WO2000076489A2 - Method of inhibiting amyloid protein aggregation and imaging amyloid deposits - Google Patents

Method of inhibiting amyloid protein aggregation and imaging amyloid deposits Download PDF

Info

Publication number
WO2000076489A2
WO2000076489A2 PCT/US2000/015071 US0015071W WO0076489A2 WO 2000076489 A2 WO2000076489 A2 WO 2000076489A2 US 0015071 W US0015071 W US 0015071W WO 0076489 A2 WO0076489 A2 WO 0076489A2
Authority
WO
WIPO (PCT)
Prior art keywords
phenylamino
phenyl
benzoic acid
ethyl
dichloro
Prior art date
Application number
PCT/US2000/015071
Other languages
French (fr)
Other versions
WO2000076489A3 (en
Inventor
Corinne Elizabeth Augelli-Szafran
Mark Robert Barvian
Christopher Franklin Bigge
Shelly Ann Glase
Shunichiro Hachiya
John Steven Keily
Takenori Kimura
Yingjie Lai
Annette Theresa Sakkab
Mark James Suto
Lary Craswell Walker
Tomoyuki Yasunaga
Nian Zhuang
Original Assignee
Warner-Lambert Company
Yamanouchi Pharmaceutical Company, Ltd.
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
Priority to EA200101135A priority Critical patent/EA004632B1/en
Priority to DZ003252A priority patent/DZ3252A1/fr
Application filed by Warner-Lambert Company, Yamanouchi Pharmaceutical Company, Ltd. filed Critical Warner-Lambert Company
Priority to NZ515621A priority patent/NZ515621A/en
Priority to PL00352430A priority patent/PL352430A1/en
Priority to CA002375551A priority patent/CA2375551A1/en
Priority to MXPA01012318A priority patent/MXPA01012318A/en
Priority to EP00939471A priority patent/EP1225886A2/en
Priority to HU0202508A priority patent/HUP0202508A3/en
Priority to AU54553/00A priority patent/AU775157B2/en
Priority to IL14697100A priority patent/IL146971A0/en
Priority to BR0011728-5A priority patent/BR0011728A/en
Priority to EEP200100673A priority patent/EE200100673A/en
Priority to US10/009,611 priority patent/US6972287B1/en
Priority to SK1763-2001A priority patent/SK17632001A3/en
Priority to KR1020017015879A priority patent/KR20020008224A/en
Priority to JP2001502823A priority patent/JP2003504310A/en
Priority to APAP/P/2002/002387A priority patent/AP2002002387A0/en
Publication of WO2000076489A2 publication Critical patent/WO2000076489A2/en
Priority to IS6193A priority patent/IS6193A/en
Priority to NO20015995A priority patent/NO20015995L/en
Priority to BG106293A priority patent/BG106293A/en
Priority to HR20020026A priority patent/HRP20020026A2/en
Publication of WO2000076489A3 publication Critical patent/WO2000076489A3/en
Priority to HK03100404.7A priority patent/HK1048258A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4021-aryl substituted, e.g. piretanide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/52Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C229/54Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C229/56Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino and carboxyl groups bound in ortho-position
    • C07C229/58Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino and carboxyl groups bound in ortho-position having the nitrogen atom of at least one of the amino groups further bound to a carbon atom of a six-membered aromatic ring, e.g. N-phenyl-anthranilic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/52Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C229/54Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C229/62Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring with amino groups and at least two carboxyl groups bound to carbon atoms of the same six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/52Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C229/54Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C229/64Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton with amino and carboxyl groups bound to carbon atoms of the same non-condensed six-membered aromatic ring the carbon skeleton being further substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/34Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups
    • C07C233/42Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
    • C07C233/43Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of a saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/50Compounds containing any of the groups, X being a hetero atom, Y being any atom
    • C07C311/51Y being a hydrogen or a carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/88Nitrogen atoms, e.g. allantoin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/135Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/50Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to atoms of the carbocyclic ring
    • C07D317/58Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • This invention relates to a method of inhibiting amyloid protein aggregation and imaging amyloid deposits. More particularly, this invention relates to a method of inhibiting amyloid protein aggregation in order to treat Alzheimer's disease.
  • Amyloidosis is a condition characterized by the accumulation of various insoluble, fibrillar proteins in the tissues of a patient.
  • the fibrillar proteins that comprise the accumulations or deposits are called amyloid proteins. While the particular proteins or peptides found in the deposits vary, the presence of fibrillar morphology and a large amount of ⁇ -sheet secondary structure is common to many types of amyloids.
  • An amyloid deposit is formed by the aggregation of amyloid proteins, followed by the further combination of aggregates and/or amyloid proteins.
  • amyloid deposits has been shown in various diseases, each with its particular associated protein, such as Mediterranean fever, Muckle- Wells syndrome, idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, amyloid polyneuropathy, hereditary cerebral hemorrhage with amyloidosis, Alzheimer's disease, Down's syndrome, Scrapie, Creutzfeldt- Jacob disease, Kuru, Gerstmann-Straussler- Scheinker syndrome, medullary carcinoma of the thyroid, Isolated atrial amyloid, ⁇ 2-microglobulin amyloid in dialysis patients, inclusion body myositis, ⁇ 2-amyloid deposits in muscle wasting disease, Sickle Cell Anemia, Parkinson's
  • Alzheimer's disease is a degenerative brain disorder characterized clinically by progressive loss of memory, cognition, reasoning, judgement, and emotional stability that gradually leads to mental deterioration and ultimately death. Because Alzheimer's disease and related degenerative brain disorders are a major medical issue for an increasingly aging population, the need for new treatments and methods for diagnosing the disorders are needed.
  • a simple, noninvasive method for detecting and quantitating amyloid deposits in a patient has been eagerly sought.
  • detection of amyloid deposits involves histological analysis of biopsy or autopsy materials. Both methods have major drawbacks.
  • an autopsy can only be used for a postmortem diagnosis.
  • amyloid deposits in vivo are difficult, as the deposits have many of the same physical properties (i.e., density and water content) as normal tissues. Attempts to image amyloid deposits directly using magnetic resonance imaging (MRI) and computer-assisted tomography (CAT) have been disappointing and have detected amyloid deposits only under certain favorable conditions. In addition, efforts to label amyloid deposits with antibodies, serum amyloid P protein, or other probe molecules has provided some selectivity on the periphery of tissues, but has provided for poor imaging of tissue interiors. Thus, it would be useful to have a noninvasive technique for imaging and quantitating amyloid deposits in a patient. In addition, it would be useful to have compounds that inhibit the aggregation of amyloid proteins to form amyloid deposits.
  • MRI magnetic resonance imaging
  • CAT computer-assisted tomography
  • the present invention provides a method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I wherein
  • R a is hydrogen, C ⁇ -Cg alkyl, or -CC ⁇ -C ⁇ alkyl; n is 0 to 5 inclusive;
  • Rl, R ⁇ , R3, R4 5 R5 5 R6 ? and R7 r e independently hydrogen, halogen
  • -CH CH-phenyl, -O(CH 2 ) p NR b R c , -CNR b R c , -NHCR b , -NH(CH 2 ) p NR b RC, -N(C 1 -C 6 alkyi ⁇ CH 2 ) p NR b RC,
  • R 8 is COOH, tetrazolyl, -SO 2 R d , or -CONHSO 2 R d ;
  • R b and R c are independently hydrogen, -C ⁇ -C alkyl, -(CH2) m -phenyl, or
  • R D and R c taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomo holino, decahydroisoquinoline, or pyrazolyl;
  • R d is hydrogen, -C ⁇ -C alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive; A is CH orN; R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
  • the n group is attached at the 4-position of the phenyl ring.
  • R a is hydrogen; n is 2;
  • R3 and R 4 are hydrogen.
  • R a is hydrogen
  • R! is halo
  • R2 is hydrogen or halo
  • R3, R 4 , R5. and R ⁇ are hydrogen
  • n is 2 to 5 inclusive.
  • R a is hydrogen; n is 2 or 3;
  • R 1 is -NR b R c ;
  • R 2 , R 3 , R 4 , R5, and R 7 all are hydrogen.
  • R 2 , R 3 , R 4 , R5, and R 7 all are hydrogen.
  • R a is hydrogen; n is 2;
  • R3 and R4 are hydrogen
  • R a is hydrogen; n is 3, 4, or 5;
  • R3 and R 4 are hydrogen
  • Rl, R 2 , and R 7 are independently chlorine or hydrogen.
  • R a is hydrogen; n is 2;
  • R3 and R 4 are hydrogen
  • R 5 , R 6 , and R 8 are independently hydrogen, -CO2H, -NO2, -OCH3, O
  • R a is hydrogen; n is 2;
  • R and R 4 are hydrogen; and R 5 is -CO 2 H. Also preferred is a method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I
  • R a is hydrogen; n is 1 to 5 inclusive;
  • R3 and R4 are hydrogen
  • R 1 , R 7 , and R 2 are independently chlorine, -N(CH2CH3) 2 , -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH 2 phenyl,
  • R 5 and R 6 are independently hydrogen, -CO2H, -NO 2 , -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; or the pharmaceutically acceptable salts thereof.
  • compounds of Formula I are 2- ⁇ 4-[2-(3 ,4-Dichlorophenyl)ethyl]phenylamino ⁇ -benzoic acid; 2- ⁇ 4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino ⁇ -5-nitrobenzoic acid;
  • the invention also provides the foregoing compounds wherein the benzoic acid portion is replaced with a pyridyl carboxylic acid, for example, 4-[4-(3,4- dichlorophenyl)phenylamino]-3-hydroxycarbonylpyridine.
  • a pyridyl carboxylic acid for example, 4-[4-(3,4- dichlorophenyl)phenylamino]-3-hydroxycarbonylpyridine.
  • R a is hydrogen, Cj-Cg alkyl, or -CCj-C alkyl; n is 0 to 5 inclusive;
  • Rl, R 2 , R3, R4, R5 5 R6 5 and R? are independently hydrogen, halogen,
  • -CH CH-phenyl, -O(CH 2 ) p NR R c , -CNR b R c , -NHCR b , -NH(CH 2 ) p NR b R C , -N(C j -C 6 alkyl)(CH 2 ) p NR b R c ,
  • R8 is COOH, tetrazolyl, -SO R d or -CONHSO 2 R d ;
  • R and R c are independently hydrogen, -Cj-Cg alkyl, -(CH 2 ) m -phenyl, or
  • R b and R c taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
  • R d is hydrogen, -Cj-Cg alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive;
  • A is CH orN
  • R! and R 2 when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
  • R a is hydrogen; n is 2; and
  • R3 and R4 are hydrogen.
  • R a is hydrogen; R3 and R are hydrogen; and n is 2 to 5 inclusive.
  • R a is hydrogen; n is 3, 4, or 5; R3 and R 4 are hydrogen; and
  • Rl, R 2 , and R 7 are independently chlorine or hydrogen.
  • R a is hydrogen; n is 2; R3 and R 4 are hydrogen; and
  • R 5 and R 6 are independently hydrogen, -CO 2 H, -NO 2 , -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, halogen,
  • R a is hydrogen; n is 2;
  • R3 and R 4 are hydrogen
  • R 5 is -CO2H.
  • R a is hydrogen; n is 1 to 5 inclusive;
  • R3 and R 4 are hydrogen
  • R 5 and R 6 are independently hydrogen, -CO2H, -NO 2 , -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; R 8 is COOH or tetrazolyl; or the pharmaceutically acceptable salts thereof.
  • R! is halo
  • R 2 is H or halo; and n and R ⁇ are as defined above in Formula I.
  • R! is halo
  • R 2 is H or halo; and n and R" are as defined above in Formula I.
  • R! is halo
  • R 2 is H or halo; and n and R ⁇ are as defined above in Formula I.
  • the present invention also provides the compounds:
  • Typical 2- and 3-substituted compounds are:
  • compositions of the novel compounds admixed with a pharmaceutically acceptable diluent, carrier, or excipient are also provided.
  • Also provided is a method of imaging amyloid deposits comprising: introducing into a patient a detectable quantity of a labeled compound having the Formula I or a pharmaceutically acceptable salt thereof:
  • R a is hydrogen, C ⁇ -C6 alkyl, or -CCi-Cg alkyl; n is 0 to 5 inclusive;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and R 7 are independently hydrogen, halogen, -OH, -NH 2 , NR b R C , -CO 2 H, -CO 2 C!-C 6 alkyl, -
  • -CH CH-phenyl, -O(CH 2 ) p NR b R c , -CNR b R c , -NHCR b , -NH(CH 2 ) p NR b R C , -N(C ] -C 6 alkyl)(CH 2 ) p NR b R c ,
  • R 8 is COOH, tetrazolyl, -SO 2 R d , or -CONHSO 2 R d ;
  • R and R c are independently hydrogen, -Cj-Cg alkyl, -(CH 2 ) m - phenyl, or R b and R c taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-C ⁇ -Cg alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
  • R d is hydrogen, -Ci-Cg alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive;
  • A is CH or N
  • R! and R 2 when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof; b. allowing sufficient time for the labeled compound to become associated with amyloid deposits; and c. detecting the labeled compound associated with the amyloid deposits.
  • the patient has or is suspected to have Alzheimer's disease.
  • the labeled compound is a radio labeled compound.
  • the labeled compound is detected using MRI.
  • the present invention also provides the preferred compounds: 2- ⁇ 4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino ⁇ -benzoic acid;
  • alkyl means a straight or branched chain hydrocarbon having from 1 to 12 carbon atoms.
  • Representative examples of alkyl groups are methyl, ethyl, propyl, isopropyl, isobutyl, butyl, tert-butyl, sec-butyl, pentyl, hexyl, octyl, decyl, and 1,1-dimethyloctyl.
  • Preferred alkyl groups are Cj-Cg alkyl, and especially C1-C6 alkyl.
  • alkoxy means an alkyl group attached to an oxygen atom.
  • Representative examples of alkoxy groups include methoxy, ethoxy, tert-butoxy, propoxy, and isobutoxy.
  • Preferred alkoxy groups are Cj-C ⁇ alkoxy, and especially C ⁇ -C alkoxy.
  • halogen includes chlorine, fluorine, bromine, and iodine.
  • substituted means that one or more hydrogen atom in a molecule has been replaced with another atom or group of atoms.
  • substituents include halogen, especially chloro, -OH, -CF3, -NO2, -NH 2 , -NH(C!-C 6 alkyl), -N(C!-C 6 alkyl) 2 , C ⁇ Cg alkyl, -OC!-C 6 alkyl, -CN, -CF3,
  • substituted phenyl means a phenyl ring in which from 1 to 4 hydrogen atoms have been independently replaced with a substituent, preferably one selected from the list above.
  • Typical "substituted phenyl” groups include 4-chlorophenyl, 3,4-dibromophenyl, 3-fluoro-4-methylphenyl,
  • Substituent groups represented by R , R 3 , and R ⁇ include amino(NR b R c ) and acylamino (-NHCOR b ).
  • R b and R c can be hydrogen, alkyl and phenylalkyl and substituted phenylalkyl, and typical NR b R c groups include methylamino, diethylamino, isobutyl-propylamino, benzylamino, and 3,4- dimethoxybenzylamino.
  • Examples of acylamino groups include formamido, acetamido, 2-phenylacetamido, and 2-(3-nitrophenyl)acetamido.
  • R ⁇ , R 3 , and R5 can also be aminoalkoxy (-O(CH2) p NR b R c ) such as N-methylaminomethoxy and 2-(N-benzylamino)ethoxy, as well as aminoalkylamino (-NH(CH2) p NR b R c ) such as 3-(dimethylamino)propylamino and 2-(N-ethyl-N-benzylamino)ethylamino.
  • Substituent groups such as Rl, R 3 , and R ⁇ additionally can be cyclic structures, for instance when NR b R c is part of the substituent group, and R b and R c are taken together with the nitrogen to which they are attached to form a cyclic ring selected from imidazole, pyrrole, piperidine, piperazine, 4-Cj-C6 alkylpiperazine, morpholine, thiomorpholine, pyrazole, and decahydroisoquinoline.
  • benzyl such as 2-trifluoromethylbenzyloxy and 4-aminobenzyloxy.
  • pharmaceutically acceptable salt, ester, amide, and prodrug refers to those carboxylate salts, amino acid addition salts, esters, amides, and prodrugs of the compounds of the present invention which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • salts refers to the relatively nontoxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laureate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate and laurylsulphonate salts, and the like.
  • alkali and alkaline earth metals such as sodium, lithium, potassium, calcium, magnesium, and the like
  • nontoxic ammonium, quaternary ammonium and amine cations including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like See, for example, Berge S.M., et al., Pharmaceutical Salts, J. Pharm. Sci., 66:1-19 (1977) which is incorporated herein by reference.
  • esters of the compounds of this invention examples include Cj-C6 alkyl esters wherein the alkyl group is a straight or branched chain. Acceptable esters also include C5-C 7 cycloalkyl esters as well as arylalkyl esters such as, but not limited to benzyl. C1-C4 alkyl esters are preferred. Esters of the compounds of the present invention may be prepared according to conventional methods, for example by reacting a carboxylic acid of Formula I with an alcohol such as ethanol or benzyl alcohol.
  • Examples of pharmaceutically acceptable, nontoxic amides of the compounds of this invention include amides derived from ammonia, primary C ⁇ -C alkyl amines and secondary C ⁇ -C dialkyl amines wherein the alkyl groups are straight or branched chain. In the case of secondary amines, the amine may also be in the form of a 5- or 6-membered heterocycle containing one nitrogen atom. Amides derived from ammonia, C1-C3 alkyl primary amides and
  • C1-C2 dialkyl secondary amides are preferred.
  • Amides of the compounds of the invention may be prepared according to conventional methods.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formulas, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • the compounds of the present invention can exist in different stereoisometric forms by virtue of the presence of asymmetric centers in the compounds. It is contemplated that all stereoisometric forms of the compounds, as well as mixture thereof, including racemic mixtures, form part of this invention.
  • a labeled compound of Formula I is introduced into a tissue or a patient in a detectable quantity.
  • the compound is typically part of a pharmaceutical composition and is administered to the tissue or the patient by methods well-known to those skilled in the art.
  • a compound in the methods of the present invention, can be administered either orally, rectally, parenterally (intravenous, by intramuscularly or subcutaneously), intracisternally, intravaginally, intraperitoneally, intravesically, locally (powders, ointments or drops), or as a buccal or nasal spray.
  • Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
  • a coating such as lecithin
  • surfactants for example, water, alcohol, alcohol, and the like.
  • compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispensing agents.
  • adjuvants such as preserving, wetting, emulsifying, and dispensing agents.
  • Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • isotonic agents for example sugars, sodium chloride, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid;
  • binders as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia;
  • humectants as for example, glycerol;
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate;
  • solution retarders as for example paraffin;
  • absorption accelerators as for example, quaternary ammonium compounds;
  • wetting agents such as sodium citrate or dicalcium phosphate
  • lubricants as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof.
  • the dosage forms may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft- and hard-filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethyleneglycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain opacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions which can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3 -butyl eneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan or mixtures of these substances, and the like.
  • inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, eth
  • the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administrations are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable nonirritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • suitable nonirritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants.
  • the active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers or propellants as may be required.
  • Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
  • the labeled compound is introduced into a patient in a detectable quantity and after sufficient time has passed for the compound to become associated with amyloid deposits, the labeled compound is detected noninvasively inside the patient.
  • a labeled compound of Formula I is introduced into a patient, sufficient time is allowed for the compound to become associated with amyloid deposits, and then a sample of tissue from the patient is removed and the labeled compound in the tissue is detected apart from the patient.
  • a tissue sample is removed from a patient and a labeled compound of Formula I is introduced into the tissue sample. After a sufficient amount of time for the compound to become bound to amyloid deposits, the compound is detected.
  • the administration of the labeled compound to a patient can be by a general or local administration route.
  • the labeled compound may be administered to the patient such that it is delivered throughout the body.
  • the labeled compound can be administered to a specific organ or tissue of interest. For example, it is desirable to locate and quantitate amyloid deposits in the brain in order to diagnose or track the progress of Alzheimer's disease in a patient.
  • tissue means a part of a patient's body. Examples of tissues include the brain, heart, liver, blood vessels, and arteries.
  • a detectable quantity is a quantity of labeled compound necessary to be detected by the detection method chosen. The amount of a labeled compound to be introduced into a patient in order to provide for detection can readily be determined by those skilled in the art. For example, increasing amounts of the labeled compound can be given to a patient until the compound is detected by the detection method of choice. A label is introduced into the compounds to provide for detection of the compounds.
  • patient means humans and other animals. Those skilled in the art are also familiar with determining the amount of time sufficient for a compound to become associated with amyloid deposits. The amount of time necessary can easily be determined by introducing a detectable amount of a labeled compound of Formula I into a patient and then detecting the labeled compound at various times after administration.
  • association means a chemical interaction between the labeled compound and the amyloid deposit. Examples of associations include covalent bonds, ionic bonds, hydrophilic-hydrophilic interactions, hydrophobic- hydrophobic interactions, and complexes.
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • the label that is introduced into the compound will depend on the detection method desired. For example, if PET is selected as a detection method, the compound must possess a positron-emitting atom, such as ⁇ C or 1 8 F.
  • a suitable label in a compound of Formula I is an atom such as 1 3 C, l ⁇ N, 0 r l" which can be detected using magnetic resonance imaging (MRI) which is also sometimes called nuclear magnetic resonance
  • MRI magnetic resonance imaging
  • the labeled compounds of Formula I may also be detected by MRI using paramagnetic contrast agents.
  • Another example of detection is electron paramagnetic resonance (EPR).
  • EPR electron paramagnetic resonance
  • EPR probes which are well-known in the art, such as nitroxides, can be used.
  • the imaging of amyloid deposits can also be carried out quantitatively so that the amount of amyloid deposits can be determined.
  • the present invention also provides a method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, by administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein inhibiting amount of a compound of Formula I.
  • an amyloid inhibiting amount by simply administering a compound of Formula I to a patient in increasing amounts until the growth of amyloid deposits is decreased or stopped. The rate of growth can be assessed using imaging or by taking a tissue sample from a patient and observing the amyloid deposits therein.
  • a patient in need of inhibition of the aggregation of amyloid proteins is a patient having a disease or condition in which amyloid proteins aggregate.
  • diseases and conditions include Mediterranean fever, Muckle- Wells syndrome, idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, amyloid polyneuropathy, hereditary cerebral hemorrhage with amyloidosis, Alzheimer's disease, Down's syndrome,
  • the radioisotope can be any radioisotope. However, 3 H, 123 L 125 ⁇ 131 T ; 11 and l 8 F are preferred. Those skilled in the art are familiar with the procedure used to introduce a radioisotope into a compound. For example, a compound of Formula I wherein one carbon atom is ⁇ C or ⁇ C is readily prepared.
  • the compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1 ,000 mg per day. For a normal human adult having a body weight of about 70 kg, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is sufficient. The specific dosage used, however, can vary.
  • the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used.
  • the determination of optimum dosages for a particular patient is well- known to those skilled in the art.
  • the examples presented below are intended to illustrate particular embodiments of the invention and are not intended to limit the scope of the specification, including the claims, in any manner.
  • Schemes 6 through 9 show synthetic routes that can be used to obtain the desired starting amines (IV), (VIII), (XV), and (XXI).
  • Scheme 2 depicts the synthesis of amine (NIII) which contains a three methylene tether. Condensation of aldehyde (I) and nitro-ketone (N) in the presence of sodium hydroxide gives the desired alpha, beta-unsaturated ketone, which upon standard hydrogenation conditions (Raney nickel) gives (Nil) and then Wolff-Kishner conditions yields the desired amine (VIII).
  • Scheme 3 is very similar to Scheme 2, except that the aldehyde (I) is condensed with a substituted aniline (IX).
  • Scheme 4 illustrates standard Wittig conditions in which the starting materials (XII) and (XIII) are obtained via aldol condensation and ylide chemistry, respectively.
  • Standard reduction conditions e.g., Raney nickel
  • (XIN) yields the desired amine (XV).
  • Scheme 5 illustrates the synthesis of amine (XXI) which contains a 5-methylene tether. Wittig reaction of the bromophosporane (XVII), which is formed from the corresponding substituted bromide (XVI), and nitro aldehyde
  • Scheme 6 illustrates one route to obtain compounds of Formula I. Either by Buchwald coupling (Method A) followed by saponification or utilizing the
  • Protecting groups will also be used when reactive functional groups such as amino and carboxylic acids are present, so as to avoid unwanted side reactions.
  • Carboxy groups typically are converted to esters (e.g., tert-butyl, benzyl), and amino groups generally are acylated (e.g., acetyl or trimethylsilyl).
  • esters e.g., tert-butyl, benzyl
  • amino groups generally are acylated (e.g., acetyl or trimethylsilyl).
  • Scheme 7 illustrates the synthesis of compounds of Formula I by reacting amines such as (IN), (NIII), and (XXI) with fluoro-nitro intermediate (XXIN), in the presence of a base (e.g., LHMDS or Et3 ⁇ ) to give ester (XXV).
  • a base e.g., LHMDS or Et3 ⁇
  • ester can then be saponified using standard conditions, such as sodium hydroxide.
  • amine (XV) can be coupled with readily available fluoro- substituted carboxylic acids [e.g., (XXVI) or (XXNII)] in the presence of various bases (such as DBU or triethylamine) to yield compounds of Formula I.
  • fluoro- substituted carboxylic acids e.g., (XXVI) or (XXNII)
  • bases such as DBU or triethylamine
  • Scheme 9 depicts coupling of amine (VIII) with readily available methyl ester (XXVIII) in the presence of a base, such as imidazole, to give ester (XXIX). This ester can then be saponified as usual to give compounds of Formula I.
  • Scheme 10 illustrates the synthesis of fluoro-intermediate (XXIV) which is obtained by nitration of readily available methyl ester (XXX) to give (XXVIII). Treatment of (XXVIII) with potassium cyanide gives (XXIV).
  • Scheme 11 the synthesis of compounds related to Example 18 is illustrated. Reaction of the potassium salts of ortho-substituted benzoic acids (XXVI) with substituted anilines (XXVII) in the presence of potassium carbonate and cupric acetate yields various iodo-substituted aminobenzoic acids (XXVIII). Reaction of (XXVIII) with substituted boronic acids and palladium chloride gives the desired substituted aminobenzoic acids (XXX).
  • the benzoic acids are readily converted to esters and amides, as well as salts and other prodrugs by routine processes.
  • the benzoic acid can be reacted with oxalylchloride to form the acid chloride, which then readily reacts with a sulfonamide such as methanesulfonamide to produce the corresponding invention compound where R 8 is -CONHSO 2 CH3.
  • a sulfonamide such as methanesulfonamide
  • R is an ester forming group such as alkyl or benzyl.
  • Step A (Scheme 1): Preparation of l,2-Dichloro-4-[2-(4-nitrophenyl)ethenyl]- benzene A mixture ofp-nitrophenylacetic acid (51.23 g, 0.28 mol) and
  • Step B (Scheme 1): Preparation of 4-[2-(3,4-Dichlorophenyl)ethyl]benzenamine A sample of l,2-dichloro-4-[2-(4-nitrophenyl)ethenyl]benzene (98.0 g,
  • Step C (Scheme 6): Preparation of 2- ⁇ 4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino ⁇ -benzoic acid
  • Example 1 An orange solid (31.95 g, 0.083 mol, 77.6%). mp 175.0-177.0°C.
  • Example 1 An orange solid (2.3 g, 0.006 mol, 17.5%). mp 165.0-173.0°C. Analysis for C2iH 17 N 1 O 2 Cl2: Calcd: C, 65.30; H, 4.44; N, 3.63.
  • Step C (Scheme 6): Preparation of 2- ⁇ 4-[2-(3,4-Dichloro-phenyl)- ethyl]phenylamino ⁇ -5-nitrobenzoic acid methyl ester
  • Step A (Scheme 1): Preparation of l,2-Dimethoxy-4-[2-(4-nitrophenyl)ethenyl]- benzene The title compound was prepared from /.-nitrophenylacetic acid (25.0 g,
  • Step B (Scheme 1): Preparation of 4-[2-(3,4-Dimethoxy-phenyl)ethyl]- phenylamine l,2-Dimethoxy-4-[2-(4-nitrophenyl)ethenyl]benzene (12.1 g, 0.042 mol) was reduced in the presence of 10% Pd-C (2.0 g) in dimethylformamide (DMF) (120 mL) at 25°C under a hydrogen atmosphere. The reaction mixture was concentrated in vacuo to give a solid. The solid was recrystallized from MeOH (400 mL) to yield a white crystalline product, 6.8 g (0.026 mol, 63%) of the desired product, mp 115-116°C.
  • DMF dimethylformamide
  • Step C (Scheme 6): Preparation of 2- ⁇ 4-[2-(3,4-Dimethoxy-phenyl)ethyl]- phenylamino ⁇ benzoic acid
  • the title compound was prepared from 4-[2-(3,4-dimethoxy-phenyl)- ethyl]phenylamine (9.25 g, 0.036 mol), 2-chlorobenzoic acid (5.2 g, 0.036 mol), anhydrous potassium carbonate (15.0 g, 0.11 mol), copper powder (0.45 g,
  • Step D Preparation of 2- ⁇ 4-[2-(3,4-Dihydroxy-phenyl)-ethyl]-phenylamino ⁇ - benzoic acid
  • Step A (Scheme 1): Preparation of l,l-Dibutylamino-4-[2-(4- nitrophenyl)ethenyl]benzene
  • Step B (Scheme 1): Preparation of 4-[2-(4,4-Dibutylaminophenyl)ethyl]- phenylamine
  • Step C (Scheme 6): Preparation of 2- ⁇ 4-[2-(4-Dibutylamino-phenyl)- ethyljphenylamino ⁇ -benzoic acid
  • Step A (Scheme 1): Preparation of l,2,3-Trimethoxy-5-[2-(4- nitrophenyl)ethenyl]benzene
  • the title compound was prepared from .-nitrophenylacetic acid (18.6 g,
  • Step B (Scheme 1): Preparation of 4-[2-(3,4,5-Trimethoxy-phenyl)ethyl]- phenylamine
  • Step C (Scheme 6): Preparation of 2- ⁇ 4-[2-(3,4,5-Trimethoxy-phenyl)- ethyl]phenylamino ⁇ -benzoic acid methyl ester
  • Step D Preparation of 2- ⁇ 4-[2-(3,4,5-Trihydroxyphenyl)ethyl]phenylamino ⁇ - benzoic acid
  • the title compound was prepared from 2- ⁇ 4-[2-(3,4,5-trimethoxy-phenyl)- ethyljphenylamino ⁇ benzoic acid (0.50 g, 1.23 mmol) in CH2C-2 (40 mL) and
  • Step A (Scheme 1): Preparation of trans - 1 -Chloro-2-trifluoromethyl-4-[2-(4- nitrophenyl)ethenyl]benzene
  • Step B (Scheme 1): Preparation of 4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]- benzenamine
  • This procedure yielded a white solid, 20.0 g (66.73 mmol, 97%) of the desired product, mp 62-64°C.
  • EXAMPLE 28 2- ⁇ 4-[3-(3 -Dimethylaminophenyl)propyl]phenylamino ⁇ benzoic acid. mp l l5-117°C.
  • EXAMPLE 38 2- ⁇ 4-[2-(3-Dibutylaminophenyl)ethyl]phenylamino ⁇ benzoic acid monohydrochloride. mp 175- 180°C.
  • EXAMPLE 54 2- ⁇ 4-[3-(3 -Piperidin- 1 yl-phenyl)-propyl] -phenylamino ⁇ -benzoic acid, mp 59-61°C.
  • EXAMPLE 58 4- ⁇ 5- [( 1 -Butyl- 1 ,2,3 ,4-tetrahydroquinolin-6-yl)methylidene]-4-oxo-2-thioxo- thiazolidin-3-yl ⁇ butanoic acid, mp 152-154°C. MS: 417 (M"l+), 418 (M+), 419 (M+1+).
  • EXAMPLE 64 5-Amino-2- ⁇ 4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid was prepared by reacting the product from Example 2 with hydrogen gas in the presence of Raney nickel, mp 137-142°C.
  • EXAMPLE 68 ⁇ 4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenyl ⁇ - [2-( 1 H-tetrazol-5 -yl)-phenyl] -amine was prepared as described in Example 1 , using a tetrazole fluoro intermediate that was synthesized from commercially available 2-fluorobenzonitrile and sodium azide under standard reaction conditions, mp 129 shrink, 152-157°C.
  • a Distriman pipet is used to add 1 mL (0.15 mmol, 1 eq) of each halo benzoate solution to the appropriate vials containing 1 mL (0.18 mmol, 1.2 eq) of the aniline reactants.
  • a catalyst solution is prepared by dissolving 0.025 M of
  • Typical compounds prepared by this method are as follows. The structure of the compounds are generally confirmed by mass spectral analysis.
  • EXAMPLE 84 2-(3',5'-Dibromo-3-methyl-biphenyl-4-ylamino)-benzoic acid MS: 459; MW: 461.1515.
  • EXAMPLE 85 -(4- 1 ,3-Benzodioxol-5-yl-2-methyl-phenylamino)-benzoic acid S: 347; MW: 347.3683.
  • EXAMPLE 105 4- ⁇ 4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ⁇ -nicotinic acid
  • EXAMPLE 106 2-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-2,3-dihydro-lH-isoindol-5-ylamino]- benzoic acid
  • EXAMPLE 120 2- ⁇ 4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino ⁇ -benzoate-2 -hydroxy- 1 , 1 -bis- hydroxymethyl-ethyl-ammonium; mp 185-187°C.
  • EXAMPLE 124 2- ⁇ 3-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid; mp 182- EXAMPLE 125 - ⁇ 4-[2-(2,4-Dimethoxy-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid; mp 180- 181°C.
  • EXAMPLE 130 2- ⁇ 4-[2-(2,4-Dichloro-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid; mp 181-183°C.
  • EXAMPLE 132 4- ⁇ 4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid; mp 214-215°C.
  • EXAMPLE 134 2- ⁇ 4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid; mp 153-154°C.
  • EXAMPLE 135 2- ⁇ 4-[5-(3,4-Dichloro-phenyl)-pentyl]-phenyl--mino ⁇ -benzoic acid; mp 106- 108°C.
  • EXAMPLE 138 4- ⁇ 4-[3 -(3 ,4-Dichloro-phenyl)-propyl] -phenylamino ⁇ -2-methoxy-5-nitro-benzoic acid; mp 74-78°C.
  • EXAMPLE 142 N-(2- ⁇ 4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino ⁇ -benzoyl)- benzenesulfonamide; MS 539.
  • EXAMPLE 143 2- ⁇ 4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ⁇ -5-trifluoromethyl-benzoic acid; mp 190-192°C. MS 453 (M" 1 ).
  • EXAMPLE 150 5-Dibutylamino-2- ⁇ 4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino ⁇ -benzoic acid;
  • Examples 158-163 illustrate the use of invention compounds as starting materials and intermediates in the synthesis of other invention compounds and derivatives.
  • the examples illustrate reduction of nitro groups to amino groups, alkylation of amino group, and esterification of carboxylic acid groups. These reactions are depicted in the following generalized
  • R D and R c are as defined above, and E is an ester forming group such as Cj-C6 alkyl (e.g., methyl, 2,2,2-trichloroethyl), benzyl, diphenylmethyl, or the like.
  • Cj-C6 alkyl e.g., methyl, 2,2,2-trichloroethyl
  • benzyl e.g., benzyl, diphenylmethyl, or the like.
  • EXAMPLE 162 2- ⁇ 4- [3 -(4-Diethylaminophenyl)propyl]phenylamino ⁇ benzoic acid methyl ester (1.68 g, 4.03 mmol) in EtOH (50 mL) was added aqueous 3 M-KOH solution (4.0 mL, 12.0 mmol) at room temperature, then the mixture was allowed to heat under reflux for 40 minutes. The reaction mixture was cooled to room temperature and neutralized with aqueous 1.0 M-HCl solution to pH 9.0. The mixture was concentrated under reduced pressure to remove EtOH, and the resulting aqueous solution was extracted with CHCI3 (50 mL, 3 times).
  • Representative compounds of Formula I have been evaluated in several in vitro and in vivo assays which are well-established as indicative of clinical usefulness in treating Alzheimer's disease.
  • Soluble A ⁇ (l -40) peptide (Bachem, Torrance, CA) - 2.2 mg/mL in deionized H2O (stored in aliquots at -20°C, keep on ice when thawed) will self-seed after 1 week storage. Typically, the solution should be stored until no lag phase is seen in the assay.
  • 125i-i a beled A/? (1-40) can be made in accordance with the procedure set forth by H. LeNine, III in Neurobiol. Aging, 16, 755, (1995), which is hereby incorporated by reference, or this reagent may be purchased from Amersham, Arlington
  • reaction mixture 1) Prepare reaction mixture above by mixing components and storing on ice. 2) Pipet 14.5 ⁇ L of reaction mixture into each of 50 wells on a polypropylene
  • RESULTS Materials: Stock Solutions: Assay Buffer - 50 mM sodium phosphate, pH 7.5, 100 mM NaCl, 0.02% NaN3,
  • Soluble A ⁇ (1-40) - 2.2 mg/mL in deionized H2O (store in aliquots at -20°C, keep on ice when thawed) will self-seed after 1 week storage. Typically, the solution should be stored until no lag phase is seen in the assay.
  • Final assay conditions 30 ⁇ M soluble A ⁇ (1-40) in deionized water in assay buffer.
  • Compound to be tested is dissolved in DMSO, typically 5 to 50 mM stock, such that the final concentration of DMSO is ⁇ 1% v/v in the assay.
  • reaction mixture for 50 assays comprised of 1 ⁇ L of soluble A ⁇ (l-40) + 13.5 ⁇ L assay buffer per assay. The following are the amounts of the components of the reaction mixture that result in each of the 50 assay wells: 50 ⁇ L soluble A ⁇ (1-40) 675 ⁇ L assay buffer
  • Assay Method 1 Prepare the reaction mix above by mixing the components and storing on ice.
  • This assay is used to provide a measure of inhibition by a compound against the aggregation behavior of the beta amyloid peptide.
  • HFIP hexafluoroisopropanol
  • a ⁇ (1-42) (California Peptide) was dried from its hexafluoroisopropanol (HFIP) stock solution.
  • the A ⁇ (1-42) was dissolved in dimethylsulfoxide (DMSO) and then mixed with phosphate buffered saline (PBS) (pH 7.4).
  • the mixed A ⁇ (1-42) solution was filtered with a 0.2 ⁇ m Omnipore membrane syringe filter (Millipore, Bedford, MA).
  • the compound to be tested in DMSO 50 times concentrate was put into each well (0.5 ⁇ L/well) of a 96-well plate.
  • the A ⁇ (1-42) solution was added into each well (24.5 ⁇ L/well). The plate was centrifuged at 1,000 g for 5 minutes and incubated at 37°C for 1 day (A ⁇ 1-42; final concentration 100 ⁇ M).
  • Inhibition (%) ⁇ (F(A ⁇ )-F(A ⁇ +compound) ⁇ / ⁇ F(A ⁇ )-F(solvent + compound) ⁇ x 100
  • the IC50S were calculated by a curve fitting program using the following equation. The data were obtained from two different experiments in triplicate.
  • Representative compounds of Formula I have exhibited inhibitory activities (IC50) ranging from 0.1 ⁇ M to greater than 100 ⁇ M in the foregoing assays.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen- Or Sulfur-Containing Heterocyclic Ring Compounds With Rings Of Six Or More Members (AREA)
  • Pyridine Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Medicinal Preparation (AREA)
  • Other In-Based Heterocyclic Compounds (AREA)
  • Heterocyclic Compounds That Contain Two Or More Ring Oxygen Atoms (AREA)
  • Pyrrole Compounds (AREA)

Abstract

The present invention provides a method of treating Alzheimer's disease using a compound of Formula (I). Also provided is a method of inhibiting the aggregation of amyloid proteins using a compound of the Formula (I) and a method of imaging amyloid deposits, as well as new compounds of Formula (I).

Description

METHOD OF INHIBITING AMYLOID PROTEIN AGGREGATION AND IMAGING AMYLOID DEPOSITS
FIELD OF THE INVENTION
This invention relates to a method of inhibiting amyloid protein aggregation and imaging amyloid deposits. More particularly, this invention relates to a method of inhibiting amyloid protein aggregation in order to treat Alzheimer's disease.
BACKGROUND OF THE INVENTION
Amyloidosis is a condition characterized by the accumulation of various insoluble, fibrillar proteins in the tissues of a patient. The fibrillar proteins that comprise the accumulations or deposits are called amyloid proteins. While the particular proteins or peptides found in the deposits vary, the presence of fibrillar morphology and a large amount of β-sheet secondary structure is common to many types of amyloids. An amyloid deposit is formed by the aggregation of amyloid proteins, followed by the further combination of aggregates and/or amyloid proteins.
The presence of amyloid deposits has been shown in various diseases, each with its particular associated protein, such as Mediterranean fever, Muckle- Wells syndrome, idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, amyloid polyneuropathy, hereditary cerebral hemorrhage with amyloidosis, Alzheimer's disease, Down's syndrome, Scrapie, Creutzfeldt- Jacob disease, Kuru, Gerstmann-Straussler- Scheinker syndrome, medullary carcinoma of the thyroid, Isolated atrial amyloid, β2-microglobulin amyloid in dialysis patients, inclusion body myositis, β2-amyloid deposits in muscle wasting disease, Sickle Cell Anemia, Parkinson's
Disease, and Islets of Langerhans diabetes Type II insulinoma. Alzheimer's disease is a degenerative brain disorder characterized clinically by progressive loss of memory, cognition, reasoning, judgement, and emotional stability that gradually leads to mental deterioration and ultimately death. Because Alzheimer's disease and related degenerative brain disorders are a major medical issue for an increasingly aging population, the need for new treatments and methods for diagnosing the disorders are needed.
A simple, noninvasive method for detecting and quantitating amyloid deposits in a patient has been eagerly sought. Presently, detection of amyloid deposits involves histological analysis of biopsy or autopsy materials. Both methods have major drawbacks. For example, an autopsy can only be used for a postmortem diagnosis.
The direct imaging of amyloid deposits in vivo is difficult, as the deposits have many of the same physical properties (i.e., density and water content) as normal tissues. Attempts to image amyloid deposits directly using magnetic resonance imaging (MRI) and computer-assisted tomography (CAT) have been disappointing and have detected amyloid deposits only under certain favorable conditions. In addition, efforts to label amyloid deposits with antibodies, serum amyloid P protein, or other probe molecules has provided some selectivity on the periphery of tissues, but has provided for poor imaging of tissue interiors. Thus, it would be useful to have a noninvasive technique for imaging and quantitating amyloid deposits in a patient. In addition, it would be useful to have compounds that inhibit the aggregation of amyloid proteins to form amyloid deposits.
SUMMARY OF THE INVENTION
The present invention provides a method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I
Figure imgf000004_0001
wherein
O
Ra is hydrogen, C^-Cg alkyl, or -CC\-Cβ alkyl; n is 0 to 5 inclusive;
Rl, R^, R3, R45 R55 R6? and R7 re independently hydrogen, halogen,
-OH, -NH2, NRbRc, -CO2H, -CO2C1-C6 alkyl, -NO2, -OC1-C12 alkyl, -Cj-Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl,
O O
II II
-CH=CH-phenyl, -O(CH2)pNRbRc, -CNRbRc, -NHCRb, -NH(CH2)pNRbRC, -N(C 1 -C6alkyiχCH2)pNRbRC,
Figure imgf000004_0002
R8 is COOH, tetrazolyl, -SO2Rd, or -CONHSO2Rd;
Rb and Rc are independently hydrogen, -C\-C alkyl, -(CH2)m-phenyl, or
RD and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomo holino, decahydroisoquinoline, or pyrazolyl;
Rd is hydrogen, -C\-C alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive; A is CH orN; R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
In a preferred embodiment, the n group is attached at
Figure imgf000005_0001
the 4-position of the phenyl ring.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2; and
R3 and R4 are hydrogen.
In a preferred embodiment of the method, in the compounds of Formula I Ra is hydrogen;
R! is halo;
R2 is hydrogen or halo; R3, R4, R5. and R^ are hydrogen; and n is 2 to 5 inclusive.
In another preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2 or 3;
R1 is -NRbRc; and
R2, R3, R4, R5, and R7 all are hydrogen. In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and
R1, R2, and R7 are independently chlorine, -N(CH CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl, -O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
, -N[(CH2)3CH3]2, substituted phenyl,
Figure imgf000006_0001
-OCH2-substituted phenyl, pyrrozolyl, or -N(phenyl)2-
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 3, 4, or 5;
R3 and R4 are hydrogen; and
Rl, R2, and R7 are independently chlorine or hydrogen.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and
R5, R6, and R8 are independently hydrogen, -CO2H, -NO2, -OCH3, O
II imidazolyl, -SCH3, -CN, fluorine, -CH3, -CF3, halogen,
II o -NH-C 1 -C6 alkyl, -N(C 1 -C6alkyl)2, -NH2, or pyrrolyl.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2;
R and R4 are hydrogen; and R5 is -CO2H. Also preferred is a method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I
Figure imgf000007_0001
wherein
Ra is hydrogen; n is 1 to 5 inclusive;
R3 and R4 are hydrogen;
R1, R7, and R2 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl,
-O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH-CH-phenyl, -N[(CH2)3CH3]2, substituted phenyl, -OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2; R5 and R6 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; or the pharmaceutically acceptable salts thereof.
In a preferred embodiment of the method, compounds of Formula I are 2- {4-[2-(3 ,4-Dichlorophenyl)ethyl]phenylamino } -benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid;
2- {4-[4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid;
2- {4-[2-(3 ,4-Dihydroxy-phenyl)-ethyl] -phenylamino } benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid; 2-{4-[2-(3,4,5-Trihydroxy-phenyl)-ethyl]phenylamino}benzoic acid; 2- {4- [3 -(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid;
2- {4-[3-(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-imidazo- 1 -yl- 5-nitrobenzoic acid; 2-{4-[3-(3,4-Dichlorophenyl)-propyl]phenylamino}benzoic acid;
2- {4-[4-(3 ,4-Dichlorophenyl)butyl]phenylamino } benzoic acid; 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-nitro-benzoic acid; 2-{4-[4-(3,4-Dichlorophenyl)-butyl]phenylamino}-3,5-dinitrobenzoic acid; 2-{4-[5-(3,4-Dichlorophenyl)pentyl]phenylamino}-5-nitrobenzoic acid;
2- {4-[5-(3 ,4-Dichloro-phenyl)pentyl]phenyl--mino } -4-methoxy- 5-nitrobenzoic acid;
2-[4-(3,4-Dichloro-benzyl)-phenylamino]-benzoic acid; 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid; 2- {4- [2-(3 ,4-Difluoro-phenyl)-ethyl] -phenylamino } -5-nitro-benzoic acid;
2- {4- [2-(4-Chloro-3 -trifluoromethyl-phenyl)-ethyl] -phenylamino} -benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid; 5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid; 2-(4-Phenethyl-phenylamino)-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methoxy-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-terephthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl-benzoic acid;
4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methanesulfonyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-imidazol-l-yl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-nitro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-nitro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-nitro-benzoic acid; 5-Cyano-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,6-difluoro-benzoic acid;
6-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-2,3-difluoro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-fluoro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-fluoro-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -methyl-benzoic acid; 2-{4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4-fluoro-benzoic acid;
2-{4- [2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -3 ,5-difluoro-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -trifluoromethyl- benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-pyrrol-l-yl-benzoic acid;
2- {4-[2-(4-Benzyloxy-phenyl)-ethyl]-phenylamino} -benzoic acid;
2-(4-{2-[4-(3-Dimethylamino-propoxy)-phenyl]-ethyl}-phenylamino)- benzoic acid;
2-{4-[2-(4-Diethylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Octyloxy-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-(4-{2-[4-(2-Ethoxy-l-ethoxymethyl-ethyl)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2-{4-[2-(4-Pyrrol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Styryl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2- {4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino} -benzoic acid;
2-{4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Octyl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[3-(3,5-Dichloro-phenoxy)-phenyl]-ethyl}-phenylamino)-benzoic acid;
2-(4-{2-[4-(2-Chloro-6-fluoro-benzyloxy)-phenyl]-ethyl}-phenylamino)- benzoic acid;
2- {4- [2-(4-Pyrazol- 1 -yl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- {4-[2-(4-Diphenylamino-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[4-(3,4-Dichloro-benzyloxy)-phenyl]-ethyl}-phenylamino)- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-amino-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichlorophenyl)]phenylamino}-5-nitrobenzoic acid; 2-{4-[2-[(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid;
2- {4- [2-(3 ,4-Dimethyl-phenyl)-ethyl] phenylamino } -5-nitrobenzoic acid; 2-[[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino-benzoic acid;
2- {4- [3-(4-Diethylaminophenyl)propyl]phenylamino } benzoic acid; 2-{4-[3-(4-Nitrophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3-Nitrophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(3 - Aminophenyl)propyl]phenyl amino } benzoic acid; 2- {4-[2-(4-Aminophenyl)ethyl]phenylamino } benzoic acid; 2-{4-[2-(4-Dipropylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2- {4-[2-(4-Diethylaminophenyl)ethyl]phenylamino } benzoic acid monohydrochloride monohydrate;
2- {4- [3 -(3 -Dipropyla inopheny l)propyl]phenylamino } benzoic acid; 2-{4-[3-(3-Dimethylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4-[3 -(4-Ethyl --minophenyl)propyl]phenylamino } benzoic acid; 2-(N-{4-[3-(4-Diethylaminophenyl)propyl]phenyl}-N-ethylamino)benzoic acid; 2-{4-[2-(3-Dibenzylaminophenyl)ethyl]phenylamino}benzoic acid;
2-{4-[3-(3-Diethylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4-[2-(3 -Aminophenyl)ethyl]phenylamino } benzoic acid;
2-{4-[3-(4-Dimethylaminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[2-(4-Acetylaminophenyl)ethyl]phenylamino}benzoic acid;
2-{4-[2-(3-Acetylaminophenyl)ethyl]phenylamino}benzoic acid;
2-{4-[2-(3-Dipropylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2-{4-[2-(3-Dibutylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2-{4-[3-(4-Acetylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3-Acetylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride; 2-{4-[2-(3-Piperidin-l-ylphenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2-{4-[3-(4-Dipropylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(4-Dibutylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3-Dibutylaminophenyl)propyl]phenylamino}benzoic acid; 2-(4- { 3-[4-( 1 H-Pyrrol- 1 -yl)phenyl]propyl } phenylamino)benzoic acid;
2- {4-[3 -(4-Piperidin- 1 -ylphenyl)propyl] phenylamino } benzoic acid;
2- {4-[3 -(4-Diethylcarbamoylphenyl)propyl]phenylamino } benzoic acid;
2- {4-[3 -(4-Carboxyphenyl)propyl]phenylamino } benzoic acid;
2-{4-[3-(4-Diethylaminomethylphenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Propylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4-[3 -(3 -Propylaminophenyl)propyl]phenylamino } benzoic acid;
2-{4-[3-(4-Pyrrolidin-l-yl-phenyl)-propyl]-phenylamino}-benzoic acid;
2- { 4- [3 -(3 -Piperidin- 1 yl-phenyl)-propyl] -phenylamino } -benzoic acid;
2-{4-[3-(4-[2-Diethylaminoethylamino]phenyl)-propyl]phenylamino}- benzoic acid;
2-{4-[2-(4-[Hydroxycarbonylmethylamino]phenyl)ethyl]phenylamino}- benzoic acid; 2-{4-[2-(4-[2-Diethylaminoethylamino]phenyl)ethyl]phenylamino}- benzoic acid;
2- {4- [3-(4-Morpholinophenyl)propyl]phenylamino } -benzoic acid; 2-{4-[3-(4-Piperazinylphenyl)propyl]phenylamino}-benzoic acid; and 2-[4-(3,4-Dichlorophenyl)phenylamino]benzoic acid.
The invention also provides the foregoing compounds wherein the benzoic acid portion is replaced with a pyridyl carboxylic acid, for example, 4-[4-(3,4- dichlorophenyl)phenylamino]-3-hydroxycarbonylpyridine.
Also provided is a method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, the method comprising administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein aggregation inhibiting amount of a compound of Formula I
Figure imgf000012_0001
wherein
O
Ra is hydrogen, Cj-Cg alkyl, or -CCj-C alkyl; n is 0 to 5 inclusive;
Rl, R2, R3, R4, R55 R65 and R? are independently hydrogen, halogen,
-OH, -NH2, NRbRc, -CO2H, -CO2Cι-C6 alkyl, -NO2, -OC1-C12 alkyl, -Ci-Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl,
O O
II II
-CH=CH-phenyl, -O(CH2)pNR Rc, -CNRbRc, -NHCRb, -NH(CH2)pNRbRC, -N(C j -C6alkyl)(CH2)pNRbRc,
Figure imgf000013_0001
R8 is COOH, tetrazolyl, -SO Rd or -CONHSO2Rd;
R and Rc are independently hydrogen, -Cj-Cg alkyl, -(CH2)m-phenyl, or
Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
Rd is hydrogen, -Cj-Cg alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive;
A is CH orN;
R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
In a preferred embodiment of the method, in the compounds of Formula I Ra is hydrogen; n is 2; and
R3 and R4 are hydrogen.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; R3 and R are hydrogen; and n is 2 to 5 inclusive.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2; R3 and R4 are hydrogen; and R1, R2 and R7 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl, -O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
, -N[(CH2)3CH3]2, substituted phenyl,
Figure imgf000014_0001
-OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2-
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 3, 4, or 5; R3 and R4 are hydrogen; and
Rl, R2, and R7 are independently chlorine or hydrogen.
In a preferred embodiment of the method, in the compounds of Formula I
Ra is hydrogen; n is 2; R3 and R4 are hydrogen; and
R5 and R6 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, halogen,
-NH-Ci-Cg alkyl, -N^^a-kyl^, -NH2, or pyrrolyl.
In a preferred embodiment of the method, in the compounds of Formula I Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and
R5 is -CO2H.
Also provided is a preferred method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, the method comprising administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein aggregation inhibiting amount of a compound of Formula I
Figure imgf000015_0001
wherein
Ra is hydrogen; n is 1 to 5 inclusive;
R3 and R4 are hydrogen;
R1, R7, and R2 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl, -O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl, -N[(CH2)3CH3]2, substituted phenyl, -OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2;
R5 and R6 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; R8 is COOH or tetrazolyl; or the pharmaceutically acceptable salts thereof.
The most preferred compounds provided by the invention have Formula II
Figure imgf000015_0002
and pharmaceutically acceptable salts thereof, wherein:
R! is halo;
R2 is H or halo; and n and R^ are as defined above in Formula I.
Another preferred group of compounds have Formula III
Figure imgf000016_0001
and pharmaceutically acceptable salts thereof, wherein:
R! is halo;
R2 is H or halo; and n and R" are as defined above in Formula I.
Another group of preferred invention compounds have Formula IV
Figure imgf000016_0002
and pharmaceutically acceptable salts thereof, wherein:
R! is halo;
R2 is H or halo; and n and R^ are as defined above in Formula I.
In a preferred embodiment of the method, the novel compounds of Formula I are provided which are
2-{4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino}-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid; 2- {4-[4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -4-methoxy-
5-nitrobenzoic acid;
2- {4- [2-(3 ,4-Dihydroxy-phenyl)-ethyl] -phenylamino } benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid; 2-{4-[2-(3,4,5-Trihydroxy-phenyl)-ethyl]phenylamino}benzoic acid; 2-{4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid;
2-{4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino}-4-imidazo-l-yl- 5-nitrobenzoic acid;
2-{4-[2-[-(3,4-Dichlorophenyl)-propyl]phenylamino}benzoic acid; 2-{4-[4-(3,4-Dichlorophenyl)butyl]phenylamino}benzoic acid; 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-nitro-benzoic acid; 2-{4-[4-(3,4-Dichlorophenyl)-butyl]phenylamino}-3,5-dinitrobenzoic acid;
2-{4-[5-(3,4-Dichlorophenyl)pentyl]phenylamino}-5-nitrobenzoic acid; 2-{4-[5-(3,4-Dichloro-phenyl)pentyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid;
2-[4-(3,4-Dichloro-benzyl)-phenylamino]-benzoic acid; 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid;
2-{4-[2-(3,4-Difluoro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid; 2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid; 5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid;
2-(4-Phenethyl-phenylamino)-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methoxy-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-terephthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl-benzoic acid;
4- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -isophthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methanesulfonyl- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5-imidazol- 1 -yl- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-nitro-benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -4-nitro-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -3 -nitro-benzoic acid; 5-Cyano-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,6-difluoro-benzoic acid;
6-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-2,3-difluoro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-fluoro-benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -fluoro-benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -methyl-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4-fluoro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3,5-difluoro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-trifluoromethyl- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-pyrrol-l-yl-benzoic acid;
2- {4- [2-(4-Benzyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4- { 2- [4-(3 -Dimethylamino-propoxy)-phenyl] -ethyl } -phenylamino)- benzoic acid; 2-{4-[2-(4-Diethylamino-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4-[2-(4-Octyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[4-(2-Ethoxy-l-ethoxymethyl-ethyl)-phenyl]-ethyl}- phenylamino)-benzoic acid; 2-{4-[2-(4-Pyrrol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2- {4- [2-(4-Styryl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Octyl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[3-(3,5-Dichloro-phenoxy)-phenyl]-ethyl}-phenylamino)-benzoic acid; 2-(4- { 2- [4-(2-Chloro-6-fluoro-benzyloxy)-phenyl] -ethyl } -phenylamino)- benzoic acid;
2- {4- [2-(4-Pyrazol- 1 -yl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Diphenylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- { 2- [4-(3 ,4-Dichloro-benzyloxy)-phenyl] -ethyl } -phenylamino)- benzoic acid;
2-{4-[2-[(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid; 2- {4-[2-(3 ,4-Dimethyl-phenyl)-ethyl] phenylamino } -5-nitrobenzoic acid; 2-[[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino-benzoic acid; or 2-[4-(3,4-Dichlorophenyl)phenyl]aminobenzoic acid.
The present invention also provides the compounds:
2- {4- [4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid;
2- {4- [2-(3 ,4-Dihydroxy-phenyl)-ethyl] -phenylamino }benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid;
2-{4-[2-(3,4,5-Trihydroxy-phenyl)-ethyl]phenylamino}benzoic acid;
2-{4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid;
2- {4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino} -4-imidazo- 1 -yl- 5-nitrobenzoic acid;
2- {4- [4-(3 ,4-Dichlorophenyl)butyl]phenylamino } benzoic acid;
2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-nitro-benzoic acid;
2-{4-[4-(3,4-Dichlorophenyl)-butyl]phenylamino}-3,5-dinitrobenzoic acid; 2-{4-[5-(3,4-Dichlorophenyl)pentyl]phenylamino}-5-nitrobenzoic acid;
2-{4-[5-(3,4-Dichloro-phenyl)pentyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid; 2-[4-(3,4-Dichloro-benzyl)-phenylamino]-benzoic acid;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid;
2-{4-[2-(3,4-Difluoro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid;
2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid;
5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5 -amino-benzoic acid; 2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5 -trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichlorophenyl)]phenylamino}-5-nitrobenzoic acid;
2-(4-Phenethyl-phenylamino)-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5 -methoxy-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-terephthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl-benzoic acid;
4- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -isophthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methanesulfonyl- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5-imidazol- 1 -yl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-nitro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-nitro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-nitro-benzoic acid;
5-Cyano-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,6-difluoro-benzoic acid; 6-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-2,3-difluoro-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -6-fluoro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-fluoro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-methyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-fluoro-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -3 ,5-difluoro-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-trifluoromethyl- benzoic acid; 2-{4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5 -trifluoromethyl- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-pyrrol-l-yl-benzoic acid;
2- {4-[2-(4-Benzyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[4-(3 -Dimethylamino-propoxy)-phenyl] -ethyl } -phenylamino)- benzoic acid;
2-{4-[2-(4-Diethylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4- [2-(4-Phenoxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- {4-[2-(4-Octyloxy-phenyl)-ethyl]-phenylamino} -benzoic acid; 2-(4-{2-[4-(2-Ethoxy-l-ethoxymethyl-ethyl)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2- {4-[2-(4-Pyrrol- 1 -yl-phenyl)-ethyl] -phenylamino} -benzoic acid; 2- {4-[2-(4-Styryl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl]-phenylamino}-benzoic acid;
2- {4-[2-(4-Octyl-phenyl)-ethyl]-phenylamino} -benzoic acid; 2-(4-{2-[3-(3,5 -Dichloro-phenoxy)-phenyl] -ethyl } -phenylamino)-benzoic acid;
2-(4-{2-[4-(2-Chloro-6-fluoro-benzyloxy)-phenyl]-ethyl}-phenylamino)- benzoic acid;
2-{4-[2-(4-Pyrazol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Diphenylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- { 2- [4-(3 ,4-Dichloro-benzyloxy)-phenyl] -ethyl } -phenylamino)- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5 -amino-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-trifluoromethyl- benzoic acid;
2- {4-[2-(3 ,4-Dichlorophenyl)]phenylamino } -5-nitrobenzoic acid; 2- {4-[2- [(3 ,4-Dichlorophenyl)propyl]phenylamino } -5-nitrobenzoic acid; 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid; 2-{4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenylamino}-benzoic acid;
2-{4-[3-(4-Diethylaminophenyl)propyl]phenylamino}benzoic acid; 2- {4-[3 -(4-Nitrophenyl)propyl]phenylamino } benzoic acid; 2-{4-[3-(3-Nitrophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3 - Aminophenyl)propyl] phenylamino } benzoic acid; 2-{4-[2-(4-Aminophenyl)ethyl]phenylamino}benzoic acid; 2-{4-[2-(4-Dipropylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride; 2-{4-[2-(4-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride monohydrate;
2-{4-[3-(3-Dipropylaminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(3-Dimethylaminophenyl)propyl]phenylamino}benzoic acid; 2- {4- [3-(4-Ethylaminophenyl)propyl]phenylamino } benzoic acid; 2-(N-{4-[3-(4-Diethylaminophenyl)propyl]phenyl}-N-ethylamino)benzoic acid;
2- {4- [2-(3-Dibenzylaminophenyl)ethyl]phenylamino } benzoic acid; 2-{4-[3-(3 -Diethylaminophenyl)propyl]phenylamino } benzoic acid; 2- {4- [2-(3- Aminophenyl)ethyl]phenylamino } benzoic acid; 2-{4-[3-(4-Dimethylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[2-(4-Acetylaminophenyl)ethyl]phenylamino}benzoic acid; 2-{4-[2-(3-Acetylaminophenyl)ethyl]phenylamino}benzoic acid; 2-{4-[2-(3-Dipropylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2-{4-[2-(3-Dibutylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride; 2-{4-[3-(4-Acetylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(3-Acetylaminophenyl)propyl]]phenylamino}benzoic acid; 2-{4-[3-(3-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2- {4- [2-(3 -Piperidin- 1 -ylphenyl)ethyl]phenylamino }benzoic acid monohydrochloride;
2-{4-[3-(4-Dipropylaminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Dibutylaminophenyl)propyl]phenylamino}benzoic acid; 2- {4- [3 -(3 -Dibutylaminophenyl)propyl]phenylamino } benzoic acid; 2-(4-{3-[4-(lH-Pyrrol-l-yl)phenyl]propyl}phenylamino)benzoic acid; 2-{4-[3-(4-Piperidin-l-ylphenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(4-Diethylcarbamoylphenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Carboxyphenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Diethylaminomethylphenyl)propyl]phenylamino}benzoic acid; 2- { 4- [3 -(4-Propylaminophenyl)propyl]phenylamino } benzoic acid; 2-{4-[3-(3-Propylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4- [3 -(4-Pyrrolidin- 1 -yl-phenyl)-propyl] -phenylamino } -benzoic acid; 2- {4- [3 -(3 -Piperidin- 1 y l-phenyl)-propy 1] -phenylamino } -benzoic acid; { 5- [( 1 -Butyl- 1 ,2,3 ,4-tetrahydro-6-quinolyl)methylidene] -4-oxo-2- thioxothiazolidin-3 -yl } acetic acid; {5-[(l-Butyl-2,3-dihydro-lH-indol-5-yl)methylidene]-4-oxo-2- thioxothiazolidin-3-yl}acetic acid;
3 - { 5- [( 1 -Butyl- 1 ,2,3 ,4-tetrahydroquinolin-6-yl)methylidene] -4-oxo-2- thioxothiazolidin-3-yl}propanoic acid;
4-{5-[(l-Butyl-l,2,3,4-tetrahydroquinolin-6-yl)methylidene]-4-oxo-2- thioxothiazolidin-3-yl}butanoic acid; or
2-[4-(3,4-Dichlorophenyl)phenyl]aminobenzoic acid. Also provided are the foregoing compounds wherein the terminal phenylalkyl group is attached at the 2- or 3-position of the central phenyl ring, i.e., compounds of the Formula la
Figure imgf000024_0001
Typical 2- and 3-substituted compounds are:
2-{3-[2-(3 ,4-Dichloropheny l)ethyl]phenylamino } -benzoic acid; 2- {2- [2-(3 ,4-Dichlorophenyl)ethyl]phenylamino } -benzoic acid; 2- { 3 - [3 -(4-Diethylaminophenyl)propyl]phenylamino } -benzoic acid; 2- { 3- [3 -(4-Di-n-propylaminophenyl)propyl]phenylamino } -benzoic acid; 2-{3-[3-(4-n-Propylaminophenyl)propyl]phenylamino}-benzoic acid;
2- { 3 -[3 -(4-[2-Diethylaminoethylamino]phenyl)propyl]phenylamino } - benzoic acid;
2-{2-[3-(4-[Hydroxycarbonylmethylamino]phenyl)propyl]phenylamino}- benzoic acid; 2- {2- [2-(3 - [2-Diethylaminoethylamino]phenyl)ethyl]phenylamino } - benzoic acid;
2- { 3 - [3 -(4-Morpholinophenyl)propyl]phenylamino } -benzoic acid; 2- { 3-[3 -(4-Piperazinylphenyl)propyl]phenylamino } -benzoic acid; 2- {3-[2-(4-Chlorophenyl)ethyl]phenylamino} -benzoic acid; 2-{3-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-benzoic acid; and
2-{4-[4-(4-{4-Methylpiperazinyl}phenyl)butyl]phenylamino}-benzoic acid.
Pharmaceutical formulations of the novel compounds admixed with a pharmaceutically acceptable diluent, carrier, or excipient are also provided.
Also provided is a method of imaging amyloid deposits, the method comprising: introducing into a patient a detectable quantity of a labeled compound having the Formula I or a pharmaceutically acceptable salt thereof:
Figure imgf000025_0001
wherein
O
Ra is hydrogen, Cι-C6 alkyl, or -CCi-Cg alkyl; n is 0 to 5 inclusive;
R1, R2, R3, R4, R5, R6, and R7 are independently hydrogen, halogen, -OH, -NH2, NRbRC, -CO2H, -CO2C!-C6 alkyl, -
NO , -OC!-C12 alkyl, -C^Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O- phenyl, -O-substituted phenyl,
O O
II II
-CH=CH-phenyl, -O(CH2)pNRbRc, -CNRbRc, -NHCRb, -NH(CH2)pNRbRC, -N(C ] -C6alkyl)(CH2)pNRbRc,
Figure imgf000025_0002
R8 is COOH, tetrazolyl, -SO2Rd, or -CONHSO2Rd;
R and Rc are independently hydrogen, -Cj-Cg alkyl, -(CH2)m- phenyl, or Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cι-Cg alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
Rd is hydrogen, -Ci-Cg alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive;
A is CH or N;
R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof; b. allowing sufficient time for the labeled compound to become associated with amyloid deposits; and c. detecting the labeled compound associated with the amyloid deposits.
In a preferred embodiment of the method, the patient has or is suspected to have Alzheimer's disease.
In a preferred embodiment of the method, the labeled compound is a radio labeled compound.
In a preferred embodiment of the method, the labeled compound is detected using MRI.
The present invention also provides the preferred compounds: 2-{4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino}-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid; 2-{4-[3-(3,4-Dichlorophenyl)-propyl]phenylamino}benzoic acid; 2-[[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino-benzoic acid; 2-{4-[3-(4-Diethylaminophenyl)propyl]phenylamino}-benzoic acid; and pharmaceutical formulations thereof.
Pharmaceutically acceptable acid addition salts, amides, and prodrugs of the foregoing compounds are also provided by this invention. DETAILED DESCRIPTION OF THE INVENTION
The term "alkyl" means a straight or branched chain hydrocarbon having from 1 to 12 carbon atoms. Representative examples of alkyl groups are methyl, ethyl, propyl, isopropyl, isobutyl, butyl, tert-butyl, sec-butyl, pentyl, hexyl, octyl, decyl, and 1,1-dimethyloctyl.
Preferred alkyl groups are Cj-Cg alkyl, and especially C1-C6 alkyl.
The term "alkoxy" means an alkyl group attached to an oxygen atom. Representative examples of alkoxy groups include methoxy, ethoxy, tert-butoxy, propoxy, and isobutoxy. Preferred alkoxy groups are Cj-C^ alkoxy, and especially C \ -C alkoxy.
The term "halogen" includes chlorine, fluorine, bromine, and iodine.
The term "substituted" means that one or more hydrogen atom in a molecule has been replaced with another atom or group of atoms. For example, substituents include halogen, especially chloro, -OH, -CF3, -NO2, -NH2, -NH(C!-C6alkyl), -N(C!-C6alkyl)2, C^Cg alkyl, -OC!-C6 alkyl, -CN, -CF3,
-CO2H, and -CO2C1-C6 alkyl.
The term "substituted phenyl" means a phenyl ring in which from 1 to 4 hydrogen atoms have been independently replaced with a substituent, preferably one selected from the list above. Typical "substituted phenyl" groups include 4-chlorophenyl, 3,4-dibromophenyl, 3-fluoro-4-methylphenyl,
3,4-dichlorophenyl, 3,4-methylenedioxyphenyl, and 4-dimethylaminophenyl. The symbol "-" means a covalent bond.
Substituent groups represented by R , R3, and R^, for example, include amino(NRbRc) and acylamino (-NHCORb). Rb and Rc can be hydrogen, alkyl and phenylalkyl and substituted phenylalkyl, and typical NRbRc groups include methylamino, diethylamino, isobutyl-propylamino, benzylamino, and 3,4- dimethoxybenzylamino. Examples of acylamino groups include formamido, acetamido, 2-phenylacetamido, and 2-(3-nitrophenyl)acetamido. R}, R3, and R5 can also be aminoalkoxy (-O(CH2)pNRbRc) such as N-methylaminomethoxy and 2-(N-benzylamino)ethoxy, as well as aminoalkylamino (-NH(CH2)pNRbRc) such as 3-(dimethylamino)propylamino and 2-(N-ethyl-N-benzylamino)ethylamino. Substituent groups such as Rl, R3, and R^ additionally can be cyclic structures, for instance when NRbRc is part of the substituent group, and Rb and Rc are taken together with the nitrogen to which they are attached to form a cyclic ring selected from imidazole, pyrrole, piperidine, piperazine, 4-Cj-C6 alkylpiperazine, morpholine, thiomorpholine, pyrazole, and decahydroisoquinoline.
Substituent groups such as R1, R2, R5, R6, and R7 also can be -CH=CH- phenyl (i.e., styryl), phenoxy, O-substituted phenyl such as 3-iodophenoxy, 2,4,6- trihydroxyphenoxy, 2-fluoro-3-nitrophenoxy, as well as -O-benzyl and
-O-substituted benzyl such as 2-trifluoromethylbenzyloxy and 4-aminobenzyloxy. The term "pharmaceutically acceptable salt, ester, amide, and prodrug" as used herein refers to those carboxylate salts, amino acid addition salts, esters, amides, and prodrugs of the compounds of the present invention which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "salts" refers to the relatively nontoxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laureate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate and laurylsulphonate salts, and the like. These may include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, and the like, as well as, nontoxic ammonium, quaternary ammonium and amine cations including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. (See, for example, Berge S.M., et al., Pharmaceutical Salts, J. Pharm. Sci., 66:1-19 (1977) which is incorporated herein by reference.)
Examples of pharmaceutically acceptable, nontoxic esters of the compounds of this invention include Cj-C6 alkyl esters wherein the alkyl group is a straight or branched chain. Acceptable esters also include C5-C7 cycloalkyl esters as well as arylalkyl esters such as, but not limited to benzyl. C1-C4 alkyl esters are preferred. Esters of the compounds of the present invention may be prepared according to conventional methods, for example by reacting a carboxylic acid of Formula I with an alcohol such as ethanol or benzyl alcohol. Examples of pharmaceutically acceptable, nontoxic amides of the compounds of this invention include amides derived from ammonia, primary C\-C alkyl amines and secondary C\-C dialkyl amines wherein the alkyl groups are straight or branched chain. In the case of secondary amines, the amine may also be in the form of a 5- or 6-membered heterocycle containing one nitrogen atom. Amides derived from ammonia, C1-C3 alkyl primary amides and
C1-C2 dialkyl secondary amides are preferred. Amides of the compounds of the invention may be prepared according to conventional methods.
The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formulas, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and
V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference. In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
The compounds of the present invention can exist in different stereoisometric forms by virtue of the presence of asymmetric centers in the compounds. It is contemplated that all stereoisometric forms of the compounds, as well as mixture thereof, including racemic mixtures, form part of this invention. In the first step of the present method of imaging, a labeled compound of Formula I is introduced into a tissue or a patient in a detectable quantity. The compound is typically part of a pharmaceutical composition and is administered to the tissue or the patient by methods well-known to those skilled in the art. In the methods of the present invention, a compound can be administered either orally, rectally, parenterally (intravenous, by intramuscularly or subcutaneously), intracisternally, intravaginally, intraperitoneally, intravesically, locally (powders, ointments or drops), or as a buccal or nasal spray. Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid; (b) binders, as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia; (c) humectants, as for example, glycerol; (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate; (e) solution retarders, as for example paraffin; (f) absorption accelerators, as for example, quaternary ammonium compounds; (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate; (h) adsorbents, as for example, kaolin and bentonite; and
(i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft- and hard-filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethyleneglycols, and the like.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain opacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions which can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3 -butyl eneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols, and fatty acid esters of sorbitan or mixtures of these substances, and the like. Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like. Compositions for rectal administrations are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable nonirritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants. The active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers or propellants as may be required. Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
In a preferred embodiment of the invention, the labeled compound is introduced into a patient in a detectable quantity and after sufficient time has passed for the compound to become associated with amyloid deposits, the labeled compound is detected noninvasively inside the patient. In another embodiment of the invention, a labeled compound of Formula I is introduced into a patient, sufficient time is allowed for the compound to become associated with amyloid deposits, and then a sample of tissue from the patient is removed and the labeled compound in the tissue is detected apart from the patient. In a third embodiment of the invention, a tissue sample is removed from a patient and a labeled compound of Formula I is introduced into the tissue sample. After a sufficient amount of time for the compound to become bound to amyloid deposits, the compound is detected.
The administration of the labeled compound to a patient can be by a general or local administration route. For example, the labeled compound may be administered to the patient such that it is delivered throughout the body. Alternatively, the labeled compound can be administered to a specific organ or tissue of interest. For example, it is desirable to locate and quantitate amyloid deposits in the brain in order to diagnose or track the progress of Alzheimer's disease in a patient.
The term "tissue" means a part of a patient's body. Examples of tissues include the brain, heart, liver, blood vessels, and arteries. A detectable quantity is a quantity of labeled compound necessary to be detected by the detection method chosen. The amount of a labeled compound to be introduced into a patient in order to provide for detection can readily be determined by those skilled in the art. For example, increasing amounts of the labeled compound can be given to a patient until the compound is detected by the detection method of choice. A label is introduced into the compounds to provide for detection of the compounds.
The term "patient" means humans and other animals. Those skilled in the art are also familiar with determining the amount of time sufficient for a compound to become associated with amyloid deposits. The amount of time necessary can easily be determined by introducing a detectable amount of a labeled compound of Formula I into a patient and then detecting the labeled compound at various times after administration.
The term "associated" means a chemical interaction between the labeled compound and the amyloid deposit. Examples of associations include covalent bonds, ionic bonds, hydrophilic-hydrophilic interactions, hydrophobic- hydrophobic interactions, and complexes.
Those skilled in the art are familiar with the various ways to detect labeled compounds. For example, magnetic resonance imaging (MRI), positron emission tomography (PET), or single photon emission computed tomography (SPECT) can be used to detect radiolabeled compounds. The label that is introduced into the compound will depend on the detection method desired. For example, if PET is selected as a detection method, the compound must possess a positron-emitting atom, such as ^C or 18F.
Another example of a suitable label in a compound of Formula I is an atom such as 13C, l^N, 0r l" which can be detected using magnetic resonance imaging (MRI) which is also sometimes called nuclear magnetic resonance
(NMR). In addition, the labeled compounds of Formula I may also be detected by MRI using paramagnetic contrast agents. Another example of detection is electron paramagnetic resonance (EPR). In this case, EPR probes which are well-known in the art, such as nitroxides, can be used.
The imaging of amyloid deposits can also be carried out quantitatively so that the amount of amyloid deposits can be determined.
The present invention also provides a method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, by administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein inhibiting amount of a compound of Formula I. Those skilled in the art are readily able to determine an amyloid inhibiting amount by simply administering a compound of Formula I to a patient in increasing amounts until the growth of amyloid deposits is decreased or stopped. The rate of growth can be assessed using imaging or by taking a tissue sample from a patient and observing the amyloid deposits therein. A patient in need of inhibition of the aggregation of amyloid proteins is a patient having a disease or condition in which amyloid proteins aggregate. Examples of such diseases and conditions include Mediterranean fever, Muckle- Wells syndrome, idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, amyloid polyneuropathy, hereditary cerebral hemorrhage with amyloidosis, Alzheimer's disease, Down's syndrome,
Scrapie, Creutzfeldt- Jacob disease, Kuru, Gerstmann-Straussler-Scheinker syndrome, medullary carcinoma of the thyroid, Isolated atrial amyloid, β2-microglobulin amyloid in dialysis patients, inclusion body myositis, β2-amyloid deposits in muscle wasting disease, and Islets of Langerhans diabetes Type II insulinoma.
Also provided by the present invention are compounds of Formula I wherein one or more atom in the compound has been replaced with a radioisotope
(a labeled compound). The radioisotope can be any radioisotope. However,3H, 123 L 125^ 131 T ; 11 and l 8F are preferred. Those skilled in the art are familiar with the procedure used to introduce a radioisotope into a compound. For example, a compound of Formula I wherein one carbon atom is ^C or ^C is readily prepared. The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1 ,000 mg per day. For a normal human adult having a body weight of about 70 kg, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is sufficient. The specific dosage used, however, can vary. For example, the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used. The determination of optimum dosages for a particular patient is well- known to those skilled in the art. The examples presented below are intended to illustrate particular embodiments of the invention and are not intended to limit the scope of the specification, including the claims, in any manner.
EXAMPLES
SYNTHESIS Compounds of Formula I can be prepared by several routes as illustrated in
Schemes 6 through 9. Schemes 1 through 5 show synthetic routes that can be used to obtain the desired starting amines (IV), (VIII), (XV), and (XXI).
In Scheme 1, the appropriately substituted aldehyde (I) and a nitrophenylacetic acid (II) yield olefin (III) when heated in piperidine at 150°C. Standard hydrogenation conditions, such as Raney nickel, give desired amine
(IN).
Scheme 2 depicts the synthesis of amine (NIII) which contains a three methylene tether. Condensation of aldehyde (I) and nitro-ketone (N) in the presence of sodium hydroxide gives the desired alpha, beta-unsaturated ketone, which upon standard hydrogenation conditions (Raney nickel) gives (Nil) and then Wolff-Kishner conditions yields the desired amine (VIII).
Scheme 3 is very similar to Scheme 2, except that the aldehyde (I) is condensed with a substituted aniline (IX).
Scheme 4 illustrates standard Wittig conditions in which the starting materials (XII) and (XIII) are obtained via aldol condensation and ylide chemistry, respectively. Reaction of aldehyde (XII) and bromophosphorane (XIII) in the presence of a base, such as butyl lithium, gives diene (XIV). Standard reduction conditions (e.g., Raney nickel) of (XIN) yields the desired amine (XV).
Scheme 5 illustrates the synthesis of amine (XXI) which contains a 5-methylene tether. Wittig reaction of the bromophosporane (XVII), which is formed from the corresponding substituted bromide (XVI), and nitro aldehyde
(XIX), obtained from Swern oxidation of the corresponding alcohol (XVIII), using a base (e.g., LHDMS) yields olefin (XX). Reduction of (XX) using standard conditions (Raney nickel) gives amine (XXI).
Scheme 6 illustrates one route to obtain compounds of Formula I. Either by Buchwald coupling (Method A) followed by saponification or utilizing the
Ullman reaction (Method B), compounds of Formula I can be isolated from amines such as (IV), (VIII), and (XV). Compounds of Formula I that contain hydroxy groups, such as Examples 4 and 6, require demethylation of the hydroxy protecting groups with reagents such as boron tribromide in the final step of the synthesis.
Protecting groups will also be used when reactive functional groups such as amino and carboxylic acids are present, so as to avoid unwanted side reactions. Carboxy groups typically are converted to esters (e.g., tert-butyl, benzyl), and amino groups generally are acylated (e.g., acetyl or trimethylsilyl). These and other such protecting groups are well-known to organic chemists, and are fully described by Greene and Wuts in Protective Groups in Organic Synthesis, John Wiley and Sons, New York (2nd Ed. 1991). All citations are incorporated herein by reference.
Scheme 7 illustrates the synthesis of compounds of Formula I by reacting amines such as (IN), (NIII), and (XXI) with fluoro-nitro intermediate (XXIN), in the presence of a base (e.g., LHMDS or Et3Ν) to give ester (XXV). This ester can then be saponified using standard conditions, such as sodium hydroxide.
In Scheme 8, amine (XV) can be coupled with readily available fluoro- substituted carboxylic acids [e.g., (XXVI) or (XXNII)] in the presence of various bases (such as DBU or triethylamine) to yield compounds of Formula I.
Scheme 9 depicts coupling of amine (VIII) with readily available methyl ester (XXVIII) in the presence of a base, such as imidazole, to give ester (XXIX). This ester can then be saponified as usual to give compounds of Formula I.
Scheme 10 illustrates the synthesis of fluoro-intermediate (XXIV) which is obtained by nitration of readily available methyl ester (XXX) to give (XXVIII). Treatment of (XXVIII) with potassium cyanide gives (XXIV). In Scheme 11, the synthesis of compounds related to Example 18 is illustrated. Reaction of the potassium salts of ortho-substituted benzoic acids (XXVI) with substituted anilines (XXVII) in the presence of potassium carbonate and cupric acetate yields various iodo-substituted aminobenzoic acids (XXVIII). Reaction of (XXVIII) with substituted boronic acids and palladium chloride gives the desired substituted aminobenzoic acids (XXX).
It should, of course, be recognized that several invention compounds of Formula I can be prepared from other compounds defined by Formula I, utilizing standard organic reactions such as oxidation, reduction, alkylation, condensation, elimination, and similar well-known synthetic processes. For example, compounds of Formula I wherein Ra is hydrogen are readily alkylated to form compounds wherein Ra is Cj-Cg alkyl. Compounds wherein R! is NH2 are readily acylated by reaction with an acid halide or acid anhydride to provide compounds wherein R! is -NHCOR . Similarly, compounds wherein R^ is NO2 are easily reduced to provide compounds wherein R^ is NH2. The benzoic acids (where R8 is COOH) are readily converted to esters and amides, as well as salts and other prodrugs by routine processes. For example, the benzoic acid can be reacted with oxalylchloride to form the acid chloride, which then readily reacts with a sulfonamide such as methanesulfonamide to produce the corresponding invention compound where R8 is -CONHSO2CH3. Formation of Amines
Scheme 1
Figure imgf000038_0001
Ra-Ni/THF or
Step B Pd-C/DMF
Figure imgf000038_0002
Scheme 2
Figure imgf000038_0003
Scheme 3
Figure imgf000039_0001
Scheme 4
Figure imgf000040_0001
(XII) (XIII)
Figure imgf000040_0002
(XIV)
Figure imgf000040_0003
Scheme 5
Figure imgf000041_0001
Coupling Routes
Scheme 6
Method A. Step C (l) Pd2(dba)3/BINAP
Cs2C03, toluene
Figure imgf000041_0002
(XXIII) Scheme 7
Figure imgf000042_0001
Compound of Formula I
R is an ester forming group such as alkyl or benzyl.
Scheme 8
Figure imgf000043_0001
Scheme 9
Figure imgf000044_0001
Compound of Formula I
Synthesis of Fluoro-Intermediate
Scheme 10
Figure imgf000044_0002
Scheme 11
KjCOj/MeOH K2C03, Cu(Ac)2
Figure imgf000045_0001
(XXV) (XXVI) (xxvπ)
Figure imgf000045_0003
(XXVIII) (xxrx)
Figure imgf000045_0004
EXAMPLE 1 Preparation of 2-{4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino}-benzoic acid
Step A (Scheme 1): Preparation of l,2-Dichloro-4-[2-(4-nitrophenyl)ethenyl]- benzene A mixture ofp-nitrophenylacetic acid (51.23 g, 0.28 mol) and
3,4-dichlorobenzaldehyde (49.50 g, 0.28 mol) in piperidine (50 mL) was heated to 150-160°C for 5 hours under a N2 atmosphere. After cooling the reaction mixture, the precipitate was triturated in boiling methanol (MeOH) (50 mL) and then cooled to -5°C for 12 hours. The crystalline precipitate was filtered off, rinsed with cold MeOH and dried at room temperature in a vacuum oven overnight to yield an orange solid, 22.71 g (0.077 mol, 27%) of the desired product, mp 190-191°C.
MS-294.9 (M+).
Step B (Scheme 1): Preparation of 4-[2-(3,4-Dichlorophenyl)ethyl]benzenamine A sample of l,2-dichloro-4-[2-(4-nitrophenyl)ethenyl]benzene (98.0 g,
0.33 mol) in tetrahydrofuran (THF) (1.6 L) was reduced in the presence of Raney Nickel (Ra-Ni) (20 g) at 25°C to 40°C (ΔP = 13.5 psi) under a hydrogen atmosphere. The reaction mixture was filtered, and the filtrate was concentrated in vacuo to give an orange solid, 85.0 g (0.32 mol, 95.8%) of the desired product. mp 68-70°C.
MS: 266.1 (M+).
Step C (Scheme 6): Preparation of 2-{4-[2-(3,4-Dichlorophenyl)ethyl]phenylamino}-benzoic acid
Method A A mixture of 4-[2-(3,4-dichlorophenyl)ethyl]benzenamine (28.37 g,
106.59 mmol), methyl 2-bromobenzoate (19.10 g, 88.82 mmol), cesium carbonate (40.52 g, 124.35 mmol), tris(dibenzylideneacetone-dipaladium(0) (2.44 g, 2.67 mmol) and (S)-(2,2'-bis(di-p-tolylphosphino-l,l'-binaphthyl (98%, (S)-tol- BINAP) (2.71 g, 4.00 mmol) (Ligand/Pd = 1.5) in anhydrous toluene (300 mL) was heated to 100°C for 34 hours under N2. After cooling to room temperature, the reaction mixture was diluted with ether, filtered through celite and rinsed thoroughly with ether. The filtrate was evaporated to dryness to give a brown residue (68 g). The resulted residue was dissolved in ethanol (EtOH) (50 mL) and THF (100 mL), and then 5N NaOH (aq.) (200 mL) was added, and the mixture was refluxed for 16 hours. The solvent was removed in vacuum. The residue was acidified with concentrated HCl to pH 3. The resulting precipitate was collected by filtration, triturated with boiling MeOH-H2θ (4:1) and dried in a vacuum at room temperature for 16 hours to give Example 1, an orange solid (31.95 g, 0.083 mol, 77.6%). mp 175.0-177.0°C.
Analysis for C2iH17N1O2Cl2: Calcd: C, 65.30; H, 4.44; N, 3.63. Found: C, 65.40; H, 4.54; N, 3.50.
Method B
A mixture of 2-chlorobenzoic acid (5.4 g, 0.034 mol), 4-[2-(3,4- dichlorophenyl)ethyl]benzenamine (10.0 g, 0.037 mol), anhydrous potassium carbonate (16.9 g, 0.12 mol), copper powder (4.94 g, 0.077 mol), and copper(I) chloride (0.37 g, 0.0037 mol) in dry dimethylformamide (DMF) (85 mL) was heated to reflux for 24 hours at 150°C. The reaction mixture was poured into hot H2O (150 mL) and heated to 90°C on a hot plate. Charcoal was added, and this mixture was stirred at 90°C for 5 minutes. The warm brown mixture was filtered through filter paper. The cooled filtrate was then acidified with concentrated HCl (pH 1), and the precipitate was collected by filtration, triturated with boiling MeOH-H2O (1 :2) and dried under vacuum at room temperature for 16 hours to give Example 1, an orange solid (2.3 g, 0.006 mol, 17.5%). mp 165.0-173.0°C. Analysis for C2iH17N1O2Cl2: Calcd: C, 65.30; H, 4.44; N, 3.63.
Found: C, 65.68; H, 4.58; N, 3.60. EXAMPLE 2 Preparation of 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -5-nitrobenzoic acid
Step C (Scheme 6): Preparation of 2-{4-[2-(3,4-Dichloro-phenyl)- ethyl]phenylamino}-5-nitrobenzoic acid methyl ester
A mixture of 4-[2-(3,4-dichlorophenyl)ethyl]benzenamine (600 mg, 2.25 mmol), 2-bromo-5-nitrobenzoic acid methyl ester (489 mg, 1.88 mmol), cesium carbonate (857 mg, 2.62 mmol), tris(dibenzylideneacetone-dipaladium(0) (51 mg, 0.056 mmol) and (S)-(2,2'-bis(di-p-tolylphosphino-l,r-binaphthyl (98%, (S)-tol-BINAP) (58 mg, 0.085 mmol) (Ligand/Pd = 1.5) in anhydrous toluene
(16 mL) was heated to 100°C for 12 hours under N2. After cooling, the reaction mixture was diluted with ether, filtered through celite and rinsed thoroughly with ether. The filtrate was evaporated to dryness to give a brown residue. Purification by flash chromatography (silica gel, 5% EtOAc/hexane) yielded 540 mg (1.21 mmol, 64%) of the desired product, mp 107-108°C.
Analysis for C 2H18N2Cl2O : Calcd: C, 59.34; H, 4.07; N, 6.29. Found: C, 59.03; H, 4.04; N, 5.99.
Preparation of 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid A solution of 2-{4-[2-(3,4-dichloro-phenyl)-ethyl]phenylamino}-
5-nitrobenzoic acid methyl ester (340 mg, 0.76 mmol) and IN NaOH (aq.) (4.0 mL) in EtOH (4.0 mL) and THF (4.0 mL) was heated to reflux for 16 hours. The solvent was removed in vacuum. The residue was diluted with H O and acidified with concentrated HCl to pH 1. The mixture was then extracted with methylene chloride, dried (Na SO4), filtered and concentrated in vacuo to yield a yellow solid, 329 mg (0.76 mmol, 100%) of the desired product, mp 214-217°C. Analysis for C2iH16N2Cl2O4: Calcd: C, 58.49; H, 3.74; N, 6.50. Found: C, 58.24; H, 3.81; N, 6.28. EXAMPLE 3 Preparation of 2- {4-[4-(3 ,4-Dichloro-pheny l)-ethyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid
To a cooled (-78°C) solution of 4-[2-(3,4-dichloro-phenyl)- ethyl]phenylamine (0.836 g, 3.14 mmol) in THF (20 mL), LHDMS (6.28 mL, 1 M in THF, 6.28 mmol) was added dropwise. The reaction mixture was allowed to stir at -78°C for 10 minutes. A solution of 2-flouro-4-methoxy-5-nitrobenzoic acid methyl ester (0.72 g, 3.14 mmol) in THF (30 mL) was added dropwise, and this solution was stirred for 30 minutes at -78°C. The reaction mixture was allowed to gradually warm to room temperature and stir for 2 hours under a N2 atmosphere.
The reaction mixture was diluted with ethyl acetate (EtOAc), and acidified with 5N HCl (pH 3). The organic layer was dried (Na2SO4), filtered and concentrated in vacuo to yield a brown residue. To a solution of this residue in EtOH (20 mL) and THF (40 mL), 5N NaOH (50 mL) was added, and the mixture was refluxed for overnight. The solvent was removed in vacuum, and the residue was acidified with concentrated HCl (pH 3). The precipitate was collected by filtration, triturated with boiling MeOH-H2O (1:1), and dried in a vacuum oven for 16 hours to give Example 3, an orange solid (0.70 g, 1.51 mmol, 48%). mp 208-209°C. Analysis for C22H18N2O5Cl : Calcd: C, 57.28; H, 3.93; N, 6.07. Found: C, 57.43; H, 3.69; N, 5.86.
EXAMPLE 4 Preparation of 2-{4-[2-(3,4-Dihydroxy-phenyl)-ethyl]-phenylamino}benzoic acid
Step A (Scheme 1): Preparation of l,2-Dimethoxy-4-[2-(4-nitrophenyl)ethenyl]- benzene The title compound was prepared from /.-nitrophenylacetic acid (25.0 g,
0.14 mol), and 3,4-dimethoxybenzaldehyde (21.0 g, 0.14 mol) in piperidine (5 mL) using the procedure described in Example 1 , Step A, to yield a yellow solid, 13.4 g (0.047 mol, 34%) of the desired product, mp: 133-134°C. Analysis of Ci6H15N1O4: Calcd: C, 67.36; H, 5.30; N, 4.91. Found: C, 66.81; H, 5.27; N, 4.84. Step B (Scheme 1): Preparation of 4-[2-(3,4-Dimethoxy-phenyl)ethyl]- phenylamine l,2-Dimethoxy-4-[2-(4-nitrophenyl)ethenyl]benzene (12.1 g, 0.042 mol) was reduced in the presence of 10% Pd-C (2.0 g) in dimethylformamide (DMF) (120 mL) at 25°C under a hydrogen atmosphere. The reaction mixture was concentrated in vacuo to give a solid. The solid was recrystallized from MeOH (400 mL) to yield a white crystalline product, 6.8 g (0.026 mol, 63%) of the desired product, mp 115-116°C.
Analysis for Ci6H19N1O : Calcd: C, 74.68; H, 7.44; N, 5.44. Found: C, 74.60; H, 7.39; N, 5.35.
Step C (Scheme 6): Preparation of 2-{4-[2-(3,4-Dimethoxy-phenyl)ethyl]- phenylamino} benzoic acid
The title compound was prepared from 4-[2-(3,4-dimethoxy-phenyl)- ethyl]phenylamine (9.25 g, 0.036 mol), 2-chlorobenzoic acid (5.2 g, 0.036 mol), anhydrous potassium carbonate (15.0 g, 0.11 mol), copper powder (0.45 g,
0.007 mol), and a catalytic amount of copper(I) chloride in dry DMF (75 mL) using the procedure described in Example 1, Step C, Method B. After crystallization with MeOH/H2O, 4.5 g (0.012 mol, 33%) of the desired product was obtained, mp: 137-139°C. Analysis for ^3^3^04: Calcd: C, 73.19; H, 6.14; N, 3.71. Found: C, 73.47;
H, 6.03; N, 3.78.
Step D: Preparation of 2-{4-[2-(3,4-Dihydroxy-phenyl)-ethyl]-phenylamino}- benzoic acid
To a solution of 2-{4-[2-(3,4-dimethoxy-phenyl)-ethyl]phenylamino}- benzoic acid (0.28 g, 0.74 mmol) in CH2CI2 (20 mL), BBr3 (3.5 mL, 1M in
CH2CI2, 3.5 mmol) was added at room temperature under a N2 atmosphere. The reaction mixture was allowed to stir at room temperature for 2 hours and then poured into ice water (50 mL). This mixture was extracted with EtOAc, and the organic layer was washed two times with water, dried (Na2SO4), filtered and concentrated in vacuo to yield 0.24 g (0.69 mmol, 93%) of the desired product. mp 215-217°C.
Analysis for C2lH19NO4: Calcd: C, 72.19; H, 5.48; N, 4.00. Found: C, 71.80; H, 5.46; N, 3.99.
EXAMPLE 5
Preparation of 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid
Step A (Scheme 1): Preparation of l,l-Dibutylamino-4-[2-(4- nitrophenyl)ethenyl]benzene
The title compound was prepared from^-nitrophenylacetic acid (9.92 g, 0.055 mol) and 4-dibutylamino-benzaldehyde (14.32 g, 0.055 mol) in piperidine
(5 mL) using the procedure described in Example 1 , Step A. This procedure yielded a red solid, 4.12 g (0.012 mol, 16%) of the desired product.
MS: 352.2. (M+); 353.2. (MH+).
Step B (Scheme 1): Preparation of 4-[2-(4,4-Dibutylaminophenyl)ethyl]- phenylamine
The title compound was prepared from l,l-dibutylamino-4-[2-(4- nitrophenyl)ethenyl]benzene (4.10 g, 11.63 mmol) and Ra-Ni (2.0 g) in MeOH (100 mL) at 21°C to 32°C (ΔP = 3.6 psi) under a hydrogen atmosphere using the procedure described in Example 1, Step B. This procedure yielded a colorless oil, 3.49 g (10.76 mmol, 92.6%) of the desired product.
MS: 325.3 (MH+).
Step C (Scheme 6): Preparation of 2-{4-[2-(4-Dibutylamino-phenyl)- ethyljphenylamino} -benzoic acid
The title compound was prepared from 2-chlorobenzoic acid (1.46 g, 9.36 mmol), 4-[2-(4,4-dibutylaminophenyl)ethyl]phenylamine (3.31 g,
10.20 mmol), anhydrous potassium carbonate (4.27 g, 30.88 mmol), copper powder (1.25 g, 19.65 mmol), and copper(I) chloride (0.092 g, 0.93 mmol) in dry DMF (30 mL) using the procedure described in Example 1, Step C, Method B. This procedure yielded a 0.39 g (0.87 mmol, 8.6%) of the desired product. mp l l5-117°C.
Analysis for C29H36N O2: Calcd: C, 78.34; H, 8.16; N, 6.30. Found: C, 78.15; H, 8.07; N, 6.10.
EXAMPLE 6
Preparation of 2-{4-[2-(3,4,5-Trihydroxy-phenyl)-ethyl]phenylamino}benzoic acid
Step A (Scheme 1): Preparation of l,2,3-Trimethoxy-5-[2-(4- nitrophenyl)ethenyl]benzene The title compound was prepared from .-nitrophenylacetic acid (18.6 g,
0.10 mol), 3,4,5-trimethoxy-benzaldehyde (19.6 g, 0.10 mol) and piperidine (5 mL) using the procedure described in Example 1, Step A. This procedure yielded a solid, 13.0 g (0.041 mol, 41%) of the desired product. mp:192-195°C.
Step B (Scheme 1): Preparation of 4-[2-(3,4,5-Trimethoxy-phenyl)ethyl]- phenylamine
The title compound was prepared from l,2,3-trimethoxy-5-[2-(4- nitrophenyl)ethenyl]benzene (9.5 g, 0.03 mol) and Ra-Ni (1.0 g) in THF (50 mL) at 21-26°C (ΔP = 9.6 psi) under a hydrogen atmosphere using the procedure described in Example 1, Step B. This procedure yielded a tan powder, 6.6 g (0.023 mol, 74%) of the desired product, mp 91 -93°C.
Step C (Scheme 6): Preparation of 2-{4-[2-(3,4,5-Trimethoxy-phenyl)- ethyl]phenylamino} -benzoic acid methyl ester
The title compound was prepared from 4-[2-(3,4,5-trmethoxyphenyl)- ethyljphenylamine (0.75 g, 2.61 mmol), methyl 2-bromobenzoate (0.47 g, 2.17 mmol), cesium carbonate (0.99 g, 3.04 mmol), tris(dibenzylideneacetone- dipaladium(O) (0.06 g, 0.065 mmol) and (S)-(-0-2,2'-bis(di-p-tolylphosphino-l,l'- binaphthyl (98% (S)-Tol-BINAP) (0.066 g, 0.098 mmol) (Ligand/Pd = 1.5) in anhydrous toluene (100 mL) using the procedure described in Example 1, Step C, Method A to yield a yellow oil, 0.69 g (1.63 mmol, 76%) of the desired product. Analysis for C25H27N1O5: Calcd: C, 71.24; H, 6.46; N, 3.32. Found: C, 71.53;
H, 6.24; N, 3.14.
Preparation of 2-{4-[2-(3,4,5-Trimethoxy-phenyl)ethyl]phenylamino}-benzoic acid To a solution of 2-{4-[2-(3,4,5-trimethoxyphenyl)ethyl]phenylamino}- benzoic acid methyl ester (0.62 g, 1.47 mmol) in THF-EtOH (2:1, 6 mL), IN NaOH solution (4 mL) was added, and the reaction mixture was heated to reflux for 5 hours. The reaction mixture was then concentrated in vacuo to remove the organic solvent. The residue was acidified with concentrated HCl (pH 3). This precipitate was collected by filtration, triturated with boiling MeOH-H2θ (4: 1) and dried in vacuum at room temperature for 16 hours to give the title compound as a white solid, 0.59 g (1.45 mmol, 98.5%). mp 146.0- 147.0°C. Analysis for C24H25N1O5: Calcd: C, 70.75; H, 6.18; N, 3.44. Found: C, 70.54; H, 6.43; N, 3.15.
Step D: Preparation of 2-{4-[2-(3,4,5-Trihydroxyphenyl)ethyl]phenylamino}- benzoic acid
The title compound was prepared from 2-{4-[2-(3,4,5-trimethoxy-phenyl)- ethyljphenylamino} benzoic acid (0.50 g, 1.23 mmol) in CH2C-2 (40 mL) and
BBr3 (10 mL, 1M in CH2CI2, 10.0 mmol) using the procedure described in Example 4, Step D. This procedure yielded a green solid, 0.25 g (0.68 mmol,
65%) of the desired product, mp: 160.0-162.0°C.
Analysis for C2iHι9NιO5-1.44 H2O: Calcd: C, 64.46; H, 5.64; N, 3.58. Found: C, 64.07; H, 5.27; N, 3.39.
EXAMPLE 7 Preparation of 2- {4-[2- [-(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-methoxy-
5-nitrobenzoic acid Step A' (Scheme 2): Preparation of 3-(3,4-Dichlorophenyl)-l-(4-nitro- phenyl)propenone
Sodium hydroxide (7.3 g, 0.18 mol) was dissolved in water (80 mL) and 95% EtOH (80 mL) and cooled to 10°C with an ice-H2θ bath. 3,4-Dichlorobenzaldehyde (31.8 g, 0.18 mol) was added in one portion. After the addition, the mixture was warmed to 15°C. l-(4-Nitrophenyl)ethanone (30.0 g, 0.18 mol) was added at this temperature with rigorous stirring. After stirring for 5 minutes, the reaction mixture was diluted with 95% EtOH (300 mL). The resulting tan mixture was stirred at room temperature for 30 minutes, then stirred with an ice-H2θ bath underneath the flask for 2 hours. The light brown solid was filtered off, washed with H2O, and air-dried. The solid was dissolved in hot THF
(1.5 L) and treated with charcoal. The resulting mixture was filtered off, and the filtrate was diluted with 95% EtOH (500 mL). This solution was filtered and oven-dried (40°C) to yield a light brown solid, 38.56 g (0.12 mol, 66%) of the title compound, mp 220-223 °C.
Analysis for C15H9C-2NO3: Calcd: C, 55.93; H, 2.82; Cl, 22.01; N, 4.35. Found: C, 55.79; H, 2.93; Cl, 22.16; N, 4.32.
Step B' (Scheme 2): Preparation of l-(4-Amino-phenyl)-3-(3,4- dichlorophenyl)propan- 1 -one 3-(3,4-Dichlorophenyl)-l-(4-nitro-phenyl)propenone (34.56 g, 0.11 mol) was reduced in the presence of Ra-Ni (3.0 g) in THF (250 mL) at 20°C to 32°C (ΔP = 33.4 psi) under a hydrogen atmosphere. The reaction mixture was concentrated in vacuo and recrystallized from MeOH (100 mL) to give a light yellow solid, 23.5 g (0.080 mol, 75%) of the desired product, mp 127-129°C. Analysis for Ci5H13Cl2NO: Calcd: C, 61.24; H, 4.45; N, 4.76; Cl, 24.10.
Found: C, 60.91; H, 4.60; N, 4.70; Cl, 23.98.
Step C (Scheme 2): Preparation of 4-[3-(3,4-Dichlorophenyl)propyl]phenylamine
A mixture of l-(4-aminophenyl)-3-(3,4-dichlorophenyl)propan-l-one (20.0 g, 0.068 mol), NH2NH2-H2O (16 mL), and KOH (85%, 5.6 g) in ethylene glycol (160 mL) was heated to reflux under a N2 atmosphere for 16 hours. After cooling to room temperature, the reaction mixture was poured into ice-H2θ and extracted with CH2CI2 (2 L). The layers were separated, and the organic layer was dried (Na2SO4) and concentrated in vacuo to afford an oil. Purification by flash chromatography (silica gel, CH2CI2) yielded an oil,14.00 g (0.05 mol, 73%) of the desired product.
Analysis for C15H15CI2N: Calcd: C, 64.30; H, 5.40; N, 4.99; Cl, 25.31.
Found: C, 64.21; H, 5.59; N, 5.24; Cl, 24.87.
Preparation of 2,4-Difluoro-5-nitrobenzoic acid methyl ester Fuming nitric acid 90% (8.5 mL, 0.19 mol) was added with gentle stirring to concentrated sulfuric acid 98% (125 mL) in a 1 L beaker. After stirring for 10 minutes at room temperature, 2,4-difluorobenzoic acid methyl ester (21.9 g, 0.127 mol) was added dropwise. After the addition, the reaction mixture was allowed to stir gently for 40 minutes at room temperature. The reaction mixture was then poured into ice-H2θ (1 L) and stirred for 10 minutes. The mixture was extracted with EtOAc. The layers were separated, and the organic layer was washed sequentially with IN NaCl, saturated NaHCO3, H2O and brine, dried
(Na2SO4), filtered and concentrated in vacuo to afford a yellow residue. This residue was washed with 10% EtOAc/hexane, filtered, and dried to yield a pale yellow solid, 29.0 g (0.133 mol, 82%). mp 78-80°C.
Analysis for C8H5F2NO4: Calcd: C, 44.25; H, 2.32; N, 6.45. Found: C, 44.18; H, 2.39; N, 6.14.
Preparation of 2-Fluoro-4-methoxy-5-nitrobenzoic acid methyl ester
A mixture of sodium metal (1.27 g, 0.055 mol) and MeOH ( 250 mL) was stirred at 0°C for 10 minutes. This solution was added to a solution of 2-fluoro-5- nitrobenzoic acid methyl ester (10.0 g, 0.046 mol) in MeOH (250 mL), and the mixture was stirred for 20 minutes at 0°C to 5°C. The reaction mixture was then allowed to warm to room temperature and stir for 2 hours. The mixture was then filtered to give an off-white precipitate. Recrystallization with CHCI3 (70 mL) yielded an off-white crystalline solid, 1.825 g (0.008 mol, 17%) of the title compound. Analysis for C9H8FιN1O5: Calcd: C, 47.17; H, 3.52; N, 6.11. Found: C, 47.09;
H, 3.47; N, 6.00.
Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-4-methoxy-
5-nitrobenzoic acid methyl ester
A mixture of 4-[3-(3,4-dichloro-phenyl)propyl]phenylamine (0.94 g, 3.3 mmol), 2-fluoro-4-methoxy-5-nitro-benzoic acid methyl ester (0.75 g, 3.3 mmol), and Et3N (0.46 mL) in'CT^CN (30 mL) was heated to reflux for 120 hours. The reaction mixture was cooled to room temperature, diluted with
CH C12 and washed with saturated NaHCO3. The organic layer was dried
(Na2SO4) and concentrated to give a solid. Recrystalhzation with MeOH yielded
0.67 g (1.37 mmol, 42%) of the desired product.
Analysis for C 4H22N2C12O5-0.42H2O: Calcd: C, 58.01; H, 4.63. N, 5.64; Found: C, 57.61; H, 4.51; N, 5.94.
Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-4-methoxy-
5-nitrobenzoic acid
To a solution of 2-{4-[3-(3,4-dichlorophenyl)propyl]phenylamino}-
4-methoxy-5-nitrobenzoic acid methyl ester (0.30 g, 0.061 mol) in THF (5 mL), IN NaOH (aq.) (2.5 mL) was added, and the mixture was stirred for 36 hours at room temperature. The solvent was removed, and the residue was acidified with concentrated HCl to pH 3. The precipitate was collected by filtration and dried in vacuum for 16 hours. Recrystalhzation with MeOH gave the title compound as an orange solid 0.21 g (0.043 mol, 70%). mp 200-201°C. Analysis for C23H2oN2O5C-2-0.2H2O: Calcd: C, 57.68; H, 4.29; N, 5.85;
Cl, 14.81. Found: C, 57.71; H, 4.34; N, 5.58; Cl, 14.56. EXAMPLE 8 Preparation of 2-{4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino}-4-imidazo- l-yl-5-nitrobenzoic acid
Preparation of 2-{4-[2-[-(3,4-Dichlorophenyl)propyl]phenylamino}-4-imidazo- l-yl-5-nitrobenzoic acid methyl ester
A mixture of 2,4-difluoro-5 -nitrobenzoic acid methyl ester (1.63 g, 7.5 mmol), imidazole (0.56 g, 8.25 mmol), and Et3N (1.14 mL, 8.25 mmol) in
CH3CN (50 mL) was stirred for 16 hours at room temperature. To this deep orange solution, 4-[3-(3,4-dichlorophenyl)propyl]phenylamine (2.10 g, 7.5 mmol) and triethylamine (Et3N) (1.14 mL, 8.25 mmol) was added, and the mixture was heated to reflux for overnight. The reaction mixture was cooled and concentrated in vacuo to afford a residue. This residue was diluted with CH2C12 and washed with a saturated K2HCO3 solution. The organic layer was dried (Na2SO4), filtered, and concentrated in vacuo to give a crude oil. Purification by flash chromatography (silica gel, 10% EtOAc/hexane) yielded 1.0 g (1.90 mmol, 25%) of the desired product.
MS: 524.1 (M+).
Preparation of 2-{4-[2-[-(3 ,4-Dichlorophenyl)propyl]phenylamino}-4-imidazo- l-l-yl-5 -nitrobenzoic acid The title compound was prepared from 2-{4-[2-[-(3,4- dichlorophenyl)propyl]phenylamino } -4-imidazo- 1 -yl- 5 -nitrobenzoic acid methyl ester (1.0 g, 1.9 mmol), IN NaOH (2.0 mL) in THF (30 mL) using the procedure described in Example 8. This procedure yielded an orange solid, 0.30 g (0.6 mmol, 32%) of the desired product. Analysis for C25H2θC-2N4O4-0.2H2θ: Calcd: C, 58.31; H, 3.99; N, 10.88;
Cl, 13.89. Found: C, 58.34; H, 4.07; N, 10.73; Cl, 13.41. EXAMPLE 9 Preparation of 2-{4-[3-(3,4-Dichlorophenyl)-propyl]phenylamino}-benzoic acid
Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-benzoic acid methyl ester The title compound was prepared from 4-[3-(3,4-dichlorophenyl)propyl]- phenylamine (600 mg, 2.14 mmol), 2-bromobenzoic acid methyl ester (380 mg, 1.78 mmol), cesium carbonate (812 mg, 2.49 mmol), tris(dibenzylideneacetone- dipaladium(O) (49 mg, 0.053 mmol) and (-S)-(2,2'-bis(di-p-tolylphosphino-l,r- binaphthyl (98%, (S)-tol-BINAP) (54 mg, 0.080 mmol) (Ligand/Pd = 1.5) in anhydrous toluene (15 mL) using the procedure described in Example 2, Step C.
This procedure yielded an yellow oil, 0.61 g (1.47 mmol, 69%) of the desired product.
MS: 414 (M+), 416 (MH+).
Analysis for C23H21C12O2N-0.4 H2O: Calcd: C, 65.25; H, 5.23; N, 3.30. Found: C, 65.76; H, 5.18; N, 3.10.
Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-benzoic acid
The title compound was prepared from 2-{4-[3-(3,4-dichlorophenyl)- propyl]phenylamino} benzoic acid methyl ester (0.41 g, 0.99 mmol), IN NaOH (4.0 mL) in EtOH (4 mL) and THF (4 mL) using the procedure described in Example 2. This procedure yielded a yellow solid, 0.32 g (0.80 mmol, 81%) of the desired product, mp 120-126°C.
Analysis for C22H19Cl2O2N 1-0.75 H O: Calcd: C, 64.04; H, 5.00; N, 3.39. Found: C, 64.17; H, 4.69; N, 3.18.
EXAMPLE 10 Preparation of 2-{4-[4-(3,4-Dichlorophenyl)butyl]phenylamino}benzoic acid
Preparation of (tr-.----)-3-(3,4-Dichlorophenyl)-2-propenal
A mixture of 3,4-dichlorobenzaldehyde (140.0 g, 0.8 mol) and acetaldehyde (300 mL) was cooled to 5°C. Potassium hydroxide (5.1 g, 0.091 mol) was dissolved in hot MeOH (40 mL), and the resulting solution was added to the above cooled mixture while maintaining the internal temperature at 25°C to 30°C. The mixture was allowed to stir in ice-H2O bath for 40 minutes and then treated with acetic anhydride (400 mL). After the addition, the mixture was heated to 100°C with stirring for 30 minutes and then cooled to 30°C. To this mixture, 12N HCl/H2O (102 mL/1.2 L) was added, and the resulting mixture was heated to reflux for 30 minutes and then cooled to room temperature. This heterogeneous mixture was filtered and washed with H2O to afford a brown solid.
The crude product was dissolved in EtOAc and washed with H2O, dried (Na SO4), and concentrated to dryness. Recrystallization from hexane/EtOAc (9:1) yielded 76.5 g (0.38 mol, 48%) of the title compound, mp: 91-93°C.
Analysis for C9H6Cl O: Calcd: C, 53.77; H, 3.01; Cl, 35.27. Found: C, 53.75; H, 3.10; Cl, 35.58.
Preparation of (trans), (trans)- 1 ,2-Dichloro-4-[4-(4-nitrophenyl)- 1,3- butadienyl]benzene A mixture of 4-nitro-benzyl bromide (200.0 g, 0.93 mol) and triphenylphosphine (244.0 g, 0.93 mol) in CHCI3 (1.5 L) was heated to reflux for overnight. The reaction mixture was cooled to room temperature, concentrated in vacuo to remove CHCI3 and then suspended in Et O and stirred rigorously.
The suspension was filtered, and the off-white solid was washed with Et2O, dried at 80°C for 16 hours to give 433.0 g (0.91 mol, 98%) of bromo[(4- nitrophenyl)methyl]triphenylphosphorane .A solution of bromo[(4- nitrophenyl)methyl]triphenylphosphorane (100.0 g, 0.23 mol) in dry THF (500 mL) was cooled to 5°C. n-Butyl lithium (n-BuLi) (2.4 M, 96 mL, 0.23 mol) was added dropwise to maintain the temperature between 5°C to 10°C. The cooling bath was then removed, and the reaction mixture was allowed to warm to room temperature. After 4 hours, a solution of (trans)-3-(3,4-dichlorophenyl)- 2-propenal (36.2 g, 0.18 mol) in THF (100 mL) was added dropwise, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was filtered, and the filtrate was concentrated in vacuo to give a residue. Purification by flash chromatography (silica gel, 20% EtOAc/hexane) yielded 16.0 g (0.05 mol, 28%) of the desired product, mp 125-135°C. Analysis for CiόHi 1CI2NO2: Calcd: C, 60.02; H, 3.46; N, 4.37, Cl, 22.15. Found: C, 59.77; H, 3.47; N, 4.40; Cl, 22.39.
Preparation of 4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamine
The title compound was prepared from (trans), (tr< s)-l,2-dichloro- 4-[4-(4-nitrophenyl)-l,3-butadienyl]benzene (15.42 g, 0.048 mol), Ra-Ni (1 g) at 20°C to 26°C (ΔP = 19.3 psi) under a hydrogen atmosphere in THF (75 mL) and MeOH (75 mL) using the procedure described in Example 1, Step B. This procedure yield a solid, 10.97 g (0.037 mol, 78%) of the desired product, mp
50-52°C.
Analysis of C^H^N^: Calcd: C, 65.32; H, 5.82; N, 4.76. Found: C, 65.43; H, 5.84; N, 4.61.
Preparation of 2-{4-[4-(3,4-Dichlorophenyl)butyl]phenylamino}benzoic acid The title compound, mp 98- 105°C, was prepared from 4-[4-(3,4- dichlorophenyl)butyl]phenylamine (0.50 g, 1.7 mmol), 2-chlorobenzoic acid (0.24 g, 1.56 mmol), anhydrous potassium carbonate (0.71 g, 5.15 mmol), copper powder (0.21 g, 3.28 mmol), and copper(I) chloride (0.015 g, 0.15 mmol) in dry DMF (5 mL) using the procedure described in Example 1, Step C, Method B.
EXAMPLE 11
Preparation of 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-nitro- benzoic acid
A mixture of 2-fluoro-5-nitrobenzoic acid (1.85 g, 0.01 mol), 4-[4-(3,4- dichlorophenyl)butyl]-phenylamine (2.94 g, 0.01 mol) and Et3N (2.80 mL) in acetonitrile (110 mL) was heated to reflux for 48 hours. The reaction mixture was cooled and concentrated in vacuo to remove the solvent. The residue was dissolved in CH2CI2 and washed with diluted HCl. The organic layer was dried (Na2SO4), concentrated in vacuo to give a crude solid. Purification by flash chromatography (silica gel, CH2CI2) yielded 1.40 g (0.003 mol, 30%) of the desired product.
Analysis for C23H19N2O4C-2: Calcd: C, 60.27; H, 4.18; N, 6.11; Cl, 14.47.
Found: C, 60.16; H, 4.41; N, 6.09; Cl, 15.69.
EXAMPLE 12
Preparation of 2-{4-[4-(3 ,4-Dichlorophenyl)-butyl]phenylamino} - 3,5-dinitrobenzoic acid
To a cooled (0°C) solution of 4-[4-(3,4-dichlorophenyl)butyl]- phenylamine (1.47 g, 5.0 mmol) and DBU (0.75 mL, 7.5 mmol) in acetonitrile (25 mL), a solution of 2-fluoro-2,5-dinitrobenzoic acid (1.15 g, 5.0 mmol) in acetonitrile (15 mL) was added dropwise. After stirring for 30 minutes at 0°C, the reaction mixture was neutralized with dilute HCl and extracted with EtOAc, dried (Na2SO4), filtered and concentrated in vacuo to afford a crude residue.
Recrystalhzation with EtOH yielded a bright orange solid, 2.06 g (4.1 mmol, 82%) of the title compound.
Analysis for C23H19CI2N3O6- Calcd: C, 54.77; H, 3.80; N, 8.33; Cl, 14.06. Found: C, 54.68; H, 4.00; N, 8.12; Cl, 13.81.
EXAMPLE 13 Preparation of 2-{4-[5-(3,4-Dichlorophenyl)pentyl]phenylamino}-5-nitrobenzoic acid
Preparation of Bromo[(3,4-dichlorophenyl)methyl]triphenylphosphorane
A mixture of 4-bromomethyl-l,2-dichlorobenzene (2.40 g, 0.01 mol), and triphenylphosphine (5.24 g, 0.02 mol) in toluene (30 mL) was stirred for 16 hours at room temperature. The solid was filtered, rinsed with toluene, and oven-dried at room temperature to yield a white powder, 3.95 g (0.0078 mol, 78%) of the desired product. iH NMR [dimethylsulfoxide (DMSO):ppm] -7.89-7.61 (m, 15H), 7.50 (d, J=8.3 Hz, 1H), 7.04 (t, J=2.3 Hz, 1H), 6.97 (m, 1H), 5.20 (d, J=15.9 Hz, 2H). Preparation of 4-(4-Nitrophenyl)butyraldehyde
To a cooled solution (-70°C) of oxalyl chloride (2.0 M in CH2CI2,
14.1 mL, 28.2 mmol), dimethylsulfoxide (DMSO) (4.40 g, 56.32 mmol) in CH2CI2 (20 mL) was added dropwise. The resulting reaction mixture was then stirred for 30 minutes at -70°C under a nitrogen atmosphere. A solution of
4-(4-nitrophenyl)butan-l-ol (5.00 g, 25.6 mmol) in CH2CI2 (3 mL) was added dropwise, and the reaction mixture was stirred for 1 hour at -70°C. Et3N (16 mL,
115 mmol) was added, and the reaction mixture was then allowed to gradually warm to room temperature and stir for 30 minutes. The mixture was then quenched with H2O and extracted with EtOAc. The organic layers were washed with 0.1N HCl solution, H2O, brine, dried (Na2SO4), filtered, and concentrated in vacuo to give a lightly brown oil. Purification by flash chromatography (silica gel, 50% EtOAc/hexane) yielded 3.20 g (16.56 mmol, 65%) of the desired product. !H NMR (DMSO:ppm): 9.75 (s, 1H), 8.12 (d, J=8.3 Hz, 2H), 7.30 (d, J=8.3 Hz,
2H), 2.72 (t, J=7.7 Hz, 2H), 2.47 (t, J=7.1 Hz, 2H), 1.94 (m, 2H).
Preparation of 1 ,2-Dichloro-4-[5-(4-nitrophenyl)- 1 -pentenyl] benzene
A solution of bromo[(3,4-dichlorophenyl)methyl]triphenylphosphorane (3.95 g, 7.9 mmol) in dry THF (20 mL) was cooled to 0°C. LHDMS (1.0 M/THF, 9 mL, 9.0 mol) was added dropwise to maintain the temperature at 0°C. After stirring for 30 minutes, a solution of 4-(4-nitro-phenyl)butyraldehyde (1.45 g, 7.5 mmol) in THF (5 mL) was added dropwise, and the mixture was allowed to warm to room temperature within 2 hours. The mixture was then quenched with H2O and extracted with EtOAc. The organic layers were washed with 0.1N HCl solution, H2O, brine, dried (Na2SO4), filtered, and concentrated in vacuo to give a lightly brown oil. Purification by flash chromatography (silica gel, 10% EtOAc/hexane) yielded 2.5 g (7.4 mmol, 99%) of the desired product.
MS: 335 (M+), 337 (MH+). Preparation of4-[5-(3,4-Dichlorophenyl)pentyl]phenylamine
The title compound was prepared from l,2-dichloro-4-[5-(4-nitrophenyl)- l-pentenyl]benzene (2.5 g, 7.4 mmol), Ra-Ni (1 g) in THF ( 50 mL) at 25°C to 40°C (ΔP = 9.9 psi) using the procedure described in Example 1, Step B. This procedure yielded 1.06 g (3.4 mmol, 46%) of the desired product.
!H NMR (DMSO-ppm): 7.45 (d, J=8.3 Hz, 1H), 7.41 (d, J=2.2 Hz, 1H), 7.12 (m, 1H), 6.74 (d, J=8.3 Hz, 2H), 6.40 (d, J=8.3 Hz, 2H), 4.73 (s, 2H), 2.50 (t, J=7.7 Hz, 2H), 2.31 (t, J=7.6 Hz, 2H), 1.6-1.5 (m, 4H), 1.5-1.4 (m, 2H).
Preparation of 2-{4-[5-(3,4-Dichloro-phenyl)pentyl]phenylamino}-5-nitrobenzoic acid
To a cooled (-78°C) solution of 4-[5-(3,4-dichlorophenyl)pentyl]- phenylamine (0.231 g, 0.75 mmol) in THF (2 mL), LHDMS (2.25 mL, 1 M in hexane, 2.25 mmol) was added dropwise. The reaction mixture was allowed to stir at -78°C for 10 minutes. A solution of 2-fluoro-5-nitrobenzoic acid (0.139 g, 0.75 mmol) in THF (2 mL) was added dropwise, and this solution was stirred for
30 minutes at -78°C. The reaction mixture was allowed to gradually warm to room temperature and stir for 2 hours under N2 atmosphere. The reaction mixture was diluted with EtOAc, and acidified with IN HCl (pH 3). The organic layer was dried (Na2SO4), filtered and concentrated in vacuo to yield a brown residue. Purification by flash chromatography (silica gel, 2% MeOH/CH2Cl2) then recrystalhzation with MeOH yielded 265 mg (0.56 mmol, 75%) of the desired product, mp 147-148°C.
Analysis for 024^20-2^04-0.37^0: Calcd: C, 60.05; H, 4.77; N, 5.84. Found: C, 59.67; H, 4.64; N, 5.51. EXAMPLE 14 Preparation of 2-{4-[5-(3,4-Dichloro-phenyl)pentyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid
Preparation of 2- {4-[5-(3 ,4-Dichlorophenyl)pentyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid methyl ester
The title compound was prepared from 4- [5 -(3,4- dichlorophenyl)pentyl]phenylamine (231 mg, 0.75 mmol), LHDMS (6.28 mL, 1 M in THF, 6.28 mmol) and 2-fluoro-4-methoxy-5-nitrobenzoic acid methyl ester (172 g, 0.75 mmol) in THF (5 mL) using the procedure described in Example 13. Purification by flash chromatography (silica gel, 10% EtOAc/hexane) yielded
145 mg (0.28 mmol, 37%) of the desired product.
MS: 515.2 (M+), 517.2 (MH+).
Preparation of 2-{4-[5-(3,4-Dichlorophenyl)pentyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid The title compound was prepared from 2-{4-[5-(3,4-dichlorophenyl)- pentyl]phenylamino}-4-methoxy-5-nitrobenzoic acid methyl ester (145 mg, 0.28 mmol) and IN NaOH (aq.) (0.56 mL) in THF (1.2 mL) using the procedure described in Example 2. Purification by flash chromatography (silica gel, 10% MeOH/CH2θ2), then recrystalhzation with MeOH yielded 58 mg (0.12 mmol, 41%) of the desired product, mp 192-193°C.
Analysis for C25H24CI2N2O5: Calcd: C, 59.65; H, 4.81; N, 5.56. Found: C, 59.29; H, 4.58; N, 5.36.
EXAMPLE 15 Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid
Preparation of 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid methyl ester
The title compound was prepared from 4-[3-(3,4-dichlorophenyl)propyl]- phenylamine (420 mg, 1.50 mmol), 2-bromobenzoic acid methyl ester (310 mg, 1.25 mmol), cesium carbonate (569 mg, 1.75 mmol), tris(dibenzylideneacetone- dipaladium(O) (34 mg, 0.037 mmol) and (5)-(2,2'-bis(di-p-tolylphosphino-l, - binaphthyl (98%, (S)-tol-BINAP) (38 mg, 0.056 mmol) (Ligand/Pd=1.5) in anhydrous toluene (15 mL) using the procedure described in Example 2, Step C. This procedure yielded an orange solid 0.51 g (1.11 mmol, 74%) of the desired product, mp 117-118°C.
MS: 457.1 (M+); 459.1 (MH+)
Preparation of 2- {4-[3 -(3 ,4-Dichlorophenyl)propyl]phenylamino } -5-nitrobenzoic acid The title compound was 2-{4-[3-(3,4-dichlorophenyl)- propyl]phenylamino}-5-nitrobenzoic acid methyl ester (0.50 g, 1.09 mmol), 2N NaOH (5.0 mL) in EtOH (2 mL) and THF (4 mL) using the procedure described in Example 2. This procedure yielded an orange solid, 0.49 g (1.10 mmol, 100%) of the desired product, mp 153-155°C. MS: 443.2 (M+), 445.2 (MH+)
EXAMPLE 16 Preparation of 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid
Preparation of 2- {4- [2-(3 ,4-Dimethyl-phenyl)-ethyl]phenylamino } -5-nitrobenzoic acid methyl ester
The title compound was prepared from 4-[2-(3,4-dimethylphenyl)ethyl]- benzenamine (1.0 g, 4.43 mmol), 2-bromo-5 -nitrobenzoic acid methyl ester
(0.96 g, 3.69 mmol), cesium carbonate (1.68 g, 5.17 mmol), tris(dibenzylideneacetone-dipaladium(0) (101 mg, 0.11 mmol) and (S)-(2,2'- bis(di-p-tolylphosphino-l,l'-binaphthyl (98%, (S)-tol-BINAP) (113 mg,
0.17 mmol) (Ligand/Pd = 1.5) in anhydrous toluene (32 mL) using the procedure described in Example 2, step C. This procedure yielded an yellow solid, 1.31 g
(3.24 mmol, 73%) of the desired product, mp 1 15-117°C.
MS: 405 (M+) Analysis for C24H24O4N2-0.25 H2O: Calcd: C, 71.27; H, 5.98; N, 6.93.
Found: C, 70.48; H, 6.03; N, 6.85.
Preparation of 2- {4- [2-(3 ,4-Dimethyl-phenyl)-ethyl]phenylamino } -5-nitrobenzoic acid The title compound was prepared 2-{4-[2-(3,4-dimethyl-phenyl)- ethyl]phenylamino} -5-nitrobenzoic acid methyl ester (1.12 g, 2.76 mmol), IN NaOH (50 mL) in EtOH (50 mL) and THF (50 mL) using the procedure described in Example 2. This procedure yielded a yellow solid, 1.03 g (2.63 mmol, 81%) of the desired product, mp 214-216°C. Analysis for C23H22O4N2-0.25 H2O: Calcd: C, 69.99; H, 5.74; N, 7.18.
Found: C, 69.90; H, 5.82; N, 6.81.
EXAMPLE 17 Preparation of 2- [ [4- [2-(4-Chloro-3 -trifluromethylphenyl)ethyl]phenyl] aminobenzoic acid
Step A (Scheme 1): Preparation of trans - 1 -Chloro-2-trifluoromethyl-4-[2-(4- nitrophenyl)ethenyl]benzene
A mixture of p-nitrophenylacetic acid (51.85 g, 0.29 mol) and 4-chloro-3- trifluoromethylbenzaldehyde (47.85 g, 0.23 mol) in piperidine (19.5 g, 0.23 mol) was heated under N2 atmosphere to 150°C tol60°°C for 1 hour. The reaction mixture was cooled to 80°C to 100°°C and refluxing /-PrOH (150 mL) was added.
The mixture was continued to cool to room temperature and then placed under refrigeration for 5 hours. The crystalline precipitate was filtered off, rinsed with cold /-PrOH, and dried at room temperature in a vacuum oven overnight to yield trans - 1 -chloro-2-trifluoromethyl-4- [2-(4-nitrophenyl)ethenyl]benzene as an orange solid, 22.53 g (68.75 mmol, 30%). mp 173-174°C.
MS: 327.0 (M+)
Step B (Scheme 1): Preparation of 4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]- benzenamine The title compound was prepared from tra«-y-l-chloro-2-trifluoromethyl-4- [2-(4-nitrophenyl)ethenyl]benzene (22.53 g, 0.069 mol) and Ra-Ni (22 g) in THF (0.5 L) at 18°C to 29°C (ΔP = 20.5 psi) under a hydrogen atmosphere using the procedure described in Example 1, Step B. This procedure yielded a white solid, 20.0 g (66.73 mmol, 97%) of the desired product, mp 62-64°C.
MS: 298.1 (M+)
Preparation of 2-[[4-[2-(4-Chloro-3-trifluromethylphenyl)ethyl]phenyl]- aminobenzoic acid
To a cold solution of 4-[2-(4-chloro-3-trifluoromethylphenyl)ethyl]- benzenamine (4.33 g, 14.45 mmol) in THF (50 mL) at -78°C, was added LHMDS
(43.35 mL, 43.35 mmol) (1 M/THF) dropwise. Allowed the reaction mixture to stir for 10 minutes at -78°C. A solution of 2-fluorobenzoic acid (2.02 g, 14.45 mmol) in THF (50 mL) was added dropwise. The mixture was stirred for 2 hours at -78°C, then warmed to room temperature and let stir for additional 3 hours. The reaction mixture was concentrated in vacuo (40°C) to remove the organic solvent.
This residue was acidified to pH 3 with 3N HCl (aq.). This precipitate was collected by filtration, rinsed with 10% HCl (40 mL), and dried in vacuum for overnight to give as a pale solid , 4.3 g (10.24 mmol, 70%) of the desired product, mp 150-152°C. Analysis for C22Hi7O2N1ClF3-0.59 H2O: Calcd: C, 61.39; H, 4.26; N, 3.25.
Found: C, 61.01; H, 4.34; N, 3.30.
EXAMPLE 18 Preparation of 2-[4-(3,4-Dichlorophenyl)phenylamino]benzoic acid
Preparation of o-Bromobenzoic acid potassium salt To a solution of o-bromobenzoic acid (201.03 g, 1.0 mol) in MeOH
(500 mL), K-2CO3 (69 g, 1.0 mol) was added. The mixture was concentrated to give the desired product (239.1 g, 1.0 mol, 100%). Preparation of 2-[(4-Iodophenyl)amino] benzoic acid
A mixture of o-bromobenzoic acid potassium salt (47.8 g, 0.2 mol), 4-iodoaniline (43.8 g, 0.2 mol), K2CO3 (13.8 g, 0.1 mol), and cupric acetate
(2.87 g, 6%) in diglyme (100 mL) was heated to reflux for 30 minutes. The reaction mixture was diluted with H2O (1.0 L) and filtered. The filtrate was acidified with diluted AcOH. The resulting precipitate was collected by filtration, washed with H2O and dried in a vacuum at 50°C for 16 hours. Recrystalhzation from EtOAc gave the desired product, a solid (29.7 g, 0.087 mol, 44%). mp 205-206°C. Analysis for Ci3H10N1O2I: Calcd: C,45.05; H, 2.97; N, 4.13. Found: C, 45.05;
H, 2.97; N, 3.92.
Preparation of 2-[4-(3,4-Dichlorophenyl)phenylamino]benzoic acid
A mixture of 3,4-dichlorophenylboronic acid (880 mg, 2.3 mmol), 2-[(4- iodophenyl)amino]benzoic acid (339 mg, 1 mmol), PdCl2-dpp CH2Cl2 [1,1'- bis(diphenylphosphino)ferrocene palladium (II) chloride, complexed with dichloromethane (1:1)] (67 mg, 0.082 mmol), K-2CO3 (829 mg, 6 mmol), and
H2O (2 mL) in dioxane (15 mL) was heated to reflux for 1 hour. The reaction mixture was diluted with EtOAc and filtered. The filtrate was treated with IN HCl, washed with H2O, brine, dried (Na2SO4), and concentrated in vacuum to give a yellow solid. Purification by flash chromatography (silica gel, 10%
MeOH/CH2Cl2) yielded 272 mg (0.76 mmol, 76%) of the desired product. mp >220°C.
Analysis for Ci9H13O2N1Cl2: Calcd: C, 63.23; H, 3.71; N, 3.88.
Found: C, 62.95; H, 3.73; N, 3.63. By following the general procedures described above, the following additional invention compounds were prepared: EXAMPLE 19 2-{4-[3-(4-Diethylaminophenyl)propyl]phenylamino}benzoic acid
MS: 403 (M+).
Analysis for C26H3θN2θ2-0.40 mol H2O: Calcd: C, 69.31; H, 6.87; N, 6.12. Found: C, 69.29; H, 7.04; N, 6.35.
EXAMPLE 20 2-{4-[3-(4-Nitrophenyl)propyl]phenylamino}benzoic acid, mp 150-153°C.
MS: 376 (M+).
EXAMPLE 21 2-{4-[3-(3-Nitrophenyl)propyl]phenylamino}benzoic acid, mp 164-167°C.
MS: 376 (M+).
Analysis for 022^0^04-2.20 mol H2O: Calcd: C, 63.51; H, 5.91; N, 6.73.
Found: C, 63.56; H, 5.45; N, 6.46.
EXAMPLE 22 2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid, mp 110-112°C.
MS: 347 (M+1+).
EXAMPLE 23 2-{4-[3-(3-Aminophenyl)propyl]phenylamino}benzoic acid, mp 109°C.
MS: 333 (M+1+).
EXAMPLE 24
2-{4-[2-(4-Aminophenyl)phenylamino}benzoic acid, mp 198-201°C.
MS: 333 (M+1+).
Analysis for C ιH2θN2θ2-0.1 mol H2O: Calcd: C, 75.47; H, 6.09; N, 8.38.
Found: C, 75.32; H, 6.12; N, 8.27. Found: C, 75.32; H 6.12; N, 8.27. EXAMPLE 25 2- {4-[2-(4-Dipropylaminophenyl)ethyl]phenylamino } benzoic acid monohydrochloride. mp 176-177°C
MS: 417 (M+1+). Analysis for C27H32N2O2: Calcd: C, 71.59; H, 7.34; N, 6.18; Cl, 7.83. Found: C,
71.31; H, 7.24; N, 6.19; Cl, 7.74.
EXAMPLE 26 2-{4-[2-(4-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride monohydrate MS: 389 (M+1+).
Analysis for C25H28N2O2-HCl-H2O: Calcd: C, 67.78; H, 7.05; N, 6.32; Cl, 8.00.
Found: C, 67.83; H, 7.01; N, 6.30; Cl, 7.75.
EXAMPLE 27 2-{4-[3-(3-Dipropylaminophenyl)propyl]phenylamino}benzoic acid MS: 431 (M+1+).
Analysis for C28H34N2O2-0.2 H2O: Calcd: C, 77.46; H, 7.99; N, 6.45.
Found: C, 77.43; H, 7.86; N, 6.40.
EXAMPLE 28 2- {4-[3-(3 -Dimethylaminophenyl)propyl]phenylamino } benzoic acid. mp l l5-117°C.
MS: 374 (M+), 375 (M+1+).
Analysis for 024^6^02-0.1 H2O: Calcd: C, 76.61; H, 7.02; N, 7.44. Found: C,
76.57; H, 7.21; N, 7.47.
EXAMPLE 29 2-{4-[3-(4-Ethylaminophenyl)propyl]phenylamino}benzoic acid, mp 133°C.
MS: 375 (M+1+). Analysis for C 4H26N2O2-0.1 H2O: Calcd: C, 76.61; H, 7.02; N, 7.44. Found: C,
76.62; H, 7.06; N, 7.36.
EXAMPLE 30 2-(N-{4-[3-(4-Diethylaminophenyl)propyl]phenyl}-N-ethylamino)benzoic acid MS: 431 (M+1+).
Analysis for C28H34N2O2: Calcd: C, 78.10; H, 7.96; N, 6.51. Found: C, 78.02;
H, 8.17; N, 6.50.
EXAMPLE 31 2- { 4- [2-(3 -Dibenzylaminophenyl)ethyl]phenylamino } benzoic acid. mp 95.5-97.5°C.
Analysis for C35H32N2O2: Calcd: C, 82.00; H, 6.29; N, 5.46. Found: C, 81.81; H, 6.58; N, 5.44.
EXAMPLE 32 2- {4-[3 -(3 -Diethylaminophenyl)propyl]phenylamino } benzoic acid MS: 403 (M+1+).
Analysis for C26H3θN2θ2-0.1 H2O: Calcd: C, 77.23; H, 7.53; N, 6.93. Found: C,
77.14; H, 7.82; N, 6.88.
EXAMPLE 33 2-{4-[2-(3-Aminophenyl)ethyl]phenylamino}benzoic acid, mp 182-184°C. MS: 333 (M+1+).
Analysis for C2ιH2oN2θ2-0.25 H2O: Calcd: C, 74.87; H, 6.13; N, 8.43.
Found: C, 74.86; H, 6.16; N, 8.32.
EXAMPLE 34
2-{4-[3-(4-Dimethylaminophenyl)propyl]phenylamino}benzoic acid MS: 375 (M+1+).
Analysis for 024^6^02-0.1 H O: Calcd: C, 76.61; H, 7.02; N, 7.44. Found: C, 76.52; H, 7.22; N, 7.49. EXAMPLE 35 2-{4-[2-(4-Acetylaminophenyl)ethyl]phenylamino}benzoic acid, mp 224°C.
MS: 375 (M+1+).
EXAMPLE 36 2-{4-[2-(3-Acetylaminophenyl)ethyl]phenylamino}benzoic acid, mp 213-215°C.
MS: 375 (M+1+).
EXAMPLE 37 2- {4- [2-(3 -Dipropylaminophenyl)ethyl]phenylamino } benzoic acid monohydrochloride. mp 189-193°C. MS: 417 (M+1+).
Analysis for C27H32N2O2ΗCI: Calcd: C, 71.58; H, 7.34; N, 6.18; Cl, 7.83.
Found: C, 71.48; H, 7.35; N, 6.10; Cl, 7.66.
EXAMPLE 38 2-{4-[2-(3-Dibutylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride. mp 175- 180°C.
MS: 445 (M+).
Analysis for C29H36N2O2ΗCI: Calcd: C, 72.40; H, 7.75; N, 5.82; Cl, 7.37.
Found: C, 72.61; H, 7.95; N, 5.78; Cl, 7.23.
EXAMPLE 39 2-{4-[3-(4-Acetylaminophenyl)propyl]phenylamino}benzoic acid, mp 176-178°C.
MS: 389 (M+1+).
EXAMPLE 40 2-{4-[3-(3-Acetylaminophenyl)propyl]phenylamino}benzoic acid, mp 140-145°O
MS: 389 (M+1+). EXAMPLE 41 2- {4- [2-(3 -Diethylaminophenyl)ethyl]phenylamino } benzoic acid monohydrochloride. mp 166-171°C.
MS: 389 (M+1+). Analysis for C25H28 2O2ΗCI: Calcd: C, 70.66; H, 6.88; N, 6.59; Cl, 8.34.
Found: C, 70.48; H, 6.89; N, 6.57; Cl, 18.39.
EXAMPLE 42 2-{4-[2-(3-Piperidin-l-ylphenyl)ethyl]phenylamino}benzoic acid monohydrochloride. mp 187-193°C. MS: 401 (M+1+).
Analysis for C26H28N2O2ΗCI: Calcd: C, 71.46; H, 6.69; N, 6.41; Cl, 8.11.
Found: C, 71.28; H, 6.73; N, 6.35; Cl, 8.30.
EXAMPLE 43 2- {4-[3-(4-Dipropyl--minophenyl)propyl]phenylamino } benzoic acid MS: 431 (M+1+).
Analysis for C28H34N2O2: Calcd: C, 78.10; H, 7.96; N, 6.51. Found: C, 77.91; H, 8.03; N, 6.43.
EXAMPLE 44 2- {4-[3 -(4-Dibutylaminophenyl)propyl]phenylamino } benzoic acid MS: 459 (M+1+).
Analysis for C3oH38N2O2: Calcd: C, 78.56; H, 8.35; N, 6.11. Found: C, 78.40;
H, 8.50; N, 6.19.
EXAMPLE 45 2-{4-[3-(3 -Dibutylaminophenyl)propy 1] phenylamino } benzoic acid MS: 459 (M+1+).
Analysis for C3oH38N2O2: Calcd: C, 78.56; H, 8.35; N, 6.11. Found: C, 78.40; H, 8.43; N, 6.11. EXAMPLE 46 2-(4- { 3 - [4-( 1 H-Pyrrol- 1 -yl)phenyl]propyl } phenylamino)benzoic acid, mp 131-136°C.
MS: 397 (M+1+). Analysis for C26H24N O2-0.2 H2O: Calcd: C, 78.05; H, 6.15; N, 7.00. Found: C,
77.95; H, 6.17; N, 7.08.
EXAMPLE 47 2- {4-[3-(4-Piperidin- 1 -ylphenyl)propyl]phenylamino}benzoic acid
MS: 415 (M+1+). Analysis for C27H3oN2O2-0.2 H2O: Calcd: C, 77.55; H, 7.33; N, 6.70. Found: C,
77.37; H, 7.35; N, 6.63.
EXAMPLE 48 2- {4- [3 -(4-Diethylcarbamoylphenyl)propyl]phenylamino } benzoic acid, mp 57-62°C. MS: 431 (M+1+).
Analysis for C27H3oN2O3-0.3 H2O: Calcd: C, 74.39; H, 7.07; N, 6.43. Found: C,
74.23; H, 6.97; N, 6.27.
EXAMPLE 49 2-{4-[3-(4-Carboxyphenyl)propyl]phenylamino}benzoic acid, mp 236-239°C. MS: 375 (M+).
EXAMPLE 50 2-{4-[3-(4-Diethylaminomethylphenyl)propyl]phenylamino}benzoic acid. mp 137°C.
MS: 417 (M+1+).
EXAMPLE 51
2- {4-[3 -(4-Propylaminophenyl)propyl]phenylamino } benzoic acid MS: 389 (M+1+). Analysis for C25H28 2O2-0.2 H2O: Calcd: C, 76.58; H, 7.30; N, 7.14. Found: C, 76.61; H, 7.29; N, 7.03.
EXAMPLE 52 2- {4- [3 -(3 -Propylaminophenyl)propyl]phenylamino } benzoic acid MS: 389 (M+1+).
Analysis for 025^8^02-0.1 H O: Calcd: C, 76.93; H, 7.28; N, 7.18. Found: C,
76.85; H, 7.44; N, 7.06.
EXAMPLE 53 2- {4- [3 -(4-Pyrrolidin- 1 -yl-phenyl)-propyl] -phenylamino } -benzoic acid. mp 171-177°C.
MS: 401 (M+1+).
Analysis for C26H28N2°2-°- H20: Calcd: C, 77.27; H, 7.08; N, 6.93. Found: C,
77.09; H, 6.97; N, 6.96.
EXAMPLE 54 2-{4-[3-(3 -Piperidin- 1 yl-phenyl)-propyl] -phenylamino } -benzoic acid, mp 59-61°C.
MS: 415 (M+l+).
Analysis for C 7H3oN2O2-0.3 H2O: Calcd: C, 77.22; H, 7.34; N, 6.67. Found: C,
77.18; H, 7.25; N, 6.49.
EXAMPLE 55
{5-[(l-Butyl-l,2,3,4-tetrahydro-6-quinolyl)methylidene]-4-oxo-2- thioxothiazolidin-3-yl} acetic acid, mp 222-224°C.
MS: 391 (M+1+).
EXAMPLE 56 {5-[(l-Butyl-2,3-dihydro-lH-indol-5-yl)methylidene]-4-oxo-2-thioxothiazolidin-
3-yl}acetic acid, mp >250°C.
MS: 377 (M+1+). Analysis for Cι8H2θN2θ3S2-0.4 H2O: Calcd: C, 56.34; H, 5.46; N, 7.30;
S, 16.71. Found: C, 56.27; H, 5.18; N, 7.31; S, 16.74.
EXAMPLE 57 3-{ 5-[( 1 -Butyl- 1 ,2,3,4-tetrahydroquinolin-6-yl)methylidene]-4-oxo-2-thioxo- thiazolidin-3-yl}propanoic acid, mp 214-215°C.
MS: 405 (M+1+).
EXAMPLE 58 4- { 5- [( 1 -Butyl- 1 ,2,3 ,4-tetrahydroquinolin-6-yl)methylidene]-4-oxo-2-thioxo- thiazolidin-3-yl}butanoic acid, mp 152-154°C. MS: 417 (M"l+), 418 (M+), 419 (M+1+).
Analysis for C2iH26N2°3s2'0- H20: Calcd: C, 59.74; H, 6.30; N, 6.64; S, 15.19. Found: C, 59.59; H, 6.16; N, 6.52; S, 15.38.
EXAMPLE 59 2-{4-[3-(3,4-Dichloro-phenyl)-propyl]phenylamino}-5-methyl-benzoic acid. mp 98-99°C.
MS: 414 (M+).
EXAMPLE 60 N-(2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-benzoyl)- methanesulfonamide was prepared by reacting the product from Example 9 with methanesulfonamide. mp 53-61 °C.
Analysis for C23H22Cl2N2O3S-0.13 H2O: Calcd: C, 57.58; H, 4.68; N, 5.84. Found: C, 57.20; H, 4.66; N, 5.51.
EXAMPLE 61 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid. mp 214-216°C.
Analysis for C23H22N2O4-0.25 H2O: Calcd: C, 69.99; H, 5.74; N, 7.18. Found: C, 69.90; H, 5.82; N, 6.81. EXAMPLE 62 2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid, mp 239-244°C.
MS: 439 (MH+).
EXAMPLE 63 2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid, mp 207-209°C.
Analysis for C22H16CIF3N2O4: Calcd: C, 56.85; H, 3.47; N, 6.03. Found: C, 56.75; H, 3.71; N, 5.83.
EXAMPLE 64 5-Amino-2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid was prepared by reacting the product from Example 2 with hydrogen gas in the presence of Raney nickel, mp 137-142°C.
Analysis for C2iH18C_2N2O2-0.96 mol THF: Calcd: C, 63.94; H, 4.72; N, 6.00. Found: C, 64.33; H, 4.91; N, 6.35.
EXAMPLE 65
5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid, mp 198-202°C. Analysis for C2ιH18N2O4-0.11 H2O: Calcd: C, 69.22; H, 5.04; N, 7.69. Found: C, 69.59; H, 5.27; N, 7.22.
EXAMPLE 66 2-{4-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}-benzoic acid, mp 148-150°C.
Analysis for C22H17F4NO2: Calcd: C, 65.51; H, 4.25; N, 3.47. Found: C, 65.51; H, 4.13; N, 3.46. EXAMPLE 67 2-{4-[2-(3 ,4-Difluoro-phenyl)-ethyl] -phenylamino } -5 -nitro-benzoic acid mp 203-208°C.
Analysis for C2iH16F2N2θ4: Calcd: C, 63.32; H, 4.05; N, 7.03. Found: C, 62.94; H, 4.37; N, 6.87.
EXAMPLE 68 {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenyl } - [2-( 1 H-tetrazol-5 -yl)-phenyl] -amine was prepared as described in Example 1 , using a tetrazole fluoro intermediate that was synthesized from commercially available 2-fluorobenzonitrile and sodium azide under standard reaction conditions, mp 129 shrink, 152-157°C.
Analysis for C21H17CI2N5O.I5 EtOAc-0.15 Hexane: Calcd: C, 61.80; H, 4.64; N, 16.12. Found: C, 61.61; H, 4.28; N, 15.83.
EXAMPLE 69 2-{4-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid, mp 190-193°C.
Analysis for C22H16F4N2O4: Calcd: C, 58.93; H, 3.60; N, 6.25. Found: C, 58.69; H, 3.42; N, 6.57.
EXAMPLE 70 2-(4-Phenethyl-phenylamino)-benzoic acid, mp 173-182°C. Analysis for C2ιH19NO2: Calcd: C, 79.47; H, 6.03; N, 4.41. Found: C, 79.42;
H, 5.97; N, 4.47. Found: C, 79.59; H, 6.03; N, 4.50.
EXAMPLE 71 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-fluoro-benzoic acid, mp 180-182°C. Analysis for C2iH16Cl2FNO2-0.06 H O: Calcd: C, 62.23; H, 4.01; N, 3.46.
Found: C, 61.83; H, 4.04; N, 3.29. EXAMPLE 72 2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -nicotinic acid, mp 168-171°C.
Analysis for C2θHi6Cl2N2O2: Calcd: C, 62.03; H, 4.16; N, 7.23. Found: C, 62.11; H, 4.17; N, 7.07.
EXAMPLE 73 2-{4-[2-(3-Chloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid, mp 192.5-194.5°C.
Analysis for C2iH17ClN2O4: Calcd: C, 63.56; H, 4.32; N, 7.06. Found: C, 63.83; H, 4.62; N, 6.79.
EXAMPLE 74 2-{4-[2-(4-Chloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid. mp 210-212°C.
Analysis for C2iH17ClN2O4-0.26 H2O: Calcd: C, 62.82; H, 4.40; N, 6.98. Found: C, 62.51; H, 4.34; N, 6.58.
EXAMPLE 75 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl-benzoic acid, mp 153-160°C.
Analysis for C22Hi9Cl2NO2-0.61 H2O: Calcd: C, 64.25; H, 4.96; N, 3.41. Found: C, 63.87; H, 4.64; N, 3.55.
EXAMPLE 76 2-{4-[2-(2-Chloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid. mp 236-238°C.
EXAMPLE 77 2-{4-[2-(2,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid, mp 200.5-202.5°C. Analysis for C2iH16Cl2N2O4: Calcd: C, 58.49; H, 3.74; N, 6.50.
Found: C, 58.33; H, 3.67; N, 6.29.
EXAMPLE 78 2- { 4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-trifluoromethyl-benzoic acid, mp 130-132°C.
Analysis for C22H1 C-2F3NO2: Calcd: C, 58.17; H, 3.55; N, 3.08. Found: C, 58.25; H, 3.65; N, 3.05.
EXAMPLE 79 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid. mp >260°C.
EXAMPLE 80 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-dimethyl--mino-benzoic acid, mp 75-80°C.
EXAMPLE 81 2-{4-[2-(3,5-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid, mp 191-194°C.
Analysis for C2iH17Cl2NO2: Calcd: C, 65.30; H, 4.44; N, 3.63. Found: C, 65.38; H, 4.29; N, 3.52.
EXAMPLE 82 2-(4-{2-[(4aS,8aR)-4-(Octahydro-isoquinolin-2-yl)-phenyl]-ethyl}-phenylamino)- benzoic acid was prepared according to Example 1 using a decahydroisoquinoline aldehyde which was prepared from trans-decahydroisoquinoline and para- fluorobenzaldehyde under standard reaction conditions, mp 203-206°C. Analysis for C30H34N2O2 .I2 H2O: Calcd: C, 78.89; H, 7.56; N, 6.13. Found: C, 78.49; H, 7.58; N, 5.90.
The following examples are prepared according to the foregoing methods, or by utilizing standard combinatorial synthetic methodology by reacting halo substituted benzoate esters with a substituted aniline to form the corresponding diarylamine, followed by saponification to the benzoic acid of Formula I. The reactions are carried out on 0.15 mmol scale as follows. Solutions of each halo benzoate reactant (0.18 M) in toluene are placed in 2 dram reaction vials. Each aniline reactant is dissolved in anhydrous toluene to give 0.15 M solutions. A Distriman pipet is used to add 1 mL (0.15 mmol, 1 eq) of each halo benzoate solution to the appropriate vials containing 1 mL (0.18 mmol, 1.2 eq) of the aniline reactants. A catalyst solution is prepared by dissolving 0.025 M of
Pd2(dba)3 (dipalladium-tridibenzylidene acetone) and 0.075 M of BINAP (2,2 - bis(diphenylphosphino)-l,l'-binapthyl) in toluene, and 0.25 mL of the catalyst solution is added to each reaction vial. A base, generally cesium carbonate
(68 mg, 0.21 mmol, 1.40 eq) is added to each reaction vial, and the vials are capped and placed in a shaker oven and heated at 100°C for 48 hours. The reaction mixtures are then cooled, and the reaction solvents are removed by evaporation. The solid residue is suspended in 400 μL of ethyl acetate and filtered to remove all catalyst. The filtrates are concentrated to dryness by evaporation to provide compounds of Formula I, wherein the benzoic acid portion is esterified (e.g., benzyl or methyl ester). The esters are dissolved in 500 μL of THF/ethanol (1 :1 v/v) to which is added 300 μL of 5 M sodium hydroxide. The solutions -ire shaken for 5 hours at 60°C and then cooled and concentrated to dryness by evaporation of the solvents to provide the desired compounds of Formula I.
Typical compounds prepared by this method are as follows. The structure of the compounds are generally confirmed by mass spectral analysis.
EXAMPLE 83 2-(3 ,5'-Dichloro-3-methyl-biphenyl-4-ylamino)-benzoic acid MS: 371; MW: 372.2495.
EXAMPLE 84 2-(3',5'-Dibromo-3-methyl-biphenyl-4-ylamino)-benzoic acid MS: 459; MW: 461.1515. EXAMPLE 85 -(4- 1 ,3-Benzodioxol-5-yl-2-methyl-phenylamino)-benzoic acid S: 347; MW: 347.3683.
EXAMPLE 86 -(2,2/,4'-Trichloro-biphenyl-4-ylamino)-benzoic acid S: 391; MW: 392.6678.
EXAMPLE 87 -(2-Chloro-3 ',4'-difluoro-biphenyl-4-ylamino)-benzoic acid S: 359; MW: 359.7578.
EXAMPLE 88 -(3 '-Bromo-2-chloro-biphenyl-4-ylamino)-benzoic acid S: 401; MW: 402.6737.
EXAMPLE 89 -{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid
EXAMPLE 90 -{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-nitro-benzoic acid
EXAMPLE 91 - {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -benzoic acid
EXAMPLE 92 -{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid
EXAMPLE 93 -{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid
EXAMPLE 94 -{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,5-dimethoxy-benzoic acid EXAMPLE 95 -{4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenylamino}-3-nitro-benzoic acid
EXAMPLE 96 3- {4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenylamino} -benzoic acid
EXAMPLE 97
5 - {4- [2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -isophthalic acid
EXAMPLE 98 2- {4- [2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -benzoic acid
EXAMPLE 99 4-(4-{2-[(4aS,8aR)-4-(Octahydro-isoquinolin-2-yl)-phenyl]-ethyl}-phenylamino)- benzoic acid
EXAMPLE 100 2-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-5-methoxy-benzoic acid
EXAMPLE 101 2-{4-[2-(3-Methoxy-phenyl)-ethyl]-phenylamino}-benzoic acid
EXAMPLE 102 2- { 4- [2-(3 -Bromo-phenyl)-ethyl] -phenylamino } -benzoic acid
EXAMPLE 103 2-{4-[2-(3-Fluoro-phenyl)-ethyl]-phenylamino}-benzoic acid
EXAMPLE 104
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methoxy-benzoic acid
EXAMPLE 105 4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-nicotinic acid EXAMPLE 106 2-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-2,3-dihydro-lH-isoindol-5-ylamino]- benzoic acid
EXAMPLE 107 2-{4-[2-(3-Fluoro-4-methyl-phenyl)-ethyl]-phenylamino}-benzoic acid
EXAMPLE 108 2-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-5-nitro-benzoic acid
EXAMPLE 109 4- {4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino} -benzoic acid
EXAMPLE 110
4-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-3-methoxy-6-nitro- benzoic acid
EXAMPLE 111 4-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-3-methoxy-benzoic acid
EXAMPLE 112
2-{4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenylamino}-5-methoxy-benzoic acid
EXAMPLE 113 {4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenyl}-(2-methoxy-5-nitro-phenyl)- amine
EXAMPLE 114 2-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-3-nitro-benzoic acid
EXAMPLE 115 3- {4- [3 -(4-Diethylamino-phenyl)-propyl] -phenylamino } -benzoic acid EXAMPLE 116 2-{4-[2-(3,4-Dimethoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 159- 161°C.
EXAMPLE 117 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino} -benzoic acid monosodium; mp 107-108°C.
EXAMPLE 118 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid monopotassium; mp >200°C.
EXAMPLE 119
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid calcium salt (l :l); mp >220°C.
EXAMPLE 120 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -benzoate-2 -hydroxy- 1 , 1 -bis- hydroxymethyl-ethyl-ammonium; mp 185-187°C.
EXAMPLE 121 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-methoxy-benzoic acid; mp 155-158°C.
EXAMPLE 122 2-{4-[2-(3,4-Difluoro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 184-185°C.
EXAMPLE 123 2-{3-[2-(4-Chloro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 155-157°C.
EXAMPLE 124 2-{3-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 182- EXAMPLE 125 -{4-[2-(2,4-Dimethoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 180- 181°C.
EXAMPLE 126 -{4-[2-(2-Chloro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 140- 143 °C.
EXAMPLE 127 - {4-[2-(2-Hydroxy-phenyl)-ethyl]-phenylamino} -benzoic acid; mp 218-219°C.
EXAMPLE 128 2- {4-[2-(3-Chloro-phenyl)-ethyl]-phenylamino} -benzoic acid; mp 152-154°C.
EXAMPLE 129
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-benzoic acid; mp 200-202°C.
EXAMPLE 130 2-{4-[2-(2,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 181-183°C.
EXAMPLE 131 3-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 137-138°C.
EXAMPLE 132 4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 214-215°C.
EXAMPLE 133 2-{4-[2-(3,4,5-Trimethoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 146- 147°C.
EXAMPLE 134 2-{4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; mp 153-154°C. EXAMPLE 135 2-{4-[5-(3,4-Dichloro-phenyl)-pentyl]-phenyl--mino}-benzoic acid; mp 106- 108°C.
EXAMPLE 136 2- {4- [2-(4- {2-Hydroxycarbonylphenyl_-mino }phenyl)ethyl]-phenylamino } - benzoic acid; MS 451 (M_1).
EXAMPLE 137 2-(3',5'-Dichloro-biphenyl-4-ylamino)-benzoic acid; mp >220°C.
EXAMPLE 138 4- {4-[3 -(3 ,4-Dichloro-phenyl)-propyl] -phenylamino } -2-methoxy-5-nitro-benzoic acid; mp 74-78°C.
EXAMPLE 139 2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-5-fluoro-benzoic acid; mp 122-123°C.
EXAMPLE 140
5-Amino-2-{4-[5-(3,4-dichloro-phenyl)-pentyl]-phenylamino}-benzoic acid; mp 182-184°C.
EXAMPLE 141 N-(2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-benzoyl)-C,C,C- trifluoro-methanesulfonamide; MS 531 (M").
EXAMPLE 142 N-(2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-benzoyl)- benzenesulfonamide; MS 539. EXAMPLE 143 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-trifluoromethyl-benzoic acid; mp 190-192°C. MS 453 (M"1).
EXAMPLE 144 4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid; mp 264-
266°C.
EXAMPLE 145 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-trifluoromethyl-benzoic acid; mp 134-136°C; MS 454 (M+).
EXAMPLE 146
2- { 4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -3 -trifluoromethyl-benzoic acid; MS 454 (M+).
EXAMPLE 147 2-({4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenyl}-methyl-amino)-5-dimethylamino- benzoic acid; mp 128-131°C.
EXAMPLE 148 2-( {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenyl } -methyl-amino)-benzoic acid;
MS 400 (M+).
EXAMPLE 149 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-dipropylamino-benzoic acid; MS 485 (M+).
EXAMPLE 150 5-Dibutylamino-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid;
MS 513 (M+). EXAMPLE 151 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-diethylamino-benzoic acid; mp l06-110°C.
EXAMPLE 152 2,2'-[ 1 ,2-Ethanediylbis (4, 1 -phenyleneimino)]bis-benzoic acid
EXAMPLE 153 4-[3-[4-(Diethyl--mino)phenyl]propyl]-N-(2-methoxy-5-nitrophenyl)-benzinamine
EXAMPLE 154 2- { 3 - [2-(4-Chlorophenyl)ethyl]phenyl amino } -benzoic acid
EXAMPLE 155
2- { 3 -[3-(3 ,4-Dichlorophenyl)propyl]phenylamino } -benzoic acid
EXAMPLE 156 2- { 3 - [3 -(4-Diethylaminophenyl)propyl]phenylamino } -benzoic acid
EXAMPLE 157 2-{3-[3-(4-Di-n-propylaminophenyl)propyl]phenylamino}-benzoic acid
The following Examples 158-163 illustrate the use of invention compounds as starting materials and intermediates in the synthesis of other invention compounds and derivatives. The examples illustrate reduction of nitro groups to amino groups, alkylation of amino group, and esterification of carboxylic acid groups. These reactions are depicted in the following generalized
Scheme 12. Scheme 12
Figure imgf000090_0001
reduction
Figure imgf000090_0002
Figure imgf000090_0003
alkylation
Figure imgf000090_0004
COOH COOE
esterification \ / alkylation
Figure imgf000090_0005
saponification
Figure imgf000090_0006
where RD and Rc are as defined above, and E is an ester forming group such as Cj-C6 alkyl (e.g., methyl, 2,2,2-trichloroethyl), benzyl, diphenylmethyl, or the like.
EXAMPLE 158 2- {4- [3 -(4-Nitrophenyl)propyl]phenylamino } benzoic acid
To a slurry of 4-[3-(4-nitrophenyl)propyl]aniline (4.08 g, 15.9 mmol) and 2-bromobenzoic acid (3.52 g, 17.5 mmol) in i-PrOH (100 mL) was added Cu(OAc)2 (87 mg, 0.478 mmol) and KOAc (3.44 g, 35.0 mmol) at room temperature. The resulting mixture was allowed to heat under reflux for 23 hours, then cooled to room temperature. After removing the solvent under reduced pressure, the residue was diluted with water (100 mL) and basified with aqueous 1.0 M-NaOH solution to pH 9.0. The aqueous layer was washed with Et2θ
(20 mL, twice) and acidified with aqueous 1.0 M-HC1 solution to pH 3.0. The precipitate formed was filtered by suction and dried at 60°C in vacuo, affording the title compound as a beige solid (5.75 g, 96% yield), mp 150-153°C.
MS (Fab): 376 (MH+).
EXAMPLE 159
2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid
To a solution of 2-{4-[3-(4-nitrophenyl)propyl]phenylamino}benzoic acid (Example 158) (3.0 g, 7.97 mmol) in DMF (40 mL) was added 10% Pd-C (300 mg) at room temperature under argon atmosphere. Hydrogen gas (1 atm) was introduced into the flask and the mixture was stirred for 14 hours at room temperature. The reaction mixture was filtered through a Celite pad to remove Pd-C and concentrated in vacuo. The residue was diluted with MeOH (ca. 50 mL) and concentrated in vacuo. This operation was repeated 3 times to remove any trace of DMF. The residue was diluted with MeOH again, and insoluble stuff was removed by filtration. Removing the solvent of the filtrate in vacuo afforded an oil, which was diluted with CH3CN (50 mL) and added dropwise water (100 mL) slowly. The precipitate formed was filtered and dried at 60°C in vacuo, affording the title compound as a white solid (2.34 g, 85% yield), mp 110-112°C.
MS (Fab): 347 (MH+).
EXAMPLE 160
2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid methyl ester
To a solution of 2-{4-[3-(4-aminophenyl)propyl]phenylamino}benzoic acid (Example 159) (2.34 g, 6.75 mmol) in MeOH (50 mL) was added concentrated H2SO4 (1.0 mL) at room temperature. The mixture was stirred under reflux for 3.0 days. The reaction was quenched with Et3N (10 mL) at 5°C, and the solvent was removed under reduced pressure. The residue was diluted with water (20 mL) and extracted with Et2θ (20 mL, 4 times). The combined ether layer was washed with water (10 mL) and brine (10 mL), and dried over anhydrous Na2SO4- The solvent was removed under reduced pressure and purification by column chromatography afforded crude title compound as a yellow amorphous material (2.59 g). This material was used without further purification.
EXAMPLE 161 2-{4-[3-(4-Diethylaminophenyl)propyl]phenylamino}benzoic acid methyl ester and 2-{4-[3-(4-Ethylaminophenyl)propyl]phenylamino}benzoic acid methyl ester To a solution of the crude ester described above (2.59 g, ca. 6.75 mmol) and CH3CHO (2.0 mL, 35.1 mmol) in CH3CN (50 mL) was added NaBH3CN
(1.70 g, 27.0 mmol) at 5°C, and the suspension was stirred for 30 minutes while the pH was monitored and aqueous 1.0 M-HCl solution was added to maintain the mixture moderately acidic (pH 3.0-4.0). The reaction mixture was allowed to warm up to room temperature over 1.0 hour and then basified with aqueous
1.0 M-NaOH solution to pH 9.0. The reaction mixture was concentrated under reduced pressure to remove CH3CN, and the resulting aqueous solution was acidified with aqueous 1.0 M-HCl solution to pH 3.0. The aqueous solution was extracted with CHCI3 (20 mL, 3 times), and the combined extract was washed with brine (5 mL). After drying over anhydrous Na2SO4, the solvent was removed under reduced pressure and purified by column chromatography (silica gel 60N, n-hexane/CHCl3/Εt3N 50:98:2). First eluted was the dialkylated product as a yellow amorphous material (1.07 g, 38%).
MS (Fab): 417 (MH+). Subsequently eluted was the monoalkylated product as a yellow amorphous material (0.79 g, 30%).
MS (Fab): 389 (MH+).
EXAMPLE 162 2- {4- [3 -(4-Diethylaminophenyl)propyl]phenylamino } benzoic acid To an emulsion of 2-{4-[3-(4-diethylaminophenyl)propyl]phenylamino}- benzoic acid methyl ester (1.68 g, 4.03 mmol) in EtOH (50 mL) was added aqueous 3 M-KOH solution (4.0 mL, 12.0 mmol) at room temperature, then the mixture was allowed to heat under reflux for 40 minutes. The reaction mixture was cooled to room temperature and neutralized with aqueous 1.0 M-HCl solution to pH 9.0. The mixture was concentrated under reduced pressure to remove EtOH, and the resulting aqueous solution was extracted with CHCI3 (50 mL, 3 times).
The combined extract was washed with brine (10 mL) and dried over anhydrous Na2SO4- The solvent was removed under reduced pressure and purification by column chromatography (silica gel 60N, cone NH4OH/MeOH/CHCl3 0.2:2:100 to 0.5:5:100) afforded a yellow oil. This oil was diluted with acetone, and the solution was concentrated under reduced pressure at room temperature to give the title compound as an amorphous solid (1.62 g, 99% for 0.2 hydrate).
MS (Fab): 403 (MH+). Analysis for C26H3oN2O2-0.20 H2O: Calcd: C, 76.89; H, 7.54; N, 6.90.
Found: C, 76.73; H, 7.67; N, 7.10.
EXAMPLE 163 2- {4- [3 -(4-Ethylaminophenyl)propyl]phenylamino }benzoic acid
The title compound was prepared from 2-{4-[3-(4-ethylaminophenyl)- propyl] phenylamino} benzoic acid methyl ester (from Example 161), EtOH
(10 mL), and 3 M-KOH solution (1.0 mL) using the procedure described in Example 162. This procedure yielded a yellow solid, 253 mg of the desired product (90% for 0.1 hydrate).
MS (Fab): 375 (MH+). Analysis for C24H26N2O2-0.10 H2O: Calcd: C, 76.61; H, 7.02; N, 7.44.
Found: C, 76.62; H, 7.06; N, 7.36. BIOLOGICAL EXAMPLES
Representative compounds of Formula I have been evaluated in several in vitro and in vivo assays which are well-established as indicative of clinical usefulness in treating Alzheimer's disease.
AMYLOID ASSAYS
BASSR CBeta-Amyloid Self-Seeding Radioassav
An assay for inhibitors of self-seeded amyloid fibril growth
Materials:
Stock Solutions: Assay Buffer - 50 mM sodium phosphate, pH 7.5, 100 mM NaCl, 0.02% NaN3,
1 M urea (filter and store at 4°C).
Soluble A β(l -40) peptide (Bachem, Torrance, CA) - 2.2 mg/mL in deionized H2O (stored in aliquots at -20°C, keep on ice when thawed) will self-seed after 1 week storage. Typically, the solution should be stored until no lag phase is seen in the assay.
1251-labeledAβ (1-40) - 150K-350K cpm/μL in 100% acetonitrile - 0.1% trifluoroacetic acid (TFA) - 1% β-mercaptoethanol (aliquots stored at -20°C). 125i-iabeled A/? (1-40) can be made in accordance with the procedure set forth by H. LeNine, III in Neurobiol. Aging, 16, 755, (1995), which is hereby incorporated by reference, or this reagent may be purchased from Amersham, Arlington
Heights, Illinois.
Final assay conditions: 30 μM soluble A ? (1-40) in deionized water in assay buffer + 20K-50K cpm 125ι_jat»eled A ? (1-40) per assay. Compound to be tested is dissolved in dimethylsulfoxide (DMSO), typically 5 to 50 mM stock, such that the final concentration of DMSO is <1% v/v in the assay. Assay: Reaction mixture for 50 assays (on ice) is comprised of 0.1 to 0.2 μL of
1 5I-labeled A 25I-labeled Aβ(l-40) + 1 μL of soluble Aβ( 1-40) + 13.5 μL assay buffer per assay. The following are the amounts of the components of the reaction mixture sufficient for 50 assay wells: 5-10 μL 125I-labeled -4/7(1-40) dried down
675 μL assay buffer 50 μL soluble Aβ (1-40)
Assay Method
1) Prepare reaction mixture above by mixing components and storing on ice. 2) Pipet 14.5 μL of reaction mixture into each of 50 wells on a polypropylene
U-bottom 96-well microtiter plate on ice. (Costar 3794).
3) Add 1.7 μL of diluted compound to be tested to each well in a column of eight, including solvent control. Serial 3-fold dilutions from 1 mM (100 μM final) in assay buffer - urea = 7 dilutions + zero. Each 96-well plate can therefore accommodate 11 samples + 1 Congo Red control (0.039-5 μM final in 2-fold steps).
4) Seal the plate with aluminum film (Beckman 538619) and incubate for 10 minutes on ice.
5) Raise the temperature to 37°C and incubate for 3 to 5 hours (depending on the lot of the peptide).
6) Remove the aluminum film and add 200 μL/well of ice cold assay buffer with urea, collecting the radiolabeled fibrils by vacuum filtration through 0.2 μm pore size GNWP filters in 96-well plates (Millipore MAGN Ν22, Bedford, MA). Determine the radioactivity of the filters using standard methods well-known to those skilled in the art.
BASST (Beta-Amyloid Self-seeding, ThioflavinT-
An assay for inhibitors of self-seeded amyloid fibril growth
METHODS: Materials: Stock Solutions: Assay Buffer - 50 mM sodium phosphate, pH 7.5, 100 mM NaCl, 0.02% NaN3,
1 M urea (filter and store at 4°C)
Soluble Aβ (1-40) - 2.2 mg/mL in deionized H2O (store in aliquots at -20°C, keep on ice when thawed) will self-seed after 1 week storage. Typically, the solution should be stored until no lag phase is seen in the assay.
Final assay conditions: 30 μM soluble Aβ (1-40) in deionized water in assay buffer. Compound to be tested is dissolved in DMSO, typically 5 to 50 mM stock, such that the final concentration of DMSO is <1% v/v in the assay.
Assay: Reaction mixture for 50 assays (on ice) comprised of 1 μL of soluble Aβ(l-40) + 13.5 μL assay buffer per assay. The following are the amounts of the components of the reaction mixture that result in each of the 50 assay wells: 50 μL soluble Aβ (1-40) 675 μL assay buffer
Assay Method 1) Prepare the reaction mix above by mixing the components and storing on ice.
2) Pipet 14.5 μL of reaction mixture into each of 50 wells of a polystyrene U-bottom 96-well microtiter plate (Corning 25881-96) on ice.
3) Add 1.7 μL of diluted compound to be tested to each well in a column of eight, including solvent control. Serial 3-fold dilutions from 1 mM (100 μM final) in assay buffer - urea = 7 dilutions + zero. Each 96-well plate can therefore accommodate 11 samples + 1 Congo Red control (0.039-5 μM final in 2-fold steps).
4) Seal the plate with aluminum film and incubate for 10 minutes on ice. 5) Raise the temperature to 37°C and incubate for 3 to 5 hours (depends on the lot of the peptide). 6) Remove the aluminum film and add 250 μL/well of 5 μM thioflavin T (ThT) [T-3516, Sigma-Aldrich] in 50 mM glycine-NaOH, pH 8.5. Read fluorescence on a plate reader (ex = 440 nm 20 nm ; em = 485 nm/20 nm) within 5 minutes.
BAPA (Beta- Amyloid Peptide Aggregation)
This assay is used to provide a measure of inhibition by a compound against the aggregation behavior of the beta amyloid peptide.
The purpose of this assays is to provide a higher volume method of assaying the amount of beta amyloid aggregation using an endpoint assay based on filtration. In this assay, hexafluoroisopropanol (HFIP) is used to break down the initial amyloid peptide to a monomer state and use a concentration of 33 μM which is high enough so that aggregation will occur at pH 6.0 in several hours.
METHODS:
β-Amyloid Peptide Aggregation, pH 6.0 (BAPA)
In a 96-well plate (Costar 3794), we add 25 μL 50 mM Phosphate Buffer, pH 6.0, 10 μL 0.5 mg/mL Aβ (1-40) peptide in 20% HFIP + 0.1 μL/assay radioiodinated 125I Aβ (1-40) [125I Aβ(l-40)], and 1 μL of the compound to be tested starting at 50 mM with a concentration of DMSO <1%. Then, we incubate for 2 to 4 hours at room temperature. We stop the reaction with 200 μL of 50 mM phosphate buffer, pH 6.0, and filter it through a 0.2 μm 96-well filter plate (Millipore MAGU N22). We wash the filter plate with 100 μL of the same phosphate buffer. Aggregation was detected on a Microbeta counter after impregnating the filters with Meltilex (1450-441) and is corrected for background.
BATYM ASSAY
METHODS:
Required Aβ (1-42) (California Peptide) was dried from its hexafluoroisopropanol (HFIP) stock solution. The Aβ (1-42) was dissolved in dimethylsulfoxide (DMSO) and then mixed with phosphate buffered saline (PBS) (pH 7.4). The mixed Aβ (1-42) solution was filtered with a 0.2 μm Omnipore membrane syringe filter (Millipore, Bedford, MA). The compound to be tested in DMSO (50 times concentrate) was put into each well (0.5 μL/well) of a 96-well plate. The Aβ (1-42) solution was added into each well (24.5 μL/well). The plate was centrifuged at 1,000 g for 5 minutes and incubated at 37°C for 1 day (Aβ 1-42; final concentration 100 μM).
After incubation Thioflavin T (ThT) (30 μM) solution in glycine-NaOH buffer (pH 8.5, 50 mM) was added into each well (250 μL/well), fluorescence was measured (ex = 440/20 nm, em = 485/20 nm) using a fluorescence plate reader. The inhibitory activity was calculated as the reduction of fluorescence with the following formula:
Inhibition (%) ={(F(Aβ)-F(Aβ+compound)}/{F(Aβ)-F(solvent + compound)} x 100
The IC50S were calculated by a curve fitting program using the following equation. The data were obtained from two different experiments in triplicate.
Inhibition(x) = 100-100/ {l+(x/IC50)n}, x = Concentration of tested compound (M), IC50 = (M), n = Hill coefficient.
Representative compounds of Formula I have exhibited inhibitory activities (IC50) ranging from 0.1 μM to greater than 100 μM in the foregoing assays.
The results of these assays for specific and representative compounds of the present invention are shown in Table 1 below.
Table 1. β- Amyloid Inhibitory Activity of Compounds of Formula I
Example BASSR BASST BATYM BAPA
No. (IC50 = μM) (IC50 = μM) (IC50 = μM) (IC50 = μM)
1 10(P),>100(P) 2, 4, 30,10 (P) 50, 58.8 (P) 60(P),>100(P)
(6x),>100(Q), 3(0) 57.8 (Q) 86 (Q),
>100(R), >100 (R) >60 (R),
>100 (S), 11(S),11(T) >60 (S),
52 (T) 6(Z) 11 (T)
>100(Z)
2 2.2,4.1,4.1, 1,1.5(P) 6.52 (P) 70 (P) 12, 4.5
3 4.5, 5, 5 2(P) 11.7(P) >60 (P) (all 3 V-shaped) 3(Q)
(P) 15(ppt),5(Q)
4 30, 3,4,8 26.3, 30.7 67 >100 (3x)
5 70,>100 4.5 10 74
6 15,21,20,40 4,1,3 21.5 >60
7 18,13,12,20 2 8.83 39
8 15,15,18,15 3,>100 7.17
9 20 (ppt), 30, 1,2(P) 20.1, 28.2 (P) 75 (P) 52, 40 (P) 38.6 (R)
10 70,50 4 75.7 67
11 18 (ppt), 1,1,3(P) 5.62 (P) 23 (P)
20 (ppt), 1,0.8(Q) 6.78 (Q) 9(Q)
20(2x),>100(P)
>100,21,30(Q)
12 20 (4x) 1,1 3.93 >60
13 21,>100, 0.9 6.41 6
20 (ppt),
15 (ppt), > 100
14 18 (ppt), 8, 1.0 10.9 >10 6 (ppt), 7 (ppt) (N-shaped)
15 100(3x)P 1(P) 8.52 (P) >60 (P)
100, 16 1.2 (Q) 7-26 (Q) 7(0)
(V-shaped) 7.07 (Q)
12, 15,11 (Q) Table 1. β- Amyloid Inhibitory Activity of Compounds of Formula I (cont'd)
Example BASSR BASST BATYM BAPA
No. (IC50 = μM) (IC50 = μM) (IC50 = μM) (IC50 = μM)
16 18, 7.5, 10 (P) 3, 0, 3 (P) 12 (P) 13 (P)
70, 32, 42 (Q) 1.1, 0.8, 0.6 (Q) 10.3 (Q)
17 >100 (ppt) (3x) 6.2 64.5 >60(Q),41(R)
18 >100(5x)(P) 30,>100(P) >100 (P) 9, >40, 53 (P), 12
19 3,4,>100,2.2 >100, 1, 1, 1.5 31.0,34.0 >60, 43
20 4.2 6 68.6 22
21 3 4 62.7 26
22 3 9 >100 24
23 20 2 >100 17
24 >100 20 >100 91
25 >100 4 21.1 47
26 >100 1 >100 57
27 >100 3 19.8 74
28 >100 5 42.3 27
29 >100 4 38.1 30
30 30,20 4,2 75.3 38
31 >100 1 22.6 86
32 >100 1 29.2 96
33 >100 >100 >100 >10
34 45 3 45.0 48
35 >100 100 >100 154
36 >100 >100 >100 149
37 >100 0.8 30.2 25
38 20, 10 (V) 3 23.4 184
39 >100 20 >100 21
40 >100 3.0 >100 53
41 >100 5 49.7 42
42 >100 2 55.6 30
43 >100 0.3 24.2 63
44 >100 1 26.5 52
45 >100 1 21.5 32
46 >100 6 34.3
47 >100 2 38.2
48 25 10 >100
49 >100 >100 >100
50 >100 >100 >100
51 85 0.8 39.1 Table 1. β-Amyloid Inhibitory Activity of Compounds of Formula I (cont'd)
Example BASSR BASST BATYM BAPA
No. (IC50 = μM) (IC50 = μM) (IC50 = μM) (IC50 = μM)
52 75 0.5 36.5
53 >100 0.3 30.0
54 >100 0.4 43.9
55 12 2 5.1 101
56 >100 3 11.5 30
57 4.8 1.5 4.0 50
58 3.5 1 5.1 60
59 >100 >100 >100 3
60 >100 3 40.7 8
61 18,7.5,10 3,0.3 12 13
62 >100 1.5 8.98
63 15, 15, 18 (ppt) 1 9.43 45
64 >100 5 35 >100
65 60,80 1.5 15.9 >100 (V-shaped)
66 >100 (ppt), 2.1 50.1 >100 >100 (ppt)
67 41 4 13.3 >60
68 >100, >100 1 >100 110
69 2 (V-shaped), 0.8 11.7 58 3.5 (ppt)
70 20, 100 10 >100 65
71 >100 3 >60
72 40, 15, 12 2,2.5 74.8 >60, >60
73 25, 35, 40 0.3 9.43 >60
74 6, 18, 19, 18 0.3, 0.5 8.36 >60
75 >100 2.2 46.2 >60
76 3 0.5 8.59 >60
77 18,15 8,0.3 9.49 >60
78 70 0.1 >100 8
79 3.1,50,38,70, 1,0.3,0.3,0.3 9.14 51 70, 30, 40
80 >100 4 24.8 >60
81 >100 15 48.4 73
82 >100,>100,>100 2, 0.3, 0.3
83 >100 >100 9, 47, 29
84 >100 >100 5,40,21
85 >100 18 8, 77, 45 Table 1. β-Amyloid Inhibitory Activity of Compounds of Formula I (cont'd)
Example BASSR BASST BATYM BAPA
No. (IC50 = μM) (IC50 = μM) (IC50 = μM) (IC50 = μM)
86 40 18 >10, 89,37
87 >100 50 >10, 15,32
88 >100 10 >10, 37, 27
116 >100,>100 18,30 96
117 >100 3 61.3 >100
118 >100,>100 6 >60
119 >100 3 >60
120 >100 3 >60
121 >100,>100 1
122 >100 2 >100 >60
123 >100(3x), 14 3,3 70.8
4, 18, >100, 3.2, 4 (Q) 85.2 (Q) >60(Q)
>100 (Q)
124 >100 10 62.7
125 82 10 >100 80
126 >100,>100 4,5 84 63
30, 100 (Q) 10, 4 (Q) 73.9 (Q) >60 (Q)
127 >100 (ppt) 10 >100 67
128 >100(ppt)(4x) 10,41,6 75 60 ll,>100(3x)
15, 20, 10, 7.5 (Q) 7, 3, 3 (Q) >60 (Q) >60 (Q)
15,>100(3x)Q
129 1 (V-shaped) (2 10,3,2,2 >100 >102 x) >100 (ppt)
130 >100 (3 x) 2,>100, 50 47.5 238
131 >100 10 93.5 >60
132 >100 10 >100 60
133 >100 >100 >100 >60
134 >100 2 36.5 >60
135 >100 1.2 31.2 >60
136 >100 3 >100 53
137 >100, >100 3 52
141 >100 7 56.7 >50
142 >100 2.1 26.9 55
143 >100(4x) 40,30 >100 2, >60, >60
144 15,25 40 >100 114 Table 1. β-Amyloid Inhibitory Activity of Compounds of Formula I (cont'd)
Example BASSR BASST BATYM BAPA
No. (IC50 = μM) (IC50 = μM) (IC50 = μM) (IC50 = μM)
145 10, 40, 30 4 56.8 9
146 >100 30 >100 >60
147 >100 10 93.4 >60
148 >100 >100
149 >100 >100 >100 >60
150 >100 10 >100 76
151 >100, >100 5, >100 >100 108
154 >100 3, 30 70.8
155 >100 3 44.6
156 27.8
157 25.9
A letter in parentheses after particular value indicates the particular synthetic lot of the compound tested. The terms "P," "Q," "R," "S," "T," and "Z" designate different lots of the same compound. For example, 10 (P) indicates that compound tested was from Lot P. If no lot is specified, the lot of the compound was Lot P.
The abbreviation "ppt" means precipitate and indicates that a precipitate formed at the indicated concentration. In addition, the term "V-shaped" means that inhibition was observed followed by precipitation.
A value followed by a number and x (i.e., 4x) means that the compound was tested four times, and each time the result was the same.
The invention compounds have also shown good activity in standard in vivo assays commonly used to evaluate agents to treat diseases related to aggregation of amyloid proteins, especially Alzheimer's disease and other amyloidoses. In one assay, amyloid protein is induced into the spleen of mice by subcutaneous injections of silver nitrate, Freund's complete adjuvant, and an intravenous injection of amyloid enhancing factor. Silver nitrate is administered each day through Day 11. Test compounds are administered to the mice daily starting on Day 1 through Day 11. On Day 12, the animals are sacrificed, and the spleens are removed, histologically prepared, stained with Congo red, and the percent area of the spleen occupied by birefringent, Congo red-stained amyloid is quantitated microscopically. Invention compounds evaluated in this test have inhibited splenic amyloid deposition by up to 70% relative to untreated controls.
Another in vivo assay in which the invention compounds have been evaluated uses transgenic mice. The mice bear a human β-amyloid precursor protein transgene with a prion promoter and are described by Hsiao et al.,
"Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice," Science 1966;274:99-102. These transgenic mice develop β-amyloid deposits at about 9 months of age. By 15 months, diffuse and compact senile plaques are abundant, primarily in the neocortex, olfactory bulb, and hippocampus. Invention compounds are administered orally to the mice beginning at the age of 8 months (just prior to the onset of amyloid deposits) and continuing for several months (up to about age 14-18 months). The animals are then sacrificed, and the brains are removed. The amount of amyloid in the brain is quantitated both histologically and biochemically. Invention compounds evaluated in this model have inhibited amyloid accumulation in the cortex and hippocampus by up to 49% relative to untreated controls.
The above data establishes that representative invention compounds are active in standard assays used to measure inhibition of protein aggregation. The compounds exhibit excellent specificity, for example, as shown in the BASST assay, as well as the BATYM and BAPA assays. The compounds are thus useful to clinically inhibit amyloid protein aggregation and to image amyloid deposits for diagnostic use. The compounds will be used in the form of pharmaceutical formulations, and the following examples illustrate typical compositions.
EXAMPLE 164
Tablet Formulation
Ingredient Amount
Compound of Example 1 50 mg
Lactose 80 mg
Cornstarch (for mix) 10 mg
Cornstarch (for paste) 8 mg
Magnesium Stearate (1%) 2 mg
150 mg
The compound of Example 1 is mixed with the lactose and cornstarch (for mix) and blended to uniformity to a powder. The cornstarch (for paste) is suspended in 6 mL of water and heated with stirring to form a paste. The paste is added to the mixed powder, and the mixture is granulated. The wet granules are passed through a No. 8 hard screen and dried at 50°C. The mixture is lubricated with 1% magnesium stearate and compressed into a tablet. The tablets are administered to a patient at the rate of 1 to 4 each day for prevention of amyloid protein aggregation and treatment of Alzheimer' s disease.
EXAMPLE 165 Parenteral Solution
In a solution of 700 mL of propylene glycol and 200 mL of water for injection is added 20.0 g of Compound No. 19 (Example 19). The mixture is stirred and the pH is adjusted to 5.5 with hydrochloric acid. The volume is adjusted to 1000 mL with water for injection. The solution is sterilized, filled into 5.0 mL ampoules, each containing 2.0 mL (40 mg of Compound No. 19), and sealed under nitrogen. The solution is administered by injection to a patient suffering from medullary carcinoma of the thyroid and in need of treatment. EXAMPLE 166 Patch Formulation
Ten milligrams of 2-{4-[3-(3,4-dichlorophenyl)propyl]phenylamino} benzoic acid is mixed with 1 mL of propylene glycol and 2 mg of acrylic-based polymer adhesive containing a resinous cross-linking agent. The mixture is applied to an impermeable backing (30 cm2) and applied to the upper back of a patient for sustained release treatment of amyloid polyneuropathy.
The invention and the manner and process of making and using it, are now described in such full, clear, concise, and exact terms as to enable any person skilled in the art to which it pertains, to make and use the same. It is to be understood that the foregoing describes preferred embodiments of the present invention and that modifications may be made therein without departing from the spirit or scope of the present invention as set forth in the claims. To particularly point out and distinctly claim the subject matter regarded as invention, the following claims conclude this specification.

Claims

What is claimed is:
A method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I
Figure imgf000107_0001
wherein
O
Ra is hydrogen, Cj-C6 alkyl, or -CCj-C6 alkyl; n is 0 to 5 inclusive;
Rl, R2, R3, R4, R5. 65 an(j R7 ^g independently hydrogen, halogen,
-OH, -NH2, NRbRc, -CO2H, -CO Cι-C6 alkyl, -NO2, -OC^C^ alkyl, -Ci-Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl,
O O
II II
-CH=CH-phenyl, -O(CH2)pNRbRc, -CNRbRc, -NHCRb, -NH(CH2)pNRbRc, -N(C γ -C6alkyl)(CH2)pNRbRC,
Figure imgf000107_0002
R8 is COOH, tetrazolyl, -SO Rd, or -CONHSO2Rd;
Rb and Rc are independently hydrogen, -Cj-C6 alkyl, -(CH2)m-phenyl, or
Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl; Rd is hydrogen, -Cj-C alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive;
A is CH orN;
R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
2. The method of Claim 1 wherein Ra is hydrogen; n is 2; and
R3 and R4 are hydrogen.
3. The method of Claim 1 wherein
Ra is hydrogen; R3 and R4 are hydrogen; and n is 2 to 5 inclusive.
4. The method of Claim 1 wherein Ra is hydrogen; n is 2; R3 and R4 are hydrogen; and
R1, R2, and R7 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl, -O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
I I I , -N[(CH2)3CH3]2, substituted phenyl,
-OCH2- substituted phenyl, pyrrozolyl, or -N(phenyl)2-
5. The method of Claim 1 wherein
Ra is hydrogen; n is 3, 4, or 5;
R3 and R4 are hydrogen; and Rl, R2, and R' are independently chlorine or hydrogen.
6. The method of Claim 1 wherein
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and R5, R6, and R8 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, halogen,
-NH-Cι-C6 alkyl, -NCC^Cgalkyl^, -NH2, or pyrrolyl.
7. The method of Claim 1 wherein
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and R5 is -CO2H.
8. A method of treating Alzheimer's disease, the method comprising administering to a patient having Alzheimer's disease a therapeutically effective amount of a compound of Formula I
Figure imgf000109_0001
wherein
Ra is hydrogen; n is 1 to 5 inclusive; R and R4 are hydrogen;
R1, R7, and R2 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl, -O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
-N[(CH2)3CH3]2, substituted phenyl, -OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2;
R5 and R6 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; R8 is COOH or tetrazolyl; or the pharmaceutically acceptable salts thereof.
9. The method of Claim 1 wherein the compound of Formula I is:
2-[[4-[2-(3,4-Dichlorophenyl)ethyl]phenyl]amino-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]phenylamino}-5-nitrobenzoic acid;
2- {4- [4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -4-methoxy- 5 -nitrobenzoic acid;
2-{4-[2-(3,4-Dihydroxy-phenyl)-ethyl]-phenylamino}benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid; 2-{4-[2-(3,4,5-Trihydroxy-phenyl)-ethyl]phenylamino}benzoic acid;
2- {4- [3-(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-methoxy- 5 -nitrobenzoic acid;
2- {4- [3 -(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-imidazo- l-yl-5-nitrobenzoic acid;
2- {4-[3 -(3 ,4-Dichlorophenyl)-propyl]phenylamino } benzoic acid; 2- {4-[4-(3 ,4-Dichlorophenyl)butyl]phenylamino } benzoic acid; 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-nitro- benzoic acid; 2-{4-[4-(3,4-Dichlorophenyl)-butyl]phenylamino}- 3,5-dinitrobenzoic acid;
2- {4- [5-(3 ,4-Dichlorophenyl)pentyl]phenylamino } -5-nitrobenzoic acid; 2- {4-[5-(3 ,4-Dichloro-phenyl)pentyl]phenylamino } -4-methoxy-
5-nitrobenzoic acid;
2-[4-(3,4-Dichloro-benzyl)-phenylamino]-benzoic acid; 2- {4- [2-(3 ,4-Dimethyl-phenyl)-ethyl]-phenyl amino } -5-nitrobenzoic acid; 2-{4-[2-(3,4-Difluoro-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}- benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid; 5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid;
2-(4-Phenethyl-phenylamino)-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methoxy- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-terephthalic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl- benzoic acid;
4- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -isophthalic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- methanesulfonyl-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -5-imidazol- 1 - yl-benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-nitro- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-e thyl] -phenylamino } -4-nitro- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -nitrobenzoic acid;
5-Cyano-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino}- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,6-difluoro- benzoic acid;
6-{4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -2 ,3 -difluoro- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6-fluoro- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-fluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino} -3-methyl- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-fluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 ,5-difluoro- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 - trifluoromethyl-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6- trifluoromethyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- trifluoromethyl-benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -5-pyrrol- 1 -yl- benzoic acid;
2- {4-[2-(4-Benzyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[4-(3-Dimethylamino-propoxy)-phenyl]-ethyl}- phenylamino)-benzoic acid; 2- {4- [2-(4-Diethylamino-phenyl)-ethyl] -phenylamino } -benzoic acid;
2-{4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Octyloxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[4-(2-Ethoxy-l-ethoxymethyl-ethyl)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2-{4-[2-(4-Pyrrol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4-[2-(4-Styryl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Octyl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- { 2- [3 -(3 ,5-Dichloro-phenoxy)-phenyl] -ethyl } -phenylamino)- benzoic acid;
2-(4-{2-[4-(2-Chloro-6-fluoro-benzyloxy)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2- {4-[2-(4-Pyrazol- 1 -yl-phenyl)-ethyl] -phenylamino } -benzoic acid;
2- {4-[2-(4-Diphenylamino-phenyl)-ethyl] -phenylamino } -benzoic acid;
2-(4-{2-[4-(3,4-Dichloro-benzyloxy)-phenyl]-ethyl}- phenylamino)-benzoic acid; 2-{4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5 -aminobenzoic acid;
2- { 4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5 - trifluoromethyl-benzoic acid;
2-{4-[2-(3,4-Dichlorophenyl)]phenylamino}-5-nitrobenzoic acid; 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl] phenylamino}-5- nitrobenzoic acid;
2-[[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino- benzoic acid; or
2-[4-(3,4-Dichlorophenyl)phenyl]aminobenzoic acid.
0. A method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, the method comprising administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein aggregation inhibiting amount of a compound of Formula I
Figure imgf000114_0001
wherein
O
Ra is hydrogen, C1-C6 alkyl, or -CCi-Ce alkyl; n is 0 to 5 inclusive;
Rl, R2, R3, R4, R55 R65 and R7 ^Q independently hydrogen, halogen,
-OH, -NH2, NRbRC, -CO2H, -CO2 -C6 alkyl, -NO2, -OC1-C12 alkyl, -Cj-Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl,
O O
II II
-CH=CH-phenyl, -O(CH2)pNRbRc, -CNRbRc, -NHCRb,
-NH(CH2)pNRbRC, -N(C \ -C6alkyl)(CH2)pNR RC,
Figure imgf000114_0002
R8 is COOH, tetrazolyl, -SO2Rd, or -CONHSO2Rd;
Rb and Rc are independently hydrogen, -Cj-C6 alkyl, -(CH2)m-phenyl, or
Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl; Rd is hydrogen, -Cj-C6 alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive; A is CH orN; R1 and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
11. The method of Claim 10 wherein
Ra is hydrogen; n is 2; and R3 and R4 are hydrogen.
12. The method of Claim 10 wherein
Ra is hydrogen;
R3 and R4 are hydrogen; and n is 2 to 5 inclusive.
13. The method of Claim 10 wherein
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and
R1, R2, and R7 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl,
-O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
, -N[(CH2)3CH3J2, substituted phenyl,
Figure imgf000115_0001
-OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2-
14. The method of Claim 10 wherein Ra is hydrogen; n is 3, 4, or 5;
R3 and R4 are hydrogen; and
Rl, R2, and R7 are independently chlorine or hydrogen.
15. The method of Claim 10 wherein
Ra is hydrogen; n is 2;
R and R4 are hydrogen; and
R5 and R^ are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, halogen, -NH-C2-C6 alkyl, -N(C1-C6alkyl)2, -NH2, or pyrrolyl.
16. The method of Claim 10 wherein
Ra is hydrogen; n is 2;
R3 and R4 are hydrogen; and
R8 is -CO2H.
17. A method of inhibiting the aggregation of amyloid proteins to form amyloid deposits, the method comprising administering to a patient in need of inhibition of the aggregation of amyloid protein an amyloid protein aggregation inhibiting amount of a compound of Formula I
Figure imgf000116_0001
wherein
Ra is hydrogen; n is 1 to 5 inclusive; R3 and R4 are hydrogen; R1, R7, and R2 are independently chlorine, -N(CH2CH3)2, -OH, CH3-, fluorine, -CF3, phenyl, hydrogen, -OCH2 phenyl,
-O(CH2)3N(CH3)2, -O phenyl, -O(CH2)7CH3, -CH(CH2OCH2CH3)2, pyrrolyl, -CH=CH-phenyl,
-N[(CH2)3CH3]2, substituted phenyl, -OCH2-substituted phenyl, pyrazolyl, or -N(phenyl)2; R5 and R6 are independently hydrogen, -CO2H, -NO2, -OCH3, imidazolyl, -CN, fluorine, -CH3, -CF3, or pyrrolyl; R8 is COOH or tetrazolyl;
A is CH or N;
Rl and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
18. The method of Claim 17 wherein the compound of Formula I is: 2-[[4-[2-(3,4-Dichlorophenyl)ethyl]phenyl]amino-benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -5-nitrobenzoic acid;
2- {4-[4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino} -4-methoxy- 5 -nitrobenzoic acid; 2- {4-[2-(3 ,4-Dihydroxy-phenyl)-ethyl] -phenylamino } benzoic acid;
2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid; 2- {4-[2-(3 ,4,5-Trihydroxy-phenyl)-ethyl]phenylamino } benzoic acid;
2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid;
2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}-4-imidazo- l-yl-5-nitrobenzoic acid;
2- {4-[3 -(3 ,4-Dichlorophenyl)-propyl]phenylamino } benzoic acid; 2- {4-[4-(3 ,4-Dichlorophenyl)butyl]phenylamino } benzoic acid; 2- {4- [4-(3 ,4-Dichloro-phenyl)-butyl] -phenylamino } -5-nitrobenzoic acid;
2- {4-[4-(3 ,4-Dichlorophenyl)-butyl]phenylamino } - 3,5-dinitrobenzoic acid; 2- {4-[5-(3 ,4-Dichlorophenyl)pentyl]phenylamino } -5-nitrobenzoic acid;
2-{4-[5-(3 ,4-Dichloro-phenyl)penty ljphenylamino } -4-methoxy- 5 -nitrobenzoic acid;
2-[4-(3,4-Dichloro-benzyl)-phenylamino]-benzoic acid; 2- {4-[2-(3 ,4-Dimethyl-phenyl)-ethyl] -phenylamino } -5-nitrobenzoic acid;
2-{4-[2-(3,4-Difluoro-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}- benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid; 5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid; 2-(4-Phenethyl-phenylamino)-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methoxy- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-terephthalic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl- benzoic acid; 4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- methanesulfonyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-imidazol-l- yl-benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-nitro- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4-nitro- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -3 -nitrobenzoic acid; 5 -Cyano-2- { 4- [2-(3 ,4-dichloro-phenyl)-ethyl] -phenylamino } - benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-4,6-difluoro- benzoic acid;
6-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-2,3-difluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-fluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -3 -fluoro- benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3-methyl- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4-fluoro- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3,5-difluoro- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-3- trifluoromethyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-6- trifluoromethyl-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5- trifluoromethyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-pyrrol-l-yl- benzoic acid;
2-{4-[2-(4-Benzyloxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- {2- [4-(3 -Dimethylamino-propoxy)-phenyl] -ethyl } - phenylamino)-benzoic acid;
2-{4-[2-(4-Diethylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4- [2-(4-Phenoxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- {4- [2-(4-Octyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[4-(2-Ethoxy-l-ethoxymethyl-ethyl)-phenyl]-ethyl}- phenylamino)-benzoic acid; 2-{4-[2-(4-Pyrrol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Styryl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Octyl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[3-(3,5-Dichloro-phenoxy)-phenyl]-ethyl}-phenylamino)- benzoic acid;
2-(4- { 2- [4-(2-Chloro-6-fluoro-benzyloxy)-phenyl]-ethyl } - phenylamino)-benzoic acid;
2-{4-[2-(4-Pyrazol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4-Diphenylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[4-(3,4-Dichloro-benzyloxy)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2-{4-[2-[(3,4-Dichlorophenyl)propyl]phenylamino}-5-nitrobenzoic acid;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl] phenylamino}-5- nitrobenzoic acid;
2-[[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino- benzoic acid; or
2- [4-(3,4-Dichlorophenyl)phenyl] aminobenzoic acid.
19. The compounds: 2- {4-[4-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -4-methoxy-
5 -nitrobenzoic acid;
2-{4-[2-(3,4-Dihydroxy-phenyl)-ethyl]-phenylamino}benzoic acid; 2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]phenylamino}benzoic acid; 2- {4-[2-(3 ,4,5-Trihydroxy-phenyl)-ethyl]phenylamino } benzoic acid;
2- {4- [3 -(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-methoxy- 5-nitrobenzoic acid;
2- {4- [3 -(3 ,4-Dichlorophenyl)propyl]phenylamino } -4-imidazo- l-yl-5-nitrobenzoic acid; or
2-{4-[4-(3,4-Dichlorophenyl)butyl]phenylamino}benzoic acid.
20. The compounds: 2- {4-[4-(3 ,4-Dichloro-pheny l)-butyl] -phenylamino } -5-nitrobenzoic acid;
2-{4-[4-(3,4-Dichlorophenyl)-butyl]phenylamino}-3,5- dinitrobenzoic acid;
2- {4-[5-(3 ,4-Dichlorophenyl)pentyl]phenylamino} -5-nitrobenzoic acid;
2-{4-[5-(3,4-Dichloro-phenyl)pentyl]phenylamino}-4-methoxy- 5-nitrobenzoic acid;
2- [4-(3,4-Dichloro-benzyl)-phenylamino] -benzoic acid; 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
2- {4- [2-(3 ,4-Difluoro-phenyl)-ethyl] -phenylamino } -5-nitrobenzoic acid;
2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}- benzoic acid; 2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid;
5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid. 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-amino- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- trifluoromethyl-benzoic acid; or
2- {4- [2-(3 ,4-Dichlorophenyl)]phenylamino } -5-nitrobenzoic acid.
21. The compounds:
2-(4-Phenethyl-phenylamino)-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino -5-methoxy- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino -terephthalic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino -5-methyl- benzoic acid;
4- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino -isophthalic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino methanesulfonyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ■5-imidazol- 1- yl-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino •6-nitro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino -4-nitro- benzoic acid; or
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino ■3-nitro- benzoic acid.
22. The compounds:
5-Cyano-2- {4-[2-(3 ,4-dichloro-phenyl)-ethyl]-phenylamino} - benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4,6-difluoro- benzoic acid;
6- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -2,3 -difluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -6-fluoro- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -fluoro- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -3 -methyl- benzoic acid;
2-{4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4-fluoro- benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino} -3 ,5-difluoro- benzoic acid;
2-{4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -3 - trifluoromethyl-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -6- trifluoromethyl-benzoic acid;
2-{4-[3-(4-Diethylaminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Nitrophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(3-Nitrophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Aminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(3-Aminophenyl)propyl]phenylamino}benzoic acid;
2- {4- [2-(4- Aminophenyl)phenylamino } benzoic acid; 2- {4- [2-(4-Dipropylaminophenyl)ethyl] phenylamino } benzoic acid monohydrochloride;
2-{4-[2-(4-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride monohydrate;
2- {4- [3 -(3 -Dipropylaminophenyl)propyl]phenylamino } benzoic acid;
2-{4-[3-(3 -Dimethylaminophenyl)propyl]pheny lamino } benzoic acid; 2-{4-[3-(4-Ethylaminophenyl)propyl]phenylamino}benzoic acid;
2-(N-{4-[3-(4-Diethylaminophenyl)propyl]phenyl}-N- ethylamino)benzoic acid;
2-{4-[2-(3-Dibenzylaminophenyl)ethyl]phenylamino}benzoic acid; 2- {4-[3-(3 -Diethylaminophenyl)propyl]phenylamino } benzoic acid; 2-{4-[2-(3-Aminophenyl)ethyl]phenylamino}benzoic acid;
2-{4-[3-(4-Dimethylaminophenyl)propyl]phenylamino}benzoic acid;
2-{4-[2-(4-Acetylaminophenyl)ethyl]phenylamino}benzoic acid; 2-{4-[2-(3-Acetylaminophenyl)ethyl]phenylamino}benzoic acid; 2-{4-[2-(3-Dipropylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2- {4-[2-(3 -Dibutylaminophenyl)ethyl]phenylamino } benzoic acid monohydrochloride;
2-{4-[3-(4-Acetylaminophenyl)propyl]phenylamino}benzoic acid; 2- {4- [3 -(3 - Acetylaminophenyl)propyl]phenylamino } benzoic acid; 2-{4-[2-(3-Diethylaminophenyl)ethyl]phenylamino}benzoic acid monohydrochloride; 2-{4-[2-(3-Piperidin-l-ylphenyl)ethyl]phenylamino}benzoic acid monohydrochloride;
2-{4-[3-(4-Dipropylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4-[3 -(4-Dibutylaminophenyl)propyl]phenylamino } benzoic acid; 2-{4-[3-(3-Dibutylaminophenyl)propyl]phenylamino}benzoic acid;
2-(4- { 3 - [4-( 1 H-Pyrrol- 1 -yl)phenyl]propyl } phenylamino)benzoic acid;
2-{4-[3-(4-Piperidin-l-ylphenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(4-Diethylcarbamoylphenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(4-Carboxyphenyl)propyl]phenylamino}benzoic acid;
2-{4-[3-(4-Diethylaminomethylphenyl)propyl]phenylamino} benzoic acid;
2-{4-[3-(4-Propylaminophenyl)propyl]phenylamino}benzoic acid; 2-{4-[3-(3-Propylaminophenyl)propyl]phenylamino}benzoic acid;
2- {4- [3 -(4-Pyrrolidin- 1 -y l-phenyl)-propyl] -phenylamino } -benzoic acid;
2- {4- [3 -(3 -Piperidin- 1 yl-phenyl)-propyl] -phenylamino} -benzoic acid; {5-[(l-Butyl-l,2,3,4-tetrahydro-6-quinolyl)methylidene]-4-oxo-2- thioxothiazolidin-3-yl} acetic acid;
{5-[(l-Butyl-2,3-dihydro-lH-indol-5-yl)methylidene]-4-oxo-2- thioxothiazolidin-3 -yl } acetic acid; 3-{5-[(l-Butyl-l,2,3,4-tetrahydroquinolin-6-yl)methylidene]-4- oxo-2-thioxo-thiazolidin-3-yl}propanoic acid;
4- { 5-[( 1 -Butyl- 1 ,2,3 ,4-tetrahydroquinolin-6-yl)methylidene]-4- oxo-2-thioxo-thiazolidin-3-yl}butanoic acid; 2-{4-[3-(3,4-Dichloro-phenyl)-propyl]phenylamino}-5-methyl- benzoic acid;
N-(2- {4- [3 -(3 ,4-Dichloro-pheny l)-propyl] -phenylamino } -benzoyl)- methanesulofnamime;
2- {4-[2-(3 ,4-Dimethyl-phenyl)-ethyl]phenylamino } -5-nitro- benzoic acid;
2-[4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-5-nitro-benzoic acid; 2-{4-[2-(4-Chloro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}- 5-nitro-benzoic acid;
5-Amino-2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}- benzoic acid;
5-Nitro-2-(4-phenethyl-phenylamino)-benzoic acid; 2- {4- [2-(4-Fluoro-3-trifluoromethyl-phenyl)-ethyl] -phenylamino } - benzoic acid;
2- {4- [2-(3 ,4-Difluoro-phenyl)-ethyl] -phenylamino } -5 -nitro- benzoic acid;
{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenyl}-[2-(lH-tetrazol-5-yl)- phenyl]-amine;
2-{4-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-ethyl]-phenylamino}- 5-nitro-benzoic acid; 2-(4-Phenethyl-phenylamino)-benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5-fluoro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -nicotinic acid; 2- {4- [2-(3 -Chloro-phenyl)-ethyl] -phenylamino } -5-nitro-benzoic acid;
2-{4-[2-(4-Chloro-phenyl)-ethyl]-phenylamino}-5-nitro-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-methyl- benzoic acid;
2- {4-[2-(2-Chloro-phenyl)-ethyl]-phenylamino} -5-nitro-benzoic acid; 2-{4-[2-(2,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl]-phenylamino } -6- trifluoromethyl-benzoic acid;
2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5- dimethylamino-benzoic acid;
2-{4-[2-(3,5-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4-{2-[(4aS,8aR)-4-(Octahydro-isoquinolin-2-yl)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2-(3',5'-Dichloro-3-methyl-biphenyl-4-ylamino)-benzoic acid; 2-(3',5'-Dibromo-3-methyl-biphenyl-4-ylamino)-benzoic acid; 2-(4- 1 ,3 -Benzodioxol-5-yl-2-methyl-phenylamino)-benzoic acid; 2-(2,2',4'-Trichloro-biphenyl-4-ylamino)-benzoic acid; 2-(2-Chloro-3',4'-difluoro-biphenyl-4-ylamino)-benzoic acid;
2-(3 '-Bromo-2-chloro-biphenyl-4-ylamino)-benzoic acid; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-nitro- benzoic acid;
3 - {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -benzoic acid; 5-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid;
2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- {4- [2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -4,5- dimethoxy-benzoic acid; 2- {4-[2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -3-nitro- benzoic acid; 3 - {4- [2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -benzoic acid;
5-{4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenylamino}- isophthalic acid; 2-{4-[2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -benzoic acid;
4-(4-{2-[(4aS,8aR)-4-(Octahydro-isoquinolin-2-yl)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-5- methoxy-benzoic acid;
2-{4-[2-(3-Methoxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- {4- [2-(3-Bromo-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- {4-[2-(3 -Fluoro-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -5 -methoxy- benzoic acid;
4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-nicotinic acid; 2-[2-(4-Fluoro-3-trifluoromethyl-phenyl)-2,3-dihydro-lH-isoindol- 5-ylamino] -benzoic acid; or
2- { 4- [2-(3 -Fluoro-4-methyl-phenyl)-ethyl] -phenylamino } -benzoic acid.
23. The compounds:
2- { 4- [2-(3 ,4-Dichloro-phenyl)-e thyl] -phenylamino } -5 - trifluoromethyl-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-pyrrol-l-yl- benzoic acid;
2- {4- [2-(4-Benzyloxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4- { 2- [4-(3 -Dimethylamino-propoxy)-phenyl] -ethyl } - phenylamino)-benzoic acid;
2-{4-[2-(4-Diethylamino-phenyl)-ethyl]-phenylamino}-benzoic acid;
2- {4- [2-(4-Phenoxy-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-{4-[2-(4-Octyloxy-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- { 2- [4-(2-Ethoxy- 1 -ethoxymethyl-ethyl)-phenyl] -ethyl } - phenylamino)-benzoic acid;
2-{4-[2-(4-Pyrrol-l-yl-phenyl)-ethyl]-phenylamino}-benzoic acid; or 2-{4-[2-(4-Styryl-phenyl)-ethyl]-phenylamino}-benzoic acid.
24. The compounds:
2-{4-[2-(4-Dibutylamino-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[2-(4'-Ethyl-biphenyl-4-yl)-ethyl]-phenylamino}-benzoic acid;
2- {4-[2-(4-Octyl-phenyl)-ethyl] -phenylamino } -benzoic acid; 2-(4-{2-[3-(3,5-Dichloro-phenoxy)-phenyl]-ethyl}-phenylamino)- benzoic acid;
2-(4-{2-[4-(2-Chloro-6-fluoro-benzyloxy)-phenyl]-ethyl}- phenylamino)-benzoic acid;
2- { 4- [2-(4-Pyrazol- 1 -yl-phenyl)-ethyl] -phenylamino } -benzoic acid;
2-{4-[2-(4-Diphenylamino-phenyl)-ethyl]-phenylamino}-benzoic acid; 2-(4- {2- [4-(3 ,4-Dichloro-benzyloxy)-phenyl] -ethyl } - phenylamino)-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5-amino- benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- trifluoromethyl-benzoic acid;
2- {4- [2-(3 ,4-Dichlorophenyl)]phenylamino } -5-nitrobenzoic acid; 2-{4-[2-[(3 ,4-Dichloropheny l)propyl]phenylamino } -5 -nitrobenzoic acid;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl] phenylamino}-5- nitrobenzoic acid;
2-[4-(3,4-Dichlorophenyl)phenyl]aminobenzoic acid.
25. 2-[4-[2-(3,4-Dichlorophenyl)ethyl]phenyl]amino-benzoic acid or a pharmaceutically acceptable salt thereof.
26. 2-{4-[3-(3,4-Dichlorophenyl)propyl]phenylamino}benzoic acid or a pharmaceutically acceptable salt thereof.
27. A compound which is selected from:
2- {4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino} -5-nitrobenzoic acid;
4- {4- [3 -(4-Diethylamino-phenyl)-propyl] -phenylamino } -benzoic acid; 4-{4-[3-(4-Diethylamino-phenyl)-propyl]-phenylamino}-3- methoxy-benzoic acid;
2- {4- [2-(3 -Chloro-4-methyl-phenyl)-ethyl] -phenylamino } -5 - methoxy-benzoic acid;
{4-[2-(3-Chloro-4-methyl-phenyl)-ethyl]-phenyl}-(2-methoxy-5- nitro-phenyl)-amine;
2- {4- [3 -(4-Diethylamino-phenyl)-propyl] -phenylamino } -3 -nitrobenzoic acid;
3 - {4- [3 -(4-Diethy 1 amino-phenyl)-propyl] -phenylamino } -benzoic acid; 2-{4-[2-(3 ,4-Dimethoxy-phenyl)-ethyl] -phenylamino } -benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid monosodium;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid monopotassium;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid calcium salt (1:1);
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -benzoate-2- hydroxy- 1 , 1 -bis-hydroxymethyl-ethyl-ammonium; 2-{4-[4-(3,4-Dichloro-phenyl)-butyl]-phenylamino}-5-methoxy- benzoic acid; 2- 4- [2-(3 ,4-Difluoro-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- 3 -[2-(4-Chloro-phenyl)-ethyl] -phenylamino} -benzoic acid; 2- 3-[2-(3,4-Dimethyl-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- 4- [2-(2,4-Dimethoxy-phenyl)-ethyl] -phenylamino } -benzoic acid;
2- 4-[2-(2-Chloro-phenyl)-ethyl]-phenylamino}-benzoic acid; 2- 4- [2-(2-Hydroxy-phenyl)-ethyl] -phenylamino} -benzoic acid; 2- 4- [2-(3 -Chloro-phenyl)-ethyl] -phenylamino } -benzoic acid; 2- 4-(2-Biphenyl-4-yl-ethyl)-phenylamino]-benzoic acid; 2- 4-[2-(2,4-Dichloro-phenyl)-ethyl]-phenylamino}-benzoic acid; 3- 4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -benzoic acid; 4- 4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -benzoic acid; 2- 4-[2-(3 ,4,5-Trimethoxy-phenyl)-ethyl] -phenylamino } -benzoic acid;
2-{4-[2-(4-Phenoxy-phenyl)-ethyl]-phenylamino}-benzoic acid;
2-{4-[5-(3,4-Dichloro-phenyl)-pentyl]-phenylamino}-benzoic acid;
2-(3',5'-Dichloro-biphenyl-4-ylamino)-benzoic acid;
4-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-2-methoxy- 5-nitro-benzoic acid;
2-{4-[3-(3 ,4-Dichloro-pheny l)-propyl] -phenylamino } -5 -fluoro- benzoic acid;
5-Amino-2-{4-[5-(3,4-dichloro-phenyl)-pentyl]-phenylamino}- benzoic acid;
N-(2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-benzoyl)- C,C,C-trifluoro-methanesulfonamide;
N-(2-{4-[3-(3,4-Dichloro-phenyl)-propyl]-phenylamino}-benzoyl)- benzenesulfonamide;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- trifluoromethyl-benzoic acid;
4-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-isophthalic acid; 2-{4-[2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -4- trifluoromethyl-benzoic acid;
2- {4- [2-(3 ,4-Dichloro-pheny l)-ethyl] -phenylamino } -3 - trifluoromethyl-benzoic acid;
2-({4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenyl}-methyl-amino)-5- dimethylamino-benzoic acid;
2-({4-[2-(3 ,4-Dichloro-pheny l)-ethy 1] -phenyl } -methyl-amino)- benzoic acid;
2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl] -phenylamino } -5- dipropylamino-benzoic acid;
5-Dibutylamino-2-{4-[2-(3,4-dichloro-phenyl)-ethyl]- phenylamino}-benzoic acid;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-phenylamino}-5- diethylamino-benzoic acid;
2,2'-[ 1,2-Ethanediylbis (4,1 -phenyl eneimino)]bis-benzoic acid; and
4- [3 -[4-(Diethylamino)phenyl]propyl] -N-(2-methoxy-5 - nitrophenyl)-benzinamine
28. A method of imaging amyloid deposits, the method comprising: a. introducing into a patient a detectable quantity of a labeled compound having the Formula I or a pharmaceutically acceptable salt thereof:
Figure imgf000131_0001
wherein
O
Ra is hydrogen, C -C alkyl, or -CC1-C6 alkyl; n is 0 to 5 inclusive; Rl, R2, R3, R4, R5; R65 and R7 are independently hydrogen, halogen,
-OH, -NH2, NRbRc, -CO2H, -CO2Cι-C6 alkyl, -NO2, -OC^C^ alkyl, -Ci -Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl, O O
II II
-CH=CH-phenyl, -O(CH2)pNRbRC, -CNRbRC, -NHCRb,
-NH(CH2)pNRbRc, -N(C ]_ -C6alkyl)(CH2)pNRbRc,
Figure imgf000132_0001
R8 is COOH, tetrazolyl, -SO2Rd, or -CONHSO2Rd;
Rb and Rc are independently hydrogen, -Cj-C6 alkyl, -(CH2)m-phenyl, or
Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pyrrolyl, imidazolyl, piperazinyl, 4-Cj-C6 alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
Rd is hydrogen, -Cj-Cό alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive; A is CH or N; R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof. b. allowing sufficient time for the labeled compound to become associated with amyloid deposits; and c. detecting the labeled compound associated with the amyloid deposits.
29. The method of Claim 28 wherein the patient has or is suspected to have Alzheimer's disease.
30. The method of Claim 28 wherein the labeled compound is a radio labeled compound.
31. The method of Claim 28 wherein the labeled compound is detected using MRI.
32. The compounds:
2-[4- [2-(3 ,4-Dichloropheny l)ethyl]phenyl] amino-benzoic acid; 2- {4-[2-(3 ,4-Dichloro-phenyl)-ethyl]phenylamino } -5-nitrobenzoic acid;
2- {4-[3 -(3 ,4-Dichlorophenyl)-propyl]phenylamino } benzoic acid; 2-[4-[2-(4-Chloro-3-trifluoromethylphenyl)ethyl]phenyl]amino- benzoic acid; and
2- {4- [3 -(4-Diethylaminophenyl)propyl]phenylamino} benzoic acid.
33. A pharmaceutical formulation comprising a compound of Claim 19 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
34. A pharmaceutical formulation comprising a compound of Claim 20 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
35. A pharmaceutical formulation comprising a compound of Claim 21 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
36. A pharmaceutical formulation comprising a compound of Claim 22 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
37. A pharmaceutical formulation comprising a compound of Claim 23 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
38. A pharmaceutical formulation comprising a compound of Claim 24 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
39. A pharmaceutical formulation comprising a compound of Claim 25 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
40. A pharmaceutical formulation comprising a compound of Claim 26 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
41. A pharmaceutical formulation comprising a compound of Claim 32 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
42. A compound of Formula I.
Figure imgf000134_0001
wherein
O
Ra is hydrogen, C -C alkyl, or -CC]-C6 alkyl; n is 0 to 5 inclusive;
Rl, R2, R3, R4, R5S R6? ^J R7 are independently hydrogen, halogen,
-OH, -NH , NRbRc, -CO2H, -CO2C!-C6 alkyl, -NO2, -O -Ci^ alkyl, -Ci-Cg alkyl, -CF3, -CN, -OCH2 phenyl, -OCH2-substituted phenyl, -(CH2)m-phenyl, -O-phenyl, -O-substituted phenyl,
O O
II II -CH=CH-phenyl, -O(CH2)pNRbRC, -CNRbRc, -NHCR ,
-NH(CH2)pNRbRC, -N(C \ -C6alkyl)(CH2)pNRbRC,
Figure imgf000135_0001
R8 is COOH, tetrazolyl, -SO2Rd, or -CONHSO2Rd;
Rb and Rc are independently hydrogen, -Cj-C6 alkyl, -(CH2) -phenyl, or Rb and Rc taken together with the nitrogen atom to which they are attached form a cyclic ring selected from piperidinyl, pynolyl, imidazolyl, piperazinyl, 4-Cι-C6 alkylpiperazinyl, morpholino, thiomorpholino, decahydroisoquinoline, or pyrazolyl;
Rd is hydrogen, -Ci -C6 alkyl, -CF3, or phenyl; m is 0 to 5 inclusive; p is 1 to 5 inclusive; A is CH orN;
R! and R2, when adjacent to one another, can be methylene-dioxy; or the pharmaceutically acceptable salts thereof.
43. A pharmaceutical formulation comprising a compound of Claim 42 admixed with a pharmaceutically acceptable diluent, excipient, or carrier therefor.
PCT/US2000/015071 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits WO2000076489A2 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
US10/009,611 US6972287B1 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
EEP200100673A EE200100673A (en) 1999-06-10 2000-05-31 Method for Inhibiting Amyloid Protein Aggregation and Imaging for Amyloid Deposits
NZ515621A NZ515621A (en) 1999-06-10 2000-05-31 Medicaments for inhibiting amyloid protein aggregation and methods of imaging amyloid deposits
DZ003252A DZ3252A1 (en) 1999-06-10 2000-05-31
CA002375551A CA2375551A1 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
MXPA01012318A MXPA01012318A (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits.
SK1763-2001A SK17632001A3 (en) 1999-06-10 2000-05-31 A drug for the treatment of Alzheimer's disease, method of inhibiting amyloid protein aggregation and imaging amyloid deposits
HU0202508A HUP0202508A3 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
AU54553/00A AU775157B2 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
IL14697100A IL146971A0 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
BR0011728-5A BR0011728A (en) 1999-06-10 2000-05-31 Method of inhibiting aggregation of amyloid protein and image of amyloid deposits
EA200101135A EA004632B1 (en) 1999-06-10 2000-05-31 Amyloid protein aggregation inhibitors, use thereof, composition based thereon and method for imaging amyloid deposits
PL00352430A PL352430A1 (en) 1999-06-10 2000-05-31 Method of inhibiting agreggation of amyloid proteins and visualising amyloid deposits
EP00939471A EP1225886A2 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
KR1020017015879A KR20020008224A (en) 1999-06-10 2000-05-31 Method of Inhibiting Amyloid Protein Aggregation and Imaging Amyloid Deposits
JP2001502823A JP2003504310A (en) 1999-06-10 2000-05-31 Amyloid protein aggregation inhibition method and amyloid deposit imaging method
APAP/P/2002/002387A AP2002002387A0 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid aggregation and imaging amyloid deposits.
IS6193A IS6193A (en) 1999-06-10 2001-12-07 A method for inhibiting the clustering of amyloid proteins and to obtain a picture of their deposits
NO20015995A NO20015995L (en) 1999-06-10 2001-12-07 Method of inhibiting amyloid-protein aggregation and imaging of amyloid deposits
BG106293A BG106293A (en) 1999-06-10 2002-01-09 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
HR20020026A HRP20020026A2 (en) 1999-06-10 2002-01-10 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
HK03100404.7A HK1048258A1 (en) 1999-06-10 2003-01-16 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13855099P 1999-06-10 1999-06-10
US60/138,550 1999-06-10

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/009,611 A-371-Of-International US6972287B1 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
US10/858,912 Division US20040220235A1 (en) 1999-06-10 2004-06-02 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits

Publications (2)

Publication Number Publication Date
WO2000076489A2 true WO2000076489A2 (en) 2000-12-21
WO2000076489A3 WO2000076489A3 (en) 2002-05-30

Family

ID=22482537

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/015071 WO2000076489A2 (en) 1999-06-10 2000-05-31 Method of inhibiting amyloid protein aggregation and imaging amyloid deposits

Country Status (30)

Country Link
EP (1) EP1225886A2 (en)
JP (1) JP2003504310A (en)
KR (1) KR20020008224A (en)
CN (1) CN1378446A (en)
AP (1) AP2002002387A0 (en)
AU (1) AU775157B2 (en)
BG (1) BG106293A (en)
BR (1) BR0011728A (en)
CA (1) CA2375551A1 (en)
CR (1) CR6528A (en)
DZ (1) DZ3252A1 (en)
EA (1) EA004632B1 (en)
EE (1) EE200100673A (en)
GE (1) GEP20053423B (en)
HK (1) HK1048258A1 (en)
HR (1) HRP20020026A2 (en)
HU (1) HUP0202508A3 (en)
IL (1) IL146971A0 (en)
IS (1) IS6193A (en)
MA (1) MA26805A1 (en)
MX (1) MXPA01012318A (en)
NO (1) NO20015995L (en)
NZ (1) NZ515621A (en)
OA (1) OA11963A (en)
PL (1) PL352430A1 (en)
SK (1) SK17632001A3 (en)
TR (1) TR200103551T2 (en)
WO (1) WO2000076489A2 (en)
YU (1) YU86701A (en)
ZA (1) ZA200109794B (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1193260A1 (en) * 2000-09-29 2002-04-03 Warner-Lambert Company Phenoxazine analogs for the treatment of amyloidosis
EP1266884A1 (en) * 2000-03-22 2002-12-18 BF Research Institute, Inc. Image diagnosis probe based on substituted azobenzene or analogue thereof for disease attributable to amyloid accumulation and composition for image diagnosis containing the same
WO2008077639A1 (en) * 2006-12-22 2008-07-03 Laboratorios Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as dhodh inhibitors
WO2008099210A2 (en) 2007-02-12 2008-08-21 Merck & Co., Inc. Piperazine derivatives for treatment of ad and related conditions
WO2008128129A1 (en) * 2007-04-13 2008-10-23 The Trustees Of The University Of Pennsylvania Halo-stilbene derivatives and their use for binding and imaging of amyloid plaques
US7446222B2 (en) 2002-11-01 2008-11-04 Glaxo Group Limited Phenyl compounds
ES2327372A1 (en) * 2007-04-23 2009-10-28 Laboratorios Almirall S.A. New derivatives of the acids amino-nicotinico and amino-isonicotinico. (Machine-translation by Google Translate, not legally binding)
EP2320897A1 (en) * 2008-08-29 2011-05-18 Treventis Corporation Compositions and methods of treating amyloid disease
WO2011101787A1 (en) * 2010-02-16 2011-08-25 Università Degli Studi Di Siena Non peptidic 14-3-3 inhibitors and the use thereof
WO2012071186A1 (en) * 2010-11-24 2012-05-31 Allergan, Inc. Modulators of s1p receptors
US8501943B2 (en) 2009-03-13 2013-08-06 Almirall, S.A. Sodium salt of 5-cyclopropyl-2-{[2-(2,6-difluorophenyl)pyrimidin-5-yl]amino}benzoic acid as DHODH inhibitor
US8536165B2 (en) 2007-08-10 2013-09-17 Almirall, S.A. Azabiphenylaminobenzoic acid derivatives as DHODH inhibitors
US8598363B2 (en) 2009-10-16 2013-12-03 Almirall, S.A. Process for manufacturing 2-[(3,5-difluoro-3′-methoxy-1,1′biphenyl-4-yl)amino]nicotinic acid
US8865728B2 (en) 2008-06-20 2014-10-21 Almirall, S.A. Combinations comprising methotrexate and DHODH inhibitors
EP3680232A4 (en) * 2017-08-07 2021-08-11 Hiroshima University NOVEL ANTHRANILIC ACID-BASED COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME
US11555009B2 (en) * 2017-03-02 2023-01-17 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences 2-(substituted benzene matrix) aromatic formate FTO inhibitor, preparation method therefor, and applications thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2228367A1 (en) * 2009-03-13 2010-09-15 Almirall, S.A. Addition salts of amines containing hydroxyl and/or carboxylic groups with amino nicotinic acid derivatives as DHODH inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0459136A1 (en) * 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
US5739169A (en) * 1996-05-31 1998-04-14 Procept, Incorporated Aromatic compounds for inhibiting immune response
WO2000076969A1 (en) * 1999-06-10 2000-12-21 Warner-Lambert Company Method of inhibiting amyloid protein aggregation and imaging amyloid deposits using isoindoline derivatives

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0459136A1 (en) * 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
US5739169A (en) * 1996-05-31 1998-04-14 Procept, Incorporated Aromatic compounds for inhibiting immune response
WO2000076969A1 (en) * 1999-06-10 2000-12-21 Warner-Lambert Company Method of inhibiting amyloid protein aggregation and imaging amyloid deposits using isoindoline derivatives

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CLEAVELAND E S ET AL: "SITE OF ACTION OF TWO NOVEL PYRIMIDINE BIOSYNTHESIS INHIBITORS ACCURATELY PREDICTED BY THE COMPARE PROGRAM" BIOCHEMICAL PHARMACOLOGY, PERGAMON, OXFORD, GB, vol. 49, no. 7, 1995, pages 947-954, XP000671777 ISSN: 0006-2952 *
COOPER G J S ET AL: "PURIFICATION AND CHARACTERIZATION OF A PEPTIDE FROM AMYLOID-RICH PANCREASES OF TYPE 2 DIABETIC PATIENTS" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 84, no. 23, 1 December 1987 (1987-12-01), pages 8628-8632, XP002031853 ISSN: 0027-8424 *
HOEPPENER J W M ET AL: "EXTENSIVE ISLET AMYLOID FORMATION IS INDUCED BY DEVELOPMENT OF TYPE II DIABETES MELLITUS AND CONTRIBUTES TO ITS PROGRESSION: PATHOGENESIS OF DIABETES IN A MOUSE MODEL" DIABETOLOGIA, BERLIN, DE, vol. 42, no. 4, April 1999 (1999-04), pages 427-434, XP001009950 ISSN: 0012-186X *
HOEPPENER J W M ET AL: "THE ROLE OF ISLET AMYLOID POLYPEPTIDE(IAPP)/AMYLIN IN THE PATHOGENESIS OF TYPE 2 DIABETES MELLITUS: IMPLICATIONS FROM A TRANSGENIC MOUSE STUDY" EUROPEAN JOURNAL OF ENDOCRINOLOGY, SCANDINAVIAN UNIVERSITY PRESS, NO, vol. 130, no. SUPPL 2, 1994, page 63 XP001009957 ISSN: 0804-4643 *
STURM, ERNST ET AL: "Electrochemical oxidation of acridans" CHEM. BER. (1978), 111(1), 227-39 , 1978, XP002178697 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1266884A1 (en) * 2000-03-22 2002-12-18 BF Research Institute, Inc. Image diagnosis probe based on substituted azobenzene or analogue thereof for disease attributable to amyloid accumulation and composition for image diagnosis containing the same
EP1266884A4 (en) * 2000-03-22 2003-04-23 Bf Res Inst Inc Image diagnosis probe based on substituted azobenzene or analogue thereof for disease attributable to amyloid accumulation and composition for image diagnosis containing the same
EP1193260A1 (en) * 2000-09-29 2002-04-03 Warner-Lambert Company Phenoxazine analogs for the treatment of amyloidosis
US7446222B2 (en) 2002-11-01 2008-11-04 Glaxo Group Limited Phenyl compounds
WO2008077639A1 (en) * 2006-12-22 2008-07-03 Laboratorios Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as dhodh inhibitors
ES2319596A1 (en) * 2006-12-22 2009-05-08 Laboratorios Almirall S.A. Amino nicotinic and isonicotinic acid derivatives as dhodh inhibitors
NO341991B1 (en) * 2006-12-22 2018-03-12 Almirall Sa Amino-nicotinic and isonicotinic acid derivatives useful as DHODH inhibitors
US8691852B2 (en) 2006-12-22 2014-04-08 Amirall, S.A. Amino nicotinic and isonicotinic acid derivatives as DHODH inhibitors
AU2007338321B2 (en) * 2006-12-22 2012-01-19 Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as DHODH inhibitors
US8258308B2 (en) 2006-12-22 2012-09-04 Laboratorios Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as DHODH inhibitors
RU2469024C2 (en) * 2006-12-22 2012-12-10 Альмираль, С.А. Aminonicotinic acid and isonicotinic acid derivatives as dhodg (dihydroorotate dehydrogenase) inhibitors
WO2008099210A2 (en) 2007-02-12 2008-08-21 Merck & Co., Inc. Piperazine derivatives for treatment of ad and related conditions
WO2008128129A1 (en) * 2007-04-13 2008-10-23 The Trustees Of The University Of Pennsylvania Halo-stilbene derivatives and their use for binding and imaging of amyloid plaques
ES2327372A1 (en) * 2007-04-23 2009-10-28 Laboratorios Almirall S.A. New derivatives of the acids amino-nicotinico and amino-isonicotinico. (Machine-translation by Google Translate, not legally binding)
US8536165B2 (en) 2007-08-10 2013-09-17 Almirall, S.A. Azabiphenylaminobenzoic acid derivatives as DHODH inhibitors
US8865728B2 (en) 2008-06-20 2014-10-21 Almirall, S.A. Combinations comprising methotrexate and DHODH inhibitors
US8420640B2 (en) 2008-08-29 2013-04-16 Treventis Corporation Methods of treating amyloid disease using analogs of 1-(4-nitrophenyl) piperazine
EP2320897A4 (en) * 2008-08-29 2012-05-09 Treventis Corp Compositions and methods of treating amyloid disease
EP2320897A1 (en) * 2008-08-29 2011-05-18 Treventis Corporation Compositions and methods of treating amyloid disease
US8501943B2 (en) 2009-03-13 2013-08-06 Almirall, S.A. Sodium salt of 5-cyclopropyl-2-{[2-(2,6-difluorophenyl)pyrimidin-5-yl]amino}benzoic acid as DHODH inhibitor
US8598363B2 (en) 2009-10-16 2013-12-03 Almirall, S.A. Process for manufacturing 2-[(3,5-difluoro-3′-methoxy-1,1′biphenyl-4-yl)amino]nicotinic acid
WO2011101787A1 (en) * 2010-02-16 2011-08-25 Università Degli Studi Di Siena Non peptidic 14-3-3 inhibitors and the use thereof
US8895600B2 (en) 2010-02-16 2014-11-25 Università Degli Studi Di Siena Non peptidic 14-3-3 inhibitors and the use thereof
WO2012071186A1 (en) * 2010-11-24 2012-05-31 Allergan, Inc. Modulators of s1p receptors
US9079864B2 (en) 2010-11-24 2015-07-14 Allergan, Inc. Modulators of S1P receptors
US11555009B2 (en) * 2017-03-02 2023-01-17 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences 2-(substituted benzene matrix) aromatic formate FTO inhibitor, preparation method therefor, and applications thereof
EP3680232A4 (en) * 2017-08-07 2021-08-11 Hiroshima University NOVEL ANTHRANILIC ACID-BASED COMPOUND, AND Pin1 INHIBITOR, THERAPEUTIC AGENT FOR INFLAMMATORY DISEASES AND THERAPEUTIC AGENT FOR CANCER THAT USE THE SAME

Also Published As

Publication number Publication date
IS6193A (en) 2001-12-07
NO20015995D0 (en) 2001-12-07
ZA200109794B (en) 2003-07-01
HRP20020026A2 (en) 2003-08-31
TR200103551T2 (en) 2002-12-23
CA2375551A1 (en) 2000-12-21
JP2003504310A (en) 2003-02-04
EA200101135A1 (en) 2002-06-27
MA26805A1 (en) 2004-12-20
HUP0202508A3 (en) 2003-03-28
BR0011728A (en) 2002-02-26
YU86701A (en) 2004-09-03
CN1378446A (en) 2002-11-06
WO2000076489A3 (en) 2002-05-30
AU5455300A (en) 2001-01-02
NO20015995L (en) 2002-02-04
AP2002002387A0 (en) 2002-03-31
EP1225886A2 (en) 2002-07-31
OA11963A (en) 2006-04-17
KR20020008224A (en) 2002-01-29
GEP20053423B (en) 2005-01-25
CR6528A (en) 2004-02-23
SK17632001A3 (en) 2003-03-04
HK1048258A1 (en) 2003-03-28
BG106293A (en) 2002-06-28
NZ515621A (en) 2004-05-28
HUP0202508A2 (en) 2002-12-28
MXPA01012318A (en) 2002-07-22
PL352430A1 (en) 2003-08-25
EA004632B1 (en) 2004-06-24
EE200100673A (en) 2003-02-17
AU775157B2 (en) 2004-07-22
IL146971A0 (en) 2002-08-14
DZ3252A1 (en) 2000-12-21

Similar Documents

Publication Publication Date Title
EP1225886A2 (en) Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
AU777747B2 (en) Method of inhibiting amyloid protein aggregation and imaging amyloid deposits using isoindoline derivatives
RU2093508C1 (en) Hydroxamic acid derivatives and acid addition salts thereof (variants)
AU4172500A (en) Compounds and methods for modulation of estrogen receptors
US6972287B1 (en) Method of inhibiting amyloid protein aggregation and imaging amyloid deposits
Nikalje et al. Facile synthesis and in vivo hypoglycemic activity of novel 2, 4-thiazolidinedione derivatives
US6770133B2 (en) Stable crystal of thiazolidinedione derivative and process for producing the same
CA2405611A1 (en) Method of inhibiting amyloid protein aggregation and imaging amyloid deposits using aminoindane derivatives
JPS60112778A (en) Morpholine derivatives, manufacture and medicinal composition
Elderfield et al. Synthesis of Certain Simple 4-Aminoquinoline Derivatives1
CA2109816A1 (en) Novel amidoalkyl- and imidoalkyl-piperazines
Sanfilippo et al. Synthesis of (aryloxy) alkylamines. I. Novel antisecretory agents with H+ K+-ATPase inhibitory activity
CZ315098A3 (en) Antagonists of alpha1-adrenergic receptors
Greco et al. Benzothiazole hydroxy ureas as inhibitors of 5-lipoxygenase: Use of the hydroxyurea moiety as a replacement for hydroxamic acid
CA2374616A1 (en) Rhodanine derivatives for use in a method of inhibiting amyloid protein aggregation and imaging amyloid deposits
EL-SHORBAGI et al. Imidazo [2, 1-b] benzothiazoles. I
EP0381628B1 (en) Glycocyamidine derivatives
JP2002201186A (en) Phenoxazine analogous compound useful as amyloid deposition inhibitor, and therapy of alzheimer&#39;s disease and disease associated with amyloidosis
CA1126280A (en) Benzimidazole and benzimidazolidine derivatives with diuretic and antihypertensive activity
Kumar et al. SYNTHESIS AND CHARACTERIZATION OF NOVEL 3-(AMINOMETHYL)-5-BENZYLIDENETHIAZOLIDINE-2, 4-DIONE DERIVATIVES AS ANTICANCER AGENTS
Ismaila et al. DESIGN, SYNTHESIS AND VIRTUAL SCREENING OF CERTAIN 2-PYRAZOLIN-5-ONE AND PYRAZOLIDINE-3, 5-DIONE DERIVATIVES AS POTENTIAL PPAR [gamma] AGONISTS
JPH0657687B2 (en) Fluorodinitrobenzene derivative and method for producing the same
NO159277B (en) 2,6-BIS-HALOGENACYLAMINO-BENZO (1,2-D: 5,4-D [) - BISTIAZOLE DERIVATIVES SUCH AS INTERMEDIATES FOR THE PREPARATION OF THERAPEUTIC ACTIVE 2,6-BISAMINOBINZO- (1,2-D: 5, 4-D [) bisthiazole derivative.

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: P-867/01

Country of ref document: YU

Ref document number: 00808672.9

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AU BA BB BG BR CA CN CR CU CZ DM DZ EE GD GE HR HU ID IL IN IS JP KP KR LC LK LR LT LV MA MG MK MN MX MZ NO NZ PL RO SG SI SK SL TR TT UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 515621

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: IN/PCT/2001/01469/MU

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2000939471

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 54553/00

Country of ref document: AU

Ref document number: 200101135

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2375551

Country of ref document: CA

Ref document number: 2375551

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PV2001-4265

Country of ref document: CZ

Ref document number: 2001/09794

Country of ref document: ZA

Ref document number: 200109794

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/012318

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 17632001

Country of ref document: SK

ENP Entry into the national phase

Ref document number: 2001 502823

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2001/03551

Country of ref document: TR

WWE Wipo information: entry into national phase

Ref document number: 1020017015879

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: DZP2001000040

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 1200101278

Country of ref document: VN

ENP Entry into the national phase

Ref document number: 2000 106293

Country of ref document: BG

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20020022

Country of ref document: UZ

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: P20020026A

Country of ref document: HR

WWP Wipo information: published in national office

Ref document number: 1020017015879

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 10009611

Country of ref document: US

AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AU BA BB BG BR CA CN CR CU CZ DM DZ EE GD GE HR HU ID IL IN IS JP KP KR LC LK LR LT LV MA MG MK MN MX MZ NO NZ PL RO SG SI SK SL TR TT UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWP Wipo information: published in national office

Ref document number: 2000939471

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2001-4265

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 54553/00

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 2000939471

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1020017015879

Country of ref document: KR