) (Mol. Pharmacol. 2006, 69, 328-337), 2-((R)-3-amino-1-phenyl-propoxy)-4-chloro-5-fluorobenzonitrile (WO 01/62704), 2-((1R,3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-6-trifluoromethyl-nicotinonitrile (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-4-chloro-benzonitrile (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-5-chloro-benzonitrile (WO 2004/041794), (2S.4R)-2-amino-4-(2-chloro-5-trifluoromethyl-phenylsulphanyl)-4-thiazol-5-yl-butan-1-ol (WO 2004/041794), 2-((1R.3S)-3-amino-4-hydroxy-1-thiazol-5-yl-butylsulphanyl)-5-chloro-nicotinonitrile (WO 2004/041794), 4-((S)-3-amino-4-hydroxy-1-phenyl-butylsulphanyl)-6-methoxy-nicotinonitrile (WO 02/090332), substituted 3-phenyl-3,4-dihydro-1-isoquinolinamine such as e.g. (1S.5S.6R)-7-chloro-5-methyl-2-aza-bicyclo[4.1.0]hept-2-en-3-ylamine (ONO-1714) (Biochem. Biophys. Res. Commun. 2000, 270, 663-667), (4R,5R)-5-ethyl-4-methyl-thiazolidin-2-ylideneamine (Bioorg. Med. Chem. 2004, 12, 4101), (4R,5R)-5-ethyl-4-methyl-selenazolidin-2-ylideneamine (Bioorg. Med. Chem. Lett. 2005, 15, 1361), 4-aminotetrahydrobiopterine (Curr. Drug Metabol. 2002, 3, 119-121), (E)-3-(4-chloro-phenyl)-N-(1-{2-oxo-2-[4-(6-trifluoromethyl-pyrimidin-4-yloxy)-piperidine-1-yl]-ethylcarbamoyl}-2-pyridin-2-yl-ethyl)-acrylamide (FR260330) (Eur. J. Pharmacol. 2005, 509, 71-76), 3-(2,4-difluoro-phenyl)-6-[2-(4-imidazol-1-ylmethyl-phenoxy)-ethoxy]-2-phenyl-pyridine (PPA250) (J. Pharmaco Exp. Ther. 2002, 303, 52-57), 3-{[(benzo[1,3]dioxo1-5-ylmethyl)-carbamoyl]-methyl}-4-(2-imidazol-1-yl-pyrimidin-4-yl)-piperazine-1-carboxylate (BBS-1) (Drugs Future 2004, 29, 45-52), (R)-1-(2-imidazol-1-yl-6-methyl-pyrimidin-4-yl)-pyrrolidine-2-carboxylic acid (2-benzo[1,3]dioxo1-5-yl-ethyl)-amide (BBS-2) (Drugs Future 2004, 29, 45-52) and the pharmaceutical salts, prodrugs or solvates thereof.
  • Examples of iNOS-inhibitors within the scope of the present invention may also include antisense oligonucleotides, particularly those antisense oligonucleotides which bind iNOS-coding nucleic acids. For example, WO 01/52902 describes antisense oligonucleotides, particularly antisense oligonucleotides, which bind iNOS coding nucleic acids, for modulating the expression of iNOS. iNOS-antisense oligonucleotides as described particularly in WO 01/52902 may therefore also be combined with the PDE4-inhibitors of the present invention on account of their similar effect to the iNOS-inhibitors.
  • Suitable HMG-CoA reductase inhibitors (also called statins) which may be preferably used in double or triple combinations with the compounds of formula 1 are selected from among Atorvastatin, Cerivastatin, Flurvastatin, Lovastatin, Pitavastatin, Pravastatin, Rosuvastatin, Simvastatin, optionally in form of their pharmaceutically available acid addition salts, prodrugs, solvates or hydrates thereof.
  • 8. FORMULATIONS
  • Suitable forms for administration are for example tablets, capsules, solutions, syrups, emulsions or inhalable powders or aerosols. The content of the pharmaceutically effective compound(s) in each case should be in the range from 0.1 to 90 wt. %, preferably 0.5 to 50 wt. % of the total composition, i.e. in amounts which are sufficient to achieve the dosage range specified hereinafter.
  • The preparations may be administered orally in the form of a tablet, as a powder, as a powder in a capsule (e.g. a hard gelatine capsule), as a solution or suspension. When administered by inhalation the active substance combination may be given as a powder, as an aqueous or aqueous-ethanolic solution or using a propellant gas formulation.
  • Preferably, therefore, pharmaceutical formulations are characterised by the content of one or more compounds of formula 1 according to the preferred embodiments above.
  • It is particularly preferable if the compounds of formula 1 are administered orally, and it is also particularly preferable if they are administered once or twice a day. Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate. The tablets may also comprise several layers.
  • Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arabic, talc, titanium dioxide or sugar. To achieve delayed release or prevent incompatibilities the core may also consist of a number of layers. Similarly the tablet coating may consist of a number of layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.
  • Syrups containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavour enhancer, e.g. a flavouring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.
  • Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gelatine capsules. Suitable suppositories may be made for example by mixing with carriers provided for this purpose, such as neutral fats or polyethyleneglycol or the derivatives thereof.
  • Excipients which may be used include, for example, water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose), emulsifiers (e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone) and lubricants (e.g. magnesium stearate, talc, stearic acid and sodium lauryl sulphate).
  • For oral administration the tablets may, of course, contain, apart from the abovementioned carriers, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additives such as starch, preferably potato starch, gelatine and the like. Moreover, lubricants such as magnesium stearate, sodium lauryl sulphate and talc may be used at the same time for the tabletting process. In the case of aqueous suspensions the active substances may be combined with various flavour enhancers or colourings in addition to the excipients mentioned above.
  • It is also preferred if the compounds of formula 1 are administered by inhalation, particularly preferably if they are administered once or twice a day. For this purpose, the compounds of formula 1 have to be made available in forms suitable for inhalation. Inhalable preparations include inhalable powders, propellant-containing metered-dose aerosols or propellant-free inhalable solutions, which are optionally present in admixture with conventional physiologically acceptable excipients.
  • Within the scope of the present invention, the term propellant-free inhalable solutions also includes concentrates or sterile ready-to-use inhalable solutions. The preparations which may be used according to the invention are described in more detail in the next part of the specification.
  • Inhalable Powders
  • If the active substances of formula 1 are present in admixture with physiologically acceptable excipients, the following physiologically acceptable excipients may be used to prepare the inhalable powders according to the invention: monosaccharides (e.g. glucose or arabinose), disaccharides (e.g. lactose, saccharose, maltose), oligo- and polysaccharides (e.g. dextran), polyalcohols (e.g. sorbitol, mannitol, xylitol), salts (e.g. sodium chloride, calcium carbonate) or mixtures of these excipients with one another. Preferably, mono- or disaccharides are used, while the use of lactose or glucose is preferred, particularly, but not exclusively, in the form of their hydrates. For the purposes of the invention, lactose is the particularly preferred excipient, while lactose monohydrate is most particularly preferred. Methods of preparing the inhalable powders according to the invention by grinding and micronising and by finally mixing the components together are known from the prior art.
  • Propellant-containing Inhalable Aerosols
  • The propellant-containing inhalable aerosols which may be used according to the invention may contain the compounds of formula 1 dissolved in the propellant gas or in dispersed form. The propellant gases which may be used to prepare the inhalation aerosols according to the invention are known from the prior art. Suitable propellant gases are selected from among hydrocarbons such as n-propane, n-butane or isobutane and halohydrocarbons such as preferably fluorinated derivatives of methane, ethane, propane, butane, cyclopropane or cyclobutane. The propellant gases mentioned above may be used on their own or in mixtures thereof. Particularly preferred propellant gases are fluorinated alkane derivatives selected from TG134a (1,1,1,2-tetrafluoroethane), TG227 (1,1,1,2,3,3,3-heptafluoropropane) and mixtures thereof. The propellant-driven inhalation aerosols used within the scope of the use according to the invention may also contain other ingredients such as co-solvents, stabilisers, surfactants, antioxidants, lubricants and pH adjusters. All these ingredients are known in the art.
  • Propellant-free Inhalable Solutions
  • The compounds of formula 1 according to the invention are preferably used to prepare propellant-free inhalable solutions and inhalable suspensions. Solvents used for this purpose include aqueous or alcoholic, preferably ethanolic solutions. The solvent may be water on its own or a mixture of water and ethanol. The solutions or suspensions are adjusted to a pH of 2 to 7, preferably 2 to 5, using suitable acids. The pH may be adjusted using acids selected from inorganic or organic acids. Examples of particularly suitable inorganic acids include hydrochloric acid, hydrobromic acid, nitric acid, sulphuric acid and/or phosphoric acid. Examples of particularly suitable organic acids include ascorbic acid, citric acid, malic acid, tartaric acid, maleic acid, succinic acid, fumaric acid, acetic acid, formic acid and/or propionic acid etc. Preferred inorganic acids are hydrochloric and sulphuric acids. It is also possible to use the acids which have already formed an acid addition salt with one of the active substances. Of the organic acids, ascorbic acid, fumaric acid and citric acid are preferred. If desired, mixtures of the above acids may also be used, particularly in the case of acids which have other properties in addition to their acidifying qualities, e.g. as flavourings, antioxidants or complexing agents, such as citric acid or ascorbic acid, for example. According to the invention, it is particularly preferred to use hydrochloric acid to adjust the pH.
  • Co-solvents and/or other excipients may be added to the propellant-free inhalable solutions used for the purpose according to the invention. Preferred co-solvents are those which contain hydroxyl groups or other polar groups, e.g. alcohols—particularly isopropyl alcohol, glycols—particularly propyleneglycol, polyethyleneglycol, polypropyleneglycol, glycolether, glycerol, polyoxyethylene alcohols and polyoxyethylene fatty acid esters. The terms excipients and additives in this context denote any pharmacologically acceptable substance which is not an active substance but which can be formulated with the active substance or substances in the pharmacologically suitable solvent in order to improve the qualitative properties of the active substance formulation. Preferably, these substances have no pharmacological effect or, in connection with the desired therapy, no appreciable or at least no undesirable pharmacological effect. The excipients and additives include, for example, surfactants such as soya lecithin, oleic acid, sorbitan esters, such as polysorbates, polyvinylpyrrolidone, other stabilisers, complexing agents, antioxidants and/or preservatives which guarantee or prolong the shelf life of the finished pharmaceutical formulation, flavourings, vitamins and/or other additives known in the art. The additives also include pharmacologically acceptable salts such as sodium chloride as isotonic agents. The preferred excipients include antioxidants such as ascorbic acid, for example, provided that it has not already been used to adjust the pH, vitamin A, vitamin E, tocopherols and similar vitamins or provitamins occurring in the human body. Preservatives may be used to protect the formulation from contamination with pathogens. Suitable preservatives are those which are known in the art, particularly cetyl pyridinium chloride, benzalkonium chloride or benzoic acid or benzoates such as sodium benzoate in the concentration known from the prior art.
  • For the treatment forms described above, ready-to-use packs of a medicament for the treatment of respiratory complaints are provided, containing an enclosed description including for example the words respiratory disease, COPD or asthma, together with a imidazolyl-pyrimidine according to formula 1 and one or more combination partners selected from those described above.

Claims (2)

1. An intermediate compound selected from the group consisting of formula 6
Figure US20180072744A1-20180315-C00232
of formula 7
Figure US20180072744A1-20180315-C00233
of formula 8
Figure US20180072744A1-20180315-C00234
and of formula 11
Figure US20180072744A1-20180315-C00235
wherein
Hal is Cl or Br
D is CH,
E is selected from the group consisting of C and N,
T is C,
G is selected from the group consisting of C and N,
and wherein each of the broken (dotted) double bonds in ring 1 are either a single bond or a double bond under the proviso that all single and double bonds of ring 1 are arranged in such a way that they all form together with ring 2 an aromatic ring system,
and wherein
Het is selected from the group consisting of
a five- to six-membered monocyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O,
and a nine- to eleven-membered bicyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
Hetaryl is selected from the group consisting of
a five- to six-membered monocyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
and a nine- to eleven-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
and wherein
R1 is selected from the group consisting of
C6-10-aryl, Het and Hetaryl;
which is optionally further substituted by one, two or three substituents Z,
whereby each Z is a substituent selected from the group consisting of
—OH, oxo, —CN, halogen, —C1-6-alkyl, —O—C1-6-alkyl, —C1-6-haloalkyl, three- to seven-membered cycloalkyl, Het, Hetaryl, —CO—N(CH3)2, —CO—NHCH3, —CO—NH2, —(C1-3-alkylene)-O—(C1-3-alkyl), and —O-Het,
which is optionally further substituted by one, two or three substituents X,
whereby each X is selected from the group consisting of halogen, —OH, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,—NH(CH3), and —N(CH3)2,
whereby substituent X is optionally further substituted by one, two or three substituents selected from the group consisting of from oxo, —OH, halogen and C1-3-alkyl,
and wherein PG is a protecting group selected from the group consisting of benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl.
2. An intermediate compound selected from the group consisting of formula 6
Figure US20180072744A1-20180315-C00236
of formula 7
Figure US20180072744A1-20180315-C00237
of formula 8
Figure US20180072744A1-20180315-C00238
and of formula 11
Figure US20180072744A1-20180315-C00239
wherein R1 is
Figure US20180072744A1-20180315-C00240
and wherein
Hal is Cl or Br,
X is selected from the group consisting of halogen, —OH, oxo, —C1-4-alkyl, —O—C1-4-alkyl, —C1-4-haloalkyl, —O—(C1-4-alkylene)-Het, Het, Hetaryl, —NH2,—NH(CH3), and —N(CH3)2,
whereby substituent X is optionally further substituted by one, two or three substituents of a group selected from oxo, —OH, halogen and C1-3-alkyl,
wherein each Het is selected from the group consisting of:
a five- to six-membered monocyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O,
and a nine- to eleven-membered bicyclic heterocycle with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
and wherein each Hetaryl is selected from the group consisting of:
a five- to six-membered monocyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
and a nine- to eleven-membered bicyclic heteroaromate with 1, 2, 3 or 4 heteroatoms each independently from one another selected from the group consisting of N, S and O;
and wherein PG is a protecting group selected from the group consisting of benzyl, 1-phenylethyl, 1-(4-methoxyphenyl)ethyl.
US15/817,435 2014-03-19 2017-11-20 Heteroaryl syk inhibitors Abandoned US20180072744A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/817,435 US20180072744A1 (en) 2014-03-19 2017-11-20 Heteroaryl syk inhibitors
US16/535,337 US10947243B2 (en) 2014-03-19 2019-08-08 Heteroaryl SYK inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201461955487P 2014-03-19 2014-03-19
PCT/EP2015/055228 WO2015140051A1 (en) 2014-03-19 2015-03-12 Heteroaryl sik inhibitors
US201615027518A 2016-04-06 2016-04-06
US15/817,435 US20180072744A1 (en) 2014-03-19 2017-11-20 Heteroaryl syk inhibitors

