US20150374711A1 - 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof - Google Patents

2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof Download PDF

Info

Publication number
US20150374711A1
US20150374711A1 US14/847,089 US201514847089A US2015374711A1 US 20150374711 A1 US20150374711 A1 US 20150374711A1 US 201514847089 A US201514847089 A US 201514847089A US 2015374711 A1 US2015374711 A1 US 2015374711A1
Authority
US
United States
Prior art keywords
group
abcc1
amyloidopathy
transporter
halogen atom
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/847,089
Inventor
Jens Pahnke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immungenetics AG
Original Assignee
Immungenetics AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immungenetics AG filed Critical Immungenetics AG
Priority to US14/847,089 priority Critical patent/US20150374711A1/en
Publication of US20150374711A1 publication Critical patent/US20150374711A1/en
Assigned to IMMUNGENETICS AG reassignment IMMUNGENETICS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PAHNKE, JENS
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/141,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems
    • C07D279/18[b, e]-condensed with two six-membered rings
    • C07D279/22[b, e]-condensed with two six-membered rings with carbon atoms directly attached to the ring nitrogen atom
    • C07D279/24[b, e]-condensed with two six-membered rings with carbon atoms directly attached to the ring nitrogen atom with hydrocarbon radicals, substituted by amino radicals, attached to the ring nitrogen atom
    • C07D279/28[b, e]-condensed with two six-membered rings with carbon atoms directly attached to the ring nitrogen atom with hydrocarbon radicals, substituted by amino radicals, attached to the ring nitrogen atom with other substituents attached to the ring system
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/141,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems
    • C07D279/18[b, e]-condensed with two six-membered rings
    • C07D279/22[b, e]-condensed with two six-membered rings with carbon atoms directly attached to the ring nitrogen atom
    • C07D279/24[b, e]-condensed with two six-membered rings with carbon atoms directly attached to the ring nitrogen atom with hydrocarbon radicals, substituted by amino radicals, attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4709Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2835Movement disorders, e.g. Parkinson, Huntington, Tourette

Definitions

  • AD Alzheimer's dementia
  • CAA cerebral ⁇ -amyloidopathy
  • a ⁇ proteostasis i.e. the equilibrium of production and degradation/removal by means of receptors or proteases is disturbed in AD and CAA.
  • ABSC transporters cellular transporters
  • Parkinson's disease the protein ⁇ -synuclein accumulates, which inter alia regulates the dopamine release in the substantia nigra.
  • ⁇ -synucleinopathy it is known that ABC transporters play a crucial role for transport (Kortekaas et al., Ann Neural 2005, 57, 176-179).
  • A-G which can alternatingly transport various substrates (metabolites, medicaments, peptides, proteins, ions) and are even able to replace each other in the transport function (e.g. ABCB1 and ABCC1, Tao et al. Cancer Chemotherapy and Pharmacology, 64, 5, 961-969).
  • ABC transporter a common structural element of the ABC transporter is an ATP-binding cassette and a transport pore
  • ABCC1 is an important protein/peptide transporter, in particular A ⁇ transporter, which has extraordinary functional effects on the cerebral protein accumulation.
  • ABCC1 is also an important ⁇ -synuclein transporter.
  • the invention provides substances which suitably influence the ABCC1 transporter to thus be able to treat neurodegenerative diseases, in particular ⁇ -amyloidopathiesor ⁇ -synucleopathies.
  • the invention provides 2-(R 2 -thio)-10-[3-(4-R 1 -piperazin-1-yl)propyl]-10H-phenothiazines.
  • R 1 and R 2 are the same or different and each independently of one another are C 1 -C 6 alkyl groups, which independently of one another optionally comprise another substituent selected from alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio groups and halogen atoms, wherein the respective alkyl groups optionally comprise at least one further halogen atom and the residue
  • R 3 is located at one of the positions 6-9 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio or alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR 4 R 5 or OR 6 group, wherein R 4 , R 5 and R 6 are the same or different and each independently of one another are selected from hydrogen and C 1 -C 3 alkyl groups and the residue
  • R 7 is located at one of the positions 1, 2 or 4 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio or alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR 8 R 9 or OR 10 group, wherein R 8 , R 9 and R 10 are the same or different and each independently of one another are selected from hydrogen and C 1 -C 3 alkyl group, for treating a ⁇ -amyloidopathy or an ⁇ -synucleinopathy accompanied by a cerebral protein deposit.
  • the invention also provides a method with which ⁇ -synucleinopathies and also ⁇ -amyloidopathies can be diagnosed or prediagnosed. Specifically, the invention provides a method for the diagnosis or prediagnosis of a ⁇ -amyloidopathy or ⁇ -synucleopathy or for determining the risk of a proband to develop such an illness, wherein the proband already takes substances transported by the cerebral ABCC1 transporter, including the following steps:
  • step b) repeating the determination of step a) at at least one further later time point;
  • step c) comparing the quantities determined in step a) and b) with quantities which had been defined as characteristic at the same time points for probands who at the time of the sampling showed no clinical symptoms of a ⁇ -amyloidopathy or an ⁇ -synucleopathy.
  • the proband already takes at least one substance which is transported via the cerebral ABCC1 transporter means that this substance needs not to be administered. On the contrary it is already present in the body of the proband, for example, as a result of a drug treatment of another disease.
  • the body fluid samples of the proband which are studied are preferably samples from blood plasma, blood serum and/or cerebral spinal fluid.
  • the ⁇ -amyloidopathy is preferably an Alzheimer's dementia, the ⁇ -synucleinopathy is preferably Parkinson's disease.
  • the ⁇ -synucleinopathy can also be a dementia with Lewy bodies (DLB).
  • Substances which are transported via the cerebral ABCC1 transporter are preferably selected from antibiotics (e.g. difloxacin, grepafloxacin), virostatics/antiviral medicaments (e.g. saquinavir, ritonavir), anti-allergics/antihistamines (e.g. cimetidine), cardio-vascular medicaments (e.g. verapamil), antidepressants (e.g.
  • citalopram antihyperuricemics
  • probenecid e.g. probenecid
  • cytostatics e.g. methotrexate, etoposit, edatrexate, ZD1694
  • vitamins/vitamin analogues e.g. methotrexate, folic acid, L-leucovorin
  • antiphlogistics e.g. indomethacin
  • anti-epileptics e.g. valproic acid
  • hormones/hormone derivatives e.g. 17 ⁇ -estradiol
  • leukotrienes e.g. LTC4
  • fluorescent samples e.g.
  • toxinsor of medicaments e.g. 2,4-dinitrophenyl-SG, bimane-SG, N-ethylmaleimide-SG, doxorubicin-SG, thiotepa-SG, cyclophosphamide-SG, melphalan-SG, chlorambucil-SG, ethacrynic acid-SG, metolachlor-SG, atrazine-SG, sulforaphan-SG, aflatoxin B1-epoxide-SG, 4-nitroquinolin 1-oxide-SG, As(SG)3, etoposide-gluc, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL)-3 ⁇ -0-gluc, SN-38-gluc, 4-methylumbelliferyl- ⁇
  • This indirect analysis of the transport activity of ABCC1 transporters can be used for the diagnosis/prediagnosis of a corresponding disease.
  • the profile of the active substance concentration in body fluids preferably blood plasma, serum and/or cerebrospinal fluid can be investigated.
  • a time-dependent measurement for probands in whom there is a reduced ABCC1 transport activity compared with healthy probands shows a delayed or shifted substance concentration curve (concentration c plotted over time t), i.e. the maximum of the curve varies with time.
  • mice The relative significance of members of the ABC transporter family was then investigated in vivo using newly generated ABC transporter-deficient Alzheimer mouse models.
  • the genetically modified mice each exhibit a deficiency (knock out) at specific ABC transporters ABCG2, ABCB1 or ABCC1.
  • the A ⁇ immunohistochemistry of brain sections showed:
  • ABCB1-deficient mice showed a smaller increase in the number and size of A ⁇ -plaques than ABCC1-deficient mice.
  • enzyme-coupled immune adsorption tests were used for A ⁇ .
  • the ABCC1-deficient mice showed a significant increase in aggregated A ⁇ compared to the control mice at all measurement time points.
  • the cerebral loading with A ⁇ was greatest at an age of 25 weeks.
  • the A ⁇ values (A ⁇ 42) were 12 times higher than in the control mice.
  • Buffer-soluble A ⁇ also increased with age but after 25 weeks, at the time of the highest plaque loading, the values of the soluble A ⁇ in the ABCC1-deficient group decreased substantially.
  • ABC transporters depend inter alia on specific protein/peptide characteristics such as the specific charge.
  • the Dutch-type variant of the amyloid precursor protein Dutch mutant, APP dt ) which introduces an additional negative charge near the interface of the ⁇ -secretase of the APP and thus results in a severe cerebral amyloidangiopathy (CAA) influence the elimination of A ⁇ dt via the blood-brain barrier.
  • the Western blot analyses of brain capillaries and plexuschoroideus (CP) from control mice showed a strong expression of ABCB1 in cerebral capillary endothelial cells (BC) and of ABCC1 in CP ( FIG. 3 d ).
  • Thiethylperazine proved to be a particularly efficient activator of the ABCC1 transporter.
  • Other derivatives starting from the same scaffold also showed good results in the activation of the ABCC1 transporter.
  • the corresponding derivatives are represented in the general formula I
  • R 1 and R 2 are the same or different and each independently of one another are C 1 -C 6 alkyl groups, which independently of one another optionally comprise another substituent selected from alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio groups and halogen atoms, wherein the respective alkyl groups optionally comprise at least one further halogen atom and the residue
  • R 3 is located at one of the positions 6-9 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR 4 R 5 or OR 6 group, wherein R 4 , R 5 and R 6 are the same or different and each independently of one another are selected from hydrogen and C 1 -C 3 alkyl groups and the residue
  • R 7 is located at one of the positions 1, 2 or 4 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR 8 R 9 or OR 10 group, wherein R 8 , R 9 and R 10 are the same or different and each independently of one another are selected from hydrogen and C 1 -C 3 alkyl group.
  • the halogen atom/the halogen atoms are preferably selected from fluorine and chlorine.
  • the residues R 1 and R 2 are the same or different and each independently of one another are a C 1 -C 6 alkyl group or a C 1 -C 6 alkyl group (preferably C 1 alkyl) substituted with an acetyl group and the residues R 3 and R 7 are hydrogen or an acetyl group.
  • the residues R 1 and R 2 are the same or different and each independently of one another are a C 1 -C 3 alkyl group. It is further preferred that the residues R 3 and R 7 are hydrogen.
  • the residue R 1 is a methyl group
  • the residue R 2 is an ethyl group
  • the residues R 3 and R 7 are hydrogen (thiethylperazine, Torecan®).
  • further active substances preferably 1-benzohydrylpiperazines, most preferably 1-benzohydryl-4-cinnamyl piperazine (cinnarizine).
  • Various neurodegenerative diseases can be treated with the 2-(R 2 -thio)-10-[3-(4-R 1 -piperazin-1 -yl)propyl]-10H-phenothiazine derivatives according to the invention or can be diagnosed by means of the indirect analysis described above.
  • the neurodegenerative disease is a ⁇ -amyloidopathy, in particular Alzheimer's dementia (AD).
  • AD Alzheimer's dementia
  • Another embodiment relates to the case that the neurodegenerative disease is an ⁇ -synucleinopathy, in particular Parkinson's disease (PD).
  • Both diseases, i.e. ⁇ -amyloidopathy and ⁇ -synucleinopathies are characterized by cerebral protein deposits which can be treated by means of an activation of the ABCC1 transporter or can be diagnosed by means of its activity.
  • LBD Lewy body dementia
  • Another embodiment relates to the case that the neurodegenerative disease is Huntington's disease (HD).
  • Another embodiment relates to the case that the neurodegenerative disease is a prion disease, in particular Creutzfeld-Jacob disease (CJD) orfatalfamilialinsomnia (FFI).
  • Another embodiment relates to the case that the neurodegenerative disease is a tauopathy, in particular cortico-basal degeneration (CBD), Steel-Richardson-Olszewski syndrome (PSP, progressivesupranuclearpalsy) or Pick's disease (PiD).
  • CBD cortico-basal degeneration
  • PSP Steel-Richardson-Olszewski syndrome
  • PiD Pick's disease
  • FTLD neurodegenerative disease frontotemporalde generation
  • FTLD neurodegenerative disease frontotemporalde generation
  • ALS amyotrophiclateralsclerosis
  • SCA spinocerebellarataxia
  • SPG spasticparaparesis
  • MS multiple sclerosis
  • ADEM Devic's syndrome
  • FIG. 1 a shows that the cortical density of neuritic plaques in ABCC1-deficient mice (ABCCI ko) is increased by ⁇ 75%;
  • FIG. 1 b,c shows that the mean plaque size is increased (+34%) as a result of the larger number of plaques (+63%) having a size of more than 700 ⁇ m 2 and a lower frequency of smaller plaques ( ⁇ 24%).
  • Error bars standard error (n ⁇ 3);
  • FIG. 1 d shows that the IHC staining in ABCG2-deficient (ABCG2ko), ABCB1-deficient (ABCBIko), ABCC1-deficient (ABCCIko) mice and in control mice shows a higher surface density of A ⁇ in ABCC1-deficient animals.
  • Typical plaques of the same size are shown in section, scaling bars represent500 ⁇ m (overview) and 50 ⁇ m (section) (*p ⁇ 0.05);
  • FIG. 2 a shows that the plaque density in the cortex (coverage) in specific ABC-transporter knockout mice is increased.
  • FIG. 2 c shows that the total increase in the plaque size is associated with fewer smaller plaques and more larger plaques (>700 ⁇ m 2 ) whereas the number of medium-size plaques remains at the same value, error bars, standard error (n ⁇ 5), *p ⁇ 0.05;
  • FIGS. 3 a - 3 e show that the deficiency of ABCC1 promotes the accumulation of A ⁇ and A ⁇ dt and that the activation of ABCC1 (by administration of Torecan) reduces the A ⁇ values;
  • FIG. 3 a shows that at an age of 25 weeks ABCC1 deficiency leads to a marked increase ( ⁇ 12 times) in insoluble A ⁇ ;
  • FIG. 3 b shows that the quantity of buffer-soluble A ⁇ 42 at an age of 25 weeks is noticeably reduced compared with 22 weeks ( ⁇ 56%). This is probably due to the deposition in insoluble deposits. At the same age the area covered by A ⁇ deposits which is measured in the immunohistochemistry is increased by 83% (error bars, standard error n ⁇ 5, p ⁇ 0.05);
  • FIG. 3 d shows that the expression of ABCC1 can be seen predominantly in the plexus choroideus (CP) whereas ABCD1 is principally expressed in the capillaries of the brain (BP);
  • APP-transgenic mice (APP, APP dt ) were obtained from The Jackson Laboratory (Bar Harbor, USA) and the University of Tübingen (Tübingen, Germany).
  • the NEP-deficient mice were obtained from the Riken Brain Research Institute (Saitama, Japan).
  • ABCG2-, ABCB1-, and ABCC1-deficient mice were obtained from Taconic-Farms (Denmark). All transgenic and knockout mouse lines were hybridised for at least 9 generations in the genetic FVB-background. The mice were held in a 12 h/12 h light/dark cycle at 23° C. with free access to food and water.
  • mice were killed by cervical dislocation and perfused transcardially with PBS (phosphate-buffered, physiological saline solution).
  • PBS phosphate-buffered, physiological saline solution
  • the brain was removed and one hemisphere was stored in buffered 4% paraformaldehydefor paraffin embedding andimmunohistochemistry.
  • the other hemisphere was shock-frozen in liquid nitrogen and stored at ⁇ 80° C. for biochemical analyses.
  • ELISA kits (TH40HS, TK42HS) from The Genetics Company (TGC, Schlieren, Switzerland) were used for the quantification of A ⁇ . Brain hemispheres were homogenised using PreCellys24 (12 s, 6,500 rpm). After addingcarbonate buffer (pH 8.0) the homogenisates were mixed using PreCellys (5 s, 5,000 rpm) and centrifuged for 90 min at 4° C. and 24,000 g, in order to separate insoluble from soluble A ⁇ species. The remaining supernatant (buffer-soluble fraction) was mixed with 8M guanidine hydrochloride in a ratio of 1:1.6.
  • the pellet was dissolved in 8 volumes of 5M guanidine hydrochloride, agitated at room temperature for 3 h and centrifuged at 24,000 g for 20 min at 4° C. The remaining supernatant formed the guanidine-soluble fraction (GuaHCl). Protein contents of all the samples were measured three times, using a Nanodrop1000 spectrophotometer (ThermoFisher Scientific, Wilmington, USA). The ELISAswere carried out according to the manufacturer's instructions using suitable dilutions.
  • Tissue homogenisates were prepared for the Western Blots.
  • the total protein concentrations of the extracts were determined using a BCA assay (Pierce, part of Thermo Fisher Scientific, Rockford, USA). After electrophoresis of 10 ⁇ g total protein per trace, the proteins were blotted onto PVDF membranes. After blocking in 5% dry milk in TBST-buffer (50 mMTris pH 7.4, 150 mM NaCl, 0.1% Tween20) for 1 h at room temperature, the blots were studied either on ABCB1 (1:500, D-11, Santa Cruz), ABCC1 (1:200, Alexis Bio) or ⁇ -actin (1:20.000, Sigma) overnight at 4° C. Anti-mouse-HRP, anti-rat-HRP or anti-hare-HRP were used as detection antibodies. An Amersham ECL Plus Detection kit and a Roper Cool Snap HQ 2 camera were used for visualisation.
  • Formalin-fixed brains were embedded in paraffin and cut into 4 ⁇ m thick sections. After removing the paraffin, the sections were further treated with a BondMaxTM Autostainer (Menarini/Leica, Germany). Immunostaining was initiated after blocking of endogenic peroxidase (5 min) and epitoperetrieval for 5 min using 95% formic acid (for antibody 6F3D, Dako, Germany) and 70% formic acid (for antibody 4G8, Millipore, Germany). Primary antibodies were routinely incubated at room temperature for 30 min with the following dilutions: 6F3D (1:100), 4G8 (1:500).
  • Brain sections of APP dt were stained with 4G8-antibody. At least two non-consecutive sections were studied for CAA of the meningeal vessels in a masked manner. All meningeal vessels were counted manually and the severity of the CAA was categorised as follows:
  • the average number of vessels for each category was calculated relative to the total number of identified vessels.
  • Endothelial cells of mouse brain capillaries were prepared as described in Coisne et al. (Coisne, C. et al. Mouse syngenic in vitro blood-brain barrier model: a new tool to examine inflammatory events in cerebral endothelium. Laboratory Investigation; 85, 734-746 (2005)). At least 3-4 week old mice were beheaded and the brains removed. Following dissection of the brain stem, the white matter and the meninges, the tissue was homogenised in two volumes of wash buffer B (WBB) (Hanks buffered salt solution (HBBS), 10 mM HEPES, 0.1% BSA) using a 15 ml glassdouncer (Wheaton Industries, Millville, N.J.; USA).
  • WBB wash buffer B
  • HBBS heads buffered salt solution
  • 10 mM HEPES 0.1% BSA

Abstract

The invention relates to 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]- 10H-phenothiazine according to general formula I, for treating a β-amyloidopathy or an α-synucleinopathy accompanied by a cerebral protein deposit and a reduced activity of the cerebral ABCC1-transporter. The invention also relates to a method for the diagnosis or prediagnosis of a β-amyloidopathy or an α-synucleopathy accompanied by a cerebral protein deposit and a reduced activity of the cerebral ABCC1-transporter, or for determining the risk of a proband suffering from such an illness, the proband already having accumulated substances transported by the cerebral ABCC1 transporter.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This is a division of U.S. patent application Ser. No. 13/821,515 filed Mar. 3, 2013, which is the U.S. national phase of PCT/EP2011/064893 filed Aug. 30, 2011, which claims the Convention priority of DE 10 201 0 044 561.4 filed Sep. 7, 2010.
  • BACKGROUND OF THE INVENTION
  • The accumulation of proteins or protein fragments (peptides) in the brain is a significant feature of age-dependent neurodegenerative diseases. In Alzheimer's dementia (Alzheimer's disease, AD) and cerebral β-amyloidopathy (CAA) the aggregation of β-amyloid peptides (Aβ) is a trigger factor, the basic mechanism being unknown. The Aβ proteostasis, i.e. the equilibrium of production and degradation/removal by means of receptors or proteases is disturbed in AD and CAA. However, so far little attention has been paid to the removal of Aβ peptides by cellular transporters (ABC transporters). In Parkinson's disease, the protein α-synuclein accumulates, which inter alia regulates the dopamine release in the substantia nigra. In Parkinson's disease α-synucleinopathy it is known that ABC transporters play a crucial role for transport (Kortekaas et al., Ann Neural 2005, 57, 176-179). Here there are several subfamilies A-G which can alternatingly transport various substrates (metabolites, medicaments, peptides, proteins, ions) and are even able to replace each other in the transport function (e.g. ABCB1 and ABCC1, Tao et al. Cancer Chemotherapy and Pharmacology, 64, 5, 961-969).
  • It has been shown by means of various genetically modified mouse models that the ABC transporter (a common structural element of the ABC transporter is an ATP-binding cassette and a transport pore) ABCC1 is an important protein/peptide transporter, in particular Aβ transporter, which has extraordinary functional effects on the cerebral protein accumulation. ABCC1 is also an important α-synuclein transporter.
  • The investigations of the transporter activity are shown as an example hereinafter for Aβ transport.
  • In order to determine the ABCC1 activity in vivo, in APP-expressing, transgenic mice, the ABCB1, ABCG2 or ABCC1 transporter was removed genetically (knockout mice) in each case.
  • Here it was found that:
  • i) the quantity of Aβ in the mice lacking the ABCC1 transporter was increased by a factor of 12,
  • ii) loss of the ABCB1 transporter only results in a three-fold increase and
  • iii) loss of ABCG2 has no Aβ-accumulating effect.
  • DESCRIPTION OF THE INVENTION
  • Accordingly, the invention provides substances which suitably influence the ABCC1 transporter to thus be able to treat neurodegenerative diseases, in particular β-amyloidopathiesor α-synucleopathies. Specifically, the invention provides 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]-10H-phenothiazines.
  • In other words, the object was solved by 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]-10H-phenothiazines according to the general formula I
  • Figure US20150374711A1-20151231-C00001
  • wherein the residues
  • R1 and R2 are the same or different and each independently of one another are C1-C6 alkyl groups, which independently of one another optionally comprise another substituent selected from alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio groups and halogen atoms, wherein the respective alkyl groups optionally comprise at least one further halogen atom and the residue
  • R3 is located at one of the positions 6-9 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio or alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR4R5 or OR6 group, wherein R4, R5 and R6 are the same or different and each independently of one another are selected from hydrogen and C1-C3 alkyl groups and the residue
  • R7 is located at one of the positions 1, 2 or 4 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio or alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR8R9 or OR10 group, wherein R8, R9 and R10 are the same or different and each independently of one another are selected from hydrogen and C1-C3 alkyl group, for treating a β-amyloidopathy or an α-synucleinopathy accompanied by a cerebral protein deposit.
  • Furthermore, both in the case of α-synucleinopathies and in the case of β-amyloidopathies, there is a need to identify or to diagnose or prediagnose these diseases.
  • The invention also provides a method with which α-synucleinopathies and also β-amyloidopathies can be diagnosed or prediagnosed. Specifically, the invention provides a method for the diagnosis or prediagnosis of a β-amyloidopathy or α-synucleopathy or for determining the risk of a proband to develop such an illness, wherein the proband already takes substances transported by the cerebral ABCC1 transporter, including the following steps:
  • a) determining the quantity of ingested substance in body fluid samples of the proband at a specific time point;
  • b) repeating the determination of step a) at at least one further later time point;
  • c) comparing the quantities determined in step a) and b) with quantities which had been defined as characteristic at the same time points for probands who at the time of the sampling showed no clinical symptoms of a β-amyloidopathy or an α-synucleopathy.
  • The fact that the proband already takes at least one substance which is transported via the cerebral ABCC1 transporter means that this substance needs not to be administered. On the contrary it is already present in the body of the proband, for example, as a result of a drug treatment of another disease. The body fluid samples of the proband which are studied are preferably samples from blood plasma, blood serum and/or cerebral spinal fluid.
  • The β-amyloidopathy is preferably an Alzheimer's dementia, the α-synucleinopathy is preferably Parkinson's disease. Optionally, the α-synucleinopathy can also be a dementia with Lewy bodies (DLB). Substances which are transported via the cerebral ABCC1 transporter are preferably selected from antibiotics (e.g. difloxacin, grepafloxacin), virostatics/antiviral medicaments (e.g. saquinavir, ritonavir), anti-allergics/antihistamines (e.g. cimetidine), cardio-vascular medicaments (e.g. verapamil), antidepressants (e.g. citalopram), antihyperuricemics (e.g. probenecid), cytostatics (e.g. methotrexate, etoposit, edatrexate, ZD1694), vitamins/vitamin analogues (e.g. methotrexate, folic acid, L-leucovorin), antiphlogistics (e.g. indomethacin), anti-epileptics (e.g. valproic acid), hormones/hormone derivatives (e.g. 17β-estradiol), leukotrienes (e.g. LTC4), fluorescent samples (e.g. calcein, Fluo-3, BCECF, SNARF), GSH-, sulphate-orglucuronide-coupledmetabolites of natural substances (endogenously produced), toxinsor of medicaments (e.g. 2,4-dinitrophenyl-SG, bimane-SG, N-ethylmaleimide-SG, doxorubicin-SG, thiotepa-SG, cyclophosphamide-SG, melphalan-SG, chlorambucil-SG, ethacrynic acid-SG, metolachlor-SG, atrazine-SG, sulforaphan-SG, aflatoxin B1-epoxide-SG, 4-nitroquinolin 1-oxide-SG, As(SG)3, etoposide-gluc, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL)-3β-0-gluc, SN-38-gluc, 4-methylumbelliferyl-β-d-gluc, 6-hydroxy-5,7-dimethyl-2-methylamino-4-(3-pyridylmethyl)-benzothiazolsulfate (E3040S)-gluc, leukotriene C4, prostaglandin A2-SG, 15-deoxy-Δ12,14 prostaglandin J2-SG, hydroxynonenal-SG, 17β-estradiol-17β-d-gluc, glucuronosylbilirubin, bis-lucuronosylbilirubin, hyodeoxycholate-6-α-gluc, estron-3-sulfate, dehydroepiandrosteronesulfate, sulfatolithocholate) (see also Deeley R G et al.: Substrate recognition and transport by multi drug resistance protein 1 (ABCC1), FEBS letters 2006, 580 (4), pp. 1103-1111.)
  • This indirect analysis of the transport activity of ABCC1 transporters can be used for the diagnosis/prediagnosis of a corresponding disease. In probands who already take ABCC1-transportable substances by other routes, the profile of the active substance concentration in body fluids, preferably blood plasma, serum and/or cerebrospinal fluid can be investigated. A time-dependent measurement for probands in whom there is a reduced ABCC1 transport activity compared with healthy probands shows a delayed or shifted substance concentration curve (concentration c plotted over time t), i.e. the maximum of the curve varies with time.
  • When a shifted curve is obtained compared with the healthy case, this is an indication of a changed ABCC1 transport activity. This means that substances such as Aβ or α-synuclein are transported less efficiently and is therefore an indication of a corresponding disease.
  • Both the mouse model and the pharmacological influencing of the ABCC1 show that this is an important cellular transmembrane transporter for the Aβ protein and imply that the blood-brain barrier and the plexus choroideus occupy a key position for the Aβ release from the brain. It could be shown that the selective pharmacological activation of the ABCC1 transporter significantly reduces the cerebral loading with Aβ and can thus be used therapeutically for the treatment of diseases with disturbed brain proteostasis. Furthermore, the analysis of the transporter activity of the ABCC1 transporter as described above can be used for the indirect or direct diagnosis/prediagnosis of a corresponding disease. Direct analysis would be possible via the administration of substances which are transported via the ABCC1 transporter, and the determination thereof. The indirect analysis has already been described further above.
  • Changes to export mechanisms which are related to ABC transporters can substantially influence the temporal aggregation profile of Aβ and other brain proteins. Consequently an influencing of the function of the ABCC1 transporter has a positive effect on the risk of developing neurodegenerative diseases, particularly Alzheimer's disease. “Treatment of neurodegenerative diseases” in this sense comprises the prevention and also the treatment of pre-existing diseases.
  • The role of the ABC transporter in the Aβ release was initially studied in such a manner that it was demonstrated that ABCC1 is able to transport Aβ. For this purpose in vitro transwell assays with endothelial cells (endothelial cell transwell assay, ECTA) of primary cultivated capillary endothelial cells from mouse brains (cell culture approach) were used.
  • Primary cultures of endothelial cells from brain capillaries of ABCB1-deficient, ABCC1-deficient (knock out) mice and control mice (C57B1/6, FVB/N) were used to study the Aβ-specific transport activity. The transport of Aβ from the abluminal (brain) into the luminal (blood) compartment is impaired in ABCB1-deficient and ABCC1-deficient endothelial cells. The mean Aβ transport rate during the first six hours after administration of Aβ peptides (Aβ42) was 2.2 pg/min for the control cells. In contrast to this, the ABCC1-deficient cells only reached half the transport capacity (1.0 pg/min). In the ABCB1-deficient cells the Aβ transport was almost non-existent (0.3 pg/min). Further investigations of capillary endothelial cells and cells from the plexus choroideus revealed that the ABCB1 transporter is strongly expressed in brain capillary endothelial cells whereas the endothelial ABCC1 expression in brain capillaries is lower.
  • The relative significance of members of the ABC transporter family was then investigated in vivo using newly generated ABC transporter-deficient Alzheimer mouse models. The genetically modified mice each exhibit a deficiency (knock out) at specific ABC transporters ABCG2, ABCB1 or ABCC1.
  • The Aβ immunohistochemistry of brain sections showed:
  • i) significant increases in the cortical number and the size of Aβ-positive plaques in ABCC1-deficient mice compared to control mice (see FIGS. 1 and 2 a-c).
  • ii) ABCB1-deficient mice showed a smaller increase in the number and size of Aβ-plaques than ABCC1-deficient mice.
  • iii) No significant difference could be determined between control mice and ABCG2-deficient mice (FIG. 2 a-c).
  • In order to determine the quantity of buffer-soluble Aβ (mostly monomers and smaller oligomers) and of guanidine-soluble Aβ (mostly fibrillar or aggregated material), enzyme-coupled immune adsorption tests (enzyme-linked immuno absorbent assays, ELISAs) were used for Aβ.
  • In agreement with the morphological results from the immunohistochemistry, the ABCC1-deficient mice showed a significant increase in aggregated Aβ compared to the control mice at all measurement time points. The cerebral loading with Aβ was greatest at an age of 25 weeks. At this time point, the Aβ values (Aβ42) were 12 times higher than in the control mice. Buffer-soluble Aβ also increased with age but after 25 weeks, at the time of the highest plaque loading, the values of the soluble Aβ in the ABCC1-deficient group decreased substantially.
  • Further investigations were carried out which provided further proof for the relationship between the possibly lacking removal by ABCC1 and the aggregation of Aβ.
  • The transport kinetics of ABC transporters depend inter alia on specific protein/peptide characteristics such as the specific charge. The Dutch-type variant of the amyloid precursor protein (Dutch mutant, APPdt) which introduces an additional negative charge near the interface of the α-secretase of the APP and thus results in a severe cerebral amyloidangiopathy (CAA) influence the elimination of Aβdt via the blood-brain barrier. The Western blot analyses of brain capillaries and plexuschoroideus (CP) from control mice showed a strong expression of ABCB1 in cerebral capillary endothelial cells (BC) and of ABCC1 in CP (FIG. 3 d). Since ABC transporters play an important role in the elimination of Aβ, it was assumed that ABC-transporter-deficient (at the blood-brain barrier and at the blood plexus choroideus barrier) APPdt-transgenic mice exhibit an increased accumulation of Aβdt in meningeal vessels. The degree of CAA in the ABC-deficient APPdt mice was quantified at the age of 24 months. In agreement with the assumption, at least 51% of the vessels were severely impaired (>75% of the vessel wall loaded with Aβ) in the ABCC1-deficient animals compared to 23% in the controls (FIG. 3 c).
  • On the basis of these results, it was investigated how far the content of soluble Aβ in the brain could be reduced/influenced by active-substance-mediated activation of ABC transporters. Mice with amyloid deposits were treated for 30 days with the anti-emeticthiethylperazine (torecan, 2-(ethylthio)-10-[3-(4-methylpiperazin-1-yl)propyl]-10H-phenothiazine). 3 mg/kg body weight was administered intramuscularly twice daily. The preventative treatment began before the mice exhibited senile plaques. ELISA measurements of the treated animals showed a reduction in the quantity of Aβ of at least 31% in the treated mice compared to vehicle-treated animals (vehicle=water) (FIG. 3 e). The results are reproduced graphically in FIGS. 3 a-3 e.
  • The capacity to remove Aβ proved to be a key factor in the regulation of the intracerebral accumulation of Aβ.
  • Thiethylperazine (Torecan®) proved to be a particularly efficient activator of the ABCC1 transporter. Other derivatives starting from the same scaffold also showed good results in the activation of the ABCC1 transporter. The corresponding derivatives are represented in the general formula I
  • Figure US20150374711A1-20151231-C00002
  • wherein the residues
  • R1 and R2 are the same or different and each independently of one another are C1-C6 alkyl groups, which independently of one another optionally comprise another substituent selected from alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio groups and halogen atoms, wherein the respective alkyl groups optionally comprise at least one further halogen atom and the residue
  • R3 is located at one of the positions 6-9 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR4R5 or OR6 group, wherein R4, R5 and R6 are the same or different and each independently of one another are selected from hydrogen and C1-C3 alkyl groups and the residue
  • R7 is located at one of the positions 1, 2 or 4 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl (preferably acetyl), amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR8R9 or OR10 group, wherein R8, R9 and R10 are the same or different and each independently of one another are selected from hydrogen and C1-C3 alkyl group.
  • These derivatives are accordingly well suited to the treatment of neurodegenerative diseases, in particular β-amyloidopathies or α-synucleinopathies where the treatment, as already mentioned, comprises both the prevention and the treatment of pre-existing diseases. The halogen atom/the halogen atoms are preferably selected from fluorine and chlorine. The acyl groups (—(C=0)-R) of the residues R1,2,3,7 are preferably acetyl groups (—C(=0)CH3). preferably the residues R1 and R2 are the same or different and each independently of one another are a C1-C6 alkyl group or a C1-C6 alkyl group (preferably C1 alkyl) substituted with an acetyl group and the residues R3 and R7 are hydrogen or an acetyl group. In a preferred embodiment the residues R1 and R2 are the same or different and each independently of one another are a C1-C3 alkyl group. It is further preferred that the residues R3 and R7 are hydrogen. It is particularly preferred that the residue R1 is a methyl group, the residue R2 is an ethyl group and the residues R3 and R7 are hydrogen (thiethylperazine, Torecan®). When used for the treatment of neurodegenerative diseases, it has proved advantageous to add further active substances, preferably 1-benzohydrylpiperazines, most preferably 1-benzohydryl-4-cinnamyl piperazine (cinnarizine).
  • Various neurodegenerative diseases can be treated with the 2-(R2-thio)-10-[3-(4-R1-piperazin-1 -yl)propyl]-10H-phenothiazine derivatives according to the invention or can be diagnosed by means of the indirect analysis described above. In a particularly preferred embodiment, the neurodegenerative disease is a β-amyloidopathy, in particular Alzheimer's dementia (AD). Another embodiment relates to the case that the neurodegenerative disease is an α-synucleinopathy, in particular Parkinson's disease (PD). Both diseases, i.e. β-amyloidopathy and α-synucleinopathies are characterized by cerebral protein deposits which can be treated by means of an activation of the ABCC1 transporter or can be diagnosed by means of its activity.
  • Other diseases which can also be treated by activation of the ABCC1 transporter or which can be diagnosed by means of the ABCC1 transporter activity are mentioned hereinafter. Another treatable disease is thus Lewy body dementia (LBD). This is also characterized by cerebral protein aggregation, i.e. is an α-synucleinopathy like Parkinson's disease.
  • Another embodiment relates to the case that the neurodegenerative disease is Huntington's disease (HD). Another embodiment relates to the case that the neurodegenerative disease is a prion disease, in particular Creutzfeld-Jacob disease (CJD) orfatalfamilialinsomnia (FFI). Another embodiment relates to the case that the neurodegenerative disease is a tauopathy, in particular cortico-basal degeneration (CBD), Steel-Richardson-Olszewski syndrome (PSP, progressivesupranuclearpalsy) or Pick's disease (PiD). Another embodiment relates to the case that the neurodegenerative disease frontotemporalde generation (FTLD), in particular ubiquitin-positive degeneration, TDP43-positive degeneration or forubiquitin and TDP43-negative degenerations. Another embodiment relates to the case that the neurodegenerative disease is an amyotrophiclateralsclerosis (ALS). Another embodiment relates to the case that the neurodegenerative disease is a spinocerebellarataxia (SCA) or spasticparaparesis (SPG). Another embodiment relates to the case that the neurodegenerative/neuroimmunological disease is multiple sclerosis (MS) or an MS-related syndrome, in particular ADEM or Devic's syndrome.
  • DESCRIPTION OF THE FIGURES
  • In the figures
  • FIG. 1 a shows that the cortical density of neuritic plaques in ABCC1-deficient mice (ABCCI ko) is increased by ˜75%;
  • FIG. 1 b,c shows that the mean plaque size is increased (+34%) as a result of the larger number of plaques (+63%) having a size of more than 700 μm2 and a lower frequency of smaller plaques (−24%). Error bars, standard error (n≧3);
  • FIG. 1 d shows that the IHC staining in ABCG2-deficient (ABCG2ko), ABCB1-deficient (ABCBIko), ABCC1-deficient (ABCCIko) mice and in control mice shows a higher surface density of Aβ in ABCC1-deficient animals. Typical plaques of the same size are shown in section, scaling bars represent500 μm (overview) and 50 μm (section) (*p<0.05);
  • FIG. 2 a shows that the plaque density in the cortex (coverage) in specific ABC-transporter knockout mice is increased. In particular ABCC1-deficient (ABCCIko) mice show an increased Aβ-amyloid loading (light-grey bars, in each case on the outside right in the individual groupings), w=week on the abscissa;
  • FIG. 2 b shows that the total plaque size in ABCC1-deficient (ABCCIko) and ABCB1-deficient (ABCBIko) mice at the age of 25 weeks is increased, w=week on the abscissa;
  • FIG. 2 c shows that the total increase in the plaque size is associated with fewer smaller plaques and more larger plaques (>700 μm2) whereas the number of medium-size plaques remains at the same value, error bars, standard error (n≧5), *p<0.05;
  • FIGS. 3 a-3 e show that the deficiency of ABCC1 promotes the accumulation of Aβ and Aβdt and that the activation of ABCC1 (by administration of Torecan) reduces the Aβ values; where
  • FIG. 3 a shows that at an age of 25 weeks ABCC1 deficiency leads to a marked increase (˜12 times) in insoluble Aβ; and
  • FIG. 3 b shows that the quantity of buffer-soluble Aβ42 at an age of 25 weeks is noticeably reduced compared with 22 weeks (−56%). This is probably due to the deposition in insoluble deposits. At the same age the area covered by Aβ deposits which is measured in the immunohistochemistry is increased by 83% (error bars, standard error n≧5, p<0.05);
  • FIG. 3 c shows that 53% of the blood vessels are severely impaired by CAA (>75% of the vessel walls exhibit An). This relates to ABCC1-deficient mice (ABCC1ko) compared to 23% in the controls (n=3);
  • FIG. 3 d shows that the expression of ABCC1 can be seen predominantly in the plexus choroideus (CP) whereas ABCD1 is principally expressed in the capillaries of the brain (BP);
  • FIG. 3 e shows that the activation of ABCC1 by thiethylperazine (Torecan) lowers the Aβ values in mice (−28%), error bars, standard error (n=4, *p<0.05).
  • EXAMPLES Animals
  • APP-transgenic mice (APP, APPdt) were obtained from The Jackson Laboratory (Bar Harbor, USA) and the University of Tübingen (Tübingen, Germany). The NEP-deficient mice were obtained from the Riken Brain Research Institute (Saitama, Japan). ABCG2-, ABCB1-, and ABCC1-deficient mice were obtained from Taconic-Farms (Denmark). All transgenic and knockout mouse lines were hybridised for at least 9 generations in the genetic FVB-background. The mice were held in a 12 h/12 h light/dark cycle at 23° C. with free access to food and water.
  • Methods Tissue Preparation
  • For the tissue preparation the mice were killed by cervical dislocation and perfused transcardially with PBS (phosphate-buffered, physiological saline solution). The brain was removed and one hemisphere was stored in buffered 4% paraformaldehydefor paraffin embedding andimmunohistochemistry. The other hemisphere was shock-frozen in liquid nitrogen and stored at −80° C. for biochemical analyses.
  • ELISA
  • ELISA kits (TH40HS, TK42HS) from The Genetics Company (TGC, Schlieren, Switzerland) were used for the quantification of Aβ. Brain hemispheres were homogenised using PreCellys24 (12 s, 6,500 rpm). After addingcarbonate buffer (pH 8.0) the homogenisates were mixed using PreCellys (5 s, 5,000 rpm) and centrifuged for 90 min at 4° C. and 24,000 g, in order to separate insoluble from soluble Aβ species. The remaining supernatant (buffer-soluble fraction) was mixed with 8M guanidine hydrochloride in a ratio of 1:1.6. To extract the aggregated Aβ species, the pellet was dissolved in 8 volumes of 5M guanidine hydrochloride, agitated at room temperature for 3 h and centrifuged at 24,000 g for 20 min at 4° C. The remaining supernatant formed the guanidine-soluble fraction (GuaHCl). Protein contents of all the samples were measured three times, using a Nanodrop1000 spectrophotometer (ThermoFisher Scientific, Wilmington, USA). The ELISAswere carried out according to the manufacturer's instructions using suitable dilutions.
  • Western Blots
  • Tissue homogenisates were prepared for the Western Blots. The total protein concentrations of the extracts were determined using a BCA assay (Pierce, part of Thermo Fisher Scientific, Rockford, USA). After electrophoresis of 10 μg total protein per trace, the proteins were blotted onto PVDF membranes. After blocking in 5% dry milk in TBST-buffer (50 mMTris pH 7.4, 150 mM NaCl, 0.1% Tween20) for 1 h at room temperature, the blots were studied either on ABCB1 (1:500, D-11, Santa Cruz), ABCC1 (1:200, Alexis Bio) or β-actin (1:20.000, Sigma) overnight at 4° C. Anti-mouse-HRP, anti-rat-HRP or anti-hare-HRP were used as detection antibodies. An Amersham ECL Plus Detection kit and a Roper Cool Snap HQ2 camera were used for visualisation.
  • Immunohistochemistry (IHC)
  • Formalin-fixed brains were embedded in paraffin and cut into 4 μm thick sections. After removing the paraffin, the sections were further treated with a BondMax™ Autostainer (Menarini/Leica, Germany). Immunostaining was initiated after blocking of endogenic peroxidase (5 min) and epitoperetrieval for 5 min using 95% formic acid (for antibody 6F3D, Dako, Germany) and 70% formic acid (for antibody 4G8, Millipore, Germany). Primary antibodies were routinely incubated at room temperature for 30 min with the following dilutions: 6F3D (1:100), 4G8 (1:500). Primary antibodies were detected with the BondMax™ Bond Polymer Ref inedetection kit and according to the DAB R30 standard protocol. The sections were completely digitised with a resolution of 230 nm using a MiraxDesk/MiraxMidi scanner and then analysed automatically using the AxioVision software package (Zeiss, Germany).
  • Assessment of the Severity of the CAA
  • Brain sections of APPdt were stained with 4G8-antibody. At least two non-consecutive sections were studied for CAA of the meningeal vessels in a masked manner. All meningeal vessels were counted manually and the severity of the CAA was categorised as follows:
  • Category I: not adversely affected
  • Category II: ≦25% of the periphery positively stained
  • Category III: ≦50% of the periphery positively stained
  • Category IV: ≦75% of the periphery positively stained
  • Category V: ≦100% of the periphery positively stained
  • The average number of vessels for each category was calculated relative to the total number of identified vessels.
  • Endothelial Cell Transwell Assay (ECTA)
  • Endothelial cells of mouse brain capillaries were prepared as described in Coisne et al. (Coisne, C. et al. Mouse syngenic in vitro blood-brain barrier model: a new tool to examine inflammatory events in cerebral endothelium. Laboratory Investigation; 85, 734-746 (2005)). At least 3-4 week old mice were beheaded and the brains removed. Following dissection of the brain stem, the white matter and the meninges, the tissue was homogenised in two volumes of wash buffer B (WBB) (Hanks buffered salt solution (HBBS), 10 mM HEPES, 0.1% BSA) using a 15 ml glassdouncer (Wheaton Industries, Millville, N.J.; USA). One volume of 30% dextran solution was added to the homogenisate. This was centrifuged twice at 3,000 g and 4° C. The pellet containing the vessels was resuspended in WBB and large vessels were broken up manually by harsh pipetting of the solution. Vacuum filtration through 60 μm membranes (SEFAR, Switzerland) was used to separate large vessels from the capillaries. After combined treatment with collagenase/dispase (HBSS, 10 mM HEPES, 0.15 g/ml TCLK, 10 μg/ml DNAse-I, 1 mg/ml collagenase/dispase (Roche) single cell suspension was achieved by further harsh pipetting of the solution. Endothelial cells were inserted into Matrigel-coated Trans well inserts (0.4 μm pores, Greiner Bio-One, Germany) having a density of 120,000 cells per insert and allowed to grow on a supporting glial culture.
  • Sulphur yellow was used to determine the paracellular flux during the assay. The culture medium of the abluminal compartment was replaced with a solution containing 10 ng Ass42 (1.6 nM final concentration). Samplesfrom the luminal compartment were then taken after 2 h, 6 h or 24 h and the Aβ content was determined with ELISA (TK42-highsense, TGC, Switzerland). The transport rate was described in Coisneet al. (Coisne, C. et al. Mouse syngenic in vitro blood-brain barrier model: a new tool to examine inflammatory events in cerebral endothelium. Laboratory Investigation; 85, 734-746 (2005)).
  • ELISA Statistics
  • The Lilliefors goodness-of-fit test (alpha=0.05) was applied to the ELISA data and to the log-transformed ELISA data to distinguish between the assumption of normally distributed sample data and the assumption of log-normally distributed sample data. Despite the small sample size, the null hypothesis (H0) was dismissed for both sets of data for 5 of 44 samples. In agreement with the observation of predominantly positive (skew) and strictly positive sample data, the assumption of normally distributed data was rejected. Mean confidence intervals were calculated assuming a basic log-normal distribution. The Wilcoxon rank-sum test was applied to compare the ELISA data of the various mouse strains for each time point.

Claims (11)

1. A method for treating a β-amyloidopathy or an α-synucleinopathy neurodegenerative disease, the method comprising:
administering to a patient exhibiting a β-amyloidopathy or an α-synucleinopathy accompanied by a cerebral protein deposit and a reduced activity of the cerebral ABCC1-transporter a 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]-10H-phenothiazine according to the general formula I, thereby activating the cerebral ABCC1-transporter of the patient;
Figure US20150374711A1-20151231-C00003
wherein the residues
R1 and R2 are the same or different and each independently of one another is a C1-C6 alkyl group, which independently of one another optionally comprises another substituent selected from the group consisting of alkyl, aryl, acyl, amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, and alkylthio groups and halogen atoms, wherein the respective alkyl groups optionally comprise at least one further halogen atom and the residue;
R3 is located at one of the positions 6-9 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl, amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR4R5 or OR6 group, wherein R4, R5, and R6 are the same or different and each independently of one another is selected from the group consisting of hydrogen and C1-C3 alkyl groups and the residue; and
R7 is located at one of the positions 1, 2, or 4 of the phenothiazine ring system and is a hydrogen atom or an alkyl, aryl, acyl, amino, nitro, sulfonyl, hydroxyl, alkoxy, aryloxy, arylthio, or alkylthio group or a halogen atom, wherein the respective alkyl groups optionally comprise at least one further halogen atom or an NR8R9 or OR10 group, wherein R8, R9, and R10 are the same or different and each independently of one another is selected from the group consisting of hydrogen and C1-C3 alkyl groups.
2. The method of claim 1, wherein each halogen atom is selected from the group consisting of fluorine and chlorine.
3. The method of claim 1, wherein R1 and R2 are the same or different and each independently of one another is a C1-C3 alkyl group.
4. The method of claim 1, wherein the residues R3 and R7 are hydrogen.
5. The method of claim 1, wherein the residue R1 is a methyl group, the residue R2 is an ethyl group, and the residues R3 and R7 are hydrogen.
6. The method of claim 1, further comprising: administering to the patient a further active substance along with the 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]-10H-phenothiazine.
7. The method of claim 6, wherein a 1-benzohydrylpiperazine is the further active substance.
8. The method of claim 7, wherein 1-benzohydryl-4-cinnamyl piperazine is the 1-benzohydrylpiperazine.
9. The method of claim 1, wherein the patient exhibits the β-amyloidopathy, and the β-amyloidopathy is Alzheimer's dementia.
10. The method of claim 1, wherein the patient exhibits the α-synucleinopathy, and the α-synucleinopathy is Parkinson's disease or Lewy body dementia.
11. The method of claim 1, comprising administering the 2-(R2-thio)-10-[3-(4-R1-piperazin-1-yl)propyl]-10H-phenothiazine to the patient prior to the patient exhibiting senile plaques.
US14/847,089 2010-09-07 2015-09-08 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof Abandoned US20150374711A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/847,089 US20150374711A1 (en) 2010-09-07 2015-09-08 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
DE102010044561 2010-09-07
DE102010044561.4 2010-09-07
PCT/EP2011/064893 WO2012031941A2 (en) 2010-09-07 2011-08-30 2-(R²-THIO)-10-[3-(4-R1-PIPERAZIN-1-YL)PROPYL]-10H-PHENOTHIAZINE FOR TREATING A β-AMYLOIDOPATHY OR AN ALPHA-SYNUCLEOPATHY, AND METHOD FOR THE DIAGNOSIS OR PREDIAGNOSIS THEREOF
US201313821515A 2013-04-03 2013-04-03
US14/847,089 US20150374711A1 (en) 2010-09-07 2015-09-08 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2011/064893 Division WO2012031941A2 (en) 2010-09-07 2011-08-30 2-(R²-THIO)-10-[3-(4-R1-PIPERAZIN-1-YL)PROPYL]-10H-PHENOTHIAZINE FOR TREATING A β-AMYLOIDOPATHY OR AN ALPHA-SYNUCLEOPATHY, AND METHOD FOR THE DIAGNOSIS OR PREDIAGNOSIS THEREOF
US13/821,515 Division US20130184268A1 (en) 2010-09-07 2011-08-30 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl) propyl]-10h-phenothiazine for treating a beta-amyloidopathy or an alpha-synucleopathy, and method for the diagnosis or prediagnosis thereof

Publications (1)

Publication Number Publication Date
US20150374711A1 true US20150374711A1 (en) 2015-12-31

Family

ID=44645670

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/821,515 Abandoned US20130184268A1 (en) 2010-09-07 2011-08-30 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl) propyl]-10h-phenothiazine for treating a beta-amyloidopathy or an alpha-synucleopathy, and method for the diagnosis or prediagnosis thereof
US14/499,918 Active 2031-09-30 US9370523B2 (en) 2010-09-07 2014-09-29 Method for diagnosing or prediagnosing a beta-amyloidopathy or an alpha-synucleopathy
US14/847,089 Abandoned US20150374711A1 (en) 2010-09-07 2015-09-08 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl)propyl]-10h-phenothiazines for treating a b-amyloidopathy or an a-synucleopathy, and method for the diagnosis or prediagnosis thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/821,515 Abandoned US20130184268A1 (en) 2010-09-07 2011-08-30 2-(r2-thio)-10-[3-(4-r1-piperazin-1-yl) propyl]-10h-phenothiazine for treating a beta-amyloidopathy or an alpha-synucleopathy, and method for the diagnosis or prediagnosis thereof
US14/499,918 Active 2031-09-30 US9370523B2 (en) 2010-09-07 2014-09-29 Method for diagnosing or prediagnosing a beta-amyloidopathy or an alpha-synucleopathy

Country Status (13)

Country Link
US (3) US20130184268A1 (en)
EP (3) EP2693216B1 (en)
JP (2) JP6084924B2 (en)
CN (2) CN106243060A (en)
BR (1) BR112013005472A2 (en)
CA (2) CA2870626C (en)
DE (2) DE102010062810B4 (en)
DK (2) DK2614060T3 (en)
ES (2) ES2701453T3 (en)
MX (1) MX357521B (en)
PL (2) PL2614060T3 (en)
RU (2) RU2016118021A (en)
WO (1) WO2012031941A2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023131606A1 (en) * 2022-01-04 2023-07-13 Immungenetics Ag Specific dosage of phenothiazine compounds for use in treatment or prevention of alzheimer's dementia

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3621097A (en) * 1970-03-30 1971-11-16 Jan Marcel Didier Aron Samuel Method and compositions for treatment of mental illness
US20030073715A1 (en) * 1994-04-22 2003-04-17 Ragab El-Rashidy Apomorphine-containing dosage form for ameliorating male erectile dysfunction
US20060276460A1 (en) * 2003-04-25 2006-12-07 Noelle Callizot Use of piperazine phenothiazine derivatives in the manufacture of a medicament with neuroprotector and/or neurotrophic effects on cns and/or pns
US7402564B1 (en) * 2006-11-10 2008-07-22 Cara Therapeutics, Inc. Synthetic peptide amides
WO2008092898A1 (en) * 2007-02-01 2008-08-07 Pad Pharma Limited Treatment of protein aggregation diseases
US20120136007A1 (en) * 2009-05-14 2012-05-31 The General Hospital Corporation Methods and compositions for treating degenerative and ischemic disorders

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH365379A (en) * 1956-04-18 1962-11-15 Sandoz Ag Process for the preparation of new phenothiazines substituted in the 3-position by a monovalent sulfur function
BE568701A (en) * 1957-06-18
US4471116A (en) * 1982-07-28 1984-09-11 Hoffmann-La Roche Inc. Substituted (10H-phenothiazin-10-L)-propyl-1-piperazines
CA2196529A1 (en) * 1994-08-08 1996-02-22 Peter Davies Methods for treating and/or preventing alzheimer's disease using phenothiazines and/or thioxanthenes
DE4430091A1 (en) * 1994-08-25 1996-02-29 Bayer Ag Use of N-substituted phenothiazines
DE102005014142B4 (en) * 2005-03-23 2006-11-09 Hennig Arzneimittel Gmbh & Co. Kg Pellet-shaped delayed-release preparation against dizziness
WO2007062862A2 (en) * 2005-12-02 2007-06-07 Ludwig Maximilians Universität München Use of calmodulin inhibitors for the treatment of neurodegenerative disorders
US7956183B2 (en) * 2006-07-11 2011-06-07 Wista Laboratories Ltd. Methods of synthesis and/or purification of diaminophenothiazinium compounds
TW200848063A (en) * 2007-04-23 2008-12-16 Combinatorx Inc Methods and compositions for the treatment of neurodegenerative disorders
PL2282779T3 (en) * 2008-04-29 2013-08-30 Pharnext New therapeutic approaches for treating alzheimer disease and related disorders through a modulation of cell stress response
HUE033726T2 (en) * 2008-04-29 2017-12-28 Pharnext New therapeutic approaches for treating alzheimer disease and related disorders through a modulation of angiogenesis
MX2010011878A (en) * 2008-04-29 2011-04-11 Pharnext Combination compositions for treating alzheimer disease and related disorders with zonisamide and acamprosate.

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3621097A (en) * 1970-03-30 1971-11-16 Jan Marcel Didier Aron Samuel Method and compositions for treatment of mental illness
US20030073715A1 (en) * 1994-04-22 2003-04-17 Ragab El-Rashidy Apomorphine-containing dosage form for ameliorating male erectile dysfunction
US20060276460A1 (en) * 2003-04-25 2006-12-07 Noelle Callizot Use of piperazine phenothiazine derivatives in the manufacture of a medicament with neuroprotector and/or neurotrophic effects on cns and/or pns
US7402564B1 (en) * 2006-11-10 2008-07-22 Cara Therapeutics, Inc. Synthetic peptide amides
WO2008092898A1 (en) * 2007-02-01 2008-08-07 Pad Pharma Limited Treatment of protein aggregation diseases
US20120136007A1 (en) * 2009-05-14 2012-05-31 The General Hospital Corporation Methods and compositions for treating degenerative and ischemic disorders

Also Published As

Publication number Publication date
CN106243060A (en) 2016-12-21
JP6426663B2 (en) 2018-11-21
CA2870626A1 (en) 2012-03-15
CN103237802B (en) 2017-06-09
MX2013002612A (en) 2013-08-01
JP6084924B2 (en) 2017-02-22
ES2701453T3 (en) 2019-02-22
BR112013005472A2 (en) 2020-11-10
EP2614060A2 (en) 2013-07-17
DE102011053114A1 (en) 2012-03-08
EP3097914A3 (en) 2016-12-28
EP2693216B1 (en) 2018-09-12
CA2811454C (en) 2017-03-28
DE102010062810A1 (en) 2012-03-08
RU2013110018A (en) 2014-10-20
CN103237802A (en) 2013-08-07
JP2017019776A (en) 2017-01-26
CA2811454A1 (en) 2012-03-15
EP2614060B1 (en) 2016-08-31
CA2870626C (en) 2018-08-14
PL2693216T3 (en) 2019-04-30
US20130184268A1 (en) 2013-07-18
EP2693216A1 (en) 2014-02-05
DK2614060T3 (en) 2017-01-02
US20150024418A1 (en) 2015-01-22
WO2012031941A2 (en) 2012-03-15
DE102010062810B4 (en) 2014-03-13
ES2605705T3 (en) 2017-03-15
EP3097914A2 (en) 2016-11-30
WO2012031941A3 (en) 2012-09-20
MX357521B (en) 2018-07-12
DK2693216T3 (en) 2019-01-14
RU2587154C2 (en) 2016-06-20
PL2614060T3 (en) 2017-05-31
RU2016118021A (en) 2018-11-01
US9370523B2 (en) 2016-06-21
JP2013537881A (en) 2013-10-07
RU2016118021A3 (en) 2020-02-26

Similar Documents

Publication Publication Date Title
US6008221A (en) Method for treating Alzheimer&#39;s disease with folic acid
JP7419486B2 (en) How to inhibit τ phosphorylation
JP6566867B2 (en) Alzheimer&#39;s disease assay in living patients
JP2002515437A (en) Composition comprising an ionophore for modulating amyloidosis
US20100303805A1 (en) Identification of small molecules recognized by antibodies in subjects with neurodegenerative diseases
US9370523B2 (en) Method for diagnosing or prediagnosing a beta-amyloidopathy or an alpha-synucleopathy
KR20210117762A (en) A screening method for plk-1 inhibitor and plk-1 inhibitor therefrom, use of the same
Kuret Detection and reduction of neurofibrillary lesions
US20080167387A1 (en) Microglia Facilitated Amyloidogenesis Assay
US20240117306A1 (en) Method of inhibiting tau phosphorylation
WO2024061658A1 (en) Thiazole derivatives as tau aggregation inhibitors
US20130035302A1 (en) Use of yessotoxin and analogues and derivatives thereof for treating and/or preventing neurodegenerative diseases linked to tau and beta amyloid
WO2013020909A1 (en) Materials and methods for the treatment of tauopathies
US20180093960A1 (en) Method of inhibiting apolipoprotein-e expression while increasing expression of at least one of ldl-receptor protein or abca1 protein comprising administering a small compound
Best et al. Quantitative measurement of changes in Aβ40 in the rat brain and CSF following treatment with the γ-secretase inhibitor, N2-[(2S)-2-(3, 5-Difluorophenyl)-2-hydroxyethanoyl]-N1-[(7S)-5-methyl-6-oxo-6, 7-dihydro-5Hdibenzo [b, d] azepin-7-yl]-L-alaninamide (LY-411575)
WO2011051521A2 (en) Use of gambierol for treating and/or preventing neurodegenerative diseases related to tau and beta-amyloid

Legal Events

Date Code Title Description
AS Assignment

Owner name: IMMUNGENETICS AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PAHNKE, JENS;REEL/FRAME:038635/0229

Effective date: 20130319

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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