WO2005058843A1 - Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors - Google Patents

Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors Download PDF

Info

Publication number
WO2005058843A1
WO2005058843A1 PCT/EP2004/013165 EP2004013165W WO2005058843A1 WO 2005058843 A1 WO2005058843 A1 WO 2005058843A1 EP 2004013165 W EP2004013165 W EP 2004013165W WO 2005058843 A1 WO2005058843 A1 WO 2005058843A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
hydrogen
formula
compound
hydroxy
Prior art date
Application number
PCT/EP2004/013165
Other languages
French (fr)
Other versions
WO2005058843A8 (en
Inventor
Dominique Jean-Pierre Mabire
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 JP2006543409A priority Critical patent/JP4948178B2/en
Priority to KR1020067013344A priority patent/KR101149031B1/en
Priority to EA200601125A priority patent/EA010592B1/en
Priority to AU2004299183A priority patent/AU2004299183B2/en
Priority to EP04803192.6A priority patent/EP1694653B1/en
Priority to BRPI0417571-9A priority patent/BRPI0417571A/en
Priority to ES04803192.6T priority patent/ES2565581T3/en
Priority to US10/596,083 priority patent/US7652014B2/en
Application filed by Janssen Pharmaceutica N.V. filed Critical Janssen Pharmaceutica N.V.
Priority to NZ547278A priority patent/NZ547278A/en
Priority to CA2548273A priority patent/CA2548273C/en
Publication of WO2005058843A1 publication Critical patent/WO2005058843A1/en
Priority to IL176199A priority patent/IL176199A/en
Publication of WO2005058843A8 publication Critical patent/WO2005058843A8/en
Priority to NO20063129A priority patent/NO337621B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/44Benzopyrazines with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • 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
    • A61P19/08Drugs 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
    • 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
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • 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
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • 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
    • 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
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • 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
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/227Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 2
    • 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
    • C07D401/14Heterocyclic 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems

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.
  • the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is a member of the PARP enzyme family consisting of PARP-1 and several recently identified novel poly(ADP-ribosylating) enzymes.
  • PARP is also referred to as poly(adenosine 5'- diphospho-ribose) polymerase or PARS (poly(ADP-ribose) synthetase).
  • PARP-1 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, topoisomerases, 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.
  • PARP and PARG form a cycle that converts a large amount of NAD + to ADP-ribose.
  • 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.
  • 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.
  • the extensive activation of PARP can rapidly lead to cell damage or death through depletion of energy stores.
  • 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 N-methyl-4 phenylpyridine (MPP + ), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease.
  • 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 N-methyl-4 phenylpyridine (MPP + ), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease.
  • N- methyl-D-aspartate NMDA
  • AMPA N- methyl-D-aspartate
  • Kainate MGR receptors
  • open ion channels open ion channels and permit uncontrolled ion flow (e.g., Ca 2+ 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.
  • 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.
  • 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.
  • many of the compositions that are effective in blocking the receptors are also toxic to animals.
  • 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.
  • 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 trinitrobenzene 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 mo ⁇ hology 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, AIDS, 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, AIDS, 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 radiosensitizing (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 371564 published on June 6, 1990, discloses (lH-azol-1-ylmethyl) substituted quinoline, quinazoline or quinoxaline derivatives.
  • the described compounds suppress the plasma elimination of retinoic acids. More in particular 6-(cyclohexyl- lH-imidazol-l-ylmethyl)-3-methyl-2(lH)-quinoxalinone (Compound A) is disclosed.
  • This invention concerns compounds of formula (I)
  • Y is -N ⁇ or -CH ⁇
  • Q is -NH-, -0-, -C(O)-, -CH2-CH2- or -CHR 5 -, wherein R 5 is hydrogen, hydroxy, C ⁇ -6 alkyl, arylC ⁇ . 6 alkyl, Ci_ 6 alkyloxycarbonyl, Ci- ⁇ alkyloxyCi- ⁇ alkylamino or haloindazolyl;
  • R 1 is C ⁇ . alkyl or thienyl
  • R ,2 i • s . hyd .rogen or * ta dressing ⁇ ken t.oge *t ⁇ her wi :.th r R»3 may form O;
  • R is hydrogen, C 1-6 alkyl or a radical selected from • NR 6 R 7 (a-1), O-H (a-2), O-R 8 (a-3), S- R 9 (a-4), or — C ⁇ N (a-5), wherein R 6 is -CHO, C ⁇ -6 alkyl, hydroxyC 1-6 alkyl, C 1-6 alkylcarbonyl, di(d- 6 alkyl)a ⁇ mnod. 6 alkyl, C ⁇ -6 alkylcarbonylaminoC 1- 6alkyl, piperidinylC 1-6 alkyl, piperidinyld- ⁇ alkylaminocarbonyl, C 1-6 alkyloxy, thienylC ⁇ .
  • R 7 is hydrogen or C ⁇ -6 alkyl
  • R 8 is C ⁇ alkyl, d- ⁇ alkylcarbonyl or di(C ⁇ . 6 alkyl)aminoC 1-6 alkyl
  • R 9 is di(C ⁇ . 6 alkyl)aminoC 1-6 alkyl
  • R 3 is a group of formula -(CH 2 ) t -Z- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
  • each R 10 independently is hydrogen, d ⁇ alkyl, aminocarbonyl, hydroxy
  • aryl is phenyl or phenyl substituted with halo, d ⁇ alkyl or C ⁇ -6 alkyloxy
  • substituents R 10 and R 11 or the rest of the molecule can be attached to the carbon or nitrogen atom in which case one or both hydrogen atoms are replaced.
  • halo is generic to fluoro, chloro, bromo and iodo
  • C,_ 6 alkyl defines straight and branched chain saturated hydrocarbon radicals having from 1 to 6 carbon atoms such as, e.g.
  • C, .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; trihalomethyl defines methyl containing three identical or different halo substituents for example trifluoromethyl; C 2.6 alkenyl
  • addition salt comprises the salts which the compounds of formula (I) are able to form with organic or inorganic bases such as amines, alkali metal bases and earth alkaline metal bases, or quaternary ammonium bases, or with organic or inorganic acids, such as mineral acids, sulfonic acids, carboxylic acids or phosphorus containing acids.
  • organic or inorganic bases such as amines, alkali metal bases and earth alkaline metal bases, or quaternary ammonium bases
  • organic or inorganic acids such as mineral acids, sulfonic acids, carboxylic acids or phosphorus containing acids.
  • addition salt further comprises pharmaceutically acceptable salts, metal complexes and solvates and the salts thereof, that the compounds of formula (I) are able to form.
  • 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 (i.e. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids.
  • succinic i.e. butanedioic acid
  • maleic fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosal
  • 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.
  • metal complexes means a complex formed between a compound of formula (I) and one or more organic or inorganic metal salt or salts.
  • organic or inorganic salts comprise the halogenides, nitrates, sulfates, phosphates, acetates, trifluoroacetates, trichloroacetates, propionates, tartrates, sulfonates, e.g. methylsulfonates, 4-methylphenylsulfonates, salicylates, benzoates and the like of the metals of the second main group of the periodical system, e.g. the magnesium or calcium salts, of the third or fourth main group, e.g. aluminium, tin, lead as well as the first to the eighth transition groups of the periodical system such as, for example, chromium, manganese, iron, cobalt, nickel, copper, zinc and the like.
  • 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.
  • 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. As is common in pharmacology one enantiomer can have a better pharmacological activity than the other.
  • 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-, piperazine or pyridazinyl-nitrogens are N-oxidized.
  • each R 10 independently is hydrogen, C ⁇ -6 alkyl, hydroxy, d.6alkyloxyC 1-6 alkyl, morpholino, C ⁇ - 6 alkylimidazolyl, or pyridinylC ⁇ - alkyl amino ; j) each R 11 independently is hydrogen or hydroxy; and k) aryl is phenyl.
  • a second group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) n is 0; b) X is CH; c) Q is -NH-, -CH 2 -CH 2 - or -CHR 5 -, wherein R 5 is hydrogen, hydroxy, or arylC ⁇ -6 alkyl; d) R 1 is C ⁇ -6 alkyl; e) R 2 is hydrogen; f) R 3 is hydrogen, hydroxy or a group of formula (b-1); g) t is 0; h) Z is a heterocyclic ring system selected from (c-8) or (c-13); i) each R 10 independently is hydrogen; j) aryl is phenyl.
  • a third group of interesting compounds consists of those compounds of formula (I), the first group of interesting compounds or the second group of interesting compounds wherein Z is a heterocyclic ring system other than the heterocyclic ring system of formula (c-2) or (c-4
  • R 6 alkyl R 7 is hydrogen; R 8 is di(C 1-6 alkyl)aminoC 1-6 alkyl; t is 0 or 2; Z is a heterocyclic ring system selected from (c-1), (c-5), (c-6), (c-8), (c-10), (c-12) or (c-13); each R 10 independently is hydrogen, Ci- ⁇ alkyl, hydroxy, d ⁇ alkyloxyd-ealkyl, d ⁇ alkyloxyd-ealkylamino, morpholino, Ci- ⁇ alkylimidazolyl, or each R 11 independently is hydrogen or hydroxy; and aryl is phenyl.
  • a further group of preferred compounds consists of those compounds of formula (I) wherein n is 0; X is CH; Q is -NH-, -CH 2 -CH 2 - or -CHR 5 -, wherein R 5 is hydrogen, hydroxy, or arylCi- ⁇ alkyl; R 1 is C
  • An even further group of preferred compounds consists of those compounds of formula (I), the group of preferred compounds or the further group of preferred compounds wherein Z is a heterocyclic ring system other than the heterocyclic ring system of formula (c-2) or (c-4).
  • the most preferred compounds are compound No 7, compound No 2, compound No 1 and compound No 11.
  • the compounds of formula (I) can be prepared according to the general methods described in EP 371564. A number of such preparation methods will be described hereinafter in more detail. Other methods for obtaining final compounds of formula (I) are described in the examples.
  • Compounds of formula (I), wherein R is hydroxy, herein referred to as compounds of formula (I-c), can be prepared by converting the keton moiety of compounds of formula (I-a) into an hydroxy group, with an appropriate reductant, e.g., sodium borohydride in a suitable solvent, e.g. methanol and tetrahydrofuran.
  • an appropriate reductant e.g., sodium borohydride
  • a suitable solvent e.g. methanol and tetrahydrofuran.
  • Compounds of formula (I-a) can be prepared by converting compounds of formula (I-c), wherein R 2 is hydrogen, herein referred to as compounds of formula (I-c-1), in the presence of a suitable oxidant such as chromium trioxide and an acid such as sulfuric acid, in a suitable solvent such as 2-propanone.
  • a suitable oxidant such as chromium trioxide and an acid such as sulfuric acid
  • Compounds of formula (I), defined as compounds of formula (I) wherein R b is as defined in R 6 and R c is as defined in R 7 , or R b and R c taken together with the nitrogen to which they are attached, form an appropriate heterocyclic ring system as defined in Z, herein referred to as compounds of formula (I-h), can be prepared by reacting an intermediate of formula (IV) with an intermediate of formula (V).
  • the reaction can be performed in a reaction-inert solvent such as dimethylformamide or acetonitrile, and optionally in the presence of a suitable base such as, for example, sodium carbonate, potassium carbonate or thriethylamine.
  • the compounds of formula (I) may also be converted into each other via art-known reactions or functional group transformations.
  • a number of such transformations are already described hereinabove.
  • Other examples are hydrolysis of carboxylic esters to the conesponding carboxylic acid or alcohol; hydrolysis of amides to the conesponding carboxylic acids or amines; hydrolysis of nitriles to the conesponding 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 esters and ethers; primary amines may be converted into secondary or tertiary amines; double bonds may be hydrogenated to the conesponding 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.
  • compounds of formula (I), (I-a), (I-b), (I-c), (I-c-1), (I-h), (I-i), (I-j) and (I-k) can optionally be the subject of one or more of the following conversions in any desired order:
  • Intermediates of formula (VII), wherein R d and R e are appropriate radicals or taken together with the carbon to which they are attached, form an appropriate heterocyclic ring system as defined in Z, can be prepared by hydrolysing intermediates of formula (VI), wherein R 3 is a group of formula (b-1) or a radical of formula (a-1) wherein s is other than 0, herein referred to as R g , according to art-known methods, such as stirring the intermediate (VI) in an aqueous acid solution in the presence of a reaction inert solvent, e.g. tetrahydrofuran.
  • An appropriate acid is for instance hydrochloric acid.
  • Compounds of formula (I) wherein R 2 is hydrogen and R g is as defined above, herein refened to as compounds of formula (I-k), can be prepared starting from intermediates of formula (VII), by a selective hydrogenation of said intermediate with an appropriate reducing agent such as, for example with a noble catalyst, such as platinum-on- charcoal, palladium-on-charcoal and the like and an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
  • an appropriate reducing agent such as, for example with a noble catalyst, such as platinum-on- charcoal, palladium-on-charcoal and the like and an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
  • Compounds of formula (I) can be prepared by hydrolysing intermediates of fonnula (VIII), according to art-known methods, by submitting the intermediates of fonnula (VIII) to appropriate reagents, such as, tinchloride, acetic acid and hydrochloric acid, in the presence of a reaction inert solvent, e.g. tetrahydrofuran.
  • appropriate reagents such as, tinchloride, acetic acid and hydrochloric acid
  • the compounds of formula (I) wherein X is CH herein refened to as compounds of fonnula (I-j), may also be obtained by cyclizing an intermediate of formula (X).
  • the cyclization reaction of intermediates of formula (X) may be conducted according to art- known cyclizing procedures.
  • the reaction is carried out in the presence of a suitable Lewis Acid, e.g. aluminum chloride either neat or in a suitable solvent such as, for example, an aromatic hydrocarbon, e.g. benzene, chlorobenzene, methylbenzene and the like; halogenated hydrocarbons, e.g. trichloromethane, tetrachloromethane and the like; an ether, e.g. tetrahydrofuran, 1,4-dioxane and the like; or mixtures of such solvents.
  • a suitable Lewis Acid e.g. aluminum chloride either neat or in a suitable solvent
  • an aromatic hydrocarbon e.g. benzen
  • the compounds of formula (I), wherein X is N, herein refened to as compounds of fonnula (I-i) may be obtained by condensing an appropriate ortho-benzenediamine of formula (XI) with an ester of formula (XII) wherein R h is d_ 6 alkyl.
  • the condensation of the substituted ortho-diamine of formula (XI) and the ester of formula (XII) can be carried out in the presence of a carboxylic acid, e.g.
  • acetic acid and the like a mineral acid such as, for example hydrochloric acid, sulfuric acid, or a sulfonic acid such as, for example, methanesulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid and the like.
  • a mineral acid such as, for example hydrochloric acid, sulfuric acid, or a sulfonic acid such as, for example, methanesulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid and the like.
  • Intermediates of formula (XI) can be prepared by a nitro to amine reduction reaction starting with an intermediate of fonnula (XIII) in the presence of a metal catalyst such as Raney Nickel and an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
  • a metal catalyst such as Raney Nickel
  • an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
  • Intermediates of fonnula (XIII) can be prepared by hydrolysing intermediates of fonnula (XIV), according to art-known methods, such as stirring the intermediate (XIV) in an aqueous acid solution in the presence of a reaction inert solvent, e.g. tetrahydrofuran,.
  • a reaction inert solvent e.g. tetrahydrofuran
  • An appropriate acid is for instance hydrochloric acid.
  • 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 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 is a compound of formula (I)
  • Y is -N ⁇ or -CH ⁇ ;
  • Q is -NH-, -O-, -C(O)-, -CH 2 -CH 2 - or -CHR 5 -, wherein R 5 is hydrogen, hydroxy, C ⁇ -6 alkyl, arylCi- ⁇ alkyl, C 1-6 alkyloxycarbonyl, C 1-6 alkyloxyd. 6 alkylamino or haloindazolyl;
  • R 1 is d- ⁇ alkyl or thienyl
  • R 3 is hydrogen, d. 6 alkyl or a radical selected from NR 6 R 7 (a-1), O-H (a-2), O-R 8 (a-3), S- R 9 (a-4), or — C ⁇ N (a-5), wherein R 6 is -CHO, d- ⁇ alkyl, hydroxyC 1-6 alkyl, C ⁇ _ 6 alkylcarbonyl, di(C ⁇ . 6 alkyl)aminoC 1-6 alkyl, C ⁇ - 6 alkylcarbonylaminoC 1-6 alkyl, piperidinylC].
  • R 9 is dKCi- ⁇ alky aminod- ⁇ alkyl; or R 3 is a group of formula -(CH 2 ) t -Z- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
  • each R 10 independently is hydrogen, C 1-6 alkyl, aminocarbonyl, hydroxy,
  • aryl is phenyl or phenyl substituted with halo, C 1-6 alkyl or d. 6 alkyloxy.
  • 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.
  • 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.
  • inhibition of PARP activity spares the cells from energy loss, preventing, in the case of neural cells, ineversible 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 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.
  • 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 neurodegenerati on .
  • 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 tumors, 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 Iymphoma, endometrial cancer, esophageal cancer, Ewing's sarcoma gallbladder cancer, hairy cell leukemia, head &neck cancer, Hodgkin's Iymphoma, 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'
  • 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 in conjunction with radiation of x-rays.
  • 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.
  • metronidazole misonidazole
  • desmethylmisonidazole pimonidazole
  • etanidazole nimorazole
  • mitomycin C RSU 1069
  • SR 4233 EO9
  • 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 cell transfusions, 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 therapueutically effective compounds for treating cancer or other disease.
  • additional therapeutica agents that may be used in conjunction with chemosensitizers include, but are not limited to : methylating agents, toposisomerase I inhibitors and other chemothearpeutic 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-1 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 0.01 mg/kg to 100 mg/kg body weight, and in particular from 0.05 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 0.5 to 500 mg, and in particular 1 mg to 200 mg of active ingredient per unit dosage form. The following examples illustrate the present invention.
  • BuLi is defines as butyl -lithium
  • DCM is defined as dichloromethane
  • DDPE is defined as diisopropyl ether
  • DMF is defined as N,N-dimethylformamide
  • 'DMSO' is defined as dimethylsulfoxide
  • EtOAc is defined as ethyl acetate
  • EtOH is defined as ethanol
  • MEK is defined as methyl ethyl keton
  • MeOH is defined as methanol
  • THF is defined as tetrahydrofuran.
  • Aluminium chloride (0.6928 mol) was added portionwise to a solution of chloro- acetyl chloride (0.5196 mol) in DCM (50.2ml) while the temperature was kept below 30°C. 3- ethyl- 2(lH)-quinolinone (0.1732 mol) was added while the temperature was kept below 30°C. The mixture was stined and refluxed for 15 hours, cooled and poured out into ice water. The precipitate was filtered off, washed with water and taken up in DCM. The organic solution was stirred and filtered. The precipitate was dried, yielding 33.5g of intermediate 1. The filtrate was extracted. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated till dryness, yielding 20.46g of intermediate 2. bjPrepar ;ation . of .in te ⁇ nedi . ate 3
  • Example A2 a Preparation of intejrmediate.4 nBuLi 1.6M in hexane (0.0764 mol) was added dropwise at -60°C under N 2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.0694 mol) in THF (185ml). The mixture was stirred at -60°C for 1 hour and then added dropwise at -60°C to a mixture of N-methoxy-N-methyl- cycloheptanecarboxamide (0.0694 mol) in diethyl ether (100ml).
  • Nitric acid fuming (5.6ml) was added dropwise at a temperature below 30°C to a mixture of intermediate 10 (0.037 mol) in acetyl anhydride (100ml). The mixture was stined at a temperature below 30°C for 1 hour, poured out into ice water, basified with a concentrated NH t OH solution and extracted with DCM. The organic layer was separated, dried (MgSO 4 ), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-35 ⁇ m) (eluent: DCM/MeOH 99/1). The desired fractions were collected and the solvent was evaporated, yielding 4g of intermediate 11. e) . Preparation of intermediate 12
  • nBuLi 1.6M in hexane (0.0516 mol) was adde dropwise at -60°C under N 2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.043 mol) in THF (115ml). The mixture was stined at -60°C for 1 hour. A mixture of l-cyclohexyl-3-(4-methyl-l- piperazinyl)- 1-propanone, (0.043 mol) in THF (103ml) was added dropwise at -60°C.
  • Example A7 a) Preparation of intermediate 28 nBuLi 1.6M in hexane (0.224 mol) was added at -78°C under N 2 flow to a solution of 6-bromo-3-ethyl-2-methoxy- quinoline (0.188 mol) in THF (500ml). The mixture was stined at -78°C for 1 hour. A mixture of 3-cyclohexene-l-carboxaldehyde (0.182 mol) in THF (500ml) was added dropwise at -78°C. The mixture was stined at -78°C for 2 hours, then brought to 0°C, hydrolized and extracted with EtOAc.
  • nBuLi 1.6M in hexane (21.32ml) was added dropwise at -70°C under N 2 flow to a mixture of 1 -methyl- lH-imidazole (0.0341 mol) in T ⁇ F (28ml). The mixture was stined at -70°C for 30 min. Chlorotriethyl- silane (0.0341 mol) was added. The mixture was brought to room temperature. nBuLi 1.6 M in hexane (21.32ml) was added dropwise at -70°C. The mixture was stined at -70°C for 1 hour and then brought to -15°C.
  • Table F-l lists the compounds that were prepared according to one of the above Examples. Table F-l
  • SPA In vitro Scintillation Proximity Assay
  • the assay relies upon the well established SPA technology for the detection of poly(ADP-ribosyl)ation of biotinylated target proteins, i.e histones.
  • This ribosylation is induced using nicked DNA activated PARP-1 enzyme and [ 3 H]- nicotinamide 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 °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 °C).
  • the PARP-1 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-1 enzyme.
  • the reaction was terminated by adding 100 ⁇ l 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 transfened to a TopCountNXTTM (Packard) for scintillation counting, values were expressed as counts per minute (cpm).
  • IC 5 o-values concentration of the drug, needed to reduce the PARP-1 enzyme activity to 50% of the control
  • pICso the negative log value of the IC 50 - 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 "6 M (see Tabel-2).
  • the radioactive ribosylated histones were precipitated by trichloroacetic acid (TCA) in 96- well filterplates and the incorporated [ 32 P] measured using a scintillation counter
  • TCA trichloroacetic acid
  • 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-1 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-1 inhibitory activity. Seventy-five ⁇ l of the PARP- 1 enzyme/DNA mixture together with 1 ⁇ l of compound in DMSO and 25 ⁇ l of histones-NAD /[ P]- NAD + mixture was added per well of a 96-well filterplate (0.45 ⁇ m, supplier Millipore). The final concentrations in the incubation mixture were 2 ⁇ g/ml for the histones, 0.1 mM for the NAD + , 200 ⁇ M (0.5 ⁇ C) for the [ 32 P]-NAD + and 2 ⁇ g/ml for the nicked DNA. Plates were incubated for 15 min.
  • the compounds can be further evaluated in a cellular chemo- and/or radiosensitization assay, an assay measuring inhibition of endogenous PARP-l 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 n, s, R1, R2, R3, Q, X and Y have defined meanings.