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2015/055228 Division WO2015140051A1 (en) 2014-03-19 2015-03-12 Heteroaryl sik inhibitors
US15/027,518 Division US9914735B2 (en) 2014-03-19 2015-03-12 Heteroaryl Syk inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/535,337 Continuation US10947243B2 (en) 2014-03-19 2019-08-08 Heteroaryl SYK inhibitors

Publications (1)

Publication Number Publication Date
US20180072744A1 true US20180072744A1 (en) 2018-03-15

Family

ID=52706148

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/125,599 Abandoned US20170008896A1 (en) 2014-03-19 2015-03-12 Heteroaryl syk inhibitors
US15/027,518 Active US9914735B2 (en) 2014-03-19 2015-03-12 Heteroaryl Syk inhibitors
US15/817,435 Abandoned US20180072744A1 (en) 2014-03-19 2017-11-20 Heteroaryl syk inhibitors
US16/535,337 Active US10947243B2 (en) 2014-03-19 2019-08-08 Heteroaryl SYK inhibitors

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/125,599 Abandoned US20170008896A1 (en) 2014-03-19 2015-03-12 Heteroaryl syk inhibitors
US15/027,518 Active US9914735B2 (en) 2014-03-19 2015-03-12 Heteroaryl Syk inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/535,337 Active US10947243B2 (en) 2014-03-19 2019-08-08 Heteroaryl SYK inhibitors

Country Status (32)

Country Link
US (4) US20170008896A1 (en)
EP (3) EP3119772B8 (en)
JP (3) JP6535022B2 (en)
KR (1) KR102431017B1 (en)
CN (3) CN110183440B (en)
AP (1) AP2016009373A0 (en)
AR (1) AR099785A1 (en)
AU (1) AU2015233654B2 (en)
CA (1) CA2942997C (en)
CL (1) CL2016001963A1 (en)
CY (1) CY1121937T1 (en)
DK (1) DK3119772T3 (en)
EA (1) EA032097B1 (en)
ES (1) ES2738416T3 (en)
HR (1) HRP20191364T1 (en)
HU (1) HUE046061T2 (en)
IL (1) IL247057B (en)
LT (1) LT3119772T (en)
ME (1) ME03472B (en)
MX (1) MX2016011784A (en)
MY (1) MY185837A (en)
PE (1) PE20161427A1 (en)
PH (1) PH12016501796A1 (en)
PL (1) PL3119772T3 (en)
PT (1) PT3119772T (en)
RS (1) RS59110B1 (en)
SG (1) SG11201607794QA (en)
SI (1) SI3119772T1 (en)
TW (1) TWI680967B (en)
UA (1) UA119667C2 (en)
UY (1) UY36037A (en)
WO (3) WO2015140054A1 (en)

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110183440B (en) 2014-03-19 2022-04-22 勃林格殷格翰国际有限公司 Heteroaryl SYK inhibitors
DK3347353T3 (en) 2015-09-11 2019-10-21 Boehringer Ingelheim Int PYRAZOLYL-SUBSTITUTED HETEROARYLS AND THEIR USE AS MEDICINES
AU2016348549B2 (en) 2015-11-02 2020-07-23 Janssen Pharmaceutica Nv [1,2,4]triazolo[1,5-a]pyrimidin-7-yl compound
CN105503730B (en) * 2015-12-25 2018-06-22 山东大学 Pyrazole derivatives and preparation method and application
TWI764950B (en) * 2016-11-02 2022-05-21 比利時商健生藥品公司 Pde2 inhibitors
CN109890824B (en) 2016-11-02 2022-05-24 詹森药业有限公司 [1,2,4] triazolo [1,5-a ] pyrimidine compounds as PDE2 inhibitors
ES2855032T3 (en) 2016-11-02 2021-09-23 Janssen Pharmaceutica Nv [1,2,4] triazolo [1,5-a] pyrimidine compounds as PDE2 inhibitors
JP7065951B2 (en) 2017-09-22 2022-05-12 ジュビラント エピパッド エルエルシー Heterocyclic compound as a PAD inhibitor
WO2019077631A1 (en) 2017-10-18 2019-04-25 Jubilant Biosys Limited Imidazo-pyridine compounds as pad inhibitors
EP3707135A1 (en) 2017-11-06 2020-09-16 Jubilant Prodel LLC Pyrimidine derivatives as inhibitors of pd1/pd-l1 activation
EP3704120B1 (en) 2017-11-24 2024-03-06 Jubilant Episcribe LLC Heterocyclic compounds as prmt5 inhibitors
BR112020012997A2 (en) 2017-12-26 2020-12-01 Kymera Therapeutics, Inc. irak degraders and uses thereof
KR20200131845A (en) 2018-03-13 2020-11-24 주빌런트 프로델 엘엘씨 Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
JP7460644B2 (en) 2018-10-31 2024-04-02 ギリアード サイエンシーズ, インコーポレイテッド Substituted 6-Azabenzimidazole Compounds as HPK1 Inhibitors
EP3873608A1 (en) 2018-10-31 2021-09-08 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020113233A1 (en) 2018-11-30 2020-06-04 Kymera Therapeutics, Inc. Irak degraders and uses thereof
US11633399B2 (en) 2018-12-25 2023-04-25 Sol-Gel Technologies Ltd. Treatment of skin disorders with compositions comprising an EGFR inhibitor
CN114364798A (en) 2019-03-21 2022-04-15 欧恩科斯欧公司 Combination of Dbait molecules with kinase inhibitors for the treatment of cancer
TWI826690B (en) 2019-05-23 2023-12-21 美商基利科學股份有限公司 Substituted eneoxindoles and uses thereof
WO2020264490A1 (en) * 2019-06-28 2020-12-30 Kymera Therapeutics, Inc. Irak degraders and uses thereof
AU2020378630A1 (en) 2019-11-08 2022-05-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
JP2023509366A (en) 2019-12-17 2023-03-08 カイメラ セラピューティクス, インコーポレイテッド IRAK dissolving agents and their uses
WO2021127283A2 (en) 2019-12-17 2021-06-24 Kymera Therapeutics, Inc. Irak degraders and uses thereof
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
TW202210483A (en) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Crystalline forms of irak degraders
US20220213086A1 (en) * 2020-12-25 2022-07-07 Eternity Bioscience Inc. Azole compounds as irak inhibitors, preparation methods and medicinal uses thereof
KR20220138655A (en) * 2021-04-06 2022-10-13 주식회사 온코크로스 Compound for preventing or treating kidney disease
KR20220138654A (en) * 2021-04-06 2022-10-13 주식회사 온코크로스 Compound for preventing or treating diabetes mellitus
CN115215803B (en) * 2022-09-19 2022-12-30 苏州美诺医药科技有限公司 Preparation method of 4-halogenated-1- (difluoromethyl) -1H-imidazole

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015017610A1 (en) * 2013-07-31 2015-02-05 Gilead Sciences, Inc. Syk inhibitors
US9845314B2 (en) * 2015-09-11 2017-12-19 Boehrnger Ingelheim International Gmbh Pyrazolyl-substituted heteroaryls and their use as medicaments

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4017500A (en) 1973-07-16 1977-04-12 Schering Corporation Certain 8-amino-1,7-naphthyridines
US4260759A (en) 1969-04-14 1981-04-07 Schering-Corporation Process for preparing 1-aminonaphthyridines
US3928367A (en) 1969-04-14 1975-12-23 Schering Corp 1-Amino naphthyridines
FR2260330A1 (en) 1974-02-07 1975-09-05 Innothera Lab Sa Pyridoxine N-oxy nicotinate - hypolipaemiant and hypocholesterolemiant of good therapeutic index
US4115395A (en) 1975-11-12 1978-09-19 Schering Corporation 1-Aminonaphthyridines
US6340759B1 (en) 1997-10-02 2002-01-22 Eisai Co., Ltd. Fused pyridine derivatives
JP2003520042A (en) 2000-01-24 2003-07-02 アイシス・ファーマシューティカルス・インコーポレーテッド Antisense modulation of inducible nitric oxide synthase expression
GB0004153D0 (en) 2000-02-23 2000-04-12 Astrazeneca Uk Ltd Novel use
JP2001302667A (en) 2000-04-28 2001-10-31 Bayer Ag Imidazopyrimidine derivative and triazolopyrimidine derivative
HU226830B1 (en) 2000-10-12 2009-11-30 Boehringer Ingelheim Pharma Thiotropium bromide monohydrate, method for producing the same and a pharmaceutical composition containing the same
AR035700A1 (en) 2001-05-08 2004-06-23 Astrazeneca Ab DERIVATIVES OF ARILHETEROALQUILAMINA, PHARMACEUTICAL COMPOSITION, USES OF THESE DERIVATIVES FOR THE MANUFACTURE OF MEDICINES, TREATMENT METHODS, AND PROCESS FOR THE PREPARATION OF THESE DERIVATIVES
DK1401445T3 (en) 2001-06-22 2006-11-27 Boehringer Ingelheim Pharma Crystalline anticholinergic, method of preparing it and using it to prepare a drug
US20030158195A1 (en) 2001-12-21 2003-08-21 Cywin Charles L. 1,6 naphthyridines useful as inhibitors of SYK kinase
SE0203304D0 (en) 2002-11-07 2002-11-07 Astrazeneca Ab Novel Coumpounds
US20080119515A1 (en) 2003-03-10 2008-05-22 M Arshad Siddiqui Heterocyclic Kinase Inhibitors: Methods of Use and Synthesis
CN1784229A (en) * 2003-03-10 2006-06-07 先灵公司 Heterocyclic kinase inhibitors: methods of use and synthesis
WO2006038041A1 (en) 2004-10-08 2006-04-13 Merck Sharp & Dohme Limited Besylate salts of six-membered amino-heterocycles as vanilloid-1 receptor antagonists for treating pain
US8703804B2 (en) * 2006-03-26 2014-04-22 Uti Limited Partnership Ryanodine receptor inhibitors and methods relating thereto
DE102007012645A1 (en) 2007-03-16 2008-09-18 Bayer Healthcare Ag Substituted imidazo and triazolopyrimidines
WO2010015520A1 (en) * 2008-08-05 2010-02-11 Boehringer Ingelheim International Gmbh Substituted naphthyridines and use thereof as medicines
JP5754568B2 (en) * 2008-08-05 2015-07-29 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Naphthyridine substituted with 4-dimethylamino-phenyl and its use as a medicament
TWI453207B (en) 2008-09-08 2014-09-21 Signal Pharm Llc Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
CA2786245A1 (en) 2010-01-29 2011-08-04 Boehringer Ingelheim International Gmbh Substituted naphthyridines and their use as syk kinase inhibitors
EP2683716A1 (en) 2011-03-11 2014-01-15 Glaxo Group Limited Pyrido[3,4-b]pyrazine derivatives as syk inhibitors
US8921383B2 (en) 2011-03-28 2014-12-30 Hoffmann-La Roche Inc. Thiazolopyrimidine compounds
WO2012167423A1 (en) 2011-06-08 2012-12-13 Hutchison Medipharma Limited Substituted pyridopyrazines as novel syk inhibitors
AU2012288969B2 (en) 2011-07-26 2017-02-23 Boehringer Ingelheim International Gmbh Substituted quinolines and their use as medicaments
JP5878178B2 (en) * 2011-09-30 2016-03-08 大鵬薬品工業株式会社 1,2,4-triazine-6-carboxamide derivatives
MX2014014582A (en) 2012-06-07 2015-05-11 Hoffmann La Roche Pyrazolopyrimidone and pyrazolopyridone inhibitors of tankyrase.
US20140309233A1 (en) 2012-12-18 2014-10-16 Hulow, Llc Syk kinase inhibitors as treatment for malaria
CN110183440B (en) 2014-03-19 2022-04-22 勃林格殷格翰国际有限公司 Heteroaryl SYK inhibitors
TW201617074A (en) 2014-07-14 2016-05-16 吉李德科學股份有限公司 Syk inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015017610A1 (en) * 2013-07-31 2015-02-05 Gilead Sciences, Inc. Syk inhibitors
US9845314B2 (en) * 2015-09-11 2017-12-19 Boehrnger Ingelheim International Gmbh Pyrazolyl-substituted heteroaryls and their use as medicaments

Also Published As

Publication number Publication date
SI3119772T1 (en) 2019-08-30
WO2015140051A1 (en) 2015-09-24
MX2016011784A (en) 2016-12-02
AU2015233654A1 (en) 2016-08-11
EP3119772B1 (en) 2019-05-22
JP6416275B2 (en) 2018-10-31
JP2017512779A (en) 2017-05-25
EP3119766B1 (en) 2019-05-22
PT3119772T (en) 2019-09-05
RS59110B1 (en) 2019-09-30
EP3119766A1 (en) 2017-01-25
CN110183440A (en) 2019-08-30
AP2016009373A0 (en) 2016-08-31
JP2017511805A (en) 2017-04-27
AU2015233654B2 (en) 2018-12-06
JP2017512778A (en) 2017-05-25
LT3119772T (en) 2019-08-26
CN110713489B (en) 2022-05-31
PH12016501796A1 (en) 2016-11-21
TW201623285A (en) 2016-07-01
CA2942997A1 (en) 2015-09-24
EP3119772A1 (en) 2017-01-25
DK3119772T3 (en) 2019-08-26
UY36037A (en) 2015-09-30
EP3119765A1 (en) 2017-01-25
US9914735B2 (en) 2018-03-13
MY185837A (en) 2021-06-12
JP6535022B2 (en) 2019-06-26
PL3119772T3 (en) 2019-11-29
CN110713489A (en) 2020-01-21
US20170008896A1 (en) 2017-01-12
HRP20191364T1 (en) 2019-11-01
US10947243B2 (en) 2021-03-16
UA119667C2 (en) 2019-07-25
WO2015140051A8 (en) 2015-12-30
ME03472B (en) 2020-01-20
EA032097B1 (en) 2019-04-30
SG11201607794QA (en) 2016-10-28
WO2015140055A1 (en) 2015-09-24
KR20160132999A (en) 2016-11-21
US20190359620A1 (en) 2019-11-28
EP3119772B8 (en) 2019-08-21
CY1121937T1 (en) 2020-10-14
CN106255689A (en) 2016-12-21
TWI680967B (en) 2020-01-01
IL247057B (en) 2019-10-31
US20160244446A1 (en) 2016-08-25
CA2942997C (en) 2022-09-13
CN106255689B (en) 2019-09-17
AR099785A1 (en) 2016-08-17
ES2738416T3 (en) 2020-01-22
KR102431017B1 (en) 2022-08-11
WO2015140054A1 (en) 2015-09-24
HUE046061T2 (en) 2020-02-28
EP3119765B1 (en) 2019-07-17
IL247057A0 (en) 2016-09-29
CN110183440B (en) 2022-04-22
PE20161427A1 (en) 2017-01-06
EA201691864A1 (en) 2017-06-30
JP6599884B2 (en) 2019-10-30
CL2016001963A1 (en) 2016-12-23

Similar Documents

Publication Publication Date Title
US10947243B2 (en) Heteroaryl SYK inhibitors
US9096579B2 (en) Amino-indolyl-substituted imidazolyl-pyrimidines and their use as medicaments
US9199937B2 (en) Substituted quinolines and their use as medicaments
US10155751B2 (en) Pyrazolyl-substituted heteroaryls and their use as medicaments
US9187478B2 (en) Substituted naphthyridines and their use as medicaments
BR112016019473B1 (en) SYK INHIBITOR HETEROARYLS AND PHARMACEUTICAL FORMULATIONS INCLUDING THEM AND INTERMEDIATE COMPOUNDS

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION