USRE36397E - Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity - Google Patents

Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity Download PDF

Info

Publication number
USRE36397E
USRE36397E US09/219,843 US21984398A USRE36397E US RE36397 E USRE36397 E US RE36397E US 21984398 A US21984398 A US 21984398A US RE36397 E USRE36397 E US RE36397E
Authority
US
United States
Prior art keywords
inhibitor
iaddend
iadd
benzamide
adp
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US09/219,843
Inventor
Jie Zhang
Valina L. Dawson
Ted M. Dawson
Solomon H. Snyder
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Johns Hopkins University
US Government
Original Assignee
Johns Hopkins University
US Government
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 Johns Hopkins University, US Government filed Critical Johns Hopkins University
Priority to US09/219,843 priority Critical patent/USRE36397E/en
Application granted granted Critical
Publication of USRE36397E publication Critical patent/USRE36397E/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide

Definitions

  • the invention relates to the use of inhibitors of poly(ADP-ribose) synthetase to prevent NMDA neurotoxicity.
  • Nitric oxide is a messenger molecule that regulates macrophage killing of tumor cells and bacteria (C. F. Nathan and J. B. Hibbs, Jr., Curr. Opin. Immunol. 3, 65 (1991)), blood vessel relaxation (S. Moncada, R. M. J. Palmer, E. A. Higgs, Pharmacol. Rev. 43, 109 (1991); L. J. Ignarro, Ann. Rev. Pharmacol. Toxicol. 30, 535 (1990)) and also is a neurotransmitter (D. S. Bredt and S. H. Snyder, Neuron 8, 3 (1992)).
  • Mechanisms proposed for NO neurotoxicity as well as tumoricidal and bactericidal actions include mono-ADP-ribosylation and S-nitrosylation of glyceraldehyde-3-phosphate dehydrogenase (J. Zhang and S. H. Snyder, Proc. Natl. Acad. Sci. U.S.A. 89, 9382 (1992); A. Y. Kots et al., FEBS Lett. 300, 9 (1992); S. Dimmeler, F. Lottspeich, B. Brune, J. Biol. Chem. 267, 16771 (1992); L. Molina y Vedia et al., J. Biol. Chem.
  • Another object of the invention is to provide a method of treating a neurodegenerative disease, such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
  • a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
  • a method of preventing or treating diseases caused by NMDA neurotoxicity comprises administering to a mammal in need thereof a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
  • a method of preventing or treating vascular stroke damage in a mammal, in particular a human patient comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
  • a method of treating a mammal for a neurodegenerative disease caused by NMDA neurotoxicity in particular preventing or treating a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease in a human patient, is provided, which method comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
  • a pharmaceutically acceptable formulation which comprises an inhibitor of poly(ADP-ribose) synthetase in a pharmaceutically acceptable vehicle.
  • the present invention provides the art with methods and formulations for treating neurological damage due to neurodegenerative diseases and vascular stroke.
  • FIG. 1 Activation of PARS by NO-damaged DNA.
  • B Autoradiography of PARS auto-poly(ADP-ribosyl)ation on 7.5% SDS-PAGE. Abbreviations AmNAP, 4amino-1,8-naphthalimide, DHIQ, 1,5-dihydroxyisoquinoline.
  • FIG. 2 Inhibition of NMDA and NO mediated Neurotoxicity by PARS Inhibitors. 100 ⁇ M benzamide reduces NMDA, SNP (500 ⁇ M) and SNAP (300 ⁇ M) mediated neurotoxicity.
  • Data are the means ⁇ SEM (n>8). Each data point represents 4,000-12,000 neurons counted (21). *p ⁇ 0.01, Student's t-test.
  • FIG. 3 A Model of NO-mediated Cytotoxicity.
  • NO damaged DNA activates PARS which depletes NAD by poly-ADP-ribosylating neuclear proteins.
  • Poly(ADP-ribose) is rapidly degraded by poly(ADP-ribose) glycohydrolase.
  • the futile cycle continues during the prolonged PARS activation. It takes an equivalent of four ATP's to resynthesize NAD from nicotinamide (NAm) via nicotinamide mononucleotide (NMN), a reaction that requires phosphoribosyl pyrophosphate (PRPP) and ATP.
  • NAm nicotinamide
  • NPN nicotinamide mononucleotide
  • PRPP phosphoribosyl pyrophosphate
  • NO activates PARS in association with damage to DNA.
  • PARS activation can kill cells by consuming ⁇ -nicotinamide adenine dinucleotide, the source of ADP-ribose, and ATP.
  • PARS inhibitors prevent NMDA neurotoxicity with relative potencies paralleling their enzyme inhibitory actions.
  • PARS inhibition The ability of PARS inhibition to provide substantial protection against NMDA neurotoxicity, implicates DNA damage in neuronal killing. PARS activation rapidly leads to energy depletion. For each ADP-ribose unit transferred by PARS, one molecule of NAD is consumed and an equivalent of four molecules of ATP are required to regenerate NAD from nicotinamide, PARS, whose density is up to one enzyme per 10-20 nucleosomes, can be activated 10-20 fold by DNA damage; PARS transfers 50-100 ADP-ribose moieties to each acceptor site of target proteins and its over-activation by substantial DNA damage can markedly deplete cells of NAD and ATP (J. C. Gaal, K. R. Smith, C. K.
  • Poly(ADP-ribose) synthetase inhibitors may be used to prevent, treat, arrest, or ameliorate the progression of any disease condition caused by NMDA neurotoxicity.
  • diseases include vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states.
  • vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states.
  • a patient is administered a poly(ADP-ribose) synthetase inhibitor to block damage to the brain.
  • Patients with symptoms of Alzheimer's or Huntington's disease are treated with poly(ADP-ribose) synthatase inhibitors to halt the progression of the disease.
  • the symptoms of these disease states are known by one skilled in this art.
  • Inhibitors of poly(ADP-ribose) synthetase are compounds which compete for the substrate binding site of poly(ADP-ribose) synthetase or other sites on the enzyme, and include both reversible and irreversible inhibitors.
  • the present invention contemplates the use of any physiologically acceptable inhibitor which inhibits poly(ADP-ribose) synthetase activity.
  • the effectiveness of a compound, and its relative potency as a poly(ADP-ribose) synthetase inhibitor can be tested and routinely determined by measuring inhibition of poly(ADP-ribose) synthetase activity.
  • Poly(ADP-ribose) synthetase activity can be assayed according to the method of Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986).
  • various benzamide derivatives have been found to have the ability to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. These include: 3-aminobenzamide and 4-aminobenzamide. Benzoic acid was found not to have any activity.
  • novobiocin an inhibitor of mono(ADP-ribose) synthetase was found not to have any activity in preventing neurotoxicity.
  • the dosage and length of treatment depends on the disease state being treated.
  • the duration of treatment may be a day, a week or longer and may, as in the case of a chronic progressive illness, such as Alzheimer's, last over the entire lifetime of the patient.
  • the inhibitors are administered in a therapeutically effective amount, a typical human dosage of benzamide ranging from about 0.01 mg/kg of body weight to about 10 mg/kg, in single or divided doses.
  • the dosage will vary depending on the poly(ADP-ribose) synthetase inhibitor being used and its relative potency. Dosage and length of treatment are readily determinable by the skilled practitioner based on the condition and stage of disease.
  • poly(ADP-ribose) synthetase inhibitors may be administered by any route by which drugs are conventionally administered.
  • routes of administration include intraperitoneal, intravenous, intramuscular, subcutaneous, intrathecal, intraventricular, as well as oral.
  • Typical pharmaceutically acceptable preparations for administration include sterile aqueous or nonaqueous solutions, suspensions and emulsions.
  • nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water alcoholic/aqueous and buffered media.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like.
  • Oral preparations, such as in capsule, tablets, and other form include additives such as cellulose, silica gel and stearic acid.
  • a poly(ADP-ribose) synthetase inhibitor desirably should be able to penetrate the blood brain barrier when peripherally administrated.
  • Poly(ADP-ribose) synthetase inhibitors which are unable to penetrate the blood brain barrier can be effectively administered by, for example, an intraventricular route of delivery.
  • SIN-1 3-morpholinosyndnonimine
  • SNP sodium nitroprusside
  • Each 50 ⁇ l assay mixture contained 10 ⁇ g of the nuclear protein fraction and [adenylate- 32 P] NAD (0.1 mM, 10 Ci/mmole), in the presence or absence of 4-amino-1,8-naphthalimide (20 ⁇ M), 1,5-dihydroxyisoquinoline (20 ⁇ M), benzamide (100 ⁇ M, SNP (1 mM), SIN-1 (1 mM), DNA (0.1 pg, pTrcA, In Vitrogen) and DNA that had been treated with NO gas, SNP or SIN-1. Incubation of DNA with NO gas was according to T.
  • benzamide derivatives exist with differing potencies as PARS inhibitors.
  • benzamide is the most active, 3-aminobenzamide is about half as potent, 4-aminobenzamide is 50-100 fold weaker than benzamide and benzoic acid is inactive (The IC 50 for benzamide in inhibiting PARS in vitro is 22 ⁇ M [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)].
  • IC 50 for benzamide in inhibiting PARS in vitro is 22 ⁇ M [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)].
  • benzamide would be competing with millimolar endogenous levels of NAD [R. McNerney et al., Biochim. Biophys. Acta 1009, 185 (1989)].
  • Benzamide provided the greatest degree of protection, with 3-aminobenzamide exerting somewhat less protection against NMDA neurotoxicity, while 4-aminobenzamide and benzoic acid were inactive (Table 1 ).
  • a structurally unrelated PARS inhibitor, 1,5-dihydroxyisoquinoline (10 ⁇ M) was also neuroprotective against NMDA neurotoxicity (Table 1A).
  • the absolute as well as the relative potencies of the benzamide derivatives and 1,5-dihydroxyisoquinoline in blocking NMDA toxicity correspond with their potencies as PARS inhibitors.
  • NMDA exposure initiates "delayed neurotoxicity" in which poorly characterized irreversible processes ultimately lead to calcium overload and cell death (R. D. Randall and S. A. Thayer, J. Neurosci. 12, 1882 (1992); D. W. Choi, Neuron 1, 623 (1988); D. W. Choi, J. Neurosci. 10, 2493 (1990)).
  • benzamide was added prior to the NMDA and during NMDA exposure (FIG. 2, Table 1A). However, in some experiments we have added benzamide up to 1 hr after NMDA exposure and observed similar protection (Table 1B).
  • Table 1A PARS inhibitors were applied 30 min before and during NMDA application.
  • Table 1B PARS inhibitors were applied after NMDA administration at the indicated times for 20 to 24 hr. Data are the means ⁇ SEM (n ⁇ 8). Each data point represents 4,000-12,000 neurons counted. *p ⁇ 0.001 Student's t-test.
  • Neurotoxicity was determined by exposing the neurons to the various test solutions as previously described (Dawson, supra). NADA, SNP or SNAP were applied to the cells for 5 min, then the cells were washed and replaced with MEM, 21 mM glucose overnight in the incubator. Twenty to 24 hr after exposure to test solutions, the neurons were exposed to 0.4% trypan blue in CSS to stain the residue of non-viable cells and to assess toxicity. Viable and non-viable cells were counted. At least two separate experiments utilizing four separate wells were performed for each data point shown. Significant overall values were obtained by using a one-way, between groups analysis of variance. Specific comparisons on all possible pair combinations were made with the Student's t-test for independent means.
  • Nitrite formation in a human kidney 293 cell line stably transfected with the cDNA for brain NOS was measured in response to A23187 (10 ⁇ M) as described (D. S. Bredt, C. D. Ferris and S. H. Snyder, J. Biol. Chem. 267, 10976 (1992).
  • A23187 (10 ⁇ M) elicited the formation of 17.1 ⁇ 2.1 ⁇ M nitrite in 2 hr.

Abstract

Inhibitors of poly(ADP-ribose) synthetase can be used to prevent neurotoxicity mediated through N-methyl-D-aspartate (NMDA) receptors. Poly(ADP-ribose) synthetase inhibitors can be used therapeutically in the treatment of vascular stroke and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and Huntington's disease.

Description

The U.S. Government has a paid-up license in this invention and the right in limited circumstances to require the patent owner to license others on reasonable terms as provided for by the terms of grants awarded by the National Institutes of Health and the U.S. Public Health Service, including USPHS grants DA-00266, Contract DA-271-90-7408, Research Scientist Award DA-00074, USPHS CIDA NS-01578 and an Intramural Research Training Award from the N.I.H.
FIELD OF THE INVENTION
The invention relates to the use of inhibitors of poly(ADP-ribose) synthetase to prevent NMDA neurotoxicity.
BACKGROUND OF THE INVENTION
Nitric oxide (NO) is a messenger molecule that regulates macrophage killing of tumor cells and bacteria (C. F. Nathan and J. B. Hibbs, Jr., Curr. Opin. Immunol. 3, 65 (1991)), blood vessel relaxation (S. Moncada, R. M. J. Palmer, E. A. Higgs, Pharmacol. Rev. 43, 109 (1991); L. J. Ignarro, Ann. Rev. Pharmacol. Toxicol. 30, 535 (1990)) and also is a neurotransmitter (D. S. Bredt and S. H. Snyder, Neuron 8, 3 (1992)). When formed in high quantities in response to actions of the excitatory neurotransmitter glutamate acting at N-methyl- D-aspartate (NMDA) receptors. NO mediates neuronal killing (V. L. Dawson et al., Proc. Natl. Acad. Sci. U.S.A. 88, 6368 (1991); V. L. Dawson et al., J. Neurosci. 13, 2651 (1993)). Toxicity due to NMDA accounts for neural damage in vascular stroke, as NO synthase (NOS) inhibitors prevent stroke damage (J. P. Nowicki, D. Duval, H. Poignet, B. Scatton, Eur. J. Pharmac. 204, 339 (1991)). Mechanisms proposed for NO neurotoxicity as well as tumoricidal and bactericidal actions include mono-ADP-ribosylation and S-nitrosylation of glyceraldehyde-3-phosphate dehydrogenase (J. Zhang and S. H. Snyder, Proc. Natl. Acad. Sci. U.S.A. 89, 9382 (1992); A. Y. Kots et al., FEBS Lett. 300, 9 (1992); S. Dimmeler, F. Lottspeich, B. Brune, J. Biol. Chem. 267, 16771 (1992); L. Molina y Vedia et al., J. Biol. Chem. 267, 24929 (1992)), inhibition of mitochondrial enzymes such as cis-aconitase (J. -C. Drapier and J. B. Hibbs, Jr., J. Clin. Invest. 78, 790 (1986)), inhibition of the mitochondrial electron transport chain (Nathan and Hibbs, supra), inhibition of ribonucleotide reductase (Lepoivre, B. Chenais, A. Yapo, G. Lemaire, J. Biol. Chem. 265, 14143 (1990); N. S. Kwon, D. J. Stuehr, C. F. Nathan, J. Exp. Med. 174, 761 (1991)), and DNA damage (D. A. Wink et al., Science 254, 1001 (1991); T. Nguyen et al., Proc. Natl. Acad. Sci. U.S.A. 89, 3030 (1992)). DNA damage activates poly(ADP-ribose) synthetase (PARS, EC 2.4.2.30) (G. de Murcia, J. Menissier-de Murcia, V. Schreiber, BioEssays 13,455 (1991); J. E. Cleaver and W. F. Morgan, Mutation Res. 257, 1 (1991); J. C. Gaal, K. R. Smith, C. K. Pearson, Trends in Biological Sciences 12, 129 (1987); N. A. Berger, Rad. Res. 101, 4 (1985)). PARS is a nuclear enzyme which, upon activation by DNA strand breaks, adds up to 100 ADP-ribose units to nuclear proteins such as histones and PARS itself.
There is a continuing need in the art for effective methods of preventing, treating or ameliorating diseases caused by NMDA neurotoxicity, such as vascular stroke, Alzheimer's disease, Huntington's disease, and Parkinson's disease.
SUMMARY OF THE INVENTION
It is an object of the invention to provide a method of treating diseases caused by NMDA neurotoxicity.
It is another object of the invention to provide a method of treating vascular stroke.
Another object of the invention is to provide a method of treating a neurodegenerative disease, such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
It is still another object of the invention to provide pharmaceutical formulations for treating neurodegenerative diseases and vascular stroke damage.
These and other objects of the invention are provided by one or more of the embodiments described below.
In one embodiment of the invention, a method of preventing or treating diseases caused by NMDA neurotoxicity is provided, which method comprises administering to a mammal in need thereof a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In another embodiment of the invention, a method of preventing or treating vascular stroke damage in a mammal, in particular a human patient, is provided, which comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In still another embodiment of the invention, a method of treating a mammal for a neurodegenerative disease caused by NMDA neurotoxicity, in particular preventing or treating a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease in a human patient, is provided, which method comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In yet another embodiment of the invention, a pharmaceutically acceptable formulation is provided which comprises an inhibitor of poly(ADP-ribose) synthetase in a pharmaceutically acceptable vehicle.
Thus the present invention provides the art with methods and formulations for treating neurological damage due to neurodegenerative diseases and vascular stroke.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1. Activation of PARS by NO-damaged DNA. (A) PARS activity, (means ±S.E.M., n=3), after different treatments. (B) Autoradiography of PARS auto-poly(ADP-ribosyl)ation on 7.5% SDS-PAGE. Abbreviations AmNAP, 4amino-1,8-naphthalimide, DHIQ, 1,5-dihydroxyisoquinoline.
FIG. 2. Inhibition of NMDA and NO mediated Neurotoxicity by PARS Inhibitors. 100 μM benzamide reduces NMDA, SNP (500 μM) and SNAP (300 μM) mediated neurotoxicity.
Data are the means ±SEM (n>8). Each data point represents 4,000-12,000 neurons counted (21). *p≦0.01, Student's t-test.
FIG. 3. A Model of NO-mediated Cytotoxicity. NO damaged DNA activates PARS which depletes NAD by poly-ADP-ribosylating neuclear proteins. Poly(ADP-ribose) is rapidly degraded by poly(ADP-ribose) glycohydrolase. The futile cycle continues during the prolonged PARS activation. It takes an equivalent of four ATP's to resynthesize NAD from nicotinamide (NAm) via nicotinamide mononucleotide (NMN), a reaction that requires phosphoribosyl pyrophosphate (PRPP) and ATP.
DETAILED DESCRIPTION OF THE INVENTION
It is a discovery of the present invention that NO activates PARS in association with damage to DNA. PARS activation can kill cells by consuming β-nicotinamide adenine dinucleotide, the source of ADP-ribose, and ATP. Moreover, PARS inhibitors prevent NMDA neurotoxicity with relative potencies paralleling their enzyme inhibitory actions.
Our observation that NO activates PARS fits with other studies indicating that NO damages DNA (D. A. Wink et al., Science 254, 1001 (1991); T. Nguyen et al., Proc. Natl. Acad. Sci. U.S.A. 89, 3030 (1992)). The protection against NMDA neurotoxicity provided by PARS inhibitors appears to reflect PARS inhibition, as the relative potencies of these drugs in blocking neurotoxicity parallel their activities as PARS inhibitors.
The ability of PARS inhibition to provide substantial protection against NMDA neurotoxicity, implicates DNA damage in neuronal killing. PARS activation rapidly leads to energy depletion. For each ADP-ribose unit transferred by PARS, one molecule of NAD is consumed and an equivalent of four molecules of ATP are required to regenerate NAD from nicotinamide, PARS, whose density is up to one enzyme per 10-20 nucleosomes, can be activated 10-20 fold by DNA damage; PARS transfers 50-100 ADP-ribose moieties to each acceptor site of target proteins and its over-activation by substantial DNA damage can markedly deplete cells of NAD and ATP (J. C. Gaal, K. R. Smith, C. K. Pearson, Trends in Biological Sciences 12, 129 (1987); N. A. Berger, Rad. Res. 101, 4 (1985)). Neurotoxicity is similarly associated with a change in the NADH/NAD redox state and energy depletion. (See, for example, O. H. Lowry et al., J. Biol. Chem. 239, 18 (1964); D. Uematsu et al., Brain Research 482, 129 (1989); but also see K. M. Raley and P. Lipton, Neurosci. Lett. 110, 118 (1990)). The protection against NMDA neurotoxicity afforded by PARS inhibition supports a mechanism of cell death in which DNA damage overwhelms repair mechanisms leading to energy depletion by activation of PARS (FIG. 3).
Two major effects of glutamate neurotoxicity suggested to participate in neuronal killing are the formation of free radicals and energy depletion (B. Meldrum and J. Garthwaite, Trends Pharmacol. Sci. 11, 379 (1990)). Our findings indicate that both participate, with NO, a free radical, triggering DNA damage, which in turn activates PARS, ultimately depleting energy sources from the cell. This mechanism of cell death might also account for the tumoricidal effects of NO formed by macrophages, which fits with observations that 3-aminobenzamide protects pancreatic islet cells from macrophage cytotoxicity (B. Kallman et al., Life Sci. 51, 671 (1992)).
Poly(ADP-ribose) synthetase inhibitors may be used to prevent, treat, arrest, or ameliorate the progression of any disease condition caused by NMDA neurotoxicity. Such conditions include vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states. For example, following the symptoms of a stroke, a patient is administered a poly(ADP-ribose) synthetase inhibitor to block damage to the brain. Patients with symptoms of Alzheimer's or Huntington's disease are treated with poly(ADP-ribose) synthatase inhibitors to halt the progression of the disease. The symptoms of these disease states are known by one skilled in this art.
Inhibitors of poly(ADP-ribose) synthetase are compounds which compete for the substrate binding site of poly(ADP-ribose) synthetase or other sites on the enzyme, and include both reversible and irreversible inhibitors. The present invention contemplates the use of any physiologically acceptable inhibitor which inhibits poly(ADP-ribose) synthetase activity. The effectiveness of a compound, and its relative potency as a poly(ADP-ribose) synthetase inhibitor, can be tested and routinely determined by measuring inhibition of poly(ADP-ribose) synthetase activity. Poly(ADP-ribose) synthetase activity can be assayed according to the method of Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986).
Both benzamide and 1,5-dihydroxy-isoquinoline, two poly(ADP-ribose) synthetase inhibitors, have been found to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. In addition, various benzamide derivatives have been found to have the ability to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. These include: 3-aminobenzamide and 4-aminobenzamide. Benzoic acid was found not to have any activity. In addition, novobiocin, an inhibitor of mono(ADP-ribose) synthetase was found not to have any activity in preventing neurotoxicity.
The dosage and length of treatment depends on the disease state being treated. The duration of treatment may be a day, a week or longer and may, as in the case of a chronic progressive illness, such as Alzheimer's, last over the entire lifetime of the patient. The inhibitors are administered in a therapeutically effective amount, a typical human dosage of benzamide ranging from about 0.01 mg/kg of body weight to about 10 mg/kg, in single or divided doses. The dosage will vary depending on the poly(ADP-ribose) synthetase inhibitor being used and its relative potency. Dosage and length of treatment are readily determinable by the skilled practitioner based on the condition and stage of disease.
In therapeutic use, poly(ADP-ribose) synthetase inhibitors may be administered by any route by which drugs are conventionally administered. Such routes of administration include intraperitoneal, intravenous, intramuscular, subcutaneous, intrathecal, intraventricular, as well as oral.
Typical pharmaceutically acceptable preparations for administration include sterile aqueous or nonaqueous solutions, suspensions and emulsions. Examples of nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil and injectable organic esters such as ethyl oleate. Aqueous carriers include water alcoholic/aqueous and buffered media. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like. Oral preparations, such as in capsule, tablets, and other form include additives such as cellulose, silica gel and stearic acid.
To be effective therapeutically, a poly(ADP-ribose) synthetase inhibitor desirably should be able to penetrate the blood brain barrier when peripherally administrated. Poly(ADP-ribose) synthetase inhibitors which are unable to penetrate the blood brain barrier can be effectively administered by, for example, an intraventricular route of delivery.
The following examples are provided to exemplify various aspects of the invention and are not intended to limit the scope of the invention.
EXAMPLE 1
This example demonstrates the activation of PARS by NO-treated DNA, as well as the inhibition of such activation by PARS inhibitors.
In rat brain nuclear extracts, PARS activity was almost tripled dose-dependently by adding DNA that had been pre-incubated with NO (FIG. 1A). Addition of covalently closed circular DNA by itself had no effect on PARS activity. Both 4amino-1,8-naphthalimide and 1,5-dihydroxyisoquinoline, two potent PARS inhibitors, reduced the activity to <5% of basal levels (FIG. 1A). The major protein ADP-ribosylated in the nuclear extracts was PARS itself (FIG. 1B). Similarly, DNA that had been treated with 3-morpholinosyndnonimine (SIN-1) and sodium nitroprusside (SNP), two NO donors, could stimulate poly (ADP-ribose) synthesis, which was inhibited by benzamide, another PARS inhibitor. SNP and SIN-1 had no effect on PARS by themselves.
Whole brains from 1-day old rats were homogenized in 20% (w/v) buffer A [50 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM DTT, 50 mM NaCl, 0.25M sucrose, 0.2 mM phenylmethylsulfonyl fluoride, 1 μg/ml each of chymostatin, leupeptin, pepstatin and trypsin inhibitor]. The homogenate was centrifuged at 1000 g for 15 min. The pellet was washed with buffer A and centrifuged again. The washed pellet, termed nuclear fraction, was resuspended in buffer A. PARS activity was assayed according to I. U. Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986). Each 50 μl assay mixture contained 10 μg of the nuclear protein fraction and [adenylate-32 P] NAD (0.1 mM, 10 Ci/mmole), in the presence or absence of 4-amino-1,8-naphthalimide (20 μM), 1,5-dihydroxyisoquinoline (20 μM), benzamide (100 μM, SNP (1 mM), SIN-1 (1 mM), DNA (0.1 pg, pTrcA, In Vitrogen) and DNA that had been treated with NO gas, SNP or SIN-1. Incubation of DNA with NO gas was according to T. Nguyen et al., Proc. Natl. Acad. Sci. USA 89, 3030 (1992). SNP and SIN-1 treated DNA doubled PARS activity, while both compounds had no effects on PARS by themselves. When [14 C]NAD's were used in place of [32 P]NAD in PARS assay, radioactive polymers were only formed from [adenine-14 C] NAD but not from [carbonyl-14 C]NAD.
McDonald and Moss recently demonstrated that NO-enhanced modification of GAPDH by NAD involves the transfer of the entire NAD to a thiol group rather than ADP-ribosylation (J. P. Nowicki, D. Duval, H. Poignet, B. Scatton, Eur. J. Pharmac. 204, 339 (1991)). To ensure that the polymer formed from NAD in our study was poly(ADP-ribose), we used both [14 C-adenine] NAD and [14 C-nicotinamide] NAD and found radioactivity could only be incorporated into the polymers from the former compound.
EXAMPLE 2
This example demonstrates that PARS inhibitors inhibit the neurotoxicity elicited by NMDA.
To determine directly if PARS activation participates in NMDA neurotoxicity, we monitored neurotoxicity elicited by NMDA in cerebral cortical cultures in which NO synthase inhibitors provide marked protection (V. L. Dawson et al., Proc. Natl. Acad. Sci. U.S.A. 88, 6368 (1991); V. L. Dawson et al., J. Neurosci. 13, 2651 (1993)) (FIG. 2). Increasing concentrations of NMDA progressively augment neuronal killing. Benzamide (100 μM) provided 40-50% protection at all NMDA concentrations examined. At 50 μM benzamide, no significant protection was detected, while 500 μM benzamide provided about 30% greater protection than at 100 μM (Table 1A). NO did not interact with benzamide.
To test whether NO interacts directly with benzamide, we incubated benzamide (1 mM) with SNP (1 mM) for five days at 25° C., and analyzed it by HPLC on a C18 column. SNP-treated benzamide was 100% recovered at the same elution time point with the same UV spectrum as that of unreated benzamide. When SNP-treated benzamide and benzamide were mixed and analyzed only one peak was observed.
A variety of benzamide derivatives exist with differing potencies as PARS inhibitors. In the family of benzamide and its derivatives, benzamide is the most active, 3-aminobenzamide is about half as potent, 4-aminobenzamide is 50-100 fold weaker than benzamide and benzoic acid is inactive (The IC50 for benzamide in inhibiting PARS in vitro is 22 μM [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)]. In intact cells a higher value would be anticipated as benzamide would be competing with millimolar endogenous levels of NAD [R. McNerney et al., Biochim. Biophys. Acta 1009, 185 (1989)].
Benzamide provided the greatest degree of protection, with 3-aminobenzamide exerting somewhat less protection against NMDA neurotoxicity, while 4-aminobenzamide and benzoic acid were inactive (Table 1 ). A structurally unrelated PARS inhibitor, 1,5-dihydroxyisoquinoline (10 μM), was also neuroprotective against NMDA neurotoxicity (Table 1A). The absolute as well as the relative potencies of the benzamide derivatives and 1,5-dihydroxyisoquinoline in blocking NMDA toxicity correspond with their potencies as PARS inhibitors. It is unlikely that the PARS inhibitors could prevent NMDA neurotoxicity through inhibition of mono(ADP-ribose) synthetase as their IC50 's for mono(ADP-ribose) synthetase inhibition are at least 100 times greater than their IC50 's as PARS inhibitors. Furthermore, novobiocin (1 mM) a relatively selective mono(ADP-ribose) synthetase inhibitor was ineffective against NMDA neurotoxicity (Table 1A).
Brief (5 rain) NMDA exposure initiates "delayed neurotoxicity" in which poorly characterized irreversible processes ultimately lead to calcium overload and cell death (R. D. Randall and S. A. Thayer, J. Neurosci. 12, 1882 (1992); D. W. Choi, Neuron 1, 623 (1988); D. W. Choi, J. Neurosci. 10, 2493 (1990)). In most experiments benzamide was added prior to the NMDA and during NMDA exposure (FIG. 2, Table 1A). However, in some experiments we have added benzamide up to 1 hr after NMDA exposure and observed similar protection (Table 1B).
              TABLE 1                                                     
______________________________________                                    
                       % Cell Death                                       
Treatment              (+ S.E.M.)                                         
______________________________________                                    
A. PARS Inhibitors Protect Against NMDA Neurotoxicity                     
500 μM NMDA         57.1 ± 4.7                                      
+50 μM Benzamide                    60.1 ± 12.1                     
+100 μM Benzamide                  39.5 ± 5.0*                      
+500 μM Benazmide                  21.9 ± 4.1*                      
+100 μM 3-aminobenzamide                                               
                               45.9 ± 5.1*                             
+100 μM 4-aminobenzamide                                               
                               55.8 ± 7.6                              
+1 mM Benzoic Acid                      58.9 ± 5.7                     
+10 μM 1.5 dihydroxyisoquinoline                                       
                       39.0 ± 4.5*                                     
+1 mM Novobiocin                          62.4 ± 8.9                   
B. PARS Inhibitors Rescue Neurons From NMDA Neurotoxicity                 
500 μM NMDA         74.7 ± 7.8                                      
+100 μM Benzamide                                                      
5 min                                                   35.6 ± 8.7*    
30 min                                                 40.4 ± 5.6*     
1 hour                                                 43.4               
______________________________________                                    
                       ± 3.9*                                          
Table 1A, PARS inhibitors were applied 30 min before and during NMDA application. In Table 1B, PARS inhibitors were applied after NMDA administration at the indicated times for 20 to 24 hr. Data are the means ±SEM (n≧8). Each data point represents 4,000-12,000 neurons counted. *p≦0.001 Student's t-test.
Primary neuronal cultures from cortex were prepared from fetal Sprague-Dawley rats, gestation day 13-14. Mature neurons (grater than 21 days in culture) were used in all experiments.
Neurotoxicity was determined by exposing the neurons to the various test solutions as previously described (Dawson, supra). NADA, SNP or SNAP were applied to the cells for 5 min, then the cells were washed and replaced with MEM, 21 mM glucose overnight in the incubator. Twenty to 24 hr after exposure to test solutions, the neurons were exposed to 0.4% trypan blue in CSS to stain the residue of non-viable cells and to assess toxicity. Viable and non-viable cells were counted. At least two separate experiments utilizing four separate wells were performed for each data point shown. Significant overall values were obtained by using a one-way, between groups analysis of variance. Specific comparisons on all possible pair combinations were made with the Student's t-test for independent means.
EXAMPLE 3
This example demonstrates that NO elicits neurotoxicity by activation of PARS.
To ascertain whether NO itself elicits neurotoxicity by activation of PARS, we administered the NO releasers SNP and S-nitroso-N-acetylpenicillamine (SNAP). Benzamide (100 μM) reduced SNP and SNAP neurotoxicity by 45% to 50% (FIG. 2). Benzamide does not inhibit NOS. In human kidney 293 cells stably transfected with the cDNA of brain. NOS, stimulated levels of nitrite, an indirect measure of NO production, were not affected by 100 μM benzamide, but were abolished by nitroarginic. This experiment also confirms that benzamide did not interact with NO.
Nitrite formation in a human kidney 293 cell line stably transfected with the cDNA for brain NOS was measured in response to A23187 (10 μM) as described (D. S. Bredt, C. D. Ferris and S. H. Snyder, J. Biol. Chem. 267, 10976 (1992). A23187 (10 μM) elicited the formation of 17.1±2.1 μM nitrite in 2 hr. Benzamide (100 μM) did not prevent the formation of nitrite (15.9±3.8 μM nitrite) and 100 μM nitroarginine diminished nitrite formation to 3.9±1.6 μM nitrite (n=3).

Claims (17)

We claim:
1. A method of treating a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity in a mammal comprising:
administering to a mammal which demonstrates symptoms of a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity a therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase.
2. The method of claim 1 wherein said inhibitor is administered intravenously, intraperitoneally, intramuscularly, intraventricularly, or orally.
3. The method of claim 2 wherein said inhibitor is benzamide.
4. The method of claim 1 wherein said inhibitor is benzamide. .[.5. The method of claim 1 wherein said inhibitor is a benzamide derivative selected from the group consisting of 3-aminobenzamide and
4-aminobenzamide..].6. The method of claim .[.5.]. .Iadd.1 .Iaddend.wherein said inhibitor is 3-aminobenzamide. .[.7. The method of
claim 5 wherein said inhibitor is 4-aminobenzamide..].8. The method of
claim 1 wherein said inhibitor is 1,5-dihydroxy-isoquinoline. 9. The
method of claim 1 wherein said disease is vascular stroke. 10. The method
of claim 1 wherein said disease is a neurodegenerative disease. 11. A pharmaceutical formulation comprising .Iadd.a therapeutically effective amount of .Iaddend.an inhibitor of poly(ADP-ribose) synthetase .Iadd.containing a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide .Iaddend.in a
pharmaceutically acceptable vehicle. 12. The pharmaceutical formulation of claim 11 wherein said inhibitor is benzamide. .[.13. The pharmaceutical formulation of claim 11 wherein said inhibitor is a benzamide derivative selected from the group consisting of 3-aminobenzamide and 4-amino
benzamide..].14. The pharmaceutical formulation of claim .[.13.]. .Iadd.11 .Iaddend.wherein said inhibitor is 3-aminobenzamide. .[.15. The pharmaceutical formulation of claim 13 wherein said inhibitor is
4-aminobenzamide..].16. The pharmaceutical formulation of claim 11 wherein
said inhibitor is 1,5-dihydroxy-isoquinoline. 17. The pharmaceutical formulation of claim 11 wherein said inhibitor has an IC50 for inhibiting poly(ADP-ribose) synthetase in vitro of at least .Iadd.as
potent as .Iaddend.22 μM. 18. The pharmaceutical formulation of claim 11 wherein said inhibitor has an IC50 for inhibiting poly(ADP-ribose)
synthetase in vitro of at least .Iadd.as potent as .Iaddend.33 μM. 19. The pharmaceutical formulation of claim 11 wherein said formulation is a capsule or tablet containing a single or divided dose of said inhibitor, wherein said dose is sufficient to treat .[.NMDA.]. .Iadd.glutamate
.Iaddend.neurotoxicity. 20. The pharmaceutical formulation of claim 11 which is a sterile solution, suspension or emulsion, in a single or divided dose, for administration to a mammal which demonstrates symptoms of a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity. .Iadd.21. The pharmaceutical formulation of claim 11 wherein said formulation is a capsule or tablet containing a single or divided dose of said inhibitor, wherein said dose is sufficient to treat glutamate neurotoxicity mediated through NMDA receptors. .Iaddend..Iadd.22. The pharmaceutical formulation of claim 11 which is a sterile solution, suspension or emulsion, in a single or divided dose, for administration to a mammal which demonstrates symptoms of a disease condition caused by glutamate neurotoxicity mediated through NMDA receptors. .Iaddend..Iadd.23. A method of claim 1 wherein said therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase is in the form of a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide. .Iaddend..Iadd.24. A method of treating a disease condition caused by glutamate neurotoxicity in a mammal comprising:
administering to a mammal which demonstrates symptoms of a disease condition caused by glutamate neurotoxicity mediated through NMDA receptors a therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase. .Iaddend..Iadd.25. The method of claim 24 wherein said inhibitor is administered intravenously, intraperitoneally, intramuscularly, intraventricularly, or orally. .Iaddend..Iadd.26. The method of claim 24 wherein said inhibitor is benzamide. .Iaddend..Iadd.27. The method of claim 25 wherein said inhibitor is benzamide.
.Iaddend..Iadd.8. The method of claim 24 wherein said inhibitor is 3-aminobenzamide. .Iaddend..Iadd.29. The method of claim 24 wherein said inhibitor is 1,5-dihydroxy-isoquinoline. .Iaddend..Iadd.30. The method of claim 24 wherein said disease is vascular stroke. .Iaddend..Iadd.31. The method of claim 24 wherein said disease is a neurodegenerative disease. .Iaddend..Iadd.32. A method of claim 24 wherein said therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase is in the form of a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide. .Iaddend.
US09/219,843 1994-02-04 1998-12-23 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity Expired - Lifetime USRE36397E (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/219,843 USRE36397E (en) 1994-02-04 1998-12-23 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/191,508 US5587384A (en) 1994-02-04 1994-02-04 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
US09/219,843 USRE36397E (en) 1994-02-04 1998-12-23 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/191,508 Reissue US5587384A (en) 1994-02-04 1994-02-04 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity

Publications (1)

Publication Number Publication Date
USRE36397E true USRE36397E (en) 1999-11-16

Family

ID=22705771

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/191,508 Ceased US5587384A (en) 1994-02-04 1994-02-04 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
US09/219,843 Expired - Lifetime USRE36397E (en) 1994-02-04 1998-12-23 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/191,508 Ceased US5587384A (en) 1994-02-04 1994-02-04 Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity

Country Status (7)

Country Link
US (2) US5587384A (en)
AU (1) AU1695495A (en)
FI (1) FI963040A0 (en)
IL (1) IL112496A0 (en)
NO (1) NO963253D0 (en)
WO (1) WO1995020952A1 (en)
ZA (1) ZA95861B (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6306889B1 (en) 1997-09-03 2001-10-23 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6348475B1 (en) 2000-06-01 2002-02-19 Guilford Pharmaceuticals Inc. Methods, compounds and compositions for treating gout
US6380211B1 (en) 1997-09-03 2002-04-30 Guilford Pharmaceutical Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6387902B1 (en) 1998-12-31 2002-05-14 Guilford Pharmaceuticals, Inc. Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP
US6395749B1 (en) 1998-05-15 2002-05-28 Guilford Pharmaceuticals Inc. Carboxamide compounds, methods, and compositions for inhibiting PARP activity
US6426415B1 (en) 1997-09-03 2002-07-30 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods and compositions for inhibiting parp activity
US6440455B1 (en) 1997-09-02 2002-08-27 Children's Medical Center Corporation Methods for modulating the axonal outgrowth of central nervous system neurons
US6444676B1 (en) 1999-12-20 2002-09-03 Iok-Hou Pang Use of PARP inhibitors in the treatment of glaucoma
US20020177614A1 (en) * 2001-03-23 2002-11-28 Merril Carl R. Methods for treating nuerodegenerative diseases including alzheimer's
US6514983B1 (en) 1997-09-03 2003-02-04 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6545011B2 (en) 2000-07-13 2003-04-08 Guilford Pharmaceuticals Inc. Substituted 4,9-dihydrocyclopenta[imn]phenanthridine-5-ones, derivatives thereof and their uses
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6716828B1 (en) 1999-09-01 2004-04-06 Guilford Pharmaceuticals, Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6723733B2 (en) 2000-05-19 2004-04-20 Guilford Pharmaceuticals, Inc. Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses
US20040138502A1 (en) * 2001-03-08 2004-07-15 Traynelis Stephen F Ph-dependent nmda receptor antagonists
US20050096342A1 (en) * 1995-10-23 2005-05-05 Medivation, Inc. Neurodegenerative disorders
WO2006078711A2 (en) 2005-01-19 2006-07-27 Mgi Gp, Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting parp
US20060276497A1 (en) * 2000-05-09 2006-12-07 Cephalon, Inc. Novel multicyclic compounds and the use thereof
US20070117834A1 (en) * 2005-10-04 2007-05-24 David Hung Methods and compositions for treating Huntington's disease
US20070179174A1 (en) * 2003-12-08 2007-08-02 Bachurin Sergei O Methods and compositions for slowing aging
US20070248702A1 (en) * 2004-06-22 2007-10-25 Javier Fernandez-Ruiz Use of CB2 receptors agonists for the treatment of Huntington's disease
US20080161255A1 (en) * 2003-05-29 2008-07-03 Michael Brownlee Use of Parp Inhibitors for Prevention and Treatment of Diabetic and Insulin Resistance Complications
US20080234310A1 (en) * 2003-12-08 2008-09-25 Bachurin Sergei O Methods and Compositions for Slowing Aging
US20110071088A1 (en) * 2003-12-16 2011-03-24 Childrens Medical Center Corporation Method for treating neurological disorders
WO2012027224A1 (en) 2010-08-24 2012-03-01 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
US8420680B2 (en) 2007-06-29 2013-04-16 Emory University NMDA receptor antagonists for neuroprotection
WO2013055911A1 (en) 2011-10-14 2013-04-18 Dana-Farber Cancer Institute, Inc. Znf365/zfp365 biomarker predictive of anti-cancer response
WO2013130347A1 (en) 2012-02-23 2013-09-06 The Children's Hospital Corporation Methods for predicting anti-cancer response
WO2015109264A1 (en) 2014-01-17 2015-07-23 Cedars-Sinai Medical Center Receptor targeting constructs and uses thereof
WO2015164743A2 (en) 2014-04-24 2015-10-29 Dana-Farber Cancer Institute, Inc. Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response
US9279156B2 (en) 2011-06-17 2016-03-08 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
WO2016044707A1 (en) 2014-09-18 2016-03-24 Cedars-Sinai Medical Center Compositions and methods for treating fibrosis
WO2016057367A1 (en) 2014-10-06 2016-04-14 Dana-Farber Cancer Institute, Inc. Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response
WO2016073647A2 (en) 2014-11-04 2016-05-12 University Of Southern California COMPOSITIONS AND METHODS FOR TREATING HIF-1alpha OVER-EXPRESSING CANCERS
WO2016094273A1 (en) 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
US9388472B2 (en) 2011-12-21 2016-07-12 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
WO2016138574A1 (en) 2015-03-02 2016-09-09 Sinai Health System Homologous recombination factors
WO2016144673A1 (en) 2015-03-06 2016-09-15 Dana-Farber Cancer Institute, Inc. Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers
WO2017066561A2 (en) 2015-10-16 2017-04-20 President And Fellows Of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
WO2018057618A1 (en) 2016-09-20 2018-03-29 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of aml using usp10 biomarkers and modulators
US10308986B2 (en) 2013-03-14 2019-06-04 Children's Medical Center Corporation Cancer diagnosis, treatment selection and treatment
US10400287B2 (en) 2014-08-15 2019-09-03 Myriad Genetics, Inc. Methods and materials for assessing homologous recombination deficiency
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
US11091808B2 (en) 2012-06-07 2021-08-17 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US11149316B2 (en) 2013-12-09 2021-10-19 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
WO2022086852A2 (en) 2020-10-19 2022-04-28 Dana-Farber Cancer Institute, Inc. Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
WO2022104104A2 (en) 2020-11-13 2022-05-19 Dana-Farber Cancer Institute, Inc. Personalized fusion cell vaccines
WO2022159793A2 (en) 2021-01-25 2022-07-28 Dana-Farber Cancer Institute, Inc. Methods and compositions for identifying neuroendocrine prostate cancer
WO2022159575A1 (en) 2021-01-20 2022-07-28 Bioentre Llc Ctla4-binding proteins and methods of treating cancer
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
WO2023043888A1 (en) 2021-09-15 2023-03-23 Celloram Inc. Method of treating cancer
WO2023240287A1 (en) 2022-06-10 2023-12-14 Bioentre Llc Combinations of ctla4 binding proteins and methods of treating cancer

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9404485D0 (en) * 1994-03-09 1994-04-20 Cancer Res Campaign Tech Benzamide analogues
US5589483A (en) * 1994-12-21 1996-12-31 Geron Corporation Isoquinoline poly (ADP-ribose) polymerase inhibitors to treat skin diseases associated with cellular senescence
US6358975B1 (en) 1997-08-15 2002-03-19 Johns Hopkins University Method of using selective parp inhibitors to prevent or treat neurotoxicity
AU8784698A (en) * 1997-08-15 1999-03-08 Johns Hopkins University, The Method of using selective parp inhibitors to prevent or treat neurotoxicity
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
US6333148B1 (en) 1998-05-01 2001-12-25 University Of Kentucky Research Genes encoding several poly (ADP-ribose) glycohydrolase (PARG) enzymes, the proteins and fragments thereof, and antibodies immunoreactive therewith
JP2002515488A (en) * 1998-05-15 2002-05-28 ギルフォード ファーマシューティカルズ インコーポレイテッド Carboxamide compounds, compositions, and methods for inhibiting PARP activity
US20030078212A1 (en) * 1998-10-30 2003-04-24 Jia-He Li Pharmaceutical compositions containing poly(adp-ribose) glycohydrolase inhibitors and methods of using the same
US6201020B1 (en) 1998-12-31 2001-03-13 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
EP1140936B1 (en) 1999-01-11 2004-03-17 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(adp-ribose) polymerases
US6187822B1 (en) * 1999-06-11 2001-02-13 University Of Medicine & Dentistry Of Nj Wound treatment through inhibition of adenosine diphosphate ribosyl transferase
ECSP003637A (en) 1999-08-31 2002-03-25 Agouron Pharma TRICYCLE POLY INHIBITORS (ADP-RIBOSA) POLYMERASES
US6476048B1 (en) 1999-12-07 2002-11-05 Inotek Pharamaceuticals Corporation Substituted phenanthridinones and methods of use thereof
US6277990B1 (en) 1999-12-07 2001-08-21 Inotek Corporation Substituted phenanthridinones and methods of use thereof
US6531464B1 (en) * 1999-12-07 2003-03-11 Inotek Pharmaceutical Corporation Methods for the treatment of neurodegenerative disorders using substituted phenanthridinone derivatives
US7122679B2 (en) 2000-05-09 2006-10-17 Cephalon, Inc. Multicyclic compounds and the use thereof
US7151102B2 (en) * 2000-10-30 2006-12-19 Kudos Pharmaceuticals Limited Phthalazinone derivatives
US6635786B2 (en) 2001-01-16 2003-10-21 Guilford Pharmaceuticals, Inc. Symmetrically disubstituted aromatic compounds and pharmaceutical compositions for inhibiting poly (ADP-ribose) glycohydrolase, and methods for their use
US7072771B2 (en) * 2001-06-07 2006-07-04 University Of Kentucky Research Foundation Selective PARP-1 targeting for designing chemo/radio sensitizing agents
MXPA04001353A (en) * 2001-08-15 2004-10-27 Icos Corp 2h-phthalazin-1-ones and methods for use thereof.
ES2357057T3 (en) * 2002-04-30 2011-04-15 Kudos Pharmaceuticals Limited DERIVATIVES OF FTALAZINONA.
GB0305681D0 (en) 2003-03-12 2003-04-16 Kudos Pharm Ltd Phthalazinone derivatives
US7449464B2 (en) * 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
MXPA05010563A (en) * 2003-03-31 2005-11-23 Pfizer Salts of tricyclic inhibitors of poly(adp-ribose) polymerases.
SG150548A1 (en) 2003-12-01 2009-03-30 Kudos Pharm Ltd Dna damage repair inhibitors for treatment of cancer
GB0419072D0 (en) 2004-08-26 2004-09-29 Kudos Pharm Ltd Phthalazinone derivatives
GB0428111D0 (en) * 2004-12-22 2005-01-26 Kudos Pharm Ltd Pthalazinone derivatives
GB0521373D0 (en) * 2005-10-20 2005-11-30 Kudos Pharm Ltd Pthalazinone derivatives
CN101484421A (en) * 2006-06-15 2009-07-15 库多斯药物有限公司 2 -oxybenzamide derivatives as parp inhibitors
WO2007144652A2 (en) * 2006-06-15 2007-12-21 Kudos Pharmaceuticals Limited Parp inhibitors
WO2007144637A1 (en) * 2006-06-15 2007-12-21 Kudos Pharmaceuticals Limited 2 -oxyheteroarylamide derivatives as parp inhibitors
UY30639A1 (en) * 2006-10-17 2008-05-31 Kudos Pharm Ltd SUBSTITUTED DERIVATIVES OF 2H-FTALAZIN-1-ONA, ITS CRYSTAL FORMS, PREPARATION PROCESS AND APPLICATIONS
US20080280910A1 (en) * 2007-03-22 2008-11-13 Keith Allan Menear Phthalazinone derivatives
TW200900396A (en) * 2007-04-10 2009-01-01 Kudos Pharm Ltd Phthalazinone derivatives
US20090023727A1 (en) * 2007-07-05 2009-01-22 Muhammad Hashim Javaid Phthalazinone derivatives
KR20100059950A (en) 2007-09-14 2010-06-04 아스트라제네카 아베 Phthalazinone derivatives
MX2010004028A (en) * 2007-10-17 2010-04-30 Kudos Pharm Ltd 4- [3- (4-cyclopropanecarbonyl-piperazine-i-carbonyl) -4 -fluoro-benzyl] -2h-phthalaz in-1-one.
UY31603A1 (en) 2008-01-23 2009-08-31 DERIVATIVES OF FTALAZINONA
CN102238945B (en) * 2008-10-07 2014-10-29 阿斯利康(英国)有限公司 Pharmaceutical formulation 514
TW201114756A (en) * 2009-07-15 2011-05-01 Astrazeneca Ab Phthalazinone compound
WO2011058367A2 (en) 2009-11-13 2011-05-19 Astrazeneca Ab Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor
KR101896567B1 (en) 2015-07-23 2018-09-07 인스티튜트 큐리 Combined use of deblock molecule and PARP inhibitor for cancer treatment
GB201519573D0 (en) 2015-11-05 2015-12-23 King S College London Combination
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
EP3765613A1 (en) 2018-03-13 2021-01-20 Onxeo A dbait molecule against acquired resistance in the treatment of cancer
GB201913030D0 (en) 2019-09-10 2019-10-23 Francis Crick Institute Ltd Treatment of hr deficient cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4594415A (en) * 1982-03-29 1986-06-10 Robins Roland K Azole dinucleotide compositions and methods of use
EP0355750A1 (en) * 1988-08-19 1990-02-28 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
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
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

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4594415A (en) * 1982-03-29 1986-06-10 Robins Roland K Azole dinucleotide compositions and methods of use
EP0355750A1 (en) * 1988-08-19 1990-02-28 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
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
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

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
Banasik et al., "Specific Inhibitors of Poly(ADP-Ribose)Synthetase and Mono (ADP-Ribosyl) Transferase", J Biol Chem, 267:1569-1575 (1992).
Banasik et al., Specific Inhibitors of Poly(ADP Ribose)Synthetase and Mono (ADP Ribosyl) Transferase , J Biol Chem, 267:1569 1575 (1992). *
Choi, Dennis, "Glutamate Neurotoxicity and Diseases of the Nervous System", Neuron, 1:623-634 (1988).
Choi, Dennis, "The Role of Glutamate Neurotoxicity in Hypoxic-Ischemic Neuronal Death", Ann Rev. Neurosci., 13:171-82 (1990).
Choi, Dennis, Glutamate Neurotoxicity and Diseases of the Nervous System , Neuron, 1:623 634 (1988). *
Choi, Dennis, The Role of Glutamate Neurotoxicity in Hypoxic Ischemic Neuronal Death , Ann Rev. Neurosci., 13:171 82 (1990). *
Cosi et al., J. Neuroscience Research, 39(1), 38 46, Sep. 1994. *
Cosi et al., J. Neuroscience Research, 39(1), 38-46, Sep. 1994.
Dawson et al., "Mechanisms of Nitric Oxide-Mediated Neurotoxicity in Primary Brain Cultures", J Neuroscience, 13:2651-2661 (1993).
Dawson et al., "Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures", Proc Natl Acad Sci, 88:6568-6371 (1991).
Dawson et al., Mechanisms of Nitric Oxide Mediated Neurotoxicity in Primary Brain Cultures , J Neuroscience, 13:2651 2661 (1993). *
Dawson et al., Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures , Proc Natl Acad Sci, 88:6568 6371 (1991). *
Meldrum et al., "Excitatory amino acid neurotoxicity and neurodegenerative disease", TIPS, 11:379-387 (1990).
Meldrum et al., Excitatory amino acid neurotoxicity and neurodegenerative disease , TIPS, 11:379 387 (1990). *
Nowicki et al., "Nitric Oxide Mediates Neuronal Death After Focal Cerebral Ischemia in the Mouse", European J Pharmacology, 204, 339-340 (1991).
Nowicki et al., Nitric Oxide Mediates Neuronal Death After Focal Cerebral Ischemia in the Mouse , European J Pharmacology, 204, 339 340 (1991). *
Purnell et al., "Novel inhibitors of Poly (ADP-Ribose) Synthetase", Biochem J, 185:775-777 (1980).
Purnell et al., Novel inhibitors of Poly (ADP Ribose) Synthetase , Biochem J, 185:775 777 (1980). *
Suto et al., "Dihydroisoquinolinones: the design and synthesis of a new series of potent inhibitors of poly (ADP-ribose) polymerase", Anti-cancer Drug Design, 7:107-117 (1991).
Suto et al., Dihydroisoquinolinones: the design and synthesis of a new series of potent inhibitors of poly (ADP ribose) polymerase , Anti cancer Drug Design, 7:107 117 (1991). *
Wallis et al., "Neuroprotection Against Nitric Oxide Injury with Inhibitors of ADP-ribosylation", Neuropharmacology, 5(3):245-248 (1993).
Wallis et al., Neuroprotection Against Nitric Oxide Injury with Inhibitors of ADP ribosylation , Neuropharmacology, 5(3):245 248 (1993). *
Zhang et al., "Nitric Oxide Activation of Poly(ADP-Ribose) Synthetase in Neurotoxicity", Science, 263:687-689 (1994).
Zhang et al., Nitric Oxide Activation of Poly(ADP Ribose) Synthetase in Neurotoxicity , Science, 263:687 689 (1994). *

Cited By (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050096342A1 (en) * 1995-10-23 2005-05-05 Medivation, Inc. Neurodegenerative disorders
US20060140866A1 (en) * 1995-10-23 2006-06-29 Zefirov Nikolai S Agents for treating neurodegenerative disorders
US7071206B2 (en) 1995-10-23 2006-07-04 Medivation, Inc. Agents for treating neurodegenerative disorders
US6440455B1 (en) 1997-09-02 2002-08-27 Children's Medical Center Corporation Methods for modulating the axonal outgrowth of central nervous system neurons
US20070213294A1 (en) * 1997-09-02 2007-09-13 Children's Medical Center Corporation Methods for modulating the axonal growth of central nervous system neurons
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6426415B1 (en) 1997-09-03 2002-07-30 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods and compositions for inhibiting parp activity
US6306889B1 (en) 1997-09-03 2001-10-23 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6514983B1 (en) 1997-09-03 2003-02-04 Guilford Pharmaceuticals Inc. Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage
US6380211B1 (en) 1997-09-03 2002-04-30 Guilford Pharmaceutical Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6346536B1 (en) 1997-09-03 2002-02-12 Guilford Pharmaceuticals Inc. Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same
US6395749B1 (en) 1998-05-15 2002-05-28 Guilford Pharmaceuticals Inc. Carboxamide compounds, methods, and compositions for inhibiting PARP activity
US6387902B1 (en) 1998-12-31 2002-05-14 Guilford Pharmaceuticals, Inc. Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP
US20050074470A1 (en) * 1999-09-01 2005-04-07 Guilford Pharmaceuticals, Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6716828B1 (en) 1999-09-01 2004-04-06 Guilford Pharmaceuticals, Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US7307080B2 (en) 1999-09-01 2007-12-11 Mgi Gp, Inc. Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage
US6444676B1 (en) 1999-12-20 2002-09-03 Iok-Hou Pang Use of PARP inhibitors in the treatment of glaucoma
US20060276497A1 (en) * 2000-05-09 2006-12-07 Cephalon, Inc. Novel multicyclic compounds and the 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
US6545011B2 (en) 2000-07-13 2003-04-08 Guilford Pharmaceuticals Inc. Substituted 4,9-dihydrocyclopenta[imn]phenanthridine-5-ones, derivatives thereof and their uses
US20040138502A1 (en) * 2001-03-08 2004-07-15 Traynelis Stephen F Ph-dependent nmda receptor antagonists
US7375136B2 (en) 2001-03-08 2008-05-20 Emory University pH-dependent NMDA receptor antagonists
US20020177614A1 (en) * 2001-03-23 2002-11-28 Merril Carl R. Methods for treating nuerodegenerative diseases including alzheimer's
US20080161255A1 (en) * 2003-05-29 2008-07-03 Michael Brownlee Use of Parp Inhibitors for Prevention and Treatment of Diabetic and Insulin Resistance Complications
US20070179174A1 (en) * 2003-12-08 2007-08-02 Bachurin Sergei O Methods and compositions for slowing aging
US20080234310A1 (en) * 2003-12-08 2008-09-25 Bachurin Sergei O Methods and Compositions for Slowing Aging
US20110071088A1 (en) * 2003-12-16 2011-03-24 Childrens Medical Center Corporation Method for treating neurological disorders
US8912144B2 (en) 2003-12-16 2014-12-16 Children's Medical Center Corporation Method for treating stroke via administration of NEP1-40 and inosine
US20070248702A1 (en) * 2004-06-22 2007-10-25 Javier Fernandez-Ruiz Use of CB2 receptors agonists for the treatment of Huntington's disease
EP2319847A2 (en) 2005-01-19 2011-05-11 Eisai Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP
WO2006078711A2 (en) 2005-01-19 2006-07-27 Mgi Gp, Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting parp
US20070117834A1 (en) * 2005-10-04 2007-05-24 David Hung Methods and compositions for treating Huntington's disease
US20070117835A1 (en) * 2005-10-04 2007-05-24 David Hung Methods and compositions for treating Huntington's disease
US8420680B2 (en) 2007-06-29 2013-04-16 Emory University NMDA receptor antagonists for neuroprotection
US9079852B2 (en) 2007-06-29 2015-07-14 Emory University NMDA receptor antagonists for neuroprotection
WO2012027224A1 (en) 2010-08-24 2012-03-01 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
US9512485B2 (en) 2010-08-24 2016-12-06 Dana-Farber Cancer Institute. Inc. Methods for predicting anti-cancer response
US10577662B2 (en) 2010-08-24 2020-03-03 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
EP3109325A1 (en) 2010-08-24 2016-12-28 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
US11225685B2 (en) 2011-06-17 2022-01-18 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US10626449B2 (en) 2011-06-17 2020-04-21 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US9279156B2 (en) 2011-06-17 2016-03-08 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
US9574229B2 (en) 2011-06-17 2017-02-21 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
WO2013055911A1 (en) 2011-10-14 2013-04-18 Dana-Farber Cancer Institute, Inc. Znf365/zfp365 biomarker predictive of anti-cancer response
US9388472B2 (en) 2011-12-21 2016-07-12 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
US10612098B2 (en) 2011-12-21 2020-04-07 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
US11299782B2 (en) 2012-02-23 2022-04-12 Children's Medical Center Corporation Methods for predicting anti-cancer response
WO2013130347A1 (en) 2012-02-23 2013-09-06 The Children's Hospital Corporation Methods for predicting anti-cancer response
EP3415915A1 (en) 2012-02-23 2018-12-19 Children's Medical Center Corporation Methods for predicting anti-cancer response
US10190160B2 (en) 2012-02-23 2019-01-29 Children's Medical Center Corporation Methods for predicting anti-cancer response
US11091808B2 (en) 2012-06-07 2021-08-17 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
US10308986B2 (en) 2013-03-14 2019-06-04 Children's Medical Center Corporation Cancer diagnosis, treatment selection and treatment
US11149316B2 (en) 2013-12-09 2021-10-19 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
EP3791889A1 (en) 2014-01-17 2021-03-17 Cedars-Sinai Medical Center C-met receptor targeting constructs and uses thereof
WO2015109264A1 (en) 2014-01-17 2015-07-23 Cedars-Sinai Medical Center Receptor targeting constructs and uses thereof
WO2015164743A2 (en) 2014-04-24 2015-10-29 Dana-Farber Cancer Institute, Inc. Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response
US10400287B2 (en) 2014-08-15 2019-09-03 Myriad Genetics, Inc. Methods and materials for assessing homologous recombination deficiency
WO2016044707A1 (en) 2014-09-18 2016-03-24 Cedars-Sinai Medical Center Compositions and methods for treating fibrosis
WO2016057367A1 (en) 2014-10-06 2016-04-14 Dana-Farber Cancer Institute, Inc. Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response
WO2016073647A2 (en) 2014-11-04 2016-05-12 University Of Southern California COMPOSITIONS AND METHODS FOR TREATING HIF-1alpha OVER-EXPRESSING CANCERS
WO2016094273A1 (en) 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
WO2016138574A1 (en) 2015-03-02 2016-09-09 Sinai Health System Homologous recombination factors
WO2016144673A1 (en) 2015-03-06 2016-09-15 Dana-Farber Cancer Institute, Inc. Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers
WO2017066561A2 (en) 2015-10-16 2017-04-20 President And Fellows Of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
WO2018057618A1 (en) 2016-09-20 2018-03-29 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of aml using usp10 biomarkers and modulators
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
WO2022086852A2 (en) 2020-10-19 2022-04-28 Dana-Farber Cancer Institute, Inc. Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
EP4343004A2 (en) 2020-10-19 2024-03-27 Dana-Farber Cancer Institute, Inc. Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
WO2022104104A2 (en) 2020-11-13 2022-05-19 Dana-Farber Cancer Institute, Inc. Personalized fusion cell vaccines
WO2022159575A1 (en) 2021-01-20 2022-07-28 Bioentre Llc Ctla4-binding proteins and methods of treating cancer
WO2022159793A2 (en) 2021-01-25 2022-07-28 Dana-Farber Cancer Institute, Inc. Methods and compositions for identifying neuroendocrine prostate cancer
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
WO2023043888A1 (en) 2021-09-15 2023-03-23 Celloram Inc. Method of treating cancer
WO2023240287A1 (en) 2022-06-10 2023-12-14 Bioentre Llc Combinations of ctla4 binding proteins and methods of treating cancer

Also Published As

Publication number Publication date
NO963253D0 (en) 1996-08-02
AU1695495A (en) 1995-08-21
IL112496A0 (en) 1995-03-30
ZA95861B (en) 1996-08-02
FI963040A0 (en) 1996-08-01
WO1995020952A1 (en) 1995-08-10
US5587384A (en) 1996-12-24

Similar Documents

Publication Publication Date Title
USRE36397E (en) Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
KR960008230B1 (en) Compositions for inhibition of disorders associated with oxidative damage
Slemmer et al. Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging
Zhang et al. Nitric oxide activation of poly (ADP-ribose) synthetase in neurotoxicity
Weiss et al. AMPA receptor activation potentiates zinc neurotoxicity
RU2261093C2 (en) Derivative of pyrrolidine acetamide or its combination for treatment of central nervous system disorders
Islam et al. Pathophysiological role of peroxynitrite induced DNA damage in human diseases: a special focus on poly (ADP-ribose) polymerase (PARP)
Timmings et al. Lamotrigine as an add-on drug in the management of Lennox-Gastaut syndrome
EP0581856B1 (en) Method of preventing nmda receptor complex-mediated neuronal damage
CA2149924C (en) Use of phenserine to treat cognitive disorders
Jin et al. Inhibiting ferroptosis: A novel approach for stroke therapeutics
US5025032A (en) Phenyl butyl nitrone compositions and methods for treatment of oxidative tissue damage
US4950658A (en) Method of medical treatment of Alzheimer&#39;s disease
Braida et al. Eptastigmine: ten years of pharmacology, toxicology, pharmacokinetic, and clinical studies
Gupta et al. Promising effects of emoxypine and its succinate derivative in the management of various diseases-with insights on recent patent applications
Marques et al. Muscle regeneration in dystrophic mdx mice is enhanced by isosorbide dinitrate
Smith et al. Inhibition of the betaine-GABA transporter (mGAT2/BGT-1) modulates spontaneous electrographic bursting in the medial entorhinal cortex (mEC)
Giacobini Modulation of brain acetylcholine levels with cholinesterase inhibitors as a treatment of Alzheimer disease
US6362160B1 (en) Immunophilin-binding agents prevent glutamate neurotoxicity associated with vascular stroke and neurodegenerative diseases
WO2001022960A1 (en) Treatment of carbon monoxide poisoning
Alonso et al. Nitric-oxide-related and non-related mechanisms in the acetylcholine-evoked relaxations in cat femoral arteries
JP2022539185A (en) Methylthioninium as a Cognitive Enhancer
BILOBA 17. IN VIVO STUDIES OF THE PHARMACOLOGICAL AND BIOCHEMICAL ACTIVITIES OF GINKGO BILOBA EXTRACT (EGb 761) AND ITS CONSTITUENTS
Díaz-Trelles et al. Antihistamine terfenadine inhibits calcium influx, cGMP formation, and NMDA receptor-dependent neurotoxicity following activation of L-type voltage sensitive calcium channels
RU2314828C1 (en) Means for treating spasmodic syndrome cases

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: PAT HOLDER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: LTOS); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

REFU Refund

Free format text: REFUND - PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: R1553); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FPAY Fee payment

Year of fee payment: 12