WO2013096870A1 - Compositions et procédés pour traiter la sclérose latérale amyotrophique - Google Patents

Compositions et procédés pour traiter la sclérose latérale amyotrophique Download PDF

Info

Publication number
WO2013096870A1
WO2013096870A1 PCT/US2012/071426 US2012071426W WO2013096870A1 WO 2013096870 A1 WO2013096870 A1 WO 2013096870A1 US 2012071426 W US2012071426 W US 2012071426W WO 2013096870 A1 WO2013096870 A1 WO 2013096870A1
Authority
WO
WIPO (PCT)
Prior art keywords
dose
administering
dexpramipexole
subjects
als
Prior art date
Application number
PCT/US2012/071426
Other languages
English (en)
Inventor
Michael E. Bozik
Thomas Petzinger
Evan W. INGERSOLL
Original Assignee
Knopp Neurosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Knopp Neurosciences Inc filed Critical Knopp Neurosciences Inc
Publication of WO2013096870A1 publication Critical patent/WO2013096870A1/fr

Links

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
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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

Definitions

  • Various embodiments described herein are directed to a method for treating amyotrophic lateral sclerosis (ALS) in a patient including the step of administering to the patient an effective amount of about chirally pure (6R)-2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole or a
  • treating can include slowing progression of amyotrophic lateral sclerosis (ALS), reducing intensity of symptoms associated with amyotrophic lateral sclerosis (ALS), reducing onset of symptoms associated with amyotrophic lateral sclerosis (ALS), reducing weight loss associated with amyotrophic lateral sclerosis (ALS), reversing weight loss associated with amyotrophic lateral sclerosis (ALS), delaying mortality, and combinations thereof.
  • ALS amyotrophic lateral sclerosis
  • ALS amyotrophic lateral sclerosis
  • ALS amyotrophic lateral sclerosis
  • ALS amyotrophic lateral sclerosis
  • ALS amyotrophic lateral sclerosis
  • the symptoms associated with amyotrophic lateral sclerosis may be, for example, fine motor function, gross motor function, bulbar function, respiratory function, and combinations thereof, and in other embodiments, the symptoms associated with amyotrophic lateral sclerosis (ALS) can include walking, speech, eating, swallowing, writing, climbing stairs, cutting food, turning in bed, salivation, dressing, maintaining hygiene, breathing, dyspnea, orthopnea, respiratory insufficiency, and combinations thereof.
  • the effective amount may be from about 50 mg to about 300 mg per day, and in other embodiments, the effective amount may be from about 150 mg to about 300 mg per day. In still other embodiments, the effective amount may be about 300 mg or more per day. In still other embodiments, the effective amount may be about 600 mg or more per day. In certain embodiments, the effective amount may be a stable daily dose. In some embodiments, the stable daily dose may be from about 50 mg to about 300 mg of about chirally pure (6R)-2-amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole or a pharmaceutically acceptable salt thereof.
  • the stable daily dose may be from about 50 mg to about 600 mg of about chirally pure (6R)-2-amino- 4,5,6,7-tetrahydro-6-(propylamino)benzothiazole or a pharmaceutically acceptable salt thereof.
  • the stable daily dose may be 1 to 5 unit doses per day, and in particular embodiments, each unit dose may be a solid unit dose.
  • administering may include
  • administering may include administering one unit dose two times per day wherein each unit dose is equal to about half of the stable daily dose, and in other embodiments, administering may include administering one unit dose once every 12 hours wherein each unit dose is equal to about half of the stable daily dose. In still other embodiments, administering may include administering one unit dose four times per day wherein each unit dose is equal to about one quarter of the stable daily dose. In some embodiments, administering can include administering three unit doses wherein each unit dose is about 150 mg once per day, or one unit dose is about 150 mg, wherein each dose is administered three times per day, and in further embodiments, administering may include administering one unit dose wherein each unit dose is about 450 mg once per day.
  • administering can include administering two unit doses wherein each unit dose is about 150 mg two times per day, in further embodiments, administering may include administering four unit doses wherein each unit dose is about 75 mg four times per day, in further embodiments, administering may include administering two unit doses wherein each unit dose is about 150 mg two times per day, in further embodiments, administering may include administering one unit dose wherein the unit dose is about 300 mg administered two times per day, and in further embodiments, administering may include administering one unit dose wherein each unit dose is about 600 mg once per day.
  • the method may further include the step of monitoring the patient, and in particular embodiments, the method may include the step of monitoring the patient for neutropenia. In other embodiments, monitoring may be measuring the ALS Functional Rating Scale Revised (ALSFRS-R) total score for the patient or monitoring the patients fine motor function, gross motor function, bulbar function, respiratory function, and combinations thereof. In still other embodiments, the method may include monitoring behaviors selected from the group consisting of swallowing, handwriting, speech, ability to walk, ability to climb stairs, ability to dress, ability to maintain hygiene, and combinations thereof. In some embodiments, the method may include scheduling a doctor visit every 6 months for at least 12 months.
  • ALSFRS-R ALS Functional Rating Scale Revised
  • the patient may be predisposed to amyotrophic lateral sclerosis (ALS) and is not exhibiting symptoms of amyotrophic lateral sclerosis (ALS).
  • the method may include administering about chirally pure (6R)-2-amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole or a pharmaceutically acceptable salt thereof to one or more family members of the patient.
  • the method may include administering the about chirally pure (6R)-2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole or a pharmaceutically acceptable salt thereof to a patient not exhibiting symptoms of amyotrophic lateral sclerosis (ALS), and in further embodiments, the method may include administering the about chirally pure (6R)-2-amino-4, 5,6,7- tetrahydro-6-(propylamino)benzothiazole or a pharmaceutically acceptable salt thereof to a patient that is predisposed to amyotrophic lateral sclerosis (ALS).
  • ALS amyotrophic lateral sclerosis
  • FIG. 1 is a bar graph showing the mean change from baseline in ALSFRS-R scores by subdomain for the placebo and 300 mg/day group over 12 weeks of treatment.
  • FIG. 2 shows the change from baseline in for vital capacity (VC) for the treatment groups with dotted line showing a defined threshold of treatment failure (greater than or equal to 20% decrease in VC from baseline).
  • FIG. 3 shows the change from baseline in ALSFRS-R for treatment groups after 4 weeks (left panel) and 12 weeks (right panel) of treatment with a dotted line showing a defined threshold for clinical treatment failure (greater than or equal to 6 point decline in ALSFRS-R score).
  • FIG. 4A-C show plots of the mean change in ALSFRS-R score over time and bar graphs of the mean change in baseline based on individual fine motor behaviors, handwriting (FIG. 4A), cutting food (FIG. 4B), and dressing and hygiene (FIG. 4C), tested in ALSFRS-R.
  • FIG. 5A-C show plots of the mean change in ALSFRS-R score over time and bar graphs of the mean change in baseline based on individual bulbar domain functions, swallowing (FIG. 5A), speech (FIG. 5B), and salivation (FIG. 5C), tested in ALSFRS-R.
  • FIG. 6A-C show plots of the mean change in ALSFRS-R score over time and bar graphs of the mean change in baseline based on individual gross motor behaviors, turning in bed (FIG. 6A), walking (FIG. 6B), and climbing stairs (FIG. 6C), tested in ALSFRS-R.
  • FIG. 7A-C show plots of the mean change in ALSFRS-R score over time and bar graphs of the mean change in baseline based on individual respiratory functions, dyspnea (FIG. 7A), orthopnea (FIG. 7B), and respiratory insufficiency (FIG. 8C), tested in ALSFRS-R.
  • FIG. 8 shows bar graphs illustrating the change from baseline in ALSFRS-R scores by question for the placebo and 300 mg groups in Part 1 and for the 50 mg and 300 mg groups in Part 2.
  • FIG. 9 shows box plots of change from baseline to Week 4, Week 8, and End of Part 1 for each treatment group.
  • FIG. 10 shows the change in ALSFRS-R from baseline to end for each treatment group.
  • FIG. 11 is a bar graph showing the change from baseline in ALSFRS-R for placebo and the 300 mg treatment group.
  • FIG. 12 is a schematic of Part 1 and Part 2 of the study.
  • FIG. 13 shows Kaplan-Meier Estimates for Time to Tracheostomy or Death— Double-Blind Treatment Period (Safety Population).
  • FIG. 14 show a plot of Mean (SE) ALSFRS-R Total Scores Estimated from the Linear Mixed- Effects Model for Slope (horizontal axis is weeks of active treatment starting at the Part 2, Week 4 visit).
  • FIG. 15 shows a graphic presentation of Kaplan-Meier Estimates for Time to Death (Double- Blind Treatment Period through Week 28).
  • FIG. 16 shows a Plot of Mean (SE) Rank of Joint Scores for Combined Time to Death and Changes from Baseline in ALSFRS-R Total Scores (double-blind treatment period through Week 28).
  • SE Mean
  • FIG. 17 shows a plot of Mean (SE) ALSFRS-R Total Score Estimates from the Linear Mixed Effects Model for Slope Including Imputed Values of Zero for the First Post-death Visit among Subjects who Died (Double-Blind Treatment Period through Week 28).
  • SE Mean
  • ALSFRS-R Total Score Estimates from the Linear Mixed Effects Model for Slope Including Imputed Values of Zero for the First Post-death Visit among Subjects who Died (Double-Blind Treatment Period through Week 28).
  • FIG. 18 shows a plot of Mean (SE) from Linear Mixed Effects Model Estimates for the Slope of Upright Vital Capacity (with imputed zeroes for the first post-death visit among subjects who died — time from first dose in double-blind treatment period through Week 28).
  • SE Mean
  • FIG. 19 shows Kaplan-Meier estimates for time to feeding tube placement— double-blind treatment period (safety population).
  • FIG. 20 shows the mean plasma dexpramipexole concentration in healthy volunteers after oral administration.
  • FIG. 21 shows the mean plasma dexpramipexole concentration in healthy volunteers after oral administration of a single 150 mg dose under fasted and fed conditions- linear axis.
  • FIG. 22 shows the mean plasma dexpramipexole concentration in healthy volunteers after oral administration of single 50 mg, 150 mg, and 300 mg doses on day 1, twice daily doses on day 3 through 6 and single doses on day 7 under fasted conditions- linear axis.
  • FIG. 23 shows the mean positional changes in systolic and diastolic blood pressures (standing minus supine) following 4 1/2 days of multiple doses of dexpramipexole or placebo in healthy volunteers.
  • the term "about” means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45%-55%.
  • administering a composition may be accomplished by oral administration, injection, infusion, absorption or by any method in combination with other known techniques.
  • administering may include the act of self administration of administration by another person such as a health care provider or a device.
  • improves is used to convey that the present invention changes either the appearance, form, characteristics and/or physical attributes of the tissue to which it is being provided, applied or administered.
  • Improves may also refer to the overall physical state of an individual to whom an active agent has been administered. For example, the overall physical state of an individual may "improve” if one or more symptoms of a neurodegenerative disorder are alleviated by
  • terapéutica means an agent utilized to treat, combat, ameliorate or prevent an unwanted condition or disease of a patient.
  • terapéuticaally effective amount or “therapeutic dose” as used herein are interchangeable and may refer to the amount of an active agent or pharmaceutical compound or composition that elicits a biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • a biological or medicinal response may include, for example, one or more of the following: (1) preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display pathology or symptoms of the disease, condition or disorder, (2) inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptoms of the disease, condition or disorder or arresting further development of the pathology and/or symptoms of the disease, condition or disorder, and (3) ameliorating a disease, condition or disorder in an individual that is experiencing or exhibiting the pathology or symptoms of the disease, condition or disorder or reversing the pathology and/or symptoms experienced or exhibited by the individual.
  • neuroprotectant refers to any agent that may prevent, ameliorate or slow the progression of neuronal degeneration and/or neuronal cell death.
  • treating may be taken to mean prophylaxis of a specific disorder, disease or condition, alleviation of the symptoms associated with a specific disorder, disease or condition and/or prevention of the symptoms associated with a specific disorder, disease or condition.
  • the term refers to slowing the progression of the disorder, disease or condition or alleviating the symptoms associated with the specific disorder, disease or condition.
  • the term refers to slowing the progression of the disorder, disease or condition.
  • the term refers to alleviating the symptoms associated with the specific disorder, disease or condition.
  • the term refers to restoring function which was impaired or lost due to a specific disorder, disease or condition.
  • patient generally refers to any living organism to which compounds described herein are administered and may include, but is not limited to, any nonhuman mammal, primate or human. Such “patients” may or my not be exhibiting the signs, symptoms or pathology of the particular diseased state.
  • the term "naive patient” refers to a patient that has not previously received pramipexole treatment (either (R)-pramipexole or (S)-pramipexole), particularly, (R)-pramipexole, or who has not received a titration regimen of pramipexole previous to receiving a starting dose of pramipexole.
  • enantiomers As used herein, the terms “enantiomers,” “stereoisomers,” and “optical isomers” may be used interchangeably and refer to molecules which contain an asymmetric or chiral center and are mirror images of one another. Further, the terms “enantiomers,” “stereoisomers,” or “optical isomers” describe a molecule which, in a given configuration, cannot be superimposed on its mirror image.
  • the terms “chirally pure”, “optically pure”, or “enantiomerically pure” may be taken to indicate that a composition contains at least 99.95% of a single optical isomer of a compound.
  • the term “enantiomerically enriched” may be taken to indicate that at least 51% of a composition is a single optical isomer or enantiomer.
  • the term “enantiomeric enrichment” as used herein refers to an increase in the amount of one enantiomer as compared to the other.
  • a “racemic” mixture is a mixture of about equal amounts of (6R) and (6S) enantiomers of a chiral molecule.
  • pramipexole will refer to (6S) enantiomer of 2-amino- 4,5,6,7-tetrahydro-6-(propylamino)benzothiazole unless otherwise specified.
  • composition shall mean a composition including at least one active ingredient. Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan.
  • a pharmaceutical composition may, for example, contain dexpramipexole or a pharmaceutically acceptable salt of dexpramipexole as the active ingredient.
  • a “salt” is any acid addition salt, preferably a
  • halogenic acid salts such as hydrobromic, hydrochloric, hydrofluoric and hydroiodic acid salt
  • an inorganic acid salt such as, for example, nitric, perchloric, sulfuric and phosphoric acid salt
  • an organic acid salt such as, for example, sulfonic acid salts (methanesulfonic, trifluoromethan sulfonic, ethanesulfonic, benzenesulfonic or p- toluenesulfonic), acetic, malic, fumaric, succinic, citric, benzoic, gluconic, lactic, mandelic, mucic, pamoic, pantothenic, oxalic and maleic acid salts; and an amino acid salt such as aspartic or glutamic acid salt.
  • halogenic acid salts such as hydrobromic, hydrochloric, hydrofluoric and hydroiodic acid salt
  • an inorganic acid salt such as, for example, nitric
  • the acid addition salt may be a mono- or di-acid addition salt, such as a di-hydrohalogenic, di-sulfuric, di-phosphoric or di-organic acid salt.
  • the acid addition salt is used as an achiral reagent which is not selected on the basis of any expected or known preference for interaction with or precipitation of a specific optical isomer of the products of this disclosure.
  • “Pharmaceutically acceptable salt” is meant to indicate those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a patient without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. (1977) J. Pharm. Sciences, Vol. 6. 1-19, describes pharmaceutically acceptable salts in detail.
  • the term "daily dose amount” refers to the amount of dexpramipexole or a pharmaceutically acceptable salt thereof per day or per 24 hours that is administered or prescribed to a patient. This amount can be administered in multiple unit doses or in a single unit dose, at a single time during the day or at multiple times during the day.
  • a “dose amount” or “dose” as used herein, is generally equal to the dosage of the active ingredient which may be administered per day.
  • a dose amount of dexpramipexole may be 150 mg/day, 300 mg/day, 450 mg/day or 600 mg/day.
  • unit dose may be taken to indicate a discrete therapeutic composition that contains a predetermined amount of the active compound.
  • the amount of the active compound is generally equal to the dosage of the active ingredient which may be administered one or more times per day.
  • the unit dose may contain a fraction of the desired daily dose, such as, for example, one-half or one-third the dosage.
  • a 600 mg/day dose amount of dexpramipexole may be administered as 1 unit dose of 600 mg each, 2 unit doses of 300 mg or 4 unit doses of 150 mg.
  • DAE dopaminergic activity equivalent
  • DAE corresponds to a DAE of below 0.05.
  • a dose amount of dexpramipexole having a DAE of 0.01 would, therefore, be below the DAE for the most preferable pramipexole NOAEL dose amount of 0.05 mg.
  • DAE is determined by measuring the binding affinity (IC50) or activity (EC50) at the D2 and/or D3 receptors relative to the same parameter for 1 mg of pramipexole.
  • the degree to which dosing of a molecule has demonstrable phenotypic activity resulting from affinity to particular receptors or other pharmaco-effective proteins, even when the activity results from affinities to unknown targets, can be operationally defined in terms of whether this activity contributes in a positive way ("on-target” activity) or a negative way (“off-target” activity) to a specific and desired therapeutic effect.
  • on-target activity a positive way
  • off-target activity a negative way
  • an index of activity can be generated for each of these categories (the "activity equivalent”, or "AE"), and one or more ratios generated to compare "off-target” to "on-target” activities, useful to compare potential risk- benefit ratios between molecules.
  • Dexpramipexole (6R)-2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole) is a synthetic aminobenzothiazole derivative.
  • the (6S) enantiomer of dexpramipexole commonly known as pramipexole and commercially available under the Mirapex® name, is a potent dopamine agonist, which mimics the effects of the neurotransmitter dopamine.
  • Pramipexole has also been shown to have both neuroprotective and dopaminergic activities, presumably through inhibition of lipid peroxidation, normalization of mitochondrial metabolism and/or detoxification of oxygen radicals.
  • pramipexole may have utility as an inhibitor of the cell death cascades and loss of cell viability observed in neurodegenerative diseases such as Parkinson's disease. Additionally, oxidative stress caused by an increase in oxygen and other free radicals has been associated with the fatal neurodegenerative disorder amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder involving the motor neurons of the cortex, brain stem, and spinal cord.
  • ALS amyotrophic lateral sclerosis
  • the neuroprotectant activity of both enantiomers are expected to require therapeutic doses in the range of about 10 mg/day to about 1,500 mg/day while pramipexole's agonistic effect on the D2 family of dopamine receptors only allows therapeutic doses that range between 0.5 and 5.0 mg/day.
  • significant adverse side effects have been reported.
  • the Boehringer Ingelheim product insert for Mirapex® sets the maximally tolerated dose for humans at 4.5 mg/day, and a dose of pramipexole as low as 1.5 mg has been shown to cause somnolence in humans.
  • Single dose toxicity of pramipexole after oral administration has been studied in rodents, dogs, monkeys and humans.
  • pramipexole as a mitochondria-targeted neuroprotectant is unlikely, as the high doses needed for the neuroprotective or anti -oxidative/mitochondrial normalization action are not accessible due to high dopamine receptor affinity associated with the (6S) enantiomer.
  • (6R)-2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (“dexpramipexole”) is an effective mitochondria-targeted agent that exhibits excellent neuroprotective properties when administered without adverse side effects.
  • the functional affinity difference between the pramipexole and dexpramipexole e.g.
  • dexpramipexole 10,000-20,000 fold) for dopamine receptor is much greater than previously reported.
  • higher doses of dexpramipexole can be tolerated by patients and will allow greater brain, spinal cord and mitochondrial concentrations increasing the degree to which oxidative stress and/or mitochondrial dysfunction may be reduced.
  • the neuroprotective effect of dexpramipexole may occur by at least one of three mechanisms.
  • dexpramipexole may be capable of reducing the formation of reactive oxygen species in cells with impaired mitochondrial energy production.
  • dexpramipexole may partially restore the reduced mitochondrial membrane potential that is correlated with Alzheimer's, Parkinson's, Huntington's and amyotrophic lateral sclerosis diseases.
  • dexpramipexole may block or attenuate the apoptotic cell death pathways which are produced by pharmacological models of Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis diseases and mitochondrial impairment.
  • High doses of dexpramipexole required to elicit these neuroprotective effects generally require highly pure preparations of dexpramipexole which take into account the upper limit of (6S) enantiomer contamination (0.5 mg to 5.0 mg).
  • Embodiments of the invention are generally directed to pharmaceutical compositions including an effective amount of dexpramipexole and methods for using such pharmaceutical compositions for the treatment of neurological diseases such as, for example, amyotrophic lateral sclerosis (ALS).
  • ALS amyotrophic lateral sclerosis
  • embodiments of the invention are directed to methods for treating neurological diseases including the step of administering at least about 150 mg of dexpramipexole per day to a patient in need of treatment, or, at least about 300 mg of dexpramipexole, or at least about 600 mg of dexpramipexole may be administered to a patient in need of treatment per day.
  • embodiments of the invention are also directed to pharmaceutical compositions at least including 50 mg of dexpramipexole and a pharmaceutically acceptable excipient, and in some embodiments, such pharmaceutical compositions may include at least 75 mg, 100 mg, 125 mg, 150 mg, 300 mg, 400 mg, 450 mg, 500 mg, or at least 600 mg of
  • ALS may be limb-onset ALS or bulbar-onset ALS.
  • dexpramipexole administered or incorporated into the pharmaceutical compositions may be enantiomerically pure or enantiomerically enriched to such an extent that the effects of any dopaminergic activity associated with residual (6S)-2- amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole (pramipexole) is either absent or sufficiently small to allow for high dosage administration of dexpramipexole relative to enantiomerically pure or enantiomerically enriched pramipexole.
  • 6S residual-6S-2- amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole
  • a description of methods for producing high purity dexpramipexole can be found in U.S. Application No.
  • treatment with dexpramipexole may include administering daily doses of about 100 mg or more, about 125 mg or more, about 150 mg or more, 300 mg or more, 400 mg or more, 450 mg or more, 500 mg or more, or 600 mg or more without the adverse side effects associated with dopaminergic agonism.
  • daily doses of dexpramipexole of about 150 mg or more or about 300 mg or more, or about 450 mg or more, or about 600 mg or more may be administered without an apparent impact on heart rate, blood pressure, or other cardiac activity that can be measured using, for instance, ECG or blood pressure cuff that would otherwise be indicative of treatment with a dopamine agonist.
  • adverse side-effects associated with low dose pramipexole treatment include, but are not limited to, dizziness, hallucination, nausea, hypotension, somnolence, constipation, headache, tremor, back pain, postural hypotension, hypertonia, depression, abdominal pain, anxiety, dyspepsia, flatulence, diarrhea, rash, ataxia, dry mouth, extrapyramidal syndrome, leg cramps, twitching, pharyngitis, sinusitis, sweating, rhinitis, urinary tract infection, vasodilatation, flu syndrome, increased saliva, tooth disease, dyspnea, increased cough, gait abnormalities, urinary frequency, vomiting, allergic reaction, hypertension, pruritis, hypokinesia, nervousness, dream abnormalities, chest pain, neck pain, paresthesia, tachycardia, vertigo, voice alteration, conjunctivitis, paralysis, tinnitus, lacrimation, myd
  • treatment including administration of daily doses of about 100 mg or more, about 125 mg or more, about 150 mg or more, or about 300 mg or more, or about 400 mg or more, or about 450 mg or more, or about 500 mg or more, or about 550 mg or more, or about 600 mg or more of dexpramipexole may be carried out for prolonged periods of time such as, for example, 12 weeks or more, 6 months or more, 1 year or more and, in certain embodiments, for 2, 3, 5 or 10 years or more, and in other embodiments, for an indefinite period of time of.
  • embodiments of the invention include methods of treating ALS may include administering dexpramipexole for an extended or prolonged period of time.
  • the extended period of time may be about 12 weeks or longer, about 6 months or longer, about 1 year or longer, and in other embodiments, a method of treating ALS comprises administering dexpramipexole on a maintenance dosing regimen.
  • the maintenance dosing regimen may include administering about 100 mg or more, about 125 mg or more, about 150 mg or more, about 300 mg or more, about 450 mg or more, about 500 mg or more, or about 550 mg or more, or about 600 mg or more of dexpramipexole per day without any titration (or an initial dosing regimen of less than the maintenance dose).
  • various embodiments are directed to maintenance therapy in which a dosing schedule for dexpramipexole is maintained for an extended period of time without titration or otherwise changing the dosing schedule.
  • the extended period of time may be about 12 weeks or longer, about 6 months or longer, about 1 year or longer, 2, 3, 4, 5, or 10 years or longer, and in certain embodiments, an indefinite period of time.
  • the maintenance dosing may include administering less than the initial daily dose, such as, less than about 150 mg, or less than about 300 mg, or less than about 600 mg of dexpramipexole per day.
  • the adverse effects associated with dopamine agonist treatment such as those described above may not develop after treatment with dexpramipexole has been carried out for a period of time of at least 12 weeks or more, and in some embodiments at least 6 months or 1, 2, 3, 5 or 10 years or more.
  • an initial dosing regimen may be provided.
  • the initial dosing regimen may include administering a higher dose of dexpramipexole than the maintenance dosing regimen as either a single administration or by administering an increased dosage for a limited period of time prior to beginning a maintenance dosing regimen.
  • the initial dosing regimen may be about 300 mg to about 500 mg or more of dexpramipexole per day, or from about 300 mg to about 600 mg or more of dexpramipexole per day, these initial dosing regimen may continue for 1, 2, 3, 4, 5, 6, or 7 days, up to 4 weeks, up to 8 weeks, or up to 12 weeks.
  • the patient may be administered a maintenance dosing regimen of, for example, about 100 mg or more, about 125 mg or more, about 150 mg or more, about 300 mg or more, about 400 mg or more, about 450 mg or more, about 500 mg or more, or about 550 mg or more, or about 600 mg or more of dexpramipexole for an indefinite period of time such as, for example, at least 12 weeks or more or at least 6 months or 1, 2, 3, 5 or 10 years or more.
  • patients undergoing a maintenance dosage regimen may be administered a higher dosage treatment at one or more times during the maintenance dosage regimen.
  • dexpramipexole may be administered to any individual exhibiting the symptoms of a neurodegenerative disease or individuals predisposed to neurodegenerative disease.
  • neurodegenerative diseases that may be treated using dexpramipexole include Huntington's Chorea, metabolically induced neurological damage, Alzheimer's disease, senile dementia, age associated cognitive dysfunction, vascular dementia, multi-infarct dementia, Lewy body dementia, neurodegenerative dementia, neurodegenerative movement disorder, ataxia, Friedreich's ataxia, multiple sclerosis, spinal muscular atrophy, primary lateral sclerosis, seizure disorders, motor neuron disorder or disease, inflammatory demyelinating disorder, Parkinson's disease, amyotrophic lateral sclerosis (ALS), hepatic encephalopathy, and chronic encephalitis.
  • the compositions and methods of the invention may be used to treat nearly any individual exhibiting symptoms of a neurological disease or susceptible to such diseases.
  • dexpramipexole may be used to treat ALS.
  • individuals who were diagnosed with ALS within two years or less may be treated with dexpramipexole to reduce, eliminate or slow advancement of ALS or symptoms associated with ALS such as, for example, fine motor function loss, gross motor function, loss of bulbar function, and loss of respiratory function.
  • dexpramipexole may be administered to reduce or slow the advancement of symptoms including, but not limited to, trembling, loss of muscle control, loss of ability to write, low of ability to move or roll over, loss of speech, inability to swallow, difficulty breathing, and so on.
  • individuals with advanced symptoms or who were diagnosed with ALS more than 2 years before beginning treatment may be treated with
  • dexpramipexole and such individuals may respond to treatment by exhibiting a reduction or elimination of one or more ALS related symptoms or, in certain embodiments, the rate of symptom onset or advancement may be reduced, for example, the rate of motor function loss, loss of speech and/or swallowing may be slowed.
  • a dose dependent response may be associated with treatment with dexpramipexole, and in certain embodiments, a dose dependent response may be enhanced when treatment is carried out for longer periods of time.
  • a naive patient who is administered a daily dose of, for example, about 300 mg of dexpramipexole or more, or about 450 mg or more, or about 500 mg or more, or about 600 mg or more may exhibit greater improvement in one or more symptoms of a neurological disease than a similarly situated naive patient who is administered a daily dose of dexpramipexole less than 300 mg or less than 450 mg, or less than 500 mg, or less than 600 mg. In such embodiments, this improvement resulting from higher dosage
  • treatment with higher doses of dexpramipexole may be carried out for prolonged periods of time, and the improvement associated with such dexpramipexole treatment may be realized after treatment has been carried out for a period of time of, for example, 1, 2, 3, 4, 5, 6, or 7 days, up to 1, 2, 4, 6, 8, 12, 24, or 48 weeks, up to 5, 10, 15, or 20 years, or any number of weeks between the recited values.
  • treatment with higher doses of dexpramipexole may be carried out as maintenance therapy, wherein the patient is administered such doses of dexpramipexole at the initiation of treatment and, thereinafter continue such doses of dexpramipexole over time.
  • any of the doses of dexpramipexole and/or any of the dosing regimens of dexpramipexole described herein may be used in such methods and continued administration of such doses may be continued for any of the described periods of time.
  • the observed improvement in one or more symptoms may become enhanced as treatment progresses such that after an improvement is observed further improvements in the one or more symptoms may become evident with continued treatment.
  • a lag between beginning treatment and the first observation of improvement may be due to a period in which the dexpramipexole concentration in one or more of the patient's tissues increases to a threshold level where symptom improvement is observed. Any lag before observation of improvement may vary between patients and may vary depending on, for instance, the patient's demographics or characteristics such as, for example, age, progression of the disease, and/or the time between the onset of symptoms of the disease and beginning treatment.
  • dexpramipexole may be administered to patients in need of treatment for excessive weight loss associated with ALS.
  • the precipitous weight loss that is a cardinal symptom of ALS may be associated with increased energy expenditure, skeletal muscle hypermetabolism, and the systematic wasting of muscle tissue known as cachexia.
  • the total daily dose of dexpramipexole administered may be for example, less than 150 mg to 300 mg or greater, 400 mg or greater, 450 mg or greater, 500 mg or greater, or 600 mg or greater.
  • any of the doses of dexpramipexole and/or any of the dosing regimens of dexpramipexole described herein may be used in such methods and continued administration of the such doses may be continued for any of the described periods of time.
  • dexpramipexole may be administered by titration where one or more initial doses are less than 150 mg, less than 300 mg, less than 400 mg, less than 450 mg, less than 500 mg, less than 600 mg, and so on when administered to naive patients.
  • pramipexole treatment requires titration because pramipexole has a significant adverse impact on naive patients, and titration over the course of weeks in which the dosage regimen is periodically increased to reach higher dosages purportedly limits these adverse effects.
  • no titration of dexpramipexole is required.
  • an effective daily dose of dexpramipexole is, for example, 150 mg, 300 mg, 450 mg, or 600 mg
  • the initial dose of dexpramipexole may be 150 mg, 300 mg, 450 mg, or 600 mg, of dexpramipexole, and each daily dose thereafter may be 150 mg, 300 mg, 450 mg, or 600 mg.
  • the daily dose may be considered a "stable daily dose.”
  • dexpramipexole treatment can be initiated at high levels without the need for titration.
  • a naive patient who requires a greater than about 150 mg or about 300 mg or more, or about 400 mg or more, or about 450 mg or more, or about 500 mg or more, or about 600 mg or more dose of dexpramipexole for treatment may be administered about 100 mg or more, about 125 mg or more, about 150 mg or more, 300 mg or more, 400 mg or more, 450 mg or more, 500 mg or more, or 600 mg or more of dexpramipexole during the first treatment without the onset of adverse effects as would be expected if pramipexole was administered at its terminal level during an initial treatment.
  • embodiments of the invention are directed to a method of treating a patient with ALS including administering an effective amount of dexpramipexole without titration.
  • the effective amount may be about 100 mg or more, about 125 mg or more, about 150 mg or more, about 300 mg or more, about 400 mg or more, about 450 mg or more, about 500 mg or more, or about 600 mg or more daily, in some embodiments, the effective amount may be about 300 mg or more daily, or the effective amount may be about 450 mg or more daily and in some embodiments, the effective amount may be about 600 mg or more daily.
  • the effective amount may be administered in separate equal doses twice daily.
  • the effective amount may be administered in separate equal doses of 150 mg dexpramipexole three times daily.
  • the effective amount may be administered three times daily or about every 8 hours, for example, in some embodiments, the effective amount of 450 mg may be administered 150 mg three times daily or about every 8 hours, or the effective amount may be administered once daily or about every 24 hours, for example, in some embodiments, the effective amount of 450 mg may be administered 450 mg once daily.
  • the effective amount may be administered in separate equal doses of 300 mg dexpramipexole twice daily.
  • the effective amount may be administered twice daily or about every 12 hours, for example, in some embodiments, the effective amount of 600 mg may be administered 300 mg twice daily or about every 12 hours or the effective amount may be administered once daily or about every 24 hours, for example, in some embodiments, the effective amount of 600 mg may be administered 600 mg once daily.
  • any of the doses of dexpramipexole and/or any of the dosing regimens of dexpramipexole described herein may be used in such methods and continued administration of the such doses may be continued for any of the described periods of time.
  • Embodiments of the invention are also directed to a dosage regimen for administering dexpramipexole.
  • the dosage regimen may include an initial dose dexpramipexole in one or more unit doses, then a plurality of daily doses having an equal amount of dexpramipexole as the initial dose in one or more unit doses.
  • Such embodiments are not limited by the amount of the initial dose and daily doses.
  • the initial dose and each of the plurality of daily doses may be from about 50 mg to about 300 mg or about 400 mg, or about 450 mg, or about 500 mg or about 600 mg of dexpramipexole.
  • the initial dose and each of the plurality of daily doses may be from about 100 mg or more to about 300 mg or about 400 mg, or about 450 mg, or about 500 mg, or about 600 mg of dexpramipexole, and in still other embodiments, the initial dose and each of the plurality of daily doses may be about 300 mg or more, about 400 mg or more, or about 450 mg or more, or about 500 mg or more, or about 600 mg or more of dexpramipexole.
  • the one or more unit doses of the dosage regimen may be 1 to 5 unit doses, and in such embodiments, each of the one or more unit doses may be substantially equal. In some embodiments, the one or more unit doses of the dosage regimen may be 150 mg of
  • each unit dose of the dosage regimen may be 300 mg of dexpramipexole. In some embodiments, two unit doses of 300 mg are administered daily, wherein each unit dose may be substantially equal. In other embodiments, each unit dose of the dosage regimen may be a solid unit dose.
  • dexpramipexole described herein may be used in any of the methods, and the dosing regiment may be carried out using any of the compositions described herein.
  • dexpramipexole may be administered to ALS patients, and in such embodiments, the improvements observed in ALS patients treated with dexpramipexole may be significantly better than conventional treatments such as, for example, riluzole.
  • the improvement may be signified by greater than 20% increase in ALS Functional Rating Scale, Revised (ALSFRS-R) score, when compared to baseline scores taken before treatment, and in other embodiments, this improvement may be manifested in a greater than 30% increase in ALSFRS-R score.
  • the improvement in ALSFRS-R score may become apparent in less than 9 months, and in some embodiment, less than 6, 3, or 1 month.
  • dexpramipexole may be administered for the treatment of ALS without incurring adverse events associated with, for example, riluzole, the current standard of pharmacological intervention for ALS.
  • the overall rates of adverse events may be higher among patients receiving riluzole concomitant with dexpramipexole or in conjunction with placebo. Headaches, for example, were reported by four times as many patients receiving riluzole as those not receiving riluzole.
  • dexpramipexole may be administered to improve the general health of individuals having a neurological disease, and in other embodiments, dexpramipexole may be administered to alleviate one or more specific symptoms.
  • dexpramipexole may be administered to ALS patients to improve symptoms associated with for example, fine motor, speech and swallowing or a combination thereof
  • improvements in fine motor and speech and swallowing related symptoms may become apparent in a shorter period of time following the initiation of dexpramipexole treatment than, for instance, improvements in large motor function and pulmonary related symptoms.
  • dexpramipexole may be administered to alleviate fine motor and speech and swallowing related symptoms more immediately than other ALS symptoms. Therefore, in certain embodiments, ALS patients treated with dexpramipexole may have an increased time before a feeding tube must be employed because such patients may retain the ability to masticate and swallow food stuffs under their own power.
  • dexpramipexole may be administered to slow the rate of decline of a patient exhibiting symptoms of a neurological disease and/or to reduce mortality in such patients.
  • populations of patients diagnosed with a neurological disease such as, for example, ALS, may exhibit an increased time to death, an increased survival rate, and/or a decreased frequency of death as a result of treatment with dexpramipexole.
  • dexpramipexole treatment may improve the quality of life for such patients up to death.
  • the foregoing methods may comprising administering dexpramipexole on a dosing regimen to achieve a dose dependent, steady state AUCo-12 (h x ng/mL) ranging from 836 +234 to 2803 +1635 to 6004 + 2700 at daily doses of 50 mg, 150 mg, and 300 mg, respectively, when administered in two equal doses twice daily.
  • a dose dependent, steady state AUCo-12 h x ng/mL
  • dexpramipexole treatment may be carried out in combination with other forms of treatment.
  • such combination therapy may produce synergistic effects, such that the effect of dexpramipexole is augmented wherein one or more symptoms show a dramatic improvement over pre-treatment levels.
  • dexpramipexole treatment may be carried out in combination with (simultaneously or concurrently) with riluzole without adverse effects or reduced symptom relief.
  • dexpramipexole may be administered in combination with (simultaneously or concurrently) with an additional form of treatment including, but not limited, those set forth in U.S. Provisional No. 61/113,680 filed November 12, 2008 and U.S. Provisional No. 61/090,094 filed August 19, 2009, each of which are hereby incorporated by reference in their entirety without producing adverse effects.
  • the pharmaceutical composition of dexpramipexole may achieve the effects described above by eliciting a neuroprotective, anti-oxidative, antiapoptotic, or other beneficial cellular effects without the side-effects associated with dopamine agonists commonly used to treat neurodegenerative diseases.
  • the ability to deliver clinically effective doses of dexpramipexole without dose limiting side effects may be made possible by: (i) the synthesis of dexpramipexole that is pure within limits of the detection; and (ii) dexpramipexole possesses a substantially lower affinity for dopamine receptors than its enantiomer, pramipexole.
  • Various embodiments of the invention include methods for treating a neurodegenerative disease by administering a therapeutically effective amount of dexpramipexole such as, for example, about 100 mg or more, about 125 mg or more, about 150 mg, or more, about 300 mg or more, or about 450 mg or more, or about 600 mg or more.
  • dexpramipexole may be formulated as a pharmaceutical or therapeutic composition by combining with one or more pharmaceutically acceptable carriers.
  • such pharmaceutical or therapeutic compositions may be formulated in tablet or capsule form for use in oral administration routes.
  • the compositions and amounts of non-active ingredients in such a formulation may depend on the amount of the active ingredient, and on the size and shape of the tablet or capsule. Such parameters may be readily appreciated and understood by one of skill in the art.
  • the pharmaceutical compositions of the invention may have a chiral purity for dexpramipexole of at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8%, at least 99.9%, at least 99.95%, or in some embodiments, at least 99.99%.
  • the chiral purity for dexpramipexole may be about 100%.
  • Such high chirally pure dexpramipexole allows for therapeutic and pharmaceutical compositions that may have a wide individual and daily dose range.
  • the present invention provides a composition including only dexpramipexole in a
  • compositions may further include a pharmaceutically acceptable carrier, excipient and/or diluent.
  • the amount of pramipexole, (6S)-2-amino-4,5,6,7- tetrahydro-6- (propylamino)benzothiazole, remaining in the chirally pure dexpramipexole may be an amount not exceeding about 1.0 mg, and in some embodiments, the amount of pramipexole may be an amount not exceeding about 0.75 mg, about 0.5 mg, about 0.25 mg, or about 0.125 mg. In particular embodiments, the amount of pramipexole in chirally pure dexpramipexole may be less than about 0.125 mg.
  • the amount of pramipexole that may be administered in pharmaceutical compositions containing the chirally pure dexpramipexole of various embodiments may be less than 1.0 mg/day, less than 0.5 mg/day, and in certain embodiments, less than 0.125 mg/day.
  • the amount of pramipexole in chirally pure dexpramipexole may be a non-effective dose such that any pramipexole in such compositions does not elicit a noticeable effect on patients who are administered the pharmaceutical compositions of the invention.
  • a 300 mg/day dose of dexpramipexole administered to a patient as a single unit dose containing chiral purity dexpramipexole at least about 99.8 % may contain a non-effective dose pramipexole less than 1.0 mg/day, a 300 mg/day dose of about 99.9 % chirally pure dexpramipexole may include non-effective dose amount of pramipexole less than 0.5 mg/day, and a 300 mg/day dose of about 99.98 % dexpramipexole may include non-effective dose pramipexole of less than 0.125 mg/day.
  • a 450 mg/day dose of dexpramipexole administered to a patient as a single unit dose containing chiral purity dexpramipexole at least about 99.8 % may contain a non-effective dose pramipexole less than 1.0 mg/day, a 450 mg/day dose of about 99.9 % chirally pure dexpramipexole may include non-effective dose amount of pramipexole less than 0.5 mg/day, and a 450 mg/day dose of about 99.98 % dexpramipexole may include non-effective dose pramipexole of less than 0.125 mg/day.
  • a 600 mg/day dose of dexpramipexole administered to a patient as a single unit dose containing chiral purity dexpramipexole at least about 99.8 % may contain a non-effective dose pramipexole less than 1.0 mg/day, a 600 mg/day dose of about 99.9 % chirally pure dexpramipexole may include non-effective dose amount of pramipexole less than 0.5 mg/day, and a 600 mg/day dose of about 99.98 % dexpramipexole may include non-effective dose pramipexole of less than 0.125 mg/day.
  • Chirally pure dexpramipexole may be prepared or converted to a pharmaceutically acceptable salt of dexpramipexole.
  • dexpramipexole may be formulated as (6R)-2-amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole dihydrochloride, which is a pharmaceutical salt and may improve solubility of dexpramipexole in water.
  • the conversion of (6R)-2- amino-4,5,6,7- tetrahydro-6-(propylamino)benzothiazole to an acceptable salt by any method known in the art.
  • (6R)-2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride may be prepared by a one step method in which (6R)-2-amino-4,5,6,7- tetrahydro-6- (propylamino)benzothiazole or (6R)-2-amino-4,5,6,7-tetrahydro-6- (propylamino)benzothiazole salt is reacted with concentrated HC1 in an organic solvent such as, an alcohol, at a reduced temperature of, for example, from about 0°C to about 5°C.
  • an organic solvent such as, an alcohol
  • the amount of dexpramipexole or a pharmaceutically acceptable salt thereof present in such a pharmaceutical composition suitable for oral administration may vary.
  • the amount of dexpramipexole or a pharmaceutically acceptable salt thereof in such compositions may range from about 25 mg to about 1000 mg, about 50 mg to about 1000 mg, from about 100 mg to about 1000 mg, from about 125 mg to about 1000 mg, from about 150 mg to about 1000 mg, from about 300 mg to about 1000 mg, from about 500 mg to about 1000 mg, from about 600 to about 1000 mg, in certain embodiments, the amount of dexpramipexole may range from about 60 mg to about 300 mg, or from about 60 mg to about 450 mg, and in certain embodiments, the amount of dexpramipexole may be from about 60 mg to about 600 mg.
  • Each of the compositions and pharmaceutical compositions embodied herein may be used in any of the methods or dosage regimen described herein.
  • the daily dose of dexpramipexole may be administered as a single daily dose or may be divided into two or more doses of equal or unequal amount administered throughout the day.
  • about 100 mg or more, about 125 mg or more, about 150 mg or more, 300 mg or more, 450 mg or more, 500 mg or more, or 600 mg or more of dexpramipexole may be administered in 1 to 5 doses each containing an equal amount of
  • dexpramipexole and in other embodiments, about 100 mg or more, about 125 mg or more, about 150 mg or more, 300 mg or more, 450 mg or more, 500 mg or more, or 600 mg or more of dexpramipexole may be administered in 2 or 3 doses throughout the day. In still other embodiments, about 100 mg or more, about 125 mg or more, about 150 mg or more, 300 mg or more, 450 mg or more, 500 mg or more, or 600 mg or more of dexpramipexole may be administered in 2 or 3 doses wherein the one dose contains a higher concentration of dexpramipexole.
  • one dose of a 300 mg regimen may contain 100 mg of dexpramipexole and a second dose administered at a different time during the day may contain 200 mg of dexpramipexole.
  • one dose of a 450 mg regimen may contain 300 mg of dexpramipexole and a second dose administered at a different time during the day may contain 150 mg of dexpramipexole, or one dose of a 450 mg regimen may contain 150 mg of dexpramipexole, a second dose administered at a different time during the day may contain 150 mg of dexpramipexole, and a third dose administered at a different time during the day may contain another 150 mg of dexpramipexole.
  • one dose of a 600 mg regimen may contain 300 mg of dexpramipexole and a second dose administered at a different time during the day may contain 300 mg of dexpramipexole.
  • the daily doses may be used in any of the methods or dosage regimen described herein.
  • compositions of the invention may be prepared, packaged, sold in bulk, as a single unit dose, or as multiple unit doses and can be administered in the conventional manner by any route where they are active.
  • the compositions may be administered orally, ophthalmically, intravenously, intramuscularly, intra-arterially, intramedularry, intrathecally, intraventricularly, transdermally, subcutaneously, intraperitoneally, intravesicularly, intranasally, enterally, topically, sublingually, rectally by inhalation, by depot injections, or by implants or by use of vaginal creams, suppositories, pessaries, vaginal rings, rectal suppositories, intrauterine devices, and transdermal forms such as patches and creams.
  • compositions containing dexpramipexole in a solid dosage may include, but are not limited to, tablets, capsules, cachets, pellets, pills, powders and granules; topical dosage forms which include, but are not limited to, solutions, powders, fluid emulsions, fluid suspensions, semisolids, ointments, pastes, creams, gels and jellies, and foams; and parenteral dosage forms which include, but are not limited to, solutions, suspensions, emulsions, and dry powder; comprising an effective amount of a polymer or copolymer of the present invention.
  • the active ingredients can be contained in such formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • pharmaceutically acceptable diluents fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • the means and methods for administration are known in the art and an artisan can refer to various pharmacologic references for guidance. For example, Modern Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Oilman's The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980) can be consulted
  • the compounds can be formulated readily by combining these compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP).
  • disintegrating agents can be added, such as, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • compositions may be suitable for oral administration such as, for example, a solid oral dosage form or a capsule, and in certain embodiments, the composition may be a tablet.
  • Such tablets may include any number of additionally agents such as, for example, one or more binder, one or more lubricant, one or more diluent, one or more lubricant, one or more surface active agent, one or more dispersing agent, one or more colorant, and the like.
  • Such tablets may be prepared by any method known in the art, for example, by compression or molding.
  • Compressed tablets may be prepared by compressing in a suitable machine the ingredients of the composition in a free-flowing form such as a powder or granules, and molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may be uncoated and, in other embodiments, they may be coated by known techniques.
  • the pharmaceutical compositions of the invention may be provided in a dragee cores with suitable coatings.
  • dragee cores may be prepared suing concentrated sugar solutions, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions including an effective amount of dexpramipexole prepared for oral administration may include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the coatings may delay disintegration and absorption in the gastrointestinal tract and thereby providing a sustained action over a longer period. Additionally, such coatings may be adapted for release dexpramipexole in a predetermined pattern (e.g., in order to achieve a controlled release formulation) or it may be adapted not to release the active compound until after passage of the stomach (enteric coating).
  • Suitable coatings encompassed by such embodiments may include, but are not limited to, sugar coating, film coating (e.g., hydroxypropyl methylcellulose, methyl-cellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone), or an enteric coating (e.g., methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and/or ethylcellulose).
  • film coating e.g., hydroxypropyl methylcellulose, methyl-cellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone
  • enteric coating e.g
  • a time delay material such as, for example, glyceryl monostearate or glyceryl distearate may be incorporated into the coatings of some embodiments.
  • solid tablet compositions may include a coating adapted to protect the composition from unwanted chemical changes, for example, to reduce chemical degradation prior to the release of the active drug substance.
  • composition suitable for oral administration encompassed in embodiments of the invention may include a therapeutically effective amount of dexpramipexole and a non-effective dose amount of pramipexole and may further include one or more diluent, one or more disintegrant, one or more lubricant, one or more pigment or colorant, one or more gelatin, one or more plasticizer and the like.
  • a tablet may include dexpramipexole, from about 20% to about 50% by weight of diluent in an amount, from about 10% to about 30% by weight of a second diluent, from about 2% to about 6% by weight of a disintegrant, and from about 0.01% to about 2% by weight of a lubricant, and in particular embodiments, such tablets may include an effective amount of dexpramipexole, from about 20% to about 50% by weight microcrystalline cellulose, about 10% to about 30% by weight, from about 2% to about 6% crospovidone or croscarmellose, and from about 0.01 % to about 2% by weight magnesium stearate.
  • the pharmaceutical composition may include any amount or combination of microcrystalline cellulose, mannitol, sodium, crospovidone, croscarmellose magnesium stearate, or combination thereof.
  • the pharmaceutical composition suitable for oral administration may include at least about 50 mg of dexpramipexole, and in some embodiments, such pharmaceutical compositions may include at least about 75 mg of dexpramipexole, at least about 100 mg of dexpramipexole, at least about 150 mg of dexpramipexole, at least about 200 mg of dexpramipexole, at least about 250 mg of dexpramipexole, 300 mg of dexpramipexole, at least about 450 mg of dexpramipexole, at least about 500 mg of dexpramipexole, at least about 600 mg of dexpramipexole, at least about 750 mg of dexpramipexole,
  • the pharmaceutical compositions including dexpramipexole may be prepared as suspensions, solutions or emulsions in oily or aqueous vehicles suitable for injection.
  • such liquid formulations may further include formulatory agents such as suspending, stabilizing and/or dispersing agents formulated for parenteral administration.
  • Such injectable formulations may be administered by any route, for example, subcutaneous, intravenous, intramuscular, intra-arterial or bolus injection or continuous infusion, and in embodiments in which injectable formulations are administered by continuous infusion, such infusion may be carried out for a period of about 15 minutes to about 24 hours.
  • formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • dexpramipexole may be formulated as a depot preparation, and such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Depot injections can be administered at about 1 to about 6 months or longer intervals.
  • the compounds can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions including dexpramipexole may be formulated for buccal or sublingual administration.
  • the pharmaceutical compositions may be prepared as chewable tablets, flash melts or lozenges formulated in any conventional manner.
  • compositions including dexpramipexole may be formulated for administration by inhalation.
  • pharmaceutical compositions according to the invention may be delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro ethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro ethane, carbon dioxide or other suitable gas.
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or star
  • compositions including dexpramipexole can be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • compositions including dexpramipexole may be formulated for transdermal administration.
  • such pharmaceutical compositions may be prepared to be applied to a plaster or applied by transdermal, therapeutic systems that are supplied to the patient.
  • dexpramipexole for transdermal administration may include a suitable solid or gel phase carriers or excipients such as, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as, e.g.,polyethyleneglycols.
  • pharmaceutical compositions including dexpramipexole may be administered alone as a single therapeutic agent.
  • the pharmaceutical compositions including dexpramipexole may be administered in combination with one or more other active ingredients, such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • Example 1 was a randomized, placebo-controlled, double-blind, parallel-group, multi-center study to evaluate the safety, tolerability, and clinical effects of oral administration of 3 dosage levels of dexpramipexole vs. placebo for 12 weeks in patients with ALS.
  • 80 eligible patients were to be randomized to 1 of 4 treatment groups in a 1 : 1 : 1 : 1 ratio for 12 weeks of treatment with dexpramipexole (50 mg, 150 mg, or 300 mg total daily dose) or placebo.
  • Doses were administered as 25 mg, 75 mg, or 150 mg dexpramipexole every twelve hours, or placebo every twelve hours.
  • ALSFRS-R ALS Functional Rating Scale
  • VC vital capacity
  • McGill SIS McGill Quality-of-Life Single-Item Scale
  • WOCBP Women of childbearing potential
  • Surgical sterilization (i.e., vasectomy) of the male partner was considered as one effective method of interventional contraception.
  • using the rhythm method was not considered sufficient.
  • WOCBP must have also agreed to pregnancy testing and have had a negative pregnancy test at periodic study visits.
  • Non- surgically sterilized men whose sexual partners were WOCBP must have agreed to ensure their partners used at least one highly effective contraception method (e.g., oral, injected or implanted hormonal methods, or intrauterine device) prior to study entry, for the duration of the study, and for 28 days after the last dose of study medication.
  • highly effective contraception method e.g., oral, injected or implanted hormonal methods, or intrauterine device
  • a total of 102 subjects were randomized at 20 US sites and received at least 1 dose of study medication: 27 subjects received placebo, 23 subjects received dexpramipexole 50 mg, 26 subjects received dexpramipexole 150 mg, and 26 subjects received dexpramipexole 300 mg. Enrollment by site ranged from 1 to 10 subjects. A total of 98 subjects (96%) completed Part 1 of the study. Two subjects (1 in the 50 mg dose group and 1 in the 300 mg dose group) withdrew consent and 2 subjects (1 in the placebo group and 1 in the 300 mg group) discontinued due to an adverse event.
  • the mean duration of disease at time of randomization (mean time from ALS symptom onset to Day 1 of dosing in the study) across treatment groups was 427 days (15.25 months).
  • the placebo and 150 mg groups had the longest mean durations of disease (473 and 458 days, respectively), while the 50 mg and 300 mg groups had the shortest mean durations of disease (381 and 391 days, respectively).
  • the percentage of subjects reporting at least 1 AE that had been judged by the investigator to be possibly or probably treatment- related was 22% (placebo), 17% (50 mg), 42% (150 mg), and 27% (300 mg).
  • ALSFRS-R mean scores at baseline were similar across the treatment groups.
  • the mean changes from baseline to endpoint in ALSFRS-R total scores were -3.6 (placebo), -5.0 (50 mg), -3.3 (150 mg), and -2.2 (300 mg).
  • the median changes from baseline to endpoint in ALSFRS-R scores were -4.0 (placebo), -3.0 (50 mg), -2.5 (150 mg), and -2.0 (300 mg).
  • the mean and median decline from baseline to study endpoint in the ALSFRS-R score was reduced by 39% and 50%, respectively, compared with the placebo group.
  • the primary analysis of ALSFRS-R data specified in the SAP was a linear mixed-effects analysis of the treatment effect on the slope of ALSFRS-R scores during the study.
  • the slope observed for the placebo group was -1.278, whereas the slope observed for the 300 mg group was -0.878, a 31% improvement relative to the placebo group.
  • ANCOVA of change from baseline in ALSFRS-R total scores on dose was conducted to adjust for selected baseline covariates (gender, duration of ALS symptoms at baseline, concomitant riluzole use, and baseline ALSFRS-R score).
  • the mean score on this 10-point McGill Quality of Life (QOL) scale at baseline was 7.0 (placebo), 6.8 (50 mg), 7.3 (150 mg), and 8.1 (300 mg). Median scores at baseline were 7.0 (placebo and 50 mg) and 8.0 (150 mg and 300 mg).
  • the mean change from baseline in QOL scores at endpoint were 0.0 (placebo), -0.6 (50 mg), -0.6 (150 mg), and -0.9 (300 mg).
  • the mean change from baseline at each time point in the placebo group was influenced by one outlier who reported a score of 0 at baseline (due to discomfort associated with the lumber puncture procedure) and subsequently reported a score of 10 for all on- treatment visits.
  • the parameter estimates for total plasma clearance uncorrected for bioavailability (CL/F), volume of distribution uncorrected for bioavailability (Vz/F), and half-life (t'/2) are comparable between the 2 populations.
  • CL/F total plasma clearance uncorrected for bioavailability
  • Vz/F volume of distribution uncorrected for bioavailability
  • t'/2 half-life
  • a 6-point or greater drop in ALSFRS-R total score from baseline has been used to identify subjects that failed to respond to drug treatment.
  • a 6-point or greater drop in ALSFRS-R total score from baseline to 12 weeks in Part 1 was used to define treatment failure in a post hoc analysis, a significant dose-dependent effect was observed.
  • the number of failures totaled 9 subjects (33%) in the placebo group; 8 subjects (35%) in the 50 mg/day group, 4 subjects (15%) in the 150 mg/day group, and 2 subjects (8%) in the 300 mg/day group (logistic regression analysis, p 0.014; FIG. 2).
  • the failure line is defined as anything at or below the dotted line, and the red lines are the median decline at the indicated week.
  • dexpramipexole exhibited almost no reduction in handwriting score while patients receiving placebo or smaller daily doses of dexpramipexole showed a reduction in handwriting.
  • patients receiving 300 mg/day of dexpramipexole exhibited less reduction in cutting food and dressing and hygiene scores than patients receiving placebo or lower dose dexpramipexole (FIG. 4B and 4C).
  • behaviors associated with bulbar function also exhibit a less dramatic decline in ALSFRS-R score over baseline when patients received dexpramipexole and, in particular, 300 mg/day of dexpramipexole. Of the behaviors quantified, swallowing scores appeared to be maintained better than other behaviors (FIG. 5A).
  • ALSFRS-R ALS Functional Rating Scale
  • the ALSFRS-R scored 0 48, is used to evaluate overall functional status of ALS patients in clinical trials as well as in clinical practice.
  • FIG. 9 shows a box plot of the results of ALSFRS-R total score of subjects taken at 4 week intervals for each treatment group.
  • FIG. 10 shows the change from baseline for each subject in each treatment group as indicated on the x-axis with lines indicating the median score for the group and with baseline as indicated by 0.
  • Table 3 Weight Loss in ALS Patients Treated with dexpramipexole
  • Part 1 Subjects completing Part 1 (as set-forth in EXAMPLE 1) were eligible to continue into Part 2 of the study.
  • Part 2 was a randomized, double-blind, 2-arm, parallel-group, extension study evaluating the longer-term safety, tolerability, and clinical effects of oral administration of 2 dosage levels of dexpramipexole (50 mg and 300 mg).
  • a 4- week, single-blind, placebo washout period was carried out.
  • the subjects were then re-randomized to 1 of 2 daily dosage levels of dexpramipexole (50 mg or 300 mg) and treated in Part 2 for up to 72 weeks.
  • the solid tablet formulation contained the following inactive ingredients (listed in order of percent volume): microcrystalline cellulose, mannitol, crospovidone, and magnesium stearate (vegetable source).
  • inactive ingredients listed in order of percent volume: microcrystalline cellulose, mannitol, crospovidone, and magnesium stearate (vegetable source).
  • Week 2 study drug was dispensed at baseline which was the same visit as the Part 1 Week 12 visit (beginning of the placebo washout period), at Week 4 (end of placebo washout), Week 8, Week 12, Week 20, Week 28, Week 40, Week 52, and Week 64.
  • any medication or supplement the subject used other than the study drug specified in the protocol was considered a concomitant medication whether it was a prescription medication or over- the-counter product.
  • the use of concomitant medications during this study was recorded throughout Part 2 of the study. All concomitant medications were recorded in the subject's source document and on the CRFs. Co-administration of other dopamine agonist medication(s) was not allowed during the trial.
  • Safety evaluations included physical examination, neurological examination, vital signs, 12-lead ECG, laboratory evaluations, lithium screening, and monitoring of adverse events.
  • Vital signs including systolic and diastolic blood pressure, respiratory rate, pulse rate, and temperature, were measured after the subject had rested for 5 minutes. The following guidelines were used to grade the intensity of an AE:
  • ALSFRS-R, VC, and McGill QoL-SIS scores were summarized by treatment group with the rate of change estimate derived from a linear mixed-effects model.
  • Linear decline of the ALSFRS-R over time has been shown previously. If the linearity assumption did not hold (quadratic term with a p- value ⁇ 0.05), a repeated measures mixed-effect model was to be used.
  • a mixed-model analysis was used to fit a model that included time, treatment group, and the interaction between time and treatment group simultaneously.
  • the coefficient of time (the slope, or rate of change) estimated for each treatment group was used to test for differences between the treatment groups. Coefficient of time estimate along with its standard error was reported.
  • a subject who died earlier than the comparator subject was given a comparison score of -1 ; if 2 subjects completed the study, their comparison score was based on a comparison of their ALSFRS-R change values at the end of the study; if a subject discontinued early, his comparison to each other subject was based on the comparison of their ALSFRS-R change at the latest time point at which they both had an ALSFRS-R value.
  • the Kaplan-Meier estimates of median time to death or tracheostomy and 95% confidence intervals, and the 25th and 75th quartiles and 95% confidence intervals were presented for each treatment group.
  • the comparison between the 2 treatment groups was performed using a log rank test.
  • a figure of the Kaplan-Meier estimated curve for each treatment group was also presented. The number and percentage of subjects who were hospitalized for tracheostomy or died or were censored were tabulated. If an insufficient number of events occurred, only the tabulation of subjects who were hospitalized for tracheostomy, died, or were censored was to be presented.
  • active treatment period of Part 2 the time to feeding tube placement was to be analyzed similarly to time to death or tracheostomy. If an insufficient number of events occurred, only the tabulation of subjects who had feeding tube placement or who were censored was to be presented.
  • the SAP specified that the analysis of clinical status evaluation data would be conducted on the ITT population where the ITT population consisted of data from all subjects in the safety population for whom at least 1 post baseline clinical status evaluation (McGill SIS, ALSFRS-R, or VC) was obtained.
  • McGill SIS, ALSFRS-R, or VC clinical status evaluation
  • the analysis of time to death or tracheotomy was listed under clinical status evaluation data, which would imply that this analysis be carried out on the ITT population.
  • 1 subject in the 50 mg group died after 28 days of follow-up without an evaluation for McGill SIS, ALSFRS-R, or VC.
  • a total of 92 randomized subjects took at least 1 dose of study drug during the double-blind treatment period. Forty-eight (48) subjects were randomized to 50 mg dexpramipexole and 44 subjects were randomized to 300 mg dexpramipexole. Seventy-one (71) subjects completed the study through Week 28. Twenty-one (21) subjects, 14 subjects in the 50 mg group and 7 subjects in the 300 mg group, discontinued from the study prior to Week 28. The most common reasons for discontinuing early were ALS-related death (8 subjects) and withdrawal of consent (7 subjects).
  • the medications used at baseline of the placebo washout period were consistent with the age and ALS diagnosis of the population under study. At baseline, 96 (99%) subjects were receiving one or more medications. WHO drug classes used by >20.0% of subjects overall included Vitamins (64%), Other Nervous System Drugs (58%), Psychoanaleptics (40%), Antiinflammatory and Antirheumatic Products (31%), Other Alimentary Tract and Metabolism Products (31%), Antithrombotic Agents (27%), Analgesics (26%), Lipid Modifying Agents (24%), and
  • Each item of the ALSFRS-R was scored on a 4 to 0 scale, with a 4 indicating normal function and each lower number indicating progressive worsening of function. For change from baseline, therefore, a score of zero would indicate no loss of function and increasingly negative scores would indicate greater losses of function.
  • the ALSFRS-R total scores were similar in the 4 Part 1 treatment groups, with mean scores of 35.0, 32.4, 35.8, and 36.2 for the placebo, 50 mg, 150 mg, and 300 mg groups, respectively, and median scores ranging from 34 to 37.
  • the mean change from baseline in these groups was - 1.5 (placebo), -0.7 (50 mg), -1.0 (150 mg), and -1.5 (300 mg).
  • the mean baseline value was 34.9, and the mean and median changes from baseline to the end of the 4-week placebo washout were -1.2 and -0.5, respectively.
  • FIG. 13 provides a graphic presentation of the Kaplan-Meier estimates for the time to tracheostomy or death through Week 28.
  • the mean change from baseline in ALSFRS-R total scores was attenuated in the 300 mg group compared with the 50 mg group; the mean change was -6.5 in the 50 mg group and -6.2 in the 300 mg group.
  • the treatment group difference in mean change scores are a biased estimate of the true treatment group difference due to the larger number of deaths and dropouts in the 50 mg group than in the 300 mg group.
  • a more appropriate estimate of treatment group difference is provided by the slopes estimates as specified in the SAP.
  • the slope estimates of ALSFRS-R scores from the linear mixed effects model through Week 28 of the study were -1.283 for the 50 mg group and -1.021 for the 300 mg group.
  • the SAP specified as a sensitivity analysis a generalized Gehan Wilcoxon rank test based on a joint ranking of time to survival and change from baseline in ALSFRS-R score. Analysis of frequency and time to death was described by Kaplan-Meier life-table estimates of survival time, for which treatment group differences were analyzed by log rank test.
  • a joint-rank test of survival and ALSFRS-R data was conducted to compare the global clinical outcomes between the 2 treatment groups.
  • ANCOVA analysis of co variance
  • the covariates in the ANCOVA included baseline ALSFRS-R score, time from symptom onset, site of disease onset, and concomitant use of riluzole.
  • FIG. 16 shows plots of the mean rank of joint scores for the combined time to death and changes from baseline in ALSFRS-R total scores.
  • Imputing an ALSFRS-R score of zero for the first scheduled visit after the time of death is an alternative method for adjusting the linear mixed-effects slopes model for the impact of death outcomes. This method was not prespecified in the SAP but has been used by other ALS studies.
  • the CRF design for collection of vital capacity data required that both raw vital capacity data (measured VC) and calculated/derived vital capacity data (Predicted Normal, % Predicted, and % Variability) be manually recorded on the CRF.
  • the values for Predicted Normal, % Predicted and % Variability were electronically re-calculated and compared to those data entered by the site. This review revealed that much of the manually calculated/derived data recorded on the CRFs were not accurate and/or were not expressed to 1 decimal place, the format being used for the data analysis.
  • FIG. 19 provides a graphic presentation of the Kaplan-Meier estimates for the time to feeding tube placement. Time to need for assisted ventilation was not analyzed during Part 2; sites were asked whether NIV was initiated, not if NIV was necessary by an objective threshold.
  • correlation coefficients were calculated among the following variables: baseline upright VC, baseline supine VC, baseline difference between upright VC and supine VC, baseline ALSFRS-R total score, change from baseline to- Week 12 upright VC, change from baseline to Week 12 supine VC, change from baseline to Week 12 difference between upright VC and supine VC, change from baseline to Week 12 ALSFRS-R total score.
  • the primary analysis of ALSFRS-R data was a linear mixed-effects analysis of the treatment effect on the slope of ALSFRS-R total scores during the study.
  • the slope of ALSFRS-R scores through Week 28 was -1.283 for the 50 mg group and -1.021 for the 300 mg group, a 20.4% attenuation of the slope of decline in the high-dose group relative to the low-dose group.
  • the mean change from baseline to Week 28 in upright vital capacity was -12.4% in the 50 mg group and -15.1 % in the 300 mg group; median changes were -10.4% and - 11.5%, respectively.
  • the estimates of slope for vital capacity for the 50 mg and 300 mg groups through Week 28 were -2.452 and -3.067 (unadjusted), respectively, and -4.17 and -3.42 (adjusted for deaths through Week 28), respectively.
  • the 300 mg group slope was attenuated by 18% relative to the 50 mg group slope.
  • Duration of dosing is the last dose date minus the first dose date +1.
  • Mean daily dose is the total daily dose divided by the number of days dosed.
  • Treatment-related are adverse events with a possible, probable, or unknown relationship to study
  • Treatment-related are adverse events with a possible, probable, or unknown relationship to study medication.
  • AEs Frequently reported AEs (>10% of subjects overall) during the double-blind treatment period through Week 28 were fall (22 subjects, 24%), muscular weakness (20 subjects, 22%), constipation (19 subjects, 21%), salivary hypersecretion (13 subjects, 14%), depression (11 subjects, 12%), and dyspnea (11 subjects, 12%).
  • a summary of AEs reported by >5% of subjects overall (>5 subjects) in the double- blind treatment period through Week 28 is presented by preferred term in descending order of frequency in TABLE 12.
  • Treatment-related AEs were most commonly associated with Gastrointestinal Disorders and Nervous System Disorders.
  • the most common treatment-related AEs overall included constipation (5 subjects, 5%), headache (5 subjects, 5%), and dry mouth (4 subjects, 4%).
  • Gastrointestinal AEs were more common in the 300 mg group than in the 50 mg group.
  • ALS amyotrophic lateral sclerosis
  • TEAE treatment-emergent adverse event
  • ALS amyotrophic lateral sclerosis
  • TEAE treatment-emergent adverse event
  • AEs in both treatment groups were considered by the Investigator to be mild or moderate in intensity. Severe AEs reported for more than 1 subject included respiratory failure (5 subjects) and dyspnea (2 subjects). One or more severe AEs were reported for 12 (25%) subjects in the 50 mg group (acute myocardial infarction; ileus; fatigue; disease progression; sudden death; pneumonia bacterial; rib fracture; hypernatremia; muscular weakness; muscle contractions involuntary; dyspnea; respiratory failure [4 subjects] ; respiratory distress; pulmonary embolism) and 6 (14%) subjects in the 300 mg group (neutropenia; dry mouth; cholecystitis acute; pneumonia; fall; concussion; subdural haematoma; vital capacity decreased;
  • subjects who were not taking riluzole had a higher incidence of salivary hypersecretion (22% vs. 12%) and dysphagia (17% vs. 4%) than subjects who were taking riluzole.
  • subjects who were not taking riluzole had a higher incidence of dyspnea (27% vs. 7%), headache (27% vs. 2%), dry mouth (27% vs. 10%), and upper respiratory tract infection (13% vs. 3%).
  • subjects in the 300 mg group who were taking concomitant riluzole had a higher incidence of constipation (31% vs. 13%), nausea (10% vs.
  • FIG. 13 provides a graphic presentation of the Kaplan-Meier estimates for the time to tracheostomy or death through Week 28.
  • Dry mouth and insomnia occurred at a higher incidence in the 300 mg group (16% and 14%, respectively) than in the 50 mg group (2% and 0%, respectively), while muscular weakness and peripheral edema occurred at a higher incidence in the 50 mg group (27% and 15%, respectively) than in the 300 mg group (16% and 2%, respectively).
  • the overall incidence of AEs considered by the Investigator to be possibly or probably related to study drug was 31% in the 50 mg group and 41% in the 300 mg group.
  • the majority of AEs in both treatment groups were related to ALS (50 mg: 85%; 300 mg: 80%).
  • Dexpramipexole may be a useful neuroprotective agent in the treatment of chronic and acute neurodegenerative disorders, including ALS. This was the first clinical study of dexpramipexole in subjects with ALS. Eligible subjects were ⁇ 24 months from ALS symptom onset and met the clinically possible, clinically probable— laboratory-supported, clinically probable, or clinically definite El Escorial criteria. Part 1 of the current study evaluated the safety and tolerability of 3 dose levels of dexpramipexole (50 mg, 150 mg, and 300 mg given as 25 mg Q12H, 75 mg Q12H, and 150 mg Q12H, respectively) over 12 weeks of treatment in subjects with ALS.
  • Part 1 Subjects who completed Part 1 were eligible to enroll in Part 2 of the study. At the beginning of Part 2, all subjects participated in a single- blind, 4-week placebo washout and were observed for withdrawal effects. Following completion of the placebo washout period, subjects were re-randomized in a double-blind manner to low-dose (50 mg, administered as 25 mg Q 1 2H) or high-dose (300 mg, administered as 150 mg Q12H) dexpramipexole to receive treatment for up to 76 weeks. Dexpramipexole was safe and well tolerated in ALS subjects over 24 weeks of active treatment at total daily doses of 50 mg and 300 mg. The majority of deaths (17/21) were considered to be related to ALS.
  • the mean change from baseline to Week 28 in upright vital capacity was -12.4% in the 50 mg group and -15.1% in the 300 mg group; median changes were -10.4% and -11.5%, respectively.
  • the estimates of slope for vital capacity for the 30 mg and 300 mg groups over the double-blind treatment period through Week 28 were -2.452 and -3.067 (unadjusted), respectively, and -4.17 and -3.42 (adjusted for deaths through Week 28), respectively.
  • the 300 mg group slope was attenuated by 18% relative to the 50 mg group slope, demonstrating improvement in functional decline among subjects in the 300 mg group. No treatment effects on the McGill SIS scores were noted.
  • Dexpramipexole was rapidly absorbed, with T max ranging from 1.75 hours to 2.58 hours, t m ranging from 6.40 hours to 8.05 hours under fasted conditions, and was mostly eliminated in urine as unchanged parent drug (84-90% of dose). Food had no effect on the single-dose PK of dexpramipexole.
  • a total of 54 subjects (30 subjects in Study CLOOl and 24 subjects in Study CL002) were enrolled. Healthy, non-smoking, male and female subjects 30 to 60 years of age, inclusive, with normal or clinically acceptable physical examination and electrocardiogram (ECG) findings, systolic (90 to 140 mm Hg) and diastolic (50 to 90 mm Hg) blood pressure, and resting heart rate (50 to 100 bpm) who were willing to provide signed, written informed consent were eligible for enrollment.
  • ECG electrocardiogram
  • systolic 90 to 140 mm Hg
  • diastolic 50 to 90 mm Hg
  • resting heart rate 50 to 100 bpm
  • dexpramipexole (>99.95 enantiomeric purity) was supplied as neat drug substance (no excipients) in hard gelatin capsules.
  • Matching placebo capsules contained equivalent weights of microcrystalline cellulose.
  • Capsules were administered orally with water.
  • a purity adjustment factor of 1.06 was used to adjust for the water weight (monohydrate) in the salt form of the dexpramipexole drug substance.
  • Subjects in the fasted cohorts were required to fast overnight for a minimum of 10 hours before dose administration.
  • Subjects in the food cohort were required to fast overnight for a minimum of 10 hours before dose administration, with exception of the high fat high calorie meal that was administered 30 minutes prior to drug administration.
  • Panels of ascending doses were enrolled sequentially, with at least 96 hours and 72 hours separating the initiation of each panel in the single-dose and multiple-dose studies, respectively. All available safety data were reviewed under blinded conditions to monitor for serious safety or tolerability events prior to proceeding with dose escalations.
  • EDTA Vacutainer® via an indwelling peripheral intravenous cannula or by direct venipuncture.
  • the samples were centrifuged at 3000 rpm for 10 minutes at 4°C. After centrifugation, the plasma was divided into 2 aliquots of at least 1.5 mL each, placed into
  • polypropylene containers frozen, and stored at -20 °C until they were shipped for analysis.
  • Urine collected in each interval was well mixed, the pH was recorded, the total volume (or the weight and specific gravity) was recorded, and 2 aliquots of 20 mL each were collected into polypropylene containers and stored at -20°C until they were shipped for analysis. All plasma and urine sample were shipped frozen on dry ice in 2 separate shipments per group (1 set of aliquots per shipment) to Eurofins AvTech Laboratories Inc. (Kalamazoo, Ml) for bioanalytical analysis.
  • Plasma and urine concentrations were measured using validated liquid chromatography/ mass spectrometry/mass spectrometry (LC/MS/MS) methods.
  • the lower limits of quantitation for dexpramipexole were 20 ng/mL in plasma and 0.1 ⁇ g/mL in urine.
  • the inter- and intra- day coefficients of variation (CV) were 7% to 8% and 1% to 17%, respectively, for plasma.
  • the corresponding CVs for urine were 5% to 7% and 0% to 7%.
  • the lower limits of quantitation for dexpramipexole were 2 ng/mL in plasma and 0.1 pg/mL in urine.
  • the inter- and intra- day coefficients of variation (CV) were 5% to 11% and 1% to 8%, respectively, for plasma and 6% to
  • the analytical procedure for analysis of plasma samples used a 100 1 AL aliquot of K2EDTA human plasma.
  • the plasma sample was spiked with 20 1 AL of working internal standard solution and 20 1AL of type 1 water for subject samples and QCs and 20 pL of the appropriate intermediate standard solution for standards.
  • One hundred microliters (100 ⁇ ⁇ ) of 50% ammonium hydroxide solution was added to the sample followed by vortex mixing.
  • One milliliter (1 mL) of tertbutyl methyl ether was then added and the sample was vortexed to extract the analyte and internal standard into the organic layer, followed by separation using flash freezing.
  • the organic layer was decanted, evaporated to dryness, and the sample was reconstituted with 0.5 mL of reconstitution solution (0.1 % ammonium hydroxide in 50:50 methanol; type 1 water (v/v/v)).
  • a 10 ⁇ ⁇ aliquot of this reconstituted sample was injected into an LC/MS/MS system for analysis.
  • the MS/MS transitions monitored were 212.1 m/z to 153.1 m/z for dexpramipexole and 219.2 m/z to 111.2 m/z for the internal standard, D7-pramipexole.
  • the calibration curve was linear between 2 and 2,000 ng/mL for dexpramipexole using a weighted (1/concentration) linear regression of the standard curve.
  • the analytical procedure for analysis of urine samples was essentially similar to the plasma procedure.
  • PK parameters were estimated from individual plasma and concentration data using non-compartmental analysis: maximum plasma concentration (C max ), time to Cmax (T max ), area under the curve from time zero to the final time with a concentration above the limit of quantitation (AUC 0 -i a st ), area under the curve from zero to infinity (AUQo-mf)), area under the curve over the dosing interval on Day 7 (AUQ0-12)) for the multiple-dose study, elimination rate constant ( ⁇ ), half-life (t m ), amount excreted in the urine (Ue), fraction excreted unchanged in urine (Fe), renal clearance (Cl r ), oral clearance (CL/F), and oral volume of distribution (Vz/F). Plasma concentrations, urinary excretions, and PK parameters were summarized by dose level using descriptive statistics.
  • Dexpramipexole administered as a highly chirally pure drug substance, is a promising novel amino-benzothiazole that is being developed for the treatment of ALS.
  • Preclinical studies have shown that dexpramipexole and its enantiomer pramipexole are equally neuroprotective, but, unlike pramipexole, dexpramipexole is not a clinically relevant dopamine agonist, and therefore may be dosed at much higher levels that may optimize its neuroprotective properties in the absence of dose-limiting side effects.
  • Dexpramipexole was well absorbed after oral administration, with maximum concentrations observed 2 hours after dosing. Dexpramipexole demonstrated linear PK over the range of doses studied and was nearly completely eliminated in the urine as unchanged parent drug (84-90% of dose). Single- dose absorption was not affected by administration of a high fat/high calorie meal.
  • dexpramipexole at the doses examined, lacks clinically relevant dopaminergic activity, in marked contrast to its enantiomer, pramipexole.
  • the highest unit dose of dexpramipexole administered in these studies (300 mg) was 2400-fold higher than the recommended safe starting unit dose of pramipexole (0.125 mg) and 67-fold higher than the maximum recommended daily dose (4.5 mg/day) of pramipexole in Parkinson's disease patients, a dose of pramipexole which may only be reached following a seven-week period of gradual dose titration.
  • Example 5 is a randomized, blinded, placebo-controlled, ascending dose study of the safety and pharmacokinetics of dexpramipexole in healthy volunteers.
  • each cohort will have 9 unique subjects (with at least 4 females per cohort) randomized to dexpramipexole:placebo in a 7:2 ratio.
  • Proposed doses of dexpramipexole in Part A (SAD) are 300, 450, and 600 mg, and for Part B (MAD) are 225 and 300 mg administered orally twice daily (every 12 hours [Q12h]) over 31 ⁇ 2 days for a total of 7 doses.
  • Subjects will receive a single oral dose of dexpramipexole (300, 450 or 600 mg) or placebo on Day 1.Within each SAD cohort, dosing will be staggered into 3 sub-cohorts of 3 subjects each. The first sub-cohort (2 subjects receiving dexpramipexole and 1 subject receiving placebo) will be dosed on the first day. Dosing of subsequent sub-cohorts will commence no earlier than 48 hours (+1 hour) after the final subject has been dosed in the previous sub-cohort and there are no significant safety concerns.
  • Safety evaluations will include assessment of adverse events (AEs), clinical laboratory evaluations, measurement of vital signs, standard paper 12-lead ECGs, and physical examinations. Additionally, 24-hour Holter ECG assessment will be performed on Day -1 and Day 1. Serial blood samples and pooled urine samples will be collected for pharmacokinetic (PK) assessments prior to dosing and for up to 72 hours post-dose. Subjects will be discharged on Day 4 (72 h post-dose) and will return to the clinical unit for a final follow-up visit on Day 8.
  • PK pharmacokinetic
  • Subjects will receive twice daily oral doses (Q12 h) of dexpramipexole (225 or 300 mg) or placebo on Days 1 to 3 and a single dose on the morning of Day 4.
  • Safety evaluations will include assessment of adverse events (AEs), clinical laboratory evaluations, measurement of vital signs, standard paper 12-lead ECGs, and physical examinations. Additionally, 24-hour Holter ECG assessment will be performed on Day -1 and Day 4. Serial blood samples will be collected for PK assessments prior to dosing and up to 12 hours post-dose following the first dose and up to 72 hours post-dose following the final dose. Pooled urine samples will also be collected over the 12-hour dosing interval following the final dose on Day 4.
  • Dexpramipexole will be administered orally as a round compressed oral tablet (75 mg) and as a compressed film-coated tablet (150 mg).
  • Placebo matched to dexpramipexole will be administered orally as matching tablets. Duration of Treatment:
  • Part A subjects will receive a single dose of dexpramipexole or placebo on the morning of Day 1.
  • Part B subjects will receive twice daily doses (at 12 hour intervals) of dexpramipexole or placebo on Day 1 to Day 3 and a single dose on the morning of Day 4.
  • Blood samples for determination of concentrations of dexpramipexole in plasma will be obtained in the SAD cohorts at pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 24, 36, 48, and 72 hours after dosing, and in the MAD cohorts at pre-dose and 0.5, 1, 2, 3, 4, 6, 8, and 12 hours post morning dose on Day 1, prior to the morning and evening doses on Day 2 and Day 3, and at pre-dose and 0.5, 1, 2, 3, 4, 6, 8, 12, 16, 24, 36, 48, and 72 hours post morning dose on Day 4.
  • Blood samples for determination of (S)-pramipexole in plasma will also be collected at pre-dose and 2 hours post-dose on Day 1 in the SAD and MAD cohorts and at pre-dose and 2 and 12 hours post-dose on Day 4 in the MAD cohorts.
  • Urine for determination of concentrations of dexpramipexole will be collected in the SAD cohorts at pre-dose and pooled across intervals (as available) at 0 to 2, 2 to 4, 4 to 8, 8 to 16, 16 to 24, 24 to 36, 36 to 48, and 48 to 72 hours after dosing, and in the MAD cohorts at pre-dose on Day 1, and pre-dose and pooled across intervals (as available) at 0 to 2, 2 to 4, 4 to 8, and 8 to 12 hours after the morning dose on Day 4.
  • Cmax time to maximum plasma concentration
  • T max minimum plasma concentration
  • T ⁇ time to minimum plasma concentration
  • V z /F average plasma concentration
  • AI accumulation index
  • Dexpramipexole plasma concentrations and urine amounts, as well as the PK parameters will be listed and summarized by study part (SAD or MAD), treatment group and/or study day using descriptive statistics, including arithmetic mean, standard deviation, coefficient of variation, median, minimum, maximum, and, where applicable, geometric mean and geometric coefficient of variation.
  • Plasma concentration graphs showing individual and arithmetic mean concentration curves by study part, treatment group and/or study day will be included.
  • Dexpramipexole exposure will be compared across study parts, treatment groups and/or study days graphically. Additionally, power models and analysis of variance models will be fitted to the log- transformed PK parameters (C max , AUQo-iast), AUQo-mt), and AUQ 0 - 12) , as appropriate) to evaluate dose proportionality and accumulation. Geometric least-squares means with corresponding 95% confidence interval (CI) and ratios of geometric least squares means with corresponding 90% CI will be presented for each comparison, as appropriate. Achievement of steady-state upon repeated dosing in the MAD cohorts will be evaluated graphically and using a repeated-measures linear-mixed model on the log- transformed trough (pre-dose) concentration data.
  • (S)-pramipexole concentrations in plasma at each sampling time will be listed and summarized by study part (SAD or MAD), treatment group and/or study day using descriptive statistics, including arithmetic mean, standard deviation, coefficient of variation, median, minimum, and maximum.
  • the (S)-pramipexole to dexpramipexole concentration ratio will be calculated at common sampling times and summarized by study part, treatment group and/or study day.
  • allelic frequency of each gene of interest will be calculated in all subjects who consented to participate in the PG evaluation.
  • dexpramipexole PK parameters C max Dose, AUQo-iast/Dose, AUQo-mf/Dose, and AUC ( o-i 2) /Dose, t 1 ⁇ 2 , CL/F, CL r , and V z /F
  • Geometric least- squares means with corresponding 95% CI and ratios of geometric least squares means with corresponding 90% CI will be presented for each comparison, as appropriate.

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des compositions pharmaceutiques de dexpramipexole ou un sel pharmaceutique de celui-ci et des procédés d'utilisation de telles compositions pour le traitement de SLA.
PCT/US2012/071426 2011-12-22 2012-12-21 Compositions et procédés pour traiter la sclérose latérale amyotrophique WO2013096870A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161579542P 2011-12-22 2011-12-22
US61/579,542 2011-12-22

Publications (1)

Publication Number Publication Date
WO2013096870A1 true WO2013096870A1 (fr) 2013-06-27

Family

ID=47522960

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/071426 WO2013096870A1 (fr) 2011-12-22 2012-12-21 Compositions et procédés pour traiter la sclérose latérale amyotrophique

Country Status (1)

Country Link
WO (1) WO2013096870A1 (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015003030A1 (fr) * 2013-07-02 2015-01-08 Cortice Biosciences, Inc. Méthode de traitement des maladies neurodégénératives
WO2015023786A1 (fr) 2013-08-13 2015-02-19 Knopp Biosciences Llc Compositions et méthodes de traitement de troubles des cellules plasmatiques et de troubles prolymphocytaires à cellules b
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US9512096B2 (en) 2011-12-22 2016-12-06 Knopp Biosciences, LLP Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
US9662313B2 (en) 2013-02-28 2017-05-30 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
WO2017117158A1 (fr) * 2015-12-28 2017-07-06 Steven Hoffman Méthodes de traitement de la sclérose latérale amyotrophique et de ses symptômes
US9763918B2 (en) 2013-08-13 2017-09-19 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
US9849116B2 (en) 2008-08-19 2017-12-26 Knopp Biosciences Llc Compositions and methods of using (R)-pramipexole
US10179774B2 (en) 2007-03-14 2019-01-15 Knopp Biosciences Llc Synthesis of chirally purified substituted benzothiazole diamines
US10383857B2 (en) 2013-07-12 2019-08-20 Knopp Biosciences Llc Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
US11534420B2 (en) 2019-05-14 2022-12-27 Tyme, Inc. Compositions and methods for treating cancer
US11607418B2 (en) 2020-05-14 2023-03-21 Tyme, Inc. Methods of treating SARS-CoV-2 infections

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4923508A (en) 1989-05-08 1990-05-08 Howmet Corporation Segmented induction skull melting crucible and method
WO2008113003A1 (fr) * 2007-03-14 2008-09-18 Knopp Neurosciences, Inc. Formulations à libération modifiée de (6r)-4,5,6,7-tétrahydro-n6-propyl-2,6-benzothiazole-diamine et procédés d'utilisation de celles-ci
WO2010022140A1 (fr) * 2008-08-19 2010-02-25 Knopp Neurosciences, Inc. Compositions et procédés employant du (r)-pramipexole
WO2010148409A1 (fr) * 2009-06-19 2010-12-23 Knopp Neurosciences, Inc. Compositions et méthodes pour traiter la sclérose latérale amyotrophique (als)
US9009408B2 (en) 2010-09-28 2015-04-14 Texas Instruments Incorporated Non-blocking, pipelined write allocates with allocate data merging in a multi-level cache system
US11368008B2 (en) 2019-06-20 2022-06-21 Nexpo Co., Ltd. Arc protection system and method to control arc protection system

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4923508A (en) 1989-05-08 1990-05-08 Howmet Corporation Segmented induction skull melting crucible and method
WO2008113003A1 (fr) * 2007-03-14 2008-09-18 Knopp Neurosciences, Inc. Formulations à libération modifiée de (6r)-4,5,6,7-tétrahydro-n6-propyl-2,6-benzothiazole-diamine et procédés d'utilisation de celles-ci
WO2010022140A1 (fr) * 2008-08-19 2010-02-25 Knopp Neurosciences, Inc. Compositions et procédés employant du (r)-pramipexole
WO2010148409A1 (fr) * 2009-06-19 2010-12-23 Knopp Neurosciences, Inc. Compositions et méthodes pour traiter la sclérose latérale amyotrophique (als)
US9009408B2 (en) 2010-09-28 2015-04-14 Texas Instruments Incorporated Non-blocking, pipelined write allocates with allocate data merging in a multi-level cache system
US11368008B2 (en) 2019-06-20 2022-06-21 Nexpo Co., Ltd. Arc protection system and method to control arc protection system

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BANKER; RHODES: "Modern Pharmaceutics", 1979, MARCEL DEKKER, INC
BERGE ET AL., J. PHARM. SCIENCES, vol. 6, 1977, pages 1 - 19
BOZIK MICHAEL E ET AL: "Safety, Tolerability, and Pharmacokinetics of KNS-760704 (Dexpramipexole) in Healthy Adult Subjects", JOURNAL OF CLINICAL PHARMACOLOGY, vol. 51, no. 8, August 2011 (2011-08-01), pages 1177 - 1185, XP009166998 *
GOODMAN; GILMAN: "The Pharmaceutical Basis of Therapeutics, 6th Edition,", 1980, MACMILLAN PUBLISHING CO.
MERIT CUDKOWICZ ET AL: "The effects of dexpramipexole (KNS-760704) in individuals with amyotrophic lateral sclerosis", NATURE MEDICINE, vol. 17, no. 12, 20 November 2011 (2011-11-20), pages 1652 - 1656, XP055052890, ISSN: 1078-8956, DOI: 10.1038/nm.2579 *
VALENTIN K. GRIBKOFF ET AL: "KNS-760704 [(6R)-4,5,6,7-tetrahydro-N6-propyl-2, 6-benzothiazole-diamine dihydrochloride monohydrate] for the Treatment of Amyotrophic Lateral Sclerosis", CNS NEUROSCIENCE & THERAPEUTICS, vol. 14, no. 3, 1 September 2008 (2008-09-01), pages 215 - 226, XP055052891, ISSN: 1755-5930, DOI: 10.1111/j.1755-5949.2008.00048.x *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10179774B2 (en) 2007-03-14 2019-01-15 Knopp Biosciences Llc Synthesis of chirally purified substituted benzothiazole diamines
US9849116B2 (en) 2008-08-19 2017-12-26 Knopp Biosciences Llc Compositions and methods of using (R)-pramipexole
US10208003B2 (en) 2011-12-22 2019-02-19 Knopp Biosciences Llc Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
US9512096B2 (en) 2011-12-22 2016-12-06 Knopp Biosciences, LLP Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
US9662313B2 (en) 2013-02-28 2017-05-30 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US9956206B2 (en) 2013-02-28 2018-05-01 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US10285981B2 (en) 2013-02-28 2019-05-14 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US11033529B2 (en) 2013-07-02 2021-06-15 Cortice Biosciences, Inc. Method of treating neurodegenerative disorders
WO2015003030A1 (fr) * 2013-07-02 2015-01-08 Cortice Biosciences, Inc. Méthode de traitement des maladies neurodégénératives
US10828284B2 (en) 2013-07-12 2020-11-10 Knopp Biosciences Llc Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
US10383857B2 (en) 2013-07-12 2019-08-20 Knopp Biosciences Llc Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
US10980783B2 (en) 2013-07-12 2021-04-20 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US10383856B2 (en) 2013-07-12 2019-08-20 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US11026928B2 (en) 2013-07-12 2021-06-08 Knopp Biosciences Llc Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US11612589B2 (en) 2013-07-12 2023-03-28 Areteia Therapeutics, Inc. Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
US10195183B2 (en) 2013-08-13 2019-02-05 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
AU2017276231B2 (en) * 2013-08-13 2019-05-16 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and b-cell prolymphocytic disorders
US10028940B2 (en) 2013-08-13 2018-07-24 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
AU2014306683B2 (en) * 2013-08-13 2017-10-12 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
US10456381B2 (en) 2013-08-13 2019-10-29 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
US9763918B2 (en) 2013-08-13 2017-09-19 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
US9642840B2 (en) 2013-08-13 2017-05-09 Knopp Biosciences, Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
US20160193187A1 (en) * 2013-08-13 2016-07-07 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and b-cell prolymphocytic disorders
WO2015023786A1 (fr) 2013-08-13 2015-02-19 Knopp Biosciences Llc Compositions et méthodes de traitement de troubles des cellules plasmatiques et de troubles prolymphocytaires à cellules b
WO2017117158A1 (fr) * 2015-12-28 2017-07-06 Steven Hoffman Méthodes de traitement de la sclérose latérale amyotrophique et de ses symptômes
US11534420B2 (en) 2019-05-14 2022-12-27 Tyme, Inc. Compositions and methods for treating cancer
US11607418B2 (en) 2020-05-14 2023-03-21 Tyme, Inc. Methods of treating SARS-CoV-2 infections

Similar Documents

Publication Publication Date Title
US20130245081A1 (en) Compositions and Methods for Treating Amyotrophic Lateral Sclerosis
WO2013096870A1 (fr) Compositions et procédés pour traiter la sclérose latérale amyotrophique
US8445474B2 (en) Compositions of R(+) and S(−) pramipexole and methods of using the same
JP4846063B2 (ja) 選択的s1p1レセプターアゴニストの投与法
JP6244497B1 (ja) 選択的s1p1レセプターアゴニストの投与法
US10285981B2 (en) Compositions and methods for treating amyotrophic lateral sclerosis in responders
EP2465500A1 (fr) Quantités thérapeutiquement efficaces de R(+) et S(-) pramipexole pour une utilisation dans le traitement de la maladie de Parkinson
JP2010521493A (ja) (6r)−4,5,6,7−テトラヒドロ−n6−プロピル−2,6−ベンゾチアゾール−ジアミンの放出調節製剤およびそれらの使用方法
WO2014134569A1 (fr) Compositions et procédés de traitement de la sclérose latérale amyotrophique chez les patients répondant au traitement
WO2004054574A1 (fr) Medicament solide administre par voie orale
JP6499634B2 (ja) 過活動膀胱の治療のためのソリフェナシンと唾液分泌刺激剤の組合せ
TW202108585A (zh) 用於治療地中海型貧血之pde9抑制劑
JP2010501626A (ja) 統合失調症を処置するためのビフェプルノックス用量
US20230338349A1 (en) Low dose regimen and formulation of a 5-methyl-1,2,4-oxadiazol-3-yl compound
KR102512518B1 (ko) 페마피브레이트를 함유하는 의약
CN113950328A (zh) 用于治疗亨廷顿病及其症状的包含普利多匹定及其类似物的组合物
JP2014513689A (ja) 過活動膀胱の治療のためのトロスピウムと唾液分泌刺激剤の組合せ
KR100709531B1 (ko) 당뇨병성 합병증의 예방 또는 진전저지용 의약조성물
JPWO2004096230A1 (ja) 関節リウマチ治療剤
KR20070011329A (ko) 인슐린 저항성 개선제를 함유하는 당뇨병 치료제
EP2961404A1 (fr) Compositions et procédés de traitement de la sclérose latérale amyotrophique chez les patients répondant au traitement

Legal Events

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

Ref document number: 12813231

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12813231

Country of ref document: EP

Kind code of ref document: A1