WO2007118276A1 - Method of treatment of age-related macular degeneration(amd) - Google Patents

Method of treatment of age-related macular degeneration(amd) Download PDF

Info

Publication number
WO2007118276A1
WO2007118276A1 PCT/AU2007/000490 AU2007000490W WO2007118276A1 WO 2007118276 A1 WO2007118276 A1 WO 2007118276A1 AU 2007000490 W AU2007000490 W AU 2007000490W WO 2007118276 A1 WO2007118276 A1 WO 2007118276A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
subject
amd
formula
pharmaceutically acceptable
Prior art date
Application number
PCT/AU2007/000490
Other languages
French (fr)
Inventor
Ashley Bush
Colin Louis Masters
Penelope Jane Huggins
Jack Gordon Parsons
Gaik Beng Kok
Vijaya Kenche
Mariana El Sous
Original Assignee
Prana Biotechnology Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to ES07718737.5T priority Critical patent/ES2437997T3/en
Priority to BRPI0710737-4A priority patent/BRPI0710737A2/en
Priority to JP2009504530A priority patent/JP5290147B2/en
Priority to US12/297,165 priority patent/US20100144693A1/en
Priority to AU2007240120A priority patent/AU2007240120A1/en
Priority to DK07718737.5T priority patent/DK2012789T3/en
Priority to NZ572591A priority patent/NZ572591A/en
Priority to EP07718737.5A priority patent/EP2012789B1/en
Application filed by Prana Biotechnology Ltd filed Critical Prana Biotechnology Ltd
Priority to CA002683756A priority patent/CA2683756A1/en
Publication of WO2007118276A1 publication Critical patent/WO2007118276A1/en
Priority to AU2010206074A priority patent/AU2010206074B2/en
Priority to IL207495A priority patent/IL207495A/en
Priority to US12/952,425 priority patent/US20110071167A1/en
Priority to US14/087,689 priority patent/US9163018B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/33Heterocyclic 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
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/04Chelating agents

Definitions

  • the present invention relates generally to the field of treatment and prophylaxis of retinal degenerative diseases. More particularly, the present invention contemplates a method for preventing, reducing the risk of development of, or otherwise treating or ameliorating the symptoms of, age-related macular degeneration (AMD) or related retinal conditions in mammals and in particular humans.
  • the present invention further provides therapeutic compositions enabling dose-dependent or dose-specific administration of agents useful in the treatment and prophylaxis of age-related macular degeneration or related retinal degenerative conditions.
  • disorders include very common conditions that affect older subjects - such as AMD as well as rarer, earlier-onset dystrophies that in some cases can be detected in the first decade of life.
  • Other maculopathies include North Carolina macular dystrophy, Sorsby's fundus dystrophy, Stargardt's disease, pattern dystrophy, Best disease and Malattia leventinese.
  • AMD AMD is the leading cause of permanent vision loss for individuals over age 65, currently affecting approximately 15 million Americans, AMD affects light-sensitive photoreceptor cells and pigmented epithelial cells in the macula, the center of the retina of the eye. While it may not cause total blindness, the disease destroys central vision, making reading, watching electronic monitor screens and driving impossible. It has no documented cure, has never demonstrated spontaneous remission and effective treatments are very limited.
  • the retina is a complicated network of nerve cells that changes light into nerve impulses that travel to the brain where they are interpreted as visual images.
  • the central part of the retina called the macula, is responsible for vision that is needed for reading and other detailed work. Damage to the macula results in poor vision.
  • the most common disease process that affects the macula is AMD.
  • AMD retinal photoreceptor and pigment epithelial cells in the macula die over the course of several years. The cell death and gradual visual loss usually do not begin until age 60 or older, hence the name age- related macular degeneration.
  • Dry macular degeneration although more common, typically results in a less severe, more gradual loss of vision.
  • Patients who are affected by dry AMD have gradual loss of central vision due to the death of photoreceptor cells and their close associates, retinal pigmented epithelial (RPE) cells, with deposition of a complex waxy amyloid mixture, termed 'drusen'.
  • RPE retinal pigmented epithelial
  • Photoreceptors the cells in the retina that actually 'see' light, are essential for vision.
  • Macrophagic RPE cells are necessary for photoreceptor survival, function and renewal.
  • CNV choroidal neovascularization
  • the RPE cells in the eye act as macrophages, which phagocytize and recycle components of the membranous outer segments of photoreceptors. If the mitochondria within the RPE cells are damaged, the photoreceptor recycling is inhibited, with resultant accumulation of drusen. Drusen causes a lateral stretching of the RPE monolayer and physical displacement of the RPE from its immediate vascular supply, the choriocapillaris. This displacement creates a physical barrier that may impede normal metabolite and waste diffusion between the choriocapillaris and the retina.
  • laser treatment can sometimes be given to destroy the abnormal blood vessels formed in wet AMD.
  • the laser is a beam of light that is absorbed by the pigment of blood, drugs and RPE cells, which converts to heat energy that cauterizes the abnormal blood vessels.
  • the neovascularization returns, since the stimulus has not been removed, resulting in severe loss of vision.
  • most of the patients with AMD, who have very poor vision have lost it due to sequelae of neovascularization.
  • Current medical opinion states that there is no treatment available that permanently prevents the cell death or abnormal blood vessel growth that occurs in AMD.
  • Phototherapy involves targeting light to the macular area containing the lesion of nascent defective blood vessels to inhibit or impair their function.
  • One type of phototherapy is photodynamic therapy (PDT).
  • PDT photodynamic therapy
  • a photosensitive agent is administered into the vessels of a patient, then the agent is activated at the target site of the lesion of new vessels (the macula) by directing low energy light from a laser specifically to this area.
  • the activated agent generates free radicals and other activated chemical species which destabilize and destroy the new vessels.
  • PDT has been reported to be of some benefit to patients having AMD.
  • one study (Arch. Ophthalmol. 777:1329-1345, 1999) evaluated PDT in four hundred and two eyes from patients diagnozed with AMD in at least one eye.
  • Treatment outcome was assessed by comparing the patient's ability to accurately read a conventional vision chart (one having about five letters per line) pre-treatment and post-treatment.
  • 61% of the eyes (246/402) lost fewer than 15 letters (that is, the patient lost less than about three lines on a standard visual chart), while 46% of the eyes (96/207) from patients undergoing treatment with a placebo lost fewer than 15 letters (p ⁇ 0.001).
  • photocoagulation therapy Another type of phototherapy is photocoagulation therapy.
  • photocoagulation therapy high energy light from a laser is directed specifically to the target site of the new vessels. The heat generated from the high energy laser coagulates the fluid in and around the new vessels.
  • Laser photocoagulation is not a form of PDT; it is a separate treatment approach. It uses lateral transfer of heat, applied with a cautery-like method, to coagulate fluid within and surrounding the vessel, while PDT uses an activated photosensitive agent to generate active chemicals which damage or destroy the new vessels containing the agent.
  • PDT or laser photocoagulation therapy is separately used to treat patients with AMD, neither is without drawbacks.
  • a problem with PDT is that its effects are transient; patients receiving PDT must be retreated about every three months. Furthermore, the patients require at least five retreatments within the first two years merely to stabilize their condition, and before any therapeutic effect occurs. These cumulative treatments damage the retina, further reducing the patient's visual acuity.
  • One drawback of laser photocoagulation is that it is non-selective, and does not target only the new blood vessels. It must therefore be administered so that only the lesions are targeted, and the unaffected surrounding tissues are undamaged.
  • the present invention is predicated in part on the recent determination that proteinaceous deposits on the limiting membrane of the retina, referred to as "drusen", also comprise zinc and copper and hence are proposed to be similar to an amyloid type plaque.
  • drusen proteinaceous deposits on the limiting membrane of the retina
  • the present invention contemplates the use of a metal protein attenuating compound (MPAC) to reduce the levels of or otherwise remove excess metal from drusen thereby restoring normal metal homeostasis in the retina.
  • MPAC metal protein attenuating compound
  • the present invention is particularly useful for treating or preventing or otherwise reducing the risk of development of age-related macular degeneration (AMD); however, the subject invention extends to the treatment of any retinal degenerative disorder associated with amyloid type aggregates, complexes, deposits or plaques or any condition associated with drusen which comprise excess metal.
  • the method of the present invention is useful irrespective of any inhibition of a matrix metalloproteinase and/or a dose-specific amount of MPAC may be employed.
  • a single agent may be administered or a combination of two or more agents.
  • the present agents comprise at least two fused 6-membered rings with at least a nitrogen atom at position 1 and a hydroxy or mercapto group at position 8.
  • Useful compounds are defined by Formulae I through XXVII which are described in detail below.
  • Suitable compounds include those in Table 8 such as PB-1033, PB-1076, PB- 1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically acceptable salt or derivative or functional equivalent thereof.
  • one aspect of the present invention contemplates a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue.
  • the present invention also provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue.
  • the present invention provides a method for treating a subject with age- related macular degeneration (AMD) said method comprising administering to said subject an amount of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof effective to reduce metal in retinal drusen to a level which ameliorates symptoms of AMD .
  • AMD age- related macular degeneration
  • alter metal levels and “reduce metal” is used in its broadest sense and refers to a change in the distribution of a metal in retinal drusen or surrounding tissue as well as a change in the amount or activity of metal in drusen or surrounding tissue.
  • the phrases also refer to a reduction in the amount or activity of metal in retinal drusen or surrounding tissue as well as a reduction in the amount or activity of metal in particular areas i.e. the distribution of metal in retinal drusen or surrounding tissue.
  • MPAC metal-binding protein
  • another aspect of the present invention provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue, irrespective of any effect on a matrix metalloproteinase.
  • references to "irrespective” means that one or more metalloproteinases may be inhibited or no metalloproteinases are inhibited. Still another aspect of the present invention defines a specific dosage range to optimally restore metal homeostasis in the retina.
  • this aspect of the present invention is directed to a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue wherein the effective amount is a specific dose range to optimally restore metal homeostasis in the retina.
  • Another aspect of the present invention contemplates a method for reducing levels of a metal from retinal drusen in a subject to thereby ameliorate symptoms of age-related macular degeneration (AMD) said method comprising administering to said subject an effective amount of PB- 1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof.
  • AMD age-related macular degeneration
  • the present invention also provides for the use of an MPAC in the manufacture of a medicament for the treatment of a retinal degenerative disorder in a subject.
  • the present invention contemplates the use of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof in the manufacture of a medicament for the treatment of age-related macular degeneration (AMD) in a subject.
  • AMD age-related macular degeneration
  • Combination therapy also forms part of the present invention in which two or more MPACs are administered or an MPAC and another active such as a metal chelator, cytokine, genetic molecule anti-microbial or anti-viral agent, an anti-oxidant, an antibiotic and/or an anasthetic.
  • another active such as a metal chelator, cytokine, genetic molecule anti-microbial or anti-viral agent, an anti-oxidant, an antibiotic and/or an anasthetic.
  • the preferred subject is a human although the present invention has application in the veterinary, horse racing and animal husbandry industries.
  • the present invention further provides formulations for treating, preventing or reducing the risk of developing a retinal degenerative condition or disorder comprising an MPAC as herein described.
  • PB- 1033 is a particularly useful MPAC 5
  • the present invention extends to any MPAC encompassed by the compounds of Formulae I through XXVII such as but not limited to those in Table 8 including PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof.
  • a formulation includes a single formulation, as well as two or more formulations
  • an agent or “a reagent” includes a single agent or reagent, as well as two or more agents or reagents; and so forth.
  • agent used interchangeably herein to refer to a chemical or biological entity which induces or exhibits a desired effect such as ameliorating the symptoms of a retinal degenerative disease.
  • agent also encompass pharmaceutically acceptable and pharmacologically active ingredients of those active agents specifically mentioned herein.
  • agent means agent, reagent, “compound”, “pharmacologically active agent”, “medicament”, “therapeutic”, active” and “drug”
  • active entity per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, prodrugs, metabolites, analogs, etc.
  • references to an "agent”, “chemical agent”, “compound”, “pharmacologically active agent”, “medicament”, “therapeutic”, “active” and “drug” includes combinations of two or more active agents.
  • a “combination” also includes multi-part such as a two-part composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispenzation.
  • a multi-part pharmaceutical pack may have two or more agents separately maintained.
  • this aspect of the present invention includes combination therapy.
  • Combination therapy includes the co-administration of a metal chelator and another active such as a chemical compound, cytokine, genetic molecule, anti-microbial or anti-viral agent, an antibiotic and/or an anasthetic.
  • an agent as used herein mean a sufficient amount of the agent to provide the desired therapeutic or physiological or effect or outcome.
  • Such an effect or outcome includes altering or reducing availability of metal ions and/or reducing their amount in drusen, reducing amyloid levels reducing or preventing macular degeneration or a related condition and/or treating or preventing vision impairment.
  • Undesirable effects e.g. side effects, are sometimes manifested along with the desired therapeutic effect; hence, a practitioner balances the potential benefits against the potential risks in determining what is an appropriate "effective amount”.
  • the exact amount required will vary from subject to subject, depending on the species, age and general condition of the subject, mode of administration and the like. Thus, it may not be possible to specify an exact "effective amount”. However, an appropriate "effective amount” in any individual case may be determined by one of ordinary skill in the art using only routine experimentation.
  • the effective amount is deemed the amount required to prevent or ameliorate symptoms of the retinal degeneration condition such as AMD.
  • the amount of MPAC used is the amount required to or which is effective in reducing levels of metal drusen.
  • metals include zinc and copper.
  • Effective amounts include from 1 ng/ml to 1000 mg/ml such as from about 5 ng/ml to about 500 mg/ml or about 10 ng/ml to about 100 mg/ml or amounts or ranges in between.
  • metal and “metal ion: may be used interchangedly in this context.
  • excipient or diluent a pharmaceutical vehicle comprized of a material that is not biologically or otherwise undesirable, i.e. the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction.
  • Carriers may include excipients and other additives such as diluents, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives, and the like.
  • a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
  • Treating" a subject may involve prevention of a retinal degenerative condition or other adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by ameliorating the symptoms of the condition.
  • the presence invention contemplates a reduction of amyloid type plaque formation and/or a reduction in metal content in drusen to restore normal metal homeostasis in the retina.
  • the "subject” as used herein refers to an animal, preferably a mammal and more preferably a primate including a lower primate and even more preferably a human who can benefit from the formulations and methods of the present invention.
  • a subject regardless of whether a human or non-human animal may be referred to as an individual, patient, animal, host or recipient.
  • the compounds and methods of the present invention have applications in human medicine, veterinary medicine as well as in general, domestic or wild animal husbandry.
  • an "animal” includes an avian species such as a poultry bird (including ducks, chicken, turkeys and geese), an aviary bird or game bird.
  • condition in a non-human animal may not be a naturally occurring but induced such as in an animal model.
  • the preferred animals are humans, non-human primates such as marmosets, baboons, orang-utans, lower primates such as tupia, livestock animals, laboratory test animals, companion animals or captive wild animals.
  • a human is the most preferred target.
  • non-human animal models may be used.
  • test system examples include mice, rats, rabbits, guinea pigs and hamsters. Rabbits and rodent animals, such as rats and mice, provide a convenient test system or animal model as do primates and lower primates. Livestock animals include sheep, cows, pigs, goats, horses and donkeys. Non-mammalian animals such as avian species, zebrafish, amphibians (including cane toads) and Drosophila species such as Drosophila melanogaster are also contemplated. Instead of a live animal model, a test system may also comprise a tissue culture system.
  • a "retinal degenerative condition” is a condition which is characterized by a progressive loss of vision. Conditions within the scope of this term include age-related macular degeneration (AMD), North Carolina macular dystrophy, Sorsby's fudus dystrosphy, Stargardt's disease, pattern dystrophy, Best disease and Malattia leventinese.
  • AMD age-related macular degeneration
  • North Carolina macular dystrophy North Carolina macular dystrophy
  • Sorsby's fudus dystrosphy Stargardt's disease
  • pattern dystrophy Best disease
  • Malattia leventinese Malattia leventinese.
  • AMD AMD
  • the present invention extends to any retinal degenerative disease associated with or characterized by amyloid like aggregates, deposits or plaques.
  • one aspect of the present invention contemplates a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue.
  • the altered metal levels are reduced metal levels.
  • the present invention provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue, irrespective of any effect on a matrix metalloproteinase.
  • the altered metal levels are reduced metal levels.
  • Still a further aspect of the present invention is directed to a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue wherein the effective amount is a specific dose range to optimally restore metal homeostasis in the retina.
  • the present invention also provides for the use of an MPAC in the manufacture of a medicament for the treatment of a retinal degenerative disorder in a subject.
  • the preferred agents of the present invention comprise at least two fused 6-membered rings with at least a nitrogen atom at position 1 and a hydroxy or mercapto group at position 8.
  • the agents of the present invention are collectively referred to as metal protein attenuating compounds or MPACs and have one or more of the following properties: act as ionophores (i.e. capture and transfer metals into cells), is a metal binder, crosses the blood brain barrier (BBB), exhibits reduced cell toxicity, is capable of dissolving or disrupting amyloid type protein deposits, aggregates or plaques and is stable in aqueous environments.
  • the agents have two or more, three or more or four or more or five or more ] of the above-listed properties.
  • Particularly useful compounds include those in Table 8 such as PB- 1033, PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof.
  • PB- 1033 is particularly useful although the present invention is not to be so limited.
  • the present invention further contemplates a method for treating a subject with age-related macular degeneration (AMD) said method comprising administering to said subject an amount of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof effective to reduce metal in retinal drusen to a level which ameliorates symptoms of AMD.
  • AMD age-related macular degeneration
  • the subject invention also provides a method for reducing levels of a metal from retinal drusen in a subject to thereby ameliorate symptoms of age-related macular degeneration (AMD) said method comprising administering to said subject an effective amount of PB- 1033 or a pharmaceutically acceptable salt, derivative or equivalent thereof.
  • AMD age-related macular degeneration
  • Examples of pharmaceutically acceptable chemical derivatives or functional equivalents of PB-1033 include those in Table 8 such as PB-1076, PB-1085, PB-1120, PB-1127, PB- 1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof.
  • Examples of metals include zinc and copper.
  • R is O or S
  • R 1 is independently selected from H 3 optionally substituted alkyl, optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; CN; halo; CF 3 ; SO 3 H; and OR 2 , SR 2 , SOR 2 , SO 2 R 2 , NR 2 R 3 , (CH 2 ) n NR 2 R 3 , HCNOR 2 , HCNNR 2 R 3 , CONR 2 R 3 , CSNR 2 R 3 , NCOR 2 , NCSR 2 , COR 2 , CO 2 R 2 , CSR 2 or SO 2 NR 2 R 3 in which R 2 and R 3 are independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety and n is an integer of 1 to
  • X is independently selected from CH, CO, N and NH;
  • Z is independently selected from CH, CO, N, NH and O;
  • Y is independently absent or together with the ring to which it is attached forms a 5- or 6- membered optionally substituted aryl or a 5- or 6-membered optionally substituted heterocyclyl;
  • n is an integer from 1 to 3;
  • p is an integer from 1 to 4,
  • R is O.
  • R 1 is preferably halo, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkyl, OR 2 , SR 2 , (CH 2 ) n NR 2 R 3 , CONR 2 R 3 and NCOR 2 in which n, R 2 and. R 3 are as defined above.
  • R 1 is fluorine; iodine; chlorine; optionally substituted phenyl such as 4-halophenyl, for example, 4-fluorophenyl or 4-chlorophenyl; an optionally substituted unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolyl or pyridinyl; an optionally substituted saturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolidinyl or piperazinyl; an optionally substituted saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl; optionally substituted Ci -4 alkyl such as methyl or ethyl; optionally substituted C 2-6 cycloalkyl such as cyclopropyl; optionally substituted Ci -6 alkoxy; optionally substituted thio; CH 2 NR 4 R 5 in which
  • Y is preferably an optionally substituted phenyl; an optionally substituted unsaturated 5- or 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolyl or pyridinyl; or an optionally substituted saturated 5 or 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl.
  • the preferred halo group is chlorine but other halogen atoms are encompassed by the present invention.
  • R , m, n and p are as defined above and q is an integer of 1 or 2.
  • R 1 is H or halo, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety;
  • R 2 is H; optionally substituted alkyl; optionally substituted alkenyl; optionally substituted aryl; optionally substituted heterocyclyl; optionally substituted alkoxy; an antioxidant; a targeting moiety; COR 6 or CSR 6 in which R is H, optionally substituted alkyl, optionally substituted alkenyl, hydroxy, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant, a targeting moiety, OR 7 , SR 7 or NR 7 R 8 in which R 7 and R 8 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; CN; (CH 2 ) n NR 9 R 10 , HCNOR 9 or HCNNR 9 R 10 in which R 9 and R 10 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substitute
  • R 3 , R 4 , R 5 , R and R are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, hydroxy, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, halo, SO 3 H, amine, CN, CF 3 , optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety,
  • R 4 when R 1 to R 3 , R and R are H, then R 4 is not Cl or I and R 5 is not I; (b) when R 1 to R 3 , R, R ' and R 5 are H, then R 4 is not CHO, CHOHCCl 3 ,
  • R 1 , R 5 , R' and R are H, R 2 is CO 2 H and R 3 is OH, then R 4 is not bromo, methyl, phenyl, hydroxymethyl or trifluoromethyl;
  • R 1 , R 4 , R 5 and R are H, R 2 is CO 2 H and R 3 is OH, then R' is not bromo, iodo, methyl, phenyl, propyl, phenethyl, heptyl, benzylaminomethyl, 3-aminopropyl, 3- hydroxypropyl, 4-methoxyphenyl, 3-methylphenyl, 4-chlorophenyl, 3,4-dichloro ⁇ henyl, pyridin-3-yl, furo-2-yl, 4-chlorophenyl, 3,4-dichlorophenyl, 2-chlorophenyl, 3- chlorophenyl, 2-chlorophenyl, 3-chlorophenyl, 2-methoxyphenyl or piperidin-2-yl;
  • R 1 , R 4 , R and R' are H, R 2 is CO 2 H and R 3 is OH, then R 5 is not phenyl, 3- hydroxypropyl, phenethyl, 3-aminoprop-l-yl or hex-l-yl;
  • R 1 , R, R' and R 5 are H, R 2 is CO 2 Me and R 3 is OH, then R 4 is not hydroxymethyl, phenyl or bromo;
  • R 1 , R, R 4 and R 5 are H, R 2 is CO 2 Me and R 3 is OH, then R 1 is not 4- methoxyphenyl, 3-methylphenyl, pyridin-3-yl, benzyl, bromo, 4-chlorophenyl, 3,4- dichlorophenyl, 3-hydroxypropyl or 3-tert-butoxycarbonylaminopropyl;
  • R 1 , R, R 4 and R 1 are H, R 2 is CO 2 Me and R 3 is OH, then R 5 is not phenyl or 3-tert-butoxycarbonylaminoprop- 1 -yl;
  • R 3 is not toluene-4- sulphonylamino, piperazin-1-yl, morpholin-1-yl, piperidin-1-yl, 4-methylpiperazin-l-yl, 3- benzoylaminoprop-1-yl, phenethyl, 3-tert-butoxycarbonylaminopropyl, 3-hydroxypropyl, amino or hex-1-yl;
  • R 3 is not phenyl, 4- chlorophenyl, phenethyl, 3-hydroxypropyl, amino, morpholin-1-yl, piperidin-1-yl, 4- methylpiperazin-1-yl, toluene-4-sulphonylamino, 3-benzoylaminoprop-l-yl, aminoprop-1- ynyl, hex- 1 -yl, 5-hydroxypent- 1 -yl, piperazin- 1 -yl or 2-( 1 -piperazinyl)pyrimidinyl;
  • R 4 is not phenyl, 4- chlorophenyl or phenylethyl
  • R 1 , R 5 , R 1 , R 4 , R 3 and R are H, then R 2 is not 2H-tetrazol-l-yl;
  • R 1 , R 5 , R 4 and R are H, R 2 is CO 2 H and R 3 is OH, then R' is not 3,5- dichlorophenyl or 4-fluorophenyl; and (u) at least one of R . 1 + to ⁇ ⁇ R> 5 , R and R 1 is other than H.
  • Useful compounds of Formula II are as follows:
  • R, R 1 and R 3 are as defined in Formula II above;
  • R a is H; optionally substituted C 1-6 alkyl; optionally substituted C 1-6 alkenyl; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; COR 6 a or CSR 6 a in which R 6 a is H, optionally substituted Cj -6 alkyl, optionally substituted C 2-6 alkenyl, hydroxy, optionally substituted aryl, optionally substituted heterocyclyl or OR a , SR a or NR a R a in which R 8 and R a are either the same or different and selected from H, optionally substituted Ci -6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; CN; CH 2 NR 9 a R 10 a , HCNOR 9 a or HCNNR 9 a R 10 in which R 9 a and R 10 a are either the same or different and selected from H, optionally substituted C
  • Preferred compounds of Formula III are as follows:
  • R 1 is as defined in Formula II above;
  • R 2' a is optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
  • Formula IV may represent compounds in which an antioxidant moiety is attached to the C2 position of the 8-hydroxyquinoline in such a way that exposure to a prooxidative environment, that is, hydroxy radicals, will result in a molecule with enhanced metal binding properties.
  • R 1 and R 3 are as defined in Formula II above;
  • R a is optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, hydroxy, 0R 7 a ',
  • R 7 ' a and R 8' a are either the same or different and selected from H, optionally substituted Ci -6 alkyl, optionally substituted aryl or optionally substituted heterocyclyl.
  • Formula V represents compounds in which a hydrophilic amide moiety is attached to the C2 position of the 8-hydroxyquinoline so as to generally enhance solubility while maintaining membrane permeability.
  • R 1 is as defined in Formula II above;
  • R 2 " a is CN; CH 2 NR 9 ' a R 10' a , HCNOR 9' a or HCNNR 9' a R 10 ' a in which R 9' a and R 10 ' a are either the same or different and selected from H, optionally substituted C 1-6 alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
  • R 1 is as defined in Formula II above;
  • R 1 V and R 12 a ' are either the same or different and selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted aryl and optionally substituted heterocyclyl or together form optionally substituted heterocyclyl.
  • R 1 is as defined in Formula II above;
  • R 1 V and R 1 V are either the same or different and selected from H, optionally substituted Ci -6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
  • R 1 , R ' , R, R 2 and R 3 are as defined in Formula II above;
  • R 4 b and R 5 b are either the same or different and selected from H; optionally substituted Ci -6 alkyl; optionally substituted C 2-6 alkenyl; halo; CN; CF 3 ; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; SO 3 H; SO 2 NR 13 a R 14 a in which R 13 a and R 14 a are as defined in Formula III above; or OR 15 b , SR 15 b , SO 2 R 15 b , CONR 15 b R 16 b or NR 15 b R 16 b in which R 15 b and R 16 b are either the same or different and selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 2-6 alkenyl, optionally substituted Ci -6 acyl, optionally substituted aryl or optionally substituted heterocyclyl,
  • provisos (a) to (c), (e), (g), (h), (I), (k), (o), (q), (r), and (u) as defined above.
  • R 1 , R 1 , R 5 R and R are as defined in Formula II above;
  • R 4 b' and R 5 a are as defined in Formula IX above provided that at least one is halo,
  • R is as defined in Formula II above;
  • R >4b is H or halo
  • R > 5 b " is optionally substituted aryl or optionally substituted heterocyclyl.
  • a representative example is shown below.
  • R 1 is as defined in Formula II above;
  • R " is C 1-6 alkoxy, halo, C 1-6 alkyl, C 2-6 alkenyl or Cj -6 haloalkyl;
  • R b is H or halo. A representative example is shown below.
  • R 1 is as defined in Formula II above;
  • R is as defined in Formula XIII above. • Formula XIV
  • R >2 i to ⁇ R r.5 , R and R 1 are as defined in Formula II above;
  • R b is optionally substituted Ci -6 alkyl, optionally substituted aryl, optionally substituted aryl acyl, Cj -6 alkyl acyl or optionally substituted heterocyclyl.
  • R 1 , R 2 , R 3 , R and R' are as defined in Formula II;
  • R 4 C and R 5 C is halo and the other is selected from H 5 optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, hydroxy, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, SO 3 H, amine, CN, CF 3 , optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant and a targeting moiety,
  • a preferred compound of Formula XV is as follows:
  • R 3 ' is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, halo, SO 3 H, amine, CN, CF 3 , optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety,
  • R 1 is as defined in Formula II and R 4 C is as defined in Formula XV; and R 5 c " is optionally substituted aryl or optionally substituted heterocyclyl;
  • R is as defined in Formula II, R c is as defined in Formula XV and R" is as defined in Formula XII;
  • R and R' are as defined in Fo ⁇ nula H 5
  • R 0 and R 0 are as defined in Formula XV
  • R J b is as defined in Formula XII.
  • the present invention also provides a compound of Formula XX which is a compound of Formula II with the provisos that:
  • R 1 , R 4 , R and R • are H, R 2 is CO 2 H and R 3 is OH, then R 5 is not phenyl, 3- hydroxypropyly phenethyl, 3-aminoprop-l-yl or hex-l-yl;
  • R 1 , R 4 , R ' and R 5 are H, R 2 is CO 2 H and R 3 is OH, then R is not N- morpholinomethyl, bromo or phenyl;
  • R 1 , R, R 1 and R 5 are H, R 2 is CO 2 Me and R 3 is OH, then R 4 is not hydroxymethyl, phenyl or bromo;
  • R 1 , R, R 4 and R 5 are H, R 2 is CO 2 Me and R 3 is OH, then R 1 is not 4- methoxyphenyl, 3-methylphenyl, pyridin-3-yl, benzyl, bromo, 4-chlorophenyl, 3,4- dichlorophenyl, 3-hydroxypropyl or 3-tert-butoxycarbonylaminopropyl;
  • R 1 , R, R 4 and R' are H, R 2 is CO 2 Me and R 3 is OH, then R 5 is not phenyl or 3-tert-butoxycarbonylaminoprop- 1 -yl;
  • R 3 is not toluene-4- sulphonylamino, piperazin-1-yl, morpholin-1-yl, piperidin-1-yl, 4-methylpiperazin-l-yl, 3- benzoylaminoprop-1-yl, phenethyl, 3-tert-butoxycarbonylaminopropyl, 3-hydroxypropyl, amino or hex-l-yl; (p) when R 1 , R 4 , R 1 and R 5 are H, R 2 is CO 2 Na and R 3 is OH, then R is not phenyl;
  • R 3 is not phenyl, A- chlorophenyl, phenethyl, 3-hydroxypropyl, amino, morpholin-1-yl, piperidin-1-yl, A- methylpiperazin-1-yl, toluene-4-sulphonylamino, 3-benzoylaminoprop-l-yl, aminoprop-1- ynyl, hex-l-yl, 5-hydroxypent-l-yl, piperazin-1-yl or 2-(l-piperazinyl)pyrimidinyl;
  • R 4 is not phenyl, A- chlorophenyl or phenylethyl
  • R 1 , R 5 , R 4 and R are H, R 2 is CO 2 H and R 3 is OH, then R' is not 3,5- dichlorophenyl or 4-fluorophenyl;
  • the invention provides a compound of Formula Ic, with the additional provisos that: (g) when R ⁇ l + to ⁇ D R 3 , R and R 1 are H, then R c and R 0 are both not chloro or bromo; and
  • Particularly preferred compounds include a series of so called “PB” (or PBT) compounds, some of which are referred to above, such as:
  • the 8-hydroxyl or 8-mercapto group on the above compounds may be blocked to form a prodrug, in particular an ester prodrug.
  • the 8-hydroxy or 8-mercapto represents a principal site of metabolism for the above compounds: conjugation with glucuronic acid or sulphate gives a hydrophilic species ready to be excreted.
  • R, R 1 and m are as defined for Formula I;
  • W is CH, N or NH
  • U is CH 5 CO or N
  • Preferred compounds of Formula XXI are as follows:
  • R 5 R 1 , m and q are as for Formula I.
  • R 1 is located at positions 2, 3, 5 and/or 7 and is selected from halo, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkyl and (CH 2 ) n NR 2 R 3 in which n, R 2 and R 3 are as defined above. More preferably R 1 is chlorine, optionally substituted phenyl, C 2-6 cycloalkyl, CH 2 NR 4 R 5 in which R 4 and R 5 are independently selected from H and C 1-4 alkyl or optionally substituted pyridinyl.
  • R, R 1 , m and q are as defined for Formula I.
  • R 1 may be is located at positions 2, 4, 5 and/or 7 and is selected from halo and optionally substituted heterocyclyl.
  • R 1 is chloro and/or morpholinyl. Preferred examples are shown below.
  • R, R 1 , m and q are as defined for Formula I.
  • R 1 is located at positions 2, 5 and/or 7 and is selected from halo and CH 2 NR 4 R 5 in which R 4 and R 5 are independently selected from H and Ci -4 alkyl.
  • R, R 1 , m and q are as defined for Formula I.
  • R 1 is located at positions 2 and/or 7 and is selected from optionally substituted heterocyclyl, CO 2 R 2 , (CH 2 ) n NR 2 R 3 and CONR 2 R 3 in which n, R 2 and R 3 are as defined in Formula I.
  • R 1 is located at positions 2, 3, 6 and/or 7 and is selected from halo, optionally substituted aryl and (CH 2 ) n NR 2 R 3 in which n, R 2 and R 3 are as defined for Formula I.
  • R 1 and m are as defined for Formula I.
  • R 1 is located at positions 2 and/or 7 and is selected from halo and (CH 2 ) n NR 2 2Rr>3 in which n, R and R are as defined above.
  • alkyl used either alone or in compound words such as “optionally substituted alkyl” or “alkylamino” refers to straight chain, branched chain or cyclic hydrocarbon groups having from 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 4 carbon atoms. Illustrative of such alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, neopentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • alkyl groups are C 1-4 alkyl such as methyl or ethyl and C 2-6 cycloalkyl such as cyclopropyl.
  • alkenyl used either alone or in compound words such as “optionally substituted alkenyl”, denotes linear, branched or mono- or poly-cyclic radicals having at least one carbon-carbon double bond of 2 to 20 carbon atoms, preferably 2 to 14 carbon atoms, more preferably 2 to 6 carbon atoms.
  • alkenyl radicals include allyl, ethenyl, propenyl, butenyl, iso-butenyl, 3-methyl-2-butenyl, 1-pentenyl, cyclopentenyl, 1-methyl- cyclopentenyl, 1-hexenyl, 3-hexenyl, cyclohexenyl, 1-heptenyl, 3-heptenyl, 1-octenyl, cyclooctenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 1-decenyl, 3-decenyl, l,3rbutadienyl, 1,4- pentadienyl, 1,3-cyclopentadienyl, 1,3-hexadienyl, 1 ,4-hexadienyl, 1,3-cyclohexadienyl, 1 ,4-cyclohexadienyl, 1 ,3-cyclohepta
  • alkynyl used either alone or in compound words such as “optionally substituted alkynyl” refers to straight chain or branched chain radicals having at least one carbon- carbon triple bond of 2 to 20 carbon atoms, preferably 2 to 14 carbon atoms, more preferably 2 to 6 carbon atoms.
  • Examples include ethynyl, 1-propynyl, 1- and 2-butynyl, 2-methyl-2-pro ⁇ ynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 2-hexynyl, 3-hexynyl, 4- hexynyl, 5-hexynyl, 10-undecynyl, 4-ethyl-l-octyn-3-yl, 7-dodecynyl, 9-dodecynyl, 10- dodecynyl, 3-methyl-l-dodecyn-3-yl, 2-tridecynyl, 11-tridecynyl, 3-tetradecynyl, 7- hexadecynyl, 3-octadecynyl and the like.
  • heterocyclyl group used either alone or in compound words such as “optionally substituted heterocyclyl” refers to monocyclic or polycyclic heterocyclic groups containing at least one heteroatom atom selected from nitrogen, sulphur and oxygen.
  • Suitable heterocyclic groups include N-containing heterocyclic groups, -such as, unsaturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl or tetrazolyl;
  • saturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms such as, pyrrolidinyl, imidazolidinyl, piperidino or piperazinyl; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, such as indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl or tetrazolopyridazinyl;
  • unsaturated 3 to 6-membered heteromonocyclic group containing an oxygen atom such as, pyranyl or ruryl
  • unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, oxazolyl, isoxazolyl or oxadiazolyl;
  • unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, benzoxazolyl or benzoxadiazolyl;
  • unsaturated condensed heterocyclic group containing 1 to 2 sulphur atoms and 1 to 3 nitrogen atoms such as, benzothiazolyl or benzothiadiazolyl.
  • the heterocyclyl is an unsaturated 5 or 6-membered heteromonocyclic group containing 1 to 3 nitrogen atoms such as imidazolyl or pyridinyl; a saturated 5 or 6- membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolidinyl or piperazinyl; or a saturated 5 or 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl.
  • aryl used either alone or in compound words such as “optionally substituted aryl” denotes a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused.
  • aryl embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane and biphenyl.
  • the aryl is optionally substituted phenyl such as 4-halophenyl, more preferably 4-fiuorophenyl or 4-chlorophenyl.
  • halo refers to fluorine, chlorine, bromine or iodine, preferably fluorine, iodine or chlorine, most preferably chlorine.
  • alkoxy refers to straight chain or branched oxy-containing radicals preferably each having alkyl portions of 1 to about 6 carbon atoms. Examples of alkoxy include methoxy, ethoxy, propoxy, butoxy and tert-butoxy.
  • optionally substituted thio refers to optional substituents such as radicals containing a linear or branched alkyl of 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 4 carbon atoms, attached to a divalent sulphur atom.
  • alkylthio radicals include methylthio, ethylthio, propylthio, butylthio and hexylthio.
  • optionally substituted refers to a group which may or may not be further substituted with one or more groups selected from alkyl, alkenyl, alkynyl, aryl, aldehyde, halo, haloalkyl, haloalkenyl, haloalkynyl, haloaryl, hydroxy, alkoxy, alkenyloxy, aryloxy, benzyloxy, haloalkoxy, haloalkenyloxy, haloaryloxy, nitro, nitroalkyl, nitroalkenyl, nitroalkynyl, nitroaryl, nitroheterocyclyl, amino, alkylamino, dialkylamino, alkenylamino, alkynylamino, arylamino, diarylamino, benzylamino, dibenzylamino, acyl, alkenylacyl, alkynylacyl, arylacyl, acyl, al
  • the optional substituent is C 1-6 alkyl, more preferably Cj -4 alkyl; CF 3 ; fluorine; chlorine; iodine; cyano; Ci -6 alkoxy, more preferably Ci -4 alkoxy; aryl; heterocyclyl; amino; or alkylamino.
  • antioxidant is used herein in its broadest sense and refers to a group which has the capacity to react with a reactive oxygen species such as a hydroxyl radical in such a way as to generate a non toxic product.
  • a reactive oxygen species such as a hydroxyl radical
  • examples include phenols such as 3,4,5- trimethoxyphenyl and 3,5-di-t-butyl-4-hydroxyphenyl, indole amines such as melatonin and flavonoids.
  • Other examples may be found the literature (Wright et al, J Am Chem Soc 123:1173-1183, 2001).
  • targeting moiety is used herein in its broadest sense and refers to a group which will facilitate the brain delivery of the drug by way of an active transport mechanism.
  • the targeting moiety is recognized by specific transporter enzymes integral to the blood brain barrier and these transporter enzymes then provide a mechanism for the drug to be imported into the brain.
  • transporters are sodium dependant and their substrates contain carboxylic acids such as ascorbic acid and L-glutamate. Conjugation of the targeting moiety to the drug is enacted so as to retain the acid moiety.
  • metal chelator is used herein is distinguished from the previously known concept of "chelation therapy”.
  • “Chelation therapy” is a term associated clinically with the removal of bulk metals such as in Wilson's disease, ⁇ -thallesemia and haemochromatosis.
  • salts of the above compounds are preferably pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salts include salts of pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium; acid addition salts of pharmaceutically acceptable inorganic acids such as hydrochloric, orthophosphoric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids; or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, trihalomethanesulphonic, toluenesulphonic, benzenes
  • solvates may form solvates with water or common organic solvents. Such solvates are encompassed within the scope of the instant invention.
  • pro-drug is used herein in its broadest sense to include those compounds which are converted in vivo to the above compounds.
  • Use of the pro-drug strategy optimises the delivery of the drug to its site of action, for example, the retina.
  • the term refers to the presence of a Ci -6 alkyl or arylester moiety which is designed to resist hydrolysis until the pro-drug has crossed the BBB.
  • the term refers to the attachment at position 2 of an antioxidant group, in particular the 3,4,- 5trimethoxyphenyl moiety or derivatives thereof.
  • Exposure to the prooxidative environment of the retinal may then lead to hydroxylation of the 3,4,5-trimethoxyphenyl group to give a 2-hydroxy-3,4,5-trimethoxyphenyl substituent, the hydroxyl group of which acts to enhance the binding properties of the above compounds.
  • tautomer is used herein in its broadest sense to include the above compounds which are capable of existing in a state of equilibrium between two isomeric forms. Such compounds may differ in the bond connecting two atoms or groups and the position of these atoms or groups in the compound.
  • compositions of the present invention comprise at least one of the above compounds together with one or more pharmaceutically acceptable carriers and optionally other therapeutic agents.
  • Each carrier, diluent, adjuvant and/or excipient . must be pharmaceutically "acceptable” in the sense of being compatible with the other ingredients of the composition and not injurious to the subject.
  • Compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers, diluents, adjuvants and/or excipients or finely divided solid carriers or both, and then if necessary shaping the product.
  • parenteral as used herein includes subcutaneous injections, aerosol for administration to lungs or nasal cavity, intravenous, intramuscular, intrathecal, intracranial, injection or infusion techniques. Intra-ocular administation is particularly useful.
  • the present invention also provides suitable topical, oral, and parenteral pharmaceutical formulations for use in the novel methods of treatment of the present invention.
  • the compounds of the present invention may be administered orally as tablets, aqueous or oily suspensions, lozenges, troches, powders, granules, emulsions, capsules, syrups or elixirs.
  • the composition for oral use may contain one or more agents selected from the group of sweetening agents, flavoring agents, colouring agents and preserving agents in order to produce pharmaceutically elegant and palatable preparations.
  • Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharin.
  • Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar.
  • Suitable flavoring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavoring.
  • Suitable preservatives include sodium benzoate, vitamin E 5 alphatocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • the tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, (1) inert diluents, such as calcium carbonate, lactose, calcium phosphate or sodium phosphate; (2) granulating and disintegrating agents, such as corn starch or alginic acid; (3) binding agents, such as starch, gelatin or acacia; and (4) lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium carbonate, lactose, calcium phosphate or sodium phosphate
  • granulating and disintegrating agents such as corn starch or alginic acid
  • binding agents such as starch, gelatin or acacia
  • lubricating agents such as magnesium stearate, stearic acid or talc.
  • These tablets may be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as gly
  • the above compounds as well as the pharmaceutically-active agent useful in the method of the invention can be administered, for in vivo application, parenterally by injection or by gradual perfusion over time independently or together. Administration may be intraocular, intravenously, intraarterial, intraperitoneally, intramuscularly, subcutaneously, intracavity, transdermally or infusion by, for example, osmotic pump. For in vitro studies the agents may be added or dissolved in an appropriate biologically acceptable buffer and added to a cell or tissue.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous 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 solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride
  • lactated Ringer's 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, anti-microbials, anti-oxidants, chelating agents, growth factors and inert gases and the like.
  • the present invention includes various pharmaceutical compositions useful for ameliorating disease.
  • the pharmaceutical compositions according to one embodiment of the invention are prepared by bringing an above compound, analogs, derivatives or salts thereof, or combinations of the above compounds and one or more pharmaceutically-active agents into a form suitable for administration to a subject using carriers, excipients and additives or auxiliaries.
  • carriers or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, glycerol and polyhydric alcohols.
  • Intravenous vehicles include fluid and nutrient replenishers.
  • Preservatives include antimicrobial, anti-oxidants, chelating agents and inert gases.
  • Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like, as described, for instance, in Remington's Pharmaceutical Sciences, 20th ed. Williams and Wilkins (2000) and The British National Formulary 43rd ed. (British Medical Association and Royal Pharmaceutical Society of Great Britain, 2002; http://bnf.rhn.net), the contents of which are hereby incorporated by reference.
  • the pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to routine skills in the art. See Goodman and Gilman's The Pharmacological Basis for Therapeutics (7th ed., 1985).
  • the pharmaceutical compositions are preferably prepared and administered in dose units.
  • Solid dose units may be tablets, capsules and suppositories.
  • different daily doses can be used for treatment of a subject, depending on activity of the compound, manner of administration, nature and severity of the disorder, age and body weight of the subject.
  • the administration of the daily dose can be carried out both by single administration in the form of an individual dose unit or else several smaller dose units and also by multiple administration of subdivided doses at specific intervals.
  • the pharmaceutical compositions according to the invention may be administered locally or systemically in a therapeutically effective dose. Amounts effective for this use will, of course, depend on the severity of the disease and the weight and general state of the subject.
  • dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of the cytotoxic side effects.
  • animal models may be used to determine effective dosages for treatment of the cytotoxic side effects.
  • Formulations for oral use may be in the form of hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin. They may also be in the form of soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions normally contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspension.
  • excipients may be (1) suspending agent such as sodium carboxymethyl cellulose, methyl cellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; (2) dispersing or wetting agents which may be (a) naturally occurring phosphatide such as lecithin; (b) a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate; (c) a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadecaethylenoxycetanol; (d) a condensation product of ethylene oxide with a partial ester derived from a fatty acid and hexitol such as polyoxyethylene sorbitol monooleate, or (e) a condensation product of ethylene oxide with a
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to known methods using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono-or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of i ⁇ jectables.
  • Liposome delivery systems such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines .
  • veterinary compositions which may be prepared, for example, by methods that are conventional in the art.
  • veterinary compositions include those adapted for:
  • oral administration external application, for example drenches (e.g. aqueous or nonaqueous solutions or suspensions); tablets or boluses; powders, granules or pellets for admixture with feed stuffs; pastes for application to the tongue;
  • drenches e.g. aqueous or nonaqueous solutions or suspensions
  • tablets or boluses e.g. aqueous or nonaqueous solutions or suspensions
  • pastes for application to the tongue for example drenches (e.g. aqueous or nonaqueous solutions or suspensions); tablets or boluses; powders, granules or pellets for admixture with feed stuffs; pastes for application to the tongue;
  • parenteral administration for example by subcutaneous, intramuscular or intravenous injection, e.g. as a sterile solution or suspension; or (when appropriate) by intramammary injection where a suspension or solution is introduced in the udder via the teat;
  • topical applications e.g. as a cream, ointment or spray applied to the skin;
  • Abeta is dissolved in distilled water and peptide concentration is assessed by measured absorption at 214 nm in UV spectrometer.
  • An aggregation reactive mixture (per one concentration of one testing compound) is set us as follows: Abeta:25 ⁇ M, ZnCl 2 50 ⁇ M, ThT 50 ⁇ M, PBS to make up to 500 ⁇ l. The tube is wrapped with foil and incubated at 37 degrees by rotation for 24 hours. A serial dilution of each test compound is made up in DMSO, for example: 100 ⁇ M, 500 ⁇ M, 1000 ⁇ M, 2500 ⁇ M and 5000 ⁇ M. The final concentrations are 1, 5, 10, 25 and 50 ⁇ M.
  • the BAS assay is adopted for post mortem retina. Using a trephine, 6 mm diameter regions of peripheral retina from frozen donor eyes are dissected. Following defrosting, the neuronal retina and the RPE cells are removed by gentle agitation in PBS buffer. Following removal of the RPE cells, strips of the Bruch membrane are cut from the eye.
  • Samples are then washed 3 times and the labeling visualized using a fluorescence and confocal microscope.
  • test results are in the form of fluorescence imaging from a confocal microscope of the 4 samples tested in this Example after 15 hours of sample incubation.
  • the results showed that TPEN inhibited ZPl labeling indicating the effectiveness of the assay.
  • PB-1033 also inhibited ZPl labeling.
  • the results clearly show that PB- 1033 inhibits and reduces metal ions in retinal drusen. Fluorescence microscopic photographs (which are in colour) are available upon request from the Patentee. EXAMPLE 3 Clinical trial
  • AMD patients are selected and given a test compound (including a PB compound) at a concentration of 500 mg/day for one month. Readouts are taken at baseline and then at 1 month and include:
  • the retinae are relieved of oxidative stress after MPAC treatment, it should be reflected by stabilization of these markers of retinal health.
  • a fiuorometric assay was used to test the ability of a test compound to inhibit hydrogen peroxide generation by A ⁇ in the presence of copper based on dichlorofluoroscein diacetate (DCF; Molecular Probes, Eugene OR).
  • the DCF solution (5mM) in 100% dimethyl sulphoxide (previously purged with argon for 2hr at 2O 0 C) was deacetylated in the presence of 0.25M NaOH for 30min and neutralized at pH 7.4 to a final concentration of ImM.
  • the reaction solutions contained AB 1-42 at concentrations in the range of 5OnM to l ⁇ M, copper-glycine chelate (Cu-GIy), was prepared by adding CuCl 2 to glycine in the ratio of 1 :6 and added to the AB in the proportion 2Cu-GIy : IAB ), reducing agents including dopamine (5 ⁇ M) or ascorbic acid, deacetylated DCF lOO ⁇ M, and HRP, O.l ⁇ M. 1-1 O ⁇ M EDTA or another chelator may also be present as a control for free copper, but was not required for the assay to function.
  • the reaction mixture was incubated at 37C for 60 min.
  • Catalase (4000 units/ml) and H 2 O 2 (l-2.5 ⁇ M) standards in PBS pH 7.4 may be included as positive controls. Fluorescence was recorded using a plate reader with excitation and emission filters at 485nM and 53OnM respectively. H 2 O 2 concentration may be established by comparing fluorescence with the H 2 O 2 standards. Inhibition of AB H 2 O 2 production was assayed by including a given concentration of test compound(s) in the test wells.
  • Cell viability is determined using the MTS assay. Culture medium is replaced with fresh neurobasal medium plus B27 supplements minus antioxidants. 1/1 Oth volume MTS solution (Cell Titre 96 Aqueous One, Promega Corporation) and incubated at 37 0 C, 2hrs. 200 microlitre aliquots are measured with a spectrophotometer at 560 nm.
  • Neuronal cortical cells were cultured for five days as per Assay 2 in NB media and B27 supplement.
  • test compounds were added to the neuronal cell cultures in NB media and B27 supplement minus antioxidants.
  • Test compounds were dissolved in 100% DMSO to a concentration of 2.5 mM (1OmM if excess compound was weighed out per vial - then diluted to 2.5mM).
  • 2.5mM stock solution was serially diluted 1 in 10 to give working solutions of 250 ⁇ M, 25 ⁇ M, 2.5 ⁇ M.
  • Test compounds were not added directly to cells, instead they were added to a 48 well 'Drug Plate' as comprized below:
  • the Drug Plate was incubated at 37° C for 15 mins. 200 ⁇ l of each well was added in triplicate to the corresponding cell plate. The cell plate was incubated at 37 C, for 4 days.
  • Human Brain Amyloid Solubilization Assay This assay was performed in order to assess the ability of a test compound to mobilise A ⁇ , as an example form of amyloid, from the insoluble to the soluble phase of an extract of tissue from post mortem human AD brain.
  • plaque-bearing cortex without meninges was homogenized using a DIAX 900 homogenizer (Heudolph and Co, Kelheim, Germany) or other suitable device for three
  • Tris-Tricine polyacrylamide gel electrophoresis was performed by loading appropriately diluted samples on to 10% to 20% gradient gels (Novex, San Diego, CA) followed by transfer on to 0.2- ⁇ m nitrocellulose membrane (Bio-Rad,Hercules, CA).
  • a ⁇ was detected by using monoclonal antibody W02, which detects residues 5 through 8, 17 (or another suitable antibody) in conjunction with horseradish peroxidase-conjugated rabbit anti- mouse IgG (Dako, Denmark), and visualized by using enhanced chemiluminescence (eg ECL; Amersham Life Science, Buckinghamshire, UK).
  • Each gel included three lanes containing 0.5, 1, and 2 ng of synthetic A ⁇ 40 (Keck Laboratory, Yale University, New Haven, CT) as reference standards.
  • Blot films were scanned by using a suitable imaging system such as the UVP gel documentation system, and densitometry performed using suitable software, eg UVP Labworks.
  • the dynamic range of the film/scanner was determined by using a step tablet (No. 911ST600, Kodak, Rochester NY), a calibrated film exposed by the manufacturer to provided steps of known increasing intensity.
  • the quantifiable range of signal intensity for densitometric analysis of the mono- and dimeric A ⁇ bands was based on the comparison with a curve obtained by scanning and densitometry of the step tablet. Samples in which the signal intensity is low after preliminary assay may be re-assayed by using synthetic standards of lower or higher concentration.
  • Transgenic mouse models are available for a number of neurological disorders, including Alzheimer's disease; Parkinson's disease; familial amyotrophic lateral sclerosis (ALS); Huntington's disease; and Creutzfeld- Jakob disease (CJD). It was found that one of the transgenic models for Alzheimer's disease, the APP2576 transgenic mouse also has a high incidence of cataract. These animal models were suitable for testing the methods of the invention.
  • Transgenic mice of the strain APP2576 were used. Eight to nine month old female mice were selected and divided into groups for treatment.
  • mice were sacrificed at intervals, and their brains examined to determine whether the treatment with test compounds decreased brain amyloid formation, and the identification of the most effective administration protocol.
  • mice in each group were tested over a period of up to eight months for cognitive performance, using a Morris water maze according to standard methods.
  • the general health and well-being of the animals was also measured every day by a blinded operator, using a five point integer scale which subjectively rates a combination of features, including motor activity, alertness and general health signs.
  • Turbidimetric Solubility Measurements The solubility estimate was measured at both pH 2.0 and pH 6.5. This is within the pH range that can be anticipated along the proximal gastrointestinal tract in humans.
  • Effective Log D values were measured using a chromatographic method employing a SUPELCOSIL LC-ABZ column using an octanol saturated mobile phase at pH 7.4. See F. Lombardo et al, J. Med. Chem. 2000, 43, 2922-2928.
  • Table 8 provides the properties and structures of particularly preferred PBT compounds which fall within the scope of the present invention.
  • test compound a- concentration in ⁇ M of test compound required to inhibit 50% of A Ma H 2 O 2 production
  • c- % inhibition of dityrosine oligomerization as referenced to in-house standard (set as 100% inhibition) d- extent by which test compound mobilizes A ⁇ from the insoluble to the soluble phase of an extract of tissue from a postmortem human AD brain.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Ophthalmology & Optometry (AREA)
  • Toxicology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Quinoline Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates generally to the field of treatment and prophylaxis of retinal degenerative diseases. More particularly, the present invention contemplates a method for preventing, reducing the risk of development of, or otherwise treating or ameliorating the symptoms of, age-related macular degeneration (AMD) or related retinal conditions in mammals and in particular humans. The present invention further provides therapeutic compositions enabling dose-dependent or dose-specific administration of agents useful in the treatment and prophylaxis of age-related macular degeneration or related retinal degenerative conditions.

Description

Method of treatment of age-related macular degeneration(AMD)
BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION
The present invention relates generally to the field of treatment and prophylaxis of retinal degenerative diseases. More particularly, the present invention contemplates a method for preventing, reducing the risk of development of, or otherwise treating or ameliorating the symptoms of, age-related macular degeneration (AMD) or related retinal conditions in mammals and in particular humans. The present invention further provides therapeutic compositions enabling dose-dependent or dose-specific administration of agents useful in the treatment and prophylaxis of age-related macular degeneration or related retinal degenerative conditions.
DESCRIPTION OF THE PRIOR ART
Reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in any country.
Bibliographic details of references in the subject specification are also listed at the end of the specification.
Macular degeneration is a clinical term that is used to describe a family of diseases that are all characterized by a progressive loss of central vision associated with abnormalities of
Bruch's membrane, the choroid, the neural retina and/or the retinal pigment epithelium.
These disorders include very common conditions that affect older subjects - such as AMD as well as rarer, earlier-onset dystrophies that in some cases can be detected in the first decade of life. Other maculopathies include North Carolina macular dystrophy, Sorsby's fundus dystrophy, Stargardt's disease, pattern dystrophy, Best disease and Malattia leventinese.
AMD is the leading cause of permanent vision loss for individuals over age 65, currently affecting approximately 15 million Americans, AMD affects light-sensitive photoreceptor cells and pigmented epithelial cells in the macula, the center of the retina of the eye. While it may not cause total blindness, the disease destroys central vision, making reading, watching electronic monitor screens and driving impossible. It has no documented cure, has never demonstrated spontaneous remission and effective treatments are very limited.
The retina is a complicated network of nerve cells that changes light into nerve impulses that travel to the brain where they are interpreted as visual images. The central part of the retina, called the macula, is responsible for vision that is needed for reading and other detailed work. Damage to the macula results in poor vision. The most common disease process that affects the macula is AMD. In patients with AMD, retinal photoreceptor and pigment epithelial cells in the macula die over the course of several years. The cell death and gradual visual loss usually do not begin until age 60 or older, hence the name age- related macular degeneration.
There are two types of AMD: dry macular degeneration and wet macular degeneration. Dry macular degeneration, although more common, typically results in a less severe, more gradual loss of vision. Patients who are affected by dry AMD have gradual loss of central vision due to the death of photoreceptor cells and their close associates, retinal pigmented epithelial (RPE) cells, with deposition of a complex waxy amyloid mixture, termed 'drusen'. Photoreceptors, the cells in the retina that actually 'see' light, are essential for vision. Macrophagic RPE cells are necessary for photoreceptor survival, function and renewal.
Patients with wet macular degeneration develop new blood vessels under the retina. As the photoreceptor and RPE cells slowly degenerate, there is a tendency for blood vessels to grow from their normal location in the choroid into an abnormal location beneath the retina. This abnormal new blood vessel growth is called choroidal neovascularization (CNV). The abnormal blood vessels leak and bleed, causing hemorrhage, swelling, scar tissue, and severe loss of central vision. Only 10% of patients with AMD have the wet type, but it is responsible for 90% of all blindness resulting from AMD.
The RPE cells in the eye act as macrophages, which phagocytize and recycle components of the membranous outer segments of photoreceptors. If the mitochondria within the RPE cells are damaged, the photoreceptor recycling is inhibited, with resultant accumulation of drusen. Drusen causes a lateral stretching of the RPE monolayer and physical displacement of the RPE from its immediate vascular supply, the choriocapillaris. This displacement creates a physical barrier that may impede normal metabolite and waste diffusion between the choriocapillaris and the retina.
Depending on the location, laser treatment can sometimes be given to destroy the abnormal blood vessels formed in wet AMD. Only 15% of the cases of wet AMD are eligible to have laser treatment because the blood vessels can not be located too close to the center part of the macula. The laser is a beam of light that is absorbed by the pigment of blood, drugs and RPE cells, which converts to heat energy that cauterizes the abnormal blood vessels. Frequently the neovascularization returns, since the stimulus has not been removed, resulting in severe loss of vision. In fact, most of the patients with AMD, who have very poor vision, have lost it due to sequelae of neovascularization. Current medical opinion states that there is no treatment available that permanently prevents the cell death or abnormal blood vessel growth that occurs in AMD.
To date, there are no known specific measures to prevent the occurrence of AMD. For patients already diagnozed with AMD in one or both eyes, current main treatments include light targeting (phototherapy) and/or a vitamin and mineral supplement, each of which is of debatable value. Phototherapy involves targeting light to the macular area containing the lesion of nascent defective blood vessels to inhibit or impair their function. One type of phototherapy is photodynamic therapy (PDT). In PDT, a photosensitive agent is administered into the vessels of a patient, then the agent is activated at the target site of the lesion of new vessels (the macula) by directing low energy light from a laser specifically to this area. The activated agent generates free radicals and other activated chemical species which destabilize and destroy the new vessels.
PDT has been reported to be of some benefit to patients having AMD. For example, one study, (Arch. Ophthalmol. 777:1329-1345, 1999) evaluated PDT in four hundred and two eyes from patients diagnozed with AMD in at least one eye. Treatment outcome was assessed by comparing the patient's ability to accurately read a conventional vision chart (one having about five letters per line) pre-treatment and post-treatment. At twelve months post-PDT, 61% of the eyes (246/402) lost fewer than 15 letters (that is, the patient lost less than about three lines on a standard visual chart), while 46% of the eyes (96/207) from patients undergoing treatment with a placebo lost fewer than 15 letters (p<0.001). At twenty-four months post-PDT, the visual acuity and contrast sensitivity was sustained in patients receiving PDT. A significantly greater percentage of these patients (58%) lost fewer than 15 letters, compared to patients undergoing treatment with a placebo (38%). However, only 16% of the patients receiving PDT had improved vision, compared to 7% of the patients receiving a placebo.
Another type of phototherapy is photocoagulation therapy. In photocoagulation therapy, high energy light from a laser is directed specifically to the target site of the new vessels. The heat generated from the high energy laser coagulates the fluid in and around the new vessels. Laser photocoagulation is not a form of PDT; it is a separate treatment approach. It uses lateral transfer of heat, applied with a cautery-like method, to coagulate fluid within and surrounding the vessel, while PDT uses an activated photosensitive agent to generate active chemicals which damage or destroy the new vessels containing the agent.
While either PDT or laser photocoagulation therapy is separately used to treat patients with AMD, neither is without drawbacks. A problem with PDT is that its effects are transient; patients receiving PDT must be retreated about every three months. Furthermore, the patients require at least five retreatments within the first two years merely to stabilize their condition, and before any therapeutic effect occurs. These cumulative treatments damage the retina, further reducing the patient's visual acuity. One drawback of laser photocoagulation is that it is non-selective, and does not target only the new blood vessels. It must therefore be administered so that only the lesions are targeted, and the unaffected surrounding tissues are undamaged. However, in about half of the patients with AMD, the new vessels are located in the subfoveal area, which is difficult or impossible to target with laser coagulation without damaging the sensory retina. Another drawback is that photocoagulation treatment is not permanent and recurrence rates for new vessel production are high, reaching 39-76%, usually within the first two years. However, repeated treatments can actually induce the growth of new vessels and membranes (subretinal neovascular membranes and recurrent choroidal neovascularizations) at the site of the treatment. Repeated treatments may also irreversibly damage unaffected areas of the retina, including the neurosensory retinal and RPE. Thus, the treatment itself may result in the patient having further reduced vision over a period of time. Specifically, some patients undergoing photocoagulation therapy develop scotoma, which is an area of depressed vision within the visual field, surrounded by an area of less depressed or of normal vision.
There is a need, therefore, to develop alternative methods to treat AMD or related conditions.
SUMMARY OF THE INVENTION
The present invention is predicated in part on the recent determination that proteinaceous deposits on the limiting membrane of the retina, referred to as "drusen", also comprise zinc and copper and hence are proposed to be similar to an amyloid type plaque. Hence, the present invention contemplates the use of a metal protein attenuating compound (MPAC) to reduce the levels of or otherwise remove excess metal from drusen thereby restoring normal metal homeostasis in the retina. The present invention is particularly useful for treating or preventing or otherwise reducing the risk of development of age-related macular degeneration (AMD); however, the subject invention extends to the treatment of any retinal degenerative disorder associated with amyloid type aggregates, complexes, deposits or plaques or any condition associated with drusen which comprise excess metal.
The method of the present invention is useful irrespective of any inhibition of a matrix metalloproteinase and/or a dose-specific amount of MPAC may be employed. A single agent may be administered or a combination of two or more agents.
The present agents comprise at least two fused 6-membered rings with at least a nitrogen atom at position 1 and a hydroxy or mercapto group at position 8. Useful compounds are defined by Formulae I through XXVII which are described in detail below.
Examples of suitable compounds include those in Table 8 such as PB-1033, PB-1076, PB- 1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically acceptable salt or derivative or functional equivalent thereof.
Hence, one aspect of the present invention contemplates a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue.
The present invention also provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue.
In particular, the present invention provides a method for treating a subject with age- related macular degeneration (AMD) said method comprising administering to said subject an amount of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof effective to reduce metal in retinal drusen to a level which ameliorates symptoms of AMD .
The phrases "alter metal levels" and "reduce metal" is used in its broadest sense and refers to a change in the distribution of a metal in retinal drusen or surrounding tissue as well as a change in the amount or activity of metal in drusen or surrounding tissue. The phrases also refer to a reduction in the amount or activity of metal in retinal drusen or surrounding tissue as well as a reduction in the amount or activity of metal in particular areas i.e. the distribution of metal in retinal drusen or surrounding tissue.
The selection of an MPAC is generally but not exclusively irrespective of its ability to inhibit a metalloproteinase. A defined or specific dosage amount may also be administered.
Accordingly, another aspect of the present invention provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue, irrespective of any effect on a matrix metalloproteinase.
Reference to "irrespective" means that one or more metalloproteinases may be inhibited or no metalloproteinases are inhibited. Still another aspect of the present invention defines a specific dosage range to optimally restore metal homeostasis in the retina.
Hence, this aspect of the present invention is directed to a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue wherein the effective amount is a specific dose range to optimally restore metal homeostasis in the retina.
Another aspect of the present invention contemplates a method for reducing levels of a metal from retinal drusen in a subject to thereby ameliorate symptoms of age-related macular degeneration (AMD) said method comprising administering to said subject an effective amount of PB- 1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof.
The present invention also provides for the use of an MPAC in the manufacture of a medicament for the treatment of a retinal degenerative disorder in a subject.
In particular, the present invention contemplates the use of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof in the manufacture of a medicament for the treatment of age-related macular degeneration (AMD) in a subject.
Combination therapy also forms part of the present invention in which two or more MPACs are administered or an MPAC and another active such as a metal chelator, cytokine, genetic molecule anti-microbial or anti-viral agent, an anti-oxidant, an antibiotic and/or an anasthetic.
The preferred subject is a human although the present invention has application in the veterinary, horse racing and animal husbandry industries.
The present invention further provides formulations for treating, preventing or reducing the risk of developing a retinal degenerative condition or disorder comprising an MPAC as herein described.
Whilst PB- 1033 is a particularly useful MPAC5 the present invention extends to any MPAC encompassed by the compounds of Formulae I through XXVII such as but not limited to those in Table 8 including PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof.
Abbreviations used herein are defined in Table 1.
TABLE 1 - ABBREVIATIONS
Figure imgf000010_0001
DETAILED DESCRIPTION OF THE INVENTION
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", were understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
All scientific citations, patents, patent applications and manufacturer's technical specifications referred to hereinafter are incorporated herein by reference in their entirety.
Reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in any country.
It is to be understood that unless otherwise indicated, the subject invention is not limited to specific formulation components, manufacturing methods, biological materials or reagents, dosage regimens and the like, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
As used in the subject specification, the singular forms "a", "an" and "the" include plural aspects unless the context clearly dictates otherwise. Thus, for example, reference to "a formulation" includes a single formulation, as well as two or more formulations; reference to "an agent" or "a reagent" includes a single agent or reagent, as well as two or more agents or reagents; and so forth.
The terms "agent", "reagent", "compound", "pharmacologically active agent", "medicament", "therapeutic", "active" and "drug" are used interchangeably herein to refer to a chemical or biological entity which induces or exhibits a desired effect such as ameliorating the symptoms of a retinal degenerative disease. The terms also encompass pharmaceutically acceptable and pharmacologically active ingredients of those active agents specifically mentioned herein. When the terms "agent", "reagent", "compound", "pharmacologically active agent", "medicament", "therapeutic", active" and "drug" are used, then it is to be understood that this includes the active entity per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, prodrugs, metabolites, analogs, etc.
Reference to an "agent", "chemical agent", "compound", "pharmacologically active agent", "medicament", "therapeutic", "active" and "drug" includes combinations of two or more active agents. A "combination" also includes multi-part such as a two-part composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispenzation. For example, a multi-part pharmaceutical pack may have two or more agents separately maintained. Hence, this aspect of the present invention includes combination therapy. Combination therapy includes the co-administration of a metal chelator and another active such as a chemical compound, cytokine, genetic molecule, anti-microbial or anti-viral agent, an antibiotic and/or an anasthetic.
The terms "effective amount" and "therapeutically effective amount" of an agent as used herein mean a sufficient amount of the agent to provide the desired therapeutic or physiological or effect or outcome. Such an effect or outcome includes altering or reducing availability of metal ions and/or reducing their amount in drusen, reducing amyloid levels reducing or preventing macular degeneration or a related condition and/or treating or preventing vision impairment. Undesirable effects, e.g. side effects, are sometimes manifested along with the desired therapeutic effect; hence, a practitioner balances the potential benefits against the potential risks in determining what is an appropriate "effective amount". The exact amount required will vary from subject to subject, depending on the species, age and general condition of the subject, mode of administration and the like. Thus, it may not be possible to specify an exact "effective amount". However, an appropriate "effective amount" in any individual case may be determined by one of ordinary skill in the art using only routine experimentation.
The effective amount is deemed the amount required to prevent or ameliorate symptoms of the retinal degeneration condition such as AMD. In one embodiment, the amount of MPAC used is the amount required to or which is effective in reducing levels of metal drusen. Examples of metals include zinc and copper. Effective amounts include from 1 ng/ml to 1000 mg/ml such as from about 5 ng/ml to about 500 mg/ml or about 10 ng/ml to about 100 mg/ml or amounts or ranges in between.
The terms "metal" and "metal ion: may be used interchangedly in this context.
By "pharmaceutically acceptable" carrier, excipient or diluent is meant a pharmaceutical vehicle comprized of a material that is not biologically or otherwise undesirable, i.e. the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction. Carriers may include excipients and other additives such as diluents, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives, and the like.
Similarly, a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
"Treating" a subject may involve prevention of a retinal degenerative condition or other adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by ameliorating the symptoms of the condition. In particular, the presence invention contemplates a reduction of amyloid type plaque formation and/or a reduction in metal content in drusen to restore normal metal homeostasis in the retina.
The "subject" as used herein refers to an animal, preferably a mammal and more preferably a primate including a lower primate and even more preferably a human who can benefit from the formulations and methods of the present invention. A subject regardless of whether a human or non-human animal may be referred to as an individual, patient, animal, host or recipient. The compounds and methods of the present invention have applications in human medicine, veterinary medicine as well as in general, domestic or wild animal husbandry. For convenience, an "animal" includes an avian species such as a poultry bird (including ducks, chicken, turkeys and geese), an aviary bird or game bird.
The condition in a non-human animal may not be a naturally occurring but induced such as in an animal model.
As indicated above, the preferred animals are humans, non-human primates such as marmosets, baboons, orang-utans, lower primates such as tupia, livestock animals, laboratory test animals, companion animals or captive wild animals. A human is the most preferred target. However, non-human animal models may be used.
Examples of laboratory test animals include mice, rats, rabbits, guinea pigs and hamsters. Rabbits and rodent animals, such as rats and mice, provide a convenient test system or animal model as do primates and lower primates. Livestock animals include sheep, cows, pigs, goats, horses and donkeys. Non-mammalian animals such as avian species, zebrafish, amphibians (including cane toads) and Drosophila species such as Drosophila melanogaster are also contemplated. Instead of a live animal model, a test system may also comprise a tissue culture system.
A "retinal degenerative condition" is a condition which is characterized by a progressive loss of vision. Conditions within the scope of this term include age-related macular degeneration (AMD), North Carolina macular dystrophy, Sorsby's fudus dystrosphy, Stargardt's disease, pattern dystrophy, Best disease and Malattia leventinese.
A particular condition for which the agents and the methods of the present invention can be effective is AMD. However, the present invention extends to any retinal degenerative disease associated with or characterized by amyloid like aggregates, deposits or plaques.
Hence, one aspect of the present invention contemplates a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue. In one embodiment, the altered metal levels are reduced metal levels.
In another embodiment, the present invention provides a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to alter metal levels in retinal drusen or surrounding tissue, irrespective of any effect on a matrix metalloproteinase. In one embodiment, the altered metal levels are reduced metal levels.
Still a further aspect of the present invention is directed to a method for the treatment or prophylaxis of a retinal degenerative condition or disorder in a subject, said method comprising administering to said subject an effective amount of an MPAC or formulation comprising an MPAC for a time and under conditions effective to reduce metal levels in retinal drusen or surrounding tissue wherein the effective amount is a specific dose range to optimally restore metal homeostasis in the retina.
The present invention also provides for the use of an MPAC in the manufacture of a medicament for the treatment of a retinal degenerative disorder in a subject.
The preferred agents of the present invention comprise at least two fused 6-membered rings with at least a nitrogen atom at position 1 and a hydroxy or mercapto group at position 8. The agents of the present invention are collectively referred to as metal protein attenuating compounds or MPACs and have one or more of the following properties: act as ionophores (i.e. capture and transfer metals into cells), is a metal binder, crosses the blood brain barrier (BBB), exhibits reduced cell toxicity, is capable of dissolving or disrupting amyloid type protein deposits, aggregates or plaques and is stable in aqueous environments. ] Preferably, the agents have two or more, three or more or four or more or five or more ] of the above-listed properties.
Particularly useful compounds, defined further below, include those in Table 8 such as PB- 1033, PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof. PB- 1033 is particularly useful although the present invention is not to be so limited. In this regard, the present invention further contemplates a method for treating a subject with age-related macular degeneration (AMD) said method comprising administering to said subject an amount of PB-1033 or a pharmaceutically acceptable salt, derivative or functional equivalent thereof effective to reduce metal in retinal drusen to a level which ameliorates symptoms of AMD.
The subject invention also provides a method for reducing levels of a metal from retinal drusen in a subject to thereby ameliorate symptoms of age-related macular degeneration (AMD) said method comprising administering to said subject an effective amount of PB- 1033 or a pharmaceutically acceptable salt, derivative or equivalent thereof.
Examples of pharmaceutically acceptable chemical derivatives or functional equivalents of PB-1033 include those in Table 8 such as PB-1076, PB-1085, PB-1120, PB-1127, PB- 1135, PB-1149, PB-1151, PB-1160 and PB-1168 or a pharmaceutically salt or derivative or functional equivalent thereof. Examples of metals include zinc and copper.
Hence, certain useful agents of the present invention are encompassed by compounds of
Formula I:
Figure imgf000016_0001
in which:
R is O or S;
R1 is independently selected from H3 optionally substituted alkyl, optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; CN; halo; CF3; SO3H; and OR2, SR2, SOR2, SO2R2, NR2R3, (CH2)nNR2R3, HCNOR2, HCNNR2R3, CONR2R3, CSNR2R3, NCOR2, NCSR2, COR2, CO2R2 , CSR2 or SO2NR2R3 in which R2 and R3 are independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety and n is an integer of 1 to 10;
X is independently selected from CH, CO, N and NH;
Z is independently selected from CH, CO, N, NH and O;
Y is independently absent or together with the ring to which it is attached forms a 5- or 6- membered optionally substituted aryl or a 5- or 6-membered optionally substituted heterocyclyl;
m is an integer from 1 to 3; and
p is an integer from 1 to 4,
salts, hydrates, solvates, derivatives, pro-drugs, tautomers and/or isomers thereof to a subject in need thereof,
with the provisos that:
(i) at least one of X and Z is other than CH; and
(ii) phanquinone or tautomers thereof are excluded i.e., when R is O, R1 at position 7 is OH, X is CH and Y is absent, then Z is not
Figure imgf000017_0001
Preferably, R is O. In addition, R1 is preferably halo, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkyl, OR2, SR2, (CH2)nNR2R3, CONR2R3 and NCOR2 in which n, R2 and. R3 are as defined above. More preferably R1 is fluorine; iodine; chlorine; optionally substituted phenyl such as 4-halophenyl, for example, 4-fluorophenyl or 4-chlorophenyl; an optionally substituted unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolyl or pyridinyl; an optionally substituted saturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolidinyl or piperazinyl; an optionally substituted saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl; optionally substituted Ci-4 alkyl such as methyl or ethyl; optionally substituted C2-6 cycloalkyl such as cyclopropyl; optionally substituted Ci-6 alkoxy; optionally substituted thio; CH2NR4R5 in which R4 and R5 are independently selected from H and C1-4 alkyl; or CONH(CH2)2R6 in which R6 is optionally substituted heterocyclyl.
Y is preferably an optionally substituted phenyl; an optionally substituted unsaturated 5- or 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolyl or pyridinyl; or an optionally substituted saturated 5 or 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl.
The preferred halo group is chlorine but other halogen atoms are encompassed by the present invention.
Illustrative classes of compounds of Formula I are as follows:
8-hydroxy-4(3H)-quinazolinones
8-hydroxy-quinazoline
8-hydroxy-quinoxaline
[ 1 ,6]naphthyridin-8-ol
9-hydroxypyrimido[l,6-a]pyrimidin-4-one
Figure imgf000019_0001
Figure imgf000019_0002
4-hydroxy-acridine
Figure imgf000019_0003
4,7(4, 10)-phenanthrolin-5-ol
Figure imgf000020_0001
9-hydroxypyrido[l,2-a]pyrimidin-4-one
Figure imgf000020_0002
pyrido [3 ,2-d]pyrimidin-4-ol
pyrido[2-3-d]pyridazin-8-ol
[l,7]naphthyridin-8-ol
[ 1 ,5]naphthyridine-4,8-diol
[ 1 ,5]naphthyridine-8-ol
pyrido [3 ,4-b]pyrazin-8-ol
pyrido [3 ,4-b]pyrazin-5 -ol
pyridol [4,3 -d]pyrimidin- 8-ol
Figure imgf000021_0001
4-hydroxy-4a,8a-dihydro-pyrano[3,2,b]pyridin-2-one
Figure imgf000022_0001
Figure imgf000022_0003
8-hydroxy-6H-[ 1 ,6]naphthyrin-5-one
dibenzo[a,g]quinolizin-8-one
4-hydroxy-l/f-pyrido[3,2-d]pyridin-2-one
Figure imgf000022_0002
in which R , m, n and p are as defined above and q is an integer of 1 or 2.
The above compounds also form part of more generic groups of compounds such as those encompassed by Formula II:
Figure imgf000023_0001
in which
R1 is H or halo, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety;
R2 is H; optionally substituted alkyl; optionally substituted alkenyl; optionally substituted aryl; optionally substituted heterocyclyl; optionally substituted alkoxy; an antioxidant; a targeting moiety; COR6 or CSR6 in which R is H, optionally substituted alkyl, optionally substituted alkenyl, hydroxy, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant, a targeting moiety, OR7, SR7 or NR7R8 in which R7 and R8 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; CN; (CH2)nNR9R10, HCNOR9 or HCNNR9R10 in which R9 and R10 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl and n is 1 to 4; OR11, SR11 or NR11R12 in which R11 and R12 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl or together form optionally substituted heterocyclyl; or SO2NR13R14 in which R13 and R14 are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; and
R3, R4, R5, R and R are either the same or different and selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, hydroxy, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, halo, SO3H, amine, CN, CF3, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety,
salts, hydrates, solvates, derivatives, pro-drugs, tautomers and/or isomers thereof with the provisos that:
(a) when R1 to R3, R and R are H, then R4 is not Cl or I and R5 is not I; (b) when R1 to R3, R, R ' and R5 are H, then R4 is not CHO, CHOHCCl3,
'
Figure imgf000024_0001
Figure imgf000025_0001
(c) when R1, R5, R' and R are H, R2 is CO2H and R3 is OH, then R4 is not bromo, methyl, phenyl, hydroxymethyl or trifluoromethyl;
(d) when R1, R4, R5 and R are H, R2 is CO2H and R3 is OH, then R' is not bromo, iodo, methyl, phenyl, propyl, phenethyl, heptyl, benzylaminomethyl, 3-aminopropyl, 3- hydroxypropyl, 4-methoxyphenyl, 3-methylphenyl, 4-chlorophenyl, 3,4-dichloroρhenyl, pyridin-3-yl, furo-2-yl, 4-chlorophenyl, 3,4-dichlorophenyl, 2-chlorophenyl, 3- chlorophenyl, 2-chlorophenyl, 3-chlorophenyl, 2-methoxyphenyl or piperidin-2-yl;
(e) when R1, R4, R and R' are H, R2 is CO2H and R3 is OH, then R5 is not phenyl, 3- hydroxypropyl, phenethyl, 3-aminoprop-l-yl or hex-l-yl;
(f) when R1, R4, R1 and R5 are H3 R2 is CO2H and R3 is OH, then R is not N- morpholinomethyl, bromo or phenyl;
(g) when R1, R and R1 are H, R2 is CO2H and R3 is OH, then R4 and R5 are not chloro;
(h) when R1, R4 and R' are H, R2 is CO2H and R3 is OH, then R and R5 are not bromo;
(i) when R1, R, R' and R5 are H, R2 is CO2Me and R3 is OH, then R4 is not hydroxymethyl, phenyl or bromo;
O) when R1, R, R4 and R5 are H, R2 is CO2Me and R3 is OH, then R1 is not 4- methoxyphenyl, 3-methylphenyl, pyridin-3-yl, benzyl, bromo, 4-chlorophenyl, 3,4- dichlorophenyl, 3-hydroxypropyl or 3-tert-butoxycarbonylaminopropyl; (k) when R1, R, R4 and R1 are H, R2 is CO2Me and R3 is OH, then R5 is not phenyl or 3-tert-butoxycarbonylaminoprop- 1 -yl;
(1) when R1, R5 R4, R1 and R5 are H and R2 is CO2Me5 then R3 is not toluene-4- sulphonylamino, piperazin-1-yl, morpholin-1-yl, piperidin-1-yl, 4-methylpiperazin-l-yl, 3- benzoylaminoprop-1-yl, phenethyl, 3-tert-butoxycarbonylaminopropyl, 3-hydroxypropyl, amino or hex-1-yl;
(m) when R1 , R4, R' and R5 are H, R2 is CO2Na and R3 is OH, then R is not phenyl;
(n) when R1, R, R4, R' and R5 are H and R2 is CO2H, then R3 is not phenyl, 4- chlorophenyl, phenethyl, 3-hydroxypropyl, amino, morpholin-1-yl, piperidin-1-yl, 4- methylpiperazin-1-yl, toluene-4-sulphonylamino, 3-benzoylaminoprop-l-yl, aminoprop-1- ynyl, hex- 1 -yl, 5-hydroxypent- 1 -yl, piperazin- 1 -yl or 2-( 1 -piperazinyl)pyrimidinyl;
(o) when R1, R' and R are H, R2 is CO2Me and R3 is OH, then R4 and R5 are not chloro;
(p) when R1, R4, R' and R5 are H, R2 is CO2Me and R3 is OH, then R is not bromo;
(q) when R1, R1 and R4 are H, R2 is CO2Me and R3 is OH, then R and R5 are not bromo;
(r) when R1, R, R3, R1 and R5 are H and R2 is CO2H, then R4 is not phenyl, 4- chlorophenyl or phenylethyl;
(s) when R1, R5, R1, R4, R3 and R are H, then R2 is not 2H-tetrazol-l-yl;
(t) when R1, R5, R4 and R are H, R2 is CO2H and R3 is OH, then R' is not 3,5- dichlorophenyl or 4-fluorophenyl; and (u) at least one of R . 1 + toΛ τ R> 5 , R and R1 is other than H.
Useful compounds of Formula II are as follows:
(i) Formula III
Figure imgf000027_0001
in which:
R, R1 and R3 are as defined in Formula II above; and
R a is H; optionally substituted C1-6 alkyl; optionally substituted C1-6 alkenyl; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; COR6 a or CSR6 a in which R6 a is H, optionally substituted Cj-6 alkyl, optionally substituted C2-6 alkenyl, hydroxy, optionally substituted aryl, optionally substituted heterocyclyl or OR a, SR a or NR aR a in which R 8 and R a are either the same or different and selected from H, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; CN; CH2NR9 aR10 a, HCNOR9 a or HCNNR9 aR10 in which R9 a and R10 a are either the same or different and selected from H, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl; ORn a, SRu a or NRπ aR12 a in which Rπ a and R12 a are either the same or different and selected from H, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl or together form optionally substituted heterocyclyl; or SO2NR13 aRI4 a in which R13 a and R14 a are either the same or different and selected from H or optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
Preferred compounds of Formula III are as follows:
• Formula IV
Figure imgf000028_0001
in which:
R1 is as defined in Formula II above; and
R2'a is optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
Formula IV may represent compounds in which an antioxidant moiety is attached to the C2 position of the 8-hydroxyquinoline in such a way that exposure to a prooxidative environment, that is, hydroxy radicals, will result in a molecule with enhanced metal binding properties.
Representative examples are shown below:
Figure imgf000029_0001
Foπnula V
Figure imgf000029_0002
in which:
R1 and R3 are as defined in Formula II above; and
R a is optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, hydroxy, 0R7 a',
SRV, N2R7'aR8'a, or NR7'a R8' a in which R7'a and R8' a are either the same or different and selected from H, optionally substituted Ci-6 alkyl, optionally substituted aryl or optionally substituted heterocyclyl.
Formula V represents compounds in which a hydrophilic amide moiety is attached to the C2 position of the 8-hydroxyquinoline so as to generally enhance solubility while maintaining membrane permeability.
Representative examples are shown below:
Figure imgf000031_0001
Formula VI
Figure imgf000032_0001
in which:
R1 is as defined in Formula II above; and
R2"a is CN; CH2NR9'aR10' a, HCNOR9' a or HCNNR9' aR10'a in which R9' a and R10'a are either the same or different and selected from H, optionally substituted C1-6 alkyl, optionally substituted alkenyl, optionally substituted aryl or optionally substituted heterocyclyl.
Representative examples are shown below:
Figure imgf000032_0002
• Formula VII
Figure imgf000033_0001
in which:
R1 is as defined in Formula II above; and
R1V and R12 a' are either the same or different and selected from H, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl and optionally substituted heterocyclyl or together form optionally substituted heterocyclyl.
• Formula VIII
Figure imgf000033_0002
in which:
R1 is as defined in Formula II above; and
R1V and R1V are either the same or different and selected from H, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted aryl or optionally substituted heterocyclyl. (ii) Formula IX
Figure imgf000034_0001
in which:
R1, R ' , R, R2 and R3 are as defined in Formula II above;
R4 b and R5 b are either the same or different and selected from H; optionally substituted Ci-6 alkyl; optionally substituted C2-6 alkenyl; halo; CN; CF3; optionally substituted aryl; optionally substituted heterocyclyl; an antioxidant; a targeting moiety; SO3H; SO2NR13 aR14 a in which R13 a and R14 a are as defined in Formula III above; or OR15 b, SR15 b, SO2R15 b, CONR15 bR16 b or NR15 bR16 b in which R15 b and R16 b are either the same or different and selected from H, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted Ci-6 acyl, optionally substituted aryl or optionally substituted heterocyclyl,
including provisos (a) to (c), (e), (g), (h), (I), (k), (o), (q), (r), and (u) as defined above.
Useful compounds of Formula IX are as follows: • Formula X
Figure imgf000035_0001
in which:
R1, R 1 , R5 R and R are as defined in Formula II above; and
R4b' and R5 a are as defined in Formula IX above provided that at least one is halo,
including provisos (a), (c), (g), (h), (i), (o), (q) and (u) defined above.
• Formula XI
Figure imgf000035_0002
in which:
R is as defined in Formula II above;
R >4b is H or halo; and
R > 5 b " is optionally substituted aryl or optionally substituted heterocyclyl. A representative example is shown below.
Formula XII
Figure imgf000036_0002
in which:
R1 is as defined in Formula II above;
R" is C1-6 alkoxy, halo, C1-6 alkyl, C2-6 alkenyl or Cj-6 haloalkyl; and
R b is H or halo. A representative example is shown below.
Figure imgf000037_0001
• Formula XIII
Figure imgf000037_0002
in which
R1 is as defined in Formula II above; and
R is as defined in Formula XIII above. • Formula XIV
Figure imgf000037_0003
in which:
R >2 i toΛ R r.5 , R and R1 are as defined in Formula II above; and
R b is optionally substituted Ci-6 alkyl, optionally substituted aryl, optionally substituted aryl acyl, Cj-6 alkyl acyl or optionally substituted heterocyclyl.
(iii) Formula XV
Figure imgf000038_0001
in which:
R1, R2, R3, R and R' are as defined in Formula II; and
at least one of R4 C and R5 C is halo and the other is selected from H5 optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, hydroxy, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, SO3H, amine, CN, CF3, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant and a targeting moiety,
salts, hydrates, solvates, derivatives, pro-drugs, tautomers and/or isomers thereof
with the provisos that: (a) when R1 to R3, R and R1 are H, then R4 C is not chloro or iodo and R5 C is not iodo;
(b) when R1, R5 C, R1 and R are H, R2 is CO2H and R3 is OH3 then R4 C is not bromo;
(c) when R1, R and R' are H, R2 is CO2H and R3 is OH, then R4 C and R5 C are not chloro;
(d) when R1, R4 C and R1 are H, R2 is CO2H or CO2Me and R3 is OH, then R and R5 C are not bromo;
(e) when R1, R, R' and R5 C are H5 R2 is CO2Me and R3 is OH, then R4 C is not bromo; and
(f) when R1, R and R' are H, R2 is CO2Me and R3 is OH, then R4 C and R5 C are not chloro.
A preferred compound of Formula XV is as follows:
Formula XVI
Figure imgf000040_0001
in which:
R >22, R D, r R> ι', τ R>44 C a „„nd j τ R> 5c are as defined in Formula XVI; and
R3' is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkoxy, optionally substituted acyl, optionally substituted amino, optionally substituted thio, optionally substituted sulphonyl, optionally substituted sulphinyl, optionally substituted sulphonylamino, halo, SO3H, amine, CN, CF3, optionally substituted aryl, optionally substituted heterocyclyl, an antioxidant or a targeting moiety,
with the proviso that at least one of R, R2 and R3' is other than H.
Representative examples are shown below:
Figure imgf000040_0002
in which:
R1 is as defined in Formula II and R4 C is as defined in Formula XV; and R5 c" is optionally substituted aryl or optionally substituted heterocyclyl;
Formula XVIII
Figure imgf000041_0001
in which:
R is as defined in Formula II, R c is as defined in Formula XV and R" is as defined in Formula XII; and
Formula XIX
Figure imgf000042_0001
in which:
R »25 rR>35R and R' are as defined in Foπnula H5 R0 and R0 are as defined in Formula XV and RJb is as defined in Formula XII.
Other examples of compounds contemplated herein include:
Figure imgf000043_0001
Figure imgf000043_0002
Figure imgf000044_0001
The present invention also provides a compound of Formula XX which is a compound of Formula II with the provisos that:
(a) when R1 and R3 to R5, R and R ' are H, then R2 is not H, methyl,
Figure imgf000044_0002
CO2H5 CN, CONCH2CO2H, COCH3, CH2NH2, CNOH, (pyrid-2-yl), 2- hydroxyphenyl, CHNNH2, NH-(pyrid-2-yl),
Figure imgf000044_0003
(b) when R1 and R4 to R7 are H, then R3 is not OH and R2 is not CO2H;
(c) when R1 to R3, R6 and R7 are H, then (i) when R5 is I, R4 is not Cl, SO3H or I; (ii) when R5 is H, R4 is not SO3H, NH2 or Cl; (iii) R4 and R5 are both not Cl, Br or CH3; arid (iv) when R2 to R7 are H, then R1 is not or
Figure imgf000045_0001
Figure imgf000045_0002
(d) when Rl to R3, R and R' are H, then R4 is not Cl or I and R5 is not I;
(e) when Rl to R >3\ ϋ R, T R>l1 a onnΛd O R5D are H, then R4 is not CHO, CHOHCCl3,
Figure imgf000045_0003
Figure imgf000045_0004
(f) when R1, R5, R ' and R are H, R2 is CO2H and R3 is OH, then R4 is not bromo, methyl, phenyl, hydroxymethyl or trifluoromethyl; (g) when R1, R4, R5 and R are H, R2 is CO2H and R3 is OH5 then R ' is not bromo, iodo, methyl, phenyl, propyl, phenethyl, heptyl, benzylaminomethyl, 3-aminopropyl, 3- hydroxypropyl, 4-methoxyphenyl, 3-methylphenyl, 4-chlorophenyl, 3,4-dichlorophenyl, pyridin-3-yl, furo-2-yl, 4-chlorophenyl, 3,4-dichlorophenyl, 2-chlorophenyl, 3- chlorophenyl, 2-chlorophenyl, 3-chlorophenyl, 2-methoxyphenyl or piperidin-2-yl;
(h) when R1, R4, R and R • are H, R2 is CO2H and R3 is OH, then R5 is not phenyl, 3- hydroxypropyly phenethyl, 3-aminoprop-l-yl or hex-l-yl;
(i) when R1, R4, R ' and R5 are H, R2 is CO2H and R3 is OH, then R is not N- morpholinomethyl, bromo or phenyl;
Q) when R1, R and R ' are H, R2 is CO2H and R3 is OH, then R4 and R5 are not chloro;
(k) when R1, R4 and R ' are H, R2 is CO2H and R3 is OH, then R and R5 are not bromo;
(1) when R1, R, R 1 and R5 are H, R2 is CO2Me and R3 is OH, then R4 is not hydroxymethyl, phenyl or bromo;
(m) when R1, R, R4 and R5 are H, R2 is CO2Me and R3 is OH, then R1 is not 4- methoxyphenyl, 3-methylphenyl, pyridin-3-yl, benzyl, bromo, 4-chlorophenyl, 3,4- dichlorophenyl, 3-hydroxypropyl or 3-tert-butoxycarbonylaminopropyl;
(n) when R1, R, R4 and R' are H, R2 is CO2Me and R3 is OH, then R5 is not phenyl or 3-tert-butoxycarbonylaminoprop- 1 -yl;
(o) when R1, R, R4, R' and R5 are H and R2 is CO2Me, then R3 is not toluene-4- sulphonylamino, piperazin-1-yl, morpholin-1-yl, piperidin-1-yl, 4-methylpiperazin-l-yl, 3- benzoylaminoprop-1-yl, phenethyl, 3-tert-butoxycarbonylaminopropyl, 3-hydroxypropyl, amino or hex-l-yl; (p) when R1, R4, R1 and R5 are H, R2 is CO2Na and R3 is OH, then R is not phenyl;
(q) when R1, R, R4, R' and R5 are H and R2 is CO2H, then R3 is not phenyl, A- chlorophenyl, phenethyl, 3-hydroxypropyl, amino, morpholin-1-yl, piperidin-1-yl, A- methylpiperazin-1-yl, toluene-4-sulphonylamino, 3-benzoylaminoprop-l-yl, aminoprop-1- ynyl, hex-l-yl, 5-hydroxypent-l-yl, piperazin-1-yl or 2-(l-piperazinyl)pyrimidinyl;
(r) when R1, R and R are H, R2 is CO2Me and R3 is OH, then R4 and R5 are not chloro;
(s) when R1, R4, R' and R5 are H, R2 is CO2Me and R3 is OH, then R is not bromo;
(t) when R1, R' and R4 are H, R2 is CO2Me and R3 is OH, then R and R5 are not bromo;
(u) when R1, R, R3, R' and R5 are H and R2 is CO2H, then R4 is not phenyl, A- chlorophenyl or phenylethyl;
(v) hen R1, R5, R', R4, R3 and R are H, then R2 is not 2H-tetrazol- 1 -yl;
(w) when R1, R5, R4 and R are H, R2 is CO2H and R3 is OH, then R' is not 3,5- dichlorophenyl or 4-fluorophenyl; and
(x) at least one of R1 to R5, R and R1 is other than H;
(y) when R1 to R3, R5, R' and R are H, then R4 is not chloro, NH2 or SO3H; and
(z) when R1, R3 to R5, R and R' are H, then R2 is not CH3.
Preferably, the invention provides a compound of Formula Ic, with the additional provisos that: (g) when R ι l + to~ D R 3 , R and R1 are H, then R c and R 0 are both not chloro or bromo; and
(h) when R ι lJ + to« D R 3J, r R> 5o, R and R' are H, then R% is not chloro.
Particularly preferred compounds include a series of so called "PB" (or PBT) compounds, some of which are referred to above, such as:
Figure imgf000048_0001
Figure imgf000048_0002
Figure imgf000048_0003
Figure imgf000048_0004
Figure imgf000049_0001
Figure imgf000049_0002
Figure imgf000049_0003
Figure imgf000049_0004
Figure imgf000049_0005
Figure imgf000049_0006
Figure imgf000050_0001
Figure imgf000050_0002
Figure imgf000050_0003
Figure imgf000050_0004
Figure imgf000050_0005
Figure imgf000050_0006
Figure imgf000051_0001
Figure imgf000051_0002
Figure imgf000051_0003
Figure imgf000051_0004
Figure imgf000051_0005
Figure imgf000051_0006
Figure imgf000052_0001
Figure imgf000052_0002
Figure imgf000052_0008
Figure imgf000052_0003
Figure imgf000052_0004
Figure imgf000052_0005
Figure imgf000052_0006
Figure imgf000052_0007
Figure imgf000053_0001
Figure imgf000053_0002
Figure imgf000053_0003
Figure imgf000053_0004
Figure imgf000053_0005
Figure imgf000053_0006
Figure imgf000054_0001
Figure imgf000054_0002
The 8-hydroxyl or 8-mercapto group on the above compounds may be blocked to form a prodrug, in particular an ester prodrug. The 8-hydroxy or 8-mercapto represents a principal site of metabolism for the above compounds: conjugation with glucuronic acid or sulphate gives a hydrophilic species ready to be excreted.
Other useful compounds include a compound of Formula XXI:
Figure imgf000055_0001
in which:
R, R1 and m are as defined for Formula I;
W is CH, N or NH;
U is CH5 CO or N; and
Y ' , together with the ring to which it is attached forms a 6 membered N-containing optionally substituted heterocyclyl.
Preferred compounds of Formula XXI are as follows:
(i) Formula XXII
Figure imgf000055_0002
in which R5 R1, m and q are as for Formula I.
Preferably R1 is located at positions 2, 3, 5 and/or 7 and is selected from halo, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkyl and (CH2)nNR2R3 in which n, R2 and R3 are as defined above. More preferably R1 is chlorine, optionally substituted phenyl, C2-6 cycloalkyl, CH2NR4R5 in which R4 and R5 are independently selected from H and C1-4 alkyl or optionally substituted pyridinyl.
Particularly examples are shown below.
Figure imgf000056_0001
1055 1061
Figure imgf000056_0002
(ii) Formula XXIII
Figure imgf000056_0003
in which R, R1, m and q are as defined for Formula I.
R1 may be is located at positions 2, 4, 5 and/or 7 and is selected from halo and optionally substituted heterocyclyl. Preferably, R1 is chloro and/or morpholinyl. Preferred examples are shown below.
Figure imgf000057_0001
(iii) Formula XXIV
Figure imgf000057_0002
in which R, R1, m and q are as defined for Formula I.
Preferably R1 is located at positions 2, 5 and/or 7 and is selected from halo and CH2NR4R5 in which R4 and R5 are independently selected from H and Ci-4 alkyl.
Useful examples are shown below.
Figure imgf000058_0001
Figure imgf000058_0002
(iv) Formula XXV
Figure imgf000058_0003
in which R, R1, m and q are as defined for Formula I.
Preferably R1 is located at positions 2 and/or 7 and is selected from optionally substituted heterocyclyl, CO2R2, (CH2)nNR2R3 and CONR2R3 in which n, R2 and R3 are as defined in Formula I.
Preferred examples are shown below.
Figure imgf000059_0001
Figure imgf000059_0002
(v) Formula XXVI
Figure imgf000059_0003
in which R, R1, m and q are as defined for Formula I. Preferably R1 is located at positions 2, 3, 6 and/or 7 and is selected from halo, optionally substituted aryl and (CH2)nNR2R3 in which n, R2 and R3 are as defined for Formula I.
Preferred examples are shown below.
Figure imgf000060_0001
(vi) Formula XXVII
Figure imgf000061_0001
in which R1 and m are as defined for Formula I.
Preferably R1 is located at positions 2 and/or 7 and is selected from halo and (CH2)nNR2 2Rr>3 in which n, R and R are as defined above.
Useful examples are shown below.
Figure imgf000061_0002
Reference to the above listed compounds includes their pharmaceutically acceptable salts and isomers.
The term "alkyl" used either alone or in compound words such as "optionally substituted alkyl" or "alkylamino" refers to straight chain, branched chain or cyclic hydrocarbon groups having from 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 4 carbon atoms. Illustrative of such alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, neopentyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. Preferred alkyl groups are C1-4 alkyl such as methyl or ethyl and C2-6 cycloalkyl such as cyclopropyl. The term "alkenyl" used either alone or in compound words such as "optionally substituted alkenyl", denotes linear, branched or mono- or poly-cyclic radicals having at least one carbon-carbon double bond of 2 to 20 carbon atoms, preferably 2 to 14 carbon atoms, more preferably 2 to 6 carbon atoms. Examples of alkenyl radicals include allyl, ethenyl, propenyl, butenyl, iso-butenyl, 3-methyl-2-butenyl, 1-pentenyl, cyclopentenyl, 1-methyl- cyclopentenyl, 1-hexenyl, 3-hexenyl, cyclohexenyl, 1-heptenyl, 3-heptenyl, 1-octenyl, cyclooctenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 1-decenyl, 3-decenyl, l,3rbutadienyl, 1,4- pentadienyl, 1,3-cyclopentadienyl, 1,3-hexadienyl, 1 ,4-hexadienyl, 1,3-cyclohexadienyl, 1 ,4-cyclohexadienyl, 1 ,3-cycloheptadienyl, 1 ,3,5-cycloheptatrienyl, 1 ,3,5,7-cycloocta- tetraenyl and the like.
The term "alkynyl" used either alone or in compound words such as "optionally substituted alkynyl" refers to straight chain or branched chain radicals having at least one carbon- carbon triple bond of 2 to 20 carbon atoms, preferably 2 to 14 carbon atoms, more preferably 2 to 6 carbon atoms. Examples include ethynyl, 1-propynyl, 1- and 2-butynyl, 2-methyl-2-proρynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 2-hexynyl, 3-hexynyl, 4- hexynyl, 5-hexynyl, 10-undecynyl, 4-ethyl-l-octyn-3-yl, 7-dodecynyl, 9-dodecynyl, 10- dodecynyl, 3-methyl-l-dodecyn-3-yl, 2-tridecynyl, 11-tridecynyl, 3-tetradecynyl, 7- hexadecynyl, 3-octadecynyl and the like.
The term "heterocyclyl group" used either alone or in compound words such as "optionally substituted heterocyclyl" refers to monocyclic or polycyclic heterocyclic groups containing at least one heteroatom atom selected from nitrogen, sulphur and oxygen.
Suitable heterocyclic groups include N-containing heterocyclic groups, -such as, unsaturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl or tetrazolyl;
saturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms, such as, pyrrolidinyl, imidazolidinyl, piperidino or piperazinyl; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, such as indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl or tetrazolopyridazinyl;
unsaturated 3 to 6-membered heteromonocyclic group containing an oxygen atom, such as, pyranyl or ruryl;
unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulphur atoms, such as, thienyl;
unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as, oxazolyl, isoxazolyl or oxadiazolyl;
saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as, morpholinyl;
unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as, benzoxazolyl or benzoxadiazolyl;
unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulphur atoms and 1 to 3 nitrogen atoms, such as, thiazolyl or thiadiazolyl;
saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulphur atoms and 1 to 3 nitrogen atoms, such as, thiazolidinyl; and
unsaturated condensed heterocyclic group containing 1 to 2 sulphur atoms and 1 to 3 nitrogen atoms, such as, benzothiazolyl or benzothiadiazolyl.
Preferably the heterocyclyl is an unsaturated 5 or 6-membered heteromonocyclic group containing 1 to 3 nitrogen atoms such as imidazolyl or pyridinyl; a saturated 5 or 6- membered heteromonocyclic group containing 1 to 4 nitrogen atoms such as imidazolidinyl or piperazinyl; or a saturated 5 or 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as morpholinyl.
The term "aryl" used either alone or in compound words such as "optionally substituted aryl" denotes a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused. The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane and biphenyl. Preferably, the aryl is optionally substituted phenyl such as 4-halophenyl, more preferably 4-fiuorophenyl or 4-chlorophenyl.
The term "halo" refers to fluorine, chlorine, bromine or iodine, preferably fluorine, iodine or chlorine, most preferably chlorine.
The term "alkoxy" refers to straight chain or branched oxy-containing radicals preferably each having alkyl portions of 1 to about 6 carbon atoms. Examples of alkoxy include methoxy, ethoxy, propoxy, butoxy and tert-butoxy.
The term "optionally substituted thio" refers to optional substituents such as radicals containing a linear or branched alkyl of 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 4 carbon atoms, attached to a divalent sulphur atom. Examples of alkylthio radicals include methylthio, ethylthio, propylthio, butylthio and hexylthio.
The term "optionally substituted" refers to a group which may or may not be further substituted with one or more groups selected from alkyl, alkenyl, alkynyl, aryl, aldehyde, halo, haloalkyl, haloalkenyl, haloalkynyl, haloaryl, hydroxy, alkoxy, alkenyloxy, aryloxy, benzyloxy, haloalkoxy, haloalkenyloxy, haloaryloxy, nitro, nitroalkyl, nitroalkenyl, nitroalkynyl, nitroaryl, nitroheterocyclyl, amino, alkylamino, dialkylamino, alkenylamino, alkynylamino, arylamino, diarylamino, benzylamino, dibenzylamino, acyl, alkenylacyl, alkynylacyl, arylacyl, acylamino, diacylamino, acyloxy, alkylsulphonyloxy, arylsulphenyloxy, heterocyclyl, heterocycloxy, heterocyclamino, haloheterocyclyl, alkylsulphenyl, arylsulphenyl, carboalkoxy, carboaryloxy, mercapto, alkylthio, benzylthio, acylthio, phosphorus-containing groups and the like. Preferably, the optional substituent is C1-6 alkyl, more preferably Cj-4 alkyl; CF3; fluorine; chlorine; iodine; cyano; Ci-6 alkoxy, more preferably Ci-4 alkoxy; aryl; heterocyclyl; amino; or alkylamino.
The term "antioxidant" is used herein in its broadest sense and refers to a group which has the capacity to react with a reactive oxygen species such as a hydroxyl radical in such a way as to generate a non toxic product. Examples include phenols such as 3,4,5- trimethoxyphenyl and 3,5-di-t-butyl-4-hydroxyphenyl, indole amines such as melatonin and flavonoids. Other examples may be found the literature (Wright et al, J Am Chem Soc 123:1173-1183, 2001).
The term "targeting moiety" is used herein in its broadest sense and refers to a group which will facilitate the brain delivery of the drug by way of an active transport mechanism. The targeting moiety is recognized by specific transporter enzymes integral to the blood brain barrier and these transporter enzymes then provide a mechanism for the drug to be imported into the brain. Typically such transporters are sodium dependant and their substrates contain carboxylic acids such as ascorbic acid and L-glutamate. Conjugation of the targeting moiety to the drug is enacted so as to retain the acid moiety.
The term "metal chelator" is used herein is distinguished from the previously known concept of "chelation therapy". "Chelation therapy" is a term associated clinically with the removal of bulk metals such as in Wilson's disease, β-thallesemia and haemochromatosis.
The salts of the above compounds are preferably pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as intermediates in the preparation of pharmaceutically acceptable salts. Examples of pharmaceutically acceptable salts include salts of pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium; acid addition salts of pharmaceutically acceptable inorganic acids such as hydrochloric, orthophosphoric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids; or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, trihalomethanesulphonic, toluenesulphonic, benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
In addition, some of the compounds of the present invention may form solvates with water or common organic solvents. Such solvates are encompassed within the scope of the instant invention.
The term "pro-drug" is used herein in its broadest sense to include those compounds which are converted in vivo to the above compounds. Use of the pro-drug strategy optimises the delivery of the drug to its site of action, for example, the retina. In one aspect, the term refers to the presence of a Ci-6 alkyl or arylester moiety which is designed to resist hydrolysis until the pro-drug has crossed the BBB. In a second aspect, the term refers to the attachment at position 2 of an antioxidant group, in particular the 3,4,- 5trimethoxyphenyl moiety or derivatives thereof. Exposure to the prooxidative environment of the retinalmay then lead to hydroxylation of the 3,4,5-trimethoxyphenyl group to give a 2-hydroxy-3,4,5-trimethoxyphenyl substituent, the hydroxyl group of which acts to enhance the binding properties of the above compounds.
The term "tautomer" is used herein in its broadest sense to include the above compounds which are capable of existing in a state of equilibrium between two isomeric forms. Such compounds may differ in the bond connecting two atoms or groups and the position of these atoms or groups in the compound.
The term "isomer" is used herein in its broadest sense and includes structural, geometric and stereo isomers. As the above compounds may have one or more chiral centres, it is capable of existing in enantiomeric forms.
The compositions of the present invention comprise at least one of the above compounds together with one or more pharmaceutically acceptable carriers and optionally other therapeutic agents. Each carrier, diluent, adjuvant and/or excipient . must be pharmaceutically "acceptable" in the sense of being compatible with the other ingredients of the composition and not injurious to the subject. Compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers, diluents, adjuvants and/or excipients or finely divided solid carriers or both, and then if necessary shaping the product.
The above compounds may be administered orally, topically, or parenterally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, and vehicles. The term parenteral as used herein includes subcutaneous injections, aerosol for administration to lungs or nasal cavity, intravenous, intramuscular, intrathecal, intracranial, injection or infusion techniques. Intra-ocular administation is particularly useful.
The present invention also provides suitable topical, oral, and parenteral pharmaceutical formulations for use in the novel methods of treatment of the present invention. The compounds of the present invention may be administered orally as tablets, aqueous or oily suspensions, lozenges, troches, powders, granules, emulsions, capsules, syrups or elixirs. The composition for oral use may contain one or more agents selected from the group of sweetening agents, flavoring agents, colouring agents and preserving agents in order to produce pharmaceutically elegant and palatable preparations. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharin. Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar. Suitable flavoring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavoring. Suitable preservatives include sodium benzoate, vitamin E5 alphatocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate. The tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
These excipients may be, for example, (1) inert diluents, such as calcium carbonate, lactose, calcium phosphate or sodium phosphate; (2) granulating and disintegrating agents, such as corn starch or alginic acid; (3) binding agents, such as starch, gelatin or acacia; and (4) lubricating agents, such as magnesium stearate, stearic acid or talc. These tablets may be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. Coating may also be performed using techniques described in the U.S. Pat. Nos. 4,256,108; 4,160,452; and 4,265,874 to form osmotic therapeutic tablets for control release.
The above compounds as well as the pharmaceutically-active agent useful in the method of the invention can be administered, for in vivo application, parenterally by injection or by gradual perfusion over time independently or together. Administration may be intraocular, intravenously, intraarterial, intraperitoneally, intramuscularly, subcutaneously, intracavity, transdermally or infusion by, for example, osmotic pump. For in vitro studies the agents may be added or dissolved in an appropriate biologically acceptable buffer and added to a cell or tissue.
Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous 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 solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's 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, anti-microbials, anti-oxidants, chelating agents, growth factors and inert gases and the like.
The present invention includes various pharmaceutical compositions useful for ameliorating disease. The pharmaceutical compositions according to one embodiment of the invention are prepared by bringing an above compound, analogs, derivatives or salts thereof, or combinations of the above compounds and one or more pharmaceutically-active agents into a form suitable for administration to a subject using carriers, excipients and additives or auxiliaries. Frequently used carriers or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, glycerol and polyhydric alcohols. Intravenous vehicles include fluid and nutrient replenishers. Preservatives include antimicrobial, anti-oxidants, chelating agents and inert gases. Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like, as described, for instance, in Remington's Pharmaceutical Sciences, 20th ed. Williams and Wilkins (2000) and The British National Formulary 43rd ed. (British Medical Association and Royal Pharmaceutical Society of Great Britain, 2002; http://bnf.rhn.net), the contents of which are hereby incorporated by reference. The pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to routine skills in the art. See Goodman and Gilman's The Pharmacological Basis for Therapeutics (7th ed., 1985).
The pharmaceutical compositions are preferably prepared and administered in dose units. Solid dose units may be tablets, capsules and suppositories. For treatment of a subject, depending on activity of the compound, manner of administration, nature and severity of the disorder, age and body weight of the subject, different daily doses can be used. Under certain circumstances, however, higher or lower daily doses may be appropriate. The administration of the daily dose can be carried out both by single administration in the form of an individual dose unit or else several smaller dose units and also by multiple administration of subdivided doses at specific intervals. The pharmaceutical compositions according to the invention may be administered locally or systemically in a therapeutically effective dose. Amounts effective for this use will, of course, depend on the severity of the disease and the weight and general state of the subject. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of the cytotoxic side effects. Various considerations are described, e.g., in Langer, Science, 249:1521, (1990). Formulations for oral use may be in the form of hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin. They may also be in the form of soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
Aqueous suspensions normally contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspension. Such excipients may be (1) suspending agent such as sodium carboxymethyl cellulose, methyl cellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; (2) dispersing or wetting agents which may be (a) naturally occurring phosphatide such as lecithin; (b) a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate; (c) a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadecaethylenoxycetanol; (d) a condensation product of ethylene oxide with a partial ester derived from a fatty acid and hexitol such as polyoxyethylene sorbitol monooleate, or (e) a condensation product of ethylene oxide with a partial ester derived from fatty acids and hexitol anhydrides, for example polyoxyethylene sorbitan monooleate.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to known methods using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of iηjectables.
The above compounds may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines .
The above compounds may also be presented for use in the form of veterinary compositions, which may be prepared, for example, by methods that are conventional in the art. Examples of such veterinary compositions include those adapted for:
(a) oral administration, external application, for example drenches (e.g. aqueous or nonaqueous solutions or suspensions); tablets or boluses; powders, granules or pellets for admixture with feed stuffs; pastes for application to the tongue;
(b) parenteral administration for example by subcutaneous, intramuscular or intravenous injection, e.g. as a sterile solution or suspension; or (when appropriate) by intramammary injection where a suspension or solution is introduced in the udder via the teat;
(c) topical applications, e.g. as a cream, ointment or spray applied to the skin; or
(d) intravaginally, e.g. as a pessary, cream or foam.
The present invention is further described by the following non-limiting Examples. EXAMPLE 1 PB compound mediated dissolution of aggregated abeta 1-42
Abetal-42, is available from the Keck Laboratory, Yale University School of Medicine. PBS(ρH 6.6): Sigma Cat# D-8662. Zn(ZnC12): BDH Cat# 100884E. (Dissolved in water in ImM concentration) DMSO: Ajax Cat# 2225. Thioflavin T: Sigma Cat# T-3516. (Dissolved in water in ImM concentration)
By way of example of an amyloid composition, Abeta is dissolved in distilled water and peptide concentration is assessed by measured absorption at 214 nm in UV spectrometer. An aggregation reactive mixture (per one concentration of one testing compound) is set us as follows: Abeta:25 μM, ZnCl2 50 μM, ThT 50 μM, PBS to make up to 500 μl. The tube is wrapped with foil and incubated at 37 degrees by rotation for 24 hours. A serial dilution of each test compound is made up in DMSO, for example: 100 μM, 500 μM, 1000 μM, 2500 μM and 5000 μM. The final concentrations are 1, 5, 10, 25 and 50 μM. 5 μl of each of these compounds is placed in a centrifuge tubeand 5 μl of DMSO is added to both negative and positive control tubes. 495 μl of aggregates (after 24 hours incubation) is added to the centrifuge tube. The negative control is PBS plus ZnCl2 and ThT and DMSO. The positive control is aggregates plus DMSO. The tubes are incubated at 37 degrees for further 2 hours with rotation. Samples are measured for ThT fluorescence using an LS55 (Perkin Elmer) fluorimeter in a cuvette (500 μl volume). The excitation wavelength is 450 nm and the emission wavelength is 480 nm. Data are analyzed using graph pad prism program. The tested compounds included so called "PB" compounds.
EXAMPLE 2 Post mortem screening
The BAS assay is adopted for post mortem retina. Using a trephine, 6 mm diameter regions of peripheral retina from frozen donor eyes are dissected. Following defrosting, the neuronal retina and the RPE cells are removed by gentle agitation in PBS buffer. Following removal of the RPE cells, strips of the Bruch membrane are cut from the eye.
4 samples are prepared: 1) Control
2) 100 μM TPEN
3) 100 μM PB- 1033
4) 250 μM PB- 1033
Following a 30mins incubation samples are washed 3 times with PBS and then 10 μM ZPl (fluorescence sensor for zinc) application for 10 min.
Samples are then washed 3 times and the labeling visualized using a fluorescence and confocal microscope.
A repeat of this procedure is performed with the exception that samples are incubated for a period of 15 hours before washing to determine differential metal binding over this longer period.
The test results are in the form of fluorescence imaging from a confocal microscope of the 4 samples tested in this Example after 15 hours of sample incubation. The results showed that TPEN inhibited ZPl labeling indicating the effectiveness of the assay. PB-1033 also inhibited ZPl labeling. The results clearly show that PB- 1033 inhibits and reduces metal ions in retinal drusen. Fluorescence microscopic photographs (which are in colour) are available upon request from the Patentee. EXAMPLE 3 Clinical trial
AMD patients are selected and given a test compound (including a PB compound) at a concentration of 500 mg/day for one month. Readouts are taken at baseline and then at 1 month and include:
1. microperimetry; and
2. multifocal retinography.
If the retinae are relieved of oxidative stress after MPAC treatment, it should be reflected by stabilization of these markers of retinal health.
EXAMPLE 4
Assessment of Compounds
The following Assays were used in the assessment of the compounds for suitability for use in the methods of the invention.
Assay 1. FIuorometric H2O2 Assay
A fiuorometric assay was used to test the ability of a test compound to inhibit hydrogen peroxide generation by Aβ in the presence of copper based on dichlorofluoroscein diacetate (DCF; Molecular Probes, Eugene OR). The DCF solution (5mM) in 100% dimethyl sulphoxide (previously purged with argon for 2hr at 2O0C) was deacetylated in the presence of 0.25M NaOH for 30min and neutralized at pH 7.4 to a final concentration of ImM. Horseradish peroxidase (HRP) stock solution was prepared to lμM at pH 7.4. The reactions were carried out in PBS, pH 7.4 in a 96 well plate (total volume =250μl/well). The reaction solutions contained AB 1-42 at concentrations in the range of 5OnM to lμM, copper-glycine chelate (Cu-GIy), was prepared by adding CuCl2 to glycine in the ratio of 1 :6 and added to the AB in the proportion 2Cu-GIy : IAB ), reducing agents including dopamine (5μM) or ascorbic acid, deacetylated DCF lOOμM, and HRP, O.lμM. 1-1 OμM EDTA or another chelator may also be present as a control for free copper, but was not required for the assay to function. The reaction mixture was incubated at 37C for 60 min. Catalase (4000 units/ml) and H2O2 (l-2.5μM) standards in PBS pH 7.4 may be included as positive controls. Fluorescence was recorded using a plate reader with excitation and emission filters at 485nM and 53OnM respectively. H2O2 concentration may be established by comparing fluorescence with the H2O2 standards. Inhibition of AB H2O2 production was assayed by including a given concentration of test compound(s) in the test wells.
Assay 2. Neurotoxicity Assays Primary cortical neuronal cultures Cortical cultures were prepared as previously described (White et ah, J Neuroscience 18:6207-6217, 1998). Embryonic day 14 BL6Jxl29sv mouse cortices were removed, dissected free of meninges and dissociated in 0.025% (wt/vol) trypsin. Dissociated cells were plated in 48 well culture plates at a density of 2 x 106 cells/mL in MEM with 25% (vol/vol) FCS and 5% (vol/vol) HS and incubated at 370C, 2hrs. Media was then replaced with Neurobasal media (Invitrogen Life Technologies) and B27 supplements (Invitrogen Life Technologies). Cultures were maintained at 370C in 5% CO2. Prior to experimentation, the culture medium was replaced with Neurobasal media and B27 minus antioxidants (Invitrogen Life Technologies).
Assay 3. MTS Assay for Cell Viability
Cell viability is determined using the MTS assay. Culture medium is replaced with fresh neurobasal medium plus B27 supplements minus antioxidants. 1/1 Oth volume MTS solution (Cell Titre 96 Aqueous One, Promega Corporation) and incubated at 370C, 2hrs. 200 microlitre aliquots are measured with a spectrophotometer at 560 nm.
Assay 4. Assay for Test Compound Cytoxicity
Neuronal cortical cells were cultured for five days as per Assay 2 in NB media and B27 supplement.
On day six the test compounds were added to the neuronal cell cultures in NB media and B27 supplement minus antioxidants.
Test compounds were dissolved in 100% DMSO to a concentration of 2.5 mM (1OmM if excess compound was weighed out per vial - then diluted to 2.5mM). 2.5mM stock solution was serially diluted 1 in 10 to give working solutions of 250μM, 25μM, 2.5μM. Test compounds were not added directly to cells, instead they were added to a 48 well 'Drug Plate' as comprized below:
Preparation of "Drug Plate": To a 48 well plate add:
Well 1: 576 μl NB+B27(no antioxidant)* + 24 μl 2.5μM test compound Well 2 : 576 μl NB+B27(no antioxidant) + 24 μl 25μM test compound Well 3 : 576 μl NB+B27(no antioxidant) + 24 μl 250μM test compound Well 4 : 576 μl NB+B27(no antioxidant) + 24 μl 2.5μM test compound Well 5 : 576 μl NB+B27(no antioxidant) + 24 μl 25μM test compound Well 6 : 576 μl NB+B27(no antioxidant) + 24 μl 250μM test compound Well 7 : 576 μl NB+B27(no antioxidant) + 24 μl test compound diluent** Well 8 : 600 μl NB+B27(no antioxidant)
The Drug Plate was incubated at 37° C for 15 mins. 200 μl of each well was added in triplicate to the corresponding cell plate. The cell plate was incubated at 37 C, for 4 days.
* NB media and B27 (no antioxidants) , ** PBT diluent 10% DMSO in NB+B27 (no antioxidants)
On completion of the assay, 1/10 volume MTS was added per well of plate (ie 25μl/ 250 μl). The plates were incubated at 37C for 2hrs, and then absorbance was read at 560nm.
Assay 5. Human Brain Amyloid Solubilization Assay This assay was performed in order to assess the ability of a test compound to mobilise Aβ, as an example form of amyloid, from the insoluble to the soluble phase of an extract of tissue from post mortem human AD brain.
Up to 0.5 g of plaque-bearing cortex without meninges was homogenized using a DIAX 900 homogenizer (Heudolph and Co, Kelheim, Germany) or other suitable device for three
30-second periods at full speed in 2 ml of ice-cold phosphate-buffered saline, pH 7.4. To obtain the phosphate-buffered saline-extractable fraction, the homogenate was centrifuged at 100,000 x g for 30 min and the supernatant removed. Alternatively, the tissue was freeze dried then pulverized to form a powder which was then weighed out into aliquots for extraction as above. Supernatant, either freeze-dried and resuspended or in unconcentrated form, was dissolved in 200 μl of Tris-Tricine sodium dodecyl sulfate (SDS) sample buffer pH 8.3 containing 8% SDS5 10% 2-mercaptoethanol. Aliquots (10 μl) were then boiled for 10 minutes before SDS-polyacrylamide gel electrophoresis. The insoluble fraction of the cortical samples was obtained by resuspending the initial pelleted sample in 1 ml of phosphate-buffered saline. A 50-μl aliquot of this suspension was then boiled in 200 ml of sample buffer as above.
Tris-Tricine polyacrylamide gel electrophoresis was performed by loading appropriately diluted samples on to 10% to 20% gradient gels (Novex, San Diego, CA) followed by transfer on to 0.2-μm nitrocellulose membrane (Bio-Rad,Hercules, CA). Aβ was detected by using monoclonal antibody W02, which detects residues 5 through 8, 17 (or another suitable antibody) in conjunction with horseradish peroxidase-conjugated rabbit anti- mouse IgG (Dako, Denmark), and visualized by using enhanced chemiluminescence (eg ECL; Amersham Life Science, Buckinghamshire, UK). Each gel included three lanes containing 0.5, 1, and 2 ng of synthetic Aβ40 (Keck Laboratory, Yale University, New Haven, CT) as reference standards.
Blot films were scanned by using a suitable imaging system such as the UVP gel documentation system, and densitometry performed using suitable software, eg UVP Labworks. The dynamic range of the film/scanner was determined by using a step tablet (No. 911ST600, Kodak, Rochester NY), a calibrated film exposed by the manufacturer to provided steps of known increasing intensity. The quantifiable range of signal intensity for densitometric analysis of the mono- and dimeric Aβ bands was based on the comparison with a curve obtained by scanning and densitometry of the step tablet. Samples in which the signal intensity is low after preliminary assay may be re-assayed by using synthetic standards of lower or higher concentration.
All samples were analyzed at least twice, and gel loadings and dilutions were adjusted to fit within the quantifiable region of the standard curve. The proportion of 'soluble' to 'insoluble' Aβ may be used to determine the efficiency of extraction of a test compound compared with the efficiency of a known compound. The insoluble Aβ being comprized of the pelletable fraction derived from the insoluble amyloid plaque from the above cortical samples and the soluble fraction comprising monomeric and/or oligomeric soluble Aβ. Assay 6. Effect of Administration of Test Compounds on Aβ deposits in Transgenic Animals
Transgenic mouse models are available for a number of neurological disorders, including Alzheimer's disease; Parkinson's disease; familial amyotrophic lateral sclerosis (ALS); Huntington's disease; and Creutzfeld- Jakob disease (CJD). It was found that one of the transgenic models for Alzheimer's disease, the APP2576 transgenic mouse also has a high incidence of cataract. These animal models were suitable for testing the methods of the invention.
Transgenic mice of the strain APP2576 were used. Eight to nine month old female mice were selected and divided into groups for treatment.
Mice were sacrificed at intervals, and their brains examined to determine whether the treatment with test compounds decreased brain amyloid formation, and the identification of the most effective administration protocol.
Other mice in each group were tested over a period of up to eight months for cognitive performance, using a Morris water maze according to standard methods. The general health and well-being of the animals was also measured every day by a blinded operator, using a five point integer scale which subjectively rates a combination of features, including motor activity, alertness and general health signs.
Assay 7. Solubility Assay
Stock solutions of compounds of formula I or II (ImM) were prepared in dimethyl sulfoxide. Compounds which did not dissolve were classed as not soluble (N). The DMSO stock solutions were diluted 1 in 100 into PBS pH 7.4. Compounds which gave a clear solution were classed as soluble (Y), while those compounds which gave a translucent suspension after dissolution in DMSO were classed as "crashed out" (C).
Assay 8. Physiochemical Properties
Polar Surface Area Calculations (PSA)
Polar surface area values were calculated using the web-based program available through
"Molinspiration", a package for calculation of molecular properties.
Turbidimetric Solubility Measurements The solubility estimate was measured at both pH 2.0 and pH 6.5. This is within the pH range that can be anticipated along the proximal gastrointestinal tract in humans.
The compounds were dissolved in DMSO to appropriate concentrations and then spiked into either 0.01M HCl (approx. pH = 2.0) or pH 6.5 isotonic phosphate buffer, the final DMSO concentration being 1%. Samples were then analyzed via Nephelometry to determine a solubility range (Bevan and Lloyd, Anal Chem. 72:1781-1787, 2000).
cLog P values Theoretical Log P values were determined using the ACD Log P software. The values quoted have been calculated from an untrained database and refer to the unionized species.
E Log D
Effective Log D values were measured using a chromatographic method employing a SUPELCOSIL LC-ABZ column using an octanol saturated mobile phase at pH 7.4. See F. Lombardo et al, J. Med. Chem. 2000, 43, 2922-2928.
EXAMPLE 5 Properties of PBT Compounds
Table 8 provides the properties and structures of particularly preferred PBT compounds which fall within the scope of the present invention.
Results on AMD compounds
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
a- concentration in μM of test compound required to inhibit 50% of AMa H2O2 production b- viability of primary cortical neuronal cultured cells (Neuronal cells) or M17 human neuroblastoma cells (M17 cells) in the presence of test compound at concentrations of 1 and lOμM. c- % inhibition of dityrosine oligomerization as referenced to in-house standard (set as 100% inhibition) d- extent by which test compound mobilizes A^ from the insoluble to the soluble phase of an extract of tissue from a postmortem human AD brain. Results are referenced to baseline PBS and are quoted as the max effect achieved across the concentration range followed by the concentration or concentration range at which an effect is observed e- Disaggregation of Ab«a:Zn (25:50μM) Synthetic Aggregations; 1st value = EC50(μM) , 2nd value = % Aggregate reduction at 5μM f- Visual observations during acute tox in mice or Tg mice experiment or PK studies in rats g- Confirmation of presence of compound in plasma at one or two time points (between 30 min and 4h) after single or repeat oral dose of 30 mg/kg (unless otherwise specified) h- % difference from control in insoluble/soluble brain amyloid burden and % difference from control in Amyloid plaque abundance following daily oral gavage at 30mg/kg (unless otherwise specified) over 9 weeks in 13-14 months old transgenic mice. Only statistically significant results (p<0.05) are quoted as percentage values, trends are indicated without numbers.
Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features. BIBLIOGRAPHY
Arch. Ophthalmol. 777:1329-1345, 1999
Bevan and Lloyd, Anal. Chem. 72:1781-1787, 2000
Goodman and Gilman's, The Pharmacological Basis for Therapeutics 7th ed, 1985
Langer, Science, 249:1527, 1990
Remington's Pharmaceutical Sciences, 20th ed, Williams and Wilkins, 2000
The British National Formulary 43rd ed, British Medical Association and Royal Pharmaceutical Society of Great Britain, 2002
WO 02/055081
White et al., JNeuroscience 18:6207-6217, 1998
Wright et al, JAm Chem Soc 723:1173-1183, 2001

Claims

CLAIMS:
1. A method for treating a subject with age-related macular degeneration (AMD) said method comprising administering to said subject an amount of PB-1033:
Figure imgf000105_0001
or a pharmaceutically acceptable salt, derivative or functional equivalent thereof effective to reduce metal in retinal drusen to a level which ameliorates symptoms of AMD.
2. The method of Claim 1 wherein the subject is a mammal.
3. The method of Claim 2 wherein the mammal is a human.
4. The method of Claim 1 or 2 or 3 wherein the metal is selected from the list consisting of zinc and copper.
5. The method of Claim 1 or 2 or 3 or 4 wherein derivative or functional equivalent of PB-1033 is selected from the list in Table 8 or pharmaceutically acceptable salts, derivatives or functional equivalents thereof.
6. The method of Claim 5 wherein the derivative or functional equivalent of PB-1033 is selected from the list consisting of PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or pharmaceutically acceptable salts, derivatives or functional equivalents thereof.
7. Use of PB-1033:
Figure imgf000105_0002
or a pharmaceutically acceptable salt, derivative or functional equivalent thereof in the manufacture of a medicament for the treatment of age-related macular degeneration (AMD) in a subject.
8. Use of Claim 7 wherein the subject is a mammal.
9. Use of Claim 8 wherein the mammal is human.
10. Use of Claim 7 or 8 or 9 wherein the amount of PB- 1033 or derivative of equivalent thereof is an amount which reduces levels of a metal in retinal drusen.
11. Use of Claim 10 wherein the metal is selected from the list consisting of zinc and copper.
12. Use of Claim 7 or 8 or 9 or 10 or 11 wherein derivative or functional equivalent of PB- 1033 is selected from the list in Table 8 or pharmaceutically acceptable salts, derivatives or functional equivalents thereof.
13. Use of Claim 12 wherein the derivative or equivalent of PB-1033 is selected from the list consisting of PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB-1149, PB-1151, PB-1160 and PB-1168 or pharmaceutically acceptable salts thereof.
14. A method for reducing levels of a metal from retinal drusen in a subject to thereby ameliorate symptoms of age-related macular degeneration (AMD) said method comprising administering to said subject an effective amount of PB-1033:
Figure imgf000106_0001
or a pharmaceutically acceptable salt, derivative or equivalent thereof.
15. The method of Claim 14 wherein the subject is a mammal.
16. The method of Claim 15 wherein the mammal is a human.
17. The method of Claim 14 or 15 or 16 wherein the metal is selected from the list consisting of zinc and copper.
18. The method of Claim 14 or 15 or 16 or 17 wherein derivative or functional equivalent of PB-1033 is selected from the list in Table 8 or pharmaceutically acceptable salts, derivatives or functional equivalents thereof.
19. The method of Claim 14 wherein the derivative or equivalent of PB- 1033 is selected from the list consisting of PB-1076, PB-1085, PB-1120, PB-1127, PB-1135, PB- 1149, PB-1151, PB-1160 and PB-1168 or pharmaceutically acceptable salts, derivatives or functional equivalents thereof.
PCT/AU2007/000490 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration(amd) WO2007118276A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
NZ572591A NZ572591A (en) 2006-04-14 2007-04-13 3N-substituted 8-hydroxy-quinazolin-4-ones
JP2009504530A JP5290147B2 (en) 2006-04-14 2007-04-13 Method for treating age-related macular degeneration (AMD)
US12/297,165 US20100144693A1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration (amd)
AU2007240120A AU2007240120A1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration (AMD)
DK07718737.5T DK2012789T3 (en) 2006-04-14 2007-04-13 Process for Age-Related Macular Degeneration (AMD)
ES07718737.5T ES2437997T3 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration (AMD)
EP07718737.5A EP2012789B1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration (AMD)
BRPI0710737-4A BRPI0710737A2 (en) 2006-04-14 2007-04-13 use of compounds pb-10 33 useful for the treatment of age-related macular degeneration (amd) as well as said compounds
CA002683756A CA2683756A1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration(amd)
AU2010206074A AU2010206074B2 (en) 2006-04-14 2010-07-30 Method of treatment of age-related macular degeneration (AMD)
IL207495A IL207495A (en) 2006-04-14 2010-08-09 Compounds useful in management of age-related macular degeneration (amd)
US12/952,425 US20110071167A1 (en) 2006-04-14 2010-11-23 Method of treatment of age-related macular degeneration (amd)
US14/087,689 US9163018B2 (en) 2006-04-14 2013-11-22 Method of treatment of age-related macular degeneration (AMD)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79227806P 2006-04-14 2006-04-14
US60/792,278 2006-04-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/297,165 A-371-Of-International US20100144693A1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration (amd)
US12/952,425 Continuation US20110071167A1 (en) 2006-04-14 2010-11-23 Method of treatment of age-related macular degeneration (amd)

Publications (1)

Publication Number Publication Date
WO2007118276A1 true WO2007118276A1 (en) 2007-10-25

Family

ID=38608967

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2007/000490 WO2007118276A1 (en) 2006-04-14 2007-04-13 Method of treatment of age-related macular degeneration(amd)

Country Status (14)

Country Link
US (3) US20100144693A1 (en)
EP (2) EP2012789B1 (en)
JP (1) JP5290147B2 (en)
KR (2) KR20080109096A (en)
CN (3) CN102225930A (en)
AU (2) AU2007240120A1 (en)
BR (2) BRPI0710737A2 (en)
CA (1) CA2683756A1 (en)
DK (1) DK2012789T3 (en)
ES (1) ES2437997T3 (en)
IL (1) IL207495A (en)
NZ (2) NZ572591A (en)
WO (1) WO2007118276A1 (en)
ZA (2) ZA200809493B (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2044026A1 (en) * 2006-06-22 2009-04-08 Prana Biotechnology Limited Method of treatment of glioma brain tumour
US7619091B2 (en) 2002-07-16 2009-11-17 Prana Biotechnology Limited 8-hydroxy quinoline derivatives
WO2010071944A1 (en) * 2008-12-24 2010-07-01 Prana Biotechnology Limited Quinazolinone compounds
WO2010083582A1 (en) 2009-01-26 2010-07-29 Michael Spino Use of deferiprone for treatment and prevention of iron-related eye disorders
EP2331525A4 (en) * 2008-08-11 2013-01-02 Harvard College Halofuginone analogs for inhibition of trna synthetases and uses thereof
WO2014100425A1 (en) * 2012-12-20 2014-06-26 Aldexa Therapeutics, Inc. Peri-carbinols
WO2016086261A1 (en) * 2014-12-02 2016-06-09 Prana Biotechnology Limited 4H-PYRIDO[1,2-a]PYRIMIDIN-4-ONE COMPOUNDS
US9364471B2 (en) 2005-05-26 2016-06-14 Aldeyra Therapeutics, Inc. Compositions and methods of treating retinal disease
US9687481B2 (en) 2013-01-23 2017-06-27 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US9814701B2 (en) 2009-12-11 2017-11-14 Aldeyra Therapeutics, Inc. Compositions and methods for the treatment of macular degeneration
WO2018000047A1 (en) * 2016-07-01 2018-01-04 Prana Biotechnology Limited Method of treating immunoglobulin light chain amyloidosis
US9896420B2 (en) 2011-03-10 2018-02-20 The Trustees Of Columbia University In The City Of New York N-quinolin-benzensulfonamides and related compounds for the treatment of cancer, autoimmune disorders and inflammation
US10111862B2 (en) 2013-01-25 2018-10-30 Aldeyra Therapeutics, Inc. Traps in the treatment of macular degeneration
US10155742B2 (en) 2012-01-13 2018-12-18 President And Fellows Of Harvard College Halofuginol derivatives and their use in cosmetic and pharmaceutical compositions
US10414732B2 (en) 2017-03-16 2019-09-17 Aldeyra Therapeutics, Inc. Polymorphic compounds and uses thereof
US10550085B2 (en) 2015-08-21 2020-02-04 Aldeyra Therapeutics, Inc. Deuterated compounds and uses thereof
US11040039B2 (en) 2017-10-10 2021-06-22 Aldeyra Therapeutics, Inc. Treatment of inflammatory disorders
US11129823B2 (en) 2016-05-09 2021-09-28 Aldeyra Therapeutics, Inc. Combination treatment of ocular inflammatory disorders and diseases
WO2022006439A3 (en) * 2020-07-02 2022-02-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cyclic compounds for use in treating retinal degeneration
US11312692B1 (en) 2018-08-06 2022-04-26 Aldeyra Therapeutics, Inc. Polymorphic compounds and uses thereof
US11708353B2 (en) 2018-06-08 2023-07-25 The General Hospital Corporation Inhibitors of prolyl-tRNA-synthetase

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018514817A (en) * 2015-04-29 2018-06-07 トライアペックス カンパニー リミテッド Polarizing film, method for producing the same, and polarizing lens including the same
US10781178B2 (en) * 2015-08-12 2020-09-22 The General Hospital Corporation 8-hydroxyquinoline derivatives as diagnostic and therapeutic agents
US11008290B2 (en) * 2015-09-24 2021-05-18 University Of Florida Research Foundation, Incorporated Halogenated quinoline derivatives as antimicrobial agents
CN109232396B (en) * 2018-11-27 2020-07-03 聊城大学 Amide pyridine derivative and application thereof
CN113214249B (en) * 2021-04-23 2023-09-19 成都大学 Synthesis method of pyrido [1,2-a ] pyrimidine-4-thioketone compound

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113277A2 (en) * 2003-06-23 2004-12-29 Neurochem (International) Limited Methods and compositions for treating amyloid-related diseases
WO2006083533A2 (en) * 2005-01-14 2006-08-10 The Regents Of The University Of California Compositions and methods for inhibiting drusen formation and for diagnosing or treating drusen-related disorders

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE505711A (en) 1950-09-09
US4256108A (en) 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4160452A (en) 1977-04-07 1979-07-10 Alza Corporation Osmotic system having laminated wall comprising semipermeable lamina and microporous lamina
HU178910B (en) * 1977-08-19 1982-07-28 Chinoin Gyogyszer Es Vegyeszet Process for preparing 2,3-disubstituted-4-oxo-4h-pyrido/1,2-a/-pyrimidines
US4265874A (en) 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
IT1204612B (en) 1987-05-14 1989-03-10 Bioresearch Spa PTERIDINS SUITABLE FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS FOR ANTI-MAGNESIC ACTIVITIES
US5314909A (en) * 1993-03-17 1994-05-24 Merck & Co., Inc. Use of non-steroidal antiiflammatory agents in macular degeneration
AU674402B2 (en) 1993-04-08 1996-12-19 Ideal-Standard Gmbh Sanitary water tap
US5631373A (en) 1993-11-05 1997-05-20 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University And The University Of Oregon, Eugene Oregon Alkyl, azido, alkoxy, and fluoro-substituted and fused quinoxalinediones
GB9404485D0 (en) 1994-03-09 1994-04-20 Cancer Res Campaign Tech Benzamide analogues
DE4418096A1 (en) * 1994-05-24 1995-11-30 Cassella Ag Use of pteridine derivatives as inhibitors of NO synthase
US6168776B1 (en) 1994-07-19 2001-01-02 University Of Pittsburgh Alkyl, alkenyl and alkynyl Chrysamine G derivatives for the antemortem diagnosis of Alzheimer's disease and in vivo imaging and prevention of amyloid deposition
US5801183A (en) 1995-01-27 1998-09-01 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University And The University Of Oregon Aza and aza (N-oxy) analogs of glycine/NMDA receptor antagonists
PL188838B1 (en) * 1996-08-13 2005-04-29 P N Gerolymatos Sa Application of a chelating agent clioquinole in production of a pharmaceutic agent designed for use in treatment of alzheimer disease
GB9702701D0 (en) 1997-02-01 1997-04-02 Univ Newcastle Ventures Ltd Quinazolinone compounds
US5939421A (en) 1997-07-01 1999-08-17 Signal Pharmaceuticals, Inc. Quinazoline analogs and related compounds and methods for treating inflammatory conditions
US20020025944A1 (en) * 2000-04-28 2002-02-28 Bush Ashley I. Use of clioquinol for the therapy of Alzheimer's disease
US6337332B1 (en) * 1998-09-17 2002-01-08 Pfizer Inc. Neuropeptide Y receptor antagonists
NZ510098A (en) 1998-10-22 2003-09-26 Neurosearch As Substituted phenyl derivatives, pharmaceuticals thereof and their use as blockers of the chloride channel
US6369058B1 (en) * 1999-02-04 2002-04-09 New Millennium Pharmaceutical Research Inc. Brain delivery of folic acid for the prevention of alzheimer's disease and stroke
AU7080500A (en) 1999-08-27 2001-03-26 Chemocentryx, Inc. Compounds and methods for modulating cxcr3 function
AUPR201600A0 (en) 2000-12-11 2001-01-11 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivative
ATE528303T1 (en) 2000-12-21 2011-10-15 Vertex Pharma PYRAZOLE COMPOUNDS AS PROTEIN KINASE INHIBITORS
FR2819187A1 (en) 2001-01-10 2002-07-12 Michel Xilinas USE OF CHELATING SUBSTANCES FOR THE TREATMENT AND PREVENTION OF EYE DEFERRESCENCE
ATE427948T1 (en) 2001-04-24 2009-04-15 Purdue Research Foundation FOLATE MIMETICS AND THEIR FOLATE RECEPTOR-BINDING CONJUGATES
FR2827599A1 (en) * 2001-07-20 2003-01-24 Neuro3D Treating central nervous system disorders, e.g. anxiety, depression, schizophrenia or pain, using new or known 2-substituted quinoline or quinoxaline derivatives having xanthenuric acid modulating action
US20050014780A1 (en) 2001-08-17 2005-01-20 Maligres Peter E. Process for preparing 5-sulfonamido-8-hydroxy-1,6-naphthyridine-7-carboxamides
AU2003211560A1 (en) 2002-02-20 2003-09-09 Ajinomoto Co., Inc. Novel phenylalanine derivative
AU2003228283A1 (en) 2002-03-07 2003-09-22 X-Ceptor Therapeutics, Inc. Quinazolinone modulators of nuclear receptors
AU2002950217A0 (en) * 2002-07-16 2002-09-12 Prana Biotechnology Limited 8- Hydroxy Quinoline Derivatives
AU2002951868A0 (en) * 2002-10-04 2002-10-24 Prana Biotechnology Limited Compound i
KR101122782B1 (en) * 2002-10-04 2012-04-12 프라나 바이오테크놀로지 리미티드 Neurologically-active compounds
CN1791408A (en) * 2003-04-03 2006-06-21 普拉纳生物技术有限公司 Treatment of neurological conditions
CA2563038C (en) * 2004-04-02 2013-10-29 Gaik Beng Kok Neurologically-active compounds
WO2006117660A2 (en) * 2005-05-04 2006-11-09 Clio Pharmaceutical Corporation Method for treating cancer, coronary, inflammatory and macular disease, combining the modulation of zinc- and/or copper dependent proteins
DK2044026T3 (en) * 2006-06-22 2014-06-30 Prana Biotechnology Ltd PROCEDURES FOR THE TREATMENT OF CEREBRAL GLIOMA TUMOR

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113277A2 (en) * 2003-06-23 2004-12-29 Neurochem (International) Limited Methods and compositions for treating amyloid-related diseases
WO2006083533A2 (en) * 2005-01-14 2006-08-10 The Regents Of The University Of California Compositions and methods for inhibiting drusen formation and for diagnosing or treating drusen-related disorders

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LENGYEL IMRE ET AL.: "High concentration of zinc in sub-retinal pigment epithelial deposits", EXPERIMENTAL EYE RESEARCH, vol. 84, no. 4, 9 January 2007 (2007-01-09), pages 772 - 780, XP005923935 *
See also references of EP2012789A4 *

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7619091B2 (en) 2002-07-16 2009-11-17 Prana Biotechnology Limited 8-hydroxy quinoline derivatives
US10913722B2 (en) 2005-05-26 2021-02-09 Aldeyra Therapeutics, Inc. Compositions and methods of treating retinal disease
US11724987B2 (en) 2005-05-26 2023-08-15 Aldeyra Therapeutics, Inc. Compositions and methods of treating retinal disease
US9364471B2 (en) 2005-05-26 2016-06-14 Aldeyra Therapeutics, Inc. Compositions and methods of treating retinal disease
EP2044026A4 (en) * 2006-06-22 2009-08-05 Prana Biotechnology Ltd Method of treatment of glioma brain tumour
EP2044026A1 (en) * 2006-06-22 2009-04-08 Prana Biotechnology Limited Method of treatment of glioma brain tumour
US8163760B2 (en) 2006-06-22 2012-04-24 Prana Biotechnology Limited Use of pyridopyrimidine compounds in the treatment of gliomas
EP2331525A4 (en) * 2008-08-11 2013-01-02 Harvard College Halofuginone analogs for inhibition of trna synthetases and uses thereof
JP2012513416A (en) * 2008-12-24 2012-06-14 プラナ バイオテクノロジー リミティッド Quinazolinone compounds
US8889695B2 (en) 2008-12-24 2014-11-18 Prana Biotechnology Limited Quinazolinone compounds
JP2015143249A (en) * 2008-12-24 2015-08-06 プラナ バイオテクノロジー リミティッド Quinazolinone compounds
US9145375B2 (en) 2008-12-24 2015-09-29 Prana Biotechnology Limited Quinazolinone compounds
WO2010071944A1 (en) * 2008-12-24 2010-07-01 Prana Biotechnology Limited Quinazolinone compounds
WO2010083582A1 (en) 2009-01-26 2010-07-29 Michael Spino Use of deferiprone for treatment and prevention of iron-related eye disorders
US9814701B2 (en) 2009-12-11 2017-11-14 Aldeyra Therapeutics, Inc. Compositions and methods for the treatment of macular degeneration
US9896420B2 (en) 2011-03-10 2018-02-20 The Trustees Of Columbia University In The City Of New York N-quinolin-benzensulfonamides and related compounds for the treatment of cancer, autoimmune disorders and inflammation
US10155742B2 (en) 2012-01-13 2018-12-18 President And Fellows Of Harvard College Halofuginol derivatives and their use in cosmetic and pharmaceutical compositions
US9604997B2 (en) 2012-12-20 2017-03-28 Aldeyra Therapeutics, Inc. Peri-carbinols
WO2014100425A1 (en) * 2012-12-20 2014-06-26 Aldexa Therapeutics, Inc. Peri-carbinols
US10213395B2 (en) 2013-01-23 2019-02-26 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11771664B2 (en) 2013-01-23 2023-10-03 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US10543181B2 (en) 2013-01-23 2020-01-28 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11701331B2 (en) 2013-01-23 2023-07-18 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US10588874B2 (en) 2013-01-23 2020-03-17 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US9687481B2 (en) 2013-01-23 2017-06-27 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11007157B2 (en) 2013-01-23 2021-05-18 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US10111862B2 (en) 2013-01-25 2018-10-30 Aldeyra Therapeutics, Inc. Traps in the treatment of macular degeneration
EA031505B1 (en) * 2014-12-02 2019-01-31 Прана Байотекнолоджи Лимитед 4H-PYRIDO[1,2-a]PYRIMIDIN-4-ONE COMPOUNDS
US10287285B2 (en) 2014-12-02 2019-05-14 Prana Biotechnology Limited 4H-pyrido[1,2-A]pyrimidin-4-one compounds
WO2016086261A1 (en) * 2014-12-02 2016-06-09 Prana Biotechnology Limited 4H-PYRIDO[1,2-a]PYRIMIDIN-4-ONE COMPOUNDS
US10738050B2 (en) 2014-12-02 2020-08-11 Prana Biotechnology Limited 4H-pyrido[1,2-A]pyrimidin-4-one compounds
US11046650B2 (en) 2015-08-21 2021-06-29 Aldeyra Therapeutics, Inc. Deuterated compounds and uses thereof
US11459300B2 (en) 2015-08-21 2022-10-04 Aldeyra Therapeutics, Inc. Deuterated compounds and uses thereof
US10550085B2 (en) 2015-08-21 2020-02-04 Aldeyra Therapeutics, Inc. Deuterated compounds and uses thereof
US11845722B2 (en) 2015-08-21 2023-12-19 Aldeyra Therapeutics, Inc. Deuterated compounds and uses thereof
US11129823B2 (en) 2016-05-09 2021-09-28 Aldeyra Therapeutics, Inc. Combination treatment of ocular inflammatory disorders and diseases
US11357770B2 (en) 2016-07-01 2022-06-14 Alterity Therapeutics Limited Method of treating immunoglobulin light chain amyloidosis
WO2018000047A1 (en) * 2016-07-01 2018-01-04 Prana Biotechnology Limited Method of treating immunoglobulin light chain amyloidosis
US10414732B2 (en) 2017-03-16 2019-09-17 Aldeyra Therapeutics, Inc. Polymorphic compounds and uses thereof
US11040039B2 (en) 2017-10-10 2021-06-22 Aldeyra Therapeutics, Inc. Treatment of inflammatory disorders
US11583529B2 (en) 2017-10-10 2023-02-21 Aldeyra Therapeutics, Inc. Treatment of inflammatory disorders
US11708353B2 (en) 2018-06-08 2023-07-25 The General Hospital Corporation Inhibitors of prolyl-tRNA-synthetase
US11312692B1 (en) 2018-08-06 2022-04-26 Aldeyra Therapeutics, Inc. Polymorphic compounds and uses thereof
WO2022006439A3 (en) * 2020-07-02 2022-02-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cyclic compounds for use in treating retinal degeneration

Also Published As

Publication number Publication date
EP2012789A4 (en) 2011-02-16
CN101987849B (en) 2013-05-08
AU2010206074A1 (en) 2010-08-19
JP2009533356A (en) 2009-09-17
EP2514423A2 (en) 2012-10-24
CN102225930A (en) 2011-10-26
ZA201005922B (en) 2013-09-25
BRPI0722382A2 (en) 2012-06-05
NZ572591A (en) 2012-01-12
AU2007240120A2 (en) 2008-12-18
EP2514423A3 (en) 2012-12-19
ES2437997T3 (en) 2014-01-15
NZ590271A (en) 2011-12-22
US9163018B2 (en) 2015-10-20
EP2012789B1 (en) 2013-09-25
US20100144693A1 (en) 2010-06-10
BRPI0710737A2 (en) 2011-05-10
US20140088122A1 (en) 2014-03-27
IL207495A (en) 2015-05-31
CA2683756A1 (en) 2007-10-25
CN101534826A (en) 2009-09-16
KR20100110396A (en) 2010-10-12
CN101987849A (en) 2011-03-23
DK2012789T3 (en) 2013-12-16
AU2010206074B2 (en) 2012-08-30
US20110071167A1 (en) 2011-03-24
AU2007240120A1 (en) 2007-10-25
EP2012789A1 (en) 2009-01-14
ZA200809493B (en) 2010-08-25
JP5290147B2 (en) 2013-09-18
IL207495A0 (en) 2010-12-30
KR20080109096A (en) 2008-12-16

Similar Documents

Publication Publication Date Title
US9163018B2 (en) Method of treatment of age-related macular degeneration (AMD)
KR101122782B1 (en) Neurologically-active compounds
KR101149323B1 (en) 8-hydroxy quinoline derivatives
EP1875912A1 (en) Agent for treating involuntary movement
JP2022511374A (en) Inhibitor of VAP-1
KR20090023621A (en) Reversion of malignant phenotype with 9-hydroxy ellipticine derivatives
CA2711638C (en) 3-substituted-8-hydroxypyridino[1,2-a]pyrimidin-4-one compounds and their use in treating retinal degenerative diseases
KR980008226A (en) Prevention and treatment of visual dysfunction
WO2012001438A1 (en) Use of kynurenic acid amide derivatives for the treatment of huntington&#39;s disease
KR20140107416A (en) Treatment of type i and type ii diabetes
US20120022097A1 (en) Compositions and Methods for Treating Cancer
JPH05140158A (en) Substituted pyridine derivative useful for treating glaucoma
JP2024509265A (en) Compositions and methods for treating polycythemia
AU2003265740B2 (en) Neurologically-active compounds
US20140228392A1 (en) Compositions and methods for treating cancer

Legal Events

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

Ref document number: 200780022273.4

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07718737

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2009504530

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 194683

Country of ref document: IL

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007718737

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007240120

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 572591

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 4600/KOLNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020087027935

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2007240120

Country of ref document: AU

Date of ref document: 20070413

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2683756

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 12297165

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 207495

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 1020107020805

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0710737

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20081014