US20090088448A1 - Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders - Google Patents

Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders Download PDF

Info

Publication number
US20090088448A1
US20090088448A1 US12/329,106 US32910608A US2009088448A1 US 20090088448 A1 US20090088448 A1 US 20090088448A1 US 32910608 A US32910608 A US 32910608A US 2009088448 A1 US2009088448 A1 US 2009088448A1
Authority
US
United States
Prior art keywords
alkyl
group
aryl
substituted
independently selected
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
US12/329,106
Inventor
Edmund Idris Graziani
Kevin Pong
Jerauld Skotnicki
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.)
Wyeth LLC
Original Assignee
Wyeth LLC
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 Wyeth LLC filed Critical Wyeth LLC
Priority to US12/329,106 priority Critical patent/US20090088448A1/en
Publication of US20090088448A1 publication Critical patent/US20090088448A1/en
Assigned to WYETH LLC reassignment WYETH LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: WYETH
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/14Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D223/16Benzazepines; Hydrogenated benzazepines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/12Antidiuretics, e.g. drugs for diabetes insipidus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present invention provides rapamycin derivatives and their use in the treatment of neurological disorders.
  • Ischemic stroke which accounts for 83% of all stroke cases (the remaining 17% are of the hemorrhagic-type) occurs in approximately 700,000 Americans each year, which equates to roughly 1 stroke every 45 seconds. Ischemic strokes occur as a result of an obstruction within a blood vessel supplying blood to the brain. The underlying condition for this type of obstruction is the development of fatty deposits lining the vessel walls, called atherosclerosis.
  • cerebral thrombosis which refers to a thrombus (blood clot) that develops at the clogged part of the vessel
  • cerebral embolism which refers generally to a blood clot that forms at another location in the circulatory system, usually the heart and large arteries of the upper chest and neck.
  • a portion of the blood clot breaks loose, enters the bloodstream and travels through the brain's blood vessels until it reaches vessels too small to let it pass.
  • Current therapies to treat ischemic stroke are limited. To date, the only approved drug for ischemic stroke is recombinant tissue plasminogen activator (rt-PA). rt-PA, which acts as a thrombolytic, has a limited therapeutic window of opportunity (3 hours), therefore allowing only 1-2% of all stroke patients to receive treatment. There are no marketed neuroprotectants agents for ischemic stroke.
  • Parkinson's disease is a neurodegenerative disease that is neuropathologically characterized by the selective degeneration of dopaminergic (DAergic) neurons of the substantia nigra.
  • PD is a progressive disease with a mean age at onset of 55, although 15% of patients are diagnosed before the age of 50. It is estimated that 1.5 million Americans have PD.
  • Some of the classical signs of PD are resting tremor on one side of the body, generalized slowness of movement (bradykinesia), stiffness of limbs (rigidity), gait or balance problems (postural dysfunction).
  • Current PD medications treat symptoms, whereas none prevent or retard DAergic neuron degeneration.
  • Neurotrophins are a family of proteins that have extraordinary therapeutic properties in pre-clinical models of neurodegeneration. Although experimentally promising, clinical development of neurotrophins was met with severe obstacles and setbacks, such as the inability to deliver these large proteins to target population of neurons, instability of the proteins, and non-specific activity.
  • the present invention provides novel compounds useful in treating, and in the preparation of medicaments useful in the treatment of, neurological disorders.
  • the present invention provides novel neuroprotective agents.
  • the present invention provides rapamycin derivatives, and pharmaceutically acceptable salts, prodrugs, and metabolites thereof.
  • the present invention provides methods of preparing rapamycin derivatives.
  • the present invention provides methods of treating neurological disorders, and the use of a compound of the invention in the preparation of a medicament therefor.
  • the present invention provides methods of treating complications due to stroke or head trauma, and the use of a compound of the invention in the preparation of a medicament therefor.
  • the present invention provides novel rapamycin derivatives which are useful as neuroprotective agents, particularly in compositions for use in treating neurological disorders.
  • the neurological disorder including, e.g., a neurodegenerative or neuromuscular degenerative condition, can be a result of a genetic disorder present at birth, a disorder developed during the lifespan of an individual, e.g., stroke, and/or the result of physical trauma, e.g., head, spinal injury, or injury to the peripheral nervous system.
  • a compound of the invention may be useful in ameliorating the symptoms of a preexisting neurological disorder or preventing further neuro- and/or neuromuscular degeneration.
  • the neuroprotective agents of the invention can be used to delay the onset of symptoms associated with a neurological disorder.
  • the present invention provides rapamycin derivatives of the formula I:
  • R 1 and R 2 are the same or different and are selected from among CR 16 R 17 and CR 18 R 19 .
  • R 3 and R 4 are (a) independently selected from among H, OH, O(C 1 to C 6 alkyl), O(substituted C 1 to C 6 alkyl), O(acyl), O(aryl), O(substituted aryl), and halogen; or
  • R 5 , R 6 , and R 7 are independently selected among H, OH, and OCH 3 .
  • R 8 and R 9 are connected through a (i) single bond and are CH 2 or (ii) double bond and are CH.
  • R 15 is selected from among C ⁇ O, CHOH, and CH 2 .
  • R 16 and R 17 are selected from among H, C 1 to C 6 alkyl, C 1 to C 6 substituted alkyl, aryl, substituted aryl, acyl, CF 3 , CN, and NO 2 .
  • R 18 and R 19 are independently selected from among H, C 1 to C 6 alkyl, C 1 to C 6 substituted alkyl, aryl, substituted aryl, acyl, CF 3 , CN, and NO 2 or R 17 and R 18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring. Further, n is 1 or 2.
  • Additional compounds that can be prepared according to the present invention include compounds of the following structure:
  • R 1 , R 2 , and R 6 -R 9 are defined as noted above.
  • the present invention provides compounds where R 17 and R 18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring.
  • R 17 and R 18 include where each is an acyl substituted by alkoxy, e.g., —COOEt.
  • R 16 and R 19 may each be CN.
  • the present invention provides compounds where R 8 and R 9 are bound through a single bond.
  • the present invention provides compounds where R 3 or R 4 is OH.
  • the present invention provides compounds where R 3 or R 4 is —O(acyl), preferably where the substituted acyl is —C(O)-optionally substituted alkyl, in particular where alkyl can be straight or branched and optionally substituted, e.g., by heterocyclic such as aromatic heterocyclic such as pyridyl.
  • R 3 or R 4 is —O(acyl), preferably where the substituted acyl is —C(O)-optionally substituted alkyl, in particular where alkyl can be straight or branched and optionally substituted, e.g., by heterocyclic such as aromatic heterocyclic such as pyridyl.
  • heterocyclic such as aromatic heterocyclic such as pyridyl
  • the present invention provides compounds where R 5 , R 6 , and R 7 are OCH 3 . In another embodiment, the present invention provides compounds where n is 2. In yet a further embodiment, the present invention provides compounds where R 15 is C ⁇ O.
  • the compounds of the invention can contain one or more asymmetric carbon atoms and some of the compounds can contain one or more asymmetric (chiral) centers and can thus give rise to optical isomers and diastereomers. While shown without respect to stereochemistry, when the compounds can contain one or more chiral centers, preferably at least one of the chiral centers is of S-stereochemistry.
  • the invention includes such optical isomers and diastereomers; as well as the racemic and resolved, enantiomerically pure stereoisomers; as well as other mixtures of the R and S stereoisomers, and pharmaceutically acceptable salts, hydrates, metabolites, and prodrugs thereof.
  • alkyl is used herein to refer to both straight- and branched-chain saturated aliphatic hydrocarbon groups having 1 to 10 carbon atoms, and desirably about 1 to 8 carbon atoms.
  • alkenyl is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon double bonds and containing about 2 to 10 carbon atoms. In one embodiment, the term alkenyl refers to an alkyl group having 1 or 2 carbon-carbon double bonds and having 2 to about 6 carbon atoms.
  • alkynyl group is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon triple bond and having 2 to 8 carbon atoms. In one embodiment, the term alkynyl refers to an alkyl group having 1 or 2 carbon-carbon triple bonds and having 2 to 6 carbon atoms.
  • cycloalkyl is used herein to refer to an alkyl group as previously described that is cyclic in structure and has about 4 to 10 carbon atoms, or about 5 to 8 carbon atoms.
  • substituted alkyl refers to alkyl, alkenyl, and alkynyl groups, respectively, having one or more substituents including, without limitation, halogen, CN, OH, NO 2 , amino, aryl, heterocyclic, alkoxy, aryloxy, alkylcarbonyl, alkylcarboxy, and arylthio, which groups can be optionally substituted.
  • substituents can be attached to any carbon of an alkyl, alkenyl, or alkynyl group provided that the attachment constitutes a stable chemical moiety.
  • aryl refers to an aromatic system, e.g., of 6-20 carbon atoms, which can include a single ring or multiple aromatic rings fused or linked together where at least one part of the fused or linked rings forms the conjugated aromatic system.
  • the aryl groups can include, but are not limited to, phenyl, naphthyl, biphenyl, anthryl, tetrahydronaphthyl, phenanthryl, indene, benzonaphthyl, fluorenyl, and carbazolyl.
  • substituted aryl refers to an aryl group which is substituted with one or more substituents including halogen, CN, OH, NO 2 , amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio, which groups can be optionally substituted.
  • a substituted aryl group is substituted with 1 to 4 substituents including halogen, CN, OH, NO 2 , amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio.
  • heterocyclic refers to a stable 4- to 7-membered monocyclic or multicyclic heterocyclic ring which is saturated, partially unsaturated, or wholly unsaturated, including aromatic such as pyridyl.
  • the heterocyclic ring has carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms.
  • the heterocyclic ring has 1 to 4 heteroatoms in the backbone of the ring. When the heterocyclic ring contains nitrogen or sulfur atoms in the backbone of the ring, the nitrogen or sulfur atoms can be oxidized.
  • heterocyclic also refers to multicyclic rings, e.g., of 9 to 20 ring members in which a heterocyclic ring is fused to an aryl ring.
  • the heterocyclic ring can be attached to the aryl ring through a heteroatom or carbon atom, provided the resultant heterocyclic ring structure is chemically stable.
  • heterocyclic groups include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof.
  • Oxygen-containing rings include, but are not limited to, furyl, tetrahydrofuranyl, pyranyl, pyronyl, and dioxinyl rings.
  • Nitrogen-containing rings include, without limitation, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, pyridyl, piperidinyl, 2-oxopiperidinyl, pyridazinyl, pyrimidinyl, pyrazinyl, piperazinyl, azepinyl, triazinyl, pyrrolidinyl, and azepinyl rings.
  • Sulfur-containing rings include, without limitation, thienyl and dithiolyl rings.
  • Mixed heteroatom containing rings include, but are not limited to, oxathiolyl, oxazolyl, thiazolyl, oxadiazolyl, oxatriazolyl, dioxazolyl, oxathiazolyl, oxathiolyl, oxazinyl, oxathiazinyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, oxepinyl, thiepinyl, and diazepinyl rings.
  • Fused heteroatom-containing rings include, but are not limited to, benzofuranyl, thionapthene, indolyl, benazazolyl, purindinyl, pyranopyrrolyl, isoindazolyl, indoxazinyl, benzoxazolyl, anthranilyl, benzopyranyl, quinolinyl, isoquinolinyl, benzodiazonyl, naphthylridinyl, benzothienyl, pyridopyridinyl, benzoxazinyl, xanthenyl, acridinyl, and purinyl rings.
  • substituted heterocyclic refers to a heterocyclic group having one or more substituents including halogen, CN, OH, NO 2 , amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio, which groups can be optionally substituted.
  • a substituted heterocyclic group is substituted with 1 to 4 substituents.
  • acyl refers to a —C(O)— group, which is substituted at the carbon atom.
  • the acyl group can be substituted or a terminal acyl group such as an HC(O)-group.
  • the substituents can include any substituents noted above for alkyl groups, viz. one or more substituents including, without limitation, halogen, CN, OH, NO 2 , amino, aryl, heterocyclic, alkoxy, aryloxy, alkylcarbonyl, alkylcarboxy, and arylthio, which groups can be optionally substituted. Examples include —C(O)-alkoxy (eg. —OMe or —OEt) or —C(O)-alkyl where alkyl can be straight or branched and optionally substituted e.g., by heterocyclic (such as pyridyl).
  • alkoxy refers to the O(alkyl) group, where the point of attachment is through the oxygen-atom and the alkyl group is optionally substituted.
  • aryloxy refers to the O(aryl) group, where the point of attachment is through the oxygen-atom and the aryl group is optionally substituted.
  • alkyloxy refers to the alkylOH group, where the point of attachment is through the alkyl group.
  • arylthio refers to the S(aryl) group, where the point of attachment is through the sulfur-atom and the aryl group can be optionally substituted.
  • alkylcarbonyl refers to the C(O)(alkyl) group, where the point of attachment is through the carbon-atom of the carbonyl moiety and the alkyl group is optionally substituted.
  • alkylcarboxy refers to the C(O)O(alkyl) group, where the point of attachment is through the carbon-atom of the carboxy moiety and the alkyl group is optionally substituted.
  • aminoalkyl refers to both secondary and tertiary amines where the point of attachment is through the nitrogen-atom and the alkyl groups are optionally substituted.
  • the alkyl groups can be the same or different.
  • halogen refers to Cl, Br, F, or I groups.
  • the rapamycin derivatives of formula I of the present invention are prepared from a rapamycin starting material.
  • the rapamycin starting material includes, without limitation, rapamycin, norrapamycin, deoxorapamycin, desmethylrapamycins, or desmethoxyrapamycin, or pharmaceutically acceptable salts, prodrugs, or metabolites thereof.
  • rapamycin starting material includes, without limitation, rapamycin, norrapamycin, deoxorapamycin, desmethylrapamycins, or desmethoxyrapamycin, or pharmaceutically acceptable salts, prodrugs, or metabolites thereof.
  • rapamycin starting material includes, without limitation, rapamycin, norrapamycin, deoxorapamycin, desmethylrapamycins, or desmethoxyrapamycin, or pharmaceutically acceptable salts, prodrugs, or metabolites thereof.
  • suitable rapamycin starting material that can be utilized to prepare the novel rapamycin derivatives of the present invention.
  • desmethylrapamycin refers to the class of rapamycin compounds which lack one or more methyl groups.
  • Examples of desmethylrapamycins that can be used according to the present invention include 3-desmethylrapamycin (U.S. Pat. No. 6,358,969), 7-O-desmethyl-rapamycin (U.S. Pat. No. 6,399,626), 17-desmethylrapamycin [U.S. Pat. No. 6,670,168], and 32-O-desmethylrapamycin, among others.
  • rapamycin refers to the class of rapamycin compounds which lack one or more methoxy groups and includes, without limitation, 32-desmethoxyrapamycin.
  • the rapamycin derivatives of formula I of the present invention are therefore prepared by combining a rapamycin starting material and an optionally substituted olefin.
  • olefin refers to a molecule that contains a double bond.
  • a variety of olefins can be utilized in the present invention and can readily be selected by one of skill in the art and include diethylfumarate, among others including any olefin sufficiently activated with electron-withdrawing substituents to undergo a [4+2]cycloaddition at temperatures up to 50° C.
  • One of skill in the art would readily be able to select the amount of olefin that would be effective in preparing the rapamycin derivatives of the present invention.
  • an excess of the olefin is utilized, e.g., a 3:1 ratio of olefin to rapamycin starting material.
  • a 3:1 ratio of olefin to rapamycin starting material can be utilized as determined by one of skill in the art.
  • the olefin and rapamycin starting material is combined in a solvent.
  • the solvent preferably dissolves the olefin and/or rapamycin on contact, or dissolves the olefin and rapamycin as the reaction proceeds.
  • Solvents that can be utilized in the present invention include, without limitation, dimethylformamide, dioxane such as p-dioxane, chloroform, alcohols such as methanol and ethanol, ethyl acetate, water, acetonitrile, tetrahydrofuran, dichloromethane, and toluene, or combinations thereof.
  • the solution containing the olefin, rapamycin starting material, and solvent is maintained at elevated temperatures, and preferably a temperature that does not promote decomposition of the rapamycin and olefin.
  • the solution is maintained at a temperature of about 30 to about 80° C., and preferably about 50° C.
  • the components are heated for a period of time sufficient to permit reaction between the rapamycin starting material and olefin.
  • One of skill in the art using known techniques would readily be able to monitor the progress of the reaction during heating and thereby determine the amount of time required to perform the reaction.
  • the rapamycin and olefin are combined with a solvent and maintained at a temperature of about 50° C.
  • Isolation and purification of the rapamycin derivative is well within one of skill in the art and include chromatography including, without limitation, and recrystallization, high performance liquid chromatography (HPLC) such as reverse phase HPLC, and normal phase HPLC, and size-exclusion chromatography.
  • HPLC high performance liquid chromatography
  • the rapamycin derivative can be reduced to form a more saturated rapamycin derivative.
  • a suitable reducing agent for use in the present invention.
  • reduction of the rapamycin derivative can be effected using a hydrogenation agent.
  • a suitable hydrogenation agent for use in the present invention.
  • transition metal catalysts or transition metals on a support preferably a carbon support, among others, in the presence hydrogen gas, are utilized to carry out the reduction.
  • the reduction is performed using palladium metal on carbon in the presence of hydrogen gas.
  • Reduction of the rapamycin derivative is typically carried out in a solvent.
  • solvents can be utilized in the reduction and include, without limitation, alcohols such as methanol.
  • alcohols such as methanol.
  • one of skill in the art would readily be able to select a suitable solvent for use in the present invention and depending on the hydrogenation catalyst and rapamycin derivative being reduced.
  • the amount of solvent depends on the scale of the reaction, and specifically the amount of rapamycin derivative being reduced.
  • the amount of hydrogenation agent utilized in the present invention can readily be determined by one of skill in the art. However, one of skill in the art would be able to determine and adjust the amount of hydrogenation agent necessary to perform the reduction and to form the more saturated rapamycin derivatives of the present invention. Further, a variety of apparatuses can be utilized to perform the hydrogenation of the present invention and include Parr apparatuses, among others. The selection of the particular apparatus for the hydrogenation is well within one of skill in the art.
  • rapamycin derivatives of the present invention are prepared as summarized in Scheme 1 below:
  • R 4 -R 7 , R 15 -R 19 , and n are defined above.
  • the rapamycin derivatives of the present invention can be utilized in the form of pharmaceutically acceptable salts, prodrugs, or metabolites thereof derived from pharmaceutically or physiologically acceptable acids or bases.
  • These salts include, but are not limited to, the following salts with mineral or inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid and organic acids such as acetic acid, oxalic acid, succinic acid, and maleic acid.
  • Other salts include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium in the form of esters, carbamates and other conventional “pro-drug” forms, which, when administered in such form, convert to the active moiety in vivo.
  • the rapamycin derivatives of formula I of the present invention have use in a variety of applications and include those relating to neurological disorders.
  • the compounds of the present invention are useful in treating neurological disorders including, but not limited to, Alzheimer's disease amyotrophic lateral sclerosis, epilepsy, Huntington's Disease, Parkinson's Disease, stroke, spinal cord injury, traumatic brain injury, Lewy body dementia, multiple sclerosis, Pick's disease, Niewmann-Pick disease, amyloid angiopathy, cerebral amyloid angiopathy, systemic amyloidosis, hereditary cerebral hemorrhage with amyloidosis of the Dutch type, inclusion body myositis, mild cognitive impairment and Down's syndrome, and in the preparation of medicaments therefor.
  • the rapamycin derivatives are also useful in treating complications due to stroke, head trauma, or spinal injury, or other injuries to the brain, peripheral nervous, central nervous, or neuromuscular system, and in the preparation of medicaments therefor
  • novel rapamycin derivatives are useful as neuroprotective agents.
  • the rapamycin derivatives of the present invention may also be useful as neuroregenerative agents, i.e. restoring some neurological and/or neuromuscular or other function following onset of one of the above conditions and/or injury, stroke, or other trauma, and in the preparation of medicaments therefor.
  • the dosage requirements of the rapamycin derivatives of the present invention can vary depending on the condition, severity of the symptoms presented and the particular subject being treated. One of skill in the art would readily be able to determine the amount of the rapamycin derivative required. In one embodiment, about 0.5 to 200 mg is administered. In a further embodiment, about 0.5 to 100 mg is administered. In another embodiment, about 0.5 to about 75 mg is administered. In yet a further embodiment, about 1 to about 25 mg is administered. In another embodiment, about 0.5 to about 10 mg is administered, particularly when used in combination with another agent. In yet a further embodiment, about 2 to about 5 mg is administered. In yet another embodiment, about 5 to about 15 mg is administered.
  • Treatment can be initiated with dosages of the rapamycin derivative smaller than those required to produce a desired effect and generally less than the optimum dose of the rapamycin derivative. Thereafter, the dosage can be increased until the optimum effect under the circumstances is reached. Precise dosages will be determined by the administering physician based on experience with the individual subject being treated.
  • the compositions of this invention are most desirably administered at a concentration that will generally afford effective results without causing any harmful or deleterious side effects.
  • the present invention includes methods of preparing a pharmaceutical composition containing one or more rapamycin derivatives of the present invention.
  • the composition can be administered to a mammalian subject by several different routes and is desirably administered orally in solid or liquid form.
  • Solid forms, including tablets, capsules, and caplets, containing the rapamycin derivative can be formed by blending the rapamycin derivative with one or more of the components described above.
  • the components of the composition are dry or wet blended.
  • the components are dry granulated.
  • the components are suspended or dissolved in a liquid and added to a form suitable for administration to a mammalian subject.
  • Liquid forms containing the rapamycin derivative can be formed by dissolving or suspending the rapamycin derivative in a liquid suitable for administration to a mammalian subject.
  • compositions containing the rapamycin derivative of the present invention can be prepared according to the present invention by combining the rapamycin derivative and a pharmaceutically acceptable carrier.
  • compositions described herein containing the rapamycin derivative can be formulated in any form suitable for the desired route of delivery using a pharmaceutically effective amount of the rapamycin derivative.
  • the compositions of the invention can be delivered by a route such as oral, dermal, transdermal, intrabronchial, intranasal, intravenous, intramuscular, subcutaneous, parenteral, intraperitoneal, intranasal, vaginal, rectal, sublingual, intracranial, epidural, intratracheal, or by sustained release.
  • delivery is oral.
  • the oral dosage tablet composition of this invention can also be used to make oral dosage tablets containing analogs of the rapamycin derivative, including, but not limited to, esters, carbamates, sulfates, ethers, oximes, carbonates, and the like which are known to those of skill in the art.
  • a pharmaceutically effective amount of the rapamycin derivative can vary depending on the specific compound(s), mode of delivery, severity of the condition being treated, and any other active ingredients used in the composition.
  • the dosing regimen can also be adjusted to provide the optimal therapeutic response.
  • Several divided doses can be delivered daily, e.g., in divided doses 2 to 4 times a day, or a single dose can be delivered. The dose can however be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • the delivery is on a daily, weekly, or monthly basis.
  • the delivery is on a daily delivery. However, daily dosages can be lowered or raised based on the periodic delivery.
  • the rapamycin derivatives of the present invention can be combined with one or more pharmaceutically acceptable carriers or excipients including, without limitation, solid and liquid carriers which are compatible with the compositions of the present invention.
  • Such carriers include adjuvants, syrups, elixirs, diluents, binders, lubricants, surfactants, granulating agents, disintegrating agents, emollients, metal chelators, pH adjustors, surfactants, fillers, disintegrants, and combinations thereof, among others.
  • the rapamycin derivative is combined with metal chelators, pH adjustors, surfactants, fillers, disintegrants, lubricants, and binders.
  • Adjuvants can include, without limitation, flavoring agents, coloring agents, preservatives, and supplemental antioxidants, which can include vitamin E, ascorbic acid, butylated hydroxytoluene (BHT) and butylated hydroxyanisole (BHA).
  • Binders can include, without limitation, cellulose, methylcellulose, hydroxymethylcellulose, carboxymethylcellulose calcium, carboxymethylcellulose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, microcrystalline cellulose, noncrystalline cellulose, polypropylpyrrolidone, polyvinylpyrrolidone (povidone, PVP), gelatin, gum arabic and acacia, polyethylene glycols, starch, sugars such as sucrose, kaolin, dextrose, and lactose, cholesterol, tragacanth, stearic acid, gelatin, casein, lecithin (phosphatides), cetostearyl alcohol, cetyl alcohol, cetyl esters wax, dextrates, dextrin, glyceryl monooleate, glyceryl monostearate, glyceryl palmitostearate, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polyoxyethylene stearates, polyvinyl alcohol,
  • Lubricants can include magnesium stearate, light anhydrous silicic acid, talc, stearic acid, sodium lauryl sulfate, and sodium stearyl furamate, among others.
  • the lubricant is magnesium stearate, stearic acid, or sodium stearyl furamate. In another embodiment, the lubricant is magnesium stearate.
  • Granulating agents can include, without limitation, silicon dioxide, microcrystalline cellulose, starch, calcium carbonate, pectin, crospovidone, and polyplasdone, among others.
  • Disintegrating agents or disintegrants can include croscarmellose sodium, starch, carboxymethylcellulose, substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate, calcium citrate, sodium starch glycolate, pregelatinized starch or crospovidone, among others.
  • the disintegrant is croscarmellose sodium.
  • Emollients can include, without limitation, stearyl alcohol, mink oil, cetyl alcohol, oleyl alcohol, isopropyl laurate, polyethylene glycol, olive oil, petroleum jelly, palmitic acid, oleic acid, and myristyl myristate.
  • Surfactants can include polysorbates, sorbitan esters, poloxamer, or sodium lauryl sulfate. In one embodiment, the surfactant is sodium lauryl sulfate.
  • Metal chelators can include physiologically acceptable chelating agents including edetic acid, malic acid, or fumaric acid. In one embodiment, the metal chelator is edetic acid.
  • pH adjusters can also be utilized to adjust the pH of a solution containing the rapamycin derivative to about 4 to about 6.
  • the pH of a solution containing the rapamycin derivative is adjusted to a pH of about 4.6.
  • pH adjustors can include physiologically acceptable agents including citric acid, ascorbic acid, fumaric acid, or malic acid, and salts thereof.
  • the pH adjuster is citric acid.
  • Fillers that can be used according to the present invention include anhydrous lactose, microcrystalline cellulose, mannitol, calcium phosphate, pregelatinized starch, or sucrose.
  • the filler is anhydrous lactose.
  • the filler is microcrystalline cellulose.
  • compositions containing the rapamycin derivative of the invention are delivered orally by tablet, caplet or capsule, microcapsules, dispersible powder, granule, suspension, syrup, elixir, and aerosol.
  • delivery is by tablets and hard- or liquid-filled capsules.
  • compositions containing the rapamycin derivative can be delivered intravenously, intramuscularly, subcutaneously, parenterally and intraperitoneally in the form of sterile injectable solutions, suspensions, dispersions, and powders which are fluid to the extent that easy syringe ability exits.
  • injectable compositions are sterile and stable under conditions of manufacture and storage, and free of the contaminating action of microorganisms such as bacteria and fungi.
  • compositions containing the rapamycin derivative can be delivered rectally in the form of a conventional suppository.
  • compositions containing the rapamycin derivative can be delivered vaginally in the form of a conventional suppository, cream, gel, ring, or coated intrauterine device (IUD).
  • IUD intrauterine device
  • compositions containing the rapamycin derivative can be delivered intranasally or intrabronchially in the form of an aerosol.
  • the rapamycin derivatives are administered orally as well as by intravenous, intramuscular, or subcutaneous routes.
  • Solid carriers include starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while liquid carriers include sterile water, polyethylene glycols, non-ionic surfactants and edible oils such as corn, peanut and sesame oils, as are appropriate to the nature of the active ingredient and the particular form of administration desired.
  • Adjuvants customarily employed in the preparation of pharmaceutical compositions are advantageously included, such as flavoring agents, coloring agents, preserving agents, and antioxidants, for example, vitamin E, ascorbic acid, BHT and BHA.
  • compositions from the standpoint of ease of preparation and administration are solid compositions, particularly tablets and hard-filled or liquid-filled capsules. Similarly, oral administration of the compounds is preferred.
  • rapamycin derivatives are also administered parenterally or intraperitoneally.
  • Solutions or suspensions of these active compounds as a free base or pharmacologically acceptable salt are prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions are also prepared in glycerol, liquid, polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form is sterile and fluid to the extent that easy syringe ability exits. It is stable under conditions of manufacture and storage and is preserved against the contaminating action of microorganisms such as bacterial and fungi.
  • the carrier is a solvent or dispersion medium containing, for example, water, ethanol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oil.
  • kits of the present invention can include the rapamycin derivative of the present invention and a carrier suitable for administration to a mammalian subject as discussed above.
  • the kits can also contain the reagents required to prepare the rapamycin derivatives of the present invention and include a rapamycin, an optionally substituted olefin, and a solvent.
  • kits can optionally include other reagents to form other rapamycin derivatives and include hydrogenation agents.
  • the kit can further contain instructions for performing the reactions of the present invention. Also provided in a kit can be other suitable chemicals, disposable gloves, decontamination instructions, applicator sticks or containers, and sample preparator cups.
  • Rapamycin (0.25 g, 0.274 mmol) is dissolved in 5 mL solvent with gentle heating. To this solution is added, dropwise, a solution of diethyl dicyanofumarate (0.183 g, 3 eq) in 7 mL solvent. The reaction mixture is stirred at 50 to 80° C. for 12 to 36 hours, and then the reaction mixture is chromatographed via reversed-phase HPLC to yield the product.
  • Example 2 The compound prepared according to Example 1 is dissolved in methanol in an 18 mm test-tube, and a spatula tip of Pd/C catalyst (Aldrich) is added. The mixture is hydrogenated on a Parr apparatus for 15 minutes at 2.0 atmosphere H 2 . The products are chromatographed via reversed-phase HPLC to yield give the product.
  • Pd/C catalyst Aldrich
  • Embryonic day 15 (E15) rat fetuses are collected and dissected in ice-cold phosphate-buffered saline (PBS). The ventral piece of tissue compromising the mesencephalic dopaminergic region is dissected out. Dissected pieces of tissue are pooled together and transferred to an enzymatic dissociation medium containing 20 IU/mL papain in Earle's balanced salt solution (Worthington Biochemical, Freehold, N.J., USA) and incubated for 60 minutes at 37° C.
  • the papain solution is aspirated and the tissue mechanically triturated with a fire-polished glass Pasteur pipette in complete medium (equal volumes of minimum essential medium (MEM) and F-12 nutrient mixture (GibcoBRL) supplemented with 0.1 mg/mL apotransferrin and 2.5 ⁇ g/mL insulin) containing 2,000 IU/mL DNase and 10 mg/mL ovomucoid protease inhibitor.
  • complete medium equal volumes of minimum essential medium (MEM) and F-12 nutrient mixture (GibcoBRL) supplemented with 0.1 mg/mL apotransferrin and 2.5 ⁇ g/mL insulin
  • the lysate is transferred to a scintillation vial containing Ultima GoldTM scintillation cocktail and radioactivity is determined with a liquid scintillation counter.
  • culture lysates are washed twice with buffer, incubated for 2 hours at room temperature with Optiphase SupermixTM scintillation cocktail (Wallac Scintillation Products, Gaithersburg, Md., USA), and radioactivity measured with a liquid scintillation counter.
  • Dissociated cortical neuron cultures are prepared as previously described (Pong et al., 2001). Briefly, embryonic day 15 rat fetuses are collected and dissected in ice-cold PBS. Dissected cortices are pooled together and transferred to an enzymatic dissociation medium containing papain. After 30 minutes, the tissue is mechanically triturated with a fire-polished glass Pasteur pipette. Single-cell suspensions in complete media are seeded on poly-L-ornithine and laminin coated 96-well plates. After 24 hours, cultures are treated with various concentrations of compound for 72 hours. The cultures are then fixed and stained with an anti-tubulin primary antibody (TUJ-1) and a fluorescent-tagged secondary antibody. Neurite outgrowth is determined by using the Enhanced Neurite Outgrowth (ENO) algorithm with the Cellomics ArrayScan and expressed as average neurite length or total neurite length per cell.
  • ENO Enhanced Neurite Outgrowth
  • Example 1 and 2 are anticipated to be active in cortical neuron assays with an EC 50 less than 1 ⁇ M and active in dopaminergic uptake assays with an EC 50 less than 1 ⁇ M.

Abstract

Provided is a method of treating neurodegenerative disorders comprising administering to a subject in need thereof a compound of formula I:
Figure US20090088448A1-20090402-C00001

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 11/891,879 filed Aug. 14, 2007, which is a continuation of U.S. patent application Ser. No. 11/300,941, filed Dec. 15, 2005, which claims benefit under 35 USC 119(e) of prior U.S. provisional Patent Application No. 60/638,004, filed Dec. 20, 2004.
  • BACKGROUND OF THE INVENTION
  • The present invention provides rapamycin derivatives and their use in the treatment of neurological disorders.
  • Ischemic stroke, which accounts for 83% of all stroke cases (the remaining 17% are of the hemorrhagic-type) occurs in approximately 700,000 Americans each year, which equates to roughly 1 stroke every 45 seconds. Ischemic strokes occur as a result of an obstruction within a blood vessel supplying blood to the brain. The underlying condition for this type of obstruction is the development of fatty deposits lining the vessel walls, called atherosclerosis. These fatty deposits can cause two types of obstruction: 1) cerebral thrombosis, which refers to a thrombus (blood clot) that develops at the clogged part of the vessel and 2) cerebral embolism, which refers generally to a blood clot that forms at another location in the circulatory system, usually the heart and large arteries of the upper chest and neck. A portion of the blood clot breaks loose, enters the bloodstream and travels through the brain's blood vessels until it reaches vessels too small to let it pass. Current therapies to treat ischemic stroke are limited. To date, the only approved drug for ischemic stroke is recombinant tissue plasminogen activator (rt-PA). rt-PA, which acts as a thrombolytic, has a limited therapeutic window of opportunity (3 hours), therefore allowing only 1-2% of all stroke patients to receive treatment. There are no marketed neuroprotectants agents for ischemic stroke.
  • Parkinson's disease (PD) is a neurodegenerative disease that is neuropathologically characterized by the selective degeneration of dopaminergic (DAergic) neurons of the substantia nigra. PD is a progressive disease with a mean age at onset of 55, although 15% of patients are diagnosed before the age of 50. It is estimated that 1.5 million Americans have PD. Some of the classical signs of PD are resting tremor on one side of the body, generalized slowness of movement (bradykinesia), stiffness of limbs (rigidity), gait or balance problems (postural dysfunction). Current PD medications treat symptoms, whereas none prevent or retard DAergic neuron degeneration.
  • Given their clinical importance, prototypical molecules that clearly exhibit both neuroprotective and/or neuroregenerative activities have been highly sought after. Neurotrophins are a family of proteins that have extraordinary therapeutic properties in pre-clinical models of neurodegeneration. Although experimentally promising, clinical development of neurotrophins was met with severe obstacles and setbacks, such as the inability to deliver these large proteins to target population of neurons, instability of the proteins, and non-specific activity.
  • What is needed in the art are further compounds useful in treating neurological disorders.
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides novel compounds useful in treating, and in the preparation of medicaments useful in the treatment of, neurological disorders.
  • In another aspect, the present invention provides novel neuroprotective agents.
  • In yet a further aspect, the present invention provides rapamycin derivatives, and pharmaceutically acceptable salts, prodrugs, and metabolites thereof.
  • In another aspect, the present invention provides methods of preparing rapamycin derivatives.
  • In a further aspect, the present invention provides methods of treating neurological disorders, and the use of a compound of the invention in the preparation of a medicament therefor.
  • In still another aspect, the present invention provides methods of treating complications due to stroke or head trauma, and the use of a compound of the invention in the preparation of a medicament therefor.
  • Other aspects and advantages of the present invention are described further in the following detailed description of the preferred embodiments thereof.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides novel rapamycin derivatives which are useful as neuroprotective agents, particularly in compositions for use in treating neurological disorders. The neurological disorder, including, e.g., a neurodegenerative or neuromuscular degenerative condition, can be a result of a genetic disorder present at birth, a disorder developed during the lifespan of an individual, e.g., stroke, and/or the result of physical trauma, e.g., head, spinal injury, or injury to the peripheral nervous system.
  • Thus, a compound of the invention may be useful in ameliorating the symptoms of a preexisting neurological disorder or preventing further neuro- and/or neuromuscular degeneration. In some embodiments, the neuroprotective agents of the invention can be used to delay the onset of symptoms associated with a neurological disorder.
  • I. Compounds of the Invention
  • In one embodiment, the present invention provides rapamycin derivatives of the formula I:
  • Figure US20090088448A1-20090402-C00002
  • R1 and R2 are the same or different and are selected from among CR16R17 and CR18R19. R3 and R4 are (a) independently selected from among H, OH, O(C1 to C6 alkyl), O(substituted C1 to C6 alkyl), O(acyl), O(aryl), O(substituted aryl), and halogen; or
  • (b) are taken together to form a double bond to O. R5, R6, and R7 are independently selected among H, OH, and OCH3. R8 and R9 are connected through a (i) single bond and are CH2 or (ii) double bond and are CH. R15 is selected from among C═O, CHOH, and CH2. R16 and R17 are selected from among H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2. R18 and R19 are independently selected from among H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2 or R17 and R18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring. Further, n is 1 or 2.
  • Additional compounds that can be prepared according to the present invention include compounds of the following structure:
  • Figure US20090088448A1-20090402-C00003
  • where R1, R2, and R6-R9 are defined as noted above.
  • In one embodiment, the present invention provides compounds where R17 and R18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring. Other examples of R17 and R18 include where each is an acyl substituted by alkoxy, e.g., —COOEt. R16 and R19 may each be CN. In another embodiment, the present invention provides compounds where R8 and R9 are bound through a single bond. In a further embodiment, the present invention provides compounds where R3 or R4 is OH. In yet another embodiment, the present invention provides compounds where R3 or R4 is —O(acyl), preferably where the substituted acyl is —C(O)-optionally substituted alkyl, in particular where alkyl can be straight or branched and optionally substituted, e.g., by heterocyclic such as aromatic heterocyclic such as pyridyl. An example is:
  • Figure US20090088448A1-20090402-C00004
  • In still a further embodiment, the present invention provides compounds where R5, R6, and R7 are OCH3. In another embodiment, the present invention provides compounds where n is 2. In yet a further embodiment, the present invention provides compounds where R15 is C═O.
  • The compounds of the invention can contain one or more asymmetric carbon atoms and some of the compounds can contain one or more asymmetric (chiral) centers and can thus give rise to optical isomers and diastereomers. While shown without respect to stereochemistry, when the compounds can contain one or more chiral centers, preferably at least one of the chiral centers is of S-stereochemistry. Thus, the invention includes such optical isomers and diastereomers; as well as the racemic and resolved, enantiomerically pure stereoisomers; as well as other mixtures of the R and S stereoisomers, and pharmaceutically acceptable salts, hydrates, metabolites, and prodrugs thereof.
  • The term “alkyl” is used herein to refer to both straight- and branched-chain saturated aliphatic hydrocarbon groups having 1 to 10 carbon atoms, and desirably about 1 to 8 carbon atoms. The term “alkenyl” is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon double bonds and containing about 2 to 10 carbon atoms. In one embodiment, the term alkenyl refers to an alkyl group having 1 or 2 carbon-carbon double bonds and having 2 to about 6 carbon atoms. The term “alkynyl” group is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon triple bond and having 2 to 8 carbon atoms. In one embodiment, the term alkynyl refers to an alkyl group having 1 or 2 carbon-carbon triple bonds and having 2 to 6 carbon atoms.
  • The term “cycloalkyl” is used herein to refer to an alkyl group as previously described that is cyclic in structure and has about 4 to 10 carbon atoms, or about 5 to 8 carbon atoms.
  • The terms “substituted alkyl”, “substituted alkenyl”, and “substituted alkynyl” refer to alkyl, alkenyl, and alkynyl groups, respectively, having one or more substituents including, without limitation, halogen, CN, OH, NO2, amino, aryl, heterocyclic, alkoxy, aryloxy, alkylcarbonyl, alkylcarboxy, and arylthio, which groups can be optionally substituted. These substituents can be attached to any carbon of an alkyl, alkenyl, or alkynyl group provided that the attachment constitutes a stable chemical moiety.
  • The term “aryl” as used herein refers to an aromatic system, e.g., of 6-20 carbon atoms, which can include a single ring or multiple aromatic rings fused or linked together where at least one part of the fused or linked rings forms the conjugated aromatic system. The aryl groups can include, but are not limited to, phenyl, naphthyl, biphenyl, anthryl, tetrahydronaphthyl, phenanthryl, indene, benzonaphthyl, fluorenyl, and carbazolyl.
  • The term “substituted aryl” refers to an aryl group which is substituted with one or more substituents including halogen, CN, OH, NO2, amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio, which groups can be optionally substituted. In one embodiment, a substituted aryl group is substituted with 1 to 4 substituents including halogen, CN, OH, NO2, amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio.
  • The term “heterocyclic” as used herein refers to a stable 4- to 7-membered monocyclic or multicyclic heterocyclic ring which is saturated, partially unsaturated, or wholly unsaturated, including aromatic such as pyridyl. The heterocyclic ring has carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms. In one embodiment, the heterocyclic ring has 1 to 4 heteroatoms in the backbone of the ring. When the heterocyclic ring contains nitrogen or sulfur atoms in the backbone of the ring, the nitrogen or sulfur atoms can be oxidized. The term “heterocyclic” also refers to multicyclic rings, e.g., of 9 to 20 ring members in which a heterocyclic ring is fused to an aryl ring. The heterocyclic ring can be attached to the aryl ring through a heteroatom or carbon atom, provided the resultant heterocyclic ring structure is chemically stable.
  • A variety of heterocyclic groups are known in the art and include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof. Oxygen-containing rings include, but are not limited to, furyl, tetrahydrofuranyl, pyranyl, pyronyl, and dioxinyl rings. Nitrogen-containing rings include, without limitation, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, pyridyl, piperidinyl, 2-oxopiperidinyl, pyridazinyl, pyrimidinyl, pyrazinyl, piperazinyl, azepinyl, triazinyl, pyrrolidinyl, and azepinyl rings. Sulfur-containing rings include, without limitation, thienyl and dithiolyl rings. Mixed heteroatom containing rings include, but are not limited to, oxathiolyl, oxazolyl, thiazolyl, oxadiazolyl, oxatriazolyl, dioxazolyl, oxathiazolyl, oxathiolyl, oxazinyl, oxathiazinyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, oxepinyl, thiepinyl, and diazepinyl rings. Fused heteroatom-containing rings include, but are not limited to, benzofuranyl, thionapthene, indolyl, benazazolyl, purindinyl, pyranopyrrolyl, isoindazolyl, indoxazinyl, benzoxazolyl, anthranilyl, benzopyranyl, quinolinyl, isoquinolinyl, benzodiazonyl, naphthylridinyl, benzothienyl, pyridopyridinyl, benzoxazinyl, xanthenyl, acridinyl, and purinyl rings.
  • The term “substituted heterocyclic” as used herein refers to a heterocyclic group having one or more substituents including halogen, CN, OH, NO2, amino, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy, alkyloxy, alkylcarbonyl, alkylcarboxy, aminoalkyl, and arylthio, which groups can be optionally substituted.
  • In one embodiment, a substituted heterocyclic group is substituted with 1 to 4 substituents.
  • The term “acyl” refers to a —C(O)— group, which is substituted at the carbon atom. The acyl group can be substituted or a terminal acyl group such as an HC(O)-group. The substituents can include any substituents noted above for alkyl groups, viz. one or more substituents including, without limitation, halogen, CN, OH, NO2, amino, aryl, heterocyclic, alkoxy, aryloxy, alkylcarbonyl, alkylcarboxy, and arylthio, which groups can be optionally substituted. Examples include —C(O)-alkoxy (eg. —OMe or —OEt) or —C(O)-alkyl where alkyl can be straight or branched and optionally substituted e.g., by heterocyclic (such as pyridyl).
  • The term “alkoxy” as used herein refers to the O(alkyl) group, where the point of attachment is through the oxygen-atom and the alkyl group is optionally substituted.
  • The term “aryloxy” as used herein refers to the O(aryl) group, where the point of attachment is through the oxygen-atom and the aryl group is optionally substituted.
  • The term “alkyloxy” as used herein refers to the alkylOH group, where the point of attachment is through the alkyl group.
  • The term “arylthio” as used herein refers to the S(aryl) group, where the point of attachment is through the sulfur-atom and the aryl group can be optionally substituted.
  • The term “alkylcarbonyl” as used herein refers to the C(O)(alkyl) group, where the point of attachment is through the carbon-atom of the carbonyl moiety and the alkyl group is optionally substituted.
  • The term “alkylcarboxy” as used herein refers to the C(O)O(alkyl) group, where the point of attachment is through the carbon-atom of the carboxy moiety and the alkyl group is optionally substituted.
  • The term “aminoalkyl” as used herein refers to both secondary and tertiary amines where the point of attachment is through the nitrogen-atom and the alkyl groups are optionally substituted. The alkyl groups can be the same or different.
  • The term “halogen” as used herein refers to Cl, Br, F, or I groups.
  • II. Methods of Preparing the Compounds of the Invention
  • The rapamycin derivatives of formula I of the present invention are prepared from a rapamycin starting material. In one embodiment, the rapamycin starting material includes, without limitation, rapamycin, norrapamycin, deoxorapamycin, desmethylrapamycins, or desmethoxyrapamycin, or pharmaceutically acceptable salts, prodrugs, or metabolites thereof. However, one of skill in the art would readily be able to select a suitable rapamycin starting material that can be utilized to prepare the novel rapamycin derivatives of the present invention.
  • The term “desmethylrapamycin” refers to the class of rapamycin compounds which lack one or more methyl groups. Examples of desmethylrapamycins that can be used according to the present invention include 3-desmethylrapamycin (U.S. Pat. No. 6,358,969), 7-O-desmethyl-rapamycin (U.S. Pat. No. 6,399,626), 17-desmethylrapamycin [U.S. Pat. No. 6,670,168], and 32-O-desmethylrapamycin, among others.
  • The term “desmethoxyrapamycin” refers to the class of rapamycin compounds which lack one or more methoxy groups and includes, without limitation, 32-desmethoxyrapamycin.
  • The rapamycin derivatives of formula I of the present invention are therefore prepared by combining a rapamycin starting material and an optionally substituted olefin. The term olefin refers to a molecule that contains a double bond. A variety of olefins can be utilized in the present invention and can readily be selected by one of skill in the art and include diethylfumarate, among others including any olefin sufficiently activated with electron-withdrawing substituents to undergo a [4+2]cycloaddition at temperatures up to 50° C. One of skill in the art would readily be able to select the amount of olefin that would be effective in preparing the rapamycin derivatives of the present invention. In one embodiment, an excess of the olefin is utilized, e.g., a 3:1 ratio of olefin to rapamycin starting material. However, even a 1:1 or 2:1 ratio of olefin to rapamycin starting material can be utilized as determined by one of skill in the art.
  • The olefin and rapamycin starting material is combined in a solvent. The solvent preferably dissolves the olefin and/or rapamycin on contact, or dissolves the olefin and rapamycin as the reaction proceeds. Solvents that can be utilized in the present invention include, without limitation, dimethylformamide, dioxane such as p-dioxane, chloroform, alcohols such as methanol and ethanol, ethyl acetate, water, acetonitrile, tetrahydrofuran, dichloromethane, and toluene, or combinations thereof. However, one of skill in the art would readily be able to select a suitable solvent based upon the solubility of the rapamycin starting material and olefin, as well as the reactivity of the solvent with the same. The amount of solvent utilized depends upon the scale of the reaction and specifically the amount of rapamycin starting material and olefin present in the reaction mixture. One of skill in the art would readily be able to determine the amount of solvent required.
  • Typically, the solution containing the olefin, rapamycin starting material, and solvent is maintained at elevated temperatures, and preferably a temperature that does not promote decomposition of the rapamycin and olefin. In one embodiment, the solution is maintained at a temperature of about 30 to about 80° C., and preferably about 50° C. The components are heated for a period of time sufficient to permit reaction between the rapamycin starting material and olefin. One of skill in the art using known techniques would readily be able to monitor the progress of the reaction during heating and thereby determine the amount of time required to perform the reaction. In one embodiment, the rapamycin and olefin are combined with a solvent and maintained at a temperature of about 50° C.
  • Isolation and purification of the rapamycin derivative is well within one of skill in the art and include chromatography including, without limitation, and recrystallization, high performance liquid chromatography (HPLC) such as reverse phase HPLC, and normal phase HPLC, and size-exclusion chromatography.
  • Once the rapamycin derivative is obtained, it can be reduced to form a more saturated rapamycin derivative. One of skill in the art would readily be able to select a suitable reducing agent for use in the present invention. In one embodiment, reduction of the rapamycin derivative can be effected using a hydrogenation agent. One of skill in the art would readily be able to select a suitable hydrogenation agent for use in the present invention. Typically, transition metal catalysts or transition metals on a support, preferably a carbon support, among others, in the presence hydrogen gas, are utilized to carry out the reduction. In another embodiment, the reduction is performed using palladium metal on carbon in the presence of hydrogen gas.
  • Reduction of the rapamycin derivative is typically carried out in a solvent. A variety of solvents can be utilized in the reduction and include, without limitation, alcohols such as methanol. However, one of skill in the art would readily be able to select a suitable solvent for use in the present invention and depending on the hydrogenation catalyst and rapamycin derivative being reduced. The amount of solvent depends on the scale of the reaction, and specifically the amount of rapamycin derivative being reduced.
  • The amount of hydrogenation agent utilized in the present invention can readily be determined by one of skill in the art. However, one of skill in the art would be able to determine and adjust the amount of hydrogenation agent necessary to perform the reduction and to form the more saturated rapamycin derivatives of the present invention. Further, a variety of apparatuses can be utilized to perform the hydrogenation of the present invention and include Parr apparatuses, among others. The selection of the particular apparatus for the hydrogenation is well within one of skill in the art.
  • In one embodiment, the rapamycin derivatives of the present invention are prepared as summarized in Scheme 1 below:
  • Figure US20090088448A1-20090402-C00005
  • where R4-R7, R15-R19, and n are defined above.
  • The rapamycin derivatives of the present invention can be utilized in the form of pharmaceutically acceptable salts, prodrugs, or metabolites thereof derived from pharmaceutically or physiologically acceptable acids or bases. These salts include, but are not limited to, the following salts with mineral or inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid and organic acids such as acetic acid, oxalic acid, succinic acid, and maleic acid. Other salts include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium in the form of esters, carbamates and other conventional “pro-drug” forms, which, when administered in such form, convert to the active moiety in vivo.
  • III. Methods of Using the Compounds of the Present Invention
  • The rapamycin derivatives of formula I of the present invention, including the more and less saturated rapamycin derivatives, have use in a variety of applications and include those relating to neurological disorders. The compounds of the present invention are useful in treating neurological disorders including, but not limited to, Alzheimer's disease amyotrophic lateral sclerosis, epilepsy, Huntington's Disease, Parkinson's Disease, stroke, spinal cord injury, traumatic brain injury, Lewy body dementia, multiple sclerosis, Pick's disease, Niewmann-Pick disease, amyloid angiopathy, cerebral amyloid angiopathy, systemic amyloidosis, hereditary cerebral hemorrhage with amyloidosis of the Dutch type, inclusion body myositis, mild cognitive impairment and Down's syndrome, and in the preparation of medicaments therefor. The rapamycin derivatives are also useful in treating complications due to stroke, head trauma, or spinal injury, or other injuries to the brain, peripheral nervous, central nervous, or neuromuscular system, and in the preparation of medicaments therefor.
  • The novel rapamycin derivatives are useful as neuroprotective agents. The rapamycin derivatives of the present invention may also be useful as neuroregenerative agents, i.e. restoring some neurological and/or neuromuscular or other function following onset of one of the above conditions and/or injury, stroke, or other trauma, and in the preparation of medicaments therefor.
  • The dosage requirements of the rapamycin derivatives of the present invention can vary depending on the condition, severity of the symptoms presented and the particular subject being treated. One of skill in the art would readily be able to determine the amount of the rapamycin derivative required. In one embodiment, about 0.5 to 200 mg is administered. In a further embodiment, about 0.5 to 100 mg is administered. In another embodiment, about 0.5 to about 75 mg is administered. In yet a further embodiment, about 1 to about 25 mg is administered. In another embodiment, about 0.5 to about 10 mg is administered, particularly when used in combination with another agent. In yet a further embodiment, about 2 to about 5 mg is administered. In yet another embodiment, about 5 to about 15 mg is administered.
  • Treatment can be initiated with dosages of the rapamycin derivative smaller than those required to produce a desired effect and generally less than the optimum dose of the rapamycin derivative. Thereafter, the dosage can be increased until the optimum effect under the circumstances is reached. Precise dosages will be determined by the administering physician based on experience with the individual subject being treated. In general, the compositions of this invention are most desirably administered at a concentration that will generally afford effective results without causing any harmful or deleterious side effects.
  • IV. Methods of Preparing Administrable Compositions Containing the Rapamycin Derivatives
  • In one aspect, the present invention includes methods of preparing a pharmaceutical composition containing one or more rapamycin derivatives of the present invention. The composition can be administered to a mammalian subject by several different routes and is desirably administered orally in solid or liquid form.
  • Solid forms, including tablets, capsules, and caplets, containing the rapamycin derivative can be formed by blending the rapamycin derivative with one or more of the components described above. In one embodiment, the components of the composition are dry or wet blended. In another embodiment, the components are dry granulated. In a further embodiment, the components are suspended or dissolved in a liquid and added to a form suitable for administration to a mammalian subject.
  • Liquid forms containing the rapamycin derivative can be formed by dissolving or suspending the rapamycin derivative in a liquid suitable for administration to a mammalian subject.
  • Compositions containing the rapamycin derivative of the present invention can be prepared according to the present invention by combining the rapamycin derivative and a pharmaceutically acceptable carrier.
  • The compositions described herein containing the rapamycin derivative can be formulated in any form suitable for the desired route of delivery using a pharmaceutically effective amount of the rapamycin derivative. For example, the compositions of the invention can be delivered by a route such as oral, dermal, transdermal, intrabronchial, intranasal, intravenous, intramuscular, subcutaneous, parenteral, intraperitoneal, intranasal, vaginal, rectal, sublingual, intracranial, epidural, intratracheal, or by sustained release. In one embodiment, delivery is oral.
  • The oral dosage tablet composition of this invention can also be used to make oral dosage tablets containing analogs of the rapamycin derivative, including, but not limited to, esters, carbamates, sulfates, ethers, oximes, carbonates, and the like which are known to those of skill in the art.
  • A pharmaceutically effective amount of the rapamycin derivative can vary depending on the specific compound(s), mode of delivery, severity of the condition being treated, and any other active ingredients used in the composition. The dosing regimen can also be adjusted to provide the optimal therapeutic response. Several divided doses can be delivered daily, e.g., in divided doses 2 to 4 times a day, or a single dose can be delivered. The dose can however be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. In one embodiment, the delivery is on a daily, weekly, or monthly basis. In another embodiment, the delivery is on a daily delivery. However, daily dosages can be lowered or raised based on the periodic delivery.
  • The rapamycin derivatives of the present invention can be combined with one or more pharmaceutically acceptable carriers or excipients including, without limitation, solid and liquid carriers which are compatible with the compositions of the present invention. Such carriers include adjuvants, syrups, elixirs, diluents, binders, lubricants, surfactants, granulating agents, disintegrating agents, emollients, metal chelators, pH adjustors, surfactants, fillers, disintegrants, and combinations thereof, among others. In one embodiment, the rapamycin derivative is combined with metal chelators, pH adjustors, surfactants, fillers, disintegrants, lubricants, and binders.
  • Adjuvants can include, without limitation, flavoring agents, coloring agents, preservatives, and supplemental antioxidants, which can include vitamin E, ascorbic acid, butylated hydroxytoluene (BHT) and butylated hydroxyanisole (BHA).
  • Binders can include, without limitation, cellulose, methylcellulose, hydroxymethylcellulose, carboxymethylcellulose calcium, carboxymethylcellulose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, microcrystalline cellulose, noncrystalline cellulose, polypropylpyrrolidone, polyvinylpyrrolidone (povidone, PVP), gelatin, gum arabic and acacia, polyethylene glycols, starch, sugars such as sucrose, kaolin, dextrose, and lactose, cholesterol, tragacanth, stearic acid, gelatin, casein, lecithin (phosphatides), cetostearyl alcohol, cetyl alcohol, cetyl esters wax, dextrates, dextrin, glyceryl monooleate, glyceryl monostearate, glyceryl palmitostearate, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polyoxyethylene stearates, polyvinyl alcohol, and gelatin, among others. In one embodiment, the binder is povidone, hydroxypropylmethylcellulose, carboxymethylcellulose, or gelatin. In another embodiment, the binder is povidone.
  • Lubricants can include magnesium stearate, light anhydrous silicic acid, talc, stearic acid, sodium lauryl sulfate, and sodium stearyl furamate, among others. In one embodiment, the lubricant is magnesium stearate, stearic acid, or sodium stearyl furamate. In another embodiment, the lubricant is magnesium stearate.
  • Granulating agents can include, without limitation, silicon dioxide, microcrystalline cellulose, starch, calcium carbonate, pectin, crospovidone, and polyplasdone, among others.
  • Disintegrating agents or disintegrants can include croscarmellose sodium, starch, carboxymethylcellulose, substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate, calcium citrate, sodium starch glycolate, pregelatinized starch or crospovidone, among others. In one embodiment, the disintegrant is croscarmellose sodium.
  • Emollients can include, without limitation, stearyl alcohol, mink oil, cetyl alcohol, oleyl alcohol, isopropyl laurate, polyethylene glycol, olive oil, petroleum jelly, palmitic acid, oleic acid, and myristyl myristate.
  • Surfactants can include polysorbates, sorbitan esters, poloxamer, or sodium lauryl sulfate. In one embodiment, the surfactant is sodium lauryl sulfate.
  • Metal chelators can include physiologically acceptable chelating agents including edetic acid, malic acid, or fumaric acid. In one embodiment, the metal chelator is edetic acid.
  • pH adjusters can also be utilized to adjust the pH of a solution containing the rapamycin derivative to about 4 to about 6. In one embodiment, the pH of a solution containing the rapamycin derivative is adjusted to a pH of about 4.6. pH adjustors can include physiologically acceptable agents including citric acid, ascorbic acid, fumaric acid, or malic acid, and salts thereof. In one embodiment, the pH adjuster is citric acid.
  • Fillers that can be used according to the present invention include anhydrous lactose, microcrystalline cellulose, mannitol, calcium phosphate, pregelatinized starch, or sucrose. In one embodiment, the filler is anhydrous lactose. In another embodiment, the filler is microcrystalline cellulose.
  • In one embodiment, compositions containing the rapamycin derivative of the invention are delivered orally by tablet, caplet or capsule, microcapsules, dispersible powder, granule, suspension, syrup, elixir, and aerosol. In a further embodiment, when compositions containing the rapamycin derivative are delivered orally, delivery is by tablets and hard- or liquid-filled capsules.
  • In another embodiment, the compositions containing the rapamycin derivative can be delivered intravenously, intramuscularly, subcutaneously, parenterally and intraperitoneally in the form of sterile injectable solutions, suspensions, dispersions, and powders which are fluid to the extent that easy syringe ability exits. Such injectable compositions are sterile and stable under conditions of manufacture and storage, and free of the contaminating action of microorganisms such as bacteria and fungi.
  • In a further embodiment, compositions containing the rapamycin derivative can be delivered rectally in the form of a conventional suppository.
  • In another embodiment, compositions containing the rapamycin derivative can be delivered vaginally in the form of a conventional suppository, cream, gel, ring, or coated intrauterine device (IUD).
  • In yet another embodiment, compositions containing the rapamycin derivative can be delivered intranasally or intrabronchially in the form of an aerosol.
  • The rapamycin derivatives are administered orally as well as by intravenous, intramuscular, or subcutaneous routes. Solid carriers include starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while liquid carriers include sterile water, polyethylene glycols, non-ionic surfactants and edible oils such as corn, peanut and sesame oils, as are appropriate to the nature of the active ingredient and the particular form of administration desired. Adjuvants customarily employed in the preparation of pharmaceutical compositions are advantageously included, such as flavoring agents, coloring agents, preserving agents, and antioxidants, for example, vitamin E, ascorbic acid, BHT and BHA.
  • The preferred pharmaceutical compositions from the standpoint of ease of preparation and administration are solid compositions, particularly tablets and hard-filled or liquid-filled capsules. Similarly, oral administration of the compounds is preferred.
  • The rapamycin derivatives are also administered parenterally or intraperitoneally. Solutions or suspensions of these active compounds as a free base or pharmacologically acceptable salt are prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions are also prepared in glycerol, liquid, polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form is sterile and fluid to the extent that easy syringe ability exits. It is stable under conditions of manufacture and storage and is preserved against the contaminating action of microorganisms such as bacterial and fungi. The carrier is a solvent or dispersion medium containing, for example, water, ethanol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oil.
  • V. Kits of the Invention
  • The present invention also provides kits or packages containing the rapamycin derivatives. Kits of the present invention can include the rapamycin derivative of the present invention and a carrier suitable for administration to a mammalian subject as discussed above. The kits can also contain the reagents required to prepare the rapamycin derivatives of the present invention and include a rapamycin, an optionally substituted olefin, and a solvent.
  • The kits can optionally include other reagents to form other rapamycin derivatives and include hydrogenation agents.
  • The kit can further contain instructions for performing the reactions of the present invention. Also provided in a kit can be other suitable chemicals, disposable gloves, decontamination instructions, applicator sticks or containers, and sample preparator cups.
  • The following examples are provided to illustrate the invention and do not limit the scope thereof. One skilled in the art will appreciate that although specific reagents and conditions are outlined in the following examples, modifications can be made which are meant to be encompassed by the spirit and scope of the invention.
  • EXAMPLES Example 1
  • Figure US20090088448A1-20090402-C00006
  • Rapamycin (0.25 g, 0.274 mmol) is dissolved in 5 mL solvent with gentle heating. To this solution is added, dropwise, a solution of diethyl dicyanofumarate (0.183 g, 3 eq) in 7 mL solvent. The reaction mixture is stirred at 50 to 80° C. for 12 to 36 hours, and then the reaction mixture is chromatographed via reversed-phase HPLC to yield the product.
  • Example 2
  • Figure US20090088448A1-20090402-C00007
  • The compound prepared according to Example 1 is dissolved in methanol in an 18 mm test-tube, and a spatula tip of Pd/C catalyst (Aldrich) is added. The mixture is hydrogenated on a Parr apparatus for 15 minutes at 2.0 atmosphere H2. The products are chromatographed via reversed-phase HPLC to yield give the product.
  • Example 3
  • Mesencephalic dopaminergic neuron cultures are prepared as described in Pong et al., J. Neurochem. 69: 986-994, 1997, which is incorporated herein by reference in its entirety. Embryonic day 15 (E15) rat fetuses are collected and dissected in ice-cold phosphate-buffered saline (PBS). The ventral piece of tissue compromising the mesencephalic dopaminergic region is dissected out. Dissected pieces of tissue are pooled together and transferred to an enzymatic dissociation medium containing 20 IU/mL papain in Earle's balanced salt solution (Worthington Biochemical, Freehold, N.J., USA) and incubated for 60 minutes at 37° C. After enzymatic dissociation, the papain solution is aspirated and the tissue mechanically triturated with a fire-polished glass Pasteur pipette in complete medium (equal volumes of minimum essential medium (MEM) and F-12 nutrient mixture (GibcoBRL) supplemented with 0.1 mg/mL apotransferrin and 2.5 μg/mL insulin) containing 2,000 IU/mL DNase and 10 mg/mL ovomucoid protease inhibitor.
  • For dopamine uptake experiments, single-cell suspensions in complete media are seeded on poly-L-ornithine and laminin coated 24-well plates. The cultures are maintained for seven days prior to experimentation. Cultures are pretreated with various concentrations of the compound for 24 hours, then exposed to 10 mM MPP+ for 1 hour. Following the 1 hour incubation, media is exchanged three times and fresh compound is added for an additional 48 hours.
  • After 48 hours growth of mesencephalic dopaminergic neuron cultures following MPP+ exposure, high-affinity 3H-dopamine uptake is performed using a modified method described by Prochiantz et al., Nature 293: 570-572, 1981, which is incorporated herein by reference. Cultures are washed with pre-warmed PBS containing 5.6 mM glucose and 1 mM ascorbic acid. Cultures are then incubated for 15 minutes at 37° C. with 50 nM 3H-dopamine (31 Ci/mmol, DuPont-NEN, Wilmington, Del., USA). The cultures are washed twice with buffer and lysed with 0.5 N NaOH. The lysate is transferred to a scintillation vial containing Ultima Gold™ scintillation cocktail and radioactivity is determined with a liquid scintillation counter. Alternatively, culture lysates are washed twice with buffer, incubated for 2 hours at room temperature with Optiphase Supermix™ scintillation cocktail (Wallac Scintillation Products, Gaithersburg, Md., USA), and radioactivity measured with a liquid scintillation counter.
  • Dissociated cortical neuron cultures are prepared as previously described (Pong et al., 2001). Briefly, embryonic day 15 rat fetuses are collected and dissected in ice-cold PBS. Dissected cortices are pooled together and transferred to an enzymatic dissociation medium containing papain. After 30 minutes, the tissue is mechanically triturated with a fire-polished glass Pasteur pipette. Single-cell suspensions in complete media are seeded on poly-L-ornithine and laminin coated 96-well plates. After 24 hours, cultures are treated with various concentrations of compound for 72 hours. The cultures are then fixed and stained with an anti-tubulin primary antibody (TUJ-1) and a fluorescent-tagged secondary antibody. Neurite outgrowth is determined by using the Enhanced Neurite Outgrowth (ENO) algorithm with the Cellomics ArrayScan and expressed as average neurite length or total neurite length per cell.
  • The compounds of Example 1 and 2 are anticipated to be active in cortical neuron assays with an EC50 less than 1 μM and active in dopaminergic uptake assays with an EC50 less than 1 μM.
  • All patent, patent publications, and other publications listed in this specification are incorporated herein by reference. While the invention has been described with reference to a particularly preferred embodiment, it will be appreciated that modifications can be made without departing from the spirit of the invention. Such modifications are intended to fall within the scope of the appended claims.

Claims (4)

1. A method of treating neurodegenerative disorders comprising administering to a subject in need thereof a compound of formula I:
Figure US20090088448A1-20090402-C00008
wherein:
R1 and R2 are the same or different and are selected from the group consisting of CR16R17 and CR18R19;
R3 and R4 are:
(a) independently selected from the group consisting of H, OH, O(C1 to C6 alkyl), O(substituted C1 to C6 alkyl), O(acyl), O(aryl), O(substituted aryl), and halogen; or
(b) taken together to form a double bond to O;
R5, R6, and R7 are independently selected from the group consisting of H, OH, and OCH3;
R8 and R9 are connected through a (i) single bond and are CH2 or (ii) double bond and are CH;
R15 is selected from the group consisting of C═O, CHOH, and CH2;
R16 and R17 are independently selected from the group consisting of H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2;
R18 and R19 are independently selected from the group consisting of H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2; or
R17 and R18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring;
n is 1 or 2; or
a pharmaceutically acceptable salt thereof.
2. A method of treating neurodegenerative disorders comprising administering to a subject in need thereof a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formula I:
Figure US20090088448A1-20090402-C00009
wherein:
R1 and R2 are the same or different and are selected from the group consisting of CR16R17 and CR18R19;
R3 and R4 are:
(a) independently selected from the group consisting of H, OH, O(C1 to C6 alkyl), O(substituted C1 to C6 alkyl), O(acyl), O(aryl), O(substituted aryl), and halogen; or
(b) taken together to form a double bond to O;
R5, R6, and R7 are independently selected from the group consisting of H, OH, and OCH3;
R8 and R9 are connected through a (i) single bond and are CH2 or (ii) double bond and are CH;
R15 is selected from the group consisting of C═O, CHOH, and CH2;
R16 and R17 are independently selected from the group consisting of H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2;
R18 and R19 are independently selected from the group consisting of H, C1 to C6 alkyl, C1 to C6 substituted alkyl, aryl, substituted aryl, acyl, CF3, CN, and NO2; or
R17 and R18 are taken together to form a carbon-based or heterocyclic 5- to 7-membered ring;
n is 1 or 2;
or a pharmaceutically acceptable salt thereof.
3. The method according to claim 1 or 2, wherein said neurodegenerative disorder is selected from Alzheimer's disease, amyotrophic lateral sclerosis, epilepsy, Huntington's Disease, Parkinson's Disease, stroke, spinal cord injury, traumatic brain injury, head trauma, Lewy body dementia, multiple sclerosis, Pick's disease, Niewmann-Pick disease, amyloid angiopathy, cerebral amyloid angiopathy, systemic amyloidosis, hereditary cerebral hemorrhage with amyloidosis of the Dutch type, inclusion body myositis, mild cognitive impairment, Down's syndrome, complications due to stroke and complications due to head trauma
4. A method of treatment according to claim 3 wherein the neurodegenerative disorder is complications due to stroke or head trauma.
US12/329,106 2004-12-20 2008-12-05 Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders Abandoned US20090088448A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/329,106 US20090088448A1 (en) 2004-12-20 2008-12-05 Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63800404P 2004-12-20 2004-12-20
US11/300,941 US7273874B2 (en) 2004-12-20 2005-12-15 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US11/891,879 US7476678B2 (en) 2004-12-20 2007-08-14 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US12/329,106 US20090088448A1 (en) 2004-12-20 2008-12-05 Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/891,879 Continuation US7476678B2 (en) 2004-12-20 2007-08-14 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders

Publications (1)

Publication Number Publication Date
US20090088448A1 true US20090088448A1 (en) 2009-04-02

Family

ID=36602218

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/300,941 Expired - Fee Related US7273874B2 (en) 2004-12-20 2005-12-15 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US11/891,879 Expired - Fee Related US7476678B2 (en) 2004-12-20 2007-08-14 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US12/329,106 Abandoned US20090088448A1 (en) 2004-12-20 2008-12-05 Rapamycin Derivatives and the Uses Thereof in the Treatment of Neurological Disorders

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/300,941 Expired - Fee Related US7273874B2 (en) 2004-12-20 2005-12-15 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US11/891,879 Expired - Fee Related US7476678B2 (en) 2004-12-20 2007-08-14 Rapamycin derivatives and the uses thereof in the treatment of neurological disorders

Country Status (16)

Country Link
US (3) US7273874B2 (en)
EP (1) EP1828203A2 (en)
JP (1) JP2008524232A (en)
KR (1) KR20070090931A (en)
CN (1) CN101084226A (en)
AU (1) AU2005319427B2 (en)
BR (1) BRPI0519593A2 (en)
CA (1) CA2590254A1 (en)
CR (1) CR9149A (en)
IL (1) IL183772A0 (en)
MX (1) MX2007007409A (en)
NI (1) NI200700156A (en)
NO (1) NO20072665L (en)
RU (1) RU2007119585A (en)
WO (1) WO2006068905A2 (en)
ZA (1) ZA200705123B (en)

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2344821C2 (en) 2002-07-30 2009-01-27 Уайт Rapamycine hydroxy ethers-containing parenteral compositions
EP1720886A1 (en) * 2004-03-02 2006-11-15 Wyeth Macrolides and methods for producing same
US7507752B2 (en) * 2004-06-03 2009-03-24 Wyeth Biosynthetic gene cluster for the production of a complex polyketide
JP2008524232A (en) * 2004-12-20 2008-07-10 ワイス Rapamycin derivatives and their use in the treatment of neurological disorders
MX2007007408A (en) * 2004-12-20 2007-07-12 Wyeth Corp Rapamycin analogues and the uses thereof in the treatment of neurological, proliferative, and inflammatory disorders.
WO2006096529A2 (en) * 2005-03-07 2006-09-14 Novartis Ag Genes involved in neurodegenerative conditions
AU2006322029A1 (en) * 2005-12-07 2007-06-14 Wyeth Process for the preparation of purified crystalline CCI-779
BRPI0619578A2 (en) * 2005-12-07 2011-10-04 Wyeth Corp process for preparing a rapamycin 42-ester compound, method for isolating crude rapamycin 42-ester boronate from the mother liquor, method for purifying a rapamycin 42-ester boronate, and cci-779 boronate
JP2009518648A (en) * 2005-12-07 2009-05-07 ワイス Method for preparing crystalline rapamycin and method for measuring crystallinity of rapamycin compounds using differential scanning calorimetry
BRPI0708611A2 (en) * 2006-03-07 2011-06-07 Wyeth Corp compound; use of the compound in the preparation of a medicament for treating a neurological disorder in a mammal; compound preparation method; and method of treating a neurological disorder in a mammal
TW200824713A (en) * 2006-10-18 2008-06-16 Wyeth Corp Processes for the synthesis of individual isomers of mono-PEG CCI-779
US20080276935A1 (en) 2006-11-20 2008-11-13 Lixiao Wang Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US9737640B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for medical devices
US8414909B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8998846B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8414526B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids, and/or lipids
US8414525B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8425459B2 (en) 2006-11-20 2013-04-23 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8414910B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US9700704B2 (en) 2006-11-20 2017-07-11 Lutonix, Inc. Drug releasing coatings for balloon catheters
JP2010521419A (en) * 2007-01-29 2010-06-24 ワイス エルエルシー Immunophilin ligands and methods for modulating immunophilin and calcium channel activity
JP2008305262A (en) * 2007-06-08 2008-12-18 Konica Minolta Business Technologies Inc Printer introduction method in server and thin client environment
EP2205249B1 (en) 2007-09-28 2018-11-07 Intrexon Corporation Therapeutic gene-switch constructs and bioreactors for the expression of biotherapeutic molecules, and uses thereof
US20090209527A1 (en) * 2008-02-19 2009-08-20 Wyeth Methods for Using Rapamycin Analogues in the Treatment of Neurological Disorders
WO2010024898A2 (en) 2008-08-29 2010-03-04 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
CA2937492C (en) 2008-11-11 2019-08-13 The Board Of Regents Of The University Of Texas System Inhibition of mammalian target of rapamycin
DE102008060549A1 (en) 2008-12-04 2010-06-10 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Drug-peptide construct for extracellular accumulation
US9283211B1 (en) 2009-11-11 2016-03-15 Rapamycin Holdings, Llc Oral rapamycin preparation and use for stomatitis
CN103038343A (en) 2010-03-23 2013-04-10 英特瑞克斯顿股份有限公司 Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof.
ES2372077B2 (en) * 2010-06-24 2012-05-18 Universidad De Sevilla USE OF RAPAMYCIN FOR THE TREATMENT OF COGNITIVE DEFICITS ASSOCIATED WITH THE DOWN SYNDROME.
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
CN102274219A (en) * 2011-06-27 2011-12-14 苏州大学附属第一医院 Application of rapamycin to treating early brain injury after subarachnoid hemorrhage
EP2906214A1 (en) 2012-10-12 2015-08-19 The Board of Regents of The University of Texas System Use of mtor inhibitors to treat vascular cognitive impairment
US9121859B2 (en) * 2012-12-04 2015-09-01 Siemens Healthcare Diagnostics Inc. Compounds and methods for determination of FKBP-binding immunosuppressant drugs
WO2014160328A1 (en) 2013-03-13 2014-10-02 The Board Of Regents Of The University Of Texas System Mtor inhibitors for prevention of intestinal polyp growth
US9700544B2 (en) 2013-12-31 2017-07-11 Neal K Vail Oral rapamycin nanoparticle preparations
ES2900426T3 (en) 2013-12-31 2022-03-16 Rapamycin Holdings Llc Oral preparations and use of rapamycin nanoparticles
UY37343A (en) 2016-07-25 2018-02-28 Intrexon Corp CONTROL OF THE PHENOTYPE IN PLANTS
AU2017359342B2 (en) 2016-11-09 2022-02-17 Intrexon Corporation Frataxin expression constructs
EP3848065B1 (en) 2017-05-15 2023-07-26 C. R. Bard, Inc. Medical device with drug-eluting coating and intermediate layer
US20210369684A1 (en) * 2018-02-09 2021-12-02 Torcept Therapeutics, Inc. Rapamycin analog for prevention and/or treatment of neurodegnerative conditions
WO2019157374A1 (en) * 2018-02-09 2019-08-15 Mount Tam Therapeutics, Inc. Rapamycin analog for prevention and/or treatment of neurodegenerative conditions
US11541152B2 (en) 2018-11-14 2023-01-03 Lutonix, Inc. Medical device with drug-eluting coating on modified device surface
US20220176084A1 (en) 2019-04-08 2022-06-09 Bard Peripheral Vascular, Inc. Medical device with drug-eluting coating on modified device surface
TW202206093A (en) * 2020-08-12 2022-02-16 中國醫藥大學 Use of seam3d antagonist for preventing or treating neurodegenerative disease and increasing lifespan
WO2022272154A2 (en) * 2021-06-25 2022-12-29 Apertor Pharmaceuticals, Inc. Small molecule compounds

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3491092A (en) * 1963-11-06 1970-01-20 Bayer Ag Substituted benzoxazinones and a process for their preparation
US3686238A (en) * 1970-01-19 1972-08-22 Syntex Corp Glycerol esterified with 2-naphthyl-acetic acids and fatty acids
US3738980A (en) * 1969-08-11 1973-06-12 Ciba Geigy Corp Antibiotically active compounds
US3900465A (en) * 1973-04-05 1975-08-19 Lepetit Spa 3-formylrifamycin azines
US3905981A (en) * 1973-10-12 1975-09-16 Research Corp N-dealkylation of tertiary amines
US4127720A (en) * 1977-09-21 1978-11-28 Bristol-Myers Company Pyrimido[2,1-a]isoquinoline derivatives having antiallergy activity
US5023262A (en) * 1990-08-14 1991-06-11 American Home Products Corporation Hydrogenated rapamycin derivatives
US20010036947A1 (en) * 1997-06-04 2001-11-01 Gpi Nil Holdings, Inc. Pipecolic acid derivative hair growth compositions and uses
US20020151088A1 (en) * 1993-04-23 2002-10-17 Molnar-Kimber Katherine L. Rapamycin conjugates

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1545824A1 (en) 1965-06-21 1969-12-11 Bayer Ag Process for the preparation of 2-sulfonamido- or 2-sulfonhydrazido-benzoxazinones
JPH0262875A (en) 1988-05-23 1990-03-02 Wakunaga Pharmaceut Co Ltd Novel isoindoline derivative
WO1991019495A1 (en) 1990-06-11 1991-12-26 Fujisawa Pharmaceutical Co., Ltd. Use of a macrolide compound such as fk 506 for manufacturing a medicament for treating idiopathic thrombocytopenic purpura and basedow's disease
DE4021404A1 (en) 1990-07-05 1992-01-09 Sandoz Ag New tri:cyclic hetero atom contg. cpds. for arthritis treatment - have immunosuppressive and antiinflammatory activity for treatment of skin disorders e.g. psoriasis and dermatitis and auto-immune disease
GB2249027A (en) 1990-10-23 1992-04-29 Fujisawa Pharmaceutical Co Use of macrolide compounds for hepatic failure
WO1992013862A1 (en) 1991-02-05 1992-08-20 Fujisawa Pharmaceutical Co., Ltd. Lactone compounds
US5604294A (en) 1991-09-05 1997-02-18 Luly; Jay R. Macrocyclic immunomodulators
DE69232630T2 (en) 1991-09-05 2003-10-09 Abbott Lab Macrocyclic immune modulators
ZA935112B (en) 1992-07-17 1994-02-08 Smithkline Beecham Corp Rapamycin derivatives
ZA935111B (en) 1992-07-17 1994-02-04 Smithkline Beecham Corp Rapamycin derivatives
DE69435044T2 (en) 1993-04-23 2008-09-18 Wyeth Rapamycin conjugates and antibodies
US5798355A (en) 1995-06-07 1998-08-25 Gpi Nil Holdings, Inc. Inhibitors of rotamase enzyme activity
US5525610A (en) 1994-03-31 1996-06-11 American Home Products Corporation 42-Epi-rapamycin and pharmaceutical compositions thereof
US5696135A (en) * 1995-06-07 1997-12-09 Gpi Nil Holdings, Inc. Inhibitors of rotamase enzyme activity effective at stimulating neuronal growth
TW427904B (en) 1995-12-07 2001-04-01 American Home Prod Neuroprotective agents
US5717092A (en) 1996-03-29 1998-02-10 Vertex Pharmaceuticals Inc. Compounds with improved multi-drug resistance activity
US5780484A (en) 1996-11-13 1998-07-14 Vertex Pharmaceuticals Incorporated Methods for stimulating neurite growth with piperidine compounds
EP1083872A1 (en) 1998-06-03 2001-03-21 GPI NIL Holdings, Inc. Pipecolic acid derivative hair growth compositions and uses
US6376517B1 (en) 1998-08-14 2002-04-23 Gpi Nil Holdings, Inc. Pipecolic acid derivatives for vision and memory disorders
CN1323310A (en) 1998-08-17 2001-11-21 美国家用产品公司 Photocyclized rapamycins
US6015809A (en) 1998-08-17 2000-01-18 American Home Products Corporation Photocyclized rapamycin
WO2000034239A2 (en) 1998-12-08 2000-06-15 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
AU7124000A (en) 1999-09-09 2001-04-10 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6569867B2 (en) 1999-10-01 2003-05-27 Kosan Biosciences, Inc. Polyketide derivatives
DE60033965T2 (en) * 1999-10-29 2008-01-10 Kosan Biosciences, Inc., Hayward RAPAMYCIN ANALOG
AU2001261432B2 (en) 2000-05-17 2005-06-16 Kosan Biosciences, Inc. Polyketide derivatives
CA2455311A1 (en) 2001-08-22 2003-03-06 Wyeth Rapamycin dialdehydes
CA2455308A1 (en) 2001-08-22 2003-03-06 Wyeth Rapamycin 29-enols
EP1521828B1 (en) 2002-07-16 2015-07-01 Buck Institute for Research on Aging Production of polyketides
JP2008524232A (en) 2004-12-20 2008-07-10 ワイス Rapamycin derivatives and their use in the treatment of neurological disorders
MX2007007408A (en) 2004-12-20 2007-07-12 Wyeth Corp Rapamycin analogues and the uses thereof in the treatment of neurological, proliferative, and inflammatory disorders.
BRPI0708611A2 (en) 2006-03-07 2011-06-07 Wyeth Corp compound; use of the compound in the preparation of a medicament for treating a neurological disorder in a mammal; compound preparation method; and method of treating a neurological disorder in a mammal

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3491092A (en) * 1963-11-06 1970-01-20 Bayer Ag Substituted benzoxazinones and a process for their preparation
US3738980A (en) * 1969-08-11 1973-06-12 Ciba Geigy Corp Antibiotically active compounds
US3686238A (en) * 1970-01-19 1972-08-22 Syntex Corp Glycerol esterified with 2-naphthyl-acetic acids and fatty acids
US3900465A (en) * 1973-04-05 1975-08-19 Lepetit Spa 3-formylrifamycin azines
US3905981A (en) * 1973-10-12 1975-09-16 Research Corp N-dealkylation of tertiary amines
US4127720A (en) * 1977-09-21 1978-11-28 Bristol-Myers Company Pyrimido[2,1-a]isoquinoline derivatives having antiallergy activity
US5023262A (en) * 1990-08-14 1991-06-11 American Home Products Corporation Hydrogenated rapamycin derivatives
US20020151088A1 (en) * 1993-04-23 2002-10-17 Molnar-Kimber Katherine L. Rapamycin conjugates
US20010036947A1 (en) * 1997-06-04 2001-11-01 Gpi Nil Holdings, Inc. Pipecolic acid derivative hair growth compositions and uses

Also Published As

Publication number Publication date
WO2006068905A3 (en) 2006-12-07
US7273874B2 (en) 2007-09-25
ZA200705123B (en) 2010-01-27
MX2007007409A (en) 2007-07-12
KR20070090931A (en) 2007-09-06
US20080027089A1 (en) 2008-01-31
BRPI0519593A2 (en) 2009-02-25
WO2006068905A2 (en) 2006-06-29
US7476678B2 (en) 2009-01-13
CR9149A (en) 2007-10-23
EP1828203A2 (en) 2007-09-05
AU2005319427B2 (en) 2011-11-03
AU2005319427A1 (en) 2006-06-29
NO20072665L (en) 2007-09-07
NI200700156A (en) 2008-06-17
JP2008524232A (en) 2008-07-10
IL183772A0 (en) 2007-09-20
RU2007119585A (en) 2009-01-27
CA2590254A1 (en) 2006-06-29
CN101084226A (en) 2007-12-05
US20060135550A1 (en) 2006-06-22

Similar Documents

Publication Publication Date Title
US7476678B2 (en) Rapamycin derivatives and the uses thereof in the treatment of neurological disorders
US7276498B2 (en) Rapamycin analogues and uses thereof in the treatment of neurological disorders
EP3412663B1 (en) Nitrogen-containing heterocycle and carbocycle derivatives having trka inhibitory activity
US20090209527A1 (en) Methods for Using Rapamycin Analogues in the Treatment of Neurological Disorders
US20080249124A1 (en) Wortmannin-rapalog conjugate and uses thereof
US7087628B2 (en) N-heterocyclyl hydrazides as neurotrophic agents
KR20080099317A (en) Meridamycin analogues for the treatment of neurodegenerative disorders
CN109776373B (en) Amide-substituted pyrrolidine amide derivatives and uses thereof
AU2002334836A1 (en) N-heterocyclyl hydrazides as neurotrophic agents
CA2462643A1 (en) Triazepine derivatives as neurotrophic agents
WO2017060826A1 (en) Difluoro-(2-substituted carbamoyl-1,6-diaza-bicyclo [3.2.1] oct-6-yloxy) acetic acid compounds and their use in treatment of bacterial infections
AU2002334837A1 (en) Triazepine derivatives as neurotrophic agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: WYETH LLC,NEW JERSEY

Free format text: CHANGE OF NAME;ASSIGNOR:WYETH;REEL/FRAME:024541/0922

Effective date: 20091109

Owner name: WYETH LLC, NEW JERSEY

Free format text: CHANGE OF NAME;ASSIGNOR:WYETH;REEL/FRAME:024541/0922

Effective date: 20091109

STCB Information on status: application discontinuation

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