WO2001090077A1 - Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses - Google Patents

Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses Download PDF

Info

Publication number
WO2001090077A1
WO2001090077A1 PCT/US2001/015571 US0115571W WO0190077A1 WO 2001090077 A1 WO2001090077 A1 WO 2001090077A1 US 0115571 W US0115571 W US 0115571W WO 0190077 A1 WO0190077 A1 WO 0190077A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
disease
compounds
parp
hydrogen
Prior art date
Application number
PCT/US2001/015571
Other languages
French (fr)
Inventor
Jia-He Li
Vincent J. Kalish
Jie Zhang
Larisa E. Serdyuk
Dana V. Ferraris
Ge Xiao
Paul W. Kletzly
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 AU2001264595A priority Critical patent/AU2001264595A1/en
Publication of WO2001090077A1 publication Critical patent/WO2001090077A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/10Aza-phenanthrenes
    • C07D221/12Phenanthridines

Definitions

  • the present application claims benefit of the filing of U.S. Provisional Application No. 60/205,259, filed May 19, 2000, the entire contents of which is incorporated by reference herein.
  • the present invention relates to inhibitors of the nuclear enzyme poly(adenosine 5'-diphospho-ribose) polymerase ["poly(ADP-ribose) polymerase” or "PARP”, which is also referred to as ADPRT (NAD:protein (ADP-ribosyi transferase (polymersing)) and PARS (poly(ADP-ribose) synthetase) and provides compounds and compositions containing the disclosed compounds.
  • ADPRT nuclear enzyme poly(adenosine 5'-diphospho-ribose) polymerase
  • PARS poly(ADP-ribose) synthetase
  • the present invention 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 ischemic stroke, head trauma or spinal cord injury; neurological disorders and neurodegenerative 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, myocardial infarction; to treat other conditions and/or disorders such as, for example, age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes (such as diabetes mellitus), inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis, hemorrhagic shock,
  • PARP (EC 2.4.2.30), also known as PARS (for poly(ADP-ribose) synthetase), or ADPRT (for NAD:protein (ADP-ribosyl) transferase (polymerising)) is a major nuclear protein of 116 kDa. It is mainly present in almost all eukaryotes. The enzyme synthesizes poly(ADP-ribose), a branched polymer that can consist of over 200 ADP-ribose units from NAD. The protein acceptors of poly(ADP-ribose) are directly or indirectly involved in maintaining DNA integrity.
  • PARP protein is expressed at a high level in many tissues, most notably in the immune system, heart, brain and germ-line cells. Under normal physiological conditions, there is minimal PARP activity. However, DNA damage causes an immediate activation of PARP by up to 500-fold. Among the many functions attributed to PARP is its major role in facilitating DNA repair by ADP-ribosylation and therefore co-ordinating a number of DNA repair proteins. As a result of PARP activation, NAD levels significantly decline.
  • peroxynitrite formed from a combination of nitric oxide (NO) and superoxide, appears to be a main perpetrator responsible for various reported disease conditions in vivo, e.g., during shock, stroke and inflammation
  • PARP inhibitors suppress production of inducible nitric oxide synthase (iNOS) in macrophages, P-type selectin and intercellular adhesion molecule- 1 (ICAM-1) in endothelial cells. Such activity underlies the strong anti-inflammation effects exhibited by PARP inhibitors. PARP inhibition is able to reduce necrosis by preventing translocation and infiltration of neutrophils to the injured tissues. (Zhang, J. "PARP inhibition: a novel approach to treat ischaemia/reperfusion and inflammation-related injuries", Chapter 10 in Emerging Drugs (1999) 4: 209-221 Ashley Publications Ltd., and references cited therein.)
  • PARP production is activated by damaged DNA fragments which, once activated, catalyzes the attachment of up to 100 ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself.
  • ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself.
  • PARP activation can also be used as a measure of damage following neurotoxic insults following over- exposure to any of glutamate (via NMDA receptor stimulation), reactive oxygen intermediates, amyloid ⁇ - protein, N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or its active metabolite N-methyl-4- phenylpyridine (MPP * ), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease.
  • MPTP N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine
  • MPP * active metabolite N-methyl-4- phenylpyridine
  • NMDA N-methyl-D-aspartate
  • AMP A AMP A
  • Kainate AMP A
  • MGR receptors which open ion channels and permit uncontrolled ion flow (e.g., Ca 2+ and Na + into the cells and K + out of the cells) leading to overstimulation of the neurons.
  • the over-stimulated neurons secrete more glutamate, creating a feedback loop or domino effect which ultimately results in cell damage or death via the production of proteases, lipases and free radicals.
  • Glutamate exposure and stimulation has also been implicated as a basis for compulsive disorders, particularly drug dependence.
  • Evidence includes findings in many animal species, as well as in cerebral cortical cultures treated with glutamate or NMDA, that glutamate receptor antagonists (i.e., compounds which block glutamate from binding to or activating its receptor) block neural damage following vascular stroke.
  • NMDA neurotoxicity may be prevented by treatment with nitric oxide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro.
  • NOS neuronal nitric oxide synthase
  • PARP inhibitors such as 3 -amino benzamide
  • DNA repair generally in response, for example, to hydrogen peroxide or gamma-radiation.
  • Cristovao et al. "Effect of a Poly(ADP- Ribose) Polymerase Inhibitor on DNA Breakage and Cytotoxicity Induced by Hydrogen Peroxide and ⁇ -
  • PARP inhibitors have been reported to be effective in radiosensitizing hypoxic tumor cells and effective in preventing tumor cells from recovering from potentially lethal damage of DNA after radiation therapy, presumably by their ability to prevent DNA repair.
  • Evidence also exists that PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis.
  • Salzman et al. "Role of Peroxynitrite and Poly(ADP-Ribose)Synthase Activation Experimental Colitis," Japanese J. Pharm., 75, Supp. 1:15 (1997).
  • Colitis was induced in rats by intraluminal administration of the hapten trinitrobenzene sulfonic acid in 50% ethanol.
  • Treated rats received 3- aminobenzamide, a specific inhibitor of PARP activity.
  • Inhibition of PARP activity reduced the inflammatory response and restored the morphology and the energetic status of the distal colon.
  • Southan et al. "Spontaneous Rearrangement of Aminoalkylithioureas into Mercaptoalkylguanidines, a Novel Class of Nitric Oxide Synthase Inhibitors with Selectivity Towards the Inducible Isoform", Br. J.
  • Heller et al. "Inactivation of the Poly(ADP-Ribose)Polymerase Gene Affects Oxygen Radical and Nitric Oxide Toxicity in Islet Cells," J. Biol. Chem., 270:19, 11176-80 (May 1995).
  • Heller et al. used cells from mice with inactivated PARP 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 have been shown to be useful for treating endotoxic shock or septic shock.
  • Zingarelli et al. "Protective Effects of Nicotinamide Against Nitric Oxide-Mediated Delayed Vascular Failure in Endotoxic Shock: Potential Involvement of PolyADP Ribosyl Synthetase," Shock, 5:258-64 (1996), suggests that inhibition of the DNA repair cycle triggered by poly(ADP ribose) synthetase has protective effects against vascular failure in endotoxic shock.
  • Zingarelli et al. found that nicotinamide protects against delayed, NO-mediated vascular failure in endotoxic shock.
  • nicotinamide may be related to inhibition of the NO-mediated activation of the energy-consuming DNA repair cycle, triggered by poly(ADP ribose) synthetase.
  • Cuzzocrea "Role of Peroxynitrite and Activation of Poly(ADP-Ribose) Synthetase in the Vascular Failure Induced by Zymosan-activated Plasma," Brit. J. Pharm., 122:493-503 (1997).
  • PARP inhibitors have been used to treat cancer.
  • PARP inhibitors are used for peripheral nerve injuries, and the resultant pathological pain syndrome known as neuropathic pain, such as that induced by chronic constriction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characterized by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark” neurons) occurs.
  • CCI chronic constriction injury
  • nucleoplasm cytoplasm and nucleoplasm
  • PARP inhibitors have also been used to extend the lifespan and proliferative capacity of cells including treatment of diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, AIDS, and other immune senescence diseases; and to alter gene expression of senescent cells.
  • diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, AIDS, and other immune senescence diseases.
  • diseases such as skin aging, Alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, AIDS, and other
  • 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 invention are specifically useful in ameliorating, treating and/or preventing neural tissue or cell damage, including that following focal ischemia and reperfusion injury.
  • inhibition of PARP activity spares the cell from energy loss, preventing irreversible depolarization of the neurons and, thus, provides neuroprotection.
  • the inhibition of PARP activity by use of the compounds, compositions and methods of the present invention is believed to protect cells, tissue and organs by protection against the ill-effects of reactive free radicals and nitric oxide.
  • the present invention therefore also provides methods of treating and/or preventing cells, tissue and/or organs from reactive free radical and/or nitric oxide induced damage or injury.
  • the present invention provides 6(5_Y)p__enan- or (J_Y)phenanthridin-6-one compounds which inhibit poly(ADP-ribose) polymerase (“PARP"), compositions containing these compounds and methods for making and using these PARP inhibitors to treat, prevent and/or ameliorate the effects of the conditions described herein.
  • PARP poly(ADP-ribose) polymerase
  • R 2 is hydrogen, hydroxyl, amino, nitroso, methyl, amino methyl or carboxylic acid, preferably Ri and R 2 are not both hydrogen; one of R 3 and R 4 is -QP and the other of R 3 and R 4 is one of hydrogen, methyl, trifluoromethyl, nitro, amino, halogen and 1-piperazine, wherein Q is one of
  • P is Z-(N(R 5 R ⁇ ;)), such as Z-N(R 5 R ⁇ 5 ), Z or a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of O, N and S;.
  • Z is hydrogen, a direct bond, a carbonyl, an optionally substituted -Cs straight or branched chain alkyl, cycloalkyl, carboxy, optionally substituted C ⁇ -C 6 ether, aryl or heteroaryl, alkylalkenyl, alkynyl, alkylhalo, straight or branched chain or CH 2 COOH, provided that when P is Z, Z is not hydrogen and R 5 and Re are independently hydrogen, lower alkyl, lower alkenyl, lower alkanol, heterocycle, heteroaryl, alkoxy, aryloxy, alkylamino, arylamino. CH 2 COOH, or R 5 and Re taken together form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic or cycloalkyl ring containing 1, 2 or 3 heteroatoms selected from O, N, S and combinations thereof.
  • Figure 1 shows the distribution of the cross-sectional infarct area at representative levels 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-dihydro-5-[4- (l-piperidinyl)-botoxyl] -1 (2_7)-isoquinolinone.
  • Figure 2 shows the effect of intraperitoneal administration of 3,4-dihydro-5-[4-(l-piperidinyl)-butoxy]- l(2/!_)-isoquinolinone on the infarct volume.
  • the present invention pertains to compounds, pharmaceutical compositions containing the same, methods of using the same, and process of making the same, wherein such compounds are useful as inhibitors of poly( ADP-ribose) polymerase (PARP).
  • PARP poly( ADP-ribose) polymerase
  • they treat or prevent neural tissue 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 the lifespan and proliferative capacity of cells and thus can be used to treat or prevent diseases associated therewith; they alter gene expression of senescent cells; and they radiosensitize hypoxic tumor cells.
  • the 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 thought 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 PARP activation.
  • the present invention provides compounds which inhibit poly(ADP-ribose) polymerase (“PARP”), compositions containing these compounds and methods for using these PARP inhibitors to treat, prevent and/or ameliorate the effects of the conditions described herein.
  • PARP poly(ADP-ribose) polymerase
  • the present invention provides compounds of Formula I:
  • R 2 is hydrogen, hydroxyl, amino, nitroso, methyl, amino methyl or carboxylic acid, preferably Ri and R 2 are not both hydrogen; one of R 3 and R 4 is -QP and the other of R 3 and R4 is one of hydrogen, methyl, trifluoromethyl, nitro, amino, halogen and 1-pi ⁇ erazine, wherein Q is one of
  • X is O, S , N or an N-substituted amino acid; provided that, when X is O or S, then Y is absent, when X is N, then Y is hydrogen, C ⁇ -C 6 straight or branched chain alkyl, alkoxy or alkyl amino, or Y and Z are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring which contains 1, 2 or 3 heteroatoms selected from O, N, S and mixtures combination;
  • Z is hydrogen, a direct bond, a carbonyl or an optionally substituted C ⁇ -C 5 straight or branched chain alkyl, alkenyl, alkynyl, alkylhalo, or CH 2 COOH, provided that when P is Z, Z is not hydrogen and
  • R 5 and Re are independently hydrogen, lower alkyl, lower alkenyl, lower alkanol, heterocycle, heteroaryl, alkoxy, aryloxy, alkylamino, arylamino, CH 2 COOH, or R 5 and Re taken together form a 5, 6 or 7 membered substituted or unsubstituted aryl, heteroaryl, heterocyclic or cycloalkyl ring containing 1, 2 or 3 heteroatoms selected from O, N, S and combinations thereof.
  • the present invention provides the following compounds of Formulas (II), (III), (IV) and (V):
  • the compounds of the invention are described by the compound of Formula I,
  • R 2 is hydrogen, Q is , and X is N.
  • Preferred compounds of this embodiment are further defined by A as carbon and P as Z-N R 5 R5), more preferably where R 5 and 5 form a 5 or 6 membered substituted or unsubstituted heterocycle or heteroaryl.
  • Alternative compounds of the preferred compounds of this embodiment are further defined by R 5 and Re being independently selected from hydrogen, lower alkyl, alkoxy, alkylamino and CH 2 COOH.
  • R 3 or i is hydrogen.
  • the compounds of the invention are described by the compound of Formula I,
  • Preferred embodiments include compounds wherein and P is a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of 0, N and S. Z in these compounds is preferably and aromatic ring containing 0 heteroatoms.
  • R 2 is hydrogen
  • Q is , and X is N.
  • Preferred embodiments include compounds wherein and Z and Y are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring, preferably a non-aromatic ring.
  • prefe ⁇ ed compounds of this embodiment include compounds wherein P is Z-N(R 5 Rs).
  • R 3 or R 4 is hydrogen.
  • compositions containing these prefe ⁇ ed and alternate embodiments and methods of making and using the same, as described herein, are also prefe ⁇ ed
  • the compounds of the invention exhibit an IC 50 for inhibiting PARP in vitro, as measured by the methods described herein, of about 20 ⁇ M or less, preferably less than about lO ⁇ M, more preferably less than about 1 ⁇ M, or less than O.l ⁇ M, most preferably less than about 0.01 ⁇ M.
  • Prefe ⁇ ed embodiments of the present invention include the following compounds (where compound numbers are shown next to each compound), and neutral forms thereof, where appropriate:
  • the compounds and compositions of the present invention 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 invention can be used to extend the lifespan and proliferative capacity of cells and thus can be used to treat or prevent diseases associated therewith; they alter gene expression of senescent cells; and they radiosensitize hypoxic tumor cells.
  • the compounds and compositions of the invention can be used to 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.
  • the compounds of the present invention are not limited to being useful in treating glutamate mediated neurotoxicity and/or NO-mediated biological pathways. Further, the compounds of the invention can be used to treat or prevent other tissue damage related to PARP activation, as described herein.
  • the present invention provides compounds which inhibit the in vitro and/or in vivo polymerase activity of poly( ADP-ribose) polymerase (PARP), and compositions containing the disclosed compounds.
  • PARP poly( ADP-ribose) polymerase
  • the present invention provides methods to inhibit, limit and/or control the in vitro and/or in vivo polymerase activity of poly( ADP-ribose) polymerase (PARP) in any of solutions, cells, tissues, organs or organ systems.
  • PARP poly( ADP-ribose) polymerase
  • the present invention provides methods of limiting or inhibiting PARP activity in a mammal, such as a human, either locally or systemically.
  • the present invention provides methods to treat and/or prevent diseases, syndromes and/or conditions exacerbated by or involving the increased generation of PARP. These methods involve application or administration of the compounds of the present invention to cells, tissues, organs or organ systems of a person in need of such treatment or prevention.
  • the present invention provides methods 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 during cardiac a ⁇ est when the heart, once prevented from receiving blood, begins to reperfuse and these methods involve administration of the compounds and compositions of the present invention preferably prior 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, cardiovascular disorders
  • the present invention provides in vitro or in vivo methods to extend or increase the lifespan and/or proliferation capacity of cells and thus also methods to treat and/or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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.
  • diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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
  • the present invention provides methods of treating or preventing or ameliorating the effect of cancer and/or to radiosensitize hypoxic tumor cells to render the tumor cells more susceptible to radiation therapy and thereby to prevent the tumor cells from recovering from potentially lethal damage of DNA after radiation therapy.
  • a method of this embodiment is directed to specifically and preferentially radiosensitizing tumor cells rendering the tumor cells more susceptible to radiation therapy than non-tumor cells.
  • the present invention 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, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, spinal chord injury, immune senescence, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis, hemo ⁇ hagic shock, splanchnic artery occlusion shock, multiple organ failure (such as involving any of the kidney, liver, renal, pulmonary, retinal, pancreatic and/or skeletal muscles systems), acute autoimmune thyroiditis, muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), local and
  • the compounds of the present invention maybe administered, for example, parenterally, to a person diagnosed with acute retinal ischemia or acute vascular stroke, either by intermittent or continuous intravenous administration, by either a single dose or a series of divided doses.
  • Compounds of the invention may be used in combination or sequentially.
  • the compound of the invention can be administered by intermittent or continuous administration via implantation of a biocompatible, biodegradable polymeric matrix delivery system containing a compound of formula I, II, III, IV or V, or via a subdural pump inserted to administer the compound directly to the infarct area of the brain.
  • the present invention provides methods to extend the lifespan and proliferative capacity of cells, such as. for example, in using the compounds of the invention as general mediators in the generation of oxidants, proinflammatory mediators and/or cytokines, and/or general mediators of leukocyte infiltration, calcium ion overload, phospholipid peroxidation, impaired nitric oxide metabolism and/or reduced ATP production
  • the compounds of the invention can treat 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 during cardiac a ⁇ est when 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 lifespan or proliferation of cells and thus to treat or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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.
  • diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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
  • 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, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging.
  • vascular stroke to treat or prevent cardiovascular disorders
  • other conditions and/or disorders such as age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence,
  • the compounds of the invention 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 lifespan and proliferative capacity of cells; to alter gene expression of senescent cells; and to radiosensitize tumor cells.
  • Another especially prefe ⁇ ed embodiment of the invention is a pharmaceutical composition which comprises (i) a therapeutically effective amount of the compound of formula I, II, III, IV or V; and (ii) a pharmaceutically acceptable carrier.
  • alkyl means a branched or unbranched saturated hydrocarbon chain comprising 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-pentyl, n-hexyl, and the like, unless otherwise indicated.
  • alkyl and ether chains include mercapto, carboxy, hydroxy, or phenyl, benzyl, or phenylethyl, which may themselves be substituted by hydroxy, halo, methoxy, d -C ⁇ alkyl, amine and carboxy.
  • Alkenyl means a branched or unbranched unsaturated hydrocarbon chain comprising a designated number of carbon atoms.
  • C 2 -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, butenyl. iso-butenyl, tert-butenyl, n-pentenyl, n-hexenyl, and the like, unless otherwise indicated.
  • Alkoxy means the group -OR wherein R is alkyl as herein defined.
  • R is a branched or unbranched saturated hydrocarbon chain containing 1 to 6 carbon atoms.
  • Cyclo used herein as a prefix, refers to a structure characterized by a closed ring.
  • Halo means at least one fluoro, chloro, bromo, or iodo moiety, unless otherwise indicated.
  • Amino compounds include amine (NH 2 ) as well as substituted amino groups comprising alkyls of one through six carbons.
  • “Ar”, “aryl” or “heteroaryl” means a moiety which is substituted or unsubstituted, especially a cyclic or fused cyclic ring and includes a mono-, bi-, or tricyclic. carbo- or heterocyclic ring, such as a 5.
  • ring 6, 7 or 8 membered ring, wherein the ring is either unsubstituted or substituted in, for example, one to five position(s) with halo, haloalkyl, hydroxyl, nitro, trifluoromethyl, Cx-Q straight or branched chain alkyl, C 2 -C 6 straight or branched chain alkenyl, C ⁇ -C 6 alkoxy. -C(0)-0(C ⁇ -C 6 alkyl), carboxy, C 2 -C 6 alkenyloxy, phenoxy, benzyloxy, amino, thiocarbonyl.
  • heterocyclic ring contains 1-4 heteroatom(s) selected from the group consisting of O, N, or S or their mixture; wherein aromatic or tertiary alkyl amines are optionally oxidized to a co ⁇ esponding N-oxide.
  • Heteroaryls may be attached to other rings or substituted through the heteroatom and/or carbon atom of the ring.
  • aryl or heteroaryl moieties include but are not limited to phenyl, benzyl, naphthyl, piperidino, py ⁇ olyl, py ⁇ olidinyl, pyridinyl, pyrimidinyl, purinyl, quinolinyl, isoquinolinyl. fiiryl, thiophenyl, imidazolyl, oxazolyl. thiazolyl, pyrazolyl, and thienyl.
  • Cycloalkyls optionally containing at least one heteroatom may be substituted by or fused to at least one 5 or 6 membered aryl or heteroaryl and/or substituted by at least one of amino, C1-C5 straight or branched chain alkyl, -C ⁇ alkanol, C ⁇ -C 6 straight or branched chain alkylamino, C ⁇ -C 6 alkoxy, or Q-C 6 alkenyl, or benzyl, or phenyl or phenylethyl wherein the ring may be substituted as described above for substitutions of "Phenyl".
  • Prefe ⁇ ed cycloalkyls containing at least one heteroatom include , py ⁇ olidinyl, indolyl, 2,3-dihydro-l H isoindolyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholino and thiomorpholino.
  • the compounds of the present invention possess one or more asymmetric center(s) and thus can be produced as mixtures (racemic and non-racemic) of stereoisomers, or as individual enantiomers or diastereomers.
  • the individual stereoisomers may be obtained by using an optically active starting material, by resolving a racemic or non-racemic mixture of an intermediate at some appropriate stage of the synthesis, or by resolution of the compound of any of formulas I, II, III, IV and V. It is understood that the individual stereoisomers as well as mixtures (racemic and non-racemic) of stereoisomers are encompassed by the scope of the present invention.
  • the compounds of the invention are useful in a free base form, in the form of pharmaceutically acceptable salts, pharmaceutically acceptable hydrates, pharmaceutically 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 activity and which is neither biologically nor otherwise undesirable.
  • Organic acids can be used to produce salts, hydrates, esters, or solvates such as acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, p-toluenesulfonate, bisulfate, sulfamate, sulfate, naphthylate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane-propionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate heptanoate, hexanoate, 2-hydroxy
  • Inorganic acids can be used to produce salts, hydrates, esters, or solvates such as hydrochloride, hydrobromide, hydroiodide, and thiocyanate.
  • suitable base salts, hydrates, 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, hydrates, esters, or solvates may also be formed with organic bases.
  • Organic bases suitable for the formation of pharmaceutically acceptable base addition salts, hydrates, 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 may include mono-, di-, and trialkylamines, such as methylamine, dimethylamine, triethylamine and dicyclohexylamine; mono-, di- or trihydroxyalkylamines, such as mono-, di-, and triethanolamine; amino acids, such as arginine and lysine; guanidine; N-methyl-glucosamine; N-methyl-glucamine; L-glutamine; N-methyl-piperazine; morpholine; ethylenediamine; N-benzyl-phenethylamine; (trihydroxy-methyl)aminoethane; and the like.
  • basic nitrogen-containing groups can be quatemized with agents including: lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl suifates such as dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aralkyl halides such as benzyl and phenethyl bromides.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl suifates such as dimethyl, diethyl, dibutyl and diamyl sulfates
  • long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bro
  • 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 invention in an aqueous or an aqueous alcohol solution or other suitable solvent containing the appropriate acid or base, and isolating 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 the PARP inhibitor having an acid group thereon with a base, such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained by concentrating the solution.
  • “Pharmaceutically acceptable prodrug” refers to a derivative of the inventive compounds which undergoes biotransformation prior to exhibiting its pharmacological effect(s).
  • the prodrug is formulated with the objective(s) of improved chemical stability, improved patient acceptance and compliance, improved bioavailability. prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity).
  • the prodrug can be readily prepared from the inventive compounds using metiiods known in the art, such as those described by Burger's Medicinal Chemistry and Drug Chemistry, Fifth Ed., Vol. 1, pp. 172-178, 949-982 (1995).
  • the inventive compounds can be transformed into prodrugs by converting 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 body, most drugs are substrates for chemical reactions that may change their physical properties and biologic effects. These metabolic conversions, which usually affect the polarity of the compound, alter the way in which drugs are distributed in and excreted from the body. However, in some cases, metabolism of a drug is required for therapeutic effect. For example, anticancer drugs of the antimetabolite class must be converted to their active forms after they have been transported into a cancer cell. Since most drugs undergo metabolic transformation of some kind, the biochemical reactions that play a role in drug metabolism may be numerous and diverse. The main site of drug metabolism is the liver, although other tissues may also participate.
  • neural insult refers to any damage to nervous tissue and any disability or death resulting therefrom.
  • the cause of nervous insult may be metabolic, toxic, neurotoxic, iafrogenic, thermal or chemical, and includes without limitation, ischemia, hypoxia, cerebrovascular accident, trauma, surgery, pressure, mass effect, hemmo ⁇ hage, radiation, vasospasm, neurodegenerative disease, infection, Parkinson's disease, amyotrophic lateral sclerosis (ALS), myelination/demyelination process, epilepsy, cognitive disorder, glutamate abnormality and secondary effects thereof.
  • ALS amyotrophic lateral sclerosis
  • neuroprotective refers to the effect of reducing, a ⁇ esting or ameliorating nervous insult, and protecting, resuscitating, or reviving nervous tissue that has suffered nervous insult.
  • preventing neurodegeneration includes the ability to prevent neurodegeneration in patients diagnosed as having a neurodegenerative disease or who are at risk of developing a neurodegenerative disease. The term also encompasses preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease.
  • treating refers to:
  • neural tissue damage resulting from ischemia and reperfusion injury and neurodegenerative diseases includes neurotoxicity, such as seen in vascular stroke and global and focal ischemia.
  • a feature characteristic of many of these transformations is that the metabolic products are more polar than the parent drugs, although a polar drug does sometimes yield a less polar product.
  • the specific secretory mechanisms for anions and cations in the proximal renal tubules and in the parenchymal liver cells operate upon highly polar substances.
  • phenacetin acetophenetidin
  • acetanilide is both mild analgesic and antipyretic agents, but are each transformed within the body to a more polar and more effective metabolite, p- hydroxyacetanilid (acetaminophen), which is widely used today.
  • acetanilid p- hydroxyacetanilid
  • the successive metabolites peak and decay in the plasma sequentially.
  • acetanilid is the principal plasma component.
  • the metabolite acetaminophen concentration reaches a peak.
  • the principal plasma component is a further metabolite that is inert and can be excreted from the body.
  • the plasma concentrations of one or more metabolites, as well as the drug itself, can be pharmacologically important.
  • Phase I or functionalization reactions generally consist of (1) oxidative and reductive reactions that alter and create new functional groups and (2) hydrolytic reactions that cleave esters and amides to release masked functional groups. These changes are usually in the direction of increased polarity.
  • Phase II reactions are conjugation reactions in which the drug, or often a metabolite of the drug, is coupled to an endogenous substrate, such as glucuronic acid, acetic acid, or sulfuric acid.
  • Oxidative deamination (monoamine oxidase and diamine oxidase)
  • the compounds of the present invention exhibit pharmacological activity and are, therefore, useful as pharmaceuticals.
  • the compounds exhibit central nervous and cardiac vesicular system activity. It is understood that tautomeric forms, when possible, are included in the invention.
  • PARP inhibitors are known and, thus, can be synthesized by known methods from starting materials that are known, may be available commercially, or may be prepared by methods used to prepare co ⁇ esponding compounds in the literature. See, for example, Suto et al., "Dihydroiso-quinolinones: The Design and Synthesis of a New Series of Potent Inhibitors of Poly( ADP-ribose) Polymerase", Anticancer Drug Des., 6:107-17 (1991), which discloses processes for synthesizing a number of different PARP inhibitors.
  • the PARP inhibitors used in the composition of the invention will have an IC S . for inhibiting poly( ADP-ribose) polymerase in vitro of about 20 ⁇ M or less, preferably less than about lO ⁇ M, more preferably less than about 1 ⁇ M, or preferably less than about 0.1 ⁇ M, most preferably less than about 0.01 ⁇ M.
  • the PARP inhibitor 3,4-dihydro-5-[4-(l-piperidinyl)butoxy]-l(2 ⁇ T)-isoquinolinone has been reported to inhibit PARP with an IC S0 of 40 nM by Suto et al., cited above.
  • the 6(5_Y)-phenanthridinone sulfonamides and carbamides, such as Example compounds 1-14 below, of this invention are represented by previously defined formula I-V.
  • these 6(5 )- phenanthridinone derivatives can be prepared in a conventional manner as illustrated below by Schemes 1 - 3.
  • the process sequence set forth herein does not present an exact sequence of reactions by which the compound must be made; that is, the sequence of reactions can be rea ⁇ anged in several ways, by means known to those of ordinary skill in the art, to reach the target molecule.
  • Scheme 1 below illustrates schematically the preparation of compounds Example 1 through Example 4.
  • Target compounds-sulfonamides and carbamides can be formed by reaction the amino group of compound 3 with either sulfonyl chloride or carboxylic acid halide derivatives as described in General procedure A. Formation of sulfonamides and carbamides with amino derivatives can be carried out by a variety of conditions known to those of ordinary skill in the art, including reaction with first or secondary amines using pyridine or triethyl amine as base. Typical solvents include chlorinated solvents, various ethers, and dipolar aprotic solvents like DMF.
  • Scheme 2 below illustrates schematically the preparation of compounds Example 10 through Example 12.
  • Monosulfonation of commercially available 6(5i ⁇ )-phenanthridinone 1 can be achieved using chorosulfonic acid neat to give compound 4 in high yield.
  • 6(5 ⁇ T)-phenanthridinone 1 was placed in neat chorosulfonic acid under nitrogen at 0 °C. The resulting mixture is allowed to warm to room temperature, continuously stirred for about 2 days and poured onto 100 g of ice.
  • Amindation of compoimd 4 using primary amines can be carried out by a variety of conditions similar to the process described above from compound 3 to Examples 1-9.
  • final products of compounds Examples 10-12 can be prepared from 6(5/ )-phenanthridinone 1 using General procedure A bellow.
  • the 3-substituted (5_7)phenanthridin-6-one skeleton of this invention can be constructed in a conventional manner using Schmidt method with fluoren-9-one 5 as starting material and sodium azide as an insertion agent in acidic condition (Chida, N.: Ohtsuka. M.: Ogawa, S. Tetrahedron Lett. 1991, 32, 4525).
  • the fluoren-9-one ring may be generically substituted as set forth in the drawing.
  • Such fluoren-9-one starting derivatives are known in the chemistry literature and accessible by processes known to one skilled in the art.
  • This Schmidt method normally generates a pair of regioisomer mixtures 6 and 7, which may be separated and purified by chromatography or crystallization.
  • 3-[6(5 f)Phenant__ridinone]carboxylic acid 6 can be routinely converted to 3- [6(J_T)Phenanthridinone]carbonyl chloride 7 using thionyl chloride neat at 60 °C.
  • Amidation of desired regioisomer of 3-[6(5_Y)phenanthridinone]sulfonyl chloride 6 or 3-[6(5iY)Phenanthridinone]carbonyl chloride 7 with R' substituted primary amine using triethyl amine (TEA) as base provides (5 _)phenanthridin-6-one sulfonamide and carbamide of Example 13-19.
  • N-(5,6-Dihydro-6-oxo-2-phenanthridinyl)-l,3-dihydro-2H-isoindole-2-acetamide Prepared from the compound of Example 2 and isoindoline.
  • 2-chloro-N-(5,6-dihydro- 6-oxo-2-phenanthridinyl)-acetamide 45 mg
  • N-dimethylformamide 10 mL
  • potassium phosphate 68 mg
  • isoindoline 0.025 mL
  • N-(5,6-dihydro-6-oxo-2-phenanthridinyl)-4-mo holineacetamide Prepared from the compound of Example 2 and morpholine.
  • 2-chloro-N-(5,6-dihydro- 6-oxo-2-phenanthridinyl)-acetamide 45 mg
  • N-dimethylformamide 10 mL
  • potassium phosphate 68 mg
  • morpholine 0.017 mL
  • is(2-Hydroxyethyl)an ⁇ no]ethyl]-5,6-dihycto ⁇ Prepared from the compound 2-(6(5i )-phenanthridinone)sulfonyl chloride 4 and N,N-bis(2- hydroxyethyl)ethylenediamine according to General Procedure A. Methylene chloride was used as solvent. A precipitation was formed after stirring the reactant for 20 hours. Purification of the precipitation by crystallization in p-dioxane gave a yellowish solid (40 % yield), mp 210 - 214 °C (dec).
  • N-[2-(4-Morpholinyl)ethyl] 5,6-dihydro-6-oxo-3-phenanthridine carboxamide Prepared from to 3-[6(5_ )Phenanthridinone]carbonyl chloride 7 and l-(2-aminoethyl)morpholine according to General Procedure A. DMF was used as solvent. Purification of compound by crystallization in acetic acid gave a solid (60 % yield), mp 285 -292 °C.
  • the compounds of the present invention may be useful in the free base form, in the form of base salts where possible, and in the form of addition salts, as well as in the free acid form. All these forms are within the scope of this invention. In practice, use of the salt form amounts to use of the base form.
  • Pharmaceutically acceptable salts within the scope of this invention are those derived from mineral acids such as hydrochloric acid and sulfuric acid; and organic acids such as ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, and the like, giving the hydrochloride, sulfonate, ethanesulfonate, benzenesuLfonate, p-toluenesulfonate, and the like respectively, or those derived from bases such as suitable organic and inorganic bases.
  • Examples of pharmaceutically acceptable base addition salts with compounds of the present invention include organic 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.
  • organic bases may include mono-, di-, and trialkylamines, such as methylamine, diethylamine and triethylamine; mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine; amino acids such as arginine, and lysine; guanidine; N-methylglucosamine; N-methylgiucamine; L-glutamine; N-methylpiperazine; mo ⁇ holine; ethylenedianane; N-benzylphenethylamine; tris(hydroxymethyl)antinoethane; and the like.
  • mono-, di-, and trialkylamines such as methylamine, diethylamine and triethylamine
  • mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine
  • amino acids such as arginine, and lysine
  • guanidine N-methylglucosamine
  • N-methylgiucamine N-methylgiucamine
  • the acid addition salts of the basic compounds may he prepared by dissolving the free base of the compounds of the present invention in aqueous or aqueous alcohol solution or other suitable solvents containing the appropriate acid or base and isolating the salt by evaporating the solution, or by reacting the free base of a compound of the present invention with an acid as well as reacting a compound of the present invention having an acid group thereon with a base such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained by concentration of the solution.
  • the compounds of this invention contain one or more asymmetric 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 invention exhibit pharmacological activity and are, therefore, useful as pharmaceuticals.
  • the compounds exhibit central nervous and cardiac vesicular system activity.
  • Other variations and modifications of this invention using the synthetic pathways described above will be obvious to those of ordinary skill in the art.
  • the compounds of the present invention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis; can ameliorate neural or cardiovascular tissue damage, including that following focal ischemia, myocardial infarction, and reperfusion injury; can treat various diseases and conditions caused or exacerbated by PARP activity; can extend or increase the lifespan or proliferative capacity of cells; can alter the gene expression of senescent cells; and can radiosensitize cells.
  • inhibition of PA P activity spares the cells from energy loss, preventing, in the case of neural cells, i ⁇ eversible depolarization of the neurons, and thus, provides neuroprotection. While not being bound to any one particular theory, 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.
  • the present invention further relates to a method of administering a therapeutically effective amount of the above-identified compounds in an amount sufficient to inhibit PARP activity, to treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis, to effect a neuronal activity not mediated by NMDA toxicity, to effect a neuronal activity mediated by NMDA toxicity, to treat neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age-related muscular degeneration, ADDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, inflammation, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis
  • the present invention relates to a method of treating, preventing or inhibiting a neurological disorder in an animal, which comprises administering to said animal a therapeutically effective amount of the above-identified compounds.
  • the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration.
  • Another prefe ⁇ ed embodiment is when the reperfusion injury is a vascular stroke.
  • Yet another prefe ⁇ ed embodiment is when the peripheral neuropathy is caused by Guillain-Barre syndrome.
  • Still another prefe ⁇ ed embodiment is when the demyelinating disease and neurological disorder relates to neurodegeneration.
  • Another prefe ⁇ ed embodiment is when the reperfusion injury is a vascular stroke.
  • Still another prefe ⁇ ed embodiment is when the demyelinating disease is multiple sclerosis.
  • Another prefe ⁇ ed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis.
  • Yet another prefe ⁇ ed embodiment is a method of treating, preventing or inhibiting a cardiovascular disease in an animal, such as angina pectoris, myocardial infarction, cardiovascular ischemia, and cardiovascular tissue damage related to PARP activation, by administering to said animal an effective amount of the compounds of the present invention.
  • a cardiovascular disease in an animal such as angina pectoris, myocardial infarction, cardiovascular ischemia, and cardiovascular tissue damage related to PARP activation
  • the present invention also contemplates the use of compound I, II, III, IV or V for inhibiting PARP activity, for treating, preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis, for treating, preventing or inhibiting a neurological disorder in an animal.
  • the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration.
  • Another prefe ⁇ ed embodiment is when the reperfusion injury is a vascular stroke.
  • Yet another prefe ⁇ ed embodiment is when the peripheral neuropathy is caused by Guillain-Ba ⁇ e syndrome.
  • Still another prefe ⁇ ed embodiment is when the demyelinating disease is multiple sclerosis.
  • Another prefe ⁇ ed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis.
  • the present invention also contemplates the use of compound I, II, III, IV or V in the preparation of a medicament for the treatment of any of the diseases and disorders in an animal described herein.
  • the disease or disorder is a neurological disorder.
  • the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration.
  • Another prefe ⁇ ed embodiment is when the reperfusion injury is a vascular stroke.
  • Yet another prefe ⁇ ed embodiment is when the peripheral neuropathy is caused by Guillain-Ba ⁇ e syndrome. •
  • Still another prefe ⁇ ed embodiment is when the demyelinating disease is multiple sclerosis.
  • Another prefe ⁇ ed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis.
  • preventing neurodegeneration includes the ability to prevent neurodegeneration in patients newly diagnosed as having a neurodegenerative disease, or at risk of developing a new degenerative disease and for preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease.
  • treatment covers any treatment of a disease and/or condition in an animal, particularly a human, and includes:
  • the term "neural tissue damage resulting from ischemia and reperfusion injury” includes neurotoxicity, such as seen in vascular stroke and global and focal ischemia.
  • ischemia relates to localized tissue anemia due to obstruction of the inflow of arterial blood.
  • Global ischemia occurs under conditions in which blood flow to the entire brain ceases for a period of time, such as may result from cardiac a ⁇ est.
  • Focal ischemia occurs under conditions in which a portion of the brain is deprived of its normal blood supply, such as may result from thromboembolytic occlusion of a cerebral vessel, traumatic head injury, edema, and brain tumors.
  • the term "cardiovascular disease” relates to myocardial infarction, angina pectoris. vascular or myocardial ischemia, and related conditions as would be known by those of skill in the art which involve dysfunction of or tissue damage to the heart or vasculature, and especially, but not limited to, tissue damage related to PARP activation.
  • radiosensitizer is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagnetic radiation and/or to promote the treatment of diseases which are treatable with electromagnetic radiation.
  • Diseases which are treatable with electromagnetic radiation include neoplastic diseases, benign and malignant tumors, and cancerous cells. Electromagnetic radiation treatment of other diseases not listed herein are also contemplated by the present invention.
  • electromagnetic radiation and “radiation” as used herein includes, but is not limited to, radiation having the wavelength of 10 "20 to 10° meters.
  • Prefe ⁇ ed embodiments of the present invention employ the electromagnetic radiation of: gamma- radiation (10 "20 to 10 '13 m) x-ray radiation (10 "u to 10 “9 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).
  • the compounds of the invention inhibit PARP activity and, thus, are believed to be useful for treating neural tissue damage, particularly damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in animals.
  • neural tissue refers to the various components that make up the nervous system including, without limitation, neurons, neural support cells, glia. Schwann cells, vasculature contained within and supplying these structures, the central nervous system, the brain, the brain stem, the spinal cord, the junction of the central nervous system with the peripheral nervous system, the peripheral nervous system, and allied structures.
  • an effective therapeutic amount of the compounds and compositions described above are administered to animals to effect a neuronal activity, particularly one that is not mediated by NMDA nemotoxicity.
  • Such neuronal activity may consist of stimulation of damaged neurons, promotion of neuronal regeneration, prevention of neurodegeneration and treatment of a neurological disorder.
  • the present invention further relates to a method of effecting a neuronal activity in an animal, comprising administering an effective amount of the compound of formula I, II , III, IV or V to said animal.
  • neurological disorders that are treatable by the method of using the present invention include, without limitation, trigeminal neuralgia; glossopharyngeal neuralgia; Bell's Palsy; myasthenia gravis; muscular dystrophy; amyotrophic lateral sclerosis; progressive muscular atrophy; progressive bulbar inherited muscular atrophy; herniated, ruptured or prolapsed invertebrate disk syndromes; cervical spondylosis; plexus disorders; thoracic outlet destruction syndromes; peripheral neuropathies such as those caused by lead, dapsone, ticks, po ⁇ hyria, or Guillain-Ba ⁇ e syndrome; Alzheimer's disease; Huntington's Disease and Parkinson's disease.
  • the method of the present invention is particularly useful for treating a neurological disorder selected from the group consisting of: peripheral neuropathy caused by physical injury or disease state; head trauma, such as traumatic brain injury; physical damage to the spinal cord; stroke associated with brain damage, such as vascular stroke associated with hypoxia and brain damage, focal cerebral ischemia, global cerebral ischemia, and cerebral reperfusion injury; demyelinating diseases, such as multiple sclerosis; and neurological disorders related to neurodegeneration, such as Alzheimer's Disease, Parkinson's Disease, Huntington's Disease and amyotrophic lateral sclerosis (ALS).
  • a neurological disorder selected from the group consisting of: peripheral neuropathy caused by physical injury or disease state; head trauma, such as traumatic brain injury; physical damage to the spinal cord; stroke associated with brain damage, such as vascular stroke associated with hypoxia and brain damage, focal cerebral ischemia, global cerebral ischemia, and cerebral reperfusion injury; demyelinating diseases, such as multiple sclerosis; and neurological disorders related to neurodegeneration, such as Alzheimer's Disease, Parkinson
  • Treating Other PAKP-Related Disorders The compounds, compositions and methods of the present invention are particularly useful for treating or preventing tissue damage resulting from cell death or damage due to necrosis or apoptosis.
  • 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 pectoris, myocardial infarction, cardiovascular tissue damage caused by cardiac arrest, cardiovascular tissue damage caused by 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 vasculature, especially, but not limited to, tissue damage related to PARP activation.
  • the methods of the invention are believed to be useful for treating cardiac tissue damage, particularly damage resulting from cardiac ischemia or caused by reperfusion injury in animals.
  • the methods of the invention are particularly useful for treating cardiovascular disorders selected from the group consisting of: coronary artery disease, such as atherosclerosis; angina pectoris; myocardial infarction; myocardial ischemia and cardiac a ⁇ est; cardiac bypass; and cardiogenic shock.
  • coronary artery disease such as atherosclerosis
  • angina pectoris myocardial infarction
  • myocardial ischemia and cardiac a ⁇ est cardiac bypass
  • cardiogenic shock cardiogenic shock.
  • the methods of the invention are particularly helpful in treating the acute forms of the above cardiovascular disorders.
  • the methods of the invention can be used to treat tissue damage resulting from cell damage or death due to necrosis or apoptosis, neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging; to extend the lifespan and proliferative capacity of cells; to alter gene expression of senescent cells: or to radiosens
  • the methods of the invention can be used to treat cancer and to radiosensitize tumor cells.
  • cancer is inte ⁇ reted broadly.
  • the compounds of the present invention can be "anti-cancer agents", which term also encompasses “anti-tumor cell growth agents” and "anti-neoplastic agents”.
  • the methods of the invention are useful for treating cancers and radiosensitizing tumor cells in cancers such as
  • ACTH-producing tumors acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, 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 peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non-Hodgkin's lymphoma, osteosarcoma, ovarian cancer, ovary (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer
  • radiosensitizer is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagnetic radiation and/or to promote the treatment of diseases which are treatable with electromagnetic radiation.
  • Diseases which are treatable with electromagnetic radiation include neoplastic diseases, benign and malignant tumors, and cancerous cells. Electromagnetic radiation treatment of other diseases not listed herein are also contemplated by the present invention.
  • electromagnetic radiation and “radiation” as used herein includes, but is not limited to, radiation having the wavelength of 10 "20 to 10° meters.
  • Prefe ⁇ ed embodiments of the present invention employ the electromagnetic radiation of: gamma- radiation (10 "20 to 10 “13 m) x-ray radiation (10 "n to 10 “9 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).
  • Radiosensitizers are known to increase the sensitivity of cancerous cells to the toxic effects of electromagnetic radiation.
  • hypoxic cell radiosensitizers e.g., 2-nitroimidazole compounds, and benzotriazine dioxide compounds
  • non-hypoxic cell radiosensitizers e.g., halogenated pyrimidines
  • various other potential mechanisms of action have been hypothesized for radiosensitizers in the treatment of disease.
  • radiosensitizers activated by the electromagnetic radiation of x-rays.
  • x-ray activated radiosensitizers include, but are not limited to, the following: metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, E09, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5-iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same.
  • Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the sensitizing agent.
  • photodynamic radiosensitizers include the following, but are not limited to: hematopo ⁇ hyrin derivatives, Photofrin, benzopo ⁇ hyrin derivatives, NPe6, tin etiopo ⁇ hyrin SnET2, pheoborbide-a, bacteriochlorophyll-a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same.
  • Radiosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to: compounds which promote the inco ⁇ oration of radiosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemotherapeutic agents which act on the tumor with or without additional radiation; or other therapeutically effective compounds for treating cancer or other disease.
  • radiosensitizers examples include, but are not limited to: 5-fluorouracil, leucovorin, 5' -amino-5'deoxythymidine, oxygen, carbogen, red cell transfusions, perfluorocarbons (e.g., Fluosol- DA), 2,3-DPG, BW12C, calcium channel blockers, pentoxyfylline, a ⁇ tiangiogenesis compounds, hydralazine, and LBSO.
  • chemotherapeutic agents examples include, but are not limited to: adriamycin, camptothecin, carboplatin, cisplatin, daunorubicin. docetaxel, doxorubicin, interferon (alpha, beta, gamma), interleukin 2, irinotecan. paclitaxel, topotecan, and therapeutically effective analogs and derivatives of the same.
  • compositions of the Invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising (i) a therapeutically effective amount of the compound of formula I, II, III, IV or V and (ii) a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to any carrier, diluent, excipient, suspending agent, lubricating agent, adjuvant, vehicle, delivery system, emulsifier, disintegrant, absorbent, preservative, surfactant, colorant, flavorant, or sweetener.
  • composition of the invention may be administered orally, parenterally, by inhalation spray, adso ⁇ tion, abso ⁇ tion, topically, rectally, nasally, bucally, vaginally, intraventricularly, via an implanted reservoir in dosage formulations containing conventional non-toxic pharmaceutically-acceptable carriers, or by any other convenient dosage form.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, and intracranial injection or infusion techniques.
  • the composition When administered parenterally, the composition will normally be in a unit dosage, sterile injectable form (solution, suspension or emulsion) which is preferably isotonic with the blood of the recipient with a pharmaceutically acceptable carrier.
  • sterile injectable forms are sterile 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 sterile injectable forms may also be sterile injectable solutions or suspensions in non-toxic parenterally-acceptable diluents or solvents, for example, as solutions in 1,3-butanediol.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides, corn, cottonseed, peanut, and sesame oil.
  • Fatty acids such as ethyl oleate, isopropyl myristate, and oleic acid and its glyceride derivatives, including olive oil and castor oil, especially in their polyoxyethylated versions, are useful in the preparation of injectables.
  • These oil solutions or suspensions may also contain long-chain alcohol diluents or dispersants.
  • Sterile saline is a prefe ⁇ ed carrier, and the compounds are often sufficiently water soluble to be made up as a solution for all foreseeable needs.
  • the carrier may contain minor amounts of additives, such as substances that enhance solubility, isotonicity, and chemical stability, e.g., anti-oxidants, buffers and preservatives.
  • Formulations suitable for nasal or buccal administration may comprise about 0.1% to about 5% w/w, for example 1% w/w of active ingredient.
  • the formulations for human medical use of the present invention comprise an active ingredient in association with a pharmaceutically acceptable carrier therefore and optionally other therapeutic ingredient(s).
  • the composition When administered orally, the composition will usually be formulated into unit dosage forms such as tablets, cachets, powder, granules, beads, chewable lozenges, capsules, liquids, aqueous suspensions or solutions, or similar dosage forms, using conventional equipment and techniques known in the art.
  • Such formulations typically include a solid, semisolid, or liquid carrier.
  • Exemplary carriers include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, mineral oil, cocoa butter, oil of theobroma, alginates, tragacanth, gelatin, syrup, methyl cellulose, polyoxyethylene sorbitan monolaurate, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, and the like.
  • composition of the invention is preferably administered as a capsule or tablet containing a single or divided dose of the inhibitor.
  • the composition is administered as a sterile solution, suspension, or emulsion, in a single or divided dose.
  • Tablets may contain carriers such as lactose and corn starch, and/or lubricating agents such as magnesium stearate.
  • Capsules may contain diluents including lactose and dried corn starch.
  • a tablet may be made by compressing or molding the active 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, lubricant, 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 carrier moistened with an inert liquid diluent.
  • compositions of this invention may also be administered rectally in the form of suppositories.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at room temperature, but liquid at rectal temperature, and, therefore, will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax, and polyethylene glycols.
  • compositions and methods of the invention also may utilize controlled release technology.
  • inventive compounds may be inco ⁇ orated into a hydrophobic polymer matrix for controlled release over a period of days.
  • the composition of the invention may then be molded into a solid implant, or externally applied patch, suitable for providing efficacious concentrations of the PARP inhibitors over a prolonged period of time without the need for frequent re-dosing.
  • controlled release films are well known to the art.
  • transdermal delivery systems are transdermal delivery systems.
  • polymers commonly employed for this piupose that may be used in the present invention include nondegradable ethylene-vinyl acetate copolymer an degradable lactic acid-glycolic acid copolymers which may be used externally or internally.
  • Certain hydrogels such as poly(hydroxyethylmethacrylate) or poly(vinylalcohol) also may be useful, but for shorter release cycles than the other polymer release systems, such as those mentioned above.
  • the carrier is a solid biodegradable polymer or mixture of biodegradable polymers with appropriate time release characteristics and release kinetics.
  • 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 period of time without the need for frequent re-dosing.
  • the composition of the present invention can be inco ⁇ orated into the biodegradable polymer or polymer mixture in any suitable manner known to one of ordinary skill in the art and may form a homogeneous matrix with the biodegradable polymer, or may be encapsulated in some way within the polymer, or may be molded into a solid implant.
  • the biodegradable polymer or polymer mixture is used to form a soft "depot" containing the pharmaceutical composition of the present invention that can be administered as a flowable liquid, for example, by injection, but which remains sufficiently viscous to maintain the pharmaceutical composition within the localized area around the injection site.
  • the degradation time of the depot so formed can be varied from several days to a few years, depending upon the polymer selected and its molecular weight.
  • a polymer composition in injectable form even the need to make an incision may be eliminated.
  • a flexible or flowable delivery "depot” will adjust to the shape of the space it occupies with the body with a minimum of trauma to su ⁇ ounding tissues.
  • the pharmaceutical composition of the present invention is used in amounts that are therapeutically effective, and may depend upon the desired release profile, the concentration of the 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 composition in amounts that are therapeutically effective.
  • the compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, welling, or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • the compositions are prepared according to conventional mixing, granulating, or coating methods, and contain about 0.1 to 75% by weight, preferably about 1 to 50% by weight, of the active ingredient.
  • the compounds of the present invention should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier can be effectively administered by an intraventricular route or other appropriate delivery system suitable for administration to the brain.
  • Doses of the compounds preferably include pharmaceutical dosage units comprising an efficacious quantity of active compound.
  • an efficacious quantity is meant a quantity sufficient to inhibit PARP and derive 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 neurodegenerative diseases.
  • the amount required of the active ingredient to achieve a therapeutic effect will vary with the particular compound, the route of administration, the mammal under treatment, and the particular disorder or disease being treated.
  • a suitable systematic dose of a compound of the present invention or a pharmacologically acceptable salt thereof for a mammal suffering from, or likely to suffer from, any of condition as described hereinbefore is in the range of about 0.1 mg/kg to about 100 mg/kg of the active ingredient compound, the most prefe ⁇ ed dosage being about 1 to about 10 mg/kg.
  • a specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the severity of the particular disease being treated and form of administration.
  • the compounds may also be blended with conventional excipients such as binders, including gelatin, pregelatinized starch, and the like; lubricants, such as hydrogenated vegetable oil, stearic acid, and the like; diluents, such as lactose, mannose, and sucrose; disintegrants, such as carboxymethylcellulose and sodium starch glycolate; suspending agents, such as povidone, polyvinyl alcohol, and the like; absorbants, such as silicon dioxide; preservatives, such as methylparaben, propylparaben, and sodium benzoate; surfactants, such as sodium lauryl sulfate, polysorbate 80, and the like; colorants such as F.D.& C. dyes and lakes; flavorants; and sweeteners.
  • binders including gelatin, pregelatinized starch, and the like
  • lubricants such as hydrogenated vegetable oil, stearic acid, and the like
  • diluents such as lactose, mannose
  • 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 described herein.
  • a convenient method to determine IC S0 of a PARP inhibitor compound is a PARP assay using purified recombinant human PARP from Trevigan (Gaithersburg, MD), as follows: The PARP enzyme assay is set up on ice in a volume of 100 microliters consisting of 100 mM Tris-HCl (pH 8.0), 1 mM MgCl_, 28 mM KC1, 28 mM NaCl, 0.1 mg/ml of DNase I activated herring sperm DNA (Sigma, MO), 3.0 micromolar [3H]nicotinamide adenine dinucleotide (470 mci/mmole), 7 micrograms/ml PARP enzyme, and various concentrations of the compounds to be tested.
  • the PARP enzyme assay is set up on ice in a volume of 100 microliters consisting of 100 mM Tris-HCl (pH 8.0), 1 mM MgCl_, 28 mM KC1, 28
  • the reaction is initiated by incubating the mixture at 25°C. After 15 minutes of incubation, the reaction is terminated by adding 500 microliters of ice cold 20% (w/v) trichloroacetic acid. The precipitate formed is transfe ⁇ ed onto a glass fiber filter (Packard Unifilter-GF/B) and washed three times with ethanol. After the filter is dried, the radioactivity is determined by scintillation counting.
  • the compounds of this invention were found to have potent enzymatic activity in the range of a few nM to 20 ⁇ M in IC S0 in this inhibition assay.
  • focal cerebral ischemia assay is useful for determining the PARP inhibiting effects of the compounds of the present invention.
  • the following examples demonstrate that compounds related to those of the present invention are effective in inhibiting PARP activity.
  • Focal cerebral ischemia is produced by cauterization of the right distal MCA (middle cerebral artery) with bilateral temporary common carotid artery occlusion in male Long-Evans rats for 90 minutes. All procedures performed on the animals are approved by the University Institutional Animal Care and Use Committee of the University of Pennsylvania. A total of 42 rats (weights: 230-340 g) obtained from Charles River were used in this study. The animals fasted overnight with free access to water prior to the surgical procedure.
  • the rats were then anesthetized with halothane (4% for induction and 0.8%- 1.2% for the surgical procedure) in a mixture of 70% nitrous 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 (Harvard Apparatus Limited, Kent, U.K.).
  • a catheter (PE-50) was placed into the tail artery, and arterial pressure was continuously monitored and recorded on a Grass polygraph recorder (Model 7D, Grass Instruments, Quincy, Massachusetts). Samples for blood gas analysis (arterial pH.
  • PaO_ and PaC0 2 were also taken from the tail artery catheter and measured with a blood gas analyzer (ABL 30, Radiometer, Copenhagen, Denmark). Arterial blood samples were obtained 30 minutes after MCA occlusion. The head of the animal was positioned in a stereotaxic frame, and a right parietal incision between the right lateral canthus and the external auditory meatus was made. Using a dental drill constantly cooled with saline, a 3 mm bun hole was prepared over the cortex supplied by the right MCA, 4 mm lateral to the sagittal suture and 5 mm caudal to the coronal suture.
  • the dura mater and a thin inner bone layer were kept, care being taken to position the probe over a tissue area devoid of large blood vessels.
  • 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 right parietal cortex was continuously monitored with a laser Doppler flowmeter (FloLab, Moor, Devon, U.K., and Periflux 4001, Perimed, Sweden).
  • Focal cerebral ischemia was produced by cauterization of the distal portion of the right MCA with bilateral temporary common carotid artery (CCA) occlusion by the procedure of Chen et al., "A Model of Focal Ischemic Stroke in the Rat: Reproducible Extensive Cortical Infarction", Stroke 17:738-43 (1986) and/or Liu et al., "Polyethylene Glycol-conjugated Superoxide Dismutase and Catalase Reduce Ischemic Brain Injury", Am. J. Physiol. 256:H589-93 (1989), both of which are hereby inco ⁇ orated by reference.
  • CCA common carotid artery
  • 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 observation of the rostral end of the zygomatic arch at the fusion point using a dental drill, and the dura mater overlying the MCA was cut.
  • the MCA distal to its crossing with the inferior cerebral vein was lifted by a fine stainless steel hook attached to a micromampulator and, following bilateral CCA occlusion, the MCA was cauterized with an electrocoagulator.
  • the bun hole was covered with a small piece of Gelform, and the wound was sutured to maintain the brain temperature within the normal or near- normal range.
  • the animals were given the same doses of the PARP inhibitor as in the pre-treatment. Twenty-four hours after MCA occlusion, the rats were sacrificed with an intraperitoneal injection 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 brain was then sectioned in the coronal plane at 2 mm intervals using a rodent brain matrix (RBM-4000C, ASI Instruments, Wa ⁇ en, Michigan). The brain slices were incubated in phosphate-buffered saline containing 2% 2,3,5-triphenyltetrazolium chloride (TTC) at 37°C for ten minutes.
  • TTC 2,3,5-triphenyltetrazolium chloride
  • the data are expressed as mean + standard deviation. The significance of differences between groups was determined using an analysis of variance (ANOVA) followed by Student's t-test for individual comparisons.
  • MABP mean arterial blood pressure
  • Exemplified compounds of the present invention may be 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 by the institutional Animal Studies Committee, and are in accordance with the PHS Guide for the Care and Use of Laboratory Animals, USDA Regulations, and the AVMA Panel on Euthanasia guidelines.
  • the animals are anesthetized with isofluorane (induction, 3%; maintenance, 1.25% in a mixture of 30% 0 2 and 70% N0 2 through a face mask.
  • the rectal temperature is maintained at 37 °C with a homeothermic blanket (Harvard Apparatus, South Natick, MA).
  • a homeothermic blanket Hard Apparatus, South Natick, MA.
  • an iv catheter is inserted into the left femoral vein and the line run up through the nape of the neck for connection to a tethered swivel (Instech Laboratories, Plymouth Meeting, PA) and remote infusion pump (Stoelting Inc., Wood Dale, IL).
  • the right femoral artery is cannulated for monitoring arterial blood pressure and heart rate and for obtaining blood samples for arterial blood gas.
  • the right middle cerebral artery (MCA) is then exposed by making vertical skin incision midway between the right eye and ear and overlying skull is removed with a dental drill (Chen et al, 1986). After incision of the dura, the artery is coagulated at the level of the inferior cerebral vein with a bipolar cautery unit (Valleylab NS2000, Boulder, CO), and cut to prevent spontaneous reperfusion (Takahashi et al., 1997).
  • CCAs common carotid arteries
  • 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).
  • MCAO MCAO-induced iv bolus
  • 10 mg/kg infused over 5 min a 12 hr continuous infusion of 2 mg/kg/hr (0.3 ml/hr).
  • the animals are removed from the infusion tether, briefly reanesthetized with isofluorane, and the carotid clips are removed .
  • the animal is returned to its warm cage and reconnected to the iv infusion pump for the duration of the experiment.
  • mice are sedated with ketamine and the heads removed by guillotine. Brains are removed, cooled in ice-cold saline, and sliced into 2 mm coronal sections using a rat brain matrice (Harvard 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 (MetaMo ⁇ h, Universal Imaging Co ⁇ ., West Chester, PA).
  • the total volume of infarction in the right hemisphere is calculated by summation of the direct (TTC-negative) and indirect measures of the infarct areas in the component brain slices.
  • Various doses of the compounds of the invention may be tested in this model.
  • the compounds are administered in either a single dose or a series of multiple doses, i.p. or i.v., at different times, both before or after the onset of ischemia.
  • Compounds of the invention are expected to provide protection from ischemia in the range of about 0 to 80%.
  • the experiments of the heart ischemia/reperfusion injury model is performed using female Sprague- Dawley rats weighing 250-3 OOg which are anesthetized with sodium pentobarbital at dose of 65 mg kg intraperitoneally.
  • the rectal temperature is maintained at 37°C by using a homeothermic blanket system
  • the trachea is cannulated and the rat is ventilated with Room Air by using Harvard Rodent Ventilator (Harvard Apparatus, South Natick, MA).
  • the left jugular vein is cannulated with PE-50 tubing for drug delivery.
  • the right carotid artery is cannulated for BP measurement.
  • the heart is exposed by opening the chest at the 4 th left intercostal space.
  • a main left branch of coronary artery (LAD) is occluded by 4-0 silk ligature for 30 min of ischemia and released for 90 min of reperfusion.
  • LAD main left branch of coronary artery
  • 4-0 silk ligature for 30 min of ischemia and released for 90 min of reperfusion.
  • arterial BP and EKG are monitored by Micro-Med Cardiovascular System (Louisville, KY).
  • the LAD coronary artery is re-occluded and about 2 ml of 5% Evans Blue dye is injected through i.v. line to distinguish the 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 sections with 2 mm thickness and stained in 1% TTC solution for 30 min at 37°C. The right ventricle is trimmed off. Infarct area, risk area and total left ventricular area in each face of the section are measured by using an image analysis system (BIOQUANT, Arlington. TN). The infarct size is calculated as the percent total infarct volume of the total risk volume.
  • compounds are administered according to the following three protocols: 1. Single dose of compound is given 60 min prior 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 by i.v. infusion until the 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.
  • the compounds are given in either single or multiple doses, i.p or i.v., at different times before or after the onset of ischemia.
  • the compounds of this invention are expected to have ischemia/reperfusion injury protection in the range of 10 to 40 percent in this assay.
  • the compounds of this invention are expected to protect against ischemia-induced degeneration of rat cortical neurons in vitro and thus may be useful in disorders arising from cerebral ischemia such as stroke, septic shock, or CNS degenerative disorders. They may also be useful in protecting the spinal cord following trauma.
  • the present invention is further directed to a method of prophylactic or therapeutic treatment of heart attack, cardiac a ⁇ est, cardiac bypass, hyperglycemia, diabetes, or risk of damage which comprises administering an effective amount of a compound of the present invention for PARP inhibition in unit dosage form.
  • In vitro radiosensitization may be measured with the use of a human prostate cancer cell line, PC-3s, which are plated in 6 well dishes and grown at monolayer cultures in RPMI1640 supplemented with 10% FCS. The cells are maintained at 37°C in 5% C0 2 and 95% air. The cells are exposed to a dose response (0.1 mM to 0.1 ⁇ M) of 3 different PARP inhibitors prior to i ⁇ adiation at one sublethal dose level. For all treatment groups, the six well plates are exposed at room temperature in a Seifert 250kV/15mA i ⁇ adiator with a 0.5 mm Cu/1 mm. Cell viability is examined by exclusion of 0.4% trypan blue.
  • Dye exclusion is assessed visually by microscopy and viable cell number was calculated by subtracting the number of cells from the viable cell number and dividing by the total number of cells.
  • Cell proliferation rates are calculated by the amount of 3 H-thymidine inco ⁇ oration post-i ⁇ adiation.
  • the PARP inhibitors are expected to radiosensitize the cells.
  • Gene expression alteration may be measured with human fibroblast BJ cells which, at Population Doubling (PDL) 94, are plated in regular growth medium and then changed to low serum medium to reflect physiological conditions described in Linskens, et al., Nucleic Acids Res. 23 : 16:3244-3251 (1995).
  • a medium of DMEM/199 supplemented with 0.5% bovine calf serum is used.
  • the cells are treated daily for 13 days.
  • the control cells are treated with and without the solvent used to administer the PARP inhibitor.
  • the untreated old and young control cells are tested for comparison.
  • RNA is prepared from the treated and control cells according to the techniques described in PCT Publication No. 96/13610 and Northern blotting is conducted.
  • Probes specific for senescence-related genes are analyzed, and treated and control cells compared. In analyzing the results, the lowest level of gene expression is arbitrarily set at 1 to provide a basis for comparison.
  • Three genes particularly relevant to age-related changes in the skin are collagen, collagenase and elastin. West, Arch. Derm. 130:87-95 (1994).
  • Elastin expression of the cells treated with the PARP inhibitor is expected to be significantly increased in comparison with the control cells. Elastin expression should be significantly higher in young cells compared to senescent cells, and thus treatment with 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 treatment with the PARP inhibitors.
  • Thermal hyperalgesia to radiant heat is assessed by using a paw-withdrawal test.
  • the rat is placed in a plastic cylinder on a 3-mm thick glass plate with a radiant heat source from a projection bulb placed directly under the plantar surface of the rat's hindpaw.
  • the paw-withdrawal latency is defined as the time elapsed from the onset of radiant heat stimulation to withdrawal of the rat's hindpaw.
  • Mechanical hyperalgesia is assessed by placing the rat in a cage with a bottom made of perforated metal sheet with many small square holes. Duration of paw-withdrawal is recorded after pricking the mid-plantar surface of the rat's hindpaw with the tip of a safety pin inserted through the cage bottom.
  • Mechano-allodynia 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 slowly until it bends.
  • a threshold force of response is defined as the first filament in the series to evoke at least one clear paw- withdrawal out of five applications.
  • MSU crystals monosodium mate
  • Deposition of crystals of monosodium mate (MSU crystals) in the joint articular space is the etiological cause of inflammatory pathologies such as gout and pseudogout.
  • these inflammatory diseases are associated with oedema and erythema of the joints with consequently severe pain.
  • a strong infiltration of leucocytes in the intraarticular and periarticular space leading to: 1) acute, episodic articular and periarticular inflammation, and 2) chronic articular changes, are also characteristic of this pathology. It has long been clear that neutrophils are the predominant cell type recovered from these inflammatory joints (Dieppe et al., (1979). Synovial fluid crystals. Q. J. Med.
  • Neutropliils have been shown to be activated by MSU crystals, releasing an array of mediators that may be, in part, responsible for the local and systemic inflammatory manifestations found in crystal-induced joint disorders.
  • the crystals interact with neutrophils leading to the release of lysomal enzymes (Hoffstein et al., 1975, Arth. Rheum. 18: 153-165), release of oxygen derived free radicals (Simchowitz et al., 1982, Arth. Rheum. 25: 181-188; Abramson et ⁇ /., 1982, Arthr Rheum.
  • MSU crystals have been shown to release the cytokine interleukin-l ⁇ (IL-l ⁇ ) from human neutrophils, adding this stimulus to a list of others that also release this cytokine, such as zymosan, LPS, phorbol esters, granulocyte macrophage-colony stimulating hormone (GM-CSF) and TNF-alpha. Furthermore it has also been shown that human monocytes and synoviocytes can synthesise and release various cytokines such as IL-6 and D -8 (Guerne et al, 1989, Arth. Rheum. 32: 1443-1452; Terkeltaub et al, 1991, Arth. Rheum. 34: 894-903). In addition, colchicine selectively inhibits MSU crystal- and TNF-G induced release of IL-l ⁇ (Roberge et al, 1994, J. Immunol. 152: 5485-5494).
  • IL-l ⁇ and TNF-alpha may be critical in mediating the rapid up-regulation of the major endothelial ligand for neutrophils.
  • IL-l ⁇ and TNF-alpha may be critical in mediating the rapid up-regulation of the major endothelial ligand for neutrophils.
  • rapid and prolonged expression of E-selectin in response to injection of urate crystals has been demonstrated in pig skin (Chapman et al, 1996, Br. J. Rheumatol. 35: 323-334).
  • the release of cytokines, chemokines and products of the arachidonic acid cascade system lead to the recruitment of neutrophils in this pathology, and inhibition of these leads to an attenuation of the pathology.
  • the following gout model was used to test a PARP inhibitor according to the present invention.
  • mice Male outbread Swiss albino mice (20-22 g body weight) were purchased from Banton and Kingsman (T.O. strain; Hull, Humberside) and maintained on a standard chow pellet diet with tap water ad libitum and a 12:00 h light /dark cycle. All animals were housed for 1 week prior to experimentation to allow body weight to reach 28-30 g. l,llb-dihydrobenzopyrano[4,3,2-_fe ]isoquinolin-l-one was dissolved in 100% DMSO at room temperature at a concentration of 45 mg in 2 ml.
  • mice received a single dose co ⁇ esponding to 45 mg/2 ml/kg (e.g. 60 ⁇ l for a mouse of 30 g).
  • Control mice received DMSO at 2 ml/kg i.p.
  • MSU crystal-induced neutrophil recruitment was tested as follows. In all cases, mice were treated 1 h after the treatment noted above, with MSU crystals. A homogenous suspension of MSU crystals was obtained by a 30 min rotation. Peritonitis was induced by injection of 3 mg MSU crystals in 0.5 ml PBS (0.1 M, pH 7.4), and the recruitment of neutrophils into the cavity evaluated at the 6 h time point (Getting et al, 1997, J. Pharmacol. Exp. Ther. 283: 123-130). Animals were then euthanised by C0 2 exposure and the peritoneal cavity washed with 3 ml of PBS supplemented with 3 mM EDTA and 25 U/ml heparin.
  • mice were left untreated (group A), received vehicle DMSO (2 ml/kg i.p. ; group B) or 1, 1 lb- dihydrobenzopyrano[4,3,2--/e ]isoquinolin-l-one (45 mg/kg i.p.; group C), 1 h prior to peritoneal injection of 3 mg MSU crystals at time 0.
  • Neutrophil influx in the peritoneal cavity was measured at the 6 h time-point after collection of the lavage fluids and specific staining as described in the experimental section. Values for neutrophil numbers are Iff per mouse.

Abstract

Compounds of Formula (1) wherein: one of R3 and R4 is -QP and the other of R3 and R4 is one of hydrogen, methyl, trifluoromethyl, nitro, amino, halogen and I-piperazine. Q is one of (A) and (B) and P is Z-(N(R5R6)), Z or a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring; A is carbon or S=O; X is O,S,N or an N-substituted amino acid; when X is O or S, then Y is absent. When X is N, then Y is hydrogen. C1-C6 straight or branched chain alkyl, optionally substituted alkoxy or alkyl amino, or Y and Z are taken together to form a 5,6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring. Pharamaceutical compositions, and methods of using the disclosed compounds for inhibiting PARP.

Description

SULFONAMIDE AND CARBAMIDE DERIVATIVES OF 6(5#)PHENANTHRIDINONES AND THEIR USES
The present application claims benefit of the filing of U.S. Provisional Application No. 60/205,259, filed May 19, 2000, the entire contents of which is incorporated by reference herein. The present invention relates to inhibitors of the nuclear enzyme poly(adenosine 5'-diphospho-ribose) polymerase ["poly(ADP-ribose) polymerase" or "PARP", which is also referred to as ADPRT (NAD:protein (ADP-ribosyi transferase (polymersing)) and PARS (poly(ADP-ribose) synthetase) and provides compounds and compositions containing the disclosed compounds. Moreover, the present invention 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 ischemic stroke, head trauma or spinal cord injury; neurological disorders and neurodegenerative 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, myocardial infarction; to treat other conditions and/or disorders such as, for example, age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, 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 any of the kidney, liver, renal, pulmonary, retinal, pancreatic and/or skeletal muscle systems), acute autoimmune thyroiditis, muscular dystrophy, osteoarthritis, osteoporosis, chronic and acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), local and/or remote endothelial 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 in the generation of oxidants, proinflammatory mediators and/or cytokines, and general mediators of leukocyte infiltration, calcium ion overload, phospholipid peroxidation, impaired nitric oxide metabolism and/or reduced ATP production; to alter gene expression of senescent cells; or to radiosensitize hypoxic tumor cells.
PARP (EC 2.4.2.30), also known as PARS (for poly(ADP-ribose) synthetase), or ADPRT (for NAD:protein (ADP-ribosyl) transferase (polymerising)) is a major nuclear protein of 116 kDa. It is mainly present in almost all eukaryotes. The enzyme synthesizes poly(ADP-ribose), a branched polymer that can consist of over 200 ADP-ribose units from NAD. The protein acceptors of poly(ADP-ribose) are directly or indirectly involved in maintaining DNA integrity. They include histones, topoisomerases, DNA and RNA polymerases, DNA ligases, and Ca2+- and Mg2+-dependent endonucleases. PARP protein is expressed at a high level in many tissues, most notably in the immune system, heart, brain and germ-line cells. Under normal physiological conditions, there is minimal PARP activity. However, DNA damage causes an immediate activation of PARP by up to 500-fold. Among the many functions attributed to PARP is its major role in facilitating DNA repair by ADP-ribosylation and therefore co-ordinating a number of DNA repair proteins. As a result of PARP activation, NAD levels significantly decline. While many endogenous and exogenous agents have been shown to damage DNA and activate PARP, peroxynitrite, formed from a combination of nitric oxide (NO) and superoxide, appears to be a main perpetrator responsible for various reported disease conditions in vivo, e.g., during shock, stroke and inflammation
Extensive PARP activation leads to severe depletion of NAD in cells suffering from massive DNA damage. The short life of poly(ADP-ribose) (half-life < 1 min) results in a rapid turnover rate. Once poly(ADP-ribose) is formed, it is quickly degraded by the constitutively active poly(ADP-ribose) glycohydrolase (PARG), together with phosphodiesterase and (ADP-ribose) protein lyase. PARP and PARG form a cycle that converts a large amount of NAD to ADP-ribose. In less than an hour, over-stimulation of PARP can cause a drop of NAD and ATP to less than 20% of the normal level. Such a scenario is especially detrimental during ischaemia when deprivation of oxygen has already drastically compromised cellular energy output. Subsequent free radical production during reperfusion is assumed to be a major cause of tissue damage. Part of the ATP drop, which is typical in many organs during ischaemia and reperfusion, could be linked to NAD depletion due to poly( ADP-ribose) turnover. Thus, PARP or PARG inhibition is expected to preserve the cellular energy level to potentiate the survival of ischaemic tissues after insult. Poly( ADP-ribose) synthesis is also involved in the induced expression of a number of genes essential for inflammatory response. PARP inhibitors suppress production of inducible nitric oxide synthase (iNOS) in macrophages, P-type selectin and intercellular adhesion molecule- 1 (ICAM-1) in endothelial cells. Such activity underlies the strong anti-inflammation effects exhibited by PARP inhibitors. PARP inhibition is able to reduce necrosis by preventing translocation and infiltration of neutrophils to the injured tissues. (Zhang, J. "PARP inhibition: a novel approach to treat ischaemia/reperfusion and inflammation-related injuries", Chapter 10 in Emerging Drugs (1999) 4: 209-221 Ashley Publications Ltd., and references cited therein.)
PARP production is activated by damaged DNA fragments which, once activated, catalyzes the attachment of up to 100 ADP-ribose units to a variety of nuclear proteins, including histones and PARP itself. During major cellular stresses the extensive activation of PARP can rapidly lead to cell damage or death through depletion of energy stores. As four molecules of ATP are consumed for every molecule of NAD (the source of ADP-ribose and PARP substrate) regenerated, NAD is depleted by massive PARP activation and, in the efforts to re-synthesize NAD, ATP may also be depleted.
It has been reported that PARP activation plays a key role in both NMD A- and NO-induced neurotoxicity. This has been demonstrated in cortical cultures and in hippocampal slices wherein prevention of toxicity is directly correlated to PARP inhibition potency (Zhang et al., "Nitric Oxide Activation of Poly(ADP- Ribose) Synthetase in Neurotoxicity", Science, 263:687-89 (1994) and Wallis et al., "Neuroprotection Against Nitric Oxide Injury with Inhibitors of ADP-Ribosylation", NeuroReport, 5:3, 245-48 (1993)). The potential role of PARP inhibitors in treating neurodegenerative diseases and head trauma has thus been recognized even if the exact mechanism of action has not yet been elucidated (Endres et al., "Ischemic Brain Injury is Mediated by the Activation of Poly(ADP-Ribose)Polymerase", J. Cereb. Blood Flow Metabol., 17: 1143-51 (1997) and Wallis et al., "Traumatic Neuroprotection with Inhibitors of Nitric Oxide and ADP-Ribosylation, Brain Res., 710:169-77 (1996)).
Similarly, it has been demonstrated that single injections of PARP inhibitors have reduced the infarct size caused by ischemia and reperfusion of the heart or skeletal muscle in rabbits. In these studies, a single injection of 3-amino-benzamide (10 mg/kg), either one minute before occlusion or one minute before reperfusion, caused similar reductions in infarct size in the heart (32-42%) while 1,5-dihydroxyisoquinoline (1 mg/kg), another PARP inhibitor, reduced infarct size by a comparable degree (38-48%). Thiemermann et al., "Inhibition of the Activity of Poly(ADP Ribose) Synthetase Reduces Ischemia-Reperfusion Injury 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 NMDA receptor stimulation), reactive oxygen intermediates, amyloid β - protein, N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or its active metabolite N-methyl-4- phenylpyridine (MPP*), which participate in pathological conditions such as stroke, Alzheimer's disease and Parkinson's disease. Zhang et al., "Poly(ADP-Ribose) Synthetase Activation: An Early Indicator of Neurotoxic DNA Damage", J. Neurochem., 65:3, 1411-14 (1995). Other studies have continued to explore the role of PARP activation in cerebellar granule cells in vitro and in MPTP neurotoxicity. Cosi et al., "Poly(ADP-Ribose) Polymerase (PARP) Revisited. A New Role for an Old Enzyme: PARP Involvement in Neurodegeneration and PARP Inhibitors as Possible Neuroprotective Agents", Ann. N. Y. Acad. Sci., 825:366-79 (1997); and Cosi et al., "Poly(ADP-Ribose) Polymerase Inhibitors Protect Against MPTP-induced Depletions of Striatal Dopamine and Cortical Noradrenaline in C57B1/6 Mice", Brain Res., 729:264-69 (1996). Excessive neural exposure to glutamate, which serves as the predominate central nervous system neurotransmitter and acts upon the N- methyl-D-aspartate (NMDA) receptors and other subtype receptors, most often occurs as a result of stroke or other neurodegenerative processes. Oxygen deprived neurons release glutamate in great quantities during ischemic brain insult such as during a stroke or heart attack. This excess release of glutamate in turn causes over-stimulation (excitotoxicity) of N-methyl-D-aspartate (NMDA), 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 ultimately results in cell damage or death via the production of proteases, lipases and free radicals. Excessive activation of glutamate receptors has been implicated in various neurological diseases and conditions including epilepsy, stroke, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's disease, schizophrenia, chronic pain, ischemia and neuronal loss following hypoxia, hypoglycemia, ischemia, trauma, and nervous insult. Glutamate exposure and stimulation has also been implicated as a basis for compulsive disorders, particularly drug dependence. Evidence includes findings in many animal species, as well as in cerebral cortical cultures treated with glutamate or NMDA, that glutamate receptor antagonists (i.e., compounds which block glutamate from binding to or activating its receptor) block neural damage following vascular stroke. Dawson et al., "Protection of the Brain from Ischemia", CerebrovascuJar Disease, 319-25 (H. HuntBatjer ed., 1997). Attempts to prevent excitotoxicity by blocking NMDA, AMP A, Kainate and MGR receptors have proven difficult because each receptor has multiple sites to which glutamate may bind and hence finding an effective mix of antagonists or universal antagonist to prevent binding of glutamate to all of the receptor and allow testing of this theory, has been difficult. Moreover, many of the compositions that are effective in blocking the receptors are also toxic to animals. As such, there is presently no known effective treatment for glutamate abnormalities.
The stimulation of NMDA receptors by glutamate, for example, activates the enzyme neuronal nitric oxide synthase (nNOS), leading to the formation of nitric oxide (NO), which also mediates neurotoxicity. NMDA neurotoxicity may be prevented by treatment with nitric oxide synthase (NOS) inhibitors or through targeted genetic disruption of nNOS in vitro. Dawson et al., "Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures", Proc. Natl. Acad. Sci. USA, 88:6368-71 (1991); and Dawson et al.. "Mechanisms of Nitric Oxide-mediated Neurotoxicity in Primary Brain Cultures", J. Neurosci., 13:6, 2651-61 (1993), Dawson et al., "Resistance to Neurotoxicity in Cortical Cultures from Neuronal Nitric Oxide Synthase-Deficient Mice", J. Neurosci., 16:8, 2479-87 (1996), Iadecola, "Bright and Dark Sides of Nitric Oxide in Ischemic Brain Injury", Trends Neurosci., 20:3, 132-39 (1997), Huang et al.. "Effects of Cerebral Ischemia in Mice Deficient in Neuronal Nitric Oxide Synthase", Science, 265:1883-85 (1994), Beckman et al., "Pathological Implications of Nitric Oxide, Superoxide and Peroxynitrite Formation", Biochem. Soc. Trans., 21:330-34 (1993), and Szabό 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 Peroxynitrite", Proc. Natl. Acad. Sci. USA, 93:1753-58 (1996).
It is also known that PARP inhibitors, such as 3 -amino benzamide, affect DNA repair generally in response, for example, to hydrogen peroxide or gamma-radiation. Cristovao et al., "Effect of a Poly(ADP- Ribose) Polymerase Inhibitor on DNA Breakage and Cytotoxicity Induced by Hydrogen Peroxide and γ-
Radiation," Terato., Carcino., and Muta., 16:219-27 (1996). Specifically, Cristovao et al. observed a PARP- dependent recovery 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 preventing tumor cells from recovering from potentially lethal damage of DNA after radiation therapy, presumably by their ability to prevent DNA repair. U. S. Patent Nos. 5,032,617; 5,215,738; and 5,041,653. Evidence also exists that PARP inhibitors are useful for treating inflammatory bowel disorders, such as colitis. Salzman et al., "Role of Peroxynitrite and Poly(ADP-Ribose)Synthase Activation Experimental Colitis," Japanese J. Pharm., 75, Supp. 1:15 (1997). Specifically, Colitis was induced in rats by intraluminal administration of the hapten trinitrobenzene sulfonic acid in 50% ethanol. Treated rats received 3- aminobenzamide, a specific inhibitor of PARP activity. Inhibition of PARP activity reduced the inflammatory response and restored the morphology and the energetic status of the distal colon. See also, Southan et al., "Spontaneous Rearrangement of Aminoalkylithioureas into Mercaptoalkylguanidines, a Novel Class of Nitric Oxide Synthase Inhibitors with Selectivity Towards the Inducible Isoform", Br. J. Pharm., 117:619-32 (1996); and Szabό et al., "Mercaptoethylguanidine and Guanidine Inhibitors of Nitric Oxide Synthase React with Peroxynitrite and Protect Against Peroxynitrite-induced Oxidative Damage", J. Biol. Chem., 272:9030-36 (1997).
Evidence also exists that PARP inhibitors are useful for treating arthritis. Szabό et al., "Protective Effects of an Inhibitor of Poly(ADP-Ribose)Synthetase in Collagen-Induced Arthritis," Japanese J. Pharm., 75, Supp. 1:102 (1997); Szabό 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 Peroxynitrite," Proc. Natl. Acad. Sci. USA, 93:1753-58 (March 1996); and Bauer et al., "Modification of Growth Related Enzymatic Pathways and Apparent Loss of Tumorigenicity of a ras-transformed Bovine Endothelial Cell Line by Treatment with 5-Iodo-6-amino-l,2-benzopyrone (INH2BP)", Intl. J. Oncol., 8:239-52 (1996); and Hughes et al., "Induction of T Helper Cell Hyporesponsiveness in an Experimental Model of Autoimmunity by Using Nonmitogenic Anti-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-Ribose)Polymerase Gene Affects Oxygen Radical and Nitric Oxide Toxicity in Islet Cells," J. Biol. Chem., 270:19, 11176-80 (May 1995). Heller et al. used cells from mice with inactivated PARP 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 have been shown to be useful for treating endotoxic shock or septic shock. Zingarelli et al., "Protective Effects of Nicotinamide Against Nitric Oxide-Mediated Delayed Vascular Failure in Endotoxic Shock: Potential Involvement of PolyADP Ribosyl Synthetase," Shock, 5:258-64 (1996), suggests that inhibition of the DNA repair cycle triggered by poly(ADP ribose) synthetase has protective effects against vascular failure in endotoxic shock. Zingarelli et al. found that nicotinamide protects against delayed, NO-mediated vascular failure in endotoxic shock. Zingarelli 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, triggered by poly(ADP ribose) synthetase. Cuzzocrea, "Role of Peroxynitrite and Activation of Poly(ADP-Ribose) Synthetase in the Vascular Failure Induced by Zymosan-activated Plasma," Brit. J. Pharm., 122:493-503 (1997). PARP inhibitors have been used to treat cancer. Suto et al., "Dihydroisoquinolinones: The Design and Synthesis of a New Series of Potent Inhibitors of Poly(ADP-Ribose) Polymerase", 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(5_Y)-Phenanthridinone, an Inhibitor of Poly (ADP-ribose) Polymerase, on Cultured Tumor Cells", Oncol. Res., 6:9, 399-403 (1994), discusses the inhibition of PARP activity, reduced proliferation of tumor cells, and a marked synergistic effect when tumor cells are co-treated with an alkylating drug.
Still another use for PARP inhibitors is the treatment of peripheral nerve injuries, and the resultant pathological pain syndrome known as neuropathic pain, such as that induced by chronic constriction injury (CCI) of the common sciatic nerve and in which transsynaptic alteration of spinal cord dorsal horn characterized by hyperchromatosis of cytoplasm and nucleoplasm (so-called "dark" neurons) occurs. 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, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence, age-related muscular degeneration, immune senescence, AIDS, and other immune senescence diseases; and to alter gene expression of senescent cells. WO 98/27975.
Large numbers of known PARP inhibitors have been described in Banasik et al, "Specific Inhibitors of Poly(ADP-Ribose) Synthetase and Mono(ADP-Ribosyl)-Transferase", J. Biol. Chem., 267:3, 1569-75 (1992), and in Banasik et al., "Inhibitors and Activators of ADP-Ribosylation Reactions", Molec. Cell. Biochem.,
138:185-97 (1994). However, effective use of these PARP inhibitors, in the ways discussed above, has been limited by the concurrent 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 effective and potent PARP inhibitors which produce minimal side- effects. 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 invention are specifically useful in ameliorating, treating and/or preventing neural tissue or cell damage, including that following focal ischemia and reperfusion injury. Generally, inhibition of PARP activity spares the cell from energy loss, preventing irreversible depolarization of the neurons and, thus, provides neuroprotection. While not wishing to be bound by any mechanistic theory, the inhibition of PARP activity by use of the compounds, compositions and methods of the present invention is believed to protect cells, tissue and organs by protection against the ill-effects of reactive free radicals and nitric oxide. The present invention therefore also provides methods of treating and/or preventing cells, tissue and/or organs from reactive free radical and/or nitric oxide induced damage or injury. SUMMARY OF THE INVENTION
The present invention provides 6(5_Y)p__enan- or (J_Y)phenanthridin-6-one compounds which inhibit poly(ADP-ribose) polymerase ("PARP"), compositions containing these compounds and methods for making and using these PARP inhibitors to treat, prevent and/or ameliorate the effects of the conditions described herein.
The compounds of the present invention are broadly described by the following Formula I:
Figure imgf000008_0001
(I)
or a pharmaceutically acceptable salt, hydrate, prodrug, or mixtures thereof, wherein:
Ri hydrogen or halogen;
R2 is hydrogen, hydroxyl, amino, nitroso, methyl, amino methyl or carboxylic acid, preferably Ri and R2 are not both hydrogen; one of R3 and R4 is -QP and the other of R3 and R4 is one of hydrogen, methyl, trifluoromethyl, nitro, amino, halogen and 1-piperazine, wherein Q is one of
Figure imgf000008_0002
P is Z-(N(R5R<;)), such as Z-N(R5R<5), Z or a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of O, N and S;. wherein A is carbon or S=0, X is O, S , N or an N-substituted amino acid; provided that, when X is O or S, then Y is absent, when X is N, then Y is hydrogen, -Cβ straight or branched chain alkyl, optionally substituted alkoxy or alkyl amino, or Y and Z are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring which contains 1, 2 or 3 heteroatoms selected from O, N, S and mixtures combination;
Z is hydrogen, a direct bond, a carbonyl, an optionally substituted -Cs straight or branched chain alkyl, cycloalkyl, carboxy, optionally substituted Cι-C6 ether, aryl or heteroaryl, alkylalkenyl, alkynyl, alkylhalo, straight or branched chain or CH2COOH, provided that when P is Z, Z is not hydrogen and R5 and Re are independently hydrogen, lower alkyl, lower alkenyl, lower alkanol, heterocycle, heteroaryl, alkoxy, aryloxy, alkylamino, arylamino. CH2COOH, or R5 and Re taken together form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic or cycloalkyl ring containing 1, 2 or 3 heteroatoms selected from O, N, S and combinations thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the distribution of the cross-sectional infarct area at representative levels 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-dihydro-5-[4- (l-piperidinyl)-botoxyl] -1 (2_7)-isoquinolinone.
Figure 2 shows the effect of intraperitoneal administration of 3,4-dihydro-5-[4-(l-piperidinyl)-butoxy]- l(2/!_)-isoquinolinone on the infarct volume.
DETAILED DESCRIPTION
The present invention pertains to compounds, pharmaceutical compositions containing the same, methods of using the same, and process of making the same, wherein such compounds are useful as inhibitors of poly( ADP-ribose) polymerase (PARP). As such, they treat or prevent neural tissue 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 the lifespan and proliferative capacity of cells and thus can be used to treat or prevent diseases associated therewith; they alter gene expression of senescent cells; and they radiosensitize hypoxic tumor cells. Preferably, the 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 thought 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 PARP activation. The present invention provides compounds which inhibit poly(ADP-ribose) polymerase ("PARP"), compositions containing these compounds and methods for using these PARP inhibitors to treat, prevent and/or ameliorate the effects of the conditions described herein. In one embodiment, the present invention provides compounds of Formula I:
Figure imgf000010_0001
(I)
or a pharmaceutically acceptable salt, hydrate, prodrug, or mixtures thereof, wherein:
Ri hydrogen or halogen;
R2 is hydrogen, hydroxyl, amino, nitroso, methyl, amino methyl or carboxylic acid, preferably Ri and R2 are not both hydrogen; one of R3 and R4 is -QP and the other of R3 and R4 is one of hydrogen, methyl, trifluoromethyl, nitro, amino, halogen and 1-piρerazine, wherein Q is one of
Figure imgf000010_0002
P is Z-N(R5Rg), Z or a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of O, N and S;. wherein A is carbon or S=0,
X is O, S , N or an N-substituted amino acid; provided that, when X is O or S, then Y is absent, when X is N, then Y is hydrogen, Cι-C6 straight or branched chain alkyl, alkoxy or alkyl amino, or Y and Z are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring which contains 1, 2 or 3 heteroatoms selected from O, N, S and mixtures combination;
Z is hydrogen, a direct bond, a carbonyl or an optionally substituted Cι-C5 straight or branched chain alkyl, alkenyl, alkynyl, alkylhalo, or CH2COOH, provided that when P is Z, Z is not hydrogen and
R5 and Re are independently hydrogen, lower alkyl, lower alkenyl, lower alkanol, heterocycle, heteroaryl, alkoxy, aryloxy, alkylamino, arylamino, CH2COOH, or R5 and Re taken together form a 5, 6 or 7 membered substituted or unsubstituted aryl, heteroaryl, heterocyclic or cycloalkyl ring containing 1, 2 or 3 heteroatoms selected from O, N, S and combinations thereof.
In yet a further embodiment, the present invention provides the following compounds of Formulas (II), (III), (IV) and (V):
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000012_0001
Figure imgf000012_0002
wherein Ri-R*;, A and Z are as defined above.
In one embodiment, the compounds of the invention are described by the compound of Formula I,
wherein R2 is hydrogen, Q is
Figure imgf000012_0003
, and X is N. Preferred compounds of this embodiment are further defined by A as carbon and P as Z-N R5R5), more preferably where R5 and 5 form a 5 or 6 membered substituted or unsubstituted heterocycle or heteroaryl. Alternative compounds of the preferred compounds of this embodiment are further defined by R5 and Re being independently selected from hydrogen, lower alkyl, alkoxy, alkylamino and CH2COOH. Preferably one of R3 or i is hydrogen. In another embodiment, the compounds of the invention are described by the compound of Formula I,
wherein R2 is hydrogen, Q is
Figure imgf000013_0001
, and X is N and A as S=0. Preferred embodiments include compounds wherein and P is a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of 0, N and S. Z in these compounds is preferably and aromatic ring containing 0 heteroatoms. Preferably one of R3 or ( is hydrogen.
In a further embodiment, the compounds of the invention are described by the compound of Formula I,
wherein R2 is hydrogen, Q is
Figure imgf000013_0002
, and X is N. Preferred embodiments include compounds wherein and Z and Y are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring, preferably a non-aromatic ring. Alternatively prefeπed compounds of this embodiment include compounds wherein P is Z-N(R5Rs). Preferably one of R3 or R4 is hydrogen.
Compositions containing these prefeπed and alternate embodiments and methods of making and using the same, as described herein, are also prefeπed
Preferably, the compounds of the invention exhibit an IC50 for inhibiting PARP in vitro, as measured by the methods described herein, of about 20 μM or less, preferably less than about lOμM, more preferably less than about 1 μM, or less than O.lμM, most preferably less than about 0.01 μM.
Prefeπed embodiments of the present invention include the following compounds (where compound numbers are shown next to each compound), and neutral forms thereof, where appropriate:
Figure imgf000013_0003
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000017_0001
Figure imgf000018_0001
15
Figure imgf000018_0002
Figure imgf000018_0003
Figure imgf000019_0001
3 19
Figure imgf000019_0002
Figure imgf000019_0003
Figure imgf000020_0001
Figure imgf000020_0002
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000026_0002
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Broadly, the compounds and compositions of the present invention 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 invention can be used to extend the lifespan and proliferative capacity of cells and thus can be used to treat or prevent diseases associated therewith; they alter gene expression of senescent cells; and they radiosensitize hypoxic tumor cells. Preferably, the compounds and compositions of the invention can be used to 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. The compounds of the present invention are not limited to being useful in treating glutamate mediated neurotoxicity and/or NO-mediated biological pathways. Further, the compounds of the invention can be used to treat or prevent other tissue damage related to PARP activation, as described herein. The present invention provides compounds which inhibit the in vitro and/or in vivo polymerase activity of poly( ADP-ribose) 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 activity of poly( ADP-ribose) polymerase (PARP) in any of solutions, cells, tissues, organs or organ systems. In one embodiment, the present invention provides methods of limiting or inhibiting PARP activity in a mammal, such as a human, either locally or systemically. The present invention provides methods to treat and/or prevent diseases, syndromes and/or conditions exacerbated by or involving the increased generation of PARP. These methods involve application or administration of the compounds of the present invention to cells, tissues, organs or organ systems of a person in need of such treatment or prevention.
In one embodiment, the present invention provides methods 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 during cardiac aπest when the heart, once prevented from receiving blood, begins to reperfuse and these methods involve administration of the compounds and compositions of the present invention preferably prior 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, cardiovascular disorders
In another embodiment, the present invention provides in vitro or in vivo methods to extend or increase the lifespan and/or proliferation capacity of cells and thus also methods to treat and/or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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 hypoxic tumor cells to render the tumor cells more susceptible to radiation therapy and thereby to prevent the tumor cells from recovering from potentially lethal damage of DNA after radiation therapy. A method of this embodiment is directed to specifically and preferentially radiosensitizing tumor cells rendering the tumor cells more susceptible to radiation therapy than non-tumor cells.
In yet another embodiment the present invention 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, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, spinal chord injury, immune senescence, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), acute pancreatitis, mucositis, hemoπhagic shock, splanchnic artery occlusion shock, multiple organ failure (such as involving any of the kidney, liver, renal, pulmonary, retinal, pancreatic and/or skeletal muscles systems), acute autoimmune thyroiditis, muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), local and/or remote endothelial cell dysfunction (such are recogmzed by endo-dependent relaxant responses and up-regulation of adhesion molecules), inflammation and skin aging.
The compounds of the present invention maybe administered, for example, parenterally, to a person diagnosed with acute retinal ischemia or acute vascular stroke, either by intermittent or continuous intravenous administration, by either a single dose or a series of divided doses. Compounds of the invention may be used in combination or sequentially. The compound of the invention can be administered by intermittent or continuous administration via implantation of a biocompatible, biodegradable polymeric matrix delivery system containing a compound of formula I, II, III, IV or V, or via a subdural pump inserted to administer the compound directly to the infarct area of the brain.
In a further embodiment, the present invention provides methods to extend the lifespan and proliferative capacity of cells, such as. for example, in using the compounds of the invention as general mediators in the generation of oxidants, proinflammatory mediators and/or cytokines, and/or general mediators of leukocyte infiltration, calcium ion overload, phospholipid peroxidation, impaired nitric oxide metabolism and/or reduced ATP production
For example, the compounds of the invention can treat 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 during cardiac aπest when 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 lifespan or proliferation of cells and thus to treat or prevent diseases associated therewith and induced or exacerbated by cellular senescence including skin aging, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative 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. These compounds can also be used to treat cancer and to radiosensitize hypoxic tumor cells to render the tumor cells more susceptible to radiation therapy and to prevent the tumor cells from recovering from potentially lethal damage of DNA after radiation therapy, 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, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging.
Preferably, the compounds of the invention 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 lifespan and proliferative capacity of cells; to alter gene expression of senescent cells; and to radiosensitize tumor cells.
Another especially prefeπed embodiment of the invention is a pharmaceutical composition which comprises (i) a therapeutically effective amount of the compound of formula I, II, III, IV or V; and (ii) a pharmaceutically acceptable carrier.
As used herein, "alkyl" means a branched or unbranched saturated hydrocarbon chain comprising 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-pentyl, n-hexyl, and the like, unless otherwise indicated. Optional substitutions of alkyl and ether chains include mercapto, carboxy, hydroxy, or phenyl, benzyl, or phenylethyl, which may themselves be substituted by hydroxy, halo, methoxy, d -Cβ alkyl, amine and carboxy.
"Alkenyl" means a branched or unbranched unsaturated hydrocarbon chain comprising a designated number of carbon atoms. For example, C2-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, butenyl. iso-butenyl, tert-butenyl, n-pentenyl, n-hexenyl, and the like, unless otherwise indicated.
"Alkoxy", means the group -OR wherein R is alkyl as herein defined. Preferably, R is a branched or unbranched saturated hydrocarbon chain containing 1 to 6 carbon atoms.
"Cyclo", used herein as a prefix, refers to a structure characterized by a closed ring.
"Halo" means at least one fluoro, chloro, bromo, or iodo moiety, unless otherwise indicated.
"Amino" compounds include amine (NH2) as well as substituted amino groups comprising alkyls of one through six carbons. "Ar", "aryl" or "heteroaryl" means a moiety which is substituted or unsubstituted, especially a cyclic or fused cyclic ring and includes a mono-, bi-, or tricyclic. carbo- or heterocyclic ring, such as a 5. 6, 7 or 8 membered ring, wherein the ring is either unsubstituted or substituted in, for example, one to five position(s) with halo, haloalkyl, hydroxyl, nitro, trifluoromethyl, Cx-Q straight or branched chain alkyl, C2-C6 straight or branched chain alkenyl, Cι-C6 alkoxy. -C(0)-0(Cι-C6 alkyl), carboxy, C2-C6 alkenyloxy, phenoxy, benzyloxy, amino, thiocarbonyl. ester, thioester, cyano, imino, alkylamino, aminoalkyl, sulfhydryl, thioalkyl, and sulfonyl; wherein the individual ring sizes are preferably 5-8 members; wherein the heterocyclic ring contains 1-4 heteroatom(s) selected from the group consisting of O, N, or S or their mixture; wherein aromatic or tertiary alkyl amines are optionally oxidized to a coπesponding N-oxide. Heteroaryls may be attached to other rings or substituted through the heteroatom and/or carbon atom of the ring. Particularly prefeπed aryl or heteroaryl moieties include but are not limited to phenyl, benzyl, naphthyl, piperidino, pyπolyl, pyπolidinyl, pyridinyl, pyrimidinyl, purinyl, quinolinyl, isoquinolinyl. fiiryl, thiophenyl, imidazolyl, oxazolyl. thiazolyl, pyrazolyl, and thienyl.
"Phpnvl " inHiiH- all
Figure imgf000039_0001
optionally substituted for a carbon atom of the ring. Cycloalkyls optionally containing at least one heteroatom, as described above, may be substituted by or fused to at least one 5 or 6 membered aryl or heteroaryl and/or substituted by at least one of amino, C1-C5 straight or branched chain alkyl, -Cβ alkanol, Cι-C6 straight or branched chain alkylamino, Cι-C6 alkoxy, or Q-C6 alkenyl, or benzyl, or phenyl or phenylethyl wherein the ring may be substituted as described above for substitutions of "Phenyl".
Prefeπed cycloalkyls containing at least one heteroatom include
Figure imgf000040_0001
, pyπolidinyl, indolyl, 2,3-dihydro-l H isoindolyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholino and thiomorpholino.
The compounds of the present invention possess one or more asymmetric center(s) and thus can be produced as mixtures (racemic and non-racemic) of stereoisomers, or as individual enantiomers or diastereomers. The individual stereoisomers may be obtained by using an optically active starting material, by resolving a racemic or non-racemic mixture of an intermediate at some appropriate stage of the synthesis, or by resolution of the compound of any of formulas I, II, III, IV and V. It is understood that the individual stereoisomers as well as mixtures (racemic and non-racemic) of stereoisomers are encompassed by the scope of the present invention.
The compounds of the invention are useful in a free base form, in the form of pharmaceutically acceptable salts, pharmaceutically acceptable hydrates, pharmaceutically 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 activity and which is neither biologically nor otherwise undesirable. Organic acids can be used to produce salts, hydrates, esters, or solvates such as acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, p-toluenesulfonate, bisulfate, sulfamate, sulfate, naphthylate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane-propionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate heptanoate, hexanoate, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, tosylate and undecanoate. Inorganic acids can be used to produce salts, hydrates, esters, or solvates such as hydrochloride, hydrobromide, hydroiodide, and thiocyanate. Examples of suitable base salts, hydrates, 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, hydrates, esters, or solvates may also be formed with organic bases. Organic bases suitable for the formation of pharmaceutically acceptable base addition salts, hydrates, 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 may include mono-, di-, and trialkylamines, such as methylamine, dimethylamine, triethylamine and dicyclohexylamine; mono-, di- or trihydroxyalkylamines, such as mono-, di-, and triethanolamine; amino acids, such as arginine and lysine; guanidine; N-methyl-glucosamine; N-methyl-glucamine; L-glutamine; N-methyl-piperazine; morpholine; ethylenediamine; N-benzyl-phenethylamine; (trihydroxy-methyl)aminoethane; and the like. See, for example, "Pharmaceutical Salts," J. Pharm. Sci., 66:1. 1-19 (1977). Accordingly, basic nitrogen-containing groups can be quatemized with agents including: lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl suifates such as dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, 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 invention in an aqueous or an aqueous alcohol solution or other suitable solvent containing the appropriate acid or base, and isolating 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 the PARP inhibitor having an acid group thereon with a base, such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained by concentrating the solution. "Pharmaceutically acceptable prodrug" refers to a derivative of the inventive compounds which undergoes biotransformation prior to exhibiting its pharmacological effect(s). The prodrug is formulated with the objective(s) of improved chemical stability, improved patient acceptance and compliance, improved bioavailability. prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity). The prodrug can be readily prepared from the inventive compounds using metiiods known in the art, such as those described by Burger's Medicinal Chemistry and Drug Chemistry, Fifth Ed., Vol. 1, pp. 172-178, 949-982 (1995). For example, the inventive compounds can be transformed into prodrugs by converting 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 body, most drugs are substrates for chemical reactions that may change their physical properties and biologic effects. These metabolic conversions, which usually affect the polarity of the compound, alter the way in which drugs are distributed in and excreted from the body. However, in some cases, metabolism of a drug is required for therapeutic effect. For example, anticancer drugs of the antimetabolite class must be converted to their active forms after they have been transported into a cancer cell. Since most drugs undergo metabolic transformation of some kind, the biochemical reactions that play a role in drug metabolism may be numerous and diverse. The main site of drug metabolism is the liver, although other tissues may also participate.
The term "neurodegenerative diseases" includes Alzheimer's disease, Parkinson's disease and Huntington's disease. The term "nervous insult" refers to any damage to nervous tissue and any disability or death resulting therefrom. The cause of nervous insult may be metabolic, toxic, neurotoxic, iafrogenic, thermal or chemical, and includes without limitation, ischemia, hypoxia, cerebrovascular accident, trauma, surgery, pressure, mass effect, hemmoπhage, radiation, vasospasm, neurodegenerative disease, infection, Parkinson's disease, amyotrophic lateral sclerosis (ALS), myelination/demyelination process, epilepsy, cognitive disorder, glutamate abnormality and secondary effects thereof.
The term "neuroprotective" refers to the effect of reducing, aπesting or ameliorating nervous insult, and protecting, resuscitating, or reviving nervous tissue that has suffered nervous insult.
The term "preventing neurodegeneration" includes the ability to prevent neurodegeneration in patients diagnosed as having a neurodegenerative disease or who are at risk of developing a neurodegenerative disease. The term also encompasses preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease.
The term "treating" refers to:
(i) preventing a disease, disorder or condition from occurring in an animal that may be predisposed to the disease, disorder and/or condition, but has not yet been diagnosed as having it; (ii) inhibiting the disease, disorder or condition, i.e., aπesting its development; and
(iii) relieving the disease, disorder or condition, i.e., causing regression of the disease, disorder and/or condition.
The term "neural tissue damage resulting from ischemia and reperfusion injury and neurodegenerative diseases" includes neurotoxicity, such as seen in vascular stroke and global and focal ischemia. A feature characteristic of many of these transformations is that the metabolic products are more polar than the parent drugs, although a polar drug does sometimes yield a less polar product. Substances with high lipid/water partition coefficients, which pass easily across membranes, also diffuse back readily from tubular urine 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 greatly reduced. Moreover, the specific secretory mechanisms for anions and cations in the proximal renal tubules and in the parenchymal liver cells operate upon highly polar substances.
As a specific example, phenacetin (acetophenetidin) and acetanilide are both mild analgesic and antipyretic agents, but are each transformed within the body to a more polar and more effective metabolite, p- hydroxyacetanilid (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. During the first hour, acetanilid is the principal plasma component. In the second hour, as the acetanilid level falls, the metabolite acetaminophen concentration reaches a peak. Finally, after a few hours, the principal plasma component is a further metabolite that is inert and can be excreted from the body. Thus, the plasma concentrations of one or more metabolites, as well as the drug itself, can be pharmacologically important.
The reactions involved in drug metabolism are often classified into two groups, as shown in the Table II. Phase I (or functionalization) reactions generally consist of (1) oxidative and reductive reactions that alter and create new functional groups and (2) hydrolytic reactions that cleave esters and amides to release masked functional groups. These changes are usually in the direction of increased polarity. Phase II reactions are conjugation reactions in which the drug, or often a metabolite of the drug, is coupled to an endogenous substrate, such as glucuronic acid, acetic acid, or sulfuric acid.
TABLE II Phase I Reactions (functionalization reactions):
(1) Oxidation via the hepatic microsomal P450 system: Aliphatic oxidation
Aromatic hydroxylation
N-Dealkylation
O-Dealkylation
S-Dealkylation Epoxidation
Oxidative deamination
Sulfoxide formation
Desulfuration
N-Oxidation and N-hydroxylation Dehalogenation
(2) Oxidation via nonmicrosomal mechanisms:
Alcohol and aldehyde oxidation Purine oxidation
Oxidative deamination (monoamine oxidase and diamine oxidase)
(3) Reduction:
Azo and nitro reduction
(4) Hydrolysis:
Ester and amide hydrolysis Peptide bond hydrolysis
Epoxide hydration
Phase II Reactions (conjugation reactions): (1) Glucuronidation (2) Acetylation
(3) Mercapturic acid formation
(4) Sulfate conjugation
(5) N-, 0-, and S-methylation
(6) Trans-sulfuration The compounds of the present invention exhibit pharmacological activity and are, therefore, useful as pharmaceuticals. In particular, the compounds exhibit central nervous and cardiac vesicular system activity. It is understood that tautomeric forms, when possible, are included in the invention.
Many of the PARP inhibitors are known and, thus, can be synthesized by known methods from starting materials that are known, may be available commercially, or may be prepared by methods used to prepare coπesponding compounds in the literature. See, for example, Suto et al., "Dihydroiso-quinolinones: The Design and Synthesis of a New Series of Potent Inhibitors of Poly( ADP-ribose) Polymerase", Anticancer Drug Des., 6:107-17 (1991), which discloses processes for synthesizing a number of different PARP inhibitors.
Typically, the PARP inhibitors used in the composition of the invention will have an ICS. for inhibiting poly( ADP-ribose) polymerase in vitro of about 20 μM or less, preferably less than about lOμM, more preferably less than about 1 μM, or preferably less than about 0.1 μM, most preferably less than about 0.01 μM.
The PARP inhibitor 3,4-dihydro-5-[4-(l-piperidinyl)butoxy]-l(2^T)-isoquinolinone, for example, has been reported to inhibit PARP with an ICS0 of 40 nM by Suto et al., cited above.
The 6(5_Y)-phenanthridinone sulfonamides and carbamides, such as Example compounds 1-14 below, of this invention are represented by previously defined formula I-V. As an example, these 6(5 )- phenanthridinone derivatives can be prepared in a conventional manner as illustrated below by Schemes 1 - 3. The process sequence set forth herein does not present an exact sequence of reactions by which the compound must be made; that is, the sequence of reactions can be reaπanged in several ways, by means known to those of ordinary skill in the art, to reach the target molecule. Scheme 1 below illustrates schematically the preparation of compounds Example 1 through Example 4.
Compound 3 (of this scheme), 2-amino-6(5_7)-phenanthridinone. is used as a precursor to prepare the desired N-substituted 6(5Jϊ)-phenan_hridinone sulfonamides and carbamides. Preparation of this amino derivative 3 is known in the chemistry literature and accessible by processes known to one of ordinary skill in the art. For example, nitration of commercially available compound of 6(5 )-phenanthridinone 1 provides compound 2- nitro-6(5_7)-phenanthridinone 2 which can be reduced by hydrogenation using palladium as catalyst or by hydrazine to afford 2-amino-6(5_7)-phenantl_ridinone 3. Target compounds-sulfonamides and carbamides can be formed by reaction the amino group of compound 3 with either sulfonyl chloride or carboxylic acid halide derivatives as described in General procedure A. Formation of sulfonamides and carbamides with amino derivatives can be carried out by a variety of conditions known to those of ordinary skill in the art, including reaction with first or secondary amines using pyridine or triethyl amine as base. Typical solvents include chlorinated solvents, various ethers, and dipolar aprotic solvents like DMF.
Scheme 1
Figure imgf000045_0001
Figure imgf000045_0002
Scheme 2 below illustrates schematically the preparation of compounds Example 10 through Example 12. Monosulfonation of commercially available 6(5iϊ)-phenanthridinone 1 can be achieved using chorosulfonic acid neat to give compound 4 in high yield. For example, 6(5ϊT)-phenanthridinone 1 was placed in neat chorosulfonic acid under nitrogen at 0 °C. The resulting mixture is allowed to warm to room temperature, continuously stirred for about 2 days and poured onto 100 g of ice. The residue is collected by filtration and washed with water, ethanol to afford the product 4 as solid without further purification, mp 317-320, H- MR (400 MHz, DMSO-ύk), 11.78 (br, IH), 8.50 (s, IH), 8.41 (d, J= 8.0 Hz, IH), 8.33 (d, J = 8.0 Hz, IH), 7.88 (t, J = 8.0 Hz, IH), 7.72 (d, J = 8.0 Hz, IH), 7.66 (t, J = 8.0 Hz, IH), 7.33 (d, J = 8.0 Hz, IH). Amindation of compoimd 4 using primary amines can be carried out by a variety of conditions similar to the process described above from compound 3 to Examples 1-9. Thus final products of compounds Examples 10-12 can be prepared from 6(5/ )-phenanthridinone 1 using General procedure A bellow.
Scheme 2
Figure imgf000046_0001
Examples 10-12
Scheme 3 below illustrates schematically the preparation of compounds Example 13 through Example
19. The 3-substituted (5_7)phenanthridin-6-one skeleton of this invention can be constructed in a conventional manner using Schmidt method with fluoren-9-one 5 as starting material and sodium azide as an insertion agent in acidic condition (Chida, N.: Ohtsuka. M.: Ogawa, S. Tetrahedron Lett. 1991, 32, 4525). The fluoren-9-one ring may be generically substituted as set forth in the drawing. Such fluoren-9-one starting derivatives are known in the chemistry literature and accessible by processes known to one skilled in the art. This Schmidt method normally generates a pair of regioisomer mixtures 6 and 7, which may be separated and purified by chromatography or crystallization. For example, a mixture of 2(fluoren-9-one)carboxylic acid (0.25g) and sodium azide (0.25g) in c-sulfuric acid (lOmL) was stiπed at 0 °C for 30 minutes at 25 °C for 10 hours, and then poured into ice-cold water (lOOmL). A yellow precipitation appeared upon adjusting the solution to pH 7 with sodium hydroxide. The precipitation, a regioisomer mixture of 3-[6(Jiϊ)phenanthridinone] carboxylic acid 6 and of 8-[6(5_ )phenanthridinone] carboxylic acid 7, was collected by filtration and purified on silica gel column to give a pure isomer of 3-[6(5_T)phenanthridinone] carboxylic acid 6 (0. lg), mp 300 °C (dec). H- NMR (400 MHZ, DMSO--/*), 13.19 (bs, IH), 11.88 (s, IH), 8.58 (d, IH, J = 8Hz), 8.52 (d, IH, J = 8Hz), 8.35 (d, IH, J = 8Hz), 8.01 (s, IH), 7.89 (t, IH, J= 8Hz), 7.76 (t, IH, J= 8Hz), 7.72 (d, IH, J = 8Hz). There are several ways, such as Ullman reactions (Synthesis, 1997, p.1273-1275 "Synthesis and Characterization of Some Nitrobenzanthrones: Suspected New Mutagens in Atmospheric Environment",
Hitomi Suzuki, Takeji Enya, Yoshiharu Hizamatsu) and Suzuki couplings (J. Organic. Chememistry., 1992, 57, p.379-381. "A simple asymetric sythesis of 4-arylphenylalanines vis palladium-catalyzed cross coupling reaction of arylboric acids with tyrosine triflate " W.-C. Shieh; J. A. Carlson), to construct substituted 6(57?)Phenanthr_dinone carboxylic acid and
Figure imgf000046_0002
sulfonyl chloride. Formation of substituted 6(5iϊ)Phenanthridinone using biphenyl compounds can be carried out by a variety of conditions known to those skilled in the art. 3-[6(5 f)Phenant__ridinone]carboxylic acid 6 can be routinely converted to 3- [6(J_T)Phenanthridinone]carbonyl chloride 7 using thionyl chloride neat at 60 °C. Amidation of desired regioisomer of 3-[6(5_Y)phenanthridinone]sulfonyl chloride 6 or 3-[6(5iY)Phenanthridinone]carbonyl chloride 7 with R' substituted primary amine using triethyl amine (TEA) as base provides (5 _)phenanthridin-6-one sulfonamide and carbamide of Example 13-19.
Scheme 3
Figure imgf000047_0001
General procedure A:
To a suspension of the sulfonyl chloride or compound 6 (1.1 mmol) in methylene chloride (100 mL) or p-dioxane (100 mL) is added triethyl amine (1.5 mmol) and amine compound or compound 3 (1.0 mmol) under nitrogen at 0 °C. The reaction mixture is allowed to warm to room temperature, stfrred continuously for 3 hour or until TLC showing no starting material left and poured into 100 mL of water. The organic layer is collected, washed with water, brine and concentrated in vacuo. The product is purified via crystallization or silica column chromatography using methanol/methylene chloride as eluent to afford solid product in 60 - 90% of yield. Example 1
Figure imgf000048_0001
N-(5,6-Dihycho-6-oxo-2-phenantlιricUnyl)-4-metlιyl beι_zenesulfonamide
Prepared from 2-amino-6(5i7)-phenantl_ridinone 3 and 4-methyl benzenesulfonamide according to
General Procedure A. P-dioxane was used as solvent and the reaction was run at 40 °C for 30 hours.
Purification of compound by crystallization in dioxane gave a brown solid ( 93 % yield), mp 199-203 °C. ^H- NMR (400 MHz, DMSO-_t5), 10.20 (s, IH), 8.30 (d, J = 8.0 Hz, IH), 8.15 (d, J = 8.1 Hz, IH), 7.95 (s, IH), 7.88 (t, J = 7.6, IH), 7.65 (m, 3H), 7.33 (d, J = 8.2 Hz, 2H), 7.19 (m, 2H). Anal. (C20 H16 N2 03 S), C H N.
Example 2
Figure imgf000048_0002
2-Chloro-N-(5,6-dihydro-6-oxo-2-phenanthridinyl)acetamide
Prepared from 2-amino-6(5_ )-phenanthridinone 3 and chloroacetyl chloride according to General Procedure A. P-dioxane was used as solvent and 1.1 equivalent of sodium hydroxide as base. A white precipitation was formed after stirring the mixture for 2 ours. Purification of the precipitation by crystallization in dioxane gave a white solid (53 % yield), mp 300-303 °C. iH-NMR (400 MHz, DMSO-afe), 11.71 (s, IH), 10.45 (s, IH), 8.63 (s, IH), 8.33 (d, J = 7.9 Hz, IH), 8.27 (d, J = 8.2 Hz, IH), 7.89 (t, J = 7.6 Hz, IH), 7.64 (m, 2H), 7.34 (d, J = 8.8 Hz, IH), 4.30 (s, IH). Anal. (C,_ H, , CI N2 02), C H N.
Example 3
Figure imgf000049_0001
4-[(5,6-Dihydro-6-oxo-2-phenanthridinyl)amino]-4-oxo-butanoic acid
Prepared from 2-amino-6(5_7)-phenanthridinone 3 and succinic anhydride. To a solution of 2-amino-
Figure imgf000049_0002
(150 mg, 0.71 mmol) in dioxane (30mL) was added a solution of succinic anhydride (87mg, 1.1 mmol) in dioxane (5mL) over 5 minutes. The mixture was stfrred for 16 hours and a yellowish precipitation was formed. Purification of the precipitation by crystallization in dioxane gave a light white solid (92 % yield), mp 224-228 °C. LH-NMR (400 MHz, DMSO-ek), 11.7 (s, IH), 10.1 (s, IH), 8.67 (s, IH), 8.33 (d, J = 7.8 Hz, IH), 8.24 (d, J = 8.2 Hz, IH), 7.87 (t, J = 7.0 Hz, IH), 7.66 (t, J = 7.44 Hz, IH), 7.60 (d, J = 8.7 Hz, IH), 7.30 (d, J = 8.7 Hz, IH), 2.60 ((d, J = 5.4 Hz, 2H), 2.56 (d, J = 5.4 Hz, 2H). Anal. (C17 H24 N204 0.1 H20), C H N.
Example 4
Figure imgf000050_0001
N-(5,6-Dihydro-6-oxo-2-phenanthridinyl)-2-(trimethyl-azanyl)-acetamide chloride
Prepared from the compound of Example 2 and dimethyl amine. To a water solution of trimethyl amine (70mg) was added 2-chloro-N-(5,6-dihydro-6-oxo-2-phenanthridinyl)acetamide (286 mg) in dimethylsulfoxide (0.2 mL) at room temperature over 0.5 hours. Acetone (lOmL) was added to the mixture which was continuously stiπed for 18 hours. A white precipitation was collected by filtration. Purification of the crude precipitation by crystallization in ethanol gave a white solid as a hydrogen chloride salt (lOOmg), mp
269-272 °C (dec). iH-NMR (400 MHz, DMSO-cfe), 11.76 (s, IH), 11.03 (s, IH), 8.68 (s, IH), 8.34 (d, J = 8 Hz, IH), 8.27 (d, J = 8 Hz, IH), 7.91 (t, J = 8 Hz, IH), 7.65 (m, 2H), 7.37 (d, J = 8 Hz, IH), 4.41 (s, 2H). Anal. (C18 H20 CI N3 02 0.7 H20), C H N. Example 5
Figure imgf000050_0002
N-(5,6-Dihydro-6-oxo-2-phenanthridinyl)-2-(dimethylamino)-acetamide
Prepared from the compound of Example 2 and dimethylamine. To a solution of 2-chloro-N-(5,6- cUhydro-6-oxo-2-phenant__ridinyl)-acetamide (635 mg) in N, N-dimethylformamide (100 mL) was added potassium phosphate (918 mg) and dimethylamine (1.3 ml, 2 molar soln.). The solution stiπed at room temperature for 5 hours followed by evaporation of the solvent. The light brown product was washed with water and filtered. Concentrated HC1 (0.3 mL) was added to a solution of the product in dioxane (150 mL) to precipitate the hydrogen chloride salt (400 mg), mp >300°C. Η-NMR (300 MHz, DMSO-d6), 11.63 (br s, IH), 10.92 (br s, IH), 8.63 (s, IH), 8.35 (d, J = 9.0 Hz, IH), 8.26 (d, J = 9.0 Hz, IH), 7.91 (t, J = 9.0 Hz, IH), 7.69 (m, 2H), 7.38 (d, J = 9.0 Hz, IH), 4.19 (br s, 2H), 2.91 (br s, 6H). Anal. (CI7HπN302 HC1 0.9H2O), Calc for: C, 58.67; H, 5.73; N, 12.07; CI, 10.19. Found: C. 58.91; H, 5.59; N, 11.80; CI, 9.95.
Example 6
Figure imgf000051_0001
N-(5,6-Dihydro-6-oxo-2-phenanthridinyl)-2-(dipropylamino)-acetamide
Prepared from the compound of Example 2 and dipropylamine. To a solution of 2-chloro-N-(5,6- dihydro-6-oxo-2-phenanthridinyl)-acetamide (45 mg) in N, N-dimethylformamide (10 mL) was added potassium phosphate (68 mg) and dipropylamine (0.027 mL). The solution stiπed at room temperature for 5 hours followed by evaporation of the solvent. The light brown product was washed with water and filtered (25 mg). Η-NMR (300 MHz, DMSO-d6), 11.70 (br s, IH), 9.72 (br s, Hi), 8.33 (d, J = 8.0 Hz, 2H), 7.89 (m, 1), 7.66 (m, 2H), 7.33 (m, IH), 3.21 (s, 2H), 1.50 (m, 6H), 0.89 (m, 8H).
Example 7
Figure imgf000051_0002
N-(5,6-Dihydro-6-oxo-2-phenanthridinyl)-l,3-dihydro-2H-isoindole-2-acetamide Prepared from the compound of Example 2 and isoindoline. To a solution of 2-chloro-N-(5,6-dihydro- 6-oxo-2-phenanthridinyl)-acetamide (45 mg) in N, N-dimethylformamide (10 mL) was added potassium phosphate (68 mg) and isoindoline (0.025 mL). The solution stiπed at room temperature for 5 hours followed by evaporation of the solvent. The light brown product was washed with water and filtered (35 mg). Η-NMR (300 MHz, DMSO-ds), 11.67 (br s, IH), 9.97 (br s, IH), 8.69 (s, IH), 8.31 (d, J = 9.0 Hz, 2H), 7.84 (m, 2H), 7.65 (t, J = 9.0 Hz, IH), 7.27 (m, 5H), 4.11 (s, 4H), 3.60 (s, 2H).
Example 8
Figure imgf000052_0001
2-(cyclohexylmethyl_u no)-N-(5,6-dihydro-6-oxo-2-phenanthridinyl)-acetamide
Prepared from the compound of Example 2 and N-methylcyclohexylamine. To a solution of 2-chloro- N-(5,6-dihy(fro-6-oxo-2-phenanthridinyl)-acetamide (45 mg) in N, N-dimethylformamide (10 mL) was added potassium phosphate (68 mg) and N-methylcyclohexylamine (0.025 mL). The solution stiπed at room temperature for 5 hours followed by evaporation of the solvent. The light brown product was washed with water and filtered (30 mg). Η-NMR (300 MHz, DMSO-d6), 11.69 (br s, IH), 9.76 (br s, IH), 8.65 (s, IH), 8.34 (t, 2H), 7.87 (m, 2H), 7.66 (t, IH), 7.32 (t, IH), 3.18 (s, 2H), 2.34 (s, 3H), 1.80 (m, 4H), 1.60 (m, 2H), 1.23 (m, 5H).
Example 9
Figure imgf000052_0002
N-(5,6-dihydro-6-oxo-2-phenanthridinyl)-4-mo holineacetamide Prepared from the compound of Example 2 and morpholine. To a solution of 2-chloro-N-(5,6-dihydro- 6-oxo-2-phenanthridinyl)-acetamide (45 mg) in N, N-dimethylformamide (10 mL) was added potassium phosphate (68 mg) and morpholine (0.017 mL). The solution stiπed at room temperature for 5 hours followed by evaporation of the solvent. The light brown product was washed with water and filtered (30 mg). :H-NMR (300 MHz, DMSO-d6), 11.68 (br s, IH), 9.85 (br s, IH), 8.64 (s, IH), 8.33 (d, J = 9.0 Hz, 2H), 7.89 (t, IH), 7.77 (d, J = 9.0 Hz, IH), 7.66 (t, IH), 7.32 (d, J = 9.0 Hz, IH), 3.68 (d, 4H), 3.17 (s, 2H), 2.55 (m, 4H).
Example 10
Figure imgf000053_0001
5,6-Dihydro-N-[2-(l-piperidinyl)ethyl]-6-oxo-2-phenant__ridinesulfonamide hydrogen chloride
Prepared from 2-(6(5iϊ)-phenanthridinone)sulfonyl chloride 4 and l-(2-aminoethyl)piperidine according to General Procedure A. A hydrogen chloride salt was formed upon adding IN HC1 to a solution of
5,6-ώhy(fro-N-[2-(l-piperiώnyl)ethyl]-6-oxo-2-phen_mthridinesuifonamide in dioxane at 70 °C. The mixture was cooled down to 0 °C produced a salt precipitation which was collected by filtration to give a white solid (53 % yield from 4), mp 262-267 °C. ^-NM (400 MHz, CDC13), 8.79 (s, IH), 8.56 (d, J = 8.1 Hz, IH), 8.36 (d, J = 7.2 Hz, IH), 7.93 (m, 2H), 7.75 (t, J = 7.5 Hz, IH), 7.54 (d, J = 8.7 Hz, IH), 3.41 (d, J = 12.2 Hz, 2H), 3.14 (m, 4H), 2.89 (t, 2H). 1.71 (m, 6H). Anal. (C20 H24 N3 03 S HC1 0.4 H20), C H N.
Example 11
Figure imgf000054_0001
5,6-Dihydro-N-[2-(4-mo holinyl)ethyl]-6-oxo-2-phen_mtlιridinesulfonamide
Prepared from the compound 2-(6(5i?)-phenanthridinone)sulfonyl chloride 4 and l-(2- aminoethyl)morpholine according to General Procedure A. Methylene chloride was used as solvent. Purification of the precipitation by crystallization in dioxane gave a white solid (95 % yield), mp 246-247 °C. H-NMR (400 MHz, CDCI3), 12.04 (s, 2H), 8.75 (s, IH), 8.52 (d, J = 8.2 Hz, IH), 8.35 (d, J = 8.0 Hz, IH), 7.90 (m, 2H), 7.73 (t, J = 7.5 Hz, IH), 7.50 (d, J = 8.6 Hz, IH), 3.44 (t, J = 4.5 Hz, 4H), 2.90 (t, J = 6.1 Hz, 2H), 2.30 (t, J = 6.8 Hz, IH), 2.23 (t, J = 4.3 Hz, 4H). Anal. (C19 H21 N3 O4 S 0.3 H20), C H N.
Example 12
Figure imgf000054_0002
N-[2-| is(2-Hydroxyethyl)anήno]ethyl]-5,6-dihycto^ Prepared from the compound 2-(6(5i )-phenanthridinone)sulfonyl chloride 4 and N,N-bis(2- hydroxyethyl)ethylenediamine according to General Procedure A. Methylene chloride was used as solvent. A precipitation was formed after stirring the reactant for 20 hours. Purification of the precipitation by crystallization in p-dioxane gave a yellowish solid (40 % yield), mp 210 - 214 °C (dec). ^H-NMR (400 MHz, DMSO-*), 8.72 (s, IH), 8.51 (d, J = 8.0Hz, IH), 8.35 (d, J = 8.0Hz, IH), 7.92 (t, J = 8.0 Hz, IH), 7.89 (d, J = 4.0Hz, IH), 7.73 (t, J = 8.0Hz, 1H0, 7.51 (d, J = 8.0Hz, IH), 4.34 (m, 2H), 2.85 (m, 2H), 2.43 (t, 4H). Anal. (C19 H23 N3 05 S 0.8 H20), C H N (: calcd 54.35 found 53.67).
Example 13
Figure imgf000055_0001
3-(4-Moφholinylcarbonyl)-6(5_7)-phenanthridinone
Prepared from to 3-[6(5iϊ)Phenanthridinone]carbonyl chloride 7 and morpholine according to General Procedure A. to 3-[6(5_Y)Phenanthridinone]carbonyl chloride 7 can be obtained by Schmidt insertion of commercially available 2(fluoren-9-one)carboxylic acid as described in the Scheme 3. Purification of compound by crystallization in water-acetone gave a white solid (50 % yield), mp 182-184 °C. * H-NMR (400 MHz, DMSO-*), 10.72(s, IH); 8.56(d, IH, 8.2Hz); 8.52(d, IH. 8.1Hz); 8.34(dd, IH, 1.0Hz, 7.9Hz); 7.90(t, IH, 7.8Hz); 7.69(t, IH, 7.6Hz); 7.47(d, IH, 7.4Hz); 7.33(t, IH, 7.7Hz); 3.84~3.42(m, 8H). Anal. (C18 H,6 N2 03), C H N.
Example 14
Figure imgf000055_0002
N-[2-(4-Morpholinyl)ethyl] 5,6-dihydro-6-oxo-3-phenanthridine carboxamide Prepared from to 3-[6(5_ )Phenanthridinone]carbonyl chloride 7 and l-(2-aminoethyl)morpholine according to General Procedure A. DMF was used as solvent. Purification of compound by crystallization in acetic acid gave a solid (60 % yield), mp 285 -292 °C. -NMR (400 MHz, DMSO-*), 11.82 (s, IH), 8.58 (d, IH, J=8Hz), 8.54 (t, IH, J=8Hz), 8.34 (d, IH, J=8Hz), 7.89 (t, IH, J=8Hz), 7.83 (s, IH), 7.70 (m, 2H), 3.58 (m, 5H), 3.40 (m, 2H), 3.31 (m, 5H). Anal. (C20 H21 N3 03 1C1 H), C H N.
Example 15
Figure imgf000056_0001
l-[(5,6-Dihydro-6-oxo-3-phenanthridinyl)carbonyl]-proline
Prepared from to 3-[6(5_Y)Phenanthridinone]-carbonyl chloride 7 and (S)-methylproIine in methylene chloride according to General Procedure A. The resulting l-[(5,6-dihydro-6-oxo-3-phenanthridinyl)carbonyl]- proline methyl ester was hydrolyzed with IN sodium hydroxide in dioxane. After neutralizing the mixture with
HC1, the desired compound was collected as a white solid precipitate (60 % yield), mp 250 - 253 °C. ^H-NMR (400 MHz, DMSO-*), 12.60 (bs, IH), 11.79 (s, IH), 8.55 9d, IH, J=8Hz), 8.47 (d, IH, J=8Hz), 8.34 (d, IH, J=8Hz), 7.90 (t, IH, J=8Hz), 7.70 (t, IH, J=8Hz), 7.54 (s, IH), 7.40 (d, IH, J=8Hz), 4.44 (m, IH), 3.56 (m, 2H), 2.37 (m,lH), 1.90 (m, 3H) Anal. (C,9 H16 N2 02), C H N.
Example 16
Figure imgf000057_0001
8-Fluoro-N-[2-(4-moφholinyl)ethyl] 5,6-dihydro-6-oxo-3-phenanthridinecarboxamide
Prepared from S-lS-fluoro-β^-YJphenanthridinonelcarbonyl chloride and l-(2-aminoethyl)moφholine in dioxane according to General Procedure A. 3-[8-fluoro-6(5_T)phenantlιridinone]carboxylic acid 6 was prepared from Schimdt nitrogen insertion of 2(7-fluorofluoren-9-one) carboxylic acid as described in Scheme 3. Purification of the regioisomer mixture on silica gel column gave'a desired isomer acid 6 as a solid. 3-[8-FIuoro- 6(5 _)phenanthridinone]carboxylic acid sodium salt has -NMR (400 MHz, D20) 7.63 (m, IH), 7.47 (d, IH,
J=8Hz), 7.34 (m, IH), 7.17 (m, 2H), mp 300 °C (dec). The acid 6 was further converted to 3-[8-fluoro- 6(J_ )phenantlιridinone]carbonyl chloride using thionyl chloride. After removal of the thionyl chloride, the residue was used for next step without purification.
Coupling reaction of 3-[8-fluoro-6(5_T)phenanthridinone]carbonyl chloride and l-(2- aminoethyl)moφholine afforded a brown solid, which was purified by crystallization in acetic acid, mp 285 -
290 °C. -NMR (400 MHz, DMSO-*), 11.98 (s, IH), 8.67 (m, IH), 8.55 (t, IH, J=8Hz), 8.47 (d, IH, J=12Hz), 7.99 (d, IH, J=8Hz), 7.84 (s, IH), 7.79 (t, IH, J=8Hz), 7.70 (d, IH, J=8Hz), 3.59 (m, 5H), 3.41(m, 2H), 2.45 (m, 5H). Anal. (C20 H20 F N3 03 ), C H N (calcd 65.03 found 64.55).
Example 17
Figure imgf000058_0001
N-[(5,6-Dihydro-6-oxo-3-phenanthridinyl)carbonyl]-bis(benzyl)aspartic acid ester
Prepared from to 3-[6(5_ϊ)Phenanthridinone]carbonyl chloride 7 and (S)-bisbezylaspartic acid ester in DMF according to General Procedure A. The compound was purified by silica gel column cliromatography to give a white solid (70 % yield), mp 162-164 °C. XH-NMR (400 MHz, DMSO-*), 9.40 (dd, IH, J=7.22 Hz), 8.96 (S, IH), 8.66 (dd, IH, J=8.08), 8.48 (dd. IH, J=8.08), 8.28 (dd, IH, J=10.3), 7.58 (t, IH, J=7.22), 7.40 (m. IH). 7.22 (m. lOH), 5.00 (m, 4H), 4.85 (m, IH), 3.15 (m, 2H). Anal. (CS H52 N42), C H N.
Example 18
Figure imgf000058_0002
8-Fluoro-3 -[(4-methyl- 1 -piperazinyl)sulfonyl] - 6(5_Y)-phenanthridinone
Prepared from 3-[8-fluoro-6(J_7)phenanthridinone]sulfonyl chloride 6 and N-methylpiperazine in dioxane according to General Procedure A. 3-[8-FIuoro-6(J_?)phenanthridinone]sulfonyl chloride 6 was prepared in two steps. Commercially available 2-fuloro-fluoren-9-one was placed in neat chorosulfonic acid under nitrogen at 0 °C. The resulting mixture is allowed to warm to room temperature, continuously stiπed for about 18 hours and poured onto 100 g of ice. The residue is collected by filtration and washed with water, ethanol to afford the product of 7-(2-fuloro-fluoren-9-one)sulfonyl chloride as solid. The solid was directly used for the next step without further purification.
A mixture of 7-(2-fuloro-fluoren-9-one)sulfonyl chloride (0.25g) and sodium azide (0.25g) in c-sulfuric acid (lOmL) was stiπed at 0 °C for 30 minutes and at 25 °C for 10 hours, and then poured into ice-cold water (lOOmL). A precipitation appeared upon adjusting the solution to pH 8 with sodium hydroxide. The precipitation, a regioisomer mixture of 3-[8-fluoro-6(5_:_)phenanthridinone]sulfonyl chloride and of 8-[3-fluoro- 6(5_¥)phenanthridinone]sulfonyl chloride, was collected by filtration. The filtrate was allowed to stand at O °C for 24 hours, a pure isomer of 3-[8-fluoro-6(5β)phenanthridinone]sulfonyl chloride was appeared as precipitate of a white crystal, -NMR (400 MHz, DMSO-*), 400 MHz, 11.87(s, IH); 8.59(dd, IH, J = 8.96Hz, 5.04Hz); 8.33(d, IH, J = 8.4Hz); 7.97(dd, IH, J = 9.68Hz, 2.96Hz); 7.75(t, IH, J = 8.6Hz); 7.68(s, IH); 7.47(d, IH, J = 7.76). Coupling of 3-[8-fluoro-6(5-^phenanthridinone]sulfonyl chloride and N-methylpiperazine in dioxane according to General Procedure A provided the final sulfonamide as a white solid, mp 260 -261 °C (dec). - NMR (400 MHz, DMSO-*), 400 MHz 12.05(s, IH); 8.72(dd, IH, J = 8Hz, 4Hz); 8.65(d, IH, J = 8Hz); 8.02(dd, IH, J = 8Hz, 4Hz); 7.83(td, IH, J= 8Hz, 4Hz); 7.78(s, IH); 7.54(d, IH, J = 8Hz); 2.96(s, 4H); 2.37(s, 4H); 2.13(s, 3H). Anal. (C18 H,s F N3 03 S 0.3 H20), C H N.
Example 19
Figure imgf000059_0001
8-Fluoro-N-[2-(4-moφholinyl)ethyl] 5,6-dihy<fro-6-oxo-3-phenanthridinesulfonainide Prepared from 3-[8-fluoro-6(J_ )phenanthridinone]sulfonyl chloride 6 (see Example 13) and l-(2- aminoethyl)moφholine in dioxane according to General Procedure A. Purification of the crude by recrystalization in acetic acid gave a white solid, the mp 240 - 241 °C. -NMR (400 MHz, DMSO-*), 12.08(s, IH); 8.70(dd, IH, J = 8Hz, 4Hz); 8.61(d, IH J = 8Hz); 8.01(dd, IH, J = 8Hz, 4Hz); 7.83- 7.73(m, 3H); 7.64(dd, IH, J = 8Hz, 4Hz); 3.47(t, 4H, J = 4Hz); 2.92(t, 2H, J = 8Hz); 2.3 l(t, 2H, J = 8Hz); 2.26(t, 4H, J = 4Hz). Anal. (C19 H20 F N3 O4S H2O), C H N.
Other manners, variations or sequences of preparing the compounds of the present invention will be readily apparent to those of ordinary skill in the art. The compounds of the present invention may be useful in the free base form, in the form of base salts where possible, and in the form of addition salts, as well as in the free acid form. All these forms are within the scope of this invention. In practice, use of the salt form amounts to use of the base form. Pharmaceutically acceptable salts within the scope of this invention are those derived from mineral acids such as hydrochloric acid and sulfuric acid; and organic acids such as ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, and the like, giving the hydrochloride, sulfonate, ethanesulfonate, benzenesuLfonate, p-toluenesulfonate, and the like respectively, or those derived from bases such as suitable organic and inorganic bases. Examples of pharmaceutically acceptable base addition salts with compounds of the present invention include organic 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 puφose of illustration, such organic bases may include mono-, di-, and trialkylamines, such as methylamine, diethylamine and triethylamine; mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine; amino acids such as arginine, and lysine; guanidine; N-methylglucosamine; N-methylgiucamine; L-glutamine; N-methylpiperazine; moφholine; ethylenedianane; N-benzylphenethylamine; tris(hydroxymethyl)antinoethane; and the like.
The acid addition salts of the basic compounds may he prepared by dissolving the free base of the compounds of the present invention in aqueous or aqueous alcohol solution or other suitable solvents containing the appropriate acid or base and isolating the salt by evaporating the solution, or by reacting the free base of a compound of the present invention with an acid as well as reacting a compound of the present invention having an acid group thereon with a base such that the reactions are in an organic solvent, in which case the salt separates directly or can be obtained by concentration of the solution. The compounds of this invention contain one or more asymmetric 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 invention exhibit pharmacological activity and are, therefore, useful as pharmaceuticals. In particular the compounds exhibit central nervous and cardiac vesicular system activity. Other variations and modifications of this invention using the synthetic pathways described above will be obvious to those of ordinary skill in the art. Methods of Using the Compounds of the Invention
The compounds of the present invention can treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis; can ameliorate neural or cardiovascular tissue damage, including that following focal ischemia, myocardial infarction, and reperfusion injury; can treat various diseases and conditions caused or exacerbated by PARP activity; can extend or increase the lifespan or proliferative capacity of cells; can alter the gene expression of senescent cells; and can radiosensitize cells. Generally, inhibition of PA P activity spares the cells from energy loss, preventing, in the case of neural cells, iπeversible depolarization of the neurons, and thus, provides neuroprotection. While not being bound to any one particular theory, 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 invention further relates to a method of administering a therapeutically effective amount of the above-identified compounds in an amount sufficient to inhibit PARP activity, to treat or prevent tissue damage resulting from cell damage or death due to necrosis or apoptosis, to effect a neuronal activity not mediated by NMDA toxicity, to effect a neuronal activity mediated by NMDA toxicity, to treat neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age-related muscular degeneration, ADDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, inflammation, gout, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging; to extend the lifespan and proliferative capacity of cells; to alter gene expression of senescent cells; or to radiosensitize hypoxic tumor cells. The present invention also relates to treating diseases and conditions in an animal which comprises administering to said animal a therapeutically effective amount of the above-identified compounds.
In particular, the present invention relates to a method of treating, preventing or inhibiting a neurological disorder in an animal, which comprises administering to said animal a therapeutically effective amount of the above-identified compounds. In a particularly prefeπed embodiment, the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration. Another prefeπed embodiment is when the reperfusion injury is a vascular stroke. Yet another prefeπed embodiment is when the peripheral neuropathy is caused by Guillain-Barre syndrome. Still another prefeπed embodiment is when the demyelinating disease and neurological disorder relates to neurodegeneration. Another prefeπed embodiment is when the reperfusion injury is a vascular stroke. Still another prefeπed embodiment is when the demyelinating disease is multiple sclerosis. Another prefeπed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis.
Yet another prefeπed embodiment is a method of treating, preventing or inhibiting a cardiovascular disease in an animal, such as angina pectoris, myocardial infarction, cardiovascular ischemia, and cardiovascular tissue damage related to PARP activation, by administering to said animal an effective amount of the compounds of the present invention.
The present invention also contemplates the use of compound I, II, III, IV or V for inhibiting PARP activity, for treating, preventing or inhibiting tissue damage resulting from cell damage or death due to necrosis or apoptosis, for treating, preventing or inhibiting a neurological disorder in an animal.
In a particularly prefeπed embodiment, the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration. Another prefeπed embodiment is when the reperfusion injury is a vascular stroke. Yet another prefeπed embodiment is when the peripheral neuropathy is caused by Guillain-Baπe syndrome. Still another prefeπed embodiment is when the demyelinating disease is multiple sclerosis. Another prefeπed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis. The present invention also contemplates the use of compound I, II, III, IV or V in the preparation of a medicament for the treatment of any of the diseases and disorders in an animal described herein. In a particular embodiment, the disease or disorder is a neurological disorder.
In a particularly prefeπed embodiment, the neurological disorder is selected from the group consisting of peripheral neuropathy caused by physical injury or disease state, traumatic brain injury, physical damage to the spinal cord, stroke associated with brain damage, focal ischemia, global ischemia, reperfusion injury, demyelinating disease and neurological disorder relating to neurodegeneration. Another prefeπed embodiment is when the reperfusion injury is a vascular stroke. Yet another prefeπed embodiment is when the peripheral neuropathy is caused by Guillain-Baπe syndrome.
Still another prefeπed embodiment is when the demyelinating disease is multiple sclerosis. Another prefeπed embodiment is when the neurological disorder relating to neurodegeneration is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, and amyotrophic lateral sclerosis.
The term "preventing neurodegeneration" includes the ability to prevent neurodegeneration in patients newly diagnosed as having a neurodegenerative disease, or at risk of developing a new degenerative disease and for preventing further neurodegeneration in patients who are already suffering from or have symptoms of a neurodegenerative disease. The term "treatment" as used herein covers any treatment of a disease and/or condition in an animal, particularly a human, and includes:
(i) preventing a disease and/or condition from occurring in a subject which may be predisposed to the disease and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease and/or condition, i.e., arresting its development; or
(iii) relieving the disease and/or condition, 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 vascular stroke and global and focal ischemia. As used herein, the term "neurodegenerative 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 arterial blood.
Global ischemia occurs under conditions in which blood flow to the entire brain ceases for a period of time, such as may result from cardiac aπest. Focal ischemia occurs under conditions in which a portion of the brain is deprived of its normal blood supply, such as may result from thromboembolytic occlusion of a cerebral vessel, traumatic head injury, edema, and brain tumors. The term "cardiovascular disease" relates to myocardial infarction, angina pectoris. vascular or myocardial ischemia, and related conditions as would be known by those of skill in the art which involve dysfunction of or tissue damage to the heart or vasculature, and especially, but not limited to, tissue 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 therapeutically effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagnetic radiation and/or to promote the treatment of diseases which are treatable with electromagnetic radiation. Diseases which are treatable with electromagnetic radiation include neoplastic diseases, benign and malignant tumors, and cancerous cells. Electromagnetic radiation treatment of other diseases not listed herein are also contemplated by the present invention. The terms "electromagnetic radiation" and "radiation" as used herein includes, but is not limited to, radiation having the wavelength of 10"20 to 10° meters. Prefeπed embodiments of the present invention employ the electromagnetic radiation of: gamma- radiation (10"20 to 10'13 m) x-ray radiation (10"u to 10"9 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, the compounds of the invention inhibit PARP activity and, thus, are believed to be useful for treating neural tissue damage, particularly damage resulting from cerebral ischemia and reperfusion injury or neurodegenerative diseases in animals. The term "nervous tissue" refers to the various components that make up the nervous system including, without limitation, neurons, neural support cells, glia. Schwann cells, vasculature contained within and supplying these structures, the central nervous system, the brain, the brain stem, the spinal cord, the junction of the central nervous system with the peripheral nervous system, the peripheral nervous system, and allied structures.
Further, according to the invention, an effective therapeutic amount of the compounds and compositions described above are administered to animals to effect a neuronal activity, particularly one that is not mediated by NMDA nemotoxicity. Such neuronal activity may consist of stimulation of damaged neurons, promotion of neuronal regeneration, prevention of neurodegeneration and treatment of a neurological disorder. Accordingly, the present invention further relates to a method of effecting a neuronal activity in an animal, comprising administering 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 invention include, without limitation, trigeminal neuralgia; glossopharyngeal neuralgia; Bell's Palsy; myasthenia gravis; muscular dystrophy; amyotrophic lateral sclerosis; progressive muscular atrophy; progressive bulbar inherited muscular atrophy; herniated, ruptured or prolapsed invertebrate disk syndromes; cervical spondylosis; plexus disorders; thoracic outlet destruction syndromes; peripheral neuropathies such as those caused by lead, dapsone, ticks, poφhyria, or Guillain-Baπe syndrome; Alzheimer's disease; Huntington's Disease and Parkinson's disease.
The method of the present invention is particularly useful for treating a neurological disorder selected from the group consisting of: peripheral neuropathy caused by physical injury or disease state; head trauma, such as traumatic brain injury; physical damage to the spinal cord; stroke associated with brain damage, such as vascular stroke associated with hypoxia and brain damage, focal cerebral ischemia, global cerebral ischemia, and cerebral reperfusion injury; demyelinating diseases, such as multiple sclerosis; and neurological disorders related to neurodegeneration, such as Alzheimer's Disease, Parkinson's Disease, Huntington's Disease and amyotrophic lateral sclerosis (ALS).
Treating Other PAKP-Related Disorders The compounds, compositions and methods of the present invention are particularly useful for treating or preventing tissue damage resulting from cell death or damage due to necrosis or apoptosis.
The compounds, compositions and methods of the invention can also be used to treat a cardiovascular disorder in an animal, by administering an effective 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 pectoris, myocardial infarction, cardiovascular tissue damage caused by cardiac arrest, cardiovascular tissue damage caused by 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 vasculature, especially, but not limited to, tissue damage related to PARP activation. For example, the methods of the invention are believed to be useful for treating cardiac tissue damage, particularly damage resulting from cardiac ischemia or caused by reperfusion injury in animals. The methods of the invention are particularly useful for treating cardiovascular disorders selected from the group consisting of: coronary artery disease, such as atherosclerosis; angina pectoris; myocardial infarction; myocardial ischemia and cardiac aπest; cardiac bypass; 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 invention can be used to treat tissue damage resulting from cell damage or death due to necrosis or apoptosis, neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases; to prevent or treat vascular stroke; to treat or prevent cardiovascular disorders; to treat other conditions and/or disorders such as age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, gout, arthritis, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, inflammatory bowel disorders (such as colitis and Crohn's disease), muscular dystrophy, osteoarthritis, osteoporosis, chronic and/or acute pain (such as neuropathic pain), renal failure, retinal ischemia, septic shock (such as endotoxic shock), and skin aging; to extend the lifespan and proliferative capacity of cells; to alter gene expression of senescent cells: or to radiosensitize tumor cells
Further still, the methods of the invention can be used to treat cancer and to radiosensitize tumor cells. The term "cancer" is inteφreted broadly. The compounds of the present invention can be "anti-cancer agents", which term also encompasses "anti-tumor cell growth agents" and "anti-neoplastic agents". For example, the methods of the invention are useful for treating cancers and radiosensitizing tumor cells in cancers such as
ACTH-producing tumors, acute lymphocytic leukemia, acute nonlymphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervical cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma, endometrial cancer, esophageal cancer, 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 peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non-Hodgkin's lymphoma, osteosarcoma, ovarian cancer, ovary (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, retinoblastoma, skin cancer, soft-tissue sarcoma, squamous cell carcinomas, stomach cancer, testicular cancer, thyroid cancer, trophoblastic neoplasms, uterine cancer, vaginal cancer, cancer of the vulva and Wilm's tumor.
The term "radiosensitizer", as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagnetic radiation and/or to promote the treatment of diseases which are treatable with electromagnetic radiation. Diseases which are treatable with electromagnetic radiation include neoplastic diseases, benign and malignant tumors, and cancerous cells. Electromagnetic radiation treatment of other diseases not listed herein are also contemplated by the present invention. The terms "electromagnetic radiation" and "radiation" as used herein includes, but is not limited to, radiation having the wavelength of 10"20 to 10° meters. Prefeπed embodiments of the present invention employ the electromagnetic radiation of: gamma- radiation (10"20 to 10"13 m) x-ray radiation (10"n to 10"9 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). Radiosensitizers are known to increase the sensitivity of cancerous cells to the toxic effects of electromagnetic radiation. Several mechamsms for the mode of action of radiosensitizers have been suggested in the literature including: hypoxic cell radiosensitizers ( e.g., 2-nitroimidazole compounds, and benzotriazine dioxide compounds) promote the reoxygenation of hypoxic tissue and/or catalyze the generation of damaging oxygen radicals; non-hypoxic cell radiosensitizers (e.g., halogenated pyrimidines) can be analogs of DNA bases and preferentially incoφorate into the DNA of cancer cells and thereby promote the radiation-induced breaking of DNA molecules and/or prevent the normal DNA repair mechamsms; and various other potential mechanisms of action have been hypothesized for radiosensitizers in the treatment of disease.
Many cancer treatment protocols cuπently employ radiosensitizers activated by the electromagnetic radiation of x-rays. Examples of x-ray activated radiosensitizers include, but are not limited to, the following: metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, E09, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5-iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same. Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the sensitizing agent. Examples of photodynamic radiosensitizers include the following, but are not limited to: hematopoφhyrin derivatives, Photofrin, benzopoφhyrin derivatives, NPe6, tin etiopoφhyrin SnET2, pheoborbide-a, bacteriochlorophyll-a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same. Radiosensitizers may be administered in conjunction with a therapeutically effective amount of one or more other compounds, including but not limited to: compounds which promote the incoφoration of radiosensitizers to the target cells; compounds which control the flow of therapeutics, nutrients, and/or oxygen to the target cells; chemotherapeutic agents which act on the tumor with or without additional radiation; or other therapeutically effective compounds for treating cancer or other disease. Examples of additional therapeutic agents that may be used in conjunction with radiosensitizers include, but are not limited to: 5-fluorouracil, leucovorin, 5' -amino-5'deoxythymidine, oxygen, carbogen, red cell transfusions, perfluorocarbons (e.g., Fluosol- DA), 2,3-DPG, BW12C, calcium channel blockers, pentoxyfylline, aπtiangiogenesis compounds, hydralazine, and LBSO. Examples of chemotherapeutic agents that may be used in conjunction with radiosensitizers include, but are not limited to: adriamycin, camptothecin, carboplatin, cisplatin, daunorubicin. docetaxel, doxorubicin, interferon (alpha, beta, gamma), interleukin 2, irinotecan. paclitaxel, topotecan, and therapeutically effective analogs and derivatives of the same.
Pharmaceutical Compositions of the Invention The present invention also relates to a pharmaceutical composition comprising (i) a therapeutically effective amount of the compound of formula I, II, III, IV or V and (ii) a pharmaceutically acceptable carrier.
The above discussion relating to the prefeπed embodiments' utility and administration of the compounds of the present invention also applies to the pharmaceutical composition of the present invention.
The term "pharmaceutically acceptable carrier" as used herein refers to any carrier, diluent, excipient, suspending agent, lubricating agent, adjuvant, vehicle, delivery system, emulsifier, disintegrant, absorbent, preservative, surfactant, colorant, flavorant, or sweetener.
For these puφoses, the composition of the invention may be administered orally, parenterally, by inhalation spray, adsoφtion, absoφtion, topically, rectally, nasally, bucally, vaginally, intraventricularly, via an implanted reservoir in dosage formulations containing conventional non-toxic pharmaceutically-acceptable carriers, or by any other convenient dosage form. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, and intracranial injection or infusion techniques.
When administered parenterally, the composition will normally be in a unit dosage, sterile injectable form (solution, suspension or emulsion) which is preferably isotonic with the blood of the recipient with a pharmaceutically acceptable carrier. Examples of such sterile injectable forms are sterile 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 sterile injectable forms may also be sterile injectable solutions or suspensions in non-toxic parenterally-acceptable diluents or solvents, for example, as solutions in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, saline, Ringer's solution, dextrose solution, isotonic sodium chloride solution, and Hanks' solution. In addition, sterile, fixed oils are conventionally employed as solvents or suspending mediums. For this piupose, any bland fixed oil may be employed including synthetic mono- or di-glycerides, corn, cottonseed, peanut, and sesame oil. Fatty acids such as ethyl oleate, isopropyl myristate, and oleic acid and its glyceride derivatives, including olive oil and castor oil, especially in their polyoxyethylated versions, are useful in the preparation of injectables. These oil solutions or suspensions may also contain long-chain alcohol diluents or dispersants.
Sterile saline is a prefeπed carrier, and the compounds are often sufficiently water soluble to be made up as a solution for all foreseeable needs. The carrier may contain minor amounts of additives, such as substances that enhance solubility, isotonicity, and chemical stability, e.g., anti-oxidants, buffers and preservatives. Formulations suitable for nasal or buccal administration (such as self-propelling powder dispensing formulations) may comprise about 0.1% to about 5% w/w, for example 1% w/w of active ingredient. The formulations for human medical use of the present invention comprise an active ingredient in association with a pharmaceutically acceptable carrier therefore and optionally other therapeutic ingredient(s). When administered orally, the composition will usually be formulated into unit dosage forms such as tablets, cachets, powder, granules, beads, chewable lozenges, capsules, liquids, aqueous suspensions or solutions, or similar dosage forms, using conventional equipment and techniques known in the art. Such formulations typically include a solid, semisolid, or liquid carrier. Exemplary carriers include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, mineral oil, cocoa butter, oil of theobroma, alginates, tragacanth, gelatin, syrup, methyl cellulose, polyoxyethylene sorbitan monolaurate, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, and the like.
The composition of the invention is preferably administered as a capsule or tablet containing a single or divided dose of the inhibitor. Preferably, the composition is administered as a sterile solution, suspension, or emulsion, in a single or divided dose. Tablets may contain carriers such as lactose and corn starch, and/or lubricating agents such as magnesium stearate. Capsules may contain diluents including lactose and dried corn starch.
A tablet may be made by compressing or molding the active 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, lubricant, 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 carrier moistened with an inert liquid diluent.
The compounds of this invention may also be administered rectally in the form of suppositories. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at room temperature, but liquid at rectal temperature, and, therefore, will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax, and polyethylene glycols.
Compositions and methods of the invention also may utilize controlled release technology. Thus, for example, the inventive compounds may be incoφorated into a hydrophobic polymer matrix for controlled release over a period of days. The composition of the invention may then be molded into a solid implant, or externally applied patch, suitable for providing efficacious concentrations of the PARP inhibitors over a prolonged period of time without the need for frequent re-dosing. Such controlled release films are well known to the art.
Particularly prefeπed are transdermal delivery systems. Other examples of polymers commonly employed for this piupose that may be used in the present invention include nondegradable ethylene-vinyl acetate copolymer an degradable lactic acid-glycolic acid copolymers which may be used externally or internally. Certain hydrogels such as poly(hydroxyethylmethacrylate) or poly(vinylalcohol) also may be useful, but for shorter release cycles than the other polymer release systems, such as those mentioned above. In a prefeπed embodiment, the carrier is a solid biodegradable polymer or mixture of biodegradable polymers with appropriate time release characteristics 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 period of time without the need for frequent re-dosing. The composition of the present invention can be incoφorated into the biodegradable polymer or polymer mixture in any suitable manner known to one of ordinary skill in the art and may form a homogeneous matrix 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 polymer or polymer mixture is used to form a soft "depot" containing the pharmaceutical composition of the present invention that can be administered as a flowable liquid, for example, by injection, but which remains sufficiently viscous to maintain the pharmaceutical composition within the localized area around the injection site. The degradation time of the depot so formed can be varied from several days to a few years, depending upon the polymer selected and its molecular weight. By using a polymer composition in injectable form, even the need to make an incision may be eliminated. In any event, a flexible or flowable delivery "depot" will adjust to the shape of the space it occupies with the body with a minimum of trauma to suπounding tissues. The pharmaceutical composition of the present invention is used in amounts that are therapeutically effective, and may depend upon the desired release profile, the concentration of the 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 composition in amounts that are therapeutically effective. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, welling, or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers. In addition, they may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating, 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 effective therapeutically as central nervous system targets, the compounds of the present invention should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier can be effectively administered by an intraventricular route or other appropriate delivery system suitable for administration to the brain.
Doses of the compounds preferably include pharmaceutical dosage units comprising an efficacious quantity of active compound. By an efficacious quantity is meant a quantity sufficient to inhibit PARP and derive 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 neurodegenerative diseases.
For medical use, the amount required of the active ingredient to achieve a therapeutic effect will vary with the particular compound, the route of administration, the mammal under treatment, and the particular disorder or disease being treated. A suitable systematic dose of a compound of the present invention or a pharmacologically acceptable salt thereof for a mammal suffering from, or likely to suffer from, any of condition as described hereinbefore is in the range of about 0.1 mg/kg to about 100 mg/kg of the active ingredient compound, the most prefeπed dosage being about 1 to about 10 mg/kg. It is understood, however, that a specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the severity of the particular disease being treated and form of administration.
It is understood that the ordinarily skilled physician or veterinarian will readily determine and prescribe the effective amount of the compound for prophylactic or therapeutic treatment of the condition for which treatment is administered. In so proceeding, the physician or veterinarian could employ an intravenous bolus followed by an intravenous infusion and repeated administrations, parenterally or orally, as considered appropriate. While it is possible for an active ingredient to be administered alone, it is preferable to present it as a formulation. When preparing dosage form incoφorating the compositions of the invention, the compounds may also be blended with conventional excipients such as binders, including gelatin, pregelatinized starch, and the like; lubricants, such as hydrogenated vegetable oil, stearic acid, and the like; diluents, such as lactose, mannose, and sucrose; disintegrants, such as carboxymethylcellulose and sodium starch glycolate; suspending agents, such as povidone, polyvinyl alcohol, and the like; absorbants, such as silicon dioxide; preservatives, such as methylparaben, propylparaben, and sodium benzoate; surfactants, such as sodium lauryl sulfate, polysorbate 80, and the like; colorants such as F.D.& C. dyes 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 described herein.
PARP Assays
A convenient method to determine ICS0 of a PARP inhibitor compound is a PARP assay using purified recombinant human PARP from Trevigan (Gaithersburg, MD), as follows: The PARP enzyme assay is set up on ice in a volume of 100 microliters consisting of 100 mM Tris-HCl (pH 8.0), 1 mM MgCl_, 28 mM KC1, 28 mM NaCl, 0.1 mg/ml of DNase I activated herring sperm DNA (Sigma, MO), 3.0 micromolar [3H]nicotinamide adenine dinucleotide (470 mci/mmole), 7 micrograms/ml PARP enzyme, and various concentrations of the compounds to be tested. The reaction is initiated by incubating the mixture at 25°C. After 15 minutes of incubation, the reaction is terminated by adding 500 microliters of ice cold 20% (w/v) trichloroacetic acid. The precipitate formed is transfeπed onto a glass fiber filter (Packard Unifilter-GF/B) and washed three times with ethanol. After the filter is dried, the radioactivity is determined by scintillation counting. The compounds of this invention were found to have potent enzymatic activity in the range of a few nM to 20 μM in ICS0 in this inhibition assay.
Using the PARP assay described above, approximate IC50 (μM) values were obtained as shown in the following Table III:
TABLE m
Figure imgf000071_0001
Figure imgf000072_0001
Focal cerebral ischemia
The following focal cerebral ischemia assay is useful for determining the PARP inhibiting effects of the compounds of the present invention. The following examples demonstrate that compounds related to those of the present invention are effective in inhibiting PARP activity.
Focal cerebral ischemia is produced by cauterization of the right distal MCA (middle cerebral artery) with bilateral temporary common carotid artery occlusion in male Long-Evans rats for 90 minutes. All procedures performed on the animals are approved by the University Institutional Animal Care and Use Committee of the University of Pennsylvania. A total of 42 rats (weights: 230-340 g) obtained from Charles River were used in this study. The animals fasted overnight with free access to water prior to the surgical procedure.
Two hours prior to MCA occlusion, varying amounts (control, π=14; 5 mg kg, n=7; 10 mg/kg, n=7; 20 mg/kg, n=7; and 40 mg/kg, n=7) of the compound, 3,4-dihycfro-5-[4-(l-piperidinyl)-butoxy]-l(2ff)- isoquinolinone ("DPQ") were dissolved in dimethyl sulfoxide (DMSO) using a sonicator. A volume of 1.28 ml/kg of the resulting solution was injected intraperitoneally into fourteen rats.
The rats were then anesthetized with halothane (4% for induction and 0.8%- 1.2% for the surgical procedure) in a mixture of 70% nitrous 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 (Harvard Apparatus Limited, Kent, U.K.). A catheter (PE-50) was placed into the tail artery, and arterial pressure was continuously monitored and recorded on a Grass polygraph recorder (Model 7D, Grass Instruments, Quincy, Massachusetts). Samples for blood gas analysis (arterial pH. PaO_ and PaC02) were also taken from the tail artery catheter and measured with a blood gas analyzer (ABL 30, Radiometer, Copenhagen, Denmark). Arterial blood samples were obtained 30 minutes after MCA occlusion. The head of the animal was positioned in a stereotaxic frame, and a right parietal incision between the right lateral canthus and the external auditory meatus was made. Using a dental drill constantly cooled with saline, a 3 mm bun hole was prepared over the cortex supplied by the right MCA, 4 mm lateral to the sagittal suture and 5 mm caudal to the coronal suture. The dura mater and a thin inner bone layer were kept, care being taken to position the probe over a tissue area devoid of large blood vessels. 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 right parietal cortex was continuously monitored with a laser Doppler flowmeter (FloLab, Moor, Devon, U.K., and Periflux 4001, Perimed, Stockholm, Sweden).
Focal cerebral ischemia was produced by cauterization of the distal portion of the right MCA with bilateral temporary common carotid artery (CCA) occlusion by the procedure of Chen et al., "A Model of Focal Ischemic Stroke in the Rat: Reproducible Extensive Cortical Infarction", Stroke 17:738-43 (1986) and/or Liu et al., "Polyethylene Glycol-conjugated Superoxide Dismutase and Catalase Reduce Ischemic Brain Injury", Am. J. 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 observation of the rostral end of the zygomatic arch at the fusion point using a dental drill, and the dura mater overlying the MCA was cut. The MCA distal to its crossing with the inferior cerebral vein was lifted by a fine stainless steel hook attached to a micromampulator and, following bilateral CCA occlusion, the MCA was cauterized with an electrocoagulator. The bun hole was covered with a small piece of Gelform, and the wound was sutured to maintain the brain temperature within the normal or near- normal range.
After 90 minutes of occlusion, the carotid loops were released, the tail arterial catheter was removed, and all of the wounds were sutured. Gentamicin sulfate (10 mg/ml) was topically applied to the wounds to prevent infection. The anesthetic was discontinued, and the 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. Twenty-four hours after MCA occlusion, the rats were sacrificed with an intraperitoneal injection 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 brain was then sectioned in the coronal plane at 2 mm intervals using a rodent brain matrix (RBM-4000C, ASI Instruments, Waπen, Michigan). The brain slices were incubated in phosphate-buffered saline containing 2% 2,3,5-triphenyltetrazolium chloride (TTC) at 37°C for ten minutes. Color photographs were taken of the posterior surface of the stained slices and were used to determine the damaged area at each cross-sectional level using a computer-based image analyzer (NIH Image 1.59). To avoid artifacts due to edema, the damaged area was calculated by subtracting 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 Method for Measuring Brain Infarct Volume", J. Cereb. Blood Flow Metabol. 10:290-93 (1990), the disclosure of which is hereby incoφorated by reference. The total volume of infarction was calculated by summation of the damaged volume of the brain slices. The cauterization of the distal portion of the right MCA with bilateral temporary CCA occlusion consistently produced a well-recognized cortical infarct in the right MCA territory of each test animal. There was an apparent uniformity 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-sectional infarct area at representative levels along the rostrocaudal axis was measured from the interaural line in non-treated animals and in animals treated with 10 mg kg of 3,4-dihydro-5-[4-(l-piperidinyl)-butoxy]-l( _τr)-isoquinolinone. The area of damage was expressed as mean ± standard deviation. Significant differences between the 10 mg-treated group and the control group were indicated (*p<0.02, **p<0.01, **p<0.001). The 5 mg/kg and 20 mg/kg curves fell approximately halfway between the control and the 10 mg kg curves, whereas the 40 mg/kg curve was close to the control. The 5, 20 and 40 mg/kg curves were omitted for clarity.
PARP inhibition led to a significant decrease in the damaged volume in the 5 mg/kg-treated group (106.7 ± 23.2 mm3, p<0.001), the 10 mg/kg-treated group (76.4 + 16.8 mm3, p<0.001), and the 20 mg/kg- treated group (110.2 + 42.0 mm3, p<0.01), compared to the control 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 variance (ANOVA) followed by Student's t-test for individual comparisons.
There was no significant difference between the 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 intraperitoneal administration of 3,4-dihydro-5-[4-(l-piperidinyl)-butoxy]- l(2_7)-isoquinolinone on the infarct volume was depicted graphically. The volumes of infarct were expressed as mean + standard deviation. Significant differences between the treated groups and the control group were indicated (*p<0.01, **p<0.001). It is not clear why a high dose (40 mg/kg) of the PARP inhibitor, 3,4-dihydro-5- [4-(l-ρiperidinyl)-butoxy]-l(2_Y)-isoquinolinone, was less neuroprotective. The U-shaped dose-response curve may suggest dual effects of the compound.
However, overall, the in vivo administration of the inhibitor led to a substantial reduction in infarct volume in the focal cerebral ischemia model in the 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 arterial 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 five groups, as shown below in Table IV. A "steady state" MABP was taken following completion of the surgical preparation, just prior to occlusion; an "ischemia" MABP was taken as the average MABP during occlusion. TABLE IV
Figure imgf000075_0001
= Significantly different from the steady state value, p<0.05. ** = Significantly different from the steady state value, pθ.01. There were no significant differences in any physiological parameter, including mean arterial blood pressure (MABP), prior to MCA and CCA occlusion among the five groups. Although MABP was significantly elevated following occlusion in all five groups, there were no significant differences in MABP during the occlusion period among the groups.
Since the blood flow values obtained from the laser doppler were in arbitrary units, only percent changes from the baseline (prior to occlusion) were reported. Right MCA and bilateral CCA occlusion produced a significant decrease in relative blood flow in the right parietal cortex to 20.8 ± 7.7 % 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 ± 11.2 % in the 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 entire occlusion period in any group.
Following release of the carotid occlusions, a good recovery of blood flow (sometimes hyperemia) was observed in the right MCA territory of all animals. Reperfusion of the ischemic tissue resulted in the formation of NO and peroxynitrite, in addition to oxygen-derived free radicals. All of these radicals have been shown to cause DNA strand breaks and to activate PARP. This example provided evidence that the related compounds of the present invention are effective in inhibiting PARP activity.
Exemplified compounds of the present invention may be 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 by the institutional Animal Studies Committee, and are in accordance with the PHS Guide for the Care and Use of Laboratory Animals, USDA Regulations, and the AVMA Panel on Euthanasia guidelines.
The animals are anesthetized with isofluorane (induction, 3%; maintenance, 1.25% in a mixture of 30% 02 and 70% N02 through a face mask. The rectal temperature is maintained at 37 °C with a homeothermic blanket (Harvard Apparatus, South Natick, MA). First, an iv catheter is inserted into the left femoral vein and the line run up through the nape of the neck for connection to a tethered swivel (Instech Laboratories, Plymouth Meeting, PA) and remote infusion pump (Stoelting Inc., Wood Dale, IL). In some cases, the right femoral artery is cannulated for monitoring arterial blood pressure and heart rate and for obtaining blood samples for arterial blood gas. The right middle cerebral artery (MCA) is then exposed by making vertical skin incision midway between the right eye and ear and overlying skull is removed with a dental drill (Chen et al, 1986). After incision of the dura, the artery is coagulated at the level of the inferior cerebral vein with a bipolar cautery unit (Valleylab NS2000, Boulder, CO), and cut to prevent spontaneous reperfusion (Takahashi et al., 1997). Both common carotid arteries (CCAs) that had been previously isolated and freed of soft tissues and nerves are then ligated using non-traumatic aneurysm clips. After the wounds are closed with surgical clips, the animals are allowed to recover from anesthesia and returned to their 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 removed from the infusion tether, briefly reanesthetized with isofluorane, and the carotid clips are removed . The animal is returned to its warm cage and reconnected to the iv infusion pump for the duration of the experiment.
At 24 lirs after permanent MCAO, animals are sedated with ketamine and the heads removed by guillotine. Brains are removed, cooled in ice-cold saline, and sliced into 2 mm coronal sections using a rat brain matrice (Harvard 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 right hemisphere is calculated by summation of the direct (TTC-negative) and indirect measures of the infarct areas in the component brain slices. The infarcted volumes in vehicle and drug-treated groups (n=8) are tested for significant statistical difference using an unpaired Student-t test.
Various doses of the compounds of the invention may be tested in this model. The compounds are administered in either a single dose or a series of multiple doses, i.p. or i.v., at different times, both before or after the onset of ischemia. Compounds of the invention are expected to provide protection from ischemia in the range of about 0 to 80%. Heart Ischemia Reperfusion Injury
The experiments of the heart ischemia/reperfusion injury model is performed using female Sprague- Dawley rats weighing 250-3 OOg which are anesthetized with sodium pentobarbital at dose of 65 mg kg intraperitoneally. The rectal temperature is maintained at 37°C by using a homeothermic blanket system
(Harvard Apparatus, South Natick, MA). The trachea is cannulated and the rat is ventilated with Room Air by using Harvard Rodent Ventilator (Harvard Apparatus, South Natick, MA). The left jugular vein is cannulated with PE-50 tubing for drug delivery. The right carotid artery is cannulated for BP measurement. The heart is exposed by opening the chest at the 4th left intercostal space. A main left branch of coronary artery (LAD) is occluded by 4-0 silk ligature for 30 min of ischemia and released for 90 min of reperfusion. During the experiment, arterial BP and EKG are monitored by Micro-Med Cardiovascular System (Louisville, KY).
At the end of reperfusion, the LAD coronary artery is re-occluded and about 2 ml of 5% Evans Blue dye is injected through i.v. line to distinguish the 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 sections with 2 mm thickness and stained in 1% TTC solution for 30 min at 37°C. The right ventricle is trimmed off. Infarct area, risk area and total left ventricular area in each face of the section are measured by using an image analysis system (BIOQUANT, Nashville. TN). The infarct size is calculated as the percent total infarct volume of the total risk volume.
For drug treated group, compounds are administered according to the following three protocols: 1. Single dose of compound is given 60 min prior 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 by i.v. infusion until the 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 drug-treated group, there is a coπesponding vehicle treated group with n=6 or n=8. The difference between vehicle and drug treated groups is compared by using unpaired t-test with p<0.05. Various doses of compounds are tested in this model. The compounds are given in either single or multiple doses, i.p or i.v., at different times before or after the onset of ischemia. The compounds of this invention are expected to have ischemia/reperfusion injury protection in the range of 10 to 40 percent in this assay.
As a result of the PARP inhibition activity, the compounds of this invention are expected to protect against ischemia-induced degeneration of rat cortical neurons in vitro and thus may be useful in disorders arising from cerebral ischemia such as stroke, septic shock, or CNS degenerative disorders. They may also be useful in protecting the spinal cord following trauma. As an experimental result of ischemia/reperfusion injury in rats, the present invention is further directed to a method of prophylactic or therapeutic treatment of heart attack, cardiac aπest, cardiac bypass, hyperglycemia, diabetes, or risk of damage which comprises administering an effective amount of a compound of the present invention for PARP inhibition in unit dosage form. In vitro Radiosensitization
In vitro radiosensitization may be measured with the use of a human prostate cancer cell line, PC-3s, which are plated in 6 well dishes and grown at monolayer cultures in RPMI1640 supplemented with 10% FCS. The cells are maintained at 37°C in 5% C02 and 95% air. The cells are exposed to a dose response (0.1 mM to 0.1 μM) of 3 different PARP inhibitors prior to iπadiation at one sublethal dose level. For all treatment groups, the six well plates are exposed at room temperature in a Seifert 250kV/15mA iπadiator with a 0.5 mm Cu/1 mm. Cell viability is examined by exclusion of 0.4% trypan blue. Dye exclusion is assessed visually by microscopy and viable cell number was calculated by subtracting the number of cells from the viable cell number and dividing by the total number of cells. Cell proliferation rates are calculated by the amount of 3H-thymidine incoφoration post-iπadiation. The PARP inhibitors are expected to radiosensitize the cells.
Measuring Altered Gene Expression in mRNA Senescent Cells
Gene expression alteration may be measured with human fibroblast BJ cells which, at Population Doubling (PDL) 94, are plated in regular growth medium and then changed to low serum medium to reflect physiological conditions described in Linskens, et al., Nucleic Acids Res. 23 : 16:3244-3251 (1995). A medium of DMEM/199 supplemented with 0.5% bovine calf serum is used. The cells are treated daily for 13 days. The control cells are treated with and without the solvent used to administer the PARP inhibitor. The untreated old and young control cells are tested for comparison. RNA is prepared from the treated and control cells according to the techniques described in PCT Publication No. 96/13610 and Northern blotting is conducted. Probes specific for senescence-related genes are analyzed, and treated and control cells compared. In analyzing the results, the lowest level of gene expression is arbitrarily set at 1 to provide a basis for comparison. Three genes particularly relevant to age-related changes in the skin are collagen, collagenase and elastin. West, Arch. Derm. 130:87-95 (1994). Elastin expression of the cells treated with the PARP inhibitor is expected to be significantly increased in comparison with the control cells. Elastin expression should be significantly higher in young cells compared to senescent cells, and thus treatment with 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 treatment with the PARP inhibitors.
Neuroprotective Effects of PARP Inhibitors on Chronic Constriction Injury (CCI) in Rats Adult male Sprague-Dawley rats, 300-350 g, are anesthetized with intraperitoneal 50 mg/kg sodium pentobarbital. Nerve ligation is performed by exposing one side of the rat's sciatic nerves 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 intratliecal catheter and inserting of a gentamicin sulfate-flushed polyethylene (PE-10) tube into the subarachnoid space through an incision at the cisterna magna. The caudal end of the catheter is gently threaded to the lumbar enlargement and the rostral end is seemed with dental cement to a screw embedded in the skull and the skin wound is closed with wound clips.
Thermal hyperalgesia to radiant heat is assessed by using a paw-withdrawal test. The rat is placed in a plastic cylinder on a 3-mm thick glass plate with a radiant heat source from a projection bulb placed directly under the plantar surface of the rat's hindpaw. The paw-withdrawal latency is defined as the time elapsed from the onset of radiant heat stimulation to withdrawal of the rat's hindpaw.
Mechanical hyperalgesia is assessed by placing the rat in a cage with a bottom made of perforated metal sheet with many small square holes. Duration of paw-withdrawal is recorded after pricking the mid-plantar surface of the rat's hindpaw with the tip of a safety pin inserted through the cage bottom.
Mechano-allodynia 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 slowly until it bends. A threshold force of response is defined as the first filament in the series to evoke at least one clear paw- withdrawal out of five applications.
Dark neurons are observed bilaterally within the spinal cord dorsal horn, particularly in laminae I-II, of rats 8 days after unilateral sciatic nerve ligation as compared with sham operated rats. Various 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.
Treatment and prevention of gout and symptoms of gout
Deposition of crystals of monosodium mate (MSU crystals) in the joint articular space is the etiological cause of inflammatory pathologies such as gout and pseudogout. Clinically, these inflammatory diseases are associated with oedema and erythema of the joints with consequently severe pain. A strong infiltration of leucocytes in the intraarticular and periarticular space leading to: 1) acute, episodic articular and periarticular inflammation, and 2) chronic articular changes, are also characteristic of this pathology. It has long been clear that neutrophils are the predominant cell type recovered from these inflammatory joints (Dieppe et al., (1979). Synovial fluid crystals. Q. J. Med. XLVHI: 533-553; Terkletaub, (1991). Monocyte-derived neutrophil chemotactic factor/interleukin-8 is a potential mediator of crystal-induced inflammation. Arth. Rheum. 34: 894- 903.). A better understanding of the inflammatory processes elicited by MSU crystals, and the fact that there is a clear relationship between these crystals and gouty arthritis, has prompted the characterisation of experimental models of crystal-induced inflammation. Examples of models where crystal challenge has led to cell recruitment into specific cavities, are canine joints (Phelps & McCarty, 1966, Ann Int. Med. 9: 115-125), rat pleurisy (Deporter et al, 1919, Br. J. Pharmacol. 65: 163-165; Sedgwick et al., 1985, Agents Actions 17: 209-213), and utilisation of a pre-formed rat air-pouch (Brookes et al, 1987). The latter experimental system has shown that neutrophil accumulation was related to generation of chemoattractants such as LTB4, which was subsequently inhibited by colchicine (Brooks et al., 1987, Br. J. Pharmacol. 90: 413-419).
Neutropliils have been shown to be activated by MSU crystals, releasing an array of mediators that may be, in part, responsible for the local and systemic inflammatory manifestations found in crystal-induced joint disorders. The crystals interact with neutrophils leading to the release of lysomal enzymes (Hoffstein et al., 1975, Arth. Rheum. 18: 153-165), release of oxygen derived free radicals (Simchowitz et al., 1982, Arth. Rheum. 25: 181-188; Abramson et α/., 1982, Arthr Rheum. 25: 174-180), induction of phospholipase A2 (PLA2) in leucocytes (Bomalaski et al, 1990, J. Immunol 145: 3391-3397), and activation of synthesis of 5- lipoxygenase products (Poubelle et al, 1987, Biochem. Biophys. Res. Commun. 149: 649-657). In vitro, MSU crystals have been shown to release the cytokine interleukin-lβ (IL-lβ) from human neutrophils, adding this stimulus to a list of others that also release this cytokine, such as zymosan, LPS, phorbol esters, granulocyte macrophage-colony stimulating hormone (GM-CSF) and TNF-alpha. Furthermore it has also been shown that human monocytes and synoviocytes can synthesise and release various cytokines such as IL-6 and D -8 (Guerne et al, 1989, Arth. Rheum. 32: 1443-1452; Terkeltaub et al, 1991, Arth. Rheum. 34: 894-903). In addition, colchicine selectively inhibits MSU crystal- and TNF-G induced release of IL-lβ (Roberge et al, 1994, J. Immunol. 152: 5485-5494).
In experimental models of gout the synthesis of a CXC chemokine selective for neutrophils, such as IL- 8, has also been observed, but not that of a CC chemokine monocyte chemoattractant protein-1 (MCP-1) (Hachicha et al, 1995, . Exp. Med. 182: 2019-2025). These results suggest that production of IL-8 and abolition of the release of MCP-1, will lead to an event where, theoretically there will be a recruitment of neutrophils but not mononuclear cells. This hypothesis is in accordance with the pathological state of gout and pseudogout, where the predominant inflammatory cell is the neutrophil (Hachicha et al, 1995). In addition MSU crystal activation of mononuclear phagocytes, which are normally found in the joint space, also induces secretion of IL-8 (Terkeltaub et al, 1991). The importance of IL-8 in this pathology has been shown in synovial fluids of patients with acute gouty arthritis where it occurs in elevated amounts (Terkeltaub et al, 1991; di
Giovine et al, 1991, J. Clin. Invest. 87: 1375-1381). The use of a neutralising antibody against IL-8 has been shown significantly to attenuate the crystal induced joint swelling at 12 h and neutrophil infiltration into arthritic joints at 12 and 24 h in a rabbit model ( shimura et al, 1997, J. Leukoc. Biol. 62: 444-449).
These studies demonstrate the importance of both the emigrating neutrophil and the chemokine IL-8, as well as the release of this and other cytokines from resident cells such as the synoviocytes, macrophages and mast cells in treating gout. Since neutrophils are not present or are extremely rare in normal synovial fluid, enhanced neutrophil-endothelial adhesion is necessary for gout to occur (Terkeltaub, 1996, In. Koopman, W.J. editor. Arthritis and allied conditions: a textbook of rheumatology. Baltimore: Williams and Wilkins: pp. 2085- 2102, and Terkeltaub, 1992, In Inflammation. Basic Principles and Clinical Correlates, ed. by J.I.Gallin, I.M. Goldstein and R.Snyderman, pp 977-981, Raven Press, New York). IL-lβ and TNF-alpha may be critical in mediating the rapid up-regulation of the major endothelial ligand for neutrophils. For instance rapid and prolonged expression of E-selectin in response to injection of urate crystals has been demonstrated in pig skin (Chapman et al, 1996, Br. J. Rheumatol. 35: 323-334). The release of cytokines, chemokines and products of the arachidonic acid cascade system lead to the recruitment of neutrophils in this pathology, and inhibition of these leads to an attenuation of the pathology.
The following gout model was used to test a PARP inhibitor according to the present invention.
Male outbread Swiss albino mice (20-22 g body weight) were purchased from Banton and Kingsman (T.O. strain; Hull, Humberside) and maintained on a standard chow pellet diet with tap water ad libitum and a 12:00 h light /dark cycle. All animals were housed for 1 week prior to experimentation to allow body weight to reach 28-30 g. l,llb-dihydrobenzopyrano[4,3,2-_fe ]isoquinolin-l-one was dissolved in 100% DMSO at room temperature at a concentration of 45 mg in 2 ml. The compound was then injected into the peritoneal cavity, so as each mouse received a single dose coπesponding to 45 mg/2 ml/kg (e.g. 60 μl for a mouse of 30 g). Control mice received DMSO at 2 ml/kg i.p. A third group of mice which were left untreated were added to control for potential effects of the vehicle. The study involved therefore, the following three groups: group A, untreated mice, n=6, group B, DMSO-treated mice, n=8. and group C, mice treated with l,llb-dihydrobenzopyrano[4,3,2- de ]isoquinolin-l-one, n=8
MSU crystal-induced neutrophil recruitment was tested as follows. In all cases, mice were treated 1 h after the treatment noted above, with MSU crystals. A homogenous suspension of MSU crystals was obtained by a 30 min rotation. Peritonitis was induced by injection of 3 mg MSU crystals in 0.5 ml PBS (0.1 M, pH 7.4), and the recruitment of neutrophils into the cavity evaluated at the 6 h time point (Getting et al, 1997, J. Pharmacol. Exp. Ther. 283: 123-130). Animals were then euthanised by C02 exposure and the peritoneal cavity washed with 3 ml of PBS supplemented with 3 mM EDTA and 25 U/ml heparin.
An aliquot (100 μl) of the lavage fluid was then diluted 1:10 in Turk's solution (0.01% crystal violet in 3% acetic acid). The samples were then vortexed and 10 μl of the stained cell solution were placed in a Neubauer haematocymometer and neutrophils numbers counted using a light microscope (Olympus B061). Cell-free supernatants have been prepared by centrifugation and stored for potential future analysis.
Data are shown for single mice, and also shown as mean ± S.E. of (n) mice per group. Statistical differences were determined by ANOVA, plus Bonfeπoni test. A P value <0.05 was taken as significant. TABLE V reports the number of neutrophils as measured 6 h post-MSU crystal injection in the three experimental groups. TABLE V:
Effect of l,llb-dihydrobenzopyrano[4,3,2-fife ]isoquinoIin-l-one on MSU crystal induced neutrophil migration as evaluated at the 6 h time-point.
Mouse Group Neutrophil Group Neutrophil Group Neutrophil
No. Numbers Numbers Numbers
1 A 4.9 B 6.0 C 5.1
2 A 5.4 B 6.6 C 2.1
3 A 6.3 B 7.5 c 2.4
4 A 6.9 B 7.8 c 2.4
5 A 5.7 B 5.1 c 3.0
6 A 6.0 B 5.7 c 3.0
7 B 5.7 c 2.7
8 B 6.0 c 2.1
Legend: Mice were left untreated (group A), received vehicle DMSO (2 ml/kg i.p. ; group B) or 1, 1 lb- dihydrobenzopyrano[4,3,2--/e ]isoquinolin-l-one (45 mg/kg i.p.; group C), 1 h prior to peritoneal injection of 3 mg MSU crystals at time 0. Neutrophil influx in the peritoneal cavity was measured at the 6 h time-point after collection of the lavage fluids and specific staining as described in the experimental section. Values for neutrophil numbers are Iff per mouse.
TABLE VI illusfrates these data as mean =fc S.E. It can be seen that DMSO produced a modest not. significant increase in cell migration (+7%). In confrast, the exemplary compound of the present invention , at the dose of 45 mg/kg, significantly reduced cell influx, with a calculated 55% of inhibition vs. the vehicle group.
TABLE VI: Accumulation of data for the effect of the exemplified compound of the present invention (means).
Experimental Group Stimulus Neutrophils
(106 per mouse)
A MSU crystals (3 mg) 5.87 ± 0.28 (6)
B MSU crystals (3 mg) 6.30 ± 0.33 (8)
C MSU crystals (3 mg) 2.85 ± 0.34 (8) *
Legend: as in TABLE IV. Values are mean ± S.E. of (n) mice per group. *P<0.05 vs. group B. The invention being thus described, it will be obvious that the same may be varied in many ways. Such variations are not to be regarded as a departure from the spirit and scope of the invention, and all such modifications are intended to be included within the scope of the following claims. All references cited herein are incoφorated in their entirety by reference herein.