Description

SUBSTITUTED 6-CYCLOHEXYLALKYL SUBSTITUTED 2-QUINOLINONES AND 2-QUINOXALINONES AS POLY(ADP-RIBOSE) POLYMERASE 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-1) is a member of the PARP enzyme family consisting of PARP-1 and several recently identified novel poly(ADP-ribosylating) enzymes. PARP is also referred to as poly(adenosine 5'- diphospho-ribose) polymerase or PARS (poly(ADP-ribose) synthetase).
PARP-1 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, topoisomerases, 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.
Among the many functions attributed to PARP, and especially PARP-1, 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 NAD+ 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 NMD A- 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 in infarct size in the heart (32-42%) while 1,5- dihydroxyisoquinoline (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 trinitrobenzene 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 moφhology 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, AIDS, 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 radiosensitizing (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.
A recent comprehensive review of the state of the art has been published by Li and Zhang in IDrugs 2001, 4(7): 804-812.
There continues to be a need for effective and potent PARP inhibitors, and more particularly PARP-1 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
EP 371564, published on June 6, 1990, discloses (lH-azol-1-ylmethyl) substituted quinoline, quinazoline or quinoxaline derivatives. The described compounds suppress the plasma elimination of retinoic acids. More in particular 6-(cyclohexyl- lH-imidazol-l-ylmethyl)-3-methyl-2(lH)-quinoxalinone (Compound A) is disclosed.
Figure imgf000007_0001
Compound A
Description of the invention
This invention concerns compounds of formula (I)
Figure imgf000007_0002
the N-oxide forms, the addition salts and the stereo-chemically isomeric forms thereof, wherein
n is O or 1; s is O or 1;
X is -Ν= or -CR4=, wherein R4 is hydrogen or taken together with R1 may form a bivalent radical of formula -CH=CH-CH=CH-;
Y is -N< or -CH<;
Q is -NH-, -0-, -C(O)-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, Cι-6alkyl, arylCι.6alkyl, Ci_6alkyloxycarbonyl, Ci-βalkyloxyCi-όalkylamino or haloindazolyl;
R1 is Cι. alkyl or thienyl; R ,2 i s . hyd .rogen or * ta„ιken t.oge *tιher wi :.th r R»3 may form =O;
R is hydrogen, C1-6alkyl or a radical selected from • NR6R7 (a-1), O-H (a-2), O-R8 (a-3), S- R9 (a-4), or — C≡N (a-5), wherein R6 is -CHO, Cι-6alkyl, hydroxyC1-6alkyl, C1-6alkylcarbonyl, di(d-6alkyl)aιmnod.6alkyl, Cι-6alkylcarbonylaminoC1-6alkyl, piperidinylC1-6alkyl, piperidinyld-όalkylaminocarbonyl, C1-6alkyloxy,
Figure imgf000008_0001
thienylCι.6alkyl, pyrrolyld-δalkyl, arylC1-6alkylpiperidinyl, arylcarbonyld^alkyl, arylcarbonylpiperidinylCi-όalkyl, haloindozolylpiperidinylC1-6alkyl, or arylCι-6alkyl(C1-6alkyl)aminoC1-6alkyl; and R7 is hydrogen or Cι-6alkyl; R8 is C όalkyl, d-όalkylcarbonyl or di(Cι.6alkyl)aminoC1-6alkyl; and R9 is di(Cι.6alkyl)aminoC1-6alkyl; or R3 is a group of formula -(CH2)t-Z- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
Figure imgf000008_0002
(c-1) (c-2) (c-3) (c-4)
Figure imgf000008_0003
(c-5) (c-6)
Figure imgf000008_0004
Figure imgf000009_0001
(c-9) (c-10) (c-11) (c-12) (c-13) wherein each R10 independently is hydrogen, d^alkyl, aminocarbonyl, hydroxy,
Figure imgf000009_0002
C ealkylo yCj.όalkyl, Ci-ealkyloxyCi-ealkylamino, di(phenylC2-6alkenyl), piperidinylCi^alkyl, C3-10cycloalkyl, C^ocycloalkyld-ealkyl, "' " ' aryloxy(hydroxy)C1-6alkyl, haloindazolyl, arylCi-βalkyl, arylC2.6alkenyl, mo holino, d^alkylimidazolyl, or pyridinyld-ealkylamino; each R11 independently is hydrogen, hydroxy, piperidinyl or aryl;
aryl is phenyl or phenyl substituted with halo, d^alkyl or Cι-6alkyloxy
with the proviso that 6-(cyclohexyl-lH-imidazol-l-ylmethyl)-3-methyl-2(lH)- quinoxalinone is not included.
Whenever the heterocyclic ring system Z contains a -CΗ2-, -CH=, or -NH- moiety the substituents R10 and R11 or the rest of the molecule can be attached to the carbon or nitrogen atom in which case one or both hydrogen atoms are replaced.
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; C,_6alkyl 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; C,.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; trihalomethyl defines methyl containing three identical or different halo substituents for example trifluoromethyl; C2.6alkenyl defines straight and branched chain hydrocarbon radicals containing one double bond and having from 2 to 6 carbon atoms such as, for example, ethenyl, 2-propenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 3-methyl-2-butenyl, and the like; and C3-1ocycloalkyl includes cyclic hydrocarbon groups having from 3 to 10 carbons, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl and the like.
The term "addition salt" comprises the salts which the compounds of formula (I) are able to form with organic or inorganic bases such as amines, alkali metal bases and earth alkaline metal bases, or quaternary ammonium bases, or with organic or inorganic acids, such as mineral acids, sulfonic acids, carboxylic acids or phosphorus containing acids.
The term "addition salt" further comprises pharmaceutically acceptable salts, metal complexes and solvates and the salts thereof, that the compounds of formula (I) are able to form.
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 (i.e. butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-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 "metal complexes" means a complex formed between a compound of formula (I) and one or more organic or inorganic metal salt or salts. Examples of said organic or inorganic salts comprise the halogenides, nitrates, sulfates, phosphates, acetates, trifluoroacetates, trichloroacetates, propionates, tartrates, sulfonates, e.g. methylsulfonates, 4-methylphenylsulfonates, salicylates, benzoates and the like of the metals of the second main group of the periodical system, e.g. the magnesium or calcium salts, of the third or fourth main group, e.g. aluminium, tin, lead as well as the first to the eighth transition groups of the periodical system such as, for example, chromium, manganese, iron, cobalt, nickel, copper, zinc 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. As is common in pharmacology one enantiomer can have a better pharmacological activity than the other. 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-, piperazine or pyridazinyl-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.
The compounds described in EP 371564 suppress the plasma elimination of retinoic acids. 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) X is -N= or -CH=; b) R1 is C1-6alkyl; c) R3 is hydrogen, d_6alkyl, a radical selected from (a-1), (a-2), (a-3) or (a-4) or a group of formula (b-1); d) R6 is di(C1-6alkyl)aminod-6alkyl or d-όalkyloxyd-όalkyl; e) R7 is hydrogen; f) R8 is di(Cι.6alkyl)aminoC1-6alkyl; g) t is 0 or 2; h) Z is a heterocyclic ring system selected from (c-1), (c-5), (c-6), (c-8), (c-10), (c-12) σr (c-13); i) each R10 independently is hydrogen, Cι-6alkyl, hydroxy, d.6alkyloxyC1-6alkyl,
Figure imgf000012_0001
morpholino, Cι-6alkylimidazolyl, or pyridinylC ι - alkyl amino ; j) each R11 independently is hydrogen or hydroxy; and k) aryl is phenyl.
A second group of interesting compounds consists of those compounds of formula (I) wherein one or more of the following restrictions apply: a) n is 0; b) X is CH; c) Q is -NH-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, or arylCι-6alkyl; d) R1 is Cι-6alkyl; e) R2 is hydrogen; f) R3 is hydrogen, hydroxy or a group of formula (b-1); g) t is 0; h) Z is a heterocyclic ring system selected from (c-8) or (c-13); i) each R10 independently is hydrogen; j) aryl is phenyl. A third group of interesting compounds consists of those compounds of formula (I), the first group of interesting compounds or the second group of interesting compounds wherein Z is a heterocyclic ring system other than the heterocyclic ring system of formula (c-2) or (c-4).
A group of preferred compounds consists of those compounds of formula (I) wherein X is -N= or -CH=; R1 is Cι.6alkyl; R3 is hydrogen, Cι.6alkyl, a radical selected from (a-1), (a-2), (a-3) or (a-4) or a group of formula (b-1); R6 is di(C1-6alkyl)aminoCι.6alkyl or Cι-6alkyloxyCι.6alkyl; R7 is hydrogen; R8 is di(C1-6alkyl)aminoC1-6alkyl; t is 0 or 2; Z is a heterocyclic ring system selected from (c-1), (c-5), (c-6), (c-8), (c-10), (c-12) or (c-13); each R10 independently is hydrogen, Ci-βalkyl, hydroxy, d^alkyloxyd-ealkyl, d^alkyloxyd-ealkylamino, morpholino, Ci-όalkylimidazolyl, or
Figure imgf000013_0001
each R11 independently is hydrogen or hydroxy; and aryl is phenyl.
A further group of preferred compounds consists of those compounds of formula (I) wherein n is 0; X is CH; Q is -NH-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, or arylCi-βalkyl; R1 is C|.6alkyl; R2 is hydrogen; R3 is hydrogen, hydroxy or a group of formula (b-1); t is 0; Z is a heterocyclic ring system selected from (c-8) or (c-13); each R10 independently is hydrogen; and aryl is phenyl.
An even further group of preferred compounds consists of those compounds of formula (I), the group of preferred compounds or the further group of preferred compounds wherein Z is a heterocyclic ring system other than the heterocyclic ring system of formula (c-2) or (c-4).
The most preferred compounds are compound No 7, compound No 2, compound No 1 and compound No 11.
Figure imgf000013_0002
Figure imgf000014_0001
The compounds of formula (I) can be prepared according to the general methods described in EP 371564. A number of such preparation methods will be described hereinafter in more detail. Other methods for obtaining final compounds of formula (I) are described in the examples.
Compounds of formula (I) wherein R2 is hydrogen and R3 is -NR7-CHO wherein R7 is hydrogen or methyl, herein refened to as compounds of formula (I-b), can be prepared starting from compounds of formula (I), wherein R2 taken together with R3 forms =O, herein refened to as compounds of fonnula (I-a), in the presence of formamide or methylformamide, here indicated as intermediates of formula (fl), and formic acid.
Figure imgf000014_0002
Compounds of formula (I), wherein R is hydroxy, herein referred to as compounds of formula (I-c), can be prepared by converting the keton moiety of compounds of formula (I-a) into an hydroxy group, with an appropriate reductant, e.g., sodium borohydride in a suitable solvent, e.g. methanol and tetrahydrofuran.
Figure imgf000014_0003
Compounds of formula (I-a) can be prepared by converting compounds of formula (I-c), wherein R2 is hydrogen, herein referred to as compounds of formula (I-c-1), in the presence of a suitable oxidant such as chromium trioxide and an acid such as sulfuric acid, in a suitable solvent such as 2-propanone.
Figure imgf000015_0001
Intermediates of formula (IV), wherein W is an appropriate leaving group such as, for example, chloro, bromo, methanesulfonyloxy or benzenesulfonyloxy can be prepared from compounds of formula (I-c-1) by treating said compounds with a suitable reagent e.g. methanesulfonyloxy chloride or benzenesulfonyloxy chloride, or a halogenating reagent such as e.g. POCl3 or SOCl2.
Figure imgf000015_0002
Compounds of formula (I), defined as compounds of formula (I) wherein Rb is as defined in R6 and Rc is as defined in R7, or Rb and Rc taken together with the nitrogen to which they are attached, form an appropriate heterocyclic ring system as defined in Z, herein referred to as compounds of formula (I-h), can be prepared by reacting an intermediate of formula (IV) with an intermediate of formula (V). The reaction can be performed in a reaction-inert solvent such as dimethylformamide or acetonitrile, and optionally in the presence of a suitable base such as, for example, sodium carbonate, potassium carbonate or thriethylamine.
Figure imgf000015_0003
The compounds of formula (I) may also be converted into each other via art-known reactions or functional group transformations. A number of such transformations are already described hereinabove. Other examples are hydrolysis of carboxylic esters to the conesponding carboxylic acid or alcohol; hydrolysis of amides to the conesponding carboxylic acids or amines; hydrolysis of nitriles to the conesponding 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 esters and ethers; primary amines may be converted into secondary or tertiary amines; double bonds may be hydrogenated to the conesponding 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.
Hence, compounds of formula (I), (I-a), (I-b), (I-c), (I-c-1), (I-h), (I-i), (I-j) and (I-k) can optionally be the subject of one or more of the following conversions in any desired order:
(i) converting a compound of formula (I) into a different compound of formula (I);
(ii) converting a compound of formula (I) into the conesponding acceptable salt or
N-oxide thereof; (iii) converting a pharmaceutically acceptable salt or N-oxide of a compound of formula (I) into the parent compound of formula (I);
(iv) preparing a stereochemical isomeric form of a compound of fonnula (I) or a pharmaceutically acceptable salt or N-oxide thereof.
Intermediates of formula (VII), wherein Rd and Re are appropriate radicals or taken together with the carbon to which they are attached, form an appropriate heterocyclic ring system as defined in Z, can be prepared by hydrolysing intermediates of formula (VI), wherein R3 is a group of formula (b-1) or a radical of formula (a-1) wherein s is other than 0, herein referred to as Rg , according to art-known methods, such as stirring the intermediate (VI) in an aqueous acid solution in the presence of a reaction inert solvent, e.g. tetrahydrofuran. An appropriate acid is for instance hydrochloric acid.
Figure imgf000017_0001
Compounds of formula (I) wherein R2 is hydrogen and Rg is as defined above, herein refened to as compounds of formula (I-k), can be prepared starting from intermediates of formula (VII), by a selective hydrogenation of said intermediate with an appropriate reducing agent such as, for example with a noble catalyst, such as platinum-on- charcoal, palladium-on-charcoal and the like and an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
Figure imgf000017_0002
(VII) G*)
Compounds of formula (I) can be prepared by hydrolysing intermediates of fonnula (VIII), according to art-known methods, by submitting the intermediates of fonnula (VIII) to appropriate reagents, such as, tinchloride, acetic acid and hydrochloric acid, in the presence of a reaction inert solvent, e.g. tetrahydrofuran.
Figure imgf000017_0003
(VII I) (!)
Compounds of formula (I) can be prepared starting from N-oxides of formula (IX) by converting the intermediates of formula (IX) in the presence of a suitable reagent such as sodium carbonate or acetic anhydride and when appropriate in a solvent such as dichloromethane.
Figure imgf000018_0001
(K) (D
The compounds of formula (I) wherein X is CH herein refened to as compounds of fonnula (I-j), may also be obtained by cyclizing an intermediate of formula (X).The cyclization reaction of intermediates of formula (X) may be conducted according to art- known cyclizing procedures. Preferably the reaction is carried out in the presence of a suitable Lewis Acid, e.g. aluminum chloride either neat or in a suitable solvent such as, for example, an aromatic hydrocarbon, e.g. benzene, chlorobenzene, methylbenzene and the like; halogenated hydrocarbons, e.g. trichloromethane, tetrachloromethane and the like; an ether, e.g. tetrahydrofuran, 1,4-dioxane and the like; or mixtures of such solvents. Somewhat elevated temperatures, preferably between 70°-100°C, and stirring may enhance the rate of the reaction.
Figure imgf000018_0002
The compounds of formula (I), wherein X is N, herein refened to as compounds of fonnula (I-i) may be obtained by condensing an appropriate ortho-benzenediamine of formula (XI) with an ester of formula (XII) wherein Rh is d_6alkyl. The condensation of the substituted ortho-diamine of formula (XI) and the ester of formula (XII) can be carried out in the presence of a carboxylic acid, e.g. acetic acid and the like, a mineral acid such as, for example hydrochloric acid, sulfuric acid, or a sulfonic acid such as, for example, methanesulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid and the like. Somewhat elevated temperatures may be appropriate to enhance the rate of the reaction and in some cases the reaction may even be carried out at the reflux temperature of the reaction mixture. The water which is liberated during the condensation may be removed from the mixture by azeotropical distillation, distillation and the like methods.
Figure imgf000019_0001
Intermediates of formula (XI) can be prepared by a nitro to amine reduction reaction starting with an intermediate of fonnula (XIII) in the presence of a metal catalyst such as Raney Nickel and an appropriate reductant such as hydrogen in a suitable solvent such as methanol.
Figure imgf000019_0002
(xm) (XI) Intermediates of fonnula (XIII) can be prepared by hydrolysing intermediates of fonnula (XIV), according to art-known methods, such as stirring the intermediate (XIV) in an aqueous acid solution in the presence of a reaction inert solvent, e.g. tetrahydrofuran,. An appropriate acid is for instance hydrochloric acid.
Figure imgf000019_0003
(xm) (χιv>
Intermediates of formula (X) can conveniently be prepared by reacting an aniline of formula (XV) with a halide of formula (XVI) in the presence of a base such as pyridine in a suitable solvent such as dichloromethane.
Figure imgf000019_0004
(XV) (X) Intermediates of formula (VITJ) wherein n is 0, R2 is hydrogen or hydroxy and when R is hydrogen then R3 is hydroxy herein refened to as intermediates of formula (VHI-a) can be prepared by treating an intermediate of formula (XVII), wherein W is halo, with an organolithium reagent such as, e.g. n-butyllithium in a reaction inert solvent, e.g. tetrahydrofuran, and subsequently reacting said intermediate with an intermediate of formula (XVIII) wherein R' is hydrogen or a radical as defined in R3.
Figure imgf000020_0001
(XVII) (XVII I) (VII I-a)
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 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 is a compound of formula (I)
Figure imgf000020_0002
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereochemically isomeric forms thereof, wherein
n is O or 1; s is O or l;
X is -Ν= or -CR4=, wherein R4 is hydrogen or taken together with R1 may form a bivalent radical of formula -CH=CH-CH=CH-;
Y is -N< or -CH<; Q is -NH-, -O-, -C(O)-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, Cι-6alkyl, arylCi-βalkyl, C1-6alkyloxycarbonyl, C1-6alkyloxyd.6alkylamino or haloindazolyl;
R1 is d-βalkyl or thienyl;
R is hydrogen or taken together with R may form =O;
R3 is hydrogen, d.6alkyl or a radical selected from NR6R7 (a-1), O-H (a-2), O-R8 (a-3), S- R9 (a-4), or — C≡N (a-5), wherein R6 is -CHO, d-βalkyl, hydroxyC1-6alkyl, Cι_6alkylcarbonyl, di(Cι.6alkyl)aminoC1-6alkyl, Cι-6alkylcarbonylaminoC1-6alkyl, piperidinylC].6alkyl, piperidinylC1-6alkylaminocarbonyl, d-6alkyloxy, Ci-βalkylo yCi-όalkyl,
Figure imgf000021_0001
pynolyld-βalkyl, arylC ealkylpiperidinyl, arylcarbonylCι_6alkyl, arylcarbonylpiperidinylCι.6alkyl, haloindozolylpiperidinylCi-όalkyl, or aryld-ealky d-ό lkytyaminod-όalkyl; and R7 is hydrogen or Cι-6alkyl; R8is d.6alkyl, d-όalkylcarbonyl or di(Cι.6alkyl)aminoCι-6alkyl; and R9is dKCi-όalky aminod-όalkyl; or R3 is a group of formula -(CH2)t-Z- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
Figure imgf000021_0002
(c-1) (c-2) (c-3) (c-4)
Figure imgf000022_0001
(c-9) (c-10) (c-11) (c-12) (c-13) wherein each R10 independently is hydrogen, C1-6alkyl, aminocarbonyl, hydroxy,
Figure imgf000022_0002
C l .6alkyloxyC i .6alkyl , C i - alkyloxyC i .ήalkylamino, di(phenylC2-6alkenyl), piperidinylCi-6alkyl, C3-ιocycloalkyl, C3-ιocycloalkylCι.6alkyl, aryloxy(hydroxy)Cι.6alkyl, haloindazolyl,
Figure imgf000022_0003
arylC2-6alkenyl, morpholino, d-βalkylimidazolyl, or pyridinylC].6alkylamino; each R11 independently is hydrogen, hydroxy, piperidinyl or aryl;
aryl is phenyl or phenyl substituted with halo, C1-6alkyl or d.6alkyloxy.
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 fonn 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, ineversible 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 neurodegenerati on .
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 tumors, 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 Iymphoma, endometrial cancer, esophageal cancer, Ewing's sarcoma gallbladder cancer, hairy cell leukemia, head &neck cancer, Hodgkin's Iymphoma, 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 Iymphoma, 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 cunently 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 cell transfusions, 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 therapueutically effective compounds for treating cancer or other disease. Examples of additional therapeutica agents that may be used in conjunction with chemosensitizers include, but are not limited to : methylating agents, toposisomerase I inhibitors and other chemothearpeutic 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-1 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 0.01 mg/kg to 100 mg/kg body weight, and in particular from 0.05 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 0.5 to 500 mg, and in particular 1 mg to 200 mg of active ingredient per unit dosage form. The following examples illustrate the present invention.
Experimental part
Hereinafter, "BuLi" is defines as butyl -lithium, "DCM" is defined as dichloromethane, "DDPE" is defined as diisopropyl ether, "DMF" is defined as N,N-dimethylformamide, 'DMSO' is defined as dimethylsulfoxide, "EtOAc" is defined as ethyl acetate, "EtOH" is defined as ethanol, "MEK" is defined as methyl ethyl keton, "MeOH" is defined as methanol and "THF" is defined as tetrahydrofuran.
A. Preparation of the intermediate compounds
Example Al a) Preparation of intermediate 1 and 2
Figure imgf000028_0001
intermediate 1 intermediate 2
Aluminium chloride (0.6928 mol) was added portionwise to a solution of chloro- acetyl chloride (0.5196 mol) in DCM (50.2ml) while the temperature was kept below 30°C. 3- ethyl- 2(lH)-quinolinone (0.1732 mol) was added while the temperature was kept below 30°C. The mixture was stined and refluxed for 15 hours, cooled and poured out into ice water. The precipitate was filtered off, washed with water and taken up in DCM. The organic solution was stirred and filtered. The precipitate was dried, yielding 33.5g of intermediate 1. The filtrate was extracted. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated till dryness, yielding 20.46g of intermediate 2. bjPrepar ;ation . of .in teπnedi .ate 3
Figure imgf000028_0002
Piperidine (0.24 mol) was added dropwise at room temperature to a solution of intermediate 1 and intermediate 2 in DMF (300ml). The mixture was stined for 5 min, poured out into water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated till dryness. The residue (39.14g) was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOΗ/ΝΗjOΗ 96/4/0.2). The pure fractions were collected and the solvent was evaporated. Part (3.7g) of the residue (13.8g) was crystallized from 2-propanone. The precipitate was filtered off, washed with diethyl ether and dried, yielding 3g of intermediate 3, melting point 190°C.
Example A2 a). Preparation of intejrmediate.4
Figure imgf000029_0001
nBuLi 1.6M in hexane (0.0764 mol) was added dropwise at -60°C under N2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.0694 mol) in THF (185ml). The mixture was stirred at -60°C for 1 hour and then added dropwise at -60°C to a mixture of N-methoxy-N-methyl- cycloheptanecarboxamide (0.0694 mol) in diethyl ether (100ml). The mixture was stined at -60°C for 1 hour, then brought to 0°C, poured out into a saturated NFLjCl solution and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The product was used without further purification, yielding 21.61g (quant.) of intermediate 4. bXPrep.aration.pf.m
Figure imgf000029_0002
A mixture of intermediate 4 (0.0694 mol) in hydrochloric acid 3Ν (317ml) and THF (159ml) was stined and refluxed overnight. The mixture was poured out on ice, basified with a concentrated NHtOH solution and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The product was used without further purification, yielding 17.59g (85%) of intermediate 5.
Figure imgf000029_0003
Sodium hydroborate (0.0296 mol) was added portionwise at 0°C under N2 flow to a mixture of intermediate 5 (0.0591 mol) in MeOH (176ml). The mixture was poured out on ice and extracted with DCM. The precipitate was filtered off and dried. The product was used without further purification, yielding 6.38g (36%) of intermediate 6. d) Prep.aration of .intenned. te.7
Figure imgf000029_0004
Intermediate 6 (0.0213 mol) was added portionwise at 0°C to thionyl chloride (32ml). The mixture was stined at room temperature overnight. The solvent was evaporated till dryness. The product was used without further purification, yielding 6.77g (quant.) of intermediate 7. Example A3 a).Prep.aratipn of intenn.ediate 8
Figure imgf000030_0001
A mixture of N-methoxy-N-methyl-4-nitro- benzeneacetamide (0.534 mol) in MeOH (1200ml) was hydrogenated at room temperature under a 3 bar pressure for 1 hour with Raney nickel (60g) as a catalyst. After uptake of H2 (3 equiv), the catalyst was filtered off and the filtrate was evaporated, yiel intermediate 8. b) Preparation of intermediate 9
Figure imgf000030_0002
Acetyl anhydride (1.36 mol) was added dropwise at room temperature to a mixture of intermediate 8 (0.525 mol) in DCM (100ml). The mixture was stined at room temperature overnight. Water was added and the mixture was extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (151.6g) was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/ΝHtOH 95/5/0.1). The pure fractions were collected and the solvent was evaporated, yielding 32g (26%) of intermediate 9. .c).Pre^aration.of intenn.e^i.ate 0 l«s*r 'Νγ'
Methyl- lithium 1.6M (74ml) was added at 0°C under N2 flow to a mixture of intermediate 9 (0.059 mol) in THF (210ml). The mixture was stirred at 0°C for 90 min, poured out into water and extracted with EtOAc. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/NHiOH 96/4/0.1). The desired fractions were collected and the solvent was evaporated, yielding 7g (33%) of intermediate 10.
Figure imgf000030_0003
Nitric acid fuming (5.6ml) was added dropwise at a temperature below 30°C to a mixture of intermediate 10 (0.037 mol) in acetyl anhydride (100ml). The mixture was stined at a temperature below 30°C for 1 hour, poured out into ice water, basified with a concentrated NHtOH solution and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-35 μm) (eluent: DCM/MeOH 99/1). The desired fractions were collected and the solvent was evaporated, yielding 4g of intermediate 11. e). Preparation of intermediate 12
Figure imgf000031_0001
Sodium hydroborate (0.0187 mol) was added portionwise at 5°C to a mixture of intermediate 11 (0.017 mol) in MeOH (50ml). The mixture was stined at 5°C, hydrolized with water and extracted with DCM. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated, yielding 4.2g (quant.) of intermediate 12. fl..PCe .arati n o nteπneώ
Figure imgf000031_0002
A mixture of intermediate 12 (0.0176 mol) in sodium hydroxide 2N (65ml), THF (25ml) and EtOH (25ml) was stirred at room temperature for 15 hours, poured out into water and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated, of intermediate 13. g) Preparation of intermediate 14
Figure imgf000031_0003
Triethylamine (0.03 mol) was added at room temperature to a mixture of intermediate 13 (0.015 mol) in DCM (40ml). Methanesulfonyl chloride (0.015 mol) was added at 0°C under N2 flow. The mixture was stined at 0°C for 1 hour and at room temperature for 3 hours. The solvent was evaporated at room temperature. The product was used without further purification, yielding (quant.) intermediate 14. fe) Prep.ar ation of .inteπnedi ate .1.5
Figure imgf000031_0004
A mixture of intermediate 14 (0.015 mol), ethyl 4-piperidinecarboxylate (0.045 mol) and potassium carbonate (0.045 mol) in acetonitrile (100ml) was stined and refluxed for 15 hours, poured out into water and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-35 μm) (eluent: DCM/MeOH/NHiOH 97/3/0.1 ). The pure fractions were collected and the solvent was evaporated, yielding 0.7g (14%) of intermediate 15.
Figure imgf000031_0005
A mixture of intermediate 15 (0.002 mol) in MeOH (50ml) was hydrogenated at room temperature under a 3 bar pressure for 1 hour with Raney nickel (0.7g) as a catalyst. After uptake of H2 (3 equiv), the catalyst was filtered through celite, washed with MeOH and a small amount of DCM and the filtrate was evaporated, yielding 0.65g (quant.) of intermediate 16.
Example A4
Preparation. of.intermediate.17
Figure imgf000032_0001
nBuLi 1.6 M in hexane (0.148 mol) was added dropwise at -60°C under N2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.114 mol) in THF (500ml) and the mixture was stined at -60°C for lh. Cyclohexanecarboxaldehyde (0.137 mol) in THF (100ml) was added dropwise at -60°C, stined at -60°C for 2h and then stined further at -40°C for lh. The mixture was poured into saturated NHtCl and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The product was used without further purification, yielding 34.13g (quant.) of intermediate 17.
Figure imgf000032_0002
A mixture of intermediate 17 (0.114 mol) in hydrochloric acid 3N (250ml) and THF (250ml) was stined and refluxed for 24 hours. The mixture was cooled and DCM (200ml) was added. The precipitate was filtered off, washed with water and dried. The product was used without further purification, yielding 12.5g (39%) of intermediate 18. c).Prcp.aration of nte
Figure imgf000032_0003
Intermediate 18 (0.035 mol) was added portionwise at 0°C to thionyl chloride
(56.23ml). The mixture was stined at room temperature for 3 hours and the solvent was evaporated. The residue was crystallized from diethyl ether. The precipitate was filtered off, washed several times with diethyl ether and recrystallized from water and DCM. The mixture was stined for 15 hours. The precipitate was filtered off and dried, yielding 7.9g (75%) of intermediate 19. . .Prep.aratipn.of.inte.
Figure imgf000032_0004
A solution of 4,4-piperidinediol, hydrochloride (0.0651 mol) and potassium carbonate (0.217 mol) in DMF (250ml) was stined at room temperature under N2 flow for 10 min. A solution of intermediate 19 (0.0434 mol) in DMF (50ml) was added slowly, and the mixture was stined at room temperature for 1 hour and then at 70°C for one hour. The mixture was cooled to room temperature and poured into water (1500ml). The precipitate was filtered off, washed several times with cold water and dried. The product was used in the next reaction step without further purification. A part (3g) of the residue (13.8g) was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOH/NHtOH 99/1/0.1). The desired fractions were collected and the solvent was evaporated. The residue (2.9g) was crystallized from MEK/DIPE, filtered off and dried, yielding 2.85g of intermediate 20.
Example A5
.a).Prep.ar.a.tipn.pf intermediate. :?!
Figure imgf000033_0001
nBuLi 1.6M in hexane (0.0516 mol) was adde dropwise at -60°C under N2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.043 mol) in THF (115ml). The mixture was stined at -60°C for 1 hour. A mixture of l-cyclohexyl-3-(4-methyl-l- piperazinyl)- 1-propanone, (0.043 mol) in THF (103ml) was added dropwise at -60°C. The mixture was stined at -60°C for 1 hour, brought to 0°C, poured out into a saturated HtCl solution and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/NHtOH 97/3/0.2 and 90/10/0.2). The pure fractions were collected and the solvent was evaporated, yielding 3.7g (20%) of intermediate 21. bXP ep.aration gf fl ' c occ
A mixture of intermediate 21 (0.00841 mol), tin(II) chloride (0.0336 mol) and hydrochloric acid 12N (0.121 mol) in acetic acid (36ml) was stined at 80°C for 16 hours. The mixture was poured out on ice, basified with a concentrated NHUOH solution and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The product was used without further purification, yielding 2.45g (74%) of intermediate 22. Example A6
Figure imgf000034_0001
Phosphoryl chloride (110.9ml) was added dropwise at 5°C to DMF (81.5ml). The mixture was stined until complete dissolution. 4-[(l-oxobutyl)amino]- benzoic acid, ethyl ester (0.34 mol) was added. The mixture was stined at 100°C for 15 hours, then cooled to room temperature and poured out into ice water. The precipitate was filtered off, washed with water and dried, yielding 42.35g (47%) of intermediate 23. b) Preparation of intermediate 24
Figure imgf000034_0002
A mixture of intermediate 23 (0.1606 m in s methanolate 30% solution in MeOH (152.8ml) and MeOH (400ml) was stined 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 24. c).. rep ration of intermediate 25 Hθ'^^ ^ ^\
Lithium aluminum tetrahydride (0.1288 mol) was added portionwise at 0°C under N2 flow to a solution of intermediate 24 (0.1288 mol) in THF (263ml). The mixture was stined 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 25.
.4) Preparatip i.pf .intermediate 26
Figure imgf000034_0003
Methanesulfonyl chloride (0.104 mol) was added dropwise at 0°C under N2 flow to a mixture of intermediate 25 (0.069 mol) and triethylamine (0.207 mol) in DCM (120ml). The mixture was stined at 0°C for 4 hours. The solvent was evaporated till dryness (without heating). The product was used without further purification, yielding 20.4g of intermediate 26. elPjrø ation nteπneώate 27
Figure imgf000034_0004
A mixture of intermediate 26 (0.0691 mol), l-(phenylmethyl)- piperazine (0.0829 mol) and potassium carbonate (0.145 mol) in acetonitrile (150ml) was stined and refluxed for 12 hours. The solvent was evaporated till dryness. The residue was taken up in DCM and water. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The residue (24.6g) was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/NHtOH 98/2/0.1). The pure fractions were collected and the solvent was evaporated. The residue (2.7g) was crystallized from DIPE. The precipitate was filtered off and dried, yielding 1.6g of intermediate 27, melting point 78°C.
Example A7 a) Preparation of intermediate 28
Figure imgf000035_0001
nBuLi 1.6M in hexane (0.224 mol) was added at -78°C under N2 flow to a solution of 6-bromo-3-ethyl-2-methoxy- quinoline (0.188 mol) in THF (500ml). The mixture was stined at -78°C for 1 hour. A mixture of 3-cyclohexene-l-carboxaldehyde (0.182 mol) in THF (500ml) was added dropwise at -78°C. The mixture was stined at -78°C for 2 hours, then brought to 0°C, hydrolized and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (58.9g) was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/EtOAc 96/4). The pure fractions were collected and the solvent was evaporated, yielding 40.5g (72%) of intermediate 28. b).P.re.p.aratipn of intemediate 29
Figure imgf000035_0002
A mixture of intermediate 28 (0.131 mol) in hydrochloric acid 3N (400ml) and THF (400ml) was stined at 60°C overnight and then basified with potassium carbonate solid. The precipitate was filtered off, washed with DCM and dried. The filtrate was extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was crystallized from DCM. The precipitate was filtered off and dried. Part (1.5g) of the residue (16.5g) was taken up in MeOH. The mixture was stined overnight. The precipitate was filtered off and dried, yielding 0.72g of intermediate 29, melting point 212°C. cj.P„rep.aratipπ.ρf. intejmeΛateJ3()
Figure imgf000035_0003
Intermediate 29 (0.053 mol) was added slowly at 0°C to thionyl chloride (150ml). The mixture was stined at room temperature for 4 hours. The solvent was evaporated till dryness. The residue was taken up several times in DCM. The solvent was evaporated. The product was used without further purification, yielding 16g (quant.) of intermediate 30. d). Preparation of intermedi i ate .31
Figure imgf000036_0001
A solution of 4,4-piperidinediol, hydrochloride (0.079 mol) and potassium carbonate (0.265 mol) in DMF (200ml) was stined at room temperature under N2 flow for 10 min. A solution of intermediate 30 (0.053 mol) in DMF (200ml) was added slowly. The mixture was stined at room temperature for 1 hour and then poured out into water. The precipitate was filtered off, washed several times with water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (19.2g) was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOHNILOH 96/4/0.2). The pure fractions were collected and the solvent was evaporated, yielding 11.4g (59%) of intermediate 31.
Example A8 a) Preparation of intermediate 32
Figure imgf000036_0002
nBuLi 1.6M in hexane (0.154 mol) was added dropwise at -60°C under N2 flow to a mixture of 6-bromo-3-ethyl-2-methoxy- quinoline (0.118 mol) in THF (314ml) and the mixture was stined at -60°C for lh. 2-thiophenecarboxaldehyde (0.142 mol) in THF (100ml) was added dropwise at -60°C. The mixture was stirred at -60°C for 2h, then at -40°C for lh, poured out into a saturated NHL4CI solution and extracted with EtOAc. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The product was used without further purification, yielding 35.37g (quant.) of intermediate 32. b.) Preparation
Figure imgf000036_0003
A mixture of intermediate 32 (0.118 mol) in hydrochloric acid 3N (426ml) and THF (274ml) was stined at 70°C for 6 hours. The mixture was poured out on ice, basified with a concentrated NILtOH solution and extracted with EtOAc. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH 98/2). The pure fractions were collected and the solvent was evaporated, yielding 9.3g (28%) of intermediate 33. c) Preparation, of inteπnedi ate 3.4
Figure imgf000037_0001
Intermediate 33 (0.0322 mol) was added portionwise at 0°C to thionyl chloride (46ml). The mixture was stined at room temperature overnight. The solvent was evaporated till dryness. The product was used without further purification, yielding 9.78g (quant.) of intermediate 34. d).Prc »aratipn f ntemeώate 35
Figure imgf000037_0002
Potassium carbonate (0.161 mol) was added to a mixture of 4,4-piperidinediol, hydrochloride (0.0483 mol) in acetonitrile (74ml). The mixture was stined under N2 flow for 15 min. A mixture of intermediate 34 (0.0322 mol) in acetonitrile (98ml) was added at room temperature. The mixture was stirred at 60°C overnight, then poured out into water and extracted with DCM. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOH/NILOH 97/3/0.1). The pure fractions were collected and the solvent was evaporated, yielding 0.46g (4%) of intermediate 35.
B. Preparation of the final compounds
Example Bl
Preparation ..of. compound.!
Figure imgf000037_0003
Sodium hydroborate (0.0318 mol) was added at 0°C under N2 flow to a solution of intermediate 3 (0.0245 mol) in MeOH (80ml). The mixture was stined for 30 min.
Water (10ml) was added. The organic solvent was evaporated. The aqueous concentrate was taken up in DCM and water and the mixture was extracted. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated till dryness. Part (3g) of the residue (7.5g) was crystallized from 2-propanone and a small amount of MeOH.
The precipitate was filtered off and dried, yielding 2.69g of compound 1, melting point
172°C. Example B2
Preparation of compound.?.
Figure imgf000038_0001
Potassium carbonate (0.107 mol) was added to a mixture of 4,4-piperidinediol hydrochloride (0.032 mol) in acetonitrile (49ml). The mixture was stined under N2 flow for 15 min. A mixture of intermediate 7 (0.0213 mol) in acetonitrile (68ml) was added. The mixture was stined at 60°C for 3 hours, then poured out into water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOH/NHtOH 98.5/1.5/0.1). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from diethyl ether. The precipitate was filtered off and dried, yielding 4.16g (51 %)of compound 2, melting point 218°C.
Example B3 Preparatipn.of.comppund.3
Figure imgf000038_0002
A mixture of intermediate 16 (0.002 mol) and ethyl 2-oxobutanoate (0.004 mol) in EtOH (15ml) was stirred and refluxed for 2.5 hours, poured out into water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (0.9g) was purified by column chromatography over silica gel (15-40 μm) (eluent: cyclohexane/2-propanol/NHtOH 85/15/1). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from diethyl ether. The precipitate was filtered off and dried, yielding 0.054g of compound 3, melting point 163°C.
Example B4
Preparation. of compound.4
Figure imgf000038_0003
A mixture of sodium hydride (0.42g) in THF (10.5ml) was stined at room temperature for 10 min. Then THF was decanted off. DMSO (32ml), then trimethylsulfoxonium iodide (0.013 mol) were added. The mixture was stined at room temperature for 1 hour. Intermediate 20 (0.0114 mol) was added slowly. The mixture was stined at room temperature overnight. Water was added and the mixture was extracted with DCM. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The residue was crystallized from 2-propanone/diethyl ether. The precipitate was filtered off and dried. The residue was recrystallized from 2-propanone/diethyl ether. The precipitate was filtered off and dried, yielding 1.54g (36%) of compound 4, melting point 200°C.
Example B5
.Preparation of cpmppund.5
Figure imgf000039_0001
A mixture of intermediate 22 (0.00623 mol) in MeOH (25ml) was hydrogenated at room temperature under a 3 bar pressure for 8 hours with Pd/C 10% (0.25g) as a catalyst. After uptake of H2 (1 equiv), the catalyst was filtered through celite and the filtrate was evaporated. The residue was taken up in water and a concentrated NHtOH solution and the mixture was extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOH/NB OH 94/6/0.3). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from 2-propanone. The precipitate was filtered off and dried, yielding 1.07g (43%) of compound 5, melting point 181°C.
Example B6
Preparation of compound 6
Figure imgf000039_0002
nBuLi 1.6M in hexane (21.32ml) was added dropwise at -70°C under N2 flow to a mixture of 1 -methyl- lH-imidazole (0.0341 mol) in TΗF (28ml). The mixture was stined at -70°C for 30 min. Chlorotriethyl- silane (0.0341 mol) was added. The mixture was brought to room temperature. nBuLi 1.6 M in hexane (21.32ml) was added dropwise at -70°C. The mixture was stined at -70°C for 1 hour and then brought to -15°C. A mixture of intermediate 20 (0.0136 mol) in THF (50ml) was added dropwise at -70°C. The mixture was allowed to warm to room temperature during the night, then poured out into a saturated NH^Cl solution and extracted with EtOAc. The organic layer was separated, dried (MgSO ), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/NHiOH 94/6/0.5). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from 2-propanone. The precipitate was filtered off and dried, yielding 5.05g (83%) of compound 6, melting point 194°C.
Example B7
Preparatipnpf comppund 7
Figure imgf000040_0001
A mixture of intermediate 27 (0.00319 mol) in hydrochloric acid 6N (70ml) was stined at 80°C for 30 min, poured out into water (50ml) and potassium carbonate solid. The mixture was stined for 10 min. The precipitate was filtered off, rinced with water and dried, yielding 0.9g (78%) of compound 7, melting point 194°C.
Example B8
Figure imgf000040_0002
A mixture of intermediate 19 (0.0164 mol) in 2-(dimethylamino)- ethanol (50ml) was stined and refluxed for 2 hours. The mixture was poured out into water and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (16g) was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOH/NILOH 94/6/0.5). The pure fractions were collected. The mixture was allowed to crystallize out for several days (precipitation resulted). The precipitate was filtered off, taken up in petroleum ether, filtered off and dried, yielding 2.8g (48%) of compound 8, melting point 122°C. b). Prep.aratipn.pf cpmpound.9 and.10
Figure imgf000041_0001
compound 9 compound 10
Compound 8 (0.02244 mol) was separated into its enantiomers by column chromatography (eluent: hexane/2-propanol 88/12; column: CHIRALPAK AD). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from hexane and petroleum ether. The precipitate was filtered off and dried, yielding 2.2g of compound 9, melting point 115°C, and 2.02g of compound 10, melting point 115°C.
Example B9
Preparation of compound ..11
Figure imgf000041_0002
Sodium cyanotrihydroborate (0.02 mol) was added portionwise at 0°C under N2 flow to a solution of intermediate 31 (0.02 mol) and 2-methoxy- ethanamine (0.024 mol) in MeOH (100ml). The mixture was stined at room temperature for 12 hours. Water was added and the mixture was extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue (9.7g) was purified by column chromatography over silica gel (20-45 μm) (eluent: DCM/MeOH/NILOH 93/7/0.5). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from 2-propanone and diethyl ether. The precipitate was filtered off and dried, yielding 0.63g of compound 11, melting point 196°C.
Example B10 reparation of .compound .1.2
Figure imgf000041_0003
Sodium cyanotrihydroborate (0.00126 mol) was added at 0°C to a mixture of intermediate 35 (0.00126 mol) and 2-methoxy- ethanamine (0.00151 mol) in MeOH (10ml). The mixture was stined at room temperature overnight, then poured out on ice and extracted with DCM. The organic layer was separated, dried (MgSO4), filtered and the solvent was evaporated. The residue was purified by column chromatography over silica gel (15-40 μm) (eluent: DCM/MeOHNHtOH 90/10/0.1). The pure fractions were collected and the solvent was evaporated. The residue was crystallized from MEK and diethyl ether. The precipitate was filtered off and dried, yielding 0.22g (41%) of compound 12.
Table F-l lists the compounds that were prepared according to one of the above Examples. Table F-l
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Pharmacological example
In vitro Scintillation Proximity Assay (SPA) for PARP-1 inhibitory activity Compounds of the present invention were tested in an in vitro assay based on SPA technology (proprietary to Amersham Pharmacia Biotech).
In principle, the assay relies upon the well established SPA technology for the detection of poly(ADP-ribosyl)ation of biotinylated target proteins, i.e histones. This ribosylation is induced using nicked DNA activated PARP-1 enzyme and [3H]- nicotinamide adenine dinucleotide ([3H]-NAD+) as ADP-ribosyl donor.
As inducer of PARP-1 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 °C, the reaction was terminated by adding 1.45 g NaCl, followed by a further incubation at 58 °C 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 °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 100 mg/ml SPA poly(vinyl 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 - 20 °C). The PARP-1 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-1 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-1 enzyme (concentration was lot dependent) was mixed with the nicked DNA and the mixture was pre-incubated for 30 min. at 4 °C. Equal parts of this histones/PVT-SPA beads solution and PARP-1 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-1 enzyme. After incubation of the mixture for 15 min. at room temperature, the reaction was terminated by adding 100 μl 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 transfened to a TopCountNXT™ (Packard) for scintillation counting, values were expressed as counts per minute (cpm). For each experiment, controls (containing PARP-1 enzyme and DMSO without compound), a blank incubation (containing DMSO but no PARP-1 enzyme or compound) and samples (containing PARP-1 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"6M. When the compounds showed activity at 10*6M, 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-1 enzyme activity. For each sample, the amount of cpm was expressed as a percentage of the mean cpm value of the controls. When appropriate, IC5o-values (concentration of the drug, needed to reduce the PARP-1 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 SPA assay. The tested compounds showed inhibitory activity at the initial test concentration of 10"6M (see Tabel-2).
In vitro filtration assay for PARP-1 inhibitory activity
Compounds of the present invention were tested in an in vitro filtration assay assessing PARP-1 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-1 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-1 inhibitory activity. Seventy-five μl of the PARP- 1 enzyme/DNA mixture together with 1 μl of compound in DMSO and 25 μl of histones-NAD /[ P]- 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 °C 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 transfened to a TopCountNXT™ (supplier: Packard) for scintillation counting and values were expressed as counts per minute (cpm). For each experiment, controls (containing PARP-1 enzyme and DMSO without compound), a blank incubation (containing DMSO but no PARP-1 enzyme or compound) and samples (containing PARP-1 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" M and 10" M. In each test, the blank value was subtracted from both the control and the sample values. The control sample represented maximal PARP-1 enzyme activity. For each sample, the amount of cpm was expressed as a percentage of the mean cpm value of the controls. When appropriate, IC5o-values (concentration of the drug, needed to reduce the PARP-1 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 pIC5o (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). Tabel-2
Figure imgf000048_0001
The compounds can be further evaluated in a cellular chemo- and/or radiosensitization assay, an assay measuring inhibition of endogenous PARP-l activity in cancer cell lines and eventually in an in vivo radiosensitization test.

Claims

CLABVIS
1. A compound of formula (I),
Figure imgf000049_0001
the N-oxide forms, the addition salts and the stereo-chemically isomeric forms thereof, wherein
n is O or l; s is O or 1;
X is -Ν= or -CR4=, wherein R4 is hydrogen or taken together with R1 may form a bivalent radical of formula -CH=CH-CH=CH-;
Y is -N< or -CH<;
Q is -NH-, -O-, -C(O)-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, d^alkyl, arylCι_6alkyl, C1-6alkyloxycarbonyl, Cϊ-όalkyloxyCi-ealkylamino or haloindazolyl;
R1 is d^alkyl or thienyl;
R is hydrogen or taken together with R may form =O;
R3is hydrogen, Ci-όalkyl or a radical selected from - NR6R7 (a-1), -O-H (a-2), -O-R8 (a-3), -S- R9 (a-4), or — C≡N (a-5), wherein R6 is -CHO, Ci-βalkyl, hydroxyCι_6alkyl, C)-6alkylcarbonyl,
Figure imgf000049_0002
Ci-6alkylcarbonylaminoCi_6alkyl, piperidinylC1-6alkyl, piperidinylCι.6alkylaminocarbonyl,
Figure imgf000050_0001
Cι_6alkyloxyC1-6alkyl, thienylC1-6alkyl, pynolylCι-6alkyl, arylCι-6alkylpiperidinyl, arylcarbonyld-όalkyl, arylcarbonylpiperidinylCι-6alkyl, haloindozolylpiperidinylC1-6alkyl, or arylC1-6alkyl(Cι-6alkyl)aminoC1-6alkyl; and R7 is hydrogen or Chalky!; R8is Cj.6alkyl, d-βalkylcarbonyl or di(C1-6alkyl)aminoCι.6alkyl; and R9is di(Cι^alkyl)aminoC1-6alkyl; or R3 is a group of formula -(CH2)t-Z- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
Figure imgf000050_0002
(c-1) (c-2) (c-3) (c-4)
Figure imgf000050_0003
(c-9) (c-10) (c-11) (c-12) (c-13) wherein each R l, aminocarbonyl, hydroxy,
Figure imgf000050_0004
C i .6alkyloxyC i -6alkyl , C i .6alkyloxyC i .βalkylamino, d phenyld-δalkenyl), piperidinylC1- alkyl, C3-10cycloalkyl, C3-ι0cycloalkylCι-6alkyl, aryloxy(hydroxy)Ci_6alkyl, haloindazolyl, arylCι-6alkyl, arylC2-6alkenyl, mo holino, Cι.6alkylimidazolyl, or pyridinyld.6alkylamino; each R11 independently is hydrogen, hydroxy, piperidinyl or aryl; aryl is phenyl or phenyl substituted with halo, C1-6alkyl or d^alkyloxy; with the proviso that 6-(cyclohexyl-lH-imidazol-l-ylmethyl)-3-methyl-2(lH)- quinoxalinone is not included.
2. A compound as claimed in claim 1 wherein X is -N= or -CΗ=; R1 is Ci^alkyl; R3 is hydrogen, Cι-6alkyl, a radical selected from (a-1), (a-2), (a-3) or (a-4) or a group of formula (b-1); R6 is di(d.6alkyl)aminoC1-6alkyl or Ci-όalkyloxyd-όalkyl; R7 is hydrogen; R8 is di(Cι.6alkyl)aminoCι-6alkyl; t is 0 or 2; Z is a heterocyclic ring system selected from (c-1), (c-5), (c-6), (c-8), (c-10), (c-12) or (c-13); each R10 independently is hydrogen, Chalky!, hydroxy, Cι.6alkyloxyCι-6alkyl, Ci-6alkyloxyCi.6alkylamino, morpholino, C1-6alkylimidazolyl, or pyridinylCι-6alkylamino; each R1 'independently is hydrogen or hydroxy; and aryl is phenyl.
3. A compound according to claim 1 and 2 wherein n is 0; X is CH; Q is -NH-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, or arylC].6alkyl; R1 is Chalky!; R2 is hydrogen; R3 is hydrogen, hydroxy or a group of formula (b-1); t is 0; Z is a heterocyclic ring system selected from (c-8) or (c-13); each R10 independently is hydrogen; and aryl is phenyl.
4. A compound according to claim 1, 2 and 3 wherein the compound is selected from
Figure imgf000051_0001
5. A compound as claimed in any of claims l to 4 for use as a medicine.
6. A pharmaceutical composition comprising pharmaceutically acceptable caniers 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 the compound is a compound of formula (I)
Figure imgf000052_0001
the N-oxide forms, the phannaceutically acceptable addition salts and the stereochemically isomeric forms thereof, wherein
n is O or 1; s is O or 1;
X is -Ν= or -CR4=, wherein R4 is hydrogen or taken together with R1 may form a bivalent radical of formula -CH=CH-CH=CH-;
Y is -N< or -CH<;
Q is -NH-, -O-, -C(O)-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, C1-6alkyl, arylCι-6alkyl, d-όalkyloxycarbonyl, Cι-6alkyloxyCι_6alkylamino or haloindazolyl;
R1 is Cι-6alkyl or thienyl;
R2 is hydrogen or taken together with R3 may form =O;
R3 is hydrogen, Cι.6alkyl or a radical selected from - NR6R7 (a-1), -O-H (a-2), -O-R8 (a-3), -S- R9 (a-4), or — C≡N (a-5), 'herein
R 6° ; i„s r
Figure imgf000053_0001
C<1-6.alkyl, hydroxyC1-6alkyl, d-6alkylcarbonyl,
Figure imgf000053_0002
Cι-6alkylcarbonylaminoCι-6alkyl, piperidinylC i - alkyl , piperidinylC -6alkyl aminocarbonyl , C -6alkyloxy , d-βalkyloxyd-βalkyl, thienyld-δalkyl,
Figure imgf000053_0003
arylC i -6alkylpiperidinyl , arylcarbonylC i -βalkyl , arylcarbonylpiperidinylC ι -6alkyl , haloindozolylpiperidinyld-ealkyl, or arylCi-όalkyKCi-ήalky aminoCi-όalkyl; and
R7 is hydrogen or Ci-βalkyl;
R8is d-βalkyl, Ci-6alkylcarbonyl or
Figure imgf000053_0004
and
Figure imgf000053_0005
is a group of formula
-(CH2)t-Z- (b-1), wherein t is 0, I or 2; Z is a heterocyclic ring system selected from
HN R
Figure imgf000053_0006
(c-1) (c-2) (c-3) ( - )
Figure imgf000053_0007
(c-9) (c-10) (c-11) (c-12) (c-13) wherein each R10 independently is hydrogen, C]-6alkyl, aminocarbonyl, hydroxy,
Figure imgf000054_0001
Ci-βalkyloxyCi-όalkyl, Cι.6alkyloxyCι.6alkylamino, di(phenylC2-6alkenyl),
Figure imgf000054_0002
C3-1ocycloalkyl, C3-1ocycloalkylCι-6alkyl, aryloxy(hydroxy)Cι-6alkyl, haloindazolyl, aryld^alkyl, arylC2-6alkenyl, morpholino, d-βalkylimidazolyl, or pyridinylCι.6alkylamino; each Ru independently is hydrogen, hydroxy, piperidinyl or aryl;
aryl is phenyl or phenyl substituted with halo, d.6alkyl,or C1-6alkyloxy.
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-1 mediated disorder
10. Use according to claim 8 and 9 wherein the treatment involves chemosensitization.
11. Use according to claims 8 and 9 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 imgf000054_0003
the N-oxide forms, the pharmaceutically acceptable addition salts and the stereochemically isomeric forms thereof, wherein
n is O or 1; s is 0 or 1 ;
X is -Ν= or -CR4=, wherein R4 is hydrogen or taken together with R1 may form a bivalent radical of formula -CH=CH-CH=CH-; Y is -N< or -CH<;
Q is -NH-, -O-, -C(O)-, -CH2-CH2- or -CHR5-, wherein R5 is hydrogen, hydroxy, d-6alkyl, aryld.6alkyl, Cι.6alkyloxycarbonyl, C 1 -6alkyloxyC 1 -6alkylamino or haloindazolyl ;
R1 is Cι.6alkyl or thienyl;
R2 is hydrogen or taken together with R3 may form =O;
R3 is hydrogen, d.6alkyl or a radical selected from - NR6R7 (a-1), -O-H (a-2), -O-R8 (a-3), -S- R9 (a-4), or — C≡N (a-5), wherein R6 is -CHO, d.6alkyl, hydroxyd-βalkyl, d.6alkylcarbonyl,
Figure imgf000055_0001
C1-6alkylcarbonylaminoCi.6alkyl, piperidinylC t-βalkyl, piperidinylCi-ealkylaminocarbonyl, C1-6alkyloxy, Cι-6alkyloxyCι-6alkyl, thienylCι-6alkyl, pynolylCi-βalkyl, arylCϊ-ealkylpiperidinyl,
Figure imgf000055_0002
arylcarbonylpiperidinylCι-6alkyl, haloindozolylpiperidinyld.6alkyl, or arylC1.6alkyl(Cι-6alkyl)aminoC1.6alkyl; and R7 is hydrogen or d-βalkyl; R8 is d ealkyl, d-6alkylcarbonyl or di(Cι. alkyl)aminoCι.6alkyl; and R9is di(C1-6alkyl)aminoCι-6alkyl; or R3 is a group of formula -(CH2)rZ- (b-1), wherein t is 0, 1 or 2; Z is a heterocyclic ring system selected from
Figure imgf000055_0003
(c-1) (c-2) (c-3) (c-4)
Figure imgf000056_0001
(c-5) (c-6) (c-7)
Figure imgf000056_0002
Figure imgf000056_0003
(c-9) (c-10) (c-11) (c-12) (c-13) wherein each R10 independently is hydrogen, Cι_6alkyl, aminocarbonyl, hydroxy,
Figure imgf000056_0004
C i -βalkyloxyC i .6alkyl , C ι -βalkyloxyC ι .6alkyl amino, di (phenylC2-6alkenyl ) , piperidinylCι-6alkyl, C3-ιocycloalkyl, C3-ι0cycloalkylCι.6alkyl, aryloxy(hydroxy)Cι-6alkyl, haloindazolyl, arylCι-6alkyl, arylC2-6alkenyl, morpholino, Cι-6alkylimidazolyl, or pyridinylCi^alkylamino; each R11 independently is hydrogen, hydroxy, piperidinyl or aryl; aryl is phenyl or phenyl substituted with halo, C1-6alkyl or C1-6alkyloxy.
13. A process for preparing a compound as claimed in claim 1, characterized by a) the hydrolysis of intermediates of formula (NTH), according to art-known methods, by submitting the intermediates of formula (VIII) to appropriate reagents, such as, tinchloride, acetic acid and hydrochloric acid, in the presence of a reaction inert solvent, e.g. tetrahydrofuran.
Figure imgf000056_0005
(VII D (I) b) the cyclization of intermediates of formula (X), according to art-known cyclizing procedures into compounds of formula (I) wherein X is CH herein refened to as compounds of formula (I-j), preferably in the presence of a suitable Lewis Acid, e.g. aluminum chloride either neat or in a suitable solvent such as, for example, an aromatic hydrocarbon, e.g. benzene, chlorobenzene, methylbenzene and the like; halogenated hydrocarbons, e.g. trichloromethane, tetrachloromethane and the like; an ether, e.g. tetrahydrofuran, 1,4-dioxane and the like or mixtures of such solvents.
Figure imgf000057_0001
c) the condensation of an appropriate ortho-benzenediamine of formula (XI) with an ester of formula (XII) into compounds of formula (I), wherein X is N and R2 taken together with R3 forms =O, herein refened to as compounds of fonnula (I-a-1), in the presence of a carboxylic acid, e.g. acetic acid and the like, a mineral acid such as, for example hydrochloric acid, sulfuric acid, or a sulfonic acid such as, for example, methanesulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid and the like.
Figure imgf000057_0002
(XD (XII) <H)
PCT/EP2004/013165 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors WO2005058843A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
ES04803192.6T ES2565581T3 (en) 2003-12-10 2004-11-18 2-Quinolinones / 2-quinoxalinones 6- (hetero) cyclohexylalkyl substituted as poly (ADP-ribose) polymerase inhibitors
EA200601125A EA010592B1 (en) 2003-12-10 2004-11-18 Substituted6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
AU2004299183A AU2004299183B2 (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
EP04803192.6A EP1694653B1 (en) 2003-12-10 2004-11-18 6-(hetero-)cyclohexylalkyl substituted 2-quinolinones/2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
BRPI0417571-9A BRPI0417571A (en) 2003-12-10 2004-11-18 2-Quinolinones and 2-quinoxalinones substituted by 6-cyclohexyl-alkyl substituted as poly (adp-ribose) polymerase inhibitors
JP2006543409A JP4948178B2 (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly (ADP-ribose) polymerase inhibitors
US10/596,083 US7652014B2 (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
KR1020067013344A KR101149031B1 (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as polyadp-ribosepolymerase inhibitors
NZ547278A NZ547278A (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as PARP inhibitors for chemosensitization or radiosensitization
CA2548273A CA2548273C (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
IL176199A IL176199A (en) 2003-12-10 2006-06-08 2-quinolinones and 2-quinoxalinones substituted by 6-cyclohexylalkyl as poly (adp-ribose) polymerase inhibitors, process for their manufacture and use thereof in the preparation of medicaments
NO20063129A NO337621B1 (en) 2003-12-10 2006-07-05 Substituted 6-cyclohexylalkyl-substituted 2-quinolinones and 2-quinoxalinones as poly (ADP-ribose) polymerase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP03078918.4 2003-12-10
EP03078918 2003-12-10

