US20140341812A1 - Beta-amyloid pet imaging agents - Google Patents

Beta-amyloid pet imaging agents Download PDF

Info

Publication number
US20140341812A1
US20140341812A1 US14/223,782 US201414223782A US2014341812A1 US 20140341812 A1 US20140341812 A1 US 20140341812A1 US 201414223782 A US201414223782 A US 201414223782A US 2014341812 A1 US2014341812 A1 US 2014341812A1
Authority
US
United States
Prior art keywords
mmol
alkyl
group
mhz
equiv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/223,782
Inventor
Lisheng Cai
Victor W. Pike
Robert B. Innis
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.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
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 US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US14/223,782 priority Critical patent/US20140341812A1/en
Publication of US20140341812A1 publication Critical patent/US20140341812A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0453Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • A61K49/0433X-ray contrast preparations containing an organic halogenated X-ray contrast-enhancing agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0455Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/041Heterocyclic compounds
    • A61K51/044Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins
    • A61K51/0459Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine, rifamycins having six-membered rings with two nitrogen atoms as the only ring hetero atoms, e.g. piperazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/64Benzothiazoles with only hydrocarbon or substituted hydrocarbon radicals attached in position 2
    • C07D277/66Benzothiazoles with only hydrocarbon or substituted hydrocarbon radicals attached in position 2 with aromatic rings or ring systems directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • AD Alzheimer's disease
  • a ⁇ amyloid- ⁇ peptide
  • Non-invasive detection and quantitation of amyloid deposits in the brain has been used to develop anti-amyloid therapies.
  • Direct imaging of amyloid load in vivo inpatients with AD is useful for the early diagnosis of AD and the development and assessment of treatment strategies.
  • compounds suitable for in vivo imaging of amyloid deposits in human brains have been developed. Among these compounds are monoclonal antibodies against A ⁇ and peptide fragments, but these have had limited uptake by the brain when tested in patients with AD, Putrescine-gadolinium-A ⁇ has been injected into transgenic mice overexpressing amyloid, and this has resulted in labeling observed with MRI.
  • Amyloid deposition can also be non-invasively imaged and quantitated with a radiotracer that readily enters the brain and selectively binds to amyloid deposits.
  • An object of the present invention is to provide radioligands that do not undergo rapid defluoridation and do not produce residual radioactivity in the brain.
  • the invention therefore resides in further analogs of imidazo[1,2-a]pyridine (IMPY) useful as radioligands for the detection of A ⁇ amyloid aggregates in Alzheimer's Disease (AD) patients.
  • IMPY imidazo[1,2-a]pyridine
  • Preferred among these analogs are imidazo[1,2-a]pyridine N-methylbenzeneamines in which a methyl group or a group similar in size to a methyl group (such as a bromine atom) is bonded to the ortho ring position nearest to the amine.
  • An object of the present invention is therefore to provide amyloid imaging agents with high specificity of binding to beta amyloid, low background noise, better entry into brains, and improved labeling efficiencies.
  • the first class are derivatives of imidazopyridinylbenzeneamine (IMPY), and are defined by Formula (I):
  • the second class are derivatives of benzothiazolylbenzeneamine (BTA), and are defined by Formula (II):
  • This invention resides in the above compounds as novel imaging agents for the imaging of amyloid deposits, new methods of synthesis of imidazopyridinylbenzeneamine (IMPY) derivatives, and methods for diagnosing Alzheimer's disease in vivo by positron emission tomography, magnetic resonance imaging and other imaging methods involving the use of the imaging agents of this invention,
  • FIG. 1 Synthesis of ketones and ⁇ -bromoketones as intermediates in the preparation of IMPY derivatives.
  • FIG. 2 Synthesis of 2-aminopyridine derivatives and 2-aminopyrazines.
  • the synthetic methods of pyridine derivatives depend on the substituents on the ring.
  • FIG. 3 Aromatic substitutions with the methoxy anion.
  • the method uses salts of copper (I) and a small amount of ester to form a stabilized tetrahedral adduct.
  • FIG. 4 Synthesis of 5-chloro-2-aminopyrazine using mild conditions
  • FIG. 5 Synthsesis of IMPY Derivatives I. Most of the IMPY derivatives were synthesized through the direct condensation of ⁇ -bromoketones and 2-amino-pyridines.
  • FIG. 6 Synthesis of IMPY Derivatives II using direct methoxylation of halide in preformed IMPY derivatives.
  • FIG. 7 Synthesis of IMPY Derivatives II using catalytic substitution of halides by thiolate-based palladium catalysts
  • Millipore water purification system comprising a combination of two filters, one RioTM, one reservoir, and one Milli-Q®, synthesis system (Bedford, Mass.). Common solvents were obtained from Fisher Scientific (Pittsburgh, Pa.), Brain tissue from deceased Alzheimer's Disease (AD) patients was obtained from Brain Bank of the Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health.
  • Analytical HPLC was performed using a reverse phase column (X-Tera C18; 5 ⁇ m; 10.0 ⁇ 250 mm; Waters) eluted with c. ammonia (0.25%) in acetonitrile-water at 6.2 mL/min.
  • the chromatography system was fitted with a continuous wavelength UV-vis detector (Beckman System Gold 168 Detector) and an autosampler (Beckman System Gold 508 Autosampler),
  • a reverse phase column X-Tera C18; 5 ⁇ m 19 ⁇ 250 mm; Waters
  • the HPLC system was fitted with a manual injector (5 mL injection loop) and a third delivery pump using acetonitrile as eluant at 1 mL/min.
  • the purity of compounds was determined with HPLC monitored for UV absorbance at 280 nm (for IMPY derivatives) or 254 nm (for other aromatic compounds) and expressed as area percentage of all peaks.
  • the 1 H and 13 C NMR spectra of all compounds were acquired on a Jeol GSX 270 (270 MHz 1 H and 68 MHz 13 C), on a Bruker DRX 300 (300 MHz 1 H and 75 MHz 13 C), on a Bruker DRX 400 (400 MHz 1 H and 100 MHz 13 C) and on a Bruker AM500 (500 MHz 1 H and 125 MHz 13 C), using the chemical shifts of residual deuterated solvent as the internal standard; chemical shift ( ⁇ ) data for the proton and carbon resonance were reported in parts per million (ppm) relative to the internal standard.
  • TLC Thin-layer chromatography
  • Silica Gel 60 F254 plates from EM Science and compounds visualized under UV light at either 250 or 360 nm.
  • Flash chromatography was carried out using a Biotage HorizonTM HPFCTM system (Charlottesville, Va., column sizes: 12 mm ⁇ 150 mm, 25 mm ⁇ 150 mm, 40 Mm ⁇ 150 mm) with hexanes and ethyl acetate (EtOAc) as eluents with chromatographic solvent proportions expressed on a volume: volume basis.
  • IR spectra were recorded using a Perkin-Elmer Spectrum One FT-IR spectrometer, and UV/vis spectra were recorded using a Lambda 40 UV/vis spectrometer.
  • Mass spectra were acquired using either Thermo Finnigan LCQ DECA LC-MS (MS-HPLC column: Luna C18; 5 ⁇ m2.0 ⁇ 150 mm; Phenomenex, flow rate; 150 ⁇ L/min, eluent; MeOH and H 2 O mixture) or Thermo Finnigan PolarisQ GC-MS (GC column: capillary RTX-5 ms 30 m ⁇ 0.25 mm, flow rate: 1 mL/min, carrier gas; He), or VG Micromass 7070E and AutoSpec-Q spectrometers. High-resolution mass spectra (HRMS) were acquired from Mass Spectrometry Laboratory, University of Illinois at Urbana-Champaign (Urbana, Ill.).
  • Elemental analyses of selected compounds were carried out by Midwest Microlab (Indianapolis, Ind.) or Galbraith Laboratories, Inc, (Knoxville, Tenn.), or Mr Stephen Boyer, SACS, University of London. The melting points were measured using Electrothermal Mel-Temp Manual Melting Point Apparatus (Fisher Scientific), and uncorrected. A CEM Discover microwave system was used for microwave synthesis (Matthews, N.C.).
  • N-(4-acetyl-2-methylphenyl)-2,2,2-trifluoroacetamide (K4), 2,2,2-trifluoro-N-o-tolylacetamide (8.0 g; 39.4 mmol) was dissolved in 20 mL CS 2 .
  • Bromoacetic bromide (8.4 g; 68.3 mmol) was added dropwise.
  • AlCl 3 (16.0 g; 120 mmol) was added in portions, with drying tube attached to the reaction flask. The reaction mixture was refluxed overnight. After reaction, the solvent was removed. Ice and water were added, and some yellow solid Precipitated.
  • N-(2-bromo-4-(2-bromoacetyl)phenyl)-2,2,2-trifluoro-N-methylacetamide(B5) CuBr 2 (6.0 g; 26.9 mmol) was suspended in 20 mL ethyl acetate. The mixture was heated to reflux. N-(4-acetyl-2-bromophenyl)-2,2,2-trifluoro-N-methylacetamide (4.5 g; 13.9 mmol) dissolved in 20 mL CHCl 3 was added. The mixture was refluxed for 4 hrs, and filtered.
  • Method b 2-Amino-4-bromopyridine (0.10 g; 0.58 mmol), sodium methoxide (0.16 g; 2.9 mmol) and copper powder nanosized activated (0.11 g; 1.74 mmol) were introduced in a microwave glass tube with 1.5 mL of anhydrous DMF and sealed. The tube was introduced in the microwave cavity and heated for 30 min at 140° C. (140C30M75W300Psi).
  • N-(2-Bromo-4-(2′-bromoacetyl)phenyl)-2,2,2-trifluoro-N-methylacetamide (4a) (4.0 g; 9.93 mmol) and 5-bromopyridin-2-amine (7b) (1.7 g; 9.83 mmol) were dissolved in MeOH (20 mL). The reaction mixture was refluxed for 8 h and solvent then removed.
  • N-(4-(6-bromoindolizin-2-yl)-2-methylphenyl)-N-methylacetamide (L16).
  • N-(4-(2′-Bromoacetyl)-2-methylphenyl)-N-methylacetamide (4c) (1.0 g; 3.52 mmol) and 5-bromopyridin-2-amine (7b) (0.73 g; 4.22 mmol) were used. Yield 37%, M.p.
  • N-(4-(6-bromoindolizin2-yl)-2-methylphenyl)-2,2,2-trifluoroacetamide (L17).
  • N-(4-(2-bromoacetyl)-2-methylphenyl)-2,2,2-trifluoroacetamide (3.0 g; 9.26 mmol) and 5-bromopyridin-2-amine (1.5 g; 8.67 mmol) were dissolved in 20 mL ethanol. The mixture was refluxed overnight, and the solvent was removed. CH 2 Cl 2 was added to precipitate a solid, which was filtered and washed with CH 2 Cl 2 , to afford 1.0 g (yield 28%) of product as a yellow powder.
  • N,N-dimethyl-4-(6-(methylthio)-H-imidazo[1,2-a]pyridin-2-yl)benzenamine (T6).
  • 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (90 mg, 0.25 mmol), 1,2-dimethyldisulfane (28 mg, 0.30 mmol), 1,1,1,2,2,2-hexamethyldistannane (97 mg, 0.30 mmol), Pd 2 (dba) 3 (22.8 mg, 0.025 mmol), DiPPF (10.5 mg, 0.025 mmol), and 4.0 mL of toluene were used.
  • a typical isolation 60 g of brain tissue were used. Initially, an equal volume of 1% SDS was added to homogenize. More 1% SDS was added later until 200 mL. The mixture was vortexed, distributed evenly among 10 Beckman ultracentrifuge tubes, and centrifuged by a Coulter type 55.2 Ti ultracentrifuge rotor using RPM: 40,000; G force; 180,000; temp: 4° C.; time: 30 min. The collected bottom pellets were further homogenized and centrifuged twice more as described above using 50 mL and 20 mL of 1% SDS each, and finally in 20 mL 1.2 M sucrose. The supernatant along the top pellet was discarded.
  • the pellets at the bottom were homogenized and centrifuged in 20 mL 1.9 M sucrose.
  • the top pellets were collected using a cell scrapper, and the bottom pellets were discarded.
  • the collected top pellets were homogenized and centrifuged using 20 mL deionized water.
  • the bottom pellets were further homogenized and centrifuged using 1.3 M sucrose solution into one centrifuge tube.
  • the bottom pellets were homogenized and centrifuged using 20 mL deionized water.
  • the collected pellets were homogenized in 30 mL of 50 mM Tris-HCl, pH 8.0, 2 mM CaCl 2 .
  • Collagenase CLS3 120 mg
  • DNAase I (10-15 mg) were added.
  • the mixture was vortexed and incubated for 24-56 hrs at 37° C. in a water bath with shaking. After incubation, the suspension was filtered through 41 ⁇ m Milllpore filter paper. The collected filtrate was centrifuged and the bottom pellets were homogenized and centrifuged one more time using the Tris-HCl solution. The supernatant was discarded, and the pellets were homogenized and centrifuged using 10 mL 1.9 M sucrose. The top pellet and the 1.9M sucrose layer were transferred to a new tube (this is to remove heavy particles such as metals and salts). Deionized water (40 mL) was added. After homogenization and centrifugation, the collected pellets were put into 500 ⁇ L of deionized water to form a homogeneous suspension. It was stored in a refrigerator or freezer for further use.
  • the amyloid suspension prepared above was diluted by a factor of 40,000 to 50,000 in PBS, and 800 ⁇ L suspension was used in each tube.
  • [ 3 H]6-OH-BTA1 with a concentration of 1 mCi/mL stock solution was diluted using ethanol to give an intermediate solution of 1 ⁇ Ci/100 ⁇ L, which was further diluted using PBS to result in a dilute stock solution of 2.7 ⁇ 10 ⁇ 2 ⁇ Ci/100 ⁇ L, and 100 ⁇ L was used in each tube (2 vials with 100 ⁇ L of [ 3 H]6-OH-BTA-1 each and scintillation fluid as references).
  • the other method to synthesize the ⁇ -bromoketones is through Friedel-Crafts reaction to introduce ⁇ -bromoacetyl group directly in the aromatic nucleus.
  • the aromatic amino group in A1 and A2 were protected using trifluoroacetyl or acetyl group, to give A3 and A4.
  • Friedel-Crafts reactions using trifluoroacetyl or bromoacetyl bromide catalyzed by AlCl 3 in CS 2 went smoothly to give B2, B3, B4, B5. 5-7
  • Imidazopyrazine L21 was synthesized from condensation of chloropyrazine P13 and bromoacetophenone B1 (eq 9).
  • the amine 67 was very unreactive though it is very soluble in acetonitrile. Long reaction time and stronger bases such as triethylamine were examined with limited success.
  • An advantage of the IMPY derivatives of the present invention relative to the prior art compound [ 11 C]PIB (6-OH-BTA-1) is that the derivatives of the present invention can be easily labeled with [ 18 F], which has a half-life of about two hours as compared with twenty minutes for [ 11 C].
  • a process for radiolabeling the IMPY derivatives with either label is shown below.
  • a method for isolating human ⁇ -amyloid plaques from brain tissue 23;24 was modified by incorporating repeated homogenization and centrifugation in 1% SDS buffer and by using DNase and collagenase for digestion. The concentration of ⁇ -amyloid plaques was gauged from the concentration of A ⁇ 1-40 and A ⁇ 1-42 monomers, using ELISA to detect dissolved plaques in formic acid.
  • Determination of the binding affinities of new ligands for ⁇ -amyloid is the first step in selecting candidate radioligands for PET studies in humans.
  • Three types of amyloid plaques have been used to assay ligand binding in vitro, namely synthetic aggregates of A ⁇ 1-40 , A ⁇ 1-42 , amyloid plaques from transgenic mice and amyloid plaques from human Alzheimer's disease (AD) brain tissue.
  • AD Alzheimer's disease
  • Tritiated-6-OH-BTA-1 was selected as the reference radioligand based on its use in human PET imaging and high affinity. 28 Displacement curves were created using non-radioactive 6-OH-BTA-1 and other novel ligands.
  • Ki is a measure (in nM units) of the binding affinity of the compound toward beta-amyloid plaques in AD brain tissue, measured through a competitive radioligand displacement in vitro assay using [ 3 H]6-OH-BTA-1 as the reference radioligand.
  • the BTA derivatives were synthesized through the condensation of 2-amino-aryl-thiols and aryl carboxylic acid under acidic conditions.
  • Roher, A. E. Structural alterations in the peptide backbone of beta-amyloid core protein may account for its deposition and stability in Alzheimer's disease. J. Biol. Chem. 1993, 268, 3072-3083.
  • a further aspect of this invention is the novel synthesis of aryl thioether derivatives of IMPY compounds by a rapid palladium-catalyzed cross-coupling under microwave conditions.
  • the cited references are identified under Reference List No. 2.
  • Aromatic thioethers of IMPY compounds are valuable as synthetic intermediates and as therapeutic drags.
  • 1;2 Homogeneous catalysts based on either copper(I) or palladium have been developed for their syntheses.
  • 2;3 Mechanistic studies of palladium-catalyzed aromatic substitution of halo or triflate groups by thiolate show that both three and four coordinate intermediates might be involved.
  • 4-6 Indeed, both mono and bidentate phosphine ligands have been developed for this type of reaction.
  • Copper(I)-based batalysts for similar syntheses are mechanistically much less clear. 13-17
  • the objects of this aspect of the invention are to achieve (a) an one-step introduction of a thioether group into a halogen position in an aryl ring; (b) selective introduction of a thioether group at an iodo position in the presence of other halogen substituents, such as bromo; (c) tolerance of functional groups, especially amino groups; and (d) fast microwave-assisted reaction conditions.
  • a thioether group into a halogen position in an aryl ring
  • tolerance of functional groups, especially amino groups especially amino groups
  • fast microwave-assisted reaction conditions Disclosed herein is a general, efficient, and operationally simple palladium-catalyzed aryl thioether synthesis that achieves these objects.
  • the aryl halide 4-(6-iodoindolizin-2-yl)-N-methylbenzenamine was used as the substrate, with either 2-mercaptoacetamide or 2-mercaptoethanol as the thiolating agent.
  • the catalyst used was CuI.
  • Method (1) 1-5 equiv CuI, 2 equiv ethylene diamine, 2 equiv t-BuOK or K 2 CO 3 in DMSO, and N-methylpyrrolidin-2-one (NMP) or dioxane were used.
  • Method (2) 1-5 equiv CuI, 2 equiv ethylene glycol, and 2 equiv t-BuOK in Py were used.
  • Method (3) 0.05 equiv CuI, and 2 equiv Cs 2 CO 3 in NMP were used.
  • a Without the thiol, the reaction generated reduced and HOCH 2 CH 2 O-substituted products.
  • b No reaction was observed when 1,2-dimethoxyethane was used instead of ethylene glycol.
  • the new ligands behave similarly to PPh 3 , giving less than 10% total conversion of aryl iodide and ratios of substitution and reduction ranging from 0.4 to 15. No further reaction progress was observed on extended reaction time.
  • BINAP-Tol, DPPF, and DiPPF are efficient ligands for the reaction. Since a number of chelating phosphines have been used in enantioseleotive hydrogenation, 21 they provide further excellent candidates for evaluation in this reaction. The yields and relative degrees of substitution and reduction for various phosphines when used in Scheme B2 are shown below in Table B3. Once again, ferrocene-based bis-phosphine, ⁇ -binaphane, provided the best combination of reactivity and selectivity.
  • Palladium catalysts based on phosphine oxide ligands are a new type of efficient, versatile, air-stable and preformed homogeneous catalysts for the C-S bond formation. 8-10 Ten of the most common catalysts were evaluated in Scheme B2. The results are shown in Table B4 which indicates that low reactivities and selectivities were generally observed.
  • R 1 SnMe 3 6 (1:0) a 16 (1:0) b 0.4 14 (1:0)
  • R 1 SnMe 3 26 (1:0)
  • e CH 2 CONH 2
  • R 1 H 100 (1:0.06)
  • R 2 CH 2 CH 2 OH
  • R 1 H 26 (1:0.67)
  • h R 2 CH 2 CONH 2 a 0.1 equiv Pd 2 (dba) 3 and 0.1 equiv L.
  • Method C1 0.1 equiv Pd 2 (dba) 3 and 0.1 equiv DiPPF were used; in Method C2, 0.2 equiv Pd 2 (dba) 3 and 0.4 equiv DiPPF were used; in Method C3, 0.2 equiv Pd 2 (dba) 3 and 0.2 equiv DiPPF were used; in Method C4, 0.2 equiv Pd 2 (dba) 3 and 0.8 equiv DiPPF were used.
  • the results shown are the isolated yield of the substitution product from reverse phase HPLC.
  • the process method for the rapid, selective and efficient substitution of bromo or iodo groups in aryl halides by tin-thiolates with microwave heating is applicable to substrates with an easily reducible iodo group, in either the presence or absence of a bromo group.
  • Lower reactivity and higher selectivity were observed as compared with those using thiols as reagents.
  • the corresponding reactions under conventional heating did not generate any appreciable amount of products except for those without any heteroatoms in the substrates (no data shown).

