US20140100372A1 - Synthesis of chirally purified substituted benzothiazole diamines - Google Patents

Synthesis of chirally purified substituted benzothiazole diamines Download PDF

Info

Publication number
US20140100372A1
US20140100372A1 US13/973,987 US201313973987A US2014100372A1 US 20140100372 A1 US20140100372 A1 US 20140100372A1 US 201313973987 A US201313973987 A US 201313973987A US 2014100372 A1 US2014100372 A1 US 2014100372A1
Authority
US
United States
Prior art keywords
tetrahydro
benzothiazole
propyl
propylamino
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/973,987
Inventor
Prasad Raje
Rajendrakumar Reddy Gadikota
Jian-Xie Chen
Olga V. Lapina
John M. McCall
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Knopp Biosciences LLC
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
Priority to US13/973,987 priority Critical patent/US20140100372A1/en
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KNOPP NEUROSCIENCES INC.
Publication of US20140100372A1 publication Critical patent/US20140100372A1/en
Assigned to KOPPER, RACHEL reassignment KOPPER, RACHEL SECURITY INTEREST Assignors: KNOPP BIOSCIENCES LLC
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GADIKOTA, RAJENDRAKUMAR REDDY
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RAJE, PRASAD
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, JIAN-XIE
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LAPINA, OLGA V.
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCCALL, JOHN M.
Priority to US15/369,230 priority patent/US10179774B2/en
Assigned to KNOPP BIOSCIENCES LLC reassignment KNOPP BIOSCIENCES LLC RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: KOPPER, RACHEL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms

Definitions

  • the compound 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is a synthetic aminobenzothiazole derivative whose (6S) enantiomer, commonly known as pramipexole and commercially available under the Mirapex® name, is a potent dopamine agonist, and thus, 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.
  • compositions of this disclosure may also be derived at least in part from the ability of the (6R) enantiomer of pramipexole to prevent neural cell death by at least one of three mechanisms.
  • the (6R) enantiomer of pramipexole may be capable of reducing the formation of reactive oxygen species in cells with impaired mitochondrial energy production.
  • the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for an (6S)-entantiomer and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is chirally purified for an (6S) entantiomer
  • the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole is (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine.
  • the step of reacting may be carried out for up to about 12 hours.
  • the step of recovering may include one or more steps selected from, but not limited to, filtering the reaction mixture to isolate a precipitate, washing a precipitate, and drying a precipitate, and the process of certain embodiments may include the step of cooling the reaction mixture to a temperature of about 25° C. after the step of reacting.
  • the propyl sulfonate may be selected from n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof, and in other embodiments, the propyl halide is selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • reaction would be expected to yield a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine that is about 60:40 R to S.
  • the temperature of reactions of such embodiments may; generally, be below the boiling temperature of the reaction mixture, more specifically, below the boiling temperature of the solvent(s) of the reaction mixture.
  • an elevated temperature may be lower than about 125° C.
  • an elevated temperature may be lower than about 100° C., and in yet another embodiment lower than about 95° C., and in still others less than about 75° C. Therefore, the reaction temperature may range from about 50° C. to about 125° C. in some embodiments, about 55° C. to about 100° C. in other embodiments, about 60° C. to about 95° C. in still other embodiments, about 60° C. to about 75° C. in yet other embodiments and in certain embodiments, from about 55° C. to about 65° C.
  • Insoluble 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be separated from the reaction solution by any method known in the art.
  • the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be collected by simple filtering. There are numerous methods for filtering a solid from a solution, and any such method may by useful in embodiments of the invention.
  • insoluble 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be isolated by centrifugation. Again, such methods are well known in the art and any such method may be used in various embodiments of the invention.

Abstract

Methods for preparing chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamines such as, for example, (6R)2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole and purifying a dominant enantiomer of substituted 4,5,6,7-tetrahydro-benzothiazole diamines from entantiomerically enriched mixtures of substituted 4,5,6,7-tetrahydro-benzothiazole diamines are provided herein.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 12/049,235, filed Mar. 14, 2008, which claims priority to U.S. Provisional Appln. No. 60/894,829, filed Mar. 14, 2007 and U.S. Provisional Appln. No. 60/894,814, filed Mar. 14, 2007, all of which are incorporated herein by reference in their entireties.
  • GOVERNMENT INTERESTS
  • Not applicable
  • PARTIES TO A JOINT RESEARCH AGREEMENT
  • Not applicable
  • INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ON A COMPACT DISC
  • Not applicable
  • BACKGROUND
  • 1. Field of Invention
  • Not applicable
  • 2. Description of Related Art
  • The compound 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is a synthetic aminobenzothiazole derivative whose (6S) enantiomer, commonly known as pramipexole and commercially available under the Mirapex® name, is a potent dopamine agonist, and thus, 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. Therefore, pramipexole may have utility as an inhibitor of the cell death cascades and loss of cell viability observed in neurodegenerative diseases and is indicated for treating Parkinson's disease, cluster headaches, restless legs syndrome and bipolar disorder with only small daily doses required and tolerated by patients activates dopamine receptors. Additionally, oxidative stress may be caused by an increase in oxygen and other free radicals, and has been associated with the fatal neurodegenerative disorder amyotrophic lateral sclerosis (ALS). ALS is a progressive neurodegenerative disorder involving the motor neurons of the cortex, brain stem, and spinal cord. About 10% of all ALS patients are familial cases, of which 20% have mutations in the superoxide dismutase 1 (SOD-1) gene. The SOD-1 enzyme may play a pivotal role in the pathogenesis and progression of familial amyotrophic lateral sclerosis (FALS). Recent studies also link the premature neuronal death associated with ALS to mutated mitochondrial genes which lead to abnormalities in functioning of the energy production pathways in mitochondria.
  • The neuroprotectant activity of both enantiomers of pramipexole have typical therapeutic doses expected to be in the range of about 10 mg/day to about 1,500 mg/day. However, the pramipexole's agonistic effect on of the D2 family of dopamine receptors only requires therapeutic doses that range between 0.5 and 5.0 mg/day, and even these relatively low doses adverse side effects have been reported. For example, 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. In rodents, death occurred at doses of 70-105 mg/kg and above which is equivalent to a human dose of 7-12 mg/kg/ or approximately 500-850 mg for a 70 kg (−150 lb) individual. In dogs, vomiting occurred at 0.0007 mg/kg and above, while monkeys displayed major excitation at 3.5 mg/kg. In human subjects, an initial single dose of pramipexole of greater than 0.20 mg was not tolerated. All species showed signs of toxicity related to exaggerated pharmacodynamic responses to the pramipexole related to dopaminergic agonism.
  • Thus, a clinical use of pramipexole as a mitochondria-targeted antioxidant 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. In contrast, (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine is an effective mitochondria-targeted neuroprotectant that exhibits excellent anti-oxidative properties when administered without adverse side effects. Thus, higher doses of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine 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 the compositions of this disclosure may also be derived at least in part from the ability of the (6R) enantiomer of pramipexole to prevent neural cell death by at least one of three mechanisms. First, the (6R) enantiomer of pramipexole may be capable of reducing the formation of reactive oxygen species in cells with impaired mitochondrial energy production. Second, the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may partially restore the reduced mitochondrial membrane potential that is correlated with Alzheimer's, Parkinson's, Huntington's and amyotrophic lateral sclerosis diseases. Third, the (6R) enantiomer of pramipexole may block 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 (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine required to elicit these neuroprotective effects generally require highly pure preparations of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine which take into account the upper limit of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine contamination (0.5 mg to 5.0 mg).
  • Processes for the preparation of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole using a simple alkylation reaction were first described in U.S. Pat. Nos. 4,843,086 and 4,886,812. Other preparations of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole have been described that involve reductive amination rather than simple alkylation and, thus, produce a mixture of the R and S optical isomers with no direct means for further purification of the optical isomers. These known processes for the production of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole are expensive, labor intensive, and utilize hydride reducing agents that pose safety risks. Furthermore, there are currently no known processes for the direct synthesis of the pure (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine from a diamine. Therefore, the R isomer must be synthesized as a mixture of the optical isomers that is purified by expensive and time consuming methods that may utilize other problematic substances. Moreover, known processes involving deamination result in a loss of enantiomeric purity, and methods useful for resolution of the optical isomers from mixtures fall short of producing chirally and chemically pure preparations of the R enantiomer or the S enantiomer.
  • BRIEF SUMMARY OF THE INVENTION
  • Embodiments of the invention presented herein are directed to a process for preparing a chirally purified substituted 4,5,6,7,-tetrahydro-benzothaizole diamine including the steps of: heating a solution comprising entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine of general formula (I):
  • Figure US20140100372A1-20140410-C00001
  • wherein:
  • R1 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkenyl or alkynyl group each having 3 to 6 carbon atoms, an alkanoyl group having 1 to 6 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the above-mentioned phenyl nuclei may be substituted by 1 or 2 halogen atoms;
  • R2 represents a hydrogen atom or an alkyl group with 1 to 4 carbon atoms;
  • R3 represents a hydrogen atom, an alkyl group with 1 to 7 carbon atoms, a cycloalkyl group having 3 to 7 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms, an alkanoyl group having 1 to 7 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the phenyl nucleus may be substituted by fluorine, chlorine or bromine atoms,
  • R4 represents a hydrogen atom, an alkyl group with 1 to 4 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms; and
  • at least one or R1, R2, R3 or R4 is a hydrogen in an organic solvent; and an alkyl sulfonate or an alkyl halide in a solvent to form a reaction mixture reacting the reaction mixture; and recovering a chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine.
  • In various embodiments, the alkyl sulfonate may be of general formula (11):
  • Figure US20140100372A1-20140410-C00002
  • wherein:
  • R′ is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl or phenyl alkyl; and
  • Z is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl or phenyl alkyl. In particular embodiments, X may be a propyl and, in some embodiments, the alkyl sulfonate may be a propyl sulfonate selected from n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof. In other embodiments, the alkyl halide may be of general formula (12):

  • R′—X  (12)
  • wherein:
  • R′ is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl or phenyl alkyl; and
  • X is any halide including, for example, fluorine, chlorine, bromine or iodide. In particular embodiments, X may be a propyl, and in some embodiments, the alkyl halide may be a propyl halide selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • The chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine of various embodiments may be at least greater than about 97% chirally pure. In some embodiments, the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine may be at least greater than about 99% chirally pure, and in other embodiments, chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is at least about 99.9% chirally pure.
  • The chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine of various embodiments may be greater than about 99%. In some embodiments, the chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine may be greater than about 99.9%, and in other embodiments, the chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine is greater than about 99.99%.
  • In some embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for an (6R) entantiomer and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is chirally purified for an (6R) entantiomer, and in particular embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole may be (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In other embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for an (6S)-entantiomer and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is chirally purified for an (6S) entantiomer, and in certain embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be entantiomerically enriched for (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole is (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In still other embodiments, the entantiomerically enriched 4,5,6,7-tetrahydrobenzothiazole diamine may be a ratio of greater than about 1:4 (6R)-entantiomer to (6S)-entantiomer to about 4:1 (6R)-entantiomer to (6S)-entantiomer.
  • The solvent of various embodiments may be selected from an organic solvent and an organic solvent mixed with water, and in some embodiments, the solvent may selected from, but not limited to, ethanol, 1-propanol, n-butanol, dihydrofuran, dimethylformamide, dimethyl, dimethylacetamide, hexamethylphosphoric triamide or mixtures or hydrates thereof.
  • In some embodiments, the steps of heating, reacting and recovering each independently include stirring. In other embodiments, the steps of heating and reacting may each independently be carried out at a temperature of from about 50° C. to about 125° C. In yet other embodiments, the step of heating may further include adding the alkyl sulfonate or alkyl halide to the heated 4,5,6,7-tetrahydro-benzothiazole diamine. In still other embodiments, the step of adding may be carried out for about 0.5 hours to about 2 hours, and in certain embodiments, about 1.0 to about 2.0 molar equivalents of the alkly sulfonate or alkyl halide may be added. In particular embodiments, the step of reacting may be carried out for up to about 12 hours. In yet other embodiments, the step of recovering may include one or more steps selected from, but not limited to, filtering the reaction mixture to isolate a precipitate, washing a precipitate, and drying a precipitate, and the process of certain embodiments may include the step of cooling the reaction mixture to a temperature of about 25° C. after the step of reacting.
  • Other embodiments of the invention include a process for preparing a chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine including the steps of heating a solution comprising an entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine of general formula (1):
  • Figure US20140100372A1-20140410-C00003
  • wherein:
      • R1 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkenyl or alkynyl group each having 3 to 6 carbon atoms, an alkanoyl group having 1 to 6 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the above-mentioned phenyl nuclei may be substituted by 1 or 2 halogen atoms;
      • R2 represents a hydrogen atom or an alkyl group with 1 to 4 carbon atoms;
      • R3 represents a hydrogen atom, an alkyl group with 1 to 7 carbon atoms, a cycloalkyl group having 3 to 7 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms, an alkanoyl group having 1 to 7 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the phenyl nucleus may be substituted by fluorine, chlorine or bromine atoms,
      • R4 represents a hydrogen atom, an alkyl group with 1 to 4 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms; and
      • at least one or R1, R2, R3 or R4 is a hydrogen in an organic solvent;
  • adding to the heated solution propyl sulfonate or a propyl halide to form a reaction mixture; and
  • reacting the reaction mixture.
  • In some embodiments, the propyl sulfonate may be selected from n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof, and in other embodiments, the propyl halide is selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • The chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine of various embodiments may be at least greater than about 97% chirally pure. In some embodiments, the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is at least greater than about 99% chirally pure, and in other embodiments, the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine is at least about 99.9% chirally pure.
  • The chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine of various embodiments may be greater than about 99%. In some embodiments, the chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine is greater than about 99.9%, and in other embodiments, the chemical purity of the substituted 4,5,6,7-tetrahydro-benzothiazole diamine is greater than about 99.99%.
  • In certain embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole may be enriched for (6R)4,5,6,7-tetrahydro-benzothiazole diamine and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine may be (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In certain other embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may be enriched for (6S) 4,5,6,7-tetrahydro-benzothiazole diamine and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine may be (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In some embodiments, the entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine may include a mixture of (6R)4,5,6,7-tetrahydro-benzothiazole diamine and (6S)4,5,6,7-tetrahydro-benzothiazole diamine and the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine may include a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In particular embodiments, the mixture may be a racemic mixture. In other embodiments, the enantiomerically enriched mixture may be a ratio of greater than about 1:4 (6R)4,5,6,7-tetrahydro-benzothiazole diamine and (6S)4,5,6,7-tetrahydro-benzothiazole diamine to about 4:1 (6R)4,5,6,7-tetrahydro-benzothiazole diamine and (6S)4,5,6,7-tetrahydro-benzothiazole diamine.
  • In some embodiments, the solvent may be selected from a polar or organic solvent and a polar or organic solvent mixed with water, and in certain embodiments, the solvent may be selected from ethanol, 1-propanol, n-butanol, dihydrofuran, dimethylformamide, dimethyl, dimethylacetamide, hexamethylphosphoric triamide or mixtures or hydrates thereof. In other embodiments, the steps of heating and reacting each independently may include stirring. In still other embodiments, the steps of heating, adding and reacting may each independently be carried out at a temperature of from about 50° C. to about 125° C. In yet other embodiments, the process may further include the step of cooling the reaction mixture to a temperature of about 25° C. after the step of reacting. In particular embodiments, the step of adding may be carried out for up to about 2 hours. Various embodiments may further include the step of recovering the chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine, and in some embodiments, recovering may include one or more steps selected from filtering the mixture to isolate a precipitate, washing a precipitate, and drying a precipitate. In particular embodiments, about 1.0 to about 2.0 molar equivalents of the propyl sulfonate or propyl halide may be added.
  • Various embodiments of the invention also include chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process including the steps of: heating a solution comprising entantiomerically enriched 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and a propyl halide or a propyl sulfonate to form a reaction mixture; reacting the reaction mixture; and recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. In some embodiments, the propyl sulfonate may be selected, from, but not limited to, n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof, and in other embodiments, the propyl halide may be selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • In various embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 97% chirally pure. In some embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 99% chirally pure, and in other embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is at least about 99.9% chirally pure.
  • The chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole of various embodiments may be greater than about 99%. In some embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is greater than about 99.9%, and in other embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is greater than about 99.99%.
  • in particular embodiments, the entantiomerically enriched 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be a ratio of greater than about 2:1 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole to (6S)2,6 diamino-4,5,6,7-tetrahydro-benzothiazole.
  • In some embodiments, the solvent may be selected from an organic solvent and an organic solvent mixed with water, and in certain embodiments, the solvent may be selected from ethanol, 1-propanol, n-butanol, dihydrofuran, dimethylformamide, dimethyl, dimethylacetamide, hexamethylphosphoric triamide or mixtures or hydrates thereof.
  • In various embodiments, the steps of heating and reacting may each independently include stirring, and in some embodiments, the steps of heating and reacting may each be independently carried out at a temperature of from about 50° C. to about 125° C. In other embodiments, the process may further include cooling the reaction mixture to a temperature of about 25° C. after the step of reacting. In yet other embodiments, the process may further include the step of adding the propyl halide or propyl sulfonate to heated entantiomerically enriched 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, and in certain embodiments, the step of adding may be carried out for up to about 2 hours. In particular embodiments, about 1.0 to about 2.0 molar equivalents of the propyl sulfonate or propyl halide may be added. In still other embodiments, the step of reacting may be carried out for up to about 12 hours. In further embodiments, the step of recovering may include one or more steps selected from filtering the mixture to isolate a precipitate, washing a precipitate, and drying a precipitate.
  • Various other embodiments of the invention include a process for preparing a chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole including the steps of heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole in an organic solvent; adding to the heated solution propyl sulfonate or a propyl halide to form a reaction mixture; and reacting the reaction mixture for up to about 12 hours. In some embodiments, the propyl sulfonate may be selected from n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof, and in other embodiments, the propyl halide is selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • The process of embodiments may result in chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole that is at least greater than about 97% chirally pure. In some embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 99% chirally pure, and in other embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least about 99.9% chirally pure.
  • The chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole of some embodiments may be greater than about 99%. In certain embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be greater than about 99.9%, and in certain other embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be greater than about 99.99%.
  • In some embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be (6R)2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In other embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In still other embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be a mixture of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In certain embodiments, the mixture is a racemic mixture. In other embodiments, the mixture may be a ratio of greater than about 1:4 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole to about 4:1 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole.
  • The organic solvent of embodiments may be selected from an organic solvent and an organic solvent is mixed with water, and in some embodiments, the organic solvent may be selected from ethanol, 1-propanol, n-butanol, dihydrofuran, dimethylformamide, dimethyl, dimethylacetamide, hexamethylphosphoric triamide or mixtures or hydrates thereof.
  • In various embodiments, the steps of heating, adding and reacting each independently may include stirring, and in some embodiments, the steps of heating, adding and reacting may each independently be carried out at a temperature of from about 50° C. to about 125° C. In certain embodiment, the process may further include the step of cooling the reaction mixture to a temperature of about 25° C. after the step of reacting. In some embodiments, about 1.0 to about 2.0 molar equivalents of the propyl sulfonate or propyl halide may be added, and the step of adding in particular embodiments may be carried out for up to about 2 hours. In still other embodiments, the process may include the step of recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole, and in some embodiments, recovering may include one or more steps selected from filtering the mixture to isolate a precipitate, washing a precipitate, and drying a precipitate.
  • Embodiments of the invention further include a process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole including the steps of heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole; adding a propyl halide or a propyl sulfonate to the heated solution slowly over from about 0.5 hours to about 2 hours to form a reaction mixture; reacting the reaction mixture; and recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
  • In some embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 97% chirally pure. In other embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 99% chirally pure, and in certain embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is at least about 99.9% chirally pure.
  • The chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole of various embodiments may be greater than about 99%. In some embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be greater than about 99.9%, and in particular embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is greater than about 99.99%.
  • In some embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In other embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole is (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In certain embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be a mixture of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In some such embodiments, the mixture may be a racemic mixture. In other such embodiments, the mixture may be a ratio of greater than about 1:4 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole to about 4:1 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole.
  • In particular embodiments, the steps of heating, reacting and cooling each independently may include stirring. In other embodiments, the step of recovering may include cooling the mixture to a temperature of about 25° C., and in still other embodiments, the step of recovering may include stirring the reaction mixture for at least about 2 hours. In yet other embodiments, the step of recovering may further include one or more steps selected from filtering the mixture to isolate a precipitate, washing a precipitate, and drying a precipitate. In various embodiments, the steps of heating, adding and reacting may each independently be carried out at a temperature of from about 50° C. to about 125° C., and in certain embodiments, the step of reacting may include stirring the reaction mixture for up to about 12 hours at from about 50° C. to about 125° C.
  • Some embodiments of the invention include a chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process including the steps of heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole; adding a propyl halide or a propyl sulfonate to the heated solution slowly over from about 0.5 hours to about 2 hours to form a reaction mixture; reacting the reaction mixture; and recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
  • In some embodiments, the propyl sulfonate is selected from n-propyl tosylate, n-propyl methoxysulfonate and combinations thereof, and in other embodiments, the propyl halide is selected from n-propyl bromide, n-propyl chloride, n-propyl fluoride, n-propyl iodide and combinations thereof.
  • In various embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may at least greater than about 97% chirally pure. In some embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be at least greater than about 99% chirally pure, and in other embodiments, the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is at least about 99.9% chirally pure.
  • In various other embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be greater than about 99%. In some embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be greater than about 99.9%, and in other embodiments, the chemical purity of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole is greater than about 99.99%.
  • In some embodiments, the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be a mixture of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. In certain embodiments, the mixture may be a racemic mixture. In other embodiments, the mixture may be a ratio of greater than about 4:1 (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole.
  • The organic solvent of various embodiments may be selected from a organic solvent and an organic solvent is mixed with water, and the organic solvent of some embodiments may be selected from ethanol, 1-propanol, n-butanol, dihydrofuran, dimethylformamide, dimethyl, dimethylacetamide, hexamethylphosphoric triamide or mixtures or hydrates thereof.
  • In particular embodiments, the steps of heating, reacting and cooling each independently may include stirring, and in some embodiments, the steps of heating, adding and reacting are each independently carried out at a temperature of from about 50° C. to about 125° C. In other embodiments, the process may further include cooling the reaction mixture to a temperature of about 25° C. after the step of reacting, and in still other embodiments, the step of adding is carried out for up to about 2 hours. In certain embodiments, the step of recovering may include one or more steps selected from filtering the mixture to isolate a precipitate, washing a precipitate, and drying a precipitate. In various embodiments, about 1.0 to about 2.0 molar equivalents of the propyl sulfonate or propyl halide may be added.
  • Yet other embodiments of the invention include a process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole including the steps of dissolving entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in an organic solvent to form a solution; heating the solution to from about 50° C. to about 125° C.; adding an acid to the solution to form a reaction mixture; and recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
  • In some embodiments, about 1 molar equivalent to about 4 molar equivalents of the acid may be added. In other embodiments, recovering may include one or more steps including cooling the reaction mixture to a temperature of about 25° C.; stirring the reaction mixture for at least about 2 hours; filtering the mixture to isolate a precipitate; washing a precipitate; and drying a precipitate.
  • Further embodiments of the invention include a chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by such a process.
  • Still other embodiments of the invention include a process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole including the steps of dissolving entantiomerically enriched 2-amino-4,5,6,7-tetmhydro-6-(propylamino)benzothiazole in an organic solvent to form a solution; heating the solution to from about 50° C. to about 125° C.; adding an achiral salt to the solution to form a reaction mixture; and recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole
  • In some embodiments, about 1 molar equivalent to about 4 molar equivalents of the achiral salt is added. In other embodiments, the step of recovering may include one or more steps selected from cooling the reaction mixture to a temperature of about 25° C.; stirring the reaction mixture for at least about 2 hours; filtering the mixture to isolate a precipitate; washing a precipitate; and drying a precipitate.
  • Further embodiments of the invention include a chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a such a process.
  • Still further embodiments of the invention include a process for preparing a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride comprising dissolving a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt in an organic solvent to form a solution; cooling the solution to a temperature of from about 0° C. to about 5° C.; adding concentrated HCl and an organic solvent to the cooled solution; and stirring the solution at a temperature of about 0° C. to about 5° C.
  • DESCRIPTION OF DRAWINGS
  • For a fuller understanding of the nature and advantages of the present invention, reference should be made to the following detailed description taken in connection with the accompanying drawings, in which:
  • FIG. 1A shows a reaction scheme illustrating the alkylation of 4,5,6,7-tetrahydro-benzothiazole diamine.
  • FIG. 1B shows a reaction scheme illustrating the entantiomeric purification of one 4,5,6,7-tetrahydro-benzothiazole diamine from an entantiomeric mixture of 4,5,6,7-tetrahydro-benzothiazole diamines.
  • FIG. 2 shows a reaction scheme illustrating the preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine-dihydrochloride and (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine-difumerate.
  • FIG. 3A shows an exemplary HPLC trace of a mixture of (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine that is entantiomerically enriched for (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and a corresponding data table.
  • FIG. 3B shows an exemplary HPLC trace of chirally purified (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and a corresponding data table.
  • FIG. 4A shows an exemplary HPLC trace of Sample 118.
  • FIG. 4B shows an exemplary HPLC trace of Sample 105.
  • FIG. 4C shows an exemplary HPLC trace of Sample 061.
  • FIG. 4D show an exemplary HPLC trace of Sample 326A.
  • FIG. 5 shows an exemplary UV spectrum of a propyl tosylate peak eluted from an SPE column.
  • FIG. 6A shows an exemplary HPLC trace of a propyl tosylate standard.
  • FIG. 6B shows an exemplary HPLC trace of a (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine sample.
  • FIG. 7A shows an exemplary HPLC trace of a standard (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine.
  • FIG. 7B shows an exemplary HPLC trace of a sample (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine.
  • DETAILED DESCRIPTION
  • Before the present compositions and methods are described, it is to be understood that this invention is not limited to the particular processes, compositions, or methodologies described, as these may vary. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
  • It must be noted that, as used herein, and in the appended claims, the singular forms “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. Although any methods similar or equivalent to those described herein can be used in the practice or testing of embodiments of the present invention, the preferred methods are now described. All publications and references mentioned herein are incorporated by reference. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
  • As used herein, 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%.
  • “Optional” or “optionally” may be taken to mean that the subsequently described structure, event or circumstance may or may not occur, and that the description includes instances where the event occurs and instances where it does not.
  • “Administering” when used in conjunction with a therapeutic means to administer a therapeutic directly into or onto a target tissue or to administer a therapeutic to a patient whereby the therapeutic positively impacts the tissue to which it is targeted. “Administering” a composition may be accomplished by oral administration, injection, infusion, absorption or by any method in combination with other known techniques. Such combination techniques include heating, radiation and ultrasound.
  • The term “target”, as used herein, refers to the material for which either deactivation, rupture, disruption or destruction or preservation, maintenance, restoration or improvement of function or state is desired. For example, diseased cells, pathogens, or infectious material may be considered undesirable material in a diseased subject and may be a target for therapy.
  • Generally speaking, the term “tissue” refers to any aggregation of similarly specialized cells which are united in the performance of a particular function.
  • The term “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 administration of an active agent.
  • As used herein, the term “therapeutic” means an agent utilized to treat, combat, ameliorate or prevent an unwanted condition or disease of a patient.
  • The terms “therapeutically 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.
  • As used herein, the term “neuroprotectant” refers to any agent that may prevent, ameliorate or slow the progression of neuronal degeneration and/or neuronal cell death.
  • The term “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 “patient” generally refers to any living organism to which to compounds described herein are administered and may include, but is not limited to, any non-human mammal, primate or human. Such “patients” may or my not be exhibiting the signs, symptoms or pathology of the particular diseased state.
  • 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.
  • As used herein, the terms “optically pure” or “entantiomerically pure” may be taken to indicate that a composition contains at least 99.95% of a single optical isomer of a compound. The term “entantiomerically enriched” may be taken to indicate that at least 51% of a composition is a single optical isomer or enantiomer. The term “entantiomeric enrichment” as used herein refers to an increase in the amount of one entantiomer as compared to the other. A “racemic” mixture is a mixture of equal amounts of (6R) and (6S) enantiomers of a chiral molecule.
  • Throughout this disclosure, the word “pramipexole” will refer to (6S) enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole unless otherwise specified.
  • The term “trituration” may be taken to indicate a method of solidifying a chemical compound. Trituration involves agitating the compound by stirring, beating or a method of the like until the chemical compound forms a crystalline solid or precipitate. This solid may act to seed the remaining chemical compound in solution, causing it to precipitate or crystallize from solution.
  • The term “pharmaceutical composition” shall mean a composition including at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). 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 (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine or a pharmaceutically acceptable salt of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine as the active ingredient. Alternatively, a pharmaceutical composition may contain (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine or a pharmaceutically acceptable salt of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine as the active ingredient.
  • For the purposes of this disclosure, a “salt” is any acid addition salt, preferably a pharmaceutically acceptable acid addition salt, including but not limited to, 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. 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. In all cases, 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.
  • Embodiments of the invention described herein are generally directed to processes for the production of an entantiomerically and/or chemically pure compound. More, specifically, embodiments of the invention are directed to production of an entantiomerically and/or chemically pure compound using a trituration step where one enantiomer of an entantiomeric mixture of R and S stereoisomers of a compound is precipitated out of solution and can be isolated by, for example, simple filtering or other means for separating a solid or crystalline compound from a solution.
  • For example, embodiments of the invention include a method for preparation of an entantiomerically and chemically pure compound using an one-pot bi-molecular nucleophilic substitution (SN2) reaction synthesis method as illustrated in the reaction scheme provided in FIG. 1A. In FIG. 1A, a 4,5,6,7-tetrahydro-benzothiazole diamine (1) is reacted with an alkyl sulfonate (11) or a alkyl halide (12), exemplified by n-propyl sulfonate or propyl halide, to generate an aminoalkyl containing compound of a 4,5,6,7-tetrahydro-benzothiazole diamine (2), and a sulfonate or halide salt. In a trituration step, one enantiomer of the 4,5,6,7-tetrahydro-benzothiazole diamine, for example, (R)(+)-4,5,6,7-tetrahydro-benzothiazole diamine (3), is precipitated based on insolubility of the enantiomers in the achiral halide or sulfonate salts produced as a result of the reaction and can be isolated. The other enantiomer remains in solution.
  • In another exemplary embodiment illustrated in FIG. 1B, chirally pure compounds, such as, (6R)4,5,6,7-tetrahydro-benzothiazole diamine (3), may be prepared from a mixture of R and S enantiomers of a compound, such as, for example, (6R)4,5,6,7-tetrahydro-benzothiazole diamine (4) and (6S)4,5,6,7-tetrahydro-benzothiazole diamine (5). In this process, trituration may result from the addition of an organic solvent and an achiral salt or acid to the mixture which may cause the formation of an acid addition salt of one enantiomer. The salt, (6R)4,5,6,7-tetrahydro-benzothiazole diamine (3), precipitates out of the solution based on insolubility of the enantiomers in the resulting solution while the other enantiomer remains in solution. The precipitated crystalline enantiomer may then be isolated.
  • The reaction illustrated in FIG. 1A is not limited to a particular 4,5,6,7-tetrahydro-benzothiazole diamine. For example, the 4,5,6,7-tetrahydro-benzothiazole diamine may be any 4,5,6,7-tetrahydro-benzothiazole diamine of formula (I):
  • Figure US20140100372A1-20140410-C00004
  • wherein:
  • R1 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkenyl or alkynyl group each having 3 to 6 carbon atoms, an alkanoyl group having 1 to 6 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the above-mentioned phenyl nuclei may be substituted by 1 or 2 halogen atoms;
  • R2 represents a hydrogen atom or an alkyl group with 1 to 4 carbon atoms;
  • R3 represents a hydrogen atom, an alkyl group with 1 to 7 carbon atoms, a cycloalkyl group having 3 to 7 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms, an alkanoyl group having 1 to 7 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the phenyl nucleus may be substituted by fluorine, chlorine or bromine atoms,
  • R4 represents a hydrogen atom, an alkyl group with 1 to 4 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms;
  • and at least one or R1, R2, R3 or R4 is a hydrogen.
  • The 4,5,6,7-tetrahydro-benzothiazole diamine of formula 1 encompasses all enantiomers at any chiral center on the molecule.
  • The reaction illustrated in FIG. 1A is not limited by the type of alkyl halide or alkyl sulfonate utilized in the reaction. For example, the alkyl sulfonates may be any alkyl sulfonate of formula (II):
  • Figure US20140100372A1-20140410-C00005
  • wherein:
  • R′ is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl, phenyl alkyl and the like; and
  • Z is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl, phenyl alkyl and the like; and the alkyl halide may be any alkyl halide of formula (12):

  • R′—X  (12)
  • wherein
  • R′ is an alkyl group having 1 to 6 carbons, or a cycloalkyl, alkenyl, alkynyl, allyl, having 1 to 10 carbon atoms, or a benzyl, chlorobenzyl, phenyl, phenyl alkyl and the like; and
  • X is any halide including, for example, chlorine, bromine or iodide. In various embodiments, R′ is an alkyl and, in particular embodiments, an n-propyl. In certain embodiments where alkyl sulfonate, the Z moiety may be toluenesulfonate (tosylate) or methoxysulfonate. For example, in various embodiments, the alkyl sulfonate may be n-propyl tosylate. In embodiments in which an alkyl halide is used, the alkyl halide may be n-propyl bromide or n-propyl chloride. In general, the alkyl sulfonate or alkyl halide may be added in a quantity corresponding to about 1.0 to about 4.0 molar equivalents of the diamine.
  • Advantages of embodiments such as those described above may include, for example, (1) the use of simple reagents for the synthesis and purification of one enantiomer enantiomeric compounds, (2) the surprising improvement of the optical and chemical purity achieved by simple trituration, and (3) such processes may be performed as a one-pot synthesis and purification reactions. When taken as a whole, the processes described above may be simpler, safer, and more efficient for the production of chirally and chemically pure compounds. Additionally, the compounds may be sufficiently chirally and chemically pure to make such compounds safe an effective for use in pharmaceutical compounds and in the treatment of disease.
  • While the methods provided above may be used to purify any enantiomeric compound known in the art, particular embodiments of the invention are directed to the production and optical purification of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole and, in particular, (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. The extremely high chiral and chemical purity of the compound produced by methods of the present invention allow for pharmaceutical compositions that may have a wide individual and daily dose range. For example, in some embodiments, (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine produced using methods embodied herein may be nearly 100% chirally pure. Such compositions include little to no (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine contamination and may be administered at high doses without the risk of the dopaminergic side effects associated with administration of high dose (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. The compositions of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may therefore be used to treat neurodegenerative diseases, or those associated with mitochondrial dysfunction or increased oxidative stress such as, for example, neurodegenerative dementias, neurodegenerative movement disorders and ataxias, seizure disorders, motor neuron disorders or diseases, inflammatory demyelinating disorders and the like in adults and children. The compositions of this disclosure may also be useful in the treatment of other disorders not listed herein, and any listing provided in this disclosure is for exemplary purposes only and is non-limiting.
  • Compositions and pharmaceutical compositions comprising entantiomerically pure (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine, such as those discussed above, are further disclosed in U.S. application Ser. No. 11/773,642 entitled “Tetrahydrobenzothiazoles and Uses Thereof” filed Apr. 10, 2007, U.S. application Ser. No. 11/957,157 entitled “Compositions and Methods of Using R(+) Pramipexole”, filed Dec. 14, 2007 and U.S. application Ser. No. 11/749,497 entitled “Pramipexole Formulations for the Treatment of Parkinson's Disease”, filed on May 16, 2007, each of which are hereby incorporated by reference in their entireties. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the present invention, the preferred methods, devices, and materials are described in detail herein.
  • Production of either enantiomer, (6R) or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be carried using the bi-molecular nucleophilic substitution (SN2) reaction described above and optical purification based on insolubility of the compound product in achiral reagents. More specifically, FIG. 2 schematically illustrates an embodiment of the process for the production of entantiomerically and chemically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole using a bi-molecular nucleophilic substitution (SN2) reaction. In a first step, 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole (7) is mixed propyl p-toluenesulfonatc (propyl tosylate), and reacted to produce 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8) and 4-methylbenzenesulfonic acid (p-toluenesulfonic acid, p-TSA). Without wishing to be bound by theory, the diamine, 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole (7), in the reaction scheme of FIG. 2 may act as the nucleophile in a nucleophilic attack on the substrate, a propyl tosylate, and the tosylate group may provide a good leaving group as depicted below:
  • Figure US20140100372A1-20140410-C00006
  • Thus, an embodiment of the invention is a process for preparing a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt (8) by a bimolecular nucleophilic substitution reaction. In a second step, the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8) may be entantiomerically purified by allowing the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8) to precipitate out of the reaction solution without the addition of any secondary agents, such as, for example, additional salt. Thus, entantiomerically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be isolated in a third step by simply filtering out the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole crystals.
  • Some embodiments of methods of the invention may include additional steps. For example, in some embodiments, p-TSA may be removed to form the entantiomerically purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8) to form a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole free base (9) and hydrochloric acid or fumaric acid may be added to the free base to form 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole-dihydrochloride (14) or 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole-difumerate (15). In other embodiments, 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole-dihydrochloride (14) may be produced by adding hydrochloric acid to the entantiomerically purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8).
  • While the reaction scheme illustrated in FIG. 2, shows the production of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine from (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, in some embodiments, the same reaction may be carried out using (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole to produce (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In other embodiments, a mixture of (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be used as starting material which may result in a mixture of (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In particular embodiments, a mixture of (6R) and (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole that is entantiomerically enriched for one entantiomer may be used as starting material. Additionally, the reaction illustrated in FIG. 2 may be carried out is an n-propyl halide or a mixture of n-propyl halide and n-propyl sulfonate.
  • In some embodiments, step one of the process may include the additional steps of heating a diamine to form a solution or melt in a heating step and adding the n-propyl halide or n-propyl sulfonate slowly over a period of time from, for example, about 0.5 hours to about 5 hours, in an additional step. In other embodiments, after the n-propyl halide or n-propyl sulfonate has been completely added to the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, the reaction may continue under heating for an additional period of time ranging from, for example, about 1 hour to about 12 hours, in a reaction step. In certain embodiments, the reaction mixture may be mixed by, for example, stirring for one or more of the steps above or the reaction mixture may be continually stirred from the heating step to the reaction step. Following the reaction step, the reaction mixture may be cooled and the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be isolated and purified.
  • In embodiments such as those exemplified above, the diamine, 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, may be a racemic mixture or entantiomerically enriched for either the S or R enantiomer, and the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole produced may be generally optically enriched for the dominant enantiomer. For example, (6R) 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole may be the dominant enantiomer in mixtures used as a starting material in reaction that produces (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. In various embodiments, any R to S ratio of diamine may be used. For example, in some embodiments, the diamine may be in a racemic mixture (i.e., R:S is about 50:50), and in such embodiments, the yield of the reaction would be expected to be a racemic mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine (i.e., about 50:50). In other embodiments, the diamine may be provided in a mixture in which one stereoisomer is in excess over the other; for example, R:S may be about 60:40. In such embodiments, the reaction would be expected to yield a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine that is about 60:40 R to S.
  • The reaction may be carried out in a melt or in a solvent or mixture of solvents, and the methods embodied herein are not limited by the type or number of solvents present during the reaction. Any solvent or mixture of solvents known in the art in which the diamine and alkyl halide or alkyl sulfonate can dissolved may be used. For example, in various embodiments, the solvents may be, for example, tetrahydrofuran, dimethylformamide, dimethly sulfoxide, dimethylacetamide, hexamethylohosphoric triamide, glacial acetic acid, pyridine, dioxan, ethanol, 1-propanol, i-propanol, n-butanol, i-butanol, or combinations thereof, for example, dioxan/water, ethanol/water, tetrahydrofuran/water and the like. In embodiments in which a combination of an organic solvent and water are used, the organic solvent may have a water content of from about 0 to about 10 volume percent. Preferably, the solvents used in the practice of this invention are standard ACS grade solvents. The selection of a solvent may enhance the reaction rate of the S N2 reaction. In some embodiments, one or more base such as, for example, sodium hydroxide, sodium hydride, potassium carbonate, sodium acetate, potassium-tertbutyloxide, triethylamine, di-isoproplyethylamine and the like, may be additionally added to the reaction mixture which may further enhance the efficiency of the reaction. When added, a base may be present in a concentration of about 0.5 to about 3.0 equivalents based on the solvent. In still other embodiments, an alkylating agent may be provided in the melt or solvent. Alkylating agents are well known in the art and may be useful in embodiments of the invention. For example, alkylating agents may include, but not limited to, methyliodide, dimethylsulfate, ethylbromide, diethylsulfate, allyliodide, benzylbromide, 2-phenylethylbromidc and methyl-p-toluenesulfonate.
  • In general, the reaction may be carried out under ambient conditions. However, the reaction temperature may vary among embodiments from between about −10° C. to about 50° C. and, in particular embodiments, from 0° C. to 30° C.
  • In further embodiments, dissolved diamine may be heated and mixed or stirred during the reaction. For example, various embodiments include the step of heating a dissolved diamine, adding a n-propyl sulfonate or n-propyl halide which may, in some embodiments, be dissolved in a solvent to form a mixture, and stirring the mixture. In other embodiments, a base such as di-isoproplyethylamine may be added to a solution including a diamine. N-propyl sulfonate or n-propyl halide may be dissolved in a solvent, and then added to the diamine/di-isoproplyethylamine solution and this reaction mixture may be stirred. The temperature of reactions of such embodiments may; generally, be below the boiling temperature of the reaction mixture, more specifically, below the boiling temperature of the solvent(s) of the reaction mixture. For example, in some embodiments, an elevated temperature may be lower than about 125° C. In others, an elevated temperature may be lower than about 100° C., and in yet another embodiment lower than about 95° C., and in still others less than about 75° C. Therefore, the reaction temperature may range from about 50° C. to about 125° C. in some embodiments, about 55° C. to about 100° C. in other embodiments, about 60° C. to about 95° C. in still other embodiments, about 60° C. to about 75° C. in yet other embodiments and in certain embodiments, from about 55° C. to about 65° C.
  • The reaction time may vary within embodiments, and may depend upon, for example, the identities of the reactants, the solvent system and the chosen temperature. For example, in some embodiments, the reaction time may be from about 0.5 hours to about 12 hours. In other embodiments, the reaction time may be from about 1 hour to about 8 hours, and in certain embodiments, the reaction time may be about 4 hours. In general, the reaction time is chosen to provide sufficient time for substantially all of the diamine to undergo alkylation. In particular embodiments, the reaction time further provides sufficient time for the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole formed to precipitate out of the reaction solution and from visible crystals.
  • Embodiments of the invention may further include the step of cooling the reaction to a temperature about room temperature (25° C.) following the reaction. In such embodiments, the reaction may be cooled for any amount of time with or without continued stirring. For example, in some embodiments, the reaction may be cooled with stirring for about 0.5 to about 4 hours or more, and in other embodiments, the reaction may be cooled with stirring for about 2 hours.
  • More specific embodiments may include the steps of: dissolving a diamine in dimethylformamide; heating the dissolved diamine to an elevated temperature; adding the n-propyl sulfonate or n-propyl halide dissolved in dimethylformamide to form a reaction mixture; and stirring the reaction mixture for about 4 hours. In another embodiment, the steps may include dissolving a diamine in dimethylformamide, heating the dissolved diamine to an elevated temperature, slowly adding to the heated dissolved diamine 1.25 molar equivalents of n-propyl sulfonate or n-propyl halide dissolved in 10 volumes of dimethylformamide and 1.25 molar equivalents of di-isoproplyethylamine with stirring the reaction over a period of about 4 hours. In yet another embodiment, 1.25 molar equivalents of di-isoproplyethylamine may be added to a diamine dissolved in dimethylformamide and the n-propyl sulfonate or n-propyl halide dissolved in dimethylformamide may be added to this mixture with stirring for about 4 hours.
  • Following synthesis, entantiomerically pure isomers of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be recovered using a trituration step in which the major isomer is isolated as a precipitated crystals, while the minor stereoisomer remains in solution. Without wishing to be bound by theory, insolubility of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product in achiral reagents, such as p-TSA, may be independent of the R or S enantiomer, such that purity of the recovered isomer may depend only on the volume of the reaction solution and starting percentage of the major isomer. Thus, in an embodiment such as is described above, an entantiomerically pure (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine (R:S equals 100:0) yield may be produced from a reaction in which diamine is provided in an R to S ratio of 60:40.
  • An unexpected advantage of the process for preparing 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole described above is the limited solubility of the sulfonate or halide salt of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in polar organic solvents which causes the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product to precipitate once formed thereby purifying the final synthesis product, 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole, from the reaction mixture. In further embodiments, the substitution reaction such as that illustrated in FIG. 2 may result in a sufficient achiral salt, p-TSA, concentration to cause the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole to become insoluble and crystallize in the reaction solution without adding additional agents, such as, for example, additional achiral salts.
  • The reaction embodied above and described in FIG. 2 may provide highly purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in a one-pot synthesis method. For example, in embodiments of the process, the chemical purity of the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared may be at least 97%, 98%, and up to 100% without additional purification steps, and in particular embodiments, the chemical purity may be from 99.90% to 100% without any additional purification steps. In still other embodiments, the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be substantially free of achiral salts such as, for example, p-TSA. For example, in some embodiments, the achiral salt concentration of the final synthesis product may be less than 3%, and in others the achiral salt concentration of the final synthesis product may be less than 1%, 0.5%, 0.1%, 0.01%, 0.001% and so on. In certain embodiments, the achiral salt concentration may be less than 1.5 ppm to less than 25 ppb or less than 0.00015% to less than 0.0000025%. Without wishing to be bound by theory, the ability to produce highly chemically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in a one-pot method may demonstrate a significant advancement in the production of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. Moreover, such purity may provide pharmaceutical grade 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole more efficiently than previous methods.
  • In various embodiments, the chiral purity for the R enantiomer prepared and purified may be greater than 99.6% about when a starting material that is entantiomerically enriched for R diamine is used. Similarly, in various other embodiments, the chiral purity for the S enantiomer produced and purified may be greater than 99.6% when a starting material that is entantiomerically enriched for S diamine is used. In some embodiments, the chiral purity for the R enantiomer prepared and purified may be greater than 99.8% about when a starting material that is entantiomerically enriched for R diamine is used. Similarly, in some other embodiments, the chiral purity for the S enantiomer produced and purified may be greater than 99.8% when a starting material that is entantiomerically enriched for S diamine is used. In particular embodiments, the chiral purity for the R enantiomer prepared and purified may be greater than 99.9% about when a starting material that is entantiomerically enriched for R diamine is used. Similarly, in particular other embodiments, the chiral purity for the S enantiomer produced and purified may be greater than 99.9% when a starting material that is entantiomerically enriched for S diamine is used.
  • Not wishing to be bound by theory, the solubility of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be the same in the trituration step of the synthesis and purification processes. For example, if a synthesis process is carried out with 90 grams of the (6R) diamine and 10 grams of the (6S)diamine, and the solubility of the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product is 10 grams for either enantiomer, then 80 grams of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine product and 0 grams of the (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine product would precipitate (assuming a 100% chemical conversion from the diamine and no change in molecular weight in going to the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product). That is, 10 grams of each enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be expected to go into solution. This would lead to a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product with a 100% chiral purity for the (6R) enantiomer. The opposite ratio of starting materials for the synthesis process (90 grams of the (6S) diamine and 10 grams of the (6R) diamine) may generate a reaction product of 90 grams of the (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and 10 grams of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. From this reaction product mixture, 80 grams of the (6S) enantiomer and 0 grams of the (6R) enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole would be expected to precipitate, leading to a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product with a 100% chiral purity for the (6S) enantiomer. Thus, the volumes which are used for a reaction may have a large potential effect on the final yield and chiral purity. That is, too large a volume will reduce the yield as more of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole enantiomer products will go into solution (but increase the chiral purity) and too small a volume will increase the yield as less of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole products will go into solution (but reduce the chiral purity).
  • Other embodiments of the invention are directed to a process for the purification of entantiomerically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole from a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine including a trituration step. In some embodiments, the mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine used in the purification methods may be prepared as described herein above. In other embodiments, the mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be obtained using another method or form a commercially available mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine.
  • In some embodiments, the trituration step may include the addition of an achiral salt to a solution containing a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. As described above, the addition of an achiral salt to a solution containing a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may cause the enantiomer having a greater concentration to become insoluble and form crystals in the solution. In some embodiments, the solution containing a mixture of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be heated to an elevated temperature such as, for example, about 50° C. to about 125° C., about 55° C. to about 100° C., about 60° C. to about 95° C. or about 60° C. to about 75° C. and an achiral salt may be added to the solution. This solution may than be cooled from the elevated temperature to about room temperature slowly. For example in one embodiment, the reaction may be cooled at a rate of about less than 25° C./hour. In another embodiment, the reaction may be slowly cooled and the reaction solution may be stirred for at least about an additional 2 hours. The rates of cooling and the time required for the additional stirring may vary with the choice of achiral salt and may be easily appreciated by one skilled in the art.
  • The crystalline (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may then be isolated, washed and dried, and in various embodiments, may result in entantiomerically pure (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine having a chemical purity of at least 97% and, in some embodiments, 98% to 100%. In such embodiments, the achiral salt may be any achiral salt listed hereinabove or any other achiral salt known in the art. Similarly, the solvent of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine solution may be any solvent described above in relation to the method for preparing 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. Either (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be purified using the process of such embodiments. However, in certain embodiments, (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine may be purified.
  • In other embodiments, entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be triturated from an acid addition solution based on the insolubility of the enantiomers in the achiral reagents. Various embodiments of this method may include the steps of dissolving an entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in a solvent at an elevated temperature such as, for example, about 50° C. to about 125° C., about 55° C. to about 100° C., about 60° C. to about 95° C. or about 60° C. to about 75° C., adding an acid to the dissolved 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole, cooling the reaction to about room temperature (25° C.) with stirring, stirring the cooled reaction mixture for an extended time at room temperature to allow formation of entantiomerically pure crystals and recovering entantiomerically pure (6R) or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine from the reaction mixture. In other embodiments, entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be dissolved in a solvent at an elevated temperature, about 0.5 equivalents to about 2.05 equivalents of an acid may be added to the solution and the solution may be cooled to room temperature. The cooled solution may then be stirred for an extended period of time and entantiomerically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be recovered. In particular embodiments, the selected acid may be p-toluenesulfonic acid (p-TSA) and the solvent may be ethanol. In other embodiments, the temperature of the solution when the acid is added may be lower than about 125° C., lower than about 100° C. or lower than about 75° C., and in certain embodiments, the temperature may be from about 65° C. to about 85° C. The cooling may generally occur slowly at, for example, a rate of about 25° C. per hour and the solution may be stirred for at least about 2 hours after 25° C. temperature has been reached. The times necessary for the reaction may vary with the identities of the reactants, the solvent system and with the chosen temperature, and may be easily appreciated by one of skill in the art. Reaction volumes may additionally dictate the degree of optical purification and the overall yield of the optically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. These volumes would be understood and appreciated by one of skill in the art. Examples of specific times, temperatures and volumes which enable the practice of this invention are given in the Examples.
  • The solvent utilized may vary in embodiments and may generally be an organic solvent such as, for example, acetonitrile, acetone, ethanol, ethyl acetate, methyl tert-butyl ether, methyl ethyl ketone, isopropyl acetate, isopropyl alcohol and combinations thereof. In a particular embodiment, the organic solvent may be ethanol.
  • The acid of various embodiments may include: halogenic acids such as, for example, hydrobromic, hydrochloric, hydrofluoric and hydroiodic acid; inorganic acids such as, for example, nitric, perchloric, sulfuric and phosphoric acid; organic acids such as, for example, sulfonic acids (methanesulfonic, trifluoromethane sulfonic, ethanesulfonic, benzenesulfonic or p-toluenesulfonic), acetic, malic, fumaric, succinic, citric, benzoic, gluconic, lactic, mandelic, mucic, pamoic, pantothenic, oxalic and maleic acid; and amino acids such as aspartic or glutamic acid. The acid may be a mono- or di-acid, such as, for example, a di-hydrohalogenic, di-sulfuric, di-phosphoric or di-organic acid. The acid of embodiments may be used as an achiral reagent which is, generally, not selected on the basis of any expected or known preference for interaction with, or precipitation of, a specific optical isomer of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole to be isolated. For example, in one embodiment, the selected acid may be p-toluenesulfonic acid. The amount of acid added may vary and is generally provided at about 1 molar equivalent to about 4 molar equivalents of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
  • Insoluble 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be separated from the reaction solution by any method known in the art. For example, in some embodiments, the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be collected by simple filtering. There are numerous methods for filtering a solid from a solution, and any such method may by useful in embodiments of the invention. In other embodiments, insoluble 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be isolated by centrifugation. Again, such methods are well known in the art and any such method may be used in various embodiments of the invention. The insoluble crystalline 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may then be washed to remove any contaminating solvent, sulfonate or halide salt, or soluble enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole from the crystals using any method available. For example, in one embodiment, the precipitated material may be washed in a volatile solvent such as an alcohol or heptane followed by vacuum drying.
  • The 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole enantiomers prepared using methods above may be purified from a starting (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine mixture that is enriched for one or the other enantiomers. For example, in some embodiments, the starting mixture may contain at least 55% or greater of either the R or S enantiomer, and in others the starting mixture may contain about 70% or greater of either the R or the S enantiomer. In still other embodiments, the starting material may contain greater than about 90% of either the R or S enantiomer. In particular embodiments, the starting mixture is enriched for (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine.
  • Without wishing to be bound by theory, the relative solubility of the optical isomers of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in the achiral salt or acid solutions allows for chiral and chemical purification that is unexpected by using a relatively easy recovery method via a simple trituration step. The enhanced enrichment resulting from the purification methods described above result in that may reach optical purity. For example, in various embodiments, the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product may be enriched to 99% optical purity or greater, 99.5% optical purity or greater, 99.8% optical purity or greater, and in certain embodiments, 99.9% optical purity or greater. In still other embodiments, the optical purity of the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be 99.95% or greater, or even 99.9.9% or greater. In particular embodiments, the optical purity may be 100%.
  • The processes disclosed herein have several advantages. First, the processes avoid the use of borane reagents such as sodium borohydride, common in the reductive amination schemes used in the prior art, which decomposes rapidly to borane and hydrogen upon acidification. Second, reductive amination schemes involve the use of a two-step procedure in which the amide is formed first, followed by a reduction step. The methods of this disclosure are one-pot synthesis and purification procedures, and therefore provide a safer, easier and more economical synthesis. Third, there is no loss of chirality during the alkylation processes of the S N2 reaction mechanism of this disclosure, as opposed to previous synthesis schemes where chiral purity is often reduced or lost altogether during synthesis. Finally, the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product of the nucleophilic substitution reaction precipitates from the reaction mixture. This may be especially true for the p-TSA salt form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. This is an unexpected advantage of the methods of the instant disclosure and provides unique methods for chiral and chemical enrichment or purification of the final 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product.
  • Additional embodiments of the invention include the conversion of either sulfonate or halide salts of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole or free base 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole into an hydrochloric acid (HCl) salt as illustrated in FIG. 2. For example, in some embodiments, solid 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole sulfonate or halide salt (8) may be re-dissolved in an alcohol, such as ethanol, and the mixture may be cooled to between about 0 and about 5° C. with continuous stirring. Concentrated HCl may then be added, followed by a solvent such as methyl tert-butyl ether (MTBE), and the mixture may be stirred for about 0.5 to about 3 hours at between about 0 and about 5° C. until or until insoluble 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride crystals (10) have formed. The reaction mixture may then be filtered, washed in an inert solvent such as MTBE/alcohol solution and dried under vacuum. A detailed example of this synthesis may be found in Example 12.
  • In another embodiment, of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole sulfonate or halide salts (8) may be converted to an HCl salt using a concentrated solution of HCl and isopropyl acetate (IPAC). In such embodiments, 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole sulfonate or halide salt (8) may be dissolved in IPAC and cooled to about 15° C. HCl (gas) may then be bubbled into the slurry for from about 0.5 hours to 3 hours to produce 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride (10), after which the mixture may be filtered, washed with an inert solvent, such as, for example, IPAC and dried under vacuum at room temperature. A detailed example of this synthesis may be found in Example 13.
  • The sulfonate or halide salts of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may alternatively be converted to the free base form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole as illustrated in FIG. 2. For example, in one embodiment, a p-TSA 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt (8) may be dissolved in dichloromethane (DCM) and water. The solution may then by brought to a pH of about 11-12 using NaOH and resulting in the formation of two phases. The aqueous phase contains 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole that may be extracted with DCM, dried over magnesium sulfate (MgSO4), filtered over Celite® and concentrated. The concentrated residue may be re-dissolved in MTBE and stirred as a slurry for several hours. The solids may then be filtered, washed with MTBE, and dried under vacuum at a temperature of about 35° C. The final product is 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole free base (9). A detailed example of this synthesis may be found in Example 14.
  • In another embodiment, the sulfonate or halide salts of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (8) may be converted to the free base form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (9) by dissolving p-TSA salt of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in water and cooling the solution to a temperature of about 10° C. NaOH may be added to the solution to increase the pH, the solution may be diluted and extracted several times in DCM. The combined organic phases are then washed, dried over MgSO4, filtered and concentrated to dryness. A detailed example of this synthesis may be found in Example 15.
  • In some embodiments, the free base form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be converted to 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride (9) by bubbling HCl gas into a cooled solution of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole free base in IPAC. Alternatively, in other embodiments, the free base form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (9) may be converted to 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride (10) by mixing with concentrated HCl at room temperature overnight. Detailed examples of such schemes may be found in Examples 16 and 17, respectively. In still other embodiments, the free base form of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole (9) may be converted to 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole fumarate (11) by the addition of about 1 to about 4 molar equivalents of fumaric acid.
  • The methods of the present disclosure require little time, utilize readily available starting materials and do not involve the use of hazardous or difficult to handle reagents. Each of the several steps of the methods disclosed as part of the present invention are high yielding and afford products with very high chemical and chiral purity. Further, the processes disclosed herein may be scaled for industrial scale manufacturing. As such, entantiomerically pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole may be manufactured in batches of greater than 1 kg or more, 10 kg or more, or even 25 kg or more as may be required to meet the needs of a large scale pharmaceutical use.
  • Embodiments of the invention also relate to a pure enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole, either (6R) or (6S), produced by processes disclosed herein. Thus, an embodiment of the invention is a chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt prepared by a process which comprises dissolving 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole in an organic solvent, reacting the 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole with a n-propyl sulfonate or a n-propyl halide under conditions sufficient to generate the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt and recovering the chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt.
  • The present invention should not be considered limited to the particular embodiments described above, but rather should be understood to cover all aspects of the invention as fairly set out in the attached claims. Various modifications, equivalent processes, as well as numerous structures to which the present invention may be applicable, will be readily apparent to those skilled in the art to which the present invention is directed upon review of the present specification. The claims are intended to cover such modifications and devices. The invention and embodiments thereof illustrating the method and materials used may be further understood by reference to the following non-limiting examples.
  • EXAMPLES
  • 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole was prepared using the S N2 substitution reaction described above. Reactions were carried out under exemplary reactions conditions A, B and C described below. Results of example syntheses using each of the several conditions which are embodiments of the present disclosure are listed in Table 1. Several example syntheses of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole using conditions A, B and C of the present disclosure are detailed in Examples 1-5, the results are presented in Table 1.
  • A: diamine dissolved in an organic solvent was heated to a reaction temperature of less than about 125° C. with continuous stirring. A solution of n-propyl sulfonate or n-propyl halide dissolved in di-isoproplyethylamine and an organic solvent was added to the dissolved, heated diamine slowly over a period of up to several hours to form a mixture, and this reaction mixture was stirred at the reaction temperature for an additional period of time of up to about 4 hours.
  • B: diamine dissolved in an organic solvent was heated to a reaction temperature of less than about 125° C. with continuous stirring. A solution of n-propyl sulfonate or n-propyl halide dissolved in dimethylformamide was added slowly over a period of up to several hours to form a reaction mixture, and this reaction mixture was stirred at the reaction temperature for an additional amount of time up to about 4 hours.
  • C: diamine was dissolved in dimethylformamide and heated to less than about 125° C. with continuous stirring. A solution of n-propyl sulfonate or n-propyl halide dissolved in dimethylformamide and di-isopropylethylamine was added to the heated diamine slowly over a period of up to several hours to form a reaction mixture. This reaction mixture was then stirred at the reaction temperature for up to about 4 hours.
  • Alternatively, di-isoproplyethylamine may be added to the heated diamine dissolved in an organic solvent prior to the addition of a solution including n-propyl sulfonate or n-propyl halide dissolved in dimethylformamide. As above, the n-propyl sulfonate or propyl halide/dimethlyformamide solution may be added slowly for a time period up to several hours with continuous stirring and the reaction mixture formed may be stirred at the reaction temperature for up to about 4 hours.
  • These products were analyzed by high pressure liquid chromatography (HPLC) for chemical and chiral purity. 1H NMR and 13C NMR was also used to confirm the structure of the product is 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole
  • Example 1 Preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition A
  • A 2.0 liter, three necked flask was equipped with an overhead stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 500 ml addition funnel. The flask was charged with 45 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine, followed by 750 ml of n-propanol. Under continuous stirring, the mixture was heated to 95° C. over 15 minutes generating a clear solution. The addition funnel was charged with a solution of 74 grams propyl tosylate and 60 ml diisopropylethyleamine in 250 ml n-propanol. This solution was added dropwise to the 2.0 liter flask with continuous stirring over a period of 4 hours. The reaction was continued with stirring for an additional 8 hours at 95° C., after which the solution was brought to room temperature, and stirring was continued for an additional 4 hours.
  • The precipitated material was collected by filtration and washed three times using 100 ml reagent grade alcohol each time. The alcohol washed precipitated cake was then washed with 100 ml heptane and dried under high vacuum for 2 hours.
  • The final weight of the dried product was 53.2 grams, representing a 52.2% yield. HPLC was used to determine the chemical purity of the (6R)-2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 98.2% and the chiral purity as greater than 99.5%. 1H NMR and 13C NMR were used to confirm the structure.
  • Example 2 Preparation of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition A
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 50 ml addition funnel. The flask was charged with 5 grams of (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 45 ml of n-propanol. Under continuous stirring, the mixture was heated to a temperature of 95° C. over 15 minutes generating a clear solution. The addition funnel was charged with a solution of 8.2 grams propyl tosylate and 6.7 ml diisopropylethyleamine in 16 ml n-propanol. This solution was added dropwise to the 250 ml flask with continuous stirring over a period of 2 hours. The reaction was continued with stirring for an additional 6 hours at 95° C., after which the solution was brought to room temperature, and stirring was continued for an additional 4 hours.
  • The precipitated material was collected by filtration and washed three times using 10 ml reagent grade alcohol each time. The alcohol washed precipitated cake was then washed with 10 ml heptane and dried under high vacuum for 2 hours.
  • The final weight of the dried product was 4.99 grams, representing a 44.2% yield. HPLC was used to determine the chemical purity of the (6S)-2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 98.0% and the chiral purity as greater than 99.6%. 1H NMR was used to confirm the structure.
  • Example 3 Preparation of racemic 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole p-TSA salt: Condition A
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 100 ml addition funnel. The flask was charged with 5 grams of racemic 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 80 ml of n-propanol. Under continuous stirring, the mixture was heated to a temperature of 95° C. over 15 minutes generating a clear solution. The addition funnel was charged with a solution of 10.1 grams propyl tosylate and 8.2 ml diisopropylethyleamine in 28 ml n-propanol. This solution was added dropwise to the 250 ml flask with continuous stirring over a period of 2 hours. The reaction was continued with stirring for an additional 6 hours at 95° C., after which the solution was brought to room temperature, and stirring was continued for an additional 6 hours.
  • The precipitated material was collected by filtration and washed two times using 25 ml reagent grade alcohol each time. The alcohol washed precipitated cake was then washed with 25 ml heptane and dried under high vacuum for 1 hour.
  • The final weight of the dried product was 5.12 grams, representing a 45% yield. HPLC was used to determine the chemical purity of the racemic 2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 97.1%, and the chiral purity showed a 1:1 mixture of the (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. 1H NMR was used to confirm the structure.
  • Example 4 Preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition B
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 50 ml addition funnel. The flask was charged with 5 grams of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 50 ml of DMF. Under continuous stirring, the mixture was heated to a temperature of 75° C. 6.3 grams propyl tosylate was added dropwise to the 250 ml flask with continuous stirring over a period of 6 hours. Progress of the reaction was monitored by analysis on HPLC.
  • The reaction was continued with stirring for an additional 12 hours at room temperature. The solution was diluted with 20 ml. MTBE and stirred for an additional hour. The precipitated material was collected by filtration and washed with 20 ml MTBE, followed by 2 washes of 20 ml each ethanol. The washed precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 4.6 grams, representing a 40% yield. HPLC was used to determine the chemical purity of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine as 94.9% and the chiral purity as greater than 99.6%. 1H NMR was used to confirm the structure.
  • Example 5 Preparation of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition B
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 50 ml addition funnel. The flask was charged with 10 grams of (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 100 ml of DMF. Under continuous stirring, the mixture was heated to a temperature of 75° C. The addition funnel was charged with a solution of 16.4 grams propyl tosylate in 20 ml DMF. This solution was added dropwise to the 250 ml flask with continuous stirring over a period of 1.5 hours. Progress of the reaction was monitored by analysis on HPLC.
  • The reaction was continued with stirring for an additional 12 hours at 75° C., after which the solution was brought to room temperature, and stirring was continued for an additional 7 hours. The solution was diluted with 100 ml MTBE and stirred for an additional hour. The precipitated material was collected by filtration and washed with 100 ml MTBE, followed by 2 washes of 50 ml each ethanol, and a wash with 50 ml heptane. The washed precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 9.81 grams, representing a 43.3% yield. HPLC was used to determine the chemical purity of the (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine as 99.4% and the chiral purity as greater than 99.8%. 1H NMR was used to confirm the structure.
  • Example 6 Preparation of racemic 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole p-TSA salt: Condition B
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 50 ml addition funnel. The flask was charged with 5 grams of racemic-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 50 ml of DMF. Under continuous stirring, the mixture was heated to a temperature of 75° C. 9.5 grams was added to the 250 ml flask with continuous stirring. Progress of the reaction was monitored by analysis on HPLC.
  • The reaction was continued with stirring for an additional 4 hours at 75° C., after which the solution was brought to room temperature, and stirring was continued for an additional 12 hours. The solution was diluted with 20 ml MTBE and stirred for an additional hour. The precipitated material was collected by filtration and washed with 50 ml MTBE, followed by 3 washes of 25 ml each ethanol, and the precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 2.9 grams, representing a 25.6% yield. HPLC was used to determine the chemical purity of the racemic 2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 98.3%, and the chiral purity showed a 1:1 mixture of the (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. 1H NMR was used to confirm the structure.
  • Example 7 Preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition C
  • A 12 L, three necked flask was equipped with an overhead stirrer, a temperature probe, a heating mantle, a claisen joint, a condenser, and a 500 ml addition funnel. The flask was charged with 250 grams of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 2 L of dimethyl formamide (DMF). Under continuous stirring, the mixture was heated to a temperature of 65° C. The addition funnel was charged with a solution of 386.6 grams propyl tosylate and 322 ml diisopropylethyleamine in 500 ml DMF. This solution was added to the 12 L flask dropwise over a period of 2.0 hours. The reaction was monitored by analysis on HPLC.
  • The reaction was continued at 65° C. for an additional 5 hours, after which the solution was gradually cooled to room temperature and stirred overnight. The solution was diluted with 2 L MTBE and stirred for an additional 0.5 hours. The precipitated material was collected by filtration and washed with 500 ml MTBE, followed by 3 washes of 500 ml each reagent alcohol. The washed precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 317.6 grams, representing a 56% yield. HPLC was used to determine the chemical purity of the (6R)-2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 98.4% and the chiral purity as greater than 99.8%. 1H NMR and 13C NMR was used to confirm the structure: 1H NMR (300 MHz, DMSO-d6) δ 8.5 (br.s, 2H), 7.5 (d, 2H), 71.2 (d, 1H), 6.8 (s, 2H), 3.4 (m, 1H), 2.95 (m, 3H), 2.6 (m, 2H, merged with DMSO peak), 2.3 (s, 3H), 2.15 (m, 1H), 1.8 (m, 1H), 1.55 (m, 2H), 0.9 (t, 3H); 13C NMR (300 MHz, DMSO-d6) δ 167.0, 145.5, 144.6, 138.4, 128.6, 125.8, 110.7, 53.9, 46.5, 25.8, 25.6, 24.5, 21.2, 19.6, 11.3.
  • Example 8 Preparation of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition C
  • A 500 ml, three necked flask was equipped with an overhead stirrer, a temperature probe, a heating mantle, a claisen joint, a condenser, and a 100 ml addition funnel. The flask was charged with 20 grams of (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 180 ml of dimethyl formamide (DMF). Under continuous stirring, the mixture was heated to a temperature of 65° C. The addition funnel was charged with a solution of 35.5 grams propyl tosylate and 32.8 ml diisopropylethylamine in 40 ml DMF. This solution was added to the 500 ml flask dropwise over a period of 2.0 hours. The reaction was monitored by analysis on HPLC.
  • The reaction was continued at 65° C. for an additional 10 hours, after which the solution was gradually cooled to room temperature and stirred for 6 hours. The solution was diluted with 220 ml MTBE and stirred for an additional 0.5 hours. The precipitated material was collected by filtration and washed with 50 ml MTBE, followed by 3 washes of 50 ml each reagent alcohol and a wash with 75 ml of heptane. The washed precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 25.4 grams, representing a 56% yield. HPLC was used to determine the chemical purity of the (6S)-2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 99.4% and the chiral purity as greater than 99.7%. 1H NMR and 13C NMR was used to confirm the structure: 1H NMR (300 MHz, DMSO-d6) δ 8.5 (br.s, 2H), 7.5 (d, 2H), 71.2 (d, 1H), 6.8 (s, 2H), 3.4 (m, 1H), 2.95 (m, 3H), 2.6 (m, 2H, merged with DMSO peak), 2.3 (s, 3H), 2.15 (m, 1H), 1.8 (m, 1H), 1.55 (m, 2H), 0.9 (t, 3H).
  • Example 9 Preparation of racemic 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole p-TSA salt: Condition C
  • A 250 ml, three necked flask was equipped with a magnetic stirrer, a temperature probe, a heating mantle, a claisen joint, a reflux condenser, and a 50 ml addition funnel. The flask was charged with 5 grams of racemic-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 45 ml of DMF. Under continuous stirring, the mixture was heated to a temperature of 65° C. The addition funnel was charged with a solution of 8.86 grams propyl tosylate and 8.2 ml of diisopropylethylamine in 10 ml. DMF. This solution was added dropwise to the 250 ml flask with continuous stirring over a period of 2 hours. Progress of the reaction was monitored by analysis on HPLC.
  • The reaction was continued with stirring for an additional 6 hours at 65° C., after which the solution was brought to room temperature. The solution was diluted with 70 ml MTBE and stirred for an additional hour. The precipitated material was collected by filtration and washed with 15 ml MTBE, followed by 2 washes of 15 ml each ethanol, and a wash with 15 ml heptane. The washed precipitated cake was dried under high vacuum.
  • The final weight of the dried product was 6.02 grams, representing a 53.1% yield. HPLC was used to determine the chemical purity of the racemic 2,6-diamino-4,5,6,7-tetrahydro-benzothiazole as 99.2%, and the chiral purity showed a 1:1 mixture of the (6R) and (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. 1H NMR was used to confirm the structure.
  • Example 10 Preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition E
  • A 1000 ml, three necked flask was equipped with an overhead stirrer, a temperature probe, a heating mantle, a claisen joint, a condenser, and a 250 ml addition funnel. The flask was charged with 25 grams of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 200 ml of dimethyl formamide (DMF). Under continuous stirring, the mixture was heated to a temperature of 75° C. The addition funnel was charged with a solution of 39.5 grams propyl tosylate and 32.5 ml diisopropylethyleamine in 50 ml DMF. This solution was added to the 1000 ml flask dropwise over a period of 1.0 hours. The reaction was monitored by analysis on HPLC.
  • The reaction was continued at 75° C. for an additional 5 hours, after which the solution was gradually cooled to room temperature and stirred overnight. The precipitated material was collected by filtration and washed with 2 washes with 0.100 ml MTBE, followed by 3 washes of 75 ml each reagent alcohol and one wash with 125 ml of heptane. The washed precipitated cake was dried under high vacuum.
  • The reaction resulted in a 47% yield. HPLC was used to determine the chiral purity as 99.8%. 1H NMR and 13C NMR was used to confirm the structure.
  • Example 11 Preparation of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition E
  • A 1000 ml, three necked flask was equipped with an overhead stirrer, a temperature probe, a heating mantle, a claisen joint, a condenser, and a 250 ml addition funnel. The flask was charged with 25 grams of (6S)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole, followed by 200 ml of dimethyl formamide (DMF). Under continuous stirring, the mixture was heated to a temperature of 75° C. The addition funnel, was charged with a solution of 39.5 grams propyl tosylate and 32.5 ml diisopropylethyleamine in 50 ml DMF. This solution was added to the 1000 ml flask dropwise over a period of 2.0 hours. The reaction was monitored by analysis on HPLC.
  • The reaction was continued at 65° C. for an additional 5 hours, after which the solution was gradually cooled to room temperature and stirred overnight. The precipitated material was collected by filtration and washed with twice with 10 ml MTBE, followed by 3 washes of 75 ml each reagent alcohol and one wash with 125 ml of heptane. The washed precipitated cake was dried under high vacuum.
  • The reaction resulted in a 47% yield. HPLC was used to determine the chiral purity as 99.8%. 1H NMR and 13C NMR was used to confirm the structure.
  • TABLE 1
    Experiments for SN2 preparation of 2-amino-4,5,6,7-tetrahydro-6-
    (propylamino)benzothiazole pTSA salt
    Condition Isomer Batch Size Results
    Ex. 1 (6R) 45 grams Yield = 53.2 grams (52%)
    Chemical Purity = 98.2%
    AUC by HPLC
    Chiral Purity = >99.5% AUC
    by HPLC
    Ex. 2 (6S) 5 grams Yield = 4.99 grams (44.2%)
    Chemical Purity = 98.0% AUC
    by HPLC
    Chiral Purity = >99.6% AUC
    by HPLC
    Ex. 3 Racemic 5 gram Yield = 5.12 grams (45%)
    Chemical Purity = 97.1% AUC
    by HPLC
    Chiral Purity = 1:1 (6R):(6S)
    by HPLC
    Ex. 4 (6R) 5 gram Yield = 4.6 grams (40%)
    Chemical Purity = 94.9% AUC
    by HPLC
    Chiral Purity = 99.6% AUC
    by HPLC
    Ex. 5 (6S) 10 gram Yield = 9.81 grams (43.3%)
    Chemical Purity = 94.9% AUC
    by HPLC
    Chiral Purity = 99.7% AUC
    by HPLC
    Ex. 6 Racemic 5 gram Yield = 2.9 grams (25.6%)
    Chemical Purity = 98.3% AUC
    by HPLC
    Chiral Purity = 1:1 (6R):(6S)
    by HPLC
    Ex. 7 (6R) 250 gram Yield = 317.6 grams (56%)
    Chemical Purity = 99.4% AUC
    by HPLC
    Chiral Purity = 99.8% AUC
    by HPLC
    Ex. 8 (6S) 20 gram Yield = 25.41 grams (56%)
    Chemical Purity = 99.4% AUC
    by HPLC
    Chiral Purity = 99.7% AUC
    by HPLC
    Ex. 9 Racemic 5 gram Yield = 6.02 grams (53.1%)
    Chemical Purity = 99.2% AUC
    by HPLC
    Chiral Purity = 1:1 (6R):(6S)
    by HPLC
    Ex. 10 (6R) 25 gram Yield = 47%
    Chiral Purity = 99.8% AUC
    by HPLC
    Ex. 11 (6S) 25 gram Yield = 47%
    Chiral Purity = 99.8% AUC
    by HPLC
  • Example 11
  • Various ratios of entantiomerically enriched mixtures of (6R)2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and (6S)2,6 diamino-4,5,6,7-tetrahydro-benzothiazole having (6R) to (6S) ratios of 80:20, 20:80, 85:15, 15:85, 90:10, 10:90, 95:5 and 5:95, were used to prepare (6R) or (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine based on the enriched species in the starting material. The following reaction conditions were used:
  • F: 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole mixtures were dissolved in 10 volumes of DMF and 1.25 equivalents of propyl tosylate at 65-67° C. The reaction is then cooled to room temperature, insoluble species were collected and washed with 8 volumes of MTBE.
  • G: 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole mixtures were dissolved in 18 volumes of DMF and 1.25 equivalents of propyl tosylate at 65-67° C. The reaction is then cooled to room temperature, insoluble species were collected and washed with 8 volumes of MTBE.
  • H: 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole mixtures were dissolved in 10 volumes of DMF and 1.25 equivalents of propyl tosylate at 65-67° C. The reaction is then cooled to room temperature and insoluble species were collected. No washing step was performed.
  • Results are compiled in Table 2:
  • TABLE 2
    Experiments for S N2 preparation of pure enantiomers of 2-amino-
    4,5,6,7-tetrahydro-6-(propylamino)benzothiazole
    Ratio of Condition G Condition H
    starting diamines Condition F (yield/ (yield/
    (6R):(6S) (yield/chiral purity) chiral purity) chiral purity)
    80:20 29%/99%   34%/98.2%
    20:80 30%/99.4% 35%/95.7%
    85:15 43%/86.8% 36%/99.8% 39%/99.9%
    15:85 52%/88.9% 27%/99.6% 37%/99.9%
    90:10 47%/95.9%
    10:90 58%/93.6%
    95:5  50%/99.6%
     5:95 47%/99.6%
  • The data in Table 2 demonstrates that both enantiomers of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole have similar, if not the same, solubility. Further, the data shows that the synthesis is equally efficient for either enantiomer of 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole. These data also demonstrate that the enantiomers behave independently of one another, in that the solubility of one enantiomer appears to be unaffected by the concentration in solution of the other. For example, the various synthesis reactions carried out using condition F all have chemical yields of about 50%, independent of the percentage of predominant diamine enantiomer of the starting material. When the volume of the organic solvent used in the synthesis reaction is increased, the chemical yield is reduced, but the chiral yield is increased. This is apparent by comparison of the reaction carried out in conditions F and G, where an 85:15 ratio of (6R):(6S) diamine produced a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product having an 86.8% chiral purity for the (6R) enantiomer when the reaction used 10 volumes of the organic solvent and a 99.8% chiral purity for the (6R) enantiomer when the reaction used 1.8 volumes of the organic solvent. Note also that the chemical yield was reduced in the reaction using a larger volume of organic solvent (43% yield in condition F and 36% yield in condition G).
  • In Table 3, condition H is the same as condition F, except that the recovery step does not incorporate dilution in MTBE. The MTBE is observed to increase 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole recovery (yield) from the synthesis reaction, but may reduce the overall chiral purity. This is born out by a comparison of the results for trials carried out in an 85:15 ratio of (6R):(6S) diamine, which produced a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product having a 86.8% chiral purity for the (6R) enantiomer when the reaction included the MTBE organic solvent and a 99.9% chiral purity for the (6R) enantiomer when the reaction did not include the MTBE organic solvent. The chemical yield was reduced by exclusion of the MTBE dilution in the recovery step; a 43% yield in condition C as opposed to a 39% yield in condition E.
  • Example 12 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride
  • (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt (50 grams; 0.13 mol) was taken into 150 ml absolute ethanol and cooled to between 0 and 5° C. with continuous stirring. Concentrated HCl (33 ml) was slowly added to the reaction while maintaining the temperature at between 0 and 5° C., and the mixture was stirred for an additional 15 minutes. MTBE (200 ml) was added to the mixture, and stirring was continued for an additional 1.5 hours at temperature. The reaction mixture was then filtered, washed twice with an MTBE/ethanol solution (2:1, 2×50 ml wash volumes), and dried under vacuum at 30° C. overnight. The final product was 34 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, indicative of 92% yield, and a 97.3% chemical purity as determined by HPLC.
  • Example 13 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride
  • (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt (10 grams; 0.026 mol) was dissolved in 200 ml IPAC and cooled to 15° C. with continuous stirring. HCl gas was bubbled into the slurry for 1 hour. The mixture was then filtered, washed with IPAC, and dried overnight under vacuum at room temperature. The final product was 6.8 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, indicative of 92% yield, and a 97% chemical purity as determined by HPLC.
  • Example 14 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine free base
  • (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt (25 grams; 0.065 mol) was dissolved in 200 ml DCM and mixed into a slurry. 10 ml of water was added and the mixture was basified with 12 ml of 6N NaOH to a pH of 11-12. The two phases were split, and the aqueous was extracted with 200 ml of DCM. The combined organic phases were dried over MgSO4, filtered over Celite® and concentrated. The residue was dissolved in 100 ml MTBE and slurried for several hours. The solids were then filtered, washed with MTBE and dried under vacuum at 35° C. The final product was 9.1 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, indicative a of 66% yield, and a 98% chemical purity as determined by HPLC.
  • Example 15 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine free base
  • Freebase formation was performed on a 200 gram scale. A 5 L, three necked, round-bottomed flask, equipped with an over head stirrer, thermometer, and addition funnel was charged with 200 g (0.522 mol) of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt and 1 L of water. The mixture was stirred and cooled to 10° C. The slurry was basified to a pH of about 11-12 by the slow addition of 200 ml of 6 N NaOH over a period of 15 min. The reaction mixture was diluted with 500 ml of brine (sodium chloride dissolved in water) and extracted with 3×1 L of dichloromethane. The combined organic phases were washed with 1.0 L of brine, dried over MgSO4, filtered and concentrated to dryness. The residue was triturated with 1 L of 1:1 IPAC:Heptane, the resulting slurry was stirred for 1 hour, filtered and the filter cake was washed with 2×250 ml of 1:1 mixture of IPAC:Heptane. The filter cake was collected and dried at 40° C. under high vacuum for 24 hours to give 94.1 grams (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine (85.5%) as a white solid. The chemical purity was 100% AUC as tested by HPLC, and the chiral purity was 100% AUC as tested by HPLC. 1H NMR and 13C NMR was used to confirm the structure: 1H NMR (300 MHz, DMSO-δ6) δ 6.6 (s, 2H), 2.8 (m, 2H), 2.5 (m, 2H, merged with DMSO peak), 2.2 (m, 1H), 1.9 (m, 1H), 1.5-1.3 (m, 4H), 0.85 (t, 3H); 13C NMR (300 MHz, DMSO-d6) δ 166.2, 144.8, 113.6, 54.2, 49.1, 30.0, 29.6, 25.2, 23.5, 12.3.
  • Example 16 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine free base to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride
  • The freebase of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine (4.8 grams; 0.022 mol) was dissolved in 200 ml of IPAC and cooled to 15° C. HCl gas was bubbled into the slurry for 1 hour. The mixture was then filtered, washed with IPAC and dried under vacuum at room temperature overnight. The final product was 6.4 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, indicative of 100% yield, and a 97% chemical purity as determined by HPLC.
  • Example 17 Conversion of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine free base to (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride
  • The freebase of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine (50 grams; 0.13 mol) was dissolved in 500 ml of IPAC. Under continuous stirring, the mixture was slowly charged with 78 ml of concentrated HCl at a temperature of 25° C. The mixture was stirred overnight at ambient conditions (˜25° C.), filtered and dried under vacuum at 40° C. The final product was 68 grams of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, indicative of 95% yield.
  • Example 18 Optical purification of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine using achiral acid addition
  • 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole entantio enriched for the (6R) enantiomer (˜300 mg) was dissolved in 10 ml of the chosen solvent at 75° C. Complete dissolution was observed in all samples. Acid addition was made at 1.05 molar equivalents for the p-TSA (solvent is ethanol; 2.97 ml of 0.5 M acid) and MSA (solvent is acetonitrile; 1.49 ml of 1.0 M acid), and 2.05 molar equivalents for the fumaric (solvent is acetonitrile; 5.84 ml of 0.5 M acid) and phosphoric (solvent is acetonitrile; 2.90 ml of 1.0 M acid). The reaction mixtures were cooled to room temperature at a rate of 25° C./hour and stirred at room temperature for an additional 19 hours.
  • The products of purification processes were then analyzed by HPLC for chemical and chiral purity. FIG. 3A shows an exemplary HPLC trace starting material. In FIG. 4A, a large (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine peak can be observed at about 6 minutes and a much smaller (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine peak can be seen at about 9 minutes. The area of these peaks provides an estimated composition for the mixture which is shown in the table below the trace, and shows the mixture as containing about 902% (6R) and 8.8% (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. FIG. 3B shows an exemplary trace of the 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole product following purification. In FIG. 3B, a large (6R)2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole peak is observed at about 6 minutes, and no (6S)2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole peak is observed. It is additionally of note that several other minor peaks are also reduced or eliminated in the product trace, and the table below the trace indicates that (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine makes up 99.9% of the product solution which is within the limits of analytical detectability. Results for each exemplary reaction are provided in Table 3:
  • TABLE 3
    Experiments for preparation of the (6R)-4,5,6,7-tetrahydro-N6-propyl-
    2,6-benzothiazole-diamine
    Salt/Acid Solvent Batch Size Results
    p-TSA ethanol 298.7 mg Yield = 489.5 mg (90.3%)
    Start Chiral Purity = 91% AUC (6R) by HPLC
    Final Chiral Purity = 100% AUC by HPLC
    MSA acetonitrile 300.0 mg Yield = 431.8 mg (98.9%)
    Start Chiral Purity = 91% AUC (6R) by HPLC
    Final Chiral Purity = 99.23% AUC by HPLC
    fumaric acetonitrile 301.0 mg Yield = 532 mg (84.2%)
    (hot ethanol) Start Chiral Purity = 91% AUC (6R) by HPLC
    Final Chiral Purity = 99.26% AUC by HPLC
    phosphoric acetonitrile 299.4 mg Yield = 592 mg (~100%)
    Start Chiral Purity = 91% AUC (6R) by HPLC
    Final Chiral Purity = 100% AUC by HPLC
  • The solids obtained by this trituration step were isolated by filtration and dried under high vacuum at room temperature. These products were analyzed by HPLC, NMR, thermal gravimetric analysis, differential scanning calorimetry, X-ray powder diffraction (XPRD), Fourier transform infrared spectroscopy and moister-sorption analysis. The XPRD patterns showed that the p-TSA, MSA and fumarate salt forms of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine were crystalline, while the phosphate salt form of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine was amorphous.
  • Example 19 Industrial Scale Resolution of Racemic Diamine
  • A 72 L, unjacketed reactor was charged with racemic 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole (4.5 kg; 26.6 mol) and 58.5 L water, and heated as a suspension to a temperature of about 60° C. to 65° C. Resolution of the enantiomers was achieved by addition of one equivalent of (D)-(−)-Tartaric acid (3991 grams; 26.6 mol) in 4.5 L of water, after which the resulting solution was heated to a temperature of about 70° C. to 75° C. and maintained at this temperature for about 1 hour. The mixture was allowed to cool to a temperature of about 20° C. to 25° C. and stirred for an additional 15 hours, after which the mixture was filtered and the solids were washed 3× with water (6.3 L each wash).
  • The wet solids, which contain the (6R) enantiomer of the diamine, were charged to the reactor followed by 54 L of water, and the mixture was heated to a temperature of about 70° C. to 75° C. for 2 hours. The mixture was allowed to cool to a temperature of about 20° C. to 25° C. and stirred for 17 hours. The mixture was then filtered and the solids were washed 2× with water (4.5 L each wash). The wet solids were transferred to a jacketed reactor and the reactor was charged with 8.1 L of water. The mixture was cooled to a temperature of about 0° C. to 5° C. and cautiously charged with concentrated 1.6 L of HCl, followed by 1.2 L of 50% NaOH to achieve a pH of about 9-10. During the addition, the temperature was maintained at about 0° C. to 5° C., and stirred for an additional hour at temperature. The resulting mixture was then filtered and the solids were washed 2× with cold (0° C. to 5° C.) water (1.1 L each wash). The solids were transferred to a jacketed reactor and were reslurried once more with 4.5 L of water at 0° C. to 5° C. The solids were filtered and dried under warm air (40° C. to 45° C.) to give 1940 grams of the product ((6R) diamine) as a white solid, with an 86% yield for the (6R) enantiomer.
  • The mother liquors of the initial resolution step, which contain the (6S) enantiomer of the diamine, were concentrated to afford diamine with a 95.5% yield for the (6S) enantiomer. Results of reactions performed using 1000, 4500 and 41.00 grams of starting material are provided in Table 4
  • TABLE 4
    Experiments for industrial scale resolution of the (6R) enantiomer of
    diamine
    Input Yield (%) of Chemical Purity Chiral Purity
    (grams) (6R) enantiomer (AUC % by HPLC) (AUC % by HPLC)
    1000 76 >99 98.3
    4500 86 >99 98.5
    4100 54 >99 98.5
  • Example 20 Industrial Scale Preparation of Propyl Tosylate
  • A 100 L glass, jacketed reactor was charged with 1-propanol (2.098 kg; 34.9 mol), triethylamine (4.585 kg; 45.3 mol; 1.3 equivalents) and DCM (20.1 L). The mixture was cooled to a temperature of about 5° C. to 15° C. and cautiously charged with a solution of p-toluenesulfonyl chloride (6 kg; 31.47 mol; 0.9 equivalents) in DCM (10.5 L) over 30 minutes. Once the addition was complete, the mixture was warmed to a temperature of about 18° C. to 22° C. and stirred for 12 hours. The reaction mixture was assayed by 1H NMR (in CDCl3) and deemed complete. HCl (6 N; 2.98 L) was cautiously charged while maintaining the temperature below 25° C. The aqueous phase was removed, and the organic phase was washed 2× with water (21 L each wash), dried with MgSO4, and filtered over Celite®. The filtered solids were then washed with DCM (4 L) and concentrated to a residue. The residue was dissolved in heptane and concentrated again to afford a final propyl tosylate product (6.385 kg, 95% yield).
  • Example 21 Industrial scale preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine p-TSA salt: Condition C
  • A 72 liter unjacketed reactor was charged with 1.84 kg (10.87 mol) of (6R)-2,6 diamino-4,5,6,7-tetrahydro-benzothiazole ((6R) diamine), followed by 14.7 L of dimethyl formamide (DMF). Under continuous stirring, the mixture was heated to a temperature of between 65° C. and 68° C. A solution of 2926 grams propyl tosylate and 1761 grams diisopropylethyleamine in 3.5 L DMF was added slowly over a period of 2 hours. The reaction was continued at 67° C. for an additional 4 hours, after which the solution was gradually cooled to room temperature (18° C. to 22° C.) and stirred for an additional 15 hours. The solution was diluted with 14.7 L of MTBE over a time period of 30 minutes, and stirred for an additional 1 hour. The precipitated material was collected by filtration and washed with 7.3 L MTBE, followed by 3 washes of 3.7 L each of ethanol, and a wash with 9.2 L heptane. The washed precipitated cake was dried under high vacuum at 30° C. to 35° C. The final weight of the dried product was 2090 grams, representing a 50% yield.
  • Example 22 Purity of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine
  • The Limit of Quantitation (LOQ) to 0.05% precision was determined from six replicate preparations of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine. The RSD was measured to be 6.3% and the S/N (signal to noise ratio) was measured to be 61:1. The pre-defined acceptance criteria at LOQ was to have a S/N>10:1 and for the RSD to be <20.0%. These results exceeded the acceptance criteria by a wide margin.
  • Signal-to-noise levels can vary for numerous reasons, including pump behavior, air in lines, extent of mobile phase degassing, system-to-system variations and electronic fluctuations. The LOD had previously been estimated to be 0.03% based upon the 0.1% preparation which produced a S/N of 30:1. Although the recent 0.05% levels produced a S/N of 61:1, based upon the overall history of the method, the estimated LOD will remain at the stated 0.03%.
  • Linearity was determined over the range 0.05%-150% of nominal (0.2 μg/mL-600 μg/mL). The correlation coefficient over this expanded range was determined to be 0.9999. This exceeded the pre-defined linearity acceptance criterion of >0.995.
  • The weight percent assay is on an as-is basis versus the current standard which has a purity of 94.0%. Each sample was prepared in duplicate with single injections. Purity data is provided in the Table 5:
  • TABLE 5
    (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine Purity Data
    %
    Sample % area ave wt/wt ave
    Sample 118 100.0 95.1
    Sample 118 100.0 100.0 95.0 95.1
    Sample 105 100.0 94.7
    Sample 105 100.0 100.0 94.7 94.7
    Sample 061 100.0 94.5
    Sample 061 100.0 100.0 94.6 94.5
    Sample 326A 100.0 95.0
    Sample 326A 100.0 100.0 94.7 94.8
  • Example chromatographs are provided in FIG. 4A-D. Specifically, FIG. 4A is an HPLC Chromatograph of Sample 118; FIG. 4B is an HPLC Chromatograph of Sample 105; FIG. 4C is an HPLC Chromatograph of Sample 061; and FIG. 4 D is an HPLC Chromatograph of Sample 326A. These data show preparation of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine at 100% purity.
  • Example 23 Propyl tosylate concentration of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine
  • A solid phase extraction (SPE) procedure was developed to remove the high levels of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine prior to analysis while maintaining recovery of propyl tosylate spiked at 1.5 ppm. The SPE cartridges (Supelco Discovery DSC-18, 6 mL, 1 g) are pre-activated and washed with 6 mL acetonitrile (MeCN) followed by 6 mL of water. Five milliliters of 100 mg/mL solutions of KNS-760704 prepared in 5:95, MeCN/water with 0.5% phosphoric acid are then introduced into the SPE cartridges. The acid sufficiently maintains the polarity of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine so that it can be readily washed from the SPE cartridge with an additional 5 mL of 5:95, MeCN/water with 0.5% phosphoric acid while retaining any propyl tosylate. Any propyl tosyate is then eluted from the SPE cartridges using 5 mL of 95:5 MeCN/water. Fortunately, due to the sensitivity gained from method development experiments, no further sample enrichment is needed and the samples are analyzed as-is. The samples are compared to a standard of 1.5 ppm (0.15 μg/mL) propyl tosylate prepared in 95:5 MeCN/water.
  • An exemplary UV spectrum of propyl tosylate eluted from the SPE cartridges is shown in FIG. 5.
  • Exemplary HPLC data from of the (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine eluted from the SPE cartridge is provided in FIGS. 6A and 6B. FIG. 6A shows an HPLC chromatograph of a propyl tosylate standard prepared from 100 mg of propyl tosylate. FIG. 6B shows an HPLC chromatograph of a 1 g sample of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine prepared by the method of embodiments of the invention. No propyl tosylate is evident based on the data provided in FIG. 6B as indicated by the absence of a peak corresponding to propyl tosylate (right).
  • Example 24 Chiral purity of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine
  • The chirality of the (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine prepared as described above was tested using a Chiralpak IA column under HPLC conditions. About 25 mg of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine were applied to the column. Exemplary HPLC traces of a (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine standard are provided in FIG. 7A and an HPLC trace of a sample (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine is provided in FIG. 7B. Test data is provided in Table 6.
  • TABLE 6
    % Recovery of (6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-
    diamine
    Level Preparation % Recovery
    100% 1 91.13
    2 91.18
    3 97.61
    Average 93.3
    % RSD 4.0
  • Although the present invention has been described in considerable detail with reference to certain preferred embodiments thereof, other versions are possible. Therefore the spirit and scope of the appended claims should not be limited to the description and the preferred versions contained within this specification.

Claims (17)

1. A process for preparing a chirally purified substituted 4,5,6,7,-tetrahydro-benzothaizole diamine comprising:
heating a solution comprising entantiomerically enriched 4,5,6,7-tetrahydro-benzothiazole diamine of general formula (I):
Figure US20140100372A1-20140410-C00007
wherein:
R1 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkenyl or alkynyl group each having 3 to 6 carbon atoms, an alkanoyl group having 1 to 6 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the above-mentioned phenyl nuclei may be substituted by 1 or 2 halogen atoms;
R2 represents a hydrogen atom or an alkyl group with 1 to 4 carbon atoms;
R3 represents a hydrogen atom, an alkyl group with 1 to 7 carbon atoms, a cycloalkyl group having 3 to 7 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms, an alkanoyl group having 1 to 7 carbon atoms, a phenyl alkyl or phenyl alkanoyl group having 1 to 3 carbon atoms in the alkyl part, whilst the phenyl nucleus may be substituted by fluorine, chlorine or bromine atoms,
R4 represents a hydrogen atom, an alkyl group with 1 to 4 carbon atoms, an alkenyl or alkynyl group having 3 to 6 carbon atoms; and
at least one or R1, R2, R3 or R4 is a hydrogen in an organic solvent; and
an alkyl sulfonate or an alkyl halide in a solvent to form a reaction mixture;
reacting the reaction mixture; and
recovering a chirally purified substituted 4,5,6,7-tetrahydro-benzothiazole diamine.
2-28. (canceled)
29. Chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process comprising:
heating a solution comprising entantiomerically enriched 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole and a propyl halide or a propyl sulfonate to form a reaction mixture;
reacting the reaction mixture; and
recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
30-52. (canceled)
53. A process for preparing a chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole comprising:
heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole in an organic solvent;
adding to the heated solution propyl sulfonate or a propyl halide to form a reaction mixture; and
reacting the reaction mixture for up to about 12 hours.
54-79. (canceled)
80. A process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole comprising:
heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole;
adding a propyl halide or a propyl sulfonate to the heated solution slowly over from about 0.5 hours to about 2 hours to form a reaction mixture;
reacting the reaction mixture; and
recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
81-101. (canceled)
102. Chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process comprising:
heating a solution comprising 2,6 diamino-4,5,6,7-tetrahydro-benzothiazole;
adding a propyl halide or a propyl sulfonate to the heated solution slowly over from about 0.5 hours to about 2 hours to form a reaction mixture;
reacting the reaction mixture; and
recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
103-125. (canceled)
126. A process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole comprising:
dissolving entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in an organic solvent to form a solution;
heating the solution to from about 50° C. to about 125° C.;
adding an acid to the solution to form a reaction mixture; and
recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
127-128. (canceled)
129. A chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process of claim 126.
130. A process for preparing chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole comprising:
dissolving entantiomerically enriched 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole in an organic solvent to form a solution;
heating the solution to from about 50° C. to about 125° C.;
adding an achiral salt to the solution to form a reaction mixture; and
recovering the chirally purified 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole.
131-132. (canceled)
133. A chirally pure 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole prepared by a process of claim 130.
134. A process for preparing a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole dihydrochloride comprising:
dissolving a 2-amino-4,5,6,7-tetrahydro-6-(propylamino)benzothiazole salt in an organic solvent to form a solution;
cooling the solution to a temperature of from about 0° C. to about 5° C.;
adding concentrated HCl and an organic solvent to the cooled solution; and
stirring the solution at a temperature of about 0° C. to about 5° C.
US13/973,987 2007-03-14 2013-08-22 Synthesis of chirally purified substituted benzothiazole diamines Abandoned US20140100372A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/973,987 US20140100372A1 (en) 2007-03-14 2013-08-22 Synthesis of chirally purified substituted benzothiazole diamines
US15/369,230 US10179774B2 (en) 2007-03-14 2016-12-05 Synthesis of chirally purified substituted benzothiazole diamines

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US89482907P 2007-03-14 2007-03-14
US89481407P 2007-03-14 2007-03-14
US12/049,235 US8519148B2 (en) 2007-03-14 2008-03-14 Synthesis of chirally purified substituted benzothiazole diamines
US13/973,987 US20140100372A1 (en) 2007-03-14 2013-08-22 Synthesis of chirally purified substituted benzothiazole diamines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/049,235 Continuation US8519148B2 (en) 2007-03-14 2008-03-14 Synthesis of chirally purified substituted benzothiazole diamines

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/369,230 Continuation US10179774B2 (en) 2007-03-14 2016-12-05 Synthesis of chirally purified substituted benzothiazole diamines

Publications (1)

Publication Number Publication Date
US20140100372A1 true US20140100372A1 (en) 2014-04-10

Family

ID=39760428

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/049,235 Active 2031-06-11 US8519148B2 (en) 2007-03-14 2008-03-14 Synthesis of chirally purified substituted benzothiazole diamines
US13/973,987 Abandoned US20140100372A1 (en) 2007-03-14 2013-08-22 Synthesis of chirally purified substituted benzothiazole diamines
US15/369,230 Active 2028-05-08 US10179774B2 (en) 2007-03-14 2016-12-05 Synthesis of chirally purified substituted benzothiazole diamines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/049,235 Active 2031-06-11 US8519148B2 (en) 2007-03-14 2008-03-14 Synthesis of chirally purified substituted benzothiazole diamines

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/369,230 Active 2028-05-08 US10179774B2 (en) 2007-03-14 2016-12-05 Synthesis of chirally purified substituted benzothiazole diamines

Country Status (9)

Country Link
US (3) US8519148B2 (en)
EP (1) EP2137171A4 (en)
JP (1) JP2010521496A (en)
CN (1) CN101715443A (en)
AU (1) AU2008224844B2 (en)
CA (1) CA2681110A1 (en)
IL (1) IL200925A0 (en)
RU (1) RU2454409C2 (en)
WO (1) WO2008113056A2 (en)

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096537B1 (en) 2014-12-31 2015-08-04 Mahesh Kandula Compositions and methods for the treatment of mucositis
US9102649B1 (en) 2014-09-29 2015-08-11 Mahesh Kandula Compositions and methods for the treatment of multiple sclerosis
US9108942B1 (en) 2014-11-05 2015-08-18 Mahesh Kandula Compositions and methods for the treatment of moderate to severe pain
US9150557B1 (en) 2014-11-05 2015-10-06 Cellix Bio Private Limited Compositions and methods for the treatment of hyperglycemia
US9173877B1 (en) 2014-11-05 2015-11-03 Cellix Bio Private Limited Compositions and methods for the treatment of local pain
US9175008B1 (en) 2014-11-05 2015-11-03 Cellix Bio Private Limited Prodrugs of anti-platelet agents
US9174931B2 (en) 2013-06-04 2015-11-03 Cellix Bio Private Limited Compositions for the treatment of diabetes and pre-diabetes
US9187427B2 (en) 2012-08-03 2015-11-17 Cellix Bio Private Limited N-substituted nicotinamide compounds and compositions for the treatment migraine and neurologic diseases
US9206111B1 (en) 2014-12-17 2015-12-08 Cellix Bio Private Limited Compositions and methods for the treatment of neurological diseases
US9227974B2 (en) 2012-05-23 2016-01-05 Cellex Bio Private Limited Compositions and methods for the treatment of respiratory disorders
US9233161B2 (en) 2012-05-10 2016-01-12 Cellix Bio Private Limited Compositions and methods for the treatment of neurological conditions
US9242939B2 (en) 2012-05-10 2016-01-26 Cellix Bio Private Limited Compositions and methods for the treatment of respiratory disorders
US9266823B2 (en) 2012-05-08 2016-02-23 Cellix Bio Private Limited Compositions and methods for the treatment of parkinson's disease
US9273061B2 (en) 2012-05-10 2016-03-01 Cellix Bio Private Limited Compositions and methods for the treatment of chronic pain
US9284287B1 (en) 2014-11-05 2016-03-15 Cellix Bio Private Limited Compositions and methods for the suppression of carbonic anhydrase activity
US9290486B1 (en) 2014-11-05 2016-03-22 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy
US9303038B2 (en) 2011-09-06 2016-04-05 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy and neurological diseases
US9309233B2 (en) 2012-05-08 2016-04-12 Cellix Bio Private Limited Compositions and methods for the treatment of blood clotting disorders
US9315478B2 (en) 2012-05-10 2016-04-19 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9315461B2 (en) 2012-05-10 2016-04-19 Cellix Bio Private Limited Compositions and methods for the treatment of neurologic diseases
US9321716B1 (en) 2014-11-05 2016-04-26 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9321775B2 (en) 2012-05-10 2016-04-26 Cellix Bio Private Limited Compositions and methods for the treatment of moderate to severe pain
US9333187B1 (en) 2013-05-15 2016-05-10 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory bowel disease
US9339484B2 (en) 2012-05-10 2016-05-17 Cellix Bio Private Limited Compositions and methods for the treatment of restless leg syndrome and fibromyalgia
US9346742B2 (en) 2012-05-10 2016-05-24 Cellix Bio Private Limited Compositions and methods for the treatment of fibromyalgia pain
US9394288B2 (en) 2012-05-10 2016-07-19 Cellix Bio Private Limited Compositions and methods for the treatment of asthma and allergy
US9399634B2 (en) 2012-05-07 2016-07-26 Cellix Bio Private Limited Compositions and methods for the treatment of depression
US9403857B2 (en) 2012-05-10 2016-08-02 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9403826B2 (en) 2012-05-08 2016-08-02 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory disorders
US9434729B2 (en) 2012-05-23 2016-09-06 Cellix Bio Private Limited Compositions and methods for the treatment of periodontitis and rheumatoid arthritis
US9434704B2 (en) 2012-05-08 2016-09-06 Cellix Bio Private Limited Compositions and methods for the treatment of neurological degenerative disorders
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US9492409B2 (en) 2012-05-23 2016-11-15 Cellix Bio Private Limited Compositions and methods for the treatment of local pain
US9499526B2 (en) 2012-05-10 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of neurologic diseases
US9499527B2 (en) 2012-05-10 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of familial amyloid polyneuropathy
US9498461B2 (en) 2012-05-23 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory bowel disease
US9512096B2 (en) 2011-12-22 2016-12-06 Knopp Biosciences, LLP Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
US9522884B2 (en) 2012-05-08 2016-12-20 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic disorders
US9573927B2 (en) 2012-05-10 2017-02-21 Cellix Bio Private Limited Compositions and methods for the treatment of severe pain
US9580383B2 (en) 2012-05-23 2017-02-28 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US9624168B2 (en) 2012-09-06 2017-04-18 Cellix Bio Private Limited Compositions and methods for the treatment inflammation and lipid disorders
US9642840B2 (en) 2013-08-13 2017-05-09 Knopp Biosciences, Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
US9642915B2 (en) 2012-05-07 2017-05-09 Cellix Bio Private Limited Compositions and methods for the treatment of neuromuscular disorders and neurodegenerative diseases
US9662313B2 (en) 2013-02-28 2017-05-30 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US9670153B2 (en) 2012-09-08 2017-06-06 Cellix Bio Private Limited Compositions and methods for the treatment of inflammation and lipid disorders
US9725404B2 (en) 2014-10-27 2017-08-08 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US9738631B2 (en) 2012-05-07 2017-08-22 Cellix Bio Private Limited Compositions and methods for the treatment of neurological disorders
US9765020B2 (en) 2012-05-23 2017-09-19 Cellix Bio Private Limited Dichlorophenyl-imino compounds and compositions, and methods for the treatment of mucositis
US9763918B2 (en) 2013-08-13 2017-09-19 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
US9771355B2 (en) 2014-09-26 2017-09-26 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy and neurological disorders
US9849116B2 (en) 2008-08-19 2017-12-26 Knopp Biosciences Llc Compositions and methods of using (R)-pramipexole
US9932294B2 (en) 2014-12-01 2018-04-03 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US10179774B2 (en) 2007-03-14 2019-01-15 Knopp Biosciences Llc Synthesis of chirally purified substituted benzothiazole diamines
US10208014B2 (en) 2014-11-05 2019-02-19 Cellix Bio Private Limited Compositions and methods for the treatment of neurological disorders
US10227301B2 (en) 2015-01-06 2019-03-12 Cellix Bio Private Limited Compositions and methods for the treatment of inflammation and pain
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

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070259930A1 (en) * 2006-04-10 2007-11-08 Knopp Neurosciences, Inc. Compositions and methods of using r(+) pramipexole
US8518926B2 (en) 2006-04-10 2013-08-27 Knopp Neurosciences, Inc. Compositions and methods of using (R)-pramipexole
KR20090021169A (en) * 2006-05-16 2009-02-27 크놉 뉴로사이언시스 인코포레이티드 Compositions of r(+) and s(-) pramipexole and methods of using the same
US8524695B2 (en) 2006-12-14 2013-09-03 Knopp Neurosciences, Inc. Modified release formulations of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and methods of using the same
RU2012101792A (en) * 2009-06-19 2013-07-27 Нопп Ньюросайенсиз, Инк. COMPOSITIONS AND METHODS FOR TREATING LATERAL AMYOTROPHIC SCLEROSIS

Family Cites Families (200)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3598122A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3797494A (en) 1969-04-01 1974-03-19 Alza Corp Bandage for the administration of drug by controlled metering through microporous materials
US3731683A (en) 1971-06-04 1973-05-08 Alza Corp Bandage for the controlled metering of topical drugs to the skin
US4144317A (en) 1975-05-30 1979-03-13 Alza Corporation Device consisting of copolymer having acetoxy groups for delivering drugs
US4031894A (en) 1975-12-08 1977-06-28 Alza Corporation Bandage for transdermally administering scopolamine to prevent nausea
US4201211A (en) 1977-07-12 1980-05-06 Alza Corporation Therapeutic system for administering clonidine transdermally
US4286592A (en) 1980-02-04 1981-09-01 Alza Corporation Therapeutic system for administering drugs to the skin
US4314557A (en) 1980-05-19 1982-02-09 Alza Corporation Dissolution controlled active agent dispenser
US4395859A (en) 1980-06-05 1983-08-02 State Of Illinois, Department Of Transportation Method and apparatus for securing an object to a support structure
US4327725A (en) 1980-11-25 1982-05-04 Alza Corporation Osmotic device with hydrogel driving member
US4379454A (en) 1981-02-17 1983-04-12 Alza Corporation Dosage for coadministering drug and percutaneous absorption enhancer
US4725272A (en) 1981-06-29 1988-02-16 Alza Corporation Novel bandage for administering beneficial drug
US4849226A (en) 1981-06-29 1989-07-18 Alza Corporation Method for increasing oxygen supply by administering vasodilator
US4435180A (en) 1982-05-25 1984-03-06 Alza Corporation Elastomeric active agent delivery system and method of use
US4559222A (en) 1983-05-04 1985-12-17 Alza Corporation Matrix composition for transdermal therapeutic system
US5082668A (en) 1983-05-11 1992-01-21 Alza Corporation Controlled-release system with constant pushing source
US4612008A (en) 1983-05-11 1986-09-16 Alza Corporation Osmotic device with dual thermodynamic activity
US4783337A (en) 1983-05-11 1988-11-08 Alza Corporation Osmotic system comprising plurality of members for dispensing drug
US4704282A (en) 1984-06-29 1987-11-03 Alza Corporation Transdermal therapeutic system having improved delivery characteristics
US4588580B2 (en) 1984-07-23 1999-02-16 Alaz Corp Transdermal administration of fentanyl and device therefor
US4626539A (en) 1984-08-10 1986-12-02 E. I. Dupont De Nemours And Company Trandermal delivery of opioids
US4568343A (en) 1984-10-09 1986-02-04 Alza Corporation Skin permeation enhancer compositions
US4573995A (en) 1984-10-09 1986-03-04 Alza Corporation Transdermal therapeutic systems for the administration of naloxone, naltrexone and nalbuphine
ATE45735T1 (en) * 1984-12-22 1989-09-15 Thomae Gmbh Dr K TETRAHYDRO-BENZTHIAZOLE, THEIR PRODUCTION AND USE AS INTERMEDIATE OR MEDICINAL PRODUCTS.
DE3447075A1 (en) * 1984-12-22 1986-07-03 Dr. Karl Thomae Gmbh, 7950 Biberach Tetrahydrobenzothiazoles, their preparation and their use as intermediates or as medicaments
US4806341A (en) 1985-02-25 1989-02-21 Rutgers, The State University Of New Jersey Transdermal absorption dosage unit for narcotic analgesics and antagonists and process for administration
US4645502A (en) 1985-05-03 1987-02-24 Alza Corporation Transdermal delivery of highly ionized fat insoluble drugs
US4904475A (en) 1985-05-03 1990-02-27 Alza Corporation Transdermal delivery of drugs from an aqueous reservoir
US4698062A (en) 1985-10-30 1987-10-06 Alza Corporation Medical device for pulsatile transdermal delivery of biologically active agents
EP0249343B1 (en) 1986-06-13 1992-01-08 Alza Corporation Moisture activation of transdermal drug delivery system
US4938759A (en) 1986-09-02 1990-07-03 Alza Corporation Transdermal delivery device having a rate controlling adhesive
US4908027A (en) 1986-09-12 1990-03-13 Alza Corporation Subsaturated transdermal therapeutic system having improved release characteristics
US5344656A (en) 1986-09-12 1994-09-06 Alza Corporation Subsaturated transdermal therapeutic system having improved release characteristics
US4816258A (en) 1987-02-26 1989-03-28 Alza Corporation Transdermal contraceptive formulations
US4788062A (en) 1987-02-26 1988-11-29 Alza Corporation Transdermal administration of progesterone, estradiol esters, and mixtures thereof
US5071656A (en) 1987-03-05 1991-12-10 Alza Corporation Delayed onset transdermal delivery device
US4943435A (en) 1987-10-05 1990-07-24 Pharmetrix Corporation Prolonged activity nicotine patch
US4917895A (en) 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US4781924A (en) 1987-11-09 1988-11-01 Alza Corporation Transdermal drug delivery device
US5004610A (en) 1988-06-14 1991-04-02 Alza Corporation Subsaturated nicotine transdermal therapeutic system
EP0387751B1 (en) 1989-03-15 1994-06-08 Nitto Denko Corporation Medicated plasters
US5024843A (en) 1989-09-05 1991-06-18 Alza Corporation Oral hypoglycemic glipizide granulation
US5591454A (en) 1989-09-05 1997-01-07 Alza Corporation Method for lowering blood glucose
US5091190A (en) 1989-09-05 1992-02-25 Alza Corporation Delivery system for administration blood-glucose lowering drug
DE3937271A1 (en) 1989-11-09 1991-05-16 Boehringer Ingelheim Kg TRANSDERMAL APPLICATION OF 2-AMINO-6-N-PROPYLAMINO-4,5,6,7-TETRAHYDROBENZOTHIAZOLE
US5069909A (en) 1990-06-20 1991-12-03 Cygnus Therapeutic Systems Transdermal administration of buprenorphine
US5122382A (en) 1990-10-29 1992-06-16 Alza Corporation Transdermal contraceptive formulations, methods and devices
US5314694A (en) 1990-10-29 1994-05-24 Alza Corporation Transdermal formulations, methods and devices
FR2688138B1 (en) 1992-03-06 1995-05-05 Rhone Poulenc Rorer Sa APPLICATION OF AMINO-2 TRIFLUOROMETHOXY-6 BENZOTHIAZOLE TO OBTAIN A MEDICINE FOR THE TREATMENT OF AMYOTROPHIC LATERAL SCLEROSIS.
ATE185694T1 (en) 1992-05-13 1999-11-15 Alza Corp OXYBUTYNIN FOR TRANSDERMAL ADMINISTRATION
US5792664A (en) 1992-05-29 1998-08-11 The Rockefeller University Methods for producing and analyzing biopolymer ladders
ATE427493T1 (en) 1992-05-29 2009-04-15 Univ Rockefeller METHOD FOR DETERMINING THE SEQUENCE OF PEPTIDES USING A MASS SPECTROMETER
PT700521E (en) 1993-05-28 2003-10-31 Baylor College Medicine METHOD AND MASS SPECTROMETER FOR DESSORING AND IONIZATION OF ANALYZES
US6566386B2 (en) 1993-08-09 2003-05-20 Nippon Zoki Pharmaceutical Co., Ltd. Immunomodulating and antiinflammatory agent
US5442117A (en) 1993-12-13 1995-08-15 Albemarle Corporation Enantiomeric resolution
US5635203A (en) 1994-09-29 1997-06-03 Alza Corporation Transdermal device having decreased delamination
US6156777A (en) 1994-12-15 2000-12-05 Pharmacia & Upjohn Company Use of pramipexole as a neuroprotective agent
US5650420A (en) 1994-12-15 1997-07-22 Pharmacia & Upjohn Company Pramipexole as a neuroprotective agent
US6262115B1 (en) 1995-05-22 2001-07-17 Alza Coporation Method for the management of incontinence
US5674895A (en) 1995-05-22 1997-10-07 Alza Corporation Dosage form comprising oxybutynin
US5912268A (en) 1995-05-22 1999-06-15 Alza Corporation Dosage form and method for treating incontinence
JP3468526B2 (en) 1995-10-26 2003-11-17 サノフィーサンテラボ Use of 1- (2-naphth-2-ylethyl) -4- (3-trifluoromethylphenyl) -1,2,3,6-tetrahydropyridine for the preparation of a remedy for amyotrophic lateral sclerosis
US6929801B2 (en) 1996-02-19 2005-08-16 Acrux Dds Pty Ltd Transdermal delivery of antiparkinson agents
US6919373B1 (en) 1996-11-12 2005-07-19 Alza Corporation Methods and devices for providing prolonged drug therapy
GB9705428D0 (en) 1997-03-15 1997-04-30 Knoll Ag Therapeutic agents
US5804215A (en) 1997-03-21 1998-09-08 L. Perrigo Company Transdermal patch disposal system and method
US5883995A (en) 1997-05-20 1999-03-16 Adc Telecommunications, Inc. Fiber connector and adapter
NZ516848A (en) 1997-06-20 2004-03-26 Ciphergen Biosystems Inc Retentate chromatography apparatus with applications in biology and medicine
JP2001517858A (en) 1997-09-23 2001-10-09 サイファーゲン・バイオシステムズ・インコーポレーテッド Secondary ion generator / detector for time-of-flight mass spectrometer
US6197339B1 (en) 1997-09-30 2001-03-06 Pharmacia & Upjohn Company Sustained release tablet formulation to treat Parkinson's disease
JP2002510604A (en) 1998-04-02 2002-04-09 アビセナ グループ, インク. Composition containing a combination of a creatine compound and a second substance
PE20000728A1 (en) 1998-06-26 2000-08-21 Cocensys Inc HETEROCYCLES 4-BENZYL PIPERIDINE ALKYLSULFOXIDE AND THEIR USE AS SUBTYPE-SELECTIVE NMDA RECEPTOR ANTAGONISTS
DE19830201A1 (en) 1998-07-07 2000-01-13 Boehringer Ingelheim Pharma Antidepressant
WO2000006162A1 (en) 1998-07-27 2000-02-10 Boehringer Ingelheim Pharma Kg Agents with antidepressant action, containing pramipexol and a further antidepressant
US20010055613A1 (en) 1998-10-21 2001-12-27 Beth A. Burnside Oral pulsed dose drug delivery system
NZ511442A (en) 1998-11-02 2003-02-28 Elan Corp Plc Multiparticulate modified release composition for multiple dosing of ADD patients with methylphenidate HCl
US6776984B1 (en) 1999-08-20 2004-08-17 George R. Schwartz Induced regeneration and repair of damaged neurons and nerve axon myelin
JP2003501418A (en) 1999-06-04 2003-01-14 エラン ファーマ インターナショナル,リミティド Compositions and methods for inhibiting cell death
DE19938825A1 (en) 1999-08-19 2001-04-26 Boehringer Ingelheim Pharma Active ingredient combination with clonidine
US6480820B1 (en) 1999-09-20 2002-11-12 Advanced Cochlear Systems, Inc. Method of processing auditory data
US6750235B1 (en) 1999-09-30 2004-06-15 The General Hospital Corporation Pramipexole as a treatment for cocaine craving
WO2001022820A1 (en) 1999-09-30 2001-04-05 The General Hospital Corporation Use of pramipexole as a treatment for cocaine craving
US20040132788A1 (en) 1999-10-11 2004-07-08 Chabrier De Lassauniere Pierre-Etienne Derivatives of heterocycles with 5 members, their preparation and their use as medicaments
US6443976B1 (en) 1999-11-30 2002-09-03 Akorn, Inc. Methods for treating conditions and illnesses associated with abnormal vasculature
CA2398255A1 (en) 2000-02-01 2001-08-09 Human Genome Sciences, Inc. Bcl-2-like polynucleotides, polypeptides, and antibodies
PE20011074A1 (en) 2000-02-23 2001-10-04 Upjohn Co USE OF PRAMIPEXOL IN THE TREATMENT OF ADDICTION DISORDERS
WO2001078652A2 (en) 2000-04-14 2001-10-25 Metabolon, Inc. Methods for drug discovery, disease treatment, and diagnosis using metabolomics
US6955821B2 (en) 2000-04-28 2005-10-18 Adams Laboratories, Inc. Sustained release formulations of guaifenesin and additional drug ingredients
CA2415815A1 (en) 2000-07-06 2002-01-17 Nigel H. Greig Tetrahydrobenzothiazole analogues as neuroprotective agents
DE10037619A1 (en) 2000-08-02 2002-02-14 Daimler Chrysler Ag Arrangement of controls
ES2187249B1 (en) * 2000-09-18 2004-09-16 Synthon Bv PROCEDURE FOR THE PREPARATION OF 2-AMINO-6- (RENT) AMINO-4,5,6,7-TETRAHYDROBENZOTIAZOLES.
AU2001296703A1 (en) 2000-10-06 2002-04-15 Xenoport, Inc. Bile acid prodrugs of l-dopa and their use in the sustained treatment of parkinsonism
US6618138B2 (en) 2000-10-12 2003-09-09 Jed Khoury Scanning fluorescent systems for various diagnostic
CN1262337C (en) 2000-11-16 2006-07-05 赛弗根生物系统股份有限公司 Method for analyzing mass spectra
US20020177626A1 (en) 2001-01-19 2002-11-28 Cook Graham D. Treatment of sleep disturbances
WO2002080957A1 (en) 2001-04-09 2002-10-17 Neurosearch A/S Adenosine a2a receptor antagonists combined with neurotrophic activity compounds in the treatment of parkinson's disease
US20030013120A1 (en) 2001-07-12 2003-01-16 Patz Edward F. System and method for differential protein expression and a diagnostic biomarker discovery system and method using same
GB0117618D0 (en) 2001-07-19 2001-09-12 Phoqus Ltd Pharmaceutical dosage form
DE10137082A1 (en) 2001-07-28 2003-02-13 Hexal Ag Stable transdermal therapeutic system containing pramipexol or ropinirol, especially for treating Parkinson's disease, comprises backing, drug-containing pressure-sensitive adhesive matrix and protective layers
US7157480B2 (en) 2001-12-11 2007-01-02 University Of Virginia Patent Foundation Use of pramipexole to treat amyotrophic lateral sclerosis
US20060281797A1 (en) 2001-12-11 2006-12-14 University Of Virginia Patent Foundation Neurorestoration with R(+) Pramipexole
TWI262083B (en) 2001-12-28 2006-09-21 Syngenta Participations Ag Microbially-expressed thermotolerant phytase for animal feed
CN101352421A (en) 2002-01-16 2009-01-28 贝林格尔英格海姆法玛两合公司 Method for preparing basically non-crystalline type telmisartan
US20030166696A1 (en) 2002-01-24 2003-09-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Pramipexole for the treatment of HIV dementia
CA2476434A1 (en) 2002-02-15 2003-08-28 Sloan-Kettering Institute For Cancer Research Method of treating trx mediated diseases
WO2003077902A1 (en) 2002-02-19 2003-09-25 Xenoport, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
JP2005526079A (en) 2002-03-15 2005-09-02 サイプレス バイオサイエンス, インコーポレイテッド NE and 5-HT reuptake inhibitors for treating visceral pain syndrome
DE10213571A1 (en) 2002-03-26 2003-10-23 Lichtwer Pharma Ag Plant extracts and their application
US20040033530A1 (en) 2002-04-08 2004-02-19 Awrey Donald E. High throughput purification, characterization and identification of recombinant proteins
EP1549301A2 (en) 2002-06-10 2005-07-06 Oklahoma Medical Research Foundation A method for using tethered bis(polyhydroxyphenyls) and o-alkyl derivatives thereof in treating inflammatory conditions of the central nervous system
KR100505260B1 (en) 2002-06-28 2005-07-29 노건웅 Method and kit for the tolerance induction to allergy-causing substances, and the tolerance achievement against the substances thereby
US20050226926A1 (en) 2002-07-25 2005-10-13 Pfizer Inc Sustained-release tablet composition of pramipexole
PE20040134A1 (en) 2002-07-25 2004-03-06 Pharmacia Corp PRAMIPEXOL ONCE-A-DAY DOSAGE FORM
US20050074865A1 (en) 2002-08-27 2005-04-07 Compound Therapeutics, Inc. Adzymes and uses thereof
AU2003272539A1 (en) 2002-09-17 2004-04-08 New York University Methods of treating age associated memory impairment (aami), mild cognitive impairment (mci), and dementias with cell cycle inhibitors
GB0221513D0 (en) 2002-09-17 2002-10-23 Generics Uk Ltd Novel compounds and processes
US20040132826A1 (en) 2002-10-25 2004-07-08 Collegium Pharmaceutical, Inc. Modified release compositions of milnacipran
GB2394951A (en) 2002-11-04 2004-05-12 Cipla Ltd One pot synthesis of 2,6-diamino-4,5,6,7-tetrahydro-benzothiazole
AU2003298837A1 (en) 2002-12-02 2004-06-23 Massachusetts Institute Of Technology Prolonged suppression of electrical activity in excitable tissues
UA79182C2 (en) 2002-12-13 2007-05-25 Basf Ag Method for the production of benzophenones
US20060154932A1 (en) 2002-12-23 2006-07-13 Nancy Hathaway Pharmaceutical compositions and method of treating parkinson's disease
DK1610791T3 (en) 2003-03-31 2011-05-09 Titan Pharmaceuticals Inc Implantable polymer dopamine agonist sustained release device
US6921953B2 (en) * 2003-04-09 2005-07-26 Micron Technology, Inc. Self-aligned, low-resistance, efficient MRAM read/write conductors
WO2005123193A2 (en) 2004-06-17 2005-12-29 Osteologix A/S Treatments comprising strontium for rheumatic and arthritic diseases and pain
US8802139B2 (en) 2003-06-26 2014-08-12 Intellipharmaceutics Corp. Proton pump-inhibitor-containing capsules which comprise subunits differently structured for a delayed release of the active ingredient
WO2005010011A2 (en) 2003-07-15 2005-02-03 Xenoport, Inc. Methods of synthesis of acyloxyalkyl compounds
DE10333393A1 (en) 2003-07-23 2005-02-24 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system with the active ingredient pramipexole
US7365086B2 (en) * 2003-07-25 2008-04-29 Synthon Ip Inc. Pramipexole acid addition salts
US20080020028A1 (en) 2003-08-20 2008-01-24 Euro-Celtique S.A. Transdermal dosage form comprising an active agent and a salt and a free-base form of an adverse agent
US20050053649A1 (en) 2003-09-08 2005-03-10 Anne-Marie Chalmers Medication delivery device
CA2542391A1 (en) 2003-10-23 2005-05-12 University Of Pittsburgh Of The Commonwealth System Of Higher Education Biomarkers for amyotrophic lateral sclerosis
US20120253047A1 (en) 2004-03-19 2012-10-04 Dipharma S.P.A. Process for the preparation of (r)-pramipexole
US7741490B2 (en) * 2004-03-19 2010-06-22 Dipharma S.P.A. Intermediates for the preparation of pramipexole
US20050220877A1 (en) 2004-03-31 2005-10-06 Patel Ashish A Bilayer tablet comprising an antihistamine and a decongestant
US7539203B2 (en) 2004-05-26 2009-05-26 Intel Corporation Multiple channel flow control with first-in-first-out (FIFO) read/write random access memory (RAM)
WO2006012277A2 (en) 2004-06-30 2006-02-02 Amr Technology, Inc. Biocatalytic process for preparing enantiomerically enriched pramipexole
EP1773793A2 (en) * 2004-07-03 2007-04-18 Merck Generics (UK) Limited Process for the preparation of pramipexole by chiral chromatography
KR20130003040A (en) 2004-08-13 2013-01-08 베링거 인겔하임 인터내셔날 게엠베하 Extended release pellet formulation containing pramipexole or a pharmaceutically acceptable salt thereof, method for manufacturing the same and use thereof
DK1781260T4 (en) 2004-08-13 2014-06-23 Boehringer Ingelheim Int Extended release tablet formulation containing pramipexole or a pharmaceutically acceptable salt thereof, process for its preparation and use thereof
TW200616604A (en) 2004-08-26 2006-06-01 Nicholas Piramal India Ltd Nitric oxide releasing prodrugs containing bio-cleavable linker
DE102004044578A1 (en) 2004-09-13 2006-03-30 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system with an adhesive layer, method for siliconizing a backing layer of the system and use of the backing layer
US20060069263A1 (en) * 2004-09-30 2006-03-30 Irina Gribun Process for the reduction of (S)-2-amino-6-propionamido-4,5,6,7-tetrahydrobenzo-thiazole
JP2006143708A (en) 2004-10-19 2006-06-08 Ono Pharmaceut Co Ltd Medicine for treating neurodegenerative disease
WO2006043532A1 (en) 2004-10-19 2006-04-27 Ono Pharmaceutical Co., Ltd. Therapeutic agent for parkinson's disease
JP5134963B2 (en) 2004-11-05 2013-01-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Bilayer tablet containing telmisartan and amlodipine
US20060121619A1 (en) 2004-12-02 2006-06-08 University Of Pittsburgh Of The Commonwealth System Of Higher Education Protein biomarkers and therapeutic targets in an animal model for amyotrophic lateral sclerosis
WO2006071469A2 (en) 2004-12-02 2006-07-06 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Modulation of the neuroendocrine system as a therapy for amyotrophic lateral sclerosis
WO2006070406A1 (en) 2004-12-29 2006-07-06 J.B. Chemicals & Pharmaceuticals Ltd Bilayer tablets of oxcarbazepine for controlled delivery and a process of preparation thereof
IL172561A0 (en) 2004-12-30 2006-04-10 Chemagis Ltd Novel process for preparing pramipexole and its optical isomeric mixture by reduction with sodium triacetoxyborohydride
WO2006076681A2 (en) 2005-01-13 2006-07-20 Sirtris Pharmaceuticals, Inc. Novel compositions for preventing and treating neurodegenerative and blood coagulation disorders
EP1877079A4 (en) 2005-04-15 2011-08-31 Human Matrix Sciences Llc Plant-derived elastin binding protein ligands and methods of using the same
KR20070122497A (en) 2005-04-22 2007-12-31 제넨테크, 인크. Method for treating dementia or alzheimer's disease with a cd20 antibody
WO2006119295A2 (en) 2005-05-02 2006-11-09 Combinatorx, Incorporated Compositions and methods for the treatment of neurodegenerative diseases
ES2264378B1 (en) * 2005-05-09 2007-11-01 Ragactives, S.L. PROCEDURE FOR THE RESOLUTION OF 2-AMINO-6PROPILAMINO-4,5,6,7-TETRAHYDROBENZOTIAZOL AND INTERMEDIATE COMPOUNDS.
CA2619217A1 (en) 2005-08-15 2007-02-22 University Of Virginia Patent Foundation Neurorestoration with r(+) pramipexole
WO2007022501A2 (en) 2005-08-18 2007-02-22 Microbia, Inc. Useful indole compounds
RU2008119454A (en) 2005-10-18 2009-11-27 Оно Фармасьютикал Ко., Лтд. (Jp) MEDICINE FOR PROTECTION OF MOTOR NEURON IN PATIENTS WITH LATERAL AMYOTROPHIC SCLEROSIS
CA2626249A1 (en) 2005-10-18 2007-04-26 Boehringer Ingelheim International Gmbh Use of pramipexol for treating moderate to severe restless legs syndrome (rls)
AU2006330858A1 (en) 2005-12-21 2007-07-05 Wyeth Protein formulations with reduced viscosity and uses thereof
PL378587A1 (en) * 2005-12-29 2007-07-09 Instytut Farmaceutyczny Methods of (S)-(-)-2-amino-6-n-propyloamino-4,5,6,7-tetrahydrobenzotiazole and/or its salts
WO2007090882A2 (en) 2006-02-10 2007-08-16 Boehringer Ingelheim International Gmbh Pharmaceutical extended release compositions comprising pramipexole
US8518926B2 (en) 2006-04-10 2013-08-27 Knopp Neurosciences, Inc. Compositions and methods of using (R)-pramipexole
US20070259930A1 (en) 2006-04-10 2007-11-08 Knopp Neurosciences, Inc. Compositions and methods of using r(+) pramipexole
KR20090021169A (en) 2006-05-16 2009-02-27 크놉 뉴로사이언시스 인코포레이티드 Compositions of r(+) and s(-) pramipexole and methods of using the same
EP2465500A1 (en) 2006-05-16 2012-06-20 Knopp Neurosciences, Inc. Therapeutically effective amounts of R(+) and S(-) pramipexole for use in the treatment of parkinson's disease
CA2661616A1 (en) 2006-08-24 2008-02-28 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for preparing pramipexole dihydrochloride tablets
WO2008042611A2 (en) 2006-09-29 2008-04-10 Centocor, Inc. Method of using il6 antagonists with mitoxantrone for prostate cancer
WO2008041240A1 (en) * 2006-10-03 2008-04-10 Cadila Healthcare Limited Process for preparing (s)-pramipexole and its intermediates
CA2667924A1 (en) 2006-10-30 2008-05-08 Boehringer Ingelheim International Gmbh The use of pramipexole or a salt thereof for the treatment of parkinson's disease
ES2611314T3 (en) 2006-12-14 2017-05-08 Knopp Biosciences Llc Compositions and procedures for using (R) -pramipexole
US8524695B2 (en) 2006-12-14 2013-09-03 Knopp Neurosciences, Inc. Modified release formulations of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and methods of using the same
WO2008104847A2 (en) 2007-02-26 2008-09-04 Wockhardt Research Centre Processes for the preparation of pramipexole and salts thereof
WO2008113056A2 (en) 2007-03-14 2008-09-18 Knopp Neurosciences, Inc. Synthesis of chirally purified substituted benzothiazole diamines
EP2136638A4 (en) 2007-03-14 2010-05-12 Knopp Neurosciences Inc Modified release formulations of (6r)-4,5,6,7-tetrahydro-n6-propyl-2,6-benzothiazole-diamine and methods of using the same
US20110189167A1 (en) 2007-04-20 2011-08-04 Flynn Daniel L Methods and Compositions for the Treatment of Myeloproliferative Diseases and other Proliferative Diseases
US20100130569A1 (en) 2007-05-11 2010-05-27 Saten Pharmaceutical Co., Ltd. Prophylactic or therapeutic agent for posterior ocular disease comprising non-ergot selective d2 receptor agonist as active ingredient
HUE036885T2 (en) 2007-05-14 2018-08-28 Astrazeneca Ab Methods of reducing basophil levels
AU2009228163B2 (en) 2008-03-28 2012-08-30 Glaxosmithkline Llc Methods of treatment
EP2334185A4 (en) 2008-08-19 2011-09-21 Knopp Neurosciences Inc Compositions and methods of using (r)-pramipexole
TWM367342U (en) 2009-05-01 2009-10-21 Advanced Connectek Inc Fiber optics adapter
US8408815B2 (en) 2009-06-18 2013-04-02 Senko Advanced Components, Inc. Optical fiber connector and adapter
RU2012101792A (en) 2009-06-19 2013-07-27 Нопп Ньюросайенсиз, Инк. COMPOSITIONS AND METHODS FOR TREATING LATERAL AMYOTROPHIC SCLEROSIS
EP2451787B1 (en) 2009-07-06 2013-04-24 Boehringer Ingelheim International GmbH Polymorph of [4,6-bis(dimethylamino)-2-(4-{[4-(trifluoromethyl)benzoyl]amino}benzyl)pyrimidin-5-yl]
CA2770023A1 (en) 2009-08-06 2011-02-10 Neuraltus Pharmaceuticals, Inc. Treatment of macrophage-related disorders
WO2011109596A1 (en) 2010-03-03 2011-09-09 Knopp Neurosciences Synthesis of chirally purified substituted benzothiazole diamines
WO2011150221A2 (en) 2010-05-26 2011-12-01 Knopp Neurosciences, Inc. Compounds and methods of modulating mitochondrial bioenergetic efficiency through an interaction with atp synthase (complex v) and its subunits
KR20140053822A (en) 2010-12-03 2014-05-08 유티믹스 바이오사이언스 인코포레이티드 Preparation and use of (+)-1-(3,4-dichlorophenyl)-3-azabicyclo[3.1.0]hexane in the treatment of conditions affected by monoamine neurotransmitters
US20130059801A1 (en) 2011-08-31 2013-03-07 Catabasis Pharmaceuticals, Inc. Fatty acid amides, compositions and methods of use
WO2013034173A1 (en) 2011-09-06 2013-03-14 Synthon Bv Pramipexole extended release tablets
WO2013096870A1 (en) 2011-12-22 2013-06-27 Knopp Neurosciences Inc Compositions and methods for treating amyotrophic lateral sclerosis
US9512096B2 (en) 2011-12-22 2016-12-06 Knopp Biosciences, LLP Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
US9085586B2 (en) 2012-07-11 2015-07-21 Nimbus Iris, Inc. IRAK inhibitors and uses thereof
US9662313B2 (en) 2013-02-28 2017-05-30 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
WO2014134569A1 (en) 2013-02-28 2014-09-04 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
PT3019167T (en) 2013-07-12 2021-03-04 Knopp Biosciences Llc Treating elevated levels of eosinophils and/or basophils
DE102013108610A1 (en) 2013-08-06 2015-02-12 Rogers Germany Gmbh Metal-ceramic substrate and method for producing a metal-ceramic substrate
EP3033081B1 (en) 2013-08-13 2021-05-12 Knopp Biosciences LLC Compositions and methods for treating chronic urticaria
CA2921378A1 (en) 2013-08-13 2015-02-19 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and b-cell prolymphocytic disorders

Cited By (71)

* 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
US9303038B2 (en) 2011-09-06 2016-04-05 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy and neurological diseases
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
US9399634B2 (en) 2012-05-07 2016-07-26 Cellix Bio Private Limited Compositions and methods for the treatment of depression
US9738631B2 (en) 2012-05-07 2017-08-22 Cellix Bio Private Limited Compositions and methods for the treatment of neurological disorders
US9642915B2 (en) 2012-05-07 2017-05-09 Cellix Bio Private Limited Compositions and methods for the treatment of neuromuscular disorders and neurodegenerative diseases
US9522884B2 (en) 2012-05-08 2016-12-20 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic disorders
US9403826B2 (en) 2012-05-08 2016-08-02 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory disorders
US9434704B2 (en) 2012-05-08 2016-09-06 Cellix Bio Private Limited Compositions and methods for the treatment of neurological degenerative disorders
US9309233B2 (en) 2012-05-08 2016-04-12 Cellix Bio Private Limited Compositions and methods for the treatment of blood clotting disorders
US9266823B2 (en) 2012-05-08 2016-02-23 Cellix Bio Private Limited Compositions and methods for the treatment of parkinson's disease
US9499527B2 (en) 2012-05-10 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of familial amyloid polyneuropathy
US9346742B2 (en) 2012-05-10 2016-05-24 Cellix Bio Private Limited Compositions and methods for the treatment of fibromyalgia pain
US9499526B2 (en) 2012-05-10 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of neurologic diseases
US9273061B2 (en) 2012-05-10 2016-03-01 Cellix Bio Private Limited Compositions and methods for the treatment of chronic pain
US9242939B2 (en) 2012-05-10 2016-01-26 Cellix Bio Private Limited Compositions and methods for the treatment of respiratory disorders
US9315478B2 (en) 2012-05-10 2016-04-19 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9315461B2 (en) 2012-05-10 2016-04-19 Cellix Bio Private Limited Compositions and methods for the treatment of neurologic diseases
US9573927B2 (en) 2012-05-10 2017-02-21 Cellix Bio Private Limited Compositions and methods for the treatment of severe pain
US9321775B2 (en) 2012-05-10 2016-04-26 Cellix Bio Private Limited Compositions and methods for the treatment of moderate to severe pain
US9403857B2 (en) 2012-05-10 2016-08-02 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9339484B2 (en) 2012-05-10 2016-05-17 Cellix Bio Private Limited Compositions and methods for the treatment of restless leg syndrome and fibromyalgia
US9233161B2 (en) 2012-05-10 2016-01-12 Cellix Bio Private Limited Compositions and methods for the treatment of neurological conditions
US9394288B2 (en) 2012-05-10 2016-07-19 Cellix Bio Private Limited Compositions and methods for the treatment of asthma and allergy
US9498461B2 (en) 2012-05-23 2016-11-22 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory bowel disease
US9765020B2 (en) 2012-05-23 2017-09-19 Cellix Bio Private Limited Dichlorophenyl-imino compounds and compositions, and methods for the treatment of mucositis
US9227974B2 (en) 2012-05-23 2016-01-05 Cellex Bio Private Limited Compositions and methods for the treatment of respiratory disorders
US9434729B2 (en) 2012-05-23 2016-09-06 Cellix Bio Private Limited Compositions and methods for the treatment of periodontitis and rheumatoid arthritis
US9492409B2 (en) 2012-05-23 2016-11-15 Cellix Bio Private Limited Compositions and methods for the treatment of local pain
US9580383B2 (en) 2012-05-23 2017-02-28 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US9403793B2 (en) 2012-07-03 2016-08-02 Cellix Bio Private Limited Compositions and methods for the treatment of moderate to severe pain
US9187427B2 (en) 2012-08-03 2015-11-17 Cellix Bio Private Limited N-substituted nicotinamide compounds and compositions for the treatment migraine and neurologic diseases
US9624168B2 (en) 2012-09-06 2017-04-18 Cellix Bio Private Limited Compositions and methods for the treatment inflammation and lipid disorders
US9670153B2 (en) 2012-09-08 2017-06-06 Cellix Bio Private Limited Compositions and methods for the treatment of inflammation and lipid disorders
US10285981B2 (en) 2013-02-28 2019-05-14 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
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
US9333187B1 (en) 2013-05-15 2016-05-10 Cellix Bio Private Limited Compositions and methods for the treatment of inflammatory bowel disease
US9174931B2 (en) 2013-06-04 2015-11-03 Cellix Bio Private Limited Compositions for the treatment of diabetes and pre-diabetes
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
US10383856B2 (en) 2013-07-12 2019-08-20 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
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
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US10980783B2 (en) 2013-07-12 2021-04-20 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
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
US9642840B2 (en) 2013-08-13 2017-05-09 Knopp Biosciences, Llc Compositions and methods for treating plasma cell disorders and B-cell prolymphocytic disorders
US10195183B2 (en) 2013-08-13 2019-02-05 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
US10456381B2 (en) 2013-08-13 2019-10-29 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
US9763918B2 (en) 2013-08-13 2017-09-19 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
US9840472B2 (en) 2013-12-07 2017-12-12 Cellix Bio Private Limited Compositions and methods for the treatment of mucositis
US9771355B2 (en) 2014-09-26 2017-09-26 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy and neurological disorders
US9102649B1 (en) 2014-09-29 2015-08-11 Mahesh Kandula Compositions and methods for the treatment of multiple sclerosis
US9988340B2 (en) 2014-09-29 2018-06-05 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US9725404B2 (en) 2014-10-27 2017-08-08 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US10208014B2 (en) 2014-11-05 2019-02-19 Cellix Bio Private Limited Compositions and methods for the treatment of neurological disorders
US9108942B1 (en) 2014-11-05 2015-08-18 Mahesh Kandula Compositions and methods for the treatment of moderate to severe pain
US9173877B1 (en) 2014-11-05 2015-11-03 Cellix Bio Private Limited Compositions and methods for the treatment of local pain
US9175008B1 (en) 2014-11-05 2015-11-03 Cellix Bio Private Limited Prodrugs of anti-platelet agents
US9284287B1 (en) 2014-11-05 2016-03-15 Cellix Bio Private Limited Compositions and methods for the suppression of carbonic anhydrase activity
US9150557B1 (en) 2014-11-05 2015-10-06 Cellix Bio Private Limited Compositions and methods for the treatment of hyperglycemia
US9321716B1 (en) 2014-11-05 2016-04-26 Cellix Bio Private Limited Compositions and methods for the treatment of metabolic syndrome
US9290486B1 (en) 2014-11-05 2016-03-22 Cellix Bio Private Limited Compositions and methods for the treatment of epilepsy
US9932294B2 (en) 2014-12-01 2018-04-03 Cellix Bio Private Limited Compositions and methods for the treatment of multiple sclerosis
US9206111B1 (en) 2014-12-17 2015-12-08 Cellix Bio Private Limited Compositions and methods for the treatment of neurological diseases
US9096537B1 (en) 2014-12-31 2015-08-04 Mahesh Kandula Compositions and methods for the treatment of mucositis
US10227301B2 (en) 2015-01-06 2019-03-12 Cellix Bio Private Limited Compositions and methods for the treatment of inflammation and pain
US10343994B2 (en) 2015-01-06 2019-07-09 Mahesh Kandula Compositions and methods for the treatment of inflammation and pain

Also Published As

Publication number Publication date
US20080227985A1 (en) 2008-09-18
US10179774B2 (en) 2019-01-15
WO2008113056A2 (en) 2008-09-18
RU2009137925A (en) 2011-04-20
CA2681110A1 (en) 2008-09-18
RU2454409C2 (en) 2012-06-27
US8519148B2 (en) 2013-08-27
WO2008113056A3 (en) 2009-08-27
AU2008224844B2 (en) 2012-08-09
EP2137171A4 (en) 2010-05-19
CN101715443A (en) 2010-05-26
EP2137171A2 (en) 2009-12-30
US20170158649A1 (en) 2017-06-08
IL200925A0 (en) 2010-05-17
AU2008224844A1 (en) 2008-09-18
JP2010521496A (en) 2010-06-24

Similar Documents

Publication Publication Date Title
US10179774B2 (en) Synthesis of chirally purified substituted benzothiazole diamines
US20130079526A1 (en) Synthesis of Chirally Purified Substituted Benzothiazole Diamines
RU2491068C2 (en) Compositions and methods of using (r)-pramipexole
JP2010521496A5 (en)
CN111936139B (en) Mono (acid) salts of 6-aminoisoquinolines and use thereof
JP2010521493A (en) (6R) -4,5,6,7-Tetrahydro-N6-propyl-2,6-benzothiazole-diamine controlled-release preparations and methods for their use
WO2008104847A2 (en) Processes for the preparation of pramipexole and salts thereof
EP0673933B1 (en) Aminoalkyl benzoxazolinones and benzothiazolinones process for their preparation and compositions containing them
US20100069451A1 (en) Salt of 3-benzyl-2-methyl-2,3,3a,4,5,6,7, 7a- octahydrobenzo[d]isoxazol-4-one
JP4714341B2 (en) Novel 2-aminothiazole fused 2-aminoindanes and 2-aminotetralins and their use
JP4426572B2 (en) Benzothiazole derivatives and their use in the treatment of diseases associated with adenosine A2A receptors
WO2006117614A1 (en) Process for the preparation of pramipexole and new anhydrous forms of its dihydrochloride
KR20060014406A (en) Benzothiazole derivatives and use thereof in the treatment of diseases related to the adenosine a2a receptor
JP4667042B2 (en) New pharmaceutical compounds
CN114874202B (en) Medicine for treating Alzheimer&#39;s disease and application thereof
US20230301984A1 (en) Cyclic compounds for use in treating retinal degeneration
CA3190418A1 (en) Heterobiaryl compounds and imaging agents for imaging huntingtin protein
CN106397413B (en) 5-hydroxytryptamine receptor agonist and preparation method and application thereof
US20120136037A1 (en) Deuterium-enriched ruboxistaurin
JP2023102778A (en) Pharmaceutical composition comprising benzoisoxazole derivative
AU2013209376A1 (en) Compositions and methods of using (r)-pramipexole

Legal Events

Date Code Title Description
AS Assignment

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KNOPP NEUROSCIENCES INC.;REEL/FRAME:032551/0311

Effective date: 20131122

AS Assignment

Owner name: KOPPER, RACHEL, PENNSYLVANIA

Free format text: SECURITY INTEREST;ASSIGNOR:KNOPP BIOSCIENCES LLC;REEL/FRAME:032992/0899

Effective date: 20140522

AS Assignment

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MCCALL, JOHN M.;REEL/FRAME:033566/0787

Effective date: 20140527

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LAPINA, OLGA V.;REEL/FRAME:033566/0761

Effective date: 20140625

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHEN, JIAN-XIE;REEL/FRAME:033566/0738

Effective date: 20140521

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GADIKOTA, RAJENDRAKUMAR REDDY;REEL/FRAME:033566/0708

Effective date: 20140522

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RAJE, PRASAD;REEL/FRAME:033566/0577

Effective date: 20140728

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: KNOPP BIOSCIENCES LLC, PENNSYLVANIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:KOPPER, RACHEL;REEL/FRAME:048455/0727

Effective date: 20190220