Publications (2)

Publication Number Publication Date
WO2005058843A1 true WO2005058843A1 (en) 2005-06-30
WO2005058843A8 WO2005058843A8 (en) 2006-06-15

Family

ID=34684557

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/013165 WO2005058843A1 (en) 2003-12-10 2004-11-18 Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors

Country Status (17)

Country Link
US (1) US7652014B2 (en)
EP (1) EP1694653B1 (en)
JP (1) JP4948178B2 (en)
KR (1) KR101149031B1 (en)
CN (2) CN1890225A (en)
AU (1) AU2004299183B2 (en)
BR (1) BRPI0417571A (en)
CA (1) CA2548273C (en)
EA (1) EA010592B1 (en)
ES (1) ES2565581T3 (en)
IL (1) IL176199A (en)
NO (1) NO337621B1 (en)
NZ (1) NZ547278A (en)
SG (1) SG151250A1 (en)
UA (1) UA91007C2 (en)
WO (1) WO2005058843A1 (en)
ZA (1) ZA200604774B (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008107478A1 (en) * 2007-03-08 2008-09-12 Janssen Pharmaceutica Nv Quinolinone derivatives as parp and tank inhibitors
WO2009053373A1 (en) * 2007-10-26 2009-04-30 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
WO2010085570A1 (en) 2009-01-23 2010-07-29 Takeda Pharmaceutical Company Limited Poly (ADP-Ribose) Polymerase (PARP) Inhibitors
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
US8377985B2 (en) 2005-07-18 2013-02-19 Bipar Sciences, Inc. Treatment of cancer
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
US8623872B2 (en) 2004-06-30 2014-01-07 Janssen Pharmaceutica, Nv Quinazolinone derivatives as PARP 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
US8980902B2 (en) 2009-07-30 2015-03-17 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
US9221804B2 (en) 2013-10-15 2015-12-29 Janssen Pharmaceutica Nv Secondary alcohol quinolinyl modulators of RORγt
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
WO2018060073A1 (en) 2016-09-29 2018-04-05 Bayer Cropscience Aktiengesellschaft Novel 5-substituted imidazole derivatives
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
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
US11325906B2 (en) 2019-07-19 2022-05-10 Astrazeneca Ab Chemical compounds

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG166019A1 (en) * 2004-01-23 2010-11-29 Janssen Pharmaceutica Nv Quinoline derivatives and use thereof as mycobacterial inhibitors
CA2977685C (en) 2015-03-02 2024-02-20 Sinai Health System Homologous recombination factors
WO2017156350A1 (en) 2016-03-09 2017-09-14 K-Gen, Inc. Methods of cancer treatment
CN106588993A (en) * 2016-09-02 2017-04-26 瑞声光电科技(常州)有限公司 Iridium complex, preparation method thereof and light-emitting device using the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0371564A2 (en) * 1988-11-29 1990-06-06 Janssen Pharmaceutica N.V. (1H-azol-1-ylmethyl)substituted quinoline, quinazoline or quinoxaline 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

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI77852C (en) * 1981-02-17 1989-05-10 Otsuka Pharma Co Ltd Process for the preparation of novel, such as cardiac drugs, useful s unsubstituted amide and (saturated heterocycle) carbonyl carbostyril derivatives.
AU532361B2 (en) * 1981-09-01 1983-09-29 Otsuka Pharmaceutical Co., Ltd. Carbostyril derivatives
KR0149162B1 (en) * 1988-11-29 1998-10-15 구스타프 반 리이트 (1h-azol-1-yemethyl)substituted quinoline, quinazoline or quinoxaline derivatives
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
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
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

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
EP0371564A2 (en) * 1988-11-29 1990-06-06 Janssen Pharmaceutica N.V. (1H-azol-1-ylmethyl)substituted quinoline, quinazoline or quinoxaline derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LI J-H ET AL: "PARP inhibitors", IDRUGS, CURRENT DRUGS LTD, GB, vol. 4, no. 7, 2001, pages 804 - 812, XP002976576, ISSN: 1369-7056 *

Cited By (56)

* 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
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
US8623872B2 (en) 2004-06-30 2014-01-07 Janssen Pharmaceutica, Nv Quinazolinone derivatives as PARP inhibitors
US10150757B2 (en) 2004-06-30 2018-12-11 Janssen Pharmaceutica Nv Quinazolinone derivatives as PARP inhibitors
US9522905B2 (en) 2004-06-30 2016-12-20 Janssen Pharmaceutica Nv Quinazolinone derivatives as PARP inhibitors
US9255080B2 (en) 2004-06-30 2016-02-09 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
WO2008107478A1 (en) * 2007-03-08 2008-09-12 Janssen Pharmaceutica Nv Quinolinone derivatives as parp and tank inhibitors
US8299256B2 (en) 2007-03-08 2012-10-30 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP and TANK inhibitors
US20130018017A1 (en) * 2007-03-08 2013-01-17 Jorge Eduardo Vialard Quinolinone derivatives as parp and tank inhibitors
AU2008223793B2 (en) * 2007-03-08 2012-08-23 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
US20100168065A1 (en) * 2007-03-08 2010-07-01 Janssen Pharmaceutica Nv Quinolinone derivatives as parp and tank inhibitors
JP2010520262A (en) * 2007-03-08 2010-06-10 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Quinolinone derivatives as PARP and TANK inhibitors
US8404713B2 (en) 2007-10-26 2013-03-26 Janssen Pharmaceutica Nv Quinolinone derivatives as PARP inhibitors
WO2009053373A1 (en) * 2007-10-26 2009-04-30 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
US8168644B2 (en) 2008-03-27 2012-05-01 Janssen Pharmaceutica Nv Quinazolinone derivatives as tubulin polymerization inhibitors
US8889866B2 (en) 2008-03-27 2014-11-18 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
US9598396B2 (en) 2008-03-27 2017-03-21 Janssen Pharmaceutica Nv Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as PARP and tubulin polymerization inhibitors
US9187497B2 (en) 2009-01-23 2015-11-17 Takeda Phamaceutical Company Limited Substituted pyrido[3,2-e]pyrrolo[1,2-a]pyrazines as inhibitors of poly(ADP-ribose)polymerase (PARP)
WO2010085570A1 (en) 2009-01-23 2010-07-29 Takeda Pharmaceutical Company Limited Poly (ADP-Ribose) Polymerase (PARP) Inhibitors
EA020301B1 (en) * 2009-01-23 2014-10-30 Такеда Фармасьютикал Компани Лимитед Poly(adp-ribose)polymerase (parp) inhibitors
US8822470B2 (en) 2009-01-23 2014-09-02 Takeda Pharmaceutical Company Limited Substituted pyrido[2,3-b]pyrazines
US8450323B2 (en) 2009-01-23 2013-05-28 Takeda Pharmaceutical Company Limited Substituted derivatives of pyrido[3,2-e][1,4]thiazino[4,3-a]pyrazine and pyrido[3,2-e][1,4]oxazino[4,3-a]pyrazine
US7928105B2 (en) 2009-01-23 2011-04-19 Takeda Pharmaceutical Company Limited Substituted 6a,7,8,9-tetrahydropyrido[3,2-e]pyrrolo[1,2-a]pyrazin-6(5H)-ones
US8124606B2 (en) 2009-01-23 2012-02-28 Takeda Pharmaceutical Company Limited Substituted 7,8,9,10-tetrahydro-5H-dipyrido[1,2-a:3′,2′-e]pyrazin-6(6aH)-ones
US8980902B2 (en) 2009-07-30 2015-03-17 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
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
US9221804B2 (en) 2013-10-15 2015-12-29 Janssen Pharmaceutica Nv Secondary alcohol quinolinyl modulators of RORγt
US9403816B2 (en) 2013-10-15 2016-08-02 Janssen Pharmaceutica Nv Phenyl 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
US9284308B2 (en) 2013-10-15 2016-03-15 Janssen Pharmaceutica Nv Methylene linked quinolinyl modulators of RORγt
US10555941B2 (en) 2013-10-15 2020-02-11 Janssen Pharmaceutica Nv Alkyl linked quinolinyl modulators of RORγt
US9328095B2 (en) 2013-10-15 2016-05-03 Janssen Pharmaceutica Nv Heteroaryl linked quinolinyl modulators of RORgammat
US10201546B2 (en) 2013-10-15 2019-02-12 Janssen Pharmaceutica Nv Quinolinyl modulators of RORγt
US10369146B2 (en) 2013-10-15 2019-08-06 Janssen Pharmaceutica Nv Phenyl linked 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
WO2018060073A1 (en) 2016-09-29 2018-04-05 Bayer Cropscience Aktiengesellschaft Novel 5-substituted imidazole derivatives
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
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
US11325906B2 (en) 2019-07-19 2022-05-10 Astrazeneca Ab Chemical compounds
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
AU2004299183B2 (en) 2010-09-23
CN102206180B (en) 2013-05-29
NZ547278A (en) 2010-01-29
AU2004299183A1 (en) 2005-06-30
IL176199A (en) 2012-03-29
ZA200604774B (en) 2007-11-28
CN102206180A (en) 2011-10-05
EP1694653B1 (en) 2016-01-20
JP2007513898A (en) 2007-05-31
EA010592B1 (en) 2008-10-30
US7652014B2 (en) 2010-01-26
EA200601125A1 (en) 2006-10-27
NO337621B1 (en) 2016-05-09
IL176199A0 (en) 2006-10-05
CA2548273A1 (en) 2005-06-30
BRPI0417571A (en) 2007-03-20
JP4948178B2 (en) 2012-06-06
UA91007C2 (en) 2010-06-25
ES2565581T3 (en) 2016-04-05
EP1694653A1 (en) 2006-08-30
SG151250A1 (en) 2009-04-30
WO2005058843A8 (en) 2006-06-15
US20090042881A1 (en) 2009-02-12
NO20063129L (en) 2006-07-05
KR101149031B1 (en) 2012-05-29
KR20060108753A (en) 2006-10-18
CA2548273C (en) 2013-04-16
CN1890225A (en) 2007-01-03

Similar Documents

Publication Publication Date Title
EP1694653B1 (en) 6-(hetero-)cyclohexylalkyl substituted 2-quinolinones/2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
AU2004295059B2 (en) 6-substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US8524714B2 (en) 7-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
AU2004295058B2 (en) 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
IL175755A (en) 6-alkenyl and 6-phenylalkyl substituted 2-quinolinones and 2-quinoxalinones, pharmaceutical compositions comprising them, processes for their preparation and uses thereof for the preparation of medicaments

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480036656.3

Country of ref document: CN

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 KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL 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): BW GH 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 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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1200600768

Country of ref document: VN

WWE Wipo information: entry into national phase

Ref document number: 2004803192

Country of ref document: EP

Ref document number: 547278

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2004299183

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 12006501064

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 10596083

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2548273

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 176199

Country of ref document: IL

Ref document number: 2006543409

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2006/04774

Country of ref document: ZA

Ref document number: 3331/DELNP/2006

Country of ref document: IN

Ref document number: 200604774

Country of ref document: ZA

Ref document number: PA/a/2006/006573

Country of ref document: MX

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 26/2005 ADD "DECLARATION UNDER RULE 4.17: - AS TO APPLICANT'S ENTITLEMENT TO APPLY FOR AND BE GRANTED A PATENT (RULE 4.17(II))."; ADD "DECLARATION UNDER RULE 4.17: - AS TO THE APPLICANT'S ENTITLEMENT TO CLAIM THE PRIORITY OF THE EARLIER APPLICATION (RULE 4.17(III)) FOR ALL DESIGNATIONS."; ADD "DECLARATION UNDER RULE 4.17: - OF INVENTORSHIP (RULE 4.17(IV)) FOR US ONLY."

ENP Entry into the national phase

Ref document number: 2004299183

Country of ref document: AU

Date of ref document: 20041118

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004299183

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1020067013344

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200601125

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 2004803192

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067013344

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0417571

Country of ref document: BR