Claims

We claim:
1. A compound of Formula I:
Figure imgf000084_0001
(I)
or a pharmaceutically acceptable salt, hydrate, prodrug, or mixtures thereof, wherein:
Ri hydrogen or halogen;
R2 is hydrogen, hydroxyl, amino, nifroso, methyl, amino methyl or carboxylic acid; one of R3 and R4 is -QP and the other of R3 and R4 is one of hydrogen, methyl, trifluoromethyl, nifro, amino, halogen and 1-piperazine, wherein Q is one of
Figure imgf000084_0002
P is Z-(N(R5R<;)), Z or a 5 or 6 membered substituted or unsubstituted aromatic or non-aromatic ring which contains 0, 1, 2 or 3 heteroatoms selected from the group consisting of O, N, S and a combination of two or three of O, N and S;. wherein A is carbon or S=0,
X is O, S , N or an N-substituted amino acid; provided that, when X is O or S, then Y is absent, when X is N, then Y is hydrogen, -Cβ straight or branched chain alkyl, optionally substituted alkoxy or alkyl amino, or Y and Z are taken together to form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic aromatic or non-aromatic ring which contains 1, 2 or 3 heteroatoms selected from O, N, S and mixtures combination;
Z is hydrogen, a direct bond, a carbonyl, an optionally substituted C1-C5 straight or branched chain alkyl, cycloalkyl, carboxy, optionally substituted Cι-C6 ether, aryl or heteroaryl, alkylalkenyl, alkynyl, alkylhalo, straight or branched chain or CH2COOH, provided that when P is Z, Z is not hydrogen and
R5 and Re are independently hydrogen, lower alkyl, lower alkenyl, lower alkanol, heterocycle, heteroaryl, alkoxy, aryloxy, alkylamino, arylamino, CH2COOH, or R5 and R5 taken together form a 5, 6 or 7 membered substituted or unsubstituted heterocyclic ring containing 1, 2 or 3 heteroatoms selected from O, N, S and combinations thereof.
2. A compound of claim 1 selected from the group consisting of
Figure imgf000085_0001
Figure imgf000085_0002
Figure imgf000086_0001
Figure imgf000086_0002
o
-A-
4. A compound of claim 1 wherein R2 is hydrogen, Q is Ϋ , and X is N and A as
S=0.
5. A compound of claim 1R2 is hydrogen, Q is
Figure imgf000086_0003
andXisN.
6. A pharmaceutical composition which comprises: (i) a therapeutically effective amount of a compound according to claim 1 and (ii) a pharmaceutically acceptable carrier.
7. The pharmaceutical composition of claim 6, wherein the carrier is a sterile solution, suspension, emulsion, a capsule or tablet.
8. The pharmaceutical composition of claim 6, wherein the carrier comprises a biodegradable polymer or a solid implant.
9. The pharmaceutical composition of claim 6 for inhibiting PARP activity, treating or preventing diseases or disorders, altering gene expression, or radiosensitizing.
10. A method of treating a disease or disorder selected from the group consisting of tissue damage resulting from cell damage or death due to necrosis or apoptosis, neuronal mediated tissue damage or diseases, neural tissue damage resulting from ischemia and reperfusion injury, neurological disorders and neurodegenerative diseases, vascular stroke, cardiovascular disorders, age-related muscular degeneration, AIDS and other immune senescence diseases, inflammation, arthritis, gout, atherosclerosis, cachexia, cancer, degenerative diseases of skeletal muscle involving replicative senescence, hyperglycemia, diabetes, head trauma, immune senescence, inflammatory bowel disorders, muscular dystrophy, osteoarthritis, osteoporosis, chronic pain, acute pain, neuropathic pain, nervous insult, peripheral nerve injury, renal failure, retinal ischemia, septic shock, and skin aging, diseases or disorders relating to lifespan or proliferative capacity of cells, and diseases or disease conditions induced or exacerbated by cellular senescence comprising administering a compound of claim 1 to an animal in need of said treatment.
11. The method of claim 10 wherein the neurodegenerative disease is selected from the group consisting of Alzheimer's Disease, Parkinson's Disease, Huntington's Disease and amyotropic lateral sclerosis.
12. The method of claim 10, wherein the cancer is selected from the group consisting of ACTH- producing tumors, acute lymphocytic leukemia, acute nonlyniphocytic leukemia, cancer of the adrenal cortex, bladder cancer, brain cancer, breast cancer, cervix cancer, chronic lymphocytic leukemia, chronic myelocytic leukemia, colorectal cancer, cutaneous T-cell lymphoma, endometrial 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 peritoneal effusion, malignant pleural effusion, melanoma, mesothelioma, multiple myeloma, neuroblastoma, non-Hodgkin's lymphoma, osteosarcoma, ovary cancer, ovary (germ cell) cancer, prostate cancer, pancreatic cancer, penile cancer, retinoblastoma, skin cancer, soft-tissue sarcoma, squamous cell carcinomas, stomach cancer, testicular cancer, thyroid cancer, frophoblastic neoplasms, cancer of the uterus, vaginal cancer, cancer of the vulva and Wilm's tumor.
13. The method of claim 10 wherein said disease is gout.
14. The method of claim 10, wherein the cardiovascular disorder is selected from the group consisting of cardiovascular tissue damage, coronary artery disease, myocardial infarction, angina pectoris and cardiogenic shock.
PCT/US2001/015571 2000-05-19 2001-05-15 Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses WO2001090077A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001264595A AU2001264595A1 (en) 2000-05-19 2001-05-15 Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20525900P 2000-05-19 2000-05-19
US60/205,259 2000-05-19

Publications (1)

Publication Number Publication Date
WO2001090077A1 true WO2001090077A1 (en) 2001-11-29

Family

ID=22761477

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/015571 WO2001090077A1 (en) 2000-05-19 2001-05-15 Sulfonamide and carbamide derivatives of 6(5h)phenanthridinones and their uses

Country Status (3)

Country Link
US (1) US6723733B2 (en)
AU (1) AU2001264595A1 (en)
WO (1) WO2001090077A1 (en)

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002045702A2 (en) * 2000-12-05 2002-06-13 Chemokine Therapeutics Corporation Tricyclic therapeutics for chemokine mediated diseases
WO2003080581A1 (en) * 2002-03-26 2003-10-02 Fujisawa Pharmaceutical Co., Ltd. Phenanthridinones as parp inhibitors
US6706767B2 (en) 2000-12-05 2004-03-16 Chemokine Therapeutics Corporation Therapeutics for chemokine mediated diseases
US6723733B2 (en) 2000-05-19 2004-04-20 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
EP1603567A2 (en) * 2003-02-28 2005-12-14 Inotek Pharmaceuticals Corporation Tetracyclic benzamide derivatives and methods of use thereof
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
WO2006010127A2 (en) 2004-07-09 2006-01-26 Medisyn Technologies, Inc. Therapeutic compound and treatments
US7151102B2 (en) 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
WO2007008994A2 (en) * 2005-07-11 2007-01-18 Wyeth Glutamate aggrecanase inhibitors
US7196085B2 (en) 2002-04-30 2007-03-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7268143B2 (en) 2001-08-31 2007-09-11 Inotek Pharmaceuticals Corporation Isoquinoline derivatives and methods of use thereof
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
EP2061765A2 (en) * 2006-09-01 2009-05-27 Cylene Pharmaceuticals, Inc. Serine-threonine protein kinase and parp modulators
US7541485B2 (en) 2005-10-13 2009-06-02 Wyeth Methods for preparing glutamic acid derivatives
US7692006B2 (en) 2006-10-17 2010-04-06 Kudos Pharmaceuticals Limited Phthalazinone derivatives
WO2010081778A1 (en) * 2009-01-17 2010-07-22 Universität Zürich Blockers of parp for the prevention and treatment of helicobacter pylori induced gastric cancer
EP2305221A1 (en) 2003-12-01 2011-04-06 Kudos Pharmaceuticals Limited Dna damage repair inhibitors for treatment of cancer
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
US7981890B2 (en) 2007-09-14 2011-07-19 Astrazeneca Ab Phthalazinone derivatives
US20110230491A1 (en) * 2003-12-05 2011-09-22 Dominique Jean-Pierre Mabire 6-substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
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
US8895576B2 (en) 2006-12-28 2014-11-25 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US8912187B2 (en) 2003-03-12 2014-12-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
CN104955806A (en) * 2013-02-06 2015-09-30 默克专利股份公司 Substituted carboxylic acid derivatives as aggrecanase inhibitors for the treatment of osteoarthritis
US9238672B2 (en) 2007-12-27 2016-01-19 Infinity Pharmaceuticals, Inc. Methods for stereoselective reduction
US9376447B2 (en) 2010-09-14 2016-06-28 Infinity Pharmaceuticals, Inc. Transfer hydrogenation of cyclopamine analogs
CN106316953A (en) * 2015-06-15 2017-01-11 浙江工业大学 Synthetic method of 6-cyanophenanthridine compounds
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
US9879293B2 (en) 2009-08-05 2018-01-30 Infinity Pharmaceuticals, Inc. Enzymatic transamination of cyclopamine analogs
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
RU2665036C2 (en) * 2012-11-14 2018-08-27 Калитор Сайенсез, ЛЛС Heteroaromatic compounds as phosphatidylinositol 3-kinase modulators and methods of 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
WO2018197461A1 (en) 2017-04-28 2018-11-01 Akribes Biomedical Gmbh A parp inhibitor in combination with a glucocorticoid and/or ascorbic acid and/or a protein growth factor for the treatment of impaired wound healing
US10369147B2 (en) 2015-06-04 2019-08-06 PellePharm, Inc. Topical formulations for delivery of hedgehog inhibitor compounds and use thereof
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
WO2021048235A1 (en) 2019-09-10 2021-03-18 The Francis Crick Institute Limited Treatment of hr deficient cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2022223022A1 (en) * 2021-04-23 2022-10-27 四川海思科制药有限公司 Fused-ring heterocycle derivative and medical application thereof
WO2022228387A1 (en) * 2021-04-26 2022-11-03 Fochon Biosciences, Ltd. Compounds as parp inhibitors

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6476048B1 (en) 1999-12-07 2002-11-05 Inotek Pharamaceuticals Corporation Substituted phenanthridinones and methods of use thereof
US20060287311A1 (en) * 2005-02-25 2006-12-21 Inotek Pharmaceuticals Corporation Tetracyclic Sulfonamide Compounds and methods of use thereof
JP2008531561A (en) * 2005-02-25 2008-08-14 イノテック ファーマシューティカルズ コーポレイション Isoquinoline compounds and methods of use thereof
CN101242822B (en) * 2005-07-18 2011-08-24 彼帕科学公司 Medicine of ovarian cancer
JP2009506060A (en) * 2005-08-24 2009-02-12 イノテック ファーマシューティカルズ コーポレイション Indenoisoquinolinone analogs and methods for their use
US20080262062A1 (en) * 2006-11-20 2008-10-23 Bipar Sciences, Inc. Method of treating diseases with parp inhibitors
US20100279327A1 (en) * 2006-06-12 2010-11-04 Bipar Sciences, Inc. Method of treating diseases with parp inhibitors
WO2008147418A1 (en) * 2006-06-12 2008-12-04 Bipar Sciences, Inc. Method of treating diseases with parp inhibitors
US8143447B2 (en) * 2006-09-05 2012-03-27 Bipar Sciences, Inc. Treatment of cancer
US7994222B2 (en) * 2006-09-05 2011-08-09 Bipar Sciences, Inc. Monitoring of the inhibition of fatty acid synthesis by iodo-nitrobenzamide compounds
JP5419699B2 (en) * 2006-10-31 2014-02-19 エボニック コーポレイション Spheroidized polymer particles
CN101674832A (en) * 2007-02-28 2010-03-17 伊诺泰克制药公司 Indenoisoquinolinone analogs and methods of use thereof
WO2009047752A1 (en) * 2007-10-09 2009-04-16 Ramot At Tel-Aviv University Ltd. Breast cancer therapy
US9486449B2 (en) 2007-10-09 2016-11-08 Malka COHEN-ARMON Cancer therapy
KR20100102609A (en) * 2007-11-12 2010-09-24 바이파 사이언스 인코포레이티드 Treatment of breast cancer with a parp inhibitor alone or in combination with anti-tumor agents
SG185954A1 (en) * 2007-11-12 2012-12-28 Bipar Sciences Inc Treatment of ovarian cancer with 4-iodo-3-nitrobenzamide in combination with anti-tumor agents
KR20100102637A (en) * 2007-12-07 2010-09-24 바이파 사이언스 인코포레이티드 Treatment of cancer with combinations of topoisomerase inhibitors and parp inhibitors
CA2713156A1 (en) * 2008-02-04 2009-08-13 Bipar Sciences, Inc. Methods of diagnosing and treating parp-mediated diseases
US20130116284A1 (en) 2010-05-10 2013-05-09 Radikal Therapeutics Inc. Lipoic acid and nitroxide derivatives and uses thereof
CN105131974B (en) * 2015-07-28 2017-04-12 江苏和成新材料有限公司 Phenanthridone-structure heterocyclic polymerizable liquid crystal compound and preparation method thereof
WO2018022851A1 (en) 2016-07-28 2018-02-01 Mitobridge, Inc. Methods of treating acute kidney injury
SG11201903842YA (en) 2016-11-02 2019-05-30 Immunogen Inc Combination treatment with antibody-drug conjugates and parp inhibitors
WO2019031470A1 (en) * 2017-08-07 2019-02-14 国立大学法人広島大学 NOVEL AMIDE COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
CN110256346A (en) * 2019-07-03 2019-09-20 浙江省医学科学院 Phenanthridines ketone compounds and preparation method and application with BRD4 albumen inhibiting effect

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998027975A1 (en) * 1996-12-20 1998-07-02 Geron Corporation Poly(adp-ribose) polymerase inhibitors to treat diseases associated with cellular senescence
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
WO2001042219A2 (en) * 1999-12-07 2001-06-14 Inotek Corporation Novel substituted phenanthridinones and methods of use thereof

Family Cites Families (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1895105A (en) 1933-01-24 Cubt bath
US2467692A (en) 1949-04-19 Naphthyridones and processes fob
US1880441A (en) 1932-10-04 Etjbole m
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
US3318682A (en) 1961-04-06 1967-05-09 Hooker Chemical Corp Herbicidal method utilizing phenyl 2, 3, 6-trichlorophenylacetate
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
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
US3950343A (en) 1973-11-06 1976-04-13 Ayerst, Mckenna And Harrison Ltd. Pyrroloisoquinoline 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
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
ES532317A0 (en) 1983-05-16 1985-11-16 Merck & Co Inc A PHARMACEUTICAL COMPOSITION
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
DE3332663A1 (en) 1983-09-10 1985-04-04 L. & C. Steinmüller GmbH, 5270 Gummersbach Process for optimising the reduction of NOx in flue gases from fossil fuel-fired furnaces
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
US5041653A (en) 1985-05-03 1991-08-20 Sri International Substituted benzamide radiosensitizers
US5215738A (en) 1985-05-03 1993-06-01 Sri International Benzamide and nicotinamide radiosensitizers
US4668506A (en) 1985-08-16 1987-05-26 Bausch & Lomb Incorporated Sustained-release formulation containing and amino acid polymer
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
EP0270306B1 (en) 1986-12-01 1991-03-27 Sumitomo Chemical Company, Limited Anthrapyridone compounds, their production process 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
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
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
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
ES2108066T3 (en) 1991-10-31 1997-12-16 Asta Medica Ag NEW PHTHALAZINES THAT IN POSITION 1 CONTAIN AN ETER GROUPING, AND PROCEDURE 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
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
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
US5391554A (en) 1992-09-09 1995-02-21 Warner-Lambert Company Dihydro- and tetrahydronaphthyridines
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
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
DE69528103T2 (en) 1994-04-05 2003-05-15 Universiteit Van Pretoria Gaut Use of riminophenazine for the treatment of MDR resistance
US5703089A (en) 1994-04-28 1997-12-30 Knoll Aktiengesellschaft Dihydrodibenzisoquinolinediones
US5589483A (en) 1994-12-21 1996-12-31 Geron Corporation Isoquinoline poly (ADP-ribose) polymerase inhibitors to treat skin diseases associated with cellular senescence
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
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
US6121278A (en) 1997-09-03 2000-09-19 Guilford Pharmaceuticals, Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
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
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
US6197785B1 (en) 1997-09-03 2001-03-06 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6201020B1 (en) 1998-12-31 2001-03-13 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
US6277990B1 (en) * 1999-12-07 2001-08-21 Inotek Corporation Substituted phenanthridinones and methods of use thereof
US6723733B2 (en) 2000-05-19 2004-04-20 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
US6348475B1 (en) 2000-06-01 2002-02-19 Guilford Pharmaceuticals Inc. Methods, compounds and compositions for treating gout

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998027975A1 (en) * 1996-12-20 1998-07-02 Geron Corporation Poly(adp-ribose) polymerase inhibitors to treat diseases associated with cellular senescence
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
WO2001042219A2 (en) * 1999-12-07 2001-06-14 Inotek Corporation Novel substituted phenanthridinones and methods of use thereof

Cited By (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6723733B2 (en) 2000-05-19 2004-04-20 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
US7151102B2 (en) 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
WO2002045702A3 (en) * 2000-12-05 2003-01-03 Chemokine Therapeutics Corp Tricyclic therapeutics for chemokine mediated diseases
US6706767B2 (en) 2000-12-05 2004-03-16 Chemokine Therapeutics Corporation Therapeutics for chemokine mediated diseases
WO2002045702A2 (en) * 2000-12-05 2002-06-13 Chemokine Therapeutics Corporation Tricyclic therapeutics for chemokine mediated diseases
US7393955B2 (en) 2001-08-31 2008-07-01 Inotek Pharmaceuticals Corporation Isoquinoline derivatives and methods of use thereof
US7268143B2 (en) 2001-08-31 2007-09-11 Inotek Pharmaceuticals Corporation Isoquinoline derivatives and methods of use thereof
WO2003080581A1 (en) * 2002-03-26 2003-10-02 Fujisawa Pharmaceutical Co., Ltd. Phenanthridinones as parp inhibitors
US7196085B2 (en) 2002-04-30 2007-03-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
CN100398519C (en) * 2003-02-28 2008-07-02 伊诺泰克制药公司 Tetracyclic benzamide derivatives and methods of use thereof
EP1603567A4 (en) * 2003-02-28 2006-10-18 Inotek Pharmaceuticals Corp Tetracyclic benzamide derivatives and methods of use thereof
US7217709B2 (en) 2003-02-28 2007-05-15 Inotek Pharmaceuticals Corporation Tetracyclic benzamide derivatives and methods of use thereof
EP1603567A2 (en) * 2003-02-28 2005-12-14 Inotek Pharmaceuticals Corporation Tetracyclic benzamide derivatives and methods of use thereof
US9169235B2 (en) 2003-03-12 2015-10-27 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US10449192B2 (en) 2003-03-12 2019-10-22 Kudo Pharmaceuticals Limited Phthalazinone derivatives
US8912187B2 (en) 2003-03-12 2014-12-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US11160803B2 (en) 2003-03-12 2021-11-02 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7662818B2 (en) 2003-03-12 2010-02-16 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US9566276B2 (en) 2003-03-12 2017-02-14 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
EP2305221A1 (en) 2003-12-01 2011-04-06 Kudos Pharmaceuticals Limited Dna damage repair inhibitors for treatment of cancer
US20110230491A1 (en) * 2003-12-05 2011-09-22 Dominique Jean-Pierre Mabire 6-substituted 2-quinolinones and 2-quinoxalinones as poly(adp-ribose) polymerase inhibitors
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
EP1778228A4 (en) * 2004-07-09 2010-08-11 Medisyn Technologies Inc Therapeutic compound and treatments
WO2006010127A2 (en) 2004-07-09 2006-01-26 Medisyn Technologies, Inc. Therapeutic compound and treatments
US8088785B2 (en) 2004-07-09 2012-01-03 Medisyn Technologies, Inc. Therapeutic compound and treatments
EP1778228A2 (en) * 2004-07-09 2007-05-02 Medisyn Technologies, Inc. Therapeutic compound and treatments
JP2008505931A (en) * 2004-07-09 2008-02-28 メディスィン テクノロジーズ, インコーポレイテッド Therapeutic compounds and treatments
WO2007008994A2 (en) * 2005-07-11 2007-01-18 Wyeth Glutamate aggrecanase inhibitors
US7998965B2 (en) 2005-07-11 2011-08-16 Wyeth Llc Glutamate aggrecanase inhibitors
WO2007008994A3 (en) * 2005-07-11 2007-07-12 Wyeth Corp Glutamate aggrecanase inhibitors
AU2006268183B2 (en) * 2005-07-11 2012-08-09 Wyeth Glutamate aggrecanase inhibitors
US7553873B2 (en) 2005-07-11 2009-06-30 Wyeth Glutamate aggrecanase inhibitors
JP2009500456A (en) * 2005-07-11 2009-01-08 ワイス Glutamate aggrecanase inhibitor
US7541485B2 (en) 2005-10-13 2009-06-02 Wyeth Methods for preparing glutamic acid derivatives
US7902193B2 (en) 2005-10-19 2011-03-08 Maybridge Limited Phthalazinone derivatives
US7470688B2 (en) 2005-10-19 2008-12-30 Maybridge Limited Phthalazinone derivatives
EP2061765A2 (en) * 2006-09-01 2009-05-27 Cylene Pharmaceuticals, Inc. Serine-threonine protein kinase and parp modulators
RU2681209C2 (en) * 2006-09-01 2019-03-05 Сенхва Байосайенсиз,Инк. Serine-threonine protein kinase and parp modulators
EP2061765A4 (en) * 2006-09-01 2012-03-21 Cylene Pharmaceuticals Inc Serine-threonine protein kinase and parp modulators
JP2010502651A (en) * 2006-09-01 2010-01-28 サイリーン ファーマシューティカルズ インコーポレーティッド Serine-threonine protein kinase and PARP regulator
US9062043B2 (en) 2006-09-01 2015-06-23 Senhwa Biosciences, Inc. Fused tricyclic compounds as serine-threonine protein kinase and PARP modulators
US7692006B2 (en) 2006-10-17 2010-04-06 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US8895576B2 (en) 2006-12-28 2014-11-25 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US10821102B2 (en) 2006-12-28 2020-11-03 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US10045970B2 (en) 2006-12-28 2018-08-14 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US10314827B2 (en) 2006-12-28 2019-06-11 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US10406139B2 (en) 2006-12-28 2019-09-10 Infinity Pharmaceuticals, Inc. Cyclopamine analogs
US9951083B2 (en) 2006-12-28 2018-04-24 Infinity Pharmaceuticals, Inc. Cyclopamine analogs
US9145422B2 (en) 2006-12-28 2015-09-29 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US9669011B2 (en) 2006-12-28 2017-06-06 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
US11007181B2 (en) 2006-12-28 2021-05-18 Infinity Pharmaceuticals, Inc. Cyclopamine analogs
US9492435B2 (en) 2006-12-28 2016-11-15 Infinity Pharmaceuticals, Inc. Cyclopamine analogs
US11602527B2 (en) 2006-12-28 2023-03-14 Infinity Pharmaceuticals, Inc. Methods of use of cyclopamine analogs
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
US9238672B2 (en) 2007-12-27 2016-01-19 Infinity Pharmaceuticals, Inc. Methods for stereoselective reduction
US8129380B2 (en) 2008-01-23 2012-03-06 Astrazeneca Ab Phthalazinone derivatives
US9050329B1 (en) 2008-09-19 2015-06-09 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
US9302984B2 (en) 2008-09-19 2016-04-05 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
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
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
WO2010081778A1 (en) * 2009-01-17 2010-07-22 Universität Zürich Blockers of parp for the prevention and treatment of helicobacter pylori induced gastric cancer
US9879293B2 (en) 2009-08-05 2018-01-30 Infinity Pharmaceuticals, Inc. Enzymatic transamination of cyclopamine analogs
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
US9394313B2 (en) 2010-09-14 2016-07-19 Infinity Pharmaceuticals, Inc. Transfer hydrogenation of cyclopamine analogs
US9879025B2 (en) 2010-09-14 2018-01-30 Infinity Pharmaceuticals, Inc. Transfer hydrogenation of cyclopamine analogs
US9376447B2 (en) 2010-09-14 2016-06-28 Infinity Pharmaceuticals, Inc. Transfer hydrogenation of cyclopamine analogs
RU2665036C9 (en) * 2012-11-14 2018-11-12 Калитор Сайенсез, ЛЛС Heteroaromatic compounds as phosphatidylinositol 3-kinase modulators and methods of use
RU2665036C2 (en) * 2012-11-14 2018-08-27 Калитор Сайенсез, ЛЛС Heteroaromatic compounds as phosphatidylinositol 3-kinase modulators and methods of use
CN104955806B (en) * 2013-02-06 2018-09-14 默克专利股份公司 The substituted carboxylic acid derivative as polyprotein xylanase inhibitor for treating osteoarthritis
CN104955806A (en) * 2013-02-06 2015-09-30 默克专利股份公司 Substituted carboxylic acid derivatives as aggrecanase inhibitors for the treatment of osteoarthritis
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
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
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
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
US11413283B2 (en) 2015-06-04 2022-08-16 PellePharm, Inc. Topical formulations for delivery of hedgehog inhibitor compounds and use thereof
US10369147B2 (en) 2015-06-04 2019-08-06 PellePharm, Inc. Topical formulations for delivery of hedgehog inhibitor compounds and use thereof
US10695344B2 (en) 2015-06-04 2020-06-30 PellePharm, Inc. Topical formulations for delivery of hedgehog inhibitor compounds and use thereof
CN106316953A (en) * 2015-06-15 2017-01-11 浙江工业大学 Synthetic method of 6-cyanophenanthridine compounds
CN106316953B (en) * 2015-06-15 2019-02-01 浙江工业大学 A kind of synthetic method of 6- cyano phenanthridines class compound
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
WO2018197461A1 (en) 2017-04-28 2018-11-01 Akribes Biomedical Gmbh A parp inhibitor in combination with a glucocorticoid and/or ascorbic acid and/or a protein growth factor for the treatment of impaired wound healing
WO2019175132A1 (en) 2018-03-13 2019-09-19 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
WO2021048235A1 (en) 2019-09-10 2021-03-18 The Francis Crick Institute Limited Treatment of hr deficient cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2022223022A1 (en) * 2021-04-23 2022-10-27 四川海思科制药有限公司 Fused-ring heterocycle derivative and medical application thereof
WO2022228387A1 (en) * 2021-04-26 2022-11-03 Fochon Biosciences, Ltd. Compounds as parp inhibitors

Also Published As

Publication number Publication date
US20020006927A1 (en) 2002-01-17
US6723733B2 (en) 2004-04-20
AU2001264595A1 (en) 2001-12-03

Similar Documents

Publication Publication Date Title
US6723733B2 (en) Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
CA2430363C (en) Compounds and their uses
US6514983B1 (en) Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6291425B1 (en) Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US7157452B2 (en) Substituted 4,9-dihydrocyclopenta{imn}phenanthridine-5-ones derivatives thereof and their uses
US6387902B1 (en) Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP
WO1999059975A1 (en) Fused tricyclic compounds which inhibit parp activity
AU9299198A (en) Alkoxy-substituted compounds, methods, and compositions for inhibiting parp activity
WO1999011622A1 (en) Amino-substituted compounds, methods, and compositions for inhibiting parp activity
WO1999011644A1 (en) Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
US7247641B2 (en) Compounds, derivatives, compositions, preparation and uses
US6545011B2 (en) Substituted 4,9-dihydrocyclopenta[imn]phenanthridine-5-ones, derivatives thereof and their uses
AU2002236521B2 (en) Compounds and their uses
AU2005202592B2 (en) Compounds, Methods and Pharmaceutical Compositions for Treating Cellular Damage, such as Neural or Cardiovascular Tissue Damage
AU2002236521A1 (en) Compounds and their uses
MXPA99011815A (en) Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
MXPA99011814A (en) Alkoxy-substituted compounds, methods, and compositions for inhi biting 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 CO 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 TR 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)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP