Language selection

Search

Patent 2692801 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2692801
(54) English Title: METHODS OF INHIBITING THE FORMATION OF AMYLOID-.BETA. DIFFUSABLE LIGANDS USING ACYLHYDRAZIDE COMPOUNDS
(54) French Title: PROCEDES CONSISTANT A INHIBER LA FORMATION DE LIGANDS DIFFUSABLES D'AMYLOIDE-.BETA. UTILISANT DES COMPOSES D'ACYLHYDRAZIDE
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07C 251/80 (2006.01)
  • A61K 31/15 (2006.01)
  • A61P 9/10 (2006.01)
  • A61P 25/28 (2006.01)
  • C07D 207/323 (2006.01)
  • C07D 211/16 (2006.01)
  • C07D 231/12 (2006.01)
  • C07D 295/192 (2006.01)
  • C07D 307/52 (2006.01)
  • C07D 333/22 (2006.01)
  • C07D 409/04 (2006.01)
  • C07D 409/12 (2006.01)
  • C07D 417/12 (2006.01)
(72) Inventors :
  • LOOK, GARY CHARLES (United States of America)
  • SCHULTZ, LAURI (United States of America)
  • POLOZOV, ALEXANDRE MIKHAYLOVICH (United States of America)
  • BHAGAT, NIKHIL (United States of America)
  • WANG, JIAN (United States of America)
  • ZEMBOWER, DAVID E. (United States of America)
  • GOURE, WILLIAM F. (United States of America)
  • PRAY, TODD (United States of America)
  • KRAFFT, GRANT A. (United States of America)
(73) Owners :
  • ACUMEN PHARMACEUTICALS, INC. (United States of America)
(71) Applicants :
  • ACUMEN PHARMACEUTICALS, INC. (United States of America)
(74) Agent: RIDOUT & MAYBEE LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2008-07-11
(87) Open to Public Inspection: 2009-01-15
Examination requested: 2013-06-03
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2008/069878
(87) International Publication Number: WO2009/009768
(85) National Entry: 2010-01-07

(30) Application Priority Data:
Application No. Country/Territory Date
11/777,264 United States of America 2007-07-12
11/777,266 United States of America 2007-07-12
60/950,724 United States of America 2007-07-19
60/950,810 United States of America 2007-07-19

Abstracts

English Abstract






Disclosed are methods of inhibiting, regulating, and/or modulating the
formation of soluble, globular, non-fibrillar,
neurotoxic amyloid .beta.1-42 oligomers from amyloid .beta.1-42 monomers using
acylhydrazidecompounds. Also disclosed are methods of
treating a patient suffering from diseases associated with the formation of
soluble, globular, non-fibrillar, neurotoxic amyloid .beta.1-42
oligomers using acylhydrazidecompounds.


French Abstract

La présente invention concerne des procédés d'inhibition, de régulation et/ou de modulation de la formation d'oligomères d'amyloïde ß1-42 neurotoxiques non-fibrillaires globulaires solubles, à partir de monomères d'amyloïde ß1-42, au moyen de composés d'acylhydrazide. La présente invention concerne également des procédés de traitement d'un patient atteint de maladies associées à la formation d'oligomères d'amyloïde ß1-42 neurotoxiques non fibrillaires globulaires solubles, au moyen de composés d'acylhydrazide.

Claims

Note: Claims are shown in the official language in which they were submitted.




WHAT IS CLAIMED IS:


1. A method for antagonizing neurotoxic ADDL formation from monomeric
A.beta.1-42 by contacting monomeric A.beta.1-42 with an effective amount of a
compound of the
formula:


Image

wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;


R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;


-116-


each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


2. A method of inhibiting, regulating and/or modulating the ADDL-induced
neuronal dysfunction and/or neurotoxicity in a neuronal cell or neuronal
tissue by inhibiting
the formation of ADDLs which method comprises contacting A.beta.1-42 monomers
which may
be in the presence of a neuronal cell with an effective amount of a compound
of the
formula:


Image

wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;

-117-


each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


3. A method of inhibiting, regulating and/or modulating formation of amyloid-
.beta.
oligomers or the activity of said oligomers in a patient suffering from or at
risk of suffering
from a disease associated with the formation of amyloid-.beta. oligomers,
wherein said method
comprises administering to said patient a therapeutically effective amount of
a compound of
the formula:


Image

wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;


-118-


R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


4. A method for treating a patient suffering from or at risk of suffering from
a
disease selected from the group consisting of Alzheimer's disease, Down's
Syndrome,
stroke, mild cognitive impairment, focal ischemia associated dementia, and
neuronal
degeneration comprising administering to said patient a therapeutically
effective amount of
a compound of the formula:


-119-




Image
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;



-120-




with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


5. A method of enhancing cognitive function in a patient who has diminished
cognitive function due to ADDL neurotoxicity which method comprises
administering to
the patient a therapeutically effective amount of a compound of the formula:

Image
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;



-121-




each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


6. The method as in claims 3 or 5, wherein the disease is associated with
formation of and/or activity of ADDLs.


7. The method as in claim 6, wherein the disease is selected from the group
consisting of Alzheimer's disease, Down's Syndrome, stroke and mild cognitive
impairment.


8. The method as in claims 3 or 5, wherein the disease associated with
insoluble
amyloid fibrils, senile plaques, and/or tangles.


9. The method as in claims 3 or 5, wherein the disease associated with over-
expression of A.beta.1-42 protein.


10. The method as in claim 9, wherein disease is selected from the group
consisting of focal ischemia associated dementia and neuronal degeneration.


11. A method of inhibiting, regulating and or modulating the binding of
neurotoxic ADDLs to spines and/or synapses of a neuronal cell which comprises
contacting
said neuronal cell with an effective amount of a compound of the formula:

Image



-122-




I.

wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;

R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.



-123-




12. A method of inhibiting, regulating and/or modulating the long term
potentiation of neuronal cells which method comprises contacting said cells
with an
effective amount of a compound of the formula:

Image
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);
each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and



-124-




m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


13. A method of treating a patient suffering from diminished cognitive
function
due to suffering from or at risk of suffering from a disease selected from the
group
consisting of Alzheimer's disease, Down's Syndrome, stroke, mild cognitive
impairment,
focal ischemia associated dementia and neuronal degeneration, the method
comprising
administering to said patient a therapeutically effective amount of a compound
of the
formula:

Image
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
X1 and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;

R1 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, C1-6 alkoxy, -N(R5)(R6), C3-10 cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R3 is selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, -
N(R5)(R6), and
R8;
R4 is selected from the group consisting of hydrogen and C1-6 alkyl;
each R5 is independently selected from the group consisting of hydrogen, C1-6
alkyl,
-C(=O)-C1-6 alkyl, and -SO2(R7);



-125-




each R6 is independently selected from the group consisting of hydrogen and C1-
6
alkyl;
R7 is selected from the group consisting of hydrogen, C1-6 alkyl, and aryl
optionally
substituted with 1 to 3 of C1-4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl,
aralkyl, aryl,
-N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
m is 0 or 1;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


14. The method as in claims 3-5 or 13, wherein the compound is administered in

a pharmaceutical composition, further comprising a pharmaceutically acceptable
excipient.

15. The method as in claim 14, wherein the compound is administered in an
amount of from about 0.05 milligram to about 1000 milligram, one or more times
per day.

16. The method as in claims 2, 11 or 12, wherein said neuronal cell are
isolated
from animal brain tissue and grown in tissue culture.


17. The method of as in one of claims 1-16 wherein the compound is of the
formula:

Image
wherein:


-126-




A2 is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R21 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
haloalkyl, -N-S(O)2-R24, and aryl;
R22 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R23 is selected from the group consisting of C1-6 alkyl, amino, and R25;
R24 is selected from the group consisting of C1-6 alkyl, and aryl optionally
substituted
with halo or C1-6 alkyl,
R25 is selected from the group consisting of aryl, heteroaryl, and
heterocyclic, all of
which may be optionally substituted with 1-3 R26 groups;
each R26 is independently selected from the group consisting of hydroxy, halo,
C1-6
alkyl, aralkyl, and aryl;
n is 0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof;
with the proviso that the compound exhibits an IC50 of about 50 µM or less
in the
FRET assay.


18. The method of as in one of claims 1-16 wherein the compound is of the
formula:

Image
wherein:
A1 is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R31 is selected from the group consisting of hydroxy, halo, nitro, C1-6 alkyl,
C1-6
alkoxy, C3-10 cycloalkyl, aryl, heteroaryl optionally substituted with 1-3 C1-
6 alkyl, amino,
-N-SO2-R34, and -N-C(=O)-R34;
R32 is selected from the group consisting of hydrogen, C1-6 alkyl, and C1-6
haloalkyl;
R33 is selected from the group consisting of aryl, biaryl, heteroaryl, and
heterocyclic,
wherein each R33 is optionally substituted with 1-4 R35 groups;



-127-




R34 is selected from the group consisting of C1-6 alkyl and aryl optionally
substituted
with halo;
R35 is selected from the group consisting of hydroxy, nitro, halo, C1-6 alkyl,
C2-6
alkenyl, C1-6 alkoxy, C1-6 haloalkoxy, C3-10 cycloalkyl, halo, amino,
alkylamino,
dialkylamino, aminoacyl, aryl-alkylene, carboxyl, carboxyl ester, and
heterocyclic; and
n is 0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.

19. The method as in one of claims 1-18, wherein the compound has an IC50 of
about 25 µM or less.


20. The method as in one of claims 1-18, wherein the compound has an IC50 of
about 10 µM or less.


21. The method as in one of claims 1-18, wherein the compound has an IC50 of
about 5 µM or less.


22. The method as in one of claims 1-16 wherein A is selected from the group
consisting of phenyl, naphthyl, benzothiophenyl, thiadiazolyl, indanyl,
thiophenyl, indolyl,
pyrazolyl, furanyl, oxoindolinyl, pyridyl, and benzodioxoyl.


23. The method as claims 1-16 or 22 wherein R1 is selected from the group
hydroxy, chloro, fluoro, bromo, iodo, methyl, methoxy, trifluoromethyl,
cyclopropyl,
phenyl, pyrrolyl, methylsulfonamido, 4-chlorophenylsulfonylamido, nitro,
benzo[d][1,3]dioxolyl, amino, thienyl, 5-chlorothienyl, and
methylcarbonylamino.


24. The method as of claims 1-16, 22, or 23 wherein A is optionally
substituted
and is selected from the group consisting of 2-(methylsulfonamido)phenyl, 1H-
indan-7-yl,
1H-indol-7-yl, 1-hydroxy-naphthalen-2-yl, 1-phenyl-5-(trifluoromethyl)-1H-
pyrazol-4-yl,
2-(4-chlorophenyl sulfonamido) phenyl, 2,4-dihydroxyphenyl, 2,6-
difluorophenyl, 2-
acetamidophenyl, 2-aminophenyl, 2-benzothiophene, 2-fluoro-6-hydroxyphenyl, 2-
hydroxy-3-methylphenyl, 2-hydroxy-4-(1H-pyrrol-1-yl)phenyl, 2-hydroxy-4-
methoxyphenyl, 2-hydroxy-5-methoxyphenyl, 2-hydroxy-5-methylphenyl, 2-hydroxy-
5-
nitrophenyl, 2-hydroxy-naphthalen-1-yl, 2-hydroxyphenyl, 2-methylfuran-3-yl, 2-




-128-




oxoindolin-7-yl, 3-(5-chlorothiophen-2-yl)-1H-pyrazol-5-yl, 3-
(benzo[d][1,3]dioxol-5-yl-
1H-pyrazol-5-yl, 3-aminophenyl, 3-chloro-1H-indol-2-yl, 3-chloro-4-methyl-
thiophen-2-yl,
3-chloro-6-fluorobenzo thiophene, 3-chlorobenzothiophen-2-yl, 3-cyclopropyl-1H-
pyrazol-
5-yl, 3-fluorophenyl, 3-hydroxy-naphthalen-2-yl, 3-methyl-1H-pyrazol-5-yl, 4-
(2,5-
dimethyl-1H-pyrrol-1-yl)phenyl, 4-bromophenyl, 4-chloro-2-hydroxyphenyl, 4-
iodo-1-
methyl-1H-pyrazol-3-yl, 4-iodophenyl, 4-methyl-1,2,3-thiadiazol-5-yl, 4-
methylphenyl, 4-
nitrophenyl, 5-bromo-2-hydroxyphenyl, 6-methyl-pyrid-3-yl, 8-hydroxynaphthalen-
1-yl,
and benzo[d][1,3]dioxol-5-yl.


25. The method as in claim 21, wherein X1 and X2 are oxygen.


26. The method as in claim 22, wherein R2 is selected from hydrogen, methyl,
or
trifluoromethyl.


27. The method as in claim 26 wherein m is 0.

28. The method as in claim 26 wherein m is 1.


29. The method as in claim 26, R3 is selected from the group consisting of
butyl,
t-butyl, and amino.


30. The method as in claim 26 wherein R3 is optionally substituted and is
selected from the group consisting of phenyl, biphenyl, thienyl, naphthyl,
furanyl,
piperazinyl, benzothiophenyl, pyrazolyl, morpholino, and piperidinyl.


31. The method as in claim 30 wherein R3 is substituted with one to four
groups
independently selected from the group consisting of hydroxy, chloro, fluoro,
bromo, iodo,
methyl, t-butyl, methoxy, ethoxy, benzyl, phenyl, cyclohexyl,
trifluoromethoxy, allyl,
aminocarbonyl, amino, ethoxycarbonyl, diethylamino, morpholino, nitro, 2,4-
difluorophenylsulfonylamino, and methylcarbonylamino.


32. The method as in one of claims 29-31 wherein R3 is selected from the group

consisting of 5-chloro-2-(2,4-difluorophenylsulfonamido)phenyl, 1-hydroxy-
napthalen-2-yl,
1-methyl-1H-pyrazol-5-yl, 2,3-dihydroxyphenyl, 2,4-dihydroxyphenyl, 2,6-
dihydroxyphenyl, 2-acetamido-5-chlorophenyl, 2-amino-5-chlorophenyl,
benzothiophen-2-



-129-




yl, 2-bromo-6-hydroxyphenyl, 2-furanyl, 2-hydroxy naphthalen-l-yl, 4-hydroxy-
3'-
methoxybiphen-3-yl, 2-hydroxy-3-methoxyphenyl, 2-hydroxy-3-methylphenyl, 2-
hydroxy-
4-methoxyphenyl, 2-hydroxy-4-methylphenyl, 2-hydroxy-4-morpholinophenyl, 2-
hydroxy-
5-methoxyphenyl, 2-hydroxy-5-methylphenyl, 2-hydroxy-5-nitrophenyl, 2-hydroxy-
5-
trifluoro methoxyphenyl, 2-hydroxy-6-methoxy-phenyl, 2-hydroxynaphthalen-l-yl,
2-
hydroxyphenyl, 3,5-dibromo-2-hydroxyphenyl, 3,5-dichloro-2-hydroxy-phenyl, 3,5-

difluoro-2-hydroxyphenyl, 3-allyl-2-hydroxyphenyl, 3-bromo-2-hydroxy-5-
methoxyphenyl,
3-bromo-5-chloro-2-hydroxyphenyl, 3-chloro-5-cyclohexyl-2-hydroxyphenyl, 3-
chloro-5-
fluoro-2-hydroxyphenyl, 3-ethoxy-2-hydroxyphenyl, 3-fluoro-2-hydroxyphenyl, 3-
hydroxy-
5-nitrobenzofuran-2-yl, 4-benzyl piperazin-1-yl, 4-diethylamino-2-
hydroxyphenyl, 4-methyl
piperazin-1-yl, 4-methylphenyl, 4-morpholino, 4-phenylpiperidin-1-yl, 5-bromo-
2-hydroxy-
3-iodophenyl, 5-bromo-2-hydroxyphenyl, 5-chloro thiophen-2-yl, 2-amino-5-
chloro-phenyl,
5-chloro-2-hydroxy-3-methoxyphenyl, 5-chloro-2-hydroxyphenyl, 5-chlorothiophen-
2-yl,
5-ethoxy-2-hydroxyphenyl, 5-methyl thiophen-2-yl, 5-tert-butyl-2-
hydroxyphenyl, 6-
bromo-5-hydroxy-2-(ethoxycarbonyl)benzofuran-4-yl, benzamid-2-yl, -NH2, tert-
butyl, and
thien-2-yl.


33. The method as in one of claims 1-18 wherein the compound is selected from
the group consisting of the compounds in Table 1A and 1B.


34. A compound selected from the group consisting of:
2-hydroxy-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide;
N'-(4-(4-benzylpiperazin-1-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

3-chloro-6-fluoro-N'-(4-(furan-2-yl)-4-oxobutan-2-ylidene)benzo[b]thiophene-2-
carbohydrazide;

N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)methanesulfonamide;
2-hydroxy-N'-(1,1,1-trifluoro-4-(4-methylpiperazin-1-yl)-4-oxobutan-2-
ylidene)benzohydrazide;

2-hydroxy-N'-(1,1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-
ylidene)benzohydrazide;



-130-




N'-(4-(benzo [b]thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

2-hydroxy-N'-(1,1,1-trifluoro-4-(1-methyl-1H-pyrazol-5-yl)-4-oxobutan-2-
ylidene)benzohydrazide;

3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(5-methylthiophen-2-yl)-4-oxobutan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide;
3-chloro-6-fluoro-N'-(1,1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide;
2-hydroxy-N'-(1,1,1-trifluoro-4-(5-methylthiophen-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide;

3-chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide;

3 -chloro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo[b]thiophene-
2-carbohydrazide;

4-chloro-N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)benzenesulfonamide;
N'-(4-(benzo[b]thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-3-chloro-6-

fluorobenzo[b]thiophene-2-carbohydrazide;
2-hydroxy-N'-(1,1,1-trifluoro-4-morpholino-4-oxobutan-2-
ylidene)benzohydrazide;
N'-(4-(5-chlorothiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

5-chloro-2-hydroxy-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;

3-chloro-4-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)thiophene-2-carbohydrazide;
N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1H-indole-7-
carbohydrazide;

3-chloro-N'-(4-(5-chlorothiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-6-

fluorobenzo[b]thiophene-2-carbohydrazide;
3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide;
2-hydroxy-3-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;



-131-




4-nitro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;
N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)benzo[b]thiophene-2-

carbohydrazide;

2-hydroxy-N'-(4-oxo-4-(4-phenylpiperidin-1-yl)butan-2-ylidene)benzohydrazide;
3-chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1H-indole-
2-
carbohydrazide;

3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide;
3-(2-(2-hydroxybenzoyl)hydrazono)butanamide;
1-phenyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-5-
(trifluoromethyl)-1H-pyrazole-4-carbohydrazide;
2,6-difluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-4-(1H-pyrrol-1-
yl)benzohydrazide;

1-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-2-naphthohydrazide;
N'-(3-chloro-5-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(2,6-dihydroxybenzylidene)-2-hydroxybenzohydrazide;

3-chloro-N'-(2-hydroxy-5-methoxybenzylidene)-1H-indole-2-carbohydrazide;
N'-(2-bromo-6-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(3-chloro-5-cyclohexyl-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(5-tert-butyl-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-((4-hydroxy-3'-methoxybiphenyl-3-yl)methylene)benzohydrazide;
4-chloro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
N'-(2-hydroxy-5-methoxybenzylidene)-2-oxoindoline-7-carbohydrazide;
2-hydroxy-N'-(2-hydroxy-6-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-(trifluoromethoxy)benzylidene)benzohydrazide;
N'-(2-hydroxy-5-methoxybenzylidene)-1H-indole-7-carbohydrazide;



-132-




3-chloro-6-fluoro-N'-((2-hydroxynaphthalen-1-yl)methylene)benzo[b]thiophene-2-
carbohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-3-methylbenzohydrazide;
2-((2-(2-hydroxybenzoyl)hydrazono)methyl)benzamide;
N'-(2-amino-5-chlorobenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide;
4-fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
3-chloro-N'-(2-hydroxy-5-methoxybenzylidene)-4-methylthiophene-2-
carbohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-methylbenzohydrazide;
N'-(3-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
5-fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
ethyl 6-bromo-5-hydroxy-4-((2-(2-hydroxybenzoyl)hydrazono)methyl)-2-
methylbenzofuran-3-carboxylate;
3-(benzo[d][1,3]dioxol-5-yl)-N'-(2-hydroxy-5-methoxybenzylidene)-1H-pyrazole-5-

carbohydrazide;

N'-(4-(diethylamino)-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-4-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-methoxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
N'-(5-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(2,3-dihydroxybenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-4-morpholinobenzylidene)benzohydrazide;
2-hydroxy-N'-((3-hydroxy-5-nitrobenzofuran-2-yl)methylene)benzohydrazide;
N'-(2,4-dihydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(5-chloro-2-hydroxy-3-methoxybenzylidene)-2-hydroxybenzohydrazide;
3-chloro-N'-(5-chloro-2-hydroxybenzylidene)-4-methylthiophene-2-
carbohydrazide;
2-amino-N'-(2-amino-5-chlorobenzylidene)benzohydrazide;


-133-




2-hydroxy-N'-(2-hydroxy-3-methoxybenzylidene)benzohydrazide;
2-fluoro-6-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-3-methylbenzylidene)benzohydrazide;
N'-((2-hydroxynaphthalen-1-yl)methylene)-3-methyl-1H-pyrazole-5-
carbohydrazide;

5-bromo-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-nitrobenzylidene)benzohydrazide;
N-(2-(2-(2-hydroxy-5-
methoxybenzylidene)hydrazinecarbonyl)phenyl)methanesulfonamide;
N'-(3,5-difluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(1-(5-chloro-2-hydroxyphenyl)-2,2,2-trifluoroethylidene)-2-
hydroxybenzohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-nitrobenzohydrazide;
8-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide;
N'-(3-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
3-(5-chlorothiophen-2-yl)-N'-(2-hydroxy-5-methoxybenzylidene)-1H-pyrazole-5-
carbohydrazide;

N'-(3-bromo-5-chloro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(3-bromo-2-hydroxy-5-methoxybenzylidene)-2-hydroxybenzohydrazide;
2-amino-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;

N'-(5-chloro-2-hydroxybenzylidene)-4-methyl-1,2,3-thiadiazole-5-
carbohydrazide;
N-(2-(2-(2-acetamido-5-chlorobenzylidene)hydrazinecarbonyl)phenyl)acetamide;
and

N-(4-chloro-2-((2-(2-hydroxybenzoyl)hydrazono)methyl)phenyl)-2,4-
difluorobenzenesulfonamide;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.



-134-

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
METHODS OF INHIBITING THE FORMATION OF AMYLOID-0 DIFFUSABLE
LIGANDS USING ACYLHYDRAZIDE COMPOUNDS

CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to United States Application Serial Nos.
11/777,264 and 11/777,266, both filed on July 12, 2007 and claims the benefit
under 35
U.S.C. 119(e) of United States Provisional Serial Nos. 60/950,724 and
60/950,810, both
filed on July 19, 2007, all of which are incorporated by reference in their
entirety.

BACKGROUND OF THE INVENTION
Field of the Invention

This invention relates to methods of inhibiting, regulating, and/or modulating
the formation of soluble, globular, non-fibrillar, neurotoxic amyloid 01_42
oligomers from
amyloid 01_42 monomers using acylhydrazide compounds. This invention also
relates to
methods of treating a patient suffering from diseases associated with the
formation of
soluble, globular, non-fibrillar, neurotoxic amyloid 01_42 oligomers by
administering
acylhydrazide compounds to the patients.

State of the Art

Alzheimer's disease (AD) is a fatal progressive dementia that has no cure at
present. Although the molecular basis of the disease is not established,
considerable
evidence now implicates neurotoxins derived from amyloid beta (A(3) peptides
and in
particular the 42-amino acid amyloid beta peptide (A(3i_42). A(3 is an
amphipathic peptide,
the abundance of which is increased by gene mutations and risk factors linked
to AD.
Fibrils formed from A(3 constitute the cores of amyloid senile plaques, which
are hallmarks
of AD brain. Analogous fibrils generated in vitro are lethal to cultured brain
neurons.
These findings provided the central rationale for the original amyloid cascade
hypothesis, a
theory in which memory loss was proposed to be the consequence of neuron death
caused
by fibrillar A(3 (Hardy and Higgins (1992) Science 256:184-185).

-1-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Despite its strong experimental support and intuitive appeal, the original
amyloid cascade hypothesis has proven inconsistent with key observations,
including the
poor correlation between dementia and amyloid senile plaque burden (Katzman
(1988) Ann.
Neurol. 23(2):138-144). Using a transgenic mouse model of AD, two surprising
findings
were obtained when the mice were treated with monoclonal antibodies against
A(3: (1)
vaccinated mice showed reversal of memory loss, with recovery evident in 24
hours; and (2)
cognitive benefits of vaccination accrued despite no change in senile plaque
levels (Dodart
et al. (2002) Nat. Neurosci 5:452-457; Kotilinek et al. (2002) J. Neurosci.
22:6331-6335).
Such findings are not consistent with a mechanism for memory loss dependent on
neuron
death caused by amyloid fibrils.

Salient flaws in the original amyloid cascade hypothesis have been
eliminated by an updated amyloid cascade hypothesis that incorporates a role
for additional
neurologically active molecules formed by A(3 self-assembly. These molecules
are amyloid
0-derived diffusible ligands (ADDLs), which assemble from A(31_42 at low
concentrations
(Lambert et al. (1998) Proc. Natl. Acad. Sci. USA 95:6448-6453). Essentially
the missing
links in the original amyloid cascade hypothesis, ADDLs rapidly inhibit long
term
potentiation (Lambert et al. (1998) Proc. Natl. Acad. Sci. USA 95:6448-6453;
Walsh et al.
(2002) Nature 416: 535-539; Wang et al. (2002) Brain Res. 924:133-140), a
classic
experimental paradigm for memory and synaptic plasticity. In the updated A(3
cascade
hypothesis memory loss stems from synapse failure, prior to neuron death, with
failure
being caused by ADDLs, not fibrils (Hardy and Selkoe (2002) Science 297:353-
356).
ADDLs occur in brain tissue and are strikingly elevated in AD brain tissue
compared to age
matched controls (Kayed et al. (2002) Science 300:486-489; Gong et al. (2003)
Proc. Natl.
Acad. Sci. USA 100:10417-10422) and in AD transgenic mice models (Kotilinek et
al.
(2002) J. Neurosci. 22:6331-6335; Chang et al. (2003) J. Mol. Neurosci. 20:305-
313).

A simplistic mechanistic approach to this theory can be illustrated as
follows:
amyloid senile - Monomeric AB 1_42 ADDLs
plaque

where formation of ADDLs is a separate pathway from formation of amyloid
senile plaque
both of which are in equilibrium with monomeric A(3i_42.

-2-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Further experiments have shown important neurological properties of
ADDLs. ADDLs were shown to have selective toxicity to hippocampal CAl neurons
compared with CA3 neurons, and the complete absence of toxicity towards
cerebellar
neurons (Kim et al. (2003) FASEB J. 17:118-120). Ventricular injection of
A(3i_42
oligomers into wild-type rats resulted in rapid, compromised behavioral models
with
complete recovery occurring within 24 hours (Cleary et al. (2005) Nat.
Neurosci. 8:79-84)
and these deficits are attributed to higher order oligomers, specifically 12-
mer oligomers
(Lesne et al. (2006) Nature 440:352-357). ADDL binding to neurons occurs with
high
specificity and is localized to post-synaptic receptors on a subset of
hippocampal neurons
(Lacor et al. (2004) J. Neurosci. 24:10191-10200). This triggers the rapid and
persistent up-
regulation of the immediate early gene product arc, translation of which is
activity
dependent at polyribosomes localized to subsets of dendritic spines (Steward
et al. (1998)
Neuron 21:741-751; Guzowski et al. (2000) J. Neurosci. 20:3993-4001). More
recently,
ADDLs have been implicated as upstream activators of tau phosphorylation and
have been
shown to interfere with animal behavior at femtomolar levels (Matsubara et al.
(2004)
Neurobiol. Aging 25:833-841).

The reversibility of memory loss in mouse models, coupled with the
neurological properties of ADDLs and their presence in an AD brain, provides
strong
support for the hypothesis that AD is a disease of ADDL-induced synaptic
failure (Lambert
et al. (1998) Proc. Natl. Acad. Sci. USA 95:6448-6453; Klein et al. (2001)
Trends Neurosci.
24:219-220; Selkoe (2002) Science 298:789-791).

The use of antibodies specific to ADDLs is a powerful way to modulate the
equilibrium between monomeric A(3i_42 and ADDLs thereby providing treatment
for disease
conditions mediated by ADDLs. However, antibody delivery is typically limited
to
injectable solutions which pose patient compliance issues as well as the
presence of an
attending clinician. Small molecules that modulate this equilibrium,
deliverable by non-
injectable means such as oral delivery, transdermal delivery, pulmonary
delivery, nasal
delivery, etc. would be particularly beneficial.

A number of small molecules developed originally as amyloid fibril blockers
are purported to possess A(3 oligomer assembly blocking properties. Some of
these

-3-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
compounds include AlzhmedTM (Gervais (2004) Neurobiol. Aging 25:S11-12),
Clioquinol
(Ritchie et al. (2003) Arch. Neurol. 60:1685-1691), substituted 0-
cyclodextrins (Yu et al.
(2002) J. Mol. Neurosci. 19:51-55), trehalose (Lui (2005) Neurobiol. Disease
20:74-81),
simple amino, carbonyl, and nitro substituted phenols (De Felice et al. (2001)
FASEB J.
March 20; De Felice et al. (2004) FASEB J. 18:1366-1372), Curcumin (Yang et
al. (2005)
J. Biol. Chem. 280(7):5892-5901), cyclohexanehexol analogs (McLaurin et al.
(2006)
Nature Med. 12:801-808), spirosterols (Lecanu et al.(2004) Steroids 69:1-16)
and tricyclic
pyrones (Maeqawa et al. (2006) J. Neurochem. 98:57-67). Two of these
compounds,
AlzhemedTM and Clioquinol, have progressed into clinical trials.

AlzhemedTM (3-amino-l-propanesulfonic acid), a so-called "GAG mimetic,"
is proposed to reduce soluble and insoluble amyloid levels by binding to A(3
monomer,
although no experimental details have appeared to confirm the proposed mode of
action.
AlzhemedTM has recently completed a 20 month open-label extension of a Phase
II trial, and
there are reports of slowed cognitive decline in some patients with mild AD,
however, no
efficacy was observed during the blinded phase of the study (Gervais (2004)
Neurobiol.
Aging 25:S11-12).

The second compound in a phase II clinical trial, Clioquinol, was shown to
stabilize the patients' cognitive ability compared to untreated patients and
showed lower
A(3i_42 levels in their plasma (Ritchie et al. (2003) Arch. Neurol. 60:1685-
1691). However,
a toxic impurity (a di-iodo form of Clioquinol) made during production has
resulted in the
study being halted and Clioquinol being replaced with an analog termed PBT2
(Blennow et
al. (2006) Lancet 368:387-403).

Lastly, an unidentified compound or compounds from an extract of ginko
biloba leaves was reported to lower the levels of A(3i_42 trimers and
tetramers and increase
the levels of high molecular weight polymers in a dose dependent manner (Yao
et al. (2001)
Brain Res. 889:181-190). Dose dependent protection against A(3 oligomer
induced toxicity
to PC-12 cells was also reported.

Of the compounds reported to block A(3 assembly or bind to A(3i_42
monomer, few appear to have high therapeutic potential. Given its very simple
structure
and hydrophilic properties, it is highly unlikely that AlzhemedTM has high and
selective
-4-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
affinity for A(31_42 monomer. Any effect that AlzhemedTM has on A(3
aggregation or
disaggregation is likely attributable to its interaction with ionic residues
near the N-terminus
of A(3i_42 . The cyclodextrins do not have either lead-like or drug-like
properties that would
recommend them for development (Oprea et al. (2001) J. Chem. Inf. Comput. Sci.
41:1308-
1315; Vieth et al. (2004) J. Med. Chem. 47:224-232), and the phenols of De
Felice contain
aldehyde and nitro functionalities that are often considered reactive and
excluded from
pharmaceutical screening libraries (Walters and Namchuk (2003) Nat. Rev. 2:259-
266). A
number of molecules containing the phenol functionality have been reported as
"frequent
hitters" in screening libraries (Roche et al. (2002) J. Med. Chem. 45:137-
142). Thus,
further evaluation of the activity and selectivity of the phenols of De Felice
is needed to
confirm that these compounds are valid hits. Some compounds with a steroidal
backbone
have been reported to be promiscuous inhibitors due to an unexpected self
aggregation
process (McGovern et al. (2002) J. Med. Chem. 45:1712-1722), which may explain
the
ambiguous spirosterol results. Finally, the active ingredient in the ginko
biloba extract is
unknown. Thus, most of the purported A(3 assembly blockers would not be
considered
compounds for therapeutic development.

Notwithstanding these putative results and as noted above, binding assays
indicate that these compounds are, at best, moderate antagonists to ADDL
formation.
Accordingly, it would be particularly beneficial to provide for small
molecules which
provide enhanced inhibition, regulation, and/or modulation of ADDL formation.
SUMMARY OF THE INVENTION

This invention is directed to the discovery that soluble, globular, non-
fibrillar, neurotoxic A(3i_42 (ADDL) formation can be antagonized by certain
compounds. It
is contemplated that by antagonizing (or inhibiting, modulating, or
regulating) ADDL
formation, these compounds may be used to treat patients suffering from
diseases mediated,
at least in part by, ADDL formation. It is further contemplated that these
compounds may
also be used to inhibit, modulate or regulate neuronal dysfunction or
neurotoxicity that is
caused by ADDLs.

In one embodiment of the invention, the methods employ compounds of the
formula:
-5-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
R2 X2

/
n A NH R3
l Rf N
Xl
m
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
Xi and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
Ri is selected from the group consisting of hydroxy, halo, nitro, Ci_6 alkyl,
Ci_6
haloalkyl, C1_6 alkoxy, -N(Rs)(R6), C3_1o cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, Ci_6 alkyl, and Ci_6
haloalkyl;
R3 is selected from the group consisting of Ci_6 alkyl, Ci_6 alkoxy, -
N(Rs)(R), and
Rg ;
R4 is selected from the group consisting of hydrogen and C1_6 alkyl;
each R 5 is independently selected from the group consisting of hydrogen, Ci_6
alkyl,
-C(=O)-Ci_6 alkyl, and -S02(R 7);
each R6 is independently selected from the group consisting of hydrogen and
C1_6
alkyl;
R' is selected from the group consisting of hydrogen, C1_6 alkyl, and aryl
optionally
substituted with 1 to 3 of Ci_4 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, biaryl, heteroaryl,
and
heterocyclic, wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1_6 alkyl, C2_6 alkenyl, C1_6 alkoxy, C1_6 haloalkoxy, C3_10 cycloalkyl,
aralkyl, aryl,
-N(Rs)(R), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
mis0orl;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.

-6-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The invention also contemplates using compounds of formula II or III:
R22 O
A2
NH ~
R21 \N R23
n
O
It
wherein:
A2 is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R 21 is selected from the group consisting of hydroxy, halo, nitro, C1_6
alkyl, C1_6
haloalkyl, -N-S(O)2-R24, and aryl;
R22 is selected from the group consisting of hydrogen, Ci_6 alkyl, and Ci_6
haloalkyl;
R23 is selected from the group consisting of C1_6 alkyl, amino, and R 25;
R24 is selected from the group consisting of C1_6 alkyl, and aryl optionally
substituted
with halo or Ci_6 alkyl,

R 25 is selected from the group consisting of aryl, heteroaryl, and
heterocyclic, all of
which may be optionally substituted with 1-3 R26 groups;
each R26 is independently selected from the group consisting of hydroxy, halo,
Ci_6
alkyl, aralkyl, and aryl;
nis0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.

R33
NH
` nA~ 32
r R31 ~ N R
O
III.
wherein:
Ai is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R31 is selected from the group consisting of hydroxy, halo, nitro, Ci_6 alkyl,
Ci_6
alkoxy, C3_10 cycloalkyl, aryl, heteroaryl optionally substituted with 1-3
Ci_6 alkyl, amino,
-N-S02-R34, and -N-C(=O)-R34;
R32 is selected from the group consisting of hydrogen, C1_6 alkyl, and C1_6
haloalkyl;
-7-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
R33 is selected from the group consisting of aryl, biaryl, heteroaryl, and
heterocyclic,
wherein each R33 is optionally substituted with 1-4 R35 groups;
R34 is selected from the group consisting of Ci_6 alkyl and aryl optionally
substituted
with halo;
R35 is selected from the group consisting of hydroxy, nitro, halo, C1_6 alkyl,
C2_6
alkenyl, C1_6 alkoxy, C1_6 haloalkoxy, C3_1o cycloalkyl, halo, amino,
alkylamino,
dialkylamino, aminoacyl, aryl-alkylene, carboxyl, carboxyl ester, and
heterocyclic; and
nis0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.
Accordingly, in one of its method aspects, this invention is directed to a
method for antagonizing neurotoxic ADDL formation from monomeric A(3i_42 by
contacting
monomeric A(3i_42 with an effective amount of a compound of formula I, II, or
III.

In yet another embodiment, the invention is directed to a method of
inhibiting, regulating and/or modulating the ADDL-induced neuronal dysfunction
and/or
neurotoxicity in a neuronal cell by inhibiting the formation of ADDLs. The
method
comprises contacting A(3i_42 monomers which may be in the presence of neuronal
cells with
an effective amount of a compound of formula I, II, or III.

In another embodiment, the invention is directed to a method of inhibiting,
regulating and/or modulating amyloid-(3 oligomer formation or the activity of
such
oligomers in a patient suffering from or at risk from suffering from a disease
associated with
the formation of A(31_42 oligomers. The method comprises administering to the
patient a
therapeutically effective amount of a compound of formula I, II, or III.

In yet another embodiment, the invention is directed to a method for treating
a patient suffering from or at risk of suffering from an ADDL-related disease
selected from
the group consisting of Alzheimer's disease, Down's Syndrome, stroke, mild
cognitive
impairment, focal ischemia associated dementia, and neuronal degeneration. The
method
comprises administering to said patient a therapeutically effective amount of
a compound of
formula I, II, or III.

-8-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878

In another embodiment, the present invention relates to a composition for use
in the treatment of a patient suffering from or at risk of suffering from a
disease selected
from the group consisting of Alzheimer's disease, Down's Syndrome, stroke,
mild cognitive
impairment, focal ischemia associated dementia, and neuronal degeneration
wherein the
composition comprises a therapeutically effective amount of a compound of
formula I, II, or
III.

One embodiment of the invention is directed to a method of enhancing
cognitive function in a patient who has diminished cognitive function due to
ADDL
neurotoxicity. The method comprises administering to the patient a
therapeutically
effective amount of a compound of formula I, II, or III.

In some embodiments of the invention, the diminished cognitive function in
a patient is due to the patient suffering from or at risk of suffering from a
disease associated
with the formation of and/or activity of ADDLs. In other embodiments, the
disease is
selected from the group consisting of Alzheimer's disease, Down's Syndrome,
stroke and
mild cognitive impairment.

In other embodiments of the invention, the diminished cognitive function in
a patient is due to the patient suffering from or at risk of suffering from a
disease associated
with insoluble amyloid fibrils, senile plaques, and/or tangles. Alternatively,
the diminished
cognitive function in a patient is due to the patient suffering from or at
risk from suffering
from a disease associated with over-expression of A(3i_42 protein. In some
embodiments, the
disease is selected from the group consisting of focal ischemia associated
dementia and
neuronal degeneration.

In another embodiment, the invention is directed to a method of inhibiting,
regulating and or modulating the binding of neurotoxic ADDLs to spines and/or
synapses of
a neuronal cell. The method comprises contacting said neuronal cell with an
effective

amount of a compound of formula I, II, or III.

In yet another embodiment, the invention is directed to a method of
inhibiting, regulating and/or modulating the long term potentiation of
neuronal cells. The
-9-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
method comprises contacting said cells with an effective amount of a compound
of formula
I, II, or III.

In another embodiment, the invention is directed to a method of treating a
patient suffering from diminished cognitive function due to the patient
suffering from or at
risk of suffering from a disease selected from the group consisting of
Alzheimer's disease,
Down's Syndrome, stroke, mild cognitive impairment, focal ischemia associated
dementia
and neuronal degeneration. The method comprises administering to said patient
a
therapeutically effective amount of a compound of formula I, II, or III.

In one embodiment of the invention, the compound is administered in an
amount of from about 0.05 milligrams to 1000 milligrams, one or more times per
day. In
another embodiment of the invention, the compound is administered in a
pharmaceutical
composition, further comprising a pharmaceutically acceptable excipient.

In one embodiment of the application, the compounds of the invention have
an IC50 of about 50 M or less when tested in the FRET assay. It is
contemplated that
compounds of this invention have an ICSO of about 50 M or less in an assay
that tests for
formation of ADDLs. In one embodiment, this assay is Example 16 described
below. In
another embodiment of the invention, the compounds have an IC50 of 25 M or
less when
tested in the FRET assay. In another embodiment of the invention, the
compounds have an
IC50 of 10 M or less. In yet another embodiment of the invention, the
compounds have an
ICSO of 5 M or less.

Also included within in the scope of the invention is a compound selected
from the group consisting of:

2-hydroxy-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide;

N'-(4-(4-benzylpiperazin-l-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

3-chloro-6-fluoro-N'-(4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;
-10-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)methanesulfonamide;
2-hydroxy-N'-(1,1,1-trifluoro-4-(4-methylpiperazin-l-yl)-4-oxobutan-2-
ylidene)benzohydrazide;
2-hydroxy-N'-(l, 1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-
ylidene)benzohydrazide;
N'-(4-(benzo [b]thiophen-2-yl)- 1, 1, 1 -trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

2-hydroxy-N'-(1,1,1-trifluoro-4-(1-methyl-1 H-pyrazol-5 -yl)-4-oxobutan-2-
ylidene)benzohydrazide;

3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(5-methylthiophen-2-yl)-4-oxobutan-
2-ylidene)benzo [b]thiophene-2-carbohydrazide;
3-chloro-6-fluoro-N'-(1,1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;

2-hydroxy-N'-(1,1,1-trifluoro-4-(5 -methylthiophen-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide;

3-chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;
3-chloro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;
4-chloro-N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)benzenesulfonamide;
N'-(4-(benzo [b]thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-3-
chloro-6-fluorobenzo [b]thiophene-2-carbohydrazide;

2-hydroxy-N'-(I, 1,1-trifluoro-4-morpholino-4-oxobutan-2-
ylidene)benzohydrazide;
N'-(4-(5 -chlorothiophen-2-yl)- 1, 1, 1 -trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide;

5-chloro-2-hydroxy-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;

3-chloro-4-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)thiophene-2-carbohydrazide;
N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1 H-indole-7-
carbohydrazide;
-11-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
3-chloro-N'-(4-(5-chlorothiophen-2-yl)- 1, 1, 1 -trifluoro-4-oxobutan-2-
ylidene)-6-fluorobenzo[b]thiophene-2-carbohydrazide;
3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;

2-hydroxy-3-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;

4-nitro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;

N'-(l,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;
2-hydroxy-N'-(4-oxo-4-(4-phenylpiperidin-l-yl)butan-2-
ylidene)benzohydrazide;
3-chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1 H-
indole-2-carbohydrazide;

3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo [b]thiophene-2-carbohydrazide;
3-(2-(2-hydroxybenzoyl)hydrazono)butanamide;
1-phenyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-5-
(trifluoromethyl)-1 H-pyrazole-4-carbohydrazide;

2,6-difluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5 -methoxybenzylidene)-4-(1 H-pyrrol-l-
yl)benzohydrazide;

1-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-2-naphthohydrazide;
N'-(3-chloro-5-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(2,6-dihydroxybenzylidene)-2-hydroxybenzohydrazide;

3-chloro-N'-(2-hydroxy-5-methoxybenzylidene)-1 H-indole-2-
carbohydrazide;

N'-(2-bromo-6-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(3-chloro-5-cyclohexyl-2-hydroxybenzylidene)-2-
hydroxybenzohydrazide;
N'-(5-tert-butyl-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
-12-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2-hydroxy-N'-((4-hydroxy-3'-methoxybiphenyl-3-
yl)methylene)benzohydrazide;
4-chloro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
N'-(2-hydroxy-5-methoxybenzylidene)-2-oxoindoline-7-carbohydrazide;

2-hydroxy-N'-(2-hydroxy-6-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-(trifluoromethoxy)benzylidene)benzohydrazide;
N'-(2-hydroxy-5-methoxybenzylidene)-1 H-indole-7-carbohydrazide;
3-chloro-6-fluoro-N'-((2-hydroxynaphthalen-l-
yl)methylene)benzo [b]thiophene-2-carbohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-3-methylbenzohydrazide;
2-((2-(2-hydroxybenzoyl)hydrazono)methyl)benzamide;
N'-(2-amino-5-chlorobenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide;
4-fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;

3-chloro-N'-(2-hydroxy-5-methoxybenzylidene)-4-methylthiophene-2-
carbohydrazide;

2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-methylbenzohydrazide;
N'-(3-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
5-fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;

ethyl 6-bromo-5-hydroxy-4-((2-(2-hydroxybenzoyl)hydrazono)methyl)-2-
methylbenzofuran-3-carboxylate;
3-(benzo [d] [ 1,3 ] dioxol-5-yl)-N'-(2-hydroxy-5-methoxybenzylidene)-1 H-
pyrazole-5-carbohydrazide;
N'-(4-(diethylamino)-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;

2-hydroxy-N'-(2-hydroxy-4-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-
methoxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;

-13-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(5-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(2,3-dihydroxybenzylidene)-2-hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-4-morpholinobenzylidene)benzohydrazide;
2-hydroxy-N'-((3-hydroxy-5-nitrobenzofuran-2-
yl)methylene)benzohydrazide;
N'-(2,4-dihydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(5-chloro-2-hydroxy-3-methoxybenzylidene)-2-hydroxybenzohydrazide;
3-chloro-N'-(5-chloro-2-hydroxybenzylidene)-4-methylthiophene-2-
carbohydrazide;

2-amino-N'-(2-amino-5-chlorobenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-3-methoxybenzylidene)benzohydrazide;
2-fluoro-6-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-3-methylbenzylidene)benzohydrazide;
N'-((2-hydroxynaphthalen-l-yl)methylene)-3-methyl-1 H-pyrazole-5-
carbohydrazide;
5-bromo-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-nitrobenzylidene)benzohydrazide;
N-(2-(2-(2-hydroxy-5-
methoxybenzylidene)hydrazinecarbonyl)phenyl)methanesulfonamide;
N'-(3,5-difluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
N'-(1-(5 -chloro-2-hydroxyphenyl)-2,2,2-trifluoroethylidene)-2-
hydroxybenzohydrazide;
2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-nitrobenzohydrazide;
8-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide;

N'-(3-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;
3 -(5 -chlorothiophen-2-yl)-N'-(2-hydroxy-5 -methoxybenzylidene)-1 H-
pyrazole-5-carbohydrazide;
N'-(3-bromo-5-chloro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide;

-14-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(3-bromo-2-hydroxy-5-methoxybenzylidene)-2-hydroxybenzohydrazide;
2-amino-N'-(2-hydroxy-5 -methoxybenzylidene)benzohydrazide;
N'-(5-chloro-2-hydroxybenzylidene)-4-methyl-1,2,3-thiadiazole-5-
carbohydrazide;

N-(2-(2-(2-acetamido-5-
chlorobenzylidene)hydrazinecarbonyl)phenyl)acetamide; and
N-(4-chloro-2-((2-(2-hydroxybenzoyl)hydrazono)methyl)phenyl)-2,4-
difluorobenzenesulfonamide;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.

DETAILED DESCRIPTION OF THE INVENTION
A. Methods of the Invention

Before the methods are described, it is to be understood that the invention is
not limited to the particular methodologies, protocols, cell lines, assays,
and reagents
described, as these may vary. It is also to be understood that the terminology
used herein is
intended to describe particular embodiments of the present invention, and is
in no way
intended to limit the scope of the present invention as set forth in the
appended claims.

It must be noted that as used herein and in the appended claims, the singular
forms "a," "an," and "the" include plural references unless the context
clearly dictates
otherwise.

Unless defined otherwise, all technical and scientific terms used herein have
the same meanings as commonly understood by one of ordinary skill in the art
to which this
invention belongs. Although any methods and materials similar or equivalent to
those
described herein can be used in the practice or testing of the present
invention, the preferred
methods, devices, and materials are now described. All publications cited
herein are
incorporated herein by reference in their entirety. Nothing herein is to be
construed as an
admission that the invention is not entitled to antedate such disclosure by
virtue of prior
invention.

-15-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The definitions used herein are limited to the application of small molecules
as they relate to ADDL aggregation or oligomerization and diseases mediated by
such.

This invention is directed to the discovery that the formation of soluble,
oligomeric, globular, non-fibrillar, neurotoxic A(3i_42 peptides (ADDLs) can
be antagonized
by compounds of formula I, II, or III. Without being limited by any theory, it
is believed
that the administration of a therapeutically effective amount of one or more
of the
compounds described herein will interact with key assembly motifs within the
A(3i_42
monomers or within critical motifs on the A(3i_42 oligomers. This interaction,
in turn, will
prevent the formation of neurotoxic ADDLs or the activity of such ligands. The
disruption
of the ADDLs or the activity of such ligands will protect long term
potentiation of neuronal
cells thereby obviating and/or reversing the neurotoxicity associated with
ADDL. In
addition, this interaction does not interfere with the formation of A(3 senile
plaques.

The term "ADDL" is conventionally defined as amyloid beta-derived
diffusable ligands which have the following characteristics: soluble,
oligomeric, globular,
non-fibrillar, neurotoxic A(3i_42 peptides (GenBank Ref. No. IZOQ_A, accessed
on
November 21, 2007).

