US20050261365A1 - Transthyretin stabilization - Google Patents

Transthyretin stabilization Download PDF

Info

Publication number
US20050261365A1
US20050261365A1 US11/134,527 US13452705A US2005261365A1 US 20050261365 A1 US20050261365 A1 US 20050261365A1 US 13452705 A US13452705 A US 13452705A US 2005261365 A1 US2005261365 A1 US 2005261365A1
Authority
US
United States
Prior art keywords
ttr
dibenzofuran
binding
inhibitors
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/134,527
Inventor
Jeffery Kelly
H. Petrassi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Scripps Research Institute
Original Assignee
Scripps Research Institute
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 Scripps Research Institute filed Critical Scripps Research Institute
Priority to US11/134,527 priority Critical patent/US20050261365A1/en
Assigned to SCRIPPS RESEARCH INSTITUTE, THE reassignment SCRIPPS RESEARCH INSTITUTE, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PETRASSI, H. MICHAEL, KELLY, JEFFERY W.
Publication of US20050261365A1 publication Critical patent/US20050261365A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE Assignors: SCRIPPS RESEARCH INSTITUTE
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/91Dibenzofurans; Hydrogenated dibenzofurans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/56Benzoxazoles; Hydrogenated benzoxazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D263/57Aryl or substituted aryl radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/40Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino groups bound to carbon atoms of at least one six-membered aromatic ring and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/42Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino groups bound to carbon atoms of at least one six-membered aromatic ring and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton with carboxyl groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by saturated carbon chains
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/52Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C229/54Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C229/56Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino and carboxyl groups bound in ortho-position
    • C07C229/58Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino and carboxyl groups bound in ortho-position having the nitrogen atom of at least one of the amino groups further bound to a carbon atom of a six-membered aromatic ring, e.g. N-phenyl-anthranilic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/52Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C229/54Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C229/60Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino and carboxyl groups bound in meta- or para- positions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C251/00Compounds containing nitrogen atoms doubly-bound to a carbon skeleton
    • C07C251/32Oximes
    • C07C251/34Oximes with oxygen atoms of oxyimino groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • C07C251/48Oximes with oxygen atoms of oxyimino groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals with the carbon atom of at least one of the oxyimino groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/24Halogenated derivatives
    • C07C39/367Halogenated derivatives polycyclic non-condensed, containing only six-membered aromatic rings as cyclic parts, e.g. halogenated poly-hydroxyphenylalkanes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/21Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing ether groups, groups, groups, or groups
    • C07C65/24Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing ether groups, groups, groups, or groups polycyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/56Benzoxazoles; Hydrogenated benzoxazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to inhibitors of transthyretin amyloid fibril formation. More particularly, the invention relates to derivatized dibenzofurans as inhibitors of transthyretin amyloid fibril formation.
  • TTR transthyretin
  • Dibenzofuran-4,6-dicarboxylic acid core structures having an aromatic substituent appended onto the dibenzofuran ring at the C1 position using three different types of linkages are disclosed herein and shown to afford exceptional amyloidogenesis inhibitors that display increased affinity and greatly increased binding selectivity to TTR over all the other plasma proteins, relative to lead compound 1 (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). It is further disclosed herein that these compounds function by imposing kinetic stabilization on the TTR tetramer.
  • Transthyretin (TTR) amyloidogenesis requires rate limiting tetramer dissociation and partial monomer denaturation to produce a misassembly competent species. This process has been followed by turbidity to identify transthyretin amyloidogenesis inhibitors including dibenzofuran-4,6-dicarboxylic acid (1).
  • An X-ray cocrystal structure of TTR•1 2 reveals that it only utilizes the outer portion of the two thyroxine binding pockets to bind to and inhibit TTR amyloidogenesis.
  • structure-based design was employed to append aryl substituents using three different chemical linkages at C1 of the dibenzofuran ring to complement the unused inner portion of the thyroxine binding pockets.
  • One aspect of the invention is directed to a compound represented by Formula I:
  • X is absent or is a diradical selected from the group consisting of —O—, —S—, and —NH—; and R 2 , R 3 , R 4 , and R 5 are radicals independently selected from the group consisting of —H, —OH, —F, —Cl, —Br, —CF 3 , and —CO 2 H.
  • the compound is represented by Formula II:
  • preferred embodiments may include species wherein R 2 is a radical selected from the group consisting of —H, —F, —Cl, and —CF 3 ; additional preferred embodiments may include species wherein R 4 is a radical selected from the group consisting of —H, —Cl, and —CO 2 H; additional preferred embodiments may include species wherein R 5 is a radical selected from the group consisting of —H, —F, and —Cl.
  • Preferred species of the subgenus of Formula II include compounds selected from the group represented by the following structures:
  • the compound is represented by Formula III:
  • preferred embodiments may include species wherein R 3 is a radical selected from the group consisting of —H, —F, —Cl, —Br, and —CF 3 ; additional preferred embodiments may include species wherein R 5 is a radical selected from the group consisting of —H, —F, —Cl, and —Br.
  • Preferred species of the subgenus of Formula III include compounds selected from the group represented by the following structures:
  • the compound is represented by Formula IV:
  • preferred embodiments may include species wherein R 2 is a radical selected from the group consisting of —H, —F, and —Cl; additional preferred embodiments may include species wherein R 3 is a radical selected from the group consisting of —H, —F, —Cl, —CF 3 , and —CO 2 H; additional preferred embodiments may include species wherein R 4 is a radical selected from the group consisting of —H, and —CO 2 H; additional preferred embodiments may include species wherein R 5 is a radical selected from the group consisting of —H, —F, —Cl, and —CF 3 .
  • Preferred species of the subgenus of Formula IV include compounds selected from the group represented by the following structures:
  • a further aspect of the invention is directed to a process comprising the step of contacting transthyretin with a concentration of a compound selected from Formulas I-IV, described above, sufficient for inhibiting amyloid fibril formation.
  • FIG. 1A illustrates an X-ray crystallographic structure of TTR•1 2 (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321).
  • FIG. 1B illustrates a line drawing representation of the design of the 1-substituted-dibenzofuran-4,6-dicarboxylic acids placed in the thyroxine binding pocket
  • X represents either an NH, O or direct C aryl -C aryl linkage
  • R represents the substituents of the aryl ring designed to complement TTR's inner binding cavity.
  • FIG. 2 illustrates a table highlighting the concentration dependent acid-substituted dibenzofuran activity against WT-TTR (3.6 ⁇ M) amyloid fibril formation (f.f.) at pH 4.4 (72 h).
  • FIG. 3 illustrates a chart showing a summary of dibenzofuran-based amyloid inhibition activity (3.6 ⁇ M) against WT-TTR (3.6 ⁇ M) fibril formation (pH 4.4, 72 h) and binding stoichiometry to TTR in human blood plasma.
  • FIG. 4 illustrates a scheme for the synthesis of 1-hydroxy-dibenzofuran-4,6-dicarboxylate dimethyl ester and the corresponding triflate.
  • FIG. 5 illustrates a scheme for the synthesis of 1-phenyl-, phenoxy-, and phenylamine-dibenzofuran-4,6-dicarboxylate dimethyl esters and the corresponding dicarboxylates.
  • FIG. 6 illustrates a chart showing dibenzofuran-based inhibitor activity (7.2 ⁇ M) against WT-TTR (3.6 ⁇ M) amyloid fibril formation (f.f.) at pH 4.4 (72 h).
  • FIG. 7 illustrates a table illustrating dibenzofuran plasma TTR binding stoichiometry plotted vs. fibril formation inhibition efficacy.
  • FIG. 8 illustrates a plot of the absorbance at 280 nm versus distance from the center in the sedimentation velocity study on TTR (3.6 ⁇ M) after being preincubated with 27 (7.2 ⁇ M) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • FIG. 9 illustrates a plot of the absorbance at 280 nm versus distance from the center in the equilibrium ultracentrifugation studies on TTR (3.6 ⁇ M) after being preincubated with 27 (7.2 ⁇ M) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • FIG. 10 illustrates a plot of the timecourse analysis of WT-TTR (3.6 ⁇ M) fibril formation mediated by partial acid denaturation in the absence ( ⁇ ) and presence of 7.2 ⁇ M ( ⁇ ) and 3.6 ⁇ M ( ⁇ ) inhibitors 25, 47, and 64, as measured by turbidity at 500 nm (see shading scheme within Figure to differentiate inhibitors).
  • FIG. 11 illustrates a plot of the timecourse analysis of WT-TTR (3.6 ⁇ M) tetramer dissociation (6.0 M urea) in the absence ( ⁇ ) and presence of 7.2 ⁇ M ( ⁇ ) and 3.6 ⁇ M ( ⁇ ) concentrations of inhibitors 25, 47, and 64 (see color scheme within Figure to differentiate inhibitors).
  • FIG. 7 shaded in gray contains dibenzofuran-based compounds that meet the criteria for high in vitro activity ( ⁇ 40% fibril formation) and high binding selectivity (>1 equiv bound to TTR in plasma).
  • the most important point is that the activity and binding selectivity of almost all of the dibenzofuran-based inhibitors, especially those in the gray box ( FIG. 7 ), are sufficient to kinetically stabilize TTR in plasma should they display desirable oral bioavailability, pharmacokinetic, and toxicity profiles.
  • inhibitor efficacy in vitro (3.6 ⁇ M) and inhibitor binding selectivity (10.8 ⁇ M) to TTR in plasma do not correlate ( FIG. 7 ).
  • Compounds that exhibit superior binding selectivity to TTR over all of the other plasma proteins should be excellent inhibitors of fibril formation.
  • excellent inhibitors need not display high TTR plasma binding selectivity.
  • Excellent inhibitors that display high TTR plasma binding selectivity are the most useful compounds in humans because these can selectively stabilize the TTR native state over the dissociative transition state and impart kinetic stabilization on TTR in a protein-rich biological fluid.
  • Their binding constants to TTR are important because the extent of kinetic stabilization is proportional to the binding constants.
  • dibenzofuran inhibitors display unprecedented binding selectivity and inhibitor potency as a group relative to inhibitors characterized heretofore (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew. Chem. 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P.
  • TTR is the tertiary carrier of T 4 , more than 99% of its binding sites are unoccupied; therefore, inhibitor binding to TTR should not perturb T 4 homeostasis.
  • the C1-substituted dibenzofuran-based TTR amyloidogenesis inhibitors are promising because of their amyloid inhibition potency in vitro, their superb binding selectivity to TTR in plasma, their slow TTR dissociation rates (which must be the case to see high plasma selectivity by the method utilized herein), their ability to impose kinetic stabilization upon the TTR tetramer, their chemical stability in plasma, and their chemical stability at low pH (making them excellent candidates for oral administration).
  • These inhibitors are useful for the treatment of TTR amyloid diseases, including SSA, FAP, and FAC, because kinetic stabilization of TTR is known to ameliorate FAP (Hammarstrom, P.; et al.
  • FIG. 1A depicts the two symmetry equivalent binding modes of 1 (green and yellow) within one of the TTR thyroxine binding sites, the surface of which is outlined in gray (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321).
  • the carboxylates at the 4 and 6 positions make electrostatic interactions with the ⁇ -NH 3 + groups of Lys 15 and 15′ at the entrance to the thyroxine binding site. Removal of one of the carboxylates renders dibenzofuran much less active, as does varying the carboxylate spacing from the aromatic ring in most cases ( FIG. 2 ).
  • the dibenzofuran ring nicely complements the shape and hydrophobicity of the outer portion of the thyroxine binding cavity.
  • Inspection of the TTR•1 2 crystal structure in FIG. 1A reveals that there is a large amount of unoccupied volume in the inner portion of the thyroxine binding site with 1 bound.
  • a substituent such as an aryl ring, could be projected into the inner portion of the binding site by attaching it to the C1 position of the dibenzofuran ring of 1.
  • such a substituent could be linked to a dibenzofuran scaffold through a heteroatom (N or O) or directly via a C aryl -C aryl bond (not shown).
  • Aromatic substituents ( FIG. 3 ) were chosen to interact with either the halogen binding pockets or hydrogen bonding substructures within the inner cavity based on the envisioned orientation of the phenyl ring in the inner binding cavity and previous SAR data from other chemical series thought to position their aryl rings similarly (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew Chem 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci.
  • Inhibition efficacy (compounds 24-38, 44-48, and 60-70) was first evaluated using recombinant TTR in a partially denaturing buffer that promotes amyloidogenesis (pH 4.4, 37° C.). As a follow up, the ability of effective inhibitors to bind to TTR selectively over all the other proteins in human plasma was assessed.
  • TTR amyloid inhibition efficacy was probed using a stagnant fibril formation assay described previously, wherein partial denaturation was triggered by acidification (pH 4.4, 37° C.) (Colon, W.; Kelly, J. W. Biochemistry 1992, 31, 8654-8660). Briefly, a test compound (7.2 or 3.6 ⁇ M) is incubated with TTR (3.6 ⁇ M) for 30 min in pH 7 buffer. Amyloidogenesis is then initiated by lowering to pH 4.4, where maximal fibril formation is observed with WT-TTR after 72 h (37° C.).
  • FIG. 6 (at 7.2 ⁇ M there is enough test compound added to occupy both of the binding sites of TTR (TTR•I 2 ), provided their dissociation constants are both in the low nM range at pH 4.4).
  • Small molecules typically bind to TTR with negative cooperativity, hence K d1 and K d2 are often are separated by one or two orders of magnitude. Therefore, when both the ligand and TTR are at equal concentrations, a population of TTR•I, TTR•I 2 and unliganded TTR is observed, dictated by the dissociation constants.
  • Inhibitor binding selectivity to TTR in human blood plasma was assessed using a previously established antibody capture method (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571).
  • inhibitors (10.8 ⁇ M, ⁇ 2-3 ⁇ the natural concentration of TTR) are incubated in human blood plasma for 24 h at 37° C. Quenched sepharose resin is then added to the plasma to remove any small molecules that would bind to the resin as opposed to TTR. TTR and any TTR-bound small molecule is then immunocaptured using a polyclonal TTR antibody covalently attached to sepharose resin.
  • the antibody-TTR complex is dissociated at high pH and analyzed by RP-HPLC.
  • the relative stoichiometry between TTR and inhibitor is then calculated from their HPLC peak areas using standard curves. Wash-associated losses are typically observed for inhibitors that have high dissociation rates; therefore, this analysis can underestimate their true binding stoichiometry, but gives faithful results for compounds exhibiting low dissociation constants and off-rates. Twenty-one inhibitors exhibit a binding stoichiometry exceeding one (two being the maximal binding stoichiometry), nineteen of which exhibit ⁇ 40% fibril formation at a concentration of 3.6 ⁇ M ( FIG. 7 , shaded box).
  • TTR quaternary structure by analytical ultracentrifugation after a preincubation period of 72 h under amyloidogenic conditions (pH 4.4, 37° C.).
  • TTR 3.6 ⁇ mM
  • FIG. 8 sedimentation velocity
  • FIG. 9 equilibrium analytical ultracentrifugation
  • TTR tetramer dissociation The ability of these inhibitors to impose kinetic stability on tetrameric TTR is best evaluated by assessing the rate of TTR tetramer dissociation (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432). Under acidic conditions tetramer dissociation leads to amyloidogenesis, whereas in the presence of chaotropes (6M urea), tetramer dissociation leads to monomer unfolding.
  • inhibitors 25, 47 and 64 representing the three structural classes of dibenzofuran-based inhibitors
  • TTR amyloidogenesis mediated by partial acidification is dramatically slowed in a dose-dependent fashion relative to control (no inhibitor) by 25, 47 and 64 ( FIG. 4A ).
  • the rate of TTR tetramer dissociation in 6M urea is easily monitored by linking the slow quaternary structural changes to rapid tertiary structural changes that are easily monitored by spectroscopic methods (Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432).
  • the pH was then adjusted to 4.4 with addition of 500 ⁇ L of acidic buffer (100 mM acetate, 100 mM KCl, 1 mM EDTA, pH 4.2), and the final 1 mL solutions were vortexed again and incubated in the dark at 37° C. without agitation.
  • acidic buffer 100 mM acetate, 100 mM KCl, 1 mM EDTA, pH 4.2
  • the solutions were vortexed and the turbidity at 500 nm was measured.
  • Control samples containing 5 ⁇ L of pure DMSO were prepared and analyzed as above for comparison. Small molecule and TTR control samples were prepared in groups of 10 to prevent disturbance of the cuvettes during incubation. Samples were discarded after their turbidities were measured.
  • urea buffer (6.67 M urea, 50 mM sodium phosphate, 100 mM KCl, 1 mM EDTA, pH 7.2), and the final 1 mL solutions were vortexed again and incubated in the dark at 25° C. without agitation.
  • the circular dichroism spectra were measured between 218 and 215 nm, with scanning every 0.5 nm and averaging for 5 s. After measurements were taken, samples were returned to their respective eppendorf tubes and incubation was continued.
  • Control samples containing 7.2 ⁇ L of 10% DMSO in EtOH were prepared and analyzed as above for comparison.
  • the CD amplitude values were averaged between 215 and 218 nm to determine the extent of ⁇ -sheet loss throughout the experiment.
  • TTR tetramer dissociation is linked to the rapid ( ⁇ 500,000 ⁇ faster) monomer denaturation as measured through this ⁇ -sheet loss (Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432).
  • the flask was cooled again to ⁇ 78° C. as described above and a 15 psi stream of CO 2 (g) was bubbled through the reaction suspension (the CO 2 was dried by passing it through a drying tube containing activated silica). Following initial addition of CO 2 (g), the cooling bath was removed and the reaction was stirred for 30 min. The reaction mixture was poured into a 1 L beaker containing ice water (50 mL). The solution was brought to pH 9 by the slow addition of 0.05 M KOH, and then cooled to 0° C. with an ice/H 2 O bath. The solution was acidified to pH 2 with 0.5 M HCl causing a white solid to precipitate.
  • the aqueous suspension (pH 2) was transferred into a 1 L separatory funnel and extracted with EtOAc (5 ⁇ 50 mL). The combined extracts were dried with MgSO 4 and concentrated under reduced pressure to afford the crude diacid as an oil.
  • the 100 mL flask containing the crude diacid was equipped with a stir bar, capped with a septum and evacuated. The flask was then back-filled with argon. Anhydrous MeOH (2 mL) and ACS reagent grade benzene (8 mL) were added via syringe.
  • Trimethylsilyidiazomethane (TMSCHN 2 ; 2.5 mL of a 2 M solution in hexanes, 5 mmol) was added slowly via syringe through the septum. Upon completion of the TMSCHN 2 addition the reaction was stirred for 10 min and the solvent removed under reduced pressure to afford a red oil. The residue was purified by flash chromatography over silica (15% EtOAc in hexanes) to afford 0.36 g (43%) of 6 as a white solid.
  • MALDI-FTMS 479.1874 m/z (M+Na) + , C 25 H 32 O 6 SiNa requires 479.1860.
  • the biaryl ether coupling was directly adapted from the procedures reported by Chan and Evans.
  • a 20 mL scintillation vial equipped with a magnetic stir bar was charged with phenol 7 (150 mg, 0.50 mmol), copper (II) acetate (91 mg, 0.5 mmol), freshly activated 4 ⁇ molecular sieves ( ⁇ 250 mg), and phenylboronic acid (180 mg, 1.5 mmol).
  • Dichloromethane (5 mL) was added followed by pyridine (201 ⁇ L, 2.5 mmol), resulting in an aqua colored suspension.
  • the cap was very loosely applied such that the reaction suspension was partly open to the atmosphere.
  • the reaction was monitored by TLC.
  • Methyl ester 9 (25 mg, 0.067 mmol) was saponified in THF: MeOH: H 2 O (3:1:1, 1 mL) in a 20 mL scintillation vial equipped with a stir bar. LiOH.H 2 O (22 mg, 0.53 mmol) was added to the suspension and the reaction was allowed to stir until completion (typically 4 h) as determined by TLC or analytical reverse phase HPLC monitoring. The reaction mixture was diluted with brine (2 mL) and acidified to pH 2 with 1 M HCl (pH paper) resulting in a biphasic solution. The upper layer (THF) was removed and the aqueous layer was extracted with THF (3 ⁇ 3 mL).
  • FIG. 1A shows an X-ray crystallographic structure of TTR•1 2 (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321).
  • the residues lining the binding site are displayed as stick models (oxygen in red, nitrogen in blue, and carbon in gray), with the protein's Connolly surface depicted in gray.
  • Compound 1 is shown in both of its C 2 symmetry equivalent binding modes (yellow and green).
  • the binding channel has 3 sets of depressions referred to as the halogen binding pockets (HBPs) because they interact with the iodines of thyroxine.
  • HBPs halogen binding pockets
  • Compound 1 occupies only the outer portion of the binding pocket and fills both HBP1 and 1′.
  • the carboxylic acids of 1 are in proximity to the e-NH 3 + of K15 and K15′.
  • FIG. 1B shows a line drawing representation of the design of the 1-substituted-dibenzofuran-4,6-dicarboxylic acids placed in the thyroxine binding pocket where X represents either an NH, O or direct C aryl -C aryl linkage. R represents the substituents of the aryl ring designed to complement TTR's inner binding cavity.
  • FIG. 2 is a table highlighting the concentration dependent acid-substituted dibenzofuran activity against WT-TTR (3.6 ⁇ M) amyloid fibril formation (f.f.) at pH 4.4 (72 h). Values represent the extent of f.f. and thus inhibitor efficacy relative to WT-TTR fibril formation in the absence of inhibitor (assigned to be 100%): complete inhibition is equivalent to 0% f.f.
  • FIG. 3 is a chart showing a summary of dibenzofuran-based amyloid inhibition activity (3.6 mM) against WT-TTR (3.6 mM) fibril formation (pH 4.4, 72 h) and binding stoichiometry to TTR in human blood plasma.
  • % Fibril formation (f.f.) values in the middle column represent the extent of f.f., and thus inhibitor efficacy, relative to WT-TTR f.f. in the absence of inhibitor (assigned to be 100%). Complete inhibition is equivalent to 0% f.f.
  • the right column depicts the observed stoichiometry of inhibitor (dosed at 10.8 mM, ⁇ 2-3 ⁇ the concentration of plasma TTR) bound to TTR in blood plasma as determined using the antibody capture method.
  • FIG. 4 is a scheme for the synthesis of 1-hydroxy-dibenzofuran-4,6-dicarboxylate dimethyl ester and the corresponding triflate: a) K 3 [Fe(CN) 6 ], KOH, H 2 O, benzene; b) AlCl 3 , toluene, 33% for both steps; c) TIPSCI, DMAP, CH 2 Cl 2 , 77%; d) sec-BuLi, Et 2 O, ⁇ 78° C., gaseous CO 2 , TMSCHN 2 , 43%; e) TBAF, THF, 97%; f) Tf 2 O, pyridine, 92%.
  • FIG. 5 is a scheme for the synthesis of 1-phenyl-, phenoxy-, and phenylamine-dibenzofuran-4,6-dicarboxylate dimethyl esters and the corresponding dicarboxylates: a) Pd 2 (DBA) 3 , ( ⁇ )binap, Cs 2 CO 3 , toluene 100° C.; b) LiOH—H 2 O, THF/MeOH/H 2 O (3:1:1); c) Cu II (OAc) 2 , pyridine, 4 ⁇ MS, CH 2 Cl 2 ; d) Pd(PPh 3 ) 4 , LiCl, aq. Na 2 CO 3 , toluene, MeOH, 80° C.
  • FIG. 6 is a chart showing dibenzofuran-based inhibitor activity (7.2 ⁇ M) against WT-TTR (3.6 ⁇ M) amyloid fibril formation (f.f.) at pH 4.4 (72 h). Values represent the extent of f.f. and thus inhibitor efficacy relative to WT-TTR fibril formation in the absence of inhibitor (assigned to be 100%): complete inhibition is equivalent to 0% f.f.
  • FIG. 7 is a table illustrating dibenzofuran plasma TTR binding stoichiometry plotted vs. fibril formation inhibition efficacy.
  • the lightly shaded area corresponds to the definitions of high activity and high selectivity ( ⁇ 40% fibril formation and a binding stoichiometry >1), while the darkly shaded area corresponds to exceptional compounds ( ⁇ 30% fibril formation and a binding stoichiometry >1.25).
  • Data points identify the three different linkers: NH ( ⁇ ), ⁇ ( ), and direct C aryl -C aryl linkage ( ⁇ ).
  • Dibenzofuran-4,6-dicarboxylic acid (1) data point ( ⁇ ) shown for comparison.
  • FIG. 8 is a plot of the absorbance at 280 nm versus distance from the center in the sedimentation velocity study on TTR (3.6 mM) after being preincubated with 27 (7.2 mM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • Velocity analysis overlay of data sets taken 15 min apart at 50,000 rpm. The data (symbols) fit to a single ideal species model (solid line) with MW 57.1 ⁇ 0.2 kDa.
  • FIG. 9 is a plot of the absorbance at 280 nm versus distance from the center in the equilibrium ultracentrifugation studies on TTR (3.6 mM) after being preincubated with 27 (7.2 mM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • Equilibrium analysis equilibrium concentration gradient observed after a 24 h application of centrifugal force to the sample employing at a speed of 17,000 rpm.
  • the data ( ⁇ ) fit to a single ideal species model (solid line) with MW 55.0 ⁇ 0.2 kDa.
  • the residuals, the difference between experimental and fitted data, are shown in the inset.
  • FIG. 10 is a plot of the timecourse analysis of WT-TTR (3.6 ⁇ M) fibril formation mediated by partial acid denaturation in the absence ( ⁇ ) and presence of 7.2 ⁇ M ( ⁇ ) and 3.6 ⁇ M ( ⁇ ) inhibitors 25, 47, and 64, as measured by turbidity at 500 nm (see color scheme within Figure to differentiate inhibitors). It is hard to discern which compound is most efficacious in the black and white plot. At the end of the plot or after 169 hours have passed, compound 47 shows the most fibrils formed followed by compound 64 followed by compound 25.
  • FIG. 11 is a plot of the timecourse analysis of WT-TTR (3.6 ⁇ M) tetramer dissociation (6.0 M urea) in the absence ( ⁇ ) and presence of 7.2 ⁇ M ( ⁇ ) and 3.6 ⁇ M ( ⁇ ) concentrations of inhibitors 25, 47, and 64 (see color scheme within Figure to differentiate inhibitors).
  • Slow tetramer dissociation is not detectable by far-UV CD spectroscopy, but this process is linked to rapid ( ⁇ 500,000 ⁇ faster) monomer denaturation as monitored by loss of ⁇ -sheet content easily followed by circular dichroism spectroscopy. It is hard to discern which compound is most efficacious in the black and white plot.
  • compound 25 is most effective in dissociating tetramers followed by compound 64 followed by compound 47.

Abstract

Dibenzofuran-4,6-dicarboxylic acid core structures having an aromatic substituent appended onto the at the C1 position using three different types of linkages are disclosed herein and shown to afford exceptional amyloidogenesis inhibitors that display increased affinity and greatly increased binding selectivity to TTR over all the other plasma proteins, relative to lead compound 1. It is further disclosed herein that these compounds function by imposing kinetic stabilization on the TTR tetramer.

Description

    TECHNICAL FIELD
  • The present invention relates to inhibitors of transthyretin amyloid fibril formation. More particularly, the invention relates to derivatized dibenzofurans as inhibitors of transthyretin amyloid fibril formation.
  • BACKGROUND
  • Several structurally distinct classes of small molecule transthyretin (TTR) stabilizers have been discovered, of which dibenzofuran-4,6-dicarboxylic acid (1) is particularly interesting, FIG. 1 (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew Chem 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P. W.; et al. Bioorg. Med. Chem. 1998, 6, 1389-1401; Baures, P. W.; et al. Bioorg. Med. Chem. 1999, 7, 1339-1347; Petrassi, H. M.; et al. J. Am. Chem. Soc. 2000, 122, 2178-2192; McCammon, M. G.; et al. Structure 2002, 10, 851-863; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Sacchettini, J. C.; et al. Nature Rev. Drug Disc. 2002, 1, 267-275; Green, N. S.; et al. J. Am. Chem. Soc. 2003, 125, 13404-13414; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374; Miller Sean, R.; et al. Lab. Inv. 2004, 84, 545-552). This inhibitor (1; 7.2 μM) decreases the extent of WT-TTR (3.6 μM) amyloid formation (pH 4.4) by 90% over a time course of 72 h. The X-ray co-crystal structure of TTR•12 reveals that it exerts its impressive activity by binding exclusively to the outer portion of each thyroxine binding site (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321). What is needed is to modify 1 with substructures that project into the inner cavity of the thyroxine binding pocket so as to increase their affinity and selectivity for binding to TTR in human blood.
  • SUMMARY
  • Dibenzofuran-4,6-dicarboxylic acid core structures having an aromatic substituent appended onto the dibenzofuran ring at the C1 position using three different types of linkages are disclosed herein and shown to afford exceptional amyloidogenesis inhibitors that display increased affinity and greatly increased binding selectivity to TTR over all the other plasma proteins, relative to lead compound 1 (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). It is further disclosed herein that these compounds function by imposing kinetic stabilization on the TTR tetramer.
  • Transthyretin (TTR) amyloidogenesis requires rate limiting tetramer dissociation and partial monomer denaturation to produce a misassembly competent species. This process has been followed by turbidity to identify transthyretin amyloidogenesis inhibitors including dibenzofuran-4,6-dicarboxylic acid (1). An X-ray cocrystal structure of TTR•12 reveals that it only utilizes the outer portion of the two thyroxine binding pockets to bind to and inhibit TTR amyloidogenesis. Herein, structure-based design was employed to append aryl substituents using three different chemical linkages at C1 of the dibenzofuran ring to complement the unused inner portion of the thyroxine binding pockets. Twenty-eight amyloidogenesis inhibitors of increased potency and dramatically increased plasma TTR binding selectivity resulted that function by imposing kinetic stabilization on the native tetrameric structure of TTR, creating a barrier that is insurmountable under physiological conditions. Since kinetic stabilization of the TTR native state by interallelic trans-suppression is known to ameliorate disease, there is reason to be optimistic that the dibenzofuran-based inhibitors will do the same. Preventing the onset of amyloidogenesis is the most conservative strategy to intervene clinically, as it remains unclear which of the TTR misassembly intermediates result in toxicity. The exceptional binding selectivity enables these inhibitors to occupy the thyroxine binding site(s) in a complex biological fluid like blood plasma, required for inhibition of amyloidogenesis in humans. It is now established that the dibenzofuran-based amyloidogenesis inhibitors have high selectivity, affinity, and efficacy.
  • One aspect of the invention is directed to a compound represented by Formula I:
    Figure US20050261365A1-20051124-C00001

    In Formula I, X is absent or is a diradical selected from the group consisting of —O—, —S—, and —NH—; and R2, R3, R4, and R5 are radicals independently selected from the group consisting of —H, —OH, —F, —Cl, —Br, —CF3, and —CO2H. In a first subgenus of this first aspect of the invention, the compound is represented by Formula II:
    Figure US20050261365A1-20051124-C00002

    Within the subgenus of Formula II, preferred embodiments may include species wherein R2 is a radical selected from the group consisting of —H, —F, —Cl, and —CF3; additional preferred embodiments may include species wherein R4 is a radical selected from the group consisting of —H, —Cl, and —CO2H; additional preferred embodiments may include species wherein R5 is a radical selected from the group consisting of —H, —F, and —Cl. Preferred species of the subgenus of Formula II include compounds selected from the group represented by the following structures:
    Figure US20050261365A1-20051124-C00003
    Figure US20050261365A1-20051124-C00004
    Figure US20050261365A1-20051124-C00005
  • In a second subgenus of this first aspect of the invention, the compound is represented by Formula III:
    Figure US20050261365A1-20051124-C00006

    Within the subgenus of Formula III, preferred embodiments may include species wherein R3 is a radical selected from the group consisting of —H, —F, —Cl, —Br, and —CF3; additional preferred embodiments may include species wherein R5 is a radical selected from the group consisting of —H, —F, —Cl, and —Br. Preferred species of the subgenus of Formula III include compounds selected from the group represented by the following structures:
    Figure US20050261365A1-20051124-C00007
  • In a third subgenus of this first aspect of the invention, the compound is represented by Formula IV:
    Figure US20050261365A1-20051124-C00008

    Within the subgenus of Formula IV, preferred embodiments may include species wherein R2 is a radical selected from the group consisting of —H, —F, and —Cl; additional preferred embodiments may include species wherein R3 is a radical selected from the group consisting of —H, —F, —Cl, —CF3, and —CO2H; additional preferred embodiments may include species wherein R4 is a radical selected from the group consisting of —H, and —CO2H; additional preferred embodiments may include species wherein R5 is a radical selected from the group consisting of —H, —F, —Cl, and —CF3.
  • Preferred species of the subgenus of Formula IV include compounds selected from the group represented by the following structures:
    Figure US20050261365A1-20051124-C00009
    Figure US20050261365A1-20051124-C00010
  • A further aspect of the invention is directed to a process comprising the step of contacting transthyretin with a concentration of a compound selected from Formulas I-IV, described above, sufficient for inhibiting amyloid fibril formation.
  • BRIEF DESCRPTION OF DRAWINGS
  • FIG. 1A illustrates an X-ray crystallographic structure of TTR•12 (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321).
  • FIG. 1B illustrates a line drawing representation of the design of the 1-substituted-dibenzofuran-4,6-dicarboxylic acids placed in the thyroxine binding pocket where X represents either an NH, O or direct Caryl-Caryl linkage. R represents the substituents of the aryl ring designed to complement TTR's inner binding cavity.
  • FIG. 2 illustrates a table highlighting the concentration dependent acid-substituted dibenzofuran activity against WT-TTR (3.6 μM) amyloid fibril formation (f.f.) at pH 4.4 (72 h).
  • FIG. 3 illustrates a chart showing a summary of dibenzofuran-based amyloid inhibition activity (3.6 μM) against WT-TTR (3.6 μM) fibril formation (pH 4.4, 72 h) and binding stoichiometry to TTR in human blood plasma.
  • FIG. 4 illustrates a scheme for the synthesis of 1-hydroxy-dibenzofuran-4,6-dicarboxylate dimethyl ester and the corresponding triflate.
  • FIG. 5 illustrates a scheme for the synthesis of 1-phenyl-, phenoxy-, and phenylamine-dibenzofuran-4,6-dicarboxylate dimethyl esters and the corresponding dicarboxylates.
  • FIG. 6 illustrates a chart showing dibenzofuran-based inhibitor activity (7.2 μM) against WT-TTR (3.6 μM) amyloid fibril formation (f.f.) at pH 4.4 (72 h).
  • FIG. 7 illustrates a table illustrating dibenzofuran plasma TTR binding stoichiometry plotted vs. fibril formation inhibition efficacy.
  • FIG. 8 illustrates a plot of the absorbance at 280 nm versus distance from the center in the sedimentation velocity study on TTR (3.6 μM) after being preincubated with 27 (7.2 μM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • FIG. 9 illustrates a plot of the absorbance at 280 nm versus distance from the center in the equilibrium ultracentrifugation studies on TTR (3.6 μM) after being preincubated with 27 (7.2 μM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor.
  • FIG. 10 illustrates a plot of the timecourse analysis of WT-TTR (3.6 μM) fibril formation mediated by partial acid denaturation in the absence (▴) and presence of 7.2 μM (⋄) and 3.6 μM (◯) inhibitors 25, 47, and 64, as measured by turbidity at 500 nm (see shading scheme within Figure to differentiate inhibitors).
  • FIG. 11 illustrates a plot of the timecourse analysis of WT-TTR (3.6 μM) tetramer dissociation (6.0 M urea) in the absence (▴) and presence of 7.2 μM (⋄) and 3.6 μM (◯) concentrations of inhibitors 25, 47, and 64 (see color scheme within Figure to differentiate inhibitors).
  • DETAILED DESCRIPTION
  • All but one of the C1-aryl substituted dibenzofurans (7.2 μM) are exceptional inhibitors of WT-TTR (3.6 μM) acid-mediated fibril formation in vitro (pH 4.4, 37° C.), even those bearing unsubstituted aryl rings (FIG. 6). The only dibenzofuran-based inhibitor that was not as effective against TTR amyloidogenesis was 34, bearing potentially four negative charges. Because all the compounds completely inhibited TTR fibril formation (within the ±5% experimental error), no structure-activity relationships (SAR) are deducible from the 7.2 μM inhibitor data. However, FIGS. 3 and 7 reveal a range of inhibitor efficacy at an inhibitor concentration equal to that of TTR (3.6 μM), allowing some SAR conclusions to be drawn, limited by the 31 analogs prepared and the experimental error. Notably, all the inhibitors (except 34) display increased potency relative to the parent compound (1) at 3.6 μM (FIG. 3). Most importantly, of the thirty inhibitors exhibiting increased potency, all but two (31 and 35) exhibit dramatically increased binding selectivity to TTR in human blood plasma. The exceptional binding selectivity exhibited by the C1-substituted inhibitors is ideal for inhibiting TTR amyloidogenesis in complex biological systems.
  • Comparing the four C1-aryl substitution patterns (H, 3-CF3, 3,5-F2, and 3,5-Cl2) found in all three inhibitor series reveals that the inhibitors having their aryl rings directly linked to C1 of the dibenzofuran skeleton, hereafter referred to as the biaryls, display slightly increased inhibitor potency relative to their biarylamine and biarylether counterparts (FIG. 3). This may be due to structural differences that enable the rings to be oriented differently within the inner binding cavity; however, we caution that this preference may not hold in a very large analog series. It could be argued with the same qualifiers that the other two series produce the most selective TTR binders in plasma; however, the SAR here is confounded by the fact that as many as one hundred proteins are competing for these inhibitors. While it is not surprising that the most potent inhibitors have 2-F or 3,5-Cl2 substituents (36, 63, and 67), likely picking up the halogen binding pockets in the thyroxine binding site, it is somewhat surprising that the 3-CO2H substituted aryl inhibitors 33 and 69 are amongst the most potent (although their plasma binding selectivity to TTR is modest). Previous crystallographic results on simple biphenyl and biphenylamine inhibitors demonstrate that it can be preferable to have the carboxyl-bearing aryl ring in the inner binding cavity (enabling H-bonding to S117 and T119) (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374).
  • Appending aryl groups to the C1 position not only increases inhibitor potency, but more importantly dramatically increases plasma binding selectivity to TTR, presumably by increasing binding affinity for TTR over the other plasma proteins. The superior binding selectivity of the C1-aryl substituted dibenzofuran-based inhibitors to TTR in plasma is clearly demonstrated by the fact that ˜⅔ of the compounds prepared display a TTR binding stoichiometry greater than one. This is exceedingly interesting as the screening hit 1, utilizing only the outer cavity of TTR for binding, displays no measurable binding selectivity to TTR in plasma. Previous experience with amyloidogenesis inhibitors of diverse chemical structure reveals that very few members display binding stoichiometries exceeding 1 (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew. Chem. 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P. W.; Peterson, S. A.; Kelly, J. W. Bioorg. Med. Chem. 1998, 6, 1389-1401; Baures, P. W.; et al. Bioorg. Med. Chem. 1999, 7, 1339-1347; Petrassi, H. M.; et al. J. Am. Chem. Soc. 2000, 122, 2178-2192; McCammon, M. G.; et al. Structure 2002, 10, 851-863; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Sacchettini, J. C.; Kelly, J. W. Nature Rev. Drug Disc. 2002, 1, 267-275; Green, N. S.; et al. J. Am. Chem. Soc. 2003, 125, 13404-13414; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374; Miller S. R.; et al. Lab. Inv. 2004, 84, 545-552). The area of FIG. 7 shaded in gray contains dibenzofuran-based compounds that meet the criteria for high in vitro activity (<40% fibril formation) and high binding selectivity (>1 equiv bound to TTR in plasma). The most important point is that the activity and binding selectivity of almost all of the dibenzofuran-based inhibitors, especially those in the gray box (FIG. 7), are sufficient to kinetically stabilize TTR in plasma should they display desirable oral bioavailability, pharmacokinetic, and toxicity profiles.
  • It is not surprising that inhibitor efficacy in vitro (3.6 μM) and inhibitor binding selectivity (10.8 μM) to TTR in plasma do not correlate (FIG. 7). Compounds that exhibit superior binding selectivity to TTR over all of the other plasma proteins should be excellent inhibitors of fibril formation. However, the converse is not necessarily true: excellent inhibitors need not display high TTR plasma binding selectivity. Excellent inhibitors that display high TTR plasma binding selectivity are the most useful compounds in humans because these can selectively stabilize the TTR native state over the dissociative transition state and impart kinetic stabilization on TTR in a protein-rich biological fluid. Their binding constants to TTR are important because the extent of kinetic stabilization is proportional to the binding constants. However, focusing on binding constants and potency in vitro can be misleading because compounds can be excellent TTR amyloidogenesis inhibitors in vitro, but bind to other plasma proteins and therefore be rendered useless in humans (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). Potent in vitro inhibitors not displaying good binding selectivity to TTR likely interact with other plasma proteins, such as albumin (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). Because the dibenzofuran inhibitors display unprecedented binding selectivity and inhibitor potency as a group relative to inhibitors characterized heretofore (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew. Chem. 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P. W.; Peterson, S. A.; Kelly, J. W. Bioorg. Med. Chem. 1998, 6, 1389-1401; Baures, P. W.; et al. Bioorg. Med. Chem. 1999, 7, 1339-1347; Petrassi, H. M.; et al. J. Am. Chem. Soc. 2000, 122, 2178-2192; McCammon, M. G.; et al. Structure 2002, 10, 851-863; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Sacchettini, J. C.; Kelly, J. W. Nature Rev. Drug Disc. 2002, 1, 267-275; Green, N. S.; et al. J. Am. Chem. Soc. 2003, 125, 13404-13414; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374; Miller S. R.; et al. Lab. Inv. 2004, 84, 545-552), these are ideal for further pharmacological evaluation. Because TTR is the tertiary carrier of T4, more than 99% of its binding sites are unoccupied; therefore, inhibitor binding to TTR should not perturb T4 homeostasis.
  • The C1-substituted dibenzofuran-based TTR amyloidogenesis inhibitors are promising because of their amyloid inhibition potency in vitro, their superb binding selectivity to TTR in plasma, their slow TTR dissociation rates (which must be the case to see high plasma selectivity by the method utilized herein), their ability to impose kinetic stabilization upon the TTR tetramer, their chemical stability in plasma, and their chemical stability at low pH (making them excellent candidates for oral administration). These inhibitors are useful for the treatment of TTR amyloid diseases, including SSA, FAP, and FAC, because kinetic stabilization of TTR is known to ameliorate FAP (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Coelho, T.; et al. J. Rheumatol. 1993, 20, 179-179; Coelho, T.; et al. Neuromuscular Disord. 1996, 6, 27-27).
  • Design and Synthesis:
  • FIG. 1A depicts the two symmetry equivalent binding modes of 1 (green and yellow) within one of the TTR thyroxine binding sites, the surface of which is outlined in gray (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321). The carboxylates at the 4 and 6 positions make electrostatic interactions with the ε-NH3 + groups of Lys 15 and 15′ at the entrance to the thyroxine binding site. Removal of one of the carboxylates renders dibenzofuran much less active, as does varying the carboxylate spacing from the aromatic ring in most cases (FIG. 2). In addition, the dibenzofuran ring nicely complements the shape and hydrophobicity of the outer portion of the thyroxine binding cavity. Inspection of the TTR•12 crystal structure in FIG. 1A reveals that there is a large amount of unoccupied volume in the inner portion of the thyroxine binding site with 1 bound. Based on this structure, it is clear that a substituent, such as an aryl ring, could be projected into the inner portion of the binding site by attaching it to the C1 position of the dibenzofuran ring of 1. As shown in FIG. 1B, such a substituent could be linked to a dibenzofuran scaffold through a heteroatom (N or O) or directly via a Caryl-Caryl bond (not shown). Aromatic substituents (FIG. 3) were chosen to interact with either the halogen binding pockets or hydrogen bonding substructures within the inner cavity based on the envisioned orientation of the phenyl ring in the inner binding cavity and previous SAR data from other chemical series thought to position their aryl rings similarly (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew Chem 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P. W.; et al. Bioorg. Med. Chem. 1998, 6, 1389-1401; Baures, P. W.; et al. Bioorg. Med. Chem. 1999, 7, 1339-1347; Petrassi, H. M.; et al. J. Am. Chem. Soc. 2000, 122, 2178-2192; McCammon, M. G.; et al. Structure 2002, 10, 851-863; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Sacchettini, J. C.; et al. Nature Rev. Drug Disc. 2002, 1, 267-275; Green, N. S.; et al. J. Am. Chem. Soc. 2003, 125, 13404-13414; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374; Miller Sean, R.; et al. Lab. Inv. 2004, 84, 545-552).
  • The synthesis of C1-substituted dibenzofuran-based inhibitors commenced with the radical phenolic homo-coupling of commercially available 2,4-ditertbutyl-6-bromophenol (2) to afford the dibenzofuran derivative 3 using potassium hexacyanoferrate (III) as previously reported (FIG. 4) (Tashiro, M. Y., et al. Synthesis 1980, 6, 495-496). This tetra-t-butyl dibenzofuran derivative was subjected to transalkylation in toluene to form 1-hydroxydibenzofuran (4) in 33% overall yield from 2 (alternative strategies for the synthesis of this intermediate have appeared) (Labiad, B.; Villemin, D. Synthesis 1989, 143-144; Lee, Y. R.; et al. Org. Lett. 2000, 2, 1387-1389). After protection of the phenol, silyl ether 5 was selectively ortho-metalated at the 4- and 6-positions with sec-butyllithium (the triisopropylsilyl group on the 1-oxygen precludes it from acting as a metalation director) (Snieckus, V. Chem. Rev. 1990, 90, 879-933). The dianion was quenched with gaseous CO2 and esterified to afford 6, which was then deprotected with TBAF and converted to triflate 8 in high overall yield.
  • Selected anilines were coupled to triflate 8 using a palladium mediated N-arylation reaction developed by Buchwald and Hartwig to afford dibenzofuran-based biarylamine analogues 9-23 (FIG. 5) (Louie, J. D., et al. J. Org. Chem. 1997, 62, 1268-1273; Wolfe, J. P. B., Stephen L. Tetrahedron Lett. 1997, 38, 6359-6362). To append an aryl ether to the C1-position of dibenzofuran, phenol 7 and several phenylboronic acids were cross-coupled using the copper-mediated biaryl ether coupling methodology of Chan and Evans, affording compounds 39-43 (Chan, D. M. T.; et al. Tetrahedron Lett. 1998, 39, 2933-2936; Evans, D. A. et al. Tetrahedron Lett. 1998, 39, 2933). Compound 8 was also subjected to Suzuki coupling conditions in the presence of several phenylboronic acids to afford the dibenzofuran-based biaryl analogues 49-59 (Suzuki, A. Modern Arene Chemistry 2002, 53-106). Saponification of the methyl esters in these precursors afforded the desired dibenzofuran-4,6-dicarboxylic acid amines (24-38), ethers (44-48), and biaryls (60-70) to be evaluated as potential TTR amyloidogenesis inhibitors.
  • Results:
  • Two of the most important characteristics of an effective small molecule amyloidogenesis inhibitor are that they must be able to bind with high affinity and selectively to TTR in blood and stabilize its native tetrameric quaternary structure (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Razavi, H.; et al. Angew Chem 2003, 42, 2758-2761; Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). Inhibition efficacy (compounds 24-38, 44-48, and 60-70) was first evaluated using recombinant TTR in a partially denaturing buffer that promotes amyloidogenesis (pH 4.4, 37° C.). As a follow up, the ability of effective inhibitors to bind to TTR selectively over all the other proteins in human plasma was assessed.
  • Evaluating the Dibenzofuran-Based Compounds as Amyloidogenesis Inhibitors.
  • TTR amyloid inhibition efficacy was probed using a stagnant fibril formation assay described previously, wherein partial denaturation was triggered by acidification (pH 4.4, 37° C.) (Colon, W.; Kelly, J. W. Biochemistry 1992, 31, 8654-8660). Briefly, a test compound (7.2 or 3.6 μM) is incubated with TTR (3.6 μM) for 30 min in pH 7 buffer. Amyloidogenesis is then initiated by lowering to pH 4.4, where maximal fibril formation is observed with WT-TTR after 72 h (37° C.). The turbidity in the presence of a potential inhibitor (Ttest) is compared to that of a solution lacking a test compound (Tcontrol) Exceptional inhibitors exhibit 0% fibril formation, whereas compounds not functioning as an inhibitor would exhibit 100% fibril formation. From experience we know that excellent inhibitors allow <10% fibril formation at a small molecule concentration of 7.2 μM and <40% fibril formation at a concentration equal to that of WT-TTR (3.6 μM) (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321; Razavi, H.; et al. Angew Chem 2003, 42, 2758-2761; Miroy, G. J.; et al. Proc. Natl. Acad. Sci. USA 1996, 93, 15051-15056; Peterson, S. A.; et al. Proc. Natl. Acad. Sci. USA 1998, 95, 12956-12960; Baures, P. W.; et al. Bioorg. Med. Chem. 1998, 6, 1389-1401; Baures, P. W.; et al. Bioorg. Med. Chem. 1999, 7, 1339-1347; Petrassi, H. M.; et al. J. Am. Chem. Soc. 2000, 122, 2178-2192; McCammon, M. G.; et al. Structure 2002, 10, 851-863; Oza, V. B.; et al. J. Med. Chem. 2002, 45, 321-332; Saccheftini, J. C.; et al. Nature Rev. Drug Disc. 2002, 1, 267-275; Green, N. S.; et al. J. Am. Chem. Soc. 2003, 125, 13404-13414; Adamski-Werner, S. L.; et al. J. Med. Chem. 2004, 47, 355-374; Miller Sean, R.; et al. Lab. Inv. 2004, 84, 545-552). Of the 31 compounds evaluated, all but one (34) completely inhibit fibril formation at a concentration twice that of TTR (7.2 μM inhibitor), FIG. 6 (at 7.2 μM there is enough test compound added to occupy both of the binding sites of TTR (TTR•I2), provided their dissociation constants are both in the low nM range at pH 4.4). Small molecules typically bind to TTR with negative cooperativity, hence Kd1 and Kd2 are often are separated by one or two orders of magnitude. Therefore, when both the ligand and TTR are at equal concentrations, a population of TTR•I, TTR•I2 and unliganded TTR is observed, dictated by the dissociation constants. It is now established by other studies that inhibitor occupancy of only one of the two TTR binding sites is sufficient to stabilize the entire tetramer against amyloidogenesis (Wiseman, R. L.; et al. J. Am. Chem. Soc. 2005, in press). This is further supported by the observation herein that twenty-six of these dibenzofurans are excellent TTR amyloidogenesis inhibitors (<40% fibril formation) at a concentration equal to that of TTR (3.6 μM each, FIG. 3). Representative small molecules from all three series (25, 26, 27, 30, 45, 47, 62; 3.6 μM) were subjected to the acid-mediated amyloidogenic conditions (pH 4.4, 37° C., 72 h) in the absence of TTR, revealing no measurable precipitation.
  • Evaluating the Plasma TTR Binding Selectivity of the Dibenzofuran-Based Inhibitors.
  • Inhibitor binding selectivity to TTR in human blood plasma was assessed using a previously established antibody capture method (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571). In this evaluation, inhibitors (10.8 μM, ˜2-3× the natural concentration of TTR) are incubated in human blood plasma for 24 h at 37° C. Quenched sepharose resin is then added to the plasma to remove any small molecules that would bind to the resin as opposed to TTR. TTR and any TTR-bound small molecule is then immunocaptured using a polyclonal TTR antibody covalently attached to sepharose resin. After washing the resin (5×10 min washes), the antibody-TTR complex is dissociated at high pH and analyzed by RP-HPLC. The relative stoichiometry between TTR and inhibitor is then calculated from their HPLC peak areas using standard curves. Wash-associated losses are typically observed for inhibitors that have high dissociation rates; therefore, this analysis can underestimate their true binding stoichiometry, but gives faithful results for compounds exhibiting low dissociation constants and off-rates. Twenty-one inhibitors exhibit a binding stoichiometry exceeding one (two being the maximal binding stoichiometry), nineteen of which exhibit <40% fibril formation at a concentration of 3.6 μM (FIG. 7, shaded box). The high plasma TTR binding selectivities observed are remarkable considering the parent dibenzofuran-4,6-dicarboxylic acid (1) displays no binding selectivity to TTR, demonstrating the importance of the C1 aryl substituent in terms of endowing binding selectivity to TTR over all the other plasma proteins.
  • Stabilization of the Tetrameric Quaternary Structure Under Amyloidogenic Conditions.
  • To ensure that these C1-arylated dibenzofurans inhibit TTR fibril formation by native state stabilization (i.e. tetramer stabilization), we studied the TTR quaternary structure by analytical ultracentrifugation after a preincubation period of 72 h under amyloidogenic conditions (pH 4.4, 37° C.). In the presence of 27 (7.2 μM), TTR (3.6 μmM) was found to have hydrodynamic molecular weights of 57.1±0.3 and 55.1±0.4 kDa by sedimentation velocity (FIG. 8) and equilibrium analytical ultracentrifugation (FIG. 9), respectively, comparable to the expected molecular weight of tetrameric TTR (55.0 kDa). In the absence of 27, TTR aggregated into very high molecular weight oligomers that sedimented rapidly in the ultracentrifugation experiment (data not shown).
  • Do the Dibenzofuran-Based Inhibitors Impose Kinetic Stabilization on TTR?
  • The ability of these inhibitors to impose kinetic stability on tetrameric TTR is best evaluated by assessing the rate of TTR tetramer dissociation (Hammarstrom, P.; et al. Science 2003, 299, 713-716; Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432). Under acidic conditions tetramer dissociation leads to amyloidogenesis, whereas in the presence of chaotropes (6M urea), tetramer dissociation leads to monomer unfolding. The influence of inhibitors 25, 47 and 64, representing the three structural classes of dibenzofuran-based inhibitors, on the rates of tetramer dissociation under acid- and urea-mediated denaturing conditions was probed. TTR amyloidogenesis mediated by partial acidification is dramatically slowed in a dose-dependent fashion relative to control (no inhibitor) by 25, 47 and 64 (FIG. 4A). The rate of TTR tetramer dissociation in 6M urea is easily monitored by linking the slow quaternary structural changes to rapid tertiary structural changes that are easily monitored by spectroscopic methods (Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432). The rate of TTR tetramer (3.6 μM) dissociation monitored by far-UV CD is markedly slowed in a dose-dependent fashion in 6M urea by 25, 47 and 64. These results are consistent with differential stabilization of the ground state vs. the dissociative transition state by the binding of 25, 47, and 64.
  • Experimental:
  • The procedures used for bacterial expression of TTR (Lai, Z.; et al. Biochemistry 1996, 35, 6470-6482), the stagnant fibril formation assay (Colon, W.; Kelly, J. W. Biochemistry 1992, 31, 8654-8660; Lai, Z.; et al. Biochemistry 1996, 35, 6470-6482), the blood plasma binding selectivity assay (Purkey, H. E.; et al. Proc. Natl. Acad. Sci. USA 2001, 98, 5566-5571), and analytical ultracentrifugation (Lashuel, H. A.; et al. Biochemistry 1998, 37, 17851-17864) have all been described in detail previously.
  • Time course analysis of WT-TTR fibril formation inhibition by compounds 25, 47, and 64. Compounds 25, 47, and 64 were dissolved in DMSO to provide 7.2 mM primary stock solutions (10× stocks), from which 5- and 10-fold DMSO dilutions yielded 1.44 mM (2×) and 0.72 mM (1×) secondary stock solutions, respectively. 495 μL of 0.4 mg/mL (7.2 μM) WT-TTR solution (10 mM sodium phosphate, 100 mM KCl, and 1 mM EDTA, pH 7.2), and 5 μL of either the 1.44 or 0.72 mM inhibitor secondary stock solutions were added to disposable UV cuvettes, vortexed briefly, then incubated for 30 min at 25° C. The pH was then adjusted to 4.4 with addition of 500 μL of acidic buffer (100 mM acetate, 100 mM KCl, 1 mM EDTA, pH 4.2), and the final 1 mL solutions were vortexed again and incubated in the dark at 37° C. without agitation. At 0, 4, 8, 12, 25, 49, 74, 100, 122, 145, and 169 h time points after acidification, the solutions were vortexed and the turbidity at 500 nm was measured. Control samples containing 5 μL of pure DMSO were prepared and analyzed as above for comparison. Small molecule and TTR control samples were prepared in groups of 10 to prevent disturbance of the cuvettes during incubation. Samples were discarded after their turbidities were measured.
  • Time course analysis of WT-TTR tetramer dissociation inhibition by compounds 25, 47, and 64 evaluated by linked-monomer unfolding in urea. Compounds 25, 47, and 64 were dissolved in DMSO to provide 10 mM primary stock solutions, from which 10-fold EtOH dilutions yielded 1 mM secondary stock solutions. 200 μL of 1.0 mg/mL (18 μM) WT-TTR solution (50 mM sodium phosphate, 100 mM KCl, and 1 mM EDTA, pH 7.2), and either 7.2 or 3.6 μL (2× and 1×, respectively) of 1 mM inhibitor secondary stock solutions were added to 2 mL eppendorf tubes, vortexed briefly, and incubated for 15 min at 25° C. 100 μL of the TTR•inhibitor solutions were added to 900 μL of urea buffer (6.67 M urea, 50 mM sodium phosphate, 100 mM KCl, 1 mM EDTA, pH 7.2), and the final 1 mL solutions were vortexed again and incubated in the dark at 25° C. without agitation. At 0, 4, 11, 24, 49, 73, 97, 122, 146, and 170 h time points after mixing with urea, the circular dichroism spectra were measured between 218 and 215 nm, with scanning every 0.5 nm and averaging for 5 s. After measurements were taken, samples were returned to their respective eppendorf tubes and incubation was continued. Control samples containing 7.2 μL of 10% DMSO in EtOH were prepared and analyzed as above for comparison. The CD amplitude values were averaged between 215 and 218 nm to determine the extent of β-sheet loss throughout the experiment. TTR tetramer dissociation is linked to the rapid (˜500,000× faster) monomer denaturation as measured through this β-sheet loss (Hammarstrom, P.; et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16427-16432).
  • Inhibitor Synthesis: Reagents for chemical synthesis were purchased from commercial suppliers and used without further purification unless otherwise stated. Thin-layer chromatography on silica gel 60 F254 coated aluminum plates (EM Sciences) or analytical reverse phase high performance liquid chromatography (HPLC) were used to monitor reaction progress. HPLC was performed using a Waters 600E multisolvent delivery system employing a Waters 486 tunable absorbance detector and a Waters 717 plus auto sampler. A C18 Western Analytical column was used (model 033-715, 150 Å pore size, 3 μm particles) for all reverse phase HPLC analyses. An acetonitrile/water/trifluoroacetic acid solvent system was used; solvent A in the proportions of 4.8%, 95%, and 0.2%, respectively, while solvent B was of 95%, 4.8%, and 0.2%, respectively. Following 2 min of isocratic flow at 100% A, a linear gradient of 0 to 100% B over 8 min was run at 1.5 mL/min. All flash chromatography was accomplished using 230-400 mesh silica gel 60 (EM Sciences). 1H- and 13C-NMR spectra were recorded at 300, 400, 500 or 600 MHz on Bruker spectrometers. Chemical shifts are reported in parts per million downfield from the internal standard (Me4Si, 0.0 ppm).
  • (Dibenzofuran-1-yloxy)-triisopropyl-silane (5). To a dry 250 mL round bottom flask was added phenol 4 (Tashiro, M. Y., et al. Synthesis 1980, 6, 495-496) (492 mg, 2.67 mmol) and a stir bar and the flask was capped with a septum. CH2Cl2 (5 mL) was added followed by DMAP (391 mg, 3.2 mmol) and triisopropylsilyl chloride (800 μL, 3.73 mmol). The resulting colorless solution became a white suspension overnight. The reaction was transferred to a 250 mL separatory funnel and washed with H2O (3×10 mL). The aqueous layers were combined and extracted with CH2Cl2 (3×30 mL). The organic layers were combined, dried with MgSO4, and concentrated under reduced pressure to afford a pale yellow oil. The oil was purified by flash chromatography over silica (100% hexanes) to afford 0.70 g (77%) of 5 as a colorless oil. MALDI-FTMS 341.1932 m/z (M+H)+, C21H29O2Si requires 341.1931.
  • 1-Triisopropylsilanyloxy-dibenzofuran-4,6-dicarboxylic acid dimethyl ester (6). Silyl ether 5 (654 mg, 1.92 mmol) was added to a dry 50 mL round bottom flask followed by Et2O (7.4 mL) and TMEDA (0.87 mL, 5.77 mmol). The flask was cooled to −78° C. in an acetone/CO2(s) bath for 10 min before adding sec-butyl lithium (4.44 mL of a 1.3 M solution in cyclohexane, 5.77 mmol) over 10 minutes. The resulting orange suspension was allowed to warm to room temperature and stirred for 24 h. The flask was cooled again to −78° C. as described above and a 15 psi stream of CO2(g) was bubbled through the reaction suspension (the CO2 was dried by passing it through a drying tube containing activated silica). Following initial addition of CO2(g), the cooling bath was removed and the reaction was stirred for 30 min. The reaction mixture was poured into a 1 L beaker containing ice water (50 mL). The solution was brought to pH 9 by the slow addition of 0.05 M KOH, and then cooled to 0° C. with an ice/H2O bath. The solution was acidified to pH 2 with 0.5 M HCl causing a white solid to precipitate. The aqueous suspension (pH 2) was transferred into a 1 L separatory funnel and extracted with EtOAc (5×50 mL). The combined extracts were dried with MgSO4 and concentrated under reduced pressure to afford the crude diacid as an oil. The 100 mL flask containing the crude diacid was equipped with a stir bar, capped with a septum and evacuated. The flask was then back-filled with argon. Anhydrous MeOH (2 mL) and ACS reagent grade benzene (8 mL) were added via syringe. Trimethylsilyidiazomethane (TMSCHN2; 2.5 mL of a 2 M solution in hexanes, 5 mmol) was added slowly via syringe through the septum. Upon completion of the TMSCHN2 addition the reaction was stirred for 10 min and the solvent removed under reduced pressure to afford a red oil. The residue was purified by flash chromatography over silica (15% EtOAc in hexanes) to afford 0.36 g (43%) of 6 as a white solid. MALDI-FTMS 479.1874 m/z (M+Na)+, C25H32O6SiNa requires 479.1860.
  • 1-Hydroxy-dibenzofuran-4,6-dicarboxylic acid dimethyl ester (7). A dry 100 mL round bottom flask was equipped with a stir bar, charged with 6 (363 mg, 0.95 mmol), capped with a septum, evacuated, and back-filled with argon. Anhydrous THF (6.3 mL) and tetra-butylammonium fluoride (1 M in THF, 1.2 mL, 1.19 mmol) were added to the reaction by syringe. The reaction was stirred for 1 h at room temperature and then poured into 30 mL of H2O in a 250 mL separatory funnel. The aqueous layer was extracted with CHCl3 (4×20 mL). The organic layers were combined, dried with MgSO4, and concentrated under reduced pressure. The residue was purified by flash chromatography over silica (30% EtOAc in hexanes) to afford 0.23 g (97%) of 7 as a white solid. LC-MS m/z 301, C16H12O6 requires 301.
  • 1-Trifluoromethanesulfonyloxy-dibenzofuran-4,6-dicarboxylic acid dimethyl ester (8). The triflation procedure previously described by Stille was used to synthesize 8 (Echavarren, A. M.; et al. J. Am. Chem. Soc. 1987, 109, 5478-5486). Phenol 7 (120 mg, 0.4 mmol) was added to a dry 10 mL round bottom flask, which was then fitted with a septum. The solvent, anhydrous pyridine (2 mL), was added by syringe through the septum. The reaction mixture was cooled to 0° C. with an ice/H2O bath. To initiate the reaction, trifluoromethanesulfonic anhydride (81 μL, 12 mmol) was added by syringe through the septum. The ice bath was removed and the reaction was allowed to warm to room temperature and stirred overnight. The reaction mixture was poured into a 250 mL beaker containing 30 mL of an ice/H2O slurry and transferred into a 125 mL separatory funnel. The aqueous layer was extracted with Et2O (4×40 mL). The organic layers were combined, washed with saturated CuSO4 (4×20 mL) and brine (2×20 mL), dried over MgSO4, and then the Et2O was removed under reduced pressure to afford a slightly yellow solid. The solid was purified by flash chromatography over silica (30% EtOAc in hexanes) to afford 159 mg (92%) of 8 as a white solid. FAB-MS (NBA/NaI) m/z 433.0215 (M+H)+, C17H12F3O8S requires 433.0205.
  • Representative Procedure for the Palladium Catalyzed Cross Coupling of 8 with Substituted Anilines.
  • The aryl coupling procedure reported by Buchwald and Hartwig was used to prepare compounds 9-23. A flame dried 10 mm by 13 cm borosilicate test tube, equipped with a stir bar and capped with a septum, was charged with 8 (140 mg, 0.324 mmol), palladium dibenzylidene acetone, Pd2(dba)3 (15 mg, 0.016 mmol), (±)-binap (15 mg, 0.024 mmol), Cs2CO3 (147 mg, 0.456 mmol), and aniline (32 μL, 0.356 mmol). Upon addition of all reagents the tube was purged with argon for 10 min. Anhydrous toluene (2.4 mL) was then added through the septum and the reaction mixture was heated to 100° C. for 36 h in an oil bath. The reaction mixture was filtered through Celite, and the solvent was removed from the filtrate under reduced pressure. The resulting dark oil was purified by flash chromatography over silica (30% EtOAc in hexanes) to afford biaryl amine 17 as a white solid (0.12 g, 68%). Refer to the supporting information for specific synthetic details and characterization data for compounds 10-23 analogous to that reported for 9 below.
  • 1-Phenylamino-dibenzofuran-4,6-dicarboxylic acid dimethyl ester (9).
  • MALDI-FTMS 375.1094 m/z (M)+, C22H17NO5 requires 375.1106.
  • Representative Procedure for the Copper-Mediated Cross-Coupling of Phenol 7 with Substituted Phenylboronic Acids to Afford 1-Phenoxydibenzofurans 39-43.
  • The biaryl ether coupling was directly adapted from the procedures reported by Chan and Evans. A 20 mL scintillation vial equipped with a magnetic stir bar was charged with phenol 7 (150 mg, 0.50 mmol), copper (II) acetate (91 mg, 0.5 mmol), freshly activated 4 Å molecular sieves (˜250 mg), and phenylboronic acid (180 mg, 1.5 mmol). Dichloromethane (5 mL) was added followed by pyridine (201 μL, 2.5 mmol), resulting in an aqua colored suspension. The cap was very loosely applied such that the reaction suspension was partly open to the atmosphere. The reaction was monitored by TLC. After completion, the reaction mixture was adsorbed onto ˜6 g of silica gel, adding silica gel to the reaction mixture, then removing the solvent under reduced pressure. Chromatography (30% EtOAc in hexanes) of the reaction mixture over silica afforded biaryl ether 39 as a white solid (29 mg, 15%). Refer to the supporting information for specific synthetic details and characterization data for compounds 40-43 analogous to that reported for 39 below.
  • 1-Phenoxy-dibenzofuran-4,6-dicarboxylic acid dimethyl ester (39)
  • MALDI-FTMS 399.0825 m/z (M+Na)+, C22H16O6Na requires 399.0839.
  • Representative Procedure for the Palladium Catalyzed Cross-Coupling of Triflate 8 with Substituted Phenylboronic Acids.
  • A flame dried 10 mm by 13 cm test tube, equipped with a stir bar and capped with a septum, was charged with 8 (100 mg, 0.23 mmol), Pd(PPh3)4 (14 mg, 0.01 mmol), LiCl (29 mg, 0.69 mmol), Na2CO3 (300 μL of a 2 M aqueous solution) and toluene (3 mL). Phenylboronic acid (43 mg, 0.35 mmol) was dissolved in EtOH (0.5 mL) and added to the reaction mixture. MeOH replaced EtOH in this procedure for all other compounds because transesterification was observed; therefore compound 49 was isolated as the diethyl ester and all other compounds as dimethyl esters. After the reagents were added, the tube was purged with argon and the reaction mixture heated to 100° C. for 12 h in an oil bath. The reaction mixture was then filtered through Celite. The solvent was removed under reduced pressure from the filtrate and the resulting dark residue was purified by flash chromatography over silica to afford biaryl 49 as a white solid (52 mg, 63%). Refer to the supporting information for specific synthetic details and characterization data for compounds 50-59 analogous to that reported for 49 below.
  • 1-Phenyl-dibenzofuran-4,6-dicarboxylic acid diethyl ester (49). MALDI-FTMS 411.1197 m/z (M+Na)+, C24H20O5Na requires 411.1203.
  • Representative Procedure for Ester Hydrolysis to Afford Final Inhibitors 24-38, 44-48, and 60-70.
  • Methyl ester 9 (25 mg, 0.067 mmol) was saponified in THF: MeOH: H2O (3:1:1, 1 mL) in a 20 mL scintillation vial equipped with a stir bar. LiOH.H2O (22 mg, 0.53 mmol) was added to the suspension and the reaction was allowed to stir until completion (typically 4 h) as determined by TLC or analytical reverse phase HPLC monitoring. The reaction mixture was diluted with brine (2 mL) and acidified to pH 2 with 1 M HCl (pH paper) resulting in a biphasic solution. The upper layer (THF) was removed and the aqueous layer was extracted with THF (3×3 mL). The combined organic layers were dried with MgSO4 and then concentrated under reduced pressure to afford diacid 24 as a white solid (21 mg, 92%). Refer to the supporting information for specific synthetic details and characterization data for compounds 25-38, 44-48, and 60-70 analogous to that reported for 24 below.
  • 1-Phenylamino-dibenzofuran-4,6-dicarboxylic acid (24). MALDI-FTMS 347.0794 m/z (M)+, C20H13NO5 requires 347.0788.
  • FIG. 1A shows an X-ray crystallographic structure of TTR•12 (Klabunde, T.; et al. Nature Struct. Biol. 2000, 7, 312-321). The residues lining the binding site are displayed as stick models (oxygen in red, nitrogen in blue, and carbon in gray), with the protein's Connolly surface depicted in gray. Compound 1 is shown in both of its C2 symmetry equivalent binding modes (yellow and green). The binding channel has 3 sets of depressions referred to as the halogen binding pockets (HBPs) because they interact with the iodines of thyroxine. Compound 1 occupies only the outer portion of the binding pocket and fills both HBP1 and 1′. The carboxylic acids of 1 are in proximity to the e-NH3 + of K15 and K15′.
  • FIG. 1B shows a line drawing representation of the design of the 1-substituted-dibenzofuran-4,6-dicarboxylic acids placed in the thyroxine binding pocket where X represents either an NH, O or direct Caryl-Caryl linkage. R represents the substituents of the aryl ring designed to complement TTR's inner binding cavity.
  • FIG. 2 is a table highlighting the concentration dependent acid-substituted dibenzofuran activity against WT-TTR (3.6 μM) amyloid fibril formation (f.f.) at pH 4.4 (72 h). Values represent the extent of f.f. and thus inhibitor efficacy relative to WT-TTR fibril formation in the absence of inhibitor (assigned to be 100%): complete inhibition is equivalent to 0% f.f.
  • FIG. 3 is a chart showing a summary of dibenzofuran-based amyloid inhibition activity (3.6 mM) against WT-TTR (3.6 mM) fibril formation (pH 4.4, 72 h) and binding stoichiometry to TTR in human blood plasma. % Fibril formation (f.f.) values in the middle column represent the extent of f.f., and thus inhibitor efficacy, relative to WT-TTR f.f. in the absence of inhibitor (assigned to be 100%). Complete inhibition is equivalent to 0% f.f. The right column depicts the observed stoichiometry of inhibitor (dosed at 10.8 mM, ˜2-3× the concentration of plasma TTR) bound to TTR in blood plasma as determined using the antibody capture method.
  • FIG. 4 is a scheme for the synthesis of 1-hydroxy-dibenzofuran-4,6-dicarboxylate dimethyl ester and the corresponding triflate: a) K3[Fe(CN)6], KOH, H2O, benzene; b) AlCl3, toluene, 33% for both steps; c) TIPSCI, DMAP, CH2Cl2, 77%; d) sec-BuLi, Et2O, −78° C., gaseous CO2, TMSCHN2, 43%; e) TBAF, THF, 97%; f) Tf2O, pyridine, 92%.
  • FIG. 5 is a scheme for the synthesis of 1-phenyl-, phenoxy-, and phenylamine-dibenzofuran-4,6-dicarboxylate dimethyl esters and the corresponding dicarboxylates: a) Pd2(DBA)3, (±)binap, Cs2CO3, toluene 100° C.; b) LiOH—H2O, THF/MeOH/H2O (3:1:1); c) CuII(OAc)2, pyridine, 4 Å MS, CH2Cl2; d) Pd(PPh3)4, LiCl, aq. Na2CO3, toluene, MeOH, 80° C.
  • FIG. 6 is a chart showing dibenzofuran-based inhibitor activity (7.2 μM) against WT-TTR (3.6 μM) amyloid fibril formation (f.f.) at pH 4.4 (72 h). Values represent the extent of f.f. and thus inhibitor efficacy relative to WT-TTR fibril formation in the absence of inhibitor (assigned to be 100%): complete inhibition is equivalent to 0% f.f.
  • FIG. 7 is a table illustrating dibenzofuran plasma TTR binding stoichiometry plotted vs. fibril formation inhibition efficacy. The lightly shaded area corresponds to the definitions of high activity and high selectivity (<40% fibril formation and a binding stoichiometry >1), while the darkly shaded area corresponds to exceptional compounds (<30% fibril formation and a binding stoichiometry >1.25). Data points identify the three different linkers: NH (▴), ◯ ( ), and direct Caryl-Caryl linkage (●). Dibenzofuran-4,6-dicarboxylic acid (1) data point (●) shown for comparison.
  • FIG. 8 is a plot of the absorbance at 280 nm versus distance from the center in the sedimentation velocity study on TTR (3.6 mM) after being preincubated with 27 (7.2 mM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor. Velocity analysis—overlay of data sets taken 15 min apart at 50,000 rpm. The data (symbols) fit to a single ideal species model (solid line) with MW 57.1±0.2 kDa.
  • FIG. 9 is a plot of the absorbance at 280 nm versus distance from the center in the equilibrium ultracentrifugation studies on TTR (3.6 mM) after being preincubated with 27 (7.2 mM) and after another incubation period where the pH was dropped to 4.4 for 72 h, a time frame that results in maximal amyloid formation in the absence of inhibitor. Equilibrium analysis—equilibrium concentration gradient observed after a 24 h application of centrifugal force to the sample employing at a speed of 17,000 rpm. The data (◯) fit to a single ideal species model (solid line) with MW 55.0±0.2 kDa. The residuals, the difference between experimental and fitted data, are shown in the inset.
  • FIG. 10 is a plot of the timecourse analysis of WT-TTR (3.6 μM) fibril formation mediated by partial acid denaturation in the absence (▴) and presence of 7.2 μM (⋄) and 3.6 μM (◯) inhibitors 25, 47, and 64, as measured by turbidity at 500 nm (see color scheme within Figure to differentiate inhibitors). It is hard to discern which compound is most efficacious in the black and white plot. At the end of the plot or after 169 hours have passed, compound 47 shows the most fibrils formed followed by compound 64 followed by compound 25.
  • FIG. 11 is a plot of the timecourse analysis of WT-TTR (3.6 μM) tetramer dissociation (6.0 M urea) in the absence (▴) and presence of 7.2 μM (⋄) and 3.6 μM (◯) concentrations of inhibitors 25, 47, and 64 (see color scheme within Figure to differentiate inhibitors). Slow tetramer dissociation is not detectable by far-UV CD spectroscopy, but this process is linked to rapid (˜500,000× faster) monomer denaturation as monitored by loss of β-sheet content easily followed by circular dichroism spectroscopy. It is hard to discern which compound is most efficacious in the black and white plot. At the end of the plot or after 169 hours have passed, compound 25 is most effective in dissociating tetramers followed by compound 64 followed by compound 47.

Claims (17)

1. A compound represented by Formula I:
Figure US20050261365A1-20051124-C00011
wherein:
X is absent or is a diradical selected from the group consisting of —O—, —S—, and —NH—; and
R2, R3, R4, and R5 are radicals independently selected from the group consisting of —H, OH, —F, —Cl, —Br, —CF3, and —CO2H.
2. A compound according to claim 1 represented by Formula II:
Figure US20050261365A1-20051124-C00012
3. A compound according to claim 2 wherein:
R2 is a radical selected from the group consisting of —H, —F, —Cl, and —CF3.
4. A compound according to claim 2 wherein:
R4 is a radical selected from the group consisting of —H, —Cl, and —CO2H.
5. A compound according to claim 2 wherein:
R5 is a radical selected from the group consisting of —H, —F, and —Cl.
6. A compound according to claim 2 selected from the group represented by the following structures:
Figure US20050261365A1-20051124-C00013
Figure US20050261365A1-20051124-C00014
Figure US20050261365A1-20051124-C00015
7. A compound according to claim 1 represented by the following structure:
Figure US20050261365A1-20051124-C00016
8. A compound according to claim 7 wherein:
R3 is a radical selected from the group consisting of —H, —F, —Cl, —Br, and —CF3.
9. A compound according to claim 7 wherein:
R5 is a radical selected from the group consisting of —H, —F, —Cl, and —Br.
10. A compound according to claim 7 selected from the group represented by the following structures:
Figure US20050261365A1-20051124-C00017
11. A compound according to claim 1 represented by the following structure:
Figure US20050261365A1-20051124-C00018
12. A compound according to claim 11 wherein:
R2 is a radical selected from the group consisting of —H, —F, and —Cl.
13. A compound according to claim 11 wherein:
R3 is a radical selected from the group consisting of —H, —F, —Cl, —CF3, and —CO2H.
14. A compound according to claim 11 wherein:
R4 is a radical selected from the group consisting of —H, and —CO2H.
15. A compound according to claim 11 wherein:
R5 is a radical selected from the group consisting of —H, —F, —Cl, and —CF3.
16. A compound according to claim 11 selected from the group represented by the following structures:
Figure US20050261365A1-20051124-C00019
Figure US20050261365A1-20051124-C00020
17. A process comprising the step of contacting transthyretin with a concentration of a compound selected from claims 1-16 sufficient for inhibiting amyloid fibril formation.
US11/134,527 2004-05-20 2005-05-20 Transthyretin stabilization Abandoned US20050261365A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/134,527 US20050261365A1 (en) 2004-05-20 2005-05-20 Transthyretin stabilization

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57372004P 2004-05-20 2004-05-20
US11/134,527 US20050261365A1 (en) 2004-05-20 2005-05-20 Transthyretin stabilization

Publications (1)

Publication Number Publication Date
US20050261365A1 true US20050261365A1 (en) 2005-11-24

Family

ID=34970266

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/134,628 Active 2026-02-28 US7868033B2 (en) 2004-05-20 2005-05-20 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US11/134,848 Abandoned US20050272704A1 (en) 2004-05-20 2005-05-20 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US11/134,527 Abandoned US20050261365A1 (en) 2004-05-20 2005-05-20 Transthyretin stabilization
US11/596,832 Abandoned US20080319175A1 (en) 2004-05-20 2005-05-20 Transthyretin Stabilization
US12/979,733 Active US8338459B2 (en) 2004-05-20 2010-12-28 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/134,628 Active 2026-02-28 US7868033B2 (en) 2004-05-20 2005-05-20 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US11/134,848 Abandoned US20050272704A1 (en) 2004-05-20 2005-05-20 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/596,832 Abandoned US20080319175A1 (en) 2004-05-20 2005-05-20 Transthyretin Stabilization
US12/979,733 Active US8338459B2 (en) 2004-05-20 2010-12-28 Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding

Country Status (12)

Country Link
US (5) US7868033B2 (en)
EP (3) EP1755585A4 (en)
JP (2) JP2007538095A (en)
KR (2) KR20070013335A (en)
CN (2) CN1976693A (en)
AU (2) AU2005244999A1 (en)
BR (2) BRPI0510142A (en)
CA (2) CA2566923A1 (en)
MX (1) MXPA06013381A (en)
RU (1) RU2371440C2 (en)
WO (3) WO2005112913A1 (en)
ZA (1) ZA200610417B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103038345A (en) * 2010-04-29 2013-04-10 Isis制药公司 Modulation of transthyretin expression

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4865958B2 (en) * 2001-05-23 2012-02-01 株式会社トクホン Analgesic anti-inflammatory patch with local action
EP1988397B8 (en) * 2002-12-19 2012-02-15 The Scripps Research Institute Compositions and uses for stabilising transthyretin and inhibiting transthyretin misfolding
WO2005112913A1 (en) * 2004-05-20 2005-12-01 The Scripps Research Institute Transthyretin stabilization
WO2008063579A2 (en) * 2006-11-17 2008-05-29 Nintendo Co., Ltd. Systems and methods for downloading video game programs
EP2509953B1 (en) 2009-12-11 2016-03-30 Genecode AS Methods of facilitating neural cell survival using gdnf family ligand (gfl) mimetics or ret signaling pathway activators
EP2635907A4 (en) 2010-11-05 2014-04-16 Univ Brandeis Ice cleaved alpha-synuclein a biomarker
MX2014003043A (en) 2011-09-16 2015-02-05 Pfizer Solid forms of a transthyretin dissociation inhibitor.
EP2770988B1 (en) * 2011-10-24 2016-07-20 Som Innovation Biotech S.L. New therapy for transthyretin-associated amyloidosis
KR102095699B1 (en) * 2011-11-18 2020-04-02 알닐람 파마슈티칼스 인코포레이티드 RNAi AGENTS, COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING TRANSTHYRETIN (TTR) ASSOCIATED DISEASES
EP2953970A4 (en) 2013-02-08 2016-06-29 Misfolding Diagnostics Inc Transthyretin antibodies and uses thereof
CN105246879A (en) 2013-05-28 2016-01-13 英派尔科技开发有限公司 Humic acid derivatives and methods of preparation and use
US9932319B2 (en) 2013-05-28 2018-04-03 Empire Technology Development Llc Antioxidant humic acid derivatives and methods of preparation and use
CN105555314B (en) 2013-06-28 2019-02-26 英派尔科技开发有限公司 Edible plasticizer for food and food package film
US11117877B2 (en) 2014-11-21 2021-09-14 Bsim Therapeutics, S.A. 2-thioxothiazolidin-4-one derivatives active as transthyretin ligands and uses thereof
BR112017027205A2 (en) 2015-06-15 2019-06-18 Bsim2 Biomolecular Simulations S A bis-furan derivatives as transthyretin stabilizers (ttr) and amyloid inhibitors for the treatment of familial amyloid polyneuropathy (fap).
AR114136A1 (en) 2017-10-10 2020-07-29 Hoffmann La Roche HETEROCYCLIC COMPOUNDS
GB2571950A (en) 2018-03-13 2019-09-18 Azad Pharma Ag New polymorph and new path to synthesize tafamidis
IL280664B2 (en) * 2018-08-06 2023-04-01 Univ Leland Stanford Junior 2-arylbenzimidazoles as ppargc1a activators for treating neurodegenerative diseases
CA3141998A1 (en) * 2019-05-31 2020-12-03 Plex Pharmaceuticals, Inc. Pharmacological agents for treating protein aggregation diseases of the eye
WO2021019448A1 (en) * 2019-08-01 2021-02-04 Honour (R&D) Process for the preparation of transthyretin dissociation inhibitor
WO2021154842A1 (en) 2020-01-28 2021-08-05 Protego Biopharma, Inc. Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
WO2022112919A1 (en) * 2020-11-25 2022-06-02 Pfizer Inc. (aza)benzothiazolyl substituted pyrazole compounds
CN112362432B (en) * 2021-01-12 2021-04-09 广州科方生物技术股份有限公司 Dissociation agent universally used in serum TT3 and TT4 and preparation method thereof

Family Cites Families (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE274490C (en)
FR769405A (en) * 1933-03-09 1934-08-25 Ig Farbenindustrie Ag Process for producing dyes for vats
DE602336C (en) 1933-03-09 1934-09-06 I G Farbenindustrie Akt Ges Process for the production of Kuepen dyes of the anthraquinone series
US3443943A (en) * 1967-07-24 1969-05-13 Polaroid Corp Photographic products and processes employing ring - closing 2 - equivalent silver halide developing agents
US3551443A (en) 1968-10-30 1970-12-29 Ciba Ltd 2-phenylbenzoxazole derivatives
US3710795A (en) 1970-09-29 1973-01-16 Alza Corp Drug-delivery device with stretched, rate-controlling membrane
GB1429184A (en) 1972-04-20 1976-03-24 Allen & Hanburys Ltd Physically anti-inflammatory steroids for use in aerosols
US4044126A (en) 1972-04-20 1977-08-23 Allen & Hanburys Limited Steroidal aerosol compositions and process for the preparation thereof
USRE29608E (en) * 1972-05-18 1978-04-11 Lilly Industries Limited Benzoxazole derivatives
USRE28819E (en) 1972-12-08 1976-05-18 Syntex (U.S.A.) Inc. Dialkylated glycol compositions and medicament preparations containing same
DE2314238A1 (en) 1973-03-22 1974-09-26 Henkel & Cie Gmbh Anti-inflammatory 2-(tetrazol-5'-yl)-benzoxazoles - prepd. from 2-cyano-benzoazoles and used in cosmetics
DE2619547A1 (en) 1976-05-04 1977-11-24 Dynamit Nobel Ag PROCESS FOR THE PREPARATION OF 2-ARYL-BENZOXAZOLEN AND 2-ARYL-BENZTHIAZOLEN
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4358603A (en) 1981-04-16 1982-11-09 Syntex (U.S.A.) Inc. Acetal stabilized prostaglandin compositions
US4416892A (en) * 1981-04-23 1983-11-22 Lilly Industries Limited Method of treating hypersensitivity disease with benzoxazole derivatives
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
DE3916729A1 (en) * 1989-05-23 1990-12-06 Behringwerke Ag FLUOROGENIC COMPOUNDS AND THEIR USE
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
JPH03258770A (en) * 1990-03-08 1991-11-19 Tanabe Seiyaku Co Ltd Benzazole derivative, preparation thereof and synthetic intermediate theroef
DE4011106A1 (en) * 1990-04-06 1991-10-10 Bayer Ag NEW HETEROCYCLIC SUBSTITUTED DIHYDROPYRIDINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN MEDICINAL PRODUCTS
US5254692A (en) 1990-04-06 1993-10-19 Bayer Aktiengesellschaft 2,6-dialkyl-4-(benzothiazol- or benzoxazol-7-yl)-1,4-dihydropyridines
IT1246382B (en) 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
US5037842A (en) 1990-06-05 1991-08-06 Pfizer Inc. Oxa- and thiazolidinedione hypoglycemic and hypocholesterolemic agents
CA2051518A1 (en) 1990-10-01 1992-04-02 Robert J. Perry Synthesis of heterocyclic compounds
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
JP3021028B2 (en) 1990-11-19 2000-03-15 株式会社日本化学工業所 Azole type cationic dye
CH681806A5 (en) 1991-03-19 1993-05-28 Ciba Geigy Ag
JPH0517458A (en) 1991-07-04 1993-01-26 Idemitsu Kosan Co Ltd Aromatic compound and its production
US5354759A (en) * 1991-09-12 1994-10-11 Fujisawa Pharmaceutical Co., Ltd. Angiotenin II antagonizing heterocyclic compounds
US6010715A (en) 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
JPH0673051A (en) 1992-08-26 1994-03-15 Asahi Glass Co Ltd Benzazole derivative
JPH0673050A (en) 1992-08-26 1994-03-15 Asahi Glass Co Ltd Heterocyclic compound
GB9218334D0 (en) 1992-08-28 1992-10-14 Ici Plc Heterocyclic compounds
DE4304650A1 (en) 1993-02-16 1994-08-18 Thomae Gmbh Dr K Condensed 5-membered heterocycles, processes for their preparation and pharmaceutical compositions containing them
JPH06239849A (en) 1993-02-16 1994-08-30 Canon Inc Optically active compound, liquid crystal composition containing the same, liquid crystal element having the same composition and display method and display device using the same
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
WO1994022846A1 (en) * 1993-03-30 1994-10-13 Pfizer Inc. Compounds enhancing antitumor activity of other cytotoxic agents
US5523092A (en) 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
JP3243887B2 (en) 1993-05-28 2002-01-07 三菱化学株式会社 Organic electroluminescent device
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
JPH0797379A (en) 1993-09-06 1995-04-11 Canon Inc Liquid crystal compound, liquid crystal composition containing the same, liquid crystal element having the same, display method and display device using the element
US5744368A (en) 1993-11-04 1998-04-28 Research Foundation Of State University Of New York Methods for the detection of soluble amyloid β-protein (βAP) or soluble transthyretin (TTR)
US5441946A (en) * 1994-04-14 1995-08-15 Rhone-Poulenc-Rorer Pharmaceuticals, Inc. Phosphonate derivatives of lipophilic amines
US5552426A (en) 1994-04-29 1996-09-03 Eli Lilly And Company Methods for treating a physiological disorder associated with β-amyloid peptide
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
DK0788511T3 (en) * 1994-10-07 2003-03-31 Fujisawa Pharmaceutical Co Cyclic hexapeptides with antibiotic activity
US5660854A (en) 1994-11-28 1997-08-26 Haynes; Duncan H Drug releasing surgical implant or dressing material
US5983134A (en) 1995-04-23 1999-11-09 Electromagnetic Bracing Systems Inc. Electrophoretic cuff apparatus drug delivery system
US5837390A (en) * 1995-05-10 1998-11-17 Sony Corporation Metal complex, method for producing the same and optical device
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US6167301A (en) 1995-08-29 2000-12-26 Flower; Ronald J. Iontophoretic drug delivery device having high-efficiency DC-to-DC energy conversion circuit
US6130217A (en) * 1995-09-20 2000-10-10 Pfizer Inc Compounds enhancing antitumor activity of other cytotoxic agents
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
JP3861930B2 (en) 1995-12-15 2006-12-27 ソニー株式会社 Metal binuclear complex, method for producing the same, and optical element
JPH09227576A (en) 1996-02-23 1997-09-02 Sony Corp Metallic multinuclear complex, its production and optical element
TW345603B (en) 1996-05-29 1998-11-21 Gmundner Fertigteile Gmbh A noise control device for tracks
JP3760508B2 (en) 1996-06-10 2006-03-29 東洋インキ製造株式会社 Organic electroluminescence device material and organic electroluminescence device using the same
US5985317A (en) 1996-09-06 1999-11-16 Theratech, Inc. Pressure sensitive adhesive matrix patches for transdermal delivery of salts of pharmaceutical agents
AU722289B2 (en) 1996-10-01 2000-07-27 Aptalis Pharmatech, Inc. Taste-masked microcapsule compositions and methods of manufacture
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
WO1998027972A2 (en) 1996-12-23 1998-07-02 Texas A & M University Anti-amyloidogenic agents
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US5948433A (en) 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
FR2767527B1 (en) 1997-08-25 1999-11-12 Pf Medicament INDOLIC PIPERAZINE DERIVATIVES USEFUL AS MEDICAMENTS AND PREPARATION METHOD
JP4395200B2 (en) 1997-10-28 2010-01-06 バンドー化学株式会社 Skin patch sheet and method for producing substrate sheet therefor
JP3279974B2 (en) 1997-12-03 2002-04-30 帝人株式会社 Antistatic polyester film
GB9725782D0 (en) 1997-12-05 1998-02-04 Pfizer Ltd Therapeutic agents
WO1999040087A2 (en) * 1998-02-04 1999-08-12 The Board Of Regents, The University Of Texas System Inhibition of human telomerase by a g-quadruplex-interaction compound
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6107491A (en) * 1998-07-20 2000-08-22 Ciba Specialty Chemicals Corporation Polymerizable diketopyrrolopyrroles
GB9816654D0 (en) * 1998-07-30 1998-09-30 Zeneca Ltd Chemical compounds
JP2000100569A (en) 1998-09-22 2000-04-07 Toray Ind Inc Luminescent element
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
JP2001055332A (en) 1999-06-07 2001-02-27 Saitama Daiichi Seiyaku Kk Iontophresis preparation containing aromatic amidine derivatives
US6256533B1 (en) 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
WO2000078733A1 (en) 1999-06-18 2000-12-28 Bayer Aktiengesellschaft Phenoxy fluoropyrimidines
JP2001064205A (en) 1999-06-25 2001-03-13 Dai Ichi Seiyaku Co Ltd Medication composition
JP2001064166A (en) 1999-06-25 2001-03-13 Dai Ichi Seiyaku Co Ltd Cataplasm
WO2001012183A1 (en) 1999-08-16 2001-02-22 Merck & Co., Inc. Heterocycle amides as cell adhesion inhibitors
GB9919673D0 (en) 1999-08-20 1999-10-20 Cancer Res Campaign Tech 2-Arlybenzazole compounds
JP2001226358A (en) 1999-10-12 2001-08-21 Japan Tobacco Inc Lpl potentiator
JP2001242165A (en) 2000-02-25 2001-09-07 Dai Ichi Seiyaku Co Ltd Reagent for collecting blood
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
RU2268258C2 (en) * 2000-03-16 2006-01-20 Ф.Хоффманн-Ля Рош Аг Derivatives of carboxylic acids as antagonists of ip
GB0007934D0 (en) 2000-03-31 2000-05-17 Darwin Discovery Ltd Chemical compounds
JP3945123B2 (en) 2000-04-10 2007-07-18 三菱化学株式会社 Organic electroluminescence device
JP2001301329A (en) 2000-04-20 2001-10-31 Mitsubishi Chemicals Corp Optical recording medium
JP2002003368A (en) 2000-06-23 2002-01-09 Saitama Daiichi Seiyaku Kk Pharmaceutical preparation for percutaneous absorption or absorption through mucous membrane
WO2002016333A2 (en) 2000-08-24 2002-02-28 University Of Pittsburgh Thioflavin derivatives and their use in diagnosis and theraphy of alzheimer's disease
WO2002036553A2 (en) 2000-11-04 2002-05-10 Aventis Pharma Limited Substituted alkanoic acids
AU2002221239A1 (en) * 2000-12-07 2002-06-18 Astrazeneca Ab Therapeutic benzimidazole compounds
EP1345914A1 (en) 2000-12-22 2003-09-24 AstraZeneca AB Therapeutic compounds
US20020160394A1 (en) 2001-01-24 2002-10-31 Bayer Corporation Regulation of transthyretin to treat obesity
GB0118357D0 (en) * 2001-07-27 2001-09-19 Syngenta Ltd Chemical compounds
AU2002334355A1 (en) 2001-09-06 2003-03-18 Prochon Biotech Ltd. Protein tyrosine kinase inhibitors
US6602619B2 (en) * 2001-10-19 2003-08-05 Lightronik Technology Inc. Organic EL device
AU2002353739A1 (en) 2001-11-28 2003-06-10 Astrazeneca Ab Therapeutic compounds
UA83620C2 (en) * 2001-12-05 2008-08-11 Уайт Substituted benzoxazoles and analogues as estrogenic agents
RU2004121898A (en) * 2001-12-19 2006-01-20 Атеродженикс, Инк. (Us) CHALKONE DERIVATIVES AND THEIR APPLICATION FOR TREATMENT OF DISEASES
WO2003053359A2 (en) * 2001-12-19 2003-07-03 Atherogenics, Inc. 1,3-bis-(substituted-phenyl)-2-propyn-1-ones and their use to treat disorders
GB0205256D0 (en) 2002-03-06 2002-04-17 Oxford Glycosciences Uk Ltd Novel compounds
NZ535603A (en) 2002-03-20 2007-10-26 Metabolex Inc Substituted phenylacetic acids
UA79755C2 (en) 2002-04-16 2007-07-25 Bayer Pharmaceuticals Corp Indane acetic acid derivatives and their use as pharmaceutical agents, intermediates, and method of preparation
GB0226822D0 (en) 2002-11-16 2002-12-24 Oxford Glycosciences Uk Ltd Novel compounds
EP1988397B8 (en) 2002-12-19 2012-02-15 The Scripps Research Institute Compositions and uses for stabilising transthyretin and inhibiting transthyretin misfolding
WO2004063155A1 (en) * 2003-01-06 2004-07-29 Eli Lilly And Company Fused heterocyclic derivatives as ppar modulators
WO2004064771A2 (en) 2003-01-14 2004-08-05 Merck & Co., Inc. Geminally di-substituted nsaid derivatives as abeta 42 lowering agents
US7223788B2 (en) * 2003-02-14 2007-05-29 Sanofi-Aventis Deutschland Gmbh Substituted N-aryl heterocycles, process for their preparation and their use as medicaments
JP2004250411A (en) 2003-02-21 2004-09-09 Bf Kenkyusho:Kk DIAGNOSTIC PROBE FOR AMYLOID beta-ACCUMULATIVE DISEASE AND THERAPEUTIC COMPOUND
EP1611115B1 (en) * 2003-03-14 2012-08-22 University of Pittsburgh - of the Commonwealth System of Higher Education Benzothiazole derivative compounds, compositions and uses
CA2438032C (en) 2003-03-14 2013-05-07 University Of Pittsburgh Benzothiazole derivative compounds, compositions and uses
MY142651A (en) 2003-03-18 2010-12-15 Merck Sharp & Dohme Biaryl substituted triazoles as sodium channel blockers
EP1608622A4 (en) 2003-03-24 2009-04-01 Merck & Co Inc Biaryl substituted 6-membered heterocyles as sodium channel blockers
JP2004302049A (en) 2003-03-31 2004-10-28 Hodogaya Chem Co Ltd Photosensitive resin composition
US7589116B2 (en) 2003-04-03 2009-09-15 Merck & Co. Inc. Biaryl substituted pyrazoles as sodium channel blockers
CN1805945A (en) 2003-04-18 2006-07-19 麦克公司 Biaryl substituted thiazoles, oxazoles and imidazoles as sodium channel blockers
JP3687660B2 (en) 2003-04-22 2005-08-24 松下電工株式会社 Massage machine
EP1620413A2 (en) 2003-04-30 2006-02-01 Cytokinetics, Inc. Compounds, compositions, and methods
WO2005112913A1 (en) * 2004-05-20 2005-12-01 The Scripps Research Institute Transthyretin stabilization

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103038345A (en) * 2010-04-29 2013-04-10 Isis制药公司 Modulation of transthyretin expression
US9816092B2 (en) 2010-04-29 2017-11-14 Ionis Phamaceuticals, Inc. Modulation of transthyretin expression
US11535849B2 (en) 2010-04-29 2022-12-27 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression

Also Published As

Publication number Publication date
EP1768968A1 (en) 2007-04-04
JP2007538103A (en) 2007-12-27
CA2566688A1 (en) 2005-12-01
ZA200610417B (en) 2008-02-27
US20050272704A1 (en) 2005-12-08
US20080319175A1 (en) 2008-12-25
EP2314584A1 (en) 2011-04-27
WO2005118511A2 (en) 2005-12-15
WO2005112913A1 (en) 2005-12-01
CA2566923A1 (en) 2005-12-01
KR20070032700A (en) 2007-03-22
US20110092545A1 (en) 2011-04-21
BRPI0510142A (en) 2007-10-02
US8338459B2 (en) 2012-12-25
BRPI0510033A (en) 2007-10-02
US20050282780A1 (en) 2005-12-22
JP2007538095A (en) 2007-12-27
RU2371440C2 (en) 2009-10-27
CN1976693A (en) 2007-06-06
WO2005118511A3 (en) 2006-02-09
EP1755585A1 (en) 2007-02-28
CN101198598A (en) 2008-06-11
EP1755585A4 (en) 2009-04-01
AU2005245470A1 (en) 2005-12-01
WO2005112913B1 (en) 2006-02-09
RU2006145074A (en) 2008-06-27
US7868033B2 (en) 2011-01-11
WO2005113523A1 (en) 2005-12-01
KR20070013335A (en) 2007-01-30
AU2005244999A1 (en) 2005-12-01
MXPA06013381A (en) 2007-03-01

Similar Documents

Publication Publication Date Title
US20050261365A1 (en) Transthyretin stabilization
US8022250B2 (en) 1-phenylalkanecarboxylic acid derivatives for the treatment of neurodegenerative diseases
TWI406856B (en) Inhibitors of human immunodeficiency virus replication
KR101326358B1 (en) (biphenyl) carboxylic acids and derivatives thereof
Thalhammer et al. Inhibition of the histone demethylase JMJD2E by 3-substituted pyridine 2, 4-dicarboxylates
Chen et al. Highly selective fluorescence turn-on chemosensor based on naphthalimide derivatives for detection of copper (II) ions
HUE027855T2 (en) Method for producing aminothiazole derivative and production intermediate
JP2022522334A (en) New thyroid imitation
JP5847710B2 (en) Compounds as hypoxic mimetics, and compositions thereof, and uses thereof
TWI468110B (en) Preparation of 6-Aryloxyquinoline Derivatives and Intermediates thereof
JP4475868B2 (en) Cephem compound and ESBL detection reagent containing the same
Tanima et al. Development of highly sensitive and selective molecules for detection of spermidine and spermine
Fiandanese et al. Stereoselective total synthesis of (S)-Virol C and (S)-1-dehydroxyvirol A
CN102372531B (en) Preparation method for aromatic carboxylic acid compounds
KR20040039430A (en) Process for producing (2-nitrophenyl)acetonitrile derivative and intermediate therefor
Suzuki et al. Synthesis and characterization of asymmetric o-and m-nitrobenzoic acids with a 1, 3-benzodioxole skeleton
Ma et al. Novel synthetic approach to fluoro-and amido-disubstituted 3-hydroxypyridin-4-ones
KR101718994B1 (en) A chemodosimeter approach to fluorescent sensing and imaging of inorganic and methylmercury species
Kaslow et al. Ortho-Substituted 2-Phenylquinolines1
Hashim et al. A convenient method for the preparation of 3-phenoxy/thiophenoxy-2 (1H)-quinolinones
TWI424842B (en) Heterocyclic compound with substituent, pharmaceutical composition, and the use thereof
JP4020663B2 (en) Process for producing aromatic compounds
Lee et al. Synthesis of 7, 8‐dihydroxychlorpromazine and analogs
CN116239482A (en) Near infrared fluorescent probe and application thereof in preparation of early diagnosis reagent for Alzheimer&#39;s disease
CN114507156A (en) Preparation method of cyanophenyl compound

Legal Events

Date Code Title Description
AS Assignment

Owner name: SCRIPPS RESEARCH INSTITUTE, THE, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KELLY, JEFFERY W.;PETRASSI, H. MICHAEL;REEL/FRAME:016832/0347;SIGNING DATES FROM 20050713 TO 20050721

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE;ASSIGNOR:SCRIPPS RESEARCH INSTITUTE;REEL/FRAME:021840/0388

Effective date: 20050616

STCB Information on status: application discontinuation

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