Abstract

Novel derivatives of imidazopyridinylbenzeneamines and novel derivatives of benzothiazolylbenzeneamines are disclosed that offer improved behavior when used as imaging agents for positron emission tomography of beta-amyloids. Also disclosed is a palladium-catalyzed reaction scheme under microwave conditions for aryl thioethers in general that provides a high ratio of substitution relative to reduction and can be used for the imidazopyridinylbenzeneamine derivatives as well as other compounds of related structure.

Description

    CROSS REFERENCE TO RELATED APPLICATION
  • This Application is a Divisional of U.S. patent application Ser. No. 12/293,340, filed Sep. 22, 2010, which is a U.S. National Stage Application of International Application No. PCT/US2007/066939, filed Apr. 19, 2007, which claims priority from U.S. Provisional Patent Application No. 60/793,807, filed Apr. 21, 2006 all of which are incorporated by reference in their entirety.
  • BACKGROUND OF THE INVENTION
  • All publications cited herein, including patents and published articles, are hereby incorporated herein by reference.
  • Alzheimer's disease (AD) is the most common cause of dementia and is characterized by progressive impairment in cognitive function and behavior. This progressive, irreversible brain disorder affects millions of lives and imposes a devastating burden on the health care around the world. Over the past two decades, significant progress has been made in deciphering the pathogenesis and developing new therapeutic approaches. The pathological features of AD include neuritic plaques composed of amyloid-β peptide (Aβ) fibrils, neurofibrillary tangles of hyper-phosphorylated tau, and neurotransmitter Deficits. Recent efforts of managing AD have been focused on the prevention of production, aggregation, and deposition of amyloid-β peptides in the brain and the acceleration of clearance of Aβ from the brain.
  • Non-invasive detection and quantitation of amyloid deposits in the brain has been used to develop anti-amyloid therapies. Direct imaging of amyloid load in vivo inpatients with AD is useful for the early diagnosis of AD and the development and assessment of treatment strategies. To this end, compounds suitable for in vivo imaging of amyloid deposits in human brains have been developed. Among these compounds are monoclonal antibodies against Aβ and peptide fragments, but these have had limited uptake by the brain when tested in patients with AD, Putrescine-gadolinium-Aβ has been injected into transgenic mice overexpressing amyloid, and this has resulted in labeling observed with MRI. Amyloid deposition can also be non-invasively imaged and quantitated with a radiotracer that readily enters the brain and selectively binds to amyloid deposits.
  • The small molecule approach for amyloid imaging has so far been the most Successful. Some of the promising compounds that have been used to image amyloid are based on Congo red, thioflavin, stilbene, and FDDNP. The binding of different derivatives of Congo red and thioflavin has been studied in human autopsy brain tissue and in transgenic Mice. Two compounds in advanced testing, fluorine-18-labelled-FDDNP (2-(1-{6-[(2-[18F]fluoroethyl)(methyl)amino]-2-naphthyl}ethylidene)malononitrile) and carbon-11-labelled-PIB (6-hydroxy-2-(4′-methylaminophenyl)benzothiazole; Pittsburg compound B), both show more binding in the brains of patients with AD than in those of healthy people. Table 1 below, which is taken from Nordberg, A., “PET imaging of amyloid in Alzheimer's disease,” The Lancet Neurology 3(9), September 2004, pp, 519-527, provides a list of compounds presently undergoing evaluation as amyloid imaging agents.
  • TABLE 1
    Prior Art Compounds Used for Imaging of Amyloid in
    the Brains of Living Organisms (reference numerals
    refer to citations in the Nordberg paper)
    Imaging
    Reference Imaging compound technique Study
    Friedland et al.40 99Tc-10H3 SPECT Patients with AD
    Shoghi-Jadid et 18F-FDDNP PET Patients with AD
    al.41
    Klunk et al.39 11C-PIB PET Patients with AD
    Mathis et al.42 11C-BTA-1 PET Baboons
    Poduslo et al.43 MION-Aβ1-40 MRI Mice PS
    Poduslo et al.43 PUT-Gd-Aβ MRI Mice PS
    Wadghiri et al.44 Gd-DTPA-Aβ1-40 NMRI Mice APP/PS
    Mathis et al.45 BTA-1 Multiphoton Mice APP/PS
    Bacskai et al.46 Thioflavin-S Multiphoton Mice APP
    Bacskai et al.46 PIB Multiphoton Mice APP
  • Two tertiary amines, [18F]FEM-IMPY [N-(2-fluoroethyl)-4-(6-iodo-H-imidazo[1,2-A]pyridin-2-yl)-N-methylbenzeneamine], and its 3-fluoropropyl analog, [18F]FPM-IMPY, have been previously evaluated by inventors herein as β-amyloid radioligands and reported in Cai, L., et al, “Synthesis and evaluation of two 18F-labeled 6-iodo-2-(4′-N,N-dimethylamino)phenylimidazo[1,2-a]pyridine derivatives as prospective radioligands for beta-amyloid in Alzheimer's disease,” J. Med. Chem. 2004, 47, 2208-2218. After intravenous injection of either radioligand into rodent or monkey there is rapid and high uptake of radioactivity into brain with an SUV (standardized uptake value, % I.D.kg/g) of about 160 followed by biphasic clearance with a fast and a very slow component. These radioligands were rapidly metabolized by processes involving de-alkylation of the tertiary aromatic amino group, culminating in defluoridation and high uptake of radioactivity in bone. Tetra-deuteration of the fluoroethyl group did not lead to a significant reduction in the residual brain radioactivity, but reduced the bone uptake of radioactivity, presumably due to an isotope effect on metabolism. An object of the present invention is to provide radioligands that do not undergo rapid defluoridation and do not produce residual radioactivity in the brain. The invention therefore resides in further analogs of imidazo[1,2-a]pyridine (IMPY) useful as radioligands for the detection of Aβ amyloid aggregates in Alzheimer's Disease (AD) patients. Preferred among these analogs are imidazo[1,2-a]pyridine N-methylbenzeneamines in which a methyl group or a group similar in size to a methyl group (such as a bromine atom) is bonded to the ortho ring position nearest to the amine.
  • The effective management of Alzheimer's disease requires tools to diagnose, monitor, treat and prevent the disease. An object of the present invention is therefore to provide amyloid imaging agents with high specificity of binding to beta amyloid, low background noise, better entry into brains, and improved labeling efficiencies.
  • THE INVENTION
  • Two novel classes of amyloid imaging agents are disclosed herein. The first class are derivatives of imidazopyridinylbenzeneamine (IMPY), and are defined by Formula (I):
  • Figure US20140341812A1-20141120-C00001
  • In Formula (I):
      • R1 is C1-C6 alkyl, or aryl-, or halo-substituted C1-C6 alkyl methyl; preferably C1-C6 alkyl or fluoro-substituted C1-C6 alkyl; and most preferably methyl, ethyl, fluoromethyl, or fluoroethyl,
      • R2 is H, C1-C6 alkyl, halo-substituted C1-C6 alkyl, halo, or C1-C6 alkylthio, and R3 is H or C1-C6 aklyl, or R2 and R3 are joined to form a thio-C1-C6 allylene linkage (—S-alkyl-) linkage or alkylene linkage between the 2-position (to which R2 is attached in the formula) on the phenyl ring and the amine nitrogen (to which R3 is attached) with the S atom attached to the 2-position carbon; and preferably R2 is hydrogen, bromo, or methylthio, R3 is hydrogen or methyl, or R2 and R3 are joined to form a thiomethylene (—SCH2—) linkage between the 2-position (to which R2 is attached in the formula) on the phenyl ring and the amine nitrogen (to which R3 is attached) with the S atom attached to the 2-position carbon, and
      • R4 is H or C1-C3 alkyl, and R5 is H or C1-C3 alkyl, or R4 and R5 are joined to form a C1-C3 alkylenethio linkage (-alkyl—S—) linkage between the amine nitrogen (to which R4 is attached) and the 6-position (to which R3 is attached in the formula) on the phenyl ring with the S atom attached to the 2-position carbon; and preferably R4 is H and R5 is H, or R4 and R5 are joined to form a methylenethio (—CH2S—) linkage between the amine nitrogen (to which R4 is attached) and the 6-position (to which R5 is attached in the formula) on the phenyl ring with the S atom attached to the 2-position carbon.
  • TABLE 1
    Examples of compounds within the scope of Formula (I) are:
    Example R1 R2 R3 R4 R5
    1.1 CH3 H CH3 H H
    1.2 C2H5 H CH3 H H
    1.3 CH3 Br H H H
    1.4 FCH2 H CH3 H H
    1.5 FCH2 Br H H H
    1.6 FCH2CH2 H CH3 H H
    1.7 FCH2CH2 Br H H H
    1.8 FCH2 SCH3 H H H
    1.9 FCH2 SCH2 H H
    1.10 FCH2 SCH2 SCH3
  • The second class are derivatives of benzothiazolylbenzeneamine (BTA), and are defined by Formula (II):
  • Figure US20140341812A1-20141120-C00002
  • In Formula (II);
      • R11 is C1-C6 alkyl, halo-substituted C1-C6 alkyl, hydroxy-substituted C1-C6 alkyl, or carbamoyl C1-C6 alkyl; preferably C1-C3 alkyl, fluoro-substituted C1-C3alkyl, hydroxy-substituted C1-C3 alkyl, or carbamoyl C1-C3 alkyl; and most preferably methyl, ethyl, fluoromethyl, fluoroethyl, hydroxymethyl, hydroxyethyl, carbamoylmethyl, or carbamoylethyl;
      • R12 is H, C1-C6 alkyl, halo-substituted C1-C6 alkyl, halo, and C1-C6 alkylthio, and R13 is a member selected from the group consisting of H and C1-C6 alkyl, or R12 and R13 are joined to form a thio-C1-C3 alkylene linkage (—SCH2—) between the 2-position (to which R12 is attached in the formula) on the phenyl ring and the amine nitrogen (to which R13 is attached) with the S atom attached to the 2-position carbon; preferably H, C1-C3 alkyl, chloro, bromo, chloro-substituted C1-C3 alkyl, bromo-substituted C1-C3 alkyl, and C1-C3 alkylthio, and R13 is a member selected from the group consisting of H and C1-C3 alkyl, or R12 and R13 are joined to form a thiomethylene linkage (—SCH2—) between the 2-position (to which R12 is attached in the formula) on the phenyl ring and the amine nitrogen (to which R13 is attached) with the S atom attached to the 2-position carbon; and most preferably R12 is hydrogen, bromo, or methylthio, R13 is hydrogen or methyl, or R12 and R13 are joined to form a thiomethylene (—SCH2—) linkage between the 2-position (to which R12 is attached in the formula) on the phenyl ring and the amine nitrogen (to which R13 is attached) with the S atom attached to the 2-position carbon, and
      • R14 is H or C1-C6 alkyl, preferably H or methyl, and
      • X is CH or N,
  • TABLE 2
    Examples of compounds within the scope of Formula (II) are:
    Example R11 R12 R13 R14 X
    2.1 CH3 H CH3 H N
    2.2 H2N—C(O)—CH2 H CH3 H CH
    2.3 HOC2H4 Br H H CH
    2.4 FCH2 H CH3 H N
    2.5 FCH2 Br H H N
    2.6 FCH2CH2 H CH3 H N
    2.7 FCH2CH2 Br H H N
    2.8 FCH2 SCH3 H H N
    2.9 FCH2 SCH2 H N
    2.10 FCH2 SCH2 CH3 N
  • This invention resides in the above compounds as novel imaging agents for the imaging of amyloid deposits, new methods of synthesis of imidazopyridinylbenzeneamine (IMPY) derivatives, and methods for diagnosing Alzheimer's disease in vivo by positron emission tomography, magnetic resonance imaging and other imaging methods involving the use of the imaging agents of this invention,
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Synthesis of ketones and α-bromoketones as intermediates in the preparation of IMPY derivatives.
  • FIG. 2. Synthesis of 2-aminopyridine derivatives and 2-aminopyrazines. The synthetic methods of pyridine derivatives depend on the substituents on the ring.
  • FIG. 3. Aromatic substitutions with the methoxy anion. The method uses salts of copper (I) and a small amount of ester to form a stabilized tetrahedral adduct.
  • FIG. 4. Synthesis of 5-chloro-2-aminopyrazine using mild conditions,
  • FIG. 5. Synthsesis of IMPY Derivatives I. Most of the IMPY derivatives were synthesized through the direct condensation of α-bromoketones and 2-amino-pyridines.
  • FIG. 6. Synthesis of IMPY Derivatives II using direct methoxylation of halide in preformed IMPY derivatives.
  • FIG. 7. Synthesis of IMPY Derivatives II using catalytic substitution of halides by thiolate-based palladium catalysts,
  • EXAMPLES
  • Materials and Methods
  • Preparation of Compounds
  • Chemicals and Reagents, Common reagents used in the syntheses were purchased from Aldrich Chemical Company, Fluka Chemical Company (Milwaukee, Wis.), Acros (Hampton, N.H.), or Strem Chemicals (Newburyport, Mass.), and were used without further purification, unless otherwise indicated. DiPPF [1;1′-bis(diisopropylphosphino)ferrocene) and hexamethylditin were from Strem Chemicals. 2-Mercaptoethanol, 5-iodopyridin-2-amine, 5-bromopyridin-2-amine, pyrazin-2-amine, 2-bromoacetyl bromide, o-toluidine, 1-(4-aminophenyl)ethanone, N-methyl-N-(trimethylstannyl)methanamine, sodium methoxide, and Pd2dba3 (dipalladium dibenzylideneacetone), were from Aldrich, 2-mercaptoacetamide and 6-aminopyridine-3-carbonitrile (P4) were from Acros. Water was purified through a Millipore water purification system, comprising a combination of two filters, one Rio™, one reservoir, and one Milli-Q®, synthesis system (Bedford, Mass.). Common solvents were obtained from Fisher Scientific (Pittsburgh, Pa.), Brain tissue from deceased Alzheimer's Disease (AD) patients was obtained from Brain Bank of the Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health.
  • 1-(4-(methylamino)phenyl)ethanone (2),2 1-(4-amino-3-bromophenyl)ethanone (3), N,2-dimethylbenzenamine (K2), 2,2,2-trifluoro-N-o-totylacetamide (A3), N-o-tolylacetamide (A4), 6-aminopyridine-2-carbonitrile (P3), 6-amino-5-bromopyridine-3-carbonitrile (P11), 5-bromo-3-iodopyridin-2-amine (B3), were synthesized as reported in the literature.
  • Instrument and general conditions. Analytical HPLC was performed using a reverse phase column (X-Tera C18; 5 μm; 10.0×250 mm; Waters) eluted with c. ammonia (0.25%) in acetonitrile-water at 6.2 mL/min. The chromatography system was fitted with a continuous wavelength UV-vis detector (Beckman System Gold 168 Detector) and an autosampler (Beckman System Gold 508 Autosampler), For semi-preparative Beckman HPLC, a reverse phase column (X-Tera C18; 5 μm 19×250 mm; Waters) was eluted at 20.0 mL/min. The HPLC system was fitted with a manual injector (5 mL injection loop) and a third delivery pump using acetonitrile as eluant at 1 mL/min. The purity of compounds was determined with HPLC monitored for UV absorbance at 280 nm (for IMPY derivatives) or 254 nm (for other aromatic compounds) and expressed as area percentage of all peaks. The 1H and 13C NMR spectra of all compounds were acquired on a Jeol GSX 270 (270 MHz 1H and 68 MHz 13C), on a Bruker DRX 300 (300 MHz 1H and 75 MHz 13C), on a Bruker DRX 400 (400 MHz 1H and 100 MHz 13C) and on a Bruker AM500 (500 MHz 1H and 125 MHz 13C), using the chemical shifts of residual deuterated solvent as the internal standard; chemical shift (δ) data for the proton and carbon resonance were reported in parts per million (ppm) relative to the internal standard. Thin-layer chromatography (TLC) was performed using Silica Gel 60 F254 plates from EM Science and compounds visualized under UV light at either 250 or 360 nm. Flash chromatography was carried out using a Biotage Horizon™ HPFC™ system (Charlottesville, Va., column sizes: 12 mm×150 mm, 25 mm×150 mm, 40 Mm×150 mm) with hexanes and ethyl acetate (EtOAc) as eluents with chromatographic solvent proportions expressed on a volume: volume basis. IR spectra were recorded using a Perkin-Elmer Spectrum One FT-IR spectrometer, and UV/vis spectra were recorded using a Lambda 40 UV/vis spectrometer. Mass spectra were acquired using either Thermo Finnigan LCQDECA LC-MS (MS-HPLC column: Luna C18; 5 μm2.0×150 mm; Phenomenex, flow rate; 150 μL/min, eluent; MeOH and H2O mixture) or Thermo Finnigan PolarisQ GC-MS (GC column: capillary RTX-5 ms 30 m×0.25 mm, flow rate: 1 mL/min, carrier gas; He), or VG Micromass 7070E and AutoSpec-Q spectrometers. High-resolution mass spectra (HRMS) were acquired from Mass Spectrometry Laboratory, University of Illinois at Urbana-Champaign (Urbana, Ill.). Elemental analyses of selected compounds were carried out by Midwest Microlab (Indianapolis, Ind.) or Galbraith Laboratories, Inc, (Knoxville, Tenn.), or Mr Stephen Boyer, SACS, University of London. The melting points were measured using Electrothermal Mel-Temp Manual Melting Point Apparatus (Fisher Scientific), and uncorrected. A CEM Discover microwave system was used for microwave synthesis (Matthews, N.C.).
  • The following syntheses are illustrated by molecular formulas and reaction schemes in the Figures attached hereto, and the structures represented by the acronyms and other symbols used below for the various intermediates and compounds are shown in the Figures.
  • 1-(3-bromo-4-(methylamino)phenyl)ethanone (K3), The compound was synthesized by bromination of the aryl amine by NBS (N-Bromosuccinimide).
  • The following are syntheses of ketones and α-bromoketones according to FIG. 1 as intermediates in the preparation of the IMPY derivatives.
  • N-(4-acetyl-2-methylphenyl)-2,2,2-trifluoroacetamide (K4), 2,2,2-trifluoro-N-o-tolylacetamide (8.0 g; 39.4 mmol) was dissolved in 20 mL CS2. Bromoacetic bromide (8.4 g; 68.3 mmol) was added dropwise. AlCl3 (16.0 g; 120 mmol) was added in portions, with drying tube attached to the reaction flask. The reaction mixture was refluxed overnight. After reaction, the solvent was removed. Ice and water were added, and some yellow solid Precipitated. The solid was isolated by filtration, and purified by silica gel column chromatography, to afford 3.8 g (yield 39%) of product as off-white solid. 1H NMR (300 MHz; CDCl3), δ8.30 (s, 1H, Ar—H), 7.91 (brs, 1H, NH), 7.81 (d, 3JHH=8.2 Hz, 1H, Ar—H), 7.36 (d, 3JHH=7.9 Hz, 1H, Ar—H), 2.60 (s, 3H, COCH3), 2.36 (s, 3H, —CH3).
  • N-(4-acetyl-2-bromophenyl)-2,2,2-trifluoroacetamide (K5), 1-(4-aminophenyl)ethanone (2.50 g; 18.5 mmol) was dissolved in 100 mL toluene. NBS (3.30 g; 18.8 mmol) was added portionwise, After 15 min stirring at RT, the organic phase was washed with water twice, and dried over Na2SO4. The solvent was removed by rotavap to get oil, which was purified by silica gel chromatography, to afford 1.7 g of a white solid, 1-(4-amino-3-bromophenyl)ethanone (1.7 g; 7.94 mmol), The intermediate was dissolved in CH2Cl2. The reaction mixture was cooled to 0° C.. Trifluoroacetic anhydride (2.0 g; 9.52 mmol) and NEt3 (1.0 g; 9.90 mmol) were added sequentially. The reaction mixture was stirred at RT for another hour. The organic phase was washed twice with water, dried over MgSO4. The solvent was removed, to afford 2.4 g (yield 42%) of product as white solid. 1H NMR (400 MHz, CDCl3) δ8.61 (s, 1H, N-H), 8.46, (d, 3JHH=8.6 Hz, 1H, Ar—H), 8.21 (d, 4JHH2.0 Hz, 1H, Ar—H), 7.95 (dd, 3JHH=8.6 Hz, 4JHH=1.9 Hz, 1H, Ar—H), 2.58 (s, 3H, CH3).
  • N-(4-acetyl-2-bromophenyl)-2,2,2-trifluoro-N-methylacetamide (K6). 1-(3-bromo-4-(methylamino)phenyl)ethanone (1.4 g; 6.14 mmol) was dissolved in 15 mL CH2Cl2. The reaction mixture was cooled to 0° C. Trifluoroacetic anhydride and Et3N were added Sequentially. The mixture mixture was stirred at RT for 3 hrs, washed with water three times, and dried over Na2SO4. The solvent was removed, and product was dried to afford 1.9 g (yield 96%) of a yellow solid. 1H NMR (300 MHz; CDCl3), δ8.13 (s, 1H, Ar—H), 7.83 (d, 3JHH=8.8 Hz, 1H, Ar—H), 7.02 (d, 3JHH=8.6 Hz, 1H, Ar—H), 2.91 (s, 3H, NCH3), 2.53 (s, 3H, COCH3).
  • N-(4-(2-bromoacetyl)-2-methylphenyl)-2,2,2-trifluoroacetamide (B2). 2,2,2-trifluoro-N-o-tolylacetamide (5.0 g; 24.6 mmol) was dissolved in 20 mL CS2. The mixture was cooled below 10° C. Bromoacetic bromide (10.1 g; 50.0 mmol) was added dropwise. AlCl3 (10.0 g; 75.0 mmol) was added in portions, with drying tube attached to the reaction Flask. The reaction mixture was refluxed overnight. After the reaction is complete as monitored by TLC, the upper CS2 layer was removed. The lower layer was added into 7.0 mL ice water mixed with HCl. The product was extracted into CH2Cl2, and dried over MgSO4. The solvent was removed, and the solid was dried to afford 5.0 g of crude product, which was purified by silica gel column chromatography, to afford 3.0 g (yield 38%) of product as yellow solid. 1H NMR (300 MHz; CDCl3), δ8.40 (s, 1H, Ar—H), 7.87 (brs+d, 3JHH=8.0 Hz, 2H, Ar—H), 7.43 (d, 3JHH=8.0 Hz, 1H, Ar—H), 4.46 (s, 2H, COCH2Br), 2.41 (s, 3H, CH3).
  • N-(4-(2-bromoacetyl)-2-methylphenyl)-N-methylacetamide (B3), 1H NMR (300 MHz; CDCl3), δ7.89 (dd, 3JHH=8.0 Hz, 4JHH=1.7 Hz, 1H, Ar—H), 7.78 (d, 4JHH=1.7 Hz, 1H, Ar—H), 7.45 (d, 3JHH=8.0 Hz, 1H, Ar—H), 4.41 (AB, 2JHH=11.7 Hz, 2H, COCH2Br), 3.20 (s, 3H, NCH3), 2.33 (s, 3H, Ar-CH3), 1.78 (s, 3H, COCH3).
  • N-(2-bromo-4-(2-bromoacetyl)phenyl)-2,2,2-trlfluoroacetamide (B4), N-(4-acetyl-2-bromophenyl)-2,2,2-trifluoroacetamide (12.1 g; 39.0 mmol) and tetrabutylammonium tribromide (18.8 g; 39.0 mmol) were dissolved separately in ethanol. After combination, and refluxing for 4 hrs, there was precipitate formation. The solvent was Removed. NaHCO3 solution was added to neutralize any residue acid. The solid was filtered, and washed with CH2Cl2, to afford 7.0 g (yield 46%) of product as pale green solid. 1H NMR (400 MHz, CDCl3) δ8.64 (s, 1H, N-H), 8.51 (d, 3JHH=8.6 Hz, 1H, Ar—H), 8.25 (d, 4JHH=2.0 Hz, 1H, Ar—H), 7.98 (dd, 3JHH=8.6 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 4.38 (s, 2H, —CH2—Br).
  • N-(2-bromo-4-(2-bromoacetyl)phenyl)-2,2,2-trifluoro-N-methylacetamide(B5). CuBr2 (6.0 g; 26.9 mmol) was suspended in 20 mL ethyl acetate. The mixture was heated to reflux. N-(4-acetyl-2-bromophenyl)-2,2,2-trifluoro-N-methylacetamide (4.5 g; 13.9 mmol) dissolved in 20 mL CHCl3 was added. The mixture was refluxed for 4 hrs, and filtered. The solvent was removed from the filtrate to get an orange solid, which was purified by silica gel column chromatography using the mixture of ethyl acetate and petroleum ether from 1:100 to 1:50, to afford 1.9 g (yield 34%) of product as yellow solid. 1H NMR (400 MHz, CDCl3) δ Isomer α (80%) 8.33 (d, 4JHH=2.0 Hz, 1H, Ar—H), 8.02 (dd, 3JHH=8.2 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 7.48 (dd, 3JHH=8.2 Hz, 5JHH=0.5 Hz, 1H, Ar—H), 4.46 (AB, 2JHH=11.8 Hz, 1H, CH2), 4.42 (AB, 2JHH=11.8 Hz, 1H, CH2), 3.36 (d, 4JHF=0.4 Hz, 3H, CH3). Isomer β (20%) 8.33 (d, 4JHH=2.0 Hz, 1H, Ar—H), 8.05 (dd, 3JHH=8.2 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 7.44 (dd, 3JHH=8.2 Hz, 5JHH=0.5 Hz, 1H, Ar—H), 4.46 (AB, 2JHH=11.8 Hz, 1H, CH2), 4.42 (AB, 2JHH=11.8 Hz, 1H, CH2), 3.49 (q, 4JHF=1.6 Hz,3H, CH3).
  • The following are syntheses of 2-aminopyridine derivatives according to FIGS. 2, 3, and 4 as further intermediates in the preparation of the IMPY derivatives and the IMPY derivatives themselves.
  • 5-methoxypyridin-2-amine (P5). Method a: 2-Amino-4-bromopyridine (0.10 g; 0.58 mmol), sodium methoxide (0.16 g; 2.9 mmol) and copper powder nanosized activated (0.11 g; 1.74 mmol) were introduced in a screw cap vial (Pyrex glass) together with 2.0 mL of anhydrous MeOH and a stirrer bar. The vial was closed and put in an oil bath at 135° C. and stirred for 14 h. The mixture was cooled, diluted with MeOH (5.0 mL) and filtered through an SPE silica gel cartridge and the product eluted with AcOEt. The fractions were collected and evaporated obtaining a crude of 92 mg of product which was further purified by FCC (DCM/AcOEt=1:1) to give 26 mg (yield 36%) of the title compound as brown oil. Method b: 2-Amino-4-bromopyridine (0.10 g; 0.58 mmol), sodium methoxide (0.16 g; 2.9 mmol) and copper powder nanosized activated (0.11 g; 1.74 mmol) were introduced in a microwave glass tube with 1.5 mL of anhydrous DMF and sealed. The tube was introduced in the microwave cavity and heated for 30 min at 140° C. (140C30M75W300Psi). Although DMF is a high boiling solvent, high pressure was observed, probably caused by the partial methanolysis of the DMF resulting in low boiling products such as methyl formate and dimethylamine. The mixture was diluted with 10 mL of 2 M NH4Cl solution and extracted 3 times with AcOEt. The organic phase was washed 2 times with 2 M NH4Cl solution and 1 time with water to remove the remaining DMF, dried on NaSO4, and filtered. After the solvent was removed, the crude product was purified by FCC (AcOEt) to afford 12 mg (yield 17%) of the title compound as brown oil. 1H NMR (270 MHz; CDCl3), δ7.74 (1 H, d, 3JHH=3.0 Hz), 7.06 (1H, dd, 3JHH=9.0 Hz, 4JHH=3.0 Hz), 6.45 (1 H, d, 3JHH=9.0 Hz), 3.95 (2H, bs, NH2), 3.74 (3H, s, OCH3); m/z (EI/MS): 124 (M+), 109 ([M-CH3]+).
  • 5-(ethylthio)pyridin-2-amine (P9). 2-Amino-5-iodo-pyridine (2.20 g; 10 mmol), sodium ethanethiolate 80% (1.7 g; 16 mmol) and copper powder (190 mg; 3.00 mmol) were loaded into a 100 mL round bottom flask under nitrogen. Ethylene glycol (40 mL; 0.25 mol) was added and the solution stirred at 150° C. for 26 h. The cooled solution was filtered and partitioned between ethyl acetate and water twice. The organic layer was dried over barium oxide, filtered and the solvent removed in vacuo to afford 1.2 g (yield 76%) of product as yellow oil. 1H-NMR (300 MHz; CDCl3), δ8.06 (1H, d, 4JHH=2.1 Hz, Ar—H), 7.48 (1H, dd, 3JHH=8.7 Hz, 4JHH=2.4 Hz, Ar—H), 6.42 (1H, dd, 3JHH=8.7 Hz, 5JHH=0.6 Hz, Ar—H), 2.68 (2H, q, 3JHH=7.3 Hz, CH2), 1.15 (3H, t, 3JHH=7.2 Hz, CH3).
  • 6-amino-3-bromopyridine-2-carbonitrile (P10). 1H NMR (300 MHz; DMSO-d6), δ7.75 (d, 3JHH=9.1 Hz, 1H, Ar—H), 6.69 (d, 3JHH=9.1 Hz, 1H, Ar—H).
  • 5-chloropyrazin-2-amine (P13). Method a: In a double necked dry flask equipped with condenser and dropping funnel, under nitrogen flow, were added 2-amino-pyrazine (0.50 g, 5.25 mmol) and 13 mL of anhydrous CHCl3 previously passed on basic Al2O3. This solution was heated at 60° C. under stirring. A solution of NCS (0.35 g, 2.12 mmol) in 7 mL of the anhydrous CHCl3 was added to the mixture through dropping funnel during 1.5 hr. After another 30 minutes, the reaction was stopped and the solvent evaporated. The residue was dissolved in methanol and absorbed on silica (2 g); This crude product was purified by FCC (Hexane/DCM/AcOEt=1:1:1) to afford 73 mg (yield 26%) of product as yellow solid. Method b: 2-Amino-pyrazine (0.10 g, 1.05 mmol) and NCS (80 mg, 0.60 mmol) were added in a microwave tube with 1.5 mL of the anhydrous CHCl3 and sealed. The tube was placed in the microwave cavity and heated at 70° C. for 10 min. (70C10M60W300 Psi). After the work-up, the crude (0.15 g) was purified by FCC (Hexane/DCM/AcOEt=1:1:1) to afford 35 mg (yield 45%) of the product as yellow solid. 1H NMR (270 MHz; d6-DMSO), δ7.99 (1H, s, H-6), 7.67 (1H, s, H-3), 6.65 (2H, bs, NH2), m/z (EI-MS): 129 (M+), 99, 94 ([M-Cl]+).
  • 4-(6-chloro-H-imidazo[1,2-a]pyridin-2-yl)-N)N-dimethylbenzenamine (L5). 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (1.21 g; 5.00 mmol) and 5-chloro-2-aminopyridine (7c) (0.668 g; 5.20 mmol) were used to give 12c (0.65 g; yield 48%); Mp: 234-236° C.; 1H NMR (400 MHz, CDCl3)δ8.10 (s, 1H, Ar—H), 7.76 (d, 3JHH=8.9 Hz, 2H, Ar—H), 7.65 (s, 2H, Ar—H), 7.14 (d, 3JHH=9.2 Hz, 1H, Ar—H), 6.71 (d, 3JHH=8.6 Hz, 2H, Ar—H), 2.95 (s, 6H, CH3). 13C13 NMR (400 MHz, CDCl3) δ149.3, 146.2, 142.6, 125.8 (s, 2C), 124.2, 121.8, 120.0, 118.8, 116.0, 111.2 (s, 2C), 105.7, 39.2 (s, 2C, NCH3), m/z (ES-MS): 275.1 (4%), 274.1 (51%), 273.1 (18%), 272.1 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C15H15N3Cl=272.0955. Found: 272.0960. Error (ppm): +1.8.
  • 4-(6-fluoro-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (L6). 2-Bromo-1-(4-(dimethylamino)-phenylethanone (11) (1.21 g; 5.00 mmol) and 5-fluoro-2-aminopyridine (7d) (0.583 g; 5.17 mmol) were stirred in reflux ethanol (50 mL) for 4 h. Pale yellow precipitate formed. NaHCO3 (0.41 g; 4.9 mmol) was added to the reaction mixture after cooling (15 min.). The mixture was refluxed again for another 2 h. After cooling, the solvents were removed. The solid was washed with water, CH2Cl2, and recrystallized from ethyl acetate, to get 0.42 g of product. Yield 33%. Mp: 228-229° C.; 1H NMR (400 MHz, CD3OD) δ8.35 (m, 3JFH=4.2 Hz, 4JHH=2.4 Hz, 5JHH=0.7 Hz, 1H, Ar—H), 7.93 (d, 4JHH=0.3 Hz, 1H, Ar—H), 7.66-7.62 (m, 2H, Ar—H), 7.41 (m, 3JHH=9.8 Hz, 4JFH=5.0 Hz, 5JHH=0.6 Hz, 1H, Ar—H), 7.14 (m, 3JHH=9.8 Hz, 3JFH=8.3 Hz, 4JHH=2.4 Hz, 1H, Ar—H), 6.74-6.70 (m, 2H, Ar—H), 2.89 (s, 6H, CH3). 13C NMR (400 MHz, CDCl3) δ153.2 (d, 1JFC=236.4 Hz, 1C, Ar), 150.0 (s, 1C, Ar), 147.5 (s, 1C, Ar), 143.1 (s, 1C, Ar), 127.0 (s, 2C, Ar), 121.2 (s, 1C, Ar), 117.2 (d, 3JFC=9.3 Hz, 1C, Ar), 116.0 (d, 2JFC=25.4 Hz,1C, Ar), 112.4 (s, 2C, Ar), 112.0 (d, 2JFC=40.6 Hz, 1C, Ar), 107.9 (d, 4JFC=1.8 Hz, 1C, Ar), 40.5 (s, 2C, CH3). m/z (ES-MS): 257.1 (7%), 256.1 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C15H15N3F=256.1250. Found: 256.1261. Error (ppm): +4.3.
  • N,N-dimethyl-4-(6-nitro-H-imidazo[1,2-a]pyridin-2-yl)benzenamine (L7). 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (0.40 g; 1.65 mmol) and 5-nitro-pyridin-2-amine (7f) (0.30 g; 2.15 mmol) were stirred in reflux anhydrous acetonitrile (25 mL) at 90-95° C. ander dinitrogen for 2 h, NaHCO3 (0.25 g; 2.97 mmol) was added to the reaction mixture after cooling (15 min). The mixture was refluxed for another 9 h. After cooling, the mixture was filtered through a Busch funnel. The precipitate was washed with acetonitrile and water, to afford 0.11 g of the red product. Yield 23%. Mp: 275-277° C.; 1H NMR (400 MHz, DMSO-d6) δ9.77 (dd, 4JHH=2.3 Hz, 5JHH=0.8 Hz, 1H, Ar—H), 8.42 (s, 1H, Ar—H), 7.91 (dd, 3JHH=9.8 Hz, 4JHH=2.3 Hz, 1H, Ar—H), 7.83-7.80 (m, 2H, Ar—H), 7.68-7.65 (m, 1H, Ar—H), 6.83-6.80 (m, 2H, Ar—H), 2.97 (s, 6H, CH3). 13C13 NMR (400 MHz, DMSO-d6) δ150.6, 148.5, 144.9, 136.0, 127.3, 126.9 (s, 2C, Ar), 120.2, 118.4, 115.2, 112.2 (s, 2C, Ar), 109.3, 40.2 (s, 2C, CH3), m/z (ES-MS): 284.1 (29%), 283.1 (100%, [M+H]+), HRMS m/z (TOF+): Calc. C15H15N4O2=283.1195. Found 283.1206. Error (ppm); +3.9.
  • 2-(4-(dimethylamino)phenyl)-H-imidazo[1,2-a]pyridine-6-carbonitrile (L8). The same method as described above was used. 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (1.21 g; 5.00 mmol) and 6-aminopyridine-3-carbonitrile (7e) (0.63 g; 5.3 mmol) were used to give 12e (0.85 g; yield 65%); Mp: 269-271° C.; 1H NMR (400 MHz, CDCl3) δ8.42 (dd, 4JHH=1.6 Hz, 5JHH0.6 Hz, 1H, Ar—H), 7.76-7.73 (m, 2H, Ar—H), 7.72 (s, 1H, Ar—H), 7.58-7.55 (m, 1H, Ar—H), 7.15 (dd, 3JHH=9.3 Hz, 4JHH=1.7 Hz, 1H, Ar—H), 6.72-6.69 (m, 2H, Ar—H), 2.95 (s, 6H, CH3). 13C13 NMR (400 MHz, CDCl3) δ149.9, 148.1, 143.7, 129.9, 126.3 (s, 2C, Ar), 123.0, 119.2, 116.6, 115.8, 111.2 (s, 2C, Ar), 106.2, 96.9, 39.3 (s, 2C, CH3). m/z (ES-MS): 364.0 (8%), 264.1 (29%), 263.1 (100%, [M+H]+). HRMS m/z (TOF+); Calc. C16H15N4=263.1297. Found: 263.1303.Error (ppm): +2.3.
  • 4-(6-methoxy-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dlmethylbenzenamine (L9). In a 2-necked round bottomed flask equipped with a condenser and under nitrogen flow were introduced the 2-amino-5-methoxypyridine (0.15 g, 1.17 mmol), the 2-bromo-4′-dimethylamino-acetophenone (0.31 g, 1.29 mmol) and 14 mL of absolute EtOH. The reaction mixture was stirred at reflux for 2 h. After it cooled down, NaHCO3 (0.15 g, 1.75 mmol) was added, the mixture was refluxed for another 6 hrs. After the solvent was removed, the residue was dissolved in AcOEt. The organic phase was washed with water, dried over MgSO4, and filtered. The solvent was removed to afford 0.271 g crude product, which was purified by FCC (DCM/AcOEt=1:1) to give 0.129 g (yield 41%) of the product as yellow solid, M.p.=177-179° C. 1H NMR (270 MHz; CDCl3), δ7.82 (d, 3JHH=8.7 Hz, 2H, Ar—H), 7.71 (s, 1H, H-3), 7.65 (s, 1H, Ar—H), 7.53 (d, 3JHH=9.6 Hz, 1H, Ar—H), 6.96 (dd, 3JHH=9.6, 4JHH=1.8, 1H, Ar—H), 6.81 (d, 3JHH=8.8, 2H, Ar—H), 3.84 (s, 3H, OCH3), 3.02 (s, 6H, NCH3); 13C{1H} NMR (125 MHz, CDCl3), δ151.0, 150.4, 149.2, 146.0, 143.0, 126.8 (Ph-2,6), 119.2 (C7), 117.0 (C8), 112.5 (Ph-3,5), 107.6 (C3), 106.2 (C5), 56.2 (OCH3), 40.5 (NCH3), m/z(EI-MS): 267 (M+), 252 (M+-CH3); Calc. C16H17N3O; C, 71.89; H, 6.41; N, 15.72. Found: C, 71.60; H, 6.62; N, 15.96.
  • 2-(4-(dimethylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ol (L12). In a dried 10 mL flask under nitrogen flow, was added the 6-methoxy-imidazopyridine (62 mg, 0.22 mmol) and 2 mL of anhydrous DCM. The stirring solution was cooled at −78° C. with a CO2/acetone bath and BBr3 (80 mg, 0.32 mmol) was added dropwise by a disposable syringe. (Cautionl BBr3 reacts violently with water). The reaction mixture was allowed to stir at RT for 2 hrs, and poured into 15 mL ice/water. The mixture of CH2Cl2 and CH3OH (15:1) was added and the mixture stirred for 10 min. The organic phase was separated, dried over MgSO4, and filtered. The solvent was removed to give 39 mg (yield 70%) of the product as yellow solid. M.p.=262-264° C., 1H NMR (270 MHz; DMSO-d6), δ9.70 (s, 1H, OH), 8.16 (s, 1H, H-3), 8.01 (s, 1H, H-5), 7.71 (d, 3JHH=8.9 Hz, 2H, Ar—H), 7.45 (d, 3JHH=9.6 Hz, 1H, Ar—H); 7.03 (d, 3JHH=9.4, 1H, Ar—H), 6.78 (d, 3JHH=8.9 Hz, 2H, Ar—H), 2.95 (s, 6H, NCH3); 13C{1H} NMR (125 MHz, DMSO-d6), δ150.2, 146.2, 143.2, 140.3, 126.4 (Ph-2,6), 120.7 (C7), 120.4, 115.3 (C8), 112.4 (Ph-3,5), 110.5 (C3), 107.8 (C5), 39.9 (NCH3). m/z (CI-MS): 254 ([M+1]+), 238 ([M-CH3]+).
  • 4-(6-(ethylthio)-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (T7). 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (1.21 g; 5.00 mmol) and 5-(ethylthio)pyridin-2-amine (7h) (0.80 g; 5.2 mmol) were used to give 12h (0.91 g; yield 61%); mp: 168-171° C.; 1H NMR (400 MHz, DMSO-d6) δ8.56 (dd, 4JHH=1.9 Hz, 5JHH=0.9 Hz, 1H, Ar—H), 8.14 (s, 1H, Ar—H), 7.78-7.74 (m, 2H, Ar—H), 7.51-7.49 (m, 1H, Ar—H), 723 (dd, 3JHH=9.3 Hz, 4JHH=1.9 Hz, 1H, Ar—H), 6.76 (m, 2H, Ar—H), 2.94 (s, 6H, CH3), 2.92 (q, 3JHH=7.3 Hz, 2H, CH2), 1.21 (t, 3JHH=7.3 Hz, 3H, CH3). 13C NMR (100 MHz, DMSO-d6) δ150.1, 145.7, 143.6, 128.0, 127.3, 126.5 (s, 2C, Ar), 121.5, 118.3, 116.1, 112.2 (s, 2C, Ar), 107.1, 40.0 (s, 2C, CH3), 28.4 (s, 1C, CH2), 14.4 (s, 1C, CH3). m/z (ES-MS): 300.1 (6%), 299.1 (39%), 298.1 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C17H20N3S=298.1378. Found: 298.1386. Error (ppm): +2.7.
  • 4-(6-bromo-8-iodoindolizin-2-yl)-N,N-dimethylbenzenamine (L13). 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (1.21 g; 5.00 mmol) and 2-amino-3-iodo-5-bromopyridine (7i) (1.58 g; 5.3 mmol) were used to give 12i (1.21 g; yield 55 %); mp: 218-220° C.; 1H NMR (400 MHz, CDCl3) δ8.17 (d, 4JHH=1.7 Hz, 1H, Ar—H), 7.85-7.81 (m, 2H, Ar—H), 7.79 (s, 1H, Ar—H), 7.66 (d, 4JHH=1.7 Hz, 1H, Ar—H), 6.79 (d, 3JHH=7.9 Hz, 2H, Ar—H), 2.99 (s, 3H, CH3), 13C NMR (100 MHz, CDCl3) δ150.5, 147.9, 143.9, 135.8, 127.5 (s, 2C, Ar), 125.4, 121.5, 112.8 (s, 2C, Ar), 108.8, 106.0, 84.0 (s, 1C, Ar-I), 40.9 (s, 1C, CH3). m/z (ES-MS): 444.9 (6%), 443.9 (99%), 442.9 (9%), 441.9 (100%, [M+H]+), HRMS m/z (TOF+): Calc. C15H14N3BrI=441.9416. Found: 441.9398. Error (ppm): −4.1.
  • 6-bromo-2-(4-(dimethylamino)phenyl)indolizine-8-carbonitrile (L14). 2-Bromo-1-(4-(dimethylamino)phenylethanone (11) (1.21 g; 5.00 mmol) and2-amino-5-bromonicotinonitrile (7j) (1.03 g; 5.2 mmol) were used to give 12j (0.77 g; yield 45%); mp: 240-246° C.; 1H NMR (400 MHz, DMSO-d6) δ9.12 (d, 4JHH=1.8 Hz, 1H, Ar—H), 8.32 (s, 1H, Ar—H), 8.13 (d, 4JHH=1.8 Hz, 1H, Ar—H), 7.81 (d, 3JHH=8.9 Hz, 2H, Ar—H), 6.79 (d, 3JHH=8.9 Hz, 2H, Ar—H), 2.96 (s, 3H, CH3), 13C NMR (100 MHz, DMSO-d6) δ150.6, 147.4, 141.3, 133.6, 131.1, 127.0 (s, 2C, Ar), 119.9, 114.8, 112.1 (s, 2C, Ar), 109.0, 103.5, 99.9, 39.9 (s, 2C, NCH3). m/z (ES-MS): 443.9 (4%), 441.9 (3%), 344.0 (11%), 343.0 (98%), 342.0 (11%), 341.0 (100%, [M+H]+), HRMS m/z (TOF+): Calc. C16H14N4Br=341.0402.Found: 341.0389. Error (ppm): −3.8.
  • N-(2-bromo-4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)phenyl)-2,2,2-trifluoro-N-methylacetamide (L15). N-(2-Bromo-4-(2′-bromoacetyl)phenyl)-2,2,2-trifluoro-N-methylacetamide (4a) (4.0 g; 9.93 mmol) and 5-bromopyridin-2-amine (7b) (1.7 g; 9.83 mmol) were dissolved in MeOH (20 mL). The reaction mixture was refluxed for 8 h and solvent then removed. A small amount of CH2Cl2 was added and the resultant precipitate filtered off to afford 13a (2.8 g; yield 59%) as a yellow solid; mp: 95-98° C.; 1H NMR (400 MHz, CD3OD) δ Isomer α (84%) 8.73-8.72 (m, 1H, Ar—H), 8.34-8.31 (m, 2H, Ar—H), 8.01 (dd, 3JHH=8.2 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 7.58 (d, 3JHH=8.3 Hz, 1H, Ar—H), 7.55-7.52 (m, 1H, Ar—H), 7.45 (dd, 3JHH=9.6 Hz, 4JHH=1.9 Hz, 1H, Ar—H), 2.66 (s, 3H, CH3). Isomer β (16%) 8.73-8.72 (m, 1H, Ar—H), 8.34-8.31 (m, 2H, Ar—H), 8.03 (dd, 3JHH=8.0Hz, 4JHH=1.9 Hz, 1H, Ar—H), 7.55-7.52 (m, 1H, Ar—H), 7.51 (d, 3JHH=8.3 Hz, 1H, Ar—H), 7.44 (dd, 3JHH=9.6 Hz, 4JHH=1.9 Hz, 1H, Ar—H), 2.66 (s, 3H, CH3). 13C NMR (100 MHz, DMSO-d6) δ Isomer α 155.5 (q, 2JCF=35.1 Hz, 1C, CO), 143.5, 142.3, 137.9, 136.3, 130.8, 129.8, 128.6, 125.8, 122.8, 117.9, 115.9 (q, 1JCF=286 Hz, 1C, CF3), 111.1, 106.5, 37.7 (s, 1C, CH3). Isomer β 155.5 (q, 2JCF=35.1 Hz, 1C, CO), 143.4, 142.6, 139.8, 135.6, 129.9, 129.4, 128.4, 127.1, 126.3, 121.3, 117.8, 115.9 (q, 1JCF=286 Hz, 1C, CF3), 110.8, 109.4, 37.7 (s, 1C, CH3). m/z (ES-MS): 479.9 (27%), 477.9 (100%, [M+H]+), 475.9 (28%), HRMS m/z (TOF+): Calc. C16H11N3OF3Br2=475.9221. Found: 475.9228. Error (ppm): +1.5.
  • N-(4-(6-bromoindolizin-2-yl)-2-methylphenyl)-N-methylacetamide (L16). N-(4-(2′-Bromoacetyl)-2-methylphenyl)-N-methylacetamide (4c) (1.0 g; 3.52 mmol) and 5-bromopyridin-2-amine (7b) (0.73 g; 4.22 mmol) were used. Yield 37%, M.p. 197-198° C.; 1H NMR (400 MHz, DMSO-d6) δ Isomer α (90%) 8.88 (dd, 4JHH=2.0 Hz, 5JHH=0.8 Hz, 1H, Ar—H), 8.39 (d, 5JHH=0.4 Hz, 1H, Ar—H), 7.89 (dd, 3JHH=7.8 Hz, 4JHH=1.8 Hz, 1H. Ar—H), 7.84 (d, 4JHH=1.8 Hz, 1H. Ar—H), 7.58-7.54 (m, 1H, Ar—H), 7.43 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.37 (dd, 3JHH=9.5 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 3.11 (s, 3H, NCH3), 2.21 (s, 3H, Ar-CH3), 1.70 (s, 3H, CH3). Isomer β (10%) 8.87 (dd, 4JHH=1.9 Hz, 5JHH=0.8 Hz, 1H, Ar—H), 8.32 (d, 5JHH=0.3 Hz, 1H, Ar—H), 7.76 (dd, 3JHH=7.8 Hz, 4JHH=1.8 Hz, 1H. Ar—H), 7.73 (d, 4JHH=1.8 Hz, 1H. Ar—H), 7.56-7.53 (m, 1H, Ar—H), 7.36 (dd, 3JHH=9.5 Hz, 4JHH=2.0 Hz, 1H, Ar—H), 7.31 (d, 3JHH=8.0 Hz, 1H, Ar—H), 3.28 (s, 3H, NCH3), 2.13 (s, 3H, Ar-CH3), 1.70 (s, 3H, CH3), 13C NMR (100 MHz, DMSO-d6) δ Isomer α 169.0 (s, 1C, CO), 144.2, 143.3, 143.2, 134.7, 133.0, 131.7, 127.9, 126.9, 125.3, 125.1, 117.6, 109.8, 106.0, 35.3 (s, 1C, NCH3), 21.6 (s, 1C, CH3), 16.7 (s, 1C, Ar-CH3). Isomer β 169.6 (s, 1C, CO), 144.6, 143.8, 143.2, 134.8, 132.2, 130.9, 127.8, 126.8, 124.5, 124.1, 117.5, 109.3, 105.9, 38.7 (s, 1C, NCH3), 21.9 (s, 1C, CH3), 17.1 (s, 1C, Ar-CH3), m/z (ES-MS); 361.0 (4%), 360.0 (100%, [M+H]+), 359.0 (47%), 358.0 (96%). HRMS m/z (TOF+); Calc. C17H17N3OBr=358.0555. Found: 358.0557. Error (ppm): +0.6.
  • N-(4-(6-bromoindolizin2-yl)-2-methylphenyl)-2,2,2-trifluoroacetamide (L17). N-(4-(2-bromoacetyl)-2-methylphenyl)-2,2,2-trifluoroacetamide (3.0 g; 9.26 mmol) and 5-bromopyridin-2-amine (1.5 g; 8.67 mmol) were dissolved in 20 mL ethanol. The mixture was refluxed overnight, and the solvent was removed. CH2Cl2 was added to precipitate a solid, which was filtered and washed with CH2Cl2, to afford 1.0 g (yield 28%) of product as a yellow powder. 1H NMR (300 MHz; DMSO-d6), δ11.1 (s, 1H, NH), 9.14 (s, 1H, Ar—H), 8.60 (s, 1H, Ar—H), 7.90-7.76 (m, 4H, Ar—H), 7.46 (d, 3JHH=8.0 Hz, 1H, Ar—H), 2.19 (s, 3H, Ar-CH3).
  • 2-bromo-4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)-N-methylbenzenamine (L18). N-(2-bromo-4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)phenyl)-2,2,2-trifluoro-N-methylacetamide (1.0 g; 2.10 mmol) and K2CO3 (2.0 g; 14.5 mmol) were suspended in 30 mL ethanol and 15 mL water. The mixture was refluxed over 8 hrs. The organic solvent was removed, and CH2Cl2 was added. The organic phase was washed with H2O twice, and dried over MgSO4. The solvent was removed to afford 0.60 g (yield 80%) of product as a yellow solid. 1H NMR (300 MHz; DMSO-d6), δ8.80 (s, 1H, Ar—H), 8.20 (s, 1H, Ar—H), 7.98 (d, 4JHH=1.8 Hz, 1H, Ar—H), 7.76 (dd, 3JHH=8.5 Hz, 4JHH=1.8 Hz, 1H, Ar—H), 7.49 (d, 3JHH=9.5 Hz, 1H, Ar—H), 7.30 (dd, 3JHH=9.4 Hz, 4JHH=1.7 Hz, 1H, Ar—H), 6.67 (d, 3JHH=8.5 Hz, 1H, Ar—H), 2.78 (s, 3H, CH3).
  • 4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)-N,2-dimethylbenzenamine (L19). N-(4-(6-Bromoimidazo[2-a]pyridin-2-yl)-2-methylphenyl)-N-methylacetamide (13c, 1.0 g; 2.79 mmol) and KOH (1.0 g; 17.8 mmol) were used, to afford 0.68 g. Yield 77%, M.p. 165-166° C.; 1H NMR (400 MHz, DMSO-6) δ8.82 (dd, 4JHH=1.8 Hz, 5JHH=0.7 Hz, 1H, Ar—H), 8.27 (s, 1H, Ar—H), 7.56 (d, 3JHH=9.5 Hz, 1H, Ar—H), 7.33 (dd, 3JHH=9.5 Hz, 4JHH=1.9 Hz, 1H, Ar—H), 7.10-7.09 (m, 2H, Ar—H), 7.01 (dd, 3JHH=8.3 Hz, 5JHH=0.32 Hz, 1H, Ar—H), 5.12 (s, 1H, N—H), 2.82 (s, 1H, CH3), 2.10 (s, 1H, CH3), 13C NMR (100 MHz, DMSO-d6) δ147.8, 146.4, 143.1, 131.9, 129.8, 127.3, 126.6, 121.8, 117.5, 113.2, 108.9, 105.7, 105.6, 30.2 (s, 1C, N—CH3), 17.5 (s, 1C, Ar-CH3), m/z (ES-MS): 319.0 (17%), 318.0 (96%), 317.0 (22%), 316.0 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C15H15N3Br=316.0449. Found: 316.0453. Error (ppm): +1.3.
  • 4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)-2-methylbenzenamine (L20). N-(4-(6-bromo-H-imidazo[1,2-a]pyridin-2-yl)-2-methylphenyl)-2,2,2-trifluoroacetamide (1.0 g; 2.51 mmol) and K2CO3 (4.0 g; 29.0 mmol) were suspended in 30 mL ethanol and 15 mL water. The mixture was refluxed overnight. The solvent was removed, and CH2Cl2 was added. The organic phase Was washed with H2O twice, and dried over MgSO4. The solvent was removed to afford a solid which was purified by silica gel column chromatography to obtain 0.60 g (yield 79%) of product as yellow solid. 1H NMR (300 MHz; DMSO-d6), δ8.81 (s, 1H, Ar—H), 8.13 (s, 1H, Ar—H), 7.49 (d, 3JHH=9.5 Hz, 1H, Ar—H), 7.30 (d, 3JHH=9.5 Hz, 1H, Ar—H), 7.25 (s, 1H, Ar—H), 7.03 (d, 3JHH=7.4 Hz, 1H, Ar—H), 6.95 (d, 3JHH=7.7 Hz, 1H, Ar—H), 2.06 (s, 3H, Ar-CH3).
  • 4-(6-chloroimidazo[1,2-a]pyrazin-2-yl)-N,N-dimethylbenzenamine (L21). In a double necked round bottomed flask equipped with condenser and under nitrogen flow were introduced 5-chloro-2-amino-pyrazine (61 mg, 0.47 mmol), 2-bromo-4′-dimethylamino-acetophenone (170 mg, 0.71 mmol) and anhydrous acetonitrile (7 mL). The reaction mixture was refluxed for 3 hrs. After it cooled down, NaHCO3 (71 mg, 0.85 mmol) was added. The reaction was refluxed for another 6 hrs. The solvent was removed and the crude product (221 mg) was purified by FCC (hex/DCM/AcOEt=2:2:1), to afford 9 mg (yield 7%) of the product as yellow solid. 1H NMR (270 MHz; CDCl3), δ8.82 (s, 1H, Ar—H), 8.09 (s, 1H, Ar—H), 7.822 (s, 1H, Ar—H), 7.818 (d, 3JHH=8.9 Hz, 2H, Ar—H), 6.77 (d, 3JHH=8.7 Hz, 2H, Ar—H), 3.02 (s, 6H, NCH3). m/z (CI-MS): 273 ([M+1]+), 261,239 ([M—Cl+1]+).
  • 4-(6-chloroimidazo[1,2-a]pyrimidin-2-yl)-N,N-dimethylbenzenamine (L22). 2-Amino-5-chloro-pyrimidine (0.150 g, 1.16 mmol), 2-bromo-4′-dimethylamino-acetophenone (0.56 g, 2.32 mmol) and 16 mL of anhydrous acetonitrile were added to a double-neck flask equipped with a condenser under dinitrogen. A small amount of anhydrous DMF (0.5 mL) was added to completely dissolve the amine. After 3 hrs of stirring and refluxing, the mixture was cooled down and NaHCO3 (0.19 g, 2.32 mmol) was added. The reaction mixture was refluxed for another 6 hrs and was filtered hot on a Busch funnel to collect the precipitate, which was washed with portions of EtOH, water, EtOH, and dried overnight, to afford 72 mg (yield 23%) of the product as yellow powder. M.p.=256-258° C. 1H NMR (270 MHz; CDCl3), δ8.37 (d, 2H, 4JHH=2.7 Hz, Ar—H), 7.88 (d, 3JHH=9.2 Hz, 2H, Ar—H), 7.67 (s, 1H, H-3), 6.79 (d, 3JHH=8.7, 2H, Ar—H), 3.01 (s, 6H, NCH3). 13C{1H} NMR(125 MHz 13C, CDCl3), δ150.5, 149.3, 147.9 (C7), 147.0, 129.6 (C5), 127.5 (Ph-2,6), 117.5, 112.8 (Ph-3,5), 106.2 (C—Cl), 104.8 (C3), 40.7 (NCH3), m/z (CI-MS): 273 ([M+1]+), 239 ([M—Cl+1]+). Calc. C14H13ClN4; C, 61.75; H, 4.82; N, 20.59. Found: C, 58.32; H, 4.88; N, 18.81.
  • General procedure for thiolate substitution: A 10 mL microwave tube was charged with Pd2dba3 (13-58 mg, 0.014-0.063 mmol, 10-20 mol % Pd), DiPPF (6-27 mg, 0.014-0.063 mmol, 10-20 mol %), IMPY derivative (0.14--0.32 mmol), and tin-thiolate (0.29-0.63 mmol). The tube was capped, and put in a microwave system for desired temperature and time as specified in the text. A small sample of the resulting suspension was analyzed by HPLC to confirm the conversion. The suspension was partitioned between CHCl3 and K2CO3 solution. The organic layer was dried over MgSO4 and filtered. The solvent was removed, and the residue was dissolved in DMSO, and loaded on a reversed phase HPLC. After proper band was collected, the solvents were removed, and the product was dried by azetrope with CH3CN to provide the desired product.
  • 2-(2-(4(methylamino)pheny)-H-imidazo[1,2-a]pyridin-6-ylthio)acetamide (T1). 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N-methylbenzenamine (100 mg, 0.29 mmol), N-methyl-N-(trimethylstannyl)methanamine (60 mg, 0.29 mmol), 2-mercaptoacetamide (26 mg, 0.29 mmol), Pd2(dba)3 (26.2 mg, 0.029 mmol), DiPPF (12 mg, 0.029 mmol), and 8.0 mL of toluene were used.
  • Mp: 198-202° C.; 1H NMR (400 MHz, DMSO-d6) δ8.68 (d, 4JHH=4.4 Hz, 1H, Ar—H), 8.11 (s, 1H, Ar—H), 7.68 (d, 3JHH=8.0 Hz, 2H, Ar—H), 7.48 (d, 3JHH=9.6 Hz, 1H, Ar—H), 7.26 (d, 3JHH=9.2 Hz, 1H, Ar—H), 7.13 (s, 1H, Ar—H), 6.59 (d, 3JHH=8.0 Hz, 2H, Ar—H), 5.86 (s, 2H, NH2), 3.53 (s, 2H, CH2S), 2.71 (s, 3H, CH3). 13C{1H} NMR (400 MHz, DMSO-d6) δ169.8 (—CO—N), 149.8, 146.2, 143.6, 128.0, 127.7, 126.6, 120.9, 118.2, 115.9, 111.6, 106.8, 38.5 (C—S), 29.6 (NCH3). m/z (LC-MS): 314.3 (21%), 313.2 (100%, [M+H]+), 255.5 (32%, [M—CH2CONH2]+). HRMS m/z (TOF+): Calc. C16H17N4OS=313.1123. Found: 313.1126. Error (ppm): +0.9.
  • 2-(2-(4-(dimethylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ylthio)acetamide (T2). 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (50 mg, 0.14 mmol), N-methyl-N-(trimethylstannyl)methanamine (30 mg, 0.14 mmol), 2-mercaptoacetamide (13 mg, 0.14 mmol), Pd2(dba)3 (13 mg, 0.014 mmol), DiPPF (6 mg, 0.014 mmol), and 5.0 mL of toluene were used.
  • Mp: 180-183° C.; 1H NMR (400 MHz, DMSO-d6) δ8.60 (s, 1H, Ar—H), 8.17 (s, 1H, Ar—H), 7,77 (d, 3JHH=8.8 Hz, 2H, Ar—H), 7.50 (d, 3JHH=7.9 Hz, 1H, Ar—H), 7.27 (d, 3JHH=9.4 Hz, 1H, Ar—H), 7.13 (brs, 2H, NH2), 6.78 (d, 3JHH=8.7 Hz, 2H, Ar—H), 3.54 (s, 2H, CH2S), 2.94 (s, 6H, N(CH3)2), 13C{1H} NMR (400 MHz, DMSO-d6) δ168.7 (—CO—N), 149.0, 144.7, 142.5, 129.4, 127.8, 126.9, 126.6, 125.3, 120.3, 117.2, 114.9, 111.1, 109.5, 106.1, 38.5 (C—S), 37.3 (NCH3). m/z (LC-MS); 329.3 (7%), 328.2 (18%), 327.2 (100%, [M+H]+), 325.1 (12%), 323.4 (7%), 321.7 (7%), 320.9 (5%), 269.4 (14%, [M—CH2CONH2]+). HRMS m/z (TOF+); Calc. C17H19N4OS=327.1280. Found: 327.1278. Error (ppm); −0.5.
  • 2-(2-(4-(methylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ylthio)ethanol (T3). 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N-methylbenzenamine (100 mg, 0.29 mmol),N-methyl-N-(trimethylstannyl)methanamine (60 mg, 0.29 mmol), 2-mercaptoethanol (23 mg, 0.29 mmol), Pd2(dba)3 (28 mg, 0.031 mmol), DiPPF (22 mg, 0.039 mmol), and 5.0 mL of toluene were used.
  • Mp: 158-161° C.; 1H NMR (400 MHz, CD3OD) δ8.51 (s, 1H, Ar—H), 7.95 (s, 1H, Ar—H), 7.68 (d, 3JHH=8.6 Hz, 2H, Ar—H), 7.45 (d, 3JHH=8.6 Hz, 1H, Ar—H), 7.34 (d, 3JHH=8.6 Hz, 1H, Ar—H), 6.67 (d, 3JHH=8.6 Hz, 2H, Ar—H), 3.69 (t, 3JHH=6.5 Hz, 2H, OCH2), 3.01 (t, 3JHH=6.5 Hz, 2H, SCH2), 2.81 (s, 3H, NCH3), 13C{1H} NMR (400 MHz, CD3OD) δ151.8, 148.0, 145.7, 130.9, 129.5, 128.1, 122.6, 121.2, 116.5, 113.4, 108.5, 61.5 (C-O), 38.8 (C-S), 30.6 (NCH3). m/z (LC-MS): 302.2 (5%), 301.3 (15%), 300.2 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C16H18N3OS=300.1171. Found: 300.1163. Error (ppm): −2.5.
  • 2-(2-(4-(dimethylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ylthio)ethanol(T4). 4-(6-Iodo-H-imidazop[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (50 mg, 0.14 mmol), N-methyl-N-(trimethylstannyl)methanamine (29 mg, 0.14 mmol), 2-mercaptoethanol (11 mg, 0.14 mmol), Pd2(dba)3 (13 mg, 0.014 mmol), DiPPF (6 mg, 0.014 mmol), and 5.0 mL of toluene were used.
  • Mp: 183-185° C.; 1H NMR (400 MHz, CD3OD) δ8.50 (s, 1H, Ar—H), 8.05 (s, 1H, Ar—H), 7.78 (d, 3JHH=8.9 Hz, 2H, Ar—H), 7.45 (d, 3JHH=8.9 Hz, 1H, Ar—H), 7.35 (d, 3JHH=8.9 Hz, 1H, Ar—H), 6.83 (d, 3JHH=8.9 Hz, 2H, Ar—H), 3.70 (t, 3JHH=6.7 Hz, 2H, OCH2), 3.03 (t, 3JHH=6.7 Hz, 2H, SCH2), 3.00 (s, 6H, NCH3), 13C{1H} NMR (400 MHz, DMSO-d6) δ150.1, 145.7, 143.6, 128.1, 127.4, 126.4, 121.5, 118.5, 116.1, 112.2, 107.1, 59.8 (C—O), 55.9 (C—S), 37.2 (NCH3). m/z (LC-MS): 316.1 (5%), 315.2 (23%), 314.2 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C17H20N3OS=314.1327. Found: 314.1317. Error (ppm): −3,4.
  • 4-(6-(4-methoxybenzylthio)-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (T5). 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (80 mg, 0.22 mmol), N-methyl-N-(trimethylstannyl)methanamine (46 mg, 0.22 mmol), (4-methoxyphenyl)methanethiol (34 mg, 0.22 mmol), Pd2(dba)3 (40 mg, 0.044 mmol), DiPPF (18.4 mg, 0.044 mmol), and 5.0 mL of toluene were used.
  • Mp: 184-186° C.; 1H NMR(400 MHz, CD3OD) δ8.43 (s, 1H, Ar—H), 8.10 (s, 1H, Ar—H), 7.75 (d, 3JHH=8.4 Hz, 2H, Ar—H), 7.47 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.19 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.16 (d, 3JHH=8.0 Hz, 2H, Ar—H), 6.83 (d, 3JHH=8.0 Hz, 1H, Ar—H), 6.77 (d, 3JHH=8.0 Hz, 1H, Ar—H), 4.13 (s, 2H, CH2S), 3.71 (s, 3H, CH3O), 2.94 (s, 6H, NCH3). 13C{1H} NMR (400 MHz, DMSO-d6) δ158.2, 150.0, 145.6, 143.5, 129.9, 129.2, 128.2, 127.7, 126.3, 121.3, 118.0, 115.8, 113.9, 112.1, 107.0, 54.8 (OCH3), 39.9 (NCH3), 38.3 (C—S). m/z (LC-MS): 392.1 (7%), 391.1 (23%), 390.1 (100%, [M+H]+), HRMS m/z (TOF+): Calc. C23H24N3OS=390.1640. Found: 390.1634. Error (ppm); −0.6.
  • N,N-dimethyl-4-(6-(methylthio)-H-imidazo[1,2-a]pyridin-2-yl)benzenamine (T6). 4-(6-Iodo-H-imidazo[1,2-a]pyridin-2-yl)-N,N-dimethylbenzenamine (90 mg, 0.25 mmol), 1,2-dimethyldisulfane (28 mg, 0.30 mmol), 1,1,1,2,2,2-hexamethyldistannane (97 mg, 0.30 mmol), Pd2(dba)3 (22.8 mg, 0.025 mmol), DiPPF (10.5 mg, 0.025 mmol), and 4.0 mL of toluene were used.
  • Mp: 155-164° C.; 1H NMR (400 MHz, CD3OD) δ8.34 (s, 1H, Ar—H), 7.96 (s, 1H, Ar—H), 7.73 (d, 3JHH=9.4 Hz, 2H, Ar—H), 7.44 (d, 3JHH=9.4 Hz, 1H, Ar—H), 7.26 (dd, 3JHH=9.4 Hz, 4JHH=2.2 Hz, 1H, Ar—H), 6.82 (d, 3JHH=8.9 Hz, 2H, Ar—H), 13C{1H} NMR (400 MHz, CD3OD) δ152.3, 147.6, 145.6, 129.0, 128.1, 125.9, 124.6, 122.8, 116.7, 113.9, 108.8, 40.9 (C−S), 17.9 (NCH3), m/z (LC-MS): 286.2 (4%), 285.3 (13%), 284.2 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C16H18N3S=284.1221. Found: 284.1215. Error (ppm): −2.4.
  • 2-(2-(3-methyl-4-(methylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ylthio)acetamide (T8). 4-(6-Bromo-H-imidazo[1,2-a]pyridin-2-yl)-N2-dimethylbenzenamine (100 mg, 0.32 mmol), N-methyl-N-(trimethylstannyl)methanamine (99 mg, 0.48 mmol), 2-mercaptoacetamide (43 mg, 0.48 mmol), Pd2(dba)3 (43 mg, 0.048 mmol), DiPPF (20 mg, 0.048 mmol), and 6.0 mL of toluene were used.
  • Mp: 181-182° C.; 1H NMR (400 MHz, CD3OD) δ8.58 (s, 1H, Ar—H), 8.10 (s, 1H, Ar—H), 7.49 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.41 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.17 (s, 1H, Ar—H), 7.12 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.06 (d, 3JHH=8.0 Hz, 1H, Ar—H), 3.46 (s, 1H, NH), 3.55 (s,2H, SCH2), 2.93 (s, 3H, NCH3), 2.16 (s, 3H, Ar-CH3). 13C{1H} NMR (400 MHz, DMSO-d6) δ168.7 (—CO—), 146.6, 144.9, 142.4, 131.0, 128.6, 127.1, 126.6, 120.5, 117.5, 115.3, 112.0, 107.5, 104.5, 37.2 (CH2S), 29.0 (NCH3), 16.3 (CH3). m/z (LC-MS): 329.2 (6%), 328.3 (22%), 327.2 (100%, [M+H]+). HRMS m/z (TOF+): Calc. C17H19N4OS=327.1280. Found: 327.1278. Error (ppm): −0.4.
  • 2-(2-(3-methyl-4-(methylamino)phenyl)-H-imidazo[1,2-a]pyridin-6-ylthio)ethanol (T9). 4-(6-Bromo-H-imidazo[1,2-a]pyridin-2-yl)-N,2-dimethylbenzenamine (100 mg, 0.32 mmol), N-methyl-N-(trimethylstannyl)methanamine (132 mg, 0.63 mmol), 2-mercaptoethanol (50 mg, 0.63 mmol), Pd2(dba)3 (58 mg, 0.063 mmol), DiPPF (27 mg, 0.063 mmol), and 6.0 mL of toluene were used.
  • Mp: 149-153° C.; 1H NMR (400 MHz, CD3OD) δ8.55 (s, 1H, Ar—H), 8.10 (s, 1H, Ar—H), 7.50 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.38 (d, 3JHH=8.0 Hz, 1H, Ar—H), 7.18 (s, 1H, Ar—H), 7.15 (d, 3JHH=8. 0 Hz, 1H, Ar—H), 7.08 (d, 3JHH=8.0 Hz, 1H, Ar—H), 3.80 (t, 3JHH=7.2 Hz, 2H, CH2O), 3.03 (t, 3JHH=7.2 Hz, 2H, CH2S), 2.94 (s, 3H, NCH3), 2.16 (s, 3H, Ar-CH3). 13C{1H} NMR (400 MHz, DMSO-d6) δ147.7, 146.0, 143.4, 132.2, 129.7, 128.2, 127.4, 121.6, 118.8, 116.5, 113.1, 108.6, 105.6, 59.5 (OCH2), 37.1 (CH2S), 30.1 (NCH3), 17.5 (CH3), m/z (LC-MS): 329.2 (6%), 328.3 (22%), 327.2 (100%, [M+H]+), m/z (LC-MS): 316.2 (8%), 315.2 (24%), 314.2 (100%, [M+H]+), HRMS m/z (TOF+); Calc. C17H20N3OS=314.1327. Found: 314.1316. Error (ppm): −3.5.
  • Isolation of human AD amyloid. Postmortem brain times were obtained from a confirmed AD patient at the age of 80. Pre- and postmortem consent was obtained from next of kin, and brains were removed at autopsy, sectioned and frozen at −70° C. Bags of brain tissue were removed from the freezer to a −20° C. freezer one day before, and then placed on ice 3 h before the experiment. While working on ice, the brain tissue was cut into small sections, and meningies from outer layer of cortex and from fissures between the lobes of the gray matter was removed. After removing the white matter, the grey matter were chopped into small pieces and placed into plastic bags. The tissue was stored in −70° C. freezer for further use.
  • In a typical isolation, 60 g of brain tissue were used. Initially, an equal volume of 1% SDS was added to homogenize. More 1% SDS was added later until 200 mL. The mixture was vortexed, distributed evenly among 10 Beckman ultracentrifuge tubes, and centrifuged by a Coulter type 55.2 Ti ultracentrifuge rotor using RPM: 40,000; G force; 180,000; temp: 4° C.; time: 30 min. The collected bottom pellets were further homogenized and centrifuged twice more as described above using 50 mL and 20 mL of 1% SDS each, and finally in 20 mL 1.2 M sucrose. The supernatant along the top pellet was discarded. The pellets at the bottom were homogenized and centrifuged in 20 mL 1.9 M sucrose. The top pellets were collected using a cell scrapper, and the bottom pellets were discarded. The collected top pellets were homogenized and centrifuged using 20 mL deionized water. The bottom pellets were further homogenized and centrifuged using 1.3 M sucrose solution into one centrifuge tube. The bottom pellets were homogenized and centrifuged using 20 mL deionized water. The collected pellets were homogenized in 30 mL of 50 mM Tris-HCl, pH 8.0, 2 mM CaCl2. Collagenase CLS3 (120 mg) and DNAase I (10-15 mg) were added. The mixture was vortexed and incubated for 24-56 hrs at 37° C. in a water bath with shaking. After incubation, the suspension was filtered through 41 μm Milllpore filter paper. The collected filtrate was centrifuged and the bottom pellets were homogenized and centrifuged one more time using the Tris-HCl solution. The supernatant was discarded, and the pellets were homogenized and centrifuged using 10 mL 1.9 M sucrose. The top pellet and the 1.9M sucrose layer were transferred to a new tube (this is to remove heavy particles such as metals and salts). Deionized water (40 mL) was added. After homogenization and centrifugation, the collected pellets were put into 500 μL of deionized water to form a homogeneous suspension. It was stored in a refrigerator or freezer for further use.
  • In vitro Binding Assay
  • The amyloid suspension prepared above was diluted by a factor of 40,000 to 50,000 in PBS, and 800 μL suspension was used in each tube. [3H]6-OH-BTA1 with a concentration of 1 mCi/mL stock solution was diluted using ethanol to give an intermediate solution of 1 μCi/100 μL, which was further diluted using PBS to result in a dilute stock solution of 2.7×10−2 μCi/100 μL, and 100 μL was used in each tube (2 vials with 100 μL of [3H]6-OH-BTA-1 each and scintillation fluid as references). Cold 6-OH-BTA-1 or other displacer was dissolved in ethanol to result in a stock solution of 1×10−3 M, which was further diluted using PBS or ethanol to result in solutions with concentrations ranging from 1×10−5 to 10−10 M, and 100 μL was used in each tube. After assembly, the tubes were vortexed, and incubated for 3 hrs at room temperature. After separation using a cell harvester, the filter paper (GF/B filter paper pretreated with 0.5% polyimine solution) was washed with 10% ethanol in PBS (3×3 mL). The filters were placed into 20 mL glass vials and 10 mL scintillation fluid each was added. After overnight incubation, the samples were counted. The data were analyzed using GraphPad Prism 4 or KaleidaGraph 3.6.
  • Results and Discussion.
  • Chemical Synthesis
  • Synthesis of Bromoketones. The nucleus of imidazo[1,2-a]pyridines is made through the condensation between a 2-amino pyridine and an α-haloketone in a manner described before.2 The reaction tolerates a variety of substituents, including amide and imine groups in both substrates. Two strategies have been developed to synthesize the α-bromoketones (FIG. 1). One is from bromination of ketones. Mono-methylation of the amino group of 1-(4-aminophenyl)ethanone follows the method established, to give 1-(4-methylaminophenyl)ethanone, KI.2 Ortho-bromination of the aromatic amine was accomplished using NBS, a mild bromination agent for activated phenyl group, to give K2and K3.3 The amino group was protected using either trifluoroacetyl or acetyl group to give K5 and K6 before bromination of the α-methyl ketone. Three bromination procedures have been reported.4 One is through tetra-n-butyl ammonium tribromide, the second is through copper (II) bromide, and the third is through the reduction of di-bromides, to afford α-bromoketones B2, B3, B4, B5. The other method to synthesize the α-bromoketones is through Friedel-Crafts reaction to introduce α-bromoacetyl group directly in the aromatic nucleus. The aromatic amino group in A1 and A2 were protected using trifluoroacetyl or acetyl group, to give A3 and A4. Friedel-Crafts reactions using trifluoroacetyl or bromoacetyl bromide catalyzed by AlCl3 in CS2 went smoothly to give B2, B3, B4, B5.5-7
  • Synthesis of 2-Aminopyridines and 2-Aminopyrazines. The synthetic methods of pyridine derivatives depend on the substituents on the ring (FIG. 2). For CN group, halide substitution using CuCN is an optimal choice, such as the synthesis of P38-10 and P4(available from Acros). Some halo group could be introduced through electrophilic substitution of either bromination or iodination, such as the synthesis of P10, P11 and P12.
  • Substitution of halide by OCH3 and SEt group is more challenging. The first attempts to prepare P5 by a nucleophilic aromatic displacement of the bromide with a methoxy group catalyzed by Cu powder in a conventional way11 or with microwave heating or an ultrasound bath were frustrated by the apparent unreactivity of the substrate. An attempt was carried out with an ultrasound high power probe (Branson 450 Sonifier) beaming the 40 dB sound waves directly in the solution, but even this technology did not enable the expected transformation (eq 1).
  • Figure US20140341812A1-20141120-C00003
  • Another literature procedure to perform aromatic substitutions with the methoxy anion was followed.12 The method uses salts of copper (I) and a small amount of ester to form a stabilized tetrahedral adduct which should act as a powerful methoxide donor (FIG. 3). Unfortunately, due to the presence of the free amino group on the bromopyridine and the ethyl acetate as co-catalyst, the reaction product was just the acetamide of the substrate (eq 2).
  • Figure US20140341812A1-20141120-C00004
  • Attempts to improve the result were made by increasing the equivalents of catalyst Used. It was believed that copper could be coordinated and thus inactivated by the nitrogen atoms of the substrate. By this means the expected product was achieved in 36% yield (eq 3). The reaction mixture, after being heated for 14 hrs, still contained a significant amount of starting material together with black polymers. The reaction was quenched to avoid further formation of polymers and degradation of the formed product. In order to decrease the reaction time, the transformation in eq 3 was carried out with microwave irradiation according to Table 3.
  • Figure US20140341812A1-20141120-C00005
  • TABLE 3
    Microwave reaction of eq 3 at 410° C.
    Starting Ratio of
    Time (h) Solvent Recovered Substitution:Reduction:Polymer
    1.0 MeOH Nothing small:small:large
    0.5 DME 70% 0:0:large
    0.5 DMF Trace 17%:0:large
  • Some modifications to this method were tried to increase the yield. 5-Iodo-2-aminopyridine was used in the hope that the weaker C-I bond could help to increase the rate of the displacement. The consumption of aromatic iodide was faster (6 h) but unfortunately only polymers formed. Decreasing the reaction temperature did not help (eq 4).
  • Figure US20140341812A1-20141120-C00006
  • A different strategy was designed using 5-pyridyl boronate and subsequently oxidized to P5 in the presence of NaOCH3. After a first attempt to prepare the boronate P6 in DMSO13 with conventional heating failed, the same coupling was tried with microwave heating using DME as the solvent.14 This procedure gave the desired boronate in 20 min instead of 20 h, but only in 10% yield due to problems found in the work-up (eq 5).
  • Figure US20140341812A1-20141120-C00007
  • The free amino group was protected to avoid undesirable coordination of the Pd catalyst and simplify the reaction work-up. In addition, 5-iodo-2-aminopyridine was used to facilitate the Pd insertion in the C-halogen bond. Therefore P1 was protected with trifluoroacetic anhydride to give trifluoroacetamide P7 in 77% yield. This was reacted with pinacolborane in dioxane at 80° C. for 3 h to give the protected pyridyl boronate P8 in 67% yield (eq 6).15
  • Figure US20140341812A1-20141120-C00008
  • Oxidation Studies did not generate any desired product. The route was Discontinued.
  • Substitution of halide by SEt group was similar to that of OCH3 group when using copper as catalysts. Although syntheses of P916;17 have been reported, the reported methods made use of copper as catalyst and methanol as solvent at 150° C. under pressure.18 Due to the inconvenience of the procedure, other solvents were evaluated to replace the low boiling point methanol. Ethylene glycol proved to be an excellent choice in this reaction, giving high yield in a short reaction time. The benefits of ethylene glycol as solvent in other copper or copper(I)-catalyzed reaction have been noted.19 Ethylene glycol was found to be the ligand and solvent when CuI was used as catalyst, to give the aromatic thio-ether, 2-amino-5-ethylthio-pyridine.
  • The synthesis of 5-chloro-2-aminopyrazine described in the literature is not a very efficient multi-step process.20 Direct chlorination of the 2-aminopyrazine using N-chlorosuccinimide was used, although the method is known to give over chlorinated by-products. Initial attempt showed that the reaction gave a lot of black polymers after a few min., with the main product being 3,5-dichloro-2-amino pyrazine. Only small amount of 5-chloro-2-aminopyrazine was isolated (eq 7). This reaction could not be scaled up because it was not reproducible. To avoid over chlorination, milder conditions were examined. No reaction was observed at RT, but a complete transformation into the dichloro derivative was observed at 50° C.
  • Figure US20140341812A1-20141120-C00009
  • When the distilled chloroform used for the reaction was pre-treated over basic Al2O3, the black polymers formed in the reaction decreased dramatically. When NCS was added slowly into the refluxing 2-aminopyrazine, the product was isolated at 26%. Moreover, when microwave irradiation was used at 70° C. for 10 min., the yield reached 45% (FIG. 4).
  • Synthesis of IMPY Derivatives I. Most of the IMPY derivatives were synthesized through the direct condensation of α-bromoketones and 2-amino-pyridines (FIG. 5).2 In selected examples, when trifluoroacetyl group was used to protect aromatic amino group, no base was needed for the condensation. The difficulty facing the synthesis of L22 came from separation. When excess amine was used, the product could not be separated from the starting amine. When excess α-bromoketone was used, a persistent amount of the starting pyrimidine existed. A small amount of DMF was added in the reaction mixture to dissolve the amine completely, and to induce the precipitation of the product at the same time. By filtering this solution hot, even before reaction complete, we were able to isolate the desired imidazopyrimidine L22 in 23% yields (eq 8).
  • Figure US20140341812A1-20141120-C00010
  • Imidazopyrazine L21 was synthesized from condensation of chloropyrazine P13 and bromoacetophenone B1 (eq 9). The amine 67 was very unreactive though it is very soluble in acetonitrile. Long reaction time and stronger bases such as triethylamine were examined with limited success.
  • Figure US20140341812A1-20141120-C00011
  • The synthesis of L12 was through demethylation of the methoxy group in L9. The most effective reagent to perform the reaction is BBr3 in DCM (eq 10).21
  • Figure US20140341812A1-20141120-C00012
  • The trifluoroacetyl group on the aromatic amino group in L15, L16, and L17, was easily removed under mild condition of K2CO3 in methanol or ethanol (FIG. 4). This provides an excellent method for the synthesis of IMPY derivatives with amino group(s).
  • Synthesis of IMPY Derivatives II. For L9, we also attempted direct methoxylation of halide in pre-formed IMPY derivatives. Unfortunately the reaction produced a low yield mixture of reduced (L11), cine (L10) and ipso (L9) substituted analogues which were difficult to separate (FIG. 6). Catalytic substitution of halides by thiolate based on palladium-catalysts was developed to overcome the reduction of the halide (FIG. 7).22 The new method has been applied to the synthesis of a number of IMPY derivatives with thiolate group.
  • An advantage of the IMPY derivatives of the present invention relative to the prior art compound [11C]PIB (6-OH-BTA-1) is that the derivatives of the present invention can be easily labeled with [18F], which has a half-life of about two hours as compared with twenty minutes for [11C]. A process for radiolabeling the IMPY derivatives with either label is shown below.
  • Figure US20140341812A1-20141120-C00013
  • Isolation of Human Amyloid
  • A method for isolating human β-amyloid plaques from brain tissue23;24 was modified by incorporating repeated homogenization and centrifugation in 1% SDS buffer and by using DNase and collagenase for digestion. The concentration of β-amyloid plaques was gauged from the concentration of Aβ1-40 and Aβ1-42 monomers, using ELISA to detect dissolved plaques in formic acid.
  • In Vitro Assay
  • Determination of the binding affinities of new ligands for β-amyloid is the first step in selecting candidate radioligands for PET studies in humans. Three types of amyloid plaques have been used to assay ligand binding in vitro, namely synthetic aggregates of Aβ1-40, Aβ1-42, amyloid plaques from transgenic mice and amyloid plaques from human Alzheimer's disease (AD) brain tissue. Results from the three types and also from different batches of synthetic amyloid plaques vary with respect to binding site concentration.25 These findings may reflect variations in binding site architecture, though no significant difference in ligand binding affinity has been detected among the three types of amyloid plaques for the binding site typical of 6-OH-BTA-1.26 Since the ultimate test for a radioligand is successful application in humans, in vitro evaluation using human AD brain tissue is highly appropriate. However, it should be noted that other proteins with similar binding sites might interfere with the binding of the ligand to β-amyloid plaques. This may then be reflected in a lower percentage of displaceable radioactivity in the binding assay.27 The use of isolated human amyloid plaques may also help to identify other native binding sites.
  • Isolated plaques were used in developing an in vitro binding assay. Tritiated-6-OH-BTA-1 was selected as the reference radioligand based on its use in human PET imaging and high affinity.28 Displacement curves were created using non-radioactive 6-OH-BTA-1 and other novel ligands.
  • The displacement of [3H]6-OH-BTA-1 by non-radioactive 6-OH-BTA-1 or other ligands resulted in classical displacement curves. Some lipophilic ligands required ethanol in the medium to increase their solubility and to effect displacement of reference radioligand. Non-radioactive 6-OH-BTA-1 achieved >95% displacement of reference radioligand, indicating that the presence of competing binding sites for the isolated amyloid plaques was negligible. This assay could therefore be used to screen compounds for amyloid binding without interference from other proteins, such as tau-tangles. This displacement curve was also analyzed using a homologous displacement mathematical model to extract the Bmax of the amyloid plaques. The measured Bmax is linear with the amount of amyloid used in the experiment. The ratio between Bmax and the amount of Aβ1-42 monomer measured by ELISA is about 1:2, somewhat less than that reported previously.25 The denaturing agents used to aid dissolution of the amyloid plaques may account for the observed difference.
  • The binding affinities for a variety of IMPY derivatives are listed in Table 3. In this table, Ki is a measure (in nM units) of the binding affinity of the compound toward beta-amyloid plaques in AD brain tissue, measured through a competitive radioligand displacement in vitro assay using [3H]6-OH-BTA-1 as the reference radioligand. The lipophilicity of the compound is represented by cLogD7.4 which is calculated through a software package (ACD/LogD version 8.0) at pH=7.4, a close mimic of physiological conditions.
  • Figure US20140341812A1-20141120-C00014
  • TABLE 4
    IMPY derivatives.
    Ligand R1 R2 R3 R4 R5 cLogD7.4 Ki (nM)
    L1 (IMPY) I H H Me Me 4.37 ± 0.88  8.9 ± 0.7
    L4 Br H H Me Me 4.11 ± 0.88  5.9 ± 0.4
    L5 Cl H H Me Me 3.93 ± 0.84 24.2 ± 5.6
    L6 F H H Me Me 3.38 ± 0.88 13.0 ± 1.6
    L7 NO2 H H Me Me 3.09 ± 1.30  7.6 ± 0.7
    L8 CN H H Me Me 2.80 ± 1.31  8.2 ± 1.0
    L9 OMe H H Me Me 3.05 ± 1.30 38.5 ± 5.0
    L12 OH H H Me Me 1.26 ± 1.29 177 ± 31
    T7 SEt H H Me Me 4.24 ± 1.31  8.3 ± 0.5
    L13 Br I H Me Me 5.17 ± 0.93 183 ± 61
    L14 Br CN H Me Me 3.28 ± 1.35  >180
    L18 Br H Br H Me 4.30 ± 0.94  7.4 ± 0.6
    L19 Br H Me H Me 4.16 ± 0.88 >1000
    L20 Br H Me H H 3.28 ± 0.87 658 ± 47
    T1 SCH2CONH2 H H H Me 1.59 ± 1.33 1840 ± 497
    T2 SCH2CONH2 H H Me Me 2.02 ± 1.33 391 ± 76
    T3 SCH2CH2OH H H H Me 2.54 ± 1.35 645 ± 75
    T4 SCH2CH2OH H H Me Me 2.96 ± 1.35 88 ± 6
    T5 SCH2C6H4OCH3 H H Me Me 4.76 ± 1.35  8.3 ± 1.8
    T6 SMe H H Me Me 3.67 ± 1.31  7.9 ± 0.6
    T8 SCH2CONH2 H Me H Me 2.12 ± 1.33 >1000
    T9 SCH2CH2OH H Me H Me 3.07 ± 1.35 >1000
    L2 I H H Me CH2CH2F 4.52 ± 0.92 31 ± 5
    L3 I H H Me CH2CH2CH2F 4.90 ± 0.92 41 ± 5
  • The BTA derivatives were synthesized through the condensation of 2-amino-aryl-thiols and aryl carboxylic acid under acidic conditions.
  • Figure US20140341812A1-20141120-C00015
  • TABLE 5
    Compound oLogD7.4 Ki (nM)
    L21 2.54 ± 1.30 88 ± 22
    L22 3.14 ± 1.43 147 ± 20 
    P1 4.36 5.7 ± 0.7
    PIB 3.31 7.23 ± 1.0 
  • Comparison of the Ki values of L1, L18 and L19 in Table 4 shows that the “isosteric effect” works partially. Since Br and Me are similar in size, the data show that a polar group increases binding. Similar analysis of the binding affinities of L1, L5, L6, and T7 shows that the thiol ether group provides optimal electronic and steric effects for binding, but the effect disappears as soon as a hydrophilic group is attached (c.f. results for T4 and T8). Increasing the size of substituents on the aromatic amino group decreases binding affinity, as reflected in L1, L2 and L3. However, there is substantial requirement for size in the 6-position (c.f. L6, L12 and L9). For L12, the combination of small and polar properties for the 6-substituent abolishes all binding affinity. Interestingly, introduction of a CN group into 8-position also completely removes the binding affinity (as in L14), reflecting-minimal tolerance for any substituents at the periphery of the IMPY skeleton.
  • The reference citations above are identified below under Reference List No. 1.
  • Reference List No. 1
  • 1. Selkoe, D. J. Alzheimer's disease; genes, proteins, and therapy, Physiol Rev, 2001, 81, 741-766.
  • 2. Cai, L.; Chin, F. T.; Pike, V. W.; Toyama, H.; Liow, J. S.; Zoghbi, S. S.; Modell, K.; Briard, E.; Shetty, H. U.; Sinclair, K.; Donohue, S.; Tipre, D.; Kung, M. P.; Dagostin, C.; Widdowson, D. A.; Green, M.; Gao, W.; Herman, M. M.; Ichise, M.; and Innis, R. B. Synthesis and evaluation of two 18F-labeled 6-iodo-2-(4′-N,N-dimethylamino)phenylimidazo[1,2-a]pyridine derivatives as prospective radioligands for beta-amyloid in Alzheimer's disease. J. Med. Chem. 2004, 47, 2208-2218.
  • 3. Hu, Jianguo, Li, Zuoy, Li, Yulin, Tang, Luoxiang, and Wu, Meijuan. Method of synthesizing 5-[1-hydroxy-2-(isopropylamino)ethyl]aminobenzonitrile. Faming Zhuanli Shenqing Gongkai Shuomingshu CN95-103024-19950403[CN1120535A19960417]. 1996. CAN130;139172.
  • 4. Cai, L.; Brouwer, C.; Sinclair, K.; Cuevas, J.; and Pike, V. W. Titanium(IV) chloride-promoted synthesis of new imidazo[1,2a]pyridine derivatives under microwave conditions. Synthesis 2005, 61, 0000.
  • 5. Hu, Z. Process for synthesizing ketoprofen. Faming Zhuanli Shenqing Gongkai Shuomingshu CN-95-109877-19950824[CN1143624A19970226]. 1997. CAN128:153928.
  • 6. Kuhla, D. E., Campbell, H. F., Studt, W. L., and Molino, B. F., Bicyolic heteroaryl thiazole compounds and their cardiotonic uses. PCT Int.Appl. WO 85-US2522-19851218[WO8603749A1-19860703]. 1986. CAN105:226538.
  • 7Otsuka-Pharmaceutical-Co, Ltd. Carbostyril derivatives and a cardiotonic composition containing them. Belg. BE82-207321-19820215[BE892148A1-19820816]. 1982. CAN98:34510.
  • 8. Ellis, G. P. and Romneyalexander, T. M. Cyanation of aromatic halides. Chem. Rev. 1987, 87, 779-794.
  • 9Liang, C. -H., Duffield, J., Romero, A., Chiu, Y. -H., Rabuka, D., Yao, S., Sucheck, S., Marby, K, Shue, Y. -K., Ichikawa, Y., and Hwang, C. -K. Preparation of macrolide erythronolide carbamates as antitumor and antibacterial agents. (Optimer Pharmaceuticals, Inc. USA. 2004-US6645[2004080391], 108. 2004, WO, CAN141;296244, Mar. 5, 2004.
  • 10. Ueno, T., Kimura, Y., and Kasuga, Y. Process for the preparation of Aminocyanopyridines. (Nippon Fine Chemical Co., Ltd. Japan and Mitsubishi Chemical Corp.). 2000-128683[2001302639], 4. 2001, JP, CAN135:331348, Apr. 28, 2000.
  • 11. Van de Poel, H.; Guillaumet, G.; and Viaud-Massuard, M. C. Synthesis of melatonin analogues derived from furo[2,3-b]- and [2,3-c]pyridines by use of a palladium-copper catalyst system. Heterocycles 2002, 57, 55-71.
  • 12. Capdevielle, P. and Maumy, M. Esters are effective cocatalysts in copper-catalyzed methanolysis of aryl bromides. Tetrahedron Letters 1993, 34, 1007-1010.
  • 13. Ishiyama, T.; Murata, M.; and Miyaura, N. Palladium(O)-catalyzed cross-coupling reaction of alkoxydiboron with haloarenes—A direct procedure for arylboronic esters. J. Org. Chem. 1995, 60, 7508-7510.
  • 14. Appukkuttan, P.; Van der Eycken, E.; and Dehaen, W. Microwave enhanced formation of electron rich arylboronates, Synlett 2003, 1204-1206.
  • 15. Baudoin, O.; Guenard, D.; and Gueritte, F. Palladium-catalyzed borylation of ortho-substituted phenyl halides and application to the one-pot synthesis of 2,2′-disubstituted biphenyls. J. Org. Chem. 2000, 65, 9268-9271.
  • 16. Sundberg, R. J.; Dahlhausen, D. J.; Manikumar, G.; Mavunkel, B.; Biswas, A.; Srinivasan, V.; King, F., Jr.; and Waid, P. Preparation of 2-aryl- and 2-(aryloxymethyl)imidazo[1,2-a]pyridines and related compounds. J. Heterocycl. Chem. 1988, 25, 129-137.
  • 17. Gol'dfarb, Y.; Stoyanovich, F. M.; Marakatkina, M. A.; and Gorushkina, G. I. Synthesis of 6-(alkylthio)imidazo[1,2-a]pyridines. Khimiya Geterotsiklicheskikh Soedinenii 1979, 634-638.
  • 18. Bochis, R. J.; Olen, L. E.; Waksmunski, F. S.; Mrozik, H.; Eskola, P.; Kulsa, P.; Wilks, G.; Taylor, J. E.; Egerton, J. R.; Ostlind, D. A.; and Olson, G. Substituted Imidazo[1,2-a]pyridine-2-carbamate anthelmintics. J. Med. Chem. 1981, 24, 1518-1521.
  • 19. Enguehard, C.; Allouchi, H.; Gueiffier, A.; and Buchwald, S. L. Easy access to novel substituted 6-aminoimidazo[1,2-a]pyridines using palladium- and copper-catalyzed animations. J. Org. Chem. 2003, 68, 4367-4370.
  • 20. Barlin, G. B.; Davies, L. P.; Ireland, S. J.; Ngu, M. M. L.; and Zhang, J. K. Imidazo[1,2-b]pyridazines .12. Syntheses and central-nervous-system activities of some substituted imidazo[1,2-b]pyridazines and related imidazo[1,2-a]pyridines, imidazo[1,2-a]pyrimidines and imidazo[1,2-a]pyrazines, Austr. J. Chem. 1992, 45, 877-888.
  • 21. Vickery, E. H.; Pahler, L. F.; and Eisenbraun, E. J. Selective O-demethylation of catechol ethers—comparison of boron tribromide and iodotrimethylsilane. J. Org. Chem. 1979, 44, 4444-4446.
  • 22. Cai, L.; Cuevas, J.; Peng, Y.; and Pike, V. W. Rapid palladium-catalyzed cross-coupling in the synthesis of aryl thioethers under microwave conditions. Org. Letters 2005.
  • 23. Soderberg, L.; Zhukareva, V.; Bogdanovic, N.; Hashimoto, T.; Winblad, B.; Iwatsubo, T.; Lee, V. M.; Trojanowski, J. Q.; and Naslund, J. Molecular identification of AMY, an Alzheimer disease amyloid-associated protein. J. Neuropathol. Exp. Neurol. 2003, 62, 1108-1117.
  • 24. Roher, A. E. Structural alterations in the peptide backbone of beta-amyloid core protein may account for its deposition and stability in Alzheimer's disease. J. Biol. Chem. 1993, 268, 3072-3083.
  • 25, Mathis, C. A.; Wang, Y.; Holt, D. P.; Huang, G. F.; Debnath, M. L.; and Klunk, W. E. Synthesis and evaluation of 11C-labeled 6-substituted 2-arylbenzothiazoles as amyloid imaging agents. J. Med. Chem. 2003, 46, 2740-2754.
  • 26. Klunk, W. E.; Wang, Y.; Huang, G. F.; Debnath, M. L.; Holt, D. P.; Shao, L.; Hamilton, R. L.; Ikonomovic, M. D.; DeKosky, S. T.; and Mathis, C. A. The binding of 2-(4′-methylaminophenyl)benzothiazole to postmortem brain homogenates is dominated by the amyloid component. J. Neurosci. 2003, 23, 2086-2092.
  • 27. Klunk, W. E.; Wang, Y.; Huang, G. F.; Debnath, M. L.; Holt, D. P.; and Mathis, C. A. Uncharged thioflavin-T derivatives bind to amyloid-beta protein with high affinity and readily enter the brain. Life Sci. 2001, 69, 1471-1484.
  • 28. Klunk, W. E.; Engler, H.; Nordberg, A.; Wang, Y.; Blomqvist, G.; Holt, D. P.; Bergstrom, M.; Savitoheva, I.; Huang, G. F.; Estrada, S.; Ausen, B.; Debnath, M. L.; Barletta, J.; Price, J. C.; Sandell, J.; Lopresti, B. J.; Wall, A.; Koivisto, P.; Antoni, G.; Mathis, C. A.; and Langstrom, B. Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B. Ann. Neurol. 2004, 55, 306-319.
  • A further aspect of this invention is the novel synthesis of aryl thioether derivatives of IMPY compounds by a rapid palladium-catalyzed cross-coupling under microwave conditions. In the following description, the cited references are identified under Reference List No. 2.
  • Aromatic thioethers of IMPY compounds are valuable as synthetic intermediates and as therapeutic drags.1;2 Homogeneous catalysts based on either copper(I) or palladium have been developed for their syntheses.2;3 Mechanistic studies of palladium-catalyzed aromatic substitution of halo or triflate groups by thiolate show that both three and four coordinate intermediates might be involved.4-6 Indeed, both mono and bidentate phosphine ligands have been developed for this type of reaction.7-12 Copper(I)-based batalysts for similar syntheses are mechanistically much less clear.13-17
  • In the synthesis of these compounds, introducing the thioether group in the last step avoids manipulation and protection of this sensitive group in the synthesis of the IMPY skeleton. The aromatic halide substrates to be substituted are generally accessible in multiple step syntheses.18 Successful substitution of aryl halides with thiolates requires the avoidance of a competing reductive removal of the halogen substituent. The objects of this aspect of the invention are to achieve (a) an one-step introduction of a thioether group into a halogen position in an aryl ring; (b) selective introduction of a thioether group at an iodo position in the presence of other halogen substituents, such as bromo; (c) tolerance of functional groups, especially amino groups; and (d) fast microwave-assisted reaction conditions. Disclosed herein is a general, efficient, and operationally simple palladium-catalyzed aryl thioether synthesis that achieves these objects.
  • Two reaction schemes are described below. The first is a copper(I)-mediated coupling of aryl iodides with thiols (Scheme B1), while the second is a palladium-catalyzed coupling of aryl iodides with tin thiolates (Scheme B2).
  • Figure US20140341812A1-20141120-C00016
  • Figure US20140341812A1-20141120-C00017
  • The aryl halide 4-(6-iodoindolizin-2-yl)-N-methylbenzenamine was used as the substrate, with either 2-mercaptoacetamide or 2-mercaptoethanol as the thiolating agent. In the copper-catalyzed reaction (Scheme B1), the catalyst used was CuI. Two ligands, ethylene glycol and ethylene diamine, were evaluated along with a variety of bases and solvents (Table B1). Low conversion of the iodo compound was observed with a catalytic amount of CuI. The reaction went to completion only when a greater than stoichiometric amount of CuI was used. For 2-mercaptoacetamide, the majority of the reaction product arose from reductive removal of the iodo group. For 2-mercaptoethanol, substitution of the iodo group was nearly quantitative for one substrate when 5 equivalents of CuI were used. However, no catalytic or general reaction was observed and so no further effort was expended on this approach for other substrates.
  • TABLE B1
    The copper(I)-mediated coupling of aryl iodides (1 or 2)
    with thiols: Percent yield of conversion followed by
    ratio of substitution to reduction in parentheses.
    Method;
    Substituents: (1) (2) (3)
    R2 = —CH2CONH2 100  81
    R = Me (1:4.0) (0:1)
    R2 = —CH2CONH2  81 100 100
    R = H (1:8.3) (1:4.1)a,b (0:1)
    R2 = —CH2CH2OH  60
    R = Me (messy)
    R2 = —CH2CH2OH  96
    R = H (1:0.038)
  • The general conditions were 130° C., 10 min, 50 W, 300 p.s.i., substrate: thiol=1: 1.2-5. In Method (1), 1-5 equiv CuI, 2 equiv ethylene diamine, 2 equiv t-BuOK or K2CO3 in DMSO, and N-methylpyrrolidin-2-one (NMP) or dioxane were used. In Method (2), 1-5 equiv CuI, 2 equiv ethylene glycol, and 2 equiv t-BuOK in Py were used. In Method (3), 0.05 equiv CuI, and 2 equiv Cs2CO3 in NMP were used. aWithout the thiol, the reaction generated reduced and HOCH2CH2O-substituted products. bNo reaction was observed when 1,2-dimethoxyethane was used instead of ethylene glycol.
  • The palladium-catalyzed coupling of aryl iodides with thiols (Scheme B2) was evaluated using the protocol originally established by Buchwald et al,7 who used aryl bromides as substrates. Variations here included the use of i) catalysts, such as (DPPF)PdCl2; ii) catalyst precursors, such as Pd2dba3 and Pd(OAc)2; iii) ligands (L) as shown in Scheme B2; iv) bases, such as NEt3, t-BuOK; and v) solvents, such as NMP, dioxane, or toluene. Multiple products in similar amounts were generated, including the desired substitution Products. Given the success of a number of bulky monophosphines in the catalysis of aryl C-N and C-O formation,6;19 a number of bulky monophosphines were evaluated, as shown below:
  • Figure US20140341812A1-20141120-C00018
  • Under the general conditions used (150° C., 10 min, 300 W, 300 p.s.i, with 0.05 equiv Pd2dba3, 0.1 equiv L, 2-4 equiv of NEt3 or t-BuOK, and ethanol or toluene as solvent), the new ligands behave similarly to PPh3, giving less than 10% total conversion of aryl iodide and ratios of substitution and reduction ranging from 0.4 to 15. No further reaction progress was observed on extended reaction time.
  • A number of thiolates have been used for the aromatic substitution reaction.1;20 When the tin thiolates were used in Scheme B2, mainly substitution products accompanied by minor reduction products were observed. Different kinds of bis-phosphine ligands were evaluated, as shown below:
  • Figure US20140341812A1-20141120-C00019
  • The results are shown in Table B2.
  • TABLE B2
    Scheme B2 with R1 = SnMe, R3 = R4 = R5 = R6 = H, R7 = Me, X = I:
    The palladium-catalyzed coupling of aryl iodides (1 or 2) with tin
    thiolates. General conditions: 150° C., 10 min, 300 W,
    300 psi, substrate:thiolate = 1:1, in toluene. Results
    shown are percent yield of conversion followed by ratio
    of substitution to reduction in parentheses.
    R2; R6:
    CH2CONH2; CH2CONH2; CH2CONH2; CH2CONH2;
    Ligand: H Me H Me
    BINAP 17 (1:0.13)a
    BINAP-Tol 81 (1:0.12)b
    DPPF 100 (1:0)c 56 (1:0.15)d
    DiPPF 51 (1:0.019)e 76 (1:0.14)f  6 (1:0.083)g
    a0.1 equiv Pd2(dba)3, 0.1 equiv L.
    b0.2 equiv Pd2(dba)3, 0.4 equiv L.
    c0.11 equiv Pd2(dba)3, 0.14 equiv L.
    d0.1 equiv Pd2(dba)3, 0.1 equiv L.
    e0.1 equiv Pd2(dba)3, 0.2 equiv L.
    f0.2 equiv Pd2(dba)3, 0.4 equiv L.
    g0.2 equiv Pd2(dba)3, 0.4 equiv L.
  • BINAP-Tol, DPPF, and DiPPF are efficient ligands for the reaction. Since a number of chelating phosphines have been used in enantioseleotive hydrogenation,21 they provide further excellent candidates for evaluation in this reaction. The yields and relative degrees of substitution and reduction for various phosphines when used in Scheme B2 are shown below in Table B3. Once again, ferrocene-based bis-phosphine,ƒ-binaphane, provided the best combination of reactivity and selectivity.
  • TABLE B3
    Scheme B2 with R1 = SnMe, R2 = CH2CONH2,
    R3 = R4 = R5 = R6 = H, R7 = Me, X = I:
    The palladium-catalyzed coupling of aryl iodide with tin thiolates.
    Ligands Products
    T-Phos 22 (1:0.91)
    TangPhos  7 (1:0.71)
    Binapine  1 (1:0.63)
    f-Binaphane 11 (1:0.077)
    C4TunePhos  8 (1:0.77)
    DuanPhos  0
    Binaphane  0

    General conditions; 150° C., 10 min, 300W, 300 psi, substrate:thiolate=1:1, 0.1 equiv Pd2(dba)3 and 0.1 equiv. ligand in toluene. Results shown are percent yield of conversion followed by ratio of substitution to reduction in parentheses. The structures of the ligand are shown in Ref. 21.
  • Palladium catalysts based on phosphine oxide ligands are a new type of efficient, versatile, air-stable and preformed homogeneous catalysts for the C-S bond formation.8-10 Ten of the most common catalysts were evaluated in Scheme B2. The results are shown in Table B4 which indicates that low reactivities and selectivities were generally observed.
  • TABLE B4
    The palladium-catalyzed coupling of aryl iodide (2) with tin thiolates,
    using Scheme B2 with R1 = SnMe, R2 = CH2CONH2,
    R3 = R4 = R5 = R6 = H, R7 = Me, X = I. General conditions:
    150° C., 10 min, 300 W, 300 psi, substrate:thiolate = 1:1.5, in toluene.
    In Method (i), 2.5 equiv K2CO3 was used; in Method (ii), 2.5 equiv
    K2CO3 and 2.5 equiv TEA were used, and in Method (iii), 2 equiv
    t-BuOK were used. The catalyst structures are shown in Refs. 8-10.
    The results shown are percent yield of conversion followed by ratio of
    substitution to reduction in parentheses.
    Method:
    Catalyst: (i) (ii) (iii)
    POPd 0 0
    PXPd 2 (1:0.63)
    PXPd7 7 (1:0.77) 1 (1:0.83) 3 (1:0.50)
    POPd2 0a,b 0 1 (1:0)
    PXPd2 2 (1:1.7)
    POPd6 0
    PXPd6 2 (1:2.5)
    POPd1 0 0 0
    POPd7 0 0
    Ph1-Phoxide 3 (1:0.59)
    aWhen the thiol was used, no reaction was observed.
    bOther thiols, such as HSCH2CH2OH, also did not react.
  • When aryl bromides were used to compare the reactivity of thiols vs. tin-thiolates, the thiols showed better reactivity but less selectivity, as in reactions with the iodides. The results are shown in Table B5.
  • TABLE B5
    The palladium-catalyzed coupling of aryl bromides with thiols or tin
    thiolates, using Scheme B2 with R3 = R4 = H, R5 = Me,
    R6 = H, R7 = Me, X = Br. General conditions: 150° C.,
    10 min, 300 W, 300 psi, substrate:thiolate = 1:1.5, in toluene.
    The results shown are percent yield of conversion, followed by
    ratio of substitution to reduction in parentheses.
    Ligand:
    R1; R2: BINAP BINAP Tol DPPF DiPPF
    R1 = SnMe3 6 (1:0)a 16 (1:0)b 0.4  14 (1:0)d
    R2 = CH2CH2OH
    R1 = SnMe3  26 (1:0)e
    R2 = CH2CONH2
    R1 = H 100 (1:0.06)
    R2 = CH2CH2OH
    R1 = H  26 (1:0.67)h
    R2 = CH2CONH2
    a0.1 equiv Pd2(dba)3 and 0.1 equiv L.
    b0.2 equiv Pd2(dba)3 and 0.2 equiv L.
    c0.1 equiv Pd2(dba)3 and 0.1 equiv L.
    d0.2 equiv Pd2(dba)3 and 0.4 equiv L.
    e0.2 equiv Pd2(dba)3 and 0.4 equiv L. General conditions for thiol reaction: 150° C., 10 min, 300 W, 300 p.s.i, 11.2 equiv t-BuOK;
    f0.2 equiv Pd(O.Ac)2 and 0.23 equiv L in dioxane,
    g0.2 equiv Pd2(dba)3 and 0.23 equiv L in toluene,
    h0.2 equiv Pd2(dba)3 and 0.23 equiv L in dioxane.
  • The combination of Pd2dba3 (dipalladium dibenzylideneacetone) as catalyst, DiPPF as ligand and tin thiolate as reagent was used to synthesize several aryl thiolates in moderate to high yield. The results are shown in Table B6.
  • TABLE B6
    The palladium-catalyzed synthesis of thioethers with Pd2dba3 as catalyst,
    DiPPF as ligand, and tin thiolate, using Scheme B2 with R1 = SnMe3,
    R3 = R4 = H, R7 = Me, X, R2, R5, and R6 are as shown in the table.
    General conditions: 150° C., 10 min, 300 W, 300 psi,
    substrate:thiolate = 1:1-1.5, in toluene. In Method C1, 0.1 equiv
    Pd2(dba)3 and 0.1 equiv DiPPF were used; in Method C2,
    0.2 equiv Pd2(dba)3 and 0.4 equiv DiPPF were used; in
    Method C3, 0.2 equiv Pd2(dba)3 and 0.2 equiv DiPPF were used;
    in Method C4, 0.2 equiv Pd2(dba)3 and 0.8 equiv DiPPF were used.
    The results shown are the isolated yield of the
    substitution product from reverse phase HPLC.
    X R5 R6 R2 Method Yield (%)
    I H H CH2CONH2 C1 78
    I H Me CH2CONH2 C2 85
    I H H CH2CH2OH C1 91
    I H Me CH2CH2OH C2 89
    I H Me CH2C6H4OCH3 C3 85a
    Br Me H CH2CONH2 C4 69
    Br Me H CH2CH2OH C4 69
    aWhen B2O3 was used with the thiol instead of tin thiolate, no reaction was observed.
  • When bromo and iodo groups were both present in the same compound, selective substitution of the iodo group was realized using the same reaction conditions developed above, using stoichiometric quantities of tin-thiolates. The results are shown in Table B7.
  • TABLE B7
    The palladium-catalyzed synthesis of thioethers with Pd2(dba)3 as
    catalyst, DiPPF as ligand, and tin thiolate, using Scheme B2 with
    R1 = SnMe3, R3R4 = CH2CH2, R5 = Br, R6 = Me, and
    R7 = Ac, R2 is as shown in the table. General conditions: 150° C.,
    10 min, 300 W, 300 psi, substrate:thiolate = 1:1, 0.1 equiv Pd2(dba)3,
    0.1 equiv DiPPF, in toluene. The results shown are the
    isolated yield of the substitution product from reverse phase HPLC.
    X R2 Yield (%)
    I CH2CONH2 60
    I CH2CH2OH 94
  • The process method for the rapid, selective and efficient substitution of bromo or iodo groups in aryl halides by tin-thiolates with microwave heating, as disclosed herein, is applicable to substrates with an easily reducible iodo group, in either the presence or absence of a bromo group. Lower reactivity and higher selectivity were observed as compared with those using thiols as reagents. The corresponding reactions under conventional heating did not generate any appreciable amount of products except for those without any heteroatoms in the substrates (no data shown).
  • Reference List No. 2
  • 1. Dickens, M. J.; Gilday, J. P.; Mowlem, T. J.; Widdowson, D. A. Tetrahedron 1991, 47, 8621-8634.
  • 2. Kondo, T.; Mitsudo, T. Chem.Rev. 2000, 100, 3205-3220.
  • 3. Baranano, D.; Mann, G.; Hartwig, J. F. Curr. Org. Chem. 1997, 1, 187-305.
  • 4. Baranano, D.; Hartwig, J. F. J. Am. Chem. Soc. 1995, 117, 2937-2938.
  • 5. Louie, J.; Hartwig, J. F. J. Am. Chem. Soc. 1995, 117, 11598-11599.
  • 6. Hartwig, J. F. Acc. Chem. Res. 1998, 31, 852-860.
  • 7. Murata, M.; Buchwald, S. L. Tetrahedron 2004, 60, 7397-7403.
  • 8. Li, G. Y. J. Org. Chem. 2002, 67, 3643-3650.
  • 9. Li, G. Y.; Zheng, G.; Noonan, A. F. J. Org. Chem. 2001, 66, 8677-8681.
  • 10. Li, G. Y. Angew.Chem.Int.Ed. 2001, 40, 1513-1516.
  • 11. Schopfer, U.; Schlapbach, A. Tetrahedron 2001, 57, 3069-3073.
  • 12. Itoh, T.; Mase, T. Organic Letters 2004, 6, 4587-4590.
  • 13. Kwong, F. Y.; Buchwald, S. L. Organic Letters 2002, 4, 3517-3520.
  • 14. Bates, C. G.; Gujadhur, R. K.; Venkataraman, D. Organic Letters 2002, 4, 2803-2806.
  • 15. Hickman, R. J. S.; Christie, B. J.; Guy, R. W.; White. T. J. Austr. J. Chem. 1985, 38, 899-904.
  • 16. Palomo, C.; Oiarbide, M.; Lopez, R.; Gomez-Bengoa, E. Tetrahedron Letters 2000, 41, 1283-1286.
  • 17. Wu, Y. J.; He, H. Synlett 2003, 1789-1790.
  • 18. Cai, L.; Chin, F. T.; Pike, V. W.; Toyama, H.; Liow, J. S.; Zoghbi, S. S.; Modell, K.; Briard, E.; Shetty, H. U.; Sinclair, K.; Donohue, S.; Tipre, D.; Kung, M. P.; Dagostin, C.; Widdowson, D. A.; Green, M.; Gao, W.; Herman, M. M.; Ichise, M.; Innis, R. B. J. Med Chem. 2004, 47, 2208-2218.
  • 19. Wolfe, J. P.; Wagaw, S.; Marcoux, J. -F.; Buchwald, S. L. Acc. Chem. Res 1998, 31, 805-818.
  • 20. Ishiyama, T.; Mori, M.; Suzuki, A.; Miyaura, N. J. Organomet. Chem. 1996, 525, 225-231.
  • 21. Tang, W.; Zhang; X. Chem. Rev. 2003, 103, 3029-3069.