The compounds described herein are useful in a method for inhibiting,
regulating and/or modulating assembly of ADDLs either in vitro or in vivo.

The term "soluble" means the ability for a given substance, the solute (an
example in the instant invention is the A(3i_42 oligomer), to dissolve in a
solvent. Within the
context of the instant invention, soluble A(3 oligomers are capable of being
fractionated by
centrifugation.

The term "oligomeric" means a protein complex of a finite number of
monomer subunits. In the context of the invention, oligomers are referred to
as trimers,
low-n-mers, dodecamers (12-mers), and large-n-multimers composed of A(31_42
peptides.
The term "oligomeric" does not include senile amyloid plaques.

The term "globular" means a large soluble protein complex, which is to be
distinguished from fibrils and amyloid plaques. Preferably, the globular
structure ranges in
-16-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
size from 4 nanometers (nm) to about 12 nm, preferably, from about 4.7 to
about 11 nm,
which can be observed upon atomic force microscope analysis (AFM) of
supernatant
fractions of A(3i_42 soluble oligomer preparations as described in US Patent
No. 6,218,506.

The term "non-fibrillar" means the A(3i_42 peptides and oligomeric
complexes that are not aligned in a morphologically distinct pattern known as
amyloid
protofibrils or amyloid fibrils.

As mentioned above, the compounds described herein are useful for
antagonizing ADDL formation in vivo and the diseases associated with ADDL
formation.
As such, the terms "disease," "disorder," and "condition" are used inclusively
and refer to

any condition mediated, at least in part, by ADDLs. In the context of this
invention the
disease may be associated with insoluble amyloid fibrils, senile plaques,
neurofibrillary
tangles, and/or the over-expression of amyloid 01_42 protein. Examples
include, but are not
limited to, Alzheimer's disease, Down's Syndrome, mild cognitive impairment,
stroke,
focal ischemia associated dementia, and neuronal degeneration. Patients
amenable to
treatment include individuals at risk of disease but not exhibiting symptoms,
as well as
patients presently exhibiting symptoms. Therefore, the compounds described
herein can be
administered prophylactically to the general population without the need for
any assessment
of the risk of the patient.

The term "amyloid fibrils" means protein aggregates sharing specific
structural traits. Histopathological techniques generally identify the
structures by apple-
green birefringence when stained with Congo red and seen under polarized
light.

The term "senile plaque" or "senile plaque formation" refers to the
extracellular deposit of amyloid in the gray matter of the brain. The deposits
are associated
with degenerative neural structures. It is understood that senile plaque is
different from and
distinguished over ADDLs.

The term "tangles" means the neurofibrillary tangles formed inside of
degenerating neurons by bundling of paired helical filaments, which assemble
from
hyperphosphorylated forms of the microtubule-associated protein know as tau.

-17-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The term "patient" refers to animals, including mammals, humans, and non-
human mammals. In certain embodiments, a patient is an animal, particularly an
animal
selected from a mammalian species including rat, rabbit, bovine, ovine,
porcine, canine,
feline, murine, equine, and primate, particularly human.

The methods are especially useful for patients who have a known genetic
risk of Alzheimer's disease. Such individuals include those having relatives
who have been
diagnosed with the disease and those whose risk is determined by analysis of
genetic or
biochemical markers. Genetic markers of risk for Alzheimer's disease include
mutations in
the APP gene, particularly mutations at position 717 and positions 670 and 671
referred to
as the Hardy and Swedish mutations respectively. Other markers of risk are
mutations in
the presenilin genes, PS 1 and PS2, and ApoE4, family history of Alzheimer's
Disease,
hypercholesterolemia or atherosclerosis. Individuals presently suffering from
Alzheimer's
disease can be recognized from characteristic dementia, as well as the
presence of risk
factors described above. In addition, a number of diagnostic test are
available for
identifying individuals who have Alzheimer's disease. These include
measurement of CSF
tau as described in Vandermeeren et al. (1993) J. Neurochem. 61:1828-1834;
Arai et al.
(1995) Ann. Neurol. 38:649-652; and Jansen et al. (1995) Neurosci. Lett.
186:189-191 and
A(31_42 levels as described in Andreasen et al. (1999) Arch. Neurol. 56:673-
680;
Vanderstichele et al., "Development of a specific diagnostic test for
measurement of 0-
amyloidl-42 in CSF", Progress in Alzheimer's and Parkinson's diseases, Fisher
et al. (eds),
New York, Plenum, pgs. 773-778; and Hulstaert et al. (1999) Neurology 52:1555-
1562.
Individuals suffering from Alzheimer's disease can also be diagnosed by NINCDS-
ADRDA
(National Institute of Neurological and Communicative Diseases and
Stroke/Alzheimer's
Disease and Related Disorders Association) criteria as described in Hogervorst
et al.
"Diagnosing dementia: Interrater Reliability Assessment and Accuracy of the
NINCDS/ADRDA Criteria versus CERAD Histopathological Criteria for Alzheimer's
Disease" University of Oxford, Oxford Project to Investigate Memory and Aging
(OPTIMA), Oxford, UK; and McKhann et al. (1984) Neurology 34(7):939-944.

In asymptomatic patients, treatment can begin at any age (e.g., 10, 20, 30
years of age). Usually, however, it is not necessary to begin treatment until
a patient
reaches about 40, 50, 60, or 70 years of age. Treatment typically entails
multiple dosages
-18-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
over a period of time. Treatment can be monitored by assaying for the presence
of ADDLs
over time.

As mentioned above, the methods described herein are useful for treating
patients. As used herein, the terms "Treating" or "treatment" of a disease
includes: (1)
preventing the disease, i.e., causing the clinical symptoms of the disease not
to develop in a
patient that may be exposed to or predisposed to the disease but does not yet
experience or
display symptoms of the disease; (2) inhibiting the disease, i.e., arresting
or reducing the
development of the disease or its clinical symptoms; or (3) relieving the
disease, i.e.,
causing regression of the disease or its clinical symptoms.

The term "suffering" as it related to the term "treatment" refers to a patient
or individual who has been diagnosed with or is predisposed to a disease. A
patient may
also be referred to being "at risk of suffering" from a disease. This patient
has not yet
developed characteristic disease pathology, however are known to be
predisposed to the
disease due to family history, being genetically predisposed to developing the
disease, or
diagnosed with a disease or disorder that predisposes them to developing the
disease to be
treated.

In addition to Alzheimer's disease, several other disease are known to be
associated with A(31-42 formation including, but are not limited to, Down's
Syndrome, stroke
and mild cognitive impairment. It is conceivable that similar to Alzheimer's
disease,
treatment of patients suffering from or at risk of suffering from these
diseases is possible
due to the parallel mechanisms of the diseases.

Similarly, over-expression of A(31-42 is associated with focal ischemia
associated dementia and neuronal degeneration. Over-expression of A(3i-42 is
believed to
result in accumulation of ADDLs, thereby inducing neurotoxicity. Treating a
patient
suffering from or at risk of suffering from one of these diseases by
administration of one or
more of the compounds described herein will ameliorate the neurotoxicity of
over-expressed
AP1-42=

The term "neurotoxicity" refers to the toxic effect of ADDLs on neuronal
cells either in vitro and/or in vivo. ADDLs bind to specific neuronal
receptors triggering
-19-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
aberrant neuronal signaling, which compromises long term potentiation and
causes memory
deficits. Thus, ADDLs alter the function of the neuronal cell in such a manner
that, while
still viable, the neuron does not properly function. Such altered
functionality is referred to
herein as "neuronal dysfunction," which is a subclass of neurotoxicity.
Persistent ADDL
signaling causes aberrant transcription and the progressive loss of synapses,
and very long
term persistent ADDL signaling and accumulated structural pathology leads to
eventual
neuron death and gross brain dystrophy.

In therapeutic applications, a pharmaceutical composition containing one or
more compounds described herein is administered to a patient suspected of, or
already
suffering from such a disease associated with the accumulation of ADDLs,
wherein said
compounds are administered in an amount sufficient to treat, or at least
partially treat, the
symptoms of the disease (biochemical, histological and/or behavioral),
including its
complication and intermediate pathological phenotypes in development of the
disease. In
prophylactic applications, a pharmaceutical composition containing one or more
compounds
described herein is administered to a patient susceptible to, or otherwise at
risk of, a disease
associated with the accumulation of ADDLs, wherein said compounds are
administered in
an amount sufficient to eliminate or reduce the risk, lessen the severity, or
delay the outset
of the disease. This includes biochemical, histological and/or behavioral
symptoms of the
disease, its complications and intermediate pathological phenotypes presenting
during
development of the disease.

In some methods, administration of the compound reduces or eliminates mild
cognitive impairment in patients that have not yet developed characteristic
Alzheimer's
pathology. In particular embodiments, a therapeutically effective amount
intends to indicate
the amount of one or more compounds described herein administered or delivered
to the
patient which is most likely to result in the desired response to treatment.

The invention is directed to enhancing cognitive function in a patient who
has diminished function. The term "cognitive function" refers to the
intellectual process by
which one becomes aware of, perceives, or comprehends ideas. Cognitive
function
embraces the quality of knowing, which includes all aspects of perception;
recognition;
conception; sensing; thinking; reasoning; remembering and imagining.
-20-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The term "diminished cognitive function" refers to memory loss, mental
slowing, intellectual decline and/or amnesia. Memory loss may be characterized
as the
difficulty or failure for immediate or delayed recall. Mental slowing is the
difficulty in
processing or completing previously learned tasks in a timely manner or in
processing new
information quickly. Intellectual decline is defined as a loss of information,
or an inability
to utilize information previously possessed or utilized by a person. Amnesia
is an extreme
loss of cognitive ability which results in partial or total inability to
recall past experiences
and impaired or total loss of the ability to speak or write. Diminished
cognitive function
may be caused by a number of disease conditions which are more thoroughly
discussed
below.

Methods of assessing cognitive function include, but are not limited to,
standardized instruments for example Folstein Mini-Mental State Examination;
Modified
Mini-Mental State Exam; Middlesex Elderly Assessment of Mental State; Short
Portable
Mental Status Questionnaire; Alzheimer's Disease Assessment Scale; Clock
Drawing Test;
Clinical Dementia Rating; Neuropsychiatric Inventory or any similarly designed
test. Using
the above listed tests, a skilled clinician would be able to assess the level
of diminished
cognitive function of a patient or enhanced cognitive function following
treatment.
Additionally, informal observations and interactions of individuals to a
patient can also be
used to assess cognitive function and include, but are not limited to, family
members,
friends, formal care givers such as nurses, and individuals who have previous
intimate
knowledge of the patient.

Mechanical measure of the neurons and neuronal tissue may also be used to
assess cognitive function including, but not limited to, Computed Tomography
(CT);
Computed Axial Tomography (CAT); Magnetic Resonance Imaging (MRI); Functional

Magnetic Resonance Imaging (fMRI); Positron Emission Tomography (PET): Single
Photon Emission Computed Tomography (SPECT); Diffuse Optical Imaging (DOI);
Diffuse Optical Tomography (DOT) or any similarly designed instrumentation.

An "effective amount" is an amount of one or more of the compounds
described herein which treats the ADDL-mediated disease. In one embodiment,
the
-21 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
compounds of this invention will decrease ADDL formation either in vitro or in
vivo by at
least 10%, 25%, 40%, 60%, 80%, 90% or 95% as compared to control.

The "therapeutically effective amount" will vary depending on the
compound, the disease and its severity and the age, weight, etc., of the
patient to be treated
all of which is within the skill of the attending clinician to assess. It is
contemplated that a
therapeutically effective amount of one or more of the compounds described
herein will
alter ADDL formation (including inhibiting or reversing formation of ADDLs) in
the
patient as compared to binding of ADDLs in the absence of treatment. As such,
impairment
of long term potentiation and subsequent memory formation is decreased. A
therapeutically
effective amount is distinguishable from an amount having a biological effect
(a
"biologically effective amount"). A compound of the present invention may have
one or
more biological effects in vitro or even in vivo, such as reduction in ADDL
formation to
some extent. A biological effect, however, may not result in any clinically
measurable
therapeutically effect as described above as determined by methods within the
skill of the
attending clinician.

The present invention is also directed ADDL inhibition in a neuronal cell
and/or neuronal tissue. A "neuronal cell" or "neuron" is a cell that transmits
and processes
signals in the brain or other parts of the nervous system. Additionally, a
neuronal cell, as
used in the invention, can be isolated from animal brain tissue and grown in
tissue culture.
The isolated cells can be comprised of an established neuronal cell line
selected from for
example, but are not limited to, MC65; HCN-2; SH-SY5Y; SK-N-AS; SK-N-FI; SK-N-
DZ;
H19-7/IGF-IR; QNR/D; QNR/K2; C8-D30; C8-S; C8-DIA; OLGA-PH-J/92; Daoy;
RSC96; SW10; RT4-D6P2T; RN33B; PC-12; DBRTG-05MG; C8-B4; SK-N-SH; B35;
R3[33-lOras3]; Neuro-2A; and HCN-lA or any genetic, chemical, and/or
biochemical
modified variants thereof. (Commercially available from American Type Culture
Collection (ATCC)). The isolated cells can also be comprised of primary cells
and/or
astrocytes isolated from neuronal tissues selected from, for example, but are
not limited to,
the hippocampus; cerebellum; cortex; hypothalamus; mid-brain; spinal cord;
striatum;
frontal lobe; temporal lobe; parietal lobe; occipital lobe and any genetic,
chemical, and/or
biochemical modified variants thereof. The isolated, cultured animal cell can
be comprised
of a neural stem cell or any differentiated, genetic, chemical, and/or
biochemical modified
-22-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
variants thereof. Additionally, a neuronal cell or neuron can be isolated and
distinguished
from other cell types by detecting expression of neuronal markers selected
from, but not
limited to, CD133, GFAP, MAP-2, MPB, Nestin, Neural tubulin, Neurofilament,
Neurosphere, Noggin, 04, 01, Synaptophysin, and Tau
(http://stemcells.nih.gov/info/scireport/appendixE.asp, accessed on November
26, 2007).
As used herein, the term "neuronal tissue" refers to any portion of the
central
nervous system including, but not limited to, the brain or spinal cord.
Neuronal tissue can
be composed of, at least in part, neuronal cells.

B. Compounds

The compounds useful in the methods of the invention contain one or more
and any combination of the following characteristics: (1) low or sub-
micromolar potency
when tested in the FRET assay described herein; (2) non-aggregating; (3)
little or no
neuronal toxicity when administered to a patient; (4) favorable solubility in
an aqueous
environment; (5) chemically tractable; (6) dose dependent characteristics; (7)
reversibly
bind to the A(3 protein; (8) capable of amyloid 0 monomer binding; (9) capable
of binding
soluble amyloid 0 oligomers.

In one embodiment of the invention, the compounds useful for treating
patients are suitable for oral delivery. In this embodiment, the compounds are
compliant
with Lipinski's rule-of-five which provides a criteria to evaluate drug
likeness. The rule
states that, in general, an orally active drug has: no more than 5 hydrogen
bond donors (OH
and NH groups); no more than 10 hydrogen bond acceptors (notably N and 0); a
molecular
weight under 500 g/mol; and a partition coefficient log P less than 5.

1. Exemplary Compounds

In one embodiment, methods of the invention employ compounds of the
formula:

- 23 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
R2 X2

'4 Y NH
( R) Rs
n
Xl
m
wherein:
A is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
Xi and X2 are independently selected from the group consisting of oxygen,
sulfur or
N-OR4;
Ri is selected from the group consisting of hydroxy, halo, nitro, C1_6 alkyl,
C1_6
haloalkyl, C1_6 alkoxy, -N(Rs)(R6), C3_1o cycloalkyl, aryl, heteroaryl,
heterocyclic, wherein
the aryl, heteroaryl, and heterocyclic group is optionally substituted with 1-
3 R9 groups;
R2 is selected from the group consisting of hydrogen, Ci_6 alkyl, and Ci_6
haloalkyl;
R3 is selected from the group consisting of C1_6 alkyl, C1_6 alkoxy, -
N(Rs)(R), and
Rg ;
R4 is selected from the group consisting of hydrogen and Ci_6 alkyl;
each R 5 is independently selected from the group consisting of hydrogen, Ci_6
alkyl,
and -S02(R7);
each R6 is independently selected from the group consisting of hydrogen and
C1_6
alkyl;
R' is selected from the group consisting of hydrogen, Ci_6 alkyl, -C(=O)-Ci_6
alkyl,
aryl optionally substituted with 1 to 3 of C14 alkyl or halo;
R8 is selected from the group consisting of aryl, biaryl, heteroaryl, and
heterocyclic,
wherein each R8 is optionally substituted with 1-4 R9 groups;
each R9 is independently selected from the group consisting of hydroxy, halo,
nitro,
C1_6 alkyl, C2_6 alkenyl, C1_6 alkoxy, C1_6 haloalkoxy, C3_10 cycloalkyl,
halo, aminoacyl,
acylamino, aralkyl, -N(R5)(R6), carboxyl, carboxyl ester, and heterocyclic;
n is 0, 1, 2, or 3; and
mis0orl;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.
In another embodiment of the invention, the methods employ compounds of
the formula:
-24-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
R22 O

NH ~
R21 A2 ~N R23
n
O
It
wherein:
A2 is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R2i is selected from the group consisting of hydroxy, halo, nitro, Ci_6 alkyl,
Ci_6
haloalkyl, -N-S(O)2-R24, and aryl;
R22 is selected from the group consisting of hydrogen, C1_6 alkyl, and C1_6
haloalkyl;
R23 is selected from the group consisting of C1_6 alkyl, amino, and R 25;
R24 is selected from the group consisting of Ci_6 alkyl, and aryl optionally
substituted
with halo or Ci_6 alkyl,
R 25 is selected from the group consisting of aryl, heteroaryl, and
heterocyclic, all of
which may be optionally substituted with 1-3 R26 groups;
each R26 is independently selected from the group consisting of hydroxy, halo,
C1_6
alkyl, aralkyl, and aryl;
n is 0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.
In yet another embodiment, methods of the invention employ a compound of
the formula:

R33
NH
1 nA~ 32
r R31 ~ N R
O
III.
wherein:
Ai is a 5-10 membered heteroaryl ring having 1 to 3 heteroatoms or an aryl
ring;
R31 is selected from the group consisting of hydroxy, halo, nitro, C1_6 alkyl,
C1_6
alkoxy, C3_1o cycloalkyl, aryl, heteroaryl optionally substituted with 1-3
Ci_6 alkyl, amino,
-N-SO2-R34, and -N-C(=O)-R34;
R32 is selected from the group consisting of hydrogen, C1_6 alkyl, and C1_6
haloalkyl;
- 25 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
R33 is selected from the group consisting of aryl, biaryl, heteroaryl, and
heterocyclic,
wherein each R33 is optionally substituted with 1-4 R35 groups;
R34 is selected from the group consisting of Ci_6 alkyl and aryl optionally
substituted
with halo;
R35 is selected from the group consisting of hydroxy, nitro, halo, C1_6 alkyl,
C2_6
alkenyl, C1_6 alkoxy, C1_6 haloalkoxy, C3_1o cycloalkyl, halo, amino,
alkylamino,
dialkylamino, aminoacyl, aryl-alkylene, carboxyl, carboxyl ester, and
heterocyclic; and
nis0, 1, 2, or 3;
or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug
thereof.
In one embodiment, the group A is selected from the group consisting of
phenyl, biphenyl, naphthyl, benzothiophenyl, thiadiazolyl, indanyl,
thiophenyl, indolyl,
pyrazolyl, furanyl, oxoindolinyl, pyridyl, and benzodioxoyl.

In some embodiments Ri is selected from the group hydroxy, chloro, fluoro,
bromo, iodo, methyl, methoxy, trifluoromethyl, cyclopropyl, phenyl, pyrrolyl,
methylsulfonamido, 4-chlorophenylsulfonylamido, nitro, benzo[d][1,3]dioxolyl,
amino,
thienyl, 5-chlorothienyl, and methylcarbonylamino.

In another embodiment, the group A is optionally substituted and is selected
from the group consisting of 2-(methylsulfonamido)phenyl, 1 H-indan-7-yl, 1 H-
indol-7-yl,
1-hydroxy-naphthalen-2-yl, 1-phenyl-5-(trifluoromethyl)-1 H-pyrazol-4-yl, 2-(4-

chlorophenyl sulfonamido) phenyl, 2,4-dihydroxyphenyl, 2,6-difluorophenyl, 2-
acetamidophenyl, 2-aminophenyl, benzothiophen-2-yl, 2-fluoro-6-hydroxyphenyl,
2-
hydroxy-3-methylphenyl, 2-hydroxy-4-(1H-pyrrol-1-yl)phenyl, 2-hydroxy-4-
methoxyphenyl, 2-hydroxy-5-methoxyphenyl, 2-hydroxy-5-methylphenyl, 2-hydroxy-
5-
nitrophenyl, 2-hydroxy-naphthalen-1-yl, 2-hydroxyphenyl, 2-methylfuran-3-yl, 2-

oxoindolin-7-yl, 3-(5-chlorothiophen-2-yl)-1H-pyrazol-5-yl, 3-
(benzo[d][1,3]dioxol-5-yl-
1H-pyrazol-5-yl, 3-aminophenyl, 3-chloro-lH-indol-2-yl, 3-chloro-4-methyl-
thiophen-2-yl,
3-chloro-6-fluorobenzo thiophene, 3-chlorobenzothiophen-2-yl, 3-cyclopropyl-lH-
pyrazol-
5-yl, 3-fluorophenyl, 3-hydroxy-naphthalen-2-yl, 3-methyl-lH-pyrazol-5-yl, 4-
(2,5-
dimethyl-lH-pyrrol-1-yl)phenyl, 4-bromophenyl, 4-chloro-2-hydroxyphenyl, 4-
iodo-l-
methyl-lH-pyrazol-3-yl, 4-iodophenyl, 4-methyl-1,2,3-thiadiazol-5-yl, 4-
methylphenyl, 4-
-26-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
nitrophenyl, 5-bromo-2-hydroxyphenyl, 6-methyl-pyrid-3-yl, 8-hydroxynaphthalen-
l-yl,
and benzo[d][1,3]dioxol-5-yl.

In some embodiments, Xi and X2 are oxygen.

In some embodiments, R2 is selected from hydrogen, methyl, or
trifluoromethyl.

In some embodiments, m is 0. In other embodiments m is 1.

In some embodiments, R3 is selected from the group consisting of butyl, t-
butyl, and amino.

In some embodiments, R3 is optionally substituted and is selected from the
group consisting of phenyl, thienyl, naphthyl, furanyl, piperazinyl,
benzothiophenyl,
pyrazolyl, morpholino, and piperidinyl.

In some embodiments, R3 when a ring, is substituted with one or more
groups consisting of hydroxy, chloro, fluoro, bromo, iodo, methyl, t-butyl,
methoxy, ethoxy,
benzyl, phenyl, cyclohexyl, trifluoromethoxy, allyl, aminocarbonyl, amino,
ethoxycarbonyl,
diethylamino, morpholino, nitro, 2,4-difluorophenylsulfonylamino, and
methylcarbonylamino.

In other embodiments, R3 is selected from the group consisting of 5-chloro-
2-(2,4-difluorophenylsulfonamido)phenyl, 1-hydroxy-napthalen-2-yl, 1-methyl-lH-
pyrazol-
5-yl, 2,3-dihydroxyphenyl, 2,4-dihydroxyphenyl, 2,6-dihydroxyphenyl, 2-
acetamido-5-
chlorophenyl, 2-amino-5-chlorophenyl, benzothiophen-2-yl, 2-bromo-6-
hydroxyphenyl, 2-
furanyl, 2-hydroxy naphthalen-l-yl, 4-hydroxy-3'-methoxybiphen-3-yl, 2-hydroxy-
3 -
methoxyphenyl, 2-hydroxy-3-methylphenyl, 2-hydroxy-4-methoxyphenyl, 2-hydroxy-
4-
methylphenyl, 2-hydroxy-4-morpholinophenyl, 2-hydroxy-5-methoxyphenyl, 2-
hydroxy-5-
methylphenyl, 2-hydroxy-5-nitrophenyl, 2-hydroxy-5-trifluoromethoxyphenyl, 2-
hydroxy-
6-methoxy-phenyl, 2-hydroxynaphthalen-1-yl, 2-hydroxyphenyl, 3,5-dibromo-2-
hydroxyphenyl, 3,5-dichloro-2-hydroxy-phenyl, 3,5-difluoro-2-hydroxyphenyl, 3-
allyl-2-
hydroxyphenyl, 3-bromo-2-hydroxy-5-methoxyphenyl, 3-bromo-5-chloro-2-
hydroxyphenyl,
3-chloro-5-cyclohexyl-2-hydroxyphenyl, 3-chloro-5-fluoro-2-hydroxyphenyl, 3-
ethoxy-2-
-27-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
hydroxyphenyl, 3-fluoro-2-hydroxyphenyl, 3-hydroxy-5-nitrobenzofuran-2-yl, 4-
benzyl
piperazin-l-yl, 4-diethylamino-2-hydroxyphenyl, 4-methyl piperazin-l-yl, 4-
methylphenyl,
4-morpholino, 4-phenylpiperidin-1-yl, 5-bromo-2-hydroxy-3-iodophenyl, 5-bromo-
2-
hydroxyphenyl, 5-chlorothiophen-2-yl, 2-amino-5-chloro-phenyl, 5-chloro-2-
hydroxy-3-
methoxyphenyl, 5-chloro-2-hydroxyphenyl, 5-chlorothiophen-2-yl, 5-ethoxy-2-
hydroxyphenyl, 5-methyl thiophen-2-yl, 5-tert-butyl-2-hydroxyphenyl, 6-bromo-5-
hydroxy-
2-(ethoxycarbonyl)benzofuran-4-yl, benzamid-2-yl, -NH2, tert-butyl, and thien-
2-yl.

Certain examples of compounds that may be useful in this invention are
presented below in Table lA and lB. It is to be understood that the
illustration of these
compounds is in no way limiting the invention to the compounds described in
these tables
and it is therefore contemplated that other compounds are suitable for use in
this invention.
It should also be noted that the following compounds may exhibit
stereoisomerism (i.e., E
and Z isomers) and the invention contemplates use of either isomer and
mixtures thereof.
Table lA

R22 O
AZ
NH
(R21 N R2s
n
0
No. Structure Compound Name
O /L 1
2-hydroxy-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-
OH ~ FF oxobutan-2-ylidene) benzohydrazide
N-N F

O
OH
H
N, 2-hydroxy-N'-(l, 1,1-trifluoro-4-oxo-4-(thiophen-
2 O N s 2-yl)butan-2-ylidene) benzohydrazide
F

- 28 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
N.N 0
I
3 ~ N'~ N'-(4-(4-benzylpiperazin-l-yl)-l,1,1 -trifluoro-4-
F F oxobutan-2-ylidene)-2-hydroxy-benzohydrazide
CI
H
4 s N, N 3-chloro-6-fluoro-N'-(4-(furan-2-yl)-4-oxobutan-
F O 1 O 2-ylidene)benzo[b]-thiophene-2-carbohydrazide
H3C
YH3
0-S-0
C NN-(2-(2-(l,l,l-trifluoro-4-oxo-4-
N , (thiophen-2-yl)butan-2-ylidene)hydrazine-
N carbonyl)phenyl) methanesulfonamide
O g
FF F
(::(;N H
2-hydroxy-N'-(l, 1, l -trifluoro-4-(4-
6 O methylpiperazin-l-yl)-4-oxobutan-2-ylidene)
F N ~ benzohydrazide
F F
N , CH 3
H
7 OHN`N O 2-hydroxy-N'-(l,1,1 -trifluoro-5,5-dimethyl-4-
0 I CH3 oxohexan-2-ylidene) benzohydrazide
F CH3
F F CH3
OH
H
g N, N N'-(4-(benzo[b]thiophen-2-yl)-1,1,1 -trifluoro-4-
0 I S oxobutan-2-ylidene)-2-hydroxy benzohydrazide
F 1 ~ \
F F

-29-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H 2-hydroxy-N'-(1,1,1-trifluoro-4-(1-methyl-lH-
N,
9 N 0 CH3 pyrazol-5-yl)-4-oxobutan-2-ylidene)
O I N benzohydrazide
F N
F F
0
S CHs
_ s 3-chloro-6-fluoro-N'-(l,1,1 -trifluoro-4-(5-
F~ c~ N F methylthiophen-2-yl)-4-oxobutan-2-
NH F F ylidene)benzo[b]thiophene-2-carbohydrazide
0
F F
F
CH3 4-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-
11 HN-N O yl)butan-2-ylidene)-1,2,3-thiadiazole-5-
N carbohydrazide
N-S S

CI
N3N O 3-chloro-6-fluoro-N'-(l,1,1 -trifluoro-5,5-
12 ~/ 5 dimethyl-4-oxohexan-2-
F O CH3 ylidene)benzo[b]thiophene-2-carbohydrazide
F F F H3C CH3

F F F

OH H O 2-hydroxy-N'-(1,1,1-trifluoro-4-(5-
13 N-N methylthiophen-2-yl)-4-oxobutan-2-ylidene)
O S benzohydrazide
CH3
CI
N_N O 3-chloro-N'-(l,1,1 -trifluoro-4-oxo-4-(thiophen-2-
14 y
1)butan-2-ylidene)benzo[b]thiophene-2-
OF 1 5 carbohydrazide
/

-30-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

/ /
O
O 3-chloro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-
15 CI F oxobutan-2-ylidene)benzo[b] thiophene-2-
/ F carbohydrazide
NH-N F
S
0
O CI

4-chloro-N-(2-(2-(l,l,l-trifluoro-4-oxo-4-
16 ,:::~; N , (thiophen-2-yl)butan-2-ylidene)
~ O hydrazinecarbonyl) phenyl) benzenesulfonamide
O S
F
F F
CI

~\ N. N O N'-(4-(benzo[b]thiohhen-2-Y1)-1>1>1-trifluoro-4-
~ /
17 F s o 1 5 oxobutan-2-ylidene)-3-chloro-6-fluorobenzo[b]
F thiophene-2-carbohydrazide
F F

OH
H
N'N 2-hydroxy-N'-(l,1,1 -trifluoro-4-morpholino-4-
18
0 oxobutan-2-ylidene) benzohydrazide
F N
F F O
F F
F
OH
19 HN-N O N'-(4-(5-chlorothiophen-2-yl)-l,l,l-trifluoro-4-
\ oxobutan-2-ylidene)-2-hydroxy benzohydrazide
O S
zz_
CI
H 5-chloro-2-hydroxy-N'-(1,1,1-trifluoro-4-oxo-4-
20 N , N O (thiophen-2-yl)butan-2-ylidene) benzohydrazide
OH O g

FF F -31-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

F F
CI F /I 3-chloro-4-methyl-N'-(l,1,1-trifluoro-4-oxo-4-
21 H3C HN-N O (thiophen-2-yl)butan-2-ylidene)thiophene-2-
~ carbohydrazide
S 0 S

F F

N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-
8%H F
22 N-N O 2-ylidene)-1 H-indole-7-carbohydrazide
O g

0
S
F CI ci 3-chloro-N'-(4-(5-chlorothiophen-2-yl)-1,1,1 -
23 N J, F trifluoro-4-oxobutan-2-ylidene)-6-
S H F F fluorobenzo[b]thiophene-2-carbohydrazide
~ N

0
CI

~ 3-chloro-6-fluoro-N'-(1,1,1-trifluoro-4-oxo-4-
24 F S HON-N (thiophen-2-yl)butan-2-
F ylidene)benzo[b]thiophene-2-carbohydrazide
F F O S
H3
OH
~ H 2-hydroxy-3-methyl-N'-(1,1,1-trifluoro-4-oxo-4-
25 N .
N 0 (thiophen-2-yl)butan-2-ylidene) benzohydrazide
O g

FF F I
O
+ F F
26 O%N F O 4-nitro-N'-(l, l, 1 -trifluoro-4-oxo-4-(thiophen-2-
H N g yl)butan-2-ylidene) benzohydrazide
/
/ `
0
27 N-NI S 4-bromo-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-
~ H O yl)butan-2-ylidene) benzohydrazide
Br F F

-32-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

N
28 g N N'-(l,l,l-trifluoro-4-oxo-4-(thiophen-2-yl)butan-
O g 2-ylidene)benzo[b]thiophene-2-carbohydrazide
F
F F
~ OH CH3 0
H
29 ~ N'N~N 2-hydroxy-N'-(4-oxo-4-(4-phenylpiperidin-l-
0 yl)butan-2-ylidene) benzohydrazide

CI

N , 3-chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-
CCN H
30 N yl)butan-2-ylidene)-1H-indole-2-carbohydrazide
H O S
F
F F

O
O 3-chloro-6-fluoro-N'-(l,1,1 -trifluoro-4-(furan-2-
31 CI F yl)-4-oxobutan-2-ylidene)benzo[b]thiophene-2-
F carbohydrazide
HN-N F

F S O
OH O
32 e N=N" NH2 3-(2-(2-hydroxybenzoyl)hydrazono)butanamide
H CH3 0
O / \
33 ~ N"N~ 5 4-methyl-N'-(l,l,l-trifluoro-4-oxo-4-(thiophen-2-
~ ~ H o yl)butan-2-ylidene) benzohydrazide
H3C F F F
N- H
v N ~ N'N 0 1-phenyl-N'-(l,1,1 -trifluoro-4-oxo-4-(thiophen-2-
34 0 S yl)butan-2-ylidene)-5-(trifluoromethyl)-1H-
F F F F pyrazole-4-carbohydrazide
F F
F
H
35 N, N O 2,6-difluoro-N'-(l,1,1 -trifluoro-4-oxo-4-
F O (thiophen-2-yl)butan-2-ylidene) benzohydrazide
F
F

- 33 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Table 1B

R13

NH ~R12
R11 A1 ~N \ rn

O
No. Structure Compound Name
\ N OH
N, 2-hydroxy-N'-(2-hydroxy-5-methoxy-
36 N oH benzylidene)-4-(1H-pyrrol-l-yl)
0 H benzohydrazide

O, CH3
OH
H
N, N OH
37 O H 3-hydroxy-N'-(2-hydroxy-5-methoxy-
benzylidene)-2-naphtho hydrazide
Ol CH3

OH
H
N'N OH 1-hydroxy-N'-(2-hydroxy-5-
38 O ~ methoxybenzylidene)-2-naphtho-
H hydrazide
O, CH3

N,
JCIY H
F N H ~ N'-(5-bromo-2-hydroxy-3-
39 H iodobenzylidene)-3-fluoro-
benzohydrazide
Br

-34-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
N
N H
0 ~ CI N'-(3-chloro-5-fluoro-2-
40 H hydroxybenzylidene)-2-hydroxy-
benzohydrazide
F

c OH
H
N,
N H N'-(2,6-dihydroxybenzylidene)-2-
41 0 H hydroxy benzohydrazide

HO
H
O
a
iLyj N H
42 0 HI Br N'-(3,5-dibromo-2-hydroxybenzylidene)-
2-hydroxybenzohydrazide
Br

ci
o~ ~ I
O=s
NH 4-chloro-N-(2-(2-((2-hydroxynaphthalen-
H 1-
43 N, N yl)methylene)hydrazinecarbonyl)phenyl)
o H benzenesulfonamide

HO
Qi~NH H
N,
ci 0 IN OH 3-chloro-N'-(2-hydroxy-5-
44 H methoxybenzylidene)-1H-indole-2-
H-indole-2-
carbohydrazide
O~CH3

- 35 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name
(::~;N H
_ N O H
45 N'-(2-bromo-6-hydroxybenzylidene)-2-
H O hydroxybenzo-hydrazide

Br
OH
H
/ N,
N OH
0 ci N'-(3-chloro-5-cyclohexyl-2-
46 H hydroxybenzylidene)-2-hydroxybenzo-
hydrazide
OH
H
N~
Uly N~
N OH
47 O H I N'-(5-tert-butyl-2-hydroxybenzylidene)-
~ 2-hydroxy-benzohydrazide
H3C CH Hs
3
OH
H
N, N OH
0 2-hydroxy-N'-((4-hydroxy-3'-
48 H methoxybiphenyl-3-yl)methylene)
benzohydrazide
,CH3

O i H
N H N'-(3,5-dibromo-2-hydroxy-
49 H3C 0 H Br benzylidene)-2-methylfuran-3-
carbohydrazide
r

-36-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
I H
/ N_ N H N'-(5-bromo-2-hydroxy-benzylidene)-2-
50 O H hydroxybenzohydrazide

r
CI \ OH
I H
/ N,
N H
51 O 4-chloro-2-hydroxy-N'-(2-hydroxy-5-
H methoxy-benzylidene) benzohydrazide
CH3
O
NH
H
52 N, N OH N'-(2-hydroxy-5-methoxy-benzylidene)-
0 2-oxoindoline-7-carbohydrazide
H I

O~CH3
H3C N~
H
53 N, N OH N'-(2-hydroxy-3-methyl-benzylidene)-6-
0 H CH3 methylnicotino-hydrazide
\
I /
~
H3C-N~ - N.
N N OH N'-((2-hydroxy-naphthalen-l-
54 0 H I yl)methylene)-4-iodo-l-methyl-lH-
~ pyrazole-3-carbohydrazide
\ OH
H
I
55 N_ N H 2-hydroxy-N'-(2-hydroxy-6-methoxy-
0 benzylidene) benzohydrazide
H
H3C,

-37-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

CHg
N H
H3C N, 4-(2,5-dimethyl-lH-pyrrol-1-yl)-N'-(2-
56 N OH hydroxy-5-methoxy-benzylidene)
0
H ~ ~ benzohydrazide
~
O,
CHg
~ OH
H
I~ N_ N OH
57 O 2-hydroxy-N'-(2-hydroxy-5-(trifluoro-
H methoxy) benzylidene) benzohydrazide
O,CF3

NH
H

N H N'-(2-hydroxy-5-methoxy-benzylidene)-
58 O H 1H-indole-7-carbohydrazide
O'CH3
OH
H
I ~ N
N H
59 N'-(5-chloro-2-hydroxy-benzylidene)-2-
0 H hydroxy-benzohydrazide

CI
H
CH2 N'-(3-allyl-2-hydroxy-benzylidene)-4-
60 O iodo-benzohydrazide
~ NW14-1

H -38-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

F CI
H
S N, N H 3-chloro-6-fluoro-N'-((2-
61 O I hydroxynaphthalen-l-yl)methylene)
H benzo[b]thiophene-2-carbohydrazide
I /
CI
O~
0=S
N H 4-chloro-N-(2-(2-(2,4-dihydroxy-
62 H benzylidene) hydrazine-
aN'N carbonyl)phenyl)benzene-sulfonamide
OH ~QH
HO H3

OH
H
N,
63 N H 2-hydroxy-N'-(2-hydroxy-5-methoxy-
H benzylidene)-3-methyl-benzohydrazide
"CH3
OH
H
N, N O N H 2 2-((2-(2-hydroxybenzoyl)hydrazono)
64 ~ methyl) benzamide
OH \
OH
H
N N NH2 ,
65 O ~ N -(2-amino-5-chloro-benzylidene)-2-
H hydroxy-benzohydrazide
CI

-39-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
c_LIINN OH
66 0 H 2-hydroxy-N'-(2-hydroxy-5-methoxy-
I benzylidene)-1-naphtho-hydrazide
O- CH3

F OH
H
\ N=N H
67 0 4-fluoro-2-hydroxy-N'-(2-hydroxy-5-
H methoxy-benzylidene) benzohydrazide
O,
CH3
OH
H
N
68 O H 2-hydroxy-N'-(2-hydroxy-5-methyl-
H benzylidene) benzohydrazide
CH3
OH
H
N,
N H
69 0 CI N'-(3,5-dichloro-2-hydroxy-
H benzylidene)-2-hydroxy-benzohydrazide
CI
H3C CI
H
S N,
N OH 3-chloro-N'-(2-hydroxy-5-methoxy-
70 0 H benzylidene)-4-methylthiophene-2-
carbohydrazide
O,
CH3

-40-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

ci
o~ I
o=~
H 4-chloro-N-(2-(2-(2-hydroxy-5-methoxy-
C(; H
71 N, N OH benzylidene) hydrazine-
o H k carbonyl)phenyl)benzene-sulfonamide
O'CH
OH
H
72 N, N OH 2-hydroxy-N'-((1-hydroxy-naphthalen-2-
0 yl)methylene) benzohydrazide

/ OH
I H
H3C \ N- N OH
73 0 H 2-hydroxy-N'-(2-hydroxy-5-methoxy-
benzylidene)-5-methyl-benzohydrazide
0
CH
OH
I H
74 \ N, N H N'-(3-fluoro-2-hydroxy-benzylidene)-2-
0 F hydroxy-benzohydrazide
H

/ OH
I H
F \ N, N OH
75 0 5-fluoro-2-hydroxy-N'-(2-hydroxy-5-
H methoxy-benzylidene) benzohydrazide
O,
CH3

-41 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

CH3
O OH
H
\ N,
N H 2-h drox N'- 2-h drox 5-methox
76 I Y Y-( Y Y- y-
O H benzylidene)-4-methoxy-benzohydrazide
O-1 CH3

a;H
0 NN OH Br ethyl6-bromo-5-hydroxy-4-((2-(2-
77 H hydroxybenzoyl)hydrazono) methyl)-2-
methyl-benzofuran-3-carboxylate
/\O \ 0
H3C
H3C
O
N-NH H

N~N OH
o 3-(benzo[d][1,3]dioxol-5-yl)-N'-(2-
78 H hydroxy-5-methoxy benzylidene)-1H-
pyrazole-5-carbohydrazide
O-
CH
H
N,
cc
79 N 2-hydroxy-N'-(1-p-tolylethylidene)
O ~ benzohydrazide
3C I

~ CH3
a;H
N
N OH
80 0 H N'-(4-(diethylamino)-2-hydroxy-
N CH3 benzylidene)-2-hydroxy-benzohydrazide
CH3

-42-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
N H
g 1 I 2-hydroxy-N'-(2-hydroxy-4-methoxy-
0 H ~ \ benzylidene) benzohydrazide

/ O CH3
OH
H
O N OH
2-hydroxy-N'-(2-hydroxy-5-methoxy-
82 CH3 0 H I O
benzylidene)-5-methoxy-benzohydrazide
O,
CH3
OH
H
NN H
83 0 H ~ 2-hydroxy-N'-(2-hydroxy-5-methoxy-
benzylidene) benzohydrazide

0
CH3
OH
H
CI N, N H
84 0 5-chloro-2-hydroxy-N'-(2-hydroxy-5-
H methoxy-benzylidene) benzohydrazide
Ol CH3

a;H
N- N OH
N'-(5-ethoxy-2-hydroxy-benzylidene)-2-
85 O H hydroxy-benzohydrazide
O,_~,CH3

- 43 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
86 N, N H N'-(2,3-dihydroxy-benzylidene)-2-
0 OH hydroxy-benzohydrazide
H
/
H
OI(NN OH
87 ~ OH 2-hydroxy-N'-(2-hydroxy-4-morpholino-
H benzylidene)-benzohydrazide
Oo

a;H
N.N
H 2-hydroxy-N'-((3-hydroxy-5-nitro-
88 O H I benzofuran-2-yl)methylene)
O benzohydrazide
NO2

OH
I H
N_ N OH
2-hydroxy-N'-((2-hydroxy-naphthalen-l-
89 O H yl)methylene) benzohydrazide

OH
H
90 N, N H N'-(2,4-dihydroxybenzylidene)-2-
0 hydroxy-benzohydrazide
a H I
OH
H
N,
91 O N H~H3 N'-(5-chloro-2-hydroxy-3-methoxy-
91 benzylidene)-2-hydroxy-benzohydrazide
CI

-44-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

H3C CI
H
N,N OH 3-chloro-N'-(5-chloro-2-
92 0 I hydroxybenzylidene)-4-methylthiophene-
H 2-carbohydrazide
CI
NH2
H
N,
N NH2 2-amino-N'-(2-amino-5-
93 O H chlorobenzylidene)benzohydrazide
CI
OH
H
94 N, N H YH3 2-hydroxy-N'-(2-hydroxy-3-
0 O methoxybenzylidene)benzohydrazide
H

OH
H
N=N H
95 F 0 I 2-fluoro-6-hydroxy-N'-(2-hydroxy-5-
H methoxybenzylidene)benzohydrazide
O,
CH3
OH
H
96 N- N OH 2-hydroxy-N'-(2-hydroxy-3-
O ~ H CH3 methylbenzylidene)benzohydrazide
~
H3C

N/ H
,
N N, N OH N'-((2-hydroxynaphthalen-l-
97 H 0 I yl)methylene)-3-methyl-lH-pyrazole-5-
H carbohydrazide
- 45 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

OH
H
Br N, N OH
gg 0 ~ 5-bromo-2-hydroxy-N'-(2-hydroxy-5-
H methoxybenzylidene)benzohydrazide
O,CH3

OH
H
N~N OH
I 2-hydroxy-N'-(2-hydroxy-5-
99 O H nitrobenzylidene)benzohydrazide
NO
~~ ~CH3
0=S
NH
N, N-(2-(2-(2-hydroxy-5-
100 N OH methoxybenzylidene)hydrazinecarbonyl)
O H ~ \ phenyl)methanesulfonamide

O,
CH3
/ OH
H
N,
\,
N H
101 ~ 0 F N'-(3,5-difluoro-2-hydroxybenzylidene)-
H 2-hydroxybenzohydrazide
/ OH

N~
\~
N H N'-(1-(5-chloro-2-hydroxyphenyl)-2,2,2-
102 O 1 trifluoroethylidene)-2-
F F hydroxybenzohydrazide
CI

-46-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

%N,l H
N , 3-cyclopropyl-N'-((2-
103 N OH hydroxynaphthalen-l-yl)methylene)-1H-
O H pyrazole-5-carbohydrazide

OH
H
O2N N I N H 2-hydroxy-N'-(2-hydroxy-5-
104 O H methoxybenzylidene)-5-
nitrobenzohydrazide

O,CH3
OH
H
N N O H 8-hydroxy-N'-(2-hydroxy-5-
105 0 1 methoxybenzylidene)-1-
H I naphthohydrazide
O'CH3

a;H
- N OH
106 0 H O~CH3 N'-(3-ethoxy-2-hydroxybenzylidene)-2-
hydroxybenzohydrazide
-47-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

CI

S

N, N, 3-(5-chlorothiophen-2-yl)-N'-(2-
107 H N H hydroxy-5-methoxybenzylidene)-1H-
0 H pyrazole-5-carbohydrazide

O-1 CH
OH
H
N
N H N'-(3-bromo-5-chloro-2-
108 O I Br hydroxybenzylidene)-2-
H hydroxybenzohydrazide
OHN
a H
N H N'-(3-bromo-2-hydroxy-5-
109 0 H V B r methoxybenzylidene)-2-
/ hydroxybenzohydrazide
O,CH3

/ NH2
H
NN OH
110 0 I 2-amino-N'-(2-hydroxy-5-
H methoxybenzylidene)benzohydrazide
O,
CH3
N CH3
, H
N'
S N, N H N'-(5-chloro-2-hydroxybenzylidene)-4-
111 O methyl-1,2,3-thiadiazole-5-
H carbohydrazide

- 48 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. Structure Compound Name

H 3C

'I H
112 N, N N`N H N'-(1-(2-hydroxyphenyl)ethylidene)-3-
H methyl-lH-pyrazole-5-carbohydrazide
~3C

OYCH3
/ NH O
~ H N-(2-(2-(2-acetamido-5-
113 N, N HN ~CH3 chlorobenzylidene)hydrazinecarbonyl)ph
0 H enyl)acetamide

OH
H O~\ t~ F
N,
N HN" S N-(4-chloro-2-((2-(2-
114 0 H F hydroxybenzoyl)hydrazono)methyl)phen
yl)-2,4-difluorobenzenesulfonamide
CI
HO OH
I H
N, N OH 2,4-dihydroxy-N'-((2-
115 0 H hydroxynaphthalen-l-
~ yl)methylene)benzohydrazide
O ~
~ NN OH N'-(2-hydroxy-4-
116 o I methylbenzylidene)benzo [d] [ 1,3 ] dioxole
H ~ -5-carbohydrazide
CH3
2. Chemical Definitions

"Alkyl" refers to monovalent saturated aliphatic hydrocarbyl groups having
from 1 to 10 carbon atoms and, in some embodiments, from 1 to 6 carbon atoms.
"CX_yalkyl" refers to alkyl groups having from x to y carbon atoms. This term
includes, by
-49-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
way of example, linear and branched hydrocarbyl groups such as methyl (CH3-),
ethyl
(CH3CH2-), n-propyl (CH3CH2CH2-), isopropyl ((CH3)2CH-), n-butyl (CH3CH2CH2CH2-
),
isobutyl ((CH3)2CHCH2-), sec-butyl ((CH3)(CH3CH2)CH-), t-butyl ((CH3)3C-), n-
pentyl
(CH3CH2CH2CH2CH2-), and neopentyl ((CH3)3CCH2-).

"Substituted alkyl" refers to an alkyl group having from 1 to 5 and, in some
embodiments, 1 to 3 or 1 to 2 substituents selected from the group consisting
of alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy,
acyl,
acylamino, acyloxy, amino, substituted amino, aminocarbonyl,
aminothiocarbonyl,
aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl,
aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl,
aryloxy, substituted
aryloxy, arylthio, substituted arylthio, azido, carboxyl, carboxyl ester,
(carboxyl
ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl,
cycloalkyloxy,
substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio,
guanidino, substituted
guanidino, halo, hydroxy, hydroxyamino, alkoxyamino, hydrazino, substituted
hydrazino,
heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy,
heteroarylthio,
substituted heteroarylthio, heterocyclic, substituted heterocyclