WO2010097092A1 - Treatment of dyskinesia related disorders - Google Patents

Treatment of dyskinesia related disorders Download PDF

Info

Publication number
WO2010097092A1
WO2010097092A1 PCT/DK2010/050051 DK2010050051W WO2010097092A1 WO 2010097092 A1 WO2010097092 A1 WO 2010097092A1 DK 2010050051 W DK2010050051 W DK 2010050051W WO 2010097092 A1 WO2010097092 A1 WO 2010097092A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
dyskinesia
dopa
disease
propyl
Prior art date
Application number
PCT/DK2010/050051
Other languages
French (fr)
Inventor
Håkan WIKSTRÖM
Morten JØRGENSEN
Niels MØRK
Jennifer Larsen
Lars Torup
Benny Bang-Andersen
Original Assignee
H. Lundbeck A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42060693&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2010097092(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to BRPI1007017A priority Critical patent/BRPI1007017A2/en
Priority to CA2751322A priority patent/CA2751322C/en
Priority to CN2010800098743A priority patent/CN102333531B/en
Priority to EP19191241.9A priority patent/EP3653210A1/en
Priority to MX2011008944A priority patent/MX340138B/en
Priority to EP10707436.1A priority patent/EP2400967B1/en
Priority to SG2011061785A priority patent/SG174163A1/en
Application filed by H. Lundbeck A/S filed Critical H. Lundbeck A/S
Priority to US13/202,768 priority patent/US20120108624A1/en
Priority to AU2010217059A priority patent/AU2010217059B2/en
Priority to JP2011551409A priority patent/JP5738775B2/en
Priority to NZ594264A priority patent/NZ594264A/en
Priority to EA201171088A priority patent/EA023778B1/en
Priority to KR1020117019748A priority patent/KR101700978B1/en
Publication of WO2010097092A1 publication Critical patent/WO2010097092A1/en
Priority to IL213502A priority patent/IL213502A/en
Priority to ZA2011/06294A priority patent/ZA201106294B/en
Priority to HK12107220.3A priority patent/HK1166472A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4741Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having oxygen as a ring hetero atom, e.g. tubocuraran derivatives, noscapine, bicuculline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs 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

Definitions

  • aspects of the subject invention relate to methods of treating Parkinson's disease while maintaining a low dyskinesia induction profile and to methods of reversing dyskinesias comprising administering therapeutically effective amount of a compound disclosed herein.
  • the present invention further relates to uses and pharmaceutical compositions of said compounds in the manufacture of medicaments in treating the same or other movement disorders such as Huntington's chorea.
  • L-DOPA Parkinson's disease
  • Dl receptors can be divided into Di and D5 receptors while D2 receptors can be divided into D 2 , D3, and D 4 receptors.
  • dopamine-replacement therapy does have limitations, especially following long-term treatment.
  • the duration period response to a dose of L-DOPA becomes progressively shorter over the years, and periods in which the patient responds to the drug become complicated by the appearance of a range of side-effects.
  • the side-effects may manifest as dyskinesias, which can be seen either when the patient is undergoing dopamine replacement therapy or even when the patient is off therapy.
  • Dyskinesias are abnormal involuntary movement disorders.
  • the abnormal movements may manifest as chorea (involuntary, rapid, irregular, jerky movements that may affect the face, arms, legs, or trunk), ballism (involuntary movements similar to chorea but of a more violent and forceful nature), dystonia (sustained muscle contractions, usually producing twisting and repetitive movements or abnormal postures or positions) and/or athetosis (repetitive involuntary, slow, sinuous, writhing movements, which are especially severe in the hands).
  • chorea involuntary, rapid, irregular, jerky movements that may affect the face, arms, legs, or trunk
  • ballism involuntary movements similar to chorea but of a more violent and forceful nature
  • dystonia sustained muscle contractions, usually producing twisting and repetitive movements or abnormal postures or
  • PD afflicted patients may cycle between "on” periods which are complicated by dyskinesia and "off' periods in which they are severely parkinsonian. As a consequence they may experience profound disability despite the fact that L-DOPA remains an effective anti-Parkinson agent throughout the course of the disease (Obeso, et al. Neurology 2000, 55, S13-23).
  • Dopamine agonists such as bromocriptine, lisuride, pramipexole, ropinirole and pergolide are less efficacious than L-DOPA, particularly in moderate-to-severe PD. However, their side-effect profile is different from that of L-DOPA.
  • Dyskinesias and other movement disorders from dysfunction of the basal ganglia are of major socio-economic importance. Many attempts have been made to develop agents to prevent and/or treat dyskinesias although such attempts have met with limited success. There is, therefore, a need to provide novel agents to treat dyskinesia.
  • 6-hydroxydopamine (6-OHDA) lesion model of parkinsonism in the rat has provided an invaluable tool in the investigation of PD at a preclinical level and for the evaluation of novel therapeutic options (Schwarting and Huston, Prog. Neurobiol. 1996, 50, 275-331).
  • 6-OHDA paradigms One of the most widely used 6-OHDA paradigms is the evaluation of rotational behavior in rats which bear a discrete degeneration of the dopaminergic nigrostriatal pathway (Ungerstedt and Aburthnott, Brain Res. 1970, 24, 485).
  • 6-OHDA is unilaterally infused into the nigrostriatal pathway, striatum or medial forebrain bundle (MFB), producing a functional imbalance between the dopaminergic nigrostriatal systems.
  • MFB medial forebrain bundle
  • Administration of drugs directly stimulating dopamine receptors, such as the dopamine metabolic precursor L-DOPA and the dopamine agonist apomorphine produces a rotational behavior directed away from the body side in which 6-OHDA has been infused.
  • the 6-OHDA model can be used to reproduce other features of PD.
  • the development of both sensitized rotational behavior as well as abnormal involuntary movements (AIMs) has been observed in rats injected with 6-OHDA either in the striatum or in the MFB, and chronically treated with L-DOPA, therefore providing further an animal model for the study of L-DOPA induced dyskinesia (Lundblad, et aL Eur. J Neurosci. 2002, 15,120-132).
  • L-DOPA but not bromocriptine induces a gradual development of AIMs.
  • rats lesioned with 6- OHDA exhibit motor deficits that share essential functional similarities with Parkinson's dyskinesia and can be used to evaluate the potential of a treatment to provide treatments for dyskinesia.
  • Compounds 10, 11 and 12 have the potential to become the first PD drugs with L-DOP A-like efficacy and a favorable profile not only in terms of both induction of dyskinesia, but also as a medication for the reversal of dyskinesias.
  • the present invention contemplates the use of the corresponding racemic trans mixture.
  • the present invention further provides methods of treating Parkinson's disease with a low dyskinesia induction profile comprising administering a therapeutically effective amount of said compound.
  • the treatment of Parkinson's disease is as efficacious as L-DOPA treatment.
  • methods of reversing dyskinesias or treating Parkinson's disease comprising administering said compound and pharmaceutical compositions thereof.
  • One aspect of the invention is concerned with the use of (4aR,l OaR)-I -n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • a separate aspect of the invention relates to the use of (4aR, 1 OaR)- 1 -n-propyl- 1 ,2,3 ,4,4a,5 , 10, 1 Oa- octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament treating Parkinson's disease.
  • Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,10a-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease.
  • a separate aspect of the invention relates to the use of (4aR,l OaR)-I -n-propyl- 1,2,3 ,4,4a,5,l 0,1 Oa- octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
  • Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,10a-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for reversing dyskinesias.
  • Another aspect is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising (4aR,l OaR)-I -n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • a pharmaceutical composition comprising racemic trans- l-n-propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (4aR, 1 OaR)- 1 -n-propyl- 1 ,2,3 ,4,4a,5 ,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
  • a separate aspect relates to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile a low dyskinesia induction profile comprising administering a therapeutically effective amount of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol .
  • Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (4aR, 1OaR)-I -n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
  • Yet another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
  • One aspect of the invention is concerned with the use of (6aR,10aR)-7-/?-propyl- 6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • a separate aspect of the invention relates to the use of (6aR,10aR)-7-/?-propyl-6,6a,7,8,9,10,10a,l 1- octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
  • Another aspect is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising (6aR,10aR)-7-n-propyl-
  • compositions comprising (6aR,10aR)-7- n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (6aR, 10aR)-7-n-propyl-6,6a,7,8,9, 10, 10a, 11 -octahydro- 1 ,3-dioxa-7-aza-cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof.
  • Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3- dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof.
  • One aspect of the invention is concerned with the use of (4aR,l OaR)-I -n-propyl-
  • a separate aspect of the invention relates to the use of (4aR,l OaR)-I -n-propyl-
  • Yet another aspect relates to the use of (4aR,10aR)-l-/?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro- lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
  • Another aspect is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising (4aR,l OaR)-I -n-propyl- 2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • compositions comprising (4aR,10aR)-l- /?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (4aR,10aR)-l-n-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof.
  • Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (4aR,10aR)-l-/?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH- benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention contain two chiral centers (denoted with * in the below formula)
  • the compounds of the invention can exist in two different diastereomeric forms, the cis- and trans- isomers, both of which can exist in two enantiomeric forms.
  • the present invention relates only to the trans racemate and the (4aR, 10aR)-enantiomer.
  • the present invention is based on the discovery that (4aR, 1OaR)-I -n- propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol (herein referred to as "Compound 10”) reversed dyskinesias induced by L-DOPA/benserazide and apomorphine in rats lesioned with 6-OHDA.
  • Compound 10 reversed dyskinesias induced by L-DOPA/benserazide and apomorphine in rats lesioned with 6-OHDA.
  • the corresponding trans racemate also falls within the scope of this invention.
  • the compound of the present invention contain two chiral centers (denoted with * in the below formula)
  • the compound of the invention can exist in two different diastereomeric forms, the cis- and trans- isomers, both of which can exist in two enantiomeric forms.
  • the present invention relates only to the trans racemate and the (6aR, 10aR)-enantiomer.
  • the present invention is based on the discovery (6aR,10aR)-7-/?-propyl- 6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene (herein referred to as "Compound 11”) reversed dyskinesias induced by L-DOPA/benserazide and apomorphine in rats lesioned with 6-OHDA.
  • the present invention is based on the discovery that (4aR, 1OaR)-I -n-propyl- 2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one (herein referred to as Compound 12) has favorable profiles in rats with unilateral 6-OHDA lesions. It induce sless dyskinesias than L-DOPA and apomorphine, and reduces L-DOPA induced dyskinesias more effectively than D2 agonists, as exemplified by pramipexole.
  • dyskinesia refers to a condition characterized by abnormal involuntary movements that are associated with disorders of brain regions known as the basal ganglia.
  • the dyskinesia may be an "L-DOP A-induced dyskinesia” that arises and is a complication of the treatment of Parkinson's disease (the most common basal ganglia disease).
  • Dyskinesia can physically manifest in two forms, chorea and dystonia.
  • Chorea consists of involuntary, continuous, purposeless, abrupt, rapid, brief, unsustained and irregular movements that flow from one part of the body to another.
  • Dystonia refers to sustained muscle contractions that cause twisting and repetitive movements or abnormal postures.
  • Treating refers to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both, and inhibit at least one physical parameter which may not be discernible to the patient. Further, “treating” or “treatment” refers to delaying the onset of the disease or disorder or at least symptoms thereof in a patient which may be exposed to or predisposed to a disease or disorder even though that patient does not yet experience or display symptoms of the disease or disorder.
  • “Therapeutically effective amount” refers to the amount of a compound that, when administered to a patient for treating a disease or disorder, is sufficient to affect such treatment for the disease or disorder.
  • the “therapeutically effective amount” will vary depending on the compound, the disease or disorder and its severity and the age and weight of the patient to be treated.
  • the phrase "while maintaining a low dyskinesia profile” refers to the dyskinesia profile as seen in patients who have been treated via continuous dopaminergic stimulation. Treatments involving continuous dopaminergic stimulation are described in Stocchi and Olanow, Neurology 2004, 2004, 62, S56-S63; and Hilary, et al, Journal of Neurology 2004, 251, 11, 1370- 1374.
  • (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza- cyclopenta[a] anthracene is referred to as Compound 11.
  • Compound 10, 11 or 12 may be used to treat dyskinesia as a monotherapy (i.e. use of the compound alone); as an adjunct to compositions to prevent dyskinetic side-effects caused by the composition (e.g. as an adjunct to L-DOPA or apomorphine given to treat parkinsonian patients) or alternatively the compound may be given in combination with other treatments which also reduce dyskinesia (e.g. opioid receptor antagonists, ( ⁇ 2-adrenoreceptor- antagonists, cannabinoid CBI- antagonists, NMDA receptor-antagonists, cholinergic receptor antagonists, histamine H3-receptor agonists, and globus pallidus/subthalamic nucleus lesion/deep brain stimulation).
  • dyskinesia e.g. opioid receptor antagonists, ( ⁇ 2-adrenoreceptor- antagonists, cannabinoid CBI- antagonists, NMDA receptor-antagonists, cholinergic receptor antagonists, histamine H3-receptor
  • the present invention is further concerned with the concurrent, separate or sequential use in the treatment of Parkinson's disease while reducing dyskinesia induced by L-DOPA or a dopamine agonist comprising administering a therapeutically effective amount of Compound 10, 11 or 12 or a pharmaceutically salt thereof.
  • the dyskinesia is associated with a basal ganglia-related movement disorder.
  • the dyskinesia is associated with Parkinson's disease.
  • One embodiment relates to dyskinesia associated with idiopathic Parkinson's disease or postencephalitic Parkinsonism.
  • the dyskinesia is associated with off-dystonia in Parkinson's disease.
  • the dyskinesia arises as a side-effect of a therapeutic agent to treat Parkinson's disease.
  • the dyskinesia is associated with dopamine replacement therapy.
  • dopamine replacement therapy agent is selected from the group consisting of rotigotine, ropinirole, pramipexole, cabergoline, bromocriptine, lisuride, pergolide, L-DOPA and apomorphine.
  • the dyskinesia is established as a result of repeated administration of L-DOPA.
  • the present invention provides for a pharmaceutical composition
  • a pharmaceutical composition comprising Compound 10, 11 or 12 or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile
  • a pharmaceutical composition comprising racemic trans-1-n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
  • the pharmaceutical composition additionally comprises a MAO-B inhibitor.
  • the MAO-B inhibitor is selegine. In a separate embodiment, the MAO-B inhibitor is rasagiline.
  • the invention in another embodiment, relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of Compound 10, 11 or 12, or a pharmaceutically acceptable acid addition salt thereof, and one or more pharmaceutically acceptable carriers, diluents and excipients.
  • the mammal is a human subject.
  • Compound 10, 11 or 12 above as the free base is suitably between 0.01 and 125 mg/day, more suitable between 0.05 and 100 mg/day, e.g. preferably between 0.1 and 50 mg/day.
  • the daily dose of Compound 10, 11 or 12 is between 1.0 and 10 mg/day.
  • the daily dose of Compound 10, 11 or 12 is less than about 1.0 mg/day.
  • the daily dose of Compound 10, 11 or 12 is about 0.10 mg/day.
  • the invention provides an oral formulation comprising from 0.001 mg to
  • the invention provides an oral formulation comprising from 0.001 mg to 0.100 mg of Compound 10, 11 or 12. In a further embodiment, the invention provides an oral formulation comprising from 0.01 mg to 1.0 mg of Compound 10, 11 or 12.
  • the invention provides an oral formulation comprising from 0.10 mg to 10 mg of Compound 10, 11 or 12.
  • Compound 10, 11 or 12 forms pharmaceutically acceptable acid addition salts with a wide variety of organic and inorganic acids. Such salts are also part of this invention.
  • a pharmaceutically acceptable acid addition salt of Compound 10, 11 or 12 is formed from a pharmaceutically acceptable acid as is well known in the art. Such salts include the pharmaceutically acceptable salts listed in Journal of Pharmaceutical Science, 1977, 66, 2-19 and are known to the skilled person.
  • Typical inorganic acids used to form such salts include hydrochloric, hydrobromic, hydriodic, nitric, sulphuric, phosphoric, hypophosphoric, metaphosphoric, pyrophosphoric, and the like.
  • Salts derived from organic acids such as aliphatic mono and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic and hydroxyalkandioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, may also be used.
  • Such pharmaceutically acceptable salts thus include the chloride, bromide, iodide, nitrate, acetate, phenylacetate, trifluoroacetate, acrylate, ascorbate, benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-acetoxybenzoate, isobutyrate, phenylbutyrate, ⁇ -hydroxybutyrate, butyne-l,4-dicarboxylate, hexyne-l,4-dicarboxylate, caprate, caprylate, cinnamate, citrate, formate, fumarate, glycollate, heptanoate, hippurate, lactate, malate, maleate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, isonicotinate, oxalate, phthalate, teraphthalate, propiolate, propionate,
  • Tablets may thus be prepared by mixing the active ingredient with ordinary adjuvants, fillers and diluents and subsequently compressing the mixture in a convenient tabletting machine.
  • adjuvants, fillers and diluents comprise microcrystalline cellulose, corn starch, potato starch, lactose, mannitol, sorbitol talcum, magnesium stearate, gelatine, lactose, gums, and the like. Any other adjuvant or additive such as colorings, aroma, preservatives, etc. may also be used provided that they are compatible with the active ingredients.
  • the tablet formulations according to the invention may be prepared by direct compression of Compound 10, 11 or 12 in admixture with conventional adjuvants or diluents.
  • a wet granulate or a melt granulate of Compound 10, 11 or 12, optionally in admixture with conventional adjuvants or diluents may be used for compression of tablets.
  • Solutions of Compound 10, 11 or 12 for injections may be prepared by dissolving the active ingredient and possible additives in a part of the solvent for injection, preferably sterile water, adjusting the solution to the desired volume, sterilization of the solution and filling in suitable ampoules or vials.
  • Any suitable additive conventionally used in the art may be added, such as tonicity agents, preservatives, antioxidants, solubilizing agents, etc.
  • FIGURE 1 Crystal structure of compound ent-l ⁇ . The absolute configuration was determined by the anomalous scattering of the 'heavy' bromine atom.
  • FIGURE 2 Dose-response curve for the concentration-dependent stimulation of intracellular Ca 2+ release by dopamine in bLVtransfected CHO-GaI 6 cells.
  • Method 14 API 150EX and Shimadzu LC8/SLC-10A LC system. Column: C-18 4.6x30mm,
  • X-ray crystal structure determination was performed as follows.
  • the crystal of the compound was cooled to 120 K using a Cryostream nitrogen gas cooler system.
  • the data were collected on a Siemens SMART Platform diffractometer with a CCD area sensitive detector.
  • the structures were solved by direct methods and refined by full-matrix least-squares against F 2 of all data.
  • the hydrogen atoms in the structures could be found in the electron density difference maps.
  • the Flack x-parameters are in the range 0.0(l)-0.05(l), indicating that the absolute structures are correct.
  • Programs used for data collection, data reduction and absorption were SMART, SAINT and SADABS [cf. "SMART and SAINT, Area Detector Control and Integration Software", Version 5.054,Bruker Analytical X-Ray Instruments Inc., Madison, USA (1998), Sheldrick “SADABS, Program for Empirical Correction of Area Detector Data” Version 2.03, University of G ⁇ ttingen, Germany (2001)].
  • the program SHELXTL [cf. Sheldrick "SHELXTL, Structure Determination Programs", Version 6.12, Bruker Analytical X-Ray Instruments Inc., Madison, USA (2001)] was used to solve the structures and for molecular graphics.
  • compound 8 can be prepared as described herein in eight steps. This material can be resolved by chiral SFC as described herein to give compounds 9 and ent-9. After cleavage of the Boc-protective group, reductive amination can be used to introduce the n-propyl group on the nitrogen atom. The resulting masked catechol amines can be deprotected under standard conditions by treatment with 48% HBr or by reaction with BBr 3 to give compounds 10 and ent-lO. Further reaction of 10 with CH 2 ClBr or a related reagent in the presence of base can be applied to give a compound of the invention (compound 11).
  • Compound 12 can be prepared as described in EP Patent No. 1274411, the contents of which are hereby incorporated by reference.
  • Compound 12 is referred to as (-)- GMC6650 in the above-identified patent.
  • Example 1 Compounds 11 and 12 convert into the catechol-containing active metabolite of compound 10 upon in-vivo administration.
  • the active metabolite (i.e. Compound 10) was found to function as a potent agonist at both the Dl and D2 receptors in-vitro. As discussed in greater detail below, the data generated from in-vivo experiments indicate that this active metabolite possesses a superior profile against other dopamine agonists and is on par with the efficacy seen with L-DOPA/apomorphine treatment.
  • the ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment.
  • the ability of the compounds to either stimulate or inhibit the D 2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D 2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl 2 , 0.9 mM CaCl 2 , 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
  • the cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl 2 ) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes.
  • 100 micro-1 S buffer 0.1 M HCl and 0.1 mM CaCl 2
  • 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate
  • D 5 assay Concentration-dependent stimulation of intracellular Ca 2+ release by dopamine in hDs-transfected CHO-Gal6 cells.
  • the cells were loaded with fluoro-4, a calcium indicator dye, for Ih.
  • Calcium response (fluorescence change) was monitored by FLIPR (fluorometric imaging plate reader) for 2.5 min. Peak responses (EC50) were averaged from duplicate wells for each data point and plotted with drug concentrations (cf. Figure 2 for dopamine).
  • Compound 10 was demonstrated to act as a D 5 agonist in this assay.
  • Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both.
  • the rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott, Brain Res., 1970, 24, 485; Setler, et al. Eur. J. Pharmacol, 1978, 50(4), 419; and Ungerstedt, et al. "Advances in Dopamine Research” (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219).
  • 6-OHDA (6- hydroxydopamine) is a neurotoxin used by neurobiologists to selectively kill dopaminergic neurons at the site of injection in the brain in experimental animals.
  • the nigrostraital dopamine cells are destroyed on one side of the brain (unilateral) by injecting 6-OHDA into the median forebrain bundle, located in front of the substantia nigra.
  • This unilateral injection combined with stimulation by dopamine agonists such as apomorphine will induce rotation behaviour as only one side of the brain is stimulated.
  • Experiments consist of determining a minimum effective dose (MED) to induce rotation for the compound in question. Once a MED has been determined, a second experiment is performed to determine the MED of the compound to overcome Nemonapride block (MEDNemonap ⁇ de).
  • Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor.
  • Compound 10 possess 'apomorphine '-like profiles with a D1/D2 ratio of about 2-4 as compared to a ratio of about 3 for apomorphine. Moreover, the duration of action observed was ca. 18h for the compound which is significantly higher than that seen with L-DOPA / apomorphine. A Dl component could not be observed for D2-agonists as exemplified by pramipexole and rotigotine.
  • Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 dopamine receptors. Pramipexole, an agonist at D2 receptors is ineffective in this model.
  • MPTP l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine
  • AMPT 250mg/kg subcutaneously
  • AMPT alpha-methyl-p- tyrosine
  • a drug that temporarily reduces brain catecholamine activity in this case especially dopamine levels.
  • Various dopaminergic compounds were tested for their ability to reverse the motility deficits produced in this model. Both L-DOPA/Benserazide, and apomorphine restored locomotion in the mice in a dose-dependent manner.
  • Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain.
  • the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
  • This model was used to evaluate whether or not Compound 10 exhibits the same superiority as L- DOPA and apomorphine over D2 agonists.
  • a dose response experiment for Compound 10 was performed and there was a dose-dependent trend for reversing the hypomotility deficits induced by severe depletion of endogenous dopamine.
  • a final experiment directly comparing the effects of apomorphine, pramipexole and Compound 10 in this model was performed and confirmed that Compound 10 was able to restore locomotion in MPTP mice treated and was superior to pramipexole.
  • Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain.
  • rats received once daily injections of the test compounds subcutaneously and were observed for 3h following injection. Each animal was observed for 1 minute every 20 min throughout the 3h period for the presence of dyskinesias using the Abnormal
  • AIMS Involuntary Movement Scale
  • Rats received drug for 14 consecutive days and were scored on days 1, 2, 3, 4, 5,
  • Example I scores of about 70. There were no differences between L-DOPA and apomorphine treated groups. Following this experiment all rats were given subcutaneous injections of compound 10 from day 15-19 in order to determine how Example I influenced the severity of dyskinesia seen in the apomorphine and L-DOPA groups. Dykinesia scoring was performed on day 19 of the experiment (corresponding to 5 days on compound 10). The data showed a partial reversal of the dyskinesias induced by L-DOPA and apomorphine to about the level of dyskinesias induced by compound 10 (which did not cause an increase in dyskinesia in group 1 as compared to the score of about 30 observed after 12 days of treatment).
  • Domperidone is an anti-dopaminergic drug that suppresses nausea and vomiting.
  • Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity.
  • AUC area under the curve
  • the assessment of motor disability was performed by a trained observer blinded to the treatment.
  • L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887).
  • the dose chosen for this challenge is at the top of the dose response curve for this drug.
  • Compound 10 dosed subcutaneously produced dose-related increases in locomotor activity and reversal of motor disability tending to produce in a response greater than for L-DOPA (12.5mg/kg, p.o.). Both test compounds produced a prolonged reversal of motor disability compared to L-DOPA and were as efficacious as L-DOPA. Compound 10 produced a prolonged reversal of motor disability compared to L-DOPA and was as efficacious as L-DOPA.
  • the ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment.
  • the cells were incubated for 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-L S buffer (0.1 M HCl and 0.1 mM CaCl 2 ) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates were shaken for 10 minutes.
  • 100 micro-L S buffer 0.1 M HCl and 0.1 mM CaCl 2
  • 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc
  • the ability of the compounds to either stimulate or inhibit the D 2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D 2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl 2 , 0.9 mM CaCl 2 , 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
  • the cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl 2 ) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes.
  • 100 micro-1 S buffer 0.1 M HCl and 0.1 mM CaCl 2
  • 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate
  • D5 assay Concentration-dependent stimulation of intracellular Ca 2+ release by dopamine in hDs-transfected CHO-Gal6 cells.
  • the cells were loaded with fluoro-4, a calcium indicator dye, for Ih.
  • Calcium response (fluorescence change) was monitored by FLIPR (fluorometric imaging plate reader) for 2.5 min. Peak responses (EC50) were averaged from duplicate wells for each data point and plotted with drug concentrations.
  • the active metabolite or Compound 10 was found to be a D5 agonist in this assay.
  • Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both.
  • the rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott, Brain Res. 24, 485 (1970); Setler, et ah, Eur. J. Pharmacol, 50(4), 419 (1978); and Ungerstedt, et al, "Advances in Dopamine Research” (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219).
  • 6- OHDA 6-hydroxydopamine
  • 6-OHDA 6-hydroxydopamine
  • Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor.
  • MEDNemonap ⁇ de is known a third experiment is run using the MEDNemonap ⁇ de dose and observing the effect of the Dl antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally, the effect of combined treatment with SCH 23390 and Nemonapride.
  • Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 dopamine receptors. Pramipexole, an agonist at D2-like receptors is ineffective in this model.
  • mice previously treated with MPTP (2xl5mg/kg subcutaneously) and that had stable lesions are used and vehicle treated mice served as normal controls.
  • MPTP l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine
  • AMPT 250mg/kg subcutaneously
  • AMPT alpha-methyl-p- tyrosine
  • brain catecholamine activity in this case especially dopamine levels.
  • rescue of locomotive deficits is attempted with The active metabolite or compound 10 and activity was recorded for an additional 1.5 hours.
  • the first 30 min of data collected after the rescue treatment was 'contaminated' due to stressing the animals with handling and injection as evidenced by increased levels in the vehicle controls therefore the data were analyzed using the last 1 hour of recorded data.
  • Various dopaminergic compounds are tested for their ability to reverse the motility deficits produced in this model. Both L-DOPA/Benserazide, and apomorphine restored locomotion in the mice in a dose-dependent manner.
  • Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain.
  • the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
  • a rat dyskinesia model reported in the literature (Lundblad, et al., Eur. JNeurosci., 2002, 15, 120) was used to examine the effects of the active metabolite vs. L-DOPA/benserazide with respect to dyskinesias that were assessed as abnormal involuntary movements (AIMs) in 'parkinsonian' rats.
  • AIMs abnormal involuntary movements
  • mice were scored according to AIM-scoring by recording dyskinesias using the Abnormal Involuntary Movement Scale (AIMS) as described previously (Lundblad, et al, Eur. J Neurosci., 2002, 15, 120).
  • Days 15 - 26 Group A animals were treated with the test drug (as group B) instead of L-DOPA/benserazide.
  • Day 15, 16, 17, 19, 22, 24 and 26 Animals scored according AIM-scoring.
  • group A animals had dyskinesia scores of 10-12, which remained constant until day 12.
  • group B animals had significantly fewer dyskinesias (scores of 2-4).
  • the degree of dyskinesias did not change during the study.
  • Group A animals After shifting group A animals from L-dopa/benserazide to the test drug, their level of dyskinesia gradually decreased to the level observed for the other group of animals.
  • Compound 11 induced significantly less dyskinesia than L-DOPA and was able to reduce the dyskinesias induced by L- DOPA.
  • Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity. The assessment of motor disability was performed by a trained observer blinded to the treatment.
  • L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887).
  • the dose chosen for this challenge is at the top of the dose response curve for this drug.
  • Compound 11 dosed p.o.
  • compound 10 dosed subcutaneously
  • Both test compounds produced a prolonged reversal of motor disability compared to L- DOPA and were as efficacious as L-DOPA.
  • Cryopreserved pooled male rat hepatocytes (Sprague Dawley) and pooled human hepatocytes from 10 donors (male and female) were purchased from In Vitro Technologies Inc., BA, USA. Cells were thawed at 37 0 C in a water bath, live cells counted and seeded in a total of 100 micro-L in Dulbecco's modified Eagle medium (high glucose) with 5 mM Hepes buffer in 96 well plates, each well containing 250.000 and 500.000 cells/mL for rat and human hepatocytes, respectively.
  • Dulbecco's modified Eagle medium high glucose
  • Incubations were started after 15 min of pre-incubation and stopped at time points of 0, 5, 15, 30 and 60 min for rats and at 0, 30, 60, 90 and 120 min for human hepatocytes. Incubations were stopped by addition of an equal volume of ice-cold acetonitrile containing 10% 1 M HCl. Following centrifugation, 20 micro-L of the supernatants were injected on a HPLC Column Atlantis dC18 3 micro-m, 150 x 2.1 mm i.d. (Waters, MA, USA). The mobile phase had the following composition: A: 5% acetonitrile, 95% H 2 O, 3.7 ml/125% aq. NH3, 1.8 mL/L formic acid.
  • Mobile phase B 100% acetonitrile and 0.1% formic acid.
  • the flow rate was 0.3 ml/min.
  • the gradient operated from 0% to 75 % B from 5 min to 20 min and the eluate was analyzed using a Q- TOFmicro mass spectrometer (Waters, MA, USA). Formation of the product/metabolite was confirmed by accurate mass measurements and comparison with a synthesized standard giving coinciding retention times. In this assay, the metabolism of Compound 11 to Compound 10 was demonstrated.
  • the ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment.
  • the cells were incubated for 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-L S buffer (0.1 M HCl and 0.1 mM CaCl 2 ) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates were shaken for 10 minutes.
  • 100 micro-L S buffer 0.1 M HCl and 0.1 mM CaCl 2
  • 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc
  • the ability of the compounds to either stimulate or inhibit the D 2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D 2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl 2 , 0.9 mM CaCl 2 , 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
  • the cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl 2 ) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes. 60 micro-L of the reaction were transferred to cAMP FlashPlates (DuPont
  • Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both.
  • the rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott; Brain Res., 24, 485 (1970); Setler, et ah, Eur. J. Pharmacol, 50(4), 419 (1978); and Ungerstedt, et ⁇ /./'Advances in Dopamine Research” (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219).
  • 6- OHDA 6-hydroxydopamine
  • 6-OHDA 6-hydroxydopamine
  • Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor.
  • MEDNemonap ⁇ de is known a third experiment is run using the MED N emonap ⁇ de dose and observing the effect of the Dl antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally, the effect of combined treatment with SCH 23390 and Nemonapride.
  • Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 receptors. Pramipexole, an agonist at D2 receptors is ineffective in this model.
  • MPTP l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine
  • AMPT 250mg/kg subcutaneously
  • AMPT alpha-methyl-p- tyrosine
  • Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it was used to prevent metabolism of L-DOPA to dopamine outside the brain.
  • the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
  • DOPA and apomorphine over D2 agonists DOPA and apomorphine over D2 agonists.
  • a dose response experiment for was performed and there was a dose-dependent trend for reversing the hypomotility deficits induced by severe depletion of endogenous dopamine.
  • a final experiment directly comparing the effects of apomorphine, pramipexole and Compound 10 was performed. It was confirmed that Compound 10 was able to restore locomotion in MPTP mice treated and was superior to pramipexole.
  • a rat dyskinesia model reported in the literature (Lundblad, et ah, Eur. J Neurosci., 2002, 15, 120) was used to examine the effects of Compound 12 vs. L-DOPA/benserazide with respect to dyskinesias that were assessed as abnormal involuntary movements (AIMs) in 'parkinsonian' rats.
  • AIMs abnormal involuntary movements
  • mice were scored according to AIM-scoring by recording dyskinesias using the Abnormal Involuntary Movement Scale (AIMS) as described previously.
  • AIMS Abnormal Involuntary Movement Scale
  • Days 15 - 26 Group A animals were treated with Compound 12 (as group B) instead of L-DOPA/benserazide.
  • Day 15, 16, 17, 19, 22, 24 and 26 Animals scored according AIM-scoring. Results
  • group A animals had dyskinesia scores of 70-80, which remained constant until day 15.
  • group B animals had significantly fewer dyskinesias (scores of 10-25).
  • the degree of dyskinesias did not change during the study.
  • group A animals After shifting group A animals from L-DOPA/benserazide to compound 12 for 10 days, their level of dyskinesia gradually decreased to scores of 30-35.
  • compound 12 induced significantly less dyskinesia than L-DOPA and was able to reduce the dyskinesias induced by L-DOPA.
  • Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity. The assessment of motor disability was performed by a trained observer blinded to the treatment.
  • L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887).
  • the dose chosen for this challenge is at the top of the dose response curve for this drug.
  • Compound 12 dosed p.o.
  • Compound 10 dosed p.o.
  • Both test compounds produced a prolonged reversal of motor disability compared to L-DOPA and were as efficacious as L-DOPA.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Psychology (AREA)
  • Inorganic Chemistry (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Disclosed herein are methods of treating Parkinsons disease while maintaining a low dyskinesia induction profile and methods of reversing dyskinesias comprising administering a therapeutically effective amount of a compound of the invention. The present invention further relates to uses and pharmaceutical compositions of said compounds in the manufacture of medicaments in treating the same.

Description

TREATMENT OF DYSKINESIA RELATED DISORDERS
FIELD OF THE INVENTION
Aspects of the subject invention relate to methods of treating Parkinson's disease while maintaining a low dyskinesia induction profile and to methods of reversing dyskinesias comprising administering therapeutically effective amount of a compound disclosed herein. The present invention further relates to uses and pharmaceutical compositions of said compounds in the manufacture of medicaments in treating the same or other movement disorders such as Huntington's chorea.
BACKGROUND ART
The use of dopamine-replacing agents in the symptomatic treatment of Parkinson's disease (PD) has undoubtedly been successful in increasing the quality of life of patients. L-DOPA, which has been used for many years and remains the gold standard for treatment of PD, alleviates motor symptoms of PD characterized by the slowness of movement (bradykinesia), rigidity and/or tremor. It is understood that L-DOPA acts as a prodrug which is bio-metabolized into dopamine (DA). DA in turn activates dopamine receptors in the brain which fall into two classes: Dl and D2 receptors. Dl receptors can be divided into Di and D5 receptors while D2 receptors can be divided into D2, D3, and D4 receptors. However, dopamine-replacement therapy does have limitations, especially following long-term treatment. The duration period response to a dose of L-DOPA becomes progressively shorter over the years, and periods in which the patient responds to the drug become complicated by the appearance of a range of side-effects.
The side-effects may manifest as dyskinesias, which can be seen either when the patient is undergoing dopamine replacement therapy or even when the patient is off therapy. Dyskinesias are abnormal involuntary movement disorders. The abnormal movements may manifest as chorea (involuntary, rapid, irregular, jerky movements that may affect the face, arms, legs, or trunk), ballism (involuntary movements similar to chorea but of a more violent and forceful nature), dystonia (sustained muscle contractions, usually producing twisting and repetitive movements or abnormal postures or positions) and/or athetosis (repetitive involuntary, slow, sinuous, writhing movements, which are especially severe in the hands). PD afflicted patients may cycle between "on" periods which are complicated by dyskinesia and "off' periods in which they are severely parkinsonian. As a consequence they may experience profound disability despite the fact that L-DOPA remains an effective anti-Parkinson agent throughout the course of the disease (Obeso, et al. Neurology 2000, 55, S13-23). Dopamine agonists such as bromocriptine, lisuride, pramipexole, ropinirole and pergolide are less efficacious than L-DOPA, particularly in moderate-to-severe PD. However, their side-effect profile is different from that of L-DOPA. It is worth noticing that DA agonists do cause less dyskinesias that L-DOPA but this is of limited value to PD patients with dyskinesias because many of them have moderate- to-severe PD and hence they need the efficacy of L-DOPA
Dyskinesias and other movement disorders from dysfunction of the basal ganglia are of major socio-economic importance. Many attempts have been made to develop agents to prevent and/or treat dyskinesias although such attempts have met with limited success. There is, therefore, a need to provide novel agents to treat dyskinesia.
The 6-hydroxydopamine (6-OHDA) lesion model of parkinsonism in the rat has provided an invaluable tool in the investigation of PD at a preclinical level and for the evaluation of novel therapeutic options (Schwarting and Huston, Prog. Neurobiol. 1996, 50, 275-331). One of the most widely used 6-OHDA paradigms is the evaluation of rotational behavior in rats which bear a discrete degeneration of the dopaminergic nigrostriatal pathway (Ungerstedt and Aburthnott, Brain Res. 1970, 24, 485). In this model, 6-OHDA is unilaterally infused into the nigrostriatal pathway, striatum or medial forebrain bundle (MFB), producing a functional imbalance between the dopaminergic nigrostriatal systems. Administration of drugs directly stimulating dopamine receptors, such as the dopamine metabolic precursor L-DOPA and the dopamine agonist apomorphine produces a rotational behavior directed away from the body side in which 6-OHDA has been infused.
In addition to motor-related deficits, the 6-OHDA model can be used to reproduce other features of PD. The development of both sensitized rotational behavior as well as abnormal involuntary movements (AIMs) has been observed in rats injected with 6-OHDA either in the striatum or in the MFB, and chronically treated with L-DOPA, therefore providing further an animal model for the study of L-DOPA induced dyskinesia (Lundblad, et aL Eur. J Neurosci. 2002, 15,120-132). During chronic treatment in this model, L-DOPA but not bromocriptine induces a gradual development of AIMs. Based on these observations, it has been accepted that rats lesioned with 6- OHDA exhibit motor deficits that share essential functional similarities with Parkinson's dyskinesia and can be used to evaluate the potential of a treatment to provide treatments for dyskinesia.
In an attempt to identify new therapies for treating dyskinesia and other related movement disorders, applicants have surprisingly found that (4aR, 1OaR)-I -n-propyl- 1,2,3 ,4,4a,5,l 0,1 Oa- octahydro-benzo[g]quinoline-6,7-diol as a potent D1/D2 agonist [herein referred to as Compound 10]; (6aR, 10aR)-7-/?-propyl-6,6a,7,8,9, 10, 1 Oa, 11-octahydro- 1 ,3-dioxa-7-aza- cyclopenta[a] anthracene [herein referred to as Compound H]; and (4aR,l OaR)-I -n-propyl- 2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one [herein referred to as Compound 12] have favorable profiles in rats with unilateral 6-OHDA lesions. They induce less dyskinesias than L-DOPA and apomorphine, and reduce L-DOPA induced dyskinesias more effectively than D2 agonists, as exemplified by pramipexole. Hence, Compounds 10, 11 and 12 have the potential to become the first PD drugs with L-DOP A-like efficacy and a favorable profile not only in terms of both induction of dyskinesia, but also as a medication for the reversal of dyskinesias.
Accordingly, it is expected that above identified compounds can be used to treat dyskinesias and other related movement disorders such as Huntington's chorea. Moreover, the present invention contemplates the use of the corresponding racemic trans mixture. The present invention further provides methods of treating Parkinson's disease with a low dyskinesia induction profile comprising administering a therapeutically effective amount of said compound. In one aspect, the treatment of Parkinson's disease is as efficacious as L-DOPA treatment. Further provided are methods of reversing dyskinesias or treating Parkinson's disease comprising administering said compound and pharmaceutical compositions thereof.
SUMMARY OF THE INVENTION
One aspect of the invention is concerned with the use of (4aR,l OaR)-I -n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile. Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
A separate aspect of the invention relates to the use of (4aR, 1 OaR)- 1 -n-propyl- 1 ,2,3 ,4,4a,5 , 10, 1 Oa- octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament treating Parkinson's disease.
Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,10a-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease.
A separate aspect of the invention relates to the use of (4aR,l OaR)-I -n-propyl- 1,2,3 ,4,4a,5,l 0,1 Oa- octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
Another aspect relates to the use of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,10a-octahydro- benzo[g]quinoline-6,7-diol in the preparation of a medicament for reversing dyskinesias.
Another aspect is directed to a pharmaceutical composition comprising (4aR,l OaR)-I -n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Separate aspects of the invention relate to a pharmaceutical composition comprising racemic trans- l-n-propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (4aR, 1 OaR)- 1 -n-propyl- 1 ,2,3 ,4,4a,5 ,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
A separate aspect relates to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile a low dyskinesia induction profile comprising administering a therapeutically effective amount of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol .
Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (4aR, 1OaR)-I -n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
Yet another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of racemic trans-1-n-propyl- 1,2,3 ,4,4a,5, 10,1 Oa-octahydro- benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
One aspect of the invention is concerned with the use of (6aR,10aR)-7-/?-propyl- 6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
A separate aspect of the invention relates to the use of (6aR,10aR)-7-/?-propyl-6,6a,7,8,9,10,10a,l 1- octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
Another aspect is directed to a pharmaceutical composition comprising (6aR,10aR)-7-n-propyl-
6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof, for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Separate aspects of the invention relate to a pharmaceutical composition comprising (6aR,10aR)-7- n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (6aR, 10aR)-7-n-propyl-6,6a,7,8,9, 10, 10a, 11 -octahydro- 1 ,3-dioxa-7-aza-cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof. Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3- dioxa-7-aza-cyclopenta[a]anthracene or a pharmaceutically acceptable salt thereof.
One aspect of the invention is concerned with the use of (4aR,l OaR)-I -n-propyl-
2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile
A separate aspect of the invention relates to the use of (4aR,l OaR)-I -n-propyl-
2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease.
Yet another aspect relates to the use of (4aR,10aR)-l-/?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro- lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
Another aspect is directed to a pharmaceutical composition comprising (4aR,l OaR)-I -n-propyl- 2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one, or a pharmaceutically acceptable salt thereof, for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Separate aspects of the invention relate to a pharmaceutical composition comprising (4aR,10aR)-l- /?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
Another aspect is directed to a method of treating Parkinson's disease while maintaining a low dyskinesia induction profile comprising administering a therapeutically effective amount of (4aR,10aR)-l-n-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof. Another aspect is directed to a method of reversing dyskinesias comprising administering a therapeutically effective amount of (4aR,10aR)-l-/?-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH- benzo[g]quinolin-6-one or a pharmaceutically acceptable salt thereof.
DETAILED DESCRIPTION
The compounds of the present invention contain two chiral centers (denoted with * in the below formula)
Figure imgf000008_0001
The compounds of the invention can exist in two different diastereomeric forms, the cis- and trans- isomers, both of which can exist in two enantiomeric forms. The present invention relates only to the trans racemate and the (4aR, 10aR)-enantiomer.
(4aS 10aR)-enantιomer
Figure imgf000008_0002
(4aS 10aS)-enantιomer
As previously indicated, the present invention is based on the discovery that (4aR, 1OaR)-I -n- propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol (herein referred to as "Compound 10") reversed dyskinesias induced by L-DOPA/benserazide and apomorphine in rats lesioned with 6-OHDA. The corresponding trans racemate also falls within the scope of this invention.
Additionally, the compound of the present invention contain two chiral centers (denoted with * in the below formula)
Figure imgf000009_0001
The compound of the invention can exist in two different diastereomeric forms, the cis- and trans- isomers, both of which can exist in two enantiomeric forms. The present invention relates only to the trans racemate and the (6aR, 10aR)-enantiomer.
Figure imgf000009_0002
as diastereomers as racemate of formula I (6aR 10aS)-enantιomer (6aS 10aR)-enantιomer trans diastereomers
Figure imgf000009_0003
of formula I
Figure imgf000009_0004
10aR)-enantιomer (6aS 10aS)-enantιomer
As previously indicated, the present invention is based on the discovery (6aR,10aR)-7-/?-propyl- 6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza-cyclopenta[a]anthracene (herein referred to as "Compound 11") reversed dyskinesias induced by L-DOPA/benserazide and apomorphine in rats lesioned with 6-OHDA.
Furthermore, the present invention is based on the discovery that (4aR, 1OaR)-I -n-propyl- 2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6-one (herein referred to as Compound 12) has favorable profiles in rats with unilateral 6-OHDA lesions. It induce sless dyskinesias than L-DOPA and apomorphine, and reduces L-DOPA induced dyskinesias more effectively than D2 agonists, as exemplified by pramipexole.
The invention is explained in greater detail below but this description is not intended to be a detailed catalog of all the different ways in which the invention may be implemented, or all the features that may be added to the instant invention. Definitions
As used herein, "dyskinesia" refers to a condition characterized by abnormal involuntary movements that are associated with disorders of brain regions known as the basal ganglia. The dyskinesia may be an "L-DOP A-induced dyskinesia" that arises and is a complication of the treatment of Parkinson's disease (the most common basal ganglia disease). Dyskinesia can physically manifest in two forms, chorea and dystonia. Chorea consists of involuntary, continuous, purposeless, abrupt, rapid, brief, unsustained and irregular movements that flow from one part of the body to another. Dystonia refers to sustained muscle contractions that cause twisting and repetitive movements or abnormal postures.
"Treating" or "treatment" refers to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both, and inhibit at least one physical parameter which may not be discernible to the patient. Further, "treating" or "treatment" refers to delaying the onset of the disease or disorder or at least symptoms thereof in a patient which may be exposed to or predisposed to a disease or disorder even though that patient does not yet experience or display symptoms of the disease or disorder.
"Therapeutically effective amount" refers to the amount of a compound that, when administered to a patient for treating a disease or disorder, is sufficient to affect such treatment for the disease or disorder. The "therapeutically effective amount" will vary depending on the compound, the disease or disorder and its severity and the age and weight of the patient to be treated.
As used herein, the phrase "while maintaining a low dyskinesia profile" refers to the dyskinesia profile as seen in patients who have been treated via continuous dopaminergic stimulation. Treatments involving continuous dopaminergic stimulation are described in Stocchi and Olanow, Neurology 2004, 2004, 62, S56-S63; and Hilary, et al, Journal of Neurology 2004, 251, 11, 1370- 1374.
As used herein, (4aR,10aR)-l-/?-propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol as a potent D1/D2 agonist is referred to as Compound 10.
As used herein, (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza- cyclopenta[a] anthracene is referred to as Compound 11. As used herein, (4aR,10aR)-l-n-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6- one [herein referred to as Compound 12.
Compound 10, 11 or 12 may be used to treat dyskinesia as a monotherapy (i.e. use of the compound alone); as an adjunct to compositions to prevent dyskinetic side-effects caused by the composition (e.g. as an adjunct to L-DOPA or apomorphine given to treat parkinsonian patients) or alternatively the compound may be given in combination with other treatments which also reduce dyskinesia (e.g. opioid receptor antagonists, (α2-adrenoreceptor- antagonists, cannabinoid CBI- antagonists, NMDA receptor-antagonists, cholinergic receptor antagonists, histamine H3-receptor agonists, and globus pallidus/subthalamic nucleus lesion/deep brain stimulation).
The present invention is further concerned with the concurrent, separate or sequential use in the treatment of Parkinson's disease while reducing dyskinesia induced by L-DOPA or a dopamine agonist comprising administering a therapeutically effective amount of Compound 10, 11 or 12 or a pharmaceutically salt thereof.
In one embodiment, the dyskinesia is associated with a basal ganglia-related movement disorder.
In another embodiment, the dyskinesia is associated with Parkinson's disease.
One embodiment relates to dyskinesia associated with idiopathic Parkinson's disease or postencephalitic Parkinsonism.
In one embodiment, the dyskinesia is associated with off-dystonia in Parkinson's disease.
In a separate embodiment, the dyskinesia arises as a side-effect of a therapeutic agent to treat Parkinson's disease.
In yet another embodiment, the dyskinesia is associated with dopamine replacement therapy. In one embodiment, dopamine replacement therapy agent is selected from the group consisting of rotigotine, ropinirole, pramipexole, cabergoline, bromocriptine, lisuride, pergolide, L-DOPA and apomorphine. In one embodiment, the dyskinesia is established as a result of repeated administration of L-DOPA.
As previously indicated, the present invention provides for a pharmaceutical composition comprising Compound 10, 11 or 12 or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile, and to a pharmaceutical composition comprising racemic trans-1-n-propyl- l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
In one embodiment, the pharmaceutical composition additionally comprises a MAO-B inhibitor.
In a one embodiment, the MAO-B inhibitor is selegine. In a separate embodiment, the MAO-B inhibitor is rasagiline.
In another embodiment, the invention relates to a pharmaceutical composition comprising a therapeutically effective amount of Compound 10, 11 or 12, or a pharmaceutically acceptable acid addition salt thereof, and one or more pharmaceutically acceptable carriers, diluents and excipients.
In a specific embodiment of the invention, the mammal is a human subject.
The therapeutically effective amount of Compound 10, 11 or 12, calculated as the daily dose of
Compound 10, 11 or 12 above as the free base, is suitably between 0.01 and 125 mg/day, more suitable between 0.05 and 100 mg/day, e.g. preferably between 0.1 and 50 mg/day.
In a specific embodiment, the daily dose of Compound 10, 11 or 12 is between 1.0 and 10 mg/day.
In another embodiment, the daily dose of Compound 10, 11 or 12 is less than about 1.0 mg/day.
In a separate embodiment, the daily dose of Compound 10, 11 or 12 is about 0.10 mg/day.
In a further embodiment, the invention provides an oral formulation comprising from 0.001 mg to
125 mg of Compound 10, 11 or 12.
In a further embodiment, the invention provides an oral formulation comprising from 0.001 mg to 0.100 mg of Compound 10, 11 or 12. In a further embodiment, the invention provides an oral formulation comprising from 0.01 mg to 1.0 mg of Compound 10, 11 or 12.
In a further embodiment, the invention provides an oral formulation comprising from 0.10 mg to 10 mg of Compound 10, 11 or 12.
Pharmaceutically Acceptable Salts
Compound 10, 11 or 12 forms pharmaceutically acceptable acid addition salts with a wide variety of organic and inorganic acids. Such salts are also part of this invention. A pharmaceutically acceptable acid addition salt of Compound 10, 11 or 12 is formed from a pharmaceutically acceptable acid as is well known in the art. Such salts include the pharmaceutically acceptable salts listed in Journal of Pharmaceutical Science, 1977, 66, 2-19 and are known to the skilled person. Typical inorganic acids used to form such salts include hydrochloric, hydrobromic, hydriodic, nitric, sulphuric, phosphoric, hypophosphoric, metaphosphoric, pyrophosphoric, and the like. Salts derived from organic acids, such as aliphatic mono and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxyalkanoic and hydroxyalkandioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, may also be used. Such pharmaceutically acceptable salts thus include the chloride, bromide, iodide, nitrate, acetate, phenylacetate, trifluoroacetate, acrylate, ascorbate, benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-acetoxybenzoate, isobutyrate, phenylbutyrate, α-hydroxybutyrate, butyne-l,4-dicarboxylate, hexyne-l,4-dicarboxylate, caprate, caprylate, cinnamate, citrate, formate, fumarate, glycollate, heptanoate, hippurate, lactate, malate, maleate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, isonicotinate, oxalate, phthalate, teraphthalate, propiolate, propionate, phenylpropionate, salicylate, sebacate, succinate, suberate, benzenesulfonate, p-bromobenzenesulfonate, chlorobenzenesulfonate, ethylsulfonate, 2-hydroxyethylsulfonate, methylsulfonate, naphthalene- 1- sulfonate, naphthalene-2-sulfonate, naphthalene- 1,5-sulfonate, p-toluenesulfonate, xylenesulfonate, tartrate, and the like.
Pharmaceutical Compositions
Methods of the preparation of solid pharmaceutical compositions are also well known in the art. Tablets may thus be prepared by mixing the active ingredient with ordinary adjuvants, fillers and diluents and subsequently compressing the mixture in a convenient tabletting machine. Examples of adjuvants, fillers and diluents comprise microcrystalline cellulose, corn starch, potato starch, lactose, mannitol, sorbitol talcum, magnesium stearate, gelatine, lactose, gums, and the like. Any other adjuvant or additive such as colorings, aroma, preservatives, etc. may also be used provided that they are compatible with the active ingredients.
In particular, the tablet formulations according to the invention may be prepared by direct compression of Compound 10, 11 or 12 in admixture with conventional adjuvants or diluents. Alternatively, a wet granulate or a melt granulate of Compound 10, 11 or 12, optionally in admixture with conventional adjuvants or diluents may be used for compression of tablets.
Solutions of Compound 10, 11 or 12 for injections may be prepared by dissolving the active ingredient and possible additives in a part of the solvent for injection, preferably sterile water, adjusting the solution to the desired volume, sterilization of the solution and filling in suitable ampoules or vials. Any suitable additive conventionally used in the art may be added, such as tonicity agents, preservatives, antioxidants, solubilizing agents, etc.
BRIEF DESCRIPTION OF THE FIGURES
FIGURE 1: Crystal structure of compound ent-lθ. The absolute configuration was determined by the anomalous scattering of the 'heavy' bromine atom.
FIGURE 2: Dose-response curve for the concentration-dependent stimulation of intracellular Ca2+ release by dopamine in bLVtransfected CHO-GaI 6 cells.
EXPERIMENTAL SECTION
Analytical LC/MS data were obtained on a PE Sciex API 150EX instrument equipped with atmospheric pressure photo ionization and a Shimadzu LC-8A/SLC-10A LC system. Purity was determined by integration of the UV (254 nm) and ELSD traces. MS instruments are from Peskier (API), equipped with APPI-source and operated in positive ion mode. The retention times in the UV-trace (RT) are expressed in min. Solvents A was made of 0.05% TFA in water, while solvent B was made of 0.035% TFA and 5% water in acetonitrile. Several different methods have been used: Method 25: API 150EX and Shimadzu LCIOAD/SLC-IOA LC system. Column: dC-18 4.6x30mm, 3μm (Atlantis, Waters). Column temperature: 40 0C. Gradient: reverse phase with ion pairing. Flow: 3.3 mL/min. Injection volume: 15 μL. Gradient: 2% B in A to 100% B over 2.4 min then 2% B in A for 0.4 min. Total run time: 2.8 min.
Method 14: API 150EX and Shimadzu LC8/SLC-10A LC system. Column: C-18 4.6x30mm,
3.5μm (Symmetry, Waters). Column temperature: rt. Gradient: reverse phase with ion pairing. Flow: 2mL/min. Injection volume: 10 μL. Gradient: 10% B in A to 100% B over 4 min then 10%
B in A for 1 min. Total run time: 5 min.
X-ray crystal structure determination was performed as follows. The crystal of the compound was cooled to 120 K using a Cryostream nitrogen gas cooler system. The data were collected on a Siemens SMART Platform diffractometer with a CCD area sensitive detector. The structures were solved by direct methods and refined by full-matrix least-squares against F2 of all data. The hydrogen atoms in the structures could be found in the electron density difference maps. The non- hydrogen atoms were refined anisotropically. All the hydrogen atoms were at calculated positions using a riding model with O-H=0.84, C-H = 0.99-1.00, N-H = 0.92-0.93 A. For all hydrogen atoms the thermal parameters were fixed [U(H) = 1.2 U for attached atom]. The Flack x-parameters are in the range 0.0(l)-0.05(l), indicating that the absolute structures are correct. Programs used for data collection, data reduction and absorption were SMART, SAINT and SADABS [cf. "SMART and SAINT, Area Detector Control and Integration Software", Version 5.054,Bruker Analytical X-Ray Instruments Inc., Madison, USA (1998), Sheldrick "SADABS, Program for Empirical Correction of Area Detector Data" Version 2.03, University of Gόttingen, Germany (2001)]. The program SHELXTL [cf. Sheldrick "SHELXTL, Structure Determination Programs", Version 6.12, Bruker Analytical X-Ray Instruments Inc., Madison, USA (2001)] was used to solve the structures and for molecular graphics.
Synthesis of the compounds of the invention (compounds 10 and 11)
Starting from compound 1 whose synthesis is described in the literature prepared as described in Taber et al, J. Am. Chem. Soc, 124(42), 12416 (2002), compound 8 can be prepared as described herein in eight steps. This material can be resolved by chiral SFC as described herein to give compounds 9 and ent-9. After cleavage of the Boc-protective group, reductive amination can be used to introduce the n-propyl group on the nitrogen atom. The resulting masked catechol amines can be deprotected under standard conditions by treatment with 48% HBr or by reaction with BBr3 to give compounds 10 and ent-lO. Further reaction of 10 with CH2ClBr or a related reagent in the presence of base can be applied to give a compound of the invention (compound 11).
Figure imgf000016_0001
rac-compound 10
Figure imgf000016_0002
Figure imgf000016_0003
compound 11 enf-compound 11 rac-compound 11 (6aR 10aR-enantιomer) (6aS 10aS-enantιomer)
Synthesis of compounds 10 and ent-10. 7-Iodo-l,2,6-trimethoxy-naphthalene (compound 2).
Figure imgf000016_0004
compound 1 compound 2
To a stirred solution of compound 1 (26.2 g; prepared as described in Taber et ah, J. Am. Chem. Soc, 124(42), 12416 (2002) in dry THF (200 mL) under argon and at -78 0C was slowly added s- butyl lithium (1.2 M in cyclohexane, 110 mL). The solution was stirred at -78°C for 3h. A solution of iodine (30.5 g) in dry THF (50 mL) was added over a period of 10 min. The resulting mixture was then stirred for another 10 min at -78 0C. The reaction mixture was quenched by the addition of sat. NH4Cl (100 mL), water (240 mL), and Et2O (240 mL). The organic layer was washed with 10% aqueous sodium sulfite solution (100 mL), dried (Na2SO4) and concentrated in vacuo. The crude material was purified by distilling off unreacted starting material. The residue was further purified by silica gel chromatography (EtO Ac/heptane ) to produce an impure solid material, which was purified by precipitation from EtO Ac/heptane affording 11.46 g of compound 2.
(iϊ/Z)-3-(3,7,8-Trimethoxy-naphthalen-2-yl)-aciylonitrile (compound 3).
Figure imgf000017_0001
compound 2 compound 3
To a suspension of compound 2 (3.41 g) in dry acetonitrile (10.7 mL) in a microwave reactor vial was added acrylonitrile (1.19 mL) Pd(OAc)2 (73 mg), and triethylamine (1.48 mL). The vial was sealed, and the mixture was heated for 40 min at 145 0C under microwave irradiation. This procedure was carried out two more times (using a total of 10.23g of compound 5). The crude reaction mixtures were combined and the catalyst was filtered off, and the filtrate was concentrated in vacuo. The residue was partitioned between Et2O (300 mL) and 2M HCl (150 mL). The organic layer was washed with brine (100 mL), dried (Na2SO4) and concentrated in vacuo. The crude material (7.34 g) was purified by silica gel chromatography (EtO Ac/heptane) to produce 5.23 g of compound 3 as a mixture of olefin isomers.
3-(3,7,8-Trimethoxy-naphthalen-2-yl)-propionitrile (compound 4).
Figure imgf000017_0002
compound 3 compound 4 Compound 3 (5.23 g) was dissolved in CHCl3 (15 mL) and 99% EtOH (100 mL). 10% Pd/C (0.8 g) was added and the solution was hydrogenated for 45 min under a hydrogen pressure of 3 bar using a Parr shaker. The catalyst was filtered off, and the filtrate was passed through a small plough of silica gel (eluent: 99% EtOH). Yield: 4.91 g compound 4 as a white solid. [3-(3,7,8-Trimethoxy-l,4-dihydro-naphthalen-2-yl)-propyl]-carbamic acid *-butyl ester
(compound 5).
Figure imgf000018_0001
Compound 4 (5.Og) was dissolved in 99% EtOH (150 mL) and the mixture was heated to reflux under nitrogen atmosphere. Sodium metal (5g) was added in small lumps over 3h. The mixture was refluxed for an addition 2h, before it was stirred at rt for 2 days. Then it was heated to reflux again, and more sodium metal (3.68 g) was added and the mixture was refluxed overnight. After cooling on an ice/water bath, the reaction was quenched by the addition of solid ammonium chloride (20 g) and water (25 mL). The resulting mixture was filtered, and the filtrate was concentrated in vacuo. The residue was partitioned between diethyl ether (50 mL) and water (50 mL). The aqueous layer was neutralized with 37% HCl and extracted with diethyl ether (2x50 mL). The combined organic extracts were washed with brine (50 mL), dried (MgSO4) and concentrated in vacuo to afford an oil. This material was dissolved in THF (50 mL) and treated with BoC2O (2.34 g) and EtsN (1.78 mL) at rt. After six days the volatiles were removed in vacuo and the residue was purified by silica gel chromatography (EtO Ac/heptane). This provided impure compound 5 (1.52 g).
Racemic 6,7-dimethoxy-2,3,4,4a,5,10-hexahydro-benzo[g]quinoline hydrochloride (compound 6).
Figure imgf000018_0002
Compound 5 (1.52 g from the previous step) was dissolved in MeOH (20 mL). 37% HCl (3.5 mL) was added, and the mixture was refluxed for 4h. The volatiles were removed in vacuo, using toluene to azeotropically remove the water. This provided impure compound 6 (0.89 g) as an yellow oil. Racemic ^rans-6,7-dimethoxy-3,4,4a,5,10,10a-hexahydro-2H-benzo[g]quinoline-l-carboxylic acid *-butyl ester (compound 8).
Figure imgf000019_0001
Compound 6 (0.89 g) was dissolved in MeOH (10 mL) and NaCNBH3 (0.19 g) was added. The reaction was stirred overnight at rt. The crude mixture was cooled on an ice/water bath, before it was quenched with 2 M HCl in Et2O (1 mL). The mixture was partitioned between Et2O (50 mL), water (50 mL), and 2 M NaOH (10 mL). The aqueous layer was extracted with diethyl ether (3x50 mL). The combined organic layers were dried (MgSO4) and concentrated in vacuo to afford the impure free amine (compound 7). This material was dissolved in THF (25 mL) and treated with BoC2O (0.68 g) and Et3N (0.86 mL) at rt for Ih. The crude mixture was concentrated in vacuo, and the residue was purified by silica gel chromatography (EtO Ac/heptane) to provide 1.18g of slightly impure racemic compound 8.
SFC-separation of the enantiomers of racemic *røns-6,7-dimethoxy-3,4,4a,5,10,10a- hexahydro-2H-benzo[g]quinoline-l-carboxylic acid *-butyl ester (compounds 9 and ent-9).
Figure imgf000019_0002
compound 8 compound 9 compound ent-9
(racemate) (4R.10R enantiomer) (4S.10S enantiomer)
Compound 8 (19.7 g) was resolved into its enantiomers using chiral SFC on a Berger SFC multigram II instrument equipped with a Chiralcel OD 21.2 x 250 mm column. Solvent system: CO2ZEtOH (85:15), Method: constant gradient with a flow rate of 50 mL/min. Fraction collection was performed by UV 230 nm detection. Fast eluting enantiomer (4aR, 1OaR enantiomer; compound 9): 9.0 g of a white solid. Slow eluting enantiomer (4aS, lOaS enantiomer; compound ent-9): 8.1 g of a white solid. (4aS,10aS)-6,7-Dimethoxy-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoIine hydrochloride
(compound ent-Ψ).
Figure imgf000020_0001
compound ent-9 compound enf-91 (4S, l OS enantiomer) (4S, 1OS enantiomer)
Compound ent-9 (0.52g) was dissolved in MeOH (15 mL) and treated with 5 M HCl in Et2O (7.5 5 mL) at rt for 2h. . The mixture was concentrated in vacuo and the solid was dried in vacuo to give compound ent-Ψ as a white solid. LC/MS (method 14): RT 1.31 min.
(4aR,10aR)-l-Propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol hydrobromide (compound 10).
Figure imgf000020_0002
compound 9 compound 10
J Q (4aR, 1OaR enantiomer) (4aR, 1OaR enantiomer)
Compound 9 (0.5 g) was dissolved in 99% EtOH (5 mL) and treated with 2M HCl in Et2O (4 mL) overnight at rt. The crude mixture was concentrated in vacuo, and the residue was partitioned between EtOAc and 10% aqueous NaOH (5 mL). The aqueous layer was extracted with EtOAc, and the combined organic layers were washed with brine, dried (MgSO4), concentrated in vacuo.
15 The residue was dissolved in 99% EtOH (5 mL) and treated with propionic aldehyde (0.52 mL), NaCNBHs (0.45 g), and AcOH (3 drops) overnight at rt. The crude mixture was portioned between sat. aqueous NaHCO3 (12.5 mL), water (12.5 mL), and EtOAc (2x25 mL). The combined organic layers were washed with brine, dried (MgSO4), and concentrated in vacuo. The residue was purified by silica gel chromatography (MeOH/EtOAc). The obtained intermediate was treated with 0 48% HBr (3 mL) at 150 0C for Ih under microwave conditions, before the crude mixture was stored at 4 0C overnight. The precipitated material was isolated by filtration and dried in vacuo. Yield of compound 10: 103 mg as a solid. LC/MS (method 25): RT 0.77 min. (4aS,10aS)-l-Propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol hydrobromide
(compound ent-10).
Figure imgf000021_0001
compound ent-9' compound enf-10 (4aS, 1OaS enantiomer) (4aS, 1OaS enantiomer)
The procedure described for compound 10 was followed starting from compound ent-Ψ (0.5 g; the
5 HCl salt was liberated by partitioning between EtOAc and 10% aqueous NaOH before the reductive amination step). Yield of compound ent-10: 70 mg as a solid. LC/MS (method 25): RT
0.70 min. A small sample of compound ent-10 was dissolved in MeOH and allowed to crystallize slowly at rt over 2 months. The formed white crystals were collected and subjected to X-ray analysis (cf. Figure 1). The absolute configuration of compound ent-10 was determined by X-ray
10 crystallography and allowed for unambiguous determination of the stereochemistry of compounds
9 and 10 and hence their derivatives.
(6aR,10aR)-7-n-Propyl-6,6a,7,8,9,10,10a,ll-octahydro-l,3-dioxa-7-aza- cyclopenta [a] anthracene hydrochloride (compound 11).
Figure imgf000021_0002
compound 10 compound 11
\ 2 (4aR 1OaR enantiomer) (6aR 1OaR enantiomer)
Compound 10 (7.80 g), Cs2CO3 (18.6 g), CH2BrCl (2.2 mL), and DMF (180 mL) were heated to 100 0C for Ih under an argon atmosphere. The crude reaction mixture was added to separatory funnel and diluted with ice/water (300 mL). The resulting mixture was extracted with Et2O (3x300 mL). The combined organic layers were washed with brine (200 mL), dried (MgSO4) and 0 concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/MeOH) to afford a pale red solid, which was dissolved in MeOH (25 mL) and precipitated as the hydrochloride salt by addition of 2 M HCl in Et2O (20 mL) and Et2O (100 mL). The precipitated product was isolated by filtration and dried in vacuo. Yield of compound 11: 5.1 g. LC/MS (method 111): RT 0.70 min. ELSD 100%. UV 97.0%. MH+: 274.0. (4aR,10aR)-n-l-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinoIin-6-one
(Compound 12)
The synthesis of Compound 12 can be prepared as described in EP Patent No. 1274411, the contents of which are hereby incorporated by reference. Compound 12 is referred to as (-)- GMC6650 in the above-identified patent.
EXPERIMENTAL SECTION
Example 1: Compounds 11 and 12 convert into the catechol-containing active metabolite of compound 10 upon in-vivo administration.
Figure imgf000022_0001
The active metabolite (i.e. Compound 10) was found to function as a potent agonist at both the Dl and D2 receptors in-vitro. As discussed in greater detail below, the data generated from in-vivo experiments indicate that this active metabolite possesses a superior profile against other dopamine agonists and is on par with the efficacy seen with L-DOPA/apomorphine treatment.
Example 2: Pharmacological Testing of Compound 10
Di cAMP assay
The ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX (3-z-butyl-l-methylxanthine) in PBS (phosphate buffered saline)) and the assay was initiated by addition of 100 micro-L of a mixture of 30 nM A68930 and test compound diluted in G buffer (antagonism) or test compound diluted in G buffer (agonism). The cells were incubated for 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-L S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates were shaken for 10 minutes. 60 micro-1 of the reaction were transferred to cAMP FlashPlates (DuPont NEN) containing 40 micro-L 60 mM Sodium acetate pH 6.2 and 100 micro-L IC mix (50 mM Sodium acetate pH 6.2, 0.1 % sodium azide, 12 mM CaCl2, 1% BSA (bovine serum albumin) and 0.15 micro-Ci/mL 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. Compound 10 was demonstrated to act as a Di agonist in this assay.
D2 cAMP assay
The ability of the compounds to either stimulate or inhibit the D2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
The cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes. 60 micro-L of the reaction were transferred to cAMP FlashPlates (DuPont NEN) containing 40 micro-L 60 mM NaOAc pH 6.2 and 100 micro-L IC mix (50 mM NaOAc pH 6.2, 0.1 % Sodium azide, 12 mM CaCl2, 1% BSA and 0.15 micro-Ci/ml 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. Compound 10 was demonstrated to act as a D2 agonist in this assay.
D5 assay Concentration-dependent stimulation of intracellular Ca2+ release by dopamine in hDs-transfected CHO-Gal6 cells. The cells were loaded with fluoro-4, a calcium indicator dye, for Ih. Calcium response (fluorescence change) was monitored by FLIPR (fluorometric imaging plate reader) for 2.5 min. Peak responses (EC50) were averaged from duplicate wells for each data point and plotted with drug concentrations (cf. Figure 2 for dopamine). Compound 10 was demonstrated to act as a D5 agonist in this assay.
6-OHDA Rat Model
Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both. The rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott, Brain Res., 1970, 24, 485; Setler, et al. Eur. J. Pharmacol, 1978, 50(4), 419; and Ungerstedt, et al. "Advances in Dopamine Research" (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219). 6-OHDA (6- hydroxydopamine) is a neurotoxin used by neurobiologists to selectively kill dopaminergic neurons at the site of injection in the brain in experimental animals. In the 6-OHDA model, the nigrostraital dopamine cells are destroyed on one side of the brain (unilateral) by injecting 6-OHDA into the median forebrain bundle, located in front of the substantia nigra. This unilateral injection combined with stimulation by dopamine agonists such as apomorphine will induce rotation behaviour as only one side of the brain is stimulated. Experiments consist of determining a minimum effective dose (MED) to induce rotation for the compound in question. Once a MED has been determined, a second experiment is performed to determine the MED of the compound to overcome Nemonapride block (MEDNemonapπde). Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor. Finally, once the MEDNemonapπde is known a third experiment is run using the MEDNemonapπde dose and observing the effect of the Dl antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally, the effect of combined treatment with SCH 23390 and Nemonapride. This third experiment confirms the activity of the compound at both receptors as either antagonist alone can only partially inhibit the rotation response induced by the test compound while the combination treatment completely blocks all rotations in the rats [Arnt and Hyttel, Psychopharmacology, 1985, 85(3), 346; and Sonsalla et al., J. Pharmacol Exp. Ther., 1988, 247(1), 180]. This model was validated using apomorphine as the proof-of-principle compound for mixed D1/D2 agonists.
In this model, Compound 10 possess 'apomorphine '-like profiles with a D1/D2 ratio of about 2-4 as compared to a ratio of about 3 for apomorphine. Moreover, the duration of action observed was ca. 18h for the compound which is significantly higher than that seen with L-DOPA / apomorphine. A Dl component could not be observed for D2-agonists as exemplified by pramipexole and rotigotine.
Superiority model
Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 dopamine receptors. Pramipexole, an agonist at D2 receptors is ineffective in this model.
The experiments were performed as follows: Mice previously treated with MPTP (2xl5mg/kg subcutaneously) and that had stable lesions are used and vehicle treated mice served as normal controls. MPTP (l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine) is a neurotoxin that causes permanent symptoms of Parkinson's disease by killing certain neurons in the substantia nigra of the brain. It is used to study the disease in monkeys and mice. On the day of the experiment, mice were treated with AMPT (250mg/kg subcutaneously) and then returned to their home cages for 1.5 hours after which they were placed in individual cages in the motility unit. AMPT (alpha-methyl-p- tyrosine) is a drug that temporarily reduces brain catecholamine activity (in this case especially dopamine levels). Three hours after the AMPT injection, rescue of locomotive deficits is attempted with Compound 10 and activity was recorded for an additional 1.5 hours. The first 30 min of data collected after the rescue treatment was 'contaminated' due to stressing the animals with handling and injection as evidenced by increased levels in the vehicle controls therefore the data were analyzed using the last 1 hour of recorded data. Various dopaminergic compounds were tested for their ability to reverse the motility deficits produced in this model. Both L-DOPA/Benserazide, and apomorphine restored locomotion in the mice in a dose-dependent manner. Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain. In contrast, the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
This model was used to evaluate whether or not Compound 10 exhibits the same superiority as L- DOPA and apomorphine over D2 agonists. A dose response experiment for Compound 10 was performed and there was a dose-dependent trend for reversing the hypomotility deficits induced by severe depletion of endogenous dopamine. A final experiment directly comparing the effects of apomorphine, pramipexole and Compound 10 in this model was performed and confirmed that Compound 10 was able to restore locomotion in MPTP mice treated and was superior to pramipexole.
Induction of dyskinesia model with naϊve 6-OHDA rats Twenty male Sprague Dawley rats with unilateral 6-OHDA lesions were used to test induction of dyskinesia by compound 10 (administered subcutaneously; n=7; group 1) compared to L- DOPA/benserazide (6mg/kg / 15mg/kg subcutaneously; n=7; group 2) and apomorphine (lmg/kg subcutaneously; n=6; group 3). Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain. Three weeks after 6-OHDA surgery, the animals were tested for their rotation response induced by 2.5mg/kg amphetamine, which induces ipsilateral circling (amphetamine increases the level dopamine in the brain via the intact neurons on the unlesioned side causing the animals to rotate in the opposite direction as compared to their response to direct agonists such as L-DOPA and apomorphine that act predominantly on the lesioned side of the brain). All animals included in this study met the criteria of greater than 350 rotations in 60 min. Rats where then randomly allocated to the three treatment groups balancing the groups for the animals' rotation response on amphetamine.
During the actual dyskinesia experiments, rats received once daily injections of the test compounds subcutaneously and were observed for 3h following injection. Each animal was observed for 1 minute every 20 min throughout the 3h period for the presence of dyskinesias using the Abnormal
Involuntary Movement Scale (AIMS) as described previously (Lundblad, et ah, Eur. J Neurosci.,
15, 120, (2002)). Rats received drug for 14 consecutive days and were scored on days 1, 2, 3, 4, 5,
8, 10 and 12. Two-way repeated measures ANOVA revealed that there was a significant treatment effect, time effect and treatment by time interaction (p<0.001, in all cases). Post hoc comparisons using Holm-Sidak method indicates that animals treated with compound 10 had significantly less dykinesia (scores of about 30) compared to animals treated with either L-DOPA or apomorphine
(scores of about 70). There were no differences between L-DOPA and apomorphine treated groups. Following this experiment all rats were given subcutaneous injections of compound 10 from day 15-19 in order to determine how Example I influenced the severity of dyskinesia seen in the apomorphine and L-DOPA groups. Dykinesia scoring was performed on day 19 of the experiment (corresponding to 5 days on compound 10). The data showed a partial reversal of the dyskinesias induced by L-DOPA and apomorphine to about the level of dyskinesias induced by compound 10 (which did not cause an increase in dyskinesia in group 1 as compared to the score of about 30 observed after 12 days of treatment).
Dyskinesia Rat Model
A separate dyskinesia study addressed the reversal of L-DOPA induced dyskinesias with either pramipexole or compound 10. Briefly, 18 animals were treated with L-DOPA/Benserazide (6/15mg/kg subcutaneously) for 7 days. Animals were observed on Days 1, 3 and 5 and AIMS were scored. The day 5 scores were then used to separate the animals into three groups of 6 animals each. Group 1 continued with daily L-DOPA treatment. Group 2 was treated with compound 10 (administered subcutaneously). Group 3 was treated with pramipexole (0.16mg/kg subcutaneously). Treatment continued daily for 10 days and the amount of dyskinesia was scored on days 1, 5, 9 and 10. Two-way repeated measures analysis of variance indicates that animals treated with compound 10 had significantly fewer dyskinesias than both the pramipexole group and the L-DOPA/Benserazide group. The pramipexole group had significantly less dyskinesias than the L-DOPA/Benserazide group. Hence, compound 10 had a superior profile over pramipexole in terms of reversing dyskinesias induced by L-DOPA.
Anti-Parkinsonian effects in MPTP-treated common marmosets The experiments were conducted using 6 MPTP treated marmosets (2.0mg/kg daily for up to 5 consecutive days dissolved in sterile 0.9% saline solution). All the animals had previously been treated with L-DOPA (12.5mg/kg p.o., plus carbidopa 12.5mg/kg p.o.) administered daily for up to 30 days in order to induce dyskinesia. Prior to the study all subjects exhibited stable motor deficits including a marked reduction of basal locomotor activity, poor coordination of movement, abnormal and/or rigid posture, reduced alertness and head checking movements. Domperidone was administered 60 min before any of the test compounds. Domperidone is an anti-dopaminergic drug that suppresses nausea and vomiting. Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity. The assessment of motor disability was performed by a trained observer blinded to the treatment. L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887). The dose chosen for this challenge is at the top of the dose response curve for this drug. Compound 10 (dosed subcutaneously) produced dose-related increases in locomotor activity and reversal of motor disability tending to produce in a response greater than for L-DOPA (12.5mg/kg, p.o.). Both test compounds produced a prolonged reversal of motor disability compared to L-DOPA and were as efficacious as L-DOPA. Compound 10 produced a prolonged reversal of motor disability compared to L-DOPA and was as efficacious as L-DOPA.
Example 3 : Pharmacological Testing of Compound 11
Di cAMP assay
The ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX (3-z-butyl-l-methylxanthine) in PBS (phosphate buffered saline)) and the assay was initiated by addition of 100 micro-L of a mixture of 30 nM A68930 and test compound diluted in G buffer (antagonism) or test compound diluted in G buffer (agonism).
The cells were incubated for 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-L S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates were shaken for 10 minutes. 60 micro-1 of the reaction were transferred to cAMP FlashPlates (DuPont NEN) containing 40 micro-L 60 mM Sodium acetate pH 6.2 and 100 micro-L IC mix (50 mM Sodium acetate pH 6.2, 0.1 % sodium azide, 12 mM CaCl2, 1% BSA (bovine serum albumin) and 0.15 micro-Ci/mL 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. The active metabolite or Compound 10 was found to be a Di agonist in this assay. D2 cAMP assay
The ability of the compounds to either stimulate or inhibit the D2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
The cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes. 60 micro-L of the reaction were transferred to cAMP FlashPlates (DuPont NEN) containing 40 micro-L 60 mM NaOAc pH 6.2 and 100 micro-L IC mix (50 mM NaOAc pH 6.2, 0.1 % Sodium azide, 12 mM CaCl2, 1% BSA and 0.15 micro-Ci/ml 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. The active metabolite or Compound 10 was found to be a D2 agonist in this assay.
D5 assay Concentration-dependent stimulation of intracellular Ca2+ release by dopamine in hDs-transfected CHO-Gal6 cells. The cells were loaded with fluoro-4, a calcium indicator dye, for Ih. Calcium response (fluorescence change) was monitored by FLIPR (fluorometric imaging plate reader) for 2.5 min. Peak responses (EC50) were averaged from duplicate wells for each data point and plotted with drug concentrations. The active metabolite or Compound 10 was found to be a D5 agonist in this assay.
6-OHDA Rat Model
Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both. The rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott, Brain Res. 24, 485 (1970); Setler, et ah, Eur. J. Pharmacol, 50(4), 419 (1978); and Ungerstedt, et al, "Advances in Dopamine Research" (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219). 6- OHDA (6-hydroxydopamine) is a neurotoxin used by neurobiologists to selectively kill dopaminergic neurons at the site of injection in the brain in experimental animals. In the 6-OHDA model the nigrostraital dopamine cells are destroyed on one side of the brain (unilateral) by injecting 6-OHDA into the median forebrain bundle, located in front of the substantia nigra. This unilateral injection combined with stimulation by dopamine agonists such as apomorphine will induce rotation behaviour as only one side of the brain is stimulated. Experiments consist of determining a minimum effective dose (MED) to induce rotation for the compound in question. Once a MED has been determined, a second experiment is performed to determine the MED of the compound to overcome Nemonapride block (MEDNemonaPπde). Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor. Finally, once the MEDNemonapπde is known a third experiment is run using the MEDNemonapπde dose and observing the effect of the Dl antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally, the effect of combined treatment with SCH 23390 and Nemonapride. This third experiment confirms the activity of the compound at both receptors as either antagonist alone can only partially inhibit the rotation response induced by the test compound while the combination treatment completely blocks all rotations in the rats (Arnt and Hyttel; Psychopharmacology, 85(3), 346 (1985); and Sonsalla, et al, J. Pharmacol Exp. Ther., 247(1), 180, (1988)). This model was validated using apomorphine as the proof-of-principle compound for mixed D1/D2 agonists.
In this model, The active metabolite or Compound 10 and Compound 11 possess ' apomorphine '- like profiles with D1/D2 ratios of about 2 as compared to a ratio of about 3 for apomorphine. Moreover, the duration of action observed was ca. 18h for the compound which is significantly higher than that seen with L-DOPA / apomorphine. A Dl component could not be observed for D2-agonists as exemplified by pramipexole and rotigotine.
Superiority model
Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 dopamine receptors. Pramipexole, an agonist at D2-like receptors is ineffective in this model.
The experiments were performed as follows: Mice previously treated with MPTP (2xl5mg/kg subcutaneously) and that had stable lesions are used and vehicle treated mice served as normal controls. MPTP (l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine) is a neurotoxin that causes permanent symptoms of Parkinson's disease by killing certain neurons in the substantia nigra of the brain. It is used to study the disease in monkeys and mice. On the day of the experiment, mice were treated with AMPT (250mg/kg subcutaneously) and then returned to their home cages for 1.5 hours after which they are placed in individual cages in the motility unit. AMPT (alpha-methyl-p- tyrosine) is a drug that temporarily reduces brain catecholamine activity (in this case especially dopamine levels). Three hours after the AMPT injection, rescue of locomotive deficits is attempted with The active metabolite or compound 10 and activity was recorded for an additional 1.5 hours. The first 30 min of data collected after the rescue treatment was 'contaminated' due to stressing the animals with handling and injection as evidenced by increased levels in the vehicle controls therefore the data were analyzed using the last 1 hour of recorded data. Various dopaminergic compounds are tested for their ability to reverse the motility deficits produced in this model. Both L-DOPA/Benserazide, and apomorphine restored locomotion in the mice in a dose-dependent manner. Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it is used to prevent metabolism of L-DOPA to dopamine outside the brain. In contrast, the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
This model was used to evaluate whether or not The active metabolite or compound 10 exhibits the same superiority as L-DOPA and apomorphine over D2 agonists. A dose response experiment for was performed and there was a dose-dependent trend for reversing the hypomotility deficits induced by severe depletion of endogenous dopamine. A final experiment directly comparing the effects of apomorphine, pramipexole and compound 10 was performed. It was confirmed that compound 10 was able to restore locomotion in MPTP mice treated and was superior to pramipexole.
Dyskinesia Rat Model
A rat dyskinesia model reported in the literature (Lundblad, et al., Eur. JNeurosci., 2002, 15, 120) was used to examine the effects of the active metabolite vs. L-DOPA/benserazide with respect to dyskinesias that were assessed as abnormal involuntary movements (AIMs) in 'parkinsonian' rats.
Study Design
Throughout the study animals received L-DOPA/benserazide (6 mg/kg and 15 mg/kg subcutaneous) or the active metabolite (Compound 10) (Group B) once daily at t = -20 min. 0 - 180 min. Animals were scored for dyskinesias. Days 1 - 14: All animals were dosed with L- DOPA/benserazide (group A) or the active metabolite (Compound 10) (Group B).
At days 1, 3, 5, 8 and 12, animals were scored according to AIM-scoring by recording dyskinesias using the Abnormal Involuntary Movement Scale (AIMS) as described previously (Lundblad, et al, Eur. J Neurosci., 2002, 15, 120). Days 15 - 26: Group A animals were treated with the test drug (as group B) instead of L-DOPA/benserazide. Day 15, 16, 17, 19, 22, 24 and 26: Animals scored according AIM-scoring.
Reversal of L-DOP A-induced dyskinesias in 6-OHDA rats
After eight days of treatment, group A animals had dyskinesia scores of 10-12, which remained constant until day 12. In comparison, group B animals had significantly fewer dyskinesias (scores of 2-4). For group B, the degree of dyskinesias did not change during the study. After shifting group A animals from L-dopa/benserazide to the test drug, their level of dyskinesia gradually decreased to the level observed for the other group of animals. Hence, Compound 11 induced significantly less dyskinesia than L-DOPA and was able to reduce the dyskinesias induced by L- DOPA.
Anti-Parkinsonian effects in MPTP-treated common marmosets
The experiments were conducted using 6 MPTP treated marmosets (2.0mg/kg daily for up to 5 consecutive days dissolved in sterile 0.9% saline solution). All the animals had previously been treated with L-DOPA (12.5mg/kg p.o., plus carbidopa 12.5mg/kg p.o.) administered daily for up to 30 days in order to induce dyskinesia. Prior to the study all subjects exhibited stable motor deficits including a marked reduction of basal locomotor activity, poor coordination of movement, abnormal and/or rigid posture, reduced alertness and head checking movements. Domperidone was administered 60 min before any of the test compounds. Domperidone is an antidopaminergic drug that suppresses nausea and vomiting. Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity. The assessment of motor disability was performed by a trained observer blinded to the treatment.
L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887). The dose chosen for this challenge is at the top of the dose response curve for this drug. Compound 11 (dosed p.o.) as well as compound 10 (dosed subcutaneously) produced dose-related increases in locomotor activity and reversal of motor disability tending to produce in a response greater than for L-DOPA (12.5mg/kg, p.o.). Both test compounds produced a prolonged reversal of motor disability compared to L- DOPA and were as efficacious as L-DOPA.
In vitro Hepatocyte Assay
Cryopreserved pooled male rat hepatocytes (Sprague Dawley) and pooled human hepatocytes from 10 donors (male and female) were purchased from In Vitro Technologies Inc., BA, USA. Cells were thawed at 37 0C in a water bath, live cells counted and seeded in a total of 100 micro-L in Dulbecco's modified Eagle medium (high glucose) with 5 mM Hepes buffer in 96 well plates, each well containing 250.000 and 500.000 cells/mL for rat and human hepatocytes, respectively. Incubations were started after 15 min of pre-incubation and stopped at time points of 0, 5, 15, 30 and 60 min for rats and at 0, 30, 60, 90 and 120 min for human hepatocytes. Incubations were stopped by addition of an equal volume of ice-cold acetonitrile containing 10% 1 M HCl. Following centrifugation, 20 micro-L of the supernatants were injected on a HPLC Column Atlantis dC18 3 micro-m, 150 x 2.1 mm i.d. (Waters, MA, USA). The mobile phase had the following composition: A: 5% acetonitrile, 95% H2O, 3.7 ml/125% aq. NH3, 1.8 mL/L formic acid. Mobile phase B: 100% acetonitrile and 0.1% formic acid. The flow rate was 0.3 ml/min. The gradient operated from 0% to 75 % B from 5 min to 20 min and the eluate was analyzed using a Q- TOFmicro mass spectrometer (Waters, MA, USA). Formation of the product/metabolite was confirmed by accurate mass measurements and comparison with a synthesized standard giving coinciding retention times. In this assay, the metabolism of Compound 11 to Compound 10 was demonstrated.
Example 4: Pharmacological Testing of Compound 12 Di cAMP assay
The ability of the compounds to either stimulate or inhibit the Di receptor mediated cAMP formation in CHO cells stably expressing the human recombinant Di receptor was measured as follows. Cells were seeded in 96-well plates at a concentration of 11000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX (3-z-butyl-l-methylxanthine) in PBS (phosphate buffered saline)) and the assay was initiated by addition of 100 micro-L of a mixture of 30 nM A68930 and test compound diluted in G buffer (antagonism) or test compound diluted in G buffer (agonism).
The cells were incubated for 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-L S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates were shaken for 10 minutes. 60 micro-1 of the reaction were transferred to cAMP FlashPlates (DuPont NEN) containing 40 micro-L 60 mM Sodium acetate pH 6.2 and 100 micro-L IC mix (50 mM Sodium acetate pH 6.2, 0.1 % sodium azide, 12 mM CaCl2, 1% BSA (bovine serum albumin) and 0.15 micro-Ci/mL 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. The active metabolite (i.e. Compound 10) was found to be a Di agonist in this assay.
D2 cAMP assay
The ability of the compounds to either stimulate or inhibit the D2 receptor mediated inhibition of cAMP formation in CHO cells transfected with the human D2 receptor was measure as follows. Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3 days prior to the experiment. On the day of the experiment the cells were washed once in preheated G buffer (1 mM MgCl2, 0.9 mM CaCl2, 1 mM IBMX in PBS) and the assay was initiated by addition of 100 micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test compound in G buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer (agonism).
The cells were incubated 20 minutes at 37 °C and the reaction was stopped by the addition of 100 micro-1 S buffer (0.1 M HCl and 0.1 mM CaCl2) and the plates were placed at 4 °C for Ih. 68 micro-L N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the plates were shaken for 10 minutes. 60 micro-L of the reaction were transferred to cAMP FlashPlates (DuPont
NEN) containing 40 micro-L 60 mM NaOAc pH 6.2 and 100 micro-L IC mix (50 mM NaOAc pH
6.2, 0.1 % Sodium azide, 12 mM CaCl2, 1% BSA and 0.15 micro-Ci/ml 125I-cAMP) were added. Following an 18h incubation at 4 °C the plates were washed once and counted in a Wallac TriLux counter. The active metabolite (i.e. Compound 10) was found to be a D2 agonist in this assay.
D5 assay
Concentration-dependent stimulation of intracellular Ca2+ release by dopamine in hLVtransfected CHO-Gal6 cells. The cells were loaded with fluoro-4, a calcium indicator dye, for Ih. Calcium response (fluorescence change) was monitored by FLIPR (fluorometric imaging plate reader) for 2.5 min. Peak responses (EC50) were averaged from duplicate wells for each data point and plotted with drug concentrations (cf. Figure 1 for dopamine). The active metabolite (i.e. Compound 10) was found to be a D5 agonist in this assay.
6-OHDA Rat Model
Dopamine agonists can have activity at either the Dl receptors, the D2 receptors, or both. The rotation response in rats with unilateral 6-OHDA lesions can be used to assess compounds for their ability to stimulate both receptor types and induce rotation (Ungerstedt and Arbuthnott; Brain Res., 24, 485 (1970); Setler, et ah, Eur. J. Pharmacol, 50(4), 419 (1978); and Ungerstedt, et α/./'Advances in Dopamine Research" (Kohsaka, Ed.), Pergamon Press, 1982, Oxford, p. 219). 6- OHDA (6-hydroxydopamine) is a neurotoxin used by neurobiologists to selectively kill dopaminergic neurons at the site of injection in the brain in experimental animals. In the 6-OHDA model the nigrostraital dopamine cells are destroyed on one side of the brain (unilateral) by injecting 6-OHDA into the median forebrain bundle, located in front of the substantia nigra. This unilateral injection combined with stimulation by dopamine agonists such as apomorphine will induce rotation behaviour as only one side of the brain is stimulated. Experiments consist of determining a minimum effective dose (MED) to induce rotation for the compound in question. Once a MED has been determined, a second experiment is performed to determine the MED of the compound to overcome Nemonapride block (MEDNemonapπde). Nemonapride is a D2 antagonist that blocks the D2 receptor, therefore any observed rotations would be dependent upon activity at the Dl receptor. Finally, once the MEDNemonapπde is known a third experiment is run using the MEDNemonapπde dose and observing the effect of the Dl antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally, the effect of combined treatment with SCH 23390 and Nemonapride. This third experiment confirms the activity of the compound at both receptors as either antagonist alone can only partially inhibit the rotation response induced by the test compound while the combination treatment completely blocks all rotations in the rats [Arnt and Hyttel, Psychopharmacology, 1985, 85(3), 346; and Sonsalla et al, J. Pharmacol Exp. Ther., 1988, 247(1), 180]. This model was validated using apomorphine as the proof-of-principle compound for mixed D1/D2 agonists.
In this model, Compounds 10 and 12 possess 'apomorphine'-like profiles with Dl / D2 ratios of about 2-4 as compared to a ratio of about 3 for apomorphine. Moreover, the duration of action observed was ca. 18h for the compound which is significantly higher than that seen with L-DOPA / apomorphine. A Dl component could not be observed for D2-agonists as exemplified by pramipexole and rotigotine.
Superiority model
Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model of severe dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 receptors. Pramipexole, an agonist at D2 receptors is ineffective in this model.
The experiments were performed as follows: Mice previously treated with MPTP (2xl5mg/kg subcutaneously) and that had stable lesions are used and vehicle treated mice served as normal controls. MPTP (l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine) is a neurotoxin that causes permanent symptoms of Parkinson's disease by killing certain neurons in the substantia nigra of the brain. It is used to study the disease in monkeys and mice. On the day of the experiment, mice were treated with AMPT (250mg/kg subcutaneously) and then returned to their home cages for 1.5 hours after which they were placed in individual cages in the motility unit. AMPT (alpha-methyl-p- tyrosine) is a drug that temporarily reduces brain catecholamine activity (in this case especially dopamine levels). Three hours after the AMPT injection, rescue of locomotive deficits was attempted with Compound 10 and activity was recorded for an additional 1.5 hours. The first 30 min of data collected after the rescue treatment was 'contaminated' due to stressing the animals with handling and injection as evidenced by increased levels in the vehicle controls therefore the data were analyzed using the last 1 hour of recorded data. Various dopaminergic compounds were tested for their ability to reverse the motility deficits produced in this model. Both L- DOPA/Benserazide, and apomorphine restored locomotion in the mice in a dose-dependent manner. Benserazide is a DOPA decarboxylase inhibitor which is unable to cross the blood-brain barrier; it was used to prevent metabolism of L-DOPA to dopamine outside the brain. In contrast, the D2 agonists, pramipexole and bromocriptine did not restore the locomotion in the mice.
This model was used to evaluate whether or not Compound 10 exhibits the same superiority as L-
DOPA and apomorphine over D2 agonists. A dose response experiment for was performed and there was a dose-dependent trend for reversing the hypomotility deficits induced by severe depletion of endogenous dopamine. A final experiment directly comparing the effects of apomorphine, pramipexole and Compound 10 was performed. It was confirmed that Compound 10 was able to restore locomotion in MPTP mice treated and was superior to pramipexole.
Dyskinesia Rat Model
A rat dyskinesia model reported in the literature (Lundblad, et ah, Eur. J Neurosci., 2002, 15, 120) was used to examine the effects of Compound 12 vs. L-DOPA/benserazide with respect to dyskinesias that were assessed as abnormal involuntary movements (AIMs) in 'parkinsonian' rats.
Study Design
Throughout the study animals received L-DOPA/benserazide (6 mg/kg and 15 mg/kg subcutaneous) or Compound 12 (group B) once daily at t = -20 min. 0 - 180 min. Animals were scored for dyskinesias. Days 1 - 14: All animals were dosed with L-DOPA/benserazide (group A) or Compound 12 (group B).
At days 1, 3, 5, 8 and 12, animals were scored according to AIM-scoring by recording dyskinesias using the Abnormal Involuntary Movement Scale (AIMS) as described previously. Days 15 - 26: Group A animals were treated with Compound 12 (as group B) instead of L-DOPA/benserazide. Day 15, 16, 17, 19, 22, 24 and 26: Animals scored according AIM-scoring. Results
After eight days of treatment, group A animals had dyskinesia scores of 70-80, which remained constant until day 15. In comparison, group B animals had significantly fewer dyskinesias (scores of 10-25). For group B, the degree of dyskinesias did not change during the study. After shifting group A animals from L-DOPA/benserazide to compound 12 for 10 days, their level of dyskinesia gradually decreased to scores of 30-35. Hence, compound 12 induced significantly less dyskinesia than L-DOPA and was able to reduce the dyskinesias induced by L-DOPA.
Anti-Parkinsonian effects in MPTP-treated common marmosets The experiments were conducted using 6 MPTP treated marmosets (2.0mg/kg daily for up to 5 consecutive days dissolved in sterile 0.9% saline solution). All the animals had previously been treated with L-DOPA (12.5mg/kg p.o., plus carbidopa 12.5mg/kg p.o.) administered daily for up to 30 days in order to induce dyskinesia. Prior to the study all subjects exhibited stable motor deficits including a marked reduction of basal locomotor activity, poor coordination of movement, abnormal and/or rigid posture, reduced alertness and head checking movements. Domperidone was administered 60 min before any of the test compounds. Locomotor Activity was assessed using test cages that are comprised of 8 photo-electric switches comprised of 8 infra-red beams which are strategically placed in the cage and interruption of a beam is recorded as one count. The total number of beam counts per time segment is then plotted as time course or displayed as area under the curve (AUC) for total activity. The assessment of motor disability was performed by a trained observer blinded to the treatment.
L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor disability as previously described (Smith, et al. Mov. Disord. 2002, 17(5), 887). The dose chosen for this challenge is at the top of the dose response curve for this drug. Compound 12 (dosed p.o.) as well as Compound 10 (dosed p.o.) produced dose-related increases in locomotor activity and reversal of motor disability tending to produce in a response greater than for L-DOPA (12.5mg/kg, p.o.). Both test compounds produced a prolonged reversal of motor disability compared to L-DOPA and were as efficacious as L-DOPA.

Claims

1. Use of (4aR,10aR)-l-/?-propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
2. Use of (4aR,10aR)-l-/?-propyl-l,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
3. Use of (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza- cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
4. Use of (6aR,10aR)-7-n-propyl-6,6a,7,8,9,10,10a,l l-octahydro-l,3-dioxa-7-aza- cyclopenta[a] anthracene or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
5. Use of (4aR,10aR)-l-n-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6- one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating Parkinson's disease while maintaining a low dyskinesia induction profile.
6. Use of (4aR,10aR)-l-n-propyl-2,3,4,4a,5,7,8,9,10,10a-decahydro-lH-benzo[g]quinolin-6- one, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for reversing dyskinesias.
7. The use of anyone of claims 1-6, wherein the dyskinesia is associated with a basal ganglia- related movement disorder.
8. The use of claim 7, wherein the dyskinesia is associated with Parkinson's disease.
9. The use of claim 8, wherein the dyskinesia is associated with idiopathic Parkinson's disease or post-encephalitic parkinsonism.
10. The use of claim 9, wherein the dyskinesia is associated with off-dystonia in Parkinson's disease.
11. The use of claim 10, wherein the dyskinesia arises as a side-effect of a therapeutic agent to treat Parkinson's disease.
12. The use of claim 11, wherein the dyskinesia is associated with dopamine replacement therapy.
13. The use of claim 12, wherein the dopamine replacement therapy agent is selected from the group consisting of rotigotine, ropinirole, pramipexole, cabergoline, bromocriptine, lisuride, pergolide, L-DOPA and apomorphine.
14. The use of claim 13, wherein the dyskinesia is established as a result of repeated administration of L-DOPA.
PCT/DK2010/050051 2007-08-31 2010-02-26 Treatment of dyskinesia related disorders WO2010097092A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
US13/202,768 US20120108624A1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
AU2010217059A AU2010217059B2 (en) 2007-08-31 2010-02-26 Treatment of dyskinesia related disorders
CA2751322A CA2751322C (en) 2009-02-27 2010-02-26 Catecholamine derivatives and prodrugs thereof for the treatment of dyskinesia related disorders
JP2011551409A JP5738775B2 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia-related disorders
MX2011008944A MX340138B (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders.
EP10707436.1A EP2400967B1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
SG2011061785A SG174163A1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
BRPI1007017A BRPI1007017A2 (en) 2009-02-27 2010-02-26 use of a compound.
KR1020117019748A KR101700978B1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
CN2010800098743A CN102333531B (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
EP19191241.9A EP3653210A1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
NZ594264A NZ594264A (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
EA201171088A EA023778B1 (en) 2009-02-27 2010-02-26 Treatment of dyskinesia related disorders
IL213502A IL213502A (en) 2009-02-27 2011-06-12 Use of d1/d2 agonist compounds in the preparation of a medicament
ZA2011/06294A ZA201106294B (en) 2009-02-27 2011-08-26 Treatment of dyskinesia related disorders
HK12107220.3A HK1166472A1 (en) 2009-02-27 2012-07-23 Treatment of dyskinesia related disorders

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US15595309P 2009-02-27 2009-02-27
US15596609P 2009-02-27 2009-02-27
US15594309P 2009-02-27 2009-02-27
DKPA200900281 2009-02-27
US61/155,966 2009-02-27
DKPA200900273 2009-02-27
DKPA200900280 2009-02-27
DKPA200900280 2009-02-27
DKPA200900273 2009-02-27
DKPA200900281 2009-02-27
US61/155,953 2009-02-27
US61/155,943 2009-02-27

Publications (1)

Publication Number Publication Date
WO2010097092A1 true WO2010097092A1 (en) 2010-09-02

Family

ID=42060693

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2010/050051 WO2010097092A1 (en) 2007-08-31 2010-02-26 Treatment of dyskinesia related disorders

Country Status (20)

Country Link
US (1) US20120108624A1 (en)
EP (2) EP2400967B1 (en)
JP (1) JP5738775B2 (en)
KR (1) KR101700978B1 (en)
CN (1) CN102333531B (en)
AR (1) AR075625A1 (en)
AU (1) AU2010217059B2 (en)
BR (1) BRPI1007017A2 (en)
CA (1) CA2751322C (en)
CL (1) CL2011002102A1 (en)
CO (1) CO6410297A2 (en)
EA (1) EA023778B1 (en)
HK (1) HK1166472A1 (en)
IL (1) IL213502A (en)
MX (1) MX340138B (en)
NZ (1) NZ594264A (en)
SG (1) SG174163A1 (en)
TW (1) TW201036949A (en)
WO (1) WO2010097092A1 (en)
ZA (1) ZA201106294B (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190160083A1 (en) * 2017-11-24 2019-05-30 H. Lundbeck A/S Catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020070099A1 (en) 2018-10-02 2020-04-09 H. Lundbeck A/S Administration of catecholamine prodrugs in combination with a 5-ht2b antagonist
WO2020234277A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S Catecholamine carbamate prodrugs for use in the treatment of parkinson s disease
WO2020234272A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S New solid forms of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
WO2020234270A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacture of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid and intermediate thereof
WO2020234274A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020234273A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacturing of (6ar,10ar)-7-propyl-6,6a,7,8,9,10,10a,11-octahydro-[1,3]dioxolo[4',5':5,6]benzo[1,2-g]quinoline and (4ar,10ar)-1-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol
WO2020234276A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020234271A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacture of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
WO2020234275A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's diseases
WO2022106352A1 (en) 2020-11-17 2022-05-27 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
RU2806075C2 (en) * 2017-11-24 2023-10-25 Х. Лундбекк А/С (H. Lundbeck A/S) New catecholamine-based prodrugs for use in treatment of parkinson's disease
WO2023208867A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL 1,2,3,4,4a,5,8,9,10,10a-DECAHYDROBENZO[G]QUINOLIN-6(7H)-ONE COMPOUNDS AND USES THEREOF
WO2023208865A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL 1,2,3,4,4a,5,6,7,8,9,10,10a-DODECAHYDROBENZO[G]QUINOLIN-6-OL COMPOUNDS AND USES THEREOF
WO2023208869A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL ESTERS OF 1,2,3,4,4a,5,6,7,8,9,10,10a-DODECAHYDROBENZO[G]QUINOLIN-6-OL COMPOUNDS AND USES THEREOF

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001078713A1 (en) * 2000-04-18 2001-10-25 Axon Biochemicals B.V. Phenylethylamines and condensed rings variants as prodrugs of catecholamines, and their use
WO2009026934A1 (en) * 2007-08-31 2009-03-05 H. Lundbeck A/S Catecholamine derivatives useful for the treatment of parkinson' s disease

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001078713A1 (en) * 2000-04-18 2001-10-25 Axon Biochemicals B.V. Phenylethylamines and condensed rings variants as prodrugs of catecholamines, and their use
EP1274411A1 (en) 2000-04-18 2003-01-15 Axon Biochemicals B.V. Phenylethylamines and condensed rings variants as prodrugs of catecholamines, and their use
WO2009026934A1 (en) * 2007-08-31 2009-03-05 H. Lundbeck A/S Catecholamine derivatives useful for the treatment of parkinson' s disease

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
ARNT; HYTTEL, PSYCHOPHANNACOLOGY, vol. 85, no. 3, 1985, pages 346
HILARY ET AL., JOURNAL OFNEUROLOGY, vol. 251, no. 11, 2004, pages 1370 - 1374
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2 - 19
LUNDBLAD ET AL., EUR. J NEUROSCI., vol. 15, 2002, pages 120
LUNDBLAD ET AL., EUR. J NEUROSCI., vol. 15, 2002, pages 120 - 132
LUNDBLAD ET AL., EUR. J NEUROSCI., vol. I5, 2002, pages 120
OBESO ET AL., NEUROLOGY, vol. 55, 2000, pages 13 - 23
SCHWARTING; HUSTON, PROG. NEUROBIOL, vol. 50, 1996, pages 275 - 33 1
SETLER ET AL., EUR. J. PHANNACOL., vol. 50, no. 4, 1978, pages 419
SETLER ET AL., EUR. J. PHARMACOL., vol. 50, no. 4, 1978, pages 419
SMITH ET AL., MOV. DISORD., vol. 17, no. 5, 2002, pages 887
SONSALLA ET AL., J PHARMACOL EXP. THER., vol. 247, no. 1, 1988, pages 180
SONSALLA ET AL., J. PHANNACOL EXP. THER., vol. 247, no. 1, 1988, pages 180
STOCCHI FABRIZIO ET AL: "Continuous dopaminergic stimulation in early and advanced Parkinson's disease.", NEUROLOGY, vol. 62, no. 1 Supplement 1, 13 January 2004 (2004-01-13), pages S56 - S63, XP002576738, ISSN: 0028-3878 *
STOCCHI; OLANOW, NEUROLOGY 2004, vol. 62, 2004, pages S56 - S63
TABER ET AL., J. AM. CHEM. SOC., vol. 124, no. 42, 2002, pages 12416
UNGERSTEDT ET AL.: "Advances in Dopamine Research", 1982, PERGAMON PRESS, pages: 219
UNGERSTEDT; ABURTHNOTT, BRAIN RES., vol. 24, 1970, pages 485
UNGERSTEDT; ARBUTHNOTT, BRAIN RES., vol. 24, 1970, pages 485

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11110110B2 (en) 2017-11-24 2021-09-07 H. Lundbeck A/S Catecholamine prodrugs for use in the treatment of Parkinson's disease
RU2806075C2 (en) * 2017-11-24 2023-10-25 Х. Лундбекк А/С (H. Lundbeck A/S) New catecholamine-based prodrugs for use in treatment of parkinson's disease
US10729710B2 (en) 2017-11-24 2020-08-04 H. Lundbeck A/S Catecholamine prodrugs for use in the treatment of Parkinson's disease
TWI816716B (en) * 2017-11-24 2023-10-01 丹麥商H 朗德貝克公司 New catecholamine prodrugs for use in the treatment of parkinson's disease
US11707476B2 (en) 2017-11-24 2023-07-25 H. Lundbeck A/S Catecholamine prodrugs for use in the treatment of parkinson's disease
AU2018371193B2 (en) * 2017-11-24 2022-12-22 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of Parkinson's disease
IL274648B1 (en) * 2017-11-24 2024-06-01 H Lundbeck As New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2019101917A1 (en) 2017-11-24 2019-05-31 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
US20190160083A1 (en) * 2017-11-24 2019-05-30 H. Lundbeck A/S Catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020070099A1 (en) 2018-10-02 2020-04-09 H. Lundbeck A/S Administration of catecholamine prodrugs in combination with a 5-ht2b antagonist
WO2020234273A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacturing of (6ar,10ar)-7-propyl-6,6a,7,8,9,10,10a,11-octahydro-[1,3]dioxolo[4',5':5,6]benzo[1,2-g]quinoline and (4ar,10ar)-1-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol
JP2022533050A (en) * 2019-05-20 2022-07-21 ハー・ルンドベック・アクチエゼルスカベット (2S, 3S, 4S, 5R, 6S) -3,4,5-trihydroxy-6-(((4aR, 10aR) -7-hydroxy-1-propyl-1,2,3,4,4a, 5) , 10,10a-Octahydrobenzo [g] quinoline-6-yl) oxy) A new solid form of tetrahydro-2H-pyran-2-carboxylic acid
US11104697B2 (en) 2019-05-20 2021-08-31 H. Lundbeck A/S Process for the manufacture of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4AR,10AR)-7-hydroxy-1- propyl-1,2,3,4,4A,5,10,10A-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
US11111263B2 (en) 2019-05-20 2021-09-07 H. Lundbeck A/S Process for the manufacture of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
WO2020234271A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacture of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
JP7509803B2 (en) 2019-05-20 2024-07-02 ハー・ルンドベック・アクチエゼルスカベット Method for preparing (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
RU2819924C2 (en) * 2019-05-20 2024-05-28 Х. Лундбекк А/С (H. Lundbeck A/S) Method of producing (6ar,10ar)-7-propyl-6,6a,7,8,9,10,10a,11-octahydro-[1,3]dioxolo[4',5':5,6]benzo[1,2-g]quinoline and (4ar,10ar)-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinoline-6,7-diol
RU2819919C2 (en) * 2019-05-20 2024-05-28 Х. Лундбекк А/С Method of producing (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
US20220185839A1 (en) 2019-05-20 2022-06-16 H. Lundbeck A/S Process for the manufacture of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
US11168056B2 (en) 2019-05-20 2021-11-09 H. Lundbeck A/S Process for the manufacturing of (6aR,10aR)-7-propyl-6,6a,7,8,9,10,10a,11-octahydro-[1,3]dioxolo[4′,5′:5,6]benzo[1,2-G]quinoline and (4aR,10aR)-1-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[G]quinoline-6,7-diol
WO2020234270A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S A process for the manufacture of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid and intermediate thereof
WO2020234272A1 (en) 2019-05-20 2020-11-26 H. Lundbeck A/S New solid forms of (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid
US11130775B2 (en) 2019-05-20 2021-09-28 H. Lundbeck A/S Solid forms of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4A,5,10,10A-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
JP7364691B2 (en) 2019-05-20 2023-10-18 ハー・ルンドベック・アクチエゼルスカベット (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5 A new solid form of ,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
US11851456B2 (en) 2019-05-20 2023-12-26 H. Lundbeck A/S Process for the manufacture of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4aR,10aR)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
RU2817700C2 (en) * 2019-05-20 2024-04-18 Х. Лундбекк А/С Method of producing (2s,3s,4s,5r,6s)-3,4,5-trihydroxy-6-(((4ar,10ar)-7-hydroxy-1-propyl-1,2,3,4,4a,5,10,10a-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2h-pyran-2-carboxylic acid and intermediate compounds for its production
US11866410B2 (en) 2019-05-20 2024-01-09 H. Lundbeck A/S Process for the manufacturing of (6AR,10AR)-7-propyl-6,6A,7,8,9,10,10A,11-octahydro-[1,3]dioxolo[4′,5′:5,6]benzo[1,2-G]quinoline and (4AR, 10AR)-1-propyl-1,2,3,4,4A,5,10,10A-octahydro-benzo[G]quinoline-6,7-diol
US11858954B2 (en) 2019-05-20 2024-01-02 H. Lundbeck A/S Solid forms of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4AR,10AR)-7-hydroxy-1-propyl-1,2,3,4,4A,5,10,10A-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
US11827665B2 (en) 2019-05-20 2023-11-28 H. Lundbeck A/S Process for the manufacture of (2S,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(((4AR,10AR)-7-hydroxy-1-propyl-1,2,3,4,4A,5,10,10A-octahydrobenzo[g]quinolin-6-yl)oxy)tetrahydro-2H-pyran-2-carboxylic acid
WO2020234276A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020234275A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's diseases
WO2020234274A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2020234277A1 (en) 2019-05-21 2020-11-26 H. Lundbeck A/S Catecholamine carbamate prodrugs for use in the treatment of parkinson s disease
JP7514859B2 (en) 2019-05-21 2024-07-11 ハー・ルンドベック・アクチエゼルスカベット Novel catecholamine prodrugs for use in the treatment of Parkinson's disease
WO2022106352A1 (en) 2020-11-17 2022-05-27 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's disease
WO2023208869A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL ESTERS OF 1,2,3,4,4a,5,6,7,8,9,10,10a-DODECAHYDROBENZO[G]QUINOLIN-6-OL COMPOUNDS AND USES THEREOF
WO2023208865A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL 1,2,3,4,4a,5,6,7,8,9,10,10a-DODECAHYDROBENZO[G]QUINOLIN-6-OL COMPOUNDS AND USES THEREOF
WO2023208867A1 (en) 2022-04-25 2023-11-02 Integrative Research Laboratories Sweden Ab NOVEL 1,2,3,4,4a,5,8,9,10,10a-DECAHYDROBENZO[G]QUINOLIN-6(7H)-ONE COMPOUNDS AND USES THEREOF

Also Published As

Publication number Publication date
EP2400967B1 (en) 2019-08-14
JP2012519157A (en) 2012-08-23
KR101700978B1 (en) 2017-01-31
IL213502A (en) 2017-02-28
CN102333531B (en) 2013-11-06
CA2751322A1 (en) 2010-09-02
CN102333531A (en) 2012-01-25
CL2011002102A1 (en) 2012-07-20
CO6410297A2 (en) 2012-03-30
JP5738775B2 (en) 2015-06-24
CA2751322C (en) 2017-10-31
AU2010217059B2 (en) 2012-08-30
AU2010217059A1 (en) 2011-08-11
US20120108624A1 (en) 2012-05-03
EA201171088A1 (en) 2012-02-28
HK1166472A1 (en) 2012-11-02
EP2400967A1 (en) 2012-01-04
TW201036949A (en) 2010-10-16
IL213502A0 (en) 2011-07-31
NZ594264A (en) 2013-11-29
BRPI1007017A2 (en) 2016-03-29
EP3653210A1 (en) 2020-05-20
MX2011008944A (en) 2011-09-15
AR075625A1 (en) 2011-04-20
ZA201106294B (en) 2012-10-31
EA023778B1 (en) 2016-07-29
MX340138B (en) 2016-06-28
SG174163A1 (en) 2011-10-28
KR20110132340A (en) 2011-12-07

Similar Documents

Publication Publication Date Title
EP2400967B1 (en) Treatment of dyskinesia related disorders
KR100412154B1 (en) Use of r-enantiomer of n-propargyl-1-aminoindan,salts,and compositions thereof
NO326878B1 (en) Use of morpholinol
TW200914426A (en) Catecholamine derivatives and prodrugs thereof
US20180071230A1 (en) Formulations, salts and polymorphs of transnorsertraline and uses thereof
JP2780834B2 (en) Methods and compositions for treating Parkinson&#39;s syndrome in mammals
US11078153B2 (en) 2-difluoro substituted 4-aminocyclopentanecarboxylic acids as inhibitors of gamma-aminobutyric acid aminotransferase and human ornithine aminotransferase
Sit Dopamine Agonists in the Treatment of Parkinsons Disease-Past, Present and Future
Van der Weide et al. The enantiomers of the D-2 dopamine receptor agonist N-0437 discriminate between pre-and postsynaptic dopamine receptors
NO885166L (en) Ergoline derivatives.
US20060189612A1 (en) Pharmaceutically active morpholinol
US10081600B2 (en) 2-alkoxy-11-hydroxyaporphine derivatives and uses thereof
Micheli et al. Beneficial effects of lisuride in Meige disease
CN105384676A (en) triple reuptake inhibitors and methods of their use
JP3926867B2 (en) Use of (α-aminomethyl-3,4-dichlorobenzyl) thioacetamide derivatives for inhibiting dopamine reuptake and novel compounds for this use
Van Oene et al. trans-Nn-propyl-7-hydroxy-octahydrobenzo (f) quinoline, a rigid 3-PPP analogue with greater potency but lack of selectivity for dopamine autoreceptors
CZ20002690A3 (en) Pharmaceutically acceptable salts and solvates of (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol and morpholinol pharmaceutical preparation and pharmaceutical preparation
KR20040023653A (en) Pharmaceutically Active Morpholinol

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080009874.3

Country of ref document: CN

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

Ref document number: 10707436

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12011501238

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2010707436

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010217059

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 594264

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2751322

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2010217059

Country of ref document: AU

Date of ref document: 20100226

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117019748

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/008944

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2011002102

Country of ref document: CL

Ref document number: 2011551409

Country of ref document: JP

Ref document number: 11109225

Country of ref document: CO

Ref document number: 6129/CHENP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: a201111022

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 201171088

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 13202768

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1007017

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI1007017

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110726