US20090270465A1 - Use of epothilone d in treating tau-associated diseases including alzheimer's disease - Google Patents

Use of epothilone d in treating tau-associated diseases including alzheimer's disease Download PDF

Info

Publication number
US20090270465A1
US20090270465A1 US12/429,492 US42949209A US2009270465A1 US 20090270465 A1 US20090270465 A1 US 20090270465A1 US 42949209 A US42949209 A US 42949209A US 2009270465 A1 US2009270465 A1 US 2009270465A1
Authority
US
United States
Prior art keywords
epothilone
brain
disease
tau
dose
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
US12/429,492
Other languages
English (en)
Inventor
Charles F. Albright
Donna Marie Barten
Francis Y. Lee
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40887113&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20090270465(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US12/429,492 priority Critical patent/US20090270465A1/en
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, FRANCIS Y., ALBRIGHT, CHARLES F., BARTEN, DONNA MARIE
Publication of US20090270465A1 publication Critical patent/US20090270465A1/en
Priority to US13/150,671 priority patent/US8673949B2/en
Priority to US14/162,286 priority patent/US20140135367A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P25/28Drugs 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
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • 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

Definitions

  • This invention relates generally to the treatment of Tau-associated diseases using epothilone D, and more specifically, to the treatment of Alzheimer's Disease using epothilone D.
  • AD Alzheimer's disease
  • Age is the greatest known risk factor for AD with an incidence of 25-50% in people aged 85 years or older.
  • AD is the fifth leading cause of death in people aged 65 and older, and most patients eventually need nursing home care. Consequently, AD has an enormous economic impact, e.g., estimated direct and indirect costs for 2005 in the US only were $148 billion. Besides the economic costs, AD has a devastating impact upon patients and their family members, causing severe emotional distress and turmoil.
  • AD Alzheimer's disease
  • a diagnosis of AD can only be confirmed post-mortem as the clinical diagnosis is based on brain neuropathology, specifically, the diagnosis requires an evaluation of brain tissue, including the existence and concentration of extracellular plaques in the brain, intracellular tangles, and brain neurodegeneration. Dementia is also a required part of the diagnosis, since plaques and tangles are observed in cognitively normal adults, although usually to a lesser extent.
  • AD N-methyl-D-aspartic acid
  • Microtubule stabilizers have been suggested as therapies to treat tauopathies including AD. See, e.g., Lee et al. (references list, infra).
  • Botowski et al. suggest use of paclitaxel (TAXOL®) to treat AD patients by stabilizing microtubules.
  • TAXOL® paclitaxel
  • Paclitaxel has proven highly effective as a microtubule-stabilizing agent in treating cancer patients; however, it presents brain-penetration and peripheral neuropathy issues when considered for AD (further described below), and has not emerged as a viable therapy to treat AD.
  • Epothilone A and epothilone B are naturally-occurring compounds that were isolated by Hofle et al. from fermentation products of the microorganism Sorangium cellulosum (e.g., WO 93/10121). Hofle et al. also discovered 37 natural epothilone variants and related compounds produced by S. cellulosum and modified strains, including epothilones C, D, E, F and other isomers and variants (e.g., U.S. Pat. No. 6,624,310).
  • Ixabepilone a semi-synthetic analog of epothilone B, for treatment of cancer.
  • Ixabepilone has the structural formula:
  • ixabepilone is (1S,3S,7S,10R,11S,12S,16R)-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[(1E)-1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-17-oxa-4-azabicyclo[14.1.0] heptadecane-5,9-dione. See also U.S. Pat. No. 6,605,599, assigned to the current assignee, Bristol-Myers Squibb Company (BMS).
  • BMS Bristol-Myers Squibb Company
  • Ixabepilone is a microtubule-stabilizing agent that has been approved by the FDA for treatment of metastatic breast cancer and is sold by BMS under the tradename IXEMPRA®.
  • Ixabepilone can be prepared as described in U.S. Pat. No. 6,605,599 or 7,172,884, incorporated herein by reference.
  • epothilone B a/k/a patupilone, or EPO-906
  • EPO-906 Phase III trials by Novartis Pharma AG
  • sagopilone or ZK-EPO
  • benzothiazolyl-7-propenyl synthetic analog of epothilone B in Phase II trials by Bayer Schering AG for treatment of various cancers including tumors of the ovary, breast, lung, prostate and melanoma.
  • sagopilone or ZK-EPO
  • epothilone D had advanced to Phase II clinical trials by Kosan Biosciences, Inc. (now a wholly-owned subsidiary of BMS) for treatment of non-small-cell lung cancer and solid tumors, and epothilone D had advanced to Phase II clinical trials for treatment of cancer by Kosan in collaboration with Hoffmann-La Roche, Inc.; however, the clinical trials with epothilone D for treating cancer were discontinued in 2007.
  • the structure for epothilone D can be represented by the following formula:
  • epothilone D compound is claimed, as composition of matter, in U.S. patent application Ser. No. 09/313,524 to Hofle et al., and described in U.S. Pat. Nos. 6,242,469 and 6,284,781 to Danishefsky et al., which application and patents were the subject of Interference No. 105,298, before the USPTO Board of Patent Appeals and Interferences.
  • BMS-310705 (Compound II herein), for cancer therapy.
  • BMS-310705 was pursued through Phase I clinical trials for treatment of ovarian cancer; it is an amino-epothilone F analog and has the chemical structure:
  • Compound II (BMS 310705) can be prepared as described in U.S. Pat. No. 6,262,094, incorporated herein.
  • BBB blood-brain barrier
  • BBB penetration is usually sought to be avoided, whereas for a drug designed to treat AD or other neurodegenerative brain diseases, good BBB penetration is necessary for the compound to be effective.
  • paclitaxel is a highly-successful cancer drug, it has not emerged as a useful therapy to treat brain diseases such as AD, as it has a low rate of brain penetration through the BBB.
  • microtubule-stabilizing drugs in treating AD and other brain diseases involve the ability of a drug to penetrate the brain, to be retained in the brain for long periods, and to selectively accumulate in the brain relative to peripheral tissues. These parameters can be measured using brain-to plasma ratios, brain half-life, and the ratio of the amount of drug retained in the brain as compared with peripheral tissues (most particularly the liver). Additionally, measuring brain penetration, retention and selective brain accumulation with microtubule-stabilizers is complex because these compounds are typically rapidly cleared from plasma but more slowly cleared from microtubule-containing tissues, making it important to set appropriate time windows for comparisons of plasma and tissue levels.
  • the brain-to-peripheral-tissue ratio is a particularly important measurement given that microtubule-stabilizing agents at certain doses are highly cytotoxic to peripheral tissues: when microtubule-stabilizing agents, such as paclitaxel, are administered at chemotherapeutic doses, a peripheral neuropathy and other side effects often occur (Postma et al. 1999). These side effects may be tolerable in treating cancer patients but a different therapeutic window and acceptable side-effect profile exists in treating patients suffering from AD and other brain diseases.
  • Lichtner et al. report brain and plasma concentration data for the above three epothilone analogs, but only for periods of up to 40 minutes.
  • Lichtner et al. are not able to report comparative data against paclitaxel on brain-to-plasma levels because their paclitaxel brain levels were below the level of detection, and they do not report data relating to brain-to-liver ratios, half-life, or brain retention for any of the compounds (e.g., concentration of drug in brain tissue over extended periods of time).
  • epothilone D achieves a surprisingly advantageous profile in treating Tau-associated diseases, including AD.
  • epothilone D exhibits a remarkable combination of advantageous properties, making the compound particularly well-suited to treat such diseases. These properties include not only a high level of brain penetration across the BBB, but also a surprisingly long half-life in the brain and a surprisingly high selective retention rate in the brain as compared with drug levels found in peripheral tissues, most notably, the liver, over extended periods of time.
  • the inventors have further discovered that surprising, therapeutic advantages in treating Tau-associated diseases, particularly, AD, can be achieved with low dosages of epothilone D, e.g., with dosages that are approximately 100-fold less than those administered to achieve chemotherapeutic effects. Consequently, the inventors have discovered methods that allow for therapies in treating Tau-associated diseases with epothilone D, particularly treatment of AD, without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of the epothilone D to be discontinued. Given the low dose as compared with chemotherapeutic treatments, any side effects are greatly reduced as compared with side effects that are induced upon administration of the epothilones and analogs for treatment of cancer.
  • the present invention provides methods of treating Tau-associated diseases including tauopathies, using epothilone D that exhibit a surprisingly advantageous therapeutic profile, and particularly, a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient.
  • the present invention further provides a pharmaceutical composition comprising epothilone D for treating Tau-associated diseases in a patient, wherein the composition exhibits a treatment profile comprising good brain penetrance, long half-life in the brain, and selective brain retention (e.g., high brain-to-liver ratio), as defined herein.
  • Preferred embodiments comprise pharmaceutical compositions for treating tauopathies, particularly, AD, comprising a therapeutically-effective amount of epothilone D and a pharmaceutically acceptable carrier. Further embodiments and aspects of the invention are set forth below.
  • FIG. 1 shows the basic design of an experiment on Tg4510 mice using epothilone D (Compound I).
  • FIG. 2 shows the results of a Morris water maze (MWM) test of the Tg4510 mice at 2.5 months, prior to dosing with epothilone D (Compound I).
  • FIG. 3 shows the results of a MWM test of the Tg4510 mice at 4.5 months, after 2 months of dosing with epothilone D (Compound I).
  • FIG. 4 shows probe data 18 hours after 5 days of training in the 4.5 month-old Tg4510 mice dosed for 2 months with epothilone D (Compound I).
  • TQ stands for target quadrant
  • AR stands for adjacent right
  • AL stands for adjacent left
  • OP stands for opposite quadrant.
  • Two measures of performance, namely % pathlength (A) and number of platform crossings (B) are described.
  • FIG. 5 shows neuronal counts in the CA1 and CA3 regions of the hippocampus in Tg4510 mice at 5.5 months following treatment with vehicle, 1 mpk epothilone D (Compound I), and 10 mpk epothilone D (Compound I).
  • FIG. 6 shows phosphorylated Tau staining of the Tg4510 mice treated with vehicle, 1 mpk epothilone D (Compound I), and 10 mpk epothilone D (Compound I) in the hippocampus. Representative sections from 3 mice per group are shown. AT8 positive staining is dark grey and black.
  • FIGS. 7A-7B show Gallyas silver staining for neurofibrillary tangles in Tg4510 mice treated with vehicle, 1 mpk epothilone D (Compound 1), or 10 mpk epothilone D (Compound I).
  • FIG. 7A shows representative micrographs of cortical staining, where the black silver stain is positive. Lighter background staining and some staining of blood vessels were observed in non-transgenic mice.
  • FIG. 7B shows the quantitation of the silver stain in both cortex and hippocampus.
  • FIGS. 8A-8D show the concentration of Compound II ( FIG. 8A ), ixabepilone ( FIG. 8B ), paclitaxel ( FIG. 8C ) and epothilone D (Compound I) ( FIG. 8D ) in the plasma, brain, and liver of mice following intravenous administration at various intervals of up to 24 hours.
  • FIG. 9 shows the concentration of epothilone D (Compound 1) and Compound III (as described in Example 7 herein) in the brain after oral administration (35 mpk) up to 5 to 24 hours after dosing.
  • FIG. 10 shows the concentration of epothilone D in the plasma, brain and liver in mice after time intervals up to one week after dosing.
  • “About” or “approximately” as used herein means within an acceptable range of standard deviation for the particular value as determined by one of ordinary skill in the art, considering the measurement in question and the instrument used to make the measurement (i.e., the limitations of the measurement system). For example, “about” can mean within one or more standard deviations. As applied to formulations and dosages, “about” can mean a deviation within 10%, more preferably within 5%, and even more preferably, within 2%, of the numbers reported.
  • Brain penetrance refers to the ability of a compound to cross the BBB. Because of the rapid peripheral clearance for most microtubule stabilizing agents, it is important to measure brain-to-plasma ratios at relatively short times post-dosing, e.g., at periods of about of 20 min to 1 h post-dosing, to assess brain penetrance itself.
  • a compound having good brain penetrance as defined herein means a compound which at 20 min to 1 h post-dosing will show a brain-to-plasma ratio of 0.5 or greater, more preferably, 0.8 or greater, and most preferably, a ratio of 1 or more (again, at a time between 20 min and 1 h post-dosing).
  • Cognitive benefits means that an improvement or lessening in decline of cognitive function for at least one patient in need of treatment is observed or reported, as characterized by cognition tests, measures of global function, and activities of daily living and behavior. Typically, cognitive benefits are measured with cognition tests designed to measure cognitive decline in a patient or group of patients.
  • Such tests include cognition tests like ADAS-cog (Alzheimer's disease Assessment Scale, cognitive subscale) and the MMSE (Mini-mental state exam); behavior tests like the NPI (Neuropsychiatric Inventory); daily living activity tests like the ADCS-ADL (Alzheimer's Disease Cooperative Study-Activities of Daily Living); and global function tests such as the CIBIC-plus (Clinician Interview Based Impression of Change), and CDR sum of boxes (Clinical Dementia Rating).
  • cognition tests like ADAS-cog (Alzheimer's disease Assessment Scale, cognitive subscale) and the MMSE (Mini-mental state exam)
  • behavior tests like the NPI (Neuropsychiatric Inventory)
  • daily living activity tests like the ADCS-ADL (Alzheimer's Disease Cooperative Study-Activities of Daily Living)
  • global function tests such as the CIBIC-plus (Clinician Interview Based Impression of Change), and CDR sum of boxes
  • Extended periods of time means period of 24 hours or more, typically 24 to 76 h.
  • “High selective retention rate” or “high selective retention” as used herein means that the drug or compound is retained in one tissue or organ, specifically the brain, at a much higher level than is found in other tissues and organs, especially the liver, as measured at an extended period of time post-dosing. More particularly as defined herein, a high selective retention rate means the concentration of drug in the brain is 4 or more times that found in the liver at 24 or more h post-dosing, more preferably, a factor of at least 6 or more, and most preferably, at a factor of at least 8 or more at 24 h or more h post-dosing. In assessing whether a compound or drug satisfies this standard of high selective retention (e.g., as recited in the claims herein), naturally non-human studies must be relied upon as human brain tissue cannot be analyzed to assess drug concentration.
  • “Impact on underlying disease” means an improvement in a measure of the biomarkers and other parameters associated with the disease process, including biochemical markers in CSF or plasma, changes in brain volume, changes in brain function as measured by functional imaging, and changes in histopathology or biochemistry that might be observed after autopsy.
  • biomarkers that may be used for AD clinical trials include analytes measured in CSF such as Tau, phosphoTau, beta-amyloid, and isoprostanes, as well as brain imaging modalities such as fluorodeoxyglucose PET and volumeteric MRI.
  • Additional biomarkers that potentially may be useful, particularly those examining synaptic activity, MT integrity/function, and oxidative stress include, but are not limited to: GABA, neuropeptide Y, alpha-synuclein, neurogranin and vasoactive intestinal peptide, tubulin, Tau fragments, ubiquitinated proteins, soluble forms of amyloid precursor protein, chromogranin B, 4-hydroxy nonenal, nitrotyrosine, and 8-hydroxy-deoxyguanidine.
  • “Intermittent” when used with reference to a dosing schedule means that there are breaks in the dosing schedule that are irregular. For example, a daily, weekly, biweekly, or monthly dosing schedule is not considered intermittent under this definition, because the break between doses is in each instance regular and defined by the dose cycle of administering the drug. However, a more elaborate dosing schedule with one or more irregular breaks would be considered intermittent, such as 5 days on, followed by 2 days off; or a dose administered on days 1, 8 and 15, of a 30 day cycle, and so forth.
  • Long half-life or “long brain half-life” as used herein means that a drug has a half-life of 20 or more h post-dosing (which is considered dose-independent), and more preferably, for a period 30 or more h post-dosing, and most preferably, for a period of 40 or more h post-dosing.
  • h post-dosing which is considered dose-independent
  • h post-dosing which is considered dose-independent
  • “Low dose” as used herein means a dose of the epothilone D compound that is significantly less than that administered to achieve chemotherapeutic effects (e.g., given a particular mode of administration, clinical trial, and/or experiment), preferably a dose that is 10-fold or less than the chemotherapeutic dose, more preferably a dose that is 50-fold or more less, and even more preferably a dose that is 100-fold or more fold less than the chemotherapeutic dose, i.e., that previously assessed as chemotherapeutically effective using the same administrative method for the given experiment, study or trial.
  • a dose administered was 100 mg/m 2 administered as a 90 min.
  • a low dose relative to this clinical trial dose would mean a cumulative one-month dose following IV administration of 30 mg/m 2 or less, more preferably a dose of 6 mg/m 2 or less, and even more preferably a dose of 3 mg/m 2 or less.
  • a low dose as compared with the above clinical trial dose when administered once every 4 weeks would be a dose of 30 mg/m 2 , more preferably a dose of 6 mg/m 2 , and even more preferably a dose of 3 mg/m 2 .
  • the relative dosages i.e., assessment whether a given dose is a “low dose” as defined herein, should be based on a comparison involving the same or similar modes of administration.
  • “Patient in need of treatment” as used herein is intended to include use of epothilone D for a patient 1) already diagnosed with a Tau-associated disease (including a tauopathy, particularly AD) at any clinical stage, including patients having mild cognitive impairment to advanced dementia; and/or 2) who has early or prodromal symptoms and signs of a Tau-associated disease (including a tauopathy, particularly AD); and/or 3) who has been diagnosed as susceptible to a Tau-associated disease (including a tauopathy, particularly AD), due to age, hereditary, or other factors for whom a course of treatment is medically recommended to delay the onset or evolution or aggravation or deterioration of the symptoms or signs of disease.
  • “Statistically significant cognitive benefits” means that there are cognitive benefits (e.g., improvement or the lessening in decline of cognitive function), following a period of 6 months to a year of treatment for at least 10% or more of patients evaluated, more preferably at least 25% or more patients, and even more preferably, 50% or more of the patient group.
  • improvement at a rate as compared with a control group is assessed and reflects an at least 10% improvement (e.g., as evaluated based on comparative test scores between placebo and control, wherein “improvement” is intended to include reduction in decline in a patient's condition), more preferably, improvement at a rate of more than 25% or more is observed, and most preferably, at a rate of 35% or more.
  • Tau-associated disease as defined herein means diseases associated with abnormalities in Tau as well as diseases that are “tauopathies.”
  • Tau-associated diseases include, but are not limited to, frontotemporal dementia, including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, agyrophilic grain disease, as well as Parkinson's disease, Down syndrome, post-encephalic Parkinsonism, myotonic dystrophy, Niemann-Pick C disease, dementia pugilistica, Blint disease, prion diseases, amyotrophic lateral sclerosis, Parkinsonism-dementia complex of Guam, multiple sclerosis, glaucoma, diabetic retinopathy, and traumatic brain injury; as well as Huntington's disease, Lewy body dementia, Charcot-Marie-Tooth disease, hereditary spastic paraplegia, and multiple system atrophy.
  • tauopathy as defined herein means a neurodegenerative disease associated with fibrillar forms of Tau protein (tangles) in brain. These diseases include AD; however, other tauopathies include, but are not limited to, frontotemporal dementia, including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and agyrophilic grain disease.
  • FTDP-17 chromosome 17
  • “Therapeutically-effective amount of epothilone D” is meant an amount of epothilone D sufficient to:
  • a Tau-associated disease preferably, a tauopathy, and more preferably, AD
  • cognitive functions such as dementia, memory loss, reduced comprehension, dexterity in performing daily living activities, and/or centrally-mediated effects such as motor deficits and vision; and/or
  • the epothilone D pharmaceutical compound is therapeutically effective in not only relieving or alleviating the symptoms of the Tau-associated disease (preferably, a tauopathy, and more preferably, AD), but also is effective in having an impact on underlying disease (i.e., as defined above).
  • epothilone D administered for the treatment of a Tau-associated disease achieves a surprising level of brain penetration, long brain half-life, and selective retention, particularly as compared with other microtubule stabilizers.
  • the inventors further have discovered that remarkably, increased therapeutic effects in treating Tau-associated diseases (particularly tauopathies, and more particularly, AD), are achieved with low doses of epothilone D.
  • a relatively low dosage of epothilone D can be administered for effective treatment of a Tau-associated disease, preferably AD.
  • the inventors have thus developed a method of treating Alzheimer's disease employing the administration of epothilone D to a patient having AD.
  • the method is expected to be therapeutically effective in treating AD in human patients while also posing significantly less serious or fewer side effects as compared with the side effects that typically occur when microtubule stabilizers are administered to human patients for chemotherapy.
  • Such side effects that are reduced or eliminated may include one or more of gastrointestinal distress (including, without limitation, nausea, diarrhea, stomatitis/mucositis, vomiting, anorexia, constipation, and/or abdominal pain), liver toxicity, neutropenia, leucopenia, myelosuppression, alopecia, myalgia/arthralgia, fatigue, musculoskeletal pain, nail disorder, pyrexia, headache, skin exfoliation, and/or neurosensory effects at various grade levels.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D compound has two or more properties selected from good brain penetrance, a long brain half-life, and a high selective retention rate, as defined herein, more preferably, where the epothilone D demonstrates all three properties of good brain penetrance, long brain half-life, and a selective retention rate, as these terms are defined herein.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D compound upon administration has properties selected from two or more of:
  • brain penetrance of 0.5 or greater, more preferably, 0.8 or greater, most preferably, 1 or greater, as measured at 20 min. to 1 h post-dosing; and/or
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D compound upon administration has properties selected from all three of:
  • brain penetrance of 0.5 or greater, more preferably, 0.8 or greater, most preferably, 1 or greater, as measured at 20 min. to 1 h post-dosing; and/or
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the method is therapeutically effective in treating AD in the patient without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said method to be discontinued.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the method provides cognitive benefits, more preferably, statistically-significant cognitive benefits, in treating AD, without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said method to be discontinued.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the method has an impact on underlying disease, more preferably, a statistically-significant impact on underlying disease, without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said method to be discontinued.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein method has an impact on underlying diseases, provides cognitive benefits, and/or is otherwise therapeutically effective, without causing side effects such as gastrointestinal side effects, leucopenia, and/or neurotoxicity, that would require use of said method to be discontinued.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the dose of epothilone D is a low dose, as defined herein.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the dose of epothilone D is between 0.001-10 mg/m 2 , or alternatively, at a dose between 0.00003-0.3 mpk, administered on a daily, weekly, or intermittent dosing cycle.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D is administered via IV, and the dose of epothilone D over a cumulative monthly dosing cycle (i.e., total dosage of compound administered over a one month cycle, regardless of schedule, e.g., weekly, bi-weekly, 3 week on, 1 week off, etc.) is in the range between 0.001-5 mg/m 2 , more preferably between 0.01-5 mg/m 2 , even more preferably between 0.01-3 mg/m 2 , yet even more preferably between 0.1-3 mg/m 2 , and most preferably between 0.1-1 mg/m 2 .
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D is administered orally, and the dose of epothilone D calculated on a daily basis is in the range between 0.001-2 mg/m 2 , more preferably between 0.01-2 mg/m 2 , even more preferably between 0.1-2 mg/m 2 , yet even more preferably between 0.2-2 mg/m 2 .
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D is administered orally, and the dose of epothilone D for a cumulative monthly basis (i.e., total dosage of compound administered over a one month cycle, regardless of schedule, e.g., daily, weekly, bi-weekly, etc.) is in the range between 0.03-60 mg/m 2 , more preferably between 0.30-60 mg/m 2 , even more preferably between 3-60 mg/m 2 , yet even more preferably between 6-60 mg/m 2 .
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D is administered orally on a dosing schedule selected from once daily, once weekly, once every two weeks, or once a month.
  • a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the epothilone D is administered orally on a dosing schedule selected from once daily, and wherein the daily dose of epothilone D is between 0.2 to 2 mg/m 2 .
  • tauopathy-associated disease there are provided methods of treating other tauopathies, besides AD, according to any one of the embodiments of the invention recited above.
  • other tauopathies may include one or more of the diseases referenced in the definition of “tauopathy-associated disease” herein.
  • one embodiment of the invention comprises use of epothilone D, according to any of the above embodiments, to treat not only AD but also a disease selected from frontotemporal dementia, including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and agyrophilic grain disease, Parkinson's disease, Down syndrome, post-encephalic Parkinsonism, myotonic dystrophy, Niemann-Pick C disease, dementia pugilistica, Blint disease, prion diseases, amyotrophic lateral sclerosis, Parkinsonism-dementia complex of Guam, multiple sclerosis, glaucoma, diabetic retinopathy and/or traumatic brain injury.
  • frontotemporal dementia including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration,
  • a preferred embodiment comprises use of epothilone D, according to any of the embodiments described herein, to treat a tauopathy, including, without limitation, a disease selected from AD, frontotemporal dementia, including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and agyrophilic grain disease.
  • a tauopathy including, without limitation, a disease selected from AD, frontotemporal dementia, including the subtype of frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and agyrophilic grain disease.
  • epothilone D for use in treating a Tau-associated disease, more preferably, a tauopathy, most preferably AD.
  • one combination of the above inventive methods may comprise a method of treating Alzheimer's disease comprising the step of administering a therapeutically effective amount of epothilone D to a patient, wherein the method is therapeutically effective in treating AD in the patient without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said method to be discontinued; and wherein the dose of epothilone D is a cumulative monthly dose of between 0.001-5 mg/m 2 , administered via IV; and/or wherein the dose of epothilone D is between 0.001 to 2 mg/m 2 , administered PO daily; and/or wherein the dose of epothilone D is selected from a dose within any one of the preferred ranges expressed above for oral or IV administration.
  • any of the recited methods of treatment may by combined with the embodiment involving epothilone D, for use in treating a tauopathy, preferably AD, in a human patient.
  • a tauopathy preferably AD
  • one embodiment of the invention comprising a combination of the above alternative embodiments, would comprise epothilone D for treating AD, wherein the use is therapeutically effective in treating AD, and wherein the epothilone D is administered to the patient at a dose between 0.001-10 mg/m 2 , or alternatively, at a dose between 0.00003-0.3 mpk, administered on a daily, weekly, or intermittent dosing cycle.
  • Yet another embodiment would comprise epothilone D, for treating a tauopathy, particularly AD, wherein the epothilone D is administered to a human patient at a low dose and is therapeutically effective in having an impact on underlying disease and/or providing cognitive benefits.
  • a pharmaceutical formulation comprising epothilone D suitable for administration to a human patient in need of treatment for a Tau-associated disease, preferably a tauopathy, more preferably, AD, wherein administration of the formulation is therapeutically effective in treating the disease in the patient without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said epothilone D formulation to be discontinued.
  • a pharmaceutical formulation comprising epothilone D suitable for administration to a human patient for treating a Tau-associated disease, preferably a tauopathy, more preferably AD, wherein administration of the formulation provides statistically-significant cognitive benefits in treating the disease, without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said epothilone D formulation to be discontinued.
  • a pharmaceutical formulation comprising epothilone D suitable for administration to a human patient for treating a Tau-associated disease, preferably a tauopathy, more preferably, AD, wherein the formulation is effective in providing an impact on underlying disease, without causing drug-induced side effects and/or drug-plasma concentration levels that would require use of said epothilone D formulation to be discontinued.
  • a pharmaceutical formulation for IV administration to a human patient wherein said formulation is suitable for delivery of a cumulative monthly dose of epothilone D in the range between 0.001-5 mg/m 2 , more preferably between 0.01-5 mg/m 2 , even more preferably between 0.01-3 mg/m 2 , yet even more preferably between 0.1-3 mg/m 2 , and most preferably between 0.1-1 mg/m 2 .
  • a pharmaceutical formulation for oral administration to a human patient wherein said formulation is suitable for delivery of a cumulative monthly oral dose of epothilone D in the range between 0.03-60 mg/m 2 , more preferably between 0.30-60 mg/m 2 , even more preferably between 3-60 mg/m 2 , yet even more preferably between 6-60 mg/m 2 .
  • a pharmaceutical formulation for administration to a human patient wherein said formulation comprises epothilone D in a pharmaceutically acceptable solvent system comprising from about 0 to 50% propylene glycol, about 1 to 10% TPGS, about 0.5 to 10% ethanol, about 0-90% water, and/or about 5 to 85% PEG such as PEG-400.
  • Tauopathies are neurodegenerative diseases associated with abnormal forms of Tau protein in brain tissue.
  • AD Alzheimer's Disease
  • neurofibrillary tangles the presence of which is one of the hallmark pathologies in AD—were found to contain fibrillar, hyperphosphorylated, conformationally-altered forms of the Tau protein.
  • other tauopathies were identified including frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and agyrophilic grain disease.
  • a link with Tau abnormalities has been associated with Parkinson's disease, Down syndrome, post-encephalic Parkinsonism, myotonic dystrophy, Niemann-Pick C disease, dementia pugilistica, Blint disease, prion diseases, amyotrophic lateral sclerosis, Parkinsonism-dementia complex of Guam, multiple sclerosis, glaucoma, diabetic retinopathy and traumatic brain injury (Avila et al. 2004; Bartosik-Psujek et al. 2006; Dickey et al. 2006; Wostyn et al. 2008).
  • Tau is a 50 to 75 kDa microtubule-associated protein (MAP) that binds and stabilizes microtubules (MTs).
  • MAP microtubule-associated protein
  • MTs microtubules
  • the splice variants contain zero, one, or two (0N, 1N, or 2N) N-terminal inserts in combination with either three repeats (3R) or four repeats (4R) of a microtubule-binding domain.
  • the repeat domains are necessary for microtubule stabilization, while proline-rich regions on either side of the repeat domains are necessary for binding to the microtubules (Preuss et al. 1997).
  • the repeat domains and proline-rich regions are phosphorylated by multiple kinases, leading to dissociation of Tau from microtubules.
  • the N-terminus of Tau extends away from the microtubule surface, where it is believed to assist in determining the spacing between microtubules and in binding of the motor protein dynactin to the microtubules (Magnani et al. 2007).
  • 3R Tau and 4R Tau there are roughly equal levels of 3R Tau and 4R Tau present. 4R Tau binds more tightly to microtubules than does 3R Tau.
  • Tau is most abundant in neurons where it is predominantly localized to axons. Tau is the major microtubule-associated protein in neuronal axons, while the MAP1 family is widely distributed in neurons, and the MAP2 family is predominantly somatodendritic. Tau stabilizes axonal microtubules, thereby facilitating transport of proteins, organelles, lipids, cellular components targeted for degradation, and cell signaling molecules bi-directionally between the cell body and the synaptic terminals. Tau dysfunction could interfere with axonal trafficking and thereby affect neuronal function and survival.
  • the Tau gene can be knocked out in mice with mild consequences, but if both Tau and MAP 1B genes are removed, the double knockout mice die as embryos. It appears that alterations in expression of some MAPs can substitute for each other in many cases, including development (Avila et al. 2004).
  • mutations in the gene encoding Tau cause tauopathies, particularly in FTDP-17 and other frontotemporal dementias.
  • Many FTDP-17 mutations decrease binding to microtubules in vitro and/or increase their propensity to form fibrils (Lace et al. 2007).
  • Other tauopathy-associated mutations alter the splice pattern of Tau to generate predominantly 3R or 4R Tau.
  • Yet another class of Tau mutations on the N-terminus alters the ability to bind to dynactin (Magnani et al. 2007). All of these mutations have the potential to interfere with normal functions of Tau.
  • AD it is thought that ⁇ -amyloid (A ⁇ ) leads to abnormalities in Tau.
  • Tau aggregated into neurofibrillary tangles is not directly pathogenic include observations of human brains and mouse models. For instance, examination of different regions and disease stages of Alzheimer's disease brains has led to the conclusion that neurons can survive and function with neurofibrillary tangles for decades (Morsch et al. 1999).
  • human Tau (hTau) transgenic mice have tangles and severe neurodegeneration, but the neurons with tangles do not show selective signs of distress and are too few in number to account for the dramatic loss in neurons observed in this model (Andorfer et al. 2005).
  • Tg4510 an inducible Tau transgenic line, shows dramatic and rapid tangle formation, neurodegeneration, and behavioral deficits when Tau-P301L is induced (Santacruz et al., 2005). When Tau-P301L expression is repressed, neurodegeneration and cognitive deficits are greatly reduced, but tangle formation continues. Further studies using these mice show that soluble Tau multimers correlate with cognitive deficits.
  • PrP T44 Tau transgenic mice were treated with paclitaxel (Zhang et al., 2005). PrP T44 mice overexpress normal human 0N3R Tau in spinal cord neurons and consequently develop motor deficits due to Tau overexpression. Paclitaxel treatment for 3 months reduced motor dysfunction and increased microtubule numbers and axonal transport in the ventral roots of the spinal cord. Although paclitaxel is poorly CNS-penetrant, it was able to influence the efferent axons from neurons in the ventral horn of the spinal cord which are outside the blood-brain barrier. Interestingly, Tau pathology in this model (spheroids) was unaffected.
  • NAP neurotrophic, anti-inflammatory, anti-apoptotic, and neuroprotective activities in many cellular and in vivo models, including middle cerebral artery occlusion (stroke model), head trauma, cholinotoxic lesions, aging, and developmental defects in fetal alcohol syndrome and apolipoprotein E deficient mice (Gozes et al. 2006; Gozes 2007).
  • NAP administration for 3 or 6 months is reported to reduce A ⁇ levels, hyperphosphorylated Tau, and sarcosyl insoluble Tau while increasing soluble Tau in 3 ⁇ Tg mice (Matsuoka et al. 2007; Matsuoka et al. 2008).
  • 3 ⁇ Tg mice overexpress APP and Tau-P301L (Oddo et al. 2003).
  • the mechanism of NAP activity is not fully defined, but there is evidence, based on binding of tubulin to a NAP affinity column and effects on microtubule formation and/or stabilization in cultured neurons, that NAP binds to microtubules (Divinski et al. 2006).
  • NAP is also known to inhibit A ⁇ aggregation, so it may be acting upstream of Tau in the 3 ⁇ Tg model.
  • NAP is not likely to act as a typical microtubule-stabilizing agent, as it is able to protect against paclitaxel-induced peripheral neuropathy in rats (U.S. Patent Application Publication No. 2006/0247168 A1).
  • microtubule-stabilizing agents such as paclitaxel
  • a peripheral neuropathy often occurs (Postma et al. 1999) that is believed to result from the over-stabilization and bundling of microtubules in peripheral nerves. Since NAP prevents paclitaxel-induced peripheral neuropathy in rats, paclitaxel and NAP are unlikely to act through identical mechanisms.
  • microtubule stabilizers could have neuroprotective effects unrelated to obvious Tau dysfunction.
  • Microtubule-stabilizing compounds protect cultured neurons from multiple toxic insults, including A ⁇ 42, oxidative stress from soluble A ⁇ 40, lysosomal disruption, calcium-induced toxicity, and glutamate-induced toxicity (Burke et al. 1994; Furukawa 1995; Sponne et al. 2003; Michaelis et al. 2005; Butler et al. 2007). It is hypothesized that microtubules play a key role not only in transport mechanisms, but also in regulation of cell signaling, particularly calcium signaling, possibly through anchoring of macromolecular signaling complexes in the vicinity of the plasma membrane (Michaelis et al.
  • Microtubule stabilizing agents have also been shown to enhance mitochondrial function by reducing reactive oxygen species generation and increasing expression of the oxidative phosphorylation genes involved in ATP production (Wagner et al. 2008). Microtubule-stabilizing agents are also known to broadly influence cell signaling during disruption of the mitotic spindle in cancer cells (Bergstralh et al. 2006).
  • microtubule stabilizers for tauopathies and other neurodegenerative diseases
  • microtubule stabilizers can cause toxicity to peripheral tissues, such as inhibition of cell proliferation, particularly in the gastrointestinal tract and hematopoietic cells, and peripheral neuropathy. It is thus highly desired to identify microtubule stabilizers with excellent brain penetration and selective retention in the brain as compared with peripheral tissues, so as to maximize the therapeutic index for tauopathies and other neurodegenerative diseases.
  • the ability of compounds to bind with a longer half life to brain tissue relative to peripheral tissues is a highly desired property.
  • the taxane series of microtubule stabilizers are substrates of multiple multi-drug resistance transporters, such as P-glycoprotein (POP), ATP-binding cassette, multidrug resistance protein, and breast cancer resistance protein.
  • POP P-glycoprotein
  • ATP-binding cassette multidrug resistance protein
  • breast cancer resistance protein multidrug resistance protein
  • These multi-drug resistance transporters prevent compounds from accumulating in tumor and brain tissue.
  • Multiple labs have worked to synthesize taxanes that are not substrates for multi-drug resistance transporters, particularly PGP, with limited success (Minderman et al. 2004; Rice et al. 2005; Ballatore et al. 2007).
  • Co-administration of a PGP inhibitor with paclitaxel has also been attempted (Fellner et al. 2002).
  • Epothilone D is a known compound which has been chemically synthesized de novo and also has been isolated from fermentations of Sorangium cellulosum strains as minor products in the fermentation of S. cellulosum .
  • Total synthesis of epothilone D is reported in U.S. Pat. No. 6,242,469 to Danishefsky et al., and additional methods for preparing epothilone D and other epothilone compounds can be found at U.S. Pat. Nos. 6,204,388, 6,288,237, 6,303,342; WO 03/072730, U.S. Pat. No. 6,410,301; U.S. Patent Application Publication No. 2002/0137152A1; U.S. Pat. No.
  • epothilone D used in methods of the present invention can be administered to a patient in various ways known in the art, typically by intravenous (IV) administration, subcutaneous administration, oral administration, and so on.
  • epothilone D can be formulated with a pharmaceutically acceptable vehicle or diluent.
  • a pharmaceutical composition comprising epothilone D can be formulated in a classical manner using solid or liquid vehicles, diluents, and additives appropriate to the desired mode of administration.
  • compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution (0.9% Sodium Chloride Injection [Normal Saline] or 5% Dextrose Injection), or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution (0.9% Sodium Chloride Injection [Normal Saline] or 5% Dextrose Injection), or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids.
  • compositions and/or methods of administering compounds of the invention may include use of co-solvents including, but not limited to ethanol, N,N dimethylacetamide, propylene glycol, glycerol and polyethylene glycols, e.g., polyethylene glycol 300 and/or polyethylene glycol 400.
  • co-solvents including, but not limited to ethanol, N,N dimethylacetamide, propylene glycol, glycerol and polyethylene glycols, e.g., polyethylene glycol 300 and/or polyethylene glycol 400.
  • Surfactants may be used to increase a compound's spreading or wetting properties by reducing its surface tension, including without limitation, d- ⁇ -Tocopheryl polyethlene glycol 1000 succinate (TPGS), Cremophor, Solutol HS 15, polysorbate 80, polysorbate 20, poloxamer, pyrrolidones such as N-alkylpyrrolidone (e.g., N-methylpyrrolidone) and/or polyvinylpyrrolidone; however, use of Cremophor has disadvantages and is not preferred.
  • the formulation may also comprise use of one or more “buffers” (e.g., an ingredient which imparts an ability to resist change in the effective acidity or alkalinity of a medium upon the addition of increments of an acid or base), including, without limitation, sodium phosphate, sodium citrate, diethanolamine, triethanolamine, L-arginine, L-lysine, L-histidine, L-alanine, glycine, sodium carbonate, tromethamine (a/k/a tris[hydroxymethyl]aminomethane or Tris), and/or mixtures thereof.
  • buffers e.g., an ingredient which imparts an ability to resist change in the effective acidity or alkalinity of a medium upon the addition of increments of an acid or base
  • buffers e.g., an ingredient which imparts an ability to resist change in the effective acidity or alkalinity of a medium upon the addition of increments of an acid or base
  • buffers e.g., an ingredient which imparts an ability to resist
  • Formulations for administering epothilone compounds including formulations that avoid use of non-ionic surfactants such as Cremophor, are described in the prior art.
  • a formulation for use in IV administration that comprises a mixture of propylene glycol and ethanol is described in U.S. Pat. No. 6,683,100.
  • Further formulations may comprise mixtures of polyethylene glycol/dehydrated alcohol, or propylene glycol or glycerol/dehydrated alcohol.
  • WO 2006/105399 (PCT/US2006/011920) to BMS, discloses formulations that include mixtures of about 30 to 70 percent by volume dehydrated alcohol for each 30 to 70 percent by volume PEG 300 and/or PEG 400, which can be diluted with saline or dextrose infusion fluids for IV administration, and may be applied for use in administering epothilone D to patients via IV administration.
  • Optimal ratios of solvents may be readily obtained by one skilled in the field.
  • Embodiments involve use of about 10 mg epothilone D and about 0.4 g of hydroxypropyl-beta-cyclodextrin combined in a 60% tert-butanol-water solution that is then lyophilized (ingredients can be reduced proportionately for preparation of individual, lower dosages units, according to the current invention).
  • the lyophilized active ingredient “cake” can then be reconstituted for IV administration with use of water, ethanol, and/or glycol, which may include propylene glycol, polyethylene glycol 400, polyoxyethylene sorbitan monooleate (sold under the trade name TWEEN 80), and related oxygenated hydrocarbons. It is understood that glycols of various chain lengths and molecular weights (e.g., polyethylene glycol 1000, other TWEEN compounds) may be used.
  • a formulation that may be used to deliver epothilone D to a patient according to the invention may comprise about 0 to 50% propylene glycol, about 1 to 10% TPGS, about 0.5 to 10% ethanol, about 0-90% water, and/or about 5 to 85% PEG such as PEG-400. More specifically, a formulation may comprise:
  • One preferred method of administering epothilone D according to the invention involves oral administration.
  • U.S. Pat. No. 6,576,651 discloses methods for oral administration of epothilones with use of one or more pharmaceutically acceptable acid-neutralizing buffers.
  • a preferred method of administration would involve use of a tablet or capsule, including a solid tablet or capsule or fluid or gelatin-filled capsule.
  • a solid tablet or capsule of epothilone D may be prepared with one or more enteric coatings. Enteric coatings have been used for many years to arrest the release of the drug from orally ingestible dosage forms.
  • the enteric coatings are resistant to stomach acid for required periods of time before they begin to disintegrate and permit slow release of the drug in the lower stomach or upper part of the small intestines.
  • enteric coatings are disclosed in U.S. Pat. Nos. 6,224,910, 5,225,202, 2,809,918, 3,835,221, 4,728,512 and 4,794,001, each of which is incorporated herein by reference.
  • enteric coated tablet directed to use of epothilone D is described in U.S. patent application Ser. No. 11/281,834, incorporated herein by reference, which may be used to formulate tablets of capsules of epothilone to practice the invention.
  • This formulation involves use of an inactive base particle, such as a sugar bead, to which the active ingredient (i.e., epothilone D), is applied, which is then encapsulated by an enteric coating polymer, and/or one or more subcoat layers. The beads are then included within a capsule.
  • Enteric coatings for use in formulating epothilone D tablets or capsules may include enteric coating polymers, such as, for example, hydroxypropyl methylcellulose phthalate, polyvinyl acetate phthalate, cellulose acetate phthalate, acrylic acid copolymers, and methacrylic acid copolymers.
  • enteric coating polymers such as, for example, hydroxypropyl methylcellulose phthalate, polyvinyl acetate phthalate, cellulose acetate phthalate, acrylic acid copolymers, and methacrylic acid copolymers.
  • EUDRAGIT® L-30-D 55 aqueous copolymer dispersion which comprises an anionic copolymer derived from methacrylic acid and ethyl acrylate with a ratio of free carboxyl groups to the ethyl ester groups of approximately 1:1, and a mean molecular weight of approximately 250,000, which is supplied as an aqueous dispersion containing 30 weight % solids.
  • EUDRAGIT® L-30-D 55 aqueous copolymer dispersion is supplied by Rohm-Pharma Co., Germany.
  • suitable materials to form the subcoat layer include starch; gelatin; sugars such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums such as acacia, sodium alginate, methyl cellulose, carboxymethylcellulose, and polyvinylpyrrolidone (PVP) polymers and copolymers such as PVP-PVA copolymers; celluloses such as ethylcellulose, hydroxypropyl cellulose, and hydroxypropyl methyl cellulose; polyethylene glycol; and waxes.
  • the subcoat layer may further comprise one or more plasticizers, such as polyethylene glycol, propylene glycol, triethyl citrate, triacitin, diethyl phthalate, tributyl se
  • the tablet or capsule of epothilone D optionally may comprise other materials such as flavoring agents, preservatives, or coloring agents as may be necessary or desired.
  • epothilone D can be determined by one of skill in the art, taking into consideration the findings described herein together with typical factors such as the body mass of the patient, the physical condition of the patient, and so on.
  • the dosage should contain epothilone D in an amount that is effective for treating Tau-associated diseases, including tauopathies such as AD.
  • a range for the dosage of epothilone D administered for the treatment of Tau-associated diseases is considered to be between 0.0001-10 mg/m 2 , more preferably between 0.001-5 mg/m 2 .
  • Other, more preferred dosage ranges for PO and IV administration are set forth above in the alternative embodiments section.
  • the units mg/m 2 are used herein, for purposes of comparison with the chemotherapeutic dosages previously administered with epothilones and their analogs. However, the units mg/m 2 can be readily converted to mpk, considering the animal species receiving (or having received) the drug and the patient's bodyweight and/or height. For example, for a human patient weighing about 70 kg, the dose range of 0.0001-10 mg/m 2 converts to about 0.00003-0.3 mpk. Further information concerning dose conversions can be found at www.rphworld.com/viewlink-25045.html, and in Freireich et al., Cancer Chemother. Reports, 50(4):219 (1966).
  • the drug can be administered daily, weekly, or on an intermittent basis.
  • the drug can be administered for three weeks on, followed by one week off, or for two weeks on, followed by one week off, or under other dosing schedules as can be determined by one skilled in the field.
  • the particular dose selected will depend upon the mode of administration and dosing regime selected.
  • One preferred schedule is a once daily oral dosing schedule.
  • each unit dose may be larger than when daily dosages are provided.
  • the dose of epothilone D that was administered to patients for treatment of cancer in certain Phase II clinical trials was 100 mg/m 2 administered as a 90 minute infusion given weekly for 3 of 4 weeks (i.e., on days 1, 8, and 15, every 4 weeks), following Phase I trials involving dose escalations of from 9 to 150 mg/m 2 for each dose.
  • the dose of drug contemplated for treatment of AD is about ten-fold less, and more likely, about 100-fold less, and in another contemplated embodiment, even more than 1000-fold less, than the therapeutic dose of epothilone D that was administered for treatment of cancer patients in clinical Phase II trials, although the dosing schedule and mode of administration will influence the dose.
  • Tg4510 an aggressive Tau transgenic mouse line
  • the creation of Tg4510 was recently described (Santacruz et al., 2005; Berger et al., 2007).
  • the Tg4510 line expressed Tau-P301L, a Tau mutant found in FTDP-17, using the calmodulin kinase II promoter.
  • the Tg4510 line was unique in several respects:
  • Epothilone D (Compound I) was dosed intraperitoneally with a 26-gauge needle, in 10% ethanol, 90% water, 10 ml/kg at 0 (vehicle), 1 mpk, and 10 mpk. A 10 ⁇ stock solution was made in 100% ethanol, and diluted just before dosing. Mice were dosed in 3 cohorts and data were combined to give a final N of 12, 9, and 15 for the vehicle, 1 mpk, and 10 mpk groups, respectively. Mice were dosed in a chemical fume hood.
  • mice were used in this study. These mice are a well-characterized, aggressive model of tauopathy that overexpress human P301L mutant Tau in the forebrain (Santacruz et al., 2005; Berger et al., 2007). The mice are characterized by accumulations of abnormal forms of Tau, including tangles similar to those observed in AD brain, behavioral deficits, and eventually neuronal loss.
  • mice were acclimated to handling with a single mock injection of phosphate buffered saline, performed within a chemical fume hood. The mice were then housed in cages kept within the chemical fume hood for 48 hours. Following the 48-hour period, the mice were transferred to clean cages and brought to a behavioral suite for testing.
  • mice were then tested in a Morris water maze (MWM) for six days.
  • the mice were distributed into treatment groups based on the results of the behavioral analysis using the rank scores for probe trial 2 annulus crossing index.
  • Mice were 11 weeks (+/ ⁇ 15 days) of age at the start of dosing and were dosed once weekly.
  • a panel of neurological and physical propensity tests (Modified SHIRPA) were performed following the first week of dosing, including analysis of body position, tremor, coat appearance, gait, touch escape, positional passivity, limb grasping, and righting reflex. Mice were additionally examined 48 hours after each weekly dose for coat appearance, limb grasping, righting reflex and for any overt stereotyped behavior. No signs of overt toxicity, weight loss, or motor deficits were observed in the course of the study.
  • mice were again tested in the MWM after the eighth dose (19 weeks of age +/ ⁇ 15 days) for six days. After behavioral testing, dosing resumed until the animals were 5.5 months of age at the time of harvest. Animals were housed and treated according to Institutional Care and Animal Use Committee and National Institutes of Health standards.
  • mice were tested in Morris water maze (MWM) on two occasions, once prior to dosing, and once two months after dosing began. The second round of water maze testing was performed in another testing room. Mice were acclimated to the experimental room for 2-3 days prior to testing. The mice were placed in a water maze of 1.5 m diameter, with a 16 cm diameter platform placed 0.5-1.0 cm under the surface of the water. The water was made opaque with non-toxic white paint and the water temperature was regulated between 22-25° C.
  • mice were given 4 trials per day of up to 90 seconds each with a 10 second rest period on the platform after each trial. If the mouse did not find the platform within 90 seconds, the mouse was gently guided to the platform and allowed to remain there for 10 seconds.
  • the testing room rooms each had large external cues to allow the mice to orient as they learned the location of the platform. Mice were placed under a heat lamp to prevent hypothermia after each trial. The interval between trials ranged from 25 to 45 minutes.
  • the mice were tracked using HVS Image Advanced Tracker VP200 software (Buckingham, UK) and the total distance traveled until reaching of the platform was determined.
  • Statistical analysis for acquisition path length from the five trials involved a repeated measures analysis of variance.
  • the statistical model included “treatment” (0, 1 mpk, or 10 mpk of epothilone D (Compound I)) as a between animal term, and the 5 trials as repeated measures on each animal. If the analysis indicated a significant effect of treatment, or a treatment-by-trial interaction, differences between the 1 mpk and 10 mpk groups were compared to the vehicle group using Dunnett's test. The probe pathlengths in each quadrant, and number of platform crossings in each quadrant, were analyzed using Dunnett's test. In all cases, 1 mpk and 10 mpk groups were compared to the vehicle group. All calculations were done in SAS, version 9.1, under the Windows XP Professional operating system.
  • mice were euthanized by cervical dislocation at 5.5 months followed by decapitation. Brains were immediately removed and divided down the midline into two hemispheres. The right hemisphere was placed into 20 mL of 4% paraformaldehyde (prepared fresh on the day of sacrifice) and stored overnight at 4° C. The following day, the brains were transferred to a tube containing 20 mL TBS (pH 7.4, 20 mM TRIS, 100 mM NaCl) and then stored at 4° C. until processing. Right hemispheres were embedded in paraffin, sectioned at 5 microns, and mounted on positively charged glass slides. The slides were dried overnight in a 60° C. oven and stored at room temperature until stained. The left hemispheres were frozen (within 2 minutes) on dry ice.
  • TBS pH 7.4, 20 mM TRIS, 100 mM NaCl
  • the Gallyas staining method was used to detect silver-positive neurofibrillary tangles and dystrophic neurites.
  • Paraffin-embedded thin sections (5 microns) mounted on glass slides were deparaffinized and rehydrated via serial incubation in xylene (two times for 10 minutes each), 100% ethanol (two times for 10 minutes each), 95% MeOH 15% H 2 O 2 (30 minutes), 95% ethanol (two times for 5 minutes each), 80% ethanol (two times for 5 minutes each), 50% ethanol (two times for 5 minutes each), and water (two times for 5 minutes each).
  • the sections were then placed into 5% periodic acid for 5 minutes, washed in dH 2 O two times for 5 minutes each time, and placed in alkaline silver iodide solution (containing 1% silver nitrate) for 1 minute.
  • the sections were washed in 0.5% acetic acid for 10 minutes, placed in freshly prepared developer solution for 15 minutes, and washed again in 0.5% acetic acid for 5 minutes. Following a rinse in deionized water, the sections were placed in 0.1% gold chloride for 5 minutes and rinsed again in deionized water. The sections were incubated in 1% sodium thiosulphate (hypo) for 5 minutes and then rinsed in tap water. Counterstain was performed in 0.1% nuclear fast red for 2 minutes. The sections were then rinsed in tap water, dehydrated in graded series of alcohol (95%, 100%, 100%) for 2 minutes, and cleared in 3 changes of xylene, 10 dips each.
  • Paraffin-embedded tin sections (5 microns) were deparaffinized and rehydrated to water in 3 changes of xylene, two changes of 100% ethanol, and 1 change of 95% ethanol, followed by rinsing in water.
  • Antigen retrieval was performed by steaming the slides in 10 mM sodium citrate buffer, pH 6.0 for 30 minutes in a Black and Decker Steamer (Model # HS900) and then cooled for 30 minutes. Endogenous peroxidase activity is removed by incubation in 0.6% hydrogen peroxide in 90% MeOH for 15 minutes. After washing in TBS, slides are blocked in 10% normal goat serum in TBS for one hour.
  • Nuclei were counterstained blue with hematoxylin, followed by dipping slides 2 times in Scott's tap water substitute (Surgipath # 02900, Richmond, Ill.) and then rinsing in tap water. The sections were then dehydrated in graded series of alcohol (95%, 100%, 100%) then cleared in 3 changes of xylene. Cover slips and Cytoseal 60 mounting medium were then added.
  • Nissl stained slides were scanned and digitized using the Aperio ScanScope (Aperio Technologies, Inc., Vista, Calif.). Images of the entire brain section were captured at high resolution and stored as files within Spectrum (Aperio software). To process images, a region of 4,000 ⁇ 4,000 pixels including the entire hippocampus was captured using the extract tool and saved as a JPEG file for importing into Metamorph (Molecular Devices, Sunnyvale, Calif.) for quantification of cell loss within the CA1 and CA3 regions of the hippocampus. A modified version of the single section dissector method (Moller et al. 1990) was utilized to quantify cell loss because of its suitability for thin, paraffin-embedded tissue sections.
  • Every fifth section was collected as the paraffin-embedded brains were cut sagitally between the Bregma and approximately 0.75 mm laterally. Three regions were drawn and counted per section using Metamorph software. The same regions were used for every image and 5 sections were counted per animal, 5 slides apart. Statistics were performed using ANOVA followed by Dunnett's post-hoc test.
  • IP intraperitoneal
  • mice were again tested in the MWM to determine the effect of treatment on cognitive performance.
  • mice were euthanized and brains were collected for subsequent analysis.
  • mice When mice were dosed once weekly intraperitoneally with 1 mpk and 10 mpk epothilone D (Compound I) for 2 or 6 months, no histopathological abnormalities were observed in multiple tissues, including liver, kidney, heart, testes, adrenal gland, bone marrow, peripheral nerve, stomach, and small and large intestines.
  • Compound I When mice were dosed once weekly intraperitoneally with 1 mpk and 10 mpk epothilone D (Compound I) for 2 or 6 months, no histopathological abnormalities were observed in multiple tissues, including liver, kidney, heart, testes, adrenal gland, bone marrow, peripheral nerve, stomach, and small and large intestines.
  • FIG. 2 shows the results of a MWM test of the Tg4510 mice at 2.5 months, prior to dosing with epothilone D (Compound I) or with vehicle. There were no statistically significant differences between the groups prior to dosing in acquisition or during probe trials, which was the basis for separating animals into groups. In other words, FIG. 2 operates as a control in showing the pre-treatment performance of each group was similar.
  • mice were then administered epothilone D (Compound I)) once weekly intraperitoneally at 1 mpk, 10 mpk, and with vehicle, and the MWM test was performed at 4.5 months, following this weekly dosing over 12 weeks.
  • the results which are reported in FIG. 3 revealed that mice treated with 1 mpk epothilone D (Compound I) were able to locate the hidden platform in the MWM more quickly (i.e., in a statistically significant manner (p ⁇ 0.01)), than could mice that were treated with the vehicle.
  • the 10 mpk treatment group showed a trend toward improvement as compared with the vehicle group.
  • FIG. 4 shows probe data 18 h after 5 days of training in the 4.5 month-old Tg4510 mice dosed for 2 months with epothilone D (Compound I) at 1 mpk, 10 mpk, and with vehicle.
  • TQ stands for target quadrant
  • AR stands for adjacent right
  • AL stands for adjacent left
  • OP stands for opposite quadrant.
  • Two measures of performance, namely % pathlength (A) and number of platform crossings (B) in each quadrant, are indicated in FIG. 4 .
  • a preference for the target quadrant indicates that the mouse remembered the location where the platform was located during the acquisition phase of the study.
  • mice performed at chance with similar results for each of TQ, AR, AL, and OP, for both the pathlength (A) and platform crossing (B) measures, and they did not show a quadrant preference.
  • the mice treated with 1 mpk showed statistically significant differences in both measures as compared with the vehicle group in memory, e.g., in recalling that the platform had been located at the TQ.
  • the 10 mpk group showed significantly greater performance compared to the vehicle group in the % pathlength measure (A) but not when using the number of platform crossings measure (B).
  • the Tg4510 mice treated with 1 mpk epothilone D had substantially more CA1 neurons than vehicle-treated animals.
  • the difference between the mice treated with vehicle and the mice treated with 1 mpk of epothilone D shows that the 1 mpk of epothilone D (Compound I) prevented neuronal loss with a statistically significant difference from vehicle (p ⁇ 0.01).
  • the mice treated with 10 mpk of epothilone D (Compound I) had CA1 neuronal levels that were intermediate between the vehicle-treated mice and the mice treated with 1 mpk of epothilone D (Compound I).
  • the effect of treatment on phosphorylated Tau staining in the CA1 region was also examined.
  • the AT8 antibody recognizes Tau that is phosphorylated on both the 202 and 205 residues. This form of hyperphosphorylated Tau is greatly enriched in AD and other Tauopathy patient brains. (Goedert et al. 1995).
  • FIG. 6 shows AT8 phosphoTau staining of the Tg4510 mice treated with vehicle, 1 mpk epothilone D (Compound I), and 10 mpk epothilone D (Compound I) as described above. PhosphoTau staining is indicated in dark black. Surprisingly, the mice treated with 1 mpk of epothilone D (Compound I), showed much less phosphoTau staining, particularly in comparison to the vehicle-treated mice. Mice treated with 10 mpk of epothilone D (Compound I) showed intermediate levels of phosphoTau staining.
  • FIG. 7A shows Gallyas silver staining for neurofibrillary tangles in the frontal cortex of the Tg4510 mice treated with vehicle, 1 mpk of epothilone D (Compound I), and 10 mpk of epothilone D (Compound I) as described above.
  • silver staining is in black (positive), and “NT” stands for non-transgenic, demonstrating some non-specific staining associated with blood vessels.
  • mice treated with 1 mpk of epothilone D had much lower levels of neurofibrillary tangles than did vehicle-treated mice; this is quantitated for all animals in the study in FIG. 7B .
  • a significant impact on underlying disease in both cortex and hippocampous was observed at the 1 mpk dose, with the 10 mpk dose again showing a trend toward improvement.
  • epothilone D (Compound I) prevented cognitive decline and improved cognitive function over time as compared with the untreated Tg4510 mice. Furthermore, neuropathological tests as measures of impact on underlying disease (i.e., cell count, phosphoTau staining, and silver staining tests), demonstrate that treatment with epothilone D prevents neuronal loss, reduces accumulation of abnormal Tau, and prevents the formation of neurofibrillary tangles at statistically significant levels as compared with untreated Tg4510 mice. Thus, the inventors herein believe they are the first to discover and demonstrate the prevention of cognitive loss, Tau pathology, and neurodegeneration upon treatment with a microtubule-stabilizing compound, namely, epothilone D.
  • a microtubule-stabilizing compound namely, epothilone D.
  • the inventors herein have discovered that the therapeutic effects achievable upon treatment with epothilone D is likely non-linearly dose dependent. Specifically, consistent dose-dependent results were repeatedly obtained in each of the behavioral and neuropathological studies reported, wherein at the lower dose (1 mpk) (about 100-fold less than the chemotherapeutic dose in tumor xenograft experiments), a significantly-enhanced beneficial effect was obtained in all measures as compared with the vehicle, while the higher dose (10 mpk), showed a trend toward effect with most measures and a statistically significant difference over vehicle in one measure of the MWM probe test.
  • ixabepilone (aza-epothilone B analog), Compound II (BMS 310705, 21-amino epothilone F), and epothilone D (Compound I) were evaluated and compared to paclitaxel after bolus IV administration into the tail veins of nude mice at dosages of 1 to 12 mpk with 3 mice/group.
  • Each of the four compounds were dosed at 5 ml/kg using 10% Cremophor, 10% ethanol, and 80% water containing 5% dextrose.
  • the plasma, brain, and liver levels of the compounds were measured at various times after a single dose using liquid chromatography with tandem mass spectrometry (LC/MS/MS) after an organic phase extraction, as reported in FIGS. 8A-8D and Table 1. Liver levels were not measured in the paclitaxel treated mice.
  • FIG. 8A shows the concentration of Compound II in the plasma, brain, and liver of mice following IV administration at 1 mpk at various times.
  • FIG. 8B shows the concentration of ixabepilone in the plasma, brain, and liver of mice following IV administration at 12 mpk at various times.
  • FIG. 8C shows the concentration of paclitaxel in the plasma and brain of mice following IV administration at 4 mpk at various times.
  • FIG. 8D shows the concentration of epothilone D (Compound I) in the plasma, brain, and liver of mice following IV administration at 5 mpk at various times.
  • Compound II and ixabepilone had modest brain levels relative to peripheral tissue as measured by the ratio of the brain-to-liver compound levels, particularly at later times after the initial distribution and clearance of plasma drug.
  • paclitaxel brain levels were low.
  • paclitaxel brain levels did not exceed plasma drug levels for at least 24 h after dosing.
  • epothilone D (Compound I) had the combined properties of remarkably better brain penetration and selective retention than the compounds tested in this experiment, as evidenced by high brain levels that exceeded liver levels at 6 and 24 h after dosing. This demonstrates unexpected retention of epothilone D (Compound I) in the target organ (brain) relative to the periphery, including the plasma and tissues, most notably the liver, which is a potential site of toxicity.
  • Table 1 reports comparative brain penetration data for four microtubule stabilizers—paclitaxel, Compound II (BMS 310705), ixabepilone, and epothilone D—after bolus IV dosing (varied mpk, as reported in the table) using nude mice, which data is also reflected in FIGS. 8A-8D .
  • the brain-to-plasma ratio generally increases with time after dosing for each compound due to the rapid loss from the plasma and retention of the drug in the brain by binding to microtubules. The brain-to-plasma ratio may then fall for compounds where there is less retention in the brain, such as is observed for Compound II showing a decrease between 6 and 24 h.
  • brain-to-plasma ratio provides a measure of the intrinsic brain penetration for a compound when data from short times after dosing (e.g., between 20-60 minutes following dosing) are compared.
  • brain-to-plasma and brain-to-liver ratios were calculated by first calculating the ratios for individual animals, and then determining the mean of the ratios; the Tables herein report the mean values thus obtained.
  • paclitaxel is poorly brain penetrant as evidenced by a brain-to-plasma ratio of 0.1 at 1 hour after dosing; ixabepilone is more brain penetrant than paclitaxel with a brain-to-plasma ratio of 0.73 at 1 hour after dosing (Table 1).
  • the brain-to-plasma ratio is a reflection of both intrinsic brain penetration and retention (half-life) in the brain.
  • the data at 6 and 24 h after dosing of epothilone D shows at least a 60-fold increase in brain-to-plasma ratio above ixabepilone, the compound with the next highest brain-to-plasma ratio in this group.
  • the brain-to-liver ratios not only provide a more singular measure of brain retention and half life, but also selective retention compared to peripheral tissues This is valuable because the liver, chosen largely because it is well perfused and tends to have higher levels than many other peripheral tissues, contains microtubules where the compound can be retained, unlike the non-cellular plasma. In contrast to the brain-to-plasma ratio where the optimal measurement time is in the 20-60 minute range, it is preferable to compare the brain-to-liver ratios at later times after dosing (e.g., 24 h or more), when the plasma levels have significantly decreased, thereby allowing a more accurate measure of the drug that is specifically retained within brain and liver cells.
  • epothilone D is highly, selectively retained in the brain relative to the liver.
  • the 24 hour brain-to-liver ratio of epothilone D is 1204, a remarkably, much higher ratio as compared with the lower ratios for ixabepilone (1.2) and Compound II (0.02) in the same set of experiments.
  • epothilone D is highly brain penetrant with substantially improved brain penetration and retention as compared with Compound II, ixabepilone, and paclitaxel.
  • the brain-to-plasma ratio of epothilone D at 6 h after dosing was 2046 in Table and 89 in Table 2, still markedly greater than the ratios at the same times for paclitaxel (0.37), and Compound II (2.1) in Table 1. Because the liver levels in the study described in Table 2 fell below the lower level of quantitation (LLQ of 49 nM in this study) at 24 hr, a brain-to-liver ratio was not quantifiable (NQ) at this time.
  • WO 03/074053 A1 broadly discloses the use of certain epothilones for the treatment of brain diseases. According to that publication, plasma and brain levels for three epothilones (not including epothilone D) were measured during the first 40 minutes following bolus IV administration at 5 mpk.
  • LC/MS analyses were carried out on a Waters instrument using a Phenomenex-Luna 3.0 ⁇ 50 mm S 10 reverse phase column employing a flow rate of 4 mL/min using a 0.1% TFA in MeOH/water gradient [0-100% in 3 min, with 4 min run time], and a UV detector set at 220 nm or Phenomenex-Luna 3.0 ⁇ 50 mm 10 u reverse phase column employing a flow rate of 5 mL/min using a 10 mM ammonium acetate acetonitrile/water gradient [5-95% in 3 min, with 4 min run time] and a UV detector set at 220 nm.
  • purifications were done on 40-63 mesh silica gel columns, or using a BIOTAGE® Horizon system, or using specified HPLC equipment and conditions.
  • Synthesis Intermediate-5 (6.8 g, 8.58 mmol) in 100 mL of ethanol at RT was added p-toluenesulfonic acid monohydrate (1.8 g, 9.44 mmol). After stirring for 6 h, saturated NaHCO 3 was added and the mixture was extracted with EtOAc. The organic layers were washed with brine, dried over Na 2 SO 4 and concentrated in vacuo. The reaction was repeated using 1 g of Synthesis Intermediate-5. The combined crude products were purified using a BIOTAGE® system (EtOAc/hexane, 10-40%) to yield Synthesis Intermediate-6 (5 g, 6.65 mmol, 68%).
  • Synthesis Intermediate-6 To a solution of Synthesis Intermediate-6 (5 g, 6.65 mmol) in 200 ml of methylene chloride at 0° was added 2,6-lutidine (7.7 mL, 66.5 mmol), followed by tert-butyldimethylsilyl trifluoromethanesulfonate (9.16 mL, 39.9 mmol). The mixture was allowed to warm to RT over 16 h, then poured into saturated NaHCO 3 and extracted with methylene chloride. The organic solvent was evaporated and the crude mixture was filtered through a layer of SiO 2 with EtOAc/hexane (10-20%) to provide Synthesis Intermediate-7 as an oil (6.9 g, 100%).
  • Synthesis Intermediate-7 To a solution of Synthesis Intermediate-7 (5 g, 5.1 mmol) in 80 mL of methylene chloride and 40 mL of MeOH at 0° was added (+/ ⁇ )-camphor-10-sulfonic acid (1.18 g, 5.1 mmol). The mixture was stirred for 6 h at 0°, poured into saturated NaHCO 3 and extracted with methylene chloride. The organic layers were washed with brine, dried over Na 2 SO 4 and concentrated to yield Synthesis Intermediate-8 as an oil (3.6 g, 4.15 mmol, 81%).
  • Synthesis Intermediate-10 As a viscous oil (2.6 g, 3.3 mmol, 100%).
  • the white solid was taken up in 5 mL of MeOH, and purified by HPLC (Varian, Dynamax PDA-2 detector; Waters C18 column; A: water with 0.05% TFA; B: acetonitrile with 0.05% TFA, isocratic). Two major peaks were collected (Peak 1, 73.4 mg, 38%; and Peak 2, 41.8 mg, 22%).
  • Peak 1 was determined to be the 13-Z (1-oxa numbering) isomer by observation of NOE between the C-14 olefinic proton and the C-13 methyl.
  • 1 H NMR 500 MHz, CDCl 3 ) ⁇ ppm 7.14 (s, 1H), 6.75 (s, 1H), 5.15-5.05 (m, 1H), 5.05-5.00 (m, 1H), 4.45-4.35 (m, 1H), 3.65-3.55 (m, 1H), 3.35-3.25 (m, 1H), 2.92 (s, 3H), 2.55-2.45 (m, 2H), 2.35-2.25 (m, 2H), 2.25-2.15 (m, 1H), 2.00 (s, 3H), 1.90-1.80 (m, 1H), 1.80-1.65 (m, 5H), 1.60-1.45 (m, 2H), 1.45-1.30 (m, 5H), 1.25-1.15 (m, 3H), 1.05-0.95 (m, 6H), 0.90-0.85
  • Peak 2 was determined to be the 13-E isomer (Compound III for Example 7 experiment, below) by absence of NOF between the C-14 olefinic proton and the C-13 methyl.
  • 1 H NMR 500 MHz, CDCl 3 ) ⁇ ppm 7.03 (s, 1H), 6.65 (s, 1H), 5.3-5.2 (m, 1H), 5.05-5.00 (m, 1H), 4.45-4.40 (m, 1H), 3.65-3.60 (m, 1H), 3.4-3.3 (m, 1H), 2.77 (s, 3H), 2.6-2.3 (m, 4H), 2.15-2.05 (m, 1H), 1.99 (s, 3H), 1.95-1.85 (m, 1H), 1.8-1.7 (m, 2H), 1.57 (s, 3H), 1.50-1.35 (m, 4H), 1.29 (s, 3H), 1.25-1.10 (m, 3H), 1.0-0.9 (m, 6H), 0.9-0.8 (t, 3H). MS (
  • mice per group (10 mpk and 35 mpk) were dosed at 10 ml/kg using 85% PEG-400, 10% TPGS, and 5.0% ethanol.
  • the plasma, brain, and liver compound levels were measured following tissue homogenization, extraction with acetonitrile, and liquid chromatography with tandem mass spectrometry (LC/MS/MS). Results from the studies are summarized in Tables 4 and 5 and also, the results of the 35 mpk study are reported in FIG. 9 .
  • Table 4 reports the concentration of epothilone D (Compound 1) and Compound III in the brain after oral administration (10 mpk) up to 24 h after dosing (for Compound III, to the extent still detectible given LLQ), and Table 5 reports and FIG. 9 plots, the concentration of epothilone D (Compound 1) and Compound III in the brain after oral administration (35 mpk) up to 5 to 24 h after dosing (again, for Compound III, to the extent detectible). (A plot was not prepared for the Table 4 data as only one brain concentration value was detectible for Compound III.) In Tables 4 and 5, below, where the values were ⁇ LLQ, the LLQ value is noted in the parenthetical.
  • the brain-to-liver ratio for oral dosing of epothilone D indicates that epothilone D is selectively retained in the brain, consistent with the data in Tables 1 and 2 after IV dosing.
  • the brain-to-liver ratio for epothilone D was 8 and 19 at 24 h after dosing at 10 mpk and 35 mpk, respectively (Tables 4 and 5). These values reflect remarkably high selective brain-to-liver retention rates for epothilone D.
  • epothilone D was calculated from multiple studies, and the results are reported in Table 6.
  • Table 6 To calculate an accurate brain half-life for a long half-life compound, measurements need to be taken for several half lives after a single dose. From the study described in Table 2, where brain concentrations were measured through 7 days after a single dose, the brain half life of epothilone D (Compound I) after IV dosing is 61 h (Table 6). The brain half life in mice after multiple routes of administration and dosages averaged 46.0+/ ⁇ 7 h (Table 6). Similarly, the brain half-life after IV dosing in rats was 31 h (Table 6).
  • FIG. 10 is provided which plots the results of a study (data reported in Table 2, above), showing brain concentration levels at time periods of up to 175 h post-dosing, following a 5 mpk bolus IV administration.
US12/429,492 2008-04-24 2009-04-24 Use of epothilone d in treating tau-associated diseases including alzheimer's disease Abandoned US20090270465A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/429,492 US20090270465A1 (en) 2008-04-24 2009-04-24 Use of epothilone d in treating tau-associated diseases including alzheimer's disease
US13/150,671 US8673949B2 (en) 2008-04-24 2011-06-01 Use of epothilone D in treating Tau-associated diseases including Alzheimer's disease
US14/162,286 US20140135367A1 (en) 2008-04-24 2014-01-23 Use of Epothilone D in Treating Tau-Associated Diseases Including Alzheimer's Disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4772908P 2008-04-24 2008-04-24
US12/429,492 US20090270465A1 (en) 2008-04-24 2009-04-24 Use of epothilone d in treating tau-associated diseases including alzheimer's disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/150,671 Continuation US8673949B2 (en) 2008-04-24 2011-06-01 Use of epothilone D in treating Tau-associated diseases including Alzheimer's disease

Publications (1)

Publication Number Publication Date
US20090270465A1 true US20090270465A1 (en) 2009-10-29

Family

ID=40887113

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/429,492 Abandoned US20090270465A1 (en) 2008-04-24 2009-04-24 Use of epothilone d in treating tau-associated diseases including alzheimer's disease
US13/150,671 Active US8673949B2 (en) 2008-04-24 2011-06-01 Use of epothilone D in treating Tau-associated diseases including Alzheimer's disease
US14/162,286 Abandoned US20140135367A1 (en) 2008-04-24 2014-01-23 Use of Epothilone D in Treating Tau-Associated Diseases Including Alzheimer's Disease

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/150,671 Active US8673949B2 (en) 2008-04-24 2011-06-01 Use of epothilone D in treating Tau-associated diseases including Alzheimer's disease
US14/162,286 Abandoned US20140135367A1 (en) 2008-04-24 2014-01-23 Use of Epothilone D in Treating Tau-Associated Diseases Including Alzheimer's Disease

Country Status (26)

Country Link
US (3) US20090270465A1 (es)
EP (1) EP2276485B1 (es)
JP (1) JP5548675B2 (es)
KR (1) KR20100137576A (es)
CN (3) CN104116736A (es)
AR (1) AR071598A1 (es)
AU (1) AU2009240538B2 (es)
BR (1) BRPI0911482A2 (es)
CA (1) CA2722371C (es)
CL (1) CL2009000990A1 (es)
CY (1) CY1115617T1 (es)
DK (1) DK2276485T3 (es)
EA (1) EA021758B1 (es)
ES (1) ES2501565T3 (es)
HK (1) HK1153152A1 (es)
HR (1) HRP20140783T1 (es)
HU (1) HUE024506T2 (es)
IL (1) IL208926A0 (es)
MX (1) MX2010011209A (es)
NZ (1) NZ588555A (es)
PL (1) PL2276485T3 (es)
PT (1) PT2276485E (es)
SI (1) SI2276485T1 (es)
TW (1) TWI472329B (es)
WO (1) WO2009132253A1 (es)
ZA (1) ZA201007460B (es)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100160372A1 (en) * 2008-11-13 2010-06-24 Link Medicine Corporation Treatment of proteinopathies using a farnesyl transferase inhibitor
US20100184803A1 (en) * 2007-03-09 2010-07-22 Link Medicine Corporation Treatment of Lysosomal Storage Diseases
US20120015999A1 (en) * 2010-06-08 2012-01-19 Vertex Pharmaceuticals Incorporated Formulations of (r)-1-(2,2-difluorobenzo[d] [1,3] dioxol-5-yl)-n-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1h-indol-5-yl)cyclopropanecarboxamide
US8232402B2 (en) 2008-03-12 2012-07-31 Link Medicine Corporation Quinolinone farnesyl transferase inhibitors for the treatment of synucleinopathies and other indications
US8343996B2 (en) 2008-11-13 2013-01-01 Astrazeneca Ab Azaquinolinone derivatives and uses thereof
US9732080B2 (en) 2006-11-03 2017-08-15 Vertex Pharmaceuticals Incorporated Azaindole derivatives as CFTR modulators
US9758510B2 (en) 2006-04-07 2017-09-12 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
EP3216456A4 (en) * 2014-11-04 2018-05-09 Well Stone Co. Tau protein production accelerator, and therapeutic or preventive agent and therapeutic or preventive food composition for diseases caused by tau protein deficiency
US9974781B2 (en) 2006-04-07 2018-05-22 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10022352B2 (en) 2006-04-07 2018-07-17 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10058546B2 (en) 2012-07-16 2018-08-28 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions of (R)-1-(2,2-difluorobenzo[D][1,3]dioxo1-5-y1)-N-(1-(2,3-dihydroxypropy1)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-y1)-1H-indol-5-y1) cyclopropanecarbox-amide and administration thereof
US10071979B2 (en) 2010-04-22 2018-09-11 Vertex Pharmaceuticals Incorporated Process of producing cycloalkylcarboxamido-indole compounds
US10081621B2 (en) 2010-03-25 2018-09-25 Vertex Pharmaceuticals Incorporated Solid forms of (R)-1(2,2-difluorobenzo[D][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide
US10206877B2 (en) 2014-04-15 2019-02-19 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
US11498974B2 (en) 2016-07-14 2022-11-15 Bioarctic Ab Brain delivery protein

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ588555A (en) 2008-04-24 2012-06-29 Bristol Myers Squibb Co Use of epothilone d in treating tau-associated diseases including alzheimer's disease
EP2811980A4 (en) * 2012-01-31 2015-12-23 Cerulean Pharma Inc POLYMER-AGENT CONJUGATES, PARTICLES, COMPOSITIONS AND METHODS OF USE THEREOF
US9834596B2 (en) 2012-07-03 2017-12-05 Washington University Antibodies to tau
TW202136296A (zh) 2014-06-27 2021-10-01 美商C2N醫療診斷有限責任公司 人類化抗-tau抗體
CN114727995A (zh) * 2020-09-02 2022-07-08 北京华昊中天生物医药股份有限公司 优替德隆的固体口服制剂
CN113005086B (zh) * 2021-02-01 2022-10-28 中国科学院遗传与发育生物学研究所 埃博霉素D和Apol8在调控神经干细胞定向神经元分化中的应用

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5580898A (en) * 1994-05-24 1996-12-03 The Trustees Of The University Of Pennsylvania Method of stabilizing microtubules
US6242469B1 (en) * 1996-12-03 2001-06-05 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6262094B1 (en) * 1999-02-22 2001-07-17 Bristol-Myers Squibb Company C-21 modified epothilones
US6380395B1 (en) * 1998-04-21 2002-04-30 Bristol-Myers Squibb Company 12, 13-cyclopropane epothilone derivatives
US6399838B1 (en) * 2000-11-17 2002-06-04 National Science Council Process for preparing 2,6-dimethylphenol
US6498257B1 (en) * 1998-04-21 2002-12-24 Bristol-Myers Squibb Company 2,3-olefinic epothilone derivatives
US6605599B1 (en) * 1997-07-08 2003-08-12 Bristol-Myers Squibb Company Epothilone derivatives
US20030186965A1 (en) * 2002-03-12 2003-10-02 Vite Gregory D. C12-cyano epothilone derivatives
US20040019088A1 (en) * 2002-03-01 2004-01-29 Schering Ag Use of Epothilones in the treatment of brain diseases associated with proliferative processes
US6719540B2 (en) * 2002-03-12 2004-04-13 Bristol-Myers Squibb Company C3-cyano epothilone derivatives
US6800653B2 (en) * 2001-06-01 2004-10-05 Bristol-Myers Squibb Compnay Epothilone derivatives
US20080207697A1 (en) * 2004-01-30 2008-08-28 Institu National De La Sante Et De La Recherche Medicale Use of Epothilones in the Treatment of Neuronal Connectivity Defects Such as Schizophrenia and Autism

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2809918A (en) 1955-10-17 1957-10-15 Victor M Hermelin Sustained release pharmaceutical preparations
DE2010416B2 (de) 1970-03-05 1979-03-29 Hoechst Ag, 6000 Frankfurt Oral anwendbare Arzneiform mit Retardwirkung
US4728512A (en) 1985-05-06 1988-03-01 American Home Products Corporation Formulations providing three distinct releases
US4794001A (en) 1986-03-04 1988-12-27 American Home Products Corporation Formulations providing three distinct releases
US5225202A (en) 1991-09-30 1993-07-06 E. R. Squibb & Sons, Inc. Enteric coated pharmaceutical compositions
DE59609305D1 (de) 1995-11-17 2002-07-11 Biotechnolog Forschung Gmbh Epothilon-Derivate und deren Herstellung
US6204388B1 (en) 1996-12-03 2001-03-20 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
KR100574454B1 (ko) 1997-12-04 2006-04-27 브리스톨-마이어즈 스퀴브 컴페니 옥시라닐 에포틸론을 환원시켜 올레핀계 에포틸론을 제조하는 방법
US6320045B1 (en) * 1997-12-04 2001-11-20 Bristol-Myers Squibb Company Process for the reduction of oxiranyl epothilones to olefinic epothilones
CA2322157C (en) 1998-02-25 2012-05-29 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
UA69413C2 (uk) 1998-05-22 2004-09-15 Брістол-Майерс Сквібб Компані Фармацевтична композиція, яка містить серцевину та ентеросолюбільну оболонку, фармацевтична композиція у вигляді сфероїдальних гранул, спосіб одержання сфероїдальних гранул та спосіб одержання фармацевтичної композиції
ATE415673T1 (de) 1998-07-27 2008-12-15 Infineon Technologies Ag Verfahren zur prüfung der echtheit von urkunden
US6410301B1 (en) 1998-11-20 2002-06-25 Kosan Biosciences, Inc. Myxococcus host cells for the production of epothilones
CA2350189A1 (en) 1998-11-20 2000-06-02 Kosan Biosciences, Inc. Recombinant methods and materials for producing epothilone and epothilone derivatives
US8618085B2 (en) * 2000-04-28 2013-12-31 Koasn Biosciences Incorporated Therapeutic formulations of desoxyepothilones
US6998256B2 (en) 2000-04-28 2006-02-14 Kosan Biosciences, Inc. Methods of obtaining epothilone D using crystallization and /or by the culture of cells in the presence of methyl oleate
JP2004516011A (ja) 2000-07-25 2004-06-03 コーサン バイオサイエンシーズ, インコーポレイテッド エポチロンのための発酵プロセス
JP3604337B2 (ja) * 2000-10-03 2004-12-22 古河電気工業株式会社 絶縁電線の製造方法
ATE349412T1 (de) 2000-10-16 2007-01-15 R & D Biopharmaceuticals Epothilon-synthesebausteine i: unsymmetrisch substituierte acyloine und acyloinderivate, verfahren zu deren herstellung sowie deren verwendung zur herstellung von epothilonen und epothilonderivaten
FR2817117B1 (fr) 2000-11-24 2005-11-04 Commissariat Energie Atomique Mammiferes non-humains transgeniques ou recombinants et leurs applications dans le criblage de medicaments utiles dans les desordres psychoactifs
WO2002058699A1 (en) 2001-01-25 2002-08-01 Bristol-Myers Squibb Company Pharmaceutical forms of epothilones for oral administration
US6660767B2 (en) * 2001-02-02 2003-12-09 The Regents Of The University Of California Coumarin compounds as microtubule stabilizing agents and therapeutic uses thereof
KR20040088531A (ko) 2002-02-25 2004-10-16 코산 바이오사이언시즈, 인코포레이티드 2차 대사 동종체 분포 조절
US7082674B2 (en) * 2002-02-27 2006-08-01 Sumitomo Heavy Industries, Ltd. Method for winding a single coil of a coil unit for a linear motor
CA2491943C (en) 2002-07-15 2012-06-19 Gerhard Hoefle Epothilone derivatives for the treatment of cancer
AU2003260723A1 (en) 2002-08-17 2004-03-03 The Queens Universlty Of Belfast Use of vinca alkaloyds, taxane, cryptophycine, ephitoline or eleutherobine for treating alzheimer
ES2439995T3 (es) 2002-09-23 2014-01-27 Bristol-Myers Squibb Company Procedimientos de preparación, aislamiento y purificación de epotilona B
EP1608626A1 (en) * 2003-03-28 2005-12-28 Cornell Research Foundation, Inc. Migrastatin analog compositions and uses thereof
US6954131B2 (en) * 2003-04-02 2005-10-11 Illinois Tool Works Inc. Electrical reactor assembly having center taps
MXPA06003835A (es) * 2003-10-09 2006-06-14 Kosan Biosciences Inc Formulaciones terapeuticas.
WO2006009178A1 (ja) 2004-07-20 2006-01-26 Toshinori Kato 生体機能診断装置、生体機能診断方法、生体用プローブ、生体用プローブ装着具、生体用プローブ支持具及び生体用プローブ装着支援具
US20060142393A1 (en) * 2004-09-16 2006-06-29 Sherman Matthew L Bis(thio-hydrazide amides) for treatment of hyperplasia
AU2007288338B2 (en) * 2006-08-21 2012-05-03 Synta Pharmaceuticals Corp. Compounds for treating proliferative disorders
JP2010501563A (ja) * 2006-08-21 2010-01-21 シンタ ファーマシューティカルズ コーポレーション 増殖性障害を治療するための化合物
EP2061451A2 (en) * 2006-08-21 2009-05-27 Synta Pharmaceuticals Corporation Bis(thiohydrazide amides) for treating melanoma
NZ588555A (en) 2008-04-24 2012-06-29 Bristol Myers Squibb Co Use of epothilone d in treating tau-associated diseases including alzheimer's disease

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5580898A (en) * 1994-05-24 1996-12-03 The Trustees Of The University Of Pennsylvania Method of stabilizing microtubules
US6242469B1 (en) * 1996-12-03 2001-06-05 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6605599B1 (en) * 1997-07-08 2003-08-12 Bristol-Myers Squibb Company Epothilone derivatives
US6380395B1 (en) * 1998-04-21 2002-04-30 Bristol-Myers Squibb Company 12, 13-cyclopropane epothilone derivatives
US6498257B1 (en) * 1998-04-21 2002-12-24 Bristol-Myers Squibb Company 2,3-olefinic epothilone derivatives
US6831090B2 (en) * 1998-04-21 2004-12-14 Bristol-Myers Squibb Company 2,3-olefinic epothilone derivatives
US6262094B1 (en) * 1999-02-22 2001-07-17 Bristol-Myers Squibb Company C-21 modified epothilones
US6399838B1 (en) * 2000-11-17 2002-06-04 National Science Council Process for preparing 2,6-dimethylphenol
US6800653B2 (en) * 2001-06-01 2004-10-05 Bristol-Myers Squibb Compnay Epothilone derivatives
US20040019088A1 (en) * 2002-03-01 2004-01-29 Schering Ag Use of Epothilones in the treatment of brain diseases associated with proliferative processes
US20030186965A1 (en) * 2002-03-12 2003-10-02 Vite Gregory D. C12-cyano epothilone derivatives
US6719540B2 (en) * 2002-03-12 2004-04-13 Bristol-Myers Squibb Company C3-cyano epothilone derivatives
US20080207697A1 (en) * 2004-01-30 2008-08-28 Institu National De La Sante Et De La Recherche Medicale Use of Epothilones in the Treatment of Neuronal Connectivity Defects Such as Schizophrenia and Autism

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10239867B2 (en) 2006-04-07 2019-03-26 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US11639347B2 (en) 2006-04-07 2023-05-02 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10987348B2 (en) 2006-04-07 2021-04-27 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US9758510B2 (en) 2006-04-07 2017-09-12 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10975061B2 (en) 2006-04-07 2021-04-13 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US9974781B2 (en) 2006-04-07 2018-05-22 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10022352B2 (en) 2006-04-07 2018-07-17 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US9732080B2 (en) 2006-11-03 2017-08-15 Vertex Pharmaceuticals Incorporated Azaindole derivatives as CFTR modulators
US20100184803A1 (en) * 2007-03-09 2010-07-22 Link Medicine Corporation Treatment of Lysosomal Storage Diseases
US8232402B2 (en) 2008-03-12 2012-07-31 Link Medicine Corporation Quinolinone farnesyl transferase inhibitors for the treatment of synucleinopathies and other indications
US8343996B2 (en) 2008-11-13 2013-01-01 Astrazeneca Ab Azaquinolinone derivatives and uses thereof
US20100160372A1 (en) * 2008-11-13 2010-06-24 Link Medicine Corporation Treatment of proteinopathies using a farnesyl transferase inhibitor
US10906891B2 (en) 2010-03-25 2021-02-02 Vertex Pharmaceuticals Incoporated Solid forms of (R)-1(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide
US11578062B2 (en) 2010-03-25 2023-02-14 Vertex Pharmaceuticals Incorporated Solid forms of (R)-1(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide
US10081621B2 (en) 2010-03-25 2018-09-25 Vertex Pharmaceuticals Incorporated Solid forms of (R)-1(2,2-difluorobenzo[D][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide
US10071979B2 (en) 2010-04-22 2018-09-11 Vertex Pharmaceuticals Incorporated Process of producing cycloalkylcarboxamido-indole compounds
US20120015999A1 (en) * 2010-06-08 2012-01-19 Vertex Pharmaceuticals Incorporated Formulations of (r)-1-(2,2-difluorobenzo[d] [1,3] dioxol-5-yl)-n-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1h-indol-5-yl)cyclopropanecarboxamide
US8563593B2 (en) * 2010-06-08 2013-10-22 Vertex Pharmaceuticals Incorporated Formulations of (R)-1-(2,2-difluorobenzo[D] [1,3] dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl)cyclopropanecarboxamide
US10058546B2 (en) 2012-07-16 2018-08-28 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions of (R)-1-(2,2-difluorobenzo[D][1,3]dioxo1-5-y1)-N-(1-(2,3-dihydroxypropy1)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-y1)-1H-indol-5-y1) cyclopropanecarbox-amide and administration thereof
US10206877B2 (en) 2014-04-15 2019-02-19 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
US11951212B2 (en) 2014-04-15 2024-04-09 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
US10980746B2 (en) 2014-04-15 2021-04-20 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
EP3216456A4 (en) * 2014-11-04 2018-05-09 Well Stone Co. Tau protein production accelerator, and therapeutic or preventive agent and therapeutic or preventive food composition for diseases caused by tau protein deficiency
US10576108B2 (en) 2014-11-04 2020-03-03 Well Stone Co. Tau protein production accelerator, and therapeutic or preventive agent and therapeutic or preventive food composition for diseases caused by tau protein deficiency
US10398740B2 (en) 2014-11-04 2019-09-03 Well Stone Co. Tau protein production accelerator, and therapeutic or preventive agent and therapeutic or preventive food composition for diseases caused by tau protein deficiency
US11498974B2 (en) 2016-07-14 2022-11-15 Bioarctic Ab Brain delivery protein

Also Published As

Publication number Publication date
AU2009240538A1 (en) 2009-10-29
EA201001695A1 (ru) 2011-08-30
TW200948360A (en) 2009-12-01
US20110230528A1 (en) 2011-09-22
CN104116736A (zh) 2014-10-29
PL2276485T3 (pl) 2014-12-31
JP5548675B2 (ja) 2014-07-16
BRPI0911482A2 (pt) 2017-08-29
MX2010011209A (es) 2010-11-12
KR20100137576A (ko) 2010-12-30
CA2722371A1 (en) 2009-10-29
AU2009240538B2 (en) 2014-08-07
DK2276485T3 (da) 2014-10-13
US20140135367A1 (en) 2014-05-15
WO2009132253A1 (en) 2009-10-29
AR071598A1 (es) 2010-06-30
EA021758B1 (ru) 2015-08-31
SI2276485T1 (sl) 2014-10-30
JP2011522782A (ja) 2011-08-04
HK1153152A1 (en) 2012-03-23
PT2276485E (pt) 2014-10-20
HRP20140783T1 (hr) 2014-09-12
NZ588555A (en) 2012-06-29
TWI472329B (zh) 2015-02-11
IL208926A0 (en) 2011-01-31
ZA201007460B (en) 2012-03-28
EP2276485B1 (en) 2014-07-16
CL2009000990A1 (es) 2010-10-15
CA2722371C (en) 2016-06-21
EP2276485A1 (en) 2011-01-26
CN102076339A (zh) 2011-05-25
HUE024506T2 (en) 2016-01-28
CY1115617T1 (el) 2017-01-04
CN104666297A (zh) 2015-06-03
US8673949B2 (en) 2014-03-18
ES2501565T3 (es) 2014-10-02

Similar Documents

Publication Publication Date Title
US8673949B2 (en) Use of epothilone D in treating Tau-associated diseases including Alzheimer's disease
US20050026811A1 (en) Rage antagonists as agents to reverse amyloidosis and diseases associated therewith
US20200237849A1 (en) Compositions and methods for treating beta-amyloid related diseases
KR20190099497A (ko) 카스파제-1 억제 및 신경학적 병태의 예방 및 치료를 위한 이의 용도
WO2023066216A1 (zh) 一种包含c21甾体皂苷的药物组合物及其应用
EP2462131B1 (en) Compositions and methods for treating beta-amyloid related diseases
US20120214836A1 (en) Sensitizer, pharmaceutical composition, kit and use for target therapy
JP2019534885A (ja) 脳アミロイド血管症の治療又は予防における使用のためのオキサジン誘導体
US20120245223A1 (en) Use of gymnodimine, analogues and derivatives for the treatment and/or prevention of neurodegenerative diseases associated with tau and b-amyloid
Ballatore et al. Microtubule stabilization
WO2023146818A2 (en) Methods of treating amyloid related brain disorders using novel compounds and antibodies
WO2011047374A1 (en) Methods of treating tauopathies
Seneci Targets and Small Molecules Against Tauopathies. Part 1: From Genes to Soluble, Aggregation-Prone Tau Proteins
US20170202792A1 (en) Method for treating tau-associated diseases
CN116253666A (zh) 中枢神经系统疾病的新疗法

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ALBRIGHT, CHARLES F.;BARTEN, DONNA MARIE;LEE, FRANCIS Y.;REEL/FRAME:022810/0134;SIGNING DATES FROM 20090518 TO 20090603

STCB Information on status: application discontinuation

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