WO2006003146A1 - Quinazolinone derivatives as parp inhibitors - Google Patents

Quinazolinone derivatives as parp inhibitors Download PDF

Info

Publication number
WO2006003146A1
WO2006003146A1 PCT/EP2005/053029 EP2005053029W WO2006003146A1 WO 2006003146 A1 WO2006003146 A1 WO 2006003146A1 EP 2005053029 W EP2005053029 W EP 2005053029W WO 2006003146 A1 WO2006003146 A1 WO 2006003146A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
formula
compound
parp
compounds
Prior art date
Application number
PCT/EP2005/053029
Other languages
French (fr)
Inventor
Jérôme Emile Georges Guillemont
Ludo Edmond Josephine Kennis
Josephus Carolus Mertens
Jacobus Alphonsus Josephus Van Dun
Maria Victorina Francisca Somers
Walter Boudewijn Leopold Wouters
Original Assignee
Janssen Pharmaceutica N.V.
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
Priority to EP05769923A priority Critical patent/EP1771422B1/en
Priority to CN2005800222595A priority patent/CN1980899B/en
Application filed by Janssen Pharmaceutica N.V. filed Critical Janssen Pharmaceutica N.V.
Priority to DE602005026391T priority patent/DE602005026391D1/en
Priority to EA200700189A priority patent/EA012837B1/en
Priority to KR1020067026804A priority patent/KR101286969B1/en
Priority to US11/569,892 priority patent/US8623872B2/en
Priority to AT05769923T priority patent/ATE498613T1/en
Priority to MXPA06014543A priority patent/MXPA06014543A/en
Priority to NZ551680A priority patent/NZ551680A/en
Priority to BRPI0512797-1A priority patent/BRPI0512797A/en
Priority to UAA200612700A priority patent/UA85593C2/en
Priority to CA2568835A priority patent/CA2568835C/en
Priority to JP2007518606A priority patent/JP4969443B2/en
Priority to AU2005259188A priority patent/AU2005259188B2/en
Publication of WO2006003146A1 publication Critical patent/WO2006003146A1/en
Priority to IL180411A priority patent/IL180411A/en
Priority to NO20070555A priority patent/NO20070555L/en
Priority to HK07111097.2A priority patent/HK1105960A1/en
Priority to US14/077,881 priority patent/US9522905B2/en
Priority to US15/350,663 priority patent/US10150757B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • C07D239/90Oxygen atoms with acyclic radicals attached in position 2 or 3
    • 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
    • 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • 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
    • 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
    • A61P17/16Emollients or protectives, e.g. against radiation
    • 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
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • 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
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C07D401/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing 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
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to inhibitors of PARP and provides compounds and compositions containing the disclosed compounds. Moreover, the present invention provides methods of using the disclosed PARP inhibitors for instance as a medicine.
  • PARP-I nuclear enzyme poly(ADP-ribose) polymerase-1
  • PARP-I nuclear enzyme poly(ADP-ribose) polymerase-1
  • PARP-2 PARP-2
  • PARP-3 Vault-PARP
  • TANKs Tankyrases
  • PARP is also referred to as poly(adenosine 5'-diphospho-ribose) polymerase or PARS (poly(ADP-ribose) synthetase).
  • PARP-I is a major nuclear protein of 116 kDa consisting of three domains : the N- terminal DNA binding domain containing two zinc fingers, the automodification domain and the C-terminal catalytic domain. It is present in almost all eukaryotes.
  • the enzyme synthesizes poly(ADP-ribose), a branched polymer that can consist of over 200 ADP-ribose units.
  • the protein acceptors of poly(ADP-ribose) are directly or indirectly involved in maintaining DNA integrity. They include histones, topoisomeras ⁇ s.j.DNA and RNA polymerases, DNA ligases, and Ca 2+ - and Mg 2+ -dependent endonucleases.
  • PARP protein is expressed at a high level in many tissues, most notably in the immune system, heart, brain and germ-line cells. Under normal physiological conditions, there is minimal PARP activity. However, DNA damage causes an immediate activation of PARP by up to 500-fold.
  • Tankyrases were identified as components of the human telomeric complex. They have also been proposed to have a role in vesicle trafficking and may serve as scaffolds for proteins involved in various other cellular processes. Telomeres, which are essential for chromosome maintenance and stability, are maintained by telomerase, a specialized reverse transcriptase. TANKs are (ADP-ribose)transferases with some features of both signalling and cytoskeletal proteins.
  • TRF-I Telomere Repeat binding Factor-1
  • TANK functions as a positive regulator of telomere length., allowing elongation of the telomeres by telomerase.
  • PARP and PARG form a cycle that converts a large amount of '1 KfAD + to ADP-ribose.
  • over-stimulation of PARP can cause a drop of NAD + and ATP to less than 20% of the normal level.
  • NAD + and ATP are especially detrimental during ischaemia when deprivation of oxygen has already drastically compromised cellular energy output.
  • Subsequent free radical production during reperfusion is assumed to be a major cause of tissue damage.
  • Part of the ATP drop which is typical in many organs during ischaemia and reperfusion, could be linked to NAD + depletion due to poly(ADP-ribose) turnover.
  • PARP or PARG inhibition is expected to preserve the cellular energy level thereby potentiating the survival of ischaemic tissues after insult.
  • PARP inhibitors suppress production of inducible nitric oxide synthase (iNOS) in macrophages, P-type selectin and intercellular adhesion molecule-1 (ICAM- 1) in endothelial cells. Such activity underlies the strong anti-inflammation effects exhibited by PARP inhibitors.
  • PARP inhibition is able to reduce necrosis by preventing translocation and infiltration of neutrophils to the injured tissues.
  • PARP is activated by damaged DNA fragments and, once activated, catalyzes the attachment of up to 100 ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself.
  • ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself.
  • ATP may also become depleted.
  • PARP activation can also be used as a measure of damage following neurotoxic insults resulting from exposure to any of the following inducers like glutamate (via NMDA receptor stimulation), reactive oxygen intermediates, amyloid ⁇ -protein, ; N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or its active metabolite
  • inducers like glutamate (via NMDA receptor stimulation), reactive oxygen intermediates, amyloid ⁇ -protein, ; N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or its active metabolite
  • MPTP N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine
  • N-methyl-4 phenylpyridine (MPP + ), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease.
  • MPP + N-methyl-4 phenylpyridine
  • Other studies have continued to explore the role of PARP activation in cerebellar granule cells in vitro and in MPTP neurotoxicity.
  • NMDA N-methyl-D-aspartate
  • AMPA AMPA
  • Kainate MGR receptors
  • ion flow e.g., Ca 2+ and Na + into the cells and K + out of the cells
  • the over-stimulated neurons secrete more glutamate, creating a feedback loop or domino effect which ultimately results in cell damage or death via the production of proteases, lipases and free radicals.
  • Excessive activation of glutamate receptors has been implicated in various neurological diseases and conditions including epilepsy, stroke, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's disease, schizophrenia, chronic pain, ischemia and neuronal loss following hypoxia, hypoglycemia, ischemia, trauma, and nervous insult. Glutamate exposure and stimulation has also been implicated as a basis for compulsive disorders, particularly drug dependence.
  • NMDA neurotoxicity may be prevented by treatment with nitric oxide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro.
  • NOS neuronal nitric oxide synthase
  • neuropathic pain such as that induced by chronic constriction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characterized by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark” neurons) occurs.
  • CCI chronic constriction injury
  • PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis. Specifically, colitis was induced in rats by intraluminal administration of the hapten trmitrobenzene sulfonic acid in 50% ethanol. Treated rats received 3- aminobenzamide, a specific inhibitor of PARP activity. Inhibition of PARP activity reduced the inflammatory response and restored the morphology and the energetic status of the distal colon.
  • PARP inhibitors are useful for treating arthritis. Further, PARP inhibitors appear to be useful for treating diabetes. PARP inhibitors have been shown to be useful for treating endotoxic shock or septic shock.
  • PARP inhibitors have also been used to extend the lifespan and proliferative capacity of cells including treatment of diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, ADDS, and other immune senescence disease; and to alter gene expression of senescent cells.
  • diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, ADDS, and other immune senescence disease.
  • PARP inhibitors such as 3-amino benzamide, affect overall DNA repair in response, for example, to hydrogen peroxide or ionizing radiation.
  • PARP inhibitors have been reported to be effective in radiosensiu ' zing (hypoxic) tumor cells and effective in preventing tumor cells from recovering from potentially lethal and sublethal damage of DNA after radiation therapy, presumably by their ability to prevent DNA strand break rejoining and by affecting several DNA damage signaling pathways.
  • the present invention provides compounds, compositions for, and methods of, inhibiting PARP activity for treating cancer and/or preventing cellular, tissue and/or organ damage resulting from cell damage or death due to, for example, necrosis or apoptosis.
  • the compounds and compositions of the present invention are especially useful in enhancing the effectiveness of chemotherapy and radiotherapy where a primary effect of the treatment is that of causing DNA damage in the targeted cells.
  • EP 669919 published on June 9, 1994, discloses dimethylbenzofurans and dimethylbenzopyrans as 5-HT3 antagonists. More in particular compounds No. 8, 4, 5, 10, 11, 12, 13, 15, 16, 17 and 14 of the present application are disclosed.
  • the disclosed compounds have gastrointestinal motility stimulating properties.
  • compounds No. 8, 6 and 9 of the present application are disclosed.
  • EP 885190 published on December 23, 1998 discloses 1,4-disubstituted piperidine derivatives having gastrokinetic properties.
  • compound No. 7 of the present application is disclosed.
  • EP 1036073 published on June 17, 1999, discloses substituted quinazolinedione derivatives. The described compounds have fundic relaxation properties.
  • EP 1355888 published on 20 June 2002 discloses quinazolinone derivatives as PARP inhibitors.
  • This invention concerns compounds of formula (I)
  • X is > ⁇ - or>CH-
  • L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-Ci_ 6 alkanediyl-, -C(O)-O-Ci_ 6 alkanediyl- or -Ci- ⁇ alkanediyl-;
  • R 1 is hydrogen, halo
  • R 2 is hydrogen, hydroxy, Q ⁇ alkyloxy or aminocarbonyl
  • R 10 is phenyl
  • R 3 is hydrogen, or Ci-galkyloxy; Z is amino, cyano or a radical selected from
  • each R 5 , R 6 , R 7 and R 8 is independently selected from hydrogen, halo, amino, Ci.6alkyl or Ci -6 alkyloxy; or
  • R 7 and R 8 taken together may form a bivalent radical of formula
  • each R 9 is independently selected from hydrogen or Ci.6alkyl
  • alkanediyl- and Z is the radical (b-2) and R 7 and R 8 taken together form a bivalent radical of formula (c-1), (c-2) or (c-3) then R 5 is other than chloro.
  • halo is generic to fluoro, chloro, bromo and iodo
  • C j ⁇ alkyl defines straight and branched chain saturated hydrocarbon radicals having from 1 to 6 carbon atoms such as, e.g.
  • C j _ 6 alkanediyl defines bivalent straight and branched chained saturated hydrocarbon radicals having from 1 to 6 carbon atoms such as, for example, methylene, 1,2-ethanediyl, 1,3-propanediyl 1,4-butanediyl, 1,5-pentanediyl, 1,6-hexanediyl and the branched isomers thereof such as, 2-methylpentanediyl, 3-methylpentanediyl, 2,2-dimethylbutanediyl, 2,3-dimethylbutanediyl and the like.
  • pharmaceutically acceptable salts means pharmaceutically acceptable acid or base addition salts.
  • the pharmaceutically acceptable acid or base addition salts as mentioned hereinabove are meant to comprise the therapeutically active non-toxic acid and non-toxic base addition salt forms which the compounds of formula (I) are able to form.
  • the compounds of formula (I) which have basic properties can be converted in their pharmaceutically acceptable acid addition salts by treating said base form with an appropriate acid.
  • Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g.
  • hydrochloric or hydrobromic acid sulfuric; nitric; phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic, malonic, succinic (Le. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic ⁇ -toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids.
  • organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic, malonic, succinic (Le. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzene
  • the compounds of formula (I) which have acidic properties may be converted in their pharmaceutically acceptable base addition salts by treating said acid form with a suitable organic or inorganic base.
  • Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • the terms acid or base addition salt also comprise the hydrates and the solvent addition forms which the compounds of formula (I) are able to form.
  • stereochemically isomeric forms of compounds of formula (I), as used hereinbefore, defines all possible compounds made up of the same atoms bonded by the same sequence of bonds but having different three-dimensional structures which are not interchangeable, which the compounds of formula (I) may possess.
  • the chemical designation of a compound encompasses the mixture of all possible stereochemically isomeric forms which said compound may possess. Said mixture may contain all diastereomers and/or enantiomers of the basic molecular structure of said compound.
  • All stereochemically isomeric forms of the compounds of formula (I) both in pure form or in admixture with each other are intended to be embraced within the scope of the present invention.
  • N-oxide forms of the compounds of formula (I) are meant to comprise those compounds of formula (I) wherein one or several nitrogen atoms are oxidized to the so-called N-oxide, particularly those N-oxides wherein one or more of the piperidine- or piperazine nitrogens are N-oxidized.
  • GB 1062357 discloses quinazolone derivatives having antihypertensive effects.
  • DE 2258561 discloses substituted pyridinone derivatives with antihypertensive " • action.
  • EP 13612 discloses substituted piperidinylalkylquinazoline derivatives that are serotonin-antagonists.
  • EP 669919 discloses dimethylbenzofurans and dimethylbenzopyrans as 5-HT3 antagonists.
  • US 5374637 discloses benzamide derivatives that have gastrointestinal motility stimulating properties.
  • EP 885190 discloses 1,4-di substituted piperidine derivatives having gastrokinetic properties.
  • EP 1036073 discloses substituted quinazolinedione derivatives that have fundic relaxation properties. Unexpectedly, it has been found that the compounds of the present invention show PARP inhibitory activity.
  • a first group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) each X is > ⁇ -; b) L is a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-C].
  • R 1 is hydrogen; d) R 2 is hydroxy, or aminocarbonyl; e) Z is amino, cyano or a radical selected from (b-1), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); f) each R 5 and R 6 is independently selected from hydrogen or amino.
  • a second group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) X is >CH-; b) L is a direct bond or a bivalent radical selected from -C(O)-, -NH-, -C(O)-Ci_6alkanediyl-, or -Cugalkanediyl-; c) Z is amino, cyano or a radical selected from (b-1), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R 5 is independently selected from hydrogen, fluoro, iodo, bromo, amino, Ci ⁇ alkyl or Ci -6 alkyloxy; e) each R 6 is independently selected from hydrogen, chloro, iodo, bromo, amino, C ⁇ alkyl or Cj. 6 alkyloxy .
  • a third group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond or a bivalent radical selected from -C(O)-, or -C(O)-NH-; b) R 2 is hydrogen, hydroxy, or Q ⁇ alkyloxy; c) Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R 5 , R 6 , R 7 and R 8 is independently selected from hydrogen, halo, C ⁇ alkyl or
  • Ci- ⁇ alkyloxy; or e) R 7 and R 8 taken together may form a bivalent radical of formula (c-1), or (c-4).
  • a fourth group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, or -C(O)-O-Ci -6 alkanediyl-; b) R 2 is hydrogen, hydroxy, or Ci -6 alkyloxy; c) Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R 5 , R , R 7 and R 8 is independently selected from hydrogen, halo, amino, Ci- 6 alkyl or Ci -6 alkyloxy; or e) R 7 and R 8 taken together may form a bivalent radical of formula (c-1), (c-2), (c-3) or (c-4).
  • a fifth group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond; b) R 1 is hydrogen, halo or Ci ⁇ alkyl; c) R 2 is hydrogen; d) R 3 is hydrogen; e) Z is a radical selected from (b-5) or (b-7); f) each R 5 is independently selected from hydrogen or halo.
  • a group of preferred compounds consists of those compounds of formula (I) wherein L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, or -C(O)-O-Ci- 6 alkanediyl-; R 2 is hydrogen, hydroxy, or Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); each R 5 , R 6 , R 7 and R 8 is independently selected from hydrogen, halo, amino, Ci ⁇ alkyl or C ⁇ alkyloxy; or R 7 and R 8 taken together may form a bivalent radical of formula (c-1), (c-2), (c-3) or (c-4).
  • a group of more preferred compounds consists of those compounds of formula (I) wherein
  • L is a direct bond
  • R 1 is hydrogen, halo or Ci ⁇ alkyl
  • R 2 is hydrogen; R 3 is hydrogen; Z is a radical selected from (b-5) or (b-7); and each R 5 is independently selected from hydrogen or halo.
  • the most preferred compounds are compounds No. 35, No. 36, No. 39, No. 1 and No. 43.
  • the compounds of formula (I) can be prepared according to the general methods described in EP 1036073, EP 885190, US 5374637, EP 669919 andEP13612.
  • the starting materials and some of the intermediates are known compounds and are 10 commercially available or may be prepared according to conventional reaction procedures generally known in the art.
  • the compounds of formula (I), can be prepared by reacting an intermediate of formula (II), with an intermediate of formula (JJT), wherein W is an appropriate leaving group such as, for example, halo, e.g. fluoro, chloro, bromo or iodo, or a sulfonyloxy radical such as methylsulfonyloxy, 4-methylphenylsulfonyloxy and the like.
  • W is an appropriate leaving group such as, for example, halo, e.g. fluoro, chloro, bromo or iodo, or a sulfonyloxy radical such as methylsulfonyloxy, 4-methylphenylsulfonyloxy and the like.
  • the reaction can be performed in a reaction-
  • inert solvent such as, for example, an alcohol, e.g. methanol, ethanol,
  • an appropriate base such as, for example, an alkali or earth alkaline metal carbonate or hydrogen carbonate,
  • 25 e.g. triethylamine or sodium carbonate
  • a small amount of an appropriate metal iodide, e.g., sodium or potassium iodide may be added to promote the reaction. Stirring may enhance the rate of the reaction.
  • the reaction may conveniently be carried out at a temperature ranging between room temperature and the reflux temperature of the reaction mixture and, if desired, the reaction may be carried out at an increased pressure.
  • the compounds of formula (I) may also be converted into each other via art-known reactions or functional group transformations. Some of such transformations are already described hereinabove. Other examples are hydrolysis of carboxylic esters to the corresponding carboxylic acid or alcohol; hydrolysis of amides to the corresponding carboxylic acids or amines; hydrolysis of nitriles to the corresponding amides; amino groups on imidazole or phenyl may be replaced by a hydrogen by art-known diazotation reactions and subsequent replacement of the diazo-group by hydrogen; alcohols may be converted into t esters and ethers; primary amines may be converted into secondary or tertiary amines; double bonds may be hydrogenated to the corresponding single bond; an iodo radical on a phenyl group may be converted in to an ester group by carbon monoxide insertion in the presence of a suitable palladium catalyst.
  • the present invention also relates to a compound of formula (I) as defined above for use as a medicine.
  • the compounds of the present invention have PARP inhibiting properties as can be seen from the experimental part hereinunder.
  • PARP poly-ADP-iibosylation activity
  • PARP encompass all proteins encoded by a parp gene, mutants thereof, and alternative slice proteins thereof. Additionally, as used herein, the term “PARP” includes PARP analogues, homologues and analogues of other animals. The term “PARP”, includes but is not limited to PARP-I .
  • PARP-2, PARP-3, Vault-PARP (PARP-4), PARP-7 (TiPARP), PARP-8, PARP-9 (BaI), PARP-10, PARP-Il, PARP-12, PARP-13, PARP-14, PARP-15, PARP-16, TANK- 1 , TANK-2, and TANK-3 may be encompassed.
  • Compounds that inhibit both PARP-I and tankyrase 2 can have advantageous properties in that they have enhanced growth inhibiting activities in cancer cells.
  • the present invention also contemplates the use of compounds in the preparation of a medicament for the treatment of any of the diseases and disorders in an animal described herein, wherein said compounds are compounds of formula (I)
  • X is > ⁇ - or >CH-
  • L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-Ci_ 6 alkanediyl-, -C(O)-O-C )-6 alkanediyl- or -Ci. 6 alkanediyl-;
  • R 1 is hydrogen, halo, Ci- ⁇ alkyloxy or Ci ⁇ alkyl
  • R 2 is hydrogen, hydroxy, C ⁇ alkyloxy or aminocarbonyl; when X is substituted with R 2 than R 2 taken together with -L-Z can form a bivalent radical of formula
  • R 10 is phenyl
  • R 3 is hydrogen, or C[. 6 alkyloxy
  • Z is amino, cyano or a radical selected from
  • each R 5 , R 6 , R 7 and R 8 is independently selected from hydrogen, halo, amino,
  • Ci ⁇ alkyl or Ci -6 alkyloxy; or R 7 and R 8 taken together may form a bivalent radical of formula - CH 2 -CR 9 2 -O- (c-1),
  • the invention also concerns the use of a compound as described above for the manufacture of a medicament for the treatment of a disorder mediated through PARP.
  • the invention concerns the use of a compound as described above for the manufacture of a medicament for the treatment of a disorder mediated through PARP.
  • Compounds that inhibit both PARP-I and TANK-2 can have advantages properties in that they have enhanced growth inhibiting activities in cancer cells.
  • the compounds of the present invention may be used as reference compounds or tracer compounds in which case one of the atoms of the molecule may be replaced with, for instance, a radioactive isotope.
  • compositions of this invention an effective amount of a particular compound, in base or acid addition salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • a pharmaceutically acceptable carrier which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • These pharmaceutical compositions are desirably in unitary dosage form suitable, preferably, for administration orally, rectally, percutaneously, or by parenteral injection.
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed.
  • the carrier will usually comprise sterile water, at least in large part, though other ingredients, to aid solubility for example, may be included.
  • Injectable solutions may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed.
  • the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not cause a significant deleterious effect to the skin. Said additives may facilitate the administration to the skin and/or may be helpful for preparing the desired compositions.
  • These compositions may be administered in various ways, e.g., as a transdermal patch, as a spot-on, as an ointment.
  • Dosage unit form as used in the specification and claims herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like, and segregated multiples thereof.
  • the compounds of the present invention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis; can ameliorate neural or cardiovascular tissue damage, including that following focal ischemia, myocardial infarction, and reperfusion injury; can treat various diseases and conditions caused or exacerbated by PARP activity; can extend or increase the lifespan or proliferative capacity of cells; can alter the gene expression of senescent cells; can radiosensitize and/or chemosensitize cells.
  • inhibition of PARP activity spares the cells from energy loss, preventing, in the case of neural cells, irreversible depolarization of the neurons, and thus, provides neuroprotection'.
  • the present invention further relates to a method of administering a therapeutically effective amount of the above-identified compounds in an amount sufficient to inhibit PARP activity, to treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis, to effect a neuronal activity not mediated by NMDA toxicity, to effect a neuronal activity mediated by NMDA toxicity, to treat neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age- related muscular degeneration, AIDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, diabetes, head trauma, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as n
  • the present invention relates to a method of treating, preventing or inhibiting a neurological disorder in an animal, which comprises administering to said animal a therapeutically effective amount of the above-identified compounds.
  • the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration.
  • the present invention also contemplates the use of compounds of formula (I) for inhibiting PARP activity, for treating, preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis, for treating, preventing or inhibiting a neurological disorder in an animal.
  • preventing neurodegeneration includes the ability to prevent neurodegeneration in patients newly diagnosed as having a neurodegenerative disease, or at risk of developing a new degenerative disease and for preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease.
  • treatment covers any treatment of a disease and/or condition in an animal, particularly a human, and includes: (i) preventing a disease and/or condition from occurring in a subject which may be predisposed to the disease and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease and/or condition, i.e., arresting its development; (iii) relieving the disease and/or condition, i.e., causing regression of the disease and/or condition.
  • radiosensitizer is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to ionizing radiation and/or to promote the treatment of diseases which are treatable with ionizing radiation.
  • Diseases which are treatable with ionizing radiation include neoplastic diseases, benign and malignant turaors, and cancerous cells. Ionizing radiation treatment of other diseases not listed herein are also contemplated by the present invention.
  • chemosensitizer is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of cells to chemotherapy and/or promote the treatment of diseases which are treatable with chemotherapeutics.
  • Diseases which are treatable with chemotherapy include neoplastic diseases, benign and malignant tmors and cancerous cells. Chemotherapy treatment of other diseases not listed herein are also contemplated by the present invention.
  • the compounds, compositions and methods of the present invention are particularly useful for treating or preventing tissue damage resulting from cell death or damage due to necrosis or apoptosis.
  • the compounds of the present invention can be "anti-cancer agents", which term also encompasses "anti-tumor cell growth agents” and "anti-neoplastic agents”.
  • the methods of the invention are useful for treating cancers and chemosensitizing and/or radiosensitizing tumor cells in cancers such as ACTH- producing tumors, acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, Ewih'g's sarcoma gallbladder cancer, hairy cell leukemia, head &neck cancer, Hodgkin's lymphoma, Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non- Hod
  • the compounds of the present invention can be used as "radiosensitizer” and/or “chemosensitizer”.
  • Radiosensitizers are known to increase the sensitivity of cancerous cells to the toxic effects of ionizing radiation.
  • hypoxic cell radiosensitizers e.g., 2- nitroimidazole compounds, and benzotriazine dioxide compounds
  • non-hypoxic cell radiosensitizers e.g., halogenated pyrimidines
  • various other potential mechanisms of action have been hypothesized for radiosensitizers in the treatment of disease.
  • radiosensitizers include, but are not limited to, the following: metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, EO9, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5- iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same.
  • Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the sensitizing agent.
  • photodynamic radiosensitizers include the following, but are not limited to: hematoporphyrin derivatives, Photofrin, benzoporphyrin derivatives, tin etioporphyrin, pheoborbide-a, bacteriochlorophyll-a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same.
  • Radiosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to: compounds which promote the incorporation of radiosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemotherapeutic agents which act on the tumor with or without additional radiation; or other therapeutically effective compounds for treating cancer or other disease.
  • radiosensitizers examples include, but are not limited to: 5-fluorouracil, leucovorin, 5' -amino- 5'deoxythymidine, oxygen, carbogen, red celltransfusions, perfluorocarbons (e.g., Fluosol 10 DA), 2,3-DPG, BW12C, calcium channel blockers, pentoxyfylline, antiangiogenesis compounds, hydralazine, and LBSO.
  • chemotherapeutic agents that may be used in conjunction with radiosensitizers include, but are not limited to: adriamycin, camptothecin, carboplatin, cisplatin, daunorubicin, docetaxel, doxorubicin, interferon (alpha, beta, gamma), interleukin 2, irinotecan, paclitaxel, topotecan, and therapeutically effective analogs and derivatives of the same.
  • Chemosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to : compounds which promote the incorporation of chemosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemothearpeutic agents which act on the tumor or other therapeutically effective compounds for treating cancer or other disease.
  • additional therapeutical agents that may be used in conjunction with chemosensitizers include, but are not limited to : methylating agents, toposisomerase I inhibitors and other chemotherapeutic agents such as cisplatin and bleomycin.
  • the compounds of formula (I) can also be used to detect or identify the PARP, and more in particular the PARP-I receptor.
  • the compounds of formula (I) can be labeled.
  • Said label can be selected from the group consisting of a radioisotope, spin label, antigen label, enzyme label fluorescent group or a chemiluminiscent group.
  • an effective amount would be from 0.001 mg/kg to 100 mg/kg body weight, and in particular from 0.005 mg/kg to 10 mg/kg body weight. It may be appropriate to administer the required dose as two, three, four or more sub-doses at appropriate intervals throughout the day. Said sub-doses may be formulated as unit dosage forms, for example, containing 0.05 to 500 mg, and in particular 0.1 mg to 200 mg of active ingredient per unit dosage form.
  • DCM dichloromethane
  • DMF is defined as NJf- dimethylformamide
  • MIK is defined as methyl isobutyl keton
  • MEK is defined as methyl ethyl keton
  • THF triethylamine
  • Table F-I lists the compounds that were prepared according to one of the above Examples.
  • SPA In vitro Scintillation Proximity Assay
  • the assay relies upon the well established SPA technology for the detection of poly(ADP-ribosy])ation of biotinylated target proteins, i.e histones.
  • This ribosylation is induced using nicked DNA activated PARP-I enzyme and [ 3 H]-nicotmamide adenine dinucleotide ([ 3 H]-NAD + ) as ADP-ribosyl donor.
  • DNAse buffer 10 mM Tris-HCl, pH 7.4; 0.5 mg/ml Bovine Serum Albumine (BSA); 5 mM MgCl 2 .6H 2 O and 1 mM KCl
  • DNAse solution lmg/ml in 0.15 M NaCl
  • the reaction mixture was cooled on ice and dialysed at 4 ° C for respectively 1.5 and 2 hours against 1.5 1 of 0.2 M KCl, and twice against 1.5 1 of 0.01 M KCl for 1.5 and 2 h respectively.
  • the mixture was aliquoted and stored at -20 0 C.
  • Histones (1 mg/ml, type II-A, supplier: Sigma) were biotinylated using the biotinylation kit of Amersham and stored aliquoted at - 20 °C.
  • a stock solution of [ 3 H]-NAD + was made by adding 120 ⁇ l of [ 3 H]-NAD + (0.1 mCi/ml, supplier: NEN) to 6 ml incubation buffer (50 mM Tris/HCl, pH 8; 0.2 mM DTT; 4 mM MgCl 2 ).
  • a solution of 4 mM NAD + (supplier: Roche) was made in incubation buffer (from a 100 mM stock solution in water stored at - 20 0 C).
  • the PARP-I enzyme was produced using art known techniques, i.e. cloning and expression of the protein starting from human liver cDNA.
  • the final concentrations in the incubation mixture were 2 ⁇ g/ml for the biotinylated histones, 2 mg/ml for the PVT-SPA beads, 2 ⁇ g/ml for the nicked DNA and between 5 - 10 ⁇ g/ml for the PARP-I enzyme.
  • the reaction was terminated by adding 100 ⁇ of 4 mM NAD + in incubation buffer (final concentration 2 mM) and plates were mixed.
  • the blank value was subtracted from both the control and the sample values.
  • the control sample represented maximal PARP-I enzyme activity.
  • the amount of cpm was expressed as a percentage of the mean cpm value of the controls.
  • IC5 0 - values concentration of the drug, needed to reduce the PARP-I enzyme activity to 50% of the control
  • pICso the negative log value of the ICso-value.
  • 4-amino-l,8- naphthalimide was included to validate the SPA assay. The tested compounds showed inhibitory activity at the initial test concentration of 10 ⁇ 5 M (see Tabel-2).
  • a mixture of histones (stock solution: 5 mg/ml in H 2 O), NAD + (stock solution: 100 mM in H 2 O), and [ 32 P]-NAD + in incubation buffer (50 mM Tris/HCl, pH 8; 0.2 mM DTT; 4 mM MgCl 2 ) was made.
  • a mixture of the PARP-I enzyme (5 - 10 ⁇ g/ml) and nicked DNA was also made. The nicked DNA was prepared as described in the in vitro SPA for PARP-I inhibitory activity.
  • SPA In vitro Scintillation Proximity Assay
  • Compounds of the present invention were tested in an in vitro assay based on SPA technology with Ni Flash plates (96 or 384 well).
  • the assay relies upon SPA technology for the detection of auto-poly(ADP-ribosyl)ation of TANK-2 protein using [ 3 H]-nicotinamide adenine dinucleotide ([ 3 H]-NAD + ) as ADP-ribosyl donor.
  • a stock solution of [ 3 H]-NAD + ZNAD was made by adding 64.6 ⁇ l of [ 3 H]-NAD + (0.1 mCi/ml, supplier: Perkin Elmer) and 46.7 ⁇ l NAD-stock (10.7 mM, stored at - 20 0 C, supplier Roche) to 1888.7 ⁇ l assay buffer (60 mM Tris/HCl, pH 7.4; 0.9 mM DTT; 6 mM MgCl 2 ).
  • the TANK-2 enzyme was produced as described in EP1238063 .
  • the amount of cpm was expressed as a percentage of the mean cpm value of the controls.
  • ICso-values concentration of the drug, needed to reduce the TANK-2 enzyme activity to 50% of the control
  • pICso the negative log value of the ICso-value
  • 3-aminobenzamide and 4-amino-l,8-naphtali ⁇ nide were included to validate the SPA assay.
  • the assay was described using 96-well plates. In the assay using 384-welI plates the same final concentrations were used and volumes were adapted. If 96-well plate results were available these results were incorporated in Table-2, otherwise the results from the 384-well plate assay were shown.
  • the compounds can be further evaluated in a cellular chemo- and/or radiosensitization assay, an assay measuring inhibition of endogenous PARP-1 activity in cancer cell lines and eventually in an in vivo radiosensitization test.

Abstract

The present invention provides compounds of formula (I), their use as PARP inhibitors as well as pharmaceutical compositions comprising said compounds of formula (I) wherein R1, R2, R3, L, X, Y and Z have defined meanings.

Description

QUINAZOLINONE DERIVATIVES AS PARP INHIBITORS
Field of the invention The present invention relates to inhibitors of PARP and provides compounds and compositions containing the disclosed compounds. Moreover, the present invention provides methods of using the disclosed PARP inhibitors for instance as a medicine.
Background of the invention The nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-I) is a member of the PARP enzyme family. This growing family of enzymes consist of PARPs such as, for example: PARP-I, PARP-2, PARP-3 and Vault-PARP; and Tankyrases (TANKs), such as, for example: TANK-I, TANK-2 and TANK-3. PARP is also referred to as poly(adenosine 5'-diphospho-ribose) polymerase or PARS (poly(ADP-ribose) synthetase).
PARP-I is a major nuclear protein of 116 kDa consisting of three domains : the N- terminal DNA binding domain containing two zinc fingers, the automodification domain and the C-terminal catalytic domain. It is present in almost all eukaryotes. The enzyme synthesizes poly(ADP-ribose), a branched polymer that can consist of over 200 ADP-ribose units. The protein acceptors of poly(ADP-ribose) are directly or indirectly involved in maintaining DNA integrity. They include histones, topoisomerasςs.j.DNA and RNA polymerases, DNA ligases, and Ca2+- and Mg2+-dependent endonucleases. PARP protein is expressed at a high level in many tissues, most notably in the immune system, heart, brain and germ-line cells. Under normal physiological conditions, there is minimal PARP activity. However, DNA damage causes an immediate activation of PARP by up to 500-fold.
Tankyrases (TANKs) were identified as components of the human telomeric complex. They have also been proposed to have a role in vesicle trafficking and may serve as scaffolds for proteins involved in various other cellular processes. Telomeres, which are essential for chromosome maintenance and stability, are maintained by telomerase, a specialized reverse transcriptase. TANKs are (ADP-ribose)transferases with some features of both signalling and cytoskeletal proteins. They contain the PARP domain, which catalyses poly-ADP-ribosylation of substrate proteins, the sterile alpha motif, which is shared with certain signalling molecules and the ANK domain, which contains 24 ankyrin repeats homologues to the cytoskeletal protein ankyrin. The ANK domain interacts with a telomeric protein, Telomere Repeat binding Factor-1 (TRF-I). These proteins were therefore named TRFl -interacting, ankyrin-related ADP-ribose polymerase (TANKs) .
One of the more specific functions of TANK is the ADP-ribosylation of TRF-I . Human telomere function requires two telomere-specific DNA binding proteins, TRF-I and TRF-2. TRF-2 protects chromosome ends, and TRF-I regulates telomere length. ADP-ribosylation inhibits the ability of TRF-I to bind to telomeric DNA. This poly-ADP-ribosylation of TRF-I releases TRF-I from the telomeres, opening up the telomeric complex and allow access to telomerase. Therefore, TANK functions as a positive regulator of telomere length., allowing elongation of the telomeres by telomerase.
Among the many functions attributed to PARP, and especially PARP-I, is its major role in facilitating DNA repair by ADP-ribosylation and therefore co-ordinating a number of DNA repair proteins. As a result of PARP activation, NAD+ levels significantly decline. Extensive PARP activation leads to severe depletion of NAD+ in cells suffering from massive DNA damage. The short half-life of poly(ADP-ribose) results in a rapid turnover rate. Once poly(ADP-ribose) is formed, it is quickly degraded by the constitutively active poly(ADP-ribose) glycohydrolase (PARG), together with phosphodiesterase and (ADP-ribose) protein lyase. PARP and PARG form a cycle that converts a large amount of '1KfAD+ to ADP-ribose. In less than an hour, over-stimulation of PARP can cause a drop of NAD+ and ATP to less than 20% of the normal level. Such a scenario is especially detrimental during ischaemia when deprivation of oxygen has already drastically compromised cellular energy output. Subsequent free radical production during reperfusion is assumed to be a major cause of tissue damage. Part of the ATP drop, which is typical in many organs during ischaemia and reperfusion, could be linked to NAD+ depletion due to poly(ADP-ribose) turnover. Thus, PARP or PARG inhibition is expected to preserve the cellular energy level thereby potentiating the survival of ischaemic tissues after insult.
Poly(ADP-ribose) synthesis is also involved in the induced expression of a number of genes essential for inflammatory response. PARP inhibitors suppress production of inducible nitric oxide synthase (iNOS) in macrophages, P-type selectin and intercellular adhesion molecule-1 (ICAM- 1) in endothelial cells. Such activity underlies the strong anti-inflammation effects exhibited by PARP inhibitors. PARP inhibition is able to reduce necrosis by preventing translocation and infiltration of neutrophils to the injured tissues.
PARP is activated by damaged DNA fragments and, once activated, catalyzes the attachment of up to 100 ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself. During major cellular stresses the extensive activation of PARP can rapidly lead to cell damage or death through depletion of energy stores. As four molecules of ATP are consumed for every molecule Of NAD+ regenerated, NAD+ is depleted by massive PARP activation, in the efforts to re-synthesize NAD+, ATP may also become depleted.
It has been reported that PARP activation plays a key role in both NMDA- and NO- induced neurotoxicity. This has been demonstrated in cortical cultures and in hippocampal slices wherein prevention of toxicity is directly correlated to PARP inhibition potency. The potential role of PARP inhibitors in treating neurodegenerative diseases and head trauma has thus been recognized even if the exact mechanism of action has not yet been elucidated.
Similarly, it has been demonstrated that single injections of PARP inhibitors have reduced the infarct size caused by ischemia and reperfusion of the heart or skeletal muscle in rabbits. In these studies, a single injection of 3-amino-benzamide (10 mg/kg), either one minute before occlusion or one minute before reperfusion, caused similar reductions irϊϊnfarct size in the heart (32-42%) while 1,5- dihydroxyisoquinoline &'"f (1 mg/kg), another PARP inhibitor, reduced infarct size by a comparable degree (38-48%) These results make it reasonable to assume that PARP inhibitors could salvage previously ischaemic heart or reperfusion injury of skeletal muscle tissue.
PARP activation can also be used as a measure of damage following neurotoxic insults resulting from exposure to any of the following inducers like glutamate (via NMDA receptor stimulation), reactive oxygen intermediates, amyloid β-protein, ; N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or its active metabolite
N-methyl-4 phenylpyridine (MPP+), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease. Other studies have continued to explore the role of PARP activation in cerebellar granule cells in vitro and in MPTP neurotoxicity. Excessive neural exposure to glutamate, which serves as the predominate central nervous system neurotransmitter and acts upon the N-methyl D-aspartate
(NMDA) receptors and other subtype receptors, most often occurs as a result of stroke or other neurodegenerative processes. Oxygen deprived neurons release glutamate in great quantities during ischaemic brain insult such as during a stroke or heart attack. This excess release of glutamate in turn causes over-stimulation (excitotoxicity) of N-methyl-D-aspartate (NMDA), AMPA, Kainate and MGR receptors, which open ion channels and permit uncontrolled ion flow (e.g., Ca2+ and Na+ into the cells and K+ out of the cells) leading to overstimulation of the neurons. The over-stimulated neurons secrete more glutamate, creating a feedback loop or domino effect which ultimately results in cell damage or death via the production of proteases, lipases and free radicals. Excessive activation of glutamate receptors has been implicated in various neurological diseases and conditions including epilepsy, stroke, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's disease, schizophrenia, chronic pain, ischemia and neuronal loss following hypoxia, hypoglycemia, ischemia, trauma, and nervous insult. Glutamate exposure and stimulation has also been implicated as a basis for compulsive disorders, particularly drug dependence. Evidence includes findings in many animal species, as well as in cerebral cortical cultures treated with glutamate or NMDA, that glutamate receptor antagonists (i.e., compounds which block glutamate from binding to or activating its receptor) block neural damage following vascular stroke. Attempts to prevent excitotoxicity by blocking NMDA, AMPA, Kainate and MGR receptors have proven difficult because each receptor has multiple sites to which glutamate may bind and hence finding an effective mix of antagonists or universal antagonist to prevent binding of glutamate to all of the receptor and allow testing of this theory, has been difficult. Moreover, many of the compositions that are effective in blocking the receptors are also toxic to animals. As such, there is presently no known effective treatment for glutamate abnormalities. The stimulation of NMDA receptors by glutamate, for example, activates the enzyme neuronal nitric oxide synthase (nNOS), leading to the formation of nitric oxide (NO), which also mediates neurotoxicity. NMDA neurotoxicity may be prevented by treatment with nitric oxide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro.
Another use for PARP inhibitors is the treatment of peripheral nerve injuries, and the resultant pathological pain syndrome known as neuropathic pain, such as that induced by chronic constriction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characterized by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark" neurons) occurs.
Evidence also exists that PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis. Specifically, colitis was induced in rats by intraluminal administration of the hapten trmitrobenzene sulfonic acid in 50% ethanol. Treated rats received 3- aminobenzamide, a specific inhibitor of PARP activity. Inhibition of PARP activity reduced the inflammatory response and restored the morphology and the energetic status of the distal colon.
Further evidence suggests that PARP inhibitors are useful for treating arthritis. Further, PARP inhibitors appear to be useful for treating diabetes. PARP inhibitors have been shown to be useful for treating endotoxic shock or septic shock.
PARP inhibitors have also been used to extend the lifespan and proliferative capacity of cells including treatment of diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, ADDS, and other immune senescence disease; and to alter gene expression of senescent cells.
It is also known that PARP inhibitors, such as 3-amino benzamide, affect overall DNA repair in response, for example, to hydrogen peroxide or ionizing radiation.
The pivotal role of PARP in the repair of DNA strand breaks is well established, especially when caused directly by ionizing radiation or, indirectly after enzymatic repair of DNA lesions induced by methylating agents, topoisomerases I inhibitors and other chemotherapeutic agents as cisplatin and bleomycin. A variety of studies using "knockout" mice, trans-dominant inhibition models (over-expression of the DNA- binding domain), antisense and small molecular weight inhibitors have demonstrated the role of PARP in repair and cell survival after induction of DNA damage. The inhibition of PARP enzymatic activity should lead to an enhanced sensitivity of the tumor cells towards DNA damaging treatments.
PARP inhibitors have been reported to be effective in radiosensiu'zing (hypoxic) tumor cells and effective in preventing tumor cells from recovering from potentially lethal and sublethal damage of DNA after radiation therapy, presumably by their ability to prevent DNA strand break rejoining and by affecting several DNA damage signaling pathways.
PARP inhibitors have been used to treat cancer. In addition, U.S. Patent No.5,177,075 discusses several isoquinolines used for enhancing the lethal effects of ionizing radiation or chemotherapeutic agents on tumor cells. Weltin et al., "Effect of 6(5 - Phenanthridinone), an Inhibitor of Poly(ADP-ribose) Polymerase, on Cultured Tumor Cells", Oncol. Res., 6:9, 399-403 (1994), discusses the inhibition of PARP activity, reduced proliferation of tumor cells, and a marked synergistic effect when tumor cells are co- treated with an alkylating drug.
Reviews of the state of the art has been published by Li and Zhang in IDrugs 2001 , 4(7): 804-812, by Ame et al in Bioassays 2004, 26: 882-883 and by Nguewa et al., in Progress in Biophysic & Molecular Biology 2005, 88: 143-172.
There continues to be a need for effective and potent PARP inhibitors, and more particularly PARP-I inhibitors which produce minimal side effects. The present invention provides compounds, compositions for, and methods of, inhibiting PARP activity for treating cancer and/or preventing cellular, tissue and/or organ damage resulting from cell damage or death due to, for example, necrosis or apoptosis. The compounds and compositions of the present invention are especially useful in enhancing the effectiveness of chemotherapy and radiotherapy where a primary effect of the treatment is that of causing DNA damage in the targeted cells.
Background prior art
GB 1062357 published on March 22, 1967 discloses quinazolone derivatives having antihypertensive effects.
DE 2258561 published on June 20,1973 discloses substituted pyridinone derivatives with antihypertensive action. EP 13612, published on November 11, 1983, discloses substituted piperidinylalkylquinazoline derivatives. The described compounds are serotonin- antagonists.
EP 669919, published on June 9, 1994, discloses dimethylbenzofurans and dimethylbenzopyrans as 5-HT3 antagonists. More in particular compounds No. 8, 4, 5, 10, 11, 12, 13, 15, 16, 17 and 14 of the present application are disclosed.
US 5374637, published on December 20, 1994, discloses benzamide derivatives.
The disclosed compounds have gastrointestinal motility stimulating properties. In particular compounds No. 8, 6 and 9 of the present application are disclosed.
EP 885190, published on December 23, 1998 discloses 1,4-disubstituted piperidine derivatives having gastrokinetic properties. In particular compound No. 7 of the present application is disclosed. EP 1036073, published on June 17, 1999, discloses substituted quinazolinedione derivatives. The described compounds have fundic relaxation properties. EP 1355888 published on 20 June 2002 discloses quinazolinone derivatives as PARP inhibitors.
Description of the invention
This invention concerns compounds of formula (I)
Figure imgf000008_0001
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereo- chemically isomeric forms thereof, wherein
the dotted lines represent optional bonds;
X is >Ν- or>CH-;
— N=Y- is -N-C(O)- or -N=CR4-, wherein R4 is hydroxy;
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-Ci_6alkanediyl-, -C(O)-O-Ci_6alkanediyl- or -Ci-βalkanediyl-;
R1 is hydrogen, halo,
Figure imgf000008_0002
R2 is hydrogen, hydroxy, Q^alkyloxy or aminocarbonyl;
when X is substituted with R2 than R2 taken together with -L-Z can form a bivalent radical of formula -C(O)-NH-CH2-NR10- (a-1)
wherein R10 is phenyl;
R3 is hydrogen, or Ci-galkyloxy; Z is amino, cyano or a radical selected from
Figure imgf000009_0001
wherein each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino, Ci.6alkyl or Ci-6alkyloxy; or
R7 and R8 taken together may form a bivalent radical of formula
Figure imgf000009_0002
-(CH2)S-O- (c-2),
-O-(CH2)2-O- (c-3) or -CH=CH-CH=CH- (c-4) wherein each R9 is independently selected from hydrogen or Ci.6alkyl;
with the proviso that when X is >N-, then Z is other than the radical (b-2) and when X is >CH- and L is -C(O)-NH- or
-C(O)-O-Ci .6alkanediyl- and Z is the radical (b-2) and R7 and R8 taken together form a bivalent radical of formula (c-1), (c-2) or (c-3) then R5 is other than chloro.
The compounds of formula (I) may also exist in their tautomeric forms. Such forms although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.
A number of terms used in the foregoing definitions and hereinafter are explained hereunder. These terms are sometimes used as such or in composite terms.
As used in the foregoing definitions and hereinafter, halo is generic to fluoro, chloro, bromo and iodo; Cj^alkyl defines straight and branched chain saturated hydrocarbon radicals having from 1 to 6 carbon atoms such as, e.g. methyl, ethyl, propyl, butyl, pentyl, hexyl, 1-methylethyl, 2-methylpropyl, 2-methyl-butyl, 2-methylpentyl and the like; Cj_6alkanediyl defines bivalent straight and branched chained saturated hydrocarbon radicals having from 1 to 6 carbon atoms such as, for example, methylene, 1,2-ethanediyl, 1,3-propanediyl 1,4-butanediyl, 1,5-pentanediyl, 1,6-hexanediyl and the branched isomers thereof such as, 2-methylpentanediyl, 3-methylpentanediyl, 2,2-dimethylbutanediyl, 2,3-dimethylbutanediyl and the like.
The term "pharmaceutically acceptable salts" means pharmaceutically acceptable acid or base addition salts. The pharmaceutically acceptable acid or base addition salts as mentioned hereinabove are meant to comprise the therapeutically active non-toxic acid and non-toxic base addition salt forms which the compounds of formula (I) are able to form. The compounds of formula (I) which have basic properties can be converted in their pharmaceutically acceptable acid addition salts by treating said base form with an appropriate acid. Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid; sulfuric; nitric; phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic, malonic, succinic (Le. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic^-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids. The compounds of formula (I) which have acidic properties may be converted in their pharmaceutically acceptable base addition salts by treating said acid form with a suitable organic or inorganic base. Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. The terms acid or base addition salt also comprise the hydrates and the solvent addition forms which the compounds of formula (I) are able to form. Examples of such forms are e.g. hydrates, alcoholates and the like. The term stereochemically isomeric forms of compounds of formula (I), as used hereinbefore, defines all possible compounds made up of the same atoms bonded by the same sequence of bonds but having different three-dimensional structures which are not interchangeable, which the compounds of formula (I) may possess. Unless otherwise mentioned or indicated, the chemical designation of a compound encompasses the mixture of all possible stereochemically isomeric forms which said compound may possess. Said mixture may contain all diastereomers and/or enantiomers of the basic molecular structure of said compound. All stereochemically isomeric forms of the compounds of formula (I) both in pure form or in admixture with each other are intended to be embraced within the scope of the present invention.
The N-oxide forms of the compounds of formula (I) are meant to comprise those compounds of formula (I) wherein one or several nitrogen atoms are oxidized to the so-called N-oxide, particularly those N-oxides wherein one or more of the piperidine- or piperazine nitrogens are N-oxidized.
Whenever used hereinafter, the term "compounds of formula (I)" is meant to include also the Ν-oxide forms, the pharmaceutically acceptable acid or base addition salts and all stereoisomeric forms.
GB 1062357 discloses quinazolone derivatives having antihypertensive effects. DE 2258561 discloses substituted pyridinone derivatives with antihypertensive " • action. EP 13612 discloses substituted piperidinylalkylquinazoline derivatives that are serotonin-antagonists. EP 669919 discloses dimethylbenzofurans and dimethylbenzopyrans as 5-HT3 antagonists. US 5374637 discloses benzamide derivatives that have gastrointestinal motility stimulating properties. EP 885190 discloses 1,4-di substituted piperidine derivatives having gastrokinetic properties. EP 1036073 discloses substituted quinazolinedione derivatives that have fundic relaxation properties. Unexpectedly, it has been found that the compounds of the present invention show PARP inhibitory activity.
A first group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) each X is >Ν-; b) L is a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-C].fialkanediyl-, -C(O)-O-Ci.ealkanediyl- or -C1-6alkanediyl-; c) R1 is hydrogen; d) R2 is hydroxy,
Figure imgf000012_0001
or aminocarbonyl; e) Z is amino, cyano or a radical selected from (b-1), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); f) each R5 and R6 is independently selected from hydrogen or amino.
A second group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) X is >CH-; b) L is a direct bond or a bivalent radical selected from -C(O)-, -NH-, -C(O)-Ci_6alkanediyl-, or -Cugalkanediyl-; c) Z is amino, cyano or a radical selected from (b-1), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R5 is independently selected from hydrogen, fluoro, iodo, bromo, amino, Ci^alkyl or Ci-6alkyloxy; e) each R6 is independently selected from hydrogen, chloro, iodo, bromo, amino, C^alkyl or Cj.6alkyloxy .
A third group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond or a bivalent radical selected from -C(O)-, or -C(O)-NH-; b) R2 is hydrogen, hydroxy, or Q^alkyloxy; c) Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R5, R6, R7 and R8 is independently selected from hydrogen, halo, C^alkyl or
Ci-βalkyloxy; or e) R7 and R8 taken together may form a bivalent radical of formula (c-1), or (c-4).
A fourth group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, or -C(O)-O-Ci-6alkanediyl-; b) R2 is hydrogen, hydroxy, or Ci-6alkyloxy; c) Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); d) each R5, R , R7 and R8 is independently selected from hydrogen, halo, amino, Ci- 6alkyl or Ci-6alkyloxy; or e) R7 and R8 taken together may form a bivalent radical of formula (c-1), (c-2), (c-3) or (c-4).
A fifth group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) L is a direct bond; b) R1 is hydrogen, halo or Ci^alkyl; c) R2 is hydrogen; d) R3 is hydrogen; e) Z is a radical selected from (b-5) or (b-7); f) each R5 is independently selected from hydrogen or halo.
A group of preferred compounds consists of those compounds of formula (I) wherein L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, or -C(O)-O-Ci-6alkanediyl-; R2 is hydrogen, hydroxy, or
Figure imgf000013_0001
Z is a radical selected from (b-2), (b-3), (b-4), (b-5), (b-6), (b-7), (b-8) or (b-9); each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino, Ci^alkyl or C^alkyloxy; or R7 and R8 taken together may form a bivalent radical of formula (c-1), (c-2), (c-3) or (c-4).
A group of more preferred compounds consists of those compounds of formula (I) wherein
L is a direct bond; R1 is hydrogen, halo or Ci^alkyl;
R2 is hydrogen; R3 is hydrogen; Z is a radical selected from (b-5) or (b-7); and each R5 is independently selected from hydrogen or halo.
The most preferred compounds are compounds No. 35, No. 36, No. 39, No. 1 and No. 43.
Figure imgf000013_0002
Compound 35 Compound 36
Figure imgf000014_0001
Compound 39 Compound 1
Figure imgf000014_0002
5 Compound 43
The compounds of formula (I) can be prepared according to the general methods described in EP 1036073, EP 885190, US 5374637, EP 669919 andEP13612. The starting materials and some of the intermediates are known compounds and are 10 commercially available or may be prepared according to conventional reaction procedures generally known in the art.
Some preparation methods will be described hereinafter in more detail. Other methods for obtaining final compounds of formula (T) are described in the examples.
,,.-.15 The compounds of formula (I), can be prepared by reacting an intermediate of formula (II), with an intermediate of formula (JJT), wherein W is an appropriate leaving group such as, for example, halo, e.g. fluoro, chloro, bromo or iodo, or a sulfonyloxy radical such as methylsulfonyloxy, 4-methylphenylsulfonyloxy and the like. The reaction can be performed in a reaction-
20 inert solvent such as, for example, an alcohol, e.g. methanol, ethanol,
2-methoxy-ethanol, propanol, butanol and the like; an ether, e.g. 4, 4-dioxane, l,l'-oxybispropane and the like; or a ketone, e.g. 4-methyl-2-pentanone, N,N-dimethylformamide, nitrobenzene and the like. The addition of an appropriate base such as, for example, an alkali or earth alkaline metal carbonate or hydrogen carbonate,
25 e.g. triethylamine or sodium carbonate, may be utilized to pick up the acid which is liberated during the course of the reaction. A small amount of an appropriate metal iodide, e.g., sodium or potassium iodide may be added to promote the reaction. Stirring may enhance the rate of the reaction. The reaction may conveniently be carried out at a temperature ranging between room temperature and the reflux temperature of the reaction mixture and, if desired, the reaction may be carried out at an increased pressure.
Figure imgf000015_0001
The compounds of formula (I) may also be converted into each other via art-known reactions or functional group transformations. Some of such transformations are already described hereinabove. Other examples are hydrolysis of carboxylic esters to the corresponding carboxylic acid or alcohol; hydrolysis of amides to the corresponding carboxylic acids or amines; hydrolysis of nitriles to the corresponding amides; amino groups on imidazole or phenyl may be replaced by a hydrogen by art-known diazotation reactions and subsequent replacement of the diazo-group by hydrogen; alcohols may be converted intot esters and ethers; primary amines may be converted into secondary or tertiary amines; double bonds may be hydrogenated to the corresponding single bond; an iodo radical on a phenyl group may be converted in to an ester group by carbon monoxide insertion in the presence of a suitable palladium catalyst.
The present invention also relates to a compound of formula (I) as defined above for use as a medicine.
The compounds of the present invention have PARP inhibiting properties as can be seen from the experimental part hereinunder.
The term "PARP" is used herein to mean a protein having poly-ADP-iibosylation activity. Within the meaning of this term, PARP encompass all proteins encoded by a parp gene, mutants thereof, and alternative slice proteins thereof. Additionally, as used herein, the term "PARP" includes PARP analogues, homologues and analogues of other animals. The term "PARP", includes but is not limited to PARP-I . Within the meaning of this term PARP-2, PARP-3, Vault-PARP (PARP-4), PARP-7 (TiPARP), PARP-8, PARP-9 (BaI), PARP-10, PARP-Il, PARP-12, PARP-13, PARP-14, PARP-15, PARP-16, TANK- 1 , TANK-2, and TANK-3 may be encompassed.
Compounds that inhibit both PARP-I and tankyrase 2 can have advantageous properties in that they have enhanced growth inhibiting activities in cancer cells.
The present invention also contemplates the use of compounds in the preparation of a medicament for the treatment of any of the diseases and disorders in an animal described herein, wherein said compounds are compounds of formula (I)
Figure imgf000016_0001
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereo- chemically isomeric forms thereof, wherein
the dotted lines represent optional bonds;
X is >Ν- or >CH-;
— N-- Υ— is -N-C(O)- or -N=CR4-, wherein R4 is hydroxy;
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-Ci_6alkanediyl-, -C(O)-O-C )-6alkanediyl- or -Ci.6alkanediyl-;
R1 is hydrogen, halo, Ci-βalkyloxy or Ci^alkyl;
R2 is hydrogen, hydroxy, C^alkyloxy or aminocarbonyl; when X is substituted with R2 than R2 taken together with -L-Z can form a bivalent radical of formula
-C(O)-NH-CH2-NR 10 - (a-1)
wherein R10 is phenyl;
R3 is hydrogen, or C[.6alkyloxy;
Z is amino, cyano or a radical selected from
Figure imgf000017_0001
(b-9) (b-10) (b-11)
wherein each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino,
Ci^alkyl or Ci-6alkyloxy; or R7 and R8 taken together may form a bivalent radical of formula - CH2-CR9 2-O- (c-1),
-(CHz)3-O- (c-2),
-O-(CH2)2-O- (c-3) or
-CH=CH-CH=CH- (c-4) wherein each R9 is independently selected from hydrogen or Chalky!. Furthermore, the invention also concerns the use of a compound as described above for the manufacture of a medicament for the treatment of a disorder mediated through PARP.
In particular, the invention concerns the use of a compound as described above for the manufacture of a medicament for the treatment of a disorder mediated through PARP.
Compounds that inhibit both PARP-I and TANK-2 can have advantages properties in that they have enhanced growth inhibiting activities in cancer cells.
In view of their PARP binding properties the compounds of the present invention may be used as reference compounds or tracer compounds in which case one of the atoms of the molecule may be replaced with, for instance, a radioactive isotope.
To prepare the pharmaceutical compositions of this invention, an effective amount of a particular compound, in base or acid addition salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirably in unitary dosage form suitable, preferably, for administration orally, rectally, percutaneously, or by parenteral injection. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. For parenteral compositions, the carrier will usually comprise sterile water, at least in large part, though other ingredients, to aid solubility for example, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. In the compositions suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not cause a significant deleterious effect to the skin. Said additives may facilitate the administration to the skin and/or may be helpful for preparing the desired compositions. These compositions may be administered in various ways, e.g., as a transdermal patch, as a spot-on, as an ointment. It is especially advantageous to formulate the aforementioned pharmaceutical compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used in the specification and claims herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like, and segregated multiples thereof.
The compounds of the present invention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis; can ameliorate neural or cardiovascular tissue damage, including that following focal ischemia, myocardial infarction, and reperfusion injury; can treat various diseases and conditions caused or exacerbated by PARP activity; can extend or increase the lifespan or proliferative capacity of cells; can alter the gene expression of senescent cells; can radiosensitize and/or chemosensitize cells. Generally, inhibition of PARP activity spares the cells from energy loss, preventing, in the case of neural cells, irreversible depolarization of the neurons, and thus, provides neuroprotection'.
For the foregoing reasons, the present invention further relates to a method of administering a therapeutically effective amount of the above-identified compounds in an amount sufficient to inhibit PARP activity, to treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis, to effect a neuronal activity not mediated by NMDA toxicity, to effect a neuronal activity mediated by NMDA toxicity, to treat neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age- related muscular degeneration, AIDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, diabetes, head trauma, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging, to extend the lifespan and proliferative capacity of cells; to alter gene expression of senescent cells; chemosensitize and/or radiosensitize (hypoxic) tumor cells. The present invention also relates to treating diseases and conditions in an animal which comprises administering to said animal a therapeutically effective amount of the above-identified compounds.
In particular, the present invention relates to a method of treating, preventing or inhibiting a neurological disorder in an animal, which comprises administering to said animal a therapeutically effective amount of the above-identified compounds. The neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration.
The present invention also contemplates the use of compounds of formula (I) for inhibiting PARP activity, for treating, preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis, for treating, preventing or inhibiting a neurological disorder in an animal.
The term "preventing neurodegeneration" includes the ability to prevent neurodegeneration in patients newly diagnosed as having a neurodegenerative disease, or at risk of developing a new degenerative disease and for preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease.
The term "treatment" as used herein covers any treatment of a disease and/or condition in an animal, particularly a human, and includes: (i) preventing a disease and/or condition from occurring in a subject which may be predisposed to the disease and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease and/or condition, i.e., arresting its development; (iii) relieving the disease and/or condition, i.e., causing regression of the disease and/or condition.
The term "radiosensitizer", as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to ionizing radiation and/or to promote the treatment of diseases which are treatable with ionizing radiation. Diseases which are treatable with ionizing radiation include neoplastic diseases, benign and malignant turaors, and cancerous cells. Ionizing radiation treatment of other diseases not listed herein are also contemplated by the present invention.
The term "chemosensitizer", as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of cells to chemotherapy and/or promote the treatment of diseases which are treatable with chemotherapeutics. Diseases which are treatable with chemotherapy include neoplastic diseases, benign and malignant tmors and cancerous cells. Chemotherapy treatment of other diseases not listed herein are also contemplated by the present invention.
The compounds, compositions and methods of the present invention are particularly useful for treating or preventing tissue damage resulting from cell death or damage due to necrosis or apoptosis. The compounds of the present invention can be "anti-cancer agents", which term also encompasses "anti-tumor cell growth agents" and "anti-neoplastic agents". For example, the methods of the invention are useful for treating cancers and chemosensitizing and/or radiosensitizing tumor cells in cancers such as ACTH- producing tumors, acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, Ewih'g's sarcoma gallbladder cancer, hairy cell leukemia, head &neck cancer, Hodgkin's lymphoma, Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non- Hodgkin's lymphoma, osteosarcoma, ovarian cancer, ovary (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, retinoblastoma, skin cancer, soft tissue sarcoma, squamous cell carcinomas, stomach cancer, testicular cancer, thyroid cancer, trophoblastic neoplasms, uterine cancer, vaginal cancer, cancer of the vulva and Wilm's tumor.
Hence the compounds of the present invention can be used as "radiosensitizer" and/or "chemosensitizer".
Radiosensitizers are known to increase the sensitivity of cancerous cells to the toxic effects of ionizing radiation. Several mechanisms for the mode of action of radiosensitizers have been suggested in the literature including: hypoxic cell radiosensitizers ( e.g., 2- nitroimidazole compounds, and benzotriazine dioxide compounds) mimicking oxygen or alternatively behave like bioreductive agents under hypoxia; non-hypoxic cell radiosensitizers (e.g., halogenated pyrimidines) can be analogs of DNA bases and preferentially incorporate into the DNA of cancer cells and thereby promote the radiation-induced breaking of DNA molecules and/or prevent the normal DNA repair mechanisms; and various other potential mechanisms of action have been hypothesized for radiosensitizers in the treatment of disease. Many cancer treatment protocols currently employ radiosensitizers in conjunction with radiation of x-rays. Examples of x-ray activated radiosensitizers include, but are not limited to, the following: metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, EO9, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5- iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same. Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the sensitizing agent. Examples of photodynamic radiosensitizers include the following, but are not limited to: hematoporphyrin derivatives, Photofrin, benzoporphyrin derivatives, tin etioporphyrin, pheoborbide-a, bacteriochlorophyll-a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same.
Radiosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to: compounds which promote the incorporation of radiosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemotherapeutic agents which act on the tumor with or without additional radiation; or other therapeutically effective compounds for treating cancer or other disease. Examples of additional therapeutic agents that may be used in conjunction with radiosensitizers include, but are not limited to: 5-fluorouracil, leucovorin, 5' -amino- 5'deoxythymidine, oxygen, carbogen, red celltransfusions, perfluorocarbons (e.g., Fluosol 10 DA), 2,3-DPG, BW12C, calcium channel blockers, pentoxyfylline, antiangiogenesis compounds, hydralazine, and LBSO. Examples of chemotherapeutic agents that may be used in conjunction with radiosensitizers include, but are not limited to: adriamycin, camptothecin, carboplatin, cisplatin, daunorubicin, docetaxel, doxorubicin, interferon (alpha, beta, gamma), interleukin 2, irinotecan, paclitaxel, topotecan, and therapeutically effective analogs and derivatives of the same. Chemosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to : compounds which promote the incorporation of chemosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemothearpeutic agents which act on the tumor or other therapeutically effective compounds for treating cancer or other disease. Examples of additional therapeutical agents that may be used in conjunction with chemosensitizers include, but are not limited to : methylating agents, toposisomerase I inhibitors and other chemotherapeutic agents such as cisplatin and bleomycin.
The compounds of formula (I) can also be used to detect or identify the PARP, and more in particular the PARP-I receptor. For that purpose the compounds of formula (I) can be labeled. Said label can be selected from the group consisting of a radioisotope, spin label, antigen label, enzyme label fluorescent group or a chemiluminiscent group.
Those skilled in the art could easily determine the effective amount from the test results presented hereinafter. In general it is contemplated that an effective amount would be from 0.001 mg/kg to 100 mg/kg body weight, and in particular from 0.005 mg/kg to 10 mg/kg body weight. It may be appropriate to administer the required dose as two, three, four or more sub-doses at appropriate intervals throughout the day. Said sub-doses may be formulated as unit dosage forms, for example, containing 0.05 to 500 mg, and in particular 0.1 mg to 200 mg of active ingredient per unit dosage form.
Experimental part Hereinafter, "DCM" is defined as dichloromethane, "DMF" is defined as NJf- dimethylformamide,"MeOH" is defined as methanol, "MIK" is defined as methyl isobutyl keton, "MEK" is defined as methyl ethyl keton, "TEA" is defined as triethylamine and "THF" is defined as tetrahydrofuran.
A. Preparation of the intermediate compounds
Example Al a) Preparation of intermediate 1
Figure imgf000024_0001
A mixture of 3-(l-piperazinyl)-l#-indazole (0.11 mol), chloro-acetonitrile (0.16 mol) and TEA (13g) in toluene (200 ml) and acetonitrile (200 ml) was stirred and refluxed for 3 hours. The cooled reaction mixture was washed with water (250 ml). The organic layer was separated , dried (MgSO4) , filtered and the solvent was evaporated. The residue was dissolved in trichloromethane and purified over silica on a glass filter (eluent: trichloromethane /MeOH 90/10). The purest fraction was collected and the solvent was evaporated. The residue was crystallized from acetonitrile. The crystals were filtered off and dried, yielding 26g (99%) of intermediate 1, melting point 1360C. bχPreparation.pf intermediate 2 K2t, N lf _J
N NH
A mixture of intermediate 1 (0.11 mol) in NBb/MeOH (600ml) was hydrogenated at 500C with Raney Nickel (4g) as a catalyst. After uptake Of H2 (2 eq), the catalyst was filtered off and the filtrate was evaporated. The residue was crystallized from acetontrile. The crystals were filtered off and dried, yielding 21g (77.5%) of intermediate 2, melting point 121°C.
Example A2 a) Preparation of intermediate 3 Λ Jl " ~* OCC
Phosphoryl chloride (110.9 ml) was added dropwise at 5°C to DMF (81.5 ml). The mixture was stirred until complete dissolution. 4-[(l-oxobutyl)amino]- benzoic acid, ethyl ester (0.34 mol) was added. The mixture was stirred at 1000C for 15 hours, then cooled to room temperature and poured out into ice water. The precipitate was filtered off, washed with water and dried, blP.rep.aration.o_f intømeΛate 4
Figure imgf000024_0002
A mixture of intermediate 3 (0.1606 mol) in sodium methylate, 30% solution in MeOH (152.8 ml) and MeOH (400ml) was stirred and refluxed for 15 hours, then cooled and poured out into ice water. The precipitate was filtered off, washed with water and taken up in DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated till dryness, yielding 31.64g (85%) of intermediate 4. .Q).Preparation.of mtermediate.5 ^^T^T^T^^
Lithium tetrahydroaluminate (0.1288 mol) was added portionwise at 00C under N2 flow to a solution of intermediate 4 (0.1288 mol) in THF (263ml). The mixture was stirred for 30 min, poured out into ice water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated till dryness, yielding 27.4g (98%) of intermediate 5. d).PreP-Kation.of Antermediate 6 JX ^~v^*\^^\
Methanesulfonyl chloride (0.104 mol) was added dropwise at 00C under N2 flow to a mixture of intermediate 5 (0.069 mol) and TEA (0.207 mol) in DCM (120ml). The mixture was stirred at 00C for 4 hours. The solvent was evaporated till dryness (without heating). The product was used without further purification, yielding 20.4g of intermediate 6.
Example A3
a) L.Preparation.of .intermediate.?
Figure imgf000025_0001
4-(2-aminoethyl)- 1-piperazinecarboxylic acid, ethyl ester (0.0586 mol) and 2- (methylthio)- 4(l#)-quinazolinone (0.0588 mol) were heated at 1800C for 2 hours while stirring upon treatment with javelle water and then taken up in DCM and MeOH. The solvent was evaporated till dryness. The residue was purified by column chromatography over silica gel (15-35 μm) (eluent: DCMTMeOHTNH4OH 94/6/0.5). The pure fractions were collected and the solvent was evaporated. The oily residue was crystallized from diethyl ether. The precipitate was filtered off and dried, yielding intermediate 7, melting point: 1380C. b}.Prep.aration.gflntermediate.8
Figure imgf000025_0002
A mixture of intermediate 7 (0.0223 mol) and potassium hydroxide (0.223 mol) in 2- propanol (100ml) was stirred and refluxed for 4 days. The solvent was evaporated till dryness. The residue was taken up in MeOH while stirring at 6O0C. The salts were filtered off. The solvent was evaporated, yielding 6.5g of intermediate 8. B. Preparation of the final compounds
Example Bl
Preparation of. compound..1
Figure imgf000026_0001
Intermediate 2 (0.000815 mol) and 6-chloro-2-(methylthio)- 4(lJϊ)-quinazolinone (0.00097 mol) were heated at 1600C for 1 hour, then taken up in water and potassium carbonate 10% and extracted with DCM/MeOH 90/10. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (0.3g) was purified by column chromatography over silica gel (15-40 μm) (eluent: DCMZMeOHZNH4OH 92/8/0.5). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from MEK and DIPE. The precipitate was filtered off and dried, yielding 0.2g (58%) of compound 1, melting point 186°C.
Example B2
Preparation of .compound 2
Figure imgf000026_0002
A mixture of l-(3-aminopropyl)-4-(4-chlorophenyl)- 4-piperidinol (0.015 mol) and 2- chloro- 4(l//)-quinazolinone (0.018 mol) in dimethylacetamide (5 ml) was stirred at 1200C for 1 hour. The reaction mixture was cooled , dissolved in DCM and washed with aqueous ammonia. The organic layer was separated , dried (MgSO4) , filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (eluent: DCM/(MeOH/NH3) 92/8). The pure fractions were collected and the solvent was evaporated. The residue was suspended in DIPE. The precipitate was filtered off and dried (vacuum; 700C), yielding 3.72g (60%) of compound 2, melting point 178.40C.
Example B3
Preparation, of.compound.3
Figure imgf000026_0003
A mixture of intermediate 6 (0.0124 mol), intermediate 8 (0.0137 mol) and potassium carbonate (0.0373 mol) in DMF (80ml) was stirred at 6O0C for 1 hour, poured out into ice water and stirred at room temperature for 30 min. The precipitate was filtered off, washed with water and taken up in 2-propanone. The precipitate was filtered off and dried, yielding 1.5g (26%) of compound 3, melting point 118°C.
Table F-I lists the compounds that were prepared according to one of the above Examples.
Figure imgf000027_0001
Ex.
[Bl]; mp.
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Pharmacological example
In vitro Scintillation Proximity Assay (SPA") for PAKP-I inhibitory activity Compounds of the present invention were tested in an in vitro assay based on SPA technology (proprietary"Jto Amersham Pharmacia Biotech).
In principle, the assay relies upon the well established SPA technology for the detection of poly(ADP-ribosy])ation of biotinylated target proteins, i.e histones. This ribosylation is induced using nicked DNA activated PARP-I enzyme and [3H]-nicotmamide adenine dinucleotide ([3H]-NAD+) as ADP-ribosyl donor.
As inducer of PARP-I enzyme activity, nicked DNA was prepared. For this, 25 mg of DNA (supplier: Sigma) was dissolved in 25 ml DNAse buffer (10 mM Tris-HCl, pH 7.4; 0.5 mg/ml Bovine Serum Albumine (BSA); 5 mM MgCl2.6H2O and 1 mM KCl) to which 50 μl DNAse solution (lmg/ml in 0.15 M NaCl) was added. After an incubation of 90 min. at 37 0C, the reaction was terminated by adding 1.45 g NaCl, followed by a further incubation at 58 0C for 15 min. The reaction mixture was cooled on ice and dialysed at 4 ° C for respectively 1.5 and 2 hours against 1.5 1 of 0.2 M KCl, and twice against 1.5 1 of 0.01 M KCl for 1.5 and 2 h respectively. The mixture was aliquoted and stored at -20 0C. Histones (1 mg/ml, type II-A, supplier: Sigma) were biotinylated using the biotinylation kit of Amersham and stored aliquoted at - 20 °C. A stock solution of 100 mg/ml SPA polyvinyl toluene) (PVT) beads (supplier: Amersham) was made in PBS. A stock solution of [3H]-NAD+ was made by adding 120 μl of [3H]-NAD+ (0.1 mCi/ml, supplier: NEN) to 6 ml incubation buffer (50 mM Tris/HCl, pH 8; 0.2 mM DTT; 4 mM MgCl2). A solution of 4 mM NAD+ (supplier: Roche) was made in incubation buffer (from a 100 mM stock solution in water stored at - 200C). The PARP-I enzyme was produced using art known techniques, i.e. cloning and expression of the protein starting from human liver cDNA. Information concerning the used protein sequence of the PARP-I enzyme including literature references can be found in the Swiss-Prot database under primary accession number P09874. Biotinylated histones and PVT-SPA beads were mixed and pre-incubated for 30 min. at room temperature. PARP-I enzyme (concentration was lot dependent) was mixed with the nicked DNA and the mixture was pre-incubated for 30 min. at 4 0C. Equal parts of this histones/PVT-SPA beads solution and PARP-I enzyme/DNA solution were mixed and 75 μl of this mixture together with 1 μl of compound in DMSO and 25 μl of [3H]-NAD+ was added per well into a 96-well microtiterplate. The final concentrations in the incubation mixture were 2 μg/ml for the biotinylated histones, 2 mg/ml for the PVT-SPA beads, 2 μg/ml for the nicked DNA and between 5 - 10 μg/ml for the PARP-I enzyme. After incubation of the mixture for 15 min. at room temperature, the reaction was terminated by adding 100 μ\ of 4 mM NAD+ in incubation buffer (final concentration 2 mM) and plates were mixed.
The beads were allowed to sediment for at least 15 min. and plates transferred to a TopCountNXT™ (Packard) for scintillation counting, values were expressed as counts per minute (cpm). For each experiment, controls (containing PARP-I enzyme and DMSO without compound), a blank incubation (containing DMSO but no PARP-I enzyme or compound) and samples (containing PARP-I enzyme and compound dissolved in DMSO) were run in parallel. All compounds tested were dissolved and eventually further diluted in DMSO. In first instance, compounds were tested at a concentration of 10"5 M. When the compounds showed activity at 10"5M, a dose- response curve was made wherein the compounds were tested at concentrations between 10"5M and 10"8M. In each test, the blank value was subtracted from both the control and the sample values. The control sample represented maximal PARP-I enzyme activity. For each sample, the amount of cpm was expressed as a percentage of the mean cpm value of the controls. When appropriate, IC50- values (concentration of the drug, needed to reduce the PARP-I enzyme activity to 50% of the control) were computed using linear interpolation between the experimental points just above and below the 50 % level. Herein the effects of test compounds are expressed as pICso (the negative log value of the ICso-value). As a reference compound, 4-amino-l,8- naphthalimide was included to validate the SPA assay. The tested compounds showed inhibitory activity at the initial test concentration of 10~5 M (see Tabel-2).
In vitro filtration assay for PARP-I inhibitory activity
Compounds of the present invention were tested in an in vitro filtration assay assessing PARP-I activity (triggered in the presence of nicked DNA) by means of its histone poly (ADP-ribosyl)ation activity using [32P]-NAD as ADP-ribosyl donor. The radioactive ribosylated histones were precipitated by trichloroacetic acid (TCA) in 96-well filterplates and the incorporated [32P] measured using a scintillation counter
A mixture of histones (stock solution: 5 mg/ml in H2O), NAD+ (stock solution: 100 mM in H2O), and [32P]-NAD+ in incubation buffer (50 mM Tris/HCl, pH 8; 0.2 mM DTT; 4 mM MgCl2) was made. A mixture of the PARP-I enzyme (5 - 10 μg/ml) and nicked DNA was also made. The nicked DNA was prepared as described in the in vitro SPA for PARP-I inhibitory activity. Seventy-five μl of the PARP-I enzyme/DNA mixture together with 1 μl of compound in DMSO and 25 μl of histones-NAD+/[32P]-NAD+ mixture was added per well of a 96-well filterplate (0.45 μm, supplier Millipore). The final concentrations in the incubation mixture were 2 μg/ml for the histones, 0.1 mM for the NAD+, 200 μM (0.5 μC) for the [32P]-NAD+ and 2 μg/ml for the nicked DNA. Plates were incubated for 15 min. at room temperature and the reaction was terminated by the addition of 10 μl ice cold 100% TCA followed by the addition of 10 μl ice-cold BSA solution (1 % in H2O). The protein fraction was allowed to precipitate for 10 min. at 4 0C and plates were vacuum filtered . The plates were subsequently washed with, for each well, 1 ml of 10 % ice cold TCA, 1 ml of 5 % ice cold TCA and 1 ml of 5 % TCA at room temperature. Finally 100 μl of scintillation solution (Microscint 40, Packard) was added to each well and the plates were transferred to a TopCountNXT™ (supplier: Packard) for scintillation counting and values were expressed as counts per minute (cpm). For each experiment, controls (containing PARP-I enzyme and DMSO without compound), a blank incubation (containing DMSO but no PARP-I enzyme or compound) and samples (containing PARP-I enzyme and compound dissolved in DMSO) were run in parallel. All compounds tested were dissolved and eventually further diluted in DMSO. In first instance, compounds were tested at a concentration of 10"5M. When the compounds showed activity at 10"5M, a dose-response curve was made wherein the compounds were tested at concentrations between 10"5M and 10"8M. In each test, the blank value was subtracted from both the control and the sample values. The control sample represented maximal PARP-I enzyme activity. For each sample, the amount of cpm was expressed as a percentage of the mean cpm value of the controls. When appropriate, ICso-values (concentration of the drug, needed to reduce the PARP-I enzyme activity to 50% of the control) were computed using linear interpolation between the experimental points just above and below the 50 % level. Herein the effects of test compounds are expressed as pICso (the negative log value of the IC50-value). As a reference compound, 4-amino-l,8-naphthalimide was included to validate the filtration assay. The tested compounds showed inhibitory activity at the initial test concentration of 10"5M (see Tabel-2).
In vitro Scintillation Proximity Assay (SPA) for TANK-2 inhibitory activity Compounds of the present invention were tested in an in vitro assay based on SPA technology with Ni Flash plates (96 or 384 well). In principle, the assay relies upon SPA technology for the detection of auto-poly(ADP-ribosyl)ation of TANK-2 protein using [3H]-nicotinamide adenine dinucleotide ([3H]-NAD+) as ADP-ribosyl donor.
A stock solution of [3H]-NAD+ZNAD was made by adding 64.6 μl of [3H]-NAD+ (0.1 mCi/ml, supplier: Perkin Elmer) and 46.7 μl NAD-stock (10.7 mM, stored at - 200C, supplier Roche) to 1888.7 μl assay buffer (60 mM Tris/HCl, pH 7.4; 0.9 mM DTT; 6 mM MgCl2). The TANK-2 enzyme was produced as described in EP1238063 . 60 μl of assay buffer, together with 1 μl of compound in DMSO, 20 μl of [3H]-NAD+/NAD and 20 μl of TANK-2 enzyme (final concentration 6 μg/ml) was added per well into a 96-well Ni-coated flash plate (Perkin Elmer). After incubation of the mixture for 120 min. at room temperature, the reaction was terminated by adding 60 μl of stopsolution (42.6 mg NAD in 6 ml H2O). The plates were covered with a plate sealer and placed in a TopCountNXT™ (Packard) for scintillation counting. Values were expressed as counts per minute (cpm). For each experiment, controls (containing TANK-2 enzyme and DMSO without compound), a blank incubation (containing DMSO but no TANK-2 enzyme or compound) and samples (containing TANK-2 enzyme and compound dissolved in DMSO) were run in parallel. All compounds tested were dissolved and eventually further diluted in DMSO. In first instance, compounds were tested at a concentration of 10"5M. When the compounds showed activity at 10"5M, a dose- response curve was made wherein the compounds were tested at concentrations between 10"5M and 10"8M. Di each test, the blank value was subtracted from both the control and the sample values. The control sample represented maximal TANK-2 enzyme activity. For each sample, the amount of cpm was expressed as a percentage of the mean cpm value of the controls. When appropriate, ICso-values (concentration of the drug, needed to reduce the TANK-2 enzyme activity to 50% of the control) were computed using linear interpolation between the experimental points just above and below the 50 % level. Herein the effects of test compounds are expressed as pICso (the negative log value of the ICso-value). As reference compounds, 3-aminobenzamide and 4-amino-l,8-naphtaliτnide were included to validate the SPA assay. Herein the assay was described using 96-well plates. In the assay using 384-welI plates the same final concentrations were used and volumes were adapted. If 96-well plate results were available these results were incorporated in Table-2, otherwise the results from the 384-well plate assay were shown.
Tabel-2
Figure imgf000034_0001
Figure imgf000035_0001
The compounds can be further evaluated in a cellular chemo- and/or radiosensitization assay, an assay measuring inhibition of endogenous PARP-1 activity in cancer cell lines and eventually in an in vivo radiosensitization test.

Claims

1. A compound of formula (I),
Figure imgf000036_0001
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereo- chemically isomeric forms thereof, wherein
the dotted lines represent optional bonds;
X is >Ν- or >CH-;
__ N^-Y- is _N_c(O)- or -N=CR4-, wherein R4 is hydroxy;
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-Ci-6alkanediyl-, -C(O)-O-Ci-6alkanediyl- or -Ci-6alkanediyl-;
R1 is hydrogen, halo, Q-βalkyloxy or Q^alkyl;
R2 is hydrogen, hydroxy, Ci^alkyloxy or aminocarbonyl;
when X is substituted with R2 than R2 taken together with -L-Z can form a bivalent radical of formula -C(O)-NH-CH2-NR10- (a-1)
wherein R10is phenyl;
R3 is hydrogen, or Ci-βalkyloxy;
Z is amino, cyano or a radical selected from
Figure imgf000037_0001
wherein each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino,
Ci-ealkyl or Ci-ealkyloxy; or R7 and R8 taken together may form a bivalent radical of formula
- CH2-CR9 Z-O- (c-1),
-(CHa)3-O- (c-2),
-O-(CH2)2-O- (c-3) or
-CH=CH-CH=CH- (c-4) wherein each R9 is independently selected from hydrogen or Ci.6alkyl;
with the proviso that when X is >N-, then Z is other than the radical (b-2) and when X is >CH- and L is -C(O)-NH- Or
-C(O)-O-Ci-6alkaπediyl- and Z is the radical (b-2) and R7 and R8 taken together form a bivalent radical of formula (c-1), (c-2) or (c-3) then R5 is other than chloro.
2. A compound as claimed in claim 1 wherein
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, or -C(O)-O-Ci-6alkanediyl-; R is hydrogen, hydroxy, or Ci.ealkyloxy; Z is a radical selected from (b-2), (b-3), (b-4), Qy-S), (b-6), Qy-T), (b-8) or (b-9); each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino,
Figure imgf000038_0001
or R7 and R8 taken together may form a bivalent radical of formula (c-1), (c-2), (c-3) or (c-4).
3. A compound according to claim 1 and 2 wherein Lis a direct bond; R1 is hydrogen, halo or Ci^alkyl; R2 is hydrogen; R3 is hydrogen; Z is a radical selected from (b-5) or (b-7); and each
R >5 i •s independently selected from hydrogen or halo.
4. A compound according to claim 1,2 and 3 wherein the compound is selected from
Figure imgf000038_0002
Figure imgf000038_0003
5. A compound as claimed in any of claims 1 to 4 for use as a medicine.
6. A pharmaceutical composition comprising pharmaceutically acceptable carriers and , as an active ingredient a therapeutically effective amount of a compound as claimed in claim 1 to 4.
7. A process of preparing a pharmaceutical composition as claimed in claim 6 wherein the pharmaceutically acceptable carriers and a compound as claimed in claim 1 to 4 are intimately mixed.
8. Use of a compound for the manufacture of a medicament for the treatment of a PARP mediated disorder, wherein said compound is a compound of formula (I)
Figure imgf000039_0001
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereo- chemically isomeric forms thereof, wherein
the dotted lines represent optional bonds;
X is >Ν- or >CH~;
—N"— Υ— is -N-C(O)- or
Figure imgf000039_0002
wherein R4 is hydroxy;
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-, -C(O)-C]-6alkanediyl-, -C(O)-O-Ci-6alkanediyl- or -Q-ealkanediyl-;
R1 is hydrogen, halo, Ci^alkyloxy or Ci-6alkyl;
R2 is hydrogen, hydroxy, Ci-βalkyloxy or aminocarbonyl;
when X is substituted with R2 than R2 taken together with -L-Z can form a bivalent radical of formula -C(O)-NH-CH2-NR10- (a-1)
wherein R10 is phenyl;
R3 is hydrogen, or Ci^alkyloxy; Z is amino, cyano or a radical selected from
Figure imgf000040_0001
wherein each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino,
Ci-6alkyl or C].6alkyloxy; or
R7 and R8 taken together may form a bivalent radical of formula
Figure imgf000040_0002
-(CH2)S-O- (c-2), -0-(CHa)2-O- (c-3) or
-CH=CH-CH=CH- (c-4) wherein each R9 is independently selected from hydrogen or Ci-6alkyl.
9. Use according to claim 8 of a PARP inhibitor of formula (I) for the manufacture of a medicament for the treatment of a PARP-I mediated disorder.
10. Use according to claim 8 and 9 wherein the treatment involves chemosensitization.
11. Use according to claims 7 and 8 wherein the treatment involves radiosensitization.
12. A combination of a compound with a chemotherapeutic agent wherein said compound is a compound of formula (I)
Figure imgf000041_0001
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereo- chemically isomeric forms thereof, wherein
the dotted lines represent optional bonds;
X is >Ν- or >CH-;
— N-- Υ— is -N-C(O)- or -N=CR4-, wherein R4 is hydroxy;
L is a direct bond or a bivalent radical selected from -C(O)-, -C(O)-NH-, -NH-,
-C(O)-Ci-6alkanediyl-, -C(O)-O-Ci-6alkanediyl- or -Ci-βalkanediyl-;
R1 is hydrogen, halo,
Figure imgf000041_0002
R2 is hydrogen, hydroxy, Ci.6alkyloxy or aminocarbonyl;
when X is substituted with R2 than R2 taken together with -L-Z can form a bivalent radical of formula
-C(O)-NH-CH2-NR10- (a-1)
wherein R10 is phenyl;
R3 is hydrogen, or Q.galkyloxy;
Z is amino, cyano or a radical selected from
Figure imgf000042_0001
(b-9) (HQ) (b-11)
wherein each R5, R6, R7 and R8 is independently selected from hydrogen, halo, amino,
Chalky! or Ci^alkyloxy; or R7 and R8 taken together may form a bivalent radical of formula
Figure imgf000042_0002
-(CH2)B-O- (o-2),
-O-(CH2)2-O- (c-3) or
-CH=CH-CH=CH- (c-4) wherein each R9 is independently selected from hydrogen or Chalky!.
13. A process for preparing a compound as claimed in claim 1, characterized by reacting an intermediate of formula (S) with an intermediate of formula (HI), wherein W is an appropriate leaving group, with the formation of a compound of formula (I-a), wherein L1 is -Q-βalkanediyl-NH- and both dotted lines can be a bond, in a reaction-inert solvent and with the addition of an appropriate base,
Figure imgf000043_0001
PCT/EP2005/053029 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors WO2006003146A1 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
UAA200612700A UA85593C2 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
BRPI0512797-1A BRPI0512797A (en) 2004-06-30 2005-06-28 quinazolinone derivatives as parp inhibitors
CA2568835A CA2568835C (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
CN2005800222595A CN1980899B (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
KR1020067026804A KR101286969B1 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
US11/569,892 US8623872B2 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as PARP inhibitors
AT05769923T ATE498613T1 (en) 2004-06-30 2005-06-28 QUINAZOLINONE DERIVATIVES AS PARP INHIBITORS
MXPA06014543A MXPA06014543A (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors.
NZ551680A NZ551680A (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as PARP inhibitors
EP05769923A EP1771422B1 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
EA200700189A EA012837B1 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors
DE602005026391T DE602005026391D1 (en) 2004-06-30 2005-06-28 CHINAZOLINONE DERIVATIVES AS PARP HEMMER
JP2007518606A JP4969443B2 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as PARP inhibitors
AU2005259188A AU2005259188B2 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as PARP inhibitors
IL180411A IL180411A (en) 2004-06-30 2006-12-28 Quinazolinone derivatives as parp inhibitors, pharmaceutical compositions containing them, uses and methods of preparation thereof
NO20070555A NO20070555L (en) 2004-06-30 2007-01-30 Quinazolinone derivatives as PARP inhibitors
HK07111097.2A HK1105960A1 (en) 2004-06-30 2007-10-15 Quinazolinone derivatives as parp inhibitors
US14/077,881 US9522905B2 (en) 2004-06-30 2013-11-12 Quinazolinone derivatives as PARP inhibitors
US15/350,663 US10150757B2 (en) 2004-06-30 2016-11-14 Quinazolinone derivatives as PARP inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04076887.1 2004-06-30
EP04076887 2004-06-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/569,892 A-371-Of-International US8623872B2 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as PARP inhibitors
US14/077,881 Division US9522905B2 (en) 2004-06-30 2013-11-12 Quinazolinone derivatives as PARP inhibitors

Publications (1)

Publication Number Publication Date
WO2006003146A1 true WO2006003146A1 (en) 2006-01-12

Family

ID=34928320

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2005/053034 WO2006003150A1 (en) 2004-06-30 2005-06-28 Substituted 2-alkyl quinazolinone derivatives as parp inhibitors
PCT/EP2005/053029 WO2006003146A1 (en) 2004-06-30 2005-06-28 Quinazolinone derivatives as parp inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/053034 WO2006003150A1 (en) 2004-06-30 2005-06-28 Substituted 2-alkyl quinazolinone derivatives as parp inhibitors

Country Status (24)

Country Link
US (5) US8623872B2 (en)
EP (2) EP1763518B1 (en)
JP (2) JP4969443B2 (en)
KR (2) KR101286969B1 (en)
CN (2) CN1980899B (en)
AR (2) AR049656A1 (en)
AT (2) ATE498613T1 (en)
AU (2) AU2005259192C1 (en)
BR (2) BRPI0512790A (en)
CA (2) CA2568835C (en)
DE (1) DE602005026391D1 (en)
EA (2) EA012416B1 (en)
ES (2) ES2380702T3 (en)
HK (2) HK1105631A1 (en)
IL (2) IL180382A (en)
MX (2) MXPA06014543A (en)
MY (2) MY140572A (en)
NO (2) NO339883B1 (en)
NZ (2) NZ551840A (en)
SG (2) SG154434A1 (en)
TW (2) TWI361188B (en)
UA (2) UA86237C2 (en)
WO (2) WO2006003150A1 (en)
ZA (2) ZA200610775B (en)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008084261A1 (en) * 2007-01-10 2008-07-17 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors
US7732491B2 (en) 2007-11-12 2010-06-08 Bipar Sciences, Inc. Treatment of breast cancer with a PARP inhibitor alone or in combination with anti-tumor agents
WO2010081778A1 (en) 2009-01-17 2010-07-22 Universität Zürich Blockers of parp for the prevention and treatment of helicobacter pylori induced gastric cancer
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
US7994222B2 (en) 2006-09-05 2011-08-09 Bipar Sciences, Inc. Monitoring of the inhibition of fatty acid synthesis by iodo-nitrobenzamide compounds
US8143447B2 (en) 2006-09-05 2012-03-27 Bipar Sciences, Inc. Treatment of cancer
US8168644B2 (en) 2008-03-27 2012-05-01 Janssen Pharmaceutica Nv Quinazolinone derivatives as tubulin polymerization inhibitors
US8299256B2 (en) 2007-03-08 2012-10-30 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP and TANK inhibitors
WO2013012723A1 (en) * 2011-07-13 2013-01-24 Novartis Ag Novel 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
US8377985B2 (en) 2005-07-18 2013-02-19 Bipar Sciences, Inc. Treatment of cancer
US8404713B2 (en) 2007-10-26 2013-03-26 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP inhibitors
US8450486B2 (en) 2003-11-20 2013-05-28 Janssen Pharmaceutica, Nv 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US8524714B2 (en) 2003-11-20 2013-09-03 Janssen Pharmaceutica, Nv 7-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US8623884B2 (en) 2004-06-30 2014-01-07 Janssen Pharmaceutica, Nv Quinazolinedione derivatives as PARP inhibitors
US8889866B2 (en) 2008-03-27 2014-11-18 Janssen Pharmaceutica, Nv Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as PARP and tubulin polymerization inhibitors
US8946221B2 (en) 2004-06-30 2015-02-03 Janssen Pharmaceutica, Nv Phthalazine derivatives as PARP inhibitors
US9163003B2 (en) 2011-07-13 2015-10-20 Novartis Ag 4-piperidinyl compounds for use as tankyrase inhibitors
US9221804B2 (en) 2013-10-15 2015-12-29 Janssen Pharmaceutica Nv Secondary alcohol quinolinyl modulators of RORγt
US9227982B2 (en) 2011-07-13 2016-01-05 Novartis Ag 4-oxo-3,5,7,8-tetrahydro-4H-pyrano[4,3-d]pyrminidinyl compounds for use as tankyrase inhibitors
US9284308B2 (en) 2013-10-15 2016-03-15 Janssen Pharmaceutica Nv Methylene linked quinolinyl modulators of RORγt
US9290476B2 (en) 2012-10-16 2016-03-22 Janssen Pharmaceutica Nv Methylene linked quinolinyl modulators of RORγt
US9303015B2 (en) 2012-10-16 2016-04-05 Janssen Pharmaceutica Nv Heteroaryl linked quinolinyl modulators of RORγt
US9309222B2 (en) 2012-10-16 2016-04-12 Janssen Pharmaceutica Nv Phenyl linked quinolinyl modulators of RORγt
US9328095B2 (en) 2013-10-15 2016-05-03 Janssen Pharmaceutica Nv Heteroaryl linked quinolinyl modulators of RORgammat
US9346782B2 (en) 2013-10-15 2016-05-24 Janssen Pharmaceutica Nv Alkyl linked quinolinyl modulators of RORγt
US9403816B2 (en) 2013-10-15 2016-08-02 Janssen Pharmaceutica Nv Phenyl linked quinolinyl modulators of RORγt
US9624225B2 (en) 2013-10-15 2017-04-18 Janssen Pharmaceutica Nv Quinolinyl modulators of RORγt
WO2018022851A1 (en) 2016-07-28 2018-02-01 Mitobridge, Inc. Methods of treating acute kidney injury
WO2018085359A1 (en) 2016-11-02 2018-05-11 Immunogen, Inc. Combination treatment with antibody-drug conjugates and parp inhibitors
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
WO2018197461A1 (en) 2017-04-28 2018-11-01 Akribes Biomedical Gmbh A parp inhibitor in combination with a glucocorticoid and/or ascorbic acid and/or a protein growth factor for the treatment of impaired wound healing
CN110167926A (en) * 2016-12-30 2019-08-23 米托布里奇公司 Poly- ADP ribose polymerase (PARP) inhibitor
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
EP3594343A1 (en) 2015-07-23 2020-01-15 Institut Curie Use of a combination of dbait molecule and parp inhibitors to treat cancer
US10555941B2 (en) 2013-10-15 2020-02-11 Janssen Pharmaceutica Nv Alkyl linked quinolinyl modulators of RORγt
US10799501B2 (en) 2015-11-05 2020-10-13 King's College Hospital Nhs Foundation Trust Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1763518B1 (en) 2004-06-30 2012-01-11 Janssen Pharmaceutica NV Substituted 2-alkyl quinazolinone derivatives as parp inhibitors
WO2006047022A1 (en) 2004-10-25 2006-05-04 Virginia Commonwealth University Nuclear sulfated oxysterol, potent regulator of cholesterol homeostasis, for therapy of hypercholesterolemia, hyperlipidemia, and atherosclerosis
KR20150018765A (en) 2012-01-20 2015-02-24 데니스 브라운 Use of substituted hexitols including dianhydrogalactitol and analogs to treat neoplastic disease and cancer stem cells including glioblastoma multforme and medulloblastoma
PT2812323T (en) * 2012-02-09 2016-07-13 Merck Patent Gmbh Tetrahydro-quinazolinone derivatives as tank and parp inhibitors
EP2870140B8 (en) 2012-07-09 2016-09-28 Lupin Limited Tetrahydroquinazolinone derivatives as parp inhibitors
EP2983674A4 (en) 2013-04-08 2017-05-10 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds
AU2014293011A1 (en) 2013-07-26 2016-03-17 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene
BR112016001645B1 (en) * 2013-07-31 2023-04-11 Merck Patent Gmbh OXOQUINAZOLINYL-BUTANAMIDE DERIVATIVES, THEIR USE, MEDICINES
TW202348234A (en) 2013-12-24 2023-12-16 維吉尼亞聯邦大學 Uses of oxygenated cholesterol sulfates (ocs)
EP3125920B1 (en) 2014-04-04 2020-12-23 Del Mar Pharmaceuticals Dianhydrogalactitol, diacetyldianhydrogalactitol or dibromodulcitol to treat non-small-cell carcinoma of the lung and ovarian cancer
MA41168A (en) * 2014-12-17 2017-10-24 Acraf NEW ANTIBACTERIAL COMPOUNDS
CA2977685C (en) 2015-03-02 2024-02-20 Sinai Health System Homologous recombination factors
US20190290656A1 (en) 2015-12-30 2019-09-26 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd Pt(iv) prodrugs
WO2017156350A1 (en) 2016-03-09 2017-09-14 K-Gen, Inc. Methods of cancer treatment
EP4101861A1 (en) 2016-08-02 2022-12-14 Virginia Commonwealth University Compositions comprising 5-cholesten-3, 25-diol, 3-sulfate (25hc3s) or pharmaceutically acceptable salt thereof and at least one cyclic oligosaccharide
MX2020011465A (en) * 2018-04-30 2020-12-07 Ribon Therapeutics Inc Pyridazinones as parp7 inhibitors.
CN109251198B (en) * 2018-09-25 2019-08-23 郝维强 A kind of drug and its preparation method and application for anesthesia
WO2020118251A2 (en) * 2018-12-07 2020-06-11 The Board Of Trustees Of The Leland Stanford Junior University Hypoxia targeting compositions and combinations thereof with a parp inhibitor and methods of use thereof
CN113480536B (en) * 2021-04-21 2024-02-23 中国人民解放军海军军医大学 Spiropiperidone derivatives
CN115778960A (en) * 2022-08-19 2023-03-14 广州六顺生物科技有限公司 Application of heteroaromatic ring derivative in preparation of medicine for treating or preventing cachexia

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2258561A1 (en) * 1971-12-02 1973-06-20 Byk Gulden Lomberg Chem Fab BASIC SUBSTITUTED PYRIMIDONE (4) DERIVATIVES, THE PROCESS FOR THEIR PRODUCTION AND THE MEDICINAL PRODUCTS CONTAINING THEM
EP0013612A2 (en) * 1979-01-08 1980-07-23 Janssen Pharmaceutica N.V. (Piperidinylalkyl)quinazoline derivatives, process for their preparation and pharmaceutical compositions containing them
US5374637A (en) * 1989-03-22 1994-12-20 Janssen Pharmaceutica N.V. N-(3-hydroxy-4-piperidinyl)(dihydrobenzofuran, dihydro-2H-benzopyran or dihydrobenzodioxin)carboxamide derivatives
EP0669919A1 (en) * 1992-11-20 1995-09-06 Janssen Pharmaceutica N.V. Use of dimethylbenzofurans and dimethylbenzopyrans as 5-ht3 antagonists
EP0885190A1 (en) * 1996-02-29 1998-12-23 Janssen Pharmaceutica N.V. Novel n-substituted 4-((4'-aminobenzoyl)-oxymethyl)-piperidines having gastric prokinetic properties
WO2002048117A1 (en) * 2000-12-11 2002-06-20 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivatives
WO2003055865A1 (en) * 2001-12-24 2003-07-10 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivative

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB732581A (en) 1952-01-18 1955-06-29 Ciba Ltd Manufacture of hydrazine compounds
DE1006423B (en) 1952-01-18 1957-04-18 Ciba Geigy Process for the preparation of hydrazinophthalazines
US3274194A (en) 1963-03-29 1966-09-20 Miles Lab Quinazolinedione derivatives
GB1062357A (en) 1965-03-23 1967-03-22 Pfizer & Co C Quinazolone derivatives
GB1293565A (en) 1969-05-03 1972-10-18 Aspro Nicholas Ltd Aminophthalazines and pharmaceutical compositions thereof
US3919425A (en) 1971-04-09 1975-11-11 Miles Lab Method of producing vasodilation using certain 3-substituted-quinazoline derivatives
US3879393A (en) 1973-06-18 1975-04-22 Miles Lab Derivatives of 1,3-disubstituted 2,4(1h,3h)-quinazolinediones
FR2436781A1 (en) 1978-09-19 1980-04-18 Berri Balzac 3-Amino-(1h,3h)-quinazoline-2,4-di:one derivs. - useful as anxiolytics and sedatives to treat hypertension
JPS5976082A (en) 1982-10-23 1984-04-28 Kyowa Hakko Kogyo Co Ltd Novel piperidine derivative
JPS60120872A (en) 1983-12-01 1985-06-28 Kyowa Hakko Kogyo Co Ltd Novel heterocyclic compound and cardiotonic agent
US5001125A (en) 1984-03-26 1991-03-19 Janssen Pharmaceutica N.V. Anti-virally active pyridazinamines
PL147465B1 (en) 1984-03-26 1989-06-30 Janssen Pharmaceutica Nv Method of obtaining novel pyridazionoamine compounds
DK623586A (en) 1985-12-27 1987-06-28 Eisai Co Ltd PIPERIDE INGREDIENTS OR SALTS THEREOF AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE COMPOUNDS
US5231184A (en) 1987-11-23 1993-07-27 Janssen Pharmaceutica N.V. Pridazinamine derivatives
US5177075A (en) 1988-08-19 1993-01-05 Warner-Lambert Company Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process
GB8827822D0 (en) 1988-11-29 1988-12-29 Janssen Pharmaceutica Nv (1h-azol-1-ylmethyl)substituted quinoline derivatives
CA2002864C (en) 1988-11-29 1999-11-16 Eddy J. E. Freyne (1h-azol-1-ylmethyl) substituted quinoline, quinazoline or quinoxaline derivatives
EP0391462A1 (en) 1989-04-05 1990-10-10 Janssen Pharmaceutica N.V. Synergistic compositions containing ketanserin
US5160727A (en) 1990-02-13 1992-11-03 Warner-Lambert Company Tumor cell sensitization method using quinazolinedione derivatives
IE913473A1 (en) 1990-10-15 1992-04-22 Fujisawa Pharmaceutical Co Quinazoline derivatives and their preparation
WO1993022309A1 (en) 1992-04-23 1993-11-11 Merrell Dow Pharmaceuticals Inc. 4-imidomethyl-1-[2'phenyl-2'oxoethyl-] piperidines as serotonin 5ht2-antagonists, their preparation and use in therapy
WO1994019342A1 (en) 1993-02-18 1994-09-01 Kyowa Hakko Kogyo Co., Ltd. Adenosine incorporation inhibitor
GB9404485D0 (en) 1994-03-09 1994-04-20 Cancer Res Campaign Tech Benzamide analogues
JPH107572A (en) 1996-06-17 1998-01-13 Sumitomo Pharmaceut Co Ltd Agent for inhibiting production of tumor necrosis factor
DE59712953D1 (en) 1996-09-10 2008-09-04 Boehringer Ingelheim Pharma Modified amino acids, pharmaceutical compositions containing these compounds and methods for their preparation
CA2291630A1 (en) 1997-05-30 1998-12-03 Tetsutaro Niizato Nitrogen-containing heterocyclic compounds and therapeutic agents for hyperlipidemia comprising the same
JPH10330377A (en) 1997-06-02 1998-12-15 Kyowa Hakko Kogyo Co Ltd Piperidine derivative
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US20030069231A1 (en) 1999-10-12 2003-04-10 Klaus Rudolf Modified aminoacids, pharmaceuticals containing these compounds and method for their production
AU9002598A (en) 1997-09-16 1999-04-05 Takeda Chemical Industries Ltd. Nitrogenous fused-ring compounds, process for the preparation of the same, and drugs
US6133277A (en) 1997-12-05 2000-10-17 Janssen Pharmaceutica N.V. (Benzodioxan, benzofuran or benzopyran) derivatives having fundic relaxation properties
JP2000191659A (en) 1999-01-04 2000-07-11 Sumitomo Pharmaceut Co Ltd Tumor necrosis factor production inhibitor
DE60006213T2 (en) 1999-01-29 2004-07-29 Abbott Laboratories, Abbott Park DIAZABICYLODERIVATIVES AS NICOTINE-ACETYLCHOLINE RECEPTOR LIGANDS
US7265115B2 (en) 1999-01-29 2007-09-04 Abbott Laboratories Diazabicyclic CNS active agents
US6566372B1 (en) 1999-08-27 2003-05-20 Ligand Pharmaceuticals Incorporated Bicyclic androgen and progesterone receptor modulator compounds and methods
NZ524945A (en) 2000-10-02 2005-01-28 Janssen Pharmaceutica Nv Metabotropic glutamate receptor antagonists
ITMI20002358A1 (en) 2000-10-31 2002-05-01 Flavio Moroni TIENO DERIVATIVES, 2, 3-C | ISOCHINOLIN-3-ONE AS INHIBITORS OF POLY (DP-RIBOSE) POLYMERASE
JP2002284699A (en) 2001-03-28 2002-10-03 Sumitomo Pharmaceut Co Ltd Curative for visual cell degenerative disease
JPWO2002094790A1 (en) 2001-05-23 2004-09-09 三菱ウェルファーマ株式会社 Fused heterocyclic compounds and their pharmaceutical uses
EP1423120A4 (en) 2001-08-15 2005-12-28 Icos Corp 2h-phthalazin-1-ones and methods for use thereof
WO2003039460A2 (en) 2001-11-07 2003-05-15 Merck & Co., Inc. Mitotic kinesin inhibitors
AUPS137402A0 (en) 2002-03-26 2002-05-09 Fujisawa Pharmaceutical Co., Ltd. Novel tricyclic compounds
UA78548C2 (en) 2002-03-29 2007-04-10 Janssen Pharmaceutica Nv Radiolabelled quinoline and quinolinone derivatives and use thereof as metabotropic ligands of glutamate receptor
US7119111B2 (en) 2002-05-29 2006-10-10 Amgen, Inc. 2-oxo-1,3,4-trihydroquinazolinyl derivatives and methods of use
AR043059A1 (en) 2002-11-12 2005-07-13 Bayer Pharmaceuticals Corp DERIVATIVES OF INDOLIL PIRAZINONA USEFUL FOR THE TREATMENT OF HYPER-PROLIFERATIVE DISORDERS
US20050075364A1 (en) 2003-07-01 2005-04-07 Kap-Sun Yeung Indole, azaindole and related heterocyclic N-substituted piperazine derivatives
WO2005054201A1 (en) 2003-11-20 2005-06-16 Janssen Pharmaceutica N.V. 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
BRPI0416817A (en) 2003-11-20 2007-03-06 Janssen Pharmaceutica Nv Substituted 2-quinolinones and 7-phenylalkyl-2-quinoxalinones as poly (adp-ribose) polymerase inhibitors
DK1689715T3 (en) 2003-12-03 2011-05-23 Ym Biosciences Australia Pty tubulin inhibitors
EA010488B1 (en) 2003-12-05 2008-10-30 Янссен Фармацевтика Н.В. 6-substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
WO2005058843A1 (en) 2003-12-10 2005-06-30 Janssen Pharmaceutica N.V. Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
PE20060285A1 (en) 2004-03-30 2006-05-08 Aventis Pharma Inc PYRIDONES SUBSTITUTE AS POL (ADP-RIBOSA) -POLYMERASE (PARP) INHIBITORS
DE102004023332A1 (en) 2004-05-12 2006-01-19 Bayer Cropscience Gmbh Quinoxaline-2-one derivatives, crop protection agents containing them, and processes for their preparation and their use
CA2568622C (en) 2004-06-01 2016-01-26 University Of Virginia Patent Foundation Dual small molecule inhibitors of cancer and angiogenesis
EP1763518B1 (en) 2004-06-30 2012-01-11 Janssen Pharmaceutica NV Substituted 2-alkyl quinazolinone derivatives as parp inhibitors
WO2006003148A1 (en) 2004-06-30 2006-01-12 Janssen Pharmaceutica N.V. Quinazolinedione derivatives as parp inhibitors
BRPI0512902A (en) 2004-06-30 2008-04-15 Janssen Pharmaceutica Nv phthalazine derivatives as parp inhibitors
AU2006214164B2 (en) 2005-02-17 2010-12-09 Synta Pharmaceuticals Corp. Isoxazole combretastin derivatives for the treatment of disorders
JP5075624B2 (en) 2005-04-28 2012-11-21 田辺三菱製薬株式会社 Cyanopyridine derivatives and their pharmaceutical use
JP2009506060A (en) 2005-08-24 2009-02-12 イノテック ファーマシューティカルズ コーポレイション Indenoisoquinolinone analogs and methods for their use
DK1984333T3 (en) 2006-02-03 2012-07-23 Bionomics Ltd Substituted benzofurans, benzothiophenes, benzoselenophenes and indoles and their use as inhibitors of tubulin polymerization
US8546368B2 (en) 2006-02-15 2013-10-01 Abbvie Inc. Pyrazoloquinolones are potent PARP inhibitors
US8198448B2 (en) 2006-07-14 2012-06-12 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8466150B2 (en) 2006-12-28 2013-06-18 Abbott Laboratories Inhibitors of poly(ADP-ribose)polymerase
ES2381446T3 (en) 2007-03-08 2012-05-28 Janssen Pharmaceutica, N.V. Quinolinone derivative as PARP and TANK inhibitors
WO2009002469A1 (en) 2007-06-25 2008-12-31 Amgen Inc. Phthalazine compounds, compositions and methods of use

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2258561A1 (en) * 1971-12-02 1973-06-20 Byk Gulden Lomberg Chem Fab BASIC SUBSTITUTED PYRIMIDONE (4) DERIVATIVES, THE PROCESS FOR THEIR PRODUCTION AND THE MEDICINAL PRODUCTS CONTAINING THEM
EP0013612A2 (en) * 1979-01-08 1980-07-23 Janssen Pharmaceutica N.V. (Piperidinylalkyl)quinazoline derivatives, process for their preparation and pharmaceutical compositions containing them
US5374637A (en) * 1989-03-22 1994-12-20 Janssen Pharmaceutica N.V. N-(3-hydroxy-4-piperidinyl)(dihydrobenzofuran, dihydro-2H-benzopyran or dihydrobenzodioxin)carboxamide derivatives
EP0669919A1 (en) * 1992-11-20 1995-09-06 Janssen Pharmaceutica N.V. Use of dimethylbenzofurans and dimethylbenzopyrans as 5-ht3 antagonists
EP0885190A1 (en) * 1996-02-29 1998-12-23 Janssen Pharmaceutica N.V. Novel n-substituted 4-((4'-aminobenzoyl)-oxymethyl)-piperidines having gastric prokinetic properties
WO2002048117A1 (en) * 2000-12-11 2002-06-20 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivatives
WO2003055865A1 (en) * 2001-12-24 2003-07-10 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivative

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8524714B2 (en) 2003-11-20 2013-09-03 Janssen Pharmaceutica, Nv 7-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US8450486B2 (en) 2003-11-20 2013-05-28 Janssen Pharmaceutica, Nv 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US9255080B2 (en) 2004-06-30 2016-02-09 Janssen Pharmaceutica Nv Quinazolinedione derivatives as PARP inhibitors
US8946221B2 (en) 2004-06-30 2015-02-03 Janssen Pharmaceutica, Nv Phthalazine derivatives as PARP inhibitors
US8623884B2 (en) 2004-06-30 2014-01-07 Janssen Pharmaceutica, Nv Quinazolinedione derivatives as PARP inhibitors
US8377985B2 (en) 2005-07-18 2013-02-19 Bipar Sciences, Inc. Treatment of cancer
US7994222B2 (en) 2006-09-05 2011-08-09 Bipar Sciences, Inc. Monitoring of the inhibition of fatty acid synthesis by iodo-nitrobenzamide compounds
US8143447B2 (en) 2006-09-05 2012-03-27 Bipar Sciences, Inc. Treatment of cancer
EA016079B1 (en) * 2007-01-10 2012-01-30 Институто Ди Ричерке Ди Биолоджиа Молеколаре П. Анджелетти Спа Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors
KR20090097191A (en) * 2007-01-10 2009-09-15 이스티투토 디 리세르쉐 디 비올로지아 몰레콜라레 피. 안젤레티에스.피.에이. Amide substituted indazoles as poly(adp-ribose)polymerase(parp) inhibitors
EP3536690A1 (en) * 2007-01-10 2019-09-11 MSD Italia S.r.l. Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors
WO2008084261A1 (en) * 2007-01-10 2008-07-17 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors
EP2336120A1 (en) * 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
KR101591656B1 (en) 2007-01-10 2016-02-19 엠에스디 이탈리아 에스.알.엘. - Amide substituted indazoles as polyADP-ribosepolymerasePARP inhibitors
EP2805945A1 (en) * 2007-01-10 2014-11-26 MSD Italia S.r.l. Amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
US8299256B2 (en) 2007-03-08 2012-10-30 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP and TANK inhibitors
US8778966B2 (en) 2007-03-08 2014-07-15 Janssen Pharmaceutica, Nv Quinolinone derivatives as PARP and tank inhibitors
US8404713B2 (en) 2007-10-26 2013-03-26 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP inhibitors
US7732491B2 (en) 2007-11-12 2010-06-08 Bipar Sciences, Inc. Treatment of breast cancer with a PARP inhibitor alone or in combination with anti-tumor agents
US8889866B2 (en) 2008-03-27 2014-11-18 Janssen Pharmaceutica, Nv Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as PARP and tubulin polymerization inhibitors
US9598396B2 (en) 2008-03-27 2017-03-21 Janssen Pharmaceutica Nv Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as PARP and tubulin polymerization inhibitors
US9150540B2 (en) 2008-03-27 2015-10-06 Janssen Pharmaceutica Nv Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as parp and tubulin polymerization inhibitors
US8168644B2 (en) 2008-03-27 2012-05-01 Janssen Pharmaceutica Nv Quinazolinone derivatives as tubulin polymerization inhibitors
WO2010081778A1 (en) 2009-01-17 2010-07-22 Universität Zürich Blockers of parp for the prevention and treatment of helicobacter pylori induced gastric cancer
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
US9227982B2 (en) 2011-07-13 2016-01-05 Novartis Ag 4-oxo-3,5,7,8-tetrahydro-4H-pyrano[4,3-d]pyrminidinyl compounds for use as tankyrase inhibitors
CN103781776A (en) * 2011-07-13 2014-05-07 诺华股份有限公司 Novel 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
US9181266B2 (en) 2011-07-13 2015-11-10 Novartis Ag 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
US9163003B2 (en) 2011-07-13 2015-10-20 Novartis Ag 4-piperidinyl compounds for use as tankyrase inhibitors
WO2013012723A1 (en) * 2011-07-13 2013-01-24 Novartis Ag Novel 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
USRE46942E1 (en) 2011-07-13 2018-07-10 Novartis Ag 4-piperidinyl compounds for use as tankyrase inhibitors
US9290476B2 (en) 2012-10-16 2016-03-22 Janssen Pharmaceutica Nv Methylene linked quinolinyl modulators of RORγt
US9303015B2 (en) 2012-10-16 2016-04-05 Janssen Pharmaceutica Nv Heteroaryl linked quinolinyl modulators of RORγt
US9309222B2 (en) 2012-10-16 2016-04-12 Janssen Pharmaceutica Nv Phenyl linked quinolinyl modulators of RORγt
US10369146B2 (en) 2013-10-15 2019-08-06 Janssen Pharmaceutica Nv Phenyl linked quinolinyl modulators of RORγt
US9284308B2 (en) 2013-10-15 2016-03-15 Janssen Pharmaceutica Nv Methylene linked quinolinyl modulators of RORγt
US9346782B2 (en) 2013-10-15 2016-05-24 Janssen Pharmaceutica Nv Alkyl linked quinolinyl modulators of RORγt
US9624225B2 (en) 2013-10-15 2017-04-18 Janssen Pharmaceutica Nv Quinolinyl modulators of RORγt
US9403816B2 (en) 2013-10-15 2016-08-02 Janssen Pharmaceutica Nv Phenyl linked quinolinyl modulators of RORγt
US9328095B2 (en) 2013-10-15 2016-05-03 Janssen Pharmaceutica Nv Heteroaryl linked quinolinyl modulators of RORgammat
US10555941B2 (en) 2013-10-15 2020-02-11 Janssen Pharmaceutica Nv Alkyl linked quinolinyl modulators of RORγt
US10201546B2 (en) 2013-10-15 2019-02-12 Janssen Pharmaceutica Nv Quinolinyl modulators of RORγt
US9221804B2 (en) 2013-10-15 2015-12-29 Janssen Pharmaceutica Nv Secondary alcohol quinolinyl modulators of RORγt
EP3594343A1 (en) 2015-07-23 2020-01-15 Institut Curie Use of a combination of dbait molecule and parp inhibitors to treat cancer
US10799501B2 (en) 2015-11-05 2020-10-13 King's College Hospital Nhs Foundation Trust Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L
WO2018022851A1 (en) 2016-07-28 2018-02-01 Mitobridge, Inc. Methods of treating acute kidney injury
WO2018085359A1 (en) 2016-11-02 2018-05-11 Immunogen, Inc. Combination treatment with antibody-drug conjugates and parp inhibitors
CN110167926A (en) * 2016-12-30 2019-08-23 米托布里奇公司 Poly- ADP ribose polymerase (PARP) inhibitor
CN110167926B (en) * 2016-12-30 2022-09-13 米托布里奇公司 Poly-ADP ribose polymerase (PARP) inhibitors
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
WO2018197461A1 (en) 2017-04-28 2018-11-01 Akribes Biomedical Gmbh A parp inhibitor in combination with a glucocorticoid and/or ascorbic acid and/or a protein growth factor for the treatment of impaired wound healing
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
IL180382A (en) 2011-11-30
ATE498613T1 (en) 2011-03-15
BRPI0512797A (en) 2008-04-08
AU2005259188A1 (en) 2006-01-12
MY140572A (en) 2009-12-31
SG154435A1 (en) 2009-08-28
ZA200610772B (en) 2008-09-25
HK1105631A1 (en) 2008-02-22
US20170158673A1 (en) 2017-06-08
KR101286969B1 (en) 2013-07-23
UA85593C2 (en) 2009-02-10
EP1771422A1 (en) 2007-04-11
EP1763518B1 (en) 2012-01-11
UA86237C2 (en) 2009-04-10
EA012416B1 (en) 2009-10-30
ATE540936T1 (en) 2012-01-15
ES2361162T3 (en) 2011-06-14
AU2005259192C1 (en) 2012-03-01
CN1976906B (en) 2010-09-01
NO20070532L (en) 2007-01-29
NZ551680A (en) 2010-02-26
TWI361689B (en) 2012-04-11
NO339883B1 (en) 2017-02-13
CA2569826A1 (en) 2006-01-12
ZA200610775B (en) 2008-03-26
US20080176876A1 (en) 2008-07-24
KR101184036B1 (en) 2012-09-18
TWI361188B (en) 2012-04-01
CN1980899B (en) 2013-07-24
US20080070915A1 (en) 2008-03-20
AU2005259188B2 (en) 2011-03-31
US7875621B2 (en) 2011-01-25
IL180411A0 (en) 2007-06-03
EA200700189A1 (en) 2007-06-29
HK1105960A1 (en) 2008-02-29
US8623872B2 (en) 2014-01-07
BRPI0512790A (en) 2008-04-08
AU2005259192A1 (en) 2006-01-12
NO20070555L (en) 2007-01-30
EP1771422B1 (en) 2011-02-16
US10150757B2 (en) 2018-12-11
IL180411A (en) 2011-12-29
US20110077245A1 (en) 2011-03-31
DE602005026391D1 (en) 2011-03-31
KR20070031944A (en) 2007-03-20
EA012837B1 (en) 2009-12-30
TW200616978A (en) 2006-06-01
MXPA06014933A (en) 2007-02-28
CN1976906A (en) 2007-06-06
MXPA06014543A (en) 2007-03-23
CA2568835C (en) 2014-05-27
ES2380702T3 (en) 2012-05-17
EP1763518A1 (en) 2007-03-21
JP2008504347A (en) 2008-02-14
SG154434A1 (en) 2009-08-28
JP4836946B2 (en) 2011-12-14
US9522905B2 (en) 2016-12-20
US20140080835A1 (en) 2014-03-20
JP2008504350A (en) 2008-02-14
WO2006003150A1 (en) 2006-01-12
JP4969443B2 (en) 2012-07-04
AU2005259192B2 (en) 2011-04-28
EA200700188A1 (en) 2007-06-29
IL180382A0 (en) 2007-06-03
KR20070043968A (en) 2007-04-26
NZ551840A (en) 2009-07-31
CN1980899A (en) 2007-06-13
CA2568835A1 (en) 2006-01-12
AR051920A1 (en) 2007-02-21
CA2569826C (en) 2013-07-16
TW200608977A (en) 2006-03-16
AR049656A1 (en) 2006-08-23
US8188103B2 (en) 2012-05-29
MY148322A (en) 2013-03-29

Similar Documents

Publication Publication Date Title
US10150757B2 (en) Quinazolinone derivatives as PARP inhibitors
US9255080B2 (en) Quinazolinedione derivatives as PARP inhibitors
EP1771175B1 (en) Phthalazine derivatives as parp inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2568835

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 551680

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 11569892

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 12006502450

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/014543

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020067026804

Country of ref document: KR

Ref document number: 200610772

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2005769923

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007518606

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 180411

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 8001/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200580022259.5

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 2005259188

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200700189

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2005259188

Country of ref document: AU

Date of ref document: 20050628

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005259188

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1020067026804

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005769923

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11569892

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0512797

Country of ref document: BR