US20190071450A1 - Bicyclic compounds for diagnosis and therapy - Google Patents

Bicyclic compounds for diagnosis and therapy Download PDF

Info

Publication number
US20190071450A1
US20190071450A1 US16/084,223 US201716084223A US2019071450A1 US 20190071450 A1 US20190071450 A1 US 20190071450A1 US 201716084223 A US201716084223 A US 201716084223A US 2019071450 A1 US2019071450 A1 US 2019071450A1
Authority
US
United States
Prior art keywords
lewy
alpha
compound
limited
synuclein aggregates
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
US16/084,223
Other languages
English (en)
Inventor
Jérôme MOLETTE
Emanuele Gabellieri
Vincent Darmency
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.)
AC Immune SA
Original Assignee
AC Immune SA
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 AC Immune SA filed Critical AC Immune SA
Publication of US20190071450A1 publication Critical patent/US20190071450A1/en
Assigned to AC IMMUNE SA reassignment AC IMMUNE SA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DARMENCY, Vincent, GABELLIERI, EMANUELE, Molette, Jérôme
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4365Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system having sulfur as a ring hetero atom, e.g. ticlopidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/0429Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K51/0431Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0455Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention relates to novel compounds that can be employed in the diagnosis, monitoring of disease progression or monitoring of drug activity, of a group of disorders and abnormalities associated with alpha-synuclein ( ⁇ -synuclein, A-synuclein, aSynuclein, A-syn, ⁇ -syn, aSyn) aggregates including, but not limited to, Lewy bodies and/or Lewy neurites, such as Parkinson's disease.
  • the instant compounds are particularly useful in determining a predisposition to such a disorder, monitoring residual disorder, or predicting the responsiveness of a patient who is suffering from such a disorder to the treatment with a certain medicament.
  • the present compounds can also be used to treat, alleviate or prevent a disorder or abnormality associated with alpha-synuclein aggregates.
  • amyloid beta amyloid beta
  • Amyloid-like proteins that form mainly intracellular aggregates, include, but are not limited to tau, alpha-synuclein, and huntingtin (htt).
  • Diseases involving alpha-synuclein aggregates are generally listed as synucleinopathies (or ⁇ -synucleinopathies) and this includes, but is not limited to, Parkinson's disease (PD).
  • PD Parkinson's disease
  • Synucleinopathies include Parkinson's disease (sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia), dementia with Lewy bodies (“pure” Lewy body dementia), sporadic Alzheimer's disease, familial Alzheimer's disease with APP mutations, familial Alzheimer's disease with PS-1, PS-2 or other mutations, familial British dementia, Lewy body variant of Alzheimer's disease and normal aging (Down syndrome).
  • Parkinson's disease sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia
  • dementia with Lewy bodies (“pure” Lewy body dementia)
  • sporadic Alzheimer's disease familial Alzheimer's disease with APP mutations
  • Synucleinopathies with neuronal and glial aggregates of alpha synuclein include multiple system atrophy (Shy-Drager syndrome, striatonigral degeneration and olivopontocerebellar atrophy).
  • Other diseases that may have alpha-synuclein-immunoreactive lesions include traumatic brain injury, chronic traumatic encephalopathy, tauopathies (Pick's disease, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration and Niemann-Pick type C1 disease), motor neuron disease, amyotrophic lateral sclerosis (sporadic, familial and ALS-dementia complex of Guam), neuroaxonal dystrophy, neurodegeneration with brain iron accumulation type 1 (Hallervorden-Spatz syndrome), prion diseases, ataxia telangiectatica, Meige's syndrome, subacute sclerosing panencephalitis, Gaucher disease as well as other lysosomal storage disorders (
  • Alpha-synuclein is a 140 amino acid natively unfolded protein (Iwai et al., Biochemistry 1995, 34(32), 10139-10145).
  • the sequence of alpha-synuclein can be divided into three main domains: 1) the N-terminal region comprising of residues 1-60, which contains the 11-mer amphipatic imperfect repeat residues with highly conserved hexamer (KTKEGV).
  • This region has been implicated in regulating alpha-synuclein binding to membranes and its internalization; 2) the hydrophobic Non Amyloid beta Component (NAC) domain spanning residues 61-95; which is essential for alpha-synuclein fibrillization; and 3) the C-terminal region spanning residues 96-140 which is highly acidic and proline-rich, has no distinct structural propensity.
  • NAC Non Amyloid beta Component
  • Alpha-synuclein has been shown to undergo several post translational modifications, including truncations, phosphorylation, ubiquitination, oxidation and/or transglutaminase covalent cross linking (Fujiwara et al., Nat Cell Biol 2002, 4(2); 160-164; Hasegawa et al., J Biol Chem 2002, 277(50), 49071-49076; Li et al., Proc Natl Acad Sci USA 2005, 102(6), 2162-2167; Oueslati et al, Prog Brain Res 2010, 183, 115-145; Schmid et al., J Biol Chem 2009, 284(19), 13128-13142). Interestingly, the majority of these modifications involve residues within the C-terminal region.
  • Tyr-125 residues can be phosphorylated by two Src family protein tyrosine kinases, c-Src and Fyn (Ellis et al., J Biol Chem 2001, 276(6), 3879-3884; Nakamura et al., Biochem Biophys Res Commun 2001, 280(4), 1085-1092). Phosphorylation by Src family kinases does not suppress or enhance the tendency of alpha-synuclein to polymerize.
  • Alpha-synuclein has proved to be an outstanding substrate for protein tyrosine kinase p72 syk (Syk) in vitro; once it is extensively Tyr-phosphorylated by Syk or tyrosine kinases with similar specificity, it loses the ability to form oligomers, suggesting a putative anti-neurodegenerative role for these tyrosine kinases (Negro et al., FASEB J 2002, 16(2), 210-212).
  • Alpha-synuclein can be Ser-phosphorylated by protein kinases CKI and CKII (Okochi et al., J Biol Chem 2000, 275(1), 390-397).
  • Ser-129 is also phosphorylated by G-protein-coupled receptor protein kinases (Pronin et al., J Biol Chem 2000, 275(34), 26515-26522). Extensive and selective phosphorylation of alpha-synuclein at Ser-129 is evident in synucleinopathy lesions, including Lewy bodies (Fujiwara et al., Nat Cell Biol 2002, 4(2); 160-164).
  • Alpha-synuclein and its mutated forms have a random coil conformation and do not form significant secondary structures in aqueous solution at low concentrations; however, at higher concentrations they are prone to self-aggregate, producing amyloid fibrils (Wood et al., J Biol Chem 1999, 274(28), 19509-19512).
  • Several differences in the aggregation behavior of the PD-linked mutants and the wild-type protein have been documented.
  • Monomeric alpha-synuclein aggregates in vitro form stable fibrils via a metastable oligomeric (i.e., protofibril) state (Volles et al., Biochemistry 2002, 41(14), 4595-4602).
  • Parkinson's disease is the most common neurodegenerative motor disorder. PD is mainly an idiopathic disease, although in at least 5% of the PD patients the pathology is linked to mutations in one or several specific genes (Lesage et al., Hum. Mol. Genet., 2009, 18, R48-59). The pathogenesis of PD remains elusive, however, growing evidence suggests a role for the pathogenic folding of the alpha-synuclein protein that leads to the formation of amyloid-like fibrils.
  • Alpha-synuclein is a natively unfolded presynaptic protein that can misfold and aggregate into larger oligomeric and fibrillar forms which are linked to the pathogenesis of PD.
  • Recent studies have implicated small soluble oligomeric and protofibrillar forms of alpha-synuclein as the most neurotoxic species (Lashuel et al., J. Mol.
  • the diagnosis of Parkinson's disease is largely clinical and depends on the presence of a specific set of symptoms and signs (the initial core feature being bradykinesia, rigidity, rest tremor and postural instability), the absence of atypical features, a slowly progressive course, and a response to drug therapy.
  • the diagnosis requires clinical skills but is open to a degree of subjectivity and error, as several other degenerative and non-degenerative diseases can mimic PD (MSA, progressive supranuclear palsy (PSP), AD, essential tremor, dystonic tremor), (Guideline No. 113: Diagnosis and pharmacological management of Parkinson's disease, January 2010. SIGN).
  • the final confirmation of the diagnosis is made by post-mortem neuropathological analysis.
  • CT computed tomography
  • MRI magnetic resonance imaging
  • Dopaminergic function in the basal ganglia can be measured with different PET and SPECT radiotracers.
  • Examples are ioflupane ( 123 I) (trade name DaTSCAN) and iometopane (Dopascan) for SPECT or fluorodeoxyglucose ( 18 F) and DTBZ for PET.
  • a pattern of reduced dopaminergic activity in the basal ganglia can aid in diagnosing PD (Brooks, J. Nucl. Med., 2010, 51, 596-609; Redmond, Neuroscientist, 2002, 8, 457-88; Wood, Nat. Rev. Neurol., 2014, 10, 305).
  • biomarkers that have been investigated in different body fluids (cerebrospinal fluid (CSF), plasma, saliva) include alpha-synuclein levels but also DJ-1, Tau and Abeta, as well as neurofilaments proteins, interleukins, osteopontin and hypocrontin (Schapira Curr Opin Neurol 2013; 26(4):395-400), but so far none of these biomarkers alone or in combination can be used as determinant diagnostic test. To our knowledge none approved alpha-synuclein agent is currently on the market or available for clinical despite a crucial needs for Parkinson's disease research and drug development (Eberling et al., J Parkinsons Dis. 2013; 3(4):565-7).
  • alpha-synuclein deposition in the brain would be a huge achievement for Parkinson's disease research and drug development.
  • the accumulation of aggregated alpha-Synuclein in the brain is a pathological hallmark of PD and a priority target for drug development given its hypothesized contribution to neurodegeneration.
  • In vivo imaging of alpha-synuclein pathology could be useful as a biomarker of the presence of the disease and disease progression and as a pharmacodynamics tool for drug development.
  • the development of an alpha-Synuclein PET imaging agent is considered as very important for the diagnosis and monitoring the effects of therapeutics targeting alpha-synuclein (Eberling, Dave and Frasier, J.
  • alpha-synuclein imaging compounds should bind with high affinity and selectivity to their target.
  • imaging compounds need to penetrate the blood brain barrier and pass into the relevant regions of the brain.
  • cell permeability is a further requirement of imaging compounds.
  • WO 2011/128455 refers to specific compounds which are suitable for treating disorders associated with amyloid proteins or amyloid-like proteins.
  • US 2012/0302755 relates to certain imaging agents for detecting neurological dysfunction. Further compounds for the diagnosis of neurodegenerative disorders on the olfactory epithelium are discussed in WO 2012/037928.
  • WO 2010/063701 refers to a certain in vivo imaging agent for use in a method to determine the presence of, or susceptibility to, Parkinson's disease, wherein the in vivo imaging agent comprises an ⁇ -synuclein binder labelled with an in vivo imaging moiety, and wherein the in vivo imaging agent binds to ⁇ -synuclein with a binding affinity.
  • US 2014/0142089 relates to a method for preventing or treating a degenerative brain disease, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising a specific compound, a pharmaceutically acceptable salt, an isomer, a solvate, a hydrate, and a combination thereof.
  • WO 2009/155017 describes aryl or heteroaryl substituted azabenzoxazole derivatives, which are stated to be useful as tracers in positron emission tomography (PET) imaging to study amyloid deposits in brain in vivo to allow diagnosis of Alzheimer's disease.
  • PET positron emission tomography
  • WO 2016/033445 refers to specific compound for imaging huntingtin protein.
  • the compounds should be suitable for determining a predisposition to such a disorder, monitoring residual disorder, or predicting the responsiveness of a patient who is suffering from such a disorder to the treatment with a certain medicament.
  • the compounds of formulae (I) and (II) display high binding affinity to different alpha-synuclein aggregates in human tissues. Moreover, the compounds of formulae (I) and (II) display high selectivity for aSyn over A ⁇ and tau pathological deposits enabling the differentiation of PD from other proteinopathies that share common clinical and pathological features.
  • these compounds display properties such as appropriate lipophilicity and molecular weight, brain uptake and pharmacokinetics, cell permeability, solubility, and autofluorescence in order to be successful imaging probes for detection and quantification of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites load in vivo, ex vivo and in vitro.
  • the present invention discloses novel compounds of formulae (I) and (II) having enhanced binding properties to alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the compounds of this invention may be labeled (e.g., radiolabeled), so that they may be used for ex vivo and in vivo imaging to detect alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the present invention provides methods for the detection of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites, ex vivo using a compound of formulae (I) and (II) or a pharmaceutical composition thereof.
  • the present invention provides compounds of formulae (I) and (II) for use as diagnostic imaging agents, particularly for presymptomatic detection of Parkinson's disease and/or other ⁇ -synucleinopathies, e.g., using positron emission tomography (PET).
  • PET positron emission tomography
  • the invention would serve as a biomarker for monitoring of topographic progression of pathology, leading to improvement of clinical diagnosis and clinical study design.
  • the present invention further provides a pharmaceutical composition comprising a compound of formula (I) or (II) and a pharmaceutically acceptable carrier or excipient.
  • R e is independently selected from the group consisting of hydrogen, —(CH 2 CH 2 —O) n —R f , —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R d , alkyl, carbocyclyl and heterocyclyl, wherein alkyl, carbocyclyl and heterocyclyl can be optionally substituted,
  • alkyl can be attached at any available position
  • LG is a leaving group
  • alkyl can be attached at any available position, and wherein
  • LG is a leaving group
  • Alkyl refers to a saturated straight or branched organic moiety consisting of carbon and hydrogen atoms.
  • suitable alkyl groups have 1 to 6 carbon atoms, preferably 1 to 4 carbon atoms, and include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl and isobutyl.
  • Carbocyclyl refers to a cyclic organic moiety consisting of carbon and hydrogen atoms. Examples of suitable carbocyclyl groups have 3 to 10 carbon atoms, preferably 3, 4, 5 or 6 carbon atoms. The carbocyclyl group can be unsaturated or saturated. The term “carbocyclyl” also covers an aromatic cyclic organic moiety (aryl group) consisting of carbon and hydrogen atoms. Examples of the carbocyclyl group include cyclopentyl, cyclohexyl and phenyl.
  • Heterocyclyl refers to a carbocyclyl group as defined above in which at least one of the carbon atoms has been replaced by a heteroatom which is, e.g., selected from N, O or S, or heteroatom (e.g., N, O and/or S)-containing moiety.
  • the heterocyclyl group can be unsaturated or saturated. It covers both heteroalkyl groups and heteroaryl groups.
  • the heterocyclyl can also be annelated, connected in a bridged manner or connected in a spiro manner such as 6-membered bicyclic rings, 7-membered bicyclic rings, 8-membered bicyclic rings, 6-membered spirocyclic rings, 7-membered spirocyclic rings or 8-membered spirocyclic rings.
  • Examples include azetidine, pyrrolidine, pyrrole, tetrahydrofuran, furan, thiolane, thiophene, imidazolidine, pyrazolidine, imidazole, pyrazole, oxazolidine, isoxazolidine, oxazole, isoxazole, thiazolidine, isothiazolidine, thiazole, isothiazole, dioxolane, dithiolane, triazole, furazan, oxadiazoles, thiadiazole, dithiazole, tetrazole, piperidine, oxane, thiane, pyridine, pyran, thiopyran, piperazine, diazine (including pyrazine and pyrimidine), morpholine, oxazine, thiomorpholine, thiazine, dioxane, dioxine, dithiane, dithiine, tri
  • heterocyclyl groups examples include azetidine, morpholine, piperazine, pyrrolidine, tetrahydrofuran, piperidine, azaspiro[3.3]heptane, etc.
  • heteroaryl groups examples include pyridine, pyrazole, etc.
  • alkenyl refers to an organic moiety consisting of carbon and hydrogen atoms which includes at least one double bond.
  • suitable alkenyl groups have 2 to 6 carbon atoms, preferably 2 to 4 carbon atoms, and include propenyl and butenyl.
  • Alkynyl refers to an organic moiety consisting of carbon and hydrogen atoms which includes at least one triple bond.
  • suitable alkynyl groups have 2 to 6 carbon atoms, preferably 2 to 4 carbon atoms, and include propinyl and butinyl.
  • Aryl refers to homocyclic aromatic organic moieties containing 1 or 2 rings consisting of carbon and hydrogen atoms which preferably have 6 to 12 carbon atoms, more preferably 5 or 6 carbon atoms. Examples are, but not limited to, phenyl, biphenyl, and naphthyl.
  • Heteroaryl refers to an aryl group as defined above in which at least one of the carbon atoms has been replaced by a heteroatom which is, e.g., selected from N, O or S, or heteroatom (e.g., N, O and/or S)-containing moiety.
  • heteroatom e.g., selected from N, O or S
  • heteroatom e.g., N, O and/or S
  • Examples of possible heteroaryl groups include pyridine, etc.
  • Hal or “halogen” refers to F, Cl, Br, and I. With respect to diagnostic and pharmaceutical applications, F (e.g., 19 F and 18 F) is particularly preferred.
  • Carbocyclylalkyl refers to a group carbocyclyl-alkyl-.
  • Heterocyclylalkyl refers to a group heterocyclyl-alkyl-.
  • “5- to 8-Membered ring system containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties” refers to ring system having 5 to 8 carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties.
  • the term is also meant to include monocyclic, bicyclic, and polycyclic versions thereof. If more than one ring is present, the rings can be annelated, connected in a bridged manner or connected in a spiro manner.
  • the ring(s) can be either carbocyclic or heterocyclic and can be saturated, unsaturated or aromatic.
  • heterocyclic groups include, but are not restricted to, azetidine (azacyclobutane), pyrrolidine (azacyclopentane), pyrrole, imidazolidine, pyrazolidine, imidazole, pyrazole, oxazolidine, isoxazolidine, oxazole, isoxazole, thiazolidine, isothiazolidine, thiazole, isothiazole, triazole, furazan, oxadiazoles, thiadiazole, dithiazole, tetrazole, imidazoline, piperidine (azacyclohexane), pyridine, piperazine, pyrrolidine, diazine (including pyrazine and pyrimidine), morpholine, thiomorpholine, thiazine, triazine, tetrazine
  • the 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties is selected from, imidazoline, dioxolane, piperidine, piperazine, pyrrolidine, tetrahydrofuran, dioxane, phenyl, pyridine, thiazole, diazines (including pyrazine and pyrimidine), and oxadiazoles, more preferably imidazoline, dioxolane, piperidine, piperazine, pyrrolidine, phenyl and pyridine.
  • the 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties can be attached at any available position.
  • the “5- to 8-membered ring system containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties” is a “5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties”, more preferably it is a 4-, 5- or 6-membered (preferably saturated) ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N.
  • Specific examples are the 5- or 6-membered rings containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N given in the above list.
  • R A , R B , R D or R E can optionally be taken together and can form a 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties.
  • the 5- to 8-membered ring can be any 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties. Examples thereof can be found in the list of examples given for the “5- to 8-membered ring system containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties”, as well as in the lists of examples of the carbocyclyl, aryl, and heterocyclyl groups.
  • the ring formed by two adjacent groups R A , R B , R D or R E , respectively, is preferably a 5- or 6-membered, saturated or unsaturated ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties.
  • Specific examples of the 5- or 6-membered, saturated or unsaturated rings containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties are given in the above list.
  • the heteroatom is preferably N and/or 0.
  • the ring preferably contains 0, 1, 2 or 3 heteroatom(s).
  • the ring preferably contains 0 or 1 heteroatom (e.g., N, O and/or S)-containing moieties.
  • Heteroatom-containing moieties are moieties which contain e.g., N, O and/or S. Examples of such moieties include —C(O)—, —C(O)O—, —C(O)N(R 50 )— and —N(R 50 )— in which R 50 is, for each occurrence, independently selected from the group consisting of H or C 1-4 alkyl, wherein C 1-4 alkyl can be optionally substituted.
  • LG leaving group
  • LG is any leaving group and means an atom or group of atoms that can be replaced by another atom or group of atoms. Examples are given e.g. in Synthesis (1982), p. 85-125, table 2, Carey and Sundberg, Organische Synthese, (1995), page 279-281, table 5.8; or Netscher, Recent Res. Dev. Org. Chem., 2003, 7, 71-83, scheme 1, 2, 10 and 15 and others). (Coenen, Fluorine-18 Labeling Methods: Features and Possibilities of Basic Reactions, (2006), in: Schubiger P.
  • the “leaving group” is selected from nitro, halogen, trimethylammonium, C 1-4 alkyl sulfonate and C 6-10 aryl sulfonate.
  • a group is defined as being “optionally substituted” (unless defined otherwise), as chemically appropriate, it can have one or more substituents selected from -Hal, —CN, —OH, —(O—CH 2 CH 2 ) n —R, —(CH 2 CH 2 —O) n —R*, —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R (with R ⁇ H or Hal and R* ⁇ H, (CH 2 CH 2 ) n Hal, CHaI 3 or CH 3 ), —C 1-6 alkyl, —C 1-6 alkoxy, —SO 2 -alkyl, —NH 2 , —NH(C 1-6 alkyl) or —N(C 1-6 alkyl) 2 , preferably -Hal, —CN, —OH, —(O—CH 2 CH 2 ) n —R, —(CH 2 CH 2 —O) n —R*, or —(CH
  • Compounds of the present invention having one or more optically active carbons can exist as racemates and racemic mixtures, stereoisomers (including diastereomeric mixtures and individual diastereomers, enantiomeric mixtures and single enantiomers, mixtures of conformers and single conformers), tautomers, atropisomers, and rotamers. All isomeric forms are included in the present invention.
  • Compounds described in this invention containing olefinic double bonds include E and Z geometric isomers. Also included in this invention are all salt forms, polymorphs, hydrates and solvates.
  • polymorphs refers to the various crystalline structures of the compounds of the present invention. This may include, but is not limited to, crystal morphologies (and amorphous materials) and all crystal lattice forms. Salts of the present invention can be crystalline and may exist as more than one polymorph. Solvates, hydrates as well as anhydrous forms of the salt are also encompassed by the invention.
  • the solvent included in the solvates is not particularly limited and can be any pharmaceutically acceptable solvent. Examples include water and C 1-4 alcohols (such as methanol or ethanol).
  • “Pharmaceutically acceptable salts” are defined as derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as, but not limited to, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as, but not limited to, acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as, but not limited to, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as, but not limited to
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two.
  • Organic solvents include, but are not limited to, nonaqueous media like ethers, ethyl acetate, ethanol, isopropanol, or acetonitrile. Lists of suitable salts can be found in Remington's Pharmaceutical Sciences, 18 th ed., Mack Publishing Company, Easton, Pa., 1990, p. 1445, the disclosure of which is hereby incorporated by reference.
  • “Pharmaceutically acceptable” is defined as those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
  • the compounds of the present invention can also be provided in the form of a prodrug, namely a compound which is metabolized in vivo to the active metabolite.
  • the patients or subjects in the present invention are typically animals, particularly mammals, more particularly humans.
  • Alpha-synuclein aggregates are multimeric beta-sheet rich assemblies of alpha-synuclein monomers that can form either soluble oligomers or soluble/insoluble protofibrils or mature fibrils which coalesce into intracellular deposits detected as a range of Lewy pathologies in Parkinson's disease and other synucleinopathies.
  • Alpha-synuclein aggregates that are composing Lewy pathologies can be detected as having the following morphologies: Lewy bodies, Lewy neurites, premature Lewy bodies or pale bodies, perikaryal deposits with diffuse, granular, punctate or pleomorphic patterns.
  • alpha-synuclein aggregates are the major component of intracellular fibrillary inclusions detected in oligodendrocytes (also referred to as glial cytoplasmic inclusions) and in neuronal somata, axons and nuclei (referred to as neuronal cytoplasmic inclusions) that are the histological hallmarks of multiple system atrophy.
  • Alpha-synuclein aggregates in Lewy pathologies often display substantial increase in post-translational modifications such as phosphorylation, ubiquitination, nitration, and truncation.
  • Lewy bodies are abnormal aggregates of protein that develop inside nerve cells in Parkinson's disease (PD), Lewy body dementia and other synucleinopathies. Lewy bodies appear as spherical masses that displace other cell components. Morphologically, Lewy bodies can be classified as being brainstem or cortical type. Classic brainstem Lewy bodies are eosinophilic cytoplasmic inclusions consisting of a dense core surrounded by a halo of 5-10-nm-wide radiating fibrils, the primary structural component of which is alpha-synuclein; cortical Lewy bodies differ by lacking a halo. The presence of Lewy bodies is a hallmark of Parkinson's disease.
  • Lewy neurites are abnormal neuronal processes in diseased neurons, containing granular material, abnormal ⁇ -synuclein filaments similar to those found in Lewy bodies, dot-like, varicose structures and axonal spheroids. Like Lewy bodies, Lewy neurites are a feature of ⁇ -synucleinopathies such as dementia with Lewy bodies, Parkinson's disease, and multiple system atrophy.
  • the present invention relates to a compound of formula (I):
  • Preferred compounds are also compounds of the formula (Ib) or (Ic):
  • V is selected from the group consisting of S, NR a and CR b R b .
  • V is S.
  • Z is selected from the group consisting of N and CR c . In one embodiment Z is N. In another embodiment, Z is CR c .
  • W is selected from the group consisting of N and CR c or W is C if W is attached to U.
  • W 1 is selected from the group consisting of N and CR c or W 1 is C if W 1 is attached to U.
  • X is selected from the group consisting of N and CR c or X is C if X is attached to U.
  • Y is selected from the group consisting of N and CR c or Y is C if Y is attached to U.
  • U is selected from the group consisting of —NR a —, —CH ⁇ CH—, —C ⁇ C— and a bond.
  • At least one of Z, X, W 1 , W and Y is N. More preferably, V is S and at least one of Z, X, W 1 , W and Y is N.
  • R a is independently selected from the group consisting of hydrogen, alkyl, and haloalkyl.
  • R a is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R b is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, and halogen.
  • R b is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R c is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, and halogen.
  • R c is preferably hydrogen.
  • R d is independently selected from the group consisting of -halogen, —OH, —O-alkyl and -hydrogen.
  • R e is independently selected from the group consisting of hydrogen, —(CH 2 CH 2 —O) n —R f , —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R d , alkyl, carbocyclyl and heterocyclyl, wherein alkyl, carbocyclyl and heterocyclyl can be optionally substituted.
  • R e is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R f is independently selected from the group consisting of hydrogen, and alkyl, wherein alkyl can be optionally substituted.
  • R f is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R A is independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R A is preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl.
  • R A is more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, and heterocyclyl.
  • R A is selected from halogen, —O(CH 2 ) 2 F, —O(CH 2 ) 2 OH, optionally substituted morpholino, optionally substituted-pyrrolidine (such as F-pyrrolidine), and optionally substituted-piperidine (such as F-piperidine), more preferably R A is optionally substituted-pyrrolidine (such as F-pyrrolidine) or optionally substituted-piperidine (such as F-piperidine). In a preferred embodiment, R A is an optionally substituted-pyrrolidine (such as F-pyrrolidine).
  • the alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl can be optionally substituted.
  • Examples of possible substituents of the alkyl group include -Hal, —CN, —OH, —O-alkyl, —CF 3 , and —OCF 3 .
  • Examples of possible substituents of the carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl include -Hal, —CN, —OH, —O-alkyl, —CF 3 , —OCF 3 , and alkyl.
  • R A can optionally be taken together and can form a 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties and wherein the 5- to 8-membered ring may be substituted.
  • the 5- to 8-membered ring include —O—CH 2 —CH 2 —O— and —O—CH 2 —O—.
  • R B is independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R B is preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CON R 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl.
  • R B is more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, and heterocyclyl.
  • R B is selected from halogen, —O—R 10 , —O(CH 2 ) 2 F, —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , optionally substituted piperidine, optionally substituted pyrrolidone, optionally substituted tetrahydropyrane, and optionally substituted azaspiro[3.3]heptane (with R 10 and R 11 being independently hydrogen or alkyl).
  • R B is an optionally substituted-pyrrolidine and an optionally substituted-piperidine.
  • the alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl can be optionally substituted.
  • Examples of possible substituents of the alkyl group include -Hal, —CN, —OH, —O-alkyl, —CF 3 , and —OCF 3 .
  • Examples of possible substituents of the carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl include -Hal, —CN, —OH, —O-alkyl, —CF 3 , —OCF 3 , and alkyl.
  • R B can optionally be taken together and can form a 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties and wherein the 5- to 8-membered ring may be substituted.
  • the 5- to 8-membered ring include —O—CH 2 —CH 2 —O— and —O—CH 2 —O—.
  • R 10 is independently selected from the group consisting of hydrogen, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl, wherein alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl can be optionally substituted.
  • R 10 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • R 11 is independently selected from the group consisting of: hydrogen, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl, wherein alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl can be optionally substituted.
  • R 11 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • R 14 is independently selected from the group consisting of hydrogen, —(CH 2 CH 2 —O) n —R f , —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R d , alkyl, carbocyclyl and heterocyclyl, wherein alkyl, carbocyclyl and heterocyclyl can be optionally substituted.
  • R 14 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • n is independently 1 to 4; preferably for each occurrence n is 1 or 2.
  • n is independently 1 to 4, preferably for each occurrence, m is 2 or 3.
  • Preferred compounds of formula (I) are the compounds given in the example section of the present application. Particularly preferred compounds are
  • the present invention refers to a compound of formula (II):
  • Another preferred embodiment of the compound of formula (II) is selected from:
  • alkyl can be attached at any available position.
  • alkyl can be attached at any available position.
  • substituents R E can be optionally substituted by one or more substituents R E .
  • preferred substituted groups include an optionally substituted heterocyclyl, particularly an optionally substituted-pyrrolidine.
  • V 2 is selected from the group consisting of S, NR a and CR b R b .
  • V 2 is S.
  • Z 2 is selected from the group consisting of N and CR c . In one embodiment Z 2 is N. In another embodiment, Z 2 is CR c .
  • R a is independently selected from the group consisting of hydrogen, alkyl, and haloalkyl.
  • R a is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R b is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, and halogen.
  • R b is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R c is independently selected from the group consisting of hydrogen, alkyl, haloalkyl, and halogen.
  • R c is preferably hydrogen.
  • R d is independently selected from the group consisting of halogen, —OH, —O-alkyl and hydrogen.
  • R e is independently selected from the group consisting of hydrogen, —(CH 2 CH 2 —O) n —R f , —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R d , alkyl, carbocyclyl and heterocyclyl, wherein alkyl, carbocyclyl and heterocyclyl can be optionally substituted.
  • R e is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R f is independently selected from the group consisting of hydrogen, and alkyl, wherein alkyl can be optionally substituted.
  • R f is preferably independently selected from the group consisting of hydrogen, and alkyl.
  • R D is independently selected from the group consisting of halogen, CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R D is preferably independently selected from the group consisting of halogen, CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R D is more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl.
  • R D is even more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, and heterocyclyl.
  • R D is alkyl, for each occurrence, R D is preferably independently selected from the group consisting of halogen, CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R D is more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl.
  • R D is even more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , and heterocyclyl.
  • the alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl can be optionally substituted.
  • Examples of possible substituents of the alkyl group include -Hal, —CN, —OH, —O-alkyl, —CF 3 , and —OCF 3 .
  • Examples of possible substituents of the carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl include -Hal, —CN, —OH, —O-alkyl, —CF 3 , —OCF 3 , and alkyl.
  • R D can optionally be taken together and can form a 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties and wherein the 5- to 8-membered ring may be substituted.
  • R E is independently selected from the group consisting of halogen, CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , ⁇ O, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl.
  • R E is preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —CONR 10 R 11 , —N(R 10 )—C(O)—R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl.
  • R E is more preferably independently selected from the group consisting of halogen, —CN, —O—R 10 , —NR 10 R 11 , —N(R 10 )—C(O)—O—R 11 , —(O—CH 2 CH 2 ) n —R d , alkyl, and heterocyclyl. Even more preferably, R E is halogen and/or F-pyrrolidine.
  • the alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl can be optionally substituted.
  • Examples of possible substituents of the alkyl group include -Hal, —CN, —OH, —O-alkyl, —CF 3 , and —OCF 3 .
  • Examples of possible substituents of the carbocyclyl, carbocyclylalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, and alkynyl include -Hal, —CN, —OH, —O-alkyl, —CF 3 , —OCF 3 , and alkyl.
  • R E can optionally be taken together and can form a 5- to 8-membered ring containing carbon atoms and optionally one or more heteroatoms selected from O, S, or N or optionally one or more heteroatom (e.g., N, O and/or S)-containing moieties and wherein the 5- to 8-membered ring may be substituted.
  • R 10 is independently selected from the group consisting of: hydrogen, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl, wherein alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl can be optionally substituted.
  • R 10 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • R 11 is independently selected from the group consisting of: hydrogen, alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl, wherein alkyl, carbocyclyl, carbocyclylalkyl, heterocyclyl, and heterocyclylalkyl can be optionally substituted.
  • R 11 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • R 14 is independently selected from the group consisting of hydrogen, —(CH 2 CH 2 —O) n —R f , —(CH 2 CH 2 —O) n —(CH 2 CH 2 )—R d , alkyl, carbocyclyl and heterocyclyl, wherein alkyl, carbocyclyl and heterocyclyl can be optionally substituted.
  • R 14 is preferably independently selected from the group consisting of hydrogen, and alkyl. Examples of the optional substituents include —OH, —O-alkyl and Hal.
  • n is independently 1 to 4; preferably for each occurrence n is 1 or 2.
  • n is independently 1 to 4, preferably for each occurrence, m is 2 or 3.
  • Preferred compounds of formula (II) are the compounds given in the example section of the present application. Particularly preferred compounds are
  • the compounds of the present invention can be detectably labeled.
  • the type of the label is not specifically limited and will depend on the detection method chosen.
  • Examples of possible labels include isotopes such as radionuclides, positron emitters, gamma emitters, as well as fluorescent, luminescent and chromogenic labels.
  • the detectably labeled compounds of the present invention which include a radioisotope, a positron emitter, or a gamma emitter, it is to be understood that the radioisotope, positron emitter, or gamma emitter is to be present in an amount which is not identical to the natural amount of the respective radioisotope, positron emitter, or gamma emitter.
  • the employed amount should allow detection thereof by the chosen detection method.
  • Suitable isotopes such as radionuclides, positron emitters and gamma emitters include 2 H, 3 H, 18 F, 123 I, 124 I, 125 I, 131 I, 11 C, 13 N, 15 O, and 77 Br, preferably 2 H, 3 H, 11 C, 13 N, 15 O, and 18 F, more preferably 2 H, 3 H and 18 F, even more preferably 18 F.
  • 18 F-labeled compounds are particularly suitable for imaging applications such as PET.
  • the corresponding compounds which include fluorine having a natural 19 F isotope are also of particular interest as they can be used as analytical standards and references during manufacturing, quality control, release and clinical use of their 18 F-analogs.
  • substitution with isotopes such as deuterium, i.e., 2 H may afford certain diagnostic and therapeutic advantages resulting from greater metabolic stability by reducing for example defluorination, increased in vivo half-life or reduced dosage requirements, while keeping or improving the original compound efficacy.
  • Isotopic variations of the compounds of the invention can generally be prepared by conventional procedures such as by the illustrative methods or by the preparations described in the Examples and Preparative Examples hereafter using appropriate isotopic variations of suitable reagents, commercially available or prepared by known synthetic techniques.
  • Radionuclides, positron emitters and gamma emitters can be included into the compounds of the present invention by methods which are usual in the field of organic synthesis. Typically, they will be introduced by using a correspondingly labeled starting material when the desired compound of the present invention is prepared. Illustrative methods of introducing detectable labels are described, for instance, in US 2012/0302755.
  • the position at which the detectable label is to be attached to the compounds of the present invention is not particularly limited.
  • the radionuclides, positron emitters and gamma emitters can be attached at any position where the corresponding non-emitting atom can also be attached.
  • 18 F can be attached at any position which is suitable for attaching F.
  • the other radionuclides, positron emitters and gamma emitters Due to the ease of synthesis, it is preferred to attach 18F, 123 I, 124 I, 125 I, 131 I and 77 Br as R A , R B , R D and R E or part of R A , R B , R D and R E .
  • 3 H is employed as a detectable label it is preferably attached in the form of —C( 3 H) 3 at any position at which a methyl group can be attached. Alternatively, 3 H per se can be attached at any available position. If 2 H is employed as a detectable label it is preferably attached in the form of —C( 2 H) 3 at any position at which a methyl group can be attached. Easily available positions include R e and R 14 , 11 C, 13 N, and 15 O can be incorporated into the compounds of the present invention at any position where C, N and O appear.
  • the compounds of the present invention are particularly suitable for imaging of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • compounds of the present invention are particularly suitable for binding to various types of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurite.
  • the compounds of the present invention are suitable for use in the diagnosis of disorders and abnormalities associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the compounds of the present invention are particularly suitable for positron emission tomography imaging of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • Diseases involving alpha-synuclein aggregates are generally listed as synucleinopathies (or ⁇ -synucleinopathies).
  • the compounds of the present invention are suitable for use in the diagnosis of disorders including, but not limited to, Parkinson's disease (sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia), dementia with Lewy bodies (“pure” Lewy body dementia), sporadic Parkinson's disease (sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia), dementia with Lewy bodies (“pure” Lewy body dementia), sporadic
  • Parkinson's disease sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia
  • dementia with Lewy bodies (“pure” Lewy body dementia)
  • Alzheimer's disease familial Alzheimer's disease with APP mutations, familial Alzheimer's disease with PS-1, PS-2 or other mutations, familial British dementia, Lewy body variant of Alzheimer's disease and normal aging (Down syndrome).
  • Synucleinopathies with neuronal and glial aggregates of alpha synuclein include multiple system atrophy (Shy-Drager syndrome, striatonigral degeneration and olivopontocerebellar atrophy).
  • alpha-synuclein-immunoreactive lesions include traumatic brain injury, chronic traumatic encephalopathy, tauopathies (Pick's disease, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration and Niemann-Pick type C1 disease), motor neuron disease, amyotrophic lateral sclerosis (sporadic, familial and ALS-dementia complex of Guam), neuroaxonal dystrophy, neurodegeneration with brain iron accumulation type 1 (Hallervorden-Spatz syndrome), prion diseases, ataxia telangiectatica, Meige's syndrome, subacute sclerosing panencephalitis, Gaucher disease as well as other lysosomal storage disorders (including Kufor-Rakeb syndrome and Sanfilippo syndrome) and rapid eye movement (REM) sleep behavior disorder.
  • traumatic brain injury chronic traumatic encephalopathy
  • tauopathies Pick's disease, frontotemporal dementia, progressive supranuclear pal
  • the compounds of the present invention are suitable for use in the diagnosis of Parkinson's disease (PD).
  • PD Parkinson's disease
  • a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites, such as Parkinson's disease, or a predisposition therefor in a subject, the method comprising:
  • the compounds of the present invention can be used for imaging of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites in any sample or a specific body part or body area of a patient which suspected to contain an alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the compounds of the present invention are able to pass the blood-brain barrier. Consequently, they are particularly suitable for imaging of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites in the brain, as well as in body fluids such as cerebrospinal fluid (CSF).
  • CSF cerebrospinal fluid
  • the compound of the present invention is preferably administered in a diagnostic composition comprising the compound of the invention.
  • a “diagnostic composition” is defined in the present invention as a composition comprising one or more compounds of the present invention in a form suitable for administration to a patient, e.g., a mammal such as a human, and which is suitable for use in the diagnosis of the specific disorder or anormality at issue.
  • a diagnostic composition further comprises a physiologically acceptable carrier, diluent, adjuvant or excipient.
  • Administration is preferably carried out as defined below. More preferably by injection of the composition as an aqueous solution.
  • Such a composition may optionally contain further ingredients such as buffers; pharmaceutically acceptable solubilisers (e.g., cyclodextrins or surfactants such as Pluronic, Tween or phospholipids); and pharmaceutically acceptable stabilisers or antioxidants (such as ascorbic acid, gentisic acid or para-aminobenzoic acid).
  • pharmaceutically acceptable solubilisers e.g., cyclodextrins or surfactants such as Pluronic, Tween or phospholipids
  • pharmaceutically acceptable stabilisers or antioxidants such as ascorbic acid, gentisic acid or para-aminobenzoic acid.
  • the invention also provides a diagnostic composition which comprises a diagnostically effective amount of a compound of the present invention in admixture with a pharmaceutically acceptable carrier, diluent, adjuvant or excipient.
  • compositions are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1975).
  • the pharmaceutical excipient can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the excipient must be acceptable in the sense of being not deleterious to the recipient thereof.
  • compositions of the present invention may comprise, for example, carriers, vehicles, diluents, solvents such as monohydric alcohols such as ethanol, isopropanol and polyhydric alcohols such as glycols and edible oils such as soybean oil, coconut oil, olive oil, safflower oil cottonseed oil, oily esters such as ethyl oleate, isopropyl myristate, binders, adjuvants, solubilizers, thickening agents, stabilizers, disintegrants, glidants, lubricating agents, buffering agents, emulsifiers, wetting agents, suspending agents, sweetening agents, colorants, flavors, coating agents, preservatives, antioxidants, processing agents, drug delivery modifiers and enhancers such as calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methylcellulose, sodium carboxy
  • the routes for administration (delivery) of the compounds of the invention include, but are not limited to, one or more of: oral (e. g. as a tablet, capsule, or as an ingestible solution), topical, mucosal (e. g. as a nasal spray or aerosol for inhalation), nasal, parenteral (e. g. by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic (including intravitreal or intracameral), transdermal, rectal, buccal, epidural and sublingual.
  • oral e. g. as a tablet, capsule, or as an ingestible solution
  • mucosal e. g. as a nasal spray or aerosol for inhalation
  • nasal parenteral (e. g. by an injectable form)
  • the compounds can be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • the compounds of the present invention are administered parenterally.
  • parenterally examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the compounds; and/or by using infusion techniques.
  • the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • the compounds of the present invention can be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray or nebulizer with the use of a suitable propellant, e. g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA134AT) or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA), carbon dioxide or other suitable gas.
  • a suitable propellant e. g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA134AT) or 1,1,1,2,
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurized container, pump, spray or nebulizer may contain a solution or suspension of the active compound, e. g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e. g. sorbitan trioleate.
  • a lubricant e. g. sorbitan trioleate.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds of the present invention can be administered in the form of a suppository or pessary, or it may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
  • the compounds may also be administered by the pulmonary or rectal routes. They may also be administered by the ocular route.
  • the compounds can be formulated as micronized suspensions in isotonic, pH was adjusted, sterile saline, or, preferably, as solutions in isotonic, pH was adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
  • the compounds of the present invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing diagnosis.
  • the dose could preferably lie in the range 0.001 ⁇ g/kg to 10 ⁇ g/kg, preferably 0.01 ⁇ g/kg to 1.0 ⁇ g/kg.
  • the dose will depend on the route of administration. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient as well as the severity of the disorder or anormality. The precise dose and route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • compositions of the invention can be produced in a manner known per se to the skilled person as described, for example, in Remington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1975).
  • the compounds of the present invention are useful as an in vitro analytical reference or an in vitro screening tool. They are also useful in in vivo diagnostic methods.
  • the compounds according to the present invention can also be provided in the form of a mixture comprising a compound according to the present invention and at least one compound selected from an imaging agent different from the compound according to the invention, a pharmaceutically acceptable carrier, a diluent and an excipient.
  • the imaging agent different from the compound according to the invention is preferably present in a diagnostically effective amount. More preferably the imaging agent different from the compound according to the invention is an abeta or tau imaging agent.
  • Diagnosis of a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites or of a predisposition to a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites in a patient may be achieved by detecting the specific binding of a compound according to the invention to the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or
  • Lewy neurites in a sample or in situ which includes:
  • the compound of the present invention can be brought into contact with the sample or the specific body part or body area suspected to contain the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites by a suitable method.
  • the compound of the present invention and a liquid sample can be simply mixed.
  • the compound of the present invention is typically administered to the patient by any suitable means.
  • routes of administration include, but are not limited to, one or more of: oral (e. g. as a tablet, capsule, or as an ingestible solution), topical, mucosal (e. g. as a nasal spray or aerosol for inhalation), nasal, parenteral (e. g.
  • an injectable form by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic (including intravitreal or intracameral), transdermal, rectal, buccal, epidural and sublingual.
  • parenteral administration can be preferred.
  • the compound of the present invention After the sample or a specific body part or body area has been brought into contact with the compound of the present invention, the compound is allowed to bind to the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the amount of time required for binding will depend on the type of test (e.g., in vitro or in vivo) and can be determined by a person skilled in the field by routine experiments.
  • the compound which has bound to the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites, can be subsequently detected by any appropriate method.
  • the specific method chosen will depend on the detectable label which has been chosen. Examples of possible methods include, but are not limited to, a fluorescence imaging technique or a nuclear imaging technique such as positron emission tomography (PET), single photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and contrast-enhanced magnetic resonance imaging (MRI). These have been described and enable visualization of amyloid biomarkers.
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • MRI magnetic resonance imaging
  • MRI contrast-enhanced magnetic resonance imaging
  • the fluorescence imaging technique and/or nuclear imaging technique can be employed for monitoring and/or visualizing the distribution of the detectably labeled compound within the sample or a specific body part or body area.
  • the presence or absence of the compound/protein aggregate complex is then optionally correlated with the presence or absence of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites in the sample or specific body part or area.
  • the amount of the compound/protein aggregate complex can be compared to a normal control value which has been determined in a sample or a specific body part or body area of a healthy subject, wherein an increase in the amount of the compound/protein aggregate complex compared to a normal control value may indicate that the patient is suffering from or is at risk of developing a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the present invention also relates to a method of determining the amount of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites in a tissue and/or a body fluid. This method comprises the steps of:
  • the sample can be tested for the presence of alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites with a compound of the present invention by bringing the sample into contact with a compound of the invention, allowing the compound of the invention to bind to the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites to form a compound/protein aggregate complex and detecting the formation of the compound/protein aggregate complex as explained above.
  • alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites
  • detecting the formation of the compound/protein aggregate complex as explained above.
  • Monitoring minimal residual disorder in a patient suffering from a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites who has been treated with a medicament with a compound according to the invention may be achieved by
  • Lewy bodies and/or Lewy neurites in the sample or specific body part or body area are Lewy bodies and/or Lewy neurites in the sample or specific body part or body area.
  • the method can further comprises steps (i) to (vi) before step (a):
  • the method can further comprise step (A) after step (d) or step (e):
  • steps (a) to (c) and optionally steps (d) and (e) of the method of monitoring minimal residual disorder can be repeated one or more times.
  • the amount of the compound/protein aggregate complex can be optionally compared at various points of time during the treatment, for instance, before and after onset of the treatment or at various points of time after the onset of the treatment.
  • a change, especially a decrease, in the amount of the compound/protein aggregate complex may indicate that the residual disorder is decreasing.
  • Predicting responsiveness of a patient suffering from a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites and being treated with a medicament can be achieved by
  • the method can further comprises steps (i) to (vi) before step (a):
  • the method can further comprise step (A) after step (d) or step (e):
  • steps (a) to (c) and optionally steps (d) and (e) of the method of predicting responsiveness can be repeated one or more times.
  • the amount of the compound/protein aggregate complex can be optionally compared at various points of time during the treatment, for instance, before and after onset of the treatment or at various points of time after the onset of the treatment.
  • a change, especially a decrease, in the amount of the compound/protein aggregate complex may indicate that the patient has a high potential of being responsive to the respective treatment.
  • the diagnostic composition can be used before, during and after, surgical procedures (e.g. deep brain stimulation (DBS)) and non-invasive brain stimulation (such as repetitive transcranial magnetic stimulation (rTMS)), for visualizing alpha-synuclein aggregates before, during and after such procedures.
  • surgical procedures e.g. deep brain stimulation (DBS)
  • non-invasive brain stimulation such as repetitive transcranial magnetic stimulation (rTMS)
  • rTMS repetitive transcranial magnetic stimulation
  • Surgical techniques including DBS, improve advanced symptoms of PD on top of the best currently used medical therapy.
  • rTMS has been closely examined as a possible treatment for PD (Ying-hui Chou et al. JAMA Neurol. 2015 Apr. 1; 72(4): 432-440).
  • the diagnostic composition can be used in a method of collecting data for monitoring residual disorder in a patient suffering from a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites who has been treated with a surgical procedure or non-invasive brain stimulation procedure, wherein the method comprises:
  • a compound according to the present invention can also be incorporated into a test kit for detecting alpha-synuclein protein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • the test kit typically comprises a container holding one or more compounds according to the present invention and instructions for using the compound for the purpose of binding to an alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites to form a compound/protein aggregate complex and detecting the formation of the compound/protein aggregate complex such that presence or absence of the compound/protein aggregate complex correlates with the presence or absence of the alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • test kit refers in general to any diagnostic kit known in the art. More specifically, the latter term refers to a diagnostic kit as described in Zrein et al., Clin. Diagn. Lab. Immunol., 1998, 5, 45-49.
  • the compounds of the present invention can also be employed in kits for the preparation of radiopharmaceutical preparations. Due to the radioactive decay, the radiopharmaceuticals are usually prepared immediately before use.
  • the kit comprises a precursor of the compound of the present invention and an agent which reacts with the precursor to introduce a radioactive label to the compound of the present invention.
  • the precursor of the compound of the present invention can, for example, be a compound having the formula (IIIa), (IIIb), (IVa) or (IVb).
  • the agent can be an agent which introduces a radioactive label such as 18 F.
  • the compounds of the present invention can be employed in treating, preventing or alleviating a disorder or abnormality associated with alpha-synuclein aggregates.
  • the compounds of the present invention are suitable for treating, preventing or alleviating a disorder or abnormality associated with alpha-synuclein aggregates including, but not limited to, Lewy bodies and/or Lewy neurites.
  • Diseases involving alpha-synuclein aggregates are generally listed as synucleinopathies (or ⁇ -synucleinopathies).
  • the compounds of the present invention are suitable for treating, preventing or alleviating disorders including, but not limited to, Parkinson's disease (sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia), dementia with Lewy bodies (“pure” Lewy body dementia), sporadic Alzheimer's disease, familial Alzheimer's disease with APP mutations, familial Alzheimer's disease with PS-1, PS-2 or other mutations, familial British dementia, Lewy body variant of Alzheimer's disease and normal aging (Down syndrome).
  • Parkinson's disease sporadic, familial with alpha-synuclein mutations, familial with mutations other than alpha-synuclein, pure autonomic failure and Lewy body dysphagia
  • dementia with Lewy bodies (“pure” Lewy body dementia)
  • sporadic Alzheimer's disease familial Alzheimer's disease with APP mutations
  • Synucleinopathies with neuronal and glial aggregates of alpha synuclein include multiple system atrophy (Shy-Drager syndrome, striatonigral degeneration and olivopontocerebellar atrophy).
  • Other diseases that may have alpha-synuclein-immunoreactive lesions include traumatic brain injury, chronic traumatic encephalopathy, tauopathies (Pick's disease, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration and Niemann-Pick type C1 disease), motor neuron disease, amyotrophic lateral sclerosis (sporadic, familial and ALS-dementia complex of Guam), neuroaxonal dystrophy, neurodegeneration with brain iron accumulation type 1 (Hallervorden-Spatz syndrome), prion diseases, ataxia telangiectatica, Meige's syndrome, subacute sclerosing panencephalitis, Gaucher disease as well as other lysosomal storage disorders (
  • the compounds of the present invention are suitable for treating, preventing or alleviating Parkinson's disease (PD).
  • PD Parkinson's disease
  • the compound of the present invention is preferably administered in a pharmaceutical composition comprising the compound of the invention.
  • a “pharmaceutical composition” is defined in the present invention as a composition comprising one or more compounds of the present invention in a form suitable for administration to a patient, e.g., a mammal such as a human, and which is suitable for treating, alleviating or preventing the specific disorder or anormality at issue.
  • a pharmaceutical composition further comprises a physiologically acceptable carrier, diluent, adjuvant or excipient.
  • the dose of the compound of the present invention will vary depending on the exact compound to be administered, the weight of the patient, and other variables as would be apparent to a physician skilled in the art.
  • the invention also provides a pharmaceutical composition which comprises a therapeutically effective amount of a compound of formulae (I) or (II) in admixture with a pharmaceutically acceptable carrier, diluent, adjuvant or excipient.
  • compositions are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1975).
  • the pharmaceutical excipient can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the excipient must be acceptable in the sense of being not deleterious to the recipient thereof.
  • compositions of the present invention may comprise, for example, carriers, vehicles, diluents, solvents such as monohydric alcohols such as ethanol, isopropanol and polyhydric alcohols such as glycols and edible oils such as soybean oil, coconut oil, olive oil, safflower oil cottonseed oil, oily esters such as ethyl oleate, isopropyl myristate, binders, adjuvants, solubilizers, thickening agents, stabilizers, disintegrants, glidants, lubricating agents, buffering agents, emulsifiers, wetting agents, suspending agents, sweetening agents, colorants, flavors, coating agents, preservatives, antioxidants, processing agents, drug delivery modifiers and enhancers such as calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methylcellulose, sodium carboxy
  • the routes for administration (delivery) of the compounds of the invention include, but are not limited to, one or more of: oral (e. g. as a tablet, capsule, or as an ingestible solution), topical, mucosal (e. g. as a nasal spray or aerosol for inhalation), nasal, parenteral (e. g. by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic (including intravitreal or intracameral), transdermal, rectal, buccal, epidural and sublingual.
  • oral e. g. as a tablet, capsule, or as an ingestible solution
  • mucosal e. g. as a nasal spray or aerosol for inhalation
  • nasal parenteral (e. g. by an injectable form)
  • the compounds can be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • the compounds of the present invention are administered parenterally, then examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the compounds; and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • the compounds of the present invention can be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray or nebulizer with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA134AT) or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA), carbon dioxide or other suitable gas.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA134AT) or 1,1,1,2,3,3,
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurized container, pump, spray or nebulizer may contain a solution or suspension of the active compound, e. g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e. g. sorbitan trioleate.
  • a lubricant e. g. sorbitan trioleate.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds of the present invention can be administered in the form of a suppository or pessary, or it may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
  • the compounds may also be administered by the pulmonary or rectal routes. They may also be administered by the ocular route.
  • the compounds can be formulated as micronized suspensions in isotonic, pH was adjusted, sterile saline, or, preferably, as solutions in isotonic, pH was adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
  • the compounds of the present invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • a proposed dose of the compounds according to the present invention for administration to a human is 0.1 mg to 1 g, preferably 1 mg to 500 mg of the active ingredient per unit dose.
  • the unit dose may be administered, for example, 1 to 4 times per day.
  • the dose will depend on the route of administration. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient as well as the severity of the condition to be treated. The precise dose and route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the compounds of the invention may also be used in combination with one or more therapeutic agents.
  • a compound of the invention is used in combination with a second therapeutic agent active against the same disease, the dose of each compound may differ from that when the compound is used alone.
  • the combinations referred to above may conveniently be presented for use in the form of a pharmaceutical formulation.
  • the individual components of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations by any convenient route.
  • administration either the compound of the invention or the second therapeutic agent may be administered first.
  • administration is simultaneous, the combination may be administered either in the same or different pharmaceutical composition.
  • the two compounds When combined in the same formulation it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the formulation.
  • they may be provided in any convenient formulation, conveniently in such manner as are known for such compounds in the art.
  • compositions of the invention can be produced in a manner known per se to the skilled person as described, for example, in Remington's Pharmaceutical Sciences, 15 th Ed., Mack Publishing Co., New Jersey (1975).
  • the compounds according to the present invention can also be provided in the form of a mixture with at least one one compound selected from a therapeutic agent different from the compound of the present invention, a pharmaceutically acceptable carrier, a diluent and an excipient.
  • the compound and/or the therapeutic agent different from the compound of the present invention are preferably present in a therapeutically effective amount.
  • the nature of the therapeutic agent different from the compound of the present invention will depend on the intended use of the mixture.
  • the therapeutic agent different from the compound of the present invention may exert its biological effect by the same or a similar mechanism as the compound according to the invention or by an unrelated mechanism of action or by a multiplicity of related and/or unrelated mechanisms of action.
  • the therapeutic agent different from the compound of the present invention may include neutron-transmission enhancers, psychotherapeutic drugs, acetylcholineesterase inhibitors, calcium-channel blockers, biogenic amines, benzodiazepine tranquillizers, acetylcholine synthesis, storage or release enhancers, acetylcholine postsynaptic receptor agonists, monoamine oxidase-A or -B inhibitors, N-methyl-D-aspartate glutamate receptor antagonists, non-steroidal anti-inflammatory drugs, antioxidants, and serotonergic receptor antagonists.
  • neutron-transmission enhancers may include neutron-transmission enhancers, psychotherapeutic drugs, acetylcholineesterase inhibitors, calcium-channel blockers, biogenic amines, benzodiazepine tranquillizers, acetylcholine synthesis, storage or release enhancers, acetylcholine postsynaptic receptor agonists, monoamine oxidase-A or
  • the therapeutic agent different from the compound of the present invention can be selected from the group consisting of a compound used in the treatment of amyloidosis, compounds against oxidative stress, anti-apoptotic compounds, metal chelators, inhibitors of DNA repair such as pirenzepin and metabolites, 3-amino-1-propanesulfonic acid (3APS), 1,3-propanedisulfonate (1,3PDS), ⁇ -secretase activators, ⁇ - and ⁇ -secretase inhibitors, tau proteins, neurotransmitter, ⁇ -sheet breakers, attractants for amyloid beta clearing/depleting cellular components, inhibitors of N-terminal truncated amyloid beta including pyroglutamated amyloid beta 3-42, anti-inflammatory molecules, or cholinesterase inhibitors (ChEIs) such as tacrine, rivastigmine, donepezil, and/or galantamine, M1 agonists, other drugs including any amyloid or tau modifying drug
  • the mixtures according to the invention may comprise niacin or memantine together with a compound according to the present invention and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.
  • mixtures are provided that comprise as a therapeutic agent different from the compound of the present invention “atypical antipsychotics” such as, for example clozapine, ziprasidone, risperidone, aripiprazole or olanzapine for the treatment of positive and negative psychotic symptoms including hallucinations, delusions, thought disorders (manifested by marked incoherence, derailment, tangentiality), and playful or disorganized behavior, as well as anhedonia, flattened affect, apathy, and social withdrawal, together with a compound according to the invention and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.
  • “atypical antipsychotics” such as, for example clozapine, ziprasidone, risperidone, aripiprazole or olanzapine for the treatment of positive and negative psychotic symptoms including hallucinations, delusions, thought disorders (manifested by marked incoherence,
  • the compounds of the present invention can be synthesized by one of the general methods shown in the following schemes. These methods are only given for illustrative purposes and should not to be construed as limiting.
  • halogenated aminopyridines are reacted with benzoyl isothiocyanate in a solvent such as acetone to afford the desired benzoyl thiourea pyridine derivatives after purification.
  • Deprotection of benzoyl groups was achieved using basic conditions.
  • the thioureas were cyclized using a copper catalyst.
  • An alternative route could be employed by cyclizing first the benzoyl thiourea pyridine derivatives followed by deprotection of the amide using acidic conditions. Finally, the amino groups were transformed into halogen using standard conditions.
  • 5-Chlorothieno[3,2-b]pyridine was treated with a strong base in a suitable solvent followed by addition of bromine to deliver the desired building block after purification.
  • the intermediate A can be further functionalized using palladium-catalyzed cross-coupling conditions such as Suzuki reaction, Buchwald-Hartwig reaction or Sonogashira reaction to afford the desired compounds after purification.
  • the final compounds could be also obtained via a one pot procedure by sequential additions of boronic acids or ester.
  • the intermediate A or even a final compound bearing a leaving group such as a fluorine atom could be subjected to S N Ar conditions to afford the desired final compounds after purification.
  • the intermediate B was subjected either to alkylation using a base and an electrophile or copper-catalyzed cross-coupling reaction with a haloaryl to afford the desired final compounds after purification.
  • Compounds having the formula (I) or (II) which are labeled by 18 F can be prepared by reacting a precursor compound, as described below, with an 18 F-fluorinating agent, so that the LG comprised in the precursor compound is replaced by 18 F.
  • any suitable 18 F-fluorinating agent can be employed. Typical examples include H 18 F, alkali or alkaline earth 18 F-fluorides (e.g., K 18 F, Rb 18 F, Cs 18 F, and Na 18 F).
  • the 18 F-fluorination agent can be used in combination with a chelating agent such as a cryptand (e.g.: 4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8.8.8]-hexacosane—Kryptofix®) or a crown ether (e.g.: 18-crown-6).
  • a cryptand e.g.: 4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8.8.8]-hexacosane—Kryptofix®
  • a crown ether e.g.: 18-crown-6
  • the 18 F-fluorinating agent can be a tetraalkylammonium salt of 18 F or a tetraalkylphosphonium salt of 18 F; e.g., tetra(C 1-6 alkyl)ammonium salt of 18 F or a tetra(C 1-6 alkyl)phosphonium salt of 18 F.
  • the 18 F-fluorination agent is K 18 F, H 18 F, Cs 18 F, Na 18 F or tetrabutylammonium [ 18 F]fluoride.
  • the intermediate A can be further functionalized using palladium-catalyzed cross-coupling conditions such as Suzuki reaction, Buchwald-Hartwig reaction or Sonogashira reaction to afford the desired precursor compounds containing a leaving group (LG) after purification.
  • the final compounds could be also obtained via a one pot procedure by sequential additions of boronic acids or esters.
  • the intermediate A or even a final compound bearing a leaving group (different from the LG group of the final precursor compounds) such as a fluorine atom could, be subjected to SNAr conditions to afford the desired precursor compounds containing a further LG after purification.
  • the derivatives containing a LG are precursor compounds to allow the introduction of the 18 F-label in the following step.
  • Preferred LG for the introduction of 18 F are: C 1-4 alkyl sulfonate C 6-10 aryl sulfonate, nitro, trimethylammonium and halogens.
  • a LG can be also employed a boronic ester.
  • Rings B, D, and E carrying a LG can be prepared following a procedure similar to the one given for Ring A above.
  • the reactions take place in the presence of a fluorinating agent and typically a solvent.
  • 18 F labeled compounds can be prepared by reacting the precursor compounds containing a LG with an 18 F-fluorinating agent, so that the LG is replaced by 18 F.
  • the reagents, solvents and conditions which can be used for the 18 F-fluorination are well-known to a skilled person in the field (L. Cai, S. Lu, V. Pike, Eur. J. Org. Chem 2008, 2853-2873; J. Fluorine Chem., 27 (1985):177-191; Coenen, Fluorine-18 Labeling Methods: Features and Possibilities of Basic Reactions, (2006), in: Schubiger P.
  • the solvents used in the 18 F-fluorination are DMF, DMSO, acetonitrile, DMA, or mixtures thereof, preferably the solvent is acetonitrile, DMSO.
  • detectably labeled compounds are intended and can be easily prepared, e.g., by using detectably labeled starting materials, such as starting materials containing C( 3 H) 3 , ( 11 C)H 3 or 18 F.
  • the mother liquors were separated and the aqueous phase was washed several times with DCM/MeOH.
  • the combined organics were washed with water (2 ⁇ 200 mL), brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • the solid was dissolved in DCM/MeOH and filtered through a plug of silica. The plug was then washed with 5% MeOH in DCM and the mother liquor was concentrated to dryness. The solid was triturated in hot EtOAc. After cooling, the solid was filtered and the mother liquor was concentrated under reduced pressure.
  • the reaction mixture was poured into 1.0 L of water and 1.0 L of aqueous saturated solution of NH 4 Cl. The suspension was stirred at room temperature for 1 hour. The solid was filtered, washed with aqueous saturated solution of NH 4 Cl (2 ⁇ 500 mL), water (2 ⁇ 500 mL) and dried to give the desired product as an off white solid (28.6 g, 94%).
  • the reaction mixture was degassed with a stream of argon for 10 minutes and heated at 70° C. for 2 h. Then the mixture was cooled at room temperature, taken up in ethyl acetate (20 mL), washed with water (2 ⁇ 20 ml) and brine (10 ml), dried over Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified on HP-Sil SNAP cartridges using a Biotage Isolera One purification system employing ethyl acetate/n-heptane gradient (20/80 ⁇ 50/50) to afford the title compound (220 mg, 74%).
  • the reaction mixture was degassed with a stream of argon for 10 minutes and heated at 100° C. for 4 h. The mixture then was cooled to room temperature, taken up in ethyl acetate (20 mL), washed with water (2 ⁇ 20 ml) and brine (10 ml), dried over Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified on HP-Sil SNAP cartridges using a Biotage Isolera One purification system employing ethyl acetate/n-heptane gradient (50/50 ⁇ 90/10) to afford the title compound (42 mg, 52%).
  • the reaction mixture was degassed with a stream of argon for 10 minutes and heated at 70° C. for 2 hours and cooled at room temperature.
  • [1,1′-bis(diphenylphosphino)ferrocene]-dichloropalladium(II) complex with dichloromethane (0.013 g, 0.016 mmol) and (6-fluoropyridin-3-yl)boronic acid (0.059 g, 0.41 mmol) were added, followed by additional nitrogen purges.
  • the reaction mixture was heated at 100° C. for 3 to 4 hours.
  • the reaction mixture was heated at 100° C. in a sand bath for 4 h, cooled to room temperature and the reaction mixture was diluted with water (50 mL). The aqueous phase was then extracted with dichloromethane (3 ⁇ 50 mL). The combined organics were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified on HP-Sil SNAP cartridges using a Biotage Isolera One purification system employing a dichloromethane/ethyl acetate gradient (90/10 ⁇ 70/30) to afford the title compound (200 mg, 80%).
  • the reaction mixture was heated at 100° C. in a sand bath for 18 h, cooled to room temperature and the solvents were removed under reduced pressure.
  • the residue was purified on HP-Sil SNAP cartridges using a Biotage Isolera One purification system employing a dichloromethane/ethyl acetate gradient (98/2 ⁇ 95/5) to afford the title compound (11 mg, 43%).
  • Brain sections were encircled with pap pen liquid blocker to reduce the volume of solution for the different incubations. Sections were fixed for 15 min at 4° C. with 4% paraformaldehyde and washed three times for 5 minutes with PBS (phosphate buffered saline) at room temperature. Test compounds were incubated on the sections at 100 ⁇ M in 50% ethanol in water for 30 min at room temperature, followed by three washes of 5 minutes with PBS.
  • PBS phosphate buffered saline
  • Sections were then saturated and permeabilized in blocking buffer (PBS, 10% NGS, 0.25% Triton) for 1 hour at room temperature and then incubated for 2 hours at room temperature with the primary antibodies against aSyn: aSyn-211 (SantaCruz Biotechnology sc-12767) or aSyn-pS129 (Abcam AB51253), and primary antibody against A ⁇ , 4G8, (Covance, SIG-39220). All primary antibodies diluted 1/250 in PBS, 5% NGS, 0.25% Triton.
  • PBS 10% NGS, 0.25% Triton
  • the sections were incubated for 30 minutes at room temperature with a secondary anti-mouse antibody labeled with AlexaFluor555 (Invitrogen A21422) or anti-rabbit antibody labeled with AlexaFluor555 (Invitrogen A21428) and further washed three times in PBS.
  • a secondary anti-mouse antibody labeled with AlexaFluor555 Invitrogen A21422
  • anti-rabbit antibody labeled with AlexaFluor555 Invitrogen A21428
  • the sections were incubated in a solution of 0.1% Sudan Black (Sigma 199664) in 70% ethanol for 15 min at room temperature, followed by four washes with PBS and mounted under cover slips using ProLong Gold Antifade reagent (Invitrogen P36930).
  • blocking buffer PBS, 10% NGS, 0.25% Triton
  • the sections were incubated for 30 minutes at room temperature with a secondary anti-mouse antibody labeled with AlexaFluor555 (Invitrogen A21422) and further washed three times in PBS.
  • AlexaFluor555 AlexaFluor555
  • the sections were incubated in a solution of 0.1% Sudan Black (Sigma 199664) in 70% ethanol for 15 min at room temperature, followed by four washes with PBS and mounted under cover slips using ProLong Gold Antifade reagent (Invitrogen P36930).
  • Approximately 300 mg of cortex from control and PD donors (purchased from Tissue Solutions Ltd., diagnosed with PD Braak stage V-VI, or healthy age-matched donors) were weighed and homogenized on ice using a glass potter in 9 ⁇ volume/weight of homogenization buffer: 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1 mM EGTA containing phosphatase inhibitors (30 mM NaF, 0.2 mM Na 3 VO 4 , 1 nM okadaic acid, 1 mM phenylmethylsulfonyl fluoride (PMSF), 5 mM Na 4 P 2 O 7 ) and protease inhibitor cocktail (CompleteTM, Roche). Samples were aliquoted and stored at ⁇ 80° C.
  • Backscattering interferometry (BSI) measurements were performed by Molecular Sensing GmbH (Idstein, Germany). Examples of this invention at 10 mM in DMSO were diluted 1:100 in PBS and then diluted again in PBS to yield 2 ⁇ M concentration of compound in PBS with 0.02% DMSO.
  • the refractive index of the assay buffer (PBS pH 7.4 containing 0.02% DMSO) and the compound were matched and then 2 ⁇ serial dilutions of the compound were done in polypropylene dilution reservoirs. A thawed aliquot of control, AD and PD brain homogenates was diluted 1/150 in PBS, pH 7.4 and used immediately.
  • Examples of this invention and brain homogenates were mixed 1:1 in 96-well PCR microplates to a final volume of 60 ⁇ L and heat sealed with foil. The assays were allowed to incubate at room temperature for 45 minutes before being run on the BSI instrument. Wells were pierced individually prior to sample injection and measurement of BSI signal (each well analyzed in triplicate).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nuclear Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
US16/084,223 2016-03-11 2017-03-10 Bicyclic compounds for diagnosis and therapy Abandoned US20190071450A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP16159878.4 2016-03-11
EP16159878 2016-03-11
EP16199577 2016-11-18
EP16199577.4 2016-11-18
PCT/EP2017/055754 WO2017153601A1 (en) 2016-03-11 2017-03-10 Bicyclic compounds for diagnosis and therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/055754 A-371-Of-International WO2017153601A1 (en) 2016-03-11 2017-03-10 Bicyclic compounds for diagnosis and therapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/894,682 Division US11623931B2 (en) 2016-03-11 2020-06-05 Bicyclic compounds for diagnosis and therapy

Publications (1)

Publication Number Publication Date
US20190071450A1 true US20190071450A1 (en) 2019-03-07

Family

ID=58488956

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/084,223 Abandoned US20190071450A1 (en) 2016-03-11 2017-03-10 Bicyclic compounds for diagnosis and therapy
US16/894,682 Active US11623931B2 (en) 2016-03-11 2020-06-05 Bicyclic compounds for diagnosis and therapy
US18/114,654 Pending US20240051967A1 (en) 2016-03-11 2023-02-27 Bicyclic compounds for diagnosis and therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/894,682 Active US11623931B2 (en) 2016-03-11 2020-06-05 Bicyclic compounds for diagnosis and therapy
US18/114,654 Pending US20240051967A1 (en) 2016-03-11 2023-02-27 Bicyclic compounds for diagnosis and therapy

Country Status (14)

Country Link
US (3) US20190071450A1 (zh)
EP (2) EP3426653B1 (zh)
JP (4) JP2019512491A (zh)
KR (3) KR102646115B1 (zh)
CN (2) CN116199700A (zh)
AU (1) AU2017231781C1 (zh)
BR (1) BR112018068066B1 (zh)
CA (2) CA3224365A1 (zh)
ES (1) ES2905539T3 (zh)
MX (1) MX2018010693A (zh)
NZ (1) NZ746901A (zh)
RU (2) RU2766139C2 (zh)
SG (2) SG11201807778YA (zh)
WO (1) WO2017153601A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210002712A1 (en) * 2019-07-05 2021-01-07 National University Corporation Chiba University Phosphatidic acid sensor
WO2024059845A1 (en) * 2022-09-15 2024-03-21 The Regents Of The University Of California Supramolecular polymer therapeutics and diagnostics

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3802548A1 (en) * 2018-06-08 2021-04-14 AC Immune SA Novel compounds for diagnosis
CN112805280A (zh) * 2018-06-27 2021-05-14 Ptc医疗公司 用于治疗亨廷顿氏病的杂芳基化合物
CN112912376A (zh) * 2018-08-20 2021-06-04 阿尔维纳斯运营股份有限公司 用于治疗神经变性疾病的具有E3泛素连接酶结合活性并靶向α-突触核蛋白的蛋白水解靶向嵌合(PROTAC)化合物
CN113874015B (zh) 2018-12-21 2024-05-24 细胞基因公司 Ripk2的噻吩并吡啶抑制剂
GB201914989D0 (en) * 2019-10-16 2019-11-27 Karin & Sten Mortstedt Cbd Solutions Ab Selective ligands for TAU Aggregates
US20230067910A1 (en) * 2019-11-19 2023-03-02 Modag Gmbh Novel compounds for the diagnosis, treatment and prevention of diseases associated with the aggregation of alpha-synuclein
BR112022019987A2 (pt) 2020-04-07 2022-11-22 Bayer Ag Tiazolpiridinas substituídas, seus sais e uso dos mesmos como substâncias herbicidamente ativas
JP2023525081A (ja) 2020-05-07 2023-06-14 エーシー・イミューン・エス・アー 診断のための新規化合物
AR123730A1 (es) * 2020-10-15 2023-01-04 Ac Immune Sa Compuestos novedosos
WO2023285661A1 (en) * 2021-07-16 2023-01-19 Ac Immune Sa Novel compounds for the diagnosis of tdp-43 proteinopathies
TW202328150A (zh) 2021-09-07 2023-07-16 德商拜耳廠股份有限公司 經取代之噻唑并吡啶、其鹽及其作為除草活性物質之用途
TW202328151A (zh) 2021-09-07 2023-07-16 德商拜耳廠股份有限公司 經取代之2,3-二氫[1,3]噻唑并[4,5-b]吡啶、其鹽及其作為除草活性物質之用途
AU2022385416A1 (en) 2021-11-10 2024-05-02 Ac Immune Sa 4h-imidazo[1,5-b]pyrazole derivatives for diagnosis
WO2023083961A1 (en) 2021-11-10 2023-05-19 Ac Immune Sa Dihydropyrrolo[3,4-c]pyrazole derivatives and their use in diagnosis
WO2023083998A1 (en) 2021-11-10 2023-05-19 Ac Immune Sa Dihydropyrrolo[3,4c]-pyrazole derivatives and their use in diagnosis
CN116262744A (zh) * 2021-12-13 2023-06-16 复旦大学 用于α-突触核蛋白聚集体成像的小分子探针
WO2023163033A1 (ja) * 2022-02-24 2023-08-31 国立研究開発法人量子科学技術研究開発機構 新規化合物、αシヌクレイン凝集体結合剤及びその利用
WO2024068948A1 (en) * 2022-09-30 2024-04-04 Ac Immune Sa Novel compounds for the diagnosis of tdp-43 proteinopathies

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05125076A (ja) * 1991-11-06 1993-05-21 Canon Inc 液晶性化合物、これを含む液晶組成物、それを有する液晶素子、それらを用いた表示方法及び表示装置
KR100446363B1 (ko) * 1997-11-11 2004-09-01 화이자 프로덕츠 인코포레이티드 항암제로 유용한 티에노피리미딘 및 티에노피리딘 유도체
ES2536449T3 (es) * 2000-08-24 2015-05-25 University Of Pittsburgh Of The Commonwealth System Of Higher Education Derivados de tioflavina para uso en diagnosis de la enfermedad de Alzheimer
US6700722B2 (en) * 2001-05-25 2004-03-02 Infineon Technologies Ag High-speed zero phase restart of a multiphase clock
US7622479B2 (en) * 2001-11-26 2009-11-24 Takeda Pharmaceutical Company Limited Bicyclic derivative, its production and use
US20040214856A1 (en) * 2003-04-23 2004-10-28 Pfizer Inc Cannabinoid receptor ligands and uses thereof
BRPI0519124A2 (pt) 2004-12-17 2008-12-23 Lilly Co Eli composto, mÉtodo para tratar, prevenir ou melhorar obesidade e doenÇas relacionadas e/ou sintomas das mesmas, composiÇço farmacÊutica, uso de composto, e, combinaÇço
AR056200A1 (es) * 2005-09-27 2007-09-26 Wyeth Corp Tieno [2,3-b]piridin-5-carbonitrilos como inhibidores de proteina quinasa
AU2007229492B2 (en) * 2006-03-28 2011-11-03 High Point Pharmaceuticals, Llc Benzothiazoles having histamine H3 receptor activity
EP2023919A4 (en) 2006-05-08 2010-12-22 Molecular Neuroimaging Llc COMPOUNDS AND AMYLOID PROBES FOR THERAPY AND IMAGING USES
CA2652524A1 (en) * 2006-06-08 2007-12-21 Eli Lilly And Company Novel mch receptor antagonists
CA2670418A1 (en) * 2006-11-24 2008-05-29 Ac Immune Sa N-(methyl)-1h-pyrazol-3-amine, n-(methyl)-pyridin-2-amine and n-(methyl)-thiazol-2-amine derivatives for the treatment of diseases associated with amyloid or amyloid-like proteins, like e.g. alzheimers's
CN101675045B (zh) * 2007-05-11 2012-11-28 弗·哈夫曼-拉罗切有限公司 作为β-淀粉样蛋白调节剂的杂芳基苯胺
US8153813B2 (en) * 2007-12-20 2012-04-10 Abbott Laboratories Benzothiazole and benzooxazole derivatives and methods of use
EP2265270A1 (en) * 2008-02-04 2010-12-29 OSI Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors
US8932557B2 (en) 2008-02-14 2015-01-13 Eli Lilly And Company Imaging agents for detecting neurological dysfunction
WO2009126635A1 (en) * 2008-04-09 2009-10-15 Abbott Laboratories 2-amino-benzothiazole derivates useful as inhibitors of rock kinases
US8530483B2 (en) * 2008-05-30 2013-09-10 Merck Sharp & Dohme Corp. Substituted azabenzoxazoles
US8420052B2 (en) * 2008-07-24 2013-04-16 Siemens Medical Solutions Usa, Inc. Imaging agents useful for identifying AD pathology
WO2010021381A1 (ja) * 2008-08-22 2010-02-25 武田薬品工業株式会社 縮合複素環誘導体およびその用途
GB0821994D0 (en) * 2008-12-02 2009-01-07 Ge Healthcare Ltd In viva imaging method
WO2010071819A1 (en) 2008-12-19 2010-06-24 Schering Corporation Bicyclic heterocyclic derivatives and methods of use thereof
AU2011232347B2 (en) * 2010-03-24 2015-08-06 Musc Foundation For Research Development Compositions and methods for the treatment of degenerative diseases
NZ602777A (en) 2010-04-16 2014-07-25 Ac Immune Sa Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins
US20110256064A1 (en) * 2010-04-16 2011-10-20 Ac Immune, S.A. Novel Compounds for the Treatment of Diseases Associated with Amyloid or Amyloid-like Proteins
GB201012889D0 (en) * 2010-08-02 2010-09-15 Univ Leuven Kath Antiviral activity of novel bicyclic heterocycles
WO2012007510A1 (en) * 2010-07-14 2012-01-19 Bayer Pharma Aktiengesellschaft Compounds for binding and imaging amyloid plaques and their use
DE102010045797A1 (de) 2010-09-20 2012-03-22 Klinikum Darmstadt Gmbh Verbindungen für die Diagnostik neurodegenerativer Erkrankungen am Riechepithel
JP5720253B2 (ja) 2011-01-06 2015-05-20 コニカミノルタ株式会社 有機エレクトロニクス素子、それを具備した表示装置及び照明装置
EP2560008B1 (en) 2011-08-18 2016-11-30 Korea Institute of Science and Technology Pharmaceutical compositions for preventing or treating degenerative brain disease and method of screening the same
US9464064B2 (en) * 2011-09-07 2016-10-11 University Of Kansas HCV helicase inhibitors and methods of use thereof
ES2711876T3 (es) * 2011-09-19 2019-05-08 C2N Diagnostics Métodos para el diagnóstico y tratamiento de las enfermedades neurológicas y neurodegenerativas, trastornos y procesos asociados
AU2012329274B2 (en) * 2011-10-27 2016-11-10 Protamed, Inc. Caffeinated compounds and compositions for treatment of amyloid diseases and synucleinopathies
WO2013156431A1 (en) * 2012-04-17 2013-10-24 Syngenta Participations Ag Pesticidally active pyridyl- and pyrimidyl- substituted thiazole and thiadiazole derivatives
BR112014032077A2 (pt) * 2012-06-22 2017-06-27 Du Pont composto, composição fungicida e método para controlar doenças de plantas
WO2014113191A1 (en) 2013-01-15 2014-07-24 Xiaohu Zhang Hedgehog pathway signaling inhibitors and therapeutic applications thereof
TWI652014B (zh) * 2013-09-13 2019-03-01 美商艾佛艾姆希公司 雜環取代之雙環唑殺蟲劑
WO2015091584A1 (en) * 2013-12-18 2015-06-25 F. Hoffmann-La Roche Ag Thiazolopyridine compounds, compositions and their use as tyk2 kinase inhibitors
PT3186233T (pt) 2014-08-29 2021-12-02 Chdi Foundation Inc Sondas para imagem de proteína de huntingtina
TR201815071T4 (tr) 2014-12-02 2018-11-21 Bayer Cropscience Ag Zararlılarla mücadele maddesi olarak bisiklik bileşikler.
WO2017069275A1 (ja) 2015-10-22 2017-04-27 田辺三菱製薬株式会社 新規二環性複素環化合物

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210002712A1 (en) * 2019-07-05 2021-01-07 National University Corporation Chiba University Phosphatidic acid sensor
WO2024059845A1 (en) * 2022-09-15 2024-03-21 The Regents Of The University Of California Supramolecular polymer therapeutics and diagnostics

Also Published As

Publication number Publication date
KR102646115B1 (ko) 2024-03-12
RU2022101103A (ru) 2022-03-29
EP4001277A2 (en) 2022-05-25
US20240051967A1 (en) 2024-02-15
CA3224365A1 (en) 2017-09-14
KR20240036709A (ko) 2024-03-20
RU2766139C2 (ru) 2022-02-08
AU2017231781B2 (en) 2021-07-15
RU2018135709A (ru) 2020-04-13
KR102464274B1 (ko) 2022-11-07
JP2019512491A (ja) 2019-05-16
JP2022001573A (ja) 2022-01-06
CA3015947A1 (en) 2017-09-14
MX2018010693A (es) 2019-03-28
CN116199700A (zh) 2023-06-02
CN109071528A (zh) 2018-12-21
JP2021167310A (ja) 2021-10-21
US20200299305A1 (en) 2020-09-24
BR112018068066B1 (pt) 2023-11-28
CN109071528B (zh) 2023-02-17
JP7297323B2 (ja) 2023-06-26
EP3426653A1 (en) 2019-01-16
KR20180117707A (ko) 2018-10-29
EP3426653B1 (en) 2021-12-15
SG11201807778YA (en) 2018-10-30
WO2017153601A1 (en) 2017-09-14
ES2905539T3 (es) 2022-04-11
JP2023116823A (ja) 2023-08-22
RU2018135709A3 (zh) 2020-06-30
SG10202006688RA (en) 2020-08-28
NZ746901A (en) 2023-06-30
EP4001277A3 (en) 2022-09-14
BR112018068066A2 (pt) 2019-01-08
US11623931B2 (en) 2023-04-11
AU2017231781A1 (en) 2018-10-25
KR20220151042A (ko) 2022-11-11
AU2017231781C1 (en) 2021-12-16
JP7383304B2 (ja) 2023-11-20

Similar Documents

Publication Publication Date Title
US11623931B2 (en) Bicyclic compounds for diagnosis and therapy
US10662193B2 (en) Carbazole and carboline compounds for use in the diagnosis, treatment, alleviation or prevention of disorders associated with amyloid or amyloid-like proteins
US20230174536A1 (en) Novel compounds for diagnosis
US20210252175A1 (en) Novel compounds for diagnosis
AU2022387830A1 (en) Dihydropyrrolo[3,4c]-pyrazole derivatives and their use in diagnosis
WO2023083961A1 (en) Dihydropyrrolo[3,4-c]pyrazole derivatives and their use in diagnosis
AU2022385416A1 (en) 4h-imidazo[1,5-b]pyrazole derivatives for diagnosis

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: AC IMMUNE SA, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOLETTE, JEROME;GABELLIERI, EMANUELE;DARMENCY, VINCENT;SIGNING DATES FROM 20190228 TO 20190305;REEL/FRAME:048595/0072

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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