WO2014122184A1 - Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds - Google Patents

Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds Download PDF

Info

Publication number
WO2014122184A1
WO2014122184A1 PCT/EP2014/052267 EP2014052267W WO2014122184A1 WO 2014122184 A1 WO2014122184 A1 WO 2014122184A1 EP 2014052267 W EP2014052267 W EP 2014052267W WO 2014122184 A1 WO2014122184 A1 WO 2014122184A1
Authority
WO
WIPO (PCT)
Prior art keywords
dihydroxyphenyl
amino
well
acceptable salts
physiologically acceptable
Prior art date
Application number
PCT/EP2014/052267
Other languages
French (fr)
Inventor
Rudolf-Giesbert Alken
Frank Schneider
Original Assignee
Cdrd Berolina Ab
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
Priority to UAA201506458A priority Critical patent/UA119845C2/en
Priority to KR1020157021874A priority patent/KR20150115807A/en
Priority to US14/765,430 priority patent/US9567289B2/en
Priority to CA2897132A priority patent/CA2897132C/en
Priority to CN201480006672.1A priority patent/CN104968643B/en
Priority to EP14707659.0A priority patent/EP2953926B1/en
Priority to BR112015016889A priority patent/BR112015016889A8/en
Priority to MX2015010087A priority patent/MX370884B/en
Priority to ES14707659T priority patent/ES2901752T3/en
Priority to AU2014214055A priority patent/AU2014214055A1/en
Priority to EA201500739A priority patent/EA028931B1/en
Priority to JP2015555754A priority patent/JP6704252B2/en
Application filed by Cdrd Berolina Ab filed Critical Cdrd Berolina Ab
Publication of WO2014122184A1 publication Critical patent/WO2014122184A1/en
Priority to IL239733A priority patent/IL239733A0/en
Priority to HK16102244.2A priority patent/HK1214241A1/en
Priority to HK16102797.3A priority patent/HK1214810A1/en
Priority to US15/402,343 priority patent/US9763904B2/en
Priority to US15/678,797 priority patent/US20180116989A1/en
Priority to US15/994,493 priority patent/US20180338943A1/en
Priority to IL260018A priority patent/IL260018B/en
Priority to AU2018247203A priority patent/AU2018247203A1/en
Priority to US16/512,684 priority patent/US20200163918A1/en
Priority to US16/992,199 priority patent/US20210094904A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/34Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C229/36Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings with at least one amino group and one carboxyl group bound to the same carbon atom of the carbon skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2813Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/2853Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers, poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C227/00Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C227/14Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton from compounds containing already amino and carboxyl groups or derivatives thereof
    • C07C227/16Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton from compounds containing already amino and carboxyl groups or derivatives thereof by reactions not involving the amino or carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention relates to position-specific asymmetric deuterium en- riched catecholamine derivatives, methods for their production and medicaments comprising said compounds, as well as their use in the treatment of Parkinson's disease.
  • L-DOPA levodopa
  • carboxylic acid esters are utilized, among other things, for the treatment of Parkinson's disease and restless leg syndrome.
  • a pharmaceutical which contains levodopa is, for example, Dopaflex®.
  • L-DOPA acts on the dopamine concentration in neurons of the brain. Unlike dopamine itself, it can pass through the blood-brain barrier and is converted into dopa- mine in the brain.
  • levodopa is administered in combination with active additives in pharmaceuticals.
  • Combinations of levodopa are used with peripheral decarboxylase inhibitors, with inhibitors of the enzyme catechol-O-methyltrans- ferase (COMT), with inhibitors of the enzyme monoamine oxidase (MAO) and with dopamine ⁇ -hydroxylase inhibitors.
  • CCT catechol-O-methyltrans- ferase
  • MAO monoamine oxidase
  • dopamine ⁇ -hydroxylase inhibitors dopamine ⁇ -hydroxylase inhibitors.
  • the decarboxylase inhibitors used are, for example: D,L- serine-2-(2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)-L-a-hydrazino- 3,4-dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4- trihydroxybenzyl) hydrazide, glycine-2-(2, 3, 4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2, 3, 4-trihydroxybenzyl) hydrazide.
  • combination preparations of levodopa and decarboxylase inhibitors include, among others: Madopar® (levodopa and benserazide hydrochloride) as well as Na- com® (levodopa and carbidopa).
  • COMT inhibitors examples include entacapone (Comtan®) and cabergoline, and frequently used MAO inhibitors are selegiline hydrochloride, mo- clobemide and tranylcypromine.
  • MAO inhibitors are selegiline hydrochloride, mo- clobemide and tranylcypromine.
  • Calcium 5-butyl picolinate and calcium 5-pentyl picolinate are described as inhibitors for dopamines-hydroxylase (DE-A 2 049 1 15).
  • Parkinson's disease is a neurodegenerative disease with a slow progressive course characterized by different symptoms and signs that may be present or develop during the progression of disease. Core symptoms are bradykinesia and at least one of the following: resting tremor, muscular rigidity and postural reflex impairment. Other symptoms that may occur during the disease progression are autonomic disturbances, sleep disturbances, disturbances in the sense of smell or sense, of temperature as well as depressive symptoms and cognitive dysfunctions.
  • the improvement of the impaired dopaminergic neurotransmission by administration of L-DOPA is the backbone of the current pharmacotherapy.
  • Pa- tients with advanced Parkinson's disease require higher doses of dopaminergics but this is limited by motor complications, like fluctuations and involuntarily movements (described as levodopa induced dyskinesia, LIDs). Fluctuations might be due to the shorter striatal persistence (half life) of dopamine especially in advanced Parkinson's disease patients, also referred to as "Parkinson's patients".
  • a clinical established approach to prolong striatal dopamine persistence is the co-administration of MAO-B inhibitors which block the main metabolic breakdown route of dopamine.
  • LIDs LIDs
  • ⁇ , ⁇ , ⁇ -03-L-DOPA exhibited higher longer-lasting striatal dopamine levels than L-DOPA.
  • ⁇ , ⁇ , ⁇ -03-L-DOPA showed improved motor activity compared to L-DOPA in several Parkinson models (Malmlof et al., Exp Neurol, 2008, 538-
  • S/S-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid ( ⁇ , ⁇ -02-L- DOPA)
  • L-2-Amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid ( ⁇ , ⁇ , ⁇ -03-L-DOPA) were shown to increase and prolong the output of striatal dopamine significantly more than L-DOPA (WO-A 2004/056724 and WO-A 2007/093450).
  • ⁇ , ⁇ , ⁇ -03-L-DOPA caused significant less dyskinesia than L-DOPA (Malmlof et al., Exp Neurol, 2010, 225: 408-415).
  • the problem to be solved according to the invention is to improve the activity of the known ⁇ , ⁇ , ⁇ -03-L-DOPA.
  • deuterated is extended to partially or completely deuterated compounds.
  • “Completely deuterated” compounds are compounds in which at least 98 mol% deuterium are present in the respective position within the chemical compound (The deviation to 100 Mol% is caused by analytical measurement deviation and experimental errors.) This means that there has been achieved an enrichment of deuterium in the respective position and that hydrogen has been replaced.
  • the respective enrichment may be performed by chemical reaction in that one uses deuterated starting materials in chemical reactions or that an hydrogen/deuterium exchange has been performed by mixing respective compounds.
  • Deuterated is therefore not related to any naturally occurring deuterium in hydrogen compounds. As it is known, deuterium is present in hydrogen in natural abundance to an extend of 0.015 mol%. Any abundance or enrichment that is greater than 0.02 mol% is understood as being “deuterated” in the sense of this present invention.
  • the problem is solved according to the invention by providing deuterated catecholamine derivatives of the general Formula I
  • R2, and R3 are independently selected from hydrogen and deuterium and wherein at least one of R2 and R3 has a deuterium enrichment in the range from 0.02 mol% to 100 mol% deuterium, and
  • deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 is at least 5 percentage points
  • R 4 is hydrogen, deuterium, Ci to C6-alkyl or C 5 to C6-cycloalkyl, deuterated Ci to C6-alkyl or C 5 to C6-cycloalkyl, or a group that is easily hydrolytically or enzymatically cleavable under physiological conditions,
  • Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 7 percentage points. Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 10 percentage points. Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R 2 and R 3 is at least 15 percentage points.
  • Deuterated catecholamine derivatives are preferred, wherein the difference between the deuterium enrichment of R 2 and R 3 is at least 20 percentage points.
  • Deuterated catecholamine derivatives, according to the invention are preferred, wherein R 4 is selected from the group comprising hydrogen, deuterium, methyl, perdeuteromethyl, ethyl, perdeuteroethyl, propyl, perdeutero- propyl, butyl, perdeuterobutyl, Ci to C 6 -alkyl, that may be branched or un- branched, or C 5 to C6-cycloalkyl, deuterated or partly deuterated Ci to Ce- alkyl, that may be branched or unbranched, or deuterated or partly deuterated C 5 to C6-cycloalkyl.
  • Deuterated catecholamine derivatives are preferred, wherein R is selected from the group comprising hydrogen, deute- rium, methyl, perdeuteromethyl, ethyl, perdeuteroethyl, propyl, perdeutero- propyl, cyclohexyl, and perdeuterocyclohexyl.
  • Deuterated catecholamine derivatives according to the invention are preferred, wherein R is hydrogen.
  • Deuterated catecholamine derivatives according to the invention are preferred, wherein R is methyl.
  • Deuterated catecholamine derivatives according to the invention are pre- ferred, wherein R is ethyl.
  • L-2-Amino-2,3,3 * - trideutero-3-(3,4-dihydroxyphenyl) propionic acid ( ⁇ , ⁇ , ⁇ * - ⁇ 3- ⁇ - ⁇ ), wherein 3 * indicates that the deuterium enrichment in one ⁇ -position is about 90 mol%.
  • This compound has according to the definition of the present inven- tion a difference in the deuterium enrichment in the ⁇ -positions of about 8 to 10 percentage points.
  • the other positions carrying deuterium are completely deuterated and show a deuterium enrichment of at least 98 mol%.
  • This compound is named Test Item D in Tables 1 and 2 as outlined in the present de- scription herein.
  • R 4 is hydrogen, deuterium, Ci to C6-alkyl or C 5 to C6-cycloalkyl, deuterated Ci to C6-alkyl or C 5 to C6-cycloalkyl, or a group that is easily hydrolytically or enzymatically cleavable under physiological conditions, as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form, in a ratio to adjust the deuterium enrichment in position R 2 or R 3 in general Formula I within the predefined range of 0.02 mol% to 100 mol% deuterium.
  • deuterated catecholamine derivatives wherein the compound according to general Formula II is se- lected from the list comprising
  • L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) methyl propionate L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) ethyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) cyclohexyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate,
  • L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) propyl propionate L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) cyclohexyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteroethyl
  • deuterated catecholamine derivatives wherein the percentage of the compound according to general Formula II is in the range of 0.1 mol% to 99.9 mol%, preferably in the range of 5 mol% to 95 mol%, especially preferred in the range of 78 mol% to 95 mol%.
  • deuterated catecholamine derivatives wherein the percentage of the compound according to general Formula II is in the range of 88 mol% to 92 mol%.
  • deuterated catecholamine derivatives wherein the percentage of the compound according to general Formula II is in the range of 78 mol% to 82 mol%. Therefore, according to the invention a mixture is preferred in which 90 mol% of L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid are admixed with 10 mol% of L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid, or in which 80 mol% of L-2-amino-2,3,3-trideutero-3-(3,4- dihydroxyphenyl) propionic acid are admixed with 20 mol% of L-2-amino-2,3- dideutero-3-(3,4-dihydroxyphenyl) propionic acid, or in which 85 mol% of L-2- amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl)
  • a further object of the present invention is a method for the preparation of deuterated catecholamine derivatives according to the present invention, by mixing
  • Ri , R 2 , R3, and R have the meaning as given above,
  • Ri , R2, R3, and R 4 have the meaning as above, as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form, wherein the deuterium enrichment of R 2 and R 3 is different from each other and that the difference between the deuterium enrichment of R 2 and R 3 has a second predefined value, (iii) in a ratio that yields a predefined difference between the deuterium enrichment of R 2 and R 3 , which is in the range from at least 5 to at least 20 percentage points.
  • a further object of the present invention is the use of the deuterated cate- cholamine derivatives according to the invention as well as physiologically acceptable salts thereof, for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy.
  • dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system at
  • an enzyme inhibitor or several enzyme inhibitors for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications,
  • the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or ⁇ -hydroxylase inhibitors.
  • the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)- 5 L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (carbidopa), L- serine-2-(2,3,4-trihydroxybenzyl) hydrazide, glycine-2-(2,3,4- trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof.
  • o Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof further characterized in that the catechol-O-methyltransferase inhibitor is selected from entacapone and cabergoline as well as physiologically acceptable salts thereof.
  • Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physio- o logically acceptable salts thereof.
  • Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the ⁇ -hydroxylase inhibitor is selected from calcium 5-butyl 5 picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof.
  • a further object of the present invention is a pharmaceutical composition,5 which contains deuterated catecholamines according to the invention as well as physiologically acceptable salts thereof, for the treatment of Parkinson's disease, of restless leg syndrome, of dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lat- eral sclerosis and of multiple system atrophy, in addition to pharmaceutically acceptable adjuvants and additives.
  • a pharmaceutical composition which comprises deuterated catecholamines according to the invention as well as physiologically accept- able salts thereof, for the treatment of Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy, as well as one or more enzyme inhibitors, in addition to phar- maceutically acceptable adjuvants and additives.
  • (2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)-L-a-hydrazino-3,4- dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4- trihydroxybenzyl) hydrazide, glycine-2-(2,3,4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically ac- ceptable salts thereof.
  • a pharmaceutical composition which comprises deuterated catecholamines according to the invention, further characterized in that the catechol-O-methyltransferase inhibitor is selected from entacapone and ca- bergoline as well as physiologically acceptable salts thereof.
  • a pharmaceutical composition which comprises deuterated catecholamines according to the invention, further characterized in that the monoamine oxidase inhibitor is selected from the group consisting of se- legiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof.
  • Still another object of the present invention is a pharmaceutical composition, which comprises a mixture of 10 mol% of L-2-amino-2,3-dideutero-3-(3,4- dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, and 90 mol% of L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, in a pharmacologically active amount, optionally in addition with pharmaceutically acceptable adjuvants and additives.
  • compositions of the present invention are very powerful in the treatment of Parkinson's disease as the asymmetric position specific deuterium enrichment can tune the known effects of position specific deuterated L-DOPA. This provides a powerful tool to adjust the treatment according to the symptoms and side effects that change during disease progression.
  • Another object of the present invention is a method for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy with a person who has been identified as a person who is in the need of the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted ty- rosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia,
  • the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or ⁇ -hydroxylase inhibitors.
  • the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (ben- serazide), (-)-L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (car- bidopa), L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide, glycine-2-(2,3,4- trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof.
  • the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (ben- serazide), (-)-L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (car- bidopa), L
  • the method wherein the catechol-O-methyltransferase inhibitor is selected from entacapone and cabergoline as well as physiologically ac- ceptable salts thereof.
  • the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof.
  • the ⁇ -hydroxylase inhibitor is selected from calcium 5-butyl picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof.
  • the preparation of the deuterated catecholamine derivatives of the present invention can be performed in at least two principal ways. One way is to mix compounds with a certain deuterium enrichment with compounds which have only hydrogen or only a highly enriched (>98 % D) deuterium substitution at a certain position. By mixing at least two compounds any required enrichment level of deuterium at any position can be obtained.
  • the other way of preparation is to add specifically enriched starting material to a certain step during the preparation process of the compounds according to the invention.
  • deuterium enriched catecholamine derivatives The preparation of deuterium enriched catecholamine derivatives is known from WO-A 2004/056724 and WO-A 2007/093450. In there, the preparation of selectively deuterated DOPA derivatives is disclosed that have a deuterium enrichment in the respective position within the molecule of at least 98 %.
  • One preferred synthetic pathway is shown in Scheme 1 .
  • the compounds according to the invention by adding non-deuterated educts 3a and/or 4 and/or 5 to the respective deuterated compounds.
  • the ratio of deuterated and non-deuterated compounds is adjusted in such a manner to obtain the desired ratio in the end product.
  • This method of production has the advantage that no further mixing steps are required. This obtained product is then by definition no longer a mixture.
  • physiologically acceptable salts of the deuterated catecholamine derivatives for the production of the physiologically acceptable salts of the deuterated catecholamine derivatives according to the invention, the usual physiologically acceptable inorganic and organic acids such as hydrochloric acid, hy- drobromic acid, phosphoric acid, sulfuric acid, oxalic acid, maleic acid, fu- marie acid, lactic acid, tartaric acid, malic acid, citric acid, salicylic acid, adipic acid and benzoic acid can be used. Additional acids that can be used are described, for example, in Fort suitse der Arzneistoffforschung, Vol. 10, pp. 224-225, Birkhauser Publishers, Basel and Stuttgart, 1966, and Journal of Pharmaceutical Sciences, Vol. 66, pp. 1 -5 (1977).
  • the acid addition salts are usually obtained in a way known in and of itself by mixing the free base or solutions thereof with the corresponding acid or solutions thereof in an organic solvent, for example, a lower alcohol, such as methanol, ethanol, n-propanol or isopropanol or a lower ketone such as acetone, methyl ethyl ketone or methyl isobutyl ketone or an ether such as diethyl ether, tetrahydrofuran or dioxane.
  • an organic solvent for example, a lower alcohol, such as methanol, ethanol, n-propanol or isopropanol or a lower ketone such as acetone, methyl ethyl ketone or methyl isobutyl ketone or an ether such as diethyl ether, tetrahydrofuran or dioxane.
  • a lower alcohol such as methanol, ethanol, n-propanol or iso
  • the acid addition salts of the compounds according to the invention can be converted to the free base in a way known in and of itself, e.g., with alkali or ion exchangers. Additional salts can be obtained from the free base by reaction with inorganic or organic acids, particularly those which are suitable for the formation of salts that can be employed therapeutically.
  • These or also other salts of the compound according to the invention such as, e.g., the picrate, may also serve for purification of the free base by converting the free base into a salt, separating this salt, and afterwards releasing the base from the salt.
  • the subject of the present invention is also pharmaceuticals for oral, buccal, sublingual, nasal, rectal, subcutaneous, intravenous or intramuscular applica- tion as well as for inhalation, which, in addition to the usual vehicle and dilution agents, also contain a compound of general Formula I or the acid addition salt thereof as an active ingredient.
  • the pharmaceuticals of the invention are produced, in the known way and with suitable dosage, with the usual solid or liquid vehicle substances or dilu- tion agents and the commonly used pharmaceutical-technical adjuvants corresponding to the desired type of application.
  • the preferred preparations consist of a form for administration which is suitable for oral application.
  • forms of administration include, for example, tablets, sucking tablets, film tab- lets, dragees, capsules, pills, powders, solutions, aerosols or suspensions or slow-release forms.
  • parenteral preparations such as injection solutions are also considered.
  • suppositories for example, have also been named as preparations.
  • Corresponding tablets can be obtained, for example, by mixing the active substance with known adjuvants, for example, inert dilution agents such as dextrose, sugar, sorbitol, mannitol, polyvinylpyrrolidone, bursting agents such as corn starch or alginic acid, binders such as starches or ge- lantins, lubricants such as magnesium stearate or talc and/or agents for achieving a slow-release effect such as carboxypolymethylene, carboxy- methylcellulose, cellulose acetate phthalate or polyvinyl acetate.
  • the tablets may also consist of several layers.
  • Dragees can also be produced correspondingly, for controlled or delayed release forms of preparation, by coating the cores produced analogously to the tablets with agents commonly used in dragee coatings, for example, polyvinylpyrrolidone or shellac, gum arabic, talc, titanium dioxide or sugar.
  • the dragee envelope may also consist of several layers, wherein the adjuvants mentioned above in the case of tablets can be used.
  • Solutions or suspensions containing the active substance used according to the invention may additionally contain agents that improve taste, such as saccharin, cyclamate or sugar, as well as, e.g., taste enhancers such as vanilla or orange extract. They may also contain suspension adjuvants such as sodium carboxymethylcellulose or preservatives such as p-hydroxybenzoate.
  • Capsules containing active substances can be produced, for example, by mixing the active substance with an inert vehicle such as lactose or sorbitol and encapsulating this mixture in gelatin capsules.
  • Suitable suppositories can be produced, for example, by mixing with vehicle agents provided therefore, such as neutral fats or polyethylene glycol or derivatives thereof.
  • vehicle agents provided therefore, such as neutral fats or polyethylene glycol or derivatives thereof.
  • mice Female Sprague-Dawley rats weighing approximately 225 g were housed on a 12-hour light/dark cycle and kept on standard laboratory diet and water ad libitum. The rats were lesioned by unilateral injection of the neurotoxin 6- OHDA. The lesion was validated by measuring the rotational activity after i.p. injection of 2.5 mg/kg D-amphetamine. The anti-Parkinson effect (effect on motor performance) was evaluated by measurement of drug induced contralateral rotations. A dose effect was established to determine the equipotent (equi-effective) dose. Dyskinesia was evaluated after repeated treatment by scoring the animals for abnormal involuntary movements. The rats were scored, by an observer blinded to the experimental design for limb, axial and orolingual involuntary movements.
  • the equipotent dose as percent of L-DOPA dose that caused the same effect on motor performance and dyskinesia observed following repeated administration of these doses is shown in Table 2.
  • ⁇ , ⁇ -02-L-DOPA [B] The effect of ⁇ , ⁇ -02-L-DOPA [B] on motor performance is significantly greater compared to ⁇ , ⁇ , ⁇ -03-L-DOPA [C] and L-DOPA [A] as reflected by a lower equipotent dose.
  • dyskinesia after ⁇ , ⁇ -02-L-DOPA [B] is not reduced in comparison to L-DOPA at equipotent dose whereas ⁇ , ⁇ , ⁇ -03-L- DOPA [C] caused significantly less dyskinesia than L-DOPA at equipotent dose.
  • test item D with almost 100% deuterium enrichment in position a and ⁇ ⁇ and 90% in position ⁇ ⁇ provides both a motor effect equivalent to the di-deuterated ⁇ , ⁇ -02-L-DOPA [B] and a reduction of dyskinesia as the triple-deuterated ⁇ , ⁇ , ⁇ -03-L-DOPA [C].
  • Test compound D is thus the optimal treatment for late stage Parkinson pa- tients suffering from motor fluctuations and LIDs and requiring high doses of L-DOPA.
  • compound D shows that asymmetric position specific deuterium enrichment can tune the known effects of position specific deuterated L- DOPA. This provides a powerful tool to adjust the treatment according to the symptoms and side effects that change during disease progression.
  • Test item D has a deuterium enrichment of 90 % in $ R position.
  • D is obtained by mixing 10 mol% L-2-Amino-2,3(S)-dideutero-3-(3,4- dihydroxyphenyl) propionic acid with 90 mol% L-2-Amino-2,3,3-trideutero-3- (3,4-dihydroxyphenyl) propionic acid (deuterium enrichment >98 % in all three positions).
  • the mixture may be processed further in order to obtain a suitable pharmaceutical product for the medication of Parkinson's disease as given in the following examples.
  • composition of the core is composition of the core:
  • Titanium oxide 3. .00 mg
  • ⁇ , ⁇ , ⁇ * - ⁇ 3- ⁇ - ⁇ (Test Item D) and highly dispersed silicon dioxide are granulated in a compulsory mixer with a solution of povidone and sorbitol.
  • the granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets.
  • the tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc.
  • composition of the core is composition of the core:
  • Titanium oxide 3. .00 mg
  • carbidopa and highly dispersed silicon di- oxide are granulated in a compulsory mixer with a solution of povidone and sorbitol.
  • the granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets.
  • the tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc.
  • composition of the core is composition of the core:
  • Titanium oxide 3 .00 mg
  • Carbidopa, sorbitol and Eudragit are microencapsulated and homogenised in a barrel mixer with tartaric acid, highly dispersed silicon dioxide, povidone, pregelatinated starch, crosscar- mellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets.
  • the tablets are film coated with hydroxypropylnnethylcellulose, Macrogol, titanium dioxide and talc.
  • composition of the core is composition of the core:
  • Titanium oxide 3 .00 mg
  • composition of the core is composition of the core:
  • Titanium oxide 3. .00 mg
  • ⁇ , ⁇ , ⁇ * - ⁇ 3- ⁇ - ⁇ (Test Item D), carbidopa, and highly dispersed silicon dioxide are granulated in a compulsory mixer with a solution of povidone and sorbitol.
  • the granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets.
  • the tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc.
  • composition of the core is composition of the core:
  • Entacapone 200 .00 mg
  • Crospovidon Type B 15. .00 mg
  • Titanium oxide 3. .00 mg

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Emergency Medicine (AREA)
  • Psychology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Inorganic Chemistry (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Herein described are deuterated catecholamine derivatives of the general Formula (I) wherein R1 is deuterium, R2, and R3 are independently selected from hydrogen and deuterium and wherein at least one of R2 and R3 has a deuterium enrichment in the range from 0.02 mol% to 100 mol% deuterium, and wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 is at least 5 percentage points, R4 is hydrogen, deuterium, C1 to C6-alkyl or C5 to C6-cycloalkyl, deuterated C1 to C6-alkyl or C5 to C6-cycloalkyl, or a group that is easily hydrolytically or enzymatically cleavable under physiological conditions, as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form. The compounds can easily be prepared by mixing deuterated and non-deuterated compounds in a predefined ratio. The compounds show anti-Parkinson effect at lower doses and show lower side effects.

Description

Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
The present invention relates to position-specific asymmetric deuterium en- riched catecholamine derivatives, methods for their production and medicaments comprising said compounds, as well as their use in the treatment of Parkinson's disease.
Known representatives of catecholamines, such as L-DOPA (levodopa) as well as their carboxylic acid esters, are utilized, among other things, for the treatment of Parkinson's disease and restless leg syndrome. Such a pharmaceutical which contains levodopa is, for example, Dopaflex®. L-DOPA acts on the dopamine concentration in neurons of the brain. Unlike dopamine itself, it can pass through the blood-brain barrier and is converted into dopa- mine in the brain.
In addition, levodopa is administered in combination with active additives in pharmaceuticals. Combinations of levodopa are used with peripheral decarboxylase inhibitors, with inhibitors of the enzyme catechol-O-methyltrans- ferase (COMT), with inhibitors of the enzyme monoamine oxidase (MAO) and with dopamine β-hydroxylase inhibitors.
In this connection, the decarboxylase inhibitors used are, for example: D,L- serine-2-(2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)-L-a-hydrazino- 3,4-dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4- trihydroxybenzyl) hydrazide, glycine-2-(2, 3, 4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2, 3, 4-trihydroxybenzyl) hydrazide. Examples of combination preparations of levodopa and decarboxylase inhibitors include, among others: Madopar® (levodopa and benserazide hydrochloride) as well as Na- com® (levodopa and carbidopa).
Examples of COMT inhibitors are entacapone (Comtan®) and cabergoline, and frequently used MAO inhibitors are selegiline hydrochloride, mo- clobemide and tranylcypromine. Calcium 5-butyl picolinate and calcium 5-pentyl picolinate are described as inhibitors for dopamines-hydroxylase (DE-A 2 049 1 15).
Parkinson's disease is a neurodegenerative disease with a slow progressive course characterized by different symptoms and signs that may be present or develop during the progression of disease. Core symptoms are bradykinesia and at least one of the following: resting tremor, muscular rigidity and postural reflex impairment. Other symptoms that may occur during the disease progression are autonomic disturbances, sleep disturbances, disturbances in the sense of smell or sense, of temperature as well as depressive symptoms and cognitive dysfunctions.
The improvement of the impaired dopaminergic neurotransmission by administration of L-DOPA is the backbone of the current pharmacotherapy. Pa- tients with advanced Parkinson's disease require higher doses of dopaminergics but this is limited by motor complications, like fluctuations and involuntarily movements (described as levodopa induced dyskinesia, LIDs). Fluctuations might be due to the shorter striatal persistence (half life) of dopamine especially in advanced Parkinson's disease patients, also referred to as "Parkinson's patients". A clinical established approach to prolong striatal dopamine persistence is the co-administration of MAO-B inhibitors which block the main metabolic breakdown route of dopamine. The induction of LIDs is associated in many patients with higher CNS dopamine levels generated by large L-DOPA doses. Currently there are different pharmacological means under development to treat existing LIDs. α,β,β-03-L-DOPA exhibited higher longer-lasting striatal dopamine levels than L-DOPA. Correspondingly to the increased availability of dopamine in the striatum, α,β,β-03-L-DOPA showed improved motor activity compared to L-DOPA in several Parkinson models (Malmlof et al., Exp Neurol, 2008, 538-
542; Malmlof et al., Exp Neurol, 2010, 225: 408-415). The equi-effective dose of α,β,β-03-L-DOPA compared to L-DOPA was about 60%. The observed longer striatal persistence of dopamine allowed the assumption that fluctuations might be reduced as well. S/S-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid (α,β-02-L- DOPA) and L-2-Amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid (α,β,β-03-L-DOPA) were shown to increase and prolong the output of striatal dopamine significantly more than L-DOPA (WO-A 2004/056724 and WO-A 2007/093450).
The highest striatal dopamine concentrations were found after administration of α,β-02-L-DOPA. Those dopamine levels were even higher than those after the administration of the triple-deuterated α,β,β-03-L-DOPA which included the same deuterated positions as the double deuterated L-DOPA.
At the equi-effective dose (same striatal dopamine levels and same motor effect as L-DOPA), α,β,β-03-L-DOPA caused significant less dyskinesia than L-DOPA (Malmlof et al., Exp Neurol, 2010, 225: 408-415).
The problem to be solved according to the invention is to improve the activity of the known α,β,β-03-L-DOPA.
As used herein and in the context of the present invention the meaning of "deuterated" is extended to partially or completely deuterated compounds. "Completely deuterated" compounds are compounds in which at least 98 mol% deuterium are present in the respective position within the chemical compound (The deviation to 100 Mol% is caused by analytical measurement deviation and experimental errors.) This means that there has been achieved an enrichment of deuterium in the respective position and that hydrogen has been replaced. The respective enrichment may be performed by chemical reaction in that one uses deuterated starting materials in chemical reactions or that an hydrogen/deuterium exchange has been performed by mixing respective compounds.
"Deuterated" is therefore not related to any naturally occurring deuterium in hydrogen compounds. As it is known, deuterium is present in hydrogen in natural abundance to an extend of 0.015 mol%. Any abundance or enrichment that is greater than 0.02 mol% is understood as being "deuterated" in the sense of this present invention. The problem is solved according to the invention by providing deuterated catecholamine derivatives of the general Formula I
O
NH2-C-C-O-R4
R2-C-R3
Figure imgf000005_0001
Ri is deuterium,
R2, and R3 are independently selected from hydrogen and deuterium and wherein at least one of R2 and R3 has a deuterium enrichment in the range from 0.02 mol% to 100 mol% deuterium, and
wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 is at least 5 percentage points,
R4 is hydrogen, deuterium, Ci to C6-alkyl or C5 to C6-cycloalkyl, deuterated Ci to C6-alkyl or C5 to C6-cycloalkyl, or a group that is easily hydrolytically or enzymatically cleavable under physiological conditions,
as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form.
Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 7 percentage points. Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 10 percentage points. Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 15 percentage points.
Deuterated catecholamine derivatives, according to the invention are preferred, wherein the difference between the deuterium enrichment of R2 and R3 is at least 20 percentage points. Deuterated catecholamine derivatives, according to the invention are preferred, wherein R4 is selected from the group comprising hydrogen, deuterium, methyl, perdeuteromethyl, ethyl, perdeuteroethyl, propyl, perdeutero- propyl, butyl, perdeuterobutyl, Ci to C6-alkyl, that may be branched or un- branched, or C5 to C6-cycloalkyl, deuterated or partly deuterated Ci to Ce- alkyl, that may be branched or unbranched, or deuterated or partly deuterated C5 to C6-cycloalkyl.
Deuterated catecholamine derivatives, according to the invention are preferred, wherein R is selected from the group comprising hydrogen, deute- rium, methyl, perdeuteromethyl, ethyl, perdeuteroethyl, propyl, perdeutero- propyl, cyclohexyl, and perdeuterocyclohexyl.
Deuterated catecholamine derivatives, according to the invention are preferred, wherein R is hydrogen.
Deuterated catecholamine derivatives, according to the invention are preferred, wherein R is methyl.
Deuterated catecholamine derivatives, according to the invention are pre- ferred, wherein R is ethyl.
Especially preferred according to the present invention is L-2-Amino-2,3,3*- trideutero-3-(3,4-dihydroxyphenyl) propionic acid (α,β,β*-ϋ3-ί-ϋΟΡΑ), wherein 3* indicates that the deuterium enrichment in one β-position is about 90 mol%. This compound has according to the definition of the present inven- tion a difference in the deuterium enrichment in the β-positions of about 8 to 10 percentage points. The other positions carrying deuterium are completely deuterated and show a deuterium enrichment of at least 98 mol%. This compound is named Test Item D in Tables 1 and 2 as outlined in the present de- scription herein.
The problem is also solved by providing deuterated catecholamine derivatives, obtainable by admixing a compound of general Formula II
Figure imgf000007_0001
(II) with a compound of general Formula III or general Formula IV
Figure imgf000007_0002
(HI) (IV) wherein, in general Formula II, III, or IV,
R4 is hydrogen, deuterium, Ci to C6-alkyl or C5 to C6-cycloalkyl, deuterated Ci to C6-alkyl or C5 to C6-cycloalkyl, or a group that is easily hydrolytically or enzymatically cleavable under physiological conditions, as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form, in a ratio to adjust the deuterium enrichment in position R2 or R3 in general Formula I within the predefined range of 0.02 mol% to 100 mol% deuterium.
Preferred are, according to the present invention, deuterated catecholamine derivatives, wherein the compound according to general Formula II is se- lected from the list comprising
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) methyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) ethyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) cyclohexyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteroethyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteropropylethyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuterocyclohexyl propionate,
as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form, and wherein the compound according to general Formula III or general Formula IV is selected from the list comprising
L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) methyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) ethyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) propyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) cyclohexyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteroethyl
propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteropropylethyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuterocyclohexyl propionate,
as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form. Especially preferred are deuterated catecholamine derivatives, wherein the percentage of the compound according to general Formula II is in the range of 0.1 mol% to 99.9 mol%, preferably in the range of 5 mol% to 95 mol%, especially preferred in the range of 78 mol% to 95 mol%. Most preferred are herein deuterated catecholamine derivatives, wherein the percentage of the compound according to general Formula II is in the range of 88 mol% to 92 mol%. Most preferred are herein also deuterated catecholamine derivatives, wherein the percentage of the compound according to general Formula II is in the range of 78 mol% to 82 mol%. Therefore, according to the invention a mixture is preferred in which 90 mol% of L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid are admixed with 10 mol% of L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid, or in which 80 mol% of L-2-amino-2,3,3-trideutero-3-(3,4- dihydroxyphenyl) propionic acid are admixed with 20 mol% of L-2-amino-2,3- dideutero-3-(3,4-dihydroxyphenyl) propionic acid, or in which 85 mol% of L-2- amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid are admixed with 15 mol% of L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid, or in which 70 mol% of L-2-amino-2,3,3-trideutero-3-(3,4- dihydroxyphenyl) propionic acid are admixed with 30 mol% of L-2-amino-2,3- dideutero-3-(3,4-dihydroxyphenyl) propionic acid.
A further object of the present invention is a method for the preparation of deuterated catecholamine derivatives according to the present invention, by mixing
(i) a compound according to general Formula I O
NH2-C— C
R2-C-R3
Figure imgf000010_0001
wherein
Ri , R2, R3, and R have the meaning as given above,
as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form,
wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 has a first predefined value, with
(ii) at least one compound according to general Formula I
Figure imgf000010_0002
wherein
Ri , R2, R3, and R4 have the meaning as above, as well as their physiologically acceptable salts and their stereoisomers, en- antiomeres or diastereomers in optically pure form, wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 has a second predefined value, (iii) in a ratio that yields a predefined difference between the deuterium enrichment of R2 and R3, which is in the range from at least 5 to at least 20 percentage points.
A further object of the present invention is the use of the deuterated cate- cholamine derivatives according to the invention as well as physiologically acceptable salts thereof, for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy.
Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, in combination with an enzyme inhibitor or several enzyme inhibitors, for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy.
Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or β-hydroxylase inhibitors. Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)- 5 L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (carbidopa), L- serine-2-(2,3,4-trihydroxybenzyl) hydrazide, glycine-2-(2,3,4- trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof. o Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof further characterized in that the catechol-O-methyltransferase inhibitor is selected from entacapone and cabergoline as well as physiologically acceptable salts thereof.
5
Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physio- o logically acceptable salts thereof.
Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, further characterized in that the β-hydroxylase inhibitor is selected from calcium 5-butyl 5 picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof.
Preferred is the use of the deuterated catecholamine derivatives according to the invention as well as physiologically acceptable salts thereof, for the pro- 0 duction of pharmaceuticals for the treatment of Parkinson's disease, restless leg syndrome, of amyotrophic lateral sclerosis and of multiple system atrophy.
A further object of the present invention is a pharmaceutical composition,5 which contains deuterated catecholamines according to the invention as well as physiologically acceptable salts thereof, for the treatment of Parkinson's disease, of restless leg syndrome, of dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lat- eral sclerosis and of multiple system atrophy, in addition to pharmaceutically acceptable adjuvants and additives.
Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention as well as physiologically accept- able salts thereof, for the treatment of Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy, as well as one or more enzyme inhibitors, in addition to phar- maceutically acceptable adjuvants and additives.
Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention, further characterized in that the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or β-hydroxylase inhibitors.
Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention, further characterized in that the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-
(2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)-L-a-hydrazino-3,4- dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4- trihydroxybenzyl) hydrazide, glycine-2-(2,3,4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically ac- ceptable salts thereof.
Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention, further characterized in that the catechol-O-methyltransferase inhibitor is selected from entacapone and ca- bergoline as well as physiologically acceptable salts thereof. Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention, further characterized in that the monoamine oxidase inhibitor is selected from the group consisting of se- legiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof.
Preferred is a pharmaceutical composition, which comprises deuterated catecholamines according to the invention, further characterized in that the β- hydroxylase inhibitor is selected from calcium 5-butyl picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof.
Still another object of the present invention is a pharmaceutical composition, which comprises a mixture of 10 mol% of L-2-amino-2,3-dideutero-3-(3,4- dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, and 90 mol% of L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, in a pharmacologically active amount, optionally in addition with pharmaceutically acceptable adjuvants and additives.
Preferred is a pharmaceutical composition, wherein the composition further comprises, in a pharmacologically active amount, carbidopa, benserazide or entacapone or a mixture of the said compounds. The pharmaceutical compositions of the present invention are very powerful in the treatment of Parkinson's disease as the asymmetric position specific deuterium enrichment can tune the known effects of position specific deuterated L-DOPA. This provides a powerful tool to adjust the treatment according to the symptoms and side effects that change during disease progression.
According to the stage of Parkinson's disease in the respective person, one can use a compounds with a deuterium enrichment adjusted to the need of the patient under treatment. This offers new opportunities for a medication that is tailor-made or customized to the patient. Another object of the present invention is a method for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy with a person who has been identified as a person who is in the need of the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted ty- rosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy, the method comprising administering to the person deuterated cate- cholamine derivatives according to the invention as given in general formula as well as physiologically acceptable salts thereof.
Preferred is the method, wherein the administering to the person is in combination with an enzyme inhibitor or several enzyme inhibitors.
Preferred is the method, wherein the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or β-hydroxylase inhibitors.
Preferred is the method, wherein the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (ben- serazide), (-)-L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (car- bidopa), L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide, glycine-2-(2,3,4- trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof.
Preferred is the method, wherein the catechol-O-methyltransferase inhibitor is selected from entacapone and cabergoline as well as physiologically ac- ceptable salts thereof. Preferred is the method, wherein the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof.
Preferred is the method, wherein the β-hydroxylase inhibitor is selected from calcium 5-butyl picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof. The preparation of the deuterated catecholamine derivatives of the present invention can be performed in at least two principal ways. One way is to mix compounds with a certain deuterium enrichment with compounds which have only hydrogen or only a highly enriched (>98 % D) deuterium substitution at a certain position. By mixing at least two compounds any required enrichment level of deuterium at any position can be obtained. The other way of preparation is to add specifically enriched starting material to a certain step during the preparation process of the compounds according to the invention.
The preparation of deuterium enriched catecholamine derivatives is known from WO-A 2004/056724 and WO-A 2007/093450. In there, the preparation of selectively deuterated DOPA derivatives is disclosed that have a deuterium enrichment in the respective position within the molecule of at least 98 %. One preferred synthetic pathway is shown in Scheme 1 .
Scheme 1
Synthetic pathway to deuterated catecholamine derivatives
Figure imgf000017_0001
200.21
C9H8N04D3
According to the present invention it is preferred to prepare the compounds according to the invention by adding non-deuterated educts 3a and/or 4 and/or 5 to the respective deuterated compounds. The ratio of deuterated and non-deuterated compounds is adjusted in such a manner to obtain the desired ratio in the end product. This method of production has the advantage that no further mixing steps are required. This obtained product is then by definition no longer a mixture.
For the production of the physiologically acceptable salts of the deuterated catecholamine derivatives according to the invention, the usual physiologically acceptable inorganic and organic acids such as hydrochloric acid, hy- drobromic acid, phosphoric acid, sulfuric acid, oxalic acid, maleic acid, fu- marie acid, lactic acid, tartaric acid, malic acid, citric acid, salicylic acid, adipic acid and benzoic acid can be used. Additional acids that can be used are described, for example, in Fortschritte der Arzneimittelforschung, Vol. 10, pp. 224-225, Birkhauser Publishers, Basel and Stuttgart, 1966, and Journal of Pharmaceutical Sciences, Vol. 66, pp. 1 -5 (1977).
The acid addition salts are usually obtained in a way known in and of itself by mixing the free base or solutions thereof with the corresponding acid or solutions thereof in an organic solvent, for example, a lower alcohol, such as methanol, ethanol, n-propanol or isopropanol or a lower ketone such as acetone, methyl ethyl ketone or methyl isobutyl ketone or an ether such as diethyl ether, tetrahydrofuran or dioxane. For better crystal precipitation, mixtures of the named solvents can also be used. In addition, physiologically acceptable aqueous solutions of acid addition salts of the compounds used according to the invention can be produced therefrom in an aqueous acid solution.
The acid addition salts of the compounds according to the invention can be converted to the free base in a way known in and of itself, e.g., with alkali or ion exchangers. Additional salts can be obtained from the free base by reaction with inorganic or organic acids, particularly those which are suitable for the formation of salts that can be employed therapeutically. These or also other salts of the compound according to the invention, such as, e.g., the picrate, may also serve for purification of the free base by converting the free base into a salt, separating this salt, and afterwards releasing the base from the salt.
The subject of the present invention is also pharmaceuticals for oral, buccal, sublingual, nasal, rectal, subcutaneous, intravenous or intramuscular applica- tion as well as for inhalation, which, in addition to the usual vehicle and dilution agents, also contain a compound of general Formula I or the acid addition salt thereof as an active ingredient.
The pharmaceuticals of the invention are produced, in the known way and with suitable dosage, with the usual solid or liquid vehicle substances or dilu- tion agents and the commonly used pharmaceutical-technical adjuvants corresponding to the desired type of application. The preferred preparations consist of a form for administration which is suitable for oral application. Such forms of administration include, for example, tablets, sucking tablets, film tab- lets, dragees, capsules, pills, powders, solutions, aerosols or suspensions or slow-release forms.
Of course, parenteral preparations such as injection solutions are also considered. In addition, suppositories, for example, have also been named as preparations. Corresponding tablets can be obtained, for example, by mixing the active substance with known adjuvants, for example, inert dilution agents such as dextrose, sugar, sorbitol, mannitol, polyvinylpyrrolidone, bursting agents such as corn starch or alginic acid, binders such as starches or ge- lantins, lubricants such as magnesium stearate or talc and/or agents for achieving a slow-release effect such as carboxypolymethylene, carboxy- methylcellulose, cellulose acetate phthalate or polyvinyl acetate. The tablets may also consist of several layers.
Dragees can also be produced correspondingly, for controlled or delayed release forms of preparation, by coating the cores produced analogously to the tablets with agents commonly used in dragee coatings, for example, polyvinylpyrrolidone or shellac, gum arabic, talc, titanium dioxide or sugar. The dragee envelope may also consist of several layers, wherein the adjuvants mentioned above in the case of tablets can be used.
Solutions or suspensions containing the active substance used according to the invention may additionally contain agents that improve taste, such as saccharin, cyclamate or sugar, as well as, e.g., taste enhancers such as vanilla or orange extract. They may also contain suspension adjuvants such as sodium carboxymethylcellulose or preservatives such as p-hydroxybenzoate.
Capsules containing active substances can be produced, for example, by mixing the active substance with an inert vehicle such as lactose or sorbitol and encapsulating this mixture in gelatin capsules. Suitable suppositories can be produced, for example, by mixing with vehicle agents provided therefore, such as neutral fats or polyethylene glycol or derivatives thereof. The production of the pharmaceutical preparations according to the invention is known in the art, and is described in handbooks known to the person skilled in the art, for example, Hager's Handbuch [Handbook] (5th ed.) 2, 622-1045; List et al., Arzneiformenlehre [Instructions for Drug Forms], Stuttgart: Wiss. Verlagsges. 1985; Sucker et al., Pharmazeutische Technologie [Pharmaceutical Technology], Stuttgart: Thieme 1991 ; Ullmann's En- zyklopadie [Encyclopedia] (5th ed.) A 19, 241 -271 ; Voigt, Pharmazeutische Technologie [Pharmaceutical Technology], Berlin: Ullstein Mosby 1995.
The following examples shall explain the present invention. The examples shall be understood only as a preferred embodiment of the invention. It is not intended to limit the present invention to the scope of the given examples. Example 1
The effects on motor performance and the development of dyskinesia following administration of deuterated L-DOPA derivatives with different deuterium enrichment at specific position of the side chain have been compared among each other and to L-DOPA in the 6-hydroxydopamine (6-OHDA) rodent model of Parkinson's disease. The tested compounds and the specific deuterium enrichment of these compounds are displayed in Table 1 .
Table 1 Test Items
Figure imgf000021_0001
(3* or β*, respectively, indicates that the position is not completely deuterated)
(PR and Ps relate to the commonly used R/S nomenclature indicating the relative positions in optically active compounds)
Female Sprague-Dawley rats weighing approximately 225 g were housed on a 12-hour light/dark cycle and kept on standard laboratory diet and water ad libitum. The rats were lesioned by unilateral injection of the neurotoxin 6- OHDA. The lesion was validated by measuring the rotational activity after i.p. injection of 2.5 mg/kg D-amphetamine. The anti-Parkinson effect (effect on motor performance) was evaluated by measurement of drug induced contralateral rotations. A dose effect was established to determine the equipotent (equi-effective) dose. Dyskinesia was evaluated after repeated treatment by scoring the animals for abnormal involuntary movements. The rats were scored, by an observer blinded to the experimental design for limb, axial and orolingual involuntary movements.
The equipotent dose as percent of L-DOPA dose that caused the same effect on motor performance and dyskinesia observed following repeated administration of these doses is shown in Table 2.
Table 2 Results
Figure imgf000022_0001
The effect of α,β-02-L-DOPA [B] on motor performance is significantly greater compared to α,β,β-03-L-DOPA [C] and L-DOPA [A] as reflected by a lower equipotent dose. However dyskinesia after α,β-02-L-DOPA [B] is not reduced in comparison to L-DOPA at equipotent dose whereas α,β,β-03-L- DOPA [C] caused significantly less dyskinesia than L-DOPA at equipotent dose. Surprisingly, test item D with almost 100% deuterium enrichment in position a and βδ and 90% in position βκ provides both a motor effect equivalent to the di-deuterated α,β-02-L-DOPA [B] and a reduction of dyskinesia as the triple-deuterated α,β,β-03-L-DOPA [C].
Test compound D is thus the optimal treatment for late stage Parkinson pa- tients suffering from motor fluctuations and LIDs and requiring high doses of L-DOPA.
The example of compound D shows that asymmetric position specific deuterium enrichment can tune the known effects of position specific deuterated L- DOPA. This provides a powerful tool to adjust the treatment according to the symptoms and side effects that change during disease progression.
According to the stage of Parkinson's disease in the respective person, one can use a compounds with a deuterium enrichment adjusted to the need of the patient under treatment. This offers new opportunities for a medication that is tailor-made or customized to the patient.
Example 2
Preparation of Test Compound D from Table 1
L-2-Amino-2,3,3*-trideutero-3-(3,4-dihydroxyphenyl) propionic acid
(α,β,β*-03-Ι_-ϋΟΡΑ)
Test item D has a deuterium enrichment of 90 % in $R position.
D is obtained by mixing 10 mol% L-2-Amino-2,3(S)-dideutero-3-(3,4- dihydroxyphenyl) propionic acid with 90 mol% L-2-Amino-2,3,3-trideutero-3- (3,4-dihydroxyphenyl) propionic acid (deuterium enrichment >98 % in all three positions).
Experimental data for C9H8.i 2H2.9NO4
Calculated: H 6.95 C 54.05 N 7.00 O 32.00
Analyt: H 7.00 C 54.02 N 7.00 0 31 .98
The degree of deuteration has also been determined by NMR spectroscopy. For that purpose NMR spectra with a 500 MHz spectrometer have been recorded. As a solvent, d6-DMSO was used. The following Table 3 shows the respective position within the compound of test item D and the integral (AUC = area under curve) of the registered spectra, reflecting the content of hydrogen at the respective positions.
Table 3 NMR results
Figure imgf000024_0001
The preparation of the starting material L-2-Amino-2,3,3-trideutero-3-(3,4- dihydroxyphenyl) propionic acid is described in WO-A 2004/056724, the preparation of the starting material L-2-Amino-2,3(S)-dideutero-3-(3,4- dihydroxyphenyl) propionic acid is described in WO-A 2007/093450.
After mixing the compounds the mixture may be processed further in order to obtain a suitable pharmaceutical product for the medication of Parkinson's disease as given in the following examples.
Example 3
Tablet with film coating containing α,β,β*-ϋ3-ί-ϋΟΡΑ
Composition of the core:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D) 40. .00 mg
Povidone 20. .00 mg
Sorbitol 7. .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13. .30 mg
Carmellose-sodium 20. .05 mg
Microcrytalline cellulose 41 . .00 mg
Magnesium stearate 2 .00 mg
Film coating:
Hydroxypropylmethylcellulose 16. .00 mg
Macrogol 400™ 2 .50 mg
Titanium oxide 3. .00 mg
Talc 3. .00 mg Preparation:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D) and highly dispersed silicon dioxide are granulated in a compulsory mixer with a solution of povidone and sorbitol. The granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets. The tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc. Example 4
Tablet with film coating containing α,β,β*-ϋ3-ί-ϋΟΡΑ and Carbidopa
Composition of the core:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D) 35. .00 mg
Carbidopa 25. .00 mg
Povidone 20. .00 mg
Sorbitol 7. .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13. .30 mg
Carmellose-sodium 20. .05 mg
Microcrytalline cellulose 41 . .00 mg
Magnesium stearate 2. .00 mg
Film coating:
Hydroxypropylmethylcellulose 16. .00 mg
Macrogol 400™ 2. .50 mg
Titanium oxide 3. .00 mg
Talc 3. .00 mg
Preparation:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D), carbidopa and highly dispersed silicon di- oxide are granulated in a compulsory mixer with a solution of povidone and sorbitol. The granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets. The tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc. Example 5
Tablet with film coating containing microencapsulated α,β,β*-ϋ3-ί-ϋΟΡΑ and Carbidopa
Composition of the core:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D) 40 .00 mg
Carbidopa 25 .00 mg
Tartaric acid 5 .00 mg
Povidone 20. .00 mg
Sorbitol 7 .00 mg
Eudragit RL™ solid 20 .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13 .30 mg
Carmellose-sodium 20 .05 mg
Microcrystalline cellulose 41 . .00 mg
Magnesium stearate 2 .00 mg
Film coating:
Hydroxypropylnnethylcellulose 16 .00 mg
Macrogol 400™ 2 .50 mg
Titanium oxide 3 .00 mg
Talc 3 .00 mg
Preparation:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D), Carbidopa, sorbitol and Eudragit are microencapsulated and homogenised in a barrel mixer with tartaric acid, highly dispersed silicon dioxide, povidone, pregelatinated starch, crosscar- mellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets. The tablets are film coated with hydroxypropylnnethylcellulose, Macrogol, titanium dioxide and talc. Example 6
Tablet with film coating containing microencapsulated α,β,β*-03-Ι_-ϋΟΡΑ and benserazide
Composition of the core:
α,β,β*-03-Ι_-ϋΟΡΑ (Test Item D) 40 .00 mg
Benserazide 25 .00 mg
Tartaric acid 5 .00 mg
Povidone 20. .00 mg
Sorbitol 7 .00 mg
Eudragit RLTM solid 20 .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13 .30 mg
Carmellose-sodium 20 .05 mg
Microcrystalline cellulose 41 . .00 mg
Magnesium stearate 2 .00 mg
Film coating:
Hydroxypropylmethylcellulose 16 .00 mg
Macrogol 400TM 2 .50 mg
Titanium oxide 3 .00 mg
Talc 3 .00 mg The preparation of the film coated tablets is as given in Example 5.
Example 7
Tablet with film coating containing α,β,β*-03-Ι_-ϋΟΡΑ and benserazide
Composition of the core:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D) 35. .00 mg
Benserazide 25. .00 mg
Povidone 20. .00 mg
Sorbitol 7. .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13. .30 mg
Carmellose-sodium 20. .05 mg
Microcrytalline cellulose 41 . .00 mg
Magnesium stearate 2 .00 mg
Film coating:
Hydroxypropylmethylcellulose 16. .00 mg
Macrogol 400™ 2 .50 mg
Titanium oxide 3. .00 mg
Talc 3. .00 mg
Preparation:
α,β,β*-ϋ3-ί-ϋΟΡΑ (Test Item D), carbidopa, and highly dispersed silicon dioxide are granulated in a compulsory mixer with a solution of povidone and sorbitol. The granules are dried, screened, mixed with pregelatinated starch, crosscarmellose sodium, carmellose sodium and microcrystalline cellulose, then combined with magnesium stearate and compressed into tablets. The tablets are film coated with hydroxypropylmethylcellulose, Macrogol, titanium dioxide and talc.
Example 8
Tablet with film coating containing α,β,β*-03-Ι_-ϋΟΡΑ and carbidopa and entacapone
Composition of the core:
α,β,β*-03-Ι_-ϋΟΡΑ (Test Item D) 40. .00 mg
Carbidopa 25. .00 mg
Entacapone 200. .00 mg
Povidon K30 20. .00 mg
Crospovidon Type B 15. .00 mg
Mannitol 9. .00 mg
Silicon dioxide, highly dispersed 2 mg
Pregelatinated starch 40. .00 mg
Crosscarmellose-sodium 13. .30 mg
Carmellose-sodium 20. .05 mg
Microcrytalline cellulose 41 . .00 mg
Magnesium stearate 2 .00 mg
Film coating:
Hydroxypropylmethylcellulose 16. .00 mg
Macrogol 400™ 2 .50 mg
Titanium oxide 3. .00 mg
Talc 3. .00 mg The preparation of the film coated tablet is performed as described in Example 3.

Claims

Claims Deuterated Catecholamine derivatives of the general Formula I
O
Ri II
NH2-C-C-O-R4
R2-C-R3
Figure imgf000030_0001
(I)
wherein Ri is deuterium,
R2, and R3 are independently selected from hydrogen and deuterium and wherein at least one of R2 and R3 has a deuterium enrichment in the range from 0.02 mol% to 100 mol% deuterium, and wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 is at least 5 percentage points,
R4 is hydrogen, deuterium, Ci to C6-alkyl or C5 to C6-cycloalkyl, deuterated Ci to C6-alkyl or C5 to C6-cycloalkyl, or a group that is easily hy- drolytically or enzymatically cleavable under physiological conditions, as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form. Deuterated catecholamine derivatives, according to claim 1 , wherein the difference between the deuterium enrichment of R2 and R3 is:
at least 7 percentage points; or
at least 10 percentage points; or
at least 15 percentage points; or
at least 20 percentage points.
Deuterated catecholamine derivatives, according to claim 1 , wherein R4 is selected from the group comprising hydrogen, deuterium, methyl, perdeuteromethyl, ethyl, perdeuteroethyl, propyl, perdeuteropropyl, butyl, perdeuterobutyl, Ci to C6-alkyl, that may be branched or un- branched, or C5 to C6-cycloalkyl, deuterated or partly deuterated Ci to C6-alkyl, that may be branched or unbranched, or deuterated or partly deuterated C5 to C6-cycloalkyl.
Deuterated catecholamine derivatives, according to claim 1 , obtainable by admixing a compound of general Formula II
Figure imgf000031_0001
(II) with a compound of general Formula III or general Formula IV
Figure imgf000032_0001
(III) (IV) wherein, in general Formula II, III, or IV,
R4 is hydrogen, deuterium, Ci to C6-alkyl or C5 to C6-cycloalkyl, deuter- ated Ci to C6-alkyl or C5 to C6-cycloalkyl, or a group that is easily hy- drolytically or enzymatically cleavable under physiological conditions, as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form, in a ratio to adjust the deuterium enrichment in position R2 or R3 in general Formula I within the predefined range of 0.02 mol% to 100 mol% deuterium.
Deuterated catecholamine derivatives, according to claim 4,
wherein the compound according to general Formula II is selected from the list comprising
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) methyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) ethyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) cyclohexyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) perdeuteroethyl propionate, L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl)
perdeuteropropylethyl propionate,
L-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl)
perdeuterocyclohexyl propionate,
as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form, and wherein the compound according to general Formula III or General Formula IV is selected from the list comprising
L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) methyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) ethyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) propyl propionate, L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) cyclohexyl
propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteromethyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteroethyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuteropropylethyl propionate,
L-2-amino-2,3-dideutero -3-(3,4-dihydroxyphenyl) perdeuterocyclohexyl propionate,
as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form.
Deuterated catecholamine derivatives, according to claim 4, wherein the percentage of the compound according to general Formula II is in the range of 0.1 mol% to 99.9 mol%, preferably in the range of 5 mol% to 95 mol%, especially preferred in the range of 78 mol% to 95 mol%.
A method for the preparation of deuterated catecholamine derivates, according to claims 1 to 3, by
mixing
(i) a compound according to general Formula I
Figure imgf000034_0001
wherein
Ri > R2, R3, and R have the meaning as given in claim 1 , as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form, wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 has a first predefined value, with
(ii) at least one compound according to general Formula I
Figure imgf000034_0002
(I)
wherein
Ri , R2, R3, and R4 have the meaning as given in claim 1 , as well as their physiologically acceptable salts and their stereoisomers, enantiomeres or diastereomers in optically pure form, wherein the deuterium enrichment of R2 and R3 is different from each other and that the difference between the deuterium enrichment of R2 and R3 has a second predefined value,
(iii) in a ratio that yields a predefined difference between the deuterium enrichment of R2 and R3, which is in the range from at least 5 to at least 20 percentage points.
A pharmaceutical composition, which comprises deuterated catecholamine derivatives according to one of claims 1 to 6 as well as physiologically acceptable salts thereof, for the treatment of Parkinson's disease, of restless leg syndrome, of dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manganese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy, in addition to pharmaceutically acceptable adjuvants and additives.
The pharmaceutical composition, according to claim 8, further comprising one or more enzyme inhibitors.
The pharmaceutical composition, according to claim 9, further characterized in that the enzyme inhibitor or the enzyme inhibitors include decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or β-hydroxylase inhibitors.
The pharmaceutical composition according to claim 10, further characterized in that the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide (ben- serazide), (-)-L-a -hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4-trihydroxybenzyl) hydrazide, glycine-2- (2,3,4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4- trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof, and in that the catechol-O-methyltransferase inhibitor is selected from entacapone and cabergoline as well as physiologically acceptable salts thereof, and in that the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof, and in that the β-hydroxylase inhibitor is selected from calcium 5-butyl pi- colinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof. 12. A pharmaceutical composition, which comprises a mixture of 10 mol% of L-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, and 90 mol% of L-2- amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) propionic acid as well as physiologically acceptable salts thereof, in a pharmacologically active amount, optionally in addition of pharmaceutically acceptable adjuvants and additives.
The pharmaceutical composition according to claim 12, further characterized, that the composition further comprises, in a pharmacologically active amount, carbidopa, benserazide or entacapone or a mixture of the said compounds.
14. Use of the deuterated catecholamine derivatives according to one of claims 1 to 6 as well as physiologically acceptable salts thereof, for the treatment of dopamine deficiency diseases or diseases which are based on disrupted tyrosine transport or disrupted tyrosine decarboxylase, such as Parkinson's disease, restless leg syndrome, dystonia, for inhibiting prolactin secretion, for stimulating the release of growth hormone, for the treatment of neurological symptoms of chronic manga- nese intoxications, of amyotrophic lateral sclerosis and of multiple system atrophy.
15. Use, according to claim 14, in combination with an enzyme inhibitor or several enzyme inhibitors. Use, according to claim 15, further characterized in that the enzyme inhibitor or the enzyme inhibitors involve decarboxylase inhibitors and/or catechol-O-methyltransferase inhibitors and/or monoamine oxidase inhibitors and/or β-hydroxylase inhibitors.
Use, according to claim 16, further characterized in that the decarboxylase inhibitor is selected from the group consisting of D,L-serine-2- (2,3,4-trihydroxybenzyl) hydrazide (benserazide), (-)-L-a-hydrazino-3,4 dihydroxy-a-methylhydrocinnamic acid (carbidopa), L-serine-2-(2,3,4- trihydroxybenzyl) hydrazide, glycine-2-(2,3,4-trihydroxybenzyl) hydrazide and L-tyrosine-2-(2,3,4-trihydroxybenzyl) hydrazide as well as physiologically acceptable salts thereof, and in that the catechol-O- methyltransferase inhibitor is selected from entacapone and cabergo- line as well as physiologically acceptable salts thereof, and in that the monoamine oxidase inhibitor is selected from the group consisting of selegiline, moclobemide and tranylcypromine as well as physiologically acceptable salts thereof, and in that the β-hydroxylase inhibitor is selected from calcium 5-butyl picolinate and calcium 5-pentyl picolinate as well as physiologically acceptable salts thereof.
Use of the deuterated catecholamine derivatives according to one of claims 1 to 6 as well as physiologically acceptable salts thereof, for the production of pharmaceuticals for the treatment of Parkinson's disease, restless leg syndrome, of amyotrophic lateral sclerosis and of multiple system atrophy.
PCT/EP2014/052267 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds WO2014122184A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
EA201500739A EA028931B1 (en) 2013-02-05 2014-02-05 Composition comprising catecholamine derivatives including l-2-amino-2,3,3-trideutero-3-(3,4-dihydroxyphenyl) and l-2-amino-2,3-dideutero-3-(3,4-dihydroxyphenyl) or physiologically acceptable salts thereof and use of the composition for treating parkinson's disease, restless leg syndrome, amyotrophic lateral sclerosis and multiple system atrophy
US14/765,430 US9567289B2 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
CA2897132A CA2897132C (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
CN201480006672.1A CN104968643B (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium-enriched catecholamine derivatives and medicaments comprising said compounds
EP14707659.0A EP2953926B1 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
BR112015016889A BR112015016889A8 (en) 2013-02-05 2014-02-05 deuterium enriched catecholamine derivatives in a specific position
MX2015010087A MX370884B (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds.
ES14707659T ES2901752T3 (en) 2013-02-05 2014-02-05 Position-Specific Asymmetric Deuterium-Enriched Catecholamine Derivatives and Medications Comprising Such Compounds
KR1020157021874A KR20150115807A (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
JP2015555754A JP6704252B2 (en) 2013-02-05 2014-02-05 Regiospecific asymmetric deuterium-enriched catecholamine derivative and medicament containing said compound
AU2014214055A AU2014214055A1 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
UAA201506458A UA119845C2 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
IL239733A IL239733A0 (en) 2013-02-05 2015-07-01 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
HK16102244.2A HK1214241A1 (en) 2013-02-05 2016-02-26 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
HK16102797.3A HK1214810A1 (en) 2013-02-05 2016-03-10 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US15/402,343 US9763904B2 (en) 2013-02-05 2017-01-10 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US15/678,797 US20180116989A1 (en) 2013-02-05 2017-08-16 Position-Specific Asymmetric Deuterium Enriched Catecholamine Derivatives And Medicaments Comprising Said Compounds
US15/994,493 US20180338943A1 (en) 2013-02-05 2018-05-31 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
IL260018A IL260018B (en) 2013-02-05 2018-06-13 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
AU2018247203A AU2018247203A1 (en) 2013-02-05 2018-10-09 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US16/512,684 US20200163918A1 (en) 2013-02-05 2019-07-16 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US16/992,199 US20210094904A1 (en) 2013-02-05 2020-08-13 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361760738P 2013-02-05 2013-02-05
US61/760,738 2013-02-05
EP13182708.1 2013-09-02
EP13182708 2013-09-02

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/765,430 A-371-Of-International US9567289B2 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US15/402,343 Continuation US9763904B2 (en) 2013-02-05 2017-01-10 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds

Publications (1)

Publication Number Publication Date
WO2014122184A1 true WO2014122184A1 (en) 2014-08-14

Family

ID=49084847

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/052267 WO2014122184A1 (en) 2013-02-05 2014-02-05 Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds

Country Status (14)

Country Link
US (2) US9567289B2 (en)
EP (1) EP2953926B1 (en)
JP (1) JP6704252B2 (en)
KR (1) KR20150115807A (en)
CN (1) CN104968643B (en)
AU (2) AU2014214055A1 (en)
BR (1) BR112015016889A8 (en)
CA (1) CA2897132C (en)
EA (1) EA028931B1 (en)
ES (1) ES2901752T3 (en)
HK (2) HK1214241A1 (en)
IL (2) IL239733A0 (en)
MX (1) MX370884B (en)
WO (1) WO2014122184A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015136003A2 (en) 2014-03-11 2015-09-17 Imphar Aktiengesellschaft Pharmaceutical composition for the treatment of parkinson's disease
WO2017060870A1 (en) 2015-10-09 2017-04-13 Hermann Russ Combination of deuterated levodopa with carbidopa and opicapone for the treatment of parkinson`s disease

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022512736A (en) * 2018-10-18 2022-02-07 オンコペプティデス エービー Compounds containing deuterium
WO2020115715A1 (en) * 2018-12-06 2020-06-11 Intrabio Ltd. Deuterated analogs of acetyl-leucine

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2049115A1 (en) 1970-10-06 1972-04-13 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai, Tokio 5-butyl-and5-pentyl-picolinic acids active - as dopamene-b-hydroxylase inhibitors
WO2004056724A1 (en) 2002-12-19 2004-07-08 Bdd Berolina Drug Development Gmbh Deuterated catecholamine derivatives and medicaments comprising said compounds
WO2007093450A2 (en) 2006-02-17 2007-08-23 Birds Pharma Gmbh Berolina Innovative Research & Development Services Deuterated catecholamine derivatives and medicaments comprising said compounds

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0904300D0 (en) * 2009-03-12 2009-04-22 Amarin Neuroscience Ltd Essential fatty acid compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2049115A1 (en) 1970-10-06 1972-04-13 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai, Tokio 5-butyl-and5-pentyl-picolinic acids active - as dopamene-b-hydroxylase inhibitors
WO2004056724A1 (en) 2002-12-19 2004-07-08 Bdd Berolina Drug Development Gmbh Deuterated catecholamine derivatives and medicaments comprising said compounds
WO2007093450A2 (en) 2006-02-17 2007-08-23 Birds Pharma Gmbh Berolina Innovative Research & Development Services Deuterated catecholamine derivatives and medicaments comprising said compounds

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Fortschritte der Arzneimittelforschung", vol. 10, 1966, BIRKHAUSER PUBLISHERS, pages: 224 - 225
"Hager's Handbuch [Handbook", pages: 622 - 1045
"Ullmann's En- zyklopädie [Encyclopedia", vol. 19, pages: 241 - 271
A.B. FOSTER: "Deuterium isotope effects in the metabolism of drugs and xenobiotics: implications for drug design", ADVANCES IN DRUG RESEARCH., vol. 14, 1985, Academic Press, London, GB, pages 1 - 40, XP009086953, ISSN: 0065-2490 *
JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 5
LIST ET AL.: "Arzneiformenlehre [Instructions for Drug Forms", 1985, WISS. VERLAGSGES
M. OBA, ET AL.: "Stereo-divergent synthesis of L-threo- and L-erythro-(2,3-2H2)amino acids using optically active dioxopiperazine as a chiral template", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1, no. 7, 1998, Royal Society of Chemistry, Cambridge, GB, pages 1275 - 1281, XP009086948, ISSN: 0300-922X, DOI: 10.1039/a708412i *
MALML6F ET AL., EXP NEUROL, 2008, pages 538 - 542
MALML6F ET AL., EXP NEUROL, vol. 225, 2010, pages 408 - 415
SUCKER ET AL.: "Pharmazeutische Technologie [Pharmaceutical Technology", 1991, THIEME
T. MALMLOF, ET AL.: "Deuterium substitutions in the L-DOPA molecule improve its anti-akinetic potency without increasing dyskinesias", EXPERIMENTAL NEUROLOGY, vol. 225, no. 2, 24 July 2010 (2010-07-24), Academic Press, New York, NY, US, pages 408 - 415, XP027274374, ISSN: 0014-4886, DOI: 10.1016/j.expneurol.2010.07.018 *
VOIGT: "Pharmazeutische Technologie [Pharmaceutical Technology", 1995, ULLSTEIN MOSBY

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015136003A2 (en) 2014-03-11 2015-09-17 Imphar Aktiengesellschaft Pharmaceutical composition for the treatment of parkinson's disease
EP3689334A1 (en) 2014-03-11 2020-08-05 Teva Pharmaceuticals International GmbH Composition comprising a dopamine agonist and an l-dopa derivative for treating parkinson's disease
WO2017060870A1 (en) 2015-10-09 2017-04-13 Hermann Russ Combination of deuterated levodopa with carbidopa and opicapone for the treatment of parkinson`s disease
JP2018530612A (en) * 2015-10-09 2018-10-18 テバ、ファーマスーティカルズ、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Teva Pharmaceuticals International Gmbh Combination of deuterated levodopa with carbidopa and opicapon for the treatment of Parkinson's disease

Also Published As

Publication number Publication date
HK1214241A1 (en) 2016-07-22
US9567289B2 (en) 2017-02-14
BR112015016889A2 (en) 2017-07-11
CA2897132C (en) 2021-05-25
EA028931B1 (en) 2018-01-31
US20200163918A1 (en) 2020-05-28
IL260018A (en) 2018-07-31
AU2018247203A1 (en) 2018-11-01
BR112015016889A8 (en) 2018-01-23
JP2016513084A (en) 2016-05-12
US20150376117A1 (en) 2015-12-31
CN104968643B (en) 2020-03-03
JP6704252B2 (en) 2020-06-03
CN104968643A (en) 2015-10-07
CA2897132A1 (en) 2014-08-14
IL260018B (en) 2020-01-30
ES2901752T3 (en) 2022-03-23
MX2015010087A (en) 2016-04-21
EP2953926A1 (en) 2015-12-16
HK1214810A1 (en) 2016-08-05
MX370884B (en) 2020-01-09
EA201500739A1 (en) 2015-11-30
IL239733A0 (en) 2015-08-31
AU2014214055A1 (en) 2015-07-16
EP2953926B1 (en) 2021-10-20
KR20150115807A (en) 2015-10-14

Similar Documents

Publication Publication Date Title
US20200163918A1 (en) Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US20240100054A1 (en) Pharmaceutical compositions comprising sepiapterin and uses thereof
DK1991522T3 (en) Deuterated CATECHOLAMINDERIVATER AND medicaments comprising THESE RELATIONS
EP3807279A1 (en) Pharmaceutically acceptable salts of sepiapterin
EP2114863B1 (en) Solid forms comprising (-)-o-desmethylvenlafaxine and uses thereof
US20200121812A1 (en) Combination of Deuterated Levodopa With Carbidopa and Opicapone For The Treatment of Parkinson's Disease
US9763904B2 (en) Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
US20210094904A1 (en) Position-specific asymmetric deuterium enriched catecholamine derivatives and medicaments comprising said compounds
AU2003289841A1 (en) Deuterated catecholamine derivatives and medicaments comprising said compounds
WO2010051239A1 (en) Tianeptine sulfate salt forms and methods of making and using the same
CA2513077A1 (en) Use of l-dopa, derivatives thereof and medicaments comprising said compounds for the prophylaxis of psychotic diseases
US20070082953A1 (en) Use of l-dopa, derivatives thereof and medicaments comprising said compounds for the prophylaxis of psychotic diseases
JP2022528355A (en) Salt form of S- (N, N-diethylcarbamoyl) glutathione

Legal Events

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

Ref document number: 14707659

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 239733

Country of ref document: IL

Ref document number: 2014707659

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2897132

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014214055

Country of ref document: AU

Date of ref document: 20140205

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015555754

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14765430

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/010087

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 201500739

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20157021874

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015016889

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015016889

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150714