Claims (10)

1-10. (canceled)
11. A compound having the formula
Figure US20140341812A1-20141120-C00020
wherein:
R11 is a member selected from the group consisting of C1-C6 alkyl, aryl- or halo-substituted C1-C6 alkyl, hydroxyl-substituted C1-C6 alkyl, and carbamoyl C1-C6alkyl;
R12 is a member selected from the group consisting of H, C1-C6 alkyl, halo-substituted C1-C6 alkyl, halo, and C1-C6 alkylthio, and R13 is a member selected from the group consisting of H and C1-C3 alkyl, or R12 and R13 are joined to form a thio-C1-C3 alkylene or alkylene linkage, and
R14 is a member selected from the group consisting of H and C1-C6 alkyl, and
X is CH or N.
12. The compound of claim 11 wherein R11 is a member selected from the group consisting of C1-C3 alkyl, fluoro-substituted C1-C3 alkyl, hydroxyl-substituted C1-C3 alkyl, and carbamoyl C1-C3 alkyl.
13. The compound of claim 11 wherein R11 is a member selected from the group consisting of methyl, ethyl, fluoromethyl, fluoroethyl, hydroxymethyl, hydroxyethyl, carbamoylmethyl, and carbamoylethyl.
14. The compound of claim 11 wherein R12 is a member selected from the group consisting of H, C1-C3 alkyl, chloro, bromo, chloro-substituted C1-C3alkyl, bromo-substituted C1-C3 alkyl, and C1-C3 alkylthio, and R 13 is a member selected from the group consisting of H and C1-C3 alkyl, or R 12 and R 13 are joined to form a thiomethylene or alkylene linkage.
15. The compound of claim 11 wherein R12 is a member selected from the group consisting of H, bromo, and methylthio, and R13 is a member selected from the group consisting of H and methyl, or R12 and R13 are joined to form a thiomethylene or alkylene linkage.
16. The compound of claim 11 wherein R14 is a member selected from the group consisting of H and methyl.
17. The compound of claim 11 wherein R11 is a member selected from the group consisting of C1-C3 alkyl, fluoro-substituted C1-C3 alkyl, hydroxyl-substituted C1-C3 alkyl, and carbamoyl C1-C3 alkyl; R12 is a member selected from the group consisting of H, C1-C3 alkyl, chloro, bromo, chloro-substituted C1-C3 alkyl, bromo-substituted C1-C3 alkyl, and C1-C3 alkylthio, and R 13 is a member selected from the group consisting of H and C1-C3 alkyl, or R 12 and R 13 are joined to form a thiomethylene or alkylene linkage; and R14 is a member selected from the group consisting of H and methyl.
18. A method for imaging amyloid deposits in a patient, said method comprising administering to said patient a compound of claim 11 and imaging portions of said patient where amyloid deposits occur when said patient is suffering from Alzheimer's disease.
19. A method for imaging amyloid deposits in a patient, said method comprising administering to said patient a compound of claim 11 and imaging portions of said patient where amyloid deposits occur when said patient is suffering from Alzheimer's disease.
US14/223,782 2006-04-21 2014-03-24 Beta-amyloid pet imaging agents Abandoned US20140341812A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/223,782 US20140341812A1 (en) 2006-04-21 2014-03-24 Beta-amyloid pet imaging agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US79380706P 2006-04-21 2006-04-21
PCT/US2007/066939 WO2007124345A2 (en) 2006-04-21 2007-04-19 Beta-amyloid pet imaging agents
US29334010A 2010-09-22 2010-09-22
US14/223,782 US20140341812A1 (en) 2006-04-21 2014-03-24 Beta-amyloid pet imaging agents

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2007/066939 Division WO2007124345A2 (en) 2006-04-21 2007-04-19 Beta-amyloid pet imaging agents
US12293340 Division 2010-09-22

Publications (1)

Publication Number Publication Date
US20140341812A1 true US20140341812A1 (en) 2014-11-20

Family

ID=38566135

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/293,340 Expired - Fee Related US8703096B2 (en) 2006-04-21 2007-04-19 Beta-amyloid PET imaging agents
US14/223,782 Abandoned US20140341812A1 (en) 2006-04-21 2014-03-24 Beta-amyloid pet imaging agents

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/293,340 Expired - Fee Related US8703096B2 (en) 2006-04-21 2007-04-19 Beta-amyloid PET imaging agents

Country Status (3)

Country Link
US (2) US8703096B2 (en)
EP (2) EP2021340B1 (en)
WO (1) WO2007124345A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI607756B (en) * 2015-12-30 2017-12-11 Neuboron Medtech Ltd Neutron capture treatment system for eliminating β amyloid

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2364597C1 (en) * 2007-12-14 2009-08-20 Андрей Александрович Иващенко HETEROCYCLIC INHIBITORS OF Hh-SYGNAL CASCADE, BASED ON THEM MEDICINAL COMPOSITIONS AND METHOD OF TREATING DISEASES INDUCED BY ABBARANT ACTIVITY OF Hh-SIGNAL SYSTEM
EP2501696B1 (en) 2009-10-15 2016-12-28 Guerbet Imaging agents and their use for the diagnostic in vivo of neurodegenerative diseases, notably alzheimer's disease and derivative diseases
EA028533B1 (en) 2011-05-27 2017-11-30 Пробиодруг Аг Radiolabelled glutaminyl cyclase inhibitors
WO2013040183A1 (en) * 2011-09-16 2013-03-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Beta-amyloid imaging agents, methods of manufacture, and methods of use thereof
CN102532119B (en) * 2011-12-08 2014-08-06 上海如絮生物科技有限公司 Benzothiazole compound and intermediate, preparation method and application thereof
US9850302B2 (en) 2013-07-12 2017-12-26 Prothena Biosciences Limited Antibodies that recognize IAPP
WO2015004633A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize islet-amyloid polypeptide (iapp)
RU2559889C1 (en) * 2014-08-12 2015-08-20 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Мордовский государственный университет им. Н.П. Огарёва" Method of obtaining 5-methoxypyridinamine-2
TWI718122B (en) 2015-01-28 2021-02-11 愛爾蘭商普羅佘納生物科技有限公司 Anti-transthyretin antibodies
TWI711631B (en) 2015-01-28 2020-12-01 愛爾蘭商普羅佘納生物科技有限公司 Anti-transthyretin antibodies
TWI718121B (en) 2015-01-28 2021-02-11 愛爾蘭商普羅佘納生物科技有限公司 Anti-transthyretin antibodies
US9926316B2 (en) * 2016-03-03 2018-03-27 The Cleveland Clinic Foundation Antitumor derivatives for differentiation therapy
JP7017013B2 (en) 2016-07-02 2022-02-08 プロセナ バイオサイエンシーズ リミテッド Anti-transthyretin antibody
WO2018007923A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
JP7076711B2 (en) 2016-07-02 2022-05-30 プロセナ バイオサイエンシーズ リミテッド Anti-transthyretin antibody
CN107629029A (en) * 2017-09-19 2018-01-26 安徽省诚联医药科技有限公司 The preparation method of Wei Patawei intermediates

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040209892A1 (en) * 2003-03-21 2004-10-21 Di Pietro Lucian V. Heterocyclic compounds and methods of use

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI77852C (en) 1981-02-17 1989-05-10 Otsuka Pharma Co Ltd Process for the preparation of novel, such as cardiac drugs, useful s unsubstituted amide and (saturated heterocycle) carbonyl carbostyril derivatives.
US4721721A (en) 1984-12-18 1988-01-26 Rorer Pharmaceutical Corporation 6-(4-thiazole) compounds, cardiotonic compositions including the same, and their uses
DE3446778A1 (en) * 1984-12-21 1986-07-03 Dr. Karl Thomae Gmbh, 7950 Biberach NEW IMIDAZO DERIVATIVES, THEIR PRODUCTION AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
CN1041198C (en) 1995-04-03 1998-12-16 北京农业大学 Method for synthesizing 5-[1-hydroxy-2-(isopropylamino)ethyl] aminobenzonitrile
CN1143624A (en) 1995-08-24 1997-02-26 胡钊侠 Process for synthesizing ketoprofen
GB9919673D0 (en) * 1999-08-20 1999-10-20 Cancer Res Campaign Tech 2-Arlybenzazole compounds
JP2001302639A (en) 2000-04-28 2001-10-31 Nippon Fine Chem Co Ltd Method for producing aminocyanopyridine
WO2004080391A2 (en) 2003-03-10 2004-09-23 Optimer Pharmaceuticals, Inc. Novel antibacterial agents
US20080219922A1 (en) * 2005-09-12 2008-09-11 Goodman Mark M Alzheimer's Disease Imaging Agents

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040209892A1 (en) * 2003-03-21 2004-10-21 Di Pietro Lucian V. Heterocyclic compounds and methods of use

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI607756B (en) * 2015-12-30 2017-12-11 Neuboron Medtech Ltd Neutron capture treatment system for eliminating β amyloid
EP3384960A4 (en) * 2015-12-30 2018-12-19 Neuboron Medtech Ltd. NEUTRON CAPTURE THERAPY SYSTEM FOR ELIMINATING AMYLOID ß-PROTEIN
US10709783B2 (en) 2015-12-30 2020-07-14 Neuboron Medtech Ltd. Neutron capture therapy system for eliminating amyloid β-protein

Also Published As

Publication number Publication date
WO2007124345A2 (en) 2007-11-01
EP2808020A1 (en) 2014-12-03
EP2021340A2 (en) 2009-02-11
EP2021340B1 (en) 2014-07-23
US20110008255A1 (en) 2011-01-13
WO2007124345A3 (en) 2008-03-27
US8703096B2 (en) 2014-04-22

Similar Documents

Publication Publication Date Title
US8703096B2 (en) Beta-amyloid PET imaging agents
CN103739605B (en) For detecting the developer of neurological disorder
Cai et al. Synthesis and Evaluation of Two 18F-Labeled 6-Iodo-2-(4 ‘-N, N-dimethylamino) phenylimidazo [1, 2-a] pyridine Derivatives as Prospective Radioligands for β-Amyloid in Alzheimer's Disease
EP2501696B1 (en) Imaging agents and their use for the diagnostic in vivo of neurodegenerative diseases, notably alzheimer's disease and derivative diseases
Cui et al. Synthesis and evaluation of novel benzothiazole derivatives based on the bithiophene structure as potential radiotracers for β-amyloid plaques in Alzheimer’s disease
Yang et al. Radiolabeled bioactive benzoheterocycles for imaging β-amyloid plaques in Alzheimer's disease
JP2011506487A (en) Translocator protein ligand
WO2007033080A2 (en) Alzheimer's disease imaging agents
JPWO2009054496A1 (en) Novel amyloid affinity compound
KR20140040722A (en) Novel compound having affinity for amyloid
Matsumura et al. Synthesis and biological evaluation of novel styryl benzimidazole derivatives as probes for imaging of neurofibrillary tangles in Alzheimer’s disease
Neumaier et al. Synthesis and evaluation of 18F-fluoroethylated benzothiazole derivatives for in vivo imaging of amyloid plaques in Alzheimer's disease
Maisonial et al. Single photon emission computed tomography/positron emission tomography imaging and targeted radionuclide therapy of melanoma: new multimodal fluorinated and iodinated radiotracers
KR20190031312A (en) Compounds for imaging tau protein aggregates
Cui et al. Novel (E)-5-styryl-2, 2′-bithiophene derivatives as ligands for β-amyloid plaques
CN111712265A (en) Diagnostic composition for PET imaging, method for the preparation thereof and use thereof in diagnostics
Fuchigami et al. Synthesis and biological evaluation of radioiodinated quinacrine-based derivatives for SPECT imaging of Aβ plaques
KR20100089858A (en) Use of novel compound having affinity for amyloid, and process for production of the same
JPWO2008059714A1 (en) Novel amyloid affinity compound
KR20200113234A (en) A novel method of preparing an imaging compound
JPWO2009057577A1 (en) Use and production methods of novel amyloid affinity compounds
TWI450728B (en) Use of 7-chloro-n,n,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4h-pyridazino[4,5-b]indole-1-acetamide as a biomarker of peripheral benzodiazepine receptor levels
WO2013040183A1 (en) Beta-amyloid imaging agents, methods of manufacture, and methods of use thereof
JP7284490B2 (en) Monoamine oxidase B imaging probe
JP5247442B2 (en) Novel amyloid affinity compound

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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