WO2001016137A1 - Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage - Google Patents

Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage Download PDF

Info

Publication number
WO2001016137A1
WO2001016137A1 PCT/US2000/023745 US0023745W WO0116137A1 WO 2001016137 A1 WO2001016137 A1 WO 2001016137A1 US 0023745 W US0023745 W US 0023745W WO 0116137 A1 WO0116137 A1 WO 0116137A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
disease
diseases
group
Prior art date
Application number
PCT/US2000/023745
Other languages
French (fr)
Inventor
Jia-He Li
Jie Zhang
Original Assignee
Guilford Pharmaceuticals Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Guilford Pharmaceuticals Inc. filed Critical Guilford Pharmaceuticals Inc.
Priority to AU70871/00A priority Critical patent/AU7087100A/en
Priority to EP00959578A priority patent/EP1212328B1/en
Priority to DE60029799T priority patent/DE60029799T2/en
Priority to CA2382317A priority patent/CA2382317C/en
Priority to MXPA02002213A priority patent/MXPA02002213A/en
Priority to JP2001519703A priority patent/JP2003508400A/en
Publication of WO2001016137A1 publication Critical patent/WO2001016137A1/en
Priority to AU2005202592A priority patent/AU2005202592B2/en

Links

Classifications

    • 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
    • C07D491/06Peri-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • the present invention relates to inhibitors of the nuclear enzyme poly(adenos ⁇ ne 5'-d ⁇ phospho-nbose) polymerase ["poiy( ADP- ⁇ bose) polymerase” or "PARP". which is also referred to as ADPRT (NAD protein (ADP-nbosyl transferase (polymersing)).
  • ADPRT NAD protein (ADP-nbosyl transferase (polymersing)
  • pADPRT poly( ADP- ⁇ bose) transferase
  • PARS polyv (ADP- ⁇ bose) synthetase
  • present ention provides methods of using the disclosed PARP inhibitors to prevent and/or treat tissue damage resulting from cell damage or death due to necrosis or apoptosis.
  • mvocardial infarction to treat other conditions and/or disorders such as. for example, age- related muscular degeneration.
  • AIDS and other immune senescence diseases arthritis, atherosclerosis, cachexia.
  • cancer degenerative diseases of skeletal muscle involving replicative senescence, diabetes (such as diabetes mellitus).
  • inflammatory bowel disorders such as colitis and Crohn's disease
  • acute pancreatitis mucositis.
  • hemorrhagic shock splanchnic artery occlusion shock, multiple organ failure (such as involving am of the kidne . li er, renal, pulmonary retinal, pancreatic and/or skeltal muscle systems), acute autoimmune th roidms. muscular dystrophy .
  • osteoa ⁇ h ⁇ tis osteoporosis, chronic and acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotouc shock), local and/or remote endothehal cell dysfunction (such are recognized by endo-dependent relaxant responses and up-regulation of adhesion molecules), inflammation and skin aging, to extend the lifespan and proliferative capacity of cells, such as. for example, as general mediators m the generation of ox dants. proinflammatory mediators and/or cytokines. and general mediators of leukocyte infiltration, calcium ion overload, phosphohpid peroxidaion. impaired nitnc oxide metabolism and/or reduced ATP production, to alter gene expression of senescent cells, or to rad osensitize hypoxic tumor cells
  • PARP (EC 2 4 2 30). also known as PARS (for poly( ADP- ⁇ bose) synthetase). or ADPRT (for NAD protein (ADP-nbosyl) transferase (polyme ⁇ sing)). or pADPRT (for polv (ADP- ⁇ bose) transferase).
  • ADP- ⁇ bose for poly( ADP- ⁇ bose) synthetase.
  • ADPRT for NAD protein (ADP-nbosyl) transferase (polyme ⁇ sing)
  • pADPRT for polv (ADP- ⁇ bose) transferase
  • ADP- ⁇ bose polv transferase
  • DP- ⁇ bose are directlv or indirecth involv ed in maintaining DNA integ ⁇ ty
  • Thev include histones. topoisomerases. DNA and RNA polymerases. DNA ligases. and Ca 2' - and Mg :" -dependent endonucleases
  • PARP protein is expressed at a high lev el in many tissues, most notably in the immune system, hea ⁇ . brain and germ-line cells Under normal ph siological conditions, there is minimal PARP activity However.
  • DNA damage causes an immediate activation of PARP bv up to 500-fold Among the many functions att ⁇ aded io P ARP is its major role in facilitaung DNA repair bv ADP- ⁇ bosy lation and therefore co-ordinating a number of DNA repair proteins As a result of PARP activation. NAD levels significantly decline While many endogenous and exogenous agents have been shown to damage DNA and acti ate PARP. peroxynit ⁇ te. formed from a combination of nit ⁇ c oxide (NO) and superoxide. appears to be a main perpetrator responsible for va ⁇ ous reported disease conditions in v IV ⁇ . e g . du ⁇ ng shock and inflammation
  • PARP and PARG form a cycle that com ens a large amount of NAD to ADP- ⁇ bose
  • over-stimulation of PARP can cause a drop of NAD and ATP to less than 20% of the normal le el
  • Such a strig ⁇ o is especiallv det ⁇ mental du ⁇ ng ischaemia when dep ⁇ vation of oxygen lias already drastically compromised cellular energy output
  • Subsequent free radical production du ⁇ ng reperfusion is assumed to be a major cause of tissue damage Part of the ATP drop, hich is typical in manv organs du ⁇ ng ischaemia and reperfusion. could be linked to NAD depletion due to polv (ADP- ⁇ bose) turnover
  • PARP or PARG inhibition is expected to preserve the cellular energy level to potentiate the sur ival of lschaemic tissues after insult
  • Polv (.ADP- ⁇ bose) sy nthesis is also inv olv ed in the induced expression of a number of genes essential for inflammatory response
  • P.ARP inhibitors suppress production of inducible nit ⁇ c oxide s nthase (iNOS ) in macrophages.
  • P-type selectin and intercellular adhesion molecule- 1 (ICAM-1 ) in endothehal 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 (Zhang. J "'PARP inhibition a novel approach to treat lschaemia/reperfusion and inflammation-related inju ⁇ es". Chapter 10 in Emerging Drugs ( 1999) 4 209-221 Ashley Publications Ltd . and references cited therein )
  • P.ARP production is activated by damaged DNA fragments which, once activated, catalyzes the attachment of up to 100 ADP- ⁇ bose units to a va ⁇ ety of nuclear proteins, including histones and PARP itself
  • NAD the source of ADP- ⁇ bose and P.ARP substrate
  • NAD is depleted by massive P.ARP activation and. in the efforts to re-synthesize NAD.
  • PARP activation can also be used as a measure of damage following neurotoxic insults following over- exposure to any of glutamate (via NMD A receptor stimulation), reactive oxygen intermediates, amvloid ⁇ - protein.
  • MPTP N-methyI-4-phenv 1-1.2.3 6-tetrahv dropy ⁇ dine
  • MPP ⁇ us active metabolite N-methyl-4- phenylpv ⁇ dine
  • PARP Polymerase
  • over-stimulated neurons secrete more glutamate. creating a feedback loop or domino effect which ultimatelv results in cell damage or death via the production of proteases, lipases and free radicals
  • Excessi e activation of glutamate receptors has been implicated in va ⁇ ous neurological diseases and conditions including epilepsy, stroke. Alzheimer s disease. Park ⁇ nson " s disease. Amvotrophic 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 manv animal species, as well as in cerebral cortical cultures treated with glutamate or NMDA. that glutamate receptor antagonists (1 e . compounds which block glutamate from binding to or activating its receptor) block neural damage following vascular stroke Davvson et al . "Protection of the Brain from Ischemia" Cerebrovascular Disease 319-25 (H Hunt Batjer ed . 1997) Attempts to prevent excitoto.xicm by blocking NMDA. AMP A.
  • NMDA receptors bv glutamate activates the enzyme neuronal ine oxide synthase (nNOS) leading to the formation of nitnc oxide (NO), which also mediates neurotoxicity NMDA neurotoxicity mav be prevented b treatment with nit ⁇ c o ide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro Daw son et al .
  • nNOS neuronal ine oxide synthase
  • NOS nit ⁇ c o ide synthase
  • Dawson et al Mechanisms of Nit ⁇ c Oxide-mediated Neurotoxicity in P ⁇ mary Brain Cultures.
  • PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis Salzman et al . "Role of Peroxv nit ⁇ te and (ADP-R ⁇ bose)S nthase Activation Expe ⁇ mental Colitis.” Japanese J Pharm . 75.
  • PARP inhibitors hav e been shown to be useful for treating endotoxic shock or septic shock Zingarelh et al . "Protective Effects of Nicotina ⁇ ude against Nit ⁇ c Oxide-Mediated Delayed Vascular Failure in Endotoxic Shock Potential Involvement of PolyADP Ribosvl Synthetase.” Shock. 5 258-64 (1996).
  • nicotinamide protects against delayed NO-mediated vascular failure in endotoxic shock Zingarelh et al also found that the actions of nicotinamide may be related to inhibition of the NO-mediated activation of the energy -consuming DNA repair cycle, t ⁇ ggered bv poly(ADP
  • PARP inhibitors have been used to treat cancer Suto et al "Dihydroisoquinolino ⁇ es The Design and Synthesis of New Se ⁇ es of Potent Inhibitors of Poly (ADP-Ribose) Poiymerase". Anticancer Drug Des . 7 107- 17 (1991) In addition. Suto et al . 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(5H)-Phenanth ⁇ d ⁇ none. an Inhibitor of Poly (ADP- ⁇ bose) Polymerase. on Cultured Tumor Cells".
  • Oncol Res . 6 9 399-403 (1994) discusses the inhibition of PARP activ lty . reduced proliferation of tumor cells, and a marked synergistic effect when tumor cells are co-treated with an alkylating drug
  • PARP inhibitors are the treatment of pe ⁇ pheral nerve inju ⁇ es. and the resultant pathological pain syndrome known as neuropatluc pain, such as that induced by chronic const ⁇ ction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characte ⁇ zed by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark” neurons) occurs Mao et al . Pain.
  • CCI chronic const ⁇ ction injury
  • 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, osteoarth ⁇ tis. osteoporosis, muscular dystrophy degenerative diseases of skeletal muscle involv ing replicative senescence, age-related muscular degeneration, immune senescence AIDS, and other immune senescence diseases, and to alter gene expression of senescent cells WO 98/279"5 Large numbers of known P.ARP inhibitors hav e been desc ⁇ bed in Banasik et al . "Specific Inhibitors of
  • the present invention provides compounds, compositions for. and methods of. inhibiting PARP activity for treating 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 mv ention are specifically useful in amelioraung, treating and/or pre enung neural Ussue or cell damage, including that following focal ischemia and reperfusion injury Generally, inhibiuon of PARP activity spares the cell from energy loss, preventing l ⁇ eversible depola ⁇ zation of the neurons and.
  • the present invenuon provides compunds which inhibit poly (ADP- ⁇ bose) polymerase (“PARP”), compositions containing these compounds and methods for using these PARP inhibitors to treat, prevent and/or ameliorate die effects of the condiuons desc ⁇ bed herein
  • PARP poly (ADP- ⁇ bose) polymerase
  • the present invenuon provides compounds of Formula I
  • X is NR n . -0-. -S-. CRi;R
  • 3 . a bond. -CR, : CR, 3 -. or -C(R 1 2Ri3)C(R 14 Ri5)-.
  • W is selected from -CN. -C(R 9 );, -(N(R 9 ) 2 ) where the R 9 subsutuents may be combined to form a heteroaryl or C 3 -C 8 cycloalkyl opuonally containing at least one hetero atom m place of a carbon atom.
  • (C ⁇ -C 9 straight or branched chain) alkyl (C ⁇ -C 9 straight or branched chain) alkyl.
  • C 3 -C cycloalk l opuonally containing at least one heteroatom in place of a carbon atom, lower (C;-C 9 straight or branched chain) alkenyl, C ⁇ -C- cycloalkenyl. lower (C 1 -C ) alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroan I. hydroxy. amino, nitro, halo, nitroso, sulfo. sulfonic acid, or carboxv.
  • Rg is hydrogen, lower (C Cg straight or branched chain) alkv l.
  • C 3 -C 3 cycloalkyl optionallv containing at least one heteroatom in place of a carbon atom lower (C : -C , straight or branched chain) alkenyl. aryl. heteroaryl. aralky l heteroaralkyl. alkaryl. alkheteroan 1. hydroxy. lower (C1 -C4) alcohol. lower (C -Cj) alkoxy. ammo, or carboxy . and R, nickname and R, , are independently hydrogen, lo er (C -C y straight or branched chain) alkyl. lower (C : -C 9 straight or branched chain) alkeny l. aryl. aralkyl. alkaryl.
  • is hydrogen or lower (C,-Co straight or branched chain) alkyl.
  • T. when present, is a divalent or t ⁇ valent organic residue independently selected from the group consisting of lower (C : -C, straight or branched chain) alk lene. lower alkenylene. arylene. aralkylene. and alkarylene. wherein one. two or three of the hydrogen atoms of said divalent or t ⁇ valent organic residue can be substituted bv a moiety selected from the group consisung of lower (C] -C 9 straight or branched chain) alkyl. cycloalkyl.
  • benzy loxy. -0-. >C 0. -S-. -SO;-. -NR, SO : -. -SO : NR,-. -NR,-. and -P0 : -, wherein any of the lower (C
  • Ci-Cx cycloalkyl opuonally containing at least one heteroatom in place of a carbon atom, lower (C : -C straight or branched chain) alkenyl. cycloalkenyl. arvl. heteroaryl. aralkyl. heteroaralky l. alkaryl. alkheteroaryl. halo, t ⁇ fluoromethyl. hydroxy. lower (C ⁇ -C 4 ) alkoxy. carboxy (such as methoxy or ethoxy). carbonyl.
  • the present invenuon provides compounds of Formula (II) or a pharmaceutically acceptable salt, hydrate, prodrug. or mixtures thereof, wherein, q is zero or one. Z is 0.
  • X is -0-. or a bond.
  • Ri . R : . R 3 . Rj. R ⁇ . R e . and R- . when present, are independently hydrogen, lower (C ⁇ -C 9 straight or branched chain) alkyl. lower (C;-C straight or branched chain) alkenyl. C 3 -C 8 cy oalkyl optionally containing at least one heteroatom in place of a carbon atom. C ⁇ -C- cycloalkenyl. lower (C,-C; ) alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroarly. hydroxy. ammo, nitro. halo, nitroso. or carboxy.
  • C 3 -C 3 cycloalky 1 optionally containing at least one heteroatom in place of a carbon atom, lower (C : - straight or branched chain) alkenyl. ary heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroar l. hydroxy. lower (C,-C ⁇ ) alcohol, lower (Ci-C,) alkoxy. amino. or carboxy. and T. when present, is a divalent or tnvalent organic radical independently selected from the group consisting of. lower alkylene. lower alken lene. C ; -C_ alkeny lox . aryiene.
  • alkheteroaryl groups can be independently substituted with one. two or three subsutuents selected from the group consisting of. lower (Ci-C ⁇ straight or branched chain) alkyl. C 3 -C « cycolalkyl optionally containing at least one heteratom. lower (C;-C 9 straight or branched chain) alkenyl. aryl. heteroar l. aralkyl. heteroaralkyl. alkaryl. alkheteroaryl, halo, t ⁇ fluoromethyl. hydroxy . lower (C,-C, ) alkoxy. carboxy (such as methoxy or ethoxy). carbonyl. lower alkyl ester (such as methylester or etliylester). amino. mtro. t ⁇ fluoromethyl. alkenyloxy. phenoxy. benzyloxy,
  • alk is lower alkylene
  • Ris and R, 9 are independently hydrogen or lower alkyl. or R,- and R, g or R, g and R, 9 taken together can be a lower alkylene to form a heterocyclic ⁇ ng; and
  • Ri. R;, R 3 . R,. R . R « and R- are independently h drogen, lower (C,-C 9 straight or branched chain) alkyl.
  • the present invenuon provides compounds of Formulas (IV) and (V)
  • R, -R are as defined above
  • Preferablv R -R * are each independently a of hvdrogen. methyl, ethyl, propyl isopropy l. butyl isobuty l h drow l. amino. niuo. nitroso. carboxy . t ⁇ fluoromethy l. phenoxy and benzy oxy
  • Prefe ⁇ ed embodiments of the present invenuon include compounds wherein X and Z are oxygen and Y is nitrogen
  • the prefe ⁇ ed forms of the following specific embodiments include, but are not limited to. compounds wherein X and Z are oxygen and Y is nitrogen
  • Preferred embodiments of the present invenuon include compounds wherein each of W is -CN. m and n are zero and p is one Of these, further prefe ⁇ ed embodiments include compounds where T is -CH;-. Z and X are oxygen. Y is N and R, to R- are hvdrogen Preferably each of T is -CH;-. Z and X are oxygen and Y is N
  • prefe ⁇ ed embodiments of the present invenuon include compounds where each of W is - CN(R 9 ) 2 ). preferably W is -N(R 9 ) 2 m and n are zero and p is one X is preferably oxygen or a bond. Z is oxygen and Y is nitrogen Of these, prefe ⁇ ed embodiments include those compounds where each R, is independently selected from hydrogen, methyl, ethyl, prop l. isopropyl. butyl, isobutyl. pentyl. lsopenryl. methoxy.
  • Prefe ⁇ ed forms of this embodiment include compounds where T is absent. -CH 2 - or - CH 2 -CH 2 - CH;S. CH : SCH 2 . CH;SCH;CH 2 . methoxy or phenoxy methyl
  • Particularly prefe ⁇ ed forms of this embodiment include compounds wherein R 0 subsutuents combine, with die N of W. to form a
  • each of W is -N(R 9 ) 2 .
  • m is one and n is one.
  • X is o.xy gen or a bond.
  • Z is oxygen and Y is mtrogen
  • prefered embodiments include those compounde where each R 9 is independently selected from hydrogen, methyl, ethyl, propyl. isopropyl. butyl, isobutyl. mehanol. ethanol. methox . ethoxy or amino or w here the R 9 substituents combine with the N to form a 5- or 6- membered substituted or unsubstituted heterocy cloalky 1.
  • Prefe ⁇ ed forms of this embodiment include compounds where T is absent.
  • -CH 2 - or -CH 2 -CH : - Particularly prefe ⁇ ed forms of this embodiment includes compounds w herein R Q substituents form with the N of W to form a
  • benz l or phenyl and or fused widi 1-2 additional benzene ⁇ ngs R, -R- are preferably hydrogen
  • Other prefe ⁇ ed embodiments of die present inv enuon includes compounds wherein W is eidier - P(0) 2 -OR 9 or -P(0)(OR 9 ) 2 , m is zero and p is one Of these, each R 9 is preferably independendy hydrogen, methyl, ethyl, propyl. isopropyl. butyl or isobutyl Preferably, in these embodiments. Y is nitrogen. Z is oxygen and X is oxygen Preferably, tn is zero and when n is one, R, 6 is preferably hydrogen T is preferably -CH 2 - or - CH 2 CH;- in these prefe ⁇ ed embodiments
  • prefe ⁇ ed embodiments of the present invenuon include compounds wherein W is either - S(0) 2 -R 9 , -S(0) 2 -OR 9 or -S(0) 2 NR 9 , m is zero and p is 1 Of these, prefe ⁇ ed embodiments include where R 9 is hydrogen, or subsututed or unsubstituted meth l, ethyl, propyl. isopropyl. butyl or isobutyl. wherein die optional subsutution is a 5- or 6- membered cycloalk l.
  • X oxygen or a bond.
  • Z is oxygen.
  • T is absent or -CH 2 -.
  • R,-R- are h drogen.
  • Y is nitrogen and R is hydrogen. methyl, ethyl, propyl. isopropyl. but l, isobutyl. benz l, benzylcarbonyl. phenyl.
  • R 9 is hydrogen, methyl, ethyl, propyl. isopropyl. butyl, isobutyl. benzyl, or benzy carbony 1
  • W is an optionally subsututed 5- or 6- membered c cloalky l opuonally containing at least one heteroatom selected from S. O or N. or a heteroaryl wherein the cycloalkyl or heteranl ma ⁇ be subsututed or attached to a further 5- or 6- membered cycloalkyl which may optionallv contain at least one heteroatom.
  • the compounds of the inv ention exhibit an IC 50 for inhibiting PARP in vitro, as measured by the methods desc ⁇ bed herein, of about 20 ⁇ M or less preferablv less than about lO ⁇ M. more preferably less than about 1 ⁇ M. most preferably less than about 0 1 ⁇ M
  • Prefe ⁇ ed embodiments of the present in ention include the following compounds, and neutral forms thereof, where approp ⁇ ate
  • the compounds and composiuons of the present invenuon can be used to treat or prevent cell damage or death due to necrosis or apoptosis.
  • cerebral ischemia and reperfusion injury or neurodegenerative diseases in an animal, such as a human The compounds and compositions of the present inv enuon can be used to extend the hfespan and proliferauve capacitv of cells and thus can be used to treat or prevent diseases associated therewith, they alter gene expression of senescent cells, and thev radiosensitize hvpoxic tumor cells Preferabl .
  • die compounds and compositons of the inv ention can be used to treat or prevent tissue damage resulung from cell damage or death due to necrosis or apoptosis. and/or effect neuronal activ ity either mediated or not mediated by NMDA toxicity
  • the compounds of the present invention are not limited to being useful in treaung glutamate mediated neurotoxicity and/or NO-mediated biological pathways Further, the compounds of the invention can be used to treat or pre ent other tissue damage related to PARP activation, as desc ⁇ bed herein
  • the present invenuon provides compounds which inhibit the in vitro and/or m vivo polymerase activity of poly (ADP- ⁇ bose) polymerase (PARP). and compositions containing the disclosed compounds
  • the present invention provides methods to inhibit, limit and or control the in vitro and/or in vivo polymerase activitv of poly (ADP- ⁇ bose) polymerase (P.ARP) in any of solutions, cells, tissues, organs or organ systems
  • P.ARP poly (ADP- ⁇ bose) polymerase
  • the present mv ention provides methods of limiting or inhibiting PARP activity in a mammal, such as a human, either locally or systemically
  • the present invenuon provides methods to treat and/or prevent diseases, syndromes and/or condiuons exacerbated by or involving die increased generation of PARP These methods involve application or admimstration of the compounds of the present invenuon to cells, tissues, organs or organ systems of a person in need of such treatment or prevention
  • the present invention provides mediods to treat and/or prevent cardiovascular tissue damage resulting from cardiac ischemia or reperfusion injury
  • Reperfusion injury for instance, occurs at the termination of cardiac bypass procedures or du ⁇ ng cardiac anest w hen the heart, once prevented from receiving blood, begins to reperfuse and these methods involve administration of the compounds and compositions of the present invenuon preferably p ⁇ or to. or immediately subsequent to reperfusion. such that reperfusion injury is prevented, treated or reduced
  • the present invention also provides methods of preventing and/or treating vascular stroke, cardiov ascular disorders
  • the present inv ention prov ides in vitro or in v ivo methods to extend or increase the hfespan and or proliferation capacitv of cells and thus also methods to treat and/or prevent diseases associated therewith and induced or exacerbated bv cellular senescence including skin aging, atherosclerosis, osteoartli ⁇ tis. osteoporosis, muscular d strophy degenerauve diseases of skeletal muscle involving rephcative senescence, age-related muscular degeneration, immune senescence.
  • the present invention provides methods of treating or preventing or ameliorating the effect of cancer and or to radiosensitize hvpoxic tumor cells to render the tumor cells more susceptible to radiation therapy and therebv to prevent the tumor cells from recove ⁇ ng from potentially lethal damage of DNA after radiation therapy
  • a method of this embodiment is directed to specificallv and preferential lv radiosensitizing tumor cells rendenng the tumor cells more susceptible to radiation therapy than non-tumor cells
  • the present in ention provides methods of preventing and/or treating vascular stroke, cardiovascular disorders, to treat other conditions and/or disorders such as age-related muscular degeneration.
  • AIDS and other immune senescence diseases arth ⁇ tis. atherosclerosis, cachexia. cancer, degenerauve diseases of skeletal muscle mv oh ing rephcativ e senescence, diabetes, head trauma, spinal chord injury , immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis.
  • pancreauc and/or skeletal muscles systems acute autoimmune thv roidius. muscular d strophy osteoa ⁇ h ⁇ tis. osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia septic shock (such as endotoxic shock), local and/or remote entothelial cell dysfunction (such are recognized bv endo-dependent relaxant responses and up-regulauon of adhesion molecules), inflammauon and skin aging
  • a person diagnosed with acute retinal ischemia or acute vascular stroke is immediately administered parenterally. either by intermittent or conunuous intravenous administration, a compound of any of formulas I. II. III. IV or V either as a single dose or a se ⁇ es of divided doses of the compound
  • die person optionally may receive the same or a different compound of the invention in the form of another parenteral dose.
  • the compound of die inv ention can be admnistered by intermittent or continuous administration via implantation of a biocompatible. biodegradable polyme ⁇ c mat ⁇ .x delivery system contaimng a compound of formula I. II. III. IV or V.
  • die present in enuon provides methods to extend the Iifespan and proliferauve capacity of cells, such as. for example, in using the compounds of the invenuon as general mediators in the generation of oxidants. proinflammatory mediators and/or cytokines. and/or general mediators of leukocyte infiltration, calcium ion ov erload, phospholipid pero.xidaion. impaired nit ⁇ c oxide metabolism and/or reduced ATP producuon
  • Figure 1 Shows the dist ⁇ buuon of the cross-secuonal infarct area at representauve lev els along the rostrocaudal axis, as measured from the interaural line in non-treated animals and in animals treated with 10 mg/kg of 3.4-d ⁇ hydro-5-l- (l-p ⁇ pe ⁇ d ⁇ nyl)-botoxyll -1 (2H)- ⁇ soqu ⁇ nol ⁇ none
  • Figure 2 Shows the effect of intrape ⁇ toneal administration of 3.4-d ⁇ hydro-5-l-( l -p ⁇ pe ⁇ d ⁇ ny l )-butoxy]- l(2H)- ⁇ soqu ⁇ nol ⁇ none on the infarct volume
  • the present invention pe ⁇ ains to compounds, pharmaceuucal compositions containing the same. methods of using the same, and process of making die same, wherein such compounds are useful as inhibitors of poly( ADP- ⁇ bose) polymerase (P.ARP) As such, thev treat or prevent neural ussue damage resulting from cell damage or death due to necrosis or apoptosis.
  • P.ARP poly( ADP- ⁇ bose) polymerase
  • die compounds of the invention treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis. and or effect neuronal activity, either mediated or not mediated by NMDA toxicity These compounds are diought to interfere with more than the glutamate neurotoxicity and NO-mediated biological pathways Further, the compounds of the invention can treat or prevent other tissue damage related to P.ARP activ auon
  • the compounds of die invenuon can treat or prevent cardiovascular tissue damage resulting from cardiac ischemia or reperfusion injury
  • Reperfusion injury for instance, occurs at the terminauo ⁇ of cardiac bypass procedures or du ⁇ ng cardiac anest w hen the heart, once prevented from receiving blood, begins to reperfuse.
  • the compounds of the present invention can also be used to extend or increase the Iifespan or proliferation of cells and thus to treat or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis. osteoarth ⁇ Us. osteoporosis, muscular d strophy, degenerative diseases of skeletal muscle inv olving replicauve senescence, age-related muscular degeneration, immune senescence.
  • AIDS and odier immune senescence diseases, and other diseases associated with cellular senescence and aging, as well as to alter the gene expression of senescent cells can also be used to treat cancer and to radiosensitize hvpoxic tumor cells to render die tumor cells more susceptible to radiation therapy and to prevent the tumor cells from recove ⁇ ng from potentially lethal damage of DNA after radiation dierapv presumably by their ability to prevent DNA repair
  • the compounds of the present invention can be used 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.
  • cancer degenerative diseases of skeletal muscle involving replicative senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as coliUs and Crohn's disease), muscular dystrophy, osteoarth ⁇ us. osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging
  • the compounds of the inv ention act as PARP inhibitors to treat or prevent tissue damage resulting from cell death or damage due to necrosis or apoptosis to treat or prevent neural tissue damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in an animal, to extend and increase the Iifespan and proliferauve capacity of cells, to alter gene expression of senescent cells, and to radiosensitize tumor cells
  • Another especiallv prefe ⁇ ed embodiment of the invention is a pharmaceutical composition which comp ⁇ ses d ) a therapeuticallv effective amount of the compound of formula I. II or III. and (n) a pharmaceuticallv acceptable earner
  • alk l means a branched or unbranched saturated hvdrocarbon chain compnsing a designated number of carbon atoms
  • C ⁇ -C 6 straight or branched alkyl hydrocarbon chain contains 1 to 6 carbon atoms, and includes but is not limited to substituents such as methyl, ethyl, propyl. iso-propyl. butyl, iso-butyl. tert-butyl. n-penty l. n-hexyl. and the like, unless othenvise indicated
  • alkenyl means a branched or unbranched unsaturated hydrocarbon chain compnsing a designated number of carbon atoms
  • C z -C 6 straight or branched alkenyl hydrocarbon chain contains 2 to 6 carbon atoms having at least one double bond, and includes but is not limited to substituents such as ethenyl. propenyl. isopropenyl. butenvl. iso-buten l. te ⁇ -butenv l. n-pentenyl. n-hexenyl. and the like, unless otherwise indicated
  • Alkox means the group -OR wherein R is alkyl as herein defined Preferably. R is a branched or unbranched saturated hydrocarbon chain contaimng 1 to 6 carbon atoms
  • Cyclo used herein as a prefix, refers to a structure characterized by a closed ⁇ ng "Halo” means at least one fluoro. chloro. bromo, or lodo moiety, unless otherwise indicated.
  • Amme amme (NH 2 ) as well as substituted ammo groups comp ⁇ sing alkyls of one through six carbons
  • aryl or “heteroaryl” means a moiety which is substituted or unsubstituted. especially a cyclic or fused cyclic ⁇ ng and includes a mono- bi-. or t ⁇ cychc. carbo- or heterocyclic ⁇ ng. wherein die ⁇ ng is either unsubsututed or substituted in one to five pos ⁇ uon(s) with halo, haloalkyl. hydroxyl. nitro. t ⁇ fluoromethy 1.
  • C 2 -C « straight or branched chain alkenyl.
  • ⁇ ng sizes are preferably 5-8 members, wherein the heterocyclic ⁇ ng contains 1-4 heteroatom(s) selected from the group consisting of O. N. or S. wherein aromatic or tertiary alkyl amines are optionally oxidized to a conesponding N-o ⁇ de Heteroaryls may be attached to other ⁇ ngs or subsututed through die heteroatom and/or carbon atom of the ⁇ ng Pa ⁇ icularl prefe ⁇ ed aryl or heteroaryl moieties include but are not limited to phen l. benz l naphthyl. pynolyl.
  • Pheny l includes all possible isome ⁇ c phen l radicals, optionally monosubstituted or multi-substituted with subsutuents selected from the group consisung of ammo t ⁇ fluoromethyl.
  • Ci-C* straight or branched chain alkyl.
  • G-C ⁇ straight or branched chain alkenvl. carbonyl. thiocarbonyl. ester, thioester alkoxy. alkenoxy. cyano. nitro. lmino. alkyiamino aminoalkyl. sulfhydryl thioalkyl. sulfonyl. hydroxy. halo, haloalkyl.
  • R 2 is selected from the group consisting of hydrogen.
  • (C ⁇ -C ) b ⁇ dging alk l wherein said b ⁇ dging alkyl forms a heterocyclic ⁇ ng starting with the nitrogen of NR, and ending with one of the carbon atoms of said alkyl or alkenv l chain, and wherein said heterocyciic nng is optionalh fused to an AT group Cycloalky l optionalh containing at least one heteroatom includes saturated C 3 -C 8 ⁇ ngs.
  • cycloalkvls containing a heteroatom include pynolidinvl. lmidazolidmyl. pyrazohdin l. pipe ⁇ dinvl. piperazinyl.
  • the compounds of the present invention possess one or more asymmet ⁇ c center(s) and thus can be produced as mixtures (racemic and non-racemic) of stereoisomers. or as individual enantiomers or diastereo ers
  • the individual stereoisomers may be obtained by using an optically acuve starting mate ⁇ al. by resolving a racemic or non-racemic mixture of an intermediate at some approp ⁇ ate stage of the svnthesis. or by resolution of the compound of anv of formulas I.
  • the compounds of the invention are useful in a free base form, m the form of pharmaceutically acceptable salts, pha ⁇ naceuticall acceptable h drates, pharmaceuucally acceptable esters, pharmaceutically acceptable solvates. pharmaceutically acceptable prodrugs. pharmaceutically acceptable metabolites and in the form of pharmaceutically acceptable stereoisomers These forms are all within the scope of the invention In practice the use of these forms amounts to use of the neutral compound
  • “Pharmaceutically acceptable salt”, " hydrate ' . "ester “ or “solvate” refers to a salt, hydrate, ester, or solvate of the inventive compounds which possesses the desired pharmacological activ ity and which is neither biologicallv nor othenvise undesirable Orgamc acids can be used to produce salts, hydrates, esters, or solvates such as acetate, adipate. alginate. aspartate. benzoate. benzenesulfonate. p-toluenesulfo ⁇ ate. bisulfate. sulfamate, sulfate naphth late butv rate, citrate camphorate.
  • camphorsulfonate cyclopentane-propionate digluconate. dodecy Isulfate ethanesulfonate. fumarate. glucoheptanoate. glvccrophosphate. hemisulfate heptanoate. hexanoate. 2-hydroxy ethanesulfonate. lactate. maleate. methanesulfonate. 2-naphthalenesulfo ⁇ ate. mcotinate. oxalate tosvlate and undecanoate Inorganic acids can be used to produce salts, hydrates, esters, or solvates such as hydrochlo ⁇ de. hydrobromide. hvdroiodide. and thiocyanate
  • suitable base salts, hvdrates. esters, or solvates include hydroxides, carbonates, and bicarbonates of ammonia, alkali metal salts such as sodium, lithium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts aluminum salts, and zinc salts Salts hvdrates. esters or solvates mav also be formed with organic bases
  • Organic bases suitable for the formation of pharmaceuticalh acceptable base addition salts, h drates esters, or solvates of the compounds of the present invention include those that are non-toxic and strong enough to form such salts, hydrates, esters, or solvates
  • the class of such organic bases mav include mono- di-. and t ⁇ alkylamines.
  • Accordingl . basic nitrogen-containing groups can be quaternized with agents including lower alk l halides such as meth l ethyl, prop l. and butyl chlo ⁇ des. bromides and iodides, dialkyl sulfates such as dimethyl, diedi l. dibutyl and diamyl sulfates. long chain halides such as dec l. lauryl. my ⁇ styl and stea l chlo ⁇ des. bromides and iodides, and aralkyl halides such as benzyl and phenethyl bromides
  • the acid addition salts, hydrates, esters, or solvates of the basic compounds may be prepared either by dissolving the free base of a PARP inhibitor of the present in ention in an aqueous or an aqueous alcohol solution or other suitable solvent containing the approp ⁇ ate acid or base, and isolaung the salt by evaporating the solution
  • the free base of the PARP inhibitor of the present invention can be reacted with an acid, as well as reacting die PARP inhibitor having an acid group thereon ith a base, such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained bv concentrating the solution
  • “Pharmaceuticalh acceptable prodnig” refers to a de ⁇ vative of the inventive compounds which undergoes biotransformauon p ⁇ or to exhibiting its pharmacological effect(s) The prodnig is fo ⁇ nulated with the object ⁇ ve(s) of improved chemical stability , improved pauent acceptance and compliance improved bioavailabihtv prolonged
  • the prodnig can be readily prepared from the invenuve compounds using methods known in die art. such as those desc ⁇ bed by Burger's Medicinal Chemistry and Drug Chemistry. Fifth Ed . Vol 1. pp 172-178. 949-982 ( 1995)
  • the invenuve compounds can be transformed into prodrugs by convening one or more of the hydroxy or carboxy groups into esters ' ⁇
  • Pharmaceutically acceptable metabolite refers to drugs that have undergone a metabolic transformation After entry into the bod ⁇ .
  • ne ous insult refers to any damage to nenous tissue and am disability or death resulting therefrom
  • the cause of nen'ous insult may be metabolic, to ic, neurotoxic. latrogenic. thermal or chemical, and includes without limitation, ischemia, h poxia. cerebrovascular accident, trauma, surgery, pressure, mass effect. hemmonhage. radiation vasospasm. neurodegenerauve disease, infection. Parkinson's disease, amyotrophic lateral sclerosis (ALS). m ehnation/demvelination process, epilepsy, cognitive disorder, glutamate abnormality and secondary effects thereof
  • neuroprotective refers to the effect of reducing, a ⁇ estmg or amelioraung nenous insult, and protecung. resuscitating, or reviving ne ous tissue that has suffered nen'ous insult
  • prev enting neurodegenunteron includes the ability to prevent neurodegenrungon in patients diagnosed as having a neurodegenerativ e disease or who are at nsk of developing a neurodegenerativ e disease The term also encompasses preventing further neurodegenrungon in patients who are already suffe ⁇ ng from or have symptoms of a neurodegen Vogelve disease
  • treating refers to (1) preventing a disease, disorder or condiuon from occumng in an animal that may be predisposed to the disease, disorder and/or condition, but has not yet been diagnosed as having it. (n) inhibiting the disease, disorder or condition. 1 e , anesting its development, and
  • neural tissue damage resulting from ischemia and reperfusion injury and neurodegen fieve diseases includes neurotoxicity.
  • phenaceun acetophenetidin
  • acetanihde are both mild analgesic and antipyretic agents but are each transformed within the body to a more polar and more effective metabolite, p- hydroxyacetani d (acetaminophen), which is widely used today
  • p- hydroxyacetani d acetaminophen
  • acetanilid is the p ⁇ ncipal plasma component
  • the metabolite acetaminophen concentrauon reaches a peak Finall .
  • the p ⁇ ncipal plasma component is a further metabolite that is inert and can be excreted from the body
  • die plasma concentrations of one or more metabolites, as well as the drug itself, can be pharmacologicalh important
  • Phase I or funcuonahzauon reactions generalh consist of ( 1 ) o idative and reducuve reactions that alter and create new functional groups and (2) hydrohlic reactions that cleave esters and amides to release masked functional groups These changes are usuallv in the direction of increased pola ⁇ ty
  • Phase II reactions are conjugation reacuons in which the drug, or often a metabolite of the drug, is coupled to an endogenous substrate, such as glucuromc acid, acetic acid, or sulfu ⁇ c acid
  • Oxidative deaminauon (monoamine oxidase and diamine oxidase)
  • the compounds of the present in enuon exhibit pharmacological activity and are. therefore, useful as pharmaceuucals
  • the compounds exhibit central nenous and cardiac vesicular system acuvity
  • tautome ⁇ c forms when possible, are included m the invenuon
  • the tautome ⁇ c forms of the following compounds are exemplary
  • the PARP inhibitors used in the composiuon of the invenuon will have an IC 50 for inhibiung poly( ADP- ⁇ bose) polymerase in vitro of about 20 ⁇ M or less, preferably less than about lO ⁇ M. more preferably less than about 1 ⁇ M, most preferably less than about 0 1 ⁇ M
  • the PARP inhibitor 3.4-d ⁇ hydro-5-[4-(l-p ⁇ pe ⁇ d ⁇ nyl)butoxy]-l(2H)- ⁇ soqu ⁇ nolmone has been reported to inhibit PARP with an IC 50 of 40 nM by Suto et al . cited above
  • the xanthene ⁇ ng may be gene ⁇ cally subsututed as set forth in formula I Such xanthene starting denvauves are known in die chemistry literary and are accessible by processes known to one skilled in the art The process sequence set forth herein does not present an exact sequence of reacuons by which the compound must be made, that is. die sequence of reacuons can be rearranged in several ways to reach the target molecule
  • the .xanthene and thioxanthene denvauves of this invenuon are represented by previously defined formulas I-V
  • the xandiene denvauves of this invenuon can be prepared in a conventional manner as illustrated below by Schemes 1 - 5
  • the xanthene ⁇ ng may be gene ⁇ cally subsututed as set forth in formula I
  • Such xanthene starting denvauves are known in the chemistn literature and accessible by processes known to one skilled in the an
  • the process sequence set forth herein does not present an exact sequence of reacuons by w hich the compound must be made, that is. the sequence of reacuons can be reananged in several ways to reach the target molecule
  • Scheme 1 below illustrates schematically the preparauon of compounds Example 1 through Examples 4
  • 9-Am ⁇ nomehty lxanthene 2 is available by reduction of 9-carboxam ⁇ de 1 using sodium boronhvdnde in dioxane
  • Other reducuon methods can be subsututed include lithium aluminum hydnde and other boronhydndes
  • the solvent can also be va ⁇ ed.
  • DMSO. tetrahy drofuran. dieth lether and other organic solvent can be used The temperature of the reaction is general between 0 C and 200 C
  • 9- a ⁇ unomethy lxanthene can be made as follows To a suned suspension of sodium boronhvdnde (1 89g.
  • the 9- ⁇ socyanoiolovlxanthenes 3 is obtained by condensauon of the amino group idi phosgene in a heated solution of toluene Odier solvents, such as p-nitrobenzene. can also be used
  • Odier solvents such as p-nitrobenzene.
  • Other fiincuonaliues include N-carbonv li ⁇ udazole.
  • N-carbony Ibenzot ⁇ azole and N-ethylfbrmate can also be applied this type of reactions
  • These functionalities can be formed by reacuons of the 9-am ⁇ nomethylxanthenes 2 with carbonyldiimidazole. carbonyldibenzotnazole and ethyl chloroformate respecuvely Step ?
  • the desired xanthane product. 3.4-d ⁇ hvdrobenzopyrano[4.3.2-de] ⁇ soqu ⁇ noh-l-one 4 can be obtained by an intramolecular F ⁇ edle-Crafts acylation using acid as catal st Zinc chlo ⁇ de. aluminum chlo ⁇ de. titanium (IV) chlo ⁇ de. hydrochlo ⁇ c acid or aceuc acid may be used, but polyphospho ⁇ c acid is often prefened for tlus type of intramolecular cycloadditions
  • the temperature can vary from 0 - 150 °C For example.
  • 3,4- dihydrobenzop rano[4.3.2-de] ⁇ soqu ⁇ nol ⁇ n-l-one 4 can be prepared as follows To a liquid of 12 g of polyphospho ⁇ c acid in a 500 mL beaker is added 9-xanthenemethyl ⁇ socyan ⁇ de 3 (2 37 g.
  • Monosulfonation of 3.4-d ⁇ hydrob nzopyrano[4.3.2-de] ⁇ soqu ⁇ nol ⁇ -l-one 4 can be achieved using chorosulfonic acid neat to give compound 5
  • the temperature can vary from -20 to 100 °C
  • Amidation of compound 5 can be earned out bv a v a ⁇ ety of conditions known to those skilled in the art.
  • 3-phenoxyphthalon ⁇ t ⁇ le 7 The starting mate ⁇ als 3-n ⁇ trophthalon ⁇ u ⁇ le 7 and or p- subsututed pheno ide 8 are either readily available from commercial sources or can be prepared by known methods by those skilled in the art
  • the desired compound can be easily obtained by coupling the orthodinit ⁇ le with the phenoxide salt in dipolar aprotic solvent, such as DMF. low dialkyl ketones. lower alkyl nit ⁇ les
  • Temperature range can be 0 - 150 C
  • a mixture of 0 1 Mol of 3-n ⁇ trophd ⁇ alomt ⁇ le and 0 14 Mol of sodium phenoxide (or p-substituted phenoxide) in 120 ml of dry DMF is mechanicalh suned for 2 5 hours at room temperature and then poured into water The precipitate is collected by filtrauon. washed with water, and d ⁇ ed under vacuum to give 19 g (0 086 Mol. 86%) of die compound 9.
  • Preparauon of 9-oxoxanthene-l -carboxylic acid 10 The formauon of orthocarboxy lie acid groups from the cyano groups can be achieved by hydrolysis of the aryl nitnle with either mineral acids, include sulfu ⁇ c acids and h drochlo ⁇ c acids, or strong inorganic base These conditions are understood by those skilled in the art.
  • An intramolecular F ⁇ edle-Crafts acylation can occur as soon as die diacids are formed to afford 9-oxoxanthene-I- carboxylic acid 10
  • a soluuon of 0 1 Mol of 3-phe ⁇ oxyphfhalon ⁇ t ⁇ le is dissolved in 150 ml of a mixture of 60 per cent sulfunc acid and glacial acid The mixture is refluxed for 6 hours and then poured into 1000 mL of ice-cold water pH value of the suspension is adjusted to 10 with sodium carbonate Impu ⁇ ties are extracted out by paruuoning of the basified soluuon idi ethyl acetate Precipitauon is formed upon adding hydrogen chlo ⁇ de (to pH5) to the aqueous soluuon. and collected b filtrauon to give a designed acid in 70% yield
  • Estenficauon of acid can be achieved by several convenuonal methods by those skilled in the art These procedures include uulizmg diazomethane or methy 1 alcohol catalyzed by mineral acids
  • ethereal diazomethane soluuon can be prepared from N-methyl-N-nitrosotolue ⁇ e-p-sulphonamide and potassium hydroxide from a water bath 9-Oxoxanthene-l -carboxylic acid (1 29g, 5 mmol) in absolute methanol is added to this ethereal diazomethane soluuon at 0 °C sol a pale yellow color formed Removal of the solvent provides the desired ester solid in 100% yield Thus the final products.
  • Examples 5 - 9, can be prepared by condensauon hydrazine with the ketonester 11 as desc ⁇ bed in General procedure C
  • BrominaUng agents include N-bromosuccinimide. bromine, complexed bromine such as pyndinium bromide, perbromide. and the like can be used to convert 7-methyl-9-oxoxanthene-l-carbox hc acid methyl ester 11 to 7-bromomethy 1-9- oxoxanthene- 1 -carbox he acid meth l ester 14 Solvents include chlo ⁇ nated hydrocarbones.
  • Monosulfonauon of the ketone ester 11 to gi e compound 16 can be achieved using neat chorosulfonic acid to react with the substrate 11
  • the temperature can vary from -20 to 100 °C
  • Amidation of compound 16 can be earned out by a variety of condiuons known to those skilled in the art. including reacuon with first or secondan amines using py ⁇ dine or tnedivl amine as base
  • Typical solvents include chlo ⁇ nated solvents, va ⁇ ous ethers, and dipolar aprotic soiv ents like DMF
  • final product of compounds can be prepared from compound 17 and hydrazine as desc ⁇ bed in General procedure C
  • a benzopyrano[4 3 2-de]phthalaz ⁇ ne ⁇ ng can be formed by condensation of the ketone ester with hydrazine
  • To a solution of the ketonester 11 or 15 ( 17 5 mmol) in absolute ethanol ( 10 mL) is added anhvdrous hvdrazine tn ethanol ( 1 mL) drop wise at room temperature
  • the soluuon is refluxed for overnight and cooled to room temperature Ice-cold water (100 mL) is added and brown solid is separated
  • the solid is collected by vacuum filtration and washed with water small amount of methanol to give a solid product in 10 - 80% of v ⁇ eld
  • the compounds of formula I may be useful in the free base form, in the fo ⁇ n of base salts where possible, and in the form of addiUon salts as ell as in the free acid form All these forms are within die scope of this invention
  • Pharmaceuticalh acceptable salts within die scope of this invention are those de ⁇ ved from mineral acids such as hydrochlo ⁇ c acid and sulfu ⁇ c acid, and organic acids such as ethanesulfomc acid, benzenesulfonic acid, p-toluenesulfomc acid, and the like, giv ing the hydrochlo ⁇ de. sulfonate. ethanesulfonate.
  • bases such as suitable organic and inorganic bases
  • pharmaceuucally acceptable base addiuon salts with compounds of the present mvenUon include orgamc bases which are nontoxic and strong enough to form such salts
  • organic bases and the use thereof are readily understood by those skilled in the art Merely for the purpose of lllustrauon.
  • such organic bases may include mono-, di- and t ⁇ alkyla ⁇ unes. such as methylamine. diethylamine and uiethylamine. mono-, di-. or tnhydroxyalk lamines such as mono-, di-.
  • amino acids such as arginine, and lysine, guanidine N-methylglucosamine. N-methylgiucamine. L-glutamine. N-methylpiperazine. morphohne, ethylenedianane N-benzylphenethylamine, tns(hydroxymethvl)anunoethane. and the like
  • the acid addition salts of the basic compounds may he prepared by dissolving the free base of the compound of formula I in aqueous or aqueous alcohol soluuon or other suitable solvents containing the approp ⁇ ate acid or base and isolaung the salt by evaporaung the solution, or by reacting the free base of the compound of formula I with an acid as well as reacung the compound of formula I having an acid group thereon with a base such that the reactions are in an orgamc solvent, m which case the salt separates direcdy or can be obtained by concentration of die soluuon
  • the invention includes the individual stereoisomers and mixtures thereof as well as the racemic compounds
  • the individual isomers may be prepared or isolated by methods known in the art
  • the compounds of the invenuon exhibit pharmacological activity and are. therefore, useful as pharmaceuticals
  • the compounds exhibit central nervous and cardiac vesicular system activity
  • Punficauon of compound by silica gel column chromatography provides sulfo ⁇ amide 17 Condensauon of compound 17 with hydrazine according to General Procedure C in ethanol affords a solid which is purified by crystalhzauon in ethanol gives a white solid product in 70 % of yield
  • To this amine compound (10 mmol) in dioxane (10 mL) is added 10 mmol of hydrochlo ⁇ c acid in dioxane at 60 °C
  • White salt solid product is formed upon cooling the solution to room temperamre The solid is collected by filtrauon.
  • va ⁇ ations and modificauons of this invention using the synthetic pathwa s desc ⁇ bed above will be obv lous to those skilled in the art
  • the compounds of the present inv ention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis.
  • the present inv ention further relates to a method of administe ⁇ ng a therapeuticallv effective amount of die above-identified compounds in an amount sufficient to inhibit PARP acuvit .
  • AIDS and other immune senescence diseases arth ⁇ tis. atherosclerosis, cache.xia. cancer, degenerative diseases of skeletal muscle involv ing replicative senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular d strophy, osteoarthnus.
  • 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 die Iifespan and proliferauv e capacitv of cells, to alter gene expression of senescent cells, or to radiosensitize hvpoxic tumor cells
  • the present inv enuon also relates to treaung diseases and condiuons in an animal which comp ⁇ ses administe ⁇ ng to said animal a therapeutically effective amount of the above-identified compounds
  • the present invenuon relates to a method of treating, preventing or lnlubiung a neurological disorder in an animal, which compnses administenng to said animal a therapeutically effecuve amount of die above-identified compounds
  • the neurological disorder is selected from the group consisting of pe ⁇ pheral neuropathy caused by physical injury or disease state, traumauc brain injury , physical damage to the spinal cord, s oke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demvehnating disease and neurological disorder relating to neurodegenerauon.
  • Another prefe ⁇ ed embodiment is when the reperfusion injury is a vascular stroke
  • Yet another prefened embodiment is when the pe ⁇ pheral neuropathy is caused by Guillain-Bane syndrome
  • Sull another preferred embodiment is when the demvehnating disease and neurological disorder relates to neurodegenerauon.
  • Another prefened embodiment is when the reperfusion injury is a vascular stroke
  • Sull anodier prefened embodiment is when the demvehnating disease is mulUple sclerosis .
  • Another prefened embodiment is when the neurological disorder relaung to neurodegen fieon is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease, and amyotrophic lateral sclerosis
  • Yet another prefened embodiment is a method of treating, preventing or inhibiting a cardiovascular disease in an animal, such as angina pecto ⁇ s. myocardial infarction, cardiovascular ischemia, and cardiovascular tissue damage related to PARP activation, bv administenng to said animal an effective amount of die compounds of die present invention
  • the present invention also contemplates the use of compound I. II. III. IV or V for inhibiting PARP acuvity . for treaung. preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis. for treating, preventing or inhibiung a neurological disorder in an animal
  • die neurological disorder is selected from the group consisung of pe ⁇ pheral 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, demyelinaung disease and neurological disorder relaung to neurodegenerauon.
  • Another prefe ⁇ ed embodiment is vv hen the reperfusion injury is a vascular stroke. Yet another prefened embodiment is when the pe ⁇ pheral neuropathv is caused by Guillam-Bane syndrome Still another prefe ⁇ ed embodiment is when the demyelinaung disease is multiple sclerosis Another prefened embodiment is when the neurological disorder relating to neurodegenerauon is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease, and am otrophic lateral sclerosis
  • the disease or disorder is a neurological disorder
  • the neurological disorder is selected from die group consisung of pe ⁇ pheral neuropathv caused b> phv sical injun or disease state, traumatic brain injury, physical damage to die spinal cord, stroke associated with brain damage, focal ischemia global ischemia, reperfusion injury, demyelinaung disease and neurological disorder relaung to neurodegenerauon
  • Another prefened embodiment is when the reperfusion injury is a vascular stroke
  • Yet another prefened embodiment is when the pe ⁇ pheral neuropathy is caused by Guillain-Bane syndrome
  • Still another prefened embodiment is when die demyelinaung disease is multiple sclerosis
  • Another prefened embodiment is when the neurological disorder relating to neurodegenunteron is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease and amyotrophic lateral sclerosis
  • the term "preventing neurodegenerauon” includes the ability to prevent neurodegenerauon in pauents ne ly diagnosed as having a neurodegenrungve disease, or at ⁇ sk of dev eloping a new degenerative disease and for prev enting further neurodegenerauon in pauents w ho are already suffe ⁇ ng from or have symptoms of a neurodegenerauve disease
  • treatment covers anv treatment of a disease and/or condiuon in an animal. particularlv a human, and includes
  • the term "neural tissue damage resulting from ischemia and reperfusion injury” includes neurotoxicity. such as seen in v ascular stroke and global and focal ischemia
  • the term "neurodegen Vogelve diseases.” includes .Alzheimer's disease. Parkinson's disease and Huntington's disease
  • ischemia relates to localized tissue anemia due to obstruction of the inflow of arte ⁇ al blood
  • Global ischemia occurs under conditions in w hich blood flow to the entire brain ceases for a pe ⁇ od of time, such as may result from cardiac anest
  • Focal ischemia occurs under conditions in which a portion of the brain is dep ⁇ ved of its normal blood supply, such as mav result from diromboemboiy ic occlusion of a cerebral vessel, traumauc head injury, edema, and brain tumors
  • cardiovascular disease relates to mvocardial infarcuon. angina pecto ⁇ s. vascular or myocardial ischemia, and related conditions as would be known by those of skill in the art which involve dysfuncuon of or tissue damage to the heart or v asculature. and especiall . but not limited to. Ussue damage related to PARP activation
  • the term '"radiosensitizer " ' as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeuticallv effectiv e amounts to increase die sensiuvity of the cells to be radiosensiuzed to electromagnetic radiauon and or to promote the treatment of diseases which are treatable with electromagnetic radiation Diseases which are ueatable with electromagnetic radiauon include neoplasUc diseases, benign and malignant tumors and cancerous cells Electromagnetic radiation treatment of other diseases not listed herein are also contemplated bv the present invention.
  • the terms " electromagnetic radiauon " and “"radiation” * as used herein includes, but is not limited to.
  • Prefened embodiments of die present inv enuon employ the electromagnetic radiauon of gamma- radiation ( 10 ':o to IO '13 m) x-ray radiauon ( 10 ' " to 10 ' ' m).
  • ultraviolet light 10 nm to 400 nm
  • visible light 400 nm to 700 nm
  • infrared radiation 700 nm to 1.0 mm
  • microwave radiation 1 mm to 30 cm).
  • die compounds of the invention inhibit PARP activity and. thus, are believed to be useful for treating neural Ussue damage, particularly damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in animals
  • nenous tissue refers to the va ⁇ ous components that make up the nen'ous system including, without limitation, neurons, neural support cells, glia. Schwann cells, vasculature contained within and supplying these structures, the central nen-ous system, die brain, the brain stem, the spinal cord, the junction of the central nenous system with the pe ⁇ pheral nen'ous system, the pe ⁇ pheral nen'ous system, and allied structures. Further, according to the invenuon.
  • an effecuve dierapeuuc amount of the compounds and compositions desc ⁇ bed above are administered to animals to effect a neuronal activity, particularly one that is not mediated by NMDA neurotoxicity.
  • Such neuronal acuvity may consist of sumulauon of damaged neurons, promouon of neuronal regeneration, prevention of neurodegenerauon and treatment of a neurological disorder.
  • the present invenuon further relates to a method of effecung a neuronal activity in an animal, comp ⁇ sing administenng an effective amount of the compound of formula I. II . III. IV or V to said animal.
  • the method of the present invenuon is particularly useful for treaung a neurological disorder selected from the group consisung of pe ⁇ pheral neuropathy caused by physical injury or disease state, head trauma, such as traumatic brain injury; physical damage to the spinal cord: suoke associated with brain damage, such as vascular stroke associated with hypoxia and brain damage, focal cerebral ischemia, global cerebral ischemia, and cerebral reperfusion injury; demyelinaung diseases, such as multiple sclerosis, and neurological disorders related to neurodegenerauon. such as .Alzheimer's Disease. Parkinson's Disease. Huntingdon's Disease and amyotrophic lateral sclerosis (ALS).
  • ALS amyotrophic lateral sclerosis
  • the compounds, composiuons and methods of the present invention are particularly useful for treating or prevenung tissue damage resulting from cell deadi or damage due to necrosis or apoptosis
  • cardiovascular disorders refers to those disorders that can either cause ischemia or are caused by reperfusion of the heart Examples include, but are not limited to coronary artery disease, angina pecto ⁇ s. myocardial infarction, cardiovascular tissue damage caused by cardiac anest. cardiovascular tissue damage caused bv cardiac bypass, cardiogenic shock, and related conditions that would be known by those of ordinary skill in the art or which involve dysfunction of or tissue damage to the heart or v asculature. especially , but not limited to. tissue damage related to P.ARP acuvation
  • the methods of die invention are believed to be useful for treating cardiac ussue damage, particularly damage resulting from cardiac ischemia or caused by reperfusion injury m animals
  • the methods of the invention are particularlv useful for ueating cardiovascular disorders selected from the group consisting of coronary arten disease such as atherosclerosis, angina pectons mvocardial infarction myocardial ischemia and cardiac anest. cardiac bvpass. and cardiogenic shock
  • the methods of the invention are particularly helpful in treating the acute forms of the above cardiovascular disorders
  • the methods of the inv ention can be used to treat tissue damage resulting from cell damage or death due to necrosis or apoptosis.
  • cancer degenerative diseases of skeletal mus ⁇ .e involv ing replicativ e senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular d strophv osteoarthnus 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 Iifespan and proliferauve capacity of cells to alter gene expression of senescent cells, or to radiosensitize tumor cells
  • the methods of the invenuon can be used to treat cancer and to radiosensitize tumor cells
  • cancer is interpreted broadly
  • the compounds of the present invention can be "anti-cancer agents", which term also encompasses “anu-tumor cell growth agents” and "anti-neoplastic agents”
  • the methods of the invenuon are useful for treaung cancers and radiosensiuzing tumor cells in cancers such as ACTH-producing tumors, acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma.
  • endometnal cancer esophageal cancer.
  • Ewing's sarcoma gallbladder cancer, hairy cell leukemia, head & neck cancer.
  • Hodgkm's lymphoma Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant pe ⁇ toneal effusion, malignant pleural effusion, melanoma, mesothelioma. multiple mveloma. neuroblastoma. non-Hodgkin's lymphoma ostcosarcoma. ova ⁇ an cancer, ovan (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, retinoblastoma.
  • Electromagnetic radiation treatment of other diseases not listed herein are also contemplated bv the present invention
  • electromagneuc radiation includes, but is not limited to.
  • Radiosensuizers are known to increase the sensitiv ity of cancerous cells to the toxic effects of electromagnetic radiation Sev eral mechanisms for the mode of action of radiosensuizers have been suggested in die literature including hvpoxic cell radiosensiuzers ( e g 2-n ⁇ tro ⁇ m ⁇ dazole compounds, and benzot ⁇ azine dioxide compounds) promote the reoxy Dockon of hvpoxic tissue and/or catalyze the generation of damaging oxygen radicals, non-hypoxic cell radiosensiuzers (e g .
  • halogenated p ⁇ midines can be analogs of DNA bases and preferential I v 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 va ⁇ ous other potential mechanisms of action have been hypothesized for radiosensiuzers in die treatment of disease
  • radiosensiuzers activ ated by the electromagnetic radiauon of x-rays
  • x-rav activated radiosensiuzers include, but are not limited to. the following metronidazole. misonidazole. desmethylmisorudazole. pimonidazole. etanidazole. ni orazole. mitomycin C. RSU 1069. SR 4233. E09. RB 6145. nicotinamide. 5-bromodeoxyu ⁇ d ⁇ ne (BUdR). 5- ⁇ ododeoxyu ⁇ dme (IUdR). bromodeoxycytidine. fluorodeoxyundine (FudR). hydroxyurea. cisplaun. and therapeuucally effective analogs and de ⁇ v atives of the same
  • Photodynamic therapy (PDT) of cancers emplov s visible light as the radiauon activator of the sensitizing agent
  • Examples of photodynamic radiosensiuzers include the following, but are not limited to hematoporphy ⁇ n denvauves. Photofnn. benzoporphy ⁇ n denvauves. NPe6. Un etioporphy ⁇ n SnET2. pheoborbide-a. bacte ⁇ ochlorophyll-a. naphthalocyanines. phthalocyamnes. zinc phdialocvanine. and therapeuucally effective analogs and denvauves of die same
  • Radiosensiuzers may be administered in conjunction with a therapeuticallv effective amount of one or more other compounds, including but not limited to compounds which promote the inco ⁇ oration of radiosensiuzers to the target cells, compounds which conuol the flow of therapeutics, nutnents. and/or oxygen to die target cells, chemotherapeutic agents which act on the tumor with or without addiuonal radiation, or other therapeutically effective compounds for treating cancer or other disease
  • additional therapeutic agents that may be used in conjunction widi radiosensiuzers include, but are not limited to 5-fluorourac ⁇ l, leucovo ⁇ n. 5' -am ⁇ no-5'deoxythym ⁇ d ⁇ ne. oxygen, carbogen.
  • chemotherapeutic agents that mav be used in conjunction with radiosensiuzers include, but are not limited to adnamycm. camptothecin. carboplatin. cisplaun. daunorubicin. docetaxel. doxorubicin. interferon (alpha, beta, gamma), interleukin 2. l ⁇ notecan. pachtaxel. topotecan. and therapeutically effective analogs and denvauves of the same
  • the present invention also relates to a pharmaceutical composition comp ⁇ sing (I) a therapeutically effective amount of the compound of formula I. II. III. IV or V and (n) a pharmaceutically acceptable earner
  • a pharmaceutical composition comp ⁇ sing I) a therapeutically effective amount of the compound of formula I. II. III. IV or V and (n) a pharmaceutically acceptable earner
  • pharmaceutically acceptable earner refers to any earner, diluent, excipient. suspending agent, lub ⁇ cating agent, adjuv ant. v elude, delivery system, emulsifier. disintegrant. absorbent, presenative. surfactant, colorant, flavorant. or sweetener
  • the composiuon of the invention may be admi stered orally .
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intrape ⁇ toneal. intrathecal. lntravent ⁇ cular, intrasternal. and intracranial mjecUon or infusion techniques When administered parenterally.
  • the composition will normallv be in a unit dosage, ste ⁇ le injectable form (solution, suspension or emulsion) v hich is preferably isotomc with die blood of the recipient with a pharmaceuucally acceptable earner
  • ste ⁇ le injectable forms are ste ⁇ le injectable aqueous or oleaginous suspensions These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents
  • the ste ⁇ le injectable forms may also be ste ⁇ le injectable solutions or suspensions in non-toxic parenterall -acceptable diluents or solvents, for example, as soluuons in 1.3-butaned ⁇ ol
  • the acceptable vehicles and solvents may be employed are water, saline.
  • Ringer's solution dextrose solution, isoto c sodium chlo ⁇ de soluuon. and Hanks' solution In addiuon.
  • ste ⁇ le. fixed oils are conventionally employed as solvents or suspending mediums
  • anv bland fixed oil may be emplo ed including synthetic mono- or di-glyce ⁇ des. com. cottonseed, peanut, and sesame oil
  • Fatty acids such as ethyl oleate. isoprop l mynstate. and oleic acid and its glycende denvauves. including olive oil
  • oil soluuons or suspensions may also contain long-chain alcohol diluents or dispersants
  • Ste ⁇ le saline is a prefe ⁇ ed earner, and the compounds are often sufficiendy water soluble to be made up as a solution for all foreseeable needs
  • the earner mav contain minor amounts of addiuv es. such as substances that enhance solubility , lsotonicity. and chemical stabihtv . e g . anti-oxidants. buffers and presenatives
  • Formulations suitable for nasal or buccal admimstration may comp ⁇ se about 0 1% to about 5% w/w . for example 1% w/w of active ingredient
  • the formulations for human medical use of die present invention comp ⁇ se an active ingredient in associauon with a pharmaceutically acceptable earner therefore and optionally other therapeutic mgred ⁇ ent(s)
  • die composiUon When administered orally, die composiUon will usually be formulated into unit dosage forms such as tablets, cachets powder, granules, beads, che able lozenges, capsules, liquids, aqueous suspensions or solutions, or similar dosage forms, using convenuonal equipment and techniques known in the art
  • Such formulations typicallv include a solid, semisohd. or liquid earner
  • Exemplary earners include lactose, dextrose, sucrose. sorbitol. mannitol starches, gum acacia calcium phosphate, mineral oil cocoa butter, oil of theobroma. alginates. tragacanth. gelatin, syrup, meth l cellulose, pol oxyethv lene sorbitan monolaurate. methyl hydroxvbenzoate. propyl hydroxybenzoate. talc, magnesium stearate. and the like
  • the composiUon of the mv enuon is preferably administered as a capsule or tablet containing a single or divided dose of the inhibitor Preferabl .
  • the composition is administered as a ste ⁇ le soluuon. suspension, or emulsion, in a single or div ided dose
  • Tablets mav contain earners such as lactose and com starch, and/or
  • Iub ⁇ cating agents such as magnesium stearate Capsules may contain diluents including lactose and d ⁇ ed com starch
  • a tablet may be made by compressing or molding the acuve ingredient optionally with one or more accessory ingredients
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lub ⁇ cant.
  • inert diluent, surface active, or dispersing agent Molded tablets may be made by molding in a suitable machine, a mixture of the powdered active ingredient and a suitable earner moistened with an inert liquid diluent
  • compositions can be prepared by mixing the drug with a suitable non-imtaUng excipient which is solid at room temperature, but liquid at rectal temperamre. and. therefore, will melt m the recmm to release the dmg
  • suitable non-imtaUng excipient which is solid at room temperature, but liquid at rectal temperamre. and. therefore, will melt m the recmm to release the dmg
  • mate ⁇ als include cocoa butter, beeswax, and pol ethylene gl cols
  • compositions of die mvention may be inco ⁇ orated into a hydrophobic pol mer mat ⁇ x for controlled release over a pe ⁇ od of days
  • the composition of die mvention may then be molded into a solid implant, or externally applied patch, suitable for providing efficacious concentrations of the PARP inhibitors over a prolonged pe ⁇ od of ume without the need for frequent re-dosing
  • Such controlled release films are well known to the art Particularly prefe ⁇ ed are uansdermal dehven systems
  • polymers commonly employed for this pu ⁇ ose that may be used in the present inv ention include nondegradable ethvlene-vinyl acetate copolymer an degradable lactic acid-glycohc acid copolymers which may be used externally or internally Certain hydrogels such as pol (hydroxy eth lmethacry late) or poly(v ⁇ nylalcohol
  • the earner is a solid biodegradable polymer or mixture of biodegradable pol mers with approp ⁇ ate time release characte ⁇ sucs and release kinetics
  • the composition of the invention may then be molded into a solid implant suitable for providing efficacious concentrations of the compounds of the invention over a prolonged pe ⁇ od of time without the need for frequent re-dosing
  • the composition of the present invention can be inco ⁇ orated into the biodegradable pol mer or polymer mixture in any suitable manner known to one of ordinary skill in the art and may form a homogeneous mauix with the biodegradable polymer, or may be encapsulated in some way within the polymer, or may be molded into a solid implant
  • the biodegradable polvmer or polymer mixture is used to form a soft " depot" containing the pharmaceutical composiUon of the present invenuon that can be administered as a flowable liquid for example, by injection but which remains sufficiently v iscous to maintain the pharmaceuucal composition within the localized area around the injection site
  • the degradation time of the depot so formed can be va ⁇ ed from several days to a few years, depending upon the polymer selected and its molecular weight
  • a flexible or flowable delivery "depot "" w ill adjust to the shape of the space it occupies w ith the body with a minimum of trauma to sunounding ussues
  • the pharmaceutical composition of the present invention is used in amounts that are therapeuticallv effecuve. and mav depend upon the desired release profile, the concentration of die pharmaceutical composition required for the sensitizing effect, and the length of time that the pharmaceutical
  • the PARP inhibitors are used in the composiUon in amounts that are therapeutically effectiv e
  • the compositions mav be ste ⁇ hzed and/or contain adjuvants, such as presen ing. stabilizing welling, or emulsifying agents, soluuon promoters, salts for regulaUng the osmouc pressure, and/or buffers In addiuon they may also contain other therapeuucally valuable substances
  • the compositions are prepared according to conventional mixing, granulaung.
  • die compounds of the present invenuon should readily penetrate die blood-brain bar ⁇ er when pe ⁇ pherally administered Compounds which cannot penetrate the blood-bram bamer can be effecuvelv administered by an intrav ent ⁇ cular route or other appropnate delivery system suitable for admimstrauon to the brain
  • Doses of the compounds preferably include pharmaceutical dosage units compnsing an efficacious quantity of active compound
  • an efficacious quanuty is meant a quantity sufficient to inhibit PARP and de ⁇ ve its beneficial effects through administration of one or more of the pharmaceutical dosage units
  • the dose is sufficient to prevent or reduce the effects of vascular stroke or other neurodegenerauve diseases
  • the amount required of the active ingredient to achieve a therapeuUc effect will vary with the particular compound, the route of admimstrauon, the mammal under treatment, and the particular disorder or disease being treated
  • a suitable systematic dose of a compound of the present invenuon or a pharmacologically acceptable salt thereof for a mammal suffenng from, or likely to suffer from, any of condition as desc ⁇ bed hereinbefore is in the range of about 0 1 mg/kg to about 100 mg/kg of the acuve ingredient compound the most prefe ⁇ ed dosage being about 1 to about 10 mg kg
  • a specific dose level for a particular patient will depend upon a va ⁇ ety of factors including the activ itv of the specific compound emplo ed, the age. body weight, general health, se . diet, time of admimstrauon. rate of excretion, drug combination, and the seventy of the particular disease being treated and form of admimstrauon
  • the compounds may also be blended with convenuonal excipients such as binders, including gelatin, pregelaumzed starch, and the like.
  • lub ⁇ cants such as hydrogenated vegetable oil. steanc acid, and the like, diluents, such as lactose, mannose. and sucrose, disintegrants. such as carboxymethy lcellulose and sodium starch glycolate.
  • suspending agents such as povidone. polyvinyl alcohol, and the like, absorbants. such as silicon dioxide, preservatives, such as methy lparaben. propy lparaben. and sodium benzoate surfactants such as sodium laury 1 sulfate. polysorbate 80. and die like, colorants such as F D & C dves and lakes, flavorants and sweeteners
  • the present invention relates to the use of compounds I. II. . III. IV or V in the preparation of a medicament for the treatment of any disease or disorder in an animal desc ⁇ bed herein
  • a convenient method to determine IC 5 0 of a PARP inhibitor compound is a PARP assay using punfied recombinant human PARP from Trevigan (Gaithersburg. MD), as follows
  • the PARP enzyme assay is set up on ice in a volume of 100 microliters consisung of 100 mM T ⁇ s-HCI (pH 8 0). 1 mM MgCl 2 . 28 mM KCl. 28 mM NaCl 0 1 mg/ml of DNase I acuvated he ⁇ ing sperm DNA (Sigma. MO). 3 0 micromolar [3H]n ⁇ cot ⁇ nam ⁇ de ade ⁇ ine dinucleoude (470 mci/mmole).
  • die radioacUvity is determined by scinullation counting
  • the compounds of this invention were found to have potent enzymauc acuvity in the range of a few nM to 20 ⁇ M in IC 50 in this inhibiuon assay
  • Focal cerebral ischemia is produced by caute ⁇ zauon of the ⁇ ght distal MCA (middle cerebral artery) with bilateral temporary common caroud artery occlusion in male Long-Evans rats for 90 minutes All procedures performed on the ammals are approved by the Umversity InsUtuuonal .Ammal Care and Use Committee of die Umversity of Pennsylvama A total of 42 rats (weights 230-340 g) obtained from Charles River were used in this study The animals fasted ov ernight with free access to water p ⁇ or to the surgical procedure
  • the rats were then anestheuzed with halodiane (4% for induction and 0 8%-I 2% for the surgical procedure) in a mixture of 70% muous oxide and 30% oxygen
  • the body temperature was monitored by a rectal probe and maintained at 37 5 + 0 5°C with a heating blanket regulated by a homeothermic blanket control unit (Han'ard Apparatus Limited. Kent. U K )
  • a catheter (PE-50) was placed into the tail artery, and arte ⁇ al pressure was continuously momtored and recorded on a Grass polygraph recorder (Model 7D. Grass Instruments. Quinc . Massachusetts) Samples for blood gas anal sis (arte ⁇ al pH.
  • PaOj and PaC0 2 were also taken from the tail artery catheter and measured with a biood gas anaivzer ( ABL 30 Radiometer. Copenhagen. Denmark) Artenal blood samples were obtained 30 minutes after MCA occlusion The head of the animal was positioned in a stereotaxic frame and a ⁇ ght panetal incision between the ⁇ ght lateral canthus and the external auditory mearus was made Using a dental d ⁇ ll constantly cooled with saline, a 3 mm bun hole was prepared ov er the co ⁇ ex supplied bv the ⁇ ght MC A.
  • the dura mater and a thin inner bone lay er were kept, care being taken to position the probe over a tissue area dev oid of large blood v essels
  • the flow probe (tip diameter of 1 mm. fiber separation of 0 25 mm) was lowered to the bottom of the cranial bun hole using a micromampulator
  • the probe was held stationary by a probe holder secured to the skull with dental cement
  • the microvascular blood flow in the ⁇ ght panetal cortex was cont uouslv monitored with a laser Doppler flow meter (FloLab. Moor. Devon. U K . and Pe ⁇ flux 4001. Pe ⁇ med. Sweden)
  • Focal cerebral ischemia was produced bv cautenzauon of the distal po ⁇ ion of the ⁇ ght MCA with bilateral te poran common caroud artery (CCA.) occlusion bv the procedure of Chen et al . "A Model of Focal Ischemic Stroke in die Rat Reproducible Extensive Cortical Infarcuon". Stroke 17 738-43 ( 1986) and/or Liu et al.. "Polyethylene Glycol-conjugated Supero.xide Dismutase and Catalase Reduce Ischemic Brain Injury ". Am I Physiol 256 H589-93 (1989). both of which are hereby inco ⁇ orated by reference
  • bilateral CCA's were isolated, and loops made from polyethylene (PE-10) catheter were carefully passed around the CCA's for later remote occlusion
  • the incision made previously for placement of the laser doppler probe was extended to allow obse auon of die rostral end of the zygomauc arch at die fusion point using a dental d ⁇ ll. and the dura mater overlying the MCA was cut
  • the MCA distal to its crossing with the lnfenor cerebral vein was lifted bv a fine stainless steel hook attached to a micromampulator and.
  • the MCA was caute ⁇ zed with an electrocoagulator
  • the bun hole was covered with a small piece of Gelform. and the wound was sutured to maintain the bram temperamre within the normal or near- normal range
  • the brain slices were incubated in phosphate-buffered saline containing 2% 2.3.5-tnphenyltetrazol ⁇ um chlonde (TTC) at 37°C for ten minutes Color photographs were taken of die poste ⁇ or surface of the stained slices and were used to determine the damaged area at each cross-secuonal level using a computer-based image analyzer (NTH Image 1.59). To avoid artifacts due to edema, the damaged area was calculated by subtracung the area of the normal tissue in the hemisphere ipsilateral to the stroke from the area of the hemisphere contralateral to the stroke, by the method of Swanson et al..
  • the animals are anesdieuzed with isofluorane (induction. 3%. maintenance. 1 25% in a mixture of 30% O; and 70%) NO; through a face mask
  • the rectal temperature is maintained at 37 "C with a homeothermic blanket (Han s ard Apparatus. Soudi N'auck. MA)
  • an iv catheter is inserted into the left femoral vein and e line run up through the nape of die neck for connection to a tethered swivel (Instech Laboratones. Pl mouth Meeung. PA) and remote infusion pump (Stoelting Inc . Wood Dale. IL)
  • the ⁇ ght femoral artery is cannulated for monito ⁇ ng arte ⁇ al blood pressure and heart rate and for obtaining blood samples for artenal blood gas
  • the ⁇ ght middle cerebral artery (MCA) is then exposed bv making ertical skin incision midway between the nght eye and ear and ov erlying skull is removed with a dental dnll (Chen et al 1986) After incision of the dura, the artery is coagulated at the level of die infenor cerebral vein with a bipolar cautery unit (Valleylab NS2000. Boulder. CO), and cut to prev ent spontaneous reperfusion (Takahashi et al..
  • the PARP inhibitor to be tested is first administered 30 min after MCAO as an iv bolus. 10 mg/kg infused over 5 min. followed by a 12 hr continuous infusion of 2 mg/kg/hr (0 3 ml/hr) Ninety minutes after the MCAO. the animals are remov ed from the infusion tether, b ⁇ efl reanesthetized with isofluorane.
  • the brain slices are incubated in phosphate-buffered saline (pH 7 4) containing 2% TTC at 37°C for 10 min and then stored in 10% neutral-buffered formalin
  • Cross-sectional area of the TTC-unstained region for each brain slice is determined using an image analyzer (MetaMo ⁇ h.
  • the total volume of infarction in the ⁇ ght hemisphere is calculated by summation of the direct (TTC-negative) and indirect measures of the infarct areas in the component brain slices
  • the compounds are administered in either a single dose or a se ⁇ es of muluple doses, i p or l v . at different times both before or after the onset of ischemia
  • Compounds of the inv ention are expected to provide protecuon from ischemia in the range of about 0 to 80%
  • the expe ⁇ ments of the heart lschemia/reperfusion injun model is performed using female Sprague- Dawlev rats weighing 250-300g w hich are anesdietized with sodium pentobarbital at dose of 65 mg/kg lntrapentoneally
  • the rectal temperature is maintained at 7°C by using a homeothermic blanket s stem (Han s ard Apparatus. South Natick MA)
  • the trachea is cannulated and the rat is ventilated with Room Air by using Hanard Rodent VenUlator (Ha ard Apparatus. South Nauck.
  • the left jugular v ein is cannulated with PE-50 tubing for dmg delivery
  • the ⁇ ght caroud art is cannulated for BP measurement
  • the heart is exposed by opemng the chest at the 4 th left intercostal space
  • a ma left branch of coronan artery (L D) is occluded bv 4-0 silk ligature for 30 m of ischemia and released for 90 min of reperfusion Du ⁇ ng the expenment.
  • a ⁇ e ⁇ al BP and EKG are monitored bv Micro-Med Cardiov ascular System (Louisv die KY)
  • the LAD coronan art is re-occluded and about 2 ml of 5% Evans Blue dye is injected through I v line to distinguish die ischemic area from non-ischemic area of the heart
  • the heart is immediately taken off and frozen in the freezer
  • the heart is sliced into five secuons with 2 mm thickness and stained in 1% TTC soluuon for 30 min at 37°C
  • the ⁇ ght vent ⁇ cle is t ⁇ mmed off Infarct area, ⁇ sk area and total left v enuicular area in each face of the secuon are measured by using an image analysis svstem (BIOQUANT. Nashville TN)
  • the infarct size is calculated as the percent total infarct volume of the total ⁇ sk volume
  • the compounds of this invention are expected to protect against ischemia-induced degenerauon of rat cortical neurons in vitro and thus may be useful in disorders a ⁇ sing from cerebral ischemia such as stroke sepuc shock or CNS degenerative disorders Thev may also be useful in protecting the spinal cord following trauma
  • the present invention is further directed to a method of prophv lactic or therapeutic treatment of heart attack cardiac anest cardiac bvpass diabetes or ⁇ sk of damage which comp ⁇ ses administenng an effective amount of a compound of the present invention for PARP inhibition in unit dosage form
  • Gene expression alteration mav be measured with human fibroblast BJ cells which at Population Doubling (PDL) 94 are plated in regular growth medium and then changed to low semm medium to reflect phvsiological conditions desc ⁇ bed in Linskens et al Nucleic Acids Res 23 16 3244-3251 (1995)
  • a medium of DMEM 199 supplemented with 0 5% bovine calf semm is used
  • the cells are treated dailv for 13 davs
  • the control cells are treated widi and without die solv ent used to administer the PARP inhibitor
  • the untreated old and voung conuol cells are tested for compa ⁇ son RNA is prepared from the treated and conuol cells according to die techniques desc ⁇ bed in PCT Pubhcauon No 96/13610 and Northern blotting is conducted
  • Probes specific for senescence-related genes are analvzed and treated and control cells compared In analvzing the results the lowest level of gene expression is arbiu
  • Gene expression alterauon mav be measured widi approximately 105 BJ cells, at PDL 95-100 which are plated and grown in 15 cm dishes
  • the growth medium is DMEM 199 supplemented with 10% bovice calf semm
  • the cells are ueated daily for 24 hours with the PARP inhibitors of (100 ⁇ g 1 mL of medium) WO 99/11645
  • the cells are washed with phosphate buffered solution (PBS), then permeabhzed with 4% paraformaldehyde for 5 minutes, then washed with PBS. and treated w ith 100% cold methanol for 10 minutes The methanol is removed and the cells are washed with PBS.
  • PBS phosphate buffered solution
  • human fibroblast cells lines (either W 138 at Population Doubling (PDL, 23 or BJ cells at PDL 1) are thawed and plated on T75 flasks and allowed to grow in normal medium (DMEM/M199 plus 10% bovine calf semm) for about a week, at which time die cells are confluent, and the cultures are therefor ready to be subdivided
  • the media is aspirated, and the cells ⁇ nsed with phosphate buffer saline (PBS) and then trypsinized
  • PBS phosphate buffer saline
  • the cells are counted with a Coulter counter and plated at a density of 10 5 cells per cnr in 6-well ussue culture plates in DMEM/199 medium supplemented with 10% bovine calf semm and varying amounts (0 lO ⁇ M.
  • Thermal hvperalgesia to radiant heat is assessed by using a paw -withdrawal test
  • the rat is placed m a plasuc cylinder on a 3-mm thick glass plate with a radiant heat source from a projection bulb placed directly under the plantar surface of die rat's hindpaw
  • the pa -withdrawal latency is defined as the time elapsed from die onset of radiant heat stimulation to withdrawal of the rat's hindpaw
  • Mechanical hvperalgesia is assessed bv placing the rat in a cage with a bottom made of perforated metal sheet w ith manv small square holes Duration of paw-withdrawal is recorded after p ⁇ cking the mid-plantar surface of the rat's hindpaw with the tip of a safetv pin inserted dirough the cage bottom
  • Mechano-allodyma is assessed by placing a rat in a cage similar to the previous test, and applying von Frey filaments in ascending order of bending force ranging from 0 07 to 76 g to the mid-plantar surface of the rat's hindpaw A von Frey filament is applied pe ⁇ endicular to the skin and depressed slo ly until it bends A threshold force of response is defined as die first filament in the se ⁇ es to evoke at least one clear paw- withdrawal out of five applications

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Biochemistry (AREA)
  • Dermatology (AREA)
  • Psychology (AREA)
  • Virology (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Obesity (AREA)
  • Endocrinology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)

Abstract

This invention relates to compounds, pharmaceutical compositions, and methods of using the disclosed compounds for inhibiting PARP.

Description

COMPOUNDS, METHODS AND PHARMACEUTICAL COMPOSITIONS FOR TREATING CELLULAR DAMAGE. SUCH AS NEURAL OR CARDIOVASCULAR TISSUE DAMAGE
FIELD OF THE INVENTION
The present invention relates to inhibitors of the nuclear enzyme poly(adenosιne 5'-dιphospho-nbose) polymerase ["poiy( ADP-πbose) polymerase" or "PARP". which is also referred to as ADPRT (NAD protein (ADP-nbosyl transferase (polymersing)). pADPRT (poly( ADP-πbose) transferase) and PARS (polv (ADP-πbose) synthetase) and provides compounds and compositions containing the disclosed compounds Moreover, the present ention provides methods of using the disclosed PARP inhibitors to prevent and/or treat tissue damage resulting from cell damage or death due to necrosis or apoptosis. neural tissue damage resulting from, for example, ischemia and reperfusion injury, such as cerebral lschemic stroke, head trauma or spinal cord injury, neurological disorders and neurodegeneram e diseases, such as. for example. Parkinson's or Alzheimer s diseases and multiple sclerosis, to prevent or treat vascular stroke, to treat or prevent cardiovascular disorders, such as. for example, mvocardial infarction, to treat other conditions and/or disorders such as. for example, age- related muscular degeneration. AIDS and other immune senescence diseases, arthritis, atherosclerosis, cachexia. cancer, degenerative diseases of skeletal muscle involving replicative senescence, diabetes (such as diabetes mellitus). inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis. hemorrhagic shock, splanchnic artery occlusion shock, multiple organ failure (such as involving am of the kidne . li er, renal, pulmonary retinal, pancreatic and/or skeltal muscle systems), acute autoimmune th roidms. muscular dystrophy . osteoaπhπtis. osteoporosis, chronic and acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotouc shock), local and/or remote endothehal cell dysfunction (such are recognized by endo-dependent relaxant responses and up-regulation of adhesion molecules), inflammation and skin aging, to extend the lifespan and proliferative capacity of cells, such as. for example, as general mediators m the generation of ox dants. proinflammatory mediators and/or cytokines. and general mediators of leukocyte infiltration, calcium ion overload, phosphohpid peroxidaion. impaired nitnc oxide metabolism and/or reduced ATP production, to alter gene expression of senescent cells, or to rad osensitize hypoxic tumor cells
BACKGROUND OF THE INVENTION
PARP (EC 2 4 2 30). also known as PARS (for poly( ADP-πbose) synthetase). or ADPRT (for NAD protein (ADP-nbosyl) transferase (polymeπsing)). or pADPRT (for polv (ADP-πbose) transferase). is a major nuclear protein of 1 16 kDa It is present in almost all cukaryotes The enz me s nthesizes poly ( ADP-πbose) a branched poivmer that can consist of over 200 ADP-πbose units from NAD The protein acceptors of pol (. DP-πbose) are directlv or indirecth involv ed in maintaining DNA integπty Thev include histones. topoisomerases. DNA and RNA polymerases. DNA ligases. and Ca2'- and Mg:"-dependent endonucleases PARP protein is expressed at a high lev el in many tissues, most notably in the immune system, heaπ. brain and germ-line cells Under normal ph siological conditions, there is minimal PARP activity However. DNA damage causes an immediate activation of PARP bv up to 500-fold Among the many functions attπbuted io P ARP is its major role in facilitaung DNA repair bv ADP-πbosy lation and therefore co-ordinating a number of DNA repair proteins As a result of PARP activation. NAD levels significantly decline While many endogenous and exogenous agents have been shown to damage DNA and acti ate PARP. peroxynitπte. formed from a combination of nitπc oxide (NO) and superoxide. appears to be a main perpetrator responsible for vaπous reported disease conditions in v IVΌ. e g . duπng shock and inflammation
Extensiv e PARP activation leads to severe depletion of NAD in cells suffeπng from massive DNA damage The short life of polv ( ADP-πbose ) (half-life < 1 mm) results in a rapid turnover rate Once pol (ADP-πbose) is formed, it is quicklv degraded by the constitutiv ely activ e pol (ADP-πbose) glycohv drolase (PARG). together with phosphodiesterase and (.ADP-πbose) protein lyase PARP and PARG form a cycle that com ens a large amount of NAD to ADP-πbose In less than an hour, over-stimulation of PARP can cause a drop of NAD and ATP to less than 20% of the normal le el Such a scenaπo is especiallv detπmental duπng ischaemia when depπvation of oxygen lias already drastically compromised cellular energy output Subsequent free radical production duπng reperfusion is assumed to be a major cause of tissue damage Part of the ATP drop, hich is typical in manv organs duπng ischaemia and reperfusion. could be linked to NAD depletion due to polv (ADP-πbose) turnover Thus. PARP or PARG inhibition is expected to preserve the cellular energy level to potentiate the sur ival of lschaemic tissues after insult
Polv (.ADP-πbose) sy nthesis is also inv olv ed in the induced expression of a number of genes essential for inflammatory response P.ARP inhibitors suppress production of inducible nitπc oxide s nthase (iNOS ) in macrophages. P-type selectin and intercellular adhesion molecule- 1 (ICAM-1 ) in endothehal 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 (Zhang. J "'PARP inhibition a novel approach to treat lschaemia/reperfusion and inflammation-related injuπes". Chapter 10 in Emerging Drugs ( 1999) 4 209-221 Ashley Publications Ltd . and references cited therein )
P.ARP production is activated by damaged DNA fragments which, once activated, catalyzes the attachment of up to 100 ADP-πbose units to a vaπety of nuclear proteins, including histones and PARP itself Duπng major cellular stresses the extensiv e activation of PARP can rapidly lead to cell damage or death through depletion of energy stores As four molecules of ATP are constuned for ev en molecule of NAD (the source of ADP-πbose and P.ARP substrate) regenerated. NAD is depleted by massive P.ARP activation and. in the efforts to re-synthesize NAD. ATP may also be depleted It has been reported that P.ARP activation pla s a kev role in both NMD A- and NO-induced neurotoxicity This has been demonstrated in conical cultures and in hippocampal slices wherein prevention of toxicirv is directly correlated to PARP inhibition potency (Zhang et al . "Nitπc Oxide Activation of Poly(ADP- Ribose) Svnthetase in Neurotoxicity". Science. 263 687-89 (1994) and Wallis et al . "Neuroprotection Against Nitπc Oxide Injury with Inhibitors of ADP-Ribosvlation". NeuroReport. 5 3. 245-48 (1993)) The potential role of PARP inhibitors in treating neurodegenerativ e diseases and head trauma has thus been recognized even if the exact mechanism of action has not yet been elucidated (Endres et al . "Ischemic Brain Injury is Mediated by the Activation of Poly(ADP-Rιbose)Polymerase". J Cereb Blood Flow Metabol . 17 1143-51 (1997) and Walhs et al . "Traumatic Neuroprotection with Inhibitors of Nitπc Oxide and ADP-Ribosy lation. Brain Res . 710 169-77 (1996))
Similarlv 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 3-amιno-benzamιde (10 mg/kg). either one minute before occlusion or one minute before reperfusion. caused similar reducuons in infarct size in the heart (32-42%) while 1.5-dιhydroxvιsoquιnohne (1 mg/kg). another PARP inhibitor reduced infarct size by a comparable degree (38-48%) Thiemermann et al . "Inhibition of the Activity of Poly(.ADP Ribose) Sv nthetase Reduces Ischemia-Reperfusion Injurv in the Heart and Skeletal Muscle". Proc Natl Acad Sci USA. 94 679-83 (1997) These results make it reasonable to suspect that PARP inhibitors could salvage previously ischemic heart or skeletal muscle tissue
PARP activation can also be used as a measure of damage following neurotoxic insults following over- exposure to any of glutamate (via NMD A receptor stimulation), reactive oxygen intermediates, amvloid β - protein. N-methyI-4-phenv 1-1.2.3 6-tetrahv dropy πdine (MPTP) or us active metabolite N-methyl-4- phenylpvπdine (MPP~). which participate in pathological conditions such as stroke. Alzheimer's disease and Parkinson's disease Zhang et al . "Poly(ADP-Rιbose) Synthetase Activation An Earlv Indicator of Neurotoxic DNA Damage". J Neurochem . 65 3. 141 1-14 ( 1995) Other studies have continued to explore the role of PARP activation in cerebellar granule cells in v uro and in MPTP neurotoxicity Cosi et al . "Poly(ADP-Rιbose)
Polymerase (PARP) Rev lsited A New Role for an Old Enzyme PARP Involv e ent in Neurodegeneration and PARP 'nhibitors as Possible Neuroprotectiv e Agents". Ann N Y Acad. Sci.. 825 366-79 (1997). and Cosi et al., "Poly(ADP-Rιbose) Polymerase Inhibitors Protect Against MPTP-induced Depletions of Stπatal Dopamine and Cortical Noradrenahne in C57B1/6 Mice", Brain Res.. 729 264-69 (1996) Excessive neural exposure to glutamate, which serves as the predominate central nervous system πeurotransmitter and acts upon the N- methyl-D-aspanate (NMDA) receptors and other subtype receptors, most often occurs as a result of stroke or other neurodegenerative processes Oxygen depπv ed neurons release glutamate in great quantities duπng ischemic brain insult such as duπng a stroke or heart attack This excess release of glutamate in turn causes over-stimulation (e citotoxicit ) of N-methv I-D-aspartate (NMDA). AMP A. 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 ultimatelv results in cell damage or death via the production of proteases, lipases and free radicals Excessi e activation of glutamate receptors has been implicated in vaπous neurological diseases and conditions including epilepsy, stroke. Alzheimer s disease. Parkιnson"s disease. Amvotrophic 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 manv animal species, as well as in cerebral cortical cultures treated with glutamate or NMDA. that glutamate receptor antagonists (1 e . compounds which block glutamate from binding to or activating its receptor) block neural damage following vascular stroke Davvson et al . "Protection of the Brain from Ischemia" Cerebrovascular Disease 319-25 (H Hunt Batjer ed . 1997) Attempts to prevent excitoto.xicm by blocking NMDA. AMP A. ainate 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 manv of the compositions that are effectiv e in blocking the receptors are also toxic to animals As such, there is presently no known effectiv e treatment for glutamate abnormalities
The stimulation of NMDA receptors bv glutamate for example, activates the enzyme neuronal ine oxide synthase (nNOS) leading to the formation of nitnc oxide (NO), which also mediates neurotoxicity NMDA neurotoxicity mav be prevented b treatment with nitπc o ide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro Daw son et al . "Nitπc Oxide Mediates Glutamate Neurotoxicity in Pπmary Cortical Cultures". Proc Natl Acad Sci USA. 88 6368-71 (1991). and Dawson et al "Mechanisms of Nitπc Oxide-mediated Neurotoxicity in Pπmary Brain Cultures". J Neurosci . 13 6. 2651-61 (1993). Dawson et al . "Resistance to Neurotoxicity in Cortical Cultures from Neuronal Nitπc Oxide Synthase-Defiαent Mice". J Neurosci . 16 8. 2479-87 ( 1996). Iadecola. "Bπght and Dark Sides of Nitnc Oxide in Ischemic Brain Injury ". Trends Neurosci . 20 3. 132-39 (1997). Huang et al . "Effects of Cerebral Ischemia in Mice Deficient in Neuronal Nitπc Oxide Svnthase" Science. 265 1883-85 (1994). Beckman et al . "Pathological Implications of Nitπc Oxide. Superoxide and Peroxynitnte Formauon". Biochem Soc Trans . 21 330-34 (1993). and Szabo et al.. "DNA Strand Breakage. Activation of Poly ( ADP-Ribose) Synthetase. and Cellular Energy Depletion are Involved in the Cytotoxicity in Macrophages and Smooth Muscle Cells Exposed to Peroxynitnte", Proc Natl Acad Sci USA. 93 1753-58 (1996) It is also known that PARP inhibitors, such as 3-ammo benzanude. affect DNA repair generally in response, for example, to hydrogen peroxide or gamma-radiation Cπsto ao et al.. "Effect of a Poly(ADP- Ribose) Polymerase Inhibitor on DNA Breakage and Cvtotoxicit Induced by Hydrogen Peroxide and γ- Radiation." Terato . Carcino . and Muta . 16 219-27 ( 1996) Specifically . Cπstovao et al observed a PARP- dependent recov ery of DNA strand breaks in leukocytes treated with hydrogen peroxide PARP inhibitors have been reported to be effective in radiosensitizing hypoxic tumor cells and effective in prev enting tumor cells from recoveπng from potenuallv lethal damage of DNA after radiation therapy, presumably by their ability to prev ent DNA repair U S Patent Nos 5.032.617. 5.215.738 and 5,041.653
E idence also exists that PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis Salzman et al . "Role of Peroxv nitπte and
Figure imgf000006_0001
(ADP-Rιbose)S nthase Activation Expeπmental Colitis." Japanese J Pharm . 75. Supp I 15 ( 1997) Specificallv Colitis was induced in rats bv intraluminal administration of the hapten tπnitrobenzene sulfonic acid in 50% ethanol Treated rats received 3- aminobenzamide a specific inhibitor of PARP activity Inhibition of PARP activitv reduced the inflammatory response and restored the morphologv and the energetic status of the distal colon See also Southan et al , "Spontaneous Rearrangement of Anunoalkv lithioureas into Mercaptoalky lguanidines. a Nov el Class of Nitπc Oxide Svnthase Inhibitors with Selectivity Towards the Inducible Isoform" Br J Pharm 117 619-32 (1996). and Szabo et al . "Mercaptoeth lguamdine and Guanidine Inhibitors of Nitπc Oxide S nthase React with Peroxv nitπte and Protect Against Peroxv nitπte-inducεd Oxidative Damage" J Biol Chem . 272 9030-36 (1997) E idence also exists that PARP inhibitors are useful for treating arthπtis Szabo et al "Protective
Effects of an Inhibitor of Polv (ADP-Ribose (Svnthetase in Collagen-Induced Arthπtis " Japanese J Pharm 75 Supp I 102 (1997), Szabo et al . "DNA Strand Breakage Activation of Poly(ADP-Rιbose)Svnthetase. and Cellular Energy Depletion are Invol ed in the Cytotoxicity in Macrophages and Smooth Muscle Cells Exposed to Perox nitnte." Proc Natl Acad Sci USA. 93 1 53-58 (March 1996). and Bauer et al . "Modification of Growth Related Enzvmatic Pathwavs and Apparent Loss of Tumoπgenicity of a ras-transformed Bovine
Endothehal Cell Line by Treatment with 5-Iodo-6-amιno-1.2-benzopyrone (INH2BP)" Intl J Oncol 8 239-52 (1996). and Hughes et al . "Induction of T Helper Cell Hyporesponsiv eness in an Expeπmental Model of Autoimmuniry by Using Nonmitogenic Antι-CD3 Monoclonal Antibody" J Immuno 153 3319-25 ( 1994) Further PARP inhibitors appear to be useful for treating diabetes Heller et al . "Inactivation of the Poly(ADP-Rιbose)Polvmerase Gene Affects Oxygen Radical and Nitπc Oxide Toxicity in Islet Cells." J Biol Chem . 270 19 11176-80 (May 1995) Heller et al used cells from mice with inactivated P RP genes and found that these mutant cells did not show NAD" depletion after exposure to DNA-damaging radicals The mutant cells were also found to be more resistant to the toxicity of NO
PARP inhibitors hav e been shown to be useful for treating endotoxic shock or septic shock Zingarelh et al . "Protective Effects of Nicotinaπude Against Nitπc Oxide-Mediated Delayed Vascular Failure in Endotoxic Shock Potential Involvement of PolyADP Ribosvl Synthetase." Shock. 5 258-64 (1996). suggests that inhibition of the DNA repair c cle tπggered by poly(ADP nbose) synthetase has protective effects against vascular failure in endotoxic shock Zingarelh et al found that nicotinamide protects against delayed NO-mediated vascular failure in endotoxic shock Zingarelh et al also found that the actions of nicotinamide may be related to inhibition of the NO-mediated activation of the energy -consuming DNA repair cycle, tπggered bv poly(ADP
- 3 πbose) s nthetase Cuzzocrea. "Role of Peroxv nitπte and Activation of Poly(ADP-Rιbose) Synthetase in the Vascular Failure Induced bv Zv mosan-activ ated Plasma." Bnt J Pharm . 122 493-503 (1997)
PARP inhibitors have been used to treat cancer Suto et al "Dihydroisoquinolinoπes The Design and Synthesis of New Seπes of Potent Inhibitors of Poly (ADP-Ribose) Poiymerase". Anticancer Drug Des . 7 107- 17 (1991) In addition. Suto et al . 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(5H)-Phenanthπdιnone. an Inhibitor of Poly (ADP-πbose) Polymerase. on Cultured Tumor Cells". Oncol Res . 6 9 399-403 (1994) discusses the inhibition of PARP activ lty . reduced proliferation of tumor cells, and a marked synergistic effect when tumor cells are co-treated with an alkylating drug Still another use for PARP inhibitors is the treatment of peπpheral nerve injuπes. and the resultant pathological pain syndrome known as neuropatluc pain, such as that induced by chronic constπction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characteπzed by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark" neurons) occurs Mao et al . Pain. 72 355- 366 (1997) 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, osteoarthπtis. osteoporosis, muscular dystrophy degenerative diseases of skeletal muscle involv ing replicative senescence, age-related muscular degeneration, immune senescence AIDS, and other immune senescence diseases, and to alter gene expression of senescent cells WO 98/279"5 Large numbers of known P.ARP inhibitors hav e been descπbed in Banasik et al . "Specific Inhibitors of
Poly(ADP-Rιbose) Svnthetase and Mono(ADP-Rιbosy l)-Transferase". J Biol Chem . 267 3. 1569-75 (1992). and in Banasik et al . "Inhibitors and Activators of ADP-Ribos lation Reactions". Molec Cell Biochem . 138 185-97 (1994) However, effective use of these P.ARP inhibitors, in the ways discussed above, has been limited bv the concuπent production of unwanted side-effects (Milam et al . "Inhibitors of Poly( Adenosine Diphosphate-Ribose) Synthesis Effect on Other Metabolic Processes" Science. 223 589-91 (1984))
There continues to be a need for effectiv e and potent PARP inhibitors w hich produce minimal side- effects The present invention prov ides compounds, compositions for. and methods of. inhibiting PARP activity for treating 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 mv ention are specifically useful in amelioraung, treating and/or pre enung neural Ussue or cell damage, including that following focal ischemia and reperfusion injury Generally, inhibiuon of PARP activity spares the cell from energy loss, preventing lπeversible depolaπzation of the neurons and. thus, prov ides neuroprotection While not wishing to be bound by am mechanistic theory the inhibition of PARP activity by use of the compounds, compositions and methods of the present invention is believed to protect cells, ussue and organs by protection against the ill-effects of reactive free radicals and nitπc oxide The present invention therefore also provides methods of treating and/or prevenung cells, tissue and/or organs from reacuve free radical and or nitπc oxide induced damage or injury
SUMMARY OF THE INVENTION
The present invenuon provides compunds which inhibit poly (ADP-πbose) polymerase ("PARP"), compositions containing these compounds and methods for using these PARP inhibitors to treat, prevent and/or ameliorate die effects of the condiuons descπbed herein
In one embodiment, the present invenuon provides compounds of Formula I
Figure imgf000008_0001
or a pharmaceutically acceptable salt hydrate, prodrug. or mixtures thereof, wherein
Figure imgf000008_0002
Y is a direct bond. >C=0. -O- -N(RI 0)-. N. or -C(Rg)p-.
X is NRn. -0-. -S-. CRi;R|3. a bond. -CR,:=CR,3-. or -C(R12Ri3)C(R14Ri5)-.
W is selected from -CN. -C(R9);, -(N(R9)2) where the R9 subsutuents may be combined to form a heteroaryl or C3-C8 cycloalkyl opuonally containing at least one hetero atom m place of a carbon atom. -P(O);- 0R9, -P(0)(OR9):, -S(0)_-R9, -S(0)3R9 -C(0)-R9, -C(0)-N(R9)2, -S(0):NR9, C3-C8 cycloalkyl opuonally containing at least one heteroatom in place of a carbon atom, and heteroaryl, R,, R:, R3, ,, R5, Rή, R-, Rg, Rι:, Rι > RM. and R15 . when present, are independently hydrogen, lower
(Cι-C9 straight or branched chain) alkyl. C3-C cycloalk l opuonally containing at least one heteroatom in place of a carbon atom, lower (C;-C9 straight or branched chain) alkenyl, C^-C- cycloalkenyl. lower (C1-C ) alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroan I. hydroxy. amino, nitro, halo, nitroso, sulfo. sulfonic acid, or carboxv. Rg is hydrogen, lower (C Cg straight or branched chain) alkv l. C3-C3 cycloalkyl optionallv containing at least one heteroatom in place of a carbon atom, lower (C:-C , straight or branched chain) alkenyl. aryl. heteroaryl. aralky l heteroaralkyl. alkaryl. alkheteroan 1. hydroxy. lower (C1 -C4) alcohol. lower (C -Cj) alkoxy. ammo, or carboxy . and R,„ and R, , are independently hydrogen, lo er (C -Cy straight or branched chain) alkyl. lower (C:-C9 straight or branched chain) alkeny l. aryl. aralkyl. alkaryl. halo, hydroxy. lower (d-C) alkoxy. am o, or carbox . each R|„ is
Figure imgf000009_0001
hydrogen or lower (C,-Co straight or branched chain) alkyl. and T. when present, is a divalent or tπvalent organic residue independently selected from the group consisting of lower (C:-C, straight or branched chain) alk lene. lower alkenylene. arylene. aralkylene. and alkarylene. wherein one. two or three of the hydrogen atoms of said divalent or tπvalent organic residue can be substituted bv a moiety selected from the group consisung of lower (C] -C9 straight or branched chain) alkyl. cycloalkyl. lower (C:-C^ straight or branched chain) alkenvl. cycloalken l. aryl. heteroary l. aralkyl. heteroar lk l. alka l. alkheteroan 1. halo, tπfluoromethv 1. hydroxy . lower (Cι-C4 ) alkoxy. amino. nitro. tπfluoromethyl. alken loxy. phenoxy . and benzy lo y . wherein one. two. or three carbon atoms in the divalent or tπvalent organic residue can be replaced by a hetero-atom-containing-moiety selected from the group consisting of phenoxy, pheno ymethyl. phenoxy carbonyl. benzy loxy. -0-. >C=0. -S-. -SO;-. -NR, SO:-. -SO:NR,-. -NR,-. and -P0:-, wherein any of the lower (C|-Cα straight or branched chain) alkyl. C3-C8 c cloalkyl optionally contaimng at least one heteroatom in place of a carbon atom, lower (C;-C9 straight or branched chain) alkenyl. aryl. heteroaryl. aralkyl. and alkary l groups can be independently substituted with one. two or three subsutuents selected from the group consisting of lower (C -C, straight or branched chain) alkyl. Ci-Cx cycloalkyl opuonally containing at least one heteroatom in place of a carbon atom, lower (C:-C straight or branched chain) alkenyl. cycloalkenyl. arvl. heteroaryl. aralkyl. heteroaralky l. alkaryl. alkheteroaryl. halo, tπfluoromethyl. hydroxy. lower (Cι-C4) alkoxy. carboxy (such as methoxy or ethoxy). carbonyl. lower alkyl ester (such as methyiester or ethylester). am o. mσo. tπfluoromethyl. alkeny loxy . phenoxy. benzyloxy, wherein one. two. or three carbon atoms of any of the lower (C1-C0 straight or branched chain) alkyl. C3-C8 cycloalkyl optionally contaimng at least one heteroatom in place of a carbon atom, lower (C:-C9 straight or branched chain) alkenyl. aryl. heteroaryl. aralkyl. and alka l groups can be replaced by a hetero-atom- containing-moiety selected from the group consisung of. -0-. >C=0. -S-. -SO;-, -NR, SO;-. -SO;NRr. N. -NR,- . and -PO;-
The dotted line between positions 1 and 2 in chemical formulas herein will be recognized to represent a single or double bond In another embodiment, the present invenuon provides compounds of Formula (II)
Figure imgf000010_0001
or a pharmaceutically acceptable salt, hydrate, prodrug. or mixtures thereof, wherein, q is zero or one.
Figure imgf000010_0002
Z is 0.
X is -0-. or a bond.
Ri . R:. R3. Rj. R<. Re. and R- . when present, are independently hydrogen, lower (Cι-C9 straight or branched chain) alkyl. lower (C;-C straight or branched chain) alkenyl. C3-C8 cy oalkyl optionally containing at least one heteroatom in place of a carbon atom. C<-C- cycloalkenyl. lower (C,-C; ) alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroarly. hydroxy. ammo, nitro. halo, nitroso. or carboxy.
R9 is hydrogen, lower (C,-C= straight or branched chain) alkyl. C3-C3 cycloalky 1 optionally containing at least one heteroatom in place of a carbon atom, lower (C:- straight or branched chain) alkenyl. ary heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroar l. hydroxy. lower (C,-Cι) alcohol, lower (Ci-C,) alkoxy. amino. or carboxy. and T. when present, is a divalent or tnvalent organic radical independently selected from the group consisting of. lower alkylene. lower alken lene. C;-C_ alkeny lox . aryiene. aralkylene. and alkarylene. wherein one. two or three of the h drogen atoms of said divalent or tπvalent organic radical can be substituted by a moiety selected from the group consisting of lower (C,-C9 straight or branched chain) alkyl. lower (C:-Cg straight or branched chain) alken l. ar l. aralkyl. alkaryl. halo, tπfluoromethvi. hydroxy . lower (Ci-Cj) alkoxy. amino. nitro. tπfluoromethyi. alkeny loxy. phenoxy. and benzyloxy. wherein one. two. or three carbon atoms in the divalent or tnvalent organic radical can be replaced by a hetero-atom-containing-moiety selected from the group consisung of: phenoxy. phenoxycarbonyl. benzy loxy. -O- , >C=0. -S-. -SO;-. -NR, S0;-, -SO.NR1-. -NR,-. and -PO;-, wherein the lower alkyl, cyioalkyl opuonally contaimng at least one heteroatom. lower (C;-C9 straight or branched chain) alkenyl. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. and alkheteroaryl groups can be independently substituted with one. two or three subsutuents selected from the group consisting of. lower (Ci-Cς straight or branched chain) alkyl. C3-C« cycolalkyl optionally containing at least one heteratom. lower (C;-C9 straight or branched chain) alkenyl. aryl. heteroar l. aralkyl. heteroaralkyl. alkaryl. alkheteroaryl, halo, tπfluoromethyl. hydroxy . lower (C,-C, ) alkoxy. carboxy (such as methoxy or ethoxy). carbonyl. lower alkyl ester (such as methylester or etliylester). amino. mtro. tπfluoromethyl. alkenyloxy. phenoxy. benzyloxy,
wherein one. two. or three carbon atoms thereof can be replaced by a hetero-atom-containing-moiety selected from the group consisting of -0-. >C=0. -S-. -SO:-, -NR, SO:-. -S02NR|-, N, -NR,-, and -PO:- In a further embodiment, the present invention provides compounds of Formula (III)
Figure imgf000011_0001
wherein
Z and X are oxy gen:
"alk" is lower alkylene:
Rr . Ris and R,9 are independently hydrogen or lower alkyl. or R,- and R,g or R,g and R,9 taken together can be a lower alkylene to form a heterocyclic πng; and
Ri. R;, R3. R,. R . R« and R- are independently h drogen, lower (C,-C9 straight or branched chain) alkyl. C3-C8 cycloalkyl optionally containing at least one hetero atom, lower (C;-C9 straight or branched chain) alkenyl. lower (C,-Cj ) alko.xy. aryl. heteroar l. aralkyl. heteroaralkyl. alkaryl. alkheteroaryl. hydroxy . amino. nitro. halo, rutroso. or carboxy
In yet a further embodiment, the present invenuon provides compounds of Formulas (IV) and (V)
Figure imgf000012_0001
wherein R, -R„ are as defined above Preferablv R -R* are each independently a of hvdrogen. methyl, ethyl, propyl isopropy l. butyl isobuty l h drow l. amino. niuo. nitroso. carboxy . tπfluoromethy l. phenoxy and benzy oxy
Prefeπed embodiments of the present invenuon include compounds wherein X and Z are oxygen and Y is nitrogen The prefeπed forms of the following specific embodiments include, but are not limited to. compounds wherein X and Z are oxygen and Y is nitrogen
Preferred embodiments of the present invenuon include compounds wherein each of W is -CN. m and n are zero and p is one Of these, further prefeπed embodiments include compounds where T is -CH;-. Z and X are oxygen. Y is N and R, to R- are hvdrogen Preferably each of T is -CH;-. Z and X are oxygen and Y is N
Further prefeπed embodiments of the present invenuon include compounds where each of W is - CN(R9)2). preferably W is -N(R9)2 m and n are zero and p is one X is preferably oxygen or a bond. Z is oxygen and Y is nitrogen Of these, prefeπed embodiments include those compounds where each R, is independently selected from hydrogen, methyl, ethyl, prop l. isopropyl. butyl, isobutyl. pentyl. lsopenryl. methoxy. ethoxy or amino or where the R9 subsutuents combine with die N to form a 5- or 6- membered subsututed or unsubstituted heterocycloalkyl. opuonally subsituted with an addiuonal ox gen or nitrogen, or combine with the N to form a 5- or 6- membered substituted or unsubstituted heteroary 1 The substituUons of these prefeπed embodiments preferably including methyl, ethyl, propyl. isopropyl butvl. isobutyl. methylester. etliylester. benzyl, phenyl. benzoxy, phenoxy. phenoxy carbonyl. one or two addiuonal heterocycloalk ls or heteroar l πngs and/or one or two fused benzene πngs Prefeπed forms of this embodiment include compounds where T is absent. -CH2- or - CH2-CH2- CH;S. CH:SCH2. CH;SCH;CH2. methoxy or phenoxy methyl Particularly prefeπed forms of this embodiment include compounds wherein R0 subsutuents combine, with die N of W. to form a
Figure imgf000013_0001
Figure imgf000013_0002
/ group, which may be optionally subsututed by die above-noted substitutions, such as. for example, pyπdyl. benzyl, phenyl methyl ester or eth l ester, and/or fused with 1-2 additional benzene πngs
In a further prefeπed embodiment of the present mv ention. each of W is -N(R9)2. m is one and n is one. X is o.xy gen or a bond. Z is oxygen and Y is mtrogen Of these, prefered embodiments include those compounde where each R9 is independently selected from hydrogen, methyl, ethyl, propyl. isopropyl. butyl, isobutyl. mehanol. ethanol. methox . ethoxy or amino or w here the R9 substituents combine with the N to form a 5- or 6- membered substituted or unsubstituted heterocy cloalky 1. optionally subsituted with an additional oxygen or mtrogen. or combine with the N to form a 5- or 6- membered subsututed or unsubstituted heteroary 1 The subsututions of these prefeπed embodiments preferably including methyl, ethyl, propyl. isopropyl. butyl, isobutyl. methyl ester, ethyl ester, benzyl, phenyl. benzoxy . phenoxy. phenoxycarbonyl and pyπdyi Prefeπed forms of this embodiment include compounds where T is absent. -CH2- or -CH2-CH:- Particularly prefeπed forms of this embodiment includes compounds w herein RQ substituents form with the N of W to form a
Figure imgf000013_0003
group, which may be optionally subsututed bv pvπd l. benz l or phenyl and or fused widi 1-2 additional benzene πngs R, -R- are preferably hydrogen
Other prefeπed embodiments of die present inv enuon includes compounds wherein W is eidier - P(0)2-OR9 or -P(0)(OR9)2, m is zero and p is one Of these, each R9 is preferably independendy hydrogen, methyl, ethyl, propyl. isopropyl. butyl or isobutyl Preferably, in these embodiments. Y is nitrogen. Z is oxygen and X is oxygen Preferably, tn is zero and when n is one, R,6 is preferably hydrogen T is preferably -CH2- or - CH2CH;- in these prefeπed embodiments
Further prefeπed embodiments of the present invenuon include compounds wherein W is either - S(0)2-R9, -S(0)2-OR9 or -S(0)2NR9, m is zero and p is 1 Of these, prefeπed embodiments include where R9 is hydrogen, or subsututed or unsubstituted meth l, ethyl, propyl. isopropyl. butyl or isobutyl. wherein die optional subsutution is a 5- or 6- membered cycloalk l. opuonally subsituted with at least one oxygen or nitrogen or a 5- or 6- membered heteroaryl Further prefeπed forms of these embodiments include compounds where X is oxygen or a bond. Z is oxygen. T is absent or -CH2-. R,-R- are h drogen. Y is nitrogen and R is hydrogen. methyl, ethyl, propyl. isopropyl. but l, isobutyl. benz l, benzylcarbonyl. phenyl. V
Figure imgf000014_0001
Preferably . R9 is hydrogen, methyl, ethyl, propyl. isopropyl. butyl, isobutyl. benzyl, or benzy carbony 1
Further prefeπed embodiments of the present invenuon include compounds wherein W is either -C(O)- R9 or -C(0)N(R9)2, and m is zero Preferably. n=l when W is C(0)R9 Of these, prefeπed embodiments include compounds where each R9 is independendy hydrogen, methyl, ethyl, propyl. isopropyl. butyl, isobutyl. hydroxyl. methanol. ethanol. -(CH(OQ))pCOOH (where Q is hydrogen, hydroxyl. ethoxy or methoxy). or amino. and T is (-CH;-)ι.j. wherein optionally one of the methen l units is replace with phenoxy. carbonyl or oxygen Prefered forms of these embodiments include compounds where X is ox gen or a bond. Z is oxygen. Y is mtrogen and R| to R- are hydrogen R|6, when present, is preferably hvdrogen
Further prefeπεd embodiments of the present invention include compounds wherein W is an optionally subsututed 5- or 6- membered c cloalky l opuonally containing at least one heteroatom selected from S. O or N. or a heteroaryl wherein the cycloalkyl or heteranl ma\ be subsututed or attached to a further 5- or 6- membered cycloalkyl which may optionallv contain at least one heteroatom. a 5- or 6- membered heteraryl or a 5- or 6- membered aryl
Preferabl . the compounds of the inv ention exhibit an IC50 for inhibiting PARP in vitro, as measured by the methods descπbed herein, of about 20 μM or less preferablv less than about lOμM. more preferably less than about 1 μM. most preferably less than about 0 1 μM
Prefeπed embodiments of the present in ention include the following compounds, and neutral forms thereof, where appropπate
Figure imgf000014_0002
Figure imgf000014_0003
- 1.
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0003
10
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000019_0002
io
Figure imgf000019_0003
Figure imgf000019_0004
Figure imgf000020_0001
Broadly , the compounds and composiuons of the present invenuon can be used to treat or prevent cell damage or death due to necrosis or apoptosis. cerebral ischemia and reperfusion injury or neurodegenerative diseases in an animal, such as a human The compounds and compositions of the present inv enuon can be used to extend the hfespan and proliferauve capacitv of cells and thus can be used to treat or prevent diseases associated therewith, they alter gene expression of senescent cells, and thev radiosensitize hvpoxic tumor cells Preferabl . die compounds and compositons of the inv ention can be used to treat or prevent tissue damage resulung from cell damage or death due to necrosis or apoptosis. and/or effect neuronal activ ity either mediated or not mediated by NMDA toxicity The compounds of the present invention are not limited to being useful in treaung glutamate mediated neurotoxicity and/or NO-mediated biological pathways Further, the compounds of the invention can be used to treat or pre ent other tissue damage related to PARP activation, as descπbed herein The present invenuon provides compounds which inhibit the in vitro and/or m vivo polymerase activity of poly (ADP-πbose) polymerase (PARP). and compositions containing the disclosed compounds
The present invention provides methods to inhibit, limit and or control the in vitro and/or in vivo polymerase activitv of poly (ADP-πbose) polymerase (P.ARP) in any of solutions, cells, tissues, organs or organ systems In one embodiment, the present mv ention provides methods of limiting or inhibiting PARP activity in a mammal, such as a human, either locally or systemically
The present invenuon provides methods to treat and/or prevent diseases, syndromes and/or condiuons exacerbated by or involving die increased generation of PARP These methods involve application or admimstration of the compounds of the present invenuon to cells, tissues, organs or organ systems of a person in need of such treatment or prevention
In one embodiment, the present invention provides mediods to treat and/or prevent cardiovascular tissue damage resulting from cardiac ischemia or reperfusion injury Reperfusion injury, for instance, occurs at the termination of cardiac bypass procedures or duπng cardiac anest w hen the heart, once prevented from receiving blood, begins to reperfuse and these methods involve administration of the compounds and compositions of the present invenuon preferably pπor to. or immediately subsequent to reperfusion. such that reperfusion injury is prevented, treated or reduced The present invention also provides methods of preventing and/or treating vascular stroke, cardiov ascular disorders
In another embodiment, the present inv ention prov ides in vitro or in v ivo methods to extend or increase the hfespan and or proliferation capacitv of cells and thus also methods to treat and/or prevent diseases associated therewith and induced or exacerbated bv cellular senescence including skin aging, atherosclerosis, osteoartliπtis. osteoporosis, muscular d strophy degenerauve diseases of skeletal muscle involving rephcative senescence, age-related muscular degeneration, immune senescence. AIDS and other immune senescence diseases, and other diseases associated with cellular senescence and aging, as well as to alter the gene expression of senescent cells In a further embodiment, the present invention provides methods of treating or preventing or ameliorating the effect of cancer and or to radiosensitize hvpoxic tumor cells to render the tumor cells more susceptible to radiation therapy and therebv to prevent the tumor cells from recoveπng from potentially lethal damage of DNA after radiation therapy A method of this embodiment is directed to specificallv and preferential lv radiosensitizing tumor cells rendenng the tumor cells more susceptible to radiation therapy than non-tumor cells
In yet another embodiment the present in ention provides methods of preventing and/or treating vascular stroke, cardiovascular disorders, to treat other conditions and/or disorders such as age-related muscular degeneration. AIDS and other immune senescence diseases, arthπtis. atherosclerosis, cachexia. cancer, degenerauve diseases of skeletal muscle mv oh ing rephcativ e senescence, diabetes, head trauma, spinal chord injury , immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis. hemoπhagic shock, splanchnic arten occlusion shock, multiple organ failure (such as involving am of the kidney , liver, renal, pulmonary , retianl. pancreauc and/or skeletal muscles systems), acute autoimmune thv roidius. muscular d strophy osteoaπhπtis. osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia septic shock (such as endotoxic shock), local and/or remote entothelial cell dysfunction (such are recognized bv endo-dependent relaxant responses and up-regulauon of adhesion molecules), inflammauon and skin aging
In one embodiment of the present invenuon. a person diagnosed with acute retinal ischemia or acute vascular stroke is immediately administered parenterally. either by intermittent or conunuous intravenous administration, a compound of any of formulas I. II. III. IV or V either as a single dose or a seπes of divided doses of the compound After this iniual treatment, and depending on the person's presenting neurological symptoms, die person optionally may receive the same or a different compound of the invention in the form of another parenteral dose. The compound of die inv ention can be admnistered by intermittent or continuous administration via implantation of a biocompatible. biodegradable polymeπc matπ.x delivery system contaimng a compound of formula I. II. III. IV or V. or via a subdural pump inserted to administer the compound direcdy to die infarct area of the brain In a further embodiment, die present in enuon provides methods to extend the Iifespan and proliferauve capacity of cells, such as. for example, in using the compounds of the invenuon as general mediators in the generation of oxidants. proinflammatory mediators and/or cytokines. and/or general mediators of leukocyte infiltration, calcium ion ov erload, phospholipid pero.xidaion. impaired nitπc oxide metabolism and/or reduced ATP producuon
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 Shows the distπbuuon of the cross-secuonal infarct area at representauve lev els along the rostrocaudal axis, as measured from the interaural line in non-treated animals and in animals treated with 10 mg/kg of 3.4-dιhydro-5-l- (l-pιpeπdιnyl)-botoxyll -1 (2H)-ιsoquιnolιnone
Figure 2 Shows the effect of intrapeπtoneal administration of 3.4-dιhydro-5-l-( l -pιpeπdιny l )-butoxy]- l(2H)-ιsoquιnolιnone on the infarct volume
DETAILED DESCRIPTION OF THE INVENTION The present invention peπains to compounds, pharmaceuucal compositions containing the same. methods of using the same, and process of making die same, wherein such compounds are useful as inhibitors of poly( ADP-πbose) polymerase (P.ARP) As such, thev treat or prevent neural ussue damage resulting from cell damage or death due to necrosis or apoptosis. cerebral ischemia and reperfusion injury or neurodegenerative diseases in an animal, they extend die Iifespan and proliferauve capacity of cells and thus can be used to treat or prevent diseases associated therewith, they alter gene expression of senescent cells, and thev radiosensiuze hvpoxic tumor cells Preferabl . die compounds of the invention treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis. and or effect neuronal activity, either mediated or not mediated by NMDA toxicity These compounds are diought to interfere with more than the glutamate neurotoxicity and NO-mediated biological pathways Further, the compounds of the invention can treat or prevent other tissue damage related to P.ARP activ auon
For example, the compounds of die invenuon can treat or prevent cardiovascular tissue damage resulting from cardiac ischemia or reperfusion injury Reperfusion injury, for instance, occurs at the terminauoπ of cardiac bypass procedures or duπng cardiac anest w hen the heart, once prevented from receiving blood, begins to reperfuse. The compounds of the present invention can also be used to extend or increase the Iifespan or proliferation of cells and thus to treat or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis. osteoarthπUs. osteoporosis, muscular d strophy, degenerative diseases of skeletal muscle inv olving replicauve senescence, age-related muscular degeneration, immune senescence. AIDS and odier immune senescence diseases, and other diseases associated with cellular senescence and aging, as well as to alter the gene expression of senescent cells These compounds can also be used to treat cancer and to radiosensitize hvpoxic tumor cells to render die tumor cells more susceptible to radiation therapy and to prevent the tumor cells from recoveπng from potentially lethal damage of DNA after radiation dierapv presumably by their ability to prevent DNA repair The compounds of the present invention can be used 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, arthπtis. atherosclerosis, cachexia. cancer, degenerative diseases of skeletal muscle involving replicative senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as coliUs and Crohn's disease), muscular dystrophy, osteoarthπus. osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging
Preferably the compounds of the inv ention act as PARP inhibitors to treat or prevent tissue damage resulting from cell death or damage due to necrosis or apoptosis to treat or prevent neural tissue damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in an animal, to extend and increase the Iifespan and proliferauve capacity of cells, to alter gene expression of senescent cells, and to radiosensitize tumor cells
Another especiallv prefeπed embodiment of the invention is a pharmaceutical composition which compπses d ) a therapeuticallv effective amount of the compound of formula I. II or III. and (n) a pharmaceuticallv acceptable earner
As used herein, "alk l" means a branched or unbranched saturated hvdrocarbon chain compnsing a designated number of carbon atoms For example. Cι-C6 straight or branched alkyl hydrocarbon chain contains 1 to 6 carbon atoms, and includes but is not limited to substituents such as methyl, ethyl, propyl. iso-propyl. butyl, iso-butyl. tert-butyl. n-penty l. n-hexyl. and the like, unless othenvise indicated
"Alkenyl" means a branched or unbranched unsaturated hydrocarbon chain compnsing a designated number of carbon atoms For example. Cz-C6 straight or branched alkenyl hydrocarbon chain contains 2 to 6 carbon atoms having at least one double bond, and includes but is not limited to substituents such as ethenyl. propenyl. isopropenyl. butenvl. iso-buten l. teπ-butenv l. n-pentenyl. n-hexenyl. and the like, unless otherwise indicated
"Alkox ". means the group -OR wherein R is alkyl as herein defined Preferably. R is a branched or unbranched saturated hydrocarbon chain contaimng 1 to 6 carbon atoms
"Cyclo". used herein as a prefix, refers to a structure characterized by a closed πng "Halo" means at least one fluoro. chloro. bromo, or lodo moiety, unless otherwise indicated. "Ammo" compounds include amme (NH2) as well as substituted ammo groups compπsing alkyls of one through six carbons
"Ar". "aryl" or "heteroaryl" means a moiety which is substituted or unsubstituted. especially a cyclic or fused cyclic πng and includes a mono- bi-. or tπcychc. carbo- or heterocyclic πng. wherein die πng is either unsubsututed or substituted in one to five posιuon(s) with halo, haloalkyl. hydroxyl. nitro. tπfluoromethy 1. Cι-C6 straight or branched chain alkyl. C2-C« straight or branched chain alkenyl. Cι-C6 alkoxy. C:-C6 alken lo.x , phenoxy. benzyloxy. amino. duocarbonyl. ester, thioester. cyano. tmino. alky amino. aminoalkyl. sulfhydryl.
. it . thioalkyl and sulfon l. wherein the individual πng sizes are preferably 5-8 members, wherein the heterocyclic πng contains 1-4 heteroatom(s) selected from the group consisting of O. N. or S. wherein aromatic or tertiary alkyl amines are optionally oxidized to a conesponding N-o\ιde Heteroaryls may be attached to other πngs or subsututed through die heteroatom and/or carbon atom of the πng Paπicularl prefeπed aryl or heteroaryl moieties include but are not limited to phen l. benz l naphthyl. pynolyl. pynolidinvl. pyπdmyl. pv nmidinvl. puπnyl. qumolinyl. lsoqu olinyl. fury l. thiophenyl. lmidazolyl. oxazolyl thiazolyl. pyrazolyl. and thienyl
"Pheny l" includes all possible isomeπc phen l radicals, optionally monosubstituted or multi-substituted with subsutuents selected from the group consisung of ammo tπfluoromethyl. Ci-C* straight or branched chain alkyl. G-Cβ straight or branched chain alkenvl. carbonyl. thiocarbonyl. ester, thioester alkoxy. alkenoxy. cyano. nitro. lmino. alkyiamino aminoalkyl. sulfhydryl thioalkyl. sulfonyl. hydroxy. halo, haloalkyl. NR2 wherein R2 is selected from the group consisting of hydrogen. (C!-C6)-straιght or branched chain alkyl. (C3-C6) straight or branched chain alkenv l or alk nyl. and (Cι-C ) bπdging alk l wherein said bπdging alkyl forms a heterocyclic πng starting with the nitrogen of NR, and ending with one of the carbon atoms of said alkyl or alkenv l chain, and wherein said heterocyciic nng is optionalh fused to an AT group Cycloalky l optionalh containing at least one heteroatom includes saturated C3-C8 πngs. preferably C, or C„ πngs herein at 1-4 heteroatoms selected from 0. N or S mav be optionalh substituted for a carbon atom of the πng C cloalk ls optionally containing at least one heteroatom. as descπbed above, may be subsututed by or fused to at least one 5 or 6 membered ar l or heteroar l Prefered cycloalkvls containing a heteroatom include pynolidinvl. lmidazolidmyl. pyrazohdin l. pipeπdinvl. piperazinyl. morpholino and thiomorpholino The compounds of the present invention possess one or more asymmetπc center(s) and thus can be produced as mixtures (racemic and non-racemic) of stereoisomers. or as individual enantiomers or diastereo ers The individual stereoisomers may be obtained by using an optically acuve starting mateπal. by resolving a racemic or non-racemic mixture of an intermediate at some appropπate stage of the svnthesis. or by resolution of the compound of anv of formulas I. II III IVand V It is understood that the indiv ldual stereoisomers as well as mixtures (racemic and non — racemic) of stereoisomers are encompassed by the scope of the present invenuon The S-stereoisomer at atom 1 of formula (I) is most prefened due to its greater acuvity "Isomers" are different compounds that have the same molecular formula and includes cyclic isomers such as (ιso)ιndole and odier isomeπc forms of cyclic moieues "Stereoisomers" are isomers that differ only in the way the atoms are aπanged in space "EnanUomers" are a pair of stereoisomers that are non-supeπmposable minor images of each other "Diastereoisomers" are stereoisomers which are not minor images of each other "Racemic mixture" means a mixture containing equal parts of individual enanuomers "Non-racemic mixture" is a mixture containing unequal parts of individual enantiomers or stereoisomers
The compounds of the invention are useful in a free base form, m the form of pharmaceutically acceptable salts, phaπnaceuticall acceptable h drates, pharmaceuucally acceptable esters, pharmaceutically acceptable solvates. pharmaceutically acceptable prodrugs. pharmaceutically acceptable metabolites and in the form of pharmaceutically acceptable stereoisomers These forms are all within the scope of the invention In practice the use of these forms amounts to use of the neutral compound
"Pharmaceutically acceptable salt", "hydrate' . "ester" or "solvate" refers to a salt, hydrate, ester, or solvate of the inventive compounds which possesses the desired pharmacological activ ity and which is neither biologicallv nor othenvise undesirable Orgamc acids can be used to produce salts, hydrates, esters, or solvates such as acetate, adipate. alginate. aspartate. benzoate. benzenesulfonate. p-toluenesulfoπate. bisulfate. sulfamate, sulfate naphth late butv rate, citrate camphorate. camphorsulfonate cyclopentane-propionate. digluconate. dodecy Isulfate ethanesulfonate. fumarate. glucoheptanoate. glvccrophosphate. hemisulfate heptanoate. hexanoate. 2-hydroxy ethanesulfonate. lactate. maleate. methanesulfonate. 2-naphthalenesulfoπate. mcotinate. oxalate tosvlate and undecanoate Inorganic acids can be used to produce salts, hydrates, esters, or solvates such as hydrochloπde. hydrobromide. hvdroiodide. and thiocyanate
Examples of suitable base salts, hvdrates. esters, or solvates include hydroxides, carbonates, and bicarbonates of ammonia, alkali metal salts such as sodium, lithium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts aluminum salts, and zinc salts Salts hvdrates. esters or solvates mav also be formed with organic bases Organic bases suitable for the formation of pharmaceuticalh acceptable base addition salts, h drates esters, or solvates of the compounds of the present invention include those that are non-toxic and strong enough to form such salts, hydrates, esters, or solvates For purposes of illustration, the class of such organic bases mav include mono- di-. and tπalkylamines. such as methylamine. dimeth laπune. tπethv lamine and dicyclohexylamine. mono-, di- or tπhydrox alkylamines. such as mono-, di- and tπethanolamine. amino acids, such as arginine and lysine. guanidine. N-methyl-glucosamine N-methv l-giucamine. L-glutamine. N-methyl-piperazine orpholine. ethylenediamine. N-benzv 1-phenethy lamine. (tnhydroxy-methv l)amιnoethane. and the like See. for example, "Pharmaceutical Salts." J Pharm Sci . 66 1. 1 -19 ( 1977) Accordingl . basic nitrogen-containing groups can be quaternized with agents including lower alk l halides such as meth l ethyl, prop l. and butyl chloπdes. bromides and iodides, dialkyl sulfates such as dimethyl, diedi l. dibutyl and diamyl sulfates. long chain halides such as dec l. lauryl. myπstyl and stea l chloπdes. bromides and iodides, and aralkyl halides such as benzyl and phenethyl bromides
The acid addition salts, hydrates, esters, or solvates of the basic compounds may be prepared either by dissolving the free base of a PARP inhibitor of the present in ention in an aqueous or an aqueous alcohol solution or other suitable solvent containing the appropπate acid or base, and isolaung the salt by evaporating the solution Alternatively the free base of the PARP inhibitor of the present invention can be reacted with an acid, as well as reacting die PARP inhibitor having an acid group thereon ith a base, such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained bv concentrating the solution "Pharmaceuticalh acceptable prodnig" refers to a deπvative of the inventive compounds which undergoes biotransformauon pπor to exhibiting its pharmacological effect(s) The prodnig is foπnulated with the objectιve(s) of improved chemical stability , improved pauent acceptance and compliance improved bioavailabihtv prolonged duration of action improved organ selecuvity improved formulauon (c g . increased hydrosolubiliry). and/or decreased side effects (e g . toxicity) The prodnig can be readily prepared from the invenuve compounds using methods known in die art. such as those descπbed by Burger's Medicinal Chemistry and Drug Chemistry. Fifth Ed . Vol 1. pp 172-178. 949-982 ( 1995) For example, the invenuve compounds can be transformed into prodrugs by convening one or more of the hydroxy or carboxy groups into esters 'Pharmaceutically acceptable metabolite" refers to drugs that have undergone a metabolic transformation After entry into the bod\. most drugs are substrates for chemical reactions that may change their ph sical properties and biologic effects These metabolic conversions, which usually affect the polaπty of the compound, alter the way in which drugs are distπbuted in and excreted from the body However, in some cases. metabolism of a drug is required for dierapeuuc effect For example, anucancer drugs of the anumetabolite class must be convened to dieir active forms after they have been transported into a cancer cell Since most drugs undergo metabolic uansformation of some kind the biochemical reactions that play a role in drug metabolism may be numerous and diverse The main sue of drug metabolism is the liver, although other tissues may also participate The term "neurodegenerau e diseases" includes Alzheimer's disease. Parkinson's disease and
Huntington's disease
The term "ne ous insult" refers to any damage to nenous tissue and am disability or death resulting therefrom The cause of nen'ous insult may be metabolic, to ic, neurotoxic. latrogenic. thermal or chemical, and includes without limitation, ischemia, h poxia. cerebrovascular accident, trauma, surgery, pressure, mass effect. hemmonhage. radiation vasospasm. neurodegenerauve disease, infection. Parkinson's disease, amyotrophic lateral sclerosis (ALS). m ehnation/demvelination process, epilepsy, cognitive disorder, glutamate abnormality and secondary effects thereof
The term "neuroprotective" refers to the effect of reducing, aπestmg or amelioraung nenous insult, and protecung. resuscitating, or reviving ne ous tissue that has suffered nen'ous insult The term "prev enting neurodegenerauon" includes the ability to prevent neurodegenerauon in patients diagnosed as having a neurodegenerativ e disease or who are at nsk of developing a neurodegenerativ e disease The term also encompasses preventing further neurodegenerauon in patients who are already suffeπng from or have symptoms of a neurodegenerauve disease The term "treating" refers to (1) preventing a disease, disorder or condiuon from occumng in an animal that may be predisposed to the disease, disorder and/or condition, but has not yet been diagnosed as having it. (n) inhibiting the disease, disorder or condition. 1 e , anesting its development, and
(in) relieving the disease, disorder or condition, i.e . causing regression of the disease, disorder and/or condiuon The term "neural tissue damage resulting from ischemia and reperfusion injury and neurodegenerauve diseases" includes neurotoxicity. such as seen in vascular stroke and global and focal ischemia A feature characteπsuc of many of these transformations is that the metabolic products are more polar than the parent drugs, although a polar drug does sometimes ield a less polar product Substances with high hpid water partition coefficients, which pass easily across membranes, also diffuse back readily from tubular uπne through the renal tubular cells into the plasma Thus, such substances tend to have a low renal clearance and a long persistence in the body If a drug is metabolized to a more polar compound, one with a lower partition coefficient, its tubular reabsorption will be gready reduced Moreover, the specific secretory mechanisms for anions and cauons in the proximal renal tubules and in the parenchymal liver cells operate upon highly polar substances
As a specific example, phenaceun (acetophenetidin) and acetanihde are both mild analgesic and antipyretic agents but are each transformed within the body to a more polar and more effective metabolite, p- hydroxyacetani d (acetaminophen), which is widely used today When a dose of acetanilid is given to a person, the successive metabolites peak and decay in the plasma sequentially Duπng the first hour, acetanilid is the pπncipal plasma component In the second hour, as the acetanilid level falls, the metabolite acetaminophen concentrauon reaches a peak Finall . after a few hours, the pπncipal plasma component is a further metabolite that is inert and can be excreted from the body Thus, die plasma concentrations of one or more metabolites, as well as the drug itself, can be pharmacologicalh important
The reactions involv ed in drug metabolism are often classified into two groups, as shown in the Table II. Phase I (or funcuonahzauon) reactions generalh consist of ( 1 ) o idative and reducuve reactions that alter and create new functional groups and (2) hydrohlic reactions that cleave esters and amides to release masked functional groups These changes are usuallv in the direction of increased polaπty
Phase II reactions are conjugation reacuons in which the drug, or often a metabolite of the drug, is coupled to an endogenous substrate, such as glucuromc acid, acetic acid, or sulfuπc acid
TABLE II Phase I Reactions (functionalization reactions) ( 1 ) Oxidation via the hepatic microsomal P450 system
Aliphauc oxidauon
Aromauc hydroxy lauon
N-Dealky lauon
O-Dealkylauon S-Dealkylauon
Epoxidauon
Oxidative deaminauon
Sulfoxide formauon
DesulfuraUon N-Oxidation and N-hy droxy lauon
Dehalogenation
(2) Oxidauon via nonmicrosomal mechanisms
Alcohol and aldehyde oxidation Puπne oxidation Oxidative deaminauon (monoamine oxidase and diamine oxidase)
(3) Reducuon Azo and mtro reducuon (4) Hydrolysis
Ester and amide hydrolysis Peptide bond hydrolysis Epoxide hvdrauon
Phase II Reactions (coniugauon reactions)
(1) Glucuronidauon
(2) Acetylauon
(3) Mercaptuπc acid formation
(4) Sulfate conjuga on
(5) N-. 0-. and S-methylauon
(6) Trans-sulfuration
The compounds of the present in enuon exhibit pharmacological activity and are. therefore, useful as pharmaceuucals In particular, the compounds exhibit central nenous and cardiac vesicular system acuvity
It is understood that tautomeπc forms, when possible, are included m the invenuon For example, the tautomeπc forms of the following compounds are exemplary
Figure imgf000028_0001
Figure imgf000028_0002
Many of the PARP inhibitors are known and. thus, can be synthesized by known methods from starting mateπals that are known, may be available commercially, or may be prepared by methods used to prepare conesponding compounds in the literature See. for example. Suto et al . "Dihydroiso-qumolinones The
Design and Synthesis of a New Seπes of Potent Inhibitors of Poly( ADP-πbose) Polymerase", Anucancer Drug
Des , 6 107-17 (1991), which discloses processes for synthesizing a number of different PARP inhibitors Typically, the PARP inhibitors used in the composiuon of the invenuon will have an IC50 for inhibiung poly( ADP-πbose) polymerase in vitro of about 20 μM or less, preferably less than about lOμM. more preferably less than about 1 μM, most preferably less than about 0 1 μM
The PARP inhibitor 3.4-dιhydro-5-[4-(l-pιpeπdιnyl)butoxy]-l(2H)-ιsoquιnolmone. for example, has been reported to inhibit PARP with an IC50 of 40 nM by Suto et al . cited above There are muluple routes which may be undertaken to prepare the compounds of the present invenuon Two of these routes for the preparation of the xanthene den auves of this invention are demonstrated below by schemes 1-3 and 4-7
The xanthene πng may be geneπcally subsututed as set forth in formula I Such xanthene starting denvauves are known in die chemistry literary and are accessible by processes known to one skilled in the art The process sequence set forth herein does not present an exact sequence of reacuons by which the compound must be made, that is. die sequence of reacuons can be rearranged in several ways to reach the target molecule
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The .xanthene and thioxanthene denvauves of this invenuon are represented by previously defined formulas I-V As an example, the xandiene denvauves of this invenuon can be prepared in a conventional manner as illustrated below by Schemes 1 - 5 The xanthene πng may be geneπcally subsututed as set forth in formula I Such xanthene starting denvauves are known in the chemistn literature and accessible by processes known to one skilled in the an The process sequence set forth herein does not present an exact sequence of reacuons by w hich the compound must be made, that is. the sequence of reacuons can be reananged in several ways to reach the target molecule Scheme 1 below illustrates schematically the preparauon of compounds Example 1 through Examples 4
Scheme 1
O 01/16137
Figure imgf000030_0001
Step 1
9-Amιnomehty lxanthene 2 is available by reduction of 9-carboxamιde 1 using sodium boronhvdnde in dioxane Other reducuon methods can be subsututed include lithium aluminum hydnde and other boronhydndes The solvent can also be vaπed. DMSO. tetrahy drofuran. dieth lether and other organic solvent can be used The temperature of the reaction is general between 0 C and 200 C For example. 9- aπunomethy lxanthene can be made as follows To a suned suspension of sodium boronhvdnde (1 89g. 50 mmol) and 9-\anthenecarboxamιde (2 25g 10 mmol) in dioxane (20mL) is added acetic acid (3 Og. 50 mmol) in dioxane (lOmL) over a peπod of 10 minutes at 10 °C The reacuon mixture is suned at reflux for 2 hours and concentrated to dryness in vacuo The excess reducu e reagent is decomposed with water and the mixture is extracted with chloroform The chloroform layer is washed with water, dπed over anhvdrous sodium sulfate and evaporated in vacuo The residue is purified by silica gel column chromatography (ethylacetate methanol, 9 1 as eluant) to give a white solid (1 6 g, 7 6 mmol) in 76 2% yield
Step 2
The 9-ιsocyanomehrvlxanthenes 3 is obtained by condensauon of the amino group idi phosgene in a heated solution of toluene Odier solvents, such as p-nitrobenzene. can also be used The new formed isocyano functionality senres as an electroplule for lntermoiecular Fπedle-Crafts reaction in next step Other fiincuonaliues include N-carbonv liπudazole. N-carbony Ibenzotπazole and N-ethylfbrmate can also be applied this type of reactions These functionalities can be formed by reacuons of the 9-amιnomethylxanthenes 2 with carbonyldiimidazole. carbonyldibenzotnazole and ethyl chloroformate respecuvely Step ?
The desired xanthane product. 3.4-dιhvdrobenzopyrano[4.3.2-de]ιsoquιnoh-l-one 4 can be obtained by an intramolecular Fπedle-Crafts acylation using acid as catal st Zinc chloπde. aluminum chloπde. titanium (IV) chloπde. hydrochloπc acid or aceuc acid may be used, but polyphosphoπc acid is often prefened for tlus type of intramolecular cycloadditions The temperature can vary from 0 - 150 °C For example. 3,4- dihydrobenzop rano[4.3.2-de]ιsoquιnolιn-l-one 4 can be prepared as follows To a liquid of 12 g of polyphosphoπc acid in a 500 mL beaker is added 9-xanthenemethylιsocyanιde 3 (2 37 g. 10 mmol) portion wise with manual sumng at 90 °C The mixture is suned for three minutes and then 100 g of more polyphosphoπc acid is added Vigorous stirπng is applied for four minutes when temperature is kept at 90 °C The mixture is allowed to cool to 60 °C and 40 g of crushed ice is added until the polyphophoπc acid is completely hvdrolyzed and a brown solid is separated The solid is collected by vacuum filtration and dien recrystal zed in chloroform to afford a unsaturated product (0 15 g. 0 63 mmol) in 6 3% yield
The solid is dissolved in glacial aceuc acid (100 mL) and the solution is placed in a hydrogenauon bomb Palladium ( 10% on carbon. 500 mg) is added The bomb is set pressure at 2000 psi The mixture of the content is suned for 20 hours under the pressure and poured dirough a fluted filter paper to remove the catalyst The solvent of the filtrate is removed in vacuo to giv e a yellow solid, which is recrystalhzed in chloroform to afford desired product (0 95 g. 4 0 mmol) in 80 % yield
Monosulfonation of 3.4-dιhydrob nzopyrano[4.3.2-de]ιsoquιnolι-l-one 4 can be achieved using chorosulfonic acid neat to give compound 5 The temperature can vary from -20 to 100 °C Amidation of compound 5 can be earned out bv a v aπety of conditions known to those skilled in the art. including reacuon widi first or secondan amines using py πdme or tπethy 1 amine as base Typical solvents include chloπnated solvents vaπous ethers, and dipolar aprotic solv ents like DMF Thus final products of compounds 6 can be prepared from 3.4-dιhydrobenzopyrano[4.3.2-de]ιsoquιnolι-l-one 4 using General procedure A and B Scheme 2 below illustrates schemaucallv the preparauon of compounds Example 5 through Example 9
Scheme 2
Figure imgf000032_0001
C
Figure imgf000032_0002
Figure imgf000032_0003
Example 5 - 9
Step 4
Preparauon of 3-phenoxyphthalonιtπle 7 The starting mateπals 3-nιtrophthalonιuιle 7 and or p- subsututed pheno ide 8 are either readily available from commercial sources or can be prepared by known methods by those skilled in the art The desired compound can be easily obtained by coupling the orthodinitπle with the phenoxide salt in dipolar aprotic solvent, such as DMF. low dialkyl ketones. lower alkyl nitπles
Temperature range can be 0 - 150 C For example, a mixture of 0 1 Mol of 3-nιtrophdιalomtπle and 0 14 Mol of sodium phenoxide (or p-substituted phenoxide) in 120 ml of dry DMF is mechanicalh suned for 2 5 hours at room temperature and then poured into water The precipitate is collected by filtrauon. washed with water, and dπed under vacuum to give 19 g (0 086 Mol. 86%) of die compound 9. mp 110-112 C
Step 5
Preparauon of 9-oxoxanthene-l -carboxylic acid 10 The formauon of orthocarboxy lie acid groups from the cyano groups can be achieved by hydrolysis of the aryl nitnle with either mineral acids, include sulfuπc acids and h drochloπc acids, or strong inorganic base These conditions are understood by those skilled in the art. An intramolecular Fπedle-Crafts acylation can occur as soon as die diacids are formed to afford 9-oxoxanthene-I- carboxylic acid 10 For example, a soluuon of 0 1 Mol of 3-pheπoxyphfhalonιtπle is dissolved in 150 ml of a mixture of 60 per cent sulfunc acid and glacial acid The mixture is refluxed for 6 hours and then poured into 1000 mL of ice-cold water pH value of the suspension is adjusted to 10 with sodium carbonate Impuπties are extracted out by paruuoning of the basified soluuon idi ethyl acetate Precipitauon is formed upon adding hydrogen chloπde (to pH5) to the aqueous soluuon. and collected b filtrauon to give a designed acid in 70% yield
Step 6
Preparauon of 9-oxoxanthene-l -carboxy lie acid methyl ester 11
Estenficauon of acid can be achieved by several convenuonal methods by those skilled in the art These procedures include uulizmg diazomethane or methy 1 alcohol catalyzed by mineral acids For example, ethereal diazomethane soluuon can be prepared from N-methyl-N-nitrosotolueπe-p-sulphonamide and potassium hydroxide from a water bath 9-Oxoxanthene-l -carboxylic acid (1 29g, 5 mmol) in absolute methanol is added to this ethereal diazomethane soluuon at 0 °C unul a pale yellow color formed Removal of the solvent provides the desired ester solid in 100% yield Thus the final products. Examples 5 - 9, can be prepared by condensauon hydrazine with the ketonester 11 as descπbed in General procedure C
Scheme 3 below illustrates schemaucally die preparauon of compounds Example 10 through Example 11
Scheme 3
General Procedure C
NH '2,INnH2,
Figure imgf000033_0002
Figure imgf000033_0001
General Procedure F
Example 10 -11
RCOOR
Figure imgf000033_0003
Step 7
Preparauon of 7-amιno-9-oxoxanthene-l -carboxylic acid methyl ester 12 Reducuon of the niuo group can be earned out under catalytic hvdrogenauon condiuons uulizmg metal catalysts descπbed in aldπchimica Acta 1979 12. or with hydrazine, metal hydπde reducing agents such as NaBILi. B2H0. and the like Suitable solvents include water, lower alcohol, ethers. THF. lower alkyl organic acids such as aceuc acid Temperature can vary from 0 - 100 °C Desired final products. Examples 11 -12. can be prepared by condensing the ketonester 12 with hydrazine using General procedure C. followed by an amide bond formation of the phthalazine deπvative 13 using General procedure F
Scheme 4 below illustrates schemaucally die preparation of Example 12 through Examples 24 Scheme 4
Figure imgf000034_0001
11 14
Example 12 - 24
Figure imgf000034_0002
Step 9
Preparauon of 7-bromomethv l-9-oxoxanthene-l -carboxy lie acid methyl ester 14 BrominaUng agents include N-bromosuccinimide. bromine, complexed bromine such as pyndinium bromide, perbromide. and the like can be used to convert 7-methyl-9-oxoxanthene-l-carbox hc acid methyl ester 11 to 7-bromomethy 1-9- oxoxanthene- 1 -carbox he acid meth l ester 14 Solvents include chloπnated hydrocarbones. dipolar aprouc solvents, and vaπous ethers Temperature can range from 0 - 100 °C For example, a suspension of 7-methyl- 9-oxo.xandιene- 1 -carboxylic acid methyl ester (0 1 Mol). N-bromosuccinimide (0 12 Mol) and benzoylperoxide
(10 mg) in dry carbon tetrachlonde (300 mL) is suned at 60 °C for 6 hours The mixture is filtered, the solid is washed successively with small amounts of chloroform, water and ether, and then dπed to leave a desired product as white solid
Displacement of the bromo group of compound 14 widi nucleophiles such as amine or alcohol, thiol using General procedure D or E provides the compound 15 The ketonester 15 can be cyc zed with hydrazine using General procedure C to give desired final products. Example 12 - 24 Scheme 5 below illustrates schemaucally the preparauon of Example 26 dirough Example 29
33 -
SUBSTITUTE SHEET (RULE 26 Scheme 5
Figure imgf000035_0002
Monosulfonauon of the ketone ester 11 to gi e compound 16 can be achieved using neat chorosulfonic acid to react with the substrate 11 The temperature can vary from -20 to 100 °C Amidation of compound 16 can be earned out by a variety of condiuons known to those skilled in the art. including reacuon with first or secondan amines using pyπdine or tnedivl amine as base Typical solvents include chloπnated solvents, vaπous ethers, and dipolar aprotic soiv ents like DMF Thus final product of compounds can be prepared from compound 17 and hydrazine as descπbed in General procedure C
General procedure A
To a liquid of chlorosulfo c acid ( 10 mL) is added the compound 5 or ketone ester (11. 7 87 mmol) under niuogen at 0 C The resulung mixture is allowed to warm to room temperature, conunuoush suned for about 2 da s and poured onto 100 g of ice The residue is collected by filtrauon and washed widi water, ethanol to afford the product 5 or 16 as solid in > 80% of yield
General procedure B
To a suspension of the sulfonyl chloπde 5 or (16. 5 67 mmol) in methylene chloπde (100 mL) is added tneth 1 amine ( 1 6 mL) and amine compound (5 mmol) under nitrogen at 0 C The reacuon mixture is allowed to warm to room temperature, suned conunuoush for 1 hour and poured into 100 mL of water The organic layer is collected, washed with water, bπne and concentrated in vacuo The product is puπfied via crystallizauon or silica column chromatographv using methanol/meth lene chlonde as eluent to afford solid product in 60 - 90% of yield General procedure C
A benzopyrano[4 3 2-de]phthalazιne πng can be formed by condensation of the ketone ester with hydrazine To a solution of the ketonester 11 or 15 ( 17 5 mmol) in absolute ethanol ( 10 mL) is added anhvdrous hvdrazine tn ethanol ( 1 mL) drop wise at room temperature The soluuon is refluxed for overnight and cooled to room temperature Ice-cold water (100 mL) is added and brown solid is separated The solid is collected by vacuum filtration and washed with water small amount of methanol to give a solid product in 10 - 80% of vιeld
General procedure D
To a solution of the bro o compound (14. 10 mmol) in dry DMF ( 100 mL) is added potassium carbonate (100 mmol) and secondan amine (10 mmol) The reaction mixture is heated to 70 C for 6 hours and cooled to 0 C Water (100 mL) is added to the reacuon mixture followed by ethvl acetate (200 mL) The organic laver is collected washed with water bπne and dπed over sodium sulfate The solvent is removed in vacuo The residue is punfied bv column chromatographv on silica gel using ethvl acetate /hexane as eluent to gi e die product 15 in 60 - 90% of vield
General procedure E
To a suned solution of the bromo compound 14 (10 mmol) and phenol deπvative or duol deπvative (10 mmol) in acetone ( 150 mL) is added potassium carbonate (100 mmol) The reaction mixture is heated to reflux for 6 hours The solvent is then removed in vacuo. and water (100 mL) and ethv lacetate ( 100 mL) are added to die residue The organic laver is collected, washed with water bnne and dπed over sodium sulfate The solvent is removed in vacuo The residue is punfied bv column chromatographv on silica gel using ethvl acetate /hexane as eluent to give the product 15 in 60 -
Figure imgf000036_0001
General procedure F
To a suspension of compound phthalazine deπvaUve 13 (10 mmol) in dn DMF (20 mL) is added succinic or aceuc anhydπde (11 mmol) The mixture is heated to 1 10 C for 3 hours and cooled to room temperature The mixture is conunuoush suned for 2 hours after addiUon of THF (100 mL) The solid is collected by filtrauon. washed with methanol to afford a desired product without further puπficauon
Other manners vaπations or sequences of prepaπng the compounds of Formula I will be readily apparent to those skilled in the art
The compounds of formula I may be useful in the free base form, in the foπn of base salts where possible, and in the form of addiUon salts as ell as in the free acid form All these forms are within die scope of this invention In practice use of the salt form amounts to use of die base form Pharmaceuticalh acceptable salts within die scope of this invention are those deπved from mineral acids such as hydrochloπc acid and sulfuπc acid, and organic acids such as ethanesulfomc acid, benzenesulfonic acid, p-toluenesulfomc acid, and the like, giv ing the hydrochloπde. sulfonate. ethanesulfonate. benzenesulfonate. p-toluenesulfonate. and the like respectiv eh . or those deπved from bases such as suitable organic and inorganic bases Examples of pharmaceuucally acceptable base addiuon salts with compounds of the present mvenUon include orgamc bases which are nontoxic and strong enough to form such salts These organic bases and the use thereof are readily understood by those skilled in the art Merely for the purpose of lllustrauon. such organic bases may include mono-, di- and tπalkylaπunes. such as methylamine. diethylamine and uiethylamine. mono-, di-. or tnhydroxyalk lamines such as mono-, di-. and tπethanolamine. amino acids such as arginine, and lysine, guanidine N-methylglucosamine. N-methylgiucamine. L-glutamine. N-methylpiperazine. morphohne, ethylenedianane N-benzylphenethylamine, tns(hydroxymethvl)anunoethane. and the like
The acid addition salts of the basic compounds may he prepared by dissolving the free base of the compound of formula I in aqueous or aqueous alcohol soluuon or other suitable solvents containing the appropπate acid or base and isolaung the salt by evaporaung the solution, or by reacting the free base of the compound of formula I with an acid as well as reacung the compound of formula I having an acid group thereon with a base such that the reactions are in an orgamc solvent, m which case the salt separates direcdy or can be obtained by concentration of die soluuon
The compounds of this invenuon contain one or more asymmetπc carbon atoms Therefore, the invention includes the individual stereoisomers and mixtures thereof as well as the racemic compounds The individual isomers may be prepared or isolated by methods known in the art
The compounds of the invenuon exhibit pharmacological activity and are. therefore, useful as pharmaceuticals In particular the compounds exhibit central nervous and cardiac vesicular system activity
Example 1
H O N
l o
OH
To a soluuon of 0 3g of compound 4 in 7 5 mL of concentrate sulfuπc acid (98%) is dropwise-added 2.0 mL of fuming sulfuπc acid at 0 °C The mixture is suned for 3 hours at 0 °C and poured into 100 mL of ice- cold water The precipitation is collected bv filtration, washed with water to give 0 17 g of a white solid, mp
>300 (dec ) [H-NMR (400 MHz. DMSO-d6) 8 20(d. 1H. 4Hz). 7 62(d. 1H, 8Hz). 7 53(s. 1H). 7 51(d. 1H, 8Hz). 7 41(t. 1H. 8Hz). 7 30(d. 1H. 8 0Hz). 7 09(d. 1H. 8 0Hz). 4 50-4 55(dd. 1H. 8Hz. 12Hz), 4 02-3 96(m, 1H). 3 46(t. 1H. 12Hz) Example 2
Prepared from compound 5 and N-(2-aminoethyl)pipeπdine according to General Procedure B.
Purification of compound by crystallizadon in water-acetone gives a white solid, mp 265 -269 °C. Η-NMR (400 MHz. DMSO-d6): 8.35(m, IH). 7.82 (s. IH). 7.75 (m. IH). 7.67 (m. IH), 7.46 - 7.43 (m. IH). 7.37 (m, IH). 4.58 - 4.54 (m. IH), 4 10 (m. IH). 3.53 - 3.47 (m. IH). 3.39 (m. 2H), 3.06 (bs. 2H). 3.02 (bs. 2H). 2.32 (s. 2H). 1.44 - 1.19 (m. 6H).
Example . '
o o s
Prepared from compound 5 and N-methy lpiperazine according to General Procedure B. Purificadon of compound by crystallizadon in aceuc acid gives a white solid, mp 300 °C (dec). ^H-NMR (400 MHz. CDCI3): 7.78-7.84 (d. J = 8 Hz. 1 H), 7.58-7.64 (d. J = 8 Hz. 1 H), 7.53 (s. 1 H). 7.32-7.40 (t. J = 8, 16 Hz. 1 H). 7.16- 7.26 (m. 2 H). 6.30 (br s. 1 H). 4.36-4.50 (q. J = 6. 14. 20 Hz. 1 H). 4.02-4.12 (m. 1 H). 3.60-3.74 (t. I = 12. 24 Hz. 1 H). 3.01 (br s. 4 H). 2.46 (br s. 4 H). 2.24. (s. 3 H).
Example 4
Figure imgf000038_0001
Prepared from compound 5 and N-acetylpiperazine according to General Procedure B. Purificadon of compound by crystallizadon in aceuc acid gives a white solid (45% yield), mp 290 °C (dec). ^-NMR (400
MHz. CDCI3): 7.88-7.93 (d. J = 8 Hz. 1 H), 7.67-7.72 (dd, J = 4, 12 Hz. 1 H), 7.62 (s. 1 H). 7.43-7.49 (t. J = 8, 16 Hz. 1 H). 7.27-7.36 (m. 2 H), 4.54-4.62 (dd. J = 8. 20 Hz. 1 H). 4.15-4.25 (m. 1 H). 3.70-3.85 (m. 3 H), 3.59(s. 2 H). 3.04 (s. 4 H). 2.06 (s. 3 H). Example 5
Figure imgf000039_0001
Prepared from the compound 11 (R = Me) and hydrazine according to General Procedure C
Puπficauon of compound by crystalhzauon in ethanol gives a whit flake solid (44 % yield), mp 332 - 334 °C -NMR (400 MHz. DMSO-dό) 12 5 (s. br, IH), 7 90 - 7 87 (tn. 3H). 7 69 (dd. J= 7 2. 1 9 Hz. IH. 7 38 (dd. J= 8 8. 1 9 Hz. IH), 7 31 (d, J = 8 4 Hz. IH). 2 39 (s. 3H)
Example 6
Figure imgf000039_0002
Prepared from the compound 11 (R = F) and hydrazine according to General Procedure C Puπficauon of compound by crystalhzauon in ethanol gives a whit solid (20 % yield), mp 355 °C (dec ) lH-NMR (400 MHz. DMSO-d6) 12 7(s. br. IH). 8 42 (d. 1=2 OHz. IH). 7 91 (m. 2H). 7 73(dd. J=2 0. 6 8 Hz. IH), 7 41(m, 2H)
Example 7
Figure imgf000040_0001
Prepared from the compound 11 (R = t-Bu) and hydrazine according to General Procedure C Puπfication of compound by crystalhzauon ethanol gives a solid (30 % yield), mp 380 °C (dec ) H-NMR (400 MHz. DMSO-d6) 8 07 (d. J = 2 5 Hz. IH), 7 91 (dd. J = 7 6, 7 0 Hz, IH), 7 89 (d. J = 7 6 Hz. IH), 7 72 ( dd. J = 7 0 2 0 Hz. IH). 7 65 (dd. J = 9 0. 2 5 Hz. IH), 7 37 (d. I = 9 0 Hz. IH), 1 38 (s. 9H)
Example 8
Figure imgf000040_0002
Prepared from the compound 11 (R = NH2) and hydrazine according to General Procedure C Puπficauon of compound by crystalhzauon in ethanol gives a yellowish solid (60 % yield), mp 300 °C (dec.) -NMR (400 MHz. DMSO-dό) 5 29-5 31 (d. 2H. J=7 8 Hz). 6 81-6 83 (dd. IH. J=8 8. 2 4 Hz), 7 14-7 16 (d. IH. J=8 8 Hz). 7 26-7 27 (d. IH. J=2 2 Hz). 7 63-7 65 (d. IH. =7 3 Hz). 7 83-7 89 (dd. J=7 5. 15 5 Hz). 7 85 (s. IH,). 12 52(s. br. IH)
Example 9
Figure imgf000040_0003
Prepared from the compound 11 (R = NO2) and 1 5 equivalent of hydrazine according to General Procedure C Puπficauon of compound by crystalhzauon in acetic acid gives a yellowish solid (20 % yield), mp > 300 °C (dec ) L H-NMR (400 MHz. DMSO-d6) 7 46-7 49 (d. IH. J=9 2Hz,). 7 75-7 77 (d. IH. J=8 1Hz), 7 97-8 01 (dd IH, J=8 1 Hz) 8 13-8 15 (d. IH. J=7 9 Hz). 8 38-8 42 (dd, IH. J=9 5 Hz), 9 07-9 08 (d. IH, =2 6 Hz)
Example 10
Figure imgf000041_0001
Prepared from the amino compound 13 and aceuc anhvdπde according to General Procedure F Purification of the compound by crystalhzauon in ethanol to gives a yellow solid in 80 % yield, mp 350 °C (dec ) [ H-NMR (400 MHz. DMSO-C-6) 2 08 (s. IH) 7 36-7 38 (d IH J=8 9). 7 66-7 70 (dd, IH. 1=2 6. 16 1
Hz). 7 68-7 69 (d IH J=2 2 Hz). 7 88-7 91 (d. IH. J=8 6 Hz). π 89 (s. IH), 8 49 (d. IH. J=2 5 Hz), 10 15 (s IH. NHCO). 12 62 (s IH)
Example 1 1
Figure imgf000041_0002
Prepared from the ammo compound 13 and succinic anhvdπde according to General Procedure F Purification of the compound by crystalhzauon in ethanol to gives a yellow solid in 20 % yield, mp 320 °C (dec ) *H-NMR (400 MHz. DMSO-d6) 2 50-2 60 (m. 4H), 7 45-7 47 (d. IH. J=8 9 Hz), 7 74-7 77 (dd. IH.
J=9 2, 2 5 Hz). 7 76-7 79 (dd, 1R J=7 2, 2 1). 7 96-7 98 (d. IH. J=9 3 Hz), 7 97 (s. IH), 8 56-8 57 (d. IH. J=2 5 Hz) 10 29(s. IH). 12 72 (s. IH)
Example 12
Figure imgf000042_0001
Prepared from die compound 14 and cysteamine according to General Procedure E Purification of compound by cr stalhzauon in ethanol gives white solid product in 30 % of yield, mp 255 - 275 °C (methylsulfonic acid salt) -NMR (400 MHz. DMSO-d6) 2 29 (s. 3H), 2 64 (t. 2H, J = 7 3 Hz), 3 01 (br s. 2H). 3 92 (s. 2H). 7 42 (d. IH. J = 8 5 Hz). 7 54 - 7 57 (dd. IH. J = 2 1 Hz, 8 6 Hz), 7 71 - 7 76 (m, 4 H). 7 89 7 95 (m, 2H). 8 0 (d. IH. J = 2 lHz). 12 69 (s. IH)
Example 13
Figure imgf000042_0002
Prepared from the compound 14 and 4-mercaptop πdιne according to General Procedure E PunficaUon of compound by crystallization in ethanol gives white solid product in 38 % of yield, mp 237 - 240 °C ^H- NMR (400 MHz. DMSO-d6) 12 68 (s. IH). 8 61 (m. 2H). 8 24 (d. IH. J = 2 2 Hz), 7 92 - 7 85 (m. 4H). 7 70 (m. 2H), 7 44 (d. IH. J = 8 5 Hz. 4 70 (s. 2H). 2 30 (s. 3H)
Example 14
Figure imgf000042_0003
Prepared from the compound 14 and hydrazine according to General Procedure D Punficauon of compound by crystalhzauon in aceuc acid gives a yellow solid product in 78 % of yield, Mp > 300 °C ^H-NMR (400 MHz. DMSO-d6) 12 64 (s. br. IH). 8 04 (d. J = 2 Hz. IH), 7 93-7 88 (m. 2H), 7.71 (dd. J = 1 9 Hz and 7 3 Hz. IH). 7 51 (dd. J = 2 1 Hz and 8 5 Hz. IH). 7 37 (d. J = 8 4 Hz, IH). 3 82 (s. 2H)
Example 15
Figure imgf000043_0001
Prepared from die compound 14 and potassium c anide according to General Procedure D Puπficauon of compound by crystalhzauon in aceuc acid gives a light yellow solid product in 70 % of yield, mp > 300 °C -NMR (400 MHz. DMSO-dό) 12 70 (s. IH. NH). 8 10 (d. J = 2 0 Hz. IH), 7 91 (m. 2H). 7 72 (dd. J = 1 8. 7.2 Hz. IH). 7 53 (dd. J = 2.2. 8 6 Hz. IH). 7 46 (d. J = 8.6 Hz. IH). 4 18 (s. 2H)
Example 16
Figure imgf000043_0002
Prepared from the compound 14 and 1-phenylριperazme according to General Procedure D Puπficauon of compound by crystalhzauon m ethanol gives a white solid product in 89 % of yield, mp > 300 °C (dec). -NMR (400 MHz. DMSO-d6). 12.63 (s. br. IH). 8 06 (d, J = 1.9 Hz. IH), 7.90 (m. 2H). 7 72 (dd. J = 7 1 and 1 9 Hz. IH), 7.53 (dd. J = 8 5 and 2.0 Hz, IH). 7.40 (d. J = 8.5. IH), 7.19 (m. 2H). 6 92 (d, I = 8 0 Hz. 2H), 6 76 (t. J = 7.3 Hz, IH). 3 61 (s. 2H), 3 14 (L J = 4.3 Hz. 2H), 2.56 (t I = 4.2 Hz, 2H).
Example 17
Figure imgf000044_0001
Prepared from the compound 14 and l-(2-pyπdyl)pιperazιne according to General Procedure D Punficauon of compound by crystallization in ethanol gives a white solid product in 89 % of yield, mp > 300 °C (dec ) lH-NMR (400 MHz. DMSO-dό) 12 64 (s. br. IH), 8 09 (m, 2H). 7 90 (m. 2H), 7 71 (dd. J = 7 2 and 1 8 Hz. IH). 7 52 (m, 2H), 7 40 (d, J = 8 5 Hz. IH). 6 80 (d. J = 8 6 Hz. IH). 6 62 (m. IH), 3 60 (s. 2H), 3 49 (s, 4H), 3 30 (s. 4H)
Example 18
Figure imgf000044_0002
Prepared from the compound 14 and pipeπd e according to General Procedure D Punficauon of compound by crystalhzauon m ethanol gives a white solid product in 69 % of yield, mp 195 - 200 °C ^H-NMR (400 MHz. DMSO-d6) 1 30 - 1 45 (m.l H). 1 53 - 1 75 (m. 3H). 1 75 - 1 90 (m. 2H). 2 35 (s. 3H), 2 82 - 3 0 (2 H). 3 30 - 3 4 (m. 2H), 4 39 (d. 2H. J = 5 2 Hz), 7 51 (d. IH. J = 8 5 Hz). 7 65 - 7 68 (dd. IH. I = 2 1 Hz. 8 5 Hz), 7 74 - 7 76 (m. IH), 7 91 - 7 96 (m. 2H). 8 29 (d. IH. J = 2 1 Hz). 9 33 (bs. IH), 12 79 (s. IH)
Example 19
Figure imgf000044_0003
Prepared from the compound 14 and N-methylbutylamine according to General Procedure D Punficauon of compound by crystalhzauon in ethanol gives a white solid product in 69 % of yield, mp 133 -135
°C !H-NMR (400 MHz. DMSO-d6) 0 91 (t. 3H, J = 7 4 Hz). 1 29 - 1 34 (m, 2H), I 65 - 1 70 (m. 2H). 2 35 (s. 3H). 2 69 (d. 3H. J = 4 8 Hz). 3 03 - 3 05 (m. IH). 3 14 (m. IH). 7 53 (d, IH. J = 8 5 Hz), 7 67 - 7 69 (dd, IH. J = 2 0 Hz. 8 5 Hz). 7 73 - 7 76 (m, IH), 7 91 - 7 96 (m. 2H). 8 30 (s. IH. J = 2 0 Hz), 9 50 (s. IH). 12 79 (s, IH)
Example 20
Figure imgf000045_0001
Prepared from the compound 14 and N-dimethylamine according to General Procedure D Puπficauon of compound by crystallizauon in ethanol gives a white solid product in 70 % of yield Η-NMR (400 MHz. DMSO-d6) 2 36 (s. 3H). 2 78 (d. 6H. J = 4 8 Hz). 4 40 (m. 2H). 7 53 (d. IH, J = 8 5 Hz). 7 65 - 7 70 (m. IH). 7 75 (m. IH). 7 92 - 7 95 (m. 2H). 8 28 (m. IH). 9 70 (s. IH). 12 78 (s. IH)
Example 21
Figure imgf000045_0002
Prepared from the compound 14 and N-methylbutylamine according to General Procedure E Puπficauon of compound by crystalhzauon m aceuc acid gives a gray solid product 59 % of yield, Mp >300 °C (dec ) -NMR (400 MHz, DMSO-d6) 12 67 (s. IH), 9 62 (s. IH), 8 17 (d. I = 2.0 Hz. IH), 7.90 (m, 2H), 7 80 (d, J = 8 8 Hz. 2H), 7 72 (dd, J = 1 7 and 7 2 Hz. IH), 7 64 (dd. I = 2 1 and 8 6 Hz. IH), 7 45 (d. J = 8 5 Hz. IH). 7 09 (d. J= 8 8 Hz. 2H), 5 27 (s. 2H), 4 42 (br s. 2H)
Example 22
Figure imgf000046_0001
Prepared from the compound 14 and 3-dιmedιylphenol according to General Procedure E Puπficauon of compound by crystallization in aceuc acid giv es a light yellow solid product in 50 % of yield, mp >300 °C (dec ) lH-NMR (400 MHz. DMSO-dό) 12 65 (s. br. IH). 8 16 (d. J = 2 0 Hz. IH), 7 91 (m. 2H), 7 72 (dd. I = 7 1 and 2 0 Hz. IH). 7 63 (dd. I = 8 6 and 2 1 Hz. IH). 7 44 (d. J = 8 5 Hz. IH), 7 07 (t. I = 7 5 Hz. IH), 6 33 (m. 3H). 5 17 (s. 2H). 2 87 (s, 6H)
Example 23
Figure imgf000046_0002
Prepared from the compound 14 and 4-(l-ιmιdazole)phenol according to General Procedure E
Purification of compound by crystalhzauon m ethanol gives a white solid product in 70 % of yield. Mp >300 °C (dec ) lH-NMR (400 MHz. DMSO-dό) 12 66 (s. br. IH). 8 18 (d. J = 2 Hz. IH), 7 91 (m. 2H). 7 73 (dd. J = 7 1 and 1 9 Hz. IH). 7 66 (dd. I = 8 5 and 2 1 Hz. IH). 7 48 (m. 3H). 7 25 (t_ J = 2 1 Hz. 2H). 7 12 (m. 2H). 6 22 (t. J = 2 1 Hz. 2H). 5 25 (s. 2H)
Example 24
Figure imgf000046_0003
Prepared from die compound 14 and 3-hydroxypyπdιne according to General Procedure E Punficauon of compound bv cr stallization in ethanol gives a yellow solid product in 57 % of yield. Mp >300 °C (dec ) 1H- NMR (400 MHz. DMSO-dό) 12 67 (s. br. IH). 8 39 (d. J = 2 9 Hz, IH), 8 19 (d. J = 2 8 Hz. 2H), 7 91 (m, 2H). 7 72 (dd. J = 7 1 and 2 0 Hz. IH). 7 66 (dd. J = 8 5 and 2 2 Hz, IH), 7 49 (m. 2H), 7 35 (m. IH). 5 30 (s. 2H)
Example 25
Figure imgf000047_0001
Prepared from the compound 11 (R = H) and 1 5 equivalent of hydrazine to give benzopyrano[4.3.2- delphthalazme according to General Procedure C To a soluuon of 0 3g of benzopyrano[4 3.2-de]phthaJazιne in
7 5 mL of concentrate sulfuπc acid is dropwise added 2 0 mL of fuming sulfunc acid at 0 °C The mixture is suned for 3 hours at 0 °C and poured into 100 mL of ice-cold water The precipitauon is collected bv filtrauon. washed with water to give of a white solid product, mp >300 °C (dec ) ΪH-NMR (400 MHz. DMSO-d6) 12 7 (s. 1 H). 8 35 (s. 1 H), 7 92 - 7 90 (m. 2 H). 7 76 - 7 71 (m. 2 H). 7 38 (d. 1 H)
Example 26
Figure imgf000047_0002
Prepared from the compound 16 and N-(2-aπunoeth l)morpholιne according to General Procedure B
Punficauon of compound by silica gel column chromatography provides sulfoπamide 17 Condensauon of compound 17 with hydrazine according to General Procedure C in ethanol affords a solid which is purified by crystalhzauon in ethanol gives a white solid product in 70 % of yield To this amine compound (10 mmol) in dioxane (10 mL) is added 10 mmol of hydrochloπc acid in dioxane at 60 °C White salt solid product is formed upon cooling the solution to room temperamre The solid is collected by filtrauon. mp 302 - 307 °C *H-NMR (400 MHz. DMSO-d6) 12 81 (s. IH). 10 90 (s. IH). 8 49 (d. IH. 2 3 Hz). 8 30 (s. IH), 7 99 - 7 92 (m, 3H). 7 78 - 7 75 (dd. IH. 2 6 Hz. 6 5 Hz), 7 65 (d. IH. 8 8 Hz). 3 95 - 3 92 (m. 2H). 3 78 - 3 72 (t. 2R 1 1 8 Hz). 3 43 • 3 39 (m 2H). 3 33 (s 4H). 3 10 - 3 08 (m. 2H)
Example 27
Figure imgf000048_0001
Prepared from the compound 16 and l-(2-amιnoethyl)pιpeπdιne according to General Procedure B Puπficauon of compound by silica gel column chromatography provides sulfonamide 17 Condensauon of compound 17 with hydrazine according to General Procedure C in ethanol-water affords a solid which is punfied by cn'stallizauon in ethanol gives a white solid product in 50 % of yield To this amine compound (10 mmol) in dioxane ( 10 mL) is added 11 mmol of hydrochloπc acid in dioxane at 60 °C White solid product is formed upon cooiing die soluuon to room temperature and collected by filtrauon mp > 300 °C (dec ) ^H-NMR (400 MHz. DMSO-d6) 12 82 (s. IH). 9 80 (s. IH). 8 49 (d. IH. 2 4 Hz). 8 26 (m. IH), 7 96-7 94 (m. 3 H). 7 78 (dd. IH. 2 6 Hz, 6 5 Hz). 7 67 (d. IH. 8 8 Hz). 3 20-3 17 (m. 2H). 3 13-3 12 (m. 2H). 2 89-2 87 (m. 2H). 1 75- 1 68 (m. 6H). 1 45-1 30 (m. 2H)
Example 28
Figure imgf000048_0002
Prepared from the compound 16 and N-methylpiperazine according to General Procedure B Punficauon of compound by silica gel column chromatography provides sulfonamide 17 Condensauon of compound 17 with hydrazine according to General Procedure C in ethanol affords a solid which is punfied by crystal zauon in ethanol gives a white solid product in 45 % of vield. Mp > 300 °C (dec ) Η-NMR (400 MHz, DMSO-d6) 12 82 (s. IH), 10 55 (s. IH). 8 36 (d. IH. 2 3 Hz), 7 96 - 7 91 (m. 3H), 7 79 (dd. IH, 2 7 Hz. 6 5 Hz). 7 71 (d. IH 8 7 Hz). 3 90 - 3 80 (bs 2H). 3 50 - 3 40 (bs. 2H), 3 20 - 3 10 (bs. 2H), 2 85 - 2 70 (m. 5H) Example 29
Figure imgf000049_0001
Prepared from the compound 16 and L-prohne according to General Procedure B Punficauon of compound by silica gel column cliromatography provides sulfonamide 17 The compound 17 is reacted with h drazine according to General Procedure C in ethanol affords a solid which is punfied by cr stalhzauon in ethanol gives a white solid product in 29 % of yield, mp > 300 (dec ) °C -NMR (400 MHz. DMSO-dό) 12 78 (s. IH). 8 41 (d. IH. J = 2 3 Hz), 7 99 - 7 92 (m. 3H). 7 79 (dd. IH. J = 5 6 Hz. 8 2 Hz), 7 63 (d. IH. J = 8 7 Hz). 4 14 (m. IH). 3 41 - 3 32 (m 2H), 1 99 - 1 90 (m. IH). 1 90 - 1 83 (m. 2H), 1 68 - 1 63 (m. IH)
Example 30
Figure imgf000049_0002
To a soluuon of compound 14 in m-xylene is added tπethyl phosphite the mixture is refluxed for 2 hours and cooled to room temperature The precipitauon is collected by filtrauon to give phosphonic ester Condensauon of this compound with hydrazine according to General Procedure C in ethanol affords a solid which is punfied by crystalhzauon in ethanol gives a hite solid product in 70 % of yield, Mp 196 -201 °C LRMS (fab. M+l) = 345 -NMR (400 MHz. DMSO-dό) 7 36 (t J = 7 8 Hz, IH), 7 23 (d, J = 7 6 Hz, IH), 7 08 (d. 7 7 Hz, IH). 6 99 (s. IH) 6 78 (d. J = 8 0 Hz. IH). 6 54 (d. J = 8 4 Hz. IH), 3 53 (d. J = 10 4 Hz. 3H), 2 82 (d. J = 20 1 Hz, 2H)
Example 31
Figure imgf000050_0001
Prepared from die compound 11 (R = COOMe) and hydrazine according to General Procedure C Punficauon of compound bv crystalhzauon in acetonitnle gives a whit solid (40 % yield), mp > 400 °C (dec ) ΪH-NMR (400 MHz. DMSO-d6) 12 73 (s br. IH). 9 99 (s. IH). 8 57 (d. J= 2 0 Hz. IH). 7 98 (dd. J= 5 5. 2 2 Hz. IH). 7 92 (m. 2H). 7 33 (dd. J= 6 5. 2 6 Hz. IH) ~ 47(d. J = 8 7 Hz. IH). 4 55 (s. 2H)
Other vaπations and modificauons of this invention using the synthetic pathwa s descπbed above will be obv lous to those skilled in the art
Methods of Usinjj the Compounds of the Inv ention
The compounds of the present inv ention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis. can ameliorate neural or cardiov ascular ussue damage, including that following focal ischemia, myocardial infarction, and reperfusion injury , can treat vanous diseases and conditions caused or exacerbated by PARP activ it . can extend or increase the Iifespan or proliferauve capacity of cells, can alter die gene expression of senescent cells, and can radiosensitize cells Generallv . inhibition of PARP acuvity spares the cells from energy loss, prev enung. in the case of neural cells, lπeversible depolaπzation of the neurons, and thus, provides neuroprotecuon While not being bound to any one particular theor . it is thought that PARP activation may play a common role in still other excitotoxic mechanisms, perhaps as yet undiscovered, in addition to the production of free radicals and NO
For the foregoing reasons, the present inv ention further relates to a method of administeπng a therapeuticallv effective amount of die above-identified compounds in an amount sufficient to inhibit PARP acuvit . to treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis. to effect a neuronal acuvity not mediated by NMDA toxicity . to effect a neuronal acuvity mediated by NMDA toxicit . to Ueat neural tissue damage resulung from ischemia and reperfusion injur . neurological disorders and neurodegenerauve 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, arthπtis. atherosclerosis, cache.xia. cancer, degenerative diseases of skeletal muscle involv ing replicative senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular d strophy, osteoarthnus. 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 die Iifespan and proliferauv e capacitv of cells, to alter gene expression of senescent cells, or to radiosensitize hvpoxic tumor cells The present inv enuon also relates to treaung diseases and condiuons in an animal which compπses administeπng to said animal a therapeutically effective amount of the above-identified compounds
In particular, the present invenuon relates to a method of treating, preventing or lnlubiung a neurological disorder in an animal, which compnses administenng to said animal a therapeutically effecuve amount of die above-identified compounds In a particularly prefened embodiment, the neurological disorder is selected from the group consisting of peπpheral neuropathy caused by physical injury or disease state, traumauc brain injury , physical damage to the spinal cord, s oke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demvehnating disease and neurological disorder relating to neurodegenerauon. Another prefeπed embodiment is when the reperfusion injury is a vascular stroke Yet another prefened embodiment is when the peπpheral neuropathy is caused by Guillain-Bane syndrome Sull another preferred embodiment is when the demvehnating disease and neurological disorder relates to neurodegenerauon. Another prefened embodiment is when the reperfusion injury is a vascular stroke Sull anodier prefened embodiment is when the demvehnating disease is mulUple sclerosis .Another prefened embodiment is when the neurological disorder relaung to neurodegenerauon is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease, and amyotrophic lateral sclerosis
Yet another prefened embodiment is a method of treating, preventing or inhibiting a cardiovascular disease in an animal, such as angina pectoπs. myocardial infarction, cardiovascular ischemia, and cardiovascular tissue damage related to PARP activation, bv administenng to said animal an effective amount of die compounds of die present invention
The present invention also contemplates the use of compound I. II. III. IV or V for inhibiting PARP acuvity . for treaung. preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis. for treating, preventing or inhibiung a neurological disorder in an animal
In a particularlv prefened embodiment, die neurological disorder is selected from the group consisung of peπpheral 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, demyelinaung disease and neurological disorder relaung to neurodegenerauon.
Another prefeπed embodiment is vv hen the reperfusion injury is a vascular stroke. Yet another prefened embodiment is when the peπpheral neuropathv is caused by Guillam-Bane syndrome Still another prefeπed embodiment is when the demyelinaung disease is multiple sclerosis Another prefened embodiment is when the neurological disorder relating to neurodegenerauon is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease, and am otrophic lateral sclerosis
The present invenuon also contemplates the use of compound I. II. III. IV or V in die preparauon of a medicament for the treatment of any of die diseases and disorders in an animal descπbed herein In a particular embodiment, the disease or disorder is a neurological disorder In a particularly prefeπed embodiment the neurological disorder is selected from die group consisung of peπpheral neuropathv caused b> phv sical injun or disease state, traumatic brain injury, physical damage to die spinal cord, stroke associated with brain damage, focal ischemia global ischemia, reperfusion injury, demyelinaung disease and neurological disorder relaung to neurodegenerauon Another prefened embodiment is when the reperfusion injury is a vascular stroke Yet another prefened embodiment is when the peπpheral neuropathy is caused by Guillain-Bane syndrome
Still another prefened embodiment is when die demyelinaung disease is multiple sclerosis Another prefened embodiment is when the neurological disorder relating to neurodegenerauon is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease and amyotrophic lateral sclerosis The term "preventing neurodegenerauon" includes the ability to prevent neurodegenerauon in pauents ne ly diagnosed as having a neurodegenerauve disease, or at πsk of dev eloping a new degenerative disease and for prev enting further neurodegenerauon in pauents w ho are already suffeπng from or have symptoms of a neurodegenerauve disease
The term "treatment" as used herein covers anv treatment of a disease and/or condiuon in an animal. particularlv a human, and includes
(0 preventing a disease and or condiuon from occurπng in a subject which may be predisposed to the disease and/or condition but has not yet been diagnosed as having it,
(n) mhibiung the disease and/or condition, l e . anesung its development, or (in) relieving the disease and/or condiuon. I e . causing regression of the disease and/or condition. As used herein, the term "neural tissue damage resulting from ischemia and reperfusion injury" includes neurotoxicity. such as seen in v ascular stroke and global and focal ischemia As used herein, the term "neurodegenerauve diseases." includes .Alzheimer's disease. Parkinson's disease and Huntington's disease
The term "ischemia" relates to localized tissue anemia due to obstruction of the inflow of arteπal blood Global ischemia occurs under conditions in w hich blood flow to the entire brain ceases for a peπod of time, such as may result from cardiac anest Focal ischemia occurs under conditions in which a portion of the brain is depπved of its normal blood supply, such as mav result from diromboemboiy ic occlusion of a cerebral vessel, traumauc head injury, edema, and brain tumors
The term "cardiovascular disease" relates to mvocardial infarcuon. angina pectoπs. vascular or myocardial ischemia, and related conditions as would be known by those of skill in the art which involve dysfuncuon of or tissue damage to the heart or v asculature. and especiall . but not limited to. Ussue damage related to PARP activation
The term '"radiosensitizer"' as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeuticallv effectiv e amounts to increase die sensiuvity of the cells to be radiosensiuzed to electromagnetic radiauon and or to promote the treatment of diseases which are treatable with electromagnetic radiation Diseases which are ueatable with electromagnetic radiauon include neoplasUc diseases, benign and malignant tumors and cancerous cells Electromagnetic radiation treatment of other diseases not listed herein are also contemplated bv the present invention. The terms " electromagnetic radiauon" and ""radiation"* as used herein includes, but is not limited to. radiation having the wavelength of 10":o to 10° meters Prefened embodiments of die present inv enuon employ the electromagnetic radiauon of gamma- radiation ( 10':o to IO'13 m) x-ray radiauon ( 10'" to 10'' m). ultraviolet light ( 10 nm to 400 nm). visible light (400 nm to 700 nm). infrared radiation (700 nm to 1.0 mm), or microwave radiation (1 mm to 30 cm).
Compositions and Methods for Effecting Neuronal Activity
Preferably, die compounds of the invention inhibit PARP activity and. thus, are believed to be useful for treating neural Ussue damage, particularly damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in animals The term "nenous tissue" refers to the vaπous components that make up the nen'ous system including, without limitation, neurons, neural support cells, glia. Schwann cells, vasculature contained within and supplying these structures, the central nen-ous system, die brain, the brain stem, the spinal cord, the junction of the central nenous system with the peπpheral nen'ous system, the peπpheral nen'ous system, and allied structures. Further, according to the invenuon. an effecuve dierapeuuc amount of the compounds and compositions descπbed above are administered to animals to effect a neuronal activity, particularly one that is not mediated by NMDA neurotoxicity. Such neuronal acuvity may consist of sumulauon of damaged neurons, promouon of neuronal regeneration, prevention of neurodegenerauon and treatment of a neurological disorder. Accordingly, the present invenuon further relates to a method of effecung a neuronal activity in an animal, compπsing administenng an effective amount of the compound of formula I. II . III. IV or V to said animal.
Examples of neurological disorders that are treatable by the method of using the present invenuon incluc s. without limitation, tπgeminal neuralgia: glossopharyngeal neuralgia; Bell's Palsy; myasthenia gravis; muscular dystrophy, amyotrophic lateral sclerosis: progressive muscular atroph . progressive bulbar inherited muscular atrophy: herniated. ruptured or prolapsed invertebrate disk syndromes: ce cal spondylosis: plexus disorders: thoracic outlet destruction syndromes, peπpheral neuropathies such as those caused by lead, dapsone, ticks, porphyria. or Guillain-Baπe syndrome: Alzheimer's disease: Hundngton's Disease and Parkinson's disease.
The method of the present invenuon is particularly useful for treaung a neurological disorder selected from the group consisung of peπpheral neuropathy caused by physical injury or disease state, head trauma, such as traumatic brain injury; physical damage to the spinal cord: suoke associated with brain damage, such as vascular stroke associated with hypoxia and brain damage, focal cerebral ischemia, global cerebral ischemia, and cerebral reperfusion injury; demyelinaung diseases, such as multiple sclerosis, and neurological disorders related to neurodegenerauon. such as .Alzheimer's Disease. Parkinson's Disease. Huntingdon's Disease and amyotrophic lateral sclerosis (ALS).
Treating Other PARP-Related Disorders The compounds, composiuons and methods of the present invention are particularly useful for treating or prevenung tissue damage resulting from cell deadi or damage due to necrosis or apoptosis
The compounds, compositions and methods of the invention can also be used to treat a cardiovascular disorder in an animal, bv administenng an effecuve amount of the compound of formula to the animal As used herein, the term "cardiovascular disorders" refers to those disorders that can either cause ischemia or are caused by reperfusion of the heart Examples include, but are not limited to coronary artery disease, angina pectoπs. myocardial infarction, cardiovascular tissue damage caused by cardiac anest. cardiovascular tissue damage caused bv cardiac bypass, cardiogenic shock, and related conditions that would be known by those of ordinary skill in the art or which involve dysfunction of or tissue damage to the heart or v asculature. especially , but not limited to. tissue damage related to P.ARP acuvation
For example, the methods of die invention are believed to be useful for treating cardiac ussue damage, particularly damage resulting from cardiac ischemia or caused by reperfusion injury m animals The methods of the invention are particularlv useful for ueating cardiovascular disorders selected from the group consisting of coronary arten disease such as atherosclerosis, angina pectons mvocardial infarction myocardial ischemia and cardiac anest. cardiac bvpass. and cardiogenic shock The methods of the invention are particularly helpful in treating the acute forms of the above cardiovascular disorders
Further, the methods of the inv ention can be used to treat tissue damage resulting from cell damage or death due to necrosis or apoptosis. neural ussue damage resulting from ischemia and reperfusion injur . neurological disorders and neurodegenerative diseases, to prevent or treat vascular stroke, to treat or prevent cardiovascular disorders, to ueat other conditions and or disorders such as age-related muscular degeneration. AIDS and other immune senescence diseases, arthπtis. atherosclerosis, cachexia. cancer, degenerative diseases of skeletal mus~.e involv ing replicativ e senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular d strophv osteoarthnus 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 Iifespan and proliferauve capacity of cells to alter gene expression of senescent cells, or to radiosensitize tumor cells
Further sull. the methods of the invenuon can be used to treat cancer and to radiosensitize tumor cells The term "cancer" is interpreted broadly The compounds of the present invention can be "anti-cancer agents", which term also encompasses "anu-tumor cell growth agents" and "anti-neoplastic agents" For example, the methods of the invenuon are useful for treaung cancers and radiosensiuzing tumor cells in cancers such as ACTH-producing tumors, acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma. endometnal cancer, esophageal cancer. Ewing's sarcoma, gallbladder cancer, hairy cell leukemia, head & neck cancer. Hodgkm's lymphoma. Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant peπtoneal effusion, malignant pleural effusion, melanoma, mesothelioma. multiple mveloma. neuroblastoma. non-Hodgkin's lymphoma ostcosarcoma. ovaπan cancer, ovan (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, retinoblastoma. skin cancer, soft-ussue sarcoma, squamous cell carcinomas stomach cancer, testicular cancer, thyroid cancer, trophoblasuc neoplasms uteπne cancer, vaginal cancer, cancer of the vulva and Wilm's tumor The teπn "radiosensiuzer". as used herein, is defined as a molecule, preferably a low molecular weight molecule administered to animals in dierapeuucallv effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagneuc radiation and/or to promote the treatment of diseases which are treatable with electromagnetic radiauon Diseases which are treatable with electromagnetic radiation include neoplasUc diseases, benign and malignant tumors, and cancerous cells Electromagnetic radiation treatment of other diseases not listed herein are also contemplated bv the present invention The terms "electromagnetic radiation" and "radiation" as used herein includes, but is not limited to. radiation having the wavelength of I0":n to 10° meters Prefened embodiments of the present inv enuon emplov the electromagneuc radiauon of gamma- radiauon ( 10":o to 10"13 m) x-ray radiauon ( 10 " to 10"" m). ultraviolet light ( 10 nm to 400 nm). visible light (400 nm to 700 nm) infrared radiation (700 nm to 1 0 mm), and microwave radiation (1 mm to 30 cm) Radiosensuizers are known to increase the sensitiv ity of cancerous cells to the toxic effects of electromagnetic radiation Sev eral mechanisms for the mode of action of radiosensuizers have been suggested in die literature including hvpoxic cell radiosensiuzers ( e g 2-nιtroιmιdazole compounds, and benzotπazine dioxide compounds) promote the reoxy genauon of hvpoxic tissue and/or catalyze the generation of damaging oxygen radicals, non-hypoxic cell radiosensiuzers (e g . halogenated p πmidines) can be analogs of DNA bases and preferential I v 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 vaπous other potential mechanisms of action have been hypothesized for radiosensiuzers in die treatment of disease
Many cancer treatment protocols cuπently emplov radiosensiuzers activ ated by the electromagnetic radiauon of x-rays Examples of x-rav activated radiosensiuzers include, but are not limited to. the following metronidazole. misonidazole. desmethylmisorudazole. pimonidazole. etanidazole. ni orazole. mitomycin C. RSU 1069. SR 4233. E09. RB 6145. nicotinamide. 5-bromodeoxyuπdιne (BUdR). 5-ιododeoxyuπdme (IUdR). bromodeoxycytidine. fluorodeoxyundine (FudR). hydroxyurea. cisplaun. and therapeuucally effective analogs and deπv atives of the same
Photodynamic therapy (PDT) of cancers emplov s visible light as the radiauon activator of the sensitizing agent Examples of photodynamic radiosensiuzers include the following, but are not limited to hematoporphyπn denvauves. Photofnn. benzoporphyπn denvauves. NPe6. Un etioporphyπn SnET2. pheoborbide-a. bacteπochlorophyll-a. naphthalocyanines. phthalocyamnes. zinc phdialocvanine. and therapeuucally effective analogs and denvauves of die same
Radiosensiuzers may be administered in conjunction with a therapeuticallv effective amount of one or more other compounds, including but not limited to compounds which promote the incoφoration of radiosensiuzers to the target cells, compounds which conuol the flow of therapeutics, nutnents. and/or oxygen to die target cells, chemotherapeutic agents which act on the tumor with or without addiuonal radiation, or other therapeutically effective compounds for treating cancer or other disease Examples of additional therapeutic agents that may be used in conjunction widi radiosensiuzers include, but are not limited to 5-fluorouracιl, leucovoπn. 5' -amιno-5'deoxythymιdιne. oxygen, carbogen. red cell transfusions, perfluorocarbons (e g . Fluosol- DA). 2.3-DPG. BW12C. calcium channel blockers. pentoxvfylhne. anuangiogenesis compounds, hydralazine, and LBSO Examples of chemotherapeutic agents that mav be used in conjunction with radiosensiuzers include, but are not limited to adnamycm. camptothecin. carboplatin. cisplaun. daunorubicin. docetaxel. doxorubicin. interferon (alpha, beta, gamma), interleukin 2. lπnotecan. pachtaxel. topotecan. and therapeutically effective analogs and denvauves of the same
Pharmaceutical Compositions of the Invention
The present invention also relates to a pharmaceutical composition compπsing (I) a therapeutically effective amount of the compound of formula I. II. III. IV or V and (n) a pharmaceutically acceptable earner The above discussion relating to the prefeπed embodiments' utility and administrauon of the compounds of the present invention also applies to the pharmaceutical composition of the present invenuon
The term "pharmaceutically acceptable earner" as used herein refers to any earner, diluent, excipient. suspending agent, lubπcating agent, adjuv ant. v elude, delivery system, emulsifier. disintegrant. absorbent, presenative. surfactant, colorant, flavorant. or sweetener
For these puφoses. the composiuon of the invention may be admi stered orally . parenterall . by inhalation spray , adsoφtion. absoφtion. topicallv . rectallv. nasally, bucally. vaginally. intraventncularly. via an implanted resen'oir in dosage formulauons containing conventional non-toxic pharmaceuucally -acceptable earners, or bv any other convenient dosage form The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intrapeπtoneal. intrathecal. lntraventπcular, intrasternal. and intracranial mjecUon or infusion techniques When administered parenterally. the composition will normallv be in a unit dosage, steπle injectable form (solution, suspension or emulsion) v hich is preferably isotomc with die blood of the recipient with a pharmaceuucally acceptable earner Examples of such steπle injectable forms are steπle injectable aqueous or oleaginous suspensions These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents The steπle injectable forms may also be steπle injectable solutions or suspensions in non-toxic parenterall -acceptable diluents or solvents, for example, as soluuons in 1.3-butanedιol Among the acceptable vehicles and solvents mat may be employed are water, saline. Ringer's solution, dextrose solution, isoto c sodium chloπde soluuon. and Hanks' solution In addiuon. steπle. fixed oils are conventionally employed as solvents or suspending mediums For this puφose. anv bland fixed oil may be emplo ed including synthetic mono- or di-glyceπdes. com. cottonseed, peanut, and sesame oil Fatty acids such as ethyl oleate. isoprop l mynstate. and oleic acid and its glycende denvauves. including olive oil
- 33 and castor oil especiallv in their polv oxyethylated versions are useful in the preparation of injectables These oil soluuons or suspensions may also contain long-chain alcohol diluents or dispersants
Steπle saline is a prefeπed earner, and the compounds are often sufficiendy water soluble to be made up as a solution for all foreseeable needs The earner mav contain minor amounts of addiuv es. such as substances that enhance solubility , lsotonicity. and chemical stabihtv . e g . anti-oxidants. buffers and presenatives
Formulations suitable for nasal or buccal admimstration (such as self-propelling powder dispensing formulations) may compπse about 0 1% to about 5% w/w . for example 1% w/w of active ingredient The formulations for human medical use of die present invention compπse an active ingredient in associauon with a pharmaceutically acceptable earner therefore and optionally other therapeutic mgredιent(s)
When administered orally, die composiUon will usually be formulated into unit dosage forms such as tablets, cachets powder, granules, beads, che able lozenges, capsules, liquids, aqueous suspensions or solutions, or similar dosage forms, using convenuonal equipment and techniques known in the art Such formulations typicallv include a solid, semisohd. or liquid earner Exemplary earners include lactose, dextrose, sucrose. sorbitol. mannitol starches, gum acacia calcium phosphate, mineral oil cocoa butter, oil of theobroma. alginates. tragacanth. gelatin, syrup, meth l cellulose, pol oxyethv lene sorbitan monolaurate. methyl hydroxvbenzoate. propyl hydroxybenzoate. talc, magnesium stearate. and the like
The composiUon of the mv enuon is preferably administered as a capsule or tablet containing a single or divided dose of the inhibitor Preferabl . the composition is administered as a steπle soluuon. suspension, or emulsion, in a single or div ided dose Tablets mav contain earners such as lactose and com starch, and/or
Iubπcating agents such as magnesium stearate Capsules may contain diluents including lactose and dπed com starch
A tablet may be made by compressing or molding the acuve ingredient optionally with one or more accessory ingredients Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubπcant. inert diluent, surface active, or dispersing agent Molded tablets may be made by molding in a suitable machine, a mixture of the powdered active ingredient and a suitable earner moistened with an inert liquid diluent
The compounds of this mv ention may also be administered rectallv in the form of suppositoπes These compositions can be prepared by mixing the drug with a suitable non-imtaUng excipient which is solid at room temperature, but liquid at rectal temperamre. and. therefore, will melt m the recmm to release the dmg Such mateπals include cocoa butter, beeswax, and pol ethylene gl cols
Composiuons and methods of die invention also may utilize controlled release technology Thus, for example, the mv entive compounds may be incoφorated into a hydrophobic pol mer matπx for controlled release over a peπod of days The composition of die mvention may then be molded into a solid implant, or externally applied patch, suitable for providing efficacious concentrations of the PARP inhibitors over a prolonged peπod of ume without the need for frequent re-dosing Such controlled release films are well known to the art Particularly prefeπed are uansdermal dehven systems Other examples of polymers commonly employed for this puφose that may be used in the present inv ention include nondegradable ethvlene-vinyl acetate copolymer an degradable lactic acid-glycohc acid copolymers which may be used externally or internally Certain hydrogels such as pol (hydroxy eth lmethacry late) or poly(vιnylalcohol) also may be useful, but for shorter release cycles than the other polymer release systems, such as those mentioned above
In a prefened embodiment, the earner is a solid biodegradable polymer or mixture of biodegradable pol mers with appropπate time release characteπsucs and release kinetics The composition of the invention may then be molded into a solid implant suitable for providing efficacious concentrations of the compounds of the invention over a prolonged peπod of time without the need for frequent re-dosing The composition of the present invention can be incoφorated into the biodegradable pol mer or polymer mixture in any suitable manner known to one of ordinary skill in the art and may form a homogeneous mauix with the biodegradable polymer, or may be encapsulated in some way within the polymer, or may be molded into a solid implant
In one embodiment, the biodegradable polvmer or polymer mixture is used to form a soft "depot" containing the pharmaceutical composiUon of the present invenuon that can be administered as a flowable liquid for example, by injection but which remains sufficiently v iscous to maintain the pharmaceuucal composition within the localized area around the injection site The degradation time of the depot so formed can be vaπed from several days to a few years, depending upon the polymer selected and its molecular weight By using a polvmer composiUon in injectable form, even the need to make an incision may be eliminated In any event, a flexible or flowable delivery "depot"" w ill adjust to the shape of the space it occupies w ith the body with a minimum of trauma to sunounding ussues The pharmaceutical composition of the present invention is used in amounts that are therapeuticallv effecuve. and mav depend upon the desired release profile, the concentration of die pharmaceutical composition required for the sensitizing effect, and the length of time that the pharmaceutical composition has to be released for treatment
The PARP inhibitors are used in the composiUon in amounts that are therapeutically effectiv e The compositions mav be steπhzed and/or contain adjuvants, such as presen ing. stabilizing welling, or emulsifying agents, soluuon promoters, salts for regulaUng the osmouc pressure, and/or buffers In addiuon they may also contain other therapeuucally valuable substances The compositions are prepared according to conventional mixing, granulaung. or coating methods, and contain about 0 1 to 75% by weight, preferably about 1 to 50% by weight, of the active ingredient To be effecuve dierapeuticall as central nervous system targets, die compounds of the present invenuon should readily penetrate die blood-brain barπer when peπpherally administered Compounds which cannot penetrate the blood-bram bamer can be effecuvelv administered by an intrav entπcular route or other appropnate delivery system suitable for admimstrauon to the brain
Doses of the compounds preferably include pharmaceutical dosage units compnsing an efficacious quantity of active compound By an efficacious quanuty is meant a quantity sufficient to inhibit PARP and deπve its beneficial effects through administration of one or more of the pharmaceutical dosage units Preferably the dose is sufficient to prevent or reduce the effects of vascular stroke or other neurodegenerauve diseases
For medical use. the amount required of the active ingredient to achieve a therapeuUc effect will vary with the particular compound, the route of admimstrauon, the mammal under treatment, and the particular disorder or disease being treated A suitable systematic dose of a compound of the present invenuon or a pharmacologically acceptable salt thereof for a mammal suffenng from, or likely to suffer from, any of condition as descπbed hereinbefore is in the range of about 0 1 mg/kg to about 100 mg/kg of the acuve ingredient compound the most prefeπed dosage being about 1 to about 10 mg kg
It is understood, however, mat a specific dose level for a particular patient will depend upon a vaπety of factors including the activ itv of the specific compound emplo ed, the age. body weight, general health, se . diet, time of admimstrauon. rate of excretion, drug combination, and the seventy of the particular disease being treated and form of admimstrauon
It is understood that the ordinaπlv skilled phvsician or vetennaπan will readily determine and prescπbe the effective amount of the compound for prophv lactic or therapeutic treatment of the condition for which ueatment is administered In so proceeding the phvsician or eteπnaπan could employ an intravenous bolus followed by an intravenous infusion and repeated admmistrauons parenterally or orally, as considered appropπate While it is possible for an active ingredient to be administered alone, it is preferable to present it as a formulation
When prepaπng dosage form incoφoraung the composiuons of die invention, the compounds may also be blended with convenuonal excipients such as binders, including gelatin, pregelaumzed starch, and the like. lubπcants. such as hydrogenated vegetable oil. steanc acid, and the like, diluents, such as lactose, mannose. and sucrose, disintegrants. such as carboxymethy lcellulose and sodium starch glycolate. suspending agents, such as povidone. polyvinyl alcohol, and the like, absorbants. such as silicon dioxide, preservatives, such as methy lparaben. propy lparaben. and sodium benzoate surfactants such as sodium laury 1 sulfate. polysorbate 80. and die like, colorants such as F D & C dves and lakes, flavorants and sweeteners
The present invention relates to the use of compounds I. II. . III. IV or V in the preparation of a medicament for the treatment of any disease or disorder in an animal descπbed herein
PARP Assays IC50
A convenient method to determine IC50 of a PARP inhibitor compound is a PARP assay using punfied recombinant human PARP from Trevigan (Gaithersburg. MD), as follows The PARP enzyme assay is set up on ice in a volume of 100 microliters consisung of 100 mM Tπs-HCI (pH 8 0). 1 mM MgCl2. 28 mM KCl. 28 mM NaCl 0 1 mg/ml of DNase I acuvated heπing sperm DNA (Sigma. MO). 3 0 micromolar [3H]nιcotιnamιde adeπine dinucleoude (470 mci/mmole). 7 micrograms/ml PARP enzvme and vanous concentrauons of the compounds to be tested The reacuon is initiated by incubating die mixture at 25°C After 15 minutes of incubation, the reacuon is terminated by adding 500 microliters of ice cold 20% (w/v) tπchloroaceuc acid. The precipitate formed is transfened onto a glass fiber filter (Packard Umfilter-GF/B) and washed three times with ethanol After the filter is dπed. die radioacUvity is determined by scinullation counting The compounds of this invention were found to have potent enzymauc acuvity in the range of a few nM to 20 μM in IC50 in this inhibiuon assay
Using the PARP assay descnbed above, approximate IC50 values were obtained for the following compounds
Figure imgf000060_0001
Figure imgf000061_0001
O 01/16137
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Focal cerebral ischemia
The following focal cerebral ischemia assay is useful for determining the PARP inhibi ng effects of the compounds of the present invenuon The following examples demonstrate that compunds related to those of the present invenuon are effecuve in inhibiung PARP acuvity
Focal cerebral ischemia is produced by cauteπzauon of the πght distal MCA (middle cerebral artery) with bilateral temporary common caroud artery occlusion in male Long-Evans rats for 90 minutes All procedures performed on the ammals are approved by the Umversity InsUtuuonal .Ammal Care and Use Committee of die Umversity of Pennsylvama A total of 42 rats (weights 230-340 g) obtained from Charles River were used in this study The animals fasted ov ernight with free access to water pπor to the surgical procedure
Two hours pπor to MCA occlusion, v n ing amounts (control. n= 14. 5 mg/kg. n=7 10 mg/kg. n=7. 20 mg/kg. π=7, and 40 mg kg. n=7) of the compound 3 4-dιhydro-5-[4-(l-pιpeπdιnyl)-butoxy]-l(2H)- isoquinolinone ("DPQ") were dissolved in dimethv l sulfoxide (DMSO) using a somcator A volume of 1 28 ml/kg of the resulting solution was injected lntrapeπtoneally into fourteen rats
The rats were then anestheuzed with halodiane (4% for induction and 0 8%-I 2% for the surgical procedure) in a mixture of 70% muous oxide and 30% oxygen The body temperature was monitored by a rectal probe and maintained at 37 5 + 0 5°C with a heating blanket regulated by a homeothermic blanket control unit (Han'ard Apparatus Limited. Kent. U K ) A catheter (PE-50) was placed into the tail artery, and arteπal pressure was continuously momtored and recorded on a Grass polygraph recorder (Model 7D. Grass Instruments. Quinc . Massachusetts) Samples for blood gas anal sis (arteπal pH. PaOj and PaC02) were also taken from the tail artery catheter and measured with a biood gas anaivzer ( ABL 30 Radiometer. Copenhagen. Denmark) Artenal blood samples were obtained 30 minutes after MCA occlusion The head of the animal was positioned in a stereotaxic frame and a πght panetal incision between the πght lateral canthus and the external auditory mearus was made Using a dental dπll constantly cooled with saline, a 3 mm bun hole was prepared ov er the coπex supplied bv the πght MC A. 4 mm lateral to the sagittal suture and 5 mm caudal to the coronal suture The dura mater and a thin inner bone lay er were kept, care being taken to position the probe over a tissue area dev oid of large blood v essels The flow probe (tip diameter of 1 mm. fiber separation of 0 25 mm) was lowered to the bottom of the cranial bun hole using a micromampulator The probe was held stationary by a probe holder secured to the skull with dental cement The microvascular blood flow in the πght panetal cortex was cont uouslv monitored with a laser Doppler flow meter (FloLab. Moor. Devon. U K . and Peπflux 4001. Peπmed. Stockholm Sweden)
Focal cerebral ischemia was produced bv cautenzauon of the distal poπion of the πght MCA with bilateral te poran common caroud artery (CCA.) occlusion bv the procedure of Chen et al . "A Model of Focal Ischemic Stroke in die Rat Reproducible Extensive Cortical Infarcuon". Stroke 17 738-43 ( 1986) and/or Liu et al.. "Polyethylene Glycol-conjugated Supero.xide Dismutase and Catalase Reduce Ischemic Brain Injury ". Am I Physiol 256 H589-93 (1989). both of which are hereby incoφorated by reference
Specifically, bilateral CCA's were isolated, and loops made from polyethylene (PE-10) catheter were carefully passed around the CCA's for later remote occlusion The incision made previously for placement of the laser doppler probe was extended to allow obse auon of die rostral end of the zygomauc arch at die fusion point using a dental dπll. and the dura mater overlying the MCA was cut The MCA distal to its crossing with the lnfenor cerebral vein was lifted bv a fine stainless steel hook attached to a micromampulator and. following bilateral CCA occlusion, the MCA was cauteπzed with an electrocoagulator The bun hole was covered with a small piece of Gelform. and the wound was sutured to maintain the bram temperamre within the normal or near- normal range
- 65 -
SUBSTITUTE SHEET (RULE 26] After 90 minutes of occlusion, the carotid loops were released, the tail arteπal catheter was removed, and all of the wounds were sutured Gentamicin sulfate ( 10 mg/ml) was topically applied to the wounds to prevent lnfecuon The anesthetic was disconunued. and die animal was returned to his cage after awakening. Water and food were allowed ad libitum Two hours after MCA occlusion, the animals were given the same doses of the PARP inhibitor as in the pre-treatment. Twent -four hours after MCA occlusion, die rats were sacrificed with an intrapeπtoneal injecuon of pentobarbital sodium ( 150 mg kg) The brain was carefully removed from the skull and cooled in ice-cold artificial CSF for five minutes The cooled bram was then sectioned in the coronal plane at 2 mm intervals using a rodent brain mauix (RBM-4000C. ASI Instruments. Waπen. Michigan) The brain slices were incubated in phosphate-buffered saline containing 2% 2.3.5-tnphenyltetrazolιum chlonde (TTC) at 37°C for ten minutes Color photographs were taken of die posteπor surface of the stained slices and were used to determine the damaged area at each cross-secuonal level using a computer-based image analyzer (NTH Image 1.59). To avoid artifacts due to edema, the damaged area was calculated by subtracung the area of the normal tissue in the hemisphere ipsilateral to the stroke from the area of the hemisphere contralateral to the stroke, by the method of Swanson et al.. "A Semiautomated Mediod for Measunng Brain Infarct Volume". J. Cereb. Blood Flow Metabol. 10:290-93 ( 1990). the disclosure of which is herebv incoφorated by reference The total volume of infarction was calculated by summauon of the damaged volume of die bram slices.
The cauterization of the distal portion of die πght MCA with bilateral temporary CCA occlusion consistently produced a well-recognized cortical infarct in the πght MCA terπtory of each test animal There was an apparent umformity in the distribution of the damaged area as measured by TTC staining in each group, as shown in Figure 1.
In Figure 1. the distribution of the cross-secuonal infarct area at representative levels along the rostrocaudal axis was measured from the interaural line in non-treated ammals and in animals treated with 10 mg/kg of 3.4-dihydro-5-[4-(l-pιpeπdinyl)-butoxy]-l(2H)-ιsoquinolinone. The area of damage was expressed as mean * standard deviation Significant differences between the 10 mg-treated group and die conuol group were indicated ( p<0 02. "p<0 01. " "pO.OOI). The 5 mg kg and 20 mg/kg curves fell approximately halfway between die control and the 10 mg kg cun'es. whereas the 40 mg/kg curve was close to the control. The 5. 20 and 40 mg/kg curves were omitted for claπty.
PARP inhibition led to a significant decrease in the damaged volume in the 5 mg/kg-treated group (106.7 + 23.2 mm3, pθ.001). the 10 mg/kg-treated group (76 4 + 16.8 mm3, pO.OOl). and the 20 mg/kg- treated group (110.2 + 42.0 mm3. p<0.01). compared to the conuol group (165.2 + 34.0 mm3). The data are expressed as mean + standard deviation. The significance of differences between groups was determined using an analysis of vaπance (ANOVA) followed by Student's t-test for individual compaπsons.
There was no significant difference between die control and the 40 mg/kg-treated group (135 6 + 44.8 mm3). However, there were significant differences between the 5 mg/kg-treated group and the 10 mg/kg-treated group (p<0 02) and between the 10 mg/kg-treated group and the 40 mg/kg-treated group (p<0 01). as shown in Figure 2
In Figure 2 the effect of intrapeπtoneal administration of 3 4-dιhydro-5-[4-(l-pιpeπdιnyI)-butoxyJ- l(2H)-ιsoquιnohnone on die infarct v olume was depicted graphically The volumes of infarct were expressed as mean + standard deviation Significant differences between die treated groups and the control group were indicated ("p<0 01. "p<0 001) It is not clear whv a high dose (40 mg/kg) of the PARP inhibitor. 3 4-dιhydro-5- [4-( 1 -pipeπdinv l)-butoxy ]-l(2H)-ιsoquιnolιnone was less neuroprotectiv e The U-shaped dose-response curve mav suggest dual effects of die compound
Howev er. overall the in v iv o adπurusuauon of the inhibitor led to a substantial reduction in infarct volume in the focal cerebral ischemia model in die rat This result indicated that the activation of PARP plays an important role in the pathogenesis of brain damage in cerebral ischemia
The values of artenal blood gases (PaO: PaC02 and pH) were within the physiological range in the control and treated groups with no significant differences in these parameters among the fiv e groups, as shown below in Table III A 'steadv state" MABP was taken following completion of the surgical preparation, just pnor to occlusion an "ischemia ' MABP was taken as die average MABP dunng occlusion
TABLE III
Figure imgf000068_0001
= Significandy different from the steady state value. p<0 05 = Significantly different from the steady state v alue. p<0 01
There were no sigmficant differences in anv phvsiological parameter, including mean artenal blood pressure (MABP), pπor to MCA and CCA occlusion among die five groups Although MABP was significantiv elevated following occlusion in all five groups there were no significant differences in MABP duπng the occlusion peπod among the groups
Since the blood flow values obtained from the laser doppler were in arbitrary umts. only percent changes from the baseline (pπor to occlusion) were reported Right MCA and bilateral CCA occlusion produced a sigmficant decrease in relative blood flow in the πght panetal cortex to 20 8 z 7 % of the baseline in the control group (n=5). 18 7 7 4 % in the 5 mg kg-treated group (n=7) 21 4 + 7 7 % in the 10 mg kg-treated group (n=7) and 19 3 1 1 2 % in die 40 mg/kg-treated group (n=7) There were no significant differences in the blood flow response to occlusion among the four groups In addition, blood flow showed no significant changes throughout the enure occlusion peπod in any group
Following release of die caroud occlusions, a good recovery of blood flow (sometimes hyperemia) was obsen-ed in the πght MCA temtor of all animals Reperfusion of the ischemic tissue resulted in the formation of NO and peroxynitnte. in addiuon to oxygen-deπved free radicals All of these radicals have been shown to cause DNA strand breaks and to acti ate PARP This example provided evidence that the related compounds of the present invention are effective in inhibiting PARP activity
Exemplified compounds of the present invenuon are tested for their ability to reduce focal cerebral ischemia in the following simplified procedure Rats are allowed free access to water and rat chow (Wayne. Chicago. IL) until surgery Housing and anesthesia concur with guidelines established bv the institutional Animal Studies Committee, and are in accordance with the PHS Guide for the Care and Use of Laboratory Ammals. USDA Regulations, and the AVMA Panel on Euthanasia guidelines
The animals are anesdieuzed with isofluorane (induction. 3%. maintenance. 1 25% in a mixture of 30% O; and 70%) NO; through a face mask The rectal temperature is maintained at 37 "C with a homeothermic blanket (Hansard Apparatus. Soudi N'auck. MA) First, an iv catheter is inserted into the left femoral vein and e line run up through the nape of die neck for connection to a tethered swivel (Instech Laboratones. Pl mouth Meeung. PA) and remote infusion pump (Stoelting Inc . Wood Dale. IL) In some cases, the πght femoral artery is cannulated for monitoπng arteπal blood pressure and heart rate and for obtaining blood samples for artenal blood gas
The πght middle cerebral artery (MCA) is then exposed bv making ertical skin incision midway between the nght eye and ear and ov erlying skull is removed with a dental dnll (Chen et al 1986) After incision of the dura, the artery is coagulated at the level of die infenor cerebral vein with a bipolar cautery unit (Valleylab NS2000. Boulder. CO), and cut to prev ent spontaneous reperfusion (Takahashi et al.. 1997) Both common carotid arteπes (CCAs) that had been previously isolated and freed of soft ussues and nen'es are then ligated using non-Uaumauc aneury sm clips After the wounds are closed with surgical clips, the ammals are allowed to recover from anesthesia and retumed to dieir cage which is warmed to 27°C with a heated water undeφad and humidified warm tent
The PARP inhibitor to be tested is first administered 30 min after MCAO as an iv bolus. 10 mg/kg infused over 5 min. followed by a 12 hr continuous infusion of 2 mg/kg/hr (0 3 ml/hr) Ninety minutes after the MCAO. the animals are remov ed from the infusion tether, bπefl reanesthetized with isofluorane. and the carotid clips are removed The ammal is returned to its warm cage and reconnected to the iv infusion pump for die durauon of the expeπment At 24 hrs after permanent MCAO animals are sedated w ith ketamine and the heads removed by guillotine Brains are remo ed, cooled in ice-cold saline and sliced into 2 mm coronal sections using a rat brain matπce (Hanard Bioscience South Natick. MA) The brain slices are incubated in phosphate-buffered saline (pH 7 4) containing 2% TTC at 37°C for 10 min and then stored in 10% neutral-buffered formalin Cross-sectional area of the TTC-unstained region for each brain slice is determined using an image analyzer (MetaMoφh. Universal Imaging Coφ West Chester PA) The total volume of infarction in the πght hemisphere is calculated by summation of the direct (TTC-negative) and indirect measures of the infarct areas in the component brain slices The lnfarcted volumes in vehicle and drug-treated groups (n=8) are tested for significant staustical difference using an unpaired Srudent-t test Vanous doses of the compounds of die inv ention are tested in this model The compounds are administered in either a single dose or a seπes of muluple doses, i p or l v . at different times both before or after the onset of ischemia Compounds of the inv ention are expected to provide protecuon from ischemia in the range of about 0 to 80%
Heart Ischemia/Reperfusion Injun
The expeπments of the heart lschemia/reperfusion injun model is performed using female Sprague- Dawlev rats weighing 250-300g w hich are anesdietized with sodium pentobarbital at dose of 65 mg/kg lntrapentoneally The rectal temperature is maintained at 7°C by using a homeothermic blanket s stem (Hansard Apparatus. South Natick MA) The trachea is cannulated and the rat is ventilated with Room Air by using Hanard Rodent VenUlator (Ha ard Apparatus. South Nauck. MA) The left jugular v ein is cannulated with PE-50 tubing for dmg delivery The πght caroud arten is cannulated for BP measurement The heart is exposed by opemng the chest at the 4th left intercostal space A ma left branch of coronan artery (L D) is occluded bv 4-0 silk ligature for 30 m of ischemia and released for 90 min of reperfusion Duπng the expenment. aπeπal BP and EKG are monitored bv Micro-Med Cardiov ascular System (Louisv die KY) At the end of reperfusion the LAD coronan arten is re-occluded and about 2 ml of 5% Evans Blue dye is injected through I v line to distinguish die ischemic area from non-ischemic area of the heart Then the heart is immediately taken off and frozen in the freezer After at least 30 min of freezing, the heart is sliced into five secuons with 2 mm thickness and stained in 1% TTC soluuon for 30 min at 37°C The πght ventπcle is tπmmed off Infarct area, πsk area and total left v enuicular area in each face of the secuon are measured by using an image analysis svstem (BIOQUANT. Nashville TN) The infarct size is calculated as the percent total infarct volume of the total πsk volume
For dmg treated group Compounds are administered according to die following three protocols 1 Single dose of compound is given 60 min pπor to the onset of ischemia through I p injection 2 Compound is delivered through I v bolus 1 min before the onset of ischemia followed bv I v infusion until die end of reperfusion 3 Compound is delivered through I v bolus 1 min before the onset of reperfusion followed by i v infusion until the end of reperfusion For each dnig-treated group, diere is a coπesponding v elucle ueated group with n=t> or n=8 The difference between vehicle and drug treated groups is compared bv using unpaired t-test with p<0 05 Vaπous doses of compounds are tested in this model The compounds are given in either single or multiple doses 1 p or 1 v at different times before or after the onset of ischemia The compounds of this invention are expected to have ischemia reperfusion injun protection in the range of 10 to 40 percent in this assav
As a result of die PARP lnhibiuon ac v itv the compounds of this invention are expected to protect against ischemia-induced degenerauon of rat cortical neurons in vitro and thus may be useful in disorders aπsing from cerebral ischemia such as stroke sepuc shock or CNS degenerative disorders Thev may also be useful in protecting the spinal cord following trauma As an expeπmental result of lschemia/reperfusion injun in rats the present invention is further directed to a method of prophv lactic or therapeutic treatment of heart attack cardiac anest cardiac bvpass diabetes or πsk of damage which compπses administenng an effective amount of a compound of the present invention for PARP inhibition in unit dosage form
In vitro Radiosensitization In v itro radiosensitization mav be measured with the use of a human prostate cancer cell line PC-3s which are plated in 6 well dishes and grown at monolaver cultures in RPM11640 supplemented with 10% FCS The cells are maintained at °C in 5% CO; and 95% air The cells are exposed to a dose response (0 1 mM to 0 1 μM) of 3 different PARP inhibitors pπor to lπadiauon at one sublethal dose level For all treatment groups, the six well plates are exposed at room temperamre in a Seifert 250kV/15mA madiator with a 0 5 mm Cu/1 mm Cell viabihtv is examined bv exclusion of 0 4% uvpan blue Dve exclusion is assessed visuallv bv microscopv and viable cell number was calculated bv subtracung die number of cells from the viable cell number and dividing bv the total number of cells Cell proliferauon rates are calculated by the amount of Η-thvmidine incoφoration post-iπadiation The P RP inhibitors are expected to radiosensitize the cells
Measuπng Altered Gene Expression in mRNA Senescent Cells
Gene expression alteration mav be measured with human fibroblast BJ cells which at Population Doubling (PDL) 94 are plated in regular growth medium and then changed to low semm medium to reflect phvsiological conditions descπbed in Linskens et al Nucleic Acids Res 23 16 3244-3251 (1995) A medium of DMEM 199 supplemented with 0 5% bovine calf semm is used The cells are treated dailv for 13 davs The control cells are treated widi and without die solv ent used to administer the PARP inhibitor The untreated old and voung conuol cells are tested for compaπson RNA is prepared from the treated and conuol cells according to die techniques descπbed in PCT Pubhcauon No 96/13610 and Northern blotting is conducted Probes specific for senescence-related genes are analvzed and treated and control cells compared In analvzing the results the lowest level of gene expression is arbiuaπlv set at 1 to provide a basis for compaπson Three genes relεv ant to age-related changes in die skin are collagen collagenase and elastin West Arch Derm 130 87-95 ( 1994) Elasun expression of die cells treated with die PARP inhibitor is expected to be significandy increased in compaπson with the control cells Elasun expression should be significantiy higher in young cells compared to senescent cells, and thus ueatment ith the PARP inhibitor should cause elastin expression levels in senescent cells to change to levels similar to those found in much younger cells Similarly, a beneficial effect should be seen in collagenase and collagen expression with ueatment with the PARP inhibitors
Measuπng .Altered Gene Expression of Protein in Senescent Cells
Gene expression alterauon mav be measured widi approximately 105 BJ cells, at PDL 95-100 which are plated and grown in 15 cm dishes The growth medium is DMEM 199 supplemented with 10% bovice calf semm The cells are ueated daily for 24 hours with the PARP inhibitors of (100 μg 1 mL of medium) WO 99/11645 The cells are washed with phosphate buffered solution (PBS), then permeabhzed with 4% paraformaldehyde for 5 minutes, then washed with PBS. and treated w ith 100% cold methanol for 10 minutes The methanol is removed and the cells are washed with PBS. and then treated with 10% semm to block nonspecific antibody binding About 1 mL of the appropπate commercially available antibody solutions (1.500 dilution Vector) is added to the cells and the mixture incubated for 1 hour The cells are πnsed and washed three times with PBS A secondarv antibody, goat anu-mouse IgG (1 mL) with a biotin tag is added along with I mL of a solution containing streptav idin conjugated to alkaline phosphatase and 1 mL of NBT reagent (Vector) The cells are washed and changes in gene expression are noted coloπmetπcally Four senescence- specific genes — collagen I. collagen III. collagenase. and interferon gamma -- in senescent cells treated with the PARP inhibitor are monitored and the results should show a decrease in interferon gamma expression with no obsenable change in the expression levels of the other tiireε gens, demonstrating that the P.ARP inhibitors can alter senescence-specific gene expression
Extending or Increasing Proliferauve Capacity and Lifespan of Cells
To demonstrate the effectiv eness of the present mediod for extending the proliferauve capacity and lifespan of cells, human fibroblast cells lines (either W 138 at Population Doubling (PDL, 23 or BJ cells at PDL 1) are thawed and plated on T75 flasks and allowed to grow in normal medium (DMEM/M199 plus 10% bovine calf semm) for about a week, at which time die cells are confluent, and the cultures are therefor ready to be subdivided At die time of subdivision, the media is aspirated, and the cells πnsed with phosphate buffer saline (PBS) and then trypsinized The cells are counted with a Coulter counter and plated at a density of 105 cells per cnr in 6-well ussue culture plates in DMEM/199 medium supplemented with 10% bovine calf semm and varying amounts (0 lOμM. and ImM from a 100X stock soluuon in DMEM/M199 medium) of a PARP inhibitor This process is repeated every 7 days until the cells appear to stop div iding The untreated (control) cells reach senescence and stop dividing after about 40 days in culture Treatment of cells with 10 μM 3-AB appears to have little or no effect in conuast to treatment with 100 μM 3-AB which appears lengthen die lifespan of the cells and treatment with 1 mM 3-AB which dramatically increases die lifespan and proliferauve capacity of the cells The cells treated with 1 mM 3-AB will sull div ide after 60 days in culture Neuroprotective Effects of PARP Inhibitors on Chronic Constriction Injury (CCD in Rats
Adult male Sprague-Dawlev rats. 300-350 g. are anesthetized with mtrapeπtoneal 50 mg/kg sodium pentobarbital Nen'e ligation is performed by exposing one side of die rat's sciauc nen'es and dissecting a 5-7 mm-long nerve segment and closing with four loose ligatures at a 1 0- 1 5-mm. followed by implanting of an intrathecal catheter and inserting of a gentamicin sulfate-flushed polyethylene (PE-10) tube into the subarachnoid space through an incision at the cistema magna The caudal end of the catheter is gently threaded to the lumbar enlargement and the rostral end is secured with dental cement to a screw embedded in the skull and the skin wound is closed with wound clips
Thermal hvperalgesia to radiant heat is assessed by using a paw -withdrawal test The rat is placed m a plasuc cylinder on a 3-mm thick glass plate with a radiant heat source from a projection bulb placed directly under the plantar surface of die rat's hindpaw The pa -withdrawal latency is defined as the time elapsed from die onset of radiant heat stimulation to withdrawal of the rat's hindpaw Mechanical hvperalgesia is assessed bv placing the rat in a cage with a bottom made of perforated metal sheet w ith manv small square holes Duration of paw-withdrawal is recorded after pπcking the mid-plantar surface of the rat's hindpaw with the tip of a safetv pin inserted dirough the cage bottom
Mechano-allodyma is assessed by placing a rat in a cage similar to the previous test, and applying von Frey filaments in ascending order of bending force ranging from 0 07 to 76 g to the mid-plantar surface of the rat's hindpaw A von Frey filament is applied peφendicular to the skin and depressed slo ly until it bends A threshold force of response is defined as die first filament in the seπes to evoke at least one clear paw- withdrawal out of five applications
Dark neurons are obsen ed bilaterally within the spinal cord dorsal horn, particularly in laminae I-II. of rats 8 days after unilateral sciatic nene ligation as compared widi sham operated rats Vanous doses of PARP inhibitors are tested in this model and shown to reduce both incidence of dark neurons and neuropathic pain behavior in CCI rats
The invention being thus descπbed. it will be obvious that the same may be vaπed in many ways Such vaπauons are not to be regarded as a departure from the spiπt and scope of the invention, and all such modificauons are intended to be included within the scope of die following claims All references cited herein are incoφorated in their entirety bv reference herein

Claims

We claim
A compound of Formula I
Figure imgf000074_0001
or a pharmaceuticalh acceptable salt, hvdrate. prodnig. or mixtures thereof wherein
Figure imgf000074_0002
p is one or two.
Y is a direct bond. >C=0. -0-. -N(Rι0)-. N. or - C(Rβ)p-.
X is NRπ. -0-. -S-. CRi;Ri3. a bond. -CRι :=CR,3-. or
Figure imgf000074_0003
W is selected from -CN. -(N(R9);) where the Ro subsutuents may be combined to form a heteroaryl or cycloalk l opuonally containing at least one hetero atom. -P(0);-OR9. -P(0)(OR9):. -S(0):-R9. -S(0)3R9 -C(O)- R9, -C(0)-N(R0);. -S(0);NR9. cycloalk l opuonally contaimng at least one heteroatom. and heteroaryl.
R], R; R3. R,. R,. R . R-. Rg, Ri . Rι3, R . and R^ are independendy hydrogen. lower alkyl. cycloalkyl opuonally containing at least one heteroatom. lower alkenyl. lower alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroaryl. hydroxy. am o. mtro. halo, nitroso. sulfo. sulfonic acid or carboxy. each Ro is independendy hydrogen, lower alkyl. cycloalkyl opuonally contaimng at least one heteroatom. lower alkenyl, aryl. heteroary l. aralkyl. heteroaralkyl. alkaryl, heteroaralkyl. hydroxy. lower alcohol, alkoxy. amino. or carboxy,
Rio and R are independendy hydrogen, lower alkyl. lower alkenyl. aryl. aralkyl. alkaryl, halo, hydroxy. alkoxy . ammo, or carboxy. each R]6 is independently hydrogen or lower alkyl. and
T. when present, is a divalent or tnvalent organic radical independently selected from the group consisung of lower alkylene. lower alkenylene. arylene. aralkylene. and alkarylene. wherein one. two or three of the hv drogen atoms of said divalent or tπvalent organic radical can be substituted bv a moiety selected from the group consisung of. lower (C| -C9 straight or branched chain) alkyl. cycloalkyl. lower (C:-C9 straight or branched chain) alkenyl. cycloalkenyl, aryl, heteroaryl, aralkyl. heteroaryalky 1. alkaryl. alkheteroaryl. halo, tnfluoromethyl. hydroxy. lower ( -C, ) alkoxy. amino. m o, tπfluoromethyl. alkenyloxy, phenoxy. and benzy loxy: wherein one. two. or three carbon atoms in the divalent or tπvalent orgamc radical can be replaced by a hetero-atom-conta ing-moiety selected from the group consisung of phenoxy. phenoxymethyl, phenoxycarbonyl. benzyloxy. -0-. >C=0. -S-. -SO;-. -NR,SO;-. -S02NR,-. -NR,-. and -PO:-, wherein any of the lower (Cι-C9 straight or branched chain) alkyl. C3-Cg cycloalkyl optionally containing at least one heteroatom in place of a carbon atom, lower (C:-C9 straight or branched chain) alkenyl. aryl. heteroaryl. aralkyl. and alkaryl groups can be independendy substimted with one. two or three subsutuents selected from die group consisung of lower (C,-Co straight or branched chain) alkyl. C3-C8 cycloalkyl opuonally containing at least one heteroatom in place of a carbon atom, lower (C:-C9 straight or branched chain) alkenyl. cycloalkenyl. aryl. heteroaryl. aralk l. heteroaralkyl. alkan l. alkheteroaryl. halo, tπfluoromethyl. hydroxy. lower (C1-C4) alkoxy . carboxy. carbonv l. lower alk l ester, amino. nitro. tnfluoromethyl. alkenyloxy . phenoxy . benzyloxy. wherein one. two. or three carbon atoms of any of the lower (Ci-Co straight or branched chain) alkyl. C3-C8 cycloalkyl optionally containing at least one heteroatom in place of a carbon atom, lower (C;-C9 straight or branched chain) alkenyl. aryl. heteroar l. aralky l. and alkar l groups can be replaced by a hetero-atom- coπtaining-moiety selected from the group consisting of -O-. >C=0. -S-. -SO;-. -NR| SO:-. -SO;NR,-. N, -NRt- . and -PO;-
2. A compound of claim 1 wherein in and n are zero, p is one. W is -CN and Ri-R- are hydrogen.
3 A compound of claim 1. wherein m and n are zero, p is one. W is -CN. T is -CH;-. Z and X are oxygen. Y is N and R, to Ri are hydrogen.
4. A compound of claim 1. wherein herein p is one and W is -(N(R9)2).
5. A compound of claim 4. selected from die group consisung of:
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000076_0002
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000079_0002
Figure imgf000080_0001
6 A compound of claim 1 wherein p is one and W is selected from the group consisung of -
P(0);-OR9 and -P(0)(OR9);
A compound of claim 6 selected from the group consisung of
Figure imgf000080_0002
8 A compound of claim 1 wherein p is one and W is selected from the group consisung of ■ S(0);-Ro. -S(0);OR9 and -S(0):NR9
9 A compound of claim 8 selected from the group consisung of
Figure imgf000080_0003
Figure imgf000081_0001
Figure imgf000082_0001
10. A compound of claim 1 wherein p is one and W is -C(0)R9 or -C(0)N(R9):.
11. A compound of claim 10 selected from the group consisung of
Figure imgf000082_0002
Figure imgf000082_0003
Figure imgf000083_0001
12 A compound of claim 1 wherein p is one and W is a heteroaryl or a cycloalkyl opuonally contaimng at least one heteroatom
13 A compound of claim 12 selected from the group consisung of
Figure imgf000084_0001
14 A compound of Formula (II)
Figure imgf000084_0002
or a pharmaceuucally acceptable salt, hydrate, prodnig. or mixtures thereof, wherein.
Figure imgf000084_0003
Z is O:
X is -0-. or a bond;
Ri, R2, R3. R-i, Rs, Re, and R- are independendy hydrogen, lower alkyl. lower alkenyl. cyioalkyl opuonally containing at least one heteroatom. lower alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl, alkheteroarly. h droxy. ammo, nitro. halo, muoso. or carboxy,
R9 is hydrogen, lower alkyl. cycloalkyl opuonally containing at least one heteroatom. lower alkenyl, ar l. heteroaryl. aralkyl, heteroaralkyl. alkaryl. alkheteroaryl, hydroxy, alkoxy. ammo, or carboxy. and T. when present, is a divalent or tnvalent orgamc radical independendy selected from the group consisung of lower alkylene. lower alkenylene. arylene. aralkylene. and alkarylene; wherein one. two or three of the hydrogen atoms of said divalent or tπvalent orgamc radical can be subsututed by a moiety selected from the group consisung of lower alkyl. lower alkenyl. aryl, aralkyl. alkaryl. halo, tπfluoromefhyl. hydroxy. alkoxy. amino. mtro. tnfluoromethyl. alkenyloxy, phenoxy, and benzyloxy: wherein one. two. or three carbon atoms in the divalent or tπvalent orgamc radical can be replaced by a hetero-atom-containing-moiety selected from the group consisting of: penoxy. phenoxycarbonvl. benzyloxy, -0-. >C=0. -S-. -SO;-. -NR, SO;-. -SO:NR,-. -NR,-. and -PO:-, wherein the lower alkyl. cyioalkyl optionally containing at least one heteroatom. lower alkenyl. aryl. heteroaryl. aralkyl. heteroaralkv 1. alkaryl. and alkheteroan 1 groups can be independently subsututed with one. two or three substituents selected from the group consisung of: lower alkyl. cycolalkyl optionally contaimng at least one heteratom. lower alkenyl. aryl, heteroaryl. aralkyl, heteroaralkyl. alkary 1. alkheteroaryl. halo, tnfluoromethyl. hydroxy, alkoxy. carboxy. carbonyl. lower alkyl ester, amino. mtro. tπfluoromethyl. alkenyloxy. phenoxy. benzyloxy. w herein one. two. or three carbon atoms thereof can be replaced by a hetero-atom-containing-moiety selected from the group consisting of. -0-. >C=0. -S-. -SO«. -NR,SO;-. -S02NR,-. -NR,-. and -PO;-
15 A compound according to Formula (III)
Figure imgf000085_0001
wherein
Z and X are oxygen:
Figure imgf000085_0002
"alk" is lower alkylene:
Rπ . Ris and R,9 are independendy hydrogen or lower alkyl: or Ri- and R,8 or Rι8 and R19 taken together can be a lower alkylene to form a heterocyclic πng; and Ri, R;. R3. R4. R . R* and R- are independendy hydrogen, lower alkyl. cycloalkyl optionally containing at least one hetero atom, lower alkenyl. lower alkoxy. ar l. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroaryl. hydroxy amino. mtro. halo, niuoso. or carboxy
16 A pharmaceutical composition which compπses (1) a therapeuucally effecuve amount of a compound according to claim 1 and (11) a pharmaceuucally acceptable earner
17 The pharmaceuucal composiUon of claim 16. wherein the earner is a steπle soluuon. suspension or emulsion, in a single or divided dose
18 The pharmaceuucal composiUon of claim 16 wherein the earner is a capsule or tablet containing a single or divided dose of said compound
19 The pharmaceuucal composiUon of claim 16 wherein the earner compπses a biodegradable polymer
20 The pharmaceuucal composition of claim 19 wherein the biodegradable polymer releases the compound of formula I over a prolonged peπod of ume
21 The pharmaceuucal composition of claim 16 wherein the earner is a solid implant
22 The pharmaceuucal composiUon of claim 16 for inhibiung PARP activity, treating or preventing diseases or disorders, alteπng gene expression or radiosensiuzing
23 The pharmaceuucal composiUon of claim 22. wherein the diseases or disorders are not mediated by NMDA toxicity
24 The pharmaceuucal composiUon of claim 22. wherein the diseases or disorders are selected from the group consisung of ussue damage resulung from cell damage or death due to necrosis or apoptosis, neuronal mediated tissue damage or diseases, neural ussue damage resulung from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases, vascular stroke, cardiovascular disorders, age- related muscular degenerauon. AIDS and other immune senescence diseases, arthπus. atherosclerosis, cachexia. cancer, degenerative diseases of skeletal muscle involving rephcauve senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders, muscular dystrophy, osteoarthnus. osteoporosis, chronic pain, acute pa . neuropathic pain, nervous insult, peπpheral nene injury, renal failure, reunal ischemia, sepuc shock, and skin aging, diseases or disorders relaung to lifespan or proliferauve capacitv of cells, and diseases or disease condiuons induced or exacerbated by cellular senescence
25 The pharmaceuucal composiUon of claim 24 wherein the neurological disorder is selected from the group consisting of peπpheral neuropathy caused by physical injury or disease state, traumatic bram injury , physical damage to the spinal cord, stroke associated with brain damage, and dem elinaung diseases
27 The pharmaceutical composition of claim 25. wherein the peπpheral neuropathy is caused by Guillain-Baπe syndrome
28 The pharmaceutical composition of claim 25. w herein the demvehnating disease is multiple sclerosis
29 The pharmaceuucal composition of claim 24 wherein the neurodegenerative disease is selected from the group consisting of Alzheimer s Disease. Parkinson's Disease Huntington s Disease and amvotropic later, sclerosis
30 The pharmaceutical composiUon of claim 24. wherein the cancer is selected from the group consisting of ACTH-producing tumors, acute lv mphocytic leukemia, acute nonlymphocyuc leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cεn
Figure imgf000087_0001
cancer, chronic lymphocytic leukemia, chrome myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma. endometπal cancer, esophageal cancer. Ew g's sarcoma, gallbladder cancer, hairy cell leukemia, head & neck cancer. Hodgkin's lymphoma. Kaposi's sarcoma kidney cancer, liv er cancer lung cancer (small and/or non-small cell), malignant peπtoneal effusion malignant pleural effusion, melanoma, mesothelioma. mulUple my eloma. neuroblastoma. non- Hodgkin's Ivmphoma. osteosarcoma. ovan cancer, ovary (germ cell) cancer prostate cancer, pancreatic cancer, penile cancer, retinoblastoma. skin cancer, soft-tissue sarcoma, squamous cell carcinomas, stomach cancer. tesUcular cancer, thyroid cancer, trophoblasuc neoplasms, cancer of the utems. vaginal cancer, cancer of the vulva and Wilm's tumor
31 The pharmaceuucal composition of claim 24. wherein the bowel disorder is colitis
32 The pharmaceuucal composition of claim 24. wherein the bowel disorder is Crohn's disease
33 The pharmaceuucal composition of claim 24. wherein the cardiovascular disorder is selected from the group consisting of cardiov ascular ussue damage coronary artery disease, myocardial infarction, angina pectoπs and cardiogenic shock 34 The pharmaceuucal composiuon of claim 24 wherein the septic shock is endotoxic shock
35 The pharmaceuucal composiUon of claim 24. wherein the disease or disease condition induced or exacerbated by cellular senescence is selected from the group consisting of skin aging, Alzheimer's disease, atherosclerosis, osteoarthnus. osteoporosis, muscular dystrophy, age-related muscular degenerauon. immune senescence, and AIDS
36 A method of inhibiung PARP acuvity. treating or prevenung diseases or disorders, alteπng gene expression, or radiosensiuzing. compπsing administenng a therapeuUcaly effective amount of a compound of claim 1
37 The method of claim 36. wherein the compound is administered as a steπle solution, suspension or emulsion, in a single or divided dose
38 The mediod of claim 36. wherein the compound is administered as a capsule or tablet contaimng a single or divided dose of said compound
39 The method of claim 36 wherein the compound is administered with a biodegradable polymer
40 The method of claim 39 wherein the biodegradable polymer releases the compound of formula I over a prolonged peπod of time
41 The mediod of claim 36 wherein the compound is admimstered with a solid implant
42 The method of claim 36. wherein the diseases or disorders are not mediated by NMDA toxicity
43 The method of claim 36. wherein the diseases or disorders are selected from the group consisung of ussue damage resulung from cell damage or death due to necrosis or apoptosis. neuronal mediated Ussue damage or diseases, neural ussue damage resulung from ischemia and reperfusion injury, neurological disorders and neurodegenerauve diseases, vascular stroke, cardiovascular disorders, age-related muscular degenerauon, AIDS and other immune senescence diseases, arthπus. atherosclerosis, cachexia. cancer, degenerauve diseases of skeletal muscle involving rephcauve senescence, diabetes, head trauma, immune senescence, inflammatory bowel disorders, muscular dystrophy, osteoarthnus. osteoporosis, chrome pa . acute pain, neuropathic pain, nen'ous insult, peπpheral nene injury, renal failure, reunal ischemia, sepuc shock, and skin aging, diseases or disorders relaung to lifespan or proliferauve capacity of cells, and diseases or disease conditions induced or exacerbated by cellular senescence
44 The method of claim 43. wherein the neurological disorder is selected from the group consisting of peπpheral neuropathy caused by ph sical injury or disease state, traumauc brain injury, physical damage to the spinal cord, stroke associated with brain damage, and demyelinaung diseases
45 The method of claim 44. wherein the peπpheral neuropathy is caused by Guillain-Bane sy ndrome
46 The method of claim 44. wherein die demyelinaung disease is multiple sclerosis
47 The method of claim 43. wherein the neurodegenerative disease is selected from the group consisting of Alzheimer's Disease. Parkinson's Disease. Hunungton's Disease and amvotropic lateral sclerosis
48 The method of claim 43 vv herein the cancer is selected from the group consisting of ACTH- produc g tumors, acute lymphocyuc leukemia, acute nonlymphocytic leukemia, cancer of die adrenal cortex, bladder cancer, brain cancer, breast cancer, cenix cancer, chronic lymphocyuc leukemia, chronic myelocyuc leukemia, colorectal cancer, cutaneous T-cell lv mphoma. endometπal cancer, esophageal cancer. Ewing's sarcoma, gallbladder cancer, hairy cell leukemia, head & neck cancer. Hodgkin's lymphoma. Kaposi's sarcoma, kidney cancer, liver cancer, lung cancer (small and/or non-small cell), malignant pentoneal effusion, malignant pleural effusion, melanoma, mesothelioma. multiple myeloma, neuroblastoma. non-Hodgkin's lymphoma. osteosarcoma. ov an cancer, ovary (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, reunoblastoma. skin cancer, soft-tissue sarcoma, squamous cell carcinomas, stomach cancer, tesucular cancer, thyroid cancer, trophoblastic neoplasms, cancer of the uterus, vaginal cancer, cancer of the vulva and Wilm's tumor
49 The method of claim 43. wherein the bowel disorder is colitis
50. The method of claim 43. wherein the bowel disorder is Crohn's disease
51 The method of claim 43 wherein the cardiovascular disorder is selected from the group consisung of cardiovascular tissue damage, coronary artery disease, my ocardial infarction, angina pectoπs and cardiogenic shock.
52 The method of claim 43. wherein the septic shock is endotoxic shock 53 The method of claim 43. wherein die disease or disease condiuon induced or exacerbated by cellular senescence is selected from die group consisung of skin aging, Alzheimer's disease, atherosclerosis, osteoarthnus. osteoporosis, muscular dystroph . age-related muscular degenerauon, immune senescence, and AIDS
54 A compound according to Formula (IV) or Formula (V)
Figure imgf000090_0001
wherein. Ri . R;. R3. R_,. R>. and R^. are independendy hydrogen. lower alkyl. lower alkenyl. cyioalkyl opuonally contaimng at least one heteroatom. lower alkoxy. aryl. heteroaryl. aralkyl. heteroaralkyl. alkaryl. alkheteroarlv. hv droxv. amino. mtro. halo, nitroso. or carboxv
PCT/US2000/023745 1997-09-03 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage WO2001016137A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU70871/00A AU7087100A (en) 1997-09-03 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage,such as neural or cardiovascular tissue damage
EP00959578A EP1212328B1 (en) 1999-09-01 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
DE60029799T DE60029799T2 (en) 1999-09-01 2000-08-30 COMPOUNDS, METHODS AND PHARMACEUTICAL PREPARATIONS FOR THE TREATMENT OF CELL DAMAGE SUCH AS, FOR EXAMPLE, DAMAGE TO NERVE OR HEART TISSUE TISSUE
CA2382317A CA2382317C (en) 1999-09-01 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
MXPA02002213A MXPA02002213A (en) 1999-09-01 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage.
JP2001519703A JP2003508400A (en) 1999-09-01 2000-08-30 Compounds, methods, and pharmaceutical compositions for treating cell damage, such as nerve or cardiovascular tissue damage
AU2005202592A AU2005202592B2 (en) 1997-09-03 2005-06-15 Compounds, Methods and Pharmaceutical Compositions for Treating Cellular Damage, such as Neural or Cardiovascular Tissue Damage

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/387,767 1999-09-01
US09/387,767 US6291425B1 (en) 1999-09-01 1999-09-01 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage

Publications (1)

Publication Number Publication Date
WO2001016137A1 true WO2001016137A1 (en) 2001-03-08

Family

ID=23531306

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/023745 WO2001016137A1 (en) 1997-09-03 2000-08-30 Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage

Country Status (9)

Country Link
US (3) US6291425B1 (en)
EP (1) EP1212328B1 (en)
JP (1) JP2003508400A (en)
AT (1) ATE334985T1 (en)
AU (1) AU7087100A (en)
CA (1) CA2382317C (en)
DE (1) DE60029799T2 (en)
MX (1) MXPA02002213A (en)
WO (1) WO2001016137A1 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002094799A2 (en) * 2001-05-22 2002-11-28 Neurogen Corporation Melanin concentrating hormone receptor ligands: substituted 1-benzyl-4-aryl piperazine analogues
US6514983B1 (en) * 1997-09-03 2003-02-04 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO2003014121A1 (en) * 2001-08-07 2003-02-20 Guilford Pharmaceuticals Inc. Compounds, derivatives, compositions, preparation and uses
US6664269B2 (en) 2001-05-08 2003-12-16 Maybridge Plc Isoquinolinone derivatives
WO2005118528A2 (en) * 2004-06-04 2005-12-15 The University Court Of The University Of Aberdeen Aryl alkyl sulfonamides as therapeutic agents for the treatment of bone conditions
WO2006035673A1 (en) * 2004-09-28 2006-04-06 Japan Science And Technology Agency Novel method of protecting ischemic cardiopathy
US7151102B2 (en) 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7196085B2 (en) 2002-04-30 2007-03-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
EP1841771A2 (en) * 2005-01-19 2007-10-10 MGI GP, Inc. Diazabenzoýde¨anthracen-3-one compounds and methods for inhibiting parp
EP1935885A3 (en) * 2001-05-22 2008-10-15 Neurogen Corporation Melanin concentrating hormone receptor ligands : substituted 1-benzyl-4-aryl piperazine analogues.
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7470688B2 (en) 2005-10-19 2008-12-30 Maybridge Limited Phthalazinone derivatives
US7692006B2 (en) 2006-10-17 2010-04-06 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7981890B2 (en) 2007-09-14 2011-07-19 Astrazeneca Ab Phthalazinone derivatives
US8129380B2 (en) 2008-01-23 2012-03-06 Astrazeneca Ab Phthalazinone derivatives
US8207167B2 (en) 2008-09-19 2012-06-26 Pimco 2664 Limited Aryl-phenyl-sulfonamide-phenylene compounds and their use
US8435968B2 (en) 2008-09-19 2013-05-07 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US8475842B2 (en) 2008-10-07 2013-07-02 Astrazeneca Ab Immediate release pharmaceutical formulation of 4-[3-(4-cyclopropanecarbonyl-piperazine-1-carbonyl)-4-fluoro-benzyl]-2H-phthalazin-1-one
US8524778B2 (en) 2007-03-21 2013-09-03 Pimco 2664 Limited Biphenyl-4-yl-sulfonic acid arylamides and their use as therapeutic agents
US8912187B2 (en) 2003-03-12 2014-12-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
CN104725389A (en) * 2015-02-11 2015-06-24 中国科学院成都生物研究所 Sparganiaceae lactone B derivative as well as preparation method and use thereof
US9624167B2 (en) 2013-06-26 2017-04-18 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamides and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)benzenesulfonamides and their therapeutic use
US10005733B2 (en) 2014-12-17 2018-06-26 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamide and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)-benzenesulfonamide compounds and their therapeutic use
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
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
US10799501B2 (en) 2015-11-05 2020-10-13 King's College Hospital Nhs Foundation Trust Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6426415B1 (en) 1997-09-03 2002-07-30 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods and compositions for inhibiting parp activity
US6291425B1 (en) * 1999-09-01 2001-09-18 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6476048B1 (en) 1999-12-07 2002-11-05 Inotek Pharamaceuticals Corporation Substituted phenanthridinones and methods of use thereof
US6531464B1 (en) 1999-12-07 2003-03-11 Inotek Pharmaceutical Corporation Methods for the treatment of neurodegenerative disorders using substituted phenanthridinone derivatives
ES2220753T3 (en) * 2000-03-20 2004-12-16 N-Gene Research Laboratories Inc. AMIDOXINE DERIVATIVES OF THE PROPENCARBOXILIC ACID, PROCEDURE FOR PREPARATION, AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US6723733B2 (en) 2000-05-19 2004-04-20 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
AU2001273099A1 (en) 2000-07-13 2002-01-30 Guilford Pharmaceuticals Inc. Substituted 4,9-dihydrocyclopenta(imn) phenanthridine-5-ones, derivatives thereof and their uses
US20020150615A1 (en) * 2001-02-12 2002-10-17 Howard Sands Injectable pharmaceutical composition comprising microdroplets of a camptothecin
US6497896B2 (en) * 2001-02-12 2002-12-24 Supergen, Inc. Method for administering camptothecins via injection of a pharmaceutical composition comprising microdroplets containing a camptothecin
US6534080B2 (en) * 2001-02-12 2003-03-18 Super Gen, Inc. Method for administering camptothecins via injection of pharmaceutical composition comprising coated particles of a camptothecin
US6509027B2 (en) * 2001-02-12 2003-01-21 Supergen, Inc. Injectable pharmaceutical composition comprising coated particles of camptothecin
ES2396334T3 (en) * 2003-05-28 2013-02-20 Eisai Inc. Compounds, methods and pharmaceutical compositions for PARP inhibition
US10669383B2 (en) * 2006-10-31 2020-06-02 Evonik Corporation Spheronized polymer particles
WO2008154129A1 (en) * 2007-06-08 2008-12-18 Bausch & Lomb Incorporated Pharmaceutical compositions and method for treating, reducing, ameliorating, alleviating, or preventing dry eye
JP5439380B2 (en) * 2007-10-03 2014-03-12 エーザイ インク. PARP inhibiting compounds, compositions and methods of use
US20100137976A1 (en) * 2008-12-02 2010-06-03 Medtronic Vascular, Inc. Systems and Methods for Treating Heart Tissue Via Localized Delivery of Parp Inhibitors
US20130116284A1 (en) 2010-05-10 2013-05-09 Radikal Therapeutics Inc. Lipoic acid and nitroxide derivatives and uses thereof
KR101827444B1 (en) 2012-02-01 2018-02-08 제일약품주식회사 Crystalline acid-added salt of tircyclo derivatives compound or hydrate thereof and preparation thereof
CN104945406B (en) * 2015-05-25 2017-10-10 苏州康润医药有限公司 Derivative, its preparation method and its application as PARP inhibitor of that non-ketone of alkene 3 of azepine

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999011649A2 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
WO1999011645A1 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Poly(adp-ribose) polymerase ('parp') inhibitors, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO1999011624A1 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Oxo-substituted compounds, process of making, and compositions and methods for inhibiting parp activity

Family Cites Families (130)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2467692A (en) 1949-04-19 Naphthyridones and processes fob
US1895105A (en) 1933-01-24 Cubt bath
US1880441A (en) 1932-10-04 Etjbole m
DE282711C (en)
BE628255A (en)
US932290A (en) 1908-03-24 1909-08-24 Basf Ag Compound of the anthracene series and process of making same.
US1001325A (en) 1910-11-02 1911-08-22 Agfa Ag Product of the anthraquinone series and process of making same.
US1253252A (en) 1914-04-15 1918-01-15 Basf Ag Bluish-green vat dyes and process of making them.
US2593798A (en) 1947-05-22 1952-04-22 Searle & Co Aminoalkylamino isoquinoline compounds
US2612503A (en) 1949-04-19 1952-09-30 Smith Kline French Lab Basic ether-substituted isoquinolines
US2638472A (en) 1949-08-26 1953-05-12 Hoffmann La Roche 1-benzyl-5, 6, 7, 8-tetrahydroisoquinoline and derivatives thereof
US2666059A (en) 1950-08-11 1954-01-12 American Cyanamid Co Anilinoisoquinolines and processes of preparing the same
US2700040A (en) 1951-05-21 1955-01-18 Smith Kline French Lab Aminoalkylaminoisoquinolines
DE963184C (en) 1955-07-24 1957-05-02 Cassella Farbwerke Mainkur Ag Process for the preparation of durable solutions of salts of 1-dialkylaminoalkoxy-3-alkyl-isoquinolines
GB810108A (en) 1955-09-13 1959-03-11 Cassella Farbwerke Mainkur Ag Phthalazine compounds basically substituted in the 1-position
US2892841A (en) 1956-04-02 1959-06-30 Grace W R & Co Process for aminating nitrogen-containing heterocyclic compounds
GB838994A (en) 1956-12-05 1960-06-22 Ici Ltd New anthradipyridazones and their use in polymeric materials
US2992220A (en) 1956-12-05 1961-07-11 Ici Ltd New anthra-1':9'(n):10'(n):5'-dipyridazones and anthra-1':9'(n):10'(n):4'-dipyridazones chemical compounds
US3247212A (en) 1963-03-19 1966-04-19 Dow Chemical Co Thio substituted 3 amino isoquinolines
US3291801A (en) 1963-05-23 1966-12-13 Grace W R & Co Novel octahydro-6(5)-phenanthridinones and preparation thereof
US3300499A (en) 1964-01-27 1967-01-24 Sterling Drug Inc 4-alkyl (or alkenyl)-1, 4-dihydro-1-oxobenzo [f] [1, 7] naphthyridine 2-carboxylic acid derivatives
CH461002A (en) 1965-08-11 1968-08-15 Ciba Geigy Process for the preparation of basic water-soluble dyes
US3403157A (en) 1965-09-01 1968-09-24 American Home Prod Benzo[6, 7]cyclohepta[1, 2, 3-d, e]isoquinoline derivatives
CH463778A (en) 1965-10-12 1968-10-15 Sandoz Ag Process for increasing the lightfastness of organic non-textile materials
DE1644596A1 (en) 1965-11-16 1971-05-06 Bayer Ag Dyes and processes for their preparation
US3557119A (en) 1967-04-11 1971-01-19 American Home Prod 2,3,7,8,9,9a-hexahydro-1h-benzo(d,e)(1,7) naphthyridine derivatives
FR7723M (en) 1967-06-22 1970-03-02
US3830816A (en) 1968-03-09 1974-08-20 Aspro Nicholas Ltd 1-cyclopropylmethyleneamino-3,4-dihydroisoquinoline and acid addition salts thereof
US3573304A (en) 1968-12-03 1971-03-30 Delalande Sa 1-dimethylamino - 3 - methyl-2(pyridyl or pyridazinyl) pentane and the corresponding non-toxic acid addition salts thereof
US3904671A (en) 1969-04-01 1975-09-09 Sterling Drug Inc Esters of 3-(hydroxy or hydroxymethyl)-4-hydroxyphenyl aminomethyl ketones
BE755131A (en) 1969-08-22 1971-02-22 Merck Patent Gmbh 1,2,3,11B-TETRAHYDROPYRIDO / 3,4,5: M, N / THIOXANTHENES, THEIR ACID ADDITIONAL SALTS AND THEIR PREPARATION METHODS
FR2081572B1 (en) 1970-03-12 1973-04-06 Rhone Poulenc Sa
US3700673A (en) 1971-02-12 1972-10-24 Morton Norwich Products Inc 3-4-dihydrobenzo(b) (1,7)naphthyridin-1(2h)-ones
US3723436A (en) 1971-02-25 1973-03-27 Sun Oil Co Process for aromatic lactams
USRE31617E (en) 1972-02-04 1984-06-26 Bristol-Myers Company Optionally substituted 1,2,3,5-tetrahydroimidezo(2,1-b)-quinazolin-2-ones and 6(H)-1,2,3,4-tetrahydropyimido(2,1-b)quinazolin-2-ones
US3838134A (en) 1972-04-03 1974-09-24 G Gauthier Phenanthridinones as antiviral agents
GB1379111A (en) 1972-04-13 1975-01-02 Aspro Nicholas Ltd Preparation of fused-ring pyridine and pyrazine derivatives
CA1000701A (en) * 1972-07-19 1976-11-30 Leslie G. Humber Process for preparing hexahydrobenzo (6,7) cyclohepta(1,2,3-de)isoquinolines
DE2355084A1 (en) 1972-11-06 1974-05-16 Guidotti & C Spa Labor COMPOUNDS HAVING A GASTIC ACID SECRETION-INHIBITING EFFECT AND PROCESS FOR THEIR PRODUCTION
US4169897A (en) 1972-12-21 1979-10-02 Richardson-Merrell Inc. 2,7-bis-basic ethers of 9-phenanthrol and 9-loweralkoxy phenanthrol
US3899529A (en) 1973-02-22 1975-08-12 Merck & Co Inc Aroyl substituted naphthalene acetic acids
CH581113A5 (en) 1973-10-08 1976-10-29 Sandoz Ag
US3950343A (en) 1973-11-06 1976-04-13 Ayerst, Mckenna And Harrison Ltd. Pyrroloisoquinoline derivatives
US3900477A (en) 1973-11-06 1975-08-19 Ayerst Mckenna & Harrison 5-amino-and 5-hydrazinodihydropyrroloisoquinoline derivatives
US3978066A (en) 1973-11-06 1976-08-31 Ayerst, Mckenna And Harrison Ltd. Certain 4,6-dihydropyrrolotriazoline-quinoline derivatives
US3932643A (en) 1974-01-07 1976-01-13 Pfizer Inc. Phenanthridines and phenanthridinones as antiviral agents
US4031097A (en) 1974-09-09 1977-06-21 Eli Lilly And Company Descarboxylysergic acid
US3991064A (en) 1975-01-17 1976-11-09 Warner-Lambert Company Benzonaphthyridines
GB1544952A (en) 1975-03-26 1979-04-25 Sandoz Ltd Isoquinolines
DE2557555C2 (en) 1975-12-20 1986-04-17 Hoechst Ag, 6230 Frankfurt Disazo compounds, process for their preparation and their use as pigments
GB1545767A (en) 1976-06-30 1979-05-16 Aspro Nicholas Ltd Isoquinoline derivatives
JPS5337696A (en) 1976-08-26 1978-04-06 Omnium Chimique Sa Hexahydrocantinonee6 derivative
DE2650226A1 (en) 1976-11-02 1978-05-11 Henkel Kgaa 1,3,4-Tri:amino-isoquinoline hair dye coupler and developer - prepd. by hydrogenating 1,3-di:amino-4-nitroso-isoquinoline using a palladium catalyst
DE2818403A1 (en) 1978-04-27 1979-11-08 Hoechst Ag NEW ISOCHINOL DERIVATIVES AND METHOD FOR THEIR PRODUCTION
US4309543A (en) 1980-03-17 1982-01-05 Dynapol Process for preparing cyclic amides
DE3025385A1 (en) 1980-07-04 1982-01-28 Boehringer Mannheim Gmbh, 6800 Mannheim NEW TRICYCLIC ARYL ETHERS, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
SE426653B (en) * 1980-12-08 1983-02-07 Alfa Laval Ab Plate evaporator
US4472401A (en) 1981-11-27 1984-09-18 Roussel Uclaf Pyrimido-quinoxalines having antiallergic properties
US4594415A (en) 1982-03-29 1986-06-10 Robins Roland K Azole dinucleotide compositions and methods of use
IT1229075B (en) 1985-04-05 1991-07-17 Fidia Farmaceutici Topical compsn. contg. hyaluronic acid deriv. as vehicle
US4757128A (en) 1986-08-01 1988-07-12 Massachusetts Institute Of Technology High molecular weight polyanhydride and preparation thereof
ES8601706A1 (en) 1983-05-16 1985-11-16 Merck & Co Inc Drug delivery system utilizing thermosetting gels.
DE3332633A1 (en) 1983-09-09 1985-04-04 Luitpold-Werk Chemisch-pharmazeutische Fabrik GmbH & Co, 8000 München Substituted carboxylic acid derivatives, process for their preparation, and pharmaceuticals
US4639454A (en) 1985-01-17 1987-01-27 E. I. Du Pont De Nemours And Company Phenylquinazolinecarboxylic acids and derivatives as cancer chemotherapeutic agents
US4742171A (en) 1985-03-13 1988-05-03 Hoechst-Roussel Pharmaceuticals Inc. Method of synthesizing 1,4-dihydro-benzo[c]-1,5-naphthyridin-2(3H)-ones
US5032617A (en) 1985-05-03 1991-07-16 Sri International Substituted benzamide radiosensitizers
US5215738A (en) 1985-05-03 1993-06-01 Sri International Benzamide and nicotinamide radiosensitizers
US5041653A (en) 1985-05-03 1991-08-20 Sri International Substituted benzamide radiosensitizers
US4713244A (en) 1985-08-16 1987-12-15 Bausch & Lomb Incorporated Sustained-release formulation containing an amino acid polymer with a lower alkyl (C1 -C4) polar solvent
US4668506A (en) 1985-08-16 1987-05-26 Bausch & Lomb Incorporated Sustained-release formulation containing and amino acid polymer
US4789724A (en) 1986-10-17 1988-12-06 Massachusetts Institute Of Technology Preparation of anhydride copolymers
DE3768922D1 (en) 1986-12-01 1991-05-02 Sumitomo Chemical Co ANTHRAPYRIDONE COMPOUNDS, THEIR PRODUCTION AND THEIR USE.
US4740581A (en) 1987-02-24 1988-04-26 Eastman Kodak Company Condensation copolymers containing copolymerized isoquinoline derivative colorants and products therefrom
US5480631A (en) 1987-11-19 1996-01-02 Vanderbilt University Radioiodinated benzamines and method of their use as radioimaging
US4925968A (en) 1987-12-23 1990-05-15 American Home Products Corporation N-acyl-N-naphthoylglycines as aldose reductase inhibitors
US5274097A (en) 1988-04-19 1993-12-28 Bayer Aktiengesellschaft 1,3-disubstituted pyrrolidines
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
ATE117553T1 (en) 1988-08-19 1995-02-15 Warner Lambert Co SUBSTITUTED DIHYDROISOCINOLINONES AND RELATED COMPOUNDS AS AMPLIFIERS OF THE LETHAL EFFECTS OF RADIATION AND CERTAIN CHEMOTHERAPEUTICS; SELECTED COMPOUNDS, ANALOGUES AND METHODS.
US4902695A (en) 1989-02-13 1990-02-20 Eli Lilly And Company Excitatory amino acid receptor antagonists
GB8908229D0 (en) 1989-04-12 1989-05-24 Smithkline Beckman Intercredit Compounds
US5665710A (en) 1990-04-30 1997-09-09 Georgetown University Method of making liposomal oligodeoxynucleotide compositions
US5719151A (en) 1990-05-04 1998-02-17 Shall; Sydney Substituted benzene compounds
US5077035A (en) 1990-05-14 1991-12-31 The University Of Michigan Radioiodinated benzovesamicol analogs for cholinergic nerve mapping
US5633282A (en) 1990-05-25 1997-05-27 British Technology Group Limited Inhibition of viral infection
US5635506A (en) 1990-06-26 1997-06-03 Research Corporation Technologies, Inc. 1, 2-dihydro-3H-dibenzisoquinoline-1,3-dione anticancer agents
WO1992000281A1 (en) 1990-06-26 1992-01-09 Research Corporation Technologies, Inc. 1,2-dihydro-3h-dibenzisoquinoline-1,3-dione anticancer agents
WO1994006771A1 (en) * 1992-09-11 1994-03-31 Research Corporation Technologies, Inc. 1,2 DIHYDRO-3-H-DIBENZ(de,h) ISOQUINOLINE 1,3 DIONE AND THEIR USE AS ANTICANCER AGENTS
US5306889A (en) * 1991-07-05 1994-04-26 Sodick Co., Ltd. Wire cut electrical discharge machining apparatus
US5516941A (en) 1991-10-22 1996-05-14 Octamer, Inc. Specific inactivators of "retroviral" (asymmetric) zinc fingers
US5464871A (en) 1993-05-12 1995-11-07 Octamer, Inc. Aromatic nitro and nitroso compounds and their metabolites useful as anti-viral and anti-tumor agents
US5753674A (en) 1991-10-22 1998-05-19 Octamer, Inc. Adenosine diphosphoribose polymerase binding nitroso aromatic compounds useful as retroviral inactivating agents, anti-retroviral agents, anti-retroviral agents and anti-tumor agents
US5473074A (en) 1991-10-22 1995-12-05 Octamer, Incorporated Adenosine diphosphoribose polymerase binding nitroso aromatic compounds useful as retroviral inactivating agents, anti-retroviral agents and anti-tumor agents-54
US5482975A (en) 1991-10-22 1996-01-09 Octamer, Inc. Adenosine diphosphoribose polymerase binding nitroso aromatic compounds useful as retroviral inactivating agents, anti-retroviral agents and anti-tumor agents
DK0539805T3 (en) 1991-10-31 1998-03-23 Asta Medica Ag Phthalazines containing an ether group at the 1-position and process for their preparation
US5264220A (en) 1991-11-12 1993-11-23 Long David M Jr Method of extending the vascular dwell-time of particulate therapeutic and particulate diagnostic agents
AU2394392A (en) * 1991-12-10 1993-07-19 Smithkline Beecham Corporation Treatment of colorectal cancer
JP2989953B2 (en) 1992-02-10 1999-12-13 富士ゼロックス株式会社 Fixing device and fixing endless belt
US5235045A (en) 1992-03-19 1993-08-10 Microbiomed Corporation Non-azo naphthalimide dyes
US5414001A (en) 1992-05-29 1995-05-09 American Cyanamid Company Antineoplastic pyrrolo[4,3,2-de]quinolin-8(1H)-ones
US5391554A (en) 1992-09-09 1995-02-21 Warner-Lambert Company Dihydro- and tetrahydronaphthyridines
JP3205402B2 (en) 1992-09-09 2001-09-04 東芝アイティー・コントロールシステム株式会社 Method and apparatus for determining crystal orientation
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
US5338851A (en) 1993-03-31 1994-08-16 Eli Lilly And Company Synthesis of cis-decahydroisoquinoline-3-carboxylic acids
US5373304A (en) * 1993-05-27 1994-12-13 Nolan; James F. Cellular phone antenna reflector
IT1265340B1 (en) 1993-07-14 1996-11-22 Istituto Biochimico Italiano SUBSTANCES INHIBITING PROTEIN ADP-RIBOSYLATION SUITABLE FOR THE PREVENTION OF COMPLICATIONS OF DIABETES MELLITUS.
US5587384A (en) 1994-02-04 1996-12-24 The Johns Hopkins University Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
US5434188A (en) 1994-03-07 1995-07-18 Warner-Lambert Company 1-ether and 1-thioether-naphthalene-2-carboxamides as inhibitors of cell adhesion and as inhibitors of the activation of HIV
GB9404485D0 (en) 1994-03-09 1994-04-20 Cancer Res Campaign Tech Benzamide analogues
US5395835A (en) 1994-03-24 1995-03-07 Warner-Lambert Company Naphthalamides as central nervous system agents
CA2144783A1 (en) 1994-04-05 1995-10-06 Universiteit Van Pretoria Mdr resistance treatment
US5703089A (en) 1994-04-28 1997-12-30 Knoll Aktiengesellschaft Dihydrodibenzisoquinolinediones
CA2188833A1 (en) 1994-04-28 1995-11-09 Miguel Fernandez Brana Dihydrodibenzo(de,h)isoquinolines derivatives and their use as anti-cancer agents
US5589483A (en) 1994-12-21 1996-12-31 Geron Corporation Isoquinoline poly (ADP-ribose) polymerase inhibitors to treat skin diseases associated with cellular senescence
DK0723956T3 (en) * 1995-01-30 2000-04-17 Hoechst Ag Basically substituted benzoylguanidines, process for their preparation, use thereof as a drug or diagnosis
US5659082A (en) 1995-04-03 1997-08-19 Centaur Pharmaceuticals, Inc. Nitro- and aminobenzamide compounds for neurodegenerative disorders
US5703116A (en) 1995-04-18 1997-12-30 Geron Corporation Telomerase Inhibitors
US5656638A (en) 1995-04-18 1997-08-12 Geron Corporation Telomerase inhibitors
US5760062A (en) 1995-04-18 1998-06-02 Geron Corporation Telomerase inhibitors
US5618813A (en) 1995-05-26 1997-04-08 Abbott Laboratories Benzo[5.6]pyrano[2.3.4-ij]quinolizine and benzo[5.6]thiopyrano[2.3.4-ij]quinolizine derivatives as antibacterial and antineoplastic agents
US5767135A (en) 1995-12-29 1998-06-16 Fernandez-Pol; Jose Alberto Antiviral agent
AU1743997A (en) 1996-12-20 1998-07-17 Geron Corporation Poly(adp-ribose) polymerase inhibitors to treat diseases associated with cellular senescence
US6235748B1 (en) 1997-09-03 2001-05-22 Guilford Pharmaceuticals Inc. Oxo-substituted compounds, process of making, and compositions and methods for inhibiting parp activity
US6197785B1 (en) * 1997-09-03 2001-03-06 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6346536B1 (en) 1997-09-03 2002-02-12 Guilford Pharmaceuticals Inc. Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same
US6291425B1 (en) 1999-09-01 2001-09-18 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6121278A (en) 1997-09-03 2000-09-19 Guilford Pharmaceuticals, Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
DE69829412T2 (en) * 1997-12-22 2005-07-28 Bayer Pharmaceuticals Corp., West Haven INHIBITION OF RAF KINASE USING SYMMETRIC AND UNSYMMETRIC SUBSTITUTED UREAS
ES2153340T3 (en) * 1997-12-22 2006-02-01 Bayer Pharmaceuticals Corp. INHIBITION OF THE QUINASA RAF USING SUBSTITUTED HETEROCICLIC UREAS.
US6201020B1 (en) 1998-12-31 2001-03-13 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
US6635506B2 (en) * 2001-11-07 2003-10-21 International Business Machines Corporation Method of fabricating micro-electromechanical switches on CMOS compatible substrates

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999011649A2 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
WO1999011645A1 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Poly(adp-ribose) polymerase ('parp') inhibitors, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
WO1999011624A1 (en) * 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Oxo-substituted compounds, process of making, and compositions and methods for inhibiting parp activity

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6514983B1 (en) * 1997-09-03 2003-02-04 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US7151102B2 (en) 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US6664269B2 (en) 2001-05-08 2003-12-16 Maybridge Plc Isoquinolinone derivatives
WO2002094799A3 (en) * 2001-05-22 2003-11-06 Neurogen Corp Melanin concentrating hormone receptor ligands: substituted 1-benzyl-4-aryl piperazine analogues
WO2002094799A2 (en) * 2001-05-22 2002-11-28 Neurogen Corporation Melanin concentrating hormone receptor ligands: substituted 1-benzyl-4-aryl piperazine analogues
EP1935885A3 (en) * 2001-05-22 2008-10-15 Neurogen Corporation Melanin concentrating hormone receptor ligands : substituted 1-benzyl-4-aryl piperazine analogues.
WO2003014121A1 (en) * 2001-08-07 2003-02-20 Guilford Pharmaceuticals Inc. Compounds, derivatives, compositions, preparation and uses
US7247641B2 (en) 2001-08-07 2007-07-24 Mgi Gp, Inc. Compounds, derivatives, compositions, preparation and uses
US7196085B2 (en) 2002-04-30 2007-03-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7662818B2 (en) 2003-03-12 2010-02-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US9169235B2 (en) 2003-03-12 2015-10-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US8912187B2 (en) 2003-03-12 2014-12-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US9566276B2 (en) 2003-03-12 2017-02-14 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US10449192B2 (en) 2003-03-12 2019-10-22 Kudo Pharmaceuticals Limited Phthalazinone derivatives
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US11160803B2 (en) 2003-03-12 2021-11-02 Kudos Pharmaceuticals Limited Phthalazinone derivatives
WO2005118528A2 (en) * 2004-06-04 2005-12-15 The University Court Of The University Of Aberdeen Aryl alkyl sulfonamides as therapeutic agents for the treatment of bone conditions
US7964643B2 (en) 2004-06-04 2011-06-21 The University Court Of The University Of Aberdeen Aryl alkyl sulfonamides as therapeutic agents for the treatment of bone conditions
WO2005118528A3 (en) * 2004-06-04 2006-01-26 Univ Aberdeen Aryl alkyl sulfonamides as therapeutic agents for the treatment of bone conditions
WO2006035673A1 (en) * 2004-09-28 2006-04-06 Japan Science And Technology Agency Novel method of protecting ischemic cardiopathy
US8883787B2 (en) 2005-01-19 2014-11-11 Eisai Inc. Diazabenzo[de] anthracen-3-one compounds and methods for inhibiting PARP
EP1841771A2 (en) * 2005-01-19 2007-10-10 MGI GP, Inc. Diazabenzoýde¨anthracen-3-one compounds and methods for inhibiting parp
US8058275B2 (en) 2005-01-19 2011-11-15 Eisai Inc. Diazabenzo[de] anthracen-3-one compounds and methods for inhibiting PARP
EP1841771A4 (en) * 2005-01-19 2008-09-24 Mgi Gp Inc Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting parp
EP2319847A3 (en) * 2005-01-19 2012-06-20 Eisai Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP
US8470825B2 (en) 2005-01-19 2013-06-25 Eisai Inc. Diazabenzo[de] anthracen-3-one compounds and methods for inhibiting PARP
US7820668B2 (en) 2005-01-19 2010-10-26 Eisai Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP
US7902193B2 (en) 2005-10-19 2011-03-08 Maybridge Limited Phthalazinone derivatives
US7470688B2 (en) 2005-10-19 2008-12-30 Maybridge Limited Phthalazinone derivatives
US7692006B2 (en) 2006-10-17 2010-04-06 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US8524778B2 (en) 2007-03-21 2013-09-03 Pimco 2664 Limited Biphenyl-4-yl-sulfonic acid arylamides and their use as therapeutic agents
US7981890B2 (en) 2007-09-14 2011-07-19 Astrazeneca Ab Phthalazinone derivatives
US8129380B2 (en) 2008-01-23 2012-03-06 Astrazeneca Ab Phthalazinone derivatives
US8207167B2 (en) 2008-09-19 2012-06-26 Pimco 2664 Limited Aryl-phenyl-sulfonamide-phenylene compounds and their use
US9050329B1 (en) 2008-09-19 2015-06-09 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US9302984B2 (en) 2008-09-19 2016-04-05 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US8822507B2 (en) 2008-09-19 2014-09-02 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US9616037B2 (en) 2008-09-19 2017-04-11 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US8435968B2 (en) 2008-09-19 2013-05-07 Pimco 2664 Limited Aryl-phenyl-sulfonamido-cycloalkyl compounds and their use
US11633396B2 (en) 2008-10-07 2023-04-25 Kudos Pharmaceuticals Limited Immediate release pharmaceutical formulation of 4-[3-(4- cyclopropanecarbonyl-piperazine-1-carbonyl)-4-fluoro-benzyl]-2H- phthalazin-1-one
US8475842B2 (en) 2008-10-07 2013-07-02 Astrazeneca Ab Immediate release pharmaceutical formulation of 4-[3-(4-cyclopropanecarbonyl-piperazine-1-carbonyl)-4-fluoro-benzyl]-2H-phthalazin-1-one
US11975001B2 (en) 2008-10-07 2024-05-07 Kudos Pharmaceuticals Limited Immediate release pharmaceutical formulation of 4-[3-(4-cyclopropanecarbonyl-piperazine-1-carbonyl)-4-fluoro-benzyl]-2H-phthalazin-1-one
US9624167B2 (en) 2013-06-26 2017-04-18 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamides and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)benzenesulfonamides and their therapeutic use
US9796670B2 (en) 2013-06-26 2017-10-24 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamides and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)benzenesulfonamides and their therapeutic use
US10029979B2 (en) 2013-06-26 2018-07-24 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamides and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)benzenesulfonamides and their therapeutic use
US10233147B2 (en) 2013-06-26 2019-03-19 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamides and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)benzenesulfonamides and their therapeutic use
US10005733B2 (en) 2014-12-17 2018-06-26 Pimco 2664 Limited N-(4-hydroxy-4-methyl-cyclohexyl)-4-phenyl-benzenesulfonamide and N-(4-hydroxy-4-methyl-cyclohexyl)-4-(2-pyridyl)-benzenesulfonamide compounds and their therapeutic use
CN104725389B (en) * 2015-02-11 2017-03-01 中国科学院成都生物研究所 SsnB derivant and its production and use
CN104725389A (en) * 2015-02-11 2015-06-24 中国科学院成都生物研究所 Sparganiaceae lactone B derivative as well as preparation method and use thereof
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
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
MXPA02002213A (en) 2002-11-07
JP2003508400A (en) 2003-03-04
US6291425B1 (en) 2001-09-18
EP1212328B1 (en) 2006-08-02
US6716828B1 (en) 2004-04-06
ATE334985T1 (en) 2006-08-15
CA2382317A1 (en) 2001-03-08
US7307080B2 (en) 2007-12-11
DE60029799D1 (en) 2006-09-14
AU7087100A (en) 2001-03-26
US20050074470A1 (en) 2005-04-07
EP1212328A1 (en) 2002-06-12
CA2382317C (en) 2010-10-12
DE60029799T2 (en) 2007-08-16

Similar Documents

Publication Publication Date Title
WO2001016137A1 (en) Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6635642B1 (en) PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6380211B1 (en) Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6514983B1 (en) Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6201020B1 (en) Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
US6387902B1 (en) Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP
US20020156050A1 (en) Carboxamine compounds, methods and compositions for inhibiting PARP activity
US20020028813A1 (en) Thioalkyl compounds, methods, and compositions for inhibiting parp activity
US20030105102A1 (en) Oxo-substituted compounds, process of making, and compositions and methods for inhibiting PARP activity
US6395749B1 (en) Carboxamide compounds, methods, and compositions for inhibiting PARP activity
WO1999011622A1 (en) Amino-substituted compounds, methods, and compositions for inhibiting parp activity
AU2005202592B2 (en) Compounds, Methods and Pharmaceutical Compositions for Treating Cellular Damage, such as Neural or Cardiovascular Tissue Damage
MXPA99011815A (en) Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
MXPA00011258A (en) Carboxamide compounds, compositions, and methods for inhibiting parp activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE 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 NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2382317

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 70871/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/002213

Country of ref document: MX

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 519703

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 2000959578

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000959578

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 2000959578

Country of ref document: EP