US20140343110A1 - Uses of 4'-desferrithiocin analogs - Google Patents

Uses of 4'-desferrithiocin analogs Download PDF

Info

Publication number
US20140343110A1
US20140343110A1 US14/363,886 US201214363886A US2014343110A1 US 20140343110 A1 US20140343110 A1 US 20140343110A1 US 201214363886 A US201214363886 A US 201214363886A US 2014343110 A1 US2014343110 A1 US 2014343110A1
Authority
US
United States
Prior art keywords
alkyl
certain embodiments
hydrogen
compound
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/363,886
Other languages
English (en)
Inventor
Raymond J. Bergeron, Jr.
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Florida Research Foundation Inc
Original Assignee
University of Florida Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Florida Research Foundation Inc filed Critical University of Florida Research Foundation Inc
Priority to US14/363,886 priority Critical patent/US20140343110A1/en
Assigned to UNIVERSITY OF FLORIDA RESEARCH FOUNDATION, INCORPORATED reassignment UNIVERSITY OF FLORIDA RESEARCH FOUNDATION, INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BERGERON, RAYMOND J., JR.
Publication of US20140343110A1 publication Critical patent/US20140343110A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF FLORIDA
Priority to US15/424,557 priority patent/US20170209420A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/08Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D277/10Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms

Definitions

  • Iron metabolism in primates is characterized by a highly efficient recycling process.
  • Brittenham “Disorders of Iron Metabolism: Iron Deficiency and Overload” In Hematology: Basic Principles and Practice ; 3rd ed.; Hoffman et al., Eds.; Churchill Livingstone: New York, 2000; 397-428. Consequently, there is no specific mechanism for eliminating this transition metal.
  • the introduction of “excess iron” into this closed metabolic loop often leads to chronic iron overload and can ultimately lead to biological damage (e.g., peroxidative tissue damage).
  • biological damage e.g., peroxidative tissue damage.
  • There are a number of ways in which excess iron is introduced including a high-iron diet, acute iron ingestion, or malabsorption of the metal. Conrad et al.
  • desferrioxamine B a hexacoordinate hydroxamate iron chelator produced by Streptomyces pilosus
  • desferrioxamine B is not ideal for chelation therapy because iron is removed with a low efficiency.
  • the oral activity of desferrioxamine B is marginal, thereby requiring parenteral administration, which can result in poor patient compliance, particularly for patients in need of long-term chelation therapy.
  • enterobactin and rhodotorulic acid exhibit unacceptable toxicity, and neither demonstrated measurable oral activity.
  • siderophores and synthetic iron chelators have been developed, most have been abandoned because their properties are not suitable for use in treating chronic iron overload.
  • macular degeneration results in the loss of central vision and is a major cause of blindness and visual impairment in older adults.
  • Subjects with macular degeneration frequently cannot read or recognize faces due to their visual impairment.
  • Stroke is caused by a lack of blood flow to an area of the brain and depending on the area of the brain affected can result in the inability to move limbs on one side of the body or can affect speech or vision.
  • Reperfusion injury is due to oxidative stress in ischemic tissue after blood flow has been restored.
  • IBD Irritable bowel disease
  • IBD is a functional bowel disease characterized by abdominal pain and discomfort, bloating, diarrhea, and/or constipation in the absence of any detectable cause.
  • IBD does not lead to more serious problems in most patients, it is a source of chronic pain and fatigue for patients who suffer with this condition.
  • closed head injury is the leading cause of death in children under 4 years of age and is the most common cause of physical disability and cognitive impairment in young people. All of these diseases need better treatments including new approaches to their treatment.
  • the present invention stems from the recognition that the pathogenesis of various diseases, including macular degeneration, closed head injury, irritable bowel disease (IBD), stroke, reperfusion injury, and other diseases and conditions, involves free iron and the generation of reactive oxygen species (ROS), including superoxide anion, hydrogen peroxide, hypochlorous acid, and hydroxyl radicals, and other longer lived, free radicals.
  • ROS reactive oxygen species
  • Such radicals are now realized to be important contributors to many diseases including macular degeneration, head injury, IBD, stroke, and reperfusion injury.
  • free iron contributes to the formation of reactive oxygen species.
  • Fe +2 ions in biological systems react with oxygen species to produce highly reactive hydroxyl radicals via the Fenton reaction (see scheme below).
  • the hydroxyl radical is a highly effective oxidizing agent, reacting at a diffusion-controlled rate with most organic species, such as nucleic acids, proteins, and lipids. Furthermore, superoxide anions or a biological reductant (e.g., ascorbic acid) can reduce the resulting Fe + 3 ion back to Fe +2 for continued peroxide reduction, thus a problematic cycle.
  • a biological reductant e.g., ascorbic acid
  • diseases or conditions that lead to bleeding and/or an inflammatory response involve the possibility that reactive oxygen species will come in contact with Fe +2 ions to produce highly reactive and damaging hydroxyl radicals. That is, the iron released from red blood cells react with oxygen species produced by inflammatory cells such as neutrophils to produce hydroxyl radicals that cause cell and tissue injury.
  • the solution therefore, is the same for conditions of focal iron overload (e.g., closed head injury, hemorrhagic stroke, IBD) as it is for global iron overload-chelation and removal of the unmanaged iron.
  • the present invention applies the use of these analogs, which have been previously only suggested for use in the treatment of global metal overload, to diseases and conditions associated with focal iron overload, such as, but not limited to, macular degeneration, stroke, IBD, closed head injury, and reperfusion injury.
  • the desferrithiocin analog useful in the present invention is of Formula (I):
  • R 1 -R 9 are as defined here.
  • desferrithiocin analogs with a polyether moiety at the 4′-position of the phenyl ring are used in the present invention (i.e., R 2 is a polyether moiety).
  • Such analogs have been found to be useful in treating diseases or conditions associated with focal iron overload, for example, where iron has been introduced into an organ, tissue, or space by bleeding or through an inflammatory response.
  • such analogs have been found in the cerebral spinal fluid (CSF) and therefore may be useful in treating neurological diseases such as closed head injury or stroke.
  • such analogs have been found to penetrate into the eye and may be useful in treating ophthalmologic diseases such as macular degeneration. All of these diseases are associated with free radical damage resulting from unmanaged or free iron in the respective tissue or organ. Therefore, the chelation and removal of the free iron in these tissues and organs would be effective in preventing or treating each of these diseases.
  • the present invention provides methods of treating and preventing diseases and conditions associated with focal iron overload and pharmaceutical compositions for use in treating such diseases and conditions.
  • the invention provides new uses for previously known compounds in the treatment of diseases and conditions associated with focal iron overload.
  • the invention also provides kits including compounds and compositions found useful in treatment of such disease and conditions.
  • the invention provides methods of preventing or treating macular degeneration by administering to a subject an effective amount of a compound of Formula (I) to prevent or treat macular degeneration.
  • Compounds of Formula (I) have been found to get into the eye and chelate and remove iron that is thought to contribute to the generation of reactive oxygen species in the eye that cause biological injury. Such reactive oxygen species are particularly detrimental in the retina of the eye.
  • the present invention also provides pharmaceutical compositions suitable for ocular administration and uses of the compounds of Formula (I) and compositions thereof for the treatment of macular degeneration.
  • the pharmaceutical composition for ocular administration is in the form of an eyedrop.
  • the compound of Formula (I) or a composition thereof may also be administered systemically for the treatment of macular degeneration.
  • the invention provides methods of removing iron from tissues or organs that have been bled into or otherwise have focal iron overload.
  • methods for the treatment of head injury including closed head injury
  • Closed head injuries that may be treated by the inventive methods and compositions may result from any number of causes including falls, blasts, sports injuries, accidents including vehicular accidents, and assaults.
  • the inventive method comprises administering to a subject an effective amount of a compound of Formula (I) to sequester iron resulting from a hemorrhage or vascular compromise in the head.
  • the subject has suffered from a closed head injury.
  • the subject has suffered from or is at risk of suffering from a stroke (e.g., a hemorrhagic stroke).
  • the present invention also provides pharmaceutical compositions for the treatment of head injury and uses of the compounds of Formula (I) and compositions thereof for the treatment of head injury.
  • the compound of Formula (I) or a composition thereof is administered systemically (e.g., orally or parenterally).
  • the invention provides methods for the treatment of stroke, particularly hemorrhagic stroke.
  • Such methods include administering a compound of Formula (I) to a subject at risk of having a stroke or having had a stroke.
  • the method comprises administering to a subject who has had or is at risk of having a stroke an effective amount of a compound of Formula (I).
  • the administered compound is thought to sequester iron resulting from hemorrhage or vascular compromise thereby preventing or at least lessening tissue damage caused by reactive oxygen species.
  • reactive oxygen species may be generated by free iron ions resulting from the bleed in the brain.
  • the subject has suffered from a hemorrhagic stroke.
  • the subject is at risk of having a hemorrhagic stroke.
  • the present invention also provides pharmaceutical compositions for the treatment of stroke and uses of the compounds of Formula (I) and compositions thereof for the treatment of stroke.
  • the compound of Formula (I) or a composition thereof is administered systemically (e.g., orally or parenterally).
  • the invention provides methods for preventing or lessening reperfusion injury.
  • Reperfusion injury is caused by reactive oxygen species that are generated when the blood supply returns to a tissue after a period of ischemia.
  • Compounds of Formula (I) or compositions thereof are administered to a subject at risk of reperfusion injury to prevent the formation of reactive oxygen species or inactivate free radical species. Ischemia may result from a number of causes including stroke, myocardial infarction, infarction of other tissues or organs, surgery (e.g., cardiac surgery), and organ donation and transplantation.
  • the present invention also provides pharmaceutical compositions for the prevention and treatment of reperfusion injury as well as the uses of compounds of Formula (I) and compositions thereof for the prevention and treatment of reperfusion injury.
  • the compounds of Formula (I) or compositions thereof may be administered locally or systemically in the prevention or treatment of reperfusion injury.
  • the present invention provides methods of treating irritable bowel disease (IBD).
  • IBD irritable bowel disease
  • the method comprises administering to a subject an effective amount of a compound of Formula (I) or a composition thereof to treat IBD.
  • the present invention also provides pharmaceutical compositions for the treatment of IBD and the uses of the compounds of Formula (I) and compositions thereof for the treatment of IBD.
  • the compounds of Formula (I) or compositions thereof may be administered locally (e.g., rectally) or systemically in the treatment of IBD.
  • kits may include one or more unit dosage forms of the compound or composition to be administered to a subject.
  • the kit may include enough unit dosage forms for a course of treatment or for a particular time period (e.g., a week, 10 days, 14 days, a month).
  • the kits may also include packaging information describing the use or prescribing information for the subject or a health care professional. Such information may be required by a regulatory agency such as the U.S. Food and Drug Administration (FDA).
  • the kit may also optionally include a device for administration of the compound or composition, for example, a dropper for ocular administration or a syringe for parenteral administration.
  • the compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, ( D )-isomers, ( L )-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • an isomer/enantiomer may, in some embodiments, be provided substantially free of the corresponding enantiomer, and may also be referred to as “optically enriched” or “enantiomerically enriched.”
  • “Optically enriched” and “enantiomerically enriched,” as used herein, means that a provided compound is made up of a significantly greater proportion of one enantiomer.
  • a compound of the present invention is made up of at least about 70% by weight of a preferred enantiomer.
  • a compound of the present invention is made up of at least about 80% by weight of a preferred enantiomer.
  • a compound of the present invention is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer.
  • Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the depicted structures that differ only in the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by 13 C or 14 C are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
  • C 1-4 is intended to encompass, C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 , and C 5-6 .
  • purified refers to a compound useful in the present invention being free of other, dissimilar compounds with which the compound is normally associated in its natural state, so that the compound comprises at least 0.5%, 1%, 5%, 10%, 20%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% of the mass, by weight, of a given sample or composition. In one embodiment, these terms refer to the compound comprising at least 95%, 98%, 99%, or 99.9% of the mass, by weight, of a given sample or composition.
  • acyl refers to a group having the general formula —C( ⁇ O)R X1 , —C( ⁇ O)OR X1 , —C( ⁇ O)—O—C( ⁇ O)R X1 , —C( ⁇ O)SR X1 , —C( ⁇ O)N(R X1 ) 2 , —C( ⁇ S)R X1 , —C( ⁇ S)N(R X1 ) 2 , and —C( ⁇ S)S(R X1 ), —C( ⁇ NR X1 )R X1 , —C( ⁇ NR X1 )OR X1 , —C( ⁇ NR X1 )SR X1 , and —C( ⁇ NR X1 )N(R X1 ) 2 , wherein R X1 is hydrogen; halogen; substituted or unsubstituted hydroxyl; substituted or unsubstituted
  • acyl groups include aldehydes (—CHO), carboxylic acids (—CO 2 H), ketones, acyl halides, esters, amides, imines, carbonates, carbamates, and ureas.
  • Acyl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyl
  • acyloxy refers to a “substituted hydroxyl” of the formula (—OR i ), wherein R i is an optionally substituted acyl group, as defined herein, and the oxygen moiety is directly attached to the parent molecule.
  • aliphatic includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, and cyclic (i.e., carbocyclic) hydrocarbons, which are optionally substituted with one or more functional groups.
  • aliphatic is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties.
  • alkyl includes straight, branched and cyclic alkyl groups.
  • alkenyl alkynyl
  • alkynyl alkenyl
  • alkynyl alkynyl
  • aliphatic is used to indicate those aliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms.
  • Aliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy,
  • alkyl refers to saturated, straight- or branched-chain hydrocarbon radicals derived from a hydrocarbon moiety containing between one and twenty carbon atoms by removal of a single hydrogen atom.
  • the alkyl group employed in the invention contains 1-20 carbon atoms.
  • the alkyl group employed contains 1-15 carbon atoms.
  • the alkyl group employed contains 1-10 carbon atoms.
  • the alkyl group employed contains 1-8 carbon atoms.
  • the alkyl group employed contains 1-5 carbon atoms.
  • alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, sec-pentyl, iso-pentyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, sec-hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, dodecyl, and the like, which may bear one or more substituents.
  • Alkyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy,
  • alkenyl denotes a monovalent group derived from a straight- or branched-chain hydrocarbon moiety having at least one carbon-carbon double bond by the removal of a single hydrogen atom.
  • the alkenyl group employed in the invention contains 2-20 carbon atoms. In some embodiments, the alkenyl group employed in the invention contains 2-15 carbon atoms. In another embodiment, the alkenyl group employed contains 2-10 carbon atoms. In still other embodiments, the alkenyl group contains 2-8 carbon atoms. In yet other embodiments, the alkenyl group contains 2-5 carbons.
  • Alkenyl groups include, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, and the like, which may bear one or more substituents.
  • Alkenyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, hetero
  • alkynyl refers to a monovalent group derived from a straight- or branched-chain hydrocarbon having at least one carbon-carbon triple bond by the removal of a single hydrogen atom.
  • the alkynyl group employed in the invention contains 2-20 carbon atoms. In some embodiments, the alkynyl group employed in the invention contains 2-15 carbon atoms. In another embodiment, the alkynyl group employed contains 2-10 carbon atoms. In still other embodiments, the alkynyl group contains 2-8 carbon atoms. In still other embodiments, the alkynyl group contains 2-5 carbon atoms.
  • alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl, and the like, which may bear one or more substituents.
  • Alkynyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alky
  • Exemplary carbon atom substituents include, but are not limited to, halogen, —CN, —NO 2 , —N 3 , —SO 2 H, —SO 3 H, —OH, —OR aa , —ON(R bb ) 2 , —N(R bb ) 2 , —N(R bb ) 3 + X ⁇ , —N(OR cc )R bb , —SH, —SR aa , —SSR cc , —C( ⁇ O)R aa , —CO 2 H, —CHO, —C(OR cc ) 2 , —CO 2 R aa , —OC( ⁇ O)R aa , —OCO 2 R aa , —C( ⁇ O)N(R bb ) 2 , —OC( ⁇ O)N(R bb ) 2 , —NR bb C
  • each instance of R aa is, independently, selected from C 1-10 alkyl, C 1-10 perhaloalkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 carbocyclyl, 3-14 membered heterocyclyl, C 6-14 aryl, and 5-14 membered heteroaryl, or two R a groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R dd groups;
  • each instance of R bb is, independently, selected from hydrogen, —OH, —OR aa , —N(R cc ) 2 , —CN, —C( ⁇ O)R aa , —C( ⁇ O)N(R cc ) 2 , —CO 2 R aa , —SO 2 R aa , —C( ⁇ NR cc )OR aa , —C( ⁇ NR cc )N(R cc ) 2 , —SO 2 N(R cc ) 2 , —SO 2 R cc , —SO 2 OR cc , —SOR aa , —C( ⁇ S)N(R cc ) 2 , —C( ⁇ O)SR cc , —C( ⁇ S)SR cc , —P( ⁇ O) 2 R aa , —P( ⁇ O)(R aa ) 2 ,
  • each instance of R cc is, independently, selected from hydrogen, C 1-10 alkyl, C 1-10 perhaloalkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 carbocyclyl, 3-14 membered heterocyclyl, C 6-14 aryl, and 5-14 membered heteroaryl, or two R cc groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R dd groups;
  • each instance of R ee is, independently, selected from C 1-6 alkyl, C 1-6 perhaloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 carbocyclyl, C 6-10 aryl, 3-10 membered heterocyclyl, and 3-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R gg groups;
  • each instance of R ff is, independently, selected from hydrogen, C 1-6 alkyl, C 1-6 perhaloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 carbocyclyl, 3-10 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, or two R ff groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R gg groups; and
  • each instance of R gg is, independently, halogen, —CN, —NO 2 , —N 3 , —SO 2 H, —SO 3 H, —OH, —OC 1-6 alkyl, —ON(C 1-6 alkyl) 2 , —N(C 1-6 alkyl) 2 , —N(C 1-6 alkyl) 3 + X ⁇ , —NH(C 1-6 alkyl) 2 + X ⁇ , —NH 2 (C 1-6 alkyl) + X ⁇ , —NH 3 + X ⁇ , —N(OC 1-6 alkyl)(C 1-6 alkyl), —N(OH)(C 1-6 alkyl), —NH(OH), —SH, —SC 1-6 alkyl, —SS(C 1-6 alkyl), —C( ⁇ O)(C 1-6 alkyl), —CO 2 H, —CO 2 (C 1-6 alkyl), —OC( ⁇ O)
  • amino refers to a group of the formula (—NH 2 ).
  • a “substituted amino” refers either to a mono-substituted amine (—NHR h ) of a disubstituted amine (—NR h 2 ), wherein the R h substituent is any substituent as described herein that results in the formation of a stable moiety (e.g., a suitable amino protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, amino, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, aryloxy
  • alkoxy refers to a “substituted hydroxyl” of the formula (—OR i ), wherein R i is an optionally substituted alkyl group as defined herein, and the oxygen moiety is directly attached to the parent molecule.
  • alkylthioxy refers to a “substituted thiol” of the formula (—SR r ), wherein R r is an optionally substituted alkyl group as defined herein, and the sulfur moiety is directly attached to the parent molecule.
  • alkylamino refers to a “substituted amino” of the formula (—NR h 2 ), wherein R h is, independently, a hydrogen or an optionally substituted alkyl group as defined herein, and the nitrogen moiety is directly attached to the parent molecule.
  • aryl refers to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which all the ring atoms are carbon, and which may be substituted or unsubstituted.
  • aryl refers to a mono, bi, or tricyclic C 4 -C 20 aromatic ring system having one, two, or three aromatic rings which include, but not limited to, phenyl, biphenyl, naphthyl, and the like, which may bear one or more substituents.
  • Aryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy, acyl
  • arylalkyl refers to an aryl substituted alkyl group, wherein the terms “aryl” and “alkyl” are defined herein, and wherein the aryl group is attached to the alkyl group, which in turn is attached to the parent molecule.
  • An exemplary arylalkyl group is benzyl.
  • aryloxy refers to a “substituted hydroxyl” of the formula (—OR i ), wherein R i is an optionally substituted aryl group as defined herein, and the oxygen moiety is directly attached to the parent molecule.
  • arylamino refers to a “substituted amino” of the formula (—NR h 2 ), wherein R h is, independently, a hydrogen or an optionally substituted aryl group as defined herein, and the nitrogen moiety is directly attached to the parent molecule.
  • arylthioxy refers to a “substituted thiol” of the formula (—SR r ), wherein R r is an optionally substituted aryl group as defined herein, and the sulfur moiety is directly attached to the parent molecule.
  • azido refers to a group of the formula (—N 3 ).
  • cyano refers to a group of the formula (—CN).
  • halo and “halogen” as used herein refer to an atom selected from fluorine (fluoro, —F), chlorine (chloro, —Cl), bromine (bromo, —Br), and iodine (iodo, —I).
  • heteroaliphatic refers to an aliphatic moiety, as defined herein, which includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, cyclic (i.e., heterocyclic), or polycyclic hydrocarbons, which are optionally substituted with one or more functional groups, and that contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • heteroaliphatic moieties are substituted by independent replacement of one or more of the hydrogen atoms thereon with one or more substituents.
  • heteroaliphatic is intended herein to include, but is not limited to, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl moieties.
  • heteroaliphatic includes the terms “heteroalkyl,” “heteroalkenyl”, “heteroalkynyl”, and the like.
  • heteroalkyl “heteroalkenyl”, “heteroalkynyl”, and the like encompass both substituted and unsubstituted groups.
  • heteroaliphatic is used to indicate those heteroaliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms.
  • Heteroaliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl
  • heteroalkyl refers to an alkyl moiety, as defined herein, which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • heteroalkenyl refers to an alkenyl moiety, as defined herein, which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • heteroalkynyl refers to an alkynyl moiety, as defined herein, which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • heteroalkylamino refers to a “substituted amino” of the formula (—NR h 2 ), wherein R h is, independently, a hydrogen or an optionally substituted heteroalkyl group, as defined herein, and the nitrogen moiety is directly attached to the parent molecule.
  • heteroalkyloxy refers to a “substituted hydroxyl” of the formula (—OR i ), wherein R i is an optionally substituted heteroalkyl group, as defined herein, and the oxygen moiety is directly attached to the parent molecule.
  • heteroalkylthioxy refers to a “substituted thiol” of the formula (—SR r ), wherein R r is an optionally substituted heteroalkyl group, as defined herein, and the sulfur moiety is directly attached to the parent molecule.
  • heterocyclic refers to a cyclic heteroaliphatic group.
  • a heterocyclic group refers to a non-aromatic, partially unsaturated or fully saturated, 3- to 12-membered ring system, which includes single rings of 3 to 8 atoms in size, and bi- and tri-cyclic ring systems which may include aromatic five- or six-membered aryl or heteroaryl groups fused to a non-aromatic ring.
  • These heterocyclic rings include those having from one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, in which the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • heterocyclic refers to a non-aromatic 5-, 6-, or 7-membered ring or polycyclic group wherein at least one ring atom is a heteroatom selected from O, S, and N (wherein the nitrogen and sulfur heteroatoms may be optionally oxidized), and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms.
  • Heterocyclyl groups include, but are not limited to, a bi- or tri-cyclic group, comprising fused five, six, or seven-membered rings having between one and three heteroatoms independently selected from the oxygen, sulfur, and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds, and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl ring.
  • heterocycles include azacyclopropanyl, azacyclobutanyl, 1,3-diazatidinyl, piperidinyl, piperazinyl, azocanyl, thiaranyl, thietanyl, tetrahydrothiophenyl, dithiolanyl, thiacyclohexanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropuranyl, dioxanyl, oxathiolanyl, morpholinyl, thioxanyl, tetrahydronaphthyl, and the like, which may bear one or more substituents.
  • Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthi
  • heteroaryl refers to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which one ring atom is selected from S, O, and N; zero, one, or two ring atoms are additional heteroatoms independently selected from S, O, and N; and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms.
  • heteroaryls include, but are not limited to pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, pyyrolizinyl, indolyl, quinolinyl, isoquinolinyl, benzoimidazolyl, indazolyl, quinolinyl, isoquinolinyl, quinolizinyl, cinnolinyl, quinazolynyl, phthalazinyl, naphthridinyl, quinoxalinyl, thiophenyl, thianaphthenyl, furanyl, benzofuranyl, benzothiazolyl, thiazolynyl, isothiazolyl, thiadiazolynyl, oxazolyl, isoxazolyl, oxadiazi
  • Heteroaryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalky
  • heteroarylene refers to a biradical derived from an heteroaryl group, as defined herein, by removal of two hydrogen atoms. Heteroarylene groups may be substituted or unsubstituted. Additionally, heteroarylene groups may be incorporated as a linker group into an alkylene, alkenylene, alkynylene, heteroalkylene, heteroalkenylene, or heteroalkynylene group, as defined herein.
  • Heteroarylene group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy
  • heteroarylamino refers to a “substituted amino” of the (—NR h 2 ), wherein R h is, independently, a hydrogen or an optionally substituted heteroaryl group, as defined herein, and the nitrogen moiety is directly attached to the parent molecule.
  • heteroaryloxy refers to a “substituted hydroxyl” of the formula (—OR i ), wherein R i is an optionally substituted heteroaryl group, as defined herein, and the oxygen moiety is directly attached to the parent molecule.
  • heteroarylthioxy refers to a “substituted thiol” of the formula (—SR r ), wherein R r is an optionally substituted heteroaryl group, as defined herein, and the sulfur moiety is directly attached to the parent molecule.
  • hydroxy refers to a group of the formula (—OH).
  • a “substituted hydroxyl” refers to a group of the formula (—OR i ), wherein R i can be any substituent which results in a stable moiety (e.g., a suitable hydroxyl protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, nitro, alkylaryl, arylalkyl, and the like, each of which may or may not be further substituted).
  • imino refers to a group of the formula ( ⁇ NR r ), wherein R r corresponds to hydrogen or any substituent as described herein, that results in the formation of a stable moiety (for example, a suitable amino protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, amino, hydroxyl, alkylaryl, arylalkyl, and the like, each of which may or may not be further substituted).
  • imino refers to ⁇ NH wherein R r is hydrogen.
  • isocyano refers to a group of the formula (—NC).
  • nitro refers to a group of the formula (—NO 2 ).
  • oxo refers to a group of the formula ( ⁇ O).
  • stable moiety preferably refers to a moiety which possess stability sufficient to allow manufacture, and which maintains its integrity for a sufficient period of time to be useful for the purposes detailed herein.
  • a “protecting group,” as used herein, is well known in the art and include those described in detail in Greene's Protective Groups in Organic Synthesis , P. G. M. Wuts and T. W. Greene, 4 th edition, Wiley-Interscience, 2006, the entirety of which is incorporated herein by reference.
  • Suitable amino-protecting groups include methyl carbamate, ethyl carbamante, 9-fluorenylmethyl carbamate (Fmoc), 9-(2-sulfo)fluorenylmethyl carbamate, 9-(2,7-dibromo)fluoroenylmethyl carbamate, 2,7-di-t-butyl-[9-(10,10-dioxo-10,10,10,10-tetrahydrothioxanthyl)]methyl carbamate (DBD-Tmoc), 4-methoxyphenacyl carbamate (Phenoc), 2,2,2-trichloroethyl carbamate (Troc), 2-trimethylsilylethyl carbamate (Teoc), 2-phenylethyl carbamate (hZ), 1-(1-adamantyl)-1-methylethyl carbamate (Adpoc), 1,1-dimethyl-2-haloethyl carbamate, 1,1-d
  • Suitable hydroxyl protecting groups include methyl, methoxylmethyl (MOM), methylthiomethyl (MTM), t-butylthiomethyl, (phenyldimethylsilyl)methoxymethyl (SMOM), benzyloxymethyl (BOM), p-methoxybenzyloxymethyl (PMBM), (4-methoxyphenoxy)methyl (p-AOM), guaiacolmethyl (GUM), t-butoxymethyl, 4-pentenyloxymethyl (POM), siloxymethyl, 2-methoxyethoxymethyl (MEM), 2,2,2-trichloroethoxymethyl, bis(2-chloroethoxy)methyl, 2-(trimethylsilyl)ethoxymethyl (SEMOR), tetrahydropyranyl (THP), 3-bromotetrahydropyranyl, tetrahydrothiopyranyl, 1-
  • the protecting groups include methylene acetal, ethylidene acetal, 1-t-butylethylidene ketal, 1-phenylethylidene ketal, (4-methoxyphenyl)ethylidene acetal, 2,2,2-trichloroethylidene acetal, acetonide, cyclopentylidene ketal, cyclohexylidene ketal, cycloheptylidene ketal, benzylidene acetal, p-methoxybenzylidene acetal, 2,4-dimethoxybenzylidene ketal, 3,4-dimethoxybenzylidene acetal, 2-nitrobenzylidene acetal, methoxymethylene acetal, ethoxymethylene acetal, dimethoxymethylene ortho ester, 1-methoxyethylidene ortho ester,
  • the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences , 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate,
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate, and aryl sulfonate.
  • the term “subject,” as used herein, refers to any animal. In certain embodiments, the subject is a mammal. In certain embodiments, the term “subject”, as used herein, refers to a human (e.g., a man, a woman, or a child). The human may be of either sex and may be at any stage of development. In certain embodiments, the subject has been diagnosed with the condition or disease to be treated (e.g., macular degeneration, stroke, IBD, closed head injury). In other embodiments, the subject is at risk of developing the condition or disease. In certain embodiments, the subject is an experimental animal (e.g., mouse, rat, dog, primate). The experimental animal may be genetically engineered. In certain embodiments, the subject is a domesticated animal (e.g., dog, cat, bird, horse, cow, goat, sheep).
  • a domesticated animal e.g., dog, cat, bird, horse, cow, goat, sheep.
  • administer refers to implanting, absorbing, ingesting, injecting, or inhaling a compound of Formula (I) or a pharmaceutical composition thereof.
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more signs or symptoms thereof, described herein.
  • treatment may be administered after one or more signs or symptoms have developed or have been observed.
  • treatment may be administered in the absence of signs or symptoms of the disease or condition.
  • treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to delay or prevent recurrence.
  • an inventive compound that, when administered to a subject, is effective to at least partially treat a condition from which the subject is suffering (e.g., chronic inflammatory disease, autoimmune disease, dry eye syndrome, fibrosis, scar formation, angiogenesis, viral infection, malaria, ischemic damage, transplant and implant rejection, neurodegenerative disease, or a cosmetic indication).
  • a condition from which the subject is suffering e.g., chronic inflammatory disease, autoimmune disease, dry eye syndrome, fibrosis, scar formation, angiogenesis, viral infection, malaria, ischemic damage, transplant and implant rejection, neurodegenerative disease, or a cosmetic indication.
  • focal iron overload refers to any disease or condition that involves the accumulation of unmanaged iron in a tissue or organ. Focal iron overload typically involves less than the subject's whole body but may involve more than one organ or tissue. Unmanaged iron in any tissue or organ is typically undesired and can be the focus of the treatments of the present invention. The treatment may involve the removal of as much iron as possible from the tissue or organ or may only involve the removal of excess iron. Examples of disease and conditions associated with focal iron overload include, but are not limited to, macular degeneration, IBD, reperfusion injury, stroke including hemorrhagic stroke, and closed head injury; however, any disease or condition of focal iron overload may be treated as described herein.
  • focal iron overload does not include diseases or conditions associated with global iron overload (e.g., global iron overload associated with chronic transfusion therapy, hereditary hemochromatosis, etc.).
  • the treatment of focal iron overload may be systemic or local administration of an effective amount of a compound of Formula (I).
  • reactive oxygen species refers to molecules or ions formed by the incomplete reduction of oxygen.
  • Reactive oxygen species include superoxide anion (O 2 ⁇ ), peroxides such as hydrogen peroxide (H 2 O 2 ), hydroxyl radical (HO ⁇ ), and hypochlorous acid (HClO). These molecules are typically chemically reactive.
  • Reactive oxygen species may be formed by any number of mechanisms (e.g. enzymatically, by ionizing radiation, by reaction oxygen with a metal).
  • the reactive oxygen species are formed by the reduction of oxygen by an iron ion such as Fe +2 .
  • closed head injury refers to any injury to the head that does not penetrate the skull. Closed head injuries may result from falls, blasts, accidents including vehicular accidents, and assaults. Closed head injuries can lead to hemorrhage or brain swelling, which can result in increased intracranial pressure, which can in turn lead to permanent brain damage or even death.
  • Various types of closed head injury include concussions, brain contusions, diffuse axonal injury, and hematomas.
  • tautomer refers to a particular isomer of a compound in which a hydrogen and double bond have changed position with respect to the other atoms of the molecule. For a pair of tautomers to exist there must be a mechanism for interconversion. Examples of tautomers include keto-enol forms, imine-enamine forms, amide-imino alcohol forms, amidine-aminidine forms, nitroso-oxime forms, thio ketone-enethiol forms, N-nitroso-hydroxyazo forms, nitro-aci-nitro forms, lactam-lactim forms, ketene-ynol forms, enamine-enamine forms, and pyridione-hydroxypyridine forms.
  • FIG. 1 shows representative colons from rats treated with desferrioxamine (DFO) (20 mg/kg) IV and sterile water (control) 30 minutes before the 4% acetic acid. Each colon is positioned such that the cecum is on the right and the rectum is on the left.
  • DFO desferrioxamine
  • FIG. 2 is a bar graph showing the concentration of Deferitrin, (S)-4′-(HO)-DADFT-PE (IV-A), and (S)-4′-(HO)-DADFT-norPE (III-A) in plasma at various time points after a 300 ⁇ mol/kg oral (po) dose of each compound in rats.
  • FIG. 3 is a bar graph showing the concentration of Deferitrin, (S)-4′-(HO)-DADFT-PE (IV-A), and (S)-4′-(HO)-DADFT-norPE (III-A) in plasma at various time points after a 300 ⁇ mol/kg subcutaneous (sc) dose of each compound in rats.
  • FIG. 4 shows the correlation between plasma concentration and concentration in the eye for (S)-4′-(HO)-DADFT-norPE (III-A) administered 300 ⁇ mol/kg subcutaneously (SC) in perfused rats.
  • FIG. 5 shows the correlation between plasma concentration and concentration in the eye for (S)-4′-(HO)-DADFT-norPE (III-A) administered 300 ⁇ mol/kg subcutaneously (SC) in non-perfused rats.
  • desferrithiocin analogs of Formula (I), particularly those with a polyether moiety at the 4′-position of the phenyl ring, are expected to be useful in the treatment of macular degeneration, closed head injury, reperfusion injury, and stroke.
  • the compounds of Formula (I) are thought to chelate iron and prevent it from participating in the generation of reactive oxygen species.
  • the compounds of Formula (I) may also act as free radical scavenger thereby limiting the damage of reactive oxygen species or other radicals.
  • the invention therefore, provides methods of treating and preventing disease and conditions associated with focal iron overload, as well as pharmaceutical compositions and kits useful in the inventive methods.
  • Desferrithiocin analogs of Formula (I) are expected to be useful in preventing and treating diseases and conditions associated with iron overload, particularly focal iron overload. Such analogs have been previously described in International PCT Applications, PCT/US2006/010945, filed Mar. 22, 2006, WO2006/017626, and PCT/US2010/002336, filed Aug. 25, 2010, published as WO2011/028255; and U.S. Ser. No. 11/973,001, filed Oct. 4, 2007, published as US2008/0214630; each of which is incorporated herein by reference. Compounds with a polyether moiety at the 4′-position of the phenyl ring are expected to be particularly useful in the methods and compositions of the present invention.
  • compounds useful in the present invention are of Formula (I):
  • R 1 is hydrogen, alkyl, or acyl
  • R 2 is hydrogen, alkyl, or —[(CH 2 ) n —O] x —[(CH 2 ) n —O] y —R′;
  • R 3 , R 4 , and R 5 are each independently hydrogen, alkyl, arylalkyl, or —OR 10 ;
  • R 6 , R 7 , and R 8 are each independently hydrogen, halogen, alkyl, or —OR 12 ;
  • R 9 is —OR 11 or —SR 11 ;
  • R 10 is hydrogen, alkyl, or acyl
  • R 11 is hydrogen or alkyl
  • R 12 is hydrogen or alkyl
  • R′ is alkyl
  • R 1 is hydrogen, alkyl, or acyl. In certain embodiments, R 1 is hydrogen. In certain embodiments, R 1 is C 1 -C 6 alkyl. In certain embodiments, R 1 is methyl. In certain embodiments, R 1 is ethyl. In certain embodiments, R 1 is propyl. In certain embodiments, R 1 is acyl. In certain embodiments, R 1 is acetyl.
  • R 2 is hydrogen, alkyl, or —[(CH 2 ) n —O] x [(CH 2 ) n —O] y —R′.
  • R 2 is hydrogen.
  • R 2 is C 1 -C 6 alkyl.
  • R 2 is methyl.
  • R 2 is ethyl.
  • R 2 is propyl.
  • R 2 is —[(CH 2 ) n —O] x —[(CH 2 ) n —O] y —R′.
  • R 2 is —[(CH 2 ) 2 —O]—CH 3 .
  • R 2 is —[(CH 2 ) 2 —O] 2 —CH 3 . In certain embodiments, R 2 is —[(CH 2 ) 2 —O] 3 —CH 3 . In certain embodiments, R 2 is —[(CH 2 ) 2 —O] 4 —CH 3 . In certain embodiments, R 2 is —[(CH 2 ) 2 —O] 5 —CH 3 .
  • n is an integer from 1 to 8, inclusive. In certain embodiments, n is 2. In certain embodiments, n is 3. In certain embodiments, n is 4. In certain embodiments, n is 5.
  • x is an integer from 1 to 8, inclusive. In certain embodiments, x is 1. In certain embodiments, x is 2. In certain embodiments, x is 3. In certain embodiments, x is 4. In certain embodiments, x is 5.
  • y is an integer from 0 to 8, inclusive. In certain embodiments, y is 0. In certain embodiments, y is 1. In certain embodiments, y is 2. In certain embodiments, y is 3. In certain embodiments, y is 4. In certain embodiments, y is 5.
  • R′ is alkyl. In certain embodiments, R′ is C 1 -C 6 alkyl. In certain embodiments, R′ is methyl. In certain embodiments, R′ is ethyl. In certain embodiments, R′ is propyl.
  • R 3 is each independently hydrogen, alkyl, arylalkyl, or —OR 10 .
  • R 3 is hydrogen.
  • R 3 is C 1 -C 6 alkyl.
  • R 3 is methyl.
  • R 3 is ethyl.
  • R 3 is propyl.
  • R 3 is —OH.
  • R 3 is —OCH 3 .
  • R 4 is each independently hydrogen, alkyl, arylalkyl, or —OR 10 .
  • R 4 is hydrogen.
  • R 4 is C 1 -C 6 alkyl.
  • R 4 is methyl.
  • R 4 is ethyl.
  • R 4 is propyl.
  • R 4 is —OH.
  • R 4 is —OCH 3 .
  • R 5 is each independently hydrogen, alkyl, arylalkyl, or —OR 10 .
  • R 5 is hydrogen.
  • R 5 is C 1 -C 6 alkyl.
  • R 5 is methyl.
  • R 5 is ethyl.
  • R 5 is propyl.
  • R 5 is —OH.
  • R 5 is —OCH 3 .
  • R 3 , R 4 , and R 5 are all hydrogen. In certain embodiments, at least one of R 3 , R 4 , and R 5 is hydrogen. In certain embodiments, at least two of R 3 , R 4 , and R 5 are hydrogen.
  • R 6 is hydrogen, halogen, alkyl, or —OR 12 .
  • R 6 is hydrogen.
  • R 6 is halogen.
  • R 6 is C 1 -C 6 alkyl.
  • R 6 is methyl.
  • R 6 is ethyl.
  • R 6 is propyl.
  • R 6 is —OH.
  • R 6 is —OCH 3 .
  • R 7 is hydrogen, halogen, alkyl, or —OR 12 .
  • R 7 is hydrogen.
  • R 7 is halogen.
  • R 7 is C 1 -C 6 alkyl.
  • R 7 is methyl.
  • R 7 is ethyl.
  • R 7 is propyl.
  • R 7 is —OH.
  • R 7 is —OCH 3 .
  • both R 6 and R 7 are hydrogen. In certain embodiments, at least one of R 6 and R 7 is hydrogen.
  • R 8 is hydrogen, halogen, alkyl, or —OR 12 .
  • R 8 is hydrogen.
  • R 8 is halogen.
  • R 8 is C 1 -C 6 alkyl.
  • R 8 is methyl.
  • R 8 is ethyl.
  • R 8 is propyl.
  • R 8 is —OH.
  • R 8 is —OCH 3 .
  • R 9 is —OR 11 or —SR 11 , wherein R 11 is hydrogen or alkyl.
  • R 9 is —OH.
  • R 9 is —OCH 3 .
  • R 9 is —OCH 2 CH 3 .
  • R 9 is —OCH(CH 3 ) 2 .
  • R 9 is —SCH 3 .
  • R 9 is —SCH 2 CH 3 .
  • R 9 is —SCH 2 CH 2 CH 3 .
  • R 9 is —SCH(CH 3 ) 2 .
  • R 9 is —SCH 2 CH(CH 3 ) 2 .
  • R 11 is hydrogen.
  • R 11 is C 1 -C 6 alkyl.
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 is methyl
  • R 3 is hydrogen
  • R 4 is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 is methyl
  • R 1 is hydrogen
  • R 3 is hydrogen
  • R 4 is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 is methyl
  • R 1 is hydrogen
  • R 3 is hydrogen
  • R 4 is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 is methyl
  • R 9 is —OH.
  • the compounds of Formula (I) may be provided in various salts forms.
  • the compound when R 9 is —OH, the compound may be provided as a carboxylate salt with a positively charged counterion.
  • the counterion is betaine, choline hydroxide, diethanolamine, diethylamine, ethanolamine, hydroxyethylmorpholine, 4-(2-hydroxyethyl morpholine), 1-(2-hydroxyethyl pyrrolidine), 1-(2-hydroxyethyl)-piperidine, 1,2-EDSA, HCl, H 2 SO 4 , MSA, p-TSA, hydroxyethyl pyrroldine, imidazone, lysine (e.g., L-lysine), arginine (e.g., L-arginine), histidine (e.g., L-histidine)N-methyl-D-glucamine (NMG), N,N′-dibenzyl-ethylenediamine, N,N′
  • the counterion is lysine. In certain embodiments, the counterion is N-methyl-D-glucamine (NMG). In certain embodiments, the counterion is tromethamine. In certain embodiments, the counterion is calcium. In certain embodiments, the counterion is magnesium. In certain embodiments, the counterion is potassium. In certain embodiments, the counterion is sodium, In certain embodiments, the counterion is zinc. In certain embodiments, the counterion is piperzine. In certain embodiments, the counterion is MgOH + . In certain embodiments, the counterion is ZnOH + .
  • a polymorph of a salt of a compound of Formula (I) is provided.
  • a polymorph of a magnesium salt of a compound of Formula (I) is provided.
  • a polymorph of a MgOH + salt of a compound of Formula (I) is provided.
  • a polymorph of a salt of a carboxylate compound of Formula (I), wherein R 9 is —OH is provided.
  • a polymorph of a magnesium salt of a carboxylate compound of Formula (I), wherein R 9 is —OH is provided.
  • a polymorph of a MgOH + salt of a carboxylate compound of Formula (I), wherein R 9 is —OH is provided.
  • the compound of Formula (I) is of Formula (III):
  • the compound of Formula (I) is of Formula (III-A):
  • a salt of a compound of Formula (III-A) is provided.
  • a magnesium hydroxide salt of Formula (III-A) is provided as shown in Formula (III-A′):
  • the compound of Formula (I) is of Formula (III-B):
  • the compound of Formula (I) is of Formula (III-C):
  • the compound of Formula (I) is of Formula (IV-A):
  • a salt of a compound of Formula (IV-A) is provided.
  • a magnesium hydroxide salt of Formula (IV-A) is provided as shown in Formula (IV-A′):
  • the compound of Formula (I) is of Formula (IV-B):
  • the compound of Formula (I) is of Formula (IV-C):
  • the compound of Formula (I) is of Formula (V-A):
  • the compound of Formula (I) is of Formula (V-B):
  • the compound of Formula (I) is of Formula (V-C):
  • the invention provides methods and pharmaceutical compositions for the treatment of macular degeneration.
  • the compounds of Formula (I) are able to get into the eye as shown in FIGS. 4 and 5 .
  • the compounds of Formula (I) are then able to chelate and remove iron from the eye thereby preventing Fe+ 2 from forming and generating reactive oxygen species.
  • the local accumulation of iron is thought to contribute to macular degeneration. Therefore, the removal of iron from the eye (including the retina) can prevent and treat macular degeneration.
  • the compound of Formula (I) or a pharmaceutical composition thereof may be administered systemically or ocularly.
  • the compound or composition is administered orally.
  • the compound or composition is administered to the eye using eyedrops or an ointment suitable for ocular administration.
  • the subject being treated for macular degeneration may be any type of animal.
  • the animal is a mammal.
  • the animal is a human.
  • the animal is a domesticated animal (e.g., dog, cat, pig, cow).
  • the animal is a research animal (e.g., mice, rat, dog, primate).
  • the exact amount of the compound of Formula (I) required to treat or prevent macular degeneration will vary from subject to subject, depending on the species, age, and general condition of the subject, the particular agent being administered, its mode of administration, and the like.
  • the compound is preferably formulated in a dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily dosage will be decided by a physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the severity of the macular degeneration; the specific compound be administered; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the particular compound being administered; the duration of the treatment; drugs used in combination or coincidental with the particular compound being administered; and like factors well known in the medical arts.
  • the daily dosage of the compound of Formula (I) for the treatment of macular degeneration in a subject may range from 0.01 mg/kg to 200 mg/kg per unit dosage. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 100 mg/kg.
  • the daily dosage ranges from 0.1 mg/kg to 50 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 20 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 1 mg/kg.
  • the compound or a composition thereof may be administered once a day to multiple times per day. In certain embodiments, a fraction of the daily dose is administered once, twice, three times, or four times daily. In other embodiments, the compound of a composition thereof is administered every other day, every third day, every week, every other week, or every month.
  • the compounds of Formula (I) and pharmaceutical compositions thereof are expected to be useful in the treatment of head injury, particularly those involving bleeding into the brain or other parts of the central nervous system.
  • the compounds of Formula (I) are thought to chelate the iron from red blood cells the blood resulting from the head injury, thereby preventing iron ions from generating reactive oxygen species.
  • a compound being used may or may not have the ability to cross the blood brain barrier.
  • the compound being used to treat a head injury in a subject is able to cross the blood brain barrier.
  • the compounds is not able to cross the blood brain barrier.
  • Certain compounds of Formula (I) have been found in the CSF after systemic administration (po and sc).
  • the injury comes in various forms and results from various causes.
  • the injury is an injury to the head that penetrates the skull.
  • the head injury being treated is a closed head injury, which does penetrate the skull. Closed head injuries results from a variety of causes including accidents including vehicular accidents, falls, and assaults. Types of closed head injuries include concussions, brain contusions, diffuse axonal injury, and hematoma.
  • the closed head injury being treated in the present invention include closed head injuries that result in blood outside the blood vessels of the brain.
  • the local accumulation of iron from the bleeding is thought to contribute to after effects of closed head injury. By assisting the clearance of iron from the brain the effects of the bleed are minimized.
  • the compound of Formula (I) or a pharmaceutical composition thereof may be administered systemically, for example, parenterally or orally.
  • the compound or composition is administered orally.
  • the compound or composition is administered parenterally (e.g., intravenously).
  • the subject being treated for a head injury may be any type of animal.
  • the animal is a mammal.
  • the animal is a human.
  • the animal is a domesticated animal (e.g., dog, cat, pig, cow).
  • the animal is a research animal (e.g., mice, rat, dog, primate).
  • the exact amount of the compound of Formula (I) required to treat a head injury will vary from subject to subject, depending on the species, age, and general condition of the subject, the particular agent being administered, its mode of administration, and the like.
  • the compound is preferably formulated in a dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily dose will be decided by a physician using sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the severity of the head injury; the specific compound be administered; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the particular compound being administered; the duration of the treatment; drugs used in combination or coincidental with the particular compound being administered; and like factors well known in the medical arts.
  • the daily dosage of the compound of Formula (I) for the treatment of a head injury in a subject may range from 0.01 mg/kg to 200 mg/kg per unit dosage. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 100 mg/kg.
  • the daily dosage ranges from 0.1 mg/kg to 50 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 20 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 1 mg/kg.
  • the compound or a composition thereof may be administered once a day to multiple times per day. In certain embodiments, a fraction of the daily dose is administered once, twice, three times, or four times daily. In other embodiments, the compound of a composition thereof is administered every other day, every third day, every week, every other week, or every month. In certain embodiments, the inventive treatment is stopped once the head injury is resolved, or it is thought the inventive treatment would no longer be beneficial. In certain embodiments, the treatment is stopped once the bleeding has been resolved in a subject with a head injury.
  • the present invention also provides for the treatment of stroke.
  • the inventive treatment typically leads to a better and/or faster recovery from stroke.
  • the stroke being treated may be either a ischemic stroke or a hemorrhagic stroke.
  • a compound of Formula (I) or composition thereof is administered to a subject to prevent or minimize the damage due to reperfusion injury after the blood supply to the affected part of the brain is restore.
  • the compound is thought to prevent the generation of reactive oxygen species by either chelating iron responsible for the generation of such species and/or quenching such radical species when they do occur.
  • hemorrhagic stroke the compound is thought to work by similar mechanisms although the sequestering of iron from the blood in the brain is probably the predominate mechanism by which the inventive treatment works.
  • the mechanism of action of the compound of Formula (I) is similar to that in the treatment of head injury.
  • the compound being used in the treatment may have the ability to cross the blood brain barrier. In certain embodiments, the compound has the ability to cross the blood brain barrier. In other embodiments, the compound does not have the ability to cross the blood brain barrier. In certain embodiments, when the subject has been diagnosed with an ischemic stroke, the compound used in the treatment can pass through the blood brain barrier.
  • the present invention may be useful in treating a subject after the subject has been diagnosed with having a stroke, or a subject who is susceptible to having a stroke may be administered a compound of Formula (I) or composition thereof to prevent or minimize the stroke's effects.
  • the compound is administered as quickly as possible after a subject has been diagnosed with having a stroke.
  • the compound is administered to the subject while the stroke is still occurring.
  • the compound or a composition thereof is administered to a subject who has a history of strokes or is susceptible to having a stroke because of the subject's underlying medical condition.
  • the compound or composition thereof may be administered once or multiple times in the treatment of stroke.
  • the compound of Formula (I) or a pharmaceutical composition thereof may be administered systemically, for example, parenterally or orally.
  • the compound or composition is administered orally.
  • the compound or composition is administered parenterally (e.g., intravenously).
  • the subject being treated for stroke may be any type of animal.
  • the animal is a mammal.
  • the animal is a human.
  • the animal is a domesticated animal (e.g., dog, cat, pig, cow).
  • the animal is a research animal (e.g., mice, rat, dog, primate).
  • the exact amount of the compound of Formula (I) required to treat a stroke will vary from subject to subject, depending on the species, age, and general condition of the subject, the particular agent being administered, its mode of administration, and the like.
  • the compound is preferably formulated in a dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily dose will be decided by a physician using sound medical judgment.
  • the daily dosage of the compound of Formula (I) for the treatment of a stroke in a subject may range from 0.01 mg/kg to 200 mg/kg per unit dosage. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 100 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 50 mg/kg.
  • the daily dosage ranges from 0.1 mg/kg to 20 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 1 mg/kg.
  • the compound or a composition thereof may be administered once a day to multiple times per day. In certain embodiments, a fraction of the daily dose is administered once, twice, three times, or four times daily. In other embodiments, the compound or a composition thereof is administered every other day, every third day, every week, every other week, or every month. Typically the compound or composition thereof is not administered after it is no longer thought to be beneficial, for example, when all the bleeding has been cleared in a hemorrhagic stroke.
  • Reactive oxygen species have been implicated in the pathogenesis of inflammatory bowel disease (IBD).
  • IBD inflammatory bowel disease
  • Grisham et al. “Neutophil-mediated mucosal injury. Role of reactive oxygen metabolites” Dig. Dis. Sci. 33:6S-15S, 1988; Allgayer “Clinical relevance of oxygen radicals in inflammatory bowel disease-facts and fashion” Klin. Klischr . 69:1001-1003, 1991; Ymamada et al. “Role of neutrophil-derived oxidants in the pathogenesis of intestinal inflammation” Klin. Sheffieldschr . 69:988-944, 1991; Babbs, “Oxygen radicals in ulcerative colitis” Free Radic. Biol. Med.
  • the present invention provides for the treatment of IBD.
  • DFO an iron chelator
  • DFO an iron chelator
  • the compounds used in the inventive treatment are thought to prevent or eliminate the generation of reactive oxygen species or other longer-lived, more stable radicals that may be responsible for the tissue damage and inflammation seen in subjects with IBD.
  • Another possible mechanism of action of the compounds useful in the invention is the chelation of metal, such as iron, which may contribute to the generation of reactive oxygen species, such as hydroxyl radicals and hydrogen peroxide, that cause cell damage.
  • the present invention may be useful in treating a subject diagnosed with IBD.
  • the treatment may be used to treat the subject long term or may be used to treat a subject with a fare up of IBD.
  • a therapeutically effective amount of a compound of Formula (I) or composition thereof is administered to a subject in need thereof to treat IBD.
  • treatment with a compound of Formula (I) leads to reduced levels of reactive oxygen species in the intestines, specifically the intestinal mucosa.
  • the compound or composition thereof may be administered to a subject once or multiple times in the treatment of IBD.
  • the compound of Formula (I) or a pharmaceutical composition thereof may be administered systemically, for example, parenterally or orally.
  • the compound or composition is administered orally.
  • the compound or composition is administered parenterally (e.g., intravenously).
  • the compound or a composition is administered rectally.
  • the subject being treated for IBD may be any type of animal.
  • the animal is a mammal.
  • the animal is a human.
  • the animal is a domesticated animal (e.g., dog, cat, pig, cow).
  • the animal is a research animal (e.g., mice, rat, dog, primate).
  • the animal is used in animal model of IBD (e.g., acetic acid-induced colitis in rats; see Fedorak et al., “Misoprostol provides a colonic mucosal protective effect during acetic acid-induced colitis in rats” Gastroenterology 98:615-625, 1990; MacPherson et al., “Experimental production of diffuse colitis in rats” Digestion 17:135-150, 1978).
  • IBD animal model of IBD
  • the exact amount of the compound of Formula (I) required to treat IBD will vary from subject to subject, depending on the species, age, and general condition of the subject, the particular agent being administered, its mode of administration, and the like.
  • the compound is preferably formulated in a dosage unit form for ease of administration and uniformity of dosage.
  • the total daily dose will be decided by a physician using sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the severity of IBD; the specific compound be administered; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the particular compound being administered; the duration of the treatment; drugs used in combination or coincidental with the particular compound being administered; and like factors well known in the medical arts.
  • the daily dosage of the compound of Formula (I) for the prevention or treatment of reperfusion injury in a subject may range from 0.01 mg/kg to 200 mg/kg per unit dosage. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 100 mg/kg.
  • the daily dosage ranges from 0.1 mg/kg to 50 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 20 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 1 mg/kg.
  • the compound or a composition thereof may be administered once a day to multiple times per day. In certain embodiments, a fraction of the daily dose is administered once, twice, three times, or four times daily. In other embodiments, the compound or a composition thereof is administered every other day, every third day, every week, every other week, or every month.
  • the present invention also provides for the treatment of reperfusion injury.
  • Reperfusion injury may occur in any area of the body where the blood supply has been compromised.
  • the reperfusion injury being treated occurs in the heart.
  • the reperfusion injury occurs in the brain, for example, as discussed above in the context of a stroke
  • the inventive treatment minimizes reperfusion injury once the blood supply to the affects organ or tissue is restored.
  • a compound of Formula (I) or composition thereof is administered to a subject who is suffering from ischemia of a tissue or organ.
  • the compound of Formula (I) is thought to prevent the generation of reactive oxygen species by either chelating iron responsible for the generation of such species and/or quenching such radical species when they do occur.
  • the present invention may be useful in treating a subject after the subject has been diagnosed with ischemia of a particular organ or tissue.
  • a therapeutically effective amount of a compound of Formula (I) or composition thereof is administered to a subject to prevent or minimize reperfusion injury.
  • the compound is administered as quickly as possible after a subject has been diagnosed with ischemia.
  • the compound is administered to the subject at risk of ischemia.
  • the compound or a composition thereof is administered to a subject who is about to undergo a procedure that may lead to ischemia of an organ or tissue (e.g., cardiac surgery).
  • the compound or a composition thereof is used to prevent reperfusion injury in a transplanted organ.
  • the compound or composition thereof is used to perfuse an isolated organ being prepared for donation.
  • the compound or composition thereof may be administered to a subject once or multiple times in the treatment of reperfusion injury.
  • the compound of Formula (I) or a pharmaceutical composition thereof may be administered systemically, for example, parenterally or orally.
  • the compound or composition is administered orally.
  • the compound or composition is administered parenterally (e.g., intravenously).
  • the compound or a composition is administered locally to the organ or tissue suffering from ischemia.
  • the subject being treated for reperfusion injury may be any type of animal.
  • the animal is a mammal.
  • the animal is a human.
  • the animal is a domesticated animal (e.g., dog, cat, pig, cow).
  • the animal is a research animal (e.g., mice, rat, dog, primate).
  • the exact amount of the compound of Formula (I) required to prevent or treat reperfusion injury will vary from subject to subject, depending on the species, age, and general condition of the subject, the particular agent being administered, its mode of administration, and the like.
  • the compound is preferably formulated in a dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily dose will be decided by a physician using sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the severity of the reperfusion injury; the specific compound be administered; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the particular compound being administered; the duration of the treatment; drugs used in combination or coincidental with the particular compound being administered; and like factors well known in the medical arts.
  • the daily dosage of the compound of Formula (I) for the prevention or treatment of reperfusion injury in a subject may range from 0.01 mg/kg to 200 mg/kg per unit dosage. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 100 mg/kg.
  • the daily dosage ranges from 0.1 mg/kg to 50 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 20 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the daily dosage ranges from 0.1 mg/kg to 1 mg/kg.
  • the compound or a composition thereof may be administered once a day to multiple times per day. In other embodiments, the compound or a composition thereof is administered every other day, every third day, every week, every other week, or every month. Typically the compound or composition thereof is not administered after it is no longer thought to be beneficial, for example, when the risk of reperfusion injury is over.
  • the present invention also provides pharmaceutical compositions of Formula (I) for the treatment of macular degeneration, closed head injury, stroke, IBD, and reperfusion injury.
  • the pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intraperitoneally, topically, bucally, ocularly, or the like, depending on the disease or condition being treated.
  • an agent of the invention may be administered orally or parenterally at dosage levels sufficient to deliver from about 0.001 mg/kg to about 200 mg/kg, about 0.001 mg/kg to about 100 mg/kg, about 0.01 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, preferably from about 0.1 mg/kg to about 40 mg/kg, preferably from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, and more preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • the desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • a compound of Formula (I) is administered at a dose that is below the dose at which the agent causes non-specific effects. In certain embodiments, a compound of Formula (I) is administered at a dose that does not cause generalized immunosuppression in a subject.
  • Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water
  • oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • agents of the invention are mixed with solubilizing agents such CREMOPHOR EL® (polyethoxylated castor oil), alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and combinations thereof.
  • solubilizing agents such as CREMOPHOR EL® (polyethoxylated castor oil), alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and combinations thereof.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • Sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol, or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active agent.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol, or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active agent.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active agent is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and gly
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active agents can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active agent may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations such as liniments, lotions, gels, applicants, oil-in-water or water-in-oil emulsions such as creams, ointments, or pastes; or solutions or suspensions such as drops.
  • Formulations for topical administration to the skin surface can be prepared by dispersing the drug with a dermatologically acceptable carrier such as a lotion, cream, ointment, or soap.
  • Useful carriers are capable of forming a film or layer over the skin to localize application and inhibit removal.
  • the agent can be dispersed in a liquid tissue adhesive or other substance known to enhance adsorption to a tissue surface.
  • hydroxypropylcellulose or fibrinogen/thrombin solutions can be used to advantage.
  • tissue-coating solutions such as pectin-containing formulations can be used.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of an agent to the body.
  • dosage forms can be made by dissolving or dispensing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the agent across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the agent in a polymer matrix or gel.
  • the carrier for a topical formulation can be in the form of a hydroalcoholic system (e.g., liquids and gels), an anhydrous oil or silicone based system, or an emulsion system, including, but not limited to, oil-in-water, water-in-oil, water-in-oil-in-water, and oil-in-water-in-silicone emulsions.
  • the emulsions can cover a broad range of consistencies including thin lotions (which can also be suitable for spray or aerosol delivery), creamy lotions, light creams, heavy creams, and the like.
  • the emulsions can also include microemulsion systems.
  • Other suitable topical carriers include anhydrous solids and semisolids (such as gels and sticks); and aqueous based mousse systems.
  • the compounds of Formula (I) and pharmaceutical compositions thereof can be employed in combination therapies, that is, the compounds and pharmaceutical compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures.
  • the particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder, or they may achieve different effects (e.g., control of any adverse effects).
  • the present invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the present invention, and in certain embodiments, includes an additional approved therapeutic agent for use as a combination therapy.
  • an additional approved therapeutic agent for use as a combination therapy can be a notice in the form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceutical products, which notice reflects approval by the agency of manufacture, use, or sale for human administration.
  • the desferrithiocin (DFT) analogs and salts thereof useful in the present invention can be prepared from readily available starting materials using methods known in the art.
  • DFT desferrithiocin
  • III-A (S)-4′-(HO)-DADFT-norPE
  • IV-A (S)-4′-(HO)-DADFT-PE
  • III-A (S)-4′-(HO)-DADFT-norPE
  • IV-A may be synthesized using methods described in International PCT Applications, PCT/US2006/010945, filed Mar. 22, 2006, published as WO 2006/107626, PCT/US2010/002336, filed Aug. 25, 2010, published as WO2011/028255, and U.S. Ser. No. 11/973,001, filed Oct. 4, 2007, each of which is incorporated herein by reference in its entirety.
  • the DFT analogs useful in the inventive methods were converted from the free acid form to the monosodium salt form. Water followed by one equivalent of sodium hydroxide was added to the DFT analog as a free acid. The resulting slurry was vortexed or sonicated until the DFT analog went into solution. More water was added, and the solution was vortexed or sonicated again. The formed yellow solution, having a pH about 7, was used as a sample solution in the following Examples. It is preferred that a fresh sample solution of the DFT analog is made shortly before the solution is used in an assay.
  • the cecum and proximal colon were returned to the abdominal cavity; the compound was allowed to remain in the gut for 30 min. Then, the cecum and proximal colon were exteriorized again. The rectal plug was removed, and the drug was gently expressed out of the colon.
  • Acetic acid (4%, 2 ml) was injected into the proximal colon over a 15- to 20-sec time period. The acid was allowed to remain in the gut until 1 min had passed (ie, 40-45 sec after the end of the acid administration).
  • the no-acid control rats received distilled water (2 ml), which was administered in the same manner as was the acetic acid. Air (10 ml) was then injected into the proximal colon to expel the acid or water.
  • the cecal/proximal colon ligature was removed, the gut was returned to the abdominal cavity, and the incisions were closed. The animals were allowed to recover overnight and were killed 24 hr later. The entire length of the colon was removed and assessed for damage both densitometrically and biochemically.
  • Gross damage was quantitated using Photoshop-based image analysis (version 5.0, Adobe Systems, Mountain View, Calif., USA) on an Apple iMac computer.
  • the Magic Wand tool in the Select menu of Photoshop was used to place the cursor on an area of obvious damage.
  • the tolerance level of the Magic Wand tool was set at 30.
  • the damaged areas were automatically selected by using the Similar command in the Select menu.
  • the Eyedropper tool was used to determine the range of the damage in the highlighted areas.
  • Individual colon images were copied to a blank Photoshop page.
  • the MagicWand tool with a tolerance set to 100, was used to select all of the pixels in the colon sample.
  • the Histogram tool which generates a graph in which each vertical line represents the number of pixels associated with a brightness level, was selected in the Image menu.
  • the Red channel was then selected; the darker (damaged areas) appear on the left side of the histogram and the lighter (normal) areas are on the right side.
  • the cursor was then placed on the histogram, the color range determined in an earlier step was selected, and the number of pixels encompassing that range and the percent damage were quantitated automatically.
  • Rats were anesthetized with ketamine/xylazine about 50 minutes after dosing. Blood of the rats was collected via cardiac puncture into vacutainer tubes containing buffered sodium citrate one hour post dose. The rats' eyes were removed. Any extraneous tissue was trimmed and discarded. The eyes were frozen. The entire eye was then processed and assessed for the concentration of the DFT analog. The whole blood was centrifuged, and the plasma was separated and frozen until the concentration of the DFT analog was determined.
  • Rats were anesthetized with ketamine/xylazine about 50 minutes after dosing. Blood of the rats was collected via cardiac puncture into vacutainer tubes containing buffered sodium citrate one hour post dose. The rats' abdomen and thorax were opened, and a portion of the sternum/ribs was removed. A 19-gauge needle was inserted into the left ventricle of the rats, and the right atrium was cut. About 100 ml of saline was perfused transcardially for five minutes. The perfusion was stopped, and the rats' eyes were removed. Any extraneous tissue was trimmed and discarded. The eyes were frozen. The entire eye was then processed and assessed for the concentration of the DFT analog. The whole blood was centrifuged, and the plasma was separated and frozen until the concentration of the DFT analog was determined.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US14/363,886 2011-12-16 2012-12-14 Uses of 4'-desferrithiocin analogs Abandoned US20140343110A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/363,886 US20140343110A1 (en) 2011-12-16 2012-12-14 Uses of 4'-desferrithiocin analogs
US15/424,557 US20170209420A1 (en) 2011-12-16 2017-02-03 Uses of 4'-desferrithiocin analogs

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161576920P 2011-12-16 2011-12-16
PCT/US2012/069795 WO2013090750A1 (fr) 2011-12-16 2012-12-14 Utilisation d'analogues de 4'-desferrithiocine
US14/363,886 US20140343110A1 (en) 2011-12-16 2012-12-14 Uses of 4'-desferrithiocin analogs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/069795 A-371-Of-International WO2013090750A1 (fr) 2011-12-16 2012-12-14 Utilisation d'analogues de 4'-desferrithiocine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/424,557 Division US20170209420A1 (en) 2011-12-16 2017-02-03 Uses of 4'-desferrithiocin analogs

Publications (1)

Publication Number Publication Date
US20140343110A1 true US20140343110A1 (en) 2014-11-20

Family

ID=48613217

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/363,886 Abandoned US20140343110A1 (en) 2011-12-16 2012-12-14 Uses of 4'-desferrithiocin analogs
US15/424,557 Abandoned US20170209420A1 (en) 2011-12-16 2017-02-03 Uses of 4'-desferrithiocin analogs
US17/321,210 Active 2032-12-24 US11931346B2 (en) 2011-12-16 2021-05-14 Uses of 4′-desferrithiocin analogs

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/424,557 Abandoned US20170209420A1 (en) 2011-12-16 2017-02-03 Uses of 4'-desferrithiocin analogs
US17/321,210 Active 2032-12-24 US11931346B2 (en) 2011-12-16 2021-05-14 Uses of 4′-desferrithiocin analogs

Country Status (12)

Country Link
US (3) US20140343110A1 (fr)
EP (2) EP3620161A1 (fr)
JP (4) JP6539046B2 (fr)
KR (1) KR102111176B1 (fr)
CN (2) CN113398126A (fr)
AU (4) AU2012352025B2 (fr)
BR (1) BR112014014544A2 (fr)
CA (1) CA2859406C (fr)
ES (1) ES2761200T3 (fr)
HK (1) HK1203355A1 (fr)
MX (1) MX368275B (fr)
WO (1) WO2013090750A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9174948B2 (en) 2007-03-15 2015-11-03 University Of Florida Research Foundation, Inc. Desferrithiocin polyether analogues
US9567309B2 (en) 2005-04-04 2017-02-14 University Of Florida Research Foundation, Inc. Desferrithiocin polyether analogues
US10010535B2 (en) 2013-11-22 2018-07-03 University Of Florida Research Foundation, Incorporated Desferrithiocin analogs and uses thereof
US10570104B2 (en) 2015-04-27 2020-02-25 University Of Florida Research Foundation, Incorporated Metabolically programmed metal chelators and uses thereof
US11931346B2 (en) 2011-12-16 2024-03-19 University Of Florida Research Foundation, Incorporated Uses of 4′-desferrithiocin analogs

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6795998B2 (ja) * 2017-02-10 2020-12-02 日本放送協会 Mtf測定装置およびそのプログラム

Family Cites Families (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3274207A (en) 1966-09-20 Processes for preparing thiazole carboxylic acids
US3809754A (en) 1968-12-31 1974-05-07 Medial De Toledo & Cie Method of treating diseases of the mucous membrane using compounds of a thiazolidine carboxylic acid and pharmaceutical preparations thereof
US3882110A (en) 1969-06-11 1975-05-06 Roussel Uclaf Novel 2-alkyl-5-thiazole-carboxylic acid derivatives
DE2045818A1 (de) 1969-09-17 1971-04-08 Godo Shusei Kabushiki Kaisha, Tokio Aeruginoinsäure und Verfahren zu ihrer Herstellung
FR2141526B1 (fr) 1971-06-14 1975-05-30 Roussel Uclaf
DE2245560A1 (de) 1972-09-16 1974-03-21 Basf Ag Verfahren zur herstellung von 4alkoxycarbonyl-2-thiazolinen
US4430344A (en) 1978-04-08 1984-02-07 Santen Pharmaceutical Co., Ltd. Antihypertensive 4-thiazolidinecarboxylic acids
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
DE3002989A1 (de) 1980-01-29 1981-07-30 Hoechst Ag, 6000 Frankfurt Hydroxyphenyl-thiazol, -thiazolin und -thiazolidin-carbonsaeuren, verfahren zu ihrer herstellung und ihre verwendung zur beeinflussung des kollagenstoffwechsels
EP0045281B1 (fr) 1980-07-28 1985-05-08 Ciba-Geigy Ag Dérivés de la thiazoline et leurs procédés de préparation
JPS5758682A (en) 1980-07-28 1982-04-08 Ciba Geigy Ag Thiazoline derivatives, manufacture and medicinal composition containing same
JPS5772975A (en) 1980-10-24 1982-05-07 Kureha Chem Ind Co Ltd 2-substituted phenylthiazole derviative, and medicine for peptic ulcer containing said derivative as active component
US4367233A (en) 1981-10-02 1983-01-04 American Home Products Corporation Inhibitors of mammalian collagenase
EP0226591B1 (fr) 1984-12-18 1992-09-23 Clark Pharmaceutical Development, Ltd. Compositions antitumorales contenant le produit de reaction d'un aldehyde cytotoxique avec de la penicillamine et leur utilisation pour la production des compositions
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
HU200177B (en) 1985-06-18 1990-04-28 Biogal Gyogyszergyar Process for producing thiazolidinecarboxylic acids and pharmaceutical compositions comprising such compounds
US4736060A (en) 1985-08-06 1988-04-05 Nippon Rikagakuyakuhin Co., Ltd. Method for optical resolution of DL-cysteine and (R,S)-1-(1-naphthyl) ethylamine
EP0214101A3 (fr) 1985-09-03 1989-05-31 Ciba-Geigy Ag Utilisation des fer(III) chelateurs du type desferrioxamine-B et desferriferrithiocine pour le traitement du paludisme
EP0214933A3 (fr) 1985-09-03 1989-05-24 Ciba-Geigy Ag Préparations à base de mélanges pour le traitement de la malaria
JPS62142168A (ja) 1985-10-16 1987-06-25 Mitsubishi Chem Ind Ltd チアゾ−ル誘導体及びそれを有効成分とするロイコトリエンきつ抗剤
DE3621540A1 (de) 1986-06-27 1988-01-07 Basf Ag Chinolinderivate, verfahren zu ihrer herstellung, diese enthaltende mikrobizide und ihre verwendung zur bekaempfung von bakterien und pilzen
CA1283827C (fr) 1986-12-18 1991-05-07 Giorgio Cirelli Dispositif pour l'injection de formules liquides
GB8704027D0 (en) 1987-02-20 1987-03-25 Owen Mumford Ltd Syringe needle combination
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
DE3735757A1 (de) 1987-10-22 1989-05-03 Degussa Optisch aktive salze aus einem substituierten thiazolidin-4-carboxylat und 3-chlor-2-hydroxypropyltrimethylammonium, deren herstellung und verwendung
ES2047149T3 (es) 1988-01-20 1994-02-16 Ciba Geigy Ag Procedimiento para la obtencion de compuestos complejos.
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
FR2638359A1 (fr) 1988-11-03 1990-05-04 Tino Dalto Guide de seringue avec reglage de la profondeur de penetration de l'aiguille dans la peau
US5182402A (en) 1988-11-07 1993-01-26 Imperial Chemical Industries Plc Herbicidal compositions
GB8826035D0 (en) 1988-11-07 1988-12-14 Ici Plc Herbicidal compositions
US5106992A (en) 1989-07-28 1992-04-21 E. R. Squibb & Sons, Inc. 3,5-dihydroxypentanoic acid derivatives useful as antihypercholesterolemic agents and method for preparing same
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
NL9001955A (nl) 1990-09-05 1992-04-01 Cedona Pharm Bv Nieuwe thiazolidinederivaten.
US5393777A (en) 1990-11-15 1995-02-28 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon Health Sciences University Deferration using anguibactin siderophore
ATE142494T1 (de) 1990-11-30 1996-09-15 Teijin Ltd 2-arylthiazolderivat sowie dieses enthaltendes arzneimittel
DK0934937T3 (da) 1990-11-30 2002-04-02 Otsuka Pharma Co Ltd Azolderivater som superoxidradikalinhibitor
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
FR2677021B1 (fr) 1991-05-31 1993-10-01 Upsa Laboratoires Nouveaux derives de thiazole antagonistes des recepteurs a l'angiotensine ii; leurs procedes de preparation, compositions pharmaceutiques les contenant.
GB9118204D0 (en) 1991-08-23 1991-10-09 Weston Terence E Needle-less injector
SE9102652D0 (sv) 1991-09-13 1991-09-13 Kabi Pharmacia Ab Injection needle arrangement
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
AU5602594A (en) 1992-11-16 1994-06-08 University Of Florida Research Foundation, Inc. 2-(pyrid-2'-yl)-2-thiazoline-4(s)-carboxylic acid derivatives
US5840739A (en) 1992-11-16 1998-11-24 University Of Florida Research Foundation, Inc. Thiazoline acid derivatives
DE4301356A1 (de) 1993-01-20 1994-07-21 Basf Ag Verfahren zur Herstellung von 2-Alkyl-4-fluormethyl-thiazolcarbonsäure-alkylestern
WO1995024176A1 (fr) 1994-03-07 1995-09-14 Bioject, Inc. Dispositif de remplissage d'ampoule
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
FR2749583B1 (fr) 1996-06-07 1998-08-21 Lipha Nouveaux derives de thiazolidine -2,4- dione substitues, leurs procedes d'obtention et les compositions pharmaceutiques en renfermant
US20020049316A1 (en) 1997-04-29 2002-04-25 Halbert Stacie Marie Protease inhibitors
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
MA26618A1 (fr) 1998-04-09 2004-12-20 Smithkline Beecham Corp Composes et compositions pharmaceutiques pour le traitement du paludisme
US6147070A (en) 1998-06-05 2000-11-14 Facchini; Francesco Methods and compositions for controlling iron stores to treat and cure disease states
US6083966A (en) 1998-08-31 2000-07-04 University Of Florida Thiazoline acid derivatives
US6159983A (en) 1998-09-18 2000-12-12 University Of Florida Method and composition for treatment of inflammatory bowel disease
CN1585639A (zh) 1998-09-21 2005-02-23 佛罗里达大学研究基金会 抗疟疾制剂
US6251927B1 (en) 1999-04-20 2001-06-26 Medinox, Inc. Methods for treatment of sickle cell anemia
DE19948434A1 (de) 1999-10-08 2001-06-07 Gruenenthal Gmbh Substanzbibliothek enthaltend bicyclische Imidazo-5-amine und/oder bicyclische Imidazo-3-amine
AU2002323368B2 (en) 2001-08-23 2006-12-07 The Government Of The United States Of America As Represented By The Secretary, Department Of Health & Human Services Methods of inhibiting formation of vascular channels and profileration using pyridinone derivatives
US7160855B2 (en) 2002-03-14 2007-01-09 Children's Hospital & Research Center At Oakland Enhancement of iron chelation therapy
US20040132789A1 (en) * 2002-08-22 2004-07-08 University Of Florida Antioxidant and radical scavenging activity of synthetic analogs of desferrithiocin
US20040044220A1 (en) 2002-08-22 2004-03-04 University Of Florida Antioxidant and radical scavenging activity of synthetic analogs of desferrithiocin
AU2003270473A1 (en) 2003-09-09 2005-04-27 University Of Florida Desferrithiocin derivatives and their use as iron chelators
WO2005023310A2 (fr) 2003-09-09 2005-03-17 University Of Florida Research Foundation, Inc. Conjugues polyamine-chelateur de metal
US7552880B2 (en) 2004-08-05 2009-06-30 Continental Automotive Systems Us, Inc. Fuel injector with a deep-drawn thin shell connector member and method of connecting components
WO2006055412A1 (fr) 2004-11-19 2006-05-26 Shiva Biomedical, Llc Procedes de traitement de la resistance a l’erythropoietine
ES2732625T3 (es) * 2005-04-04 2019-11-25 Univ Florida Análogos de poliéter de desferritiocina
US20080194518A1 (en) 2005-12-23 2008-08-14 MOOKERJEE Pradip Antimicrobial Compositions
CN101129381B (zh) 2006-08-25 2012-02-01 天津和美生物技术有限公司 含β-内酰胺类抗生素和离子螯合剂的抗生素复方
WO2008130395A2 (fr) 2006-12-12 2008-10-30 University Of Florida Agents de décorporation de l'actinide analogues de la desferrithiocine
CA2680592C (fr) 2007-03-15 2016-07-05 University Of Florida Research Foundation, Inc. Analogues de polyether de desferrithiocine
FR2922210A1 (fr) 2007-10-11 2009-04-17 Univ Louis Pasteur Etablisseme Nouveaux composes, preparation et utilisations
WO2009140215A2 (fr) 2008-05-11 2009-11-19 Geraghty, Erin Procédé pour traiter des infections bactériennes résistant au médicament et d'autres infections par du clioquinol, de la phanquinone et des composés apparentés
WO2009155088A1 (fr) 2008-05-30 2009-12-23 University Of Cincinnati Inhibition de la formation de biofilms à l'aide de chélateurs de zinc
CN103435569B (zh) 2008-07-14 2015-08-19 法罗肯生物科学公司 作为金属螯合剂的脱氮杂去铁硫辛聚醚类似物的新型盐和多晶型物
BR112012001761A2 (pt) 2009-07-27 2017-05-09 Ferrokin Bioscience Inc composto de formula i, composto farmacêutica e método de tratamento de uma condição mediada por metal em um indivduo
JP5925679B2 (ja) 2009-08-25 2016-05-25 ユニバーシティ オブ フロリダ リサーチ ファウンデーション,インコーポレイティド デスフェリチオシンポリエーテル類似体およびその使用
US9045440B2 (en) 2010-05-04 2015-06-02 Ferrokin Biosciences, Inc. Desazadesferrothiocin and desazadesferrothiocin polyether analogues as metal chelation agents
WO2012027794A2 (fr) 2010-09-01 2012-03-08 The Mental Health Research Institute Of Victoria Méthode de traitement et agents utiles pour celle-ci
AU2012347595A1 (en) 2011-12-09 2014-06-26 Ferrokin Biosciences, Inc. Oral formulations for treating metal overload
ES2761200T3 (es) * 2011-12-16 2020-05-19 Univ Florida Usos de análogos de 4'-desferritiocina
US20140323534A1 (en) 2011-12-16 2014-10-30 University Of Florida Research Foundation, Inc. Uses of 3'-desferrithiocin analogs
EP2888270B1 (fr) 2012-08-15 2019-10-02 Yissum Research Development Company of the Hebrew University of Jerusalem Ltd. Complexes de titane et de vanadium cytotoxiques
WO2014134701A1 (fr) 2013-03-07 2014-09-12 Kane Biotech Inc. Compositions antimicrobiennes-antibiofilm et procédés d'utilisation de celles-ci
AU2014352780A1 (en) 2013-11-22 2016-06-09 University Of Florida Research Foundation, Inc. Desferrithiocin analogs and uses thereof
DE102014002484A1 (de) 2014-02-19 2015-08-20 Lucas Automotive Gmbh Kraftfahrzeugbremse, insbesondere kombiniert hydraulisch und elektromechanisch betätigbare Kraftfahrzeugbremse, mit mehrstufiger Spindel
CA2984250A1 (fr) 2015-04-27 2016-11-03 University Of Florida Research Foundation, Incorporated Agents chelateurs metalliques programmes metaboliquement et leurs utilisations

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9567309B2 (en) 2005-04-04 2017-02-14 University Of Florida Research Foundation, Inc. Desferrithiocin polyether analogues
US9994535B2 (en) 2005-04-04 2018-06-12 University Of Florida Foundation, Inc. Desferrithiocin polyether analogues
US9174948B2 (en) 2007-03-15 2015-11-03 University Of Florida Research Foundation, Inc. Desferrithiocin polyether analogues
US9730917B2 (en) 2007-03-15 2017-08-15 University Of Florida Research Foundation, Incorporated Desferrithiocin polyether analogues
US11931346B2 (en) 2011-12-16 2024-03-19 University Of Florida Research Foundation, Incorporated Uses of 4′-desferrithiocin analogs
US10010535B2 (en) 2013-11-22 2018-07-03 University Of Florida Research Foundation, Incorporated Desferrithiocin analogs and uses thereof
US10570104B2 (en) 2015-04-27 2020-02-25 University Of Florida Research Foundation, Incorporated Metabolically programmed metal chelators and uses thereof

Also Published As

Publication number Publication date
AU2012352025A1 (en) 2014-07-17
MX2014007200A (es) 2014-11-25
JP2018027947A (ja) 2018-02-22
US20170209420A1 (en) 2017-07-27
MX368275B (es) 2019-09-26
CA2859406A1 (fr) 2013-06-20
AU2018202374A1 (en) 2018-04-26
KR102111176B1 (ko) 2020-05-15
AU2018202374B2 (en) 2019-12-19
JP2021185140A (ja) 2021-12-09
ES2761200T3 (es) 2020-05-19
CN104114170B (zh) 2021-07-06
EP2790697B1 (fr) 2019-10-23
US11931346B2 (en) 2024-03-19
US20220105078A1 (en) 2022-04-07
JP2019206537A (ja) 2019-12-05
AU2020201924A1 (en) 2020-04-02
EP2790697A1 (fr) 2014-10-22
BR112014014544A2 (pt) 2017-06-13
JP6539046B2 (ja) 2019-07-03
WO2013090750A1 (fr) 2013-06-20
KR20140103149A (ko) 2014-08-25
AU2012352025B2 (en) 2018-01-18
JP2015500839A (ja) 2015-01-08
CA2859406C (fr) 2020-08-25
EP2790697A4 (fr) 2015-08-12
HK1203355A1 (en) 2015-10-30
JP6987399B2 (ja) 2022-01-05
CN104114170A (zh) 2014-10-22
EP3620161A1 (fr) 2020-03-11
CN113398126A (zh) 2021-09-17
AU2021282444A1 (en) 2021-12-23

Similar Documents

Publication Publication Date Title
US11931346B2 (en) Uses of 4′-desferrithiocin analogs
US20140323534A1 (en) Uses of 3'-desferrithiocin analogs
US9284297B2 (en) Halofuginone analogs for inhibition of tRNA synthetases and uses thereof
US9073941B2 (en) Compounds and methods for treating tuberculosis infection
US8957075B2 (en) O-GlcNAc transferase inhibitors and uses thereof
US10010535B2 (en) Desferrithiocin analogs and uses thereof
US20110263569A1 (en) Ryanodine channel binders and uses thereof
US20120190659A1 (en) Angiogenesis inhibitors
JPWO2020005807A5 (fr)
US9550744B2 (en) Vitamin C prodrugs and uses thereof
US20180362574A1 (en) Methods for treating polycystic kidney disease and polycystic liver disease
WO2016154547A1 (fr) Utilisation de chélateurs de métaux dans la gestion de maladies infectieuses
US20220152036A1 (en) COMPOUNDS FOR USES IN PHARMACOLOGICAL INDUCTION OF HBF FOR TREATMENT OF SICKLE CELL DISEASE AND ß-THALASSEMIA
US20180161295A1 (en) Treatment of closed head injury and hemorrhagic stroke with hbed
WO2015077652A1 (fr) Analogues de la désazadesferrithiocine et leurs utilisations

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF FLORIDA RESEARCH FOUNDATION, INCORPO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BERGERON, RAYMOND J., JR.;REEL/FRAME:033412/0112

Effective date: 20120127

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF FLORIDA;REEL/FRAME:040569/0763

Effective date: 20140616

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION