WO2012164103A2 - Blockers of the nogo-a s1pr pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair - Google Patents

Blockers of the nogo-a s1pr pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair Download PDF

Info

Publication number
WO2012164103A2
WO2012164103A2 PCT/EP2012/060533 EP2012060533W WO2012164103A2 WO 2012164103 A2 WO2012164103 A2 WO 2012164103A2 EP 2012060533 W EP2012060533 W EP 2012060533W WO 2012164103 A2 WO2012164103 A2 WO 2012164103A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
amino
chloro
ethyl
group
Prior art date
Application number
PCT/EP2012/060533
Other languages
French (fr)
Other versions
WO2012164103A9 (en
WO2012164103A3 (en
Inventor
Martin Schwab
Björn Tews
Anissa KEMPF
Original Assignee
Universität Zürich
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 Universität Zürich filed Critical Universität Zürich
Publication of WO2012164103A2 publication Critical patent/WO2012164103A2/en
Publication of WO2012164103A3 publication Critical patent/WO2012164103A3/en
Publication of WO2012164103A9 publication Critical patent/WO2012164103A9/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic 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
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4021-aryl substituted, e.g. piretanide
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • 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
    • A61K31/4375Heterocyclic 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 six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides

Definitions

  • This invention relates to the treatment of diseases characterized by neuronal damage combined with lack of subsequent repair, using blockers of the Nogo-A Sphingosine 1 - phosphate receptor S1 PR pathway, particularly antibodies to, and small molecule inhibitors of, S1 PR receptors.
  • Damage to neuronal cells can have severe clinical outcomes depending on which neuronal cells are damaged and where. Damage to or injury of the soma or proximal part of the axon will kill neuronal cells. Damage to the distal part of the axon can only be repaired in cells of the peripheral nervous system, and in some cases even there repair is not possible.
  • Nogo-A is a protein identified to be an inhibitor of neurite outgrowth and cell spreading. Fragment analysis of the Nogo-A molecule has shown that the molecule has at least two inhibitory domains "Amino-Nogo" (rat aa 1-979), at the N-terminus and Nogo-66. Amino- Nogo-A contains the functionally active Nogo ⁇ 20 fragment (rat aa 544-725).
  • the receptor of Nogo-66 a glycosylphosphatidyl-anchored (GPI-anchored) protein, has been identified and characterized. This receptor is termed NgR1 and signals via the RhoA pathway. Besides NgR1 , other receptors homologs have been characterized, termed NgR2 and NgR3. No receptor for Amino-Nogo has been identified so far.
  • the objective of the instant invention is to provide safe and efficacious means for treating neuronal damage. This objective is attained by the subject-matter of the independent claims.
  • the present invention is based on the surprising finding that Nogo-A binds to receptors of the S1 PR family and that inhibition of these receptors suppresses the inhibitory activity of Nogo-A.
  • an inhibitor capable of binding to a member of the S1 PR receptor group comprised of S1 PR2 (Gene ID: 9294) and S1 PR5 (Gene ID: 53637) with a dissociation constant of 5x10 "7 mol/l or smaller is provided for use in treating neuronal damage.
  • an agonist of S1 PR1 (Gene ID: 1901 , S1 PR1 has an antagonistic effect on S1 PR2) is provided for use in treating neuronal damage.
  • Gene ID numbers in this document refer to entries in the Gene data base of the United States National Center for Biotechnology Information.
  • Neuronal damage in the sense of the instant invention means injury to or dysfunction of a neuronal cell caused by trauma or neuronal disease resulting in constraint or disruption of the saltatory conduction by the neuronal cell within the nervous system in the absence of repair capacity.
  • diseases characterized by neuronal damage and subsequent lack of repair include, without being restricted to, paraplegia caused by spinal cord injury, stroke, dementia, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis and paralysis and subsequent atrophy of limbs after rupture of a peripheral nerve.
  • Such an inhibitor according to the first aspect of the invention may be a small molecule pharmaceutical drug, an antibody, an antibody-fragment, an antibody-like molecule, a nucleic acid aptamer or an oligopeptide 6 to 30 amino acid residues in length.
  • Methods for generating antibodies against the members of the S1 PR receptor group are known in the art. They include, for example, immunization of mice with a S1 PR receptor or soluble parts thereof.
  • An antibody fragment may be the Fab domain of an antibody (the antigen binding region of an antibody) or a single chain antibody (scFv), a fusion protein consisting of the variable regions of light and heavy chains of an antibody connected by a peptide linker.
  • An antibody-like molecule may be a repeat protein, such as a designed ankyrin repeat protein (Molecular Partners, Zurich).
  • An antibody fragment or an antibody-like molecule may be manufactured by methods such as recombinant protein expression.
  • Suitable inhibitors according to the first aspect of the invention may also be developed by evolutive methods such as phage display, ribosome display or SELEX, wherein polypeptides or oligonucleotides are selected according to their binding affinity to a target of interest. Additionally, higher affinity inhibitors may be identified by reiterative rounds of evolution and selection of the amino acid sequence or nucleotide sequence.
  • An oligopeptide according to a first aspect of this invention may be a peptide derived from the part of Nogo-A which is recognized by a member of the S1 PR receptor group. The binding of the peptide results in inhibition of the receptor. Conversely, a peptide may also be derived from the recognition site of member of the S1 PR receptor family that competes with the receptor for Nogo-A. Binding of such an oligopeptide must not activate the signalling pathway downstream of the receptor.
  • a modulator of gene expression of G13 (Gene ID: 10672) or a member of the S1 PR group comprised of S1 PR2 and S1 PR5 is provided for use in treating neuronal damage.
  • G13 is a G-protein and strongly coupled to S1 PR2.
  • Such modulator may be single-stranded or double-stranded interfering ribonucleic acid oligomer of precursor thereof, comprising a sequence tract that is complementary to an mRNA molecule encoding G13 or a member of the S1 PR group described in the preceding paragraphs.
  • siRNA silencing or "knocking down" genes, by degradation of mRNA or other effects, is well known.
  • technologies developed for this purpose include siRNA, miRNA, shRNA, shmiRNA, and dsRNA.
  • siRNA miRNA, miRNA, shRNA, shmiRNA, and dsRNA.
  • a modulator according to the above aspect may also be an expression vector comprising a sequence encoding an interfering ribonucleic acid oligomer as described in the preceding paragraphs.
  • the sequence may be under the control of a promoter operable in mammalian cells.
  • Such expression vectors facilitate production of an interfering RNA within the cell. Methods for making and using such expression vectors are known in the art.
  • a modulator according to the above aspect of the invention may be a single- stranded or double-stranded antisense ribonucleic or deoxyribonucleic acid, comprising sequences complementary to an operon expressing of a gene encoding a member of the S1 PR group described above or G13.
  • Such operon sequences may include, without being restricted to, intron, exon, operator, ribosome binding site or enhancer sequences.
  • Such antisense molecules may be 12-50 nucleotides in length.
  • a soluble polypeptide for treating neuronal damage comprising a contiguous amino acid sequence of at least 30 amino acid residues taken from the protein sequence of a member of the S1 PR group.
  • a soluble polypeptide can be used to inhibit Nogo-A by specifically binding to it.
  • Specific binding in the sense of the invention means binding with dissociation constant of 5x10 "7 mol/l or smaller.
  • the soluble polypeptide is linked to an Fc antibody domain.
  • Such soluble polypeptide may act as a decoy receptor to capture, or to bind to and render inactive, Nogo-A or other S1 PR receptor ligands involved in neuronal damage.
  • An example of a soluble polypeptide may be the extracellular domain of the S1 PR2 or S1 PR5 receptor linked to an Fc antibody domain.
  • a compound for use in treating neuronal damage wherein the compound is selected from the group comprised of:
  • R1 is a CrC 12 alkyl
  • R2, R3 and R4 are each independently hydrogen, halogen, CrC 6 alkyl, Ci-C 6 perhaloalkyl, CrC 4 perhaloalkoxy, amino, mono- or di C C 4 alkylamino, C 3 -C 7 cycloalkyl or C 3 -C 7 cycloalkoxy, and R3 and R4 are optionally positioned at h, i, or j, but not simultaneously at the same position, wherein h, I or j denotes substitution positions in the pyrimidyl moiety, and
  • R5 is , halogen, C C 6 alkyl, C C 6 perhaloalkyl, d-C 4 perhaloalkoxy, amino, mono- or di C C 4 alkylamino, C 3 -C 7 cycloalkyl or C 3 -C 7 cycloalkoxy, and
  • - n 0, 1 , 2, 3 or 4;
  • - Q is selected from CH 2 , C(O), and C(S), and
  • Qa and Qb are independently selected from N and CH, and
  • R 1 is selected from C 6 -C 15 -aromatic carbocyclic groups, C 5 -C 15 -carbocyclic group, and a 4- to 15-membered heterocyclic group having one or more heteroatoms selected form the group consisting of oxygen, nitrogen and sulphur
  • R 1 is selected from C-i-C 8 -alkyl optionally substituted -OH, halogene, -CN, O- d-Ce-alkyl, NR 1c R 1d , carboxy-C C 8 -alkyl, and COOH, - C(R 1a R 1 b ) m C(0)CR 1c R 1d R1 e, -C(R 1a R 1 b )mC(0)NR 1c R 1d , -C(R 1a R 1b ) t NR 1c R 1d , C(R 1a R 1 b )mCR 1c R 1d R 1e , and
  • R 1 , R 1g and R 1h are independently selected from C-
  • each R 1a and R 1b are independently selected from H- OH and CrC 8 -alkyl optionally substituted by -OH and halogen, and
  • R 1c and R 1d are independently selected from H,
  • C 6 -Ci 5 -aromatic carbocyclic group C 7 -Ci 5 -aralkyl, C 3 -Ci 5 -carbocyclic group, a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
  • C C 8 -alkoxy optionally substituted by -OH, -CN, halogen, NR 4 R 5 , a C 6 -C 15 - aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
  • C 2 -C 8 -alkynyl optionally substituted by -OH, -CN, halogen, NR 4 R 5 , a C 6 -C 15 - aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
  • C 2 -C 8 -alkyl optionally substituted by -OH, -CN, halogen, NR 4 R 5 , a C 6 -Ci 5 - aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
  • R 1e is selected from H and CrC 8 -alkyl
  • R 3 is selected from C 6 -C 15 -aromatic carbocyclic group optionally substituted by -C(R 3a R 3b ) n C(0)NR 3c R 3d or -C(R 3a R 3b ) n C(0)OH, a C 7 -C 15 -aralkyl, C C 8 -alkyl substituted by a C 3 -C 15 -carbocyclic group, a C 3 -C 15 -carbocyclic group, a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, C-
  • R 3a and R 3b are independently selected form H, -OH and CrC 8 -alkyl optionally substituted by -OH and halogen;
  • R3c and R3d are independently selected from H,
  • C 6 -Ci 5 -aromatic carbocyclic group C 3 -Ci 5 -carbocyclic group, or R 3c and R3d, together with the N atom that they are attached, form a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
  • CrCs-alkyoxy optionally substituted by a C 6 -C 15 -aromatic carbocyclic group, C 3 -C 15 -carbocyclic group and a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, a CrCs-alkoxycarbonyl optionally substituted by a C 6 - C 15 -aromatic carbocyclic group, C 3 -C 15 -carbocyclic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
  • d-Cs-alkyl optionally substituted by -OH, -CN, halogen, NR 4 R 5 , a C 6 -C 15 - aromatic carbocyclic group, C 3 -C 15 -carbocyclic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
  • R4 and R5 are independently selected from H and CrC 8 -alkyl, and m and n are independently selected from an integer of 0, 1 , 2 and 3, and t is integer from 1 , 2 and 3,
  • C 6 -Ci 5 -aromatic carbocyclic group, C 7 -Ci 5 -aralkyl, C 3 -Ci 5 -carbocyclic group and 4- to 15-membered group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, unless otherwise stated, are optionally substituted by C 7 -C 15 -aralkyl, d-C 8 -alkyl,CN, d-C 8 - alkylsulfonyl, d-C 8 -haloalkylsulfonyl, halogen, d-C 8 -alkoxy, OH, d-C 8 - alkylcarbonyl, -C(0)-C 6 -C 15 -aromatic carbocyclic group, -C(0)-C 3 -C 15 -carbocyclic group, -C(0)-4- to 15-membered heterocyclic group having one or more heteroatoms selected from the
  • R-i aynd R 5 have both.
  • R 2 and R 4 have both, in each case, identical meanings are hydrogen, C-
  • identical meanings are hydrogen, C-
  • R 3 is hydrogen, C 1 -C 4 -alkoxy, F, CI, CF 3 , or an optionally mono- or
  • C-i-C 8 -alkyl group disubstituted C-i-C 8 -alkyl group, the optional substitent(s) on the said alkyl group being independently selected from the group consiting of halogen, nitro, cyano, formyl, C 1 -C 4 -alkylcarbonyl, hydroxy, C 1 -C 4 -alkoxy, formyloxy, CrC 4 - alkoxycarbonyloxy, C 1 -C 4 -alkoxycarbonyloxy, amino, C1-C4.alkylamino, formylamino, Ci-C 4 -alkylcarbonyamino and Ci-C 4 -alkoxycarbonylamino, and
  • R 6 is hydrogen, an optionally mono- or disubstituted CrC 8 -alkyl, C 2 -C 4 -alkenyl or C 3 -C 7 -cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyi group being indepentently selected from the group consisting of halogen, nitro, cyano, formyl, CrC 4 -alkylcarbonyl, hydroxy, CrC 4 -alkoxy, formyloxy, Ci-C4-alkylcarbonyloxy, CrC 4 -alkoxycarbonyloxy, amino, C 1 -C4- alkylamino, di-(Ci-C 4 -alkyl)amino with two identical or different Ci-C 4 -alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, formylamino, C 1 -C4- alkylcarbonylamino and
  • -C 4 -alkyl C-
  • -C 4 -alkoxycarbonyl, hydroxy, hydroxy-C C 4 -alkyl, C C 4 -alkoxy, d-C ⁇ alkoxy-d-C ⁇ alkyl, HO-C( 0)-, C C 4 -alkoxycarbonyl, formyloxy, CrC 4 -alkylcarbonyloxy, CrC 4 - alkoxycarbonyloxy, amino, Ci-C 4 -alkylamino, di-(
  • alkyl with two identical or different C-i-C 4 -alkyl moieties in the di-(C-i-C 4 - alkyl)amino moiety pyrrolidyl-C-
  • R 7 is hydrogen, an optionally mono- or disubstituted C-
  • R 8 is hydrogen, d-d-alkyl, d-d-alkoxy, F or CI, a compound characterized by a general formula V (described in WO 2008/129029):
  • Ri and R 5 have both, in each case, identical meanings and are CrC 6 -alkyl, d- C 6 -alkoxy, CI, F, Br or CF 3 , and
  • R 2 and R4 both, in each case, identical meanings and are CrC 6 -alkyl, Ci-C 6 - alkoxy, CI, F, Br or CF 3 , and
  • R 3 is hydrogen, CrC 4 -alkoxy, F, CI, CF 3 , or an mono- or disubstituted CrC 8 - alkyl, C 2 -C 4 -alkenyl or C 3 -C 7 -cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyl being independently selected from the group consisting of halogen, nitro, cyano, formyl C 1 -C 4 -alkylcarbonyl, hydroxy, C-
  • R 7 is hydrogen, an optionally mono- or disubstituted CrC 8 -alkyl, C 2 -C 4 -alkenyl or C 3 -C 7 -cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyi group being indepentently selected from the group consisting of halogen, nitro, cyano, formyl, Ci-C 4 -alkylcarbonyl, hydroxy, C1 -C 4 -alkoxy, formyloxy, Ci-C4-alkylcarbonyloxy, CrC 4 -alkoxycarbonyloxy, amino, d-d- alkylamino, di-(C-
  • C 1 -C 4 -alkyl C 1 -C 4 -alkyl, morpholinyl- C 1 -C 4 -alkyl, formylamino, C-
  • -C 4 - alkoxycarbonyl, hydroxy, hydroxy-CrC 4 -alkyl, CrC 4 -alkoxy, Ci-C 4 -alkoxy-Cr C 4 -alkyl, HO-C( 0)-, CrC 4 -alkoxycarbonyl, formyloxy, CrC 4 -alkylcarbonyloxy, CrC 4 -alkoxycarbonyloxy, amino, Ci-C 4 -alky
  • R8 is hydrogen, Ci-C 4 -alkyl, CrC 4 -alkoxy, F or CI, and
  • a pharmaceutical composition for treating neuronal damage comprising an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention.
  • Pharmaceutical compositions for enteral administration such as nasal, buccal, rectal or, especially, oral administration, and for parenteral administration, such as subcutaneous, intravenous, intrahepatic or intramuscular administration, are preferred.
  • the pharmaceutical compositions comprise from approximately 1 % to approximately 95% active ingredient, preferably from approximately 20% to approximately 90% active ingredient.
  • a dosage form for treating neuronal damage comprising an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention.
  • a pharmaceutical carrier or excipient may be present.
  • compositions may be sterilized and/or comprise excipients, for example preservatives, stabilizers, wetting agents and/or emulsifiers, solubilizers, viscosity-increasing agents, salts for regulating osmotic pressure and/or buffers and are prepared in a manner known per se, for example by means of conventional dissolving and lyophilizing processes.
  • suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates.
  • Binders such as starches, cellulose derivatives and/or polyvinylpyrrolidone may also be employed, Disintegrators, flow conditioners and lubricants, for example stearic acid or salts thereof and/or polyethylene glycol may be added in addition or instead.
  • Tablet cores can be provided with suitable, optionally enteric, coatings. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes to indicate different types or doses of active ingredient.
  • compositions for oral administration also include hard capsules consisting of gelatin, and soft, sealed capsules consisting of gelatin and a plasticizer, such as glycerol or sorbitol.
  • a plasticizer such as glycerol or sorbitol.
  • Capsules may contain the active ingredient in the form of granules, or dissolved or suspended in suitable liquid excipients, such as in oils.
  • Transdermal/intraperitoneal and intravenous applications are also considered, for example using a transdermal patch, which allows administration over an extended period of time, e.g. from one to twenty days.
  • Intravenous or subcutaneous applications are particularly preferred as well as intrathecal applications.
  • a method for treating neuronal damage comprising the administration of an inhibitor, modulator, soluble polypeptide or compound according to the above aspect of the invention to a patient in need thereof.
  • the treatment may be for prophylactic or therapeutic purposes.
  • an active agent as described in the preceding paragraphs is preferably provided in the form of a pharmaceutical preparation comprising the active agent in chemically pure form and optionally a pharmaceutically acceptable carrier and optionally adjuvants.
  • the active agent is used in an amount effective against the diseases characterized by neuronal damage and lack of subsequent repair.
  • the dosage of the active agent depends upon the species, the patient age, weight, and individual condition, the individual pharmacokinetic data, mode of administration, and whether the administration is for prophylactic or therapeutic purposes.
  • the daily dose administered ranges from approximately 0.1 mg/kg to approximately 1000 mg/kg, preferably from approximately 0.5 mg to approximately 100 mg/kg, of an active agent.
  • a method of manufacturing a medicament for treating neuronal damage comprising the use of an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention.
  • Medicaments according to the invention are manufactured by methods known in the art, especially by conventional mixing, coating, granulating, dissolving or lyophilizing.
  • An active agent as described in the preceding paragraphs can be administered alone or in combination with one or more other therapeutic agents, possible combination therapy taking the form of fixed combinations of a blocker of the Nogo-A S1 PR pathway and one or more other therapeutic agents known in treating neuronal damage. Administration can be staggered or combined agents can be given independently of one another, or in the form of a fixed combination.
  • ATI355 a human monoclonal antibody neutralizing Nogo-A (Novartis AG, Basel) is a possible combination partner.
  • Fig 1 shows binding of Nogo-A-A20 to S1 PR receptors and TSPAN3.
  • Fig 2 shows that Nogo-A-A20 inhibits cell spreading and neurite outgrowth via S1 PR2 and S1 PR5.
  • Fig 3 shows that Nogo-A-A20 signals via S1 PR2 and the G protein G13 in a S1 P- independent manner.
  • Fig 4 shows / ' n-v/Vo-treatment of damaged neurons with JTE013 and genetic depletion of S1 PR2.
  • Fig 5 S1 PR2 inhibition resembles the LTP phenotype of anti-Nogo-A treatment in hippocampal slice cultures.
  • the Nogo-A S1 PR pathway can be blocked by administration of blockers of S1 PR2 or S1 PR5, such as antibodies or antibody fragments directed against S1 PR2 or S1 PR5, small molecules that interfere with the binding of either Nogo-A to S1 PR2 or S1 PR5 or proteins downstream in the signal transduction pathway which are triggered either by the receptor or, soluble S1 PR2 or S1 PR5 or fragments thereof, agonists of S1 PR2 or S1 PR5 which lead to a down regulation of the receptor.
  • blockers of S1 PR2 or S1 PR5 such as antibodies or antibody fragments directed against S1 PR2 or S1 PR5, small molecules that interfere with the binding of either Nogo-A to S1 PR2 or S1 PR5 or proteins downstream in the signal transduction pathway which are triggered either by the receptor or, soluble S1 PR2 or S1 PR5 or fragments thereof, agonists of S1 PR2 or S1 PR5 which lead to a down regulation of the receptor.
  • Preferred blockers according to the invention are blockers of S1 PR2 or S1 PR5, such as antibodies or antibody fragments directed against S1 PR2, small molecules that interfere with the binding of Nogo-A to S1 PR2 or S1 PR5 or proteins downstream in the signal transduction pathway and triggered by the receptor and, soluble S1 PR2 or S1 PR5 or fragments thereof, agonists of S1 PR2 or S1 PR5 which lead to a down regulation of the receptor. Examples of blockers of the Nogo-A S1 PR pathway according to the invention are disclosed below.
  • Preferred blockers of the Nogo-A S1 PR pathway according to the invention are:
  • antibodies that bind to S1 PR2 or S1 PR5 antigen binding fragments of an antibody (e.g. Fab fragments) or antibody-like molecules (e.g. repeat proteins) which by binding to S1 PR2 or S1 PR5, block the binding between S1 PR2 or S1 PR5 and Nogo-A.
  • Such antibodies preferably bind to S1 PR2 or S1 PR5, in the region where Nogo-A would normally bind, but do not induce signaling;
  • antisense molecules for down regulation of S1 PR2 or S1 PR5.
  • These antisense molecules are 12-50 nucleotides in length and complementary to exons or introns of genes coding for S1 PR.
  • antisense molecules containing sequences complementary to promoter regions of these genes may be used.
  • antisense molecules binding in the 3' UTR -non translated regions of S1 PR genes are contemplated;
  • small molecules that inhibit binding of Nogo-A to S1 PR2 or S1 PR5, or small molecules which are S1 PR1 agonists and lead to down regulation of S1 PR2 or S1 PR5.
  • Small molecules contemplated are synthetic compounds of molecular weights up to 1000, which have suitable physiological activity and pharmacological properties to make them useful for application as medicaments. Such small synthetic molecules are, for example, found by the screening method of the present invention described below. Alternatively, such small molecules are designed by molecular modelling, taking into account possible binding sites of S1 PR2 or S1 PR5;
  • the most preferred blockers are:
  • ACT128800 ((2Z,5Z)-5-[[3-chloro-4-(2,3-dihydroxypropoxy)phenyl]methylene]-3-(o-tolyl)- 2-propylimino-thiazolidin-4-one, Actelion AG, Basel, Switzerland), a sphingosine 1 - phosphate receptor agonist;
  • APD334 (Arena Pharmaceuticals Inc., San Diego);
  • S1 P Receptor modulator CS0777 (Daiichi Sankyo Company); - GSK2018682 (Glaxo Smith Kline), an agonist of the S1 P1 receptor;
  • sphingosine 1 -phosphate receptor agonist chemical formula 2-amino-2-(2-(4-(3-benzyloxyphenylthio)-2- chlorophenyl)ethyl)-1 ,3-propanediol hydrochloride;
  • Nogo-A is a high affinity agonist of S1 PR2/5. Its effect can be reversed by the S1 PR2 specific blocker JTE-013 (Cayman Chemical Company, Ann Arbor, Ml).
  • Example 1 Noqo-A-A20 binds to S1 PR2, S1 PR5 and TSPAN3.
  • Nogo-A-A20 is immunoprecipitated with S1 PR2 and S1 PR5 from membrane preparations of cells overexpressing the corresponding receptor isoforms, and with TSPAN3 from total protein brain extracts.
  • Figure 1A shows the co-immunoprecipitation of Nogo-A-A20 and S1 PR2 (upper panel), S1 PR5 (middle panel) and TSPAN3 (lower blot) from total rat brain protein extracts. Additionally, data demonstrate that Tetraspanin-3 (TSPAN3) acts as a co-receptor of Nogo-A-A20 ( Figure 1A).
  • Figure 1 B shows the Label-free interaction measurements (OctedRed) to determine affinity range and constant for S1 PR1 , S1 PR2 and S1 PR5 using a recombinant Strep- tagged fusion protein of the extracellular N-terminal part of Nogo-A (rat aa 1-979) containing the Nogo-A-A20 region.
  • Recombinant S1 PR1 , S1 PR2, S1 PR5 and parental (control) membrane preparations were immobilized on amine-reactive biosensors (25 g/ml).
  • Nogo-A-ext was serially diluted and allowed to bind the saturated biosensor tips for 15 min at 1000 rpm.
  • Example 2 Nogo-A-A20 inhibition of cell spreading and neurite outgrowth can be blocked by S1 PR2 or S1 PR5 inhibition.
  • JTE-013 was screened for its ability to reverse Nogo-A-A20-induced inhibition of cell spreading of 3T3 fibroblasts.
  • 3T3 fibroblasts were plated on different concentrations of a Nogo-A-A20 substrate (20, 40 or 80 pmol/cm 2 ) in the presence of increasing concentrations of JTE-013 (Fig. 2A, first column DMSO, second column 0.1 ⁇ , third column 1 ⁇ , fourth column 10 ⁇ and fifth column 20 ⁇ , respectively).
  • DMSO was used as a vehicle control.
  • the endogenous expression level of S1 PR2 on 3T3 fibroblasts was confirmed by Fluorescence Activated Cell Sorting.
  • JTE-013 prevented the inhibitory effects of Nogo-A-A20 on cell spreading (Figure 2A).
  • the effect of JTE-013 was dose-dependent and almost completely reversed the spreading inhibition of 20 pmol / cm 2 Nogo-A-A20 at the highest concentration used in the assay (20 ⁇ ) ( Figure 2A, fifth column, respectively).
  • JTE-013 significantly rescued Nogo-A-A20-mediated cell spreading inhibition at a concentration of 1 ⁇ ( Figure 2A, third column, respectively).
  • Figure 2B (first vertical column DMSO, second column JTE-013, first horizontal line: control, second line Nogo-A- ⁇ 20, third line myelin) and Figure 2C (first column DMSO, second column JTE-013, respectively) show a rescue of Nogo-A-A20- and myelin-mediated inhibition of cell spreading after treatment of 3T3 cells with JTE-013 (1 ⁇ ).
  • Figure 2D first vertical column sh-control, second column shS1 PR2, first horizontal line control, second line Nogo-A- ⁇ 20, third line myelin
  • Figure 2E first column sh-control, second column S1 PR2 sh-RNS, respectively
  • Figure 2F first column WT, second column S1 PR2 " ' " , first line control, second line Nogo-A- ⁇ 20, third line myelin
  • Figure 2G first column WT, second column S1 PR2 " ' " , respectively
  • the following constructs targeting S1 PR2 mRNA transcript were used: ACCAAGGAGACGCTGGACATG (SEQ ID 01 ) and ACCAAGGA
  • Example 3 Nogo-A-A20 inhibits cell spreading and neurite outgrowth via G13, LARG and RhoA.
  • Nogo-A-A20 is internalised into neuronal and non-neuronal cells. This results in the formation of Nogo-A-A20-containing signalling endosomes which then activate RhoA. It was investigated whether S1 PR2 is also internalised upon treatment with Nogo-A-A20. Plasma membranes were prepared from 3T3 cells 15, 30 and 60 min after Nogo-A-A20 addition and were then analysed for S1 PR2 expression by western blot analysis. Cell membrane S1 PR2 levels were almost completely depleted 15 min after Nogo-A-A20 treatment; they remained low at 30 min and recovered to control levels after 60 min ( Figure 3A, upper panel).
  • the lower panel of Figure 3A shows the relative quantification of S1 PR2 signal intensities at 15, 30 and 60 min post Nogo-A-A20 incubation versus control.
  • Western blot analysis of ubiquitin- conjugates and non-ubiquitinated protein fractions of 3T3 cells transiently transfected with S1 PR2 shows that S1 PR2 is not ubiquitinated post Nogo-A-A20 stimulation versus control (Figure 3B).
  • Nogo-A-A20-induced activation of RhoA was significantly reversed by 1 ⁇ JTE-013 ( Figure 3C, first lane DMSO, second lane JTE-013, third lane DMSO, fourth lane JTE-013).
  • FIG. 3C A representative RhoA western blot analysis of RhoA-GTP and total RhoA 20 min after treatment with 1 ⁇ Nogo-A-A20 is shown.
  • Figure 3C first column DMSO, second column JTE-013, respectively
  • Figure 3D shows the corresponding quantification. Similar results were obtained when interfering with S1 PR2-signalling by shRNA against S1 PR2 shown by Figure 3D (first lane control, second lane D20, third lane control, fourth lane D20) and Figure 3D' (first column sh-control, second column sh-S1 PR2, respectively).
  • 3T3 fibroblasts were transfected with control (ctrl), G q , Gi 2 , Gi 3 and LARG siRNA, respectively.
  • Example 4 Micro-crush lesion of the optic nerve.
  • S1 PR2 is expressed in retinal ganglion cells.
  • the optic nerve micro-crush model was used to evaluate the effect of S1 PR2 receptor blockade or genetic depletion on axonal outgrowth in vivo ( Figure 4).
  • Gelfoam-containing JTE-013 or vehicle was applied onto the optic nerve at the time of the crush lesion and at 7 days post-injury.
  • alexa-594-coupled cholera-toxin beta subunit (CTb) was intraocularly injected the day preceding the perfusion. Axons growing across the lesion site were examined on longitudinal sections of optic nerves 2 weeks after injury.
  • JTE-013 Local in vivo application of JTE-013 is crucial to achieve the maximal pharmacologic efficacy because this compound is highly lipophilic and can be sequestered by myelin (data not shown).
  • JTE-013-soaked gelfoam onto the crushed optic nerve of adult mice ( Figure 4B).
  • Immunohistochemistry results reveal that retinal ganglion cells (RGCs) express S1 PR2 (Figure 4A).
  • Sprouting axons contained the growth-associated protein 43 (GAP-43), a marker indicating the active growth state of neurons.
  • GAP-43 growth-associated protein 43
  • This growth induction by JTE-013 was comparable to the blockade of the Chondroitin sulfate proteoglycan receptor Protein tyrosine phosphatase sigma and higher than in PirB-deficient mice.
  • Example 5 JTE013 treatment in LTP experiments in WT and Nogo-A KO acute slices.
  • Fig. 5A shows the expression pattern of S1 PR2 in the dentate gyrus (DG) and in the CA1 - CA3 regions of the hippocampus in adult BL/6 mice.
  • Hippocampal WT slices were treated with JTE-013 (open circles) or DMSO as vehicle control (black circles) (Fig. 5B).
  • LTP was induced by application of theta-burst stimulation (TBS, arrow): 10 trains of 4 pulses at 100 Hz, interburst interval of 200 ms, repeated 3x (arrow).
  • LTP long-term potentiation

Abstract

The present invention provides inhibitors capable of binding to a member of the S1PR receptor group comprised of S1PR2 and S1PR5 with a dissociation constant of 5x10-7 mol/l or smaller for treating neuronal damage, specifically an antibody, an antibody fragment, an antibody-like molecule, a nucleic acid aptamer or an oligopeptide with 6 to 30 amino acid residues in length. Similarly, an interfering RNA or an antisense modulator of gene expression of G13 or a member of the S1PR receptor group described above are provided for treating of neuronal damage.

Description

Blockers of the Nogo-A S1 PR pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair
Description
This invention relates to the treatment of diseases characterized by neuronal damage combined with lack of subsequent repair, using blockers of the Nogo-A Sphingosine 1 - phosphate receptor S1 PR pathway, particularly antibodies to, and small molecule inhibitors of, S1 PR receptors.
Damage to neuronal cells can have severe clinical outcomes depending on which neuronal cells are damaged and where. Damage to or injury of the soma or proximal part of the axon will kill neuronal cells. Damage to the distal part of the axon can only be repaired in cells of the peripheral nervous system, and in some cases even there repair is not possible.
Nogo-A is a protein identified to be an inhibitor of neurite outgrowth and cell spreading. Fragment analysis of the Nogo-A molecule has shown that the molecule has at least two inhibitory domains "Amino-Nogo" (rat aa 1-979), at the N-terminus and Nogo-66. Amino- Nogo-A contains the functionally active Nogo Δ20 fragment (rat aa 544-725). The receptor of Nogo-66, a glycosylphosphatidyl-anchored (GPI-anchored) protein, has been identified and characterized. This receptor is termed NgR1 and signals via the RhoA pathway. Besides NgR1 , other receptors homologs have been characterized, termed NgR2 and NgR3. No receptor for Amino-Nogo has been identified so far.
The objective of the instant invention is to provide safe and efficacious means for treating neuronal damage. This objective is attained by the subject-matter of the independent claims.
The present invention is based on the surprising finding that Nogo-A binds to receptors of the S1 PR family and that inhibition of these receptors suppresses the inhibitory activity of Nogo-A.
According to a first aspect of the invention, an inhibitor capable of binding to a member of the S1 PR receptor group comprised of S1 PR2 (Gene ID: 9294) and S1 PR5 (Gene ID: 53637) with a dissociation constant of 5x10"7 mol/l or smaller is provided for use in treating neuronal damage. According to an alternative aspect of the invention, an agonist of S1 PR1 (Gene ID: 1901 , S1 PR1 has an antagonistic effect on S1 PR2) is provided for use in treating neuronal damage.
Gene ID numbers in this document refer to entries in the Gene data base of the United States National Center for Biotechnology Information.
According to one embodiment, 1-(2,6-dichloro-4-pyridyl)-3-[(4-isopropyl-1 ,3-dimethyl- pyrazolo[3,4-b]pyridin-6-yl)amino]urea
Figure imgf000003_0001
is provided for use in treating neuronal damage.
Neuronal damage in the sense of the instant invention means injury to or dysfunction of a neuronal cell caused by trauma or neuronal disease resulting in constraint or disruption of the saltatory conduction by the neuronal cell within the nervous system in the absence of repair capacity. Examples of diseases characterized by neuronal damage and subsequent lack of repair include, without being restricted to, paraplegia caused by spinal cord injury, stroke, dementia, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis and paralysis and subsequent atrophy of limbs after rupture of a peripheral nerve.
Such an inhibitor according to the first aspect of the invention may be a small molecule pharmaceutical drug, an antibody, an antibody-fragment, an antibody-like molecule, a nucleic acid aptamer or an oligopeptide 6 to 30 amino acid residues in length.
Methods for generating antibodies against the members of the S1 PR receptor group are known in the art. They include, for example, immunization of mice with a S1 PR receptor or soluble parts thereof.
An antibody fragment may be the Fab domain of an antibody (the antigen binding region of an antibody) or a single chain antibody (scFv), a fusion protein consisting of the variable regions of light and heavy chains of an antibody connected by a peptide linker. An antibody-like molecule may be a repeat protein, such as a designed ankyrin repeat protein (Molecular Partners, Zurich). An antibody fragment or an antibody-like molecule may be manufactured by methods such as recombinant protein expression.
Suitable inhibitors according to the first aspect of the invention may also be developed by evolutive methods such as phage display, ribosome display or SELEX, wherein polypeptides or oligonucleotides are selected according to their binding affinity to a target of interest. Additionally, higher affinity inhibitors may be identified by reiterative rounds of evolution and selection of the amino acid sequence or nucleotide sequence.
An oligopeptide according to a first aspect of this invention may be a peptide derived from the part of Nogo-A which is recognized by a member of the S1 PR receptor group. The binding of the peptide results in inhibition of the receptor. Conversely, a peptide may also be derived from the recognition site of member of the S1 PR receptor family that competes with the receptor for Nogo-A. Binding of such an oligopeptide must not activate the signalling pathway downstream of the receptor.
The binding of inhibitors described in the preceding paragraphs results in blocking the S1 PR 2/5 receptor. By hindering the binding of Nogo-A, its biological activity is inhibited and neurite growth is no longer suppressed.
According to another aspect of the invention, a modulator of gene expression of G13 (Gene ID: 10672) or a member of the S1 PR group comprised of S1 PR2 and S1 PR5 is provided for use in treating neuronal damage.
G13 is a G-protein and strongly coupled to S1 PR2.
Such modulator may be single-stranded or double-stranded interfering ribonucleic acid oligomer of precursor thereof, comprising a sequence tract that is complementary to an mRNA molecule encoding G13 or a member of the S1 PR group described in the preceding paragraphs.
The art of silencing or "knocking down" genes, by degradation of mRNA or other effects, is well known. Examples of technologies developed for this purpose include siRNA, miRNA, shRNA, shmiRNA, and dsRNA. A comprehensive overview of this field can be found in Perrimon et al, Cold Spring Harbour Perspectives in Biology, 2010, 2, a003640.
A modulator according to the above aspect may also be an expression vector comprising a sequence encoding an interfering ribonucleic acid oligomer as described in the preceding paragraphs. Optionally, the sequence may be under the control of a promoter operable in mammalian cells. Such expression vectors facilitate production of an interfering RNA within the cell. Methods for making and using such expression vectors are known in the art.
Alternatively, a modulator according to the above aspect of the invention may be a single- stranded or double-stranded antisense ribonucleic or deoxyribonucleic acid, comprising sequences complementary to an operon expressing of a gene encoding a member of the S1 PR group described above or G13. Such operon sequences may include, without being restricted to, intron, exon, operator, ribosome binding site or enhancer sequences. Such antisense molecules may be 12-50 nucleotides in length.
According to one aspect of the invention, a soluble polypeptide for treating neuronal damage is provided, comprising a contiguous amino acid sequence of at least 30 amino acid residues taken from the protein sequence of a member of the S1 PR group. Such a soluble polypeptide can be used to inhibit Nogo-A by specifically binding to it. Specific binding in the sense of the invention means binding with dissociation constant of 5x10"7 mol/l or smaller. Optionally, the soluble polypeptide is linked to an Fc antibody domain. Such soluble polypeptide may act as a decoy receptor to capture, or to bind to and render inactive, Nogo-A or other S1 PR receptor ligands involved in neuronal damage. An example of a soluble polypeptide may be the extracellular domain of the S1 PR2 or S1 PR5 receptor linked to an Fc antibody domain.
According to one aspect of the invention a compound for use in treating neuronal damage is provided, wherein the compound is selected from the group comprised of:
a compound characterized by the general formula I (described in
WO/08154470):
Figure imgf000005_0001
wherein
R1 is a CrC12 alkyl, and
R2, R3 and R4 are each independently hydrogen, halogen, CrC6 alkyl, Ci-C6 perhaloalkyl, CrC4 perhaloalkoxy, amino, mono- or di C C4 alkylamino, C3-C7 cycloalkyl or C3-C7 cycloalkoxy, and R3 and R4 are optionally positioned at h, i, or j, but not simultaneously at the same position, wherein h, I or j denotes substitution positions in the pyrimidyl moiety, and
R5 is , halogen, C C6 alkyl, C C6 perhaloalkyl, d-C4 perhaloalkoxy, amino, mono- or di C C4 alkylamino, C3-C7 cycloalkyl or C3-C7 cycloalkoxy, and
- n is 0, 1 , 2, 3 or 4;
2-amino-2-[2-(4-octylphenyl)ethyl]propane-1 ,3-diol (Fingolimod):
Figure imgf000006_0001
5-[[3-chloro-4-(2,3-dihydroxypropoxy)phenyl]methyl]-3-(o-tolyl)-2- (propylamino)thiazolidin-4-one (ACT128800):
Figure imgf000006_0002
2-amino-2-[2-[4-(3-benzyloxyphenyl)sulfanyl-2-chloro-phenyl]ethyl]propane- 1 ,3-diol (KRP203):
Figure imgf000007_0001
1-[5-[(3R)-3-amino-4-hydroxy-3-methyl-butyl]-1-methyl-pyrrol-2-yl]-4-(p- tolyl)butan-1-one (CS-0777):
Figure imgf000007_0002
a compound characterized by a general formula III (described
2008/107436):
Figure imgf000007_0003
in free or salt form, wherein
- Q is selected from CH2, C(O), and C(S), and
Qa and Qb are independently selected from N and CH, and
is selected from C6-C15-aromatic carbocyclic groups, C5-C15-carbocyclic group, and a 4- to 15-membered heterocyclic group having one or more heteroatoms selected form the group consisting of oxygen, nitrogen and sulphur, and R1 is selected from C-i-C8-alkyl optionally substituted -OH, halogene, -CN, O- d-Ce-alkyl, NR1cR1d, carboxy-C C8-alkyl, and COOH, - C(R1aR1 b)mC(0)CR1cR1dR1 e, -C(R1aR1 b)mC(0)NR1cR1d, -C(R1aR1b)tNR1cR1d, C(R1aR1 b)mCR1cR1dR1e, and
R1 , R1g and R1h are independently selected from C-|-C8-alkyl, C-|-C8-hydroxyl, CrCs-alkylaminoiCrCs-alkyl), d C Cs-alky aminoiC Cs-alkyl), C C8 cyanalkyl, C6-C15-aromatic carbocyclic group, C3-C15-carbocyclic group, CrC8- haloalkyl and a 4- to 10- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
each R1a and R1b are independently selected from H- OH and CrC8-alkyl optionally substituted by -OH and halogen, and
R1c and R1d are independently selected from H,
C6-Ci5-aromatic carbocyclic group, C7-Ci5-aralkyl, C3-Ci5-carbocyclic group, a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
C C8-alkoxy optionally substituted by -OH, -CN, halogen, NR4R5, a C6-C15- aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
C2-C8-alkenyl optionally substituted by -OH, -CN, halogen, NR4R5, a C6-C15- aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
C2-C8-alkynyl optionally substituted by -OH, -CN, halogen, NR4R5, a C6-C15- aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
C2-C8-alkyl optionally substituted by -OH, -CN, halogen, NR4R5, a C6-Ci5- aromatic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
R1e is selected from H and CrC8-alkyl, and R3 is selected from C6-C15-aromatic carbocyclic group optionally substituted by -C(R3aR3b)nC(0)NR3cR3d or -C(R3aR3b)nC(0)OH, a C7-C15-aralkyl, C C8-alkyl substituted by a C3-C15-carbocyclic group, a C3-C15-carbocyclic group, a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, C-|-C8- alkylaminocarbonyl, di(C"|-C8-alkyl)aminocarbonyl, C-i-Cs-alkylamino, di(C-|-C8- alkyl)amino, -C(R3aR3b)nC(0)NR3cR3d and -C(R3aR3b)n(C(0)OH; and
R3a and R3b are independently selected form H, -OH and CrC8-alkyl optionally substituted by -OH and halogen; and
R3c and R3d are independently selected from H,
C6-Ci5-aromatic carbocyclic group, C3-Ci5-carbocyclic group, or R3c and R3d, together with the N atom that they are attached, form a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur,
CrCs-alkyoxy optionally substituted by a C6-C15-aromatic carbocyclic group, C3-C15-carbocyclic group and a 4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, a CrCs-alkoxycarbonyl optionally substituted by a C6- C15-aromatic carbocyclic group, C3-C15-carbocyclic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
d-Cs-alkyl optionally substituted by -OH, -CN, halogen, NR4R5, a C6-C15- aromatic carbocyclic group, C3-C15-carbocyclic group and a 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, and
R4 and R5 are independently selected from H and CrC8-alkyl, and m and n are independently selected from an integer of 0, 1 , 2 and 3, and t is integer from 1 , 2 and 3,
wherein said C6-Ci5-aromatic carbocyclic group, C7-Ci5-aralkyl, C3-Ci5-carbocyclic group and 4- to 15-membered group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, unless otherwise stated, are optionally substituted by C7-C15-aralkyl, d-C8-alkyl,CN, d-C8- alkylsulfonyl, d-C8-haloalkylsulfonyl, halogen, d-C8-alkoxy, OH, d-C8- alkylcarbonyl, -C(0)-C6-C15-aromatic carbocyclic group, -C(0)-C3-C15-carbocyclic group, -C(0)-4- to 15-membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur, d-C8-cyanoalkyl, d-C8-cyanoalkoxy, d-C8-halogenalkoxy, d-C8- alkylaminocarbonyl,
Figure imgf000010_0001
d-d-alkylamino, di(d-C8- alkyl)amino, COOH, Ci-C8-alkoxycarbonyl, d-C8-alkoxycarbonyl, 4- to 15- membered heterocyclic group having one or more heteroatoms selected from the group consisting of oxygen, nitrogen and sulphur optionally substituted by d-d5- aralkyl, Ci-C8-alkyl,CN, d-C8-alkylsulfonyl, Ci-C8-haloalkylsulfonyl, halogen, d- d-alkoxy, OH, Ci-C8-alkylcarbonyl, d-C8-cyanoalkyl, Ci-C8-cyanoalkoxy, Ci-C8- haloalkyl, Ci-C8-haloalkoxy, d-C8-alkylaminocarbonyl, di(Ci-C8- alkyl)aminocarbonyl, d-d-alkylamino, di(d-d-alkyl)amino, COOH, d-d- alkoxycarbonyl, a C6-Ci5-aromatic carbocyclic group optionally substituted by C7- Ci5-aralkyl, d-d-alkyl, CN, halogen, d-d-alkoxy, OH, d-d-alkylcarbonyl, d- d-alkylaminocarbonyl, di(d-C8-alkyl)aminocarbonyl, d-d-alkylamino, COOH, CF3 or d-C8-alkoxycarbonyl or a C3-C15-carbocyclic group optionally substituted by d-ds-aralkyl, C C8-alkyl, CN, halogen, C C8-alkoxy, OH, C C8- alkylcarbonyl, d-C8-alkylaminocarbonyl, diiCrCs-alky aminocarbonyl, C Cs- alkylamino, d d-d-alky amino, COOH, CF3 or d-C8-alkoxycarbonyl, a compound characterized by a general formula IV (described in WO
2008/142073 A1 ):
Figure imgf000010_0002
wherein
R-i aynd R5 have both. In each case, identical meanings and are d-C6-alkyl, C1-C6-alkoxy, CI, Br, or CF3, and R2 and R4 have both, in each case, identical meanings are hydrogen, C-|-C6- alkyl, d-Ce-alkoxy, CI, Br, or CF3, and
R3 is hydrogen, C1-C4-alkoxy, F, CI, CF3, or an optionally mono- or
disubstituted C-i-C8-alkyl group, the optional substitent(s) on the said alkyl group being independently selected from the group consiting of halogen, nitro, cyano, formyl, C1-C4-alkylcarbonyl, hydroxy, C1-C4-alkoxy, formyloxy, CrC4- alkoxycarbonyloxy, C1-C4-alkoxycarbonyloxy, amino, C1-C4.alkylamino, formylamino, Ci-C4-alkylcarbonyamino and Ci-C4-alkoxycarbonylamino, and
R6 is hydrogen, an optionally mono- or disubstituted CrC8-alkyl, C2-C4-alkenyl or C3-C7-cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyi group being indepentently selected from the group consisting of halogen, nitro, cyano, formyl, CrC4-alkylcarbonyl, hydroxy, CrC4-alkoxy, formyloxy, Ci-C4-alkylcarbonyloxy, CrC4-alkoxycarbonyloxy, amino, C1-C4- alkylamino, di-(Ci-C4-alkyl)amino with two identical or different Ci-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, formylamino, C1-C4- alkylcarbonylamino and Ci-C4-alkoxycarbonylamino, an optionally mono- or disubstituted heteroarylgroup, the optional substituent(s) on the said heteroaryl group being independently selected from the group consisting of halogen, nitro, cyano, formyl, C1-C4-alkylcarbonyl, hydroxy, C1-C4-alkoxy, C C4-alkyl, hydroxy-C1-C4. alkyl, C1-C4-alkoxy-C1-C4-alkyl, formyloxy, C1-C4- alkylcarbonyloxy, HO-C(=0)-, C1-C4-alkoxycarbonyloxy, amino, CrC4- alkylamino, di-(C1-C4-alkyl)amino with two identical or different C1-C4-alkyl moieties, pyrrolidyl, piperidiyl, morpholinyl, amino, -C1-C4-alkyl, CrC4- alkylamino-C1-C4-alkyl, di-(C1-C4-alkyl)-amino-C1-C4-alkyl with two identical or different C1-C4-alkyl moieties in the di-(C1-C4-alkyl)-amino moiety, pyrrolidyl- CrC4-alkyl, piperidyl- CrC4-alkyl, morpholinyl- CrC4-alkyl, formylamino, C C4-alkylcarbonylamino and Ci-C4-alkoxycarbonylamino, a heteroaryl- C1-C4- alkyl group which is optionally mono- or disubstituted on the heteroaryl moiety, the optional substituent(s) on the said heteroaryl moiety being independently selected from the group consisting of halogen, nitro, cyano, formyl, C1-C4- alkoxycarbonyl, hydroxy, CrC4-alkoxy, CrC4-alkyl, hydroxy- CrC4-alkyl, d- C4-alkoxy-Ci-C4-alkyl, formyloxy, CrC4-alkylcarbonyloxy, HO-C(=0)-, C1-C4- alkoxycarbonyl, amino, Ci-C4-alkylamino, di-( Ci-C4-alkyl)amino with two identical or different Ci-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- C1-C4-alkyl, C1-C4-alkylamino-C1-C4-alkyl, di-(C1-C4-alkyl)amino-C1-C4- alkyl with two identical or different C1-C4-alkyl moieties in the di-( C C4- alkyl)amino moiety, pyrrolidyl- C-|-C4-alkyl, piperidyl. C-|-C4-alkyl, morpholinyl- C-|-C4-alkyl, formylamino, C"|-C4-alkylcarbonylamino and C C4- alkoxycarbonylamino, an optionally mono- or disubstituted phenyl group, the optional substituent(s) on the said phenyl group being independently selected form the group consisting of cyano, formyl, C-|-C4-alkoxycarbonyl, hydroxy, hydroxy-C C4-alkyl, C C4-alkoxy, d-C^alkoxy-d-C^alkyl, HO-C(=0)-, C C4-alkoxycarbonyl, formyloxy, CrC4-alkylcarbonyloxy, CrC4- alkoxycarbonyloxy, amino, Ci-C4-alkylamino, di-(Ci-C4-alkyl)amino with two identical or different Ci-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- CrC4-alkyl, Ci-C4-alkylamino-Ci-C4-alkyl, di-(Ci-C4-alkyl)amino-Ci-C4- alkyl with two identical or different Ci-C4-alkyl moieties in the di-(CrC4- alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholinyl- CrC4-alkyl and Ci-C4-alkoxycarbonylamino, or an optionally mono- or disubstituted non-aromatic heterocyclyl group, the optional substituent (s) on the said heterocyclyl group being independently selected from the group consisting of C-|-C4-alkyl, hydroxy-C-|-C4-alkyl, C"|-C4-alkoxy-C-|-C4-alkyl, amino-C"|-C4-alkyl, Crd-aminoalkyl-C-i-C -alkyl,
Figure imgf000012_0001
alkyl with two identical or different C-i-C4-alkyl moieties in the di-(C-i-C4- alkyl)amino moiety, pyrrolidyl-C-|-C4-alkyl, piperidyl-C-|-C4-alkyl, morpholinyl- C-|-C4-alkyl, formyl, C-|-C4-alkylcarbonyl, formyloxy, C-|-C4-alkylcarbonyloxy, formylamino and C-|-C4-alkylcarbonylamino, and
R7 is hydrogen, an optionally mono- or disubstituted C-|-C8-alkyl, C2-C4-alkenyl or C3-C7-cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyi group being indepentently selected from the group consisting of halogen, nitro, cyano, formyl, Ci-C4-alkylcarbonyl, hydroxy, C1 -C4-alkoxy, formyloxy, Ci-C4-alkylcarbonyloxy, CrC4-alkoxycarbonyloxy, amino, CrC4- alkylamino, di-(Ci-C4-alkyl)amino with two identical or different Ci-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, formylamino, Ci-C4- alkylcarbonylamino and Ci-C4-alkoxycarbonylamino, an optionally mono- or disubstituted heteroaryl group, the optional substituent(s) on the said heteroaryl group being independently selected from the group consisting of halogen, nitro, cyano, formyl, CrC4-alkylcarbonyl, hydroxy, Ci-C4-alkoxy, C C4-alkyl, hydroxy-Ci-C4. alkyl, Ci-C4-alkoxy-CrC4-alkyl, CF3, formyloxy, Ci-C4- alkylcarbonyloxy, HO-C(=0)-, d-C -alkoxycarbonyl, d-C -alkoxycarbonyloxy, amino, d-d-alkylamino, di-(d-d-alkyl)amino with two identical or different C1-C4-alkyl moieties, pyrrolidyl, piperidiyl, morpholinyl, amino-d-d-alkyl, C C4-alkylamino-C-|-C4-alkyl, di-(d-d-alkyl)-amino-d-d-alkyl with two identical or different C-|-C4-alkyl moieties in the di-(C-|-C4-alkyl)-amino moiety, pyrrolidyl- C-|-C4-alkyl, piperidyl-d-d-alkyl, morpholinyl-d-d-alkyl, formylamino, C C4- alkylcarbonylamino and d-d-alkoxycarbonylamino, a heteroaryl-d-d-alkyl group which is optionally mono- or disubstituted on the heteroaryl moiety, the optional substituent(s) on the said heteroaryl moiety being independently selected from the group consisting of halogen, nitro, cyano, formyl, d-d- alkoxycarbonyl, hydroxy, Ci-C4-alkoxy, Ci-C4-alkyl, hydroxy-Ci-C4-alkyl, d- C4-alkoxy-CrC4-alkyl, formyloxy, CrC4-alkylcarbonyloxy, HO-C(=0)-, d-d- alkoxycarbonyl, Ci-C4-alkoxycarbonyloxy, amino, Ci-C4-alkylamino, di-( d-d- alkyl)amino with two identical or different Ci-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- CrC4-alkyl, Ci-C4-alkylamino-Ci-C4-alkyl, di-(d- C4-alkyl)amino-Ci-C4-alkyl with two identical or different Ci-C4-alkyl moieties in the di-( Ci-C4-alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholinyl-Ci-C4-alkyl, formylamino, Ci-C4-alkylcarbonylamino and d-d- alkoxycarbonylamino, an optionally mono- or disubstituted phenyl group, the optional substituent(s) on the said phenyl group being independently selected form the group consisting of cyano, formyl, C-|-C4-alkylcarbonyl, hydroxy, hydroxy-C C4-alkyl, C C4-alkoxy, d-d-alkoxy-d-d-alkyl, HO-C(=0)-, C C4-alkoxycarbonyl, formyloxy, d-d-alkylcarbonyloxy, C C4- alkoxycarbonyloxy, amino, d-d-alkylamino, di-(d-d-alkyl)amino with two identical or different C-i-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino-C"|-C4-alkyl, d-d-alkylamino-d-d-alkyl, di-(d-d-alkyl)amino-d-d- alkyl with two identical or different C-i-C4-alkyl moieties in the di-(d-d- alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholinyl- CrC4-alkyl and Ci-C4-alkoxycarbonylamino, or an optionally mono- or disubstituted non-aromatic heterocyclyl group, the optional substituent (s) on the said heterocyclyl group being independently selected from the group consisting of CrC4-alkyl, hydroxy-CrC4-alkyl, Ci-C4-alkoxy-CrC4-alkyl, amino-Ci-C4-alkyl, Ci-C4-aminoalkyl-Ci-C4-alkyl, di-(Ci-C4-alkyl)amino-Ci-C4- alkyl with two identical or different Ci-C4-alkyl moieties in the di-(d-d- alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholinyl- CrC4-alkyl, formyl, Ci-C4-alkylcarbonyl, formyloxy, CrC4-alkylcarbonyloxy, formylamino and C-|-C4-alkylcarbonylamino, and
R8 is hydrogen, d-d-alkyl, d-d-alkoxy, F or CI, a compound characterized by a general formula V (described in WO 2008/129029):
Figure imgf000014_0001
wherein
Ri and R5 have both, in each case, identical meanings and are CrC6-alkyl, d- C6-alkoxy, CI, F, Br or CF3, and
R2 and R4 both, in each case, identical meanings and are CrC6-alkyl, Ci-C6- alkoxy, CI, F, Br or CF3, and
R3 is hydrogen, CrC4-alkoxy, F, CI, CF3, or an mono- or disubstituted CrC8- alkyl, C2-C4-alkenyl or C3-C7-cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyl being independently selected from the group consisting of halogen, nitro, cyano, formyl C1-C4-alkylcarbonyl, hydroxy, C-|-C4- alkoxym formyloxy, C1-C4-alkylcarbonyloxy, C1-C4-alkoxycarbonyloxy, amino, C1-C4-alkylamino, di-(C1-C4-alkyl)amino, with two identical or different C1-C4- alkyl moieties, pyrrolidyl, piperidly, morpholinyl, formylamino, C-|-C4- alkycarbonylamino and C1-C4-alkyloxycarbonylamino, an optionally mono- or disubstituted heteroaryl group, the optional substituent(s) on the said heteroaryl being independently selected from the group consisting of halogen, nitro, cyano, formyl, CrC4-alkylcarbonyl, hydroxy, C1-C4-alkoxy, CrC4-alkyl, hydroxy-CrC4- alkyl, Ci-C4-alkoxy-Ci-C4-alkyl, formyloxy, CrC4-alkylcarbonyloxy, HO-C(=0)-, CrC4-alkoxycarbonyl, CrC4-alkoxycarbonyloxy, amino, Ci-C4-alkylamino, di- (Ci-C4-alkyl)amino with two identical or different CrC4-alkyl moieties, pyrrolidyl, piperidiyl, morpholinyl, amino, -CrC4-alkyl, di-(Ci-C4-alkyl)-amino-Ci-C4-alkyl with two identical or different CrC4-alkyl moieties in the di-(Ci-C4-alkyl)-amino moiety, pyrrolidyl- CrC4-alkyl, piperidyl- CrC4-alkyl, morpholinyl- CrC4-alkyl, formylamino, Ci-C4-alkylcarbonylamino and Ci-C4-alkoxycarbonylamino, a heteroaryl- CrC4-alkyl group which is optionally mono- or disubstituted on the heteroaryl moiety, the optional substituent(s) on the said heteroaryl moiety being independently selected from the group consisting of halogen, nitro, cyano, formyl, C1-C4-alkoxycarbonyl, hydroxy, C-|-C4-alkoxy, C-|-C4-alkyl, hydroxy- C C4-alkyl, C"|-C4-alkoxy-C-|-C4-alkyl, formyloxy, C-|-C4-alkylcarbonyloxy, HO- C(=0)-, CrC^alkoxycarbonyl, d-C -alkoxycarbonyloxy, amino, C C4- alkylamino, di-( d-d-alkyl)amino with two identical or different C-|-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- C-|-C4-alkyl, d-d-alkylamino- CrC4-alkyl, di-(Ci-C4-alkyl)amino-Ci-C4-alkyl with two identical or different d- C4-alkyl moieties in the di-( Ci-C4-alkyl)amino moiety, pyrrolidyl- CrC4-alkyl, piperidyl. CrC4-alkyl, morpholinyl- Ci-C4-alkyl, formylamino, d-d- alkylcarbonylamino and Ci-C4-alkoxycarbonylamino, an optionally mono- or disubstituted phenyl group, the optional substituent(s) on the said phenyl group being independently selected form the group consisting of cyano, formyl, d-d- alkoxycarbonyl, hydroxy, hydroxy-CrC4-alkyl, Ci-C4-alkoxy, d-d-alkoxy-d- C4-alkyl, HO-C(=0)-, Ci-C4-alkoxycarbonyl, formyloxy, CrC4-alkylcarbonyloxy, CrC4-alkoxycarbonyloxy, amino, Ci-C4-alkylamino, di-(Ci-C4-alkyl)amino with two identical or different C-i-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- C-|-C4-alkyl, d-d-alkylamino-d-d-alkyl, di-(d-d-alkyl)amino-d-d- alkyl with two identical or different C-i-C4-alkyl moieties in the di-(d-d- alkyl)amino moiety, pyrrolidyl-C-|-C4-alkyl, piperidyl-d-d-alkyl, morpholinyl-d- C4-alkyl and C-i-C4-alkoxycarbonylamino, or an optionally mono- or disubstituted non-aromatic heterocyclyl group, the optional substituent (s) on the said heterocyclyl group being independently selected from the group consisting of C-|-C4-alkyl, hydroxy-C-|-C4-alkyl, C"|-C4-alkoxy-C-|-C4-alkyl, armino-C-|-C4-alkyl, d-d-aminoalkyl-d-d-alkyl, di-(d-d-alkyl)amino-d-d-alkyl with two identical or different Ci-C4-alkyl moieties in the di-(Ci-C4-alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholinyl-Ci-C4-alkyl, formyl, d- C4-alkylcarbonyl, formyloxy, CrC4-alkylcarbonyloxy, formylamino and d-d- alkylcarbonylamino, and
R7 is hydrogen, an optionally mono- or disubstituted CrC8-alkyl, C2-C4-alkenyl or C3-C7-cycloalkyl group, the optional substituent(s) on the said alkyl, alkenyl or cycloalkyi group being indepentently selected from the group consisting of halogen, nitro, cyano, formyl, Ci-C4-alkylcarbonyl, hydroxy, C1 -C4-alkoxy, formyloxy, Ci-C4-alkylcarbonyloxy, CrC4-alkoxycarbonyloxy, amino, d-d- alkylamino, di-(C-|-C4-alkyl)amino with two identical or different C-|-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, formylamino, d-d- alkylcarbonylamino and C1-C4-alkoxycarbonylamino, an optionally mono- or disubstituted heteroarylgroup, the optional substituent(s) on the said heteroaryl group being independently selected from the group consisting of halogen, nitro, cyano, formyl, C1-C4-alkylcarbonyl, hydroxy, C1-C4-alkoxy, C1-C4-alkyl, hydroxy- C1-C4.alkyl, C1-C4-alkoxy-C1-C4-alkyl, formyloxy, C1-C4-alkylcarbonyloxy, HO- C(=0)-, C1-C4-alkoxycarbonyloxy, amino, C1-C4-alkylamino, di-(C C4- alkyl)amino with two identical or different C1-C4-alkyl moieties, pyrrolidyl, piperidiyl, morpholinyl, amino, -CrC4-alkyl, Ci-C4-alkylamino-Ci-C4-alkyl, di-(Ci- C4-alkyl)-amino-Ci-C4-alkyl with two identical or different d-C4-alkyl moieties in the di-(Ci-C4-alkyl)-amino moiety, pyrrolidyl- CrC4-alkyl, piperidyl- CrC4-alkyl, morpholinyl- CrC4-alkyl, formylamino, Ci-C4-alkylcarbonylamino and C1-C4- alkoxycarbonylamino, a heteroaryl- CrC4-alkyl group which is optionally mono- or disubstituted on the heteroaryl moiety, the optional substituent(s) on the said heteroaryl moiety being independently selected from the group consisting of halogen, nitro, cyano, formyl, CrC4-alkoxycarbonyl, hydroxy, CrC4-alkoxy, C C4-alkyl, hydroxy- CrC4-alkyl, Ci-C4-alkoxy-Ci-C4-alkyl, formyloxy, C1-C4- alkylcarbonyloxy, HO-C(=0)-, C1-C4-alkoxycarbonyl, C1-C4-alkoxycarbonyloxy, amino, C1-C4-alkylamino, di-( C1-C4-alkyl)amino with two identical or different C1-C4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- C1-C4-alkyl, CrC4- alkylamino-C1-C4-alkyl, di-(C1-C4-alkyl)amino-C1-C4-alkyl with two identical or different C1-C4-alkyl moieties in the di-( C1-C4-alkyl)amino moiety, pyrrolidyl- C C4-alkyl, piperidyl. C1-C4-alkyl, morpholinyl- C1-C4-alkyl, formylamino, C-|-C4- alkylcarbonylamino and C1-C4-alkoxycarbonylamino, an optionally mono- or disubstituted phenyl group, the optional substituent(s) on the said phenyl group being independently selected form the group consisting of cyano, formyl, C-|-C4- alkoxycarbonyl, hydroxy, hydroxy-CrC4-alkyl, CrC4-alkoxy, Ci-C4-alkoxy-Cr C4-alkyl, HO-C(=0)-, CrC4-alkoxycarbonyl, formyloxy, CrC4-alkylcarbonyloxy, CrC4-alkoxycarbonyloxy, amino, Ci-C4-alkylamino, di-(Ci-C4-alkyl)amino with two identical or different CrC4-alkyl moieties, pyrrolidyl, piperidyl, morpholinyl, amino- CrC4-alkyl, Ci-C4-alkylamino-Ci-C4-alkyl, di-(Ci-C4-alkyl)amino-Ci-C4- alkyl with two identical or different Ci-C4-alkyl moieties in the di-(CrC4- alkyl)amino moiety, pyrrolidyl-Ci-C4-alkyl, piperidyl-CrC4-alkyl, morpholidyl-d- C4-alkyl and Ci-C4-alkoxycarbonylamino, or an optionally mono- or disubstituted non-aromatic heterocyclyl group, the optional substituent (s) on the said heterocyclyl group being independently selected from the group consisting of C1-C4-alkyl, hydroxy-C1-C4-alkyl, C1-C4-alkoxy-C1-C4-alkyl, amino-C1-C4-alkyl, C1-C4-aminoalkyl-C1-C4-alkyl,
Figure imgf000017_0001
with two identical or different C-i-C4-alkyl moieties in the di-(C"i-C4-alkyl)amino moiety, pyrrolidyl-CrC -alkyl, piperidyl-CrC -alkyl, morpholinyl-d-C -alkyl, formyl, C C4-alkylcarbonyl, formyloxy, C-|-C4-alkylcarbonyloxy, formylamino and C C4- alkylcarbonylamino, and
- R8 is hydrogen, Ci-C4-alkyl, CrC4-alkoxy, F or CI, and
- X is CH or N,
In free form or salt,
2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-quinazolin-3- yl]acetic acid (WO 2008/107436):
Figure imgf000017_0002
N-(5-chloro-2,4-dimethoxy-phenyl)-2-[2,4-dioxo-3-[2-oxo-2-[2-(3- pyridyl)ethylamino]ethyl]quinazolin-1-yl]acetamide acetamide (WO
2008/107436):
Figure imgf000017_0003
2-[4-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-quinazolin- yl]phenyl]-N-phenethyl-acetamide (WO 2008/107436):
Figure imgf000018_0001
4-[6-chloro-1-[2-(3-chloro-4-ethoxy-phenyl)-2-oxo-ethyl]-2,4-dioxo-quinazolin yl]-N-cyclopentyl-butanamide (WO 2008/107436):
Figure imgf000018_0002
N-(5-chloro-2,4-dimethoxy-phenyl)-2-[2,4-dioxo-3-[2-oxo-2-
(phenethylamino)ethyl]quinazolin-1-yl]acetamide (WO 2008/107436):
Figure imgf000019_0001
- tert-butyl 2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo- quinazolin-3-yl]acetate (WO 2008/107436):
Figure imgf000019_0002
- tert-butyl N-[2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo- quinazolin-3-yl]ethyl]carbamate (WO 2008/107436):
Figure imgf000020_0001
2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-pyrido[3,2- d]pyrimidin-3-yl]acetic acid (WO 2008/107436):
Figure imgf000020_0002
2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2-oxo-4H-quinazolin yl]acetic acid (WO 2008/107436):
Figure imgf000020_0003
N-(5-chloro-2,4-dimethoxy-phenyl)-2-[3-(3-methoxybenzoyl)-7-methyl-4- 4a!8a-dihydro-1 ,8-naphthyridin-1 -yl]acetamide (WO 2008/107436):
Figure imgf000021_0001
- 2-[1-[2-[(2,6-dichloro-4-pyridyl)amino]-2-oxo-ethyl]-5-methyl-2,4-dioxo- quinazolin-3-yl]acetic acid (WO 2008/107436):
Figure imgf000021_0002
- 4-methyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1 -one (WO 2008/129029):
Figure imgf000021_0003
- 4-methyl-8-(2!4!6-trimethylanilino)-2H-isoquinolin-1 -one (WO 2008/129029):
Figure imgf000022_0001
- 8-(2,6-dimethylanilino)-2H-isoquinolin-1 -one (WO 2008/129029):
Figure imgf000022_0002
- 8-(4-fluoro-2,6-dimethyl-anilino)-4-methyl-2H-phthalazin-1 -one (WO
2008/129029):
Figure imgf000022_0003
- 4-ethyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1 -one (WO 2008/129029):
Figure imgf000023_0001
- 4-isopropyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1-one (WO 2008/129029):
Figure imgf000023_0002
- 4-(2-hydroxyethyl)-8-(2!4,6-trimethylanilino)-2H-phthalazin-1-one (WO
2008/129029):
Figure imgf000023_0003
- 8-(2,6-diethyl-4-fluoro-anilino)-4-methyl-2H-phthalazin-1 -one (WO 2008/129029):
Figure imgf000024_0001
- 8-(4-chloro-2!6-dimethyl-anilino)-4-methyl-2H-phthalazin-1-one (WO 2008/129029):
Figure imgf000024_0002
- 4-ethyl-8-(4-fluoro-2,6-dimethyl-anilino)-2H-phthalazin-1 -one (WO 2008/129029):
Figure imgf000024_0003
5-(2-propylpyrazol-3-yl)-2-(2,4,6-trimethylanilino)benzamide (WO
2008/142073):
Figure imgf000025_0001
- 5-methoxy-2-(2!4,6-trimethylanilino)benzamide (WO 2008/142073):
Figure imgf000025_0002
- 5-chloro-2-(2!4,6-trimethylanilino)benzamide (WO 2008/142073):
Figure imgf000025_0003
According to one aspect of the invention, a pharmaceutical composition for treating neuronal damage is provided, comprising an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention. Pharmaceutical compositions for enteral administration, such as nasal, buccal, rectal or, especially, oral administration, and for parenteral administration, such as subcutaneous, intravenous, intrahepatic or intramuscular administration, are preferred. The pharmaceutical compositions comprise from approximately 1 % to approximately 95% active ingredient, preferably from approximately 20% to approximately 90% active ingredient.
Similarly, a dosage form for treating neuronal damage is provided, comprising an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention. Optionally, a pharmaceutical carrier or excipient may be present.
For parenteral administration preference is given to the use of solutions of active agents like an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention, and also suspensions or dispersions, especially isotonic aqueous solutions, dispersions or suspensions which, for example, can be prepared shortly before use. Pharmaceutical compositions may be sterilized and/or comprise excipients, for example preservatives, stabilizers, wetting agents and/or emulsifiers, solubilizers, viscosity-increasing agents, salts for regulating osmotic pressure and/or buffers and are prepared in a manner known per se, for example by means of conventional dissolving and lyophilizing processes.
For oral pharmaceutical preparations suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates. Binders, such as starches, cellulose derivatives and/or polyvinylpyrrolidone may also be employed, Disintegrators, flow conditioners and lubricants, for example stearic acid or salts thereof and/or polyethylene glycol may be added in addition or instead. Tablet cores can be provided with suitable, optionally enteric, coatings. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes to indicate different types or doses of active ingredient. Pharmaceutical compositions for oral administration also include hard capsules consisting of gelatin, and soft, sealed capsules consisting of gelatin and a plasticizer, such as glycerol or sorbitol. Capsules may contain the active ingredient in the form of granules, or dissolved or suspended in suitable liquid excipients, such as in oils.
Transdermal/intraperitoneal and intravenous applications are also considered, for example using a transdermal patch, which allows administration over an extended period of time, e.g. from one to twenty days. Intravenous or subcutaneous applications are particularly preferred as well as intrathecal applications.
According to one aspect of the invention, a method for treating neuronal damage is provided, comprising the administration of an inhibitor, modulator, soluble polypeptide or compound according to the above aspect of the invention to a patient in need thereof.
The treatment may be for prophylactic or therapeutic purposes. For administration, an active agent as described in the preceding paragraphs is preferably provided in the form of a pharmaceutical preparation comprising the active agent in chemically pure form and optionally a pharmaceutically acceptable carrier and optionally adjuvants. The active agent is used in an amount effective against the diseases characterized by neuronal damage and lack of subsequent repair. The dosage of the active agent depends upon the species, the patient age, weight, and individual condition, the individual pharmacokinetic data, mode of administration, and whether the administration is for prophylactic or therapeutic purposes. For individual having a body weight of about 70 kg, the daily dose administered ranges from approximately 0.1 mg/kg to approximately 1000 mg/kg, preferably from approximately 0.5 mg to approximately 100 mg/kg, of an active agent.
According to one aspect of the invention, a method of manufacturing a medicament for treating neuronal damage is provided comprising the use of an inhibitor, modulator, soluble polypeptide or compound according to the above aspects of the invention. Medicaments according to the invention are manufactured by methods known in the art, especially by conventional mixing, coating, granulating, dissolving or lyophilizing.
An active agent as described in the preceding paragraphs can be administered alone or in combination with one or more other therapeutic agents, possible combination therapy taking the form of fixed combinations of a blocker of the Nogo-A S1 PR pathway and one or more other therapeutic agents known in treating neuronal damage. Administration can be staggered or combined agents can be given independently of one another, or in the form of a fixed combination.
ATI355, a human monoclonal antibody neutralizing Nogo-A (Novartis AG, Basel) is a possible combination partner.
Description of the figures:
Fig 1 shows binding of Nogo-A-A20 to S1 PR receptors and TSPAN3.
Fig 2 shows that Nogo-A-A20 inhibits cell spreading and neurite outgrowth via S1 PR2 and S1 PR5. Fig 3 shows that Nogo-A-A20 signals via S1 PR2 and the G protein G13 in a S1 P- independent manner.
Fig 4 shows /'n-v/Vo-treatment of damaged neurons with JTE013 and genetic depletion of S1 PR2.
Fig 5: S1 PR2 inhibition resembles the LTP phenotype of anti-Nogo-A treatment in hippocampal slice cultures.
The Nogo-A S1 PR pathway can be blocked by administration of blockers of S1 PR2 or S1 PR5, such as antibodies or antibody fragments directed against S1 PR2 or S1 PR5, small molecules that interfere with the binding of either Nogo-A to S1 PR2 or S1 PR5 or proteins downstream in the signal transduction pathway which are triggered either by the receptor or, soluble S1 PR2 or S1 PR5 or fragments thereof, agonists of S1 PR2 or S1 PR5 which lead to a down regulation of the receptor.
Preferred blockers according to the invention are blockers of S1 PR2 or S1 PR5, such as antibodies or antibody fragments directed against S1 PR2, small molecules that interfere with the binding of Nogo-A to S1 PR2 or S1 PR5 or proteins downstream in the signal transduction pathway and triggered by the receptor and, soluble S1 PR2 or S1 PR5 or fragments thereof, agonists of S1 PR2 or S1 PR5 which lead to a down regulation of the receptor. Examples of blockers of the Nogo-A S1 PR pathway according to the invention are disclosed below.
Preferred blockers of the Nogo-A S1 PR pathway according to the invention are:
- soluble S1 PR2 or S1 PR5 or fragments thereof;
- antibodies that bind to S1 PR2 or S1 PR5, antigen binding fragments of an antibody (e.g. Fab fragments) or antibody-like molecules (e.g. repeat proteins) which by binding to S1 PR2 or S1 PR5, block the binding between S1 PR2 or S1 PR5 and Nogo-A. Such antibodies preferably bind to S1 PR2 or S1 PR5, in the region where Nogo-A would normally bind, but do not induce signaling;
- Virus-like particles loaded with immunogenic sequences of S1 PR2 or S1 PR5, and therefore capable of inducing an antibody response directed against these molecules with the effect of blocking their biological activity;
- antisense molecules for down regulation of S1 PR2 or S1 PR5. These antisense molecules are 12-50 nucleotides in length and complementary to exons or introns of genes coding for S1 PR. Moreover, antisense molecules containing sequences complementary to promoter regions of these genes may be used. Finally, antisense molecules binding in the 3' UTR -non translated regions of S1 PR genes are contemplated;
- small molecules that inhibit binding of Nogo-A to S1 PR2 or S1 PR5, or small molecules which are S1 PR1 agonists and lead to down regulation of S1 PR2 or S1 PR5. Small molecules contemplated are synthetic compounds of molecular weights up to 1000, which have suitable physiological activity and pharmacological properties to make them useful for application as medicaments. Such small synthetic molecules are, for example, found by the screening method of the present invention described below. Alternatively, such small molecules are designed by molecular modelling, taking into account possible binding sites of S1 PR2 or S1 PR5;
- proteins and protein analogs which by binding to S1 PR2 or S1 PR5, prevent binding of Nogo-A to S1 PR2 or S1 PR5, for example, synthetic proteins or protein analogs which mimic the variable region scFv of binding and/or neutralizing antibodies, or antibodies that mimic a binding pocket for S1 PR2 or S1 PR5.
The most preferred blockers are:
- JTE-013 (Cayman Chemical Company, Ann Arbor, Ml, USA), a S1 PR2 blocker;
- S1 P2 Receptor Antagonists as described in WO 08/154470;
- Gilenia (Novartis AG, Basel, Switzerland), a sphingosine 1 -phosphate receptor agonist, acting on the four Sphingosine 1-Phosphate Receptors S1 P1 , S1 P3, S1 P4 and S1 P5 and leading to a down regulation of S1 PR, chemical formula 2-amino-2-[2-(4- octylphenyl)ethyl]propane-1 ,3-diol hydrochloride;
- ACT128800 ((2Z,5Z)-5-[[3-chloro-4-(2,3-dihydroxypropoxy)phenyl]methylene]-3-(o-tolyl)- 2-propylimino-thiazolidin-4-one, Actelion AG, Basel, Switzerland), a sphingosine 1 - phosphate receptor agonist;
- BAF312 (Novartis AG, Basel, Switzerland), a sphingosine 1 -phosphate receptor modulator;
- ON04641 (Ono Pharmaceutical Co., Ltd., Merck-Serono SA), a sphingosine 1 - phosphate receptor agonist;
- EXEL-9953 (Exelexis Inc.) a sphingosine-1-phosphate type 1 receptor (S1 P1 ) agonist;
- S1 P receptor agonist APD334 (Arena Pharmaceuticals Inc., San Diego);
- S1 P Receptor modulator CS0777 (Daiichi Sankyo Company); - GSK2018682 (Glaxo Smith Kline), an agonist of the S1 P1 receptor;
- S1 P Receptor Agonist BIOGEN (Biogen Idee Inc., Boston);
- KRP203 (Kyorin Co. Ltd. and Novartis AG, Basel), a sphingosine 1 -phosphate receptor agonist, chemical formula 2-amino-2-(2-(4-(3-benzyloxyphenylthio)-2- chlorophenyl)ethyl)-1 ,3-propanediol hydrochloride;
- RPC1063 (Receptos, Inc.), a S1 P1 agonist;
- Bicyclic organic compounds as described in WO 2008/107436 (Novartis AG);
- Phtalazine and isoquinoline derivatives as described in WO 2008/129029 (Novartis AG);
- Benzamides as described in WO 2008/142073 (Allergan, Inc.).
Concepts and Evidence behind the Invention
This invention is based on the finding that Nogo-A is a high affinity agonist of S1 PR2/5. Its effect can be reversed by the S1 PR2 specific blocker JTE-013 (Cayman Chemical Company, Ann Arbor, Ml).
Examples:
Example 1 : Noqo-A-A20 binds to S1 PR2, S1 PR5 and TSPAN3.
Nogo-A-A20 is immunoprecipitated with S1 PR2 and S1 PR5 from membrane preparations of cells overexpressing the corresponding receptor isoforms, and with TSPAN3 from total protein brain extracts. Figure 1A shows the co-immunoprecipitation of Nogo-A-A20 and S1 PR2 (upper panel), S1 PR5 (middle panel) and TSPAN3 (lower blot) from total rat brain protein extracts. Additionally, data demonstrate that Tetraspanin-3 (TSPAN3) acts as a co-receptor of Nogo-A-A20 (Figure 1A).
Figure 1 B shows the Label-free interaction measurements (OctedRed) to determine affinity range and constant for S1 PR1 , S1 PR2 and S1 PR5 using a recombinant Strep- tagged fusion protein of the extracellular N-terminal part of Nogo-A (rat aa 1-979) containing the Nogo-A-A20 region. Recombinant S1 PR1 , S1 PR2, S1 PR5 and parental (control) membrane preparations were immobilized on amine-reactive biosensors (25 g/ml). Nogo-A-ext was serially diluted and allowed to bind the saturated biosensor tips for 15 min at 1000 rpm. Nogo-A-ext bound stronger to S1 PR2 (-142 nM) and S1 PR5 (-109 nM) than to S1 PR1 (-382 nM). Nogo-A-ext binding to parental membrane preparations and blank surfaces was minimal and subtracted from all values. Data shown are mean ± SEM (n = 5). Using BioLayer interferometry technology, it was demonstrated for the first time that Amino-Nogo binds to S1 PR1 , S1 PR2 and S1 PR5. The Biolayer interferometry technology is a label-free technology for measuring biomolecule interactions and described in detail by Witte et al. (Comb. Chem. High Throughput Screen, 12(8), 791 -800, 2009). Binding affinities are strong and vary between the different isoforms (Figure 1 B). They can be ordered as follows: S1 PR5 > S1 PR2 > S1 PR1 .
Example 2: Nogo-A-A20 inhibition of cell spreading and neurite outgrowth can be blocked by S1 PR2 or S1 PR5 inhibition.
JTE-013 was screened for its ability to reverse Nogo-A-A20-induced inhibition of cell spreading of 3T3 fibroblasts. 3T3 fibroblasts were plated on different concentrations of a Nogo-A-A20 substrate (20, 40 or 80 pmol/cm2) in the presence of increasing concentrations of JTE-013 (Fig. 2A, first column DMSO, second column 0.1 μΜ, third column 1 μΜ, fourth column 10 μΜ and fifth column 20 μΜ, respectively). DMSO was used as a vehicle control. The endogenous expression level of S1 PR2 on 3T3 fibroblasts was confirmed by Fluorescence Activated Cell Sorting. At 1 μΜ, JTE-013 prevented the inhibitory effects of Nogo-A-A20 on cell spreading (Figure 2A). The effect of JTE-013 was dose-dependent and almost completely reversed the spreading inhibition of 20 pmol / cm2 Nogo-A-A20 at the highest concentration used in the assay (20 μΜ) (Figure 2A, fifth column, respectively). JTE-013 significantly rescued Nogo-A-A20-mediated cell spreading inhibition at a concentration of 1 μΜ (Figure 2A, third column, respectively). Figure 2B (first vertical column DMSO, second column JTE-013, first horizontal line: control, second line Nogo-A- Δ20, third line myelin) and Figure 2C (first column DMSO, second column JTE-013, respectively) show a rescue of Nogo-A-A20- and myelin-mediated inhibition of cell spreading after treatment of 3T3 cells with JTE-013 (1 μΜ). Figure 2D (first vertical column sh-control, second column shS1 PR2, first horizontal line control, second line Nogo-A- Δ20, third line myelin), Figure 2E (first column sh-control, second column S1 PR2 sh-RNS, respectively) and Figure 2F (first column WT, second column S1 PR2"'", first line control, second line Nogo-A- Δ20, third line myelin), Figure 2G (first column WT, second column S1 PR2"'", respectively) show the same rescue effects of Nogo-A-A20- and myelin- mediated inhibition of cell spreading after knockdown of S1 PR2 using an shRNA or a DNA for lentiviral expression (data not shown) against S1 PR2 in 3T3 cells or transgenic depletion of S1 PR2 in mouse embryonic fibroblasts. The following constructs targeting S1 PR2 mRNA transcript were used: ACCAAGGAGACGCTGGACATG (SEQ ID 01 ) and ACCAAGGAGACGCUGGACAUG (SEQ ID 02).
Postnatal day (P) 8 rat cerebellar granule neurons (CGNs) are frequently used as a model to study neurite outgrowth mechanisms in vitro. S1 PR2 is expressed on the cell surface of CGNs. There was a potent inhibition of neurite outgrowth of CGNs plated on a Nogo-A- Δ20 substrate (20 pmol / cm2; Figure 2H, I). In the presence of JTE-013, neurite outgrowth was significantly increased, suggesting that S1 PR2 plays a major role in Nogo-A- A20- mediated outgrowth inhibition (Figure 2H, first column control, second column JTE-013, third column a-S1 PR5, respectively). Using CGNs from S1 PR2 KO mice in this assay resulted in a similar phenotype of increased neurite outgrowth (Figure 2I, first column WT, second column S1 PR5 KO, respectively). Furthermore, inhibition of S1 PR5 by using a function-blocking antibody resulted in increased neurite outgrowth as well, indicating, that S1 PR5 may also contribute to in Nogo-A- A20-mediated outgrowth inhibition (Figure 2H). Data shown are mean ± SEM (n > 3). * p < 0.05, ** p < 0.01 , *** p < 0.001 ; unpaired Student's t-test.
Example 3: Nogo-A-A20 inhibits cell spreading and neurite outgrowth via G13, LARG and RhoA.
It has been shown recently that surface bound Nogo-A-A20 is internalised into neuronal and non-neuronal cells. This results in the formation of Nogo-A-A20-containing signalling endosomes which then activate RhoA. It was investigated whether S1 PR2 is also internalised upon treatment with Nogo-A-A20. Plasma membranes were prepared from 3T3 cells 15, 30 and 60 min after Nogo-A-A20 addition and were then analysed for S1 PR2 expression by western blot analysis. Cell membrane S1 PR2 levels were almost completely depleted 15 min after Nogo-A-A20 treatment; they remained low at 30 min and recovered to control levels after 60 min (Figure 3A, upper panel). The lower panel of Figure 3A shows the relative quantification of S1 PR2 signal intensities at 15, 30 and 60 min post Nogo-A-A20 incubation versus control. Western blot analysis of ubiquitin- conjugates and non-ubiquitinated protein fractions of 3T3 cells transiently transfected with S1 PR2 shows that S1 PR2 is not ubiquitinated post Nogo-A-A20 stimulation versus control (Figure 3B). Nogo-A-A20-induced activation of RhoA was significantly reversed by 1 μΜ JTE-013 (Figure 3C, first lane DMSO, second lane JTE-013, third lane DMSO, fourth lane JTE-013). A representative RhoA western blot analysis of RhoA-GTP and total RhoA 20 min after treatment with 1 μΜ Nogo-A-A20 is shown. Figure 3C (first column DMSO, second column JTE-013, respectively) shows the corresponding quantification. Similar results were obtained when interfering with S1 PR2-signalling by shRNA against S1 PR2 shown by Figure 3D (first lane control, second lane D20, third lane control, fourth lane D20) and Figure 3D' (first column sh-control, second column sh-S1 PR2, respectively). 3T3 fibroblasts were transfected with control (ctrl), Gq, Gi2, Gi3 and LARG siRNA, respectively. Knockdown of Gi3 and LARG abolished Nogo-A-A20-mediated cell spreading inhibition (Figure 3E, first column control siRNA, second column Gq siRNA, third column G12 siRNA, fourth column G13 siRNA, fifth column G12 siRNA + JTE-013, sixth column LARG siRNA, seventh column control, eighth column PTX). Combined treatment with siRNA against G13 and JTE-013 did not result in more cell spreading than siRNA treatment against G13 alone. Furthermore, arresting the Gi subunit with Pertussis toxin (PTX) in its GDP-bound, inactive state did not influence cell spreading behaviour on Nogo-A-A20 substrate (Figure 3E). Competitive ELISA quantification of S1 P levels in cells and corresponding media. S1 P levels did not significantly change in CGNs and 3T3 cells treated with Nogo-A-A20 versus control (Figure 3F, circles Nogo- Α- Δ20; S1 P medium, triangles Nogo- Α- Δ20; cellular S1 P, dashed line control). Data shown are mean ± SEM (n > 3). * p < 0.05, ** p < 0.01 ; unpaired Student's t-test.
Example 4: Micro-crush lesion of the optic nerve.
S1 PR2 is expressed in retinal ganglion cells. The optic nerve micro-crush model was used to evaluate the effect of S1 PR2 receptor blockade or genetic depletion on axonal outgrowth in vivo (Figure 4). Gelfoam-containing JTE-013 or vehicle was applied onto the optic nerve at the time of the crush lesion and at 7 days post-injury. To visualise optic axons, alexa-594-coupled cholera-toxin beta subunit (CTb) was intraocularly injected the day preceding the perfusion. Axons growing across the lesion site were examined on longitudinal sections of optic nerves 2 weeks after injury. The number of axons growing past the lesion site was significantly higher after JTE-013 treatment or S1 PR2 genetic depletion compared to control optic nerves (Fig. 4 C and D). White stars indicate the lesion site. Higher magnification shows that the density of axons extending in the distal optic nerves appeared bigger in the JTE-013 and S1 PR2 genetic depletion group than in control mice. Data shown in Figure D (first column vehicle [n=5], second column JTE-013 [n=7], third column S1 PR2 KO [n=6], respectively) are mean ± SEM. * p < 0.05; ** p < 0.01 ; unpaired Student's t-test. Scale bar: C: 100 μηι.
Local in vivo application of JTE-013 is crucial to achieve the maximal pharmacologic efficacy because this compound is highly lipophilic and can be sequestered by myelin (data not shown). To study the effects of S1 PR2 blockade on axonal outgrowth in vivo, we applied JTE-013-soaked gelfoam onto the crushed optic nerve of adult mice (Figure 4B). Immunohistochemistry results reveal that retinal ganglion cells (RGCs) express S1 PR2 (Figure 4A). Two weeks after injury, JTE-013 treatment or genetic depletion induced significantly more axonal growth past the lesion site than the vehicle control (Figure 4 C and D). More axons extended in the distal segment of optic nerves treated with JTE-013 than in those receiving the vehicle (Figure 4 C and D). Sprouting axons contained the growth-associated protein 43 (GAP-43), a marker indicating the active growth state of neurons. This growth induction by JTE-013 was comparable to the blockade of the Chondroitin sulfate proteoglycan receptor Protein tyrosine phosphatase sigma and higher than in PirB-deficient mice.
Example 5: JTE013 treatment in LTP experiments in WT and Nogo-A KO acute slices.
Fig. 5A shows the expression pattern of S1 PR2 in the dentate gyrus (DG) and in the CA1 - CA3 regions of the hippocampus in adult BL/6 mice. Hippocampal WT slices were treated with JTE-013 (open circles) or DMSO as vehicle control (black circles) (Fig. 5B). LTP was induced by application of theta-burst stimulation (TBS, arrow): 10 trains of 4 pulses at 100 Hz, interburst interval of 200 ms, repeated 3x (arrow). 60 min after TBS, a significant difference between JTE-013 and DMSO treatment could be observed (average level 60 min after TBS: JTE-013: 184 ± 8%; DMSO: 151 ± 8.5%; p = 0.018). Hippocampal Nogo-A KO slices were treated with JTE-013 (open circles) or DMSO (black circles) (Fig. 5C. LTP induced by TBS (arrow revealed no significant differences in Nogo-A KO mice treated with JTE-013 (average level 60 min after TBS: JTE-013: 180 ± 12%) and DMSO (181 ± 13%). Inserts show original traces from representative individual experiments; letters correspond to the time point at which the traces were taken. * p < 0.05; unpaired Student's t-test. Scale bar: A: 300 μηι (left), 30 μηι (right).
It was recently demonstrated that long-term potentiation (LTP) of CA3-CA1 synapses in mouse hippocampus was significantly increased by acute, antibody-mediated neutralisation of neuronal Nogo-A or NgR1 . LTP was analysed in the presence of the S1 PR2 specific blocker JTE-013 in WT and Nogo-A KO mice. Long-term potentiation (LTP) was significantly increased by JTE-013 treatment in WT (Figure 5B), but not Nogo- A KO mice (Figure 5C). These results suggest a functional Nogo-A-S1 PR2 interaction which restricts hippocampal synaptic plasticity independently of S1 P. Baseline synaptic transmission was not affected by the treatment with JTE-013.

Claims

Claims:
1. 1-(2,6-dichloro-4-pyridyl)-3-[(4-isopropyl-1 ,3-dimethyl-pyrazolo[3,4-b]pyridin-6- yl)amino]urea for use in prevention or therapy neuronal damage.
Figure imgf000035_0001
2. An inhibitor capable of binding to a member of the S1 PR receptor group comprised of S1 PR2 and S1 PR5 with a dissociation constant of 5x10"7 mol/l or smaller, comprising an antibody, an antibody fragment, an antibody-like molecule, a nucleic acid aptamer or an oligopeptide with 6 to 30 amino acid residues in length, for use in treating neuronal damage.
3. A modulator of gene expression of G13 or a member of the S1 PR receptor group comprised of S1 PR2 and S1 PR5 comprising
- a single-stranded or double-stranded interfering ribonucleic acid oligomer or precursor thereof, comprising a sequence tract complementary to an mRNA molecule encoding any member of said S1 PR group or G13; or
- an expression vector, comprising a sequence encoding said interfering
ribonucleic acid oligomer or precursor thereof, or
a single-stranded or double-stranded antisense ribonucleic or deoxyribonucleic acid, comprising sequences complementary to a regulatory region of a gene encoding a member of said S1 PR group or G13.
for use in treating neuronal damage.
4. A soluble polypeptide comprising a contiguous amino acid sequence of at least 30 amino acids comprised within the protein sequence of a member of the S1 PR receptor group comprised of S1 PR2 and S1 PR5 for use in treating neuronal injury.
5. A compound for use in treating neuronal damage selected from the group
comprised of a compound characterized by the general formula I:
Figure imgf000036_0001
- R1 is a C1-C12 alkyl, and
R2, R3 and R4 are each independently hydrogen, halogen, C1 -C6 alkyl, C1-C6 perhaloalkyi, C1 -C4 perhaloalkoxy, amino, mono- or di C1-C4 alkylamino, C3-C7 cycloalkyl or C3-C7 cycloalkoxy, and
R3 and R4 are optionally positioned at h, i, or j, but not simultaneously at the same position, and
- R5 is , halogen, C1 -C6 alkyl, C1 -C6 perhaloalkyi, C1-C4 perhaloalkoxy, amino, mono- or di C1-C4 alkylamino, C3-C7 cycloalkyl or C3-C7 cycloalkoxy, and
- n is 0, 1 , 2, 3 or 4;
2-amino-2-[2-(4-octylphenyl)ethyl]propane-1 ,3-diol:
Figure imgf000036_0002
5-[[3-chloro-4-(2,3-dihydroxypropoxy)phenyl]methyl]-3-(o-tolyl)-2- (propylamino)thiazolidin-4-one:
Figure imgf000037_0001
2-amino-2-[2-[4-(3-benzyloxyphenyl)sulfanyl-2-chloro-phenyl]ethyl]propane- 1 ,3-diol:
Figure imgf000037_0002
1-[5-[(3R)-3-amino-4-hydroxy-3-methyl-butyl]-1-methyl-pyrrol-2-yl]-4-(p- tolyl)butan-1-one:
Figure imgf000037_0003
[3-amino-4-(3-octylanilino)-4-oxo-butyl]phosphonic acid:
Figure imgf000038_0001
5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)^
oxadiazole:
Figure imgf000038_0002
a compound characterized by a general formula II:
Figure imgf000038_0003
wherein,
- X is NRaRb, SRb, F, CI, Br or I, and
- R1 is H or Rb
- R2 is H, F, CI, Br, I, or Rb
- Ra is H or Rb, and
Rb is branched or linear alkyl having 1 to 12 carbon atoms, wherein one or more, preferably 1 to 7 hydrogen atoms may be replaced by F, CI, Br, I, ORa, COOR3, CN, N(Ra)2 and wherein one or more, preferably 1 to 7 non- adjacent CH2-group may be replaced by O, NRa, S or S02, and/or by -CH=CH- groups, or is cycloalkyl or cycloalkylalkylene having 3 to 7 ring carbon atoms, and
- W is C=0, C=S, S02 or SO, and
- Q is NR3, -O- or -S-, and
R is hydrogen, Rb, Ar or Het, and Ar is a monocyclic or bicyclic, saturated, unsaturated or aromatic carbocyclic ring having 6 to 14 carbon atoms which may be unsubstituted, mono-, di-, or tri- substituted by F, CI, Br, I, Rb, OR3, -[C(R3)2]n-OR3, N(R3)2, -[C(R3)2]n-N(R3)2, N02, CN, COOR3, CF3, OCF3, CON(R3), NR3COA, NR3CON(R3)2, -[C(R3)2]n-Het, -[C(R3)2]n-Ar, -[C(R3)2]n- cycloalkyl, -[C(R3)2]n-CON(R3)2, -[(R3)2]n-COOR3, -[C(R3)2]n-NR3- [C(R3)2]n-C02R3; -[C(R3)2]n-NR3-[C(R3)2]n-OR3, -S02-[C(R3)2]n-C02R3, - S02-N(R3)2]n-[C02R3, -[C(R3)2]N-S02-[C(R3)]n-C02R3, -S02[C(R3)2]n- OR3, -S02N(R3)2-[C(R3)2]n-OR3, -[C(R3)2]N-S02-[C(R3)2]n-OR3,
NR3CON(R3)2, NR3S02Rb, COR3, S02N(R3)2, S02N(R3)Rb, SORb, SONR3Rb, S02Rb, and/or -0[C(R3)2]n-COOR3 and
Het is a monocyclic or bicyclic, saturated, unsaturated or aromatic heterocyclic ring having 1 to 4 N, O and/or S which may be unsubstituted, mono-, di-, or trisubstituted by F, CI, Br, I, Rb, OR3, -[C(R3)2]n-OR3, N(R3)2, -[C(R3)2]n-N(R3)2, N02, CN, COOR3, CF3, OCF3, CON(R3), NR3COA, NR3CON(R3)2, -[C(R3)2]n-Het, -[C(R3)2]n-Ar, -[C(R3)2]n- cycloalkyl, -[C(R3)2]n-CON(R3)2, -[(R3)2]n-COOR3, -[C(R3)2]n-NR3- [C(R3)2]n-C02R3; -[C(R3)2]n-NR3-[C(R3)2]n-OR3, -S02-[C(R3)2]n-C02R3, - S02-N(R3)2]n-[C02R3, -[C(R3)2]N-S02-[C(R3)]n-C02R3, -S02[C(R3)2]n- OR3, -S02N(R3)2-[C(R3)2]n-OR3, -[C(R3)2]N-S02-[C(R3)2]n-OR3,
NR3CON(R3)2, NR3S02Rb, COR3, S02N(R3)2, S02N(R3)Rb, SORb, SONR3Rb, S02Rb, and/or -0[C(R3)2]n-COOR3, and
- R1 is H or Rb, and
- R2 is H, F, CI, Br, I, or Rb, and
- R3 is is H or Rb, and
- n is 0, 1 , 2, 3, 4, 5, 6, 7 or 8;
2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-quinazolin-3- yl]acetic acid:
Figure imgf000040_0001
- N-(5-chloro-2,4-dimethoxy-phenyl)-2-[2,4-dioxo-3-[2-oxo-2-[2-(3- p ridyl)ethylamino]ethyl]quinazolin-1-yl]:
Figure imgf000040_0002
2-[4-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-quinazolin-3- yl]phenyl]-N-phenethyl-acetamide:
H
Figure imgf000040_0003
4-[6-chloro-1-[2-(3-chloro-4-ethoxy-phenyl)-2-oxo-ethyl]-2,4-dioxo-quinazolin-3- yl]-N-cyclopentyl-butanamide:
Figure imgf000041_0001
N-(5-chloro-2,4-dimethoxy-phenyl)-2-[2,4-dioxo-3-[2- (phenethylamino)ethyl]quinazolin-1-yl]acetamide:
Figure imgf000041_0002
- tert-butyl 2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo- quinazolin-3-yl]acetate:
Figure imgf000042_0001
- tert-butyl N-[2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo- quinazolin-3-yl]ethyl]carbamate:
Figure imgf000042_0002
- 2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2,4-dioxo-pyrido[3,2- d]pyrimidin-3-yl]acetic acid:
Figure imgf000043_0001
- 2-[1-[2-(5-chloro-2,4-dimethoxy-anilino)-2-oxo-ethyl]-2-oxo-4H-quinazolin-3- yl acetic acid:
Figure imgf000043_0002
N-(5-chloro-2,4-dimethoxy-phenyl)-2-[3-(3-methoxybenzoyl)-7-methyl-4- 4a,8a-dihydro-1 ,8-naphthyridin-1 -yl]acetamide:
Figure imgf000043_0003
2-[1-[2-[(2,6-dichloro-4-pyridyl)amino]-2-oxo-ethyl]-5-methyl-2,4-dioxo- quinazolin-3- l]acetic acid:
Figure imgf000044_0001
4-methyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1 -one:
Figure imgf000044_0002
4-methyl-8-(2,4,6-trimethylanilino)-2H-isoquinolin-1 -one:
Figure imgf000044_0003
8-(2,6-dimethylanilino)-2H-isoquinolin-1 -one:
Figure imgf000045_0001
8-(4-fluoro-2,6-dimethyl-anilino)-4-methyl-2H-phthalazin-1 -one:
Figure imgf000045_0002
- 4-ethyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1 -one:
Figure imgf000045_0003
4-isopropyl-8-(2,4,6-trimethylanilino)-2H-phthalazin-1-one:
Figure imgf000046_0001
- 4-(2-hydroxyethyl)-8-(2,4,6-trimethylanilino)-2H-phthalazin-1-one:
Figure imgf000046_0002
8-(4-chloro-2,6-dimethyl-anilino)-4-methyl-2H-phthalazin-1-one:
Figure imgf000047_0001
Figure imgf000048_0001
5-chloro-2-(2,4,6-trimethylanilino)benzamide:
Figure imgf000048_0002
6. A pharmaceutical composition for treating neuronal damage, comprising an
inhibitor, a modulator, a soluble polypeptide or a compound according to any of the above claims 1 to 5.
7. A dosage form for treating neuronal damage, comprising an inhibitor, a modulator, a soluble polypeptide or a compound according to any of the above claims 1 to 5, and optionally a pharmaceutically acceptable carrier and/or excipient.
8. A method for treating neuronal damage, comprising the administration of an
inhibitor, a modulator, a soluble polypeptide or a compound according to any of the above claims 1 to 5 to a patient in need thereof.
9. A method for the manufacture of a medicament for treating neuronal damage, comprising the use of an inhibitor, a modulator, a soluble polypeptide or a compound according to any of the above claims 1 to 5.
PCT/EP2012/060533 2011-06-03 2012-06-04 Blockers of the nogo-a s1pr pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair WO2012164103A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11168718 2011-06-03
EP11168718.2 2011-06-03

Publications (3)

Publication Number Publication Date
WO2012164103A2 true WO2012164103A2 (en) 2012-12-06
WO2012164103A3 WO2012164103A3 (en) 2013-01-24
WO2012164103A9 WO2012164103A9 (en) 2013-03-21

Family

ID=46354178

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/060533 WO2012164103A2 (en) 2011-06-03 2012-06-04 Blockers of the nogo-a s1pr pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair

Country Status (1)

Country Link
WO (1) WO2012164103A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016191872A1 (en) * 2015-06-01 2016-12-08 Dalhousie University S1pr2 antagonists and uses therefor
US10058543B2 (en) 2014-06-02 2018-08-28 Dalhousie University Treatment of familial exudative vitreoretinopathy through S1PR2 inhibition
CN110461322A (en) * 2017-02-03 2019-11-15 加利福尼亚大学董事会 For inhibiting the composition and method of plasma membrane protein 4
US10487082B2 (en) 2015-06-01 2019-11-26 Dalhousie University S1PR2 antagonists and uses therefor
US10858358B2 (en) 2015-06-01 2020-12-08 Dalhousie University S1PR2 antagonists and uses therefor
WO2021012018A1 (en) * 2019-07-24 2021-01-28 Cincera Therapeutics Pty Ltd Inhibitor compounds
WO2023021278A1 (en) * 2021-08-16 2023-02-23 Redx Pharma Plc Phenyl- and pyridopyrazole derivatives as inhibitors of ddr1

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008107436A1 (en) 2007-03-06 2008-09-12 Novartis Ag Bicyclic organic compounds suitable for the treatment of inflammatory or allergic conditions
WO2008129029A1 (en) 2007-04-23 2008-10-30 Novartis Ag Phthalazine and isoquinoline derivatives with slp receptor modulating activities
WO2008142073A1 (en) 2007-05-22 2008-11-27 Novartis Ag Benzamides useful as s1p receptor modulators
WO2008154470A1 (en) 2007-06-08 2008-12-18 University Of Connecticut Nhibitor of the receptor activity of the s1p2 receptor for inhibiting pathological angiogenesis in the eye

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008107436A1 (en) 2007-03-06 2008-09-12 Novartis Ag Bicyclic organic compounds suitable for the treatment of inflammatory or allergic conditions
WO2008129029A1 (en) 2007-04-23 2008-10-30 Novartis Ag Phthalazine and isoquinoline derivatives with slp receptor modulating activities
WO2008142073A1 (en) 2007-05-22 2008-11-27 Novartis Ag Benzamides useful as s1p receptor modulators
WO2008154470A1 (en) 2007-06-08 2008-12-18 University Of Connecticut Nhibitor of the receptor activity of the s1p2 receptor for inhibiting pathological angiogenesis in the eye

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PERRIMON ET AL., COLD SPRING HARBOUR PERSPECTIVES IN BIOLOGY, vol. 2, 2010, pages A003640
WITTE ET AL., COMB. CHEM. HIGH THROUGHPUT SCREEN, vol. 12, no. 8, 2009, pages 791 - 800

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10058543B2 (en) 2014-06-02 2018-08-28 Dalhousie University Treatment of familial exudative vitreoretinopathy through S1PR2 inhibition
WO2016191872A1 (en) * 2015-06-01 2016-12-08 Dalhousie University S1pr2 antagonists and uses therefor
CN107849038A (en) * 2015-06-01 2018-03-27 达尔豪西大学 S1PR2 antagonists and application thereof
US10487082B2 (en) 2015-06-01 2019-11-26 Dalhousie University S1PR2 antagonists and uses therefor
US10858358B2 (en) 2015-06-01 2020-12-08 Dalhousie University S1PR2 antagonists and uses therefor
CN110461322A (en) * 2017-02-03 2019-11-15 加利福尼亚大学董事会 For inhibiting the composition and method of plasma membrane protein 4
EP3576728A4 (en) * 2017-02-03 2020-08-12 The Regents of the University of California Compositions and methods for inhibiting reticulon 4
WO2021012018A1 (en) * 2019-07-24 2021-01-28 Cincera Therapeutics Pty Ltd Inhibitor compounds
CN114450281A (en) * 2019-07-24 2022-05-06 辛塞拉治疗私人有限公司 Inhibitor compounds
WO2023021278A1 (en) * 2021-08-16 2023-02-23 Redx Pharma Plc Phenyl- and pyridopyrazole derivatives as inhibitors of ddr1

Also Published As

Publication number Publication date
WO2012164103A9 (en) 2013-03-21
WO2012164103A3 (en) 2013-01-24

Similar Documents

Publication Publication Date Title
WO2012164103A2 (en) Blockers of the nogo-a s1pr pathway for the treatment of diseases characterized by neuronal damage and lack of subsequent repair
JP6125505B2 (en) Treatment of voltage-gated sodium channel alpha subunit related diseases with small molecules
Brandt et al. Bumepamine, a brain-permeant benzylamine derivative of bumetanide, does not inhibit NKCC1 but is more potent to enhance phenobarbital's anti-seizure efficacy
WO2017075222A1 (en) Treatment of neurological and neurodevelopmental diseases and disorders associated with aberrant ion channel expression and activity
Sun et al. Inhibitors of voltage-gated sodium channel Nav1. 7: patent applications since 2010
KR20180093930A (en) How to treat hyperalgesia
WO2006017861A2 (en) Treatment for methamphetamine addiction and reduction of methamphetamine use using serotonin antagonists
ES2911388T3 (en) Adapter-associated kinase 1 inhibitors, compositions comprising them, and methods for their use
US20200306244A1 (en) Wnt inhibitors for use in the treatment of fibrosis
US20230122940A1 (en) Combination therapy for treating cancer
WO2016118632A1 (en) Prevention and treatment of itch with an mrgpr antagonist
US20200087662A1 (en) Inhibition of poly(a) binding protein and the treatment of pain
WO2015039187A1 (en) Stem cell modulation ii
US20200289482A1 (en) Cystic fibrosis transmembrane conductance regulator modulators for treating autosomal dominant polycystic kidney disease
US10660957B2 (en) Compositions and methods for treating an Aβ-modulated disease or disorder or improving cognition in a subject
EP3965769B1 (en) Use of 2-phenyl-6-(1h-imidazol-1-yl)quinazoline for treating neurodegenerative diseases, preferably alzheimer&#39;s disease
US9993522B2 (en) Treatment of pain by inhibition of USP5 de-ubiquitinase
KR20230005175A (en) EIF4A inhibitor combinations
KR20100116177A (en) Use of hedgehog agonists in the treatment of musculoskeletal-related disorders
EP3215146B1 (en) Blt2 agonists for the treatment of pain
WO2018136933A1 (en) Inhibition of stromal interaction molecule 1 (stim1) as a co-treatment for adult onset polycystic kidney disease (adpkd)
US20220265607A1 (en) Methods and compositions for unsilencing imprinted genes
US20220071955A1 (en) Methods of Treatment, Prevention and Diagnosis
ES2412955B1 (en) USE OF TRPA1 RECEIVER ANTAGONISTS FOR THE TREATMENT OF DISEASES ASSOCIATED WITH BACTERIAL INFECTIONS.
ES2408132B1 (en) PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF THE EPIPHORE.

Legal Events

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

Ref document number: 12729399

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12729399

Country of ref document: EP

Kind code of ref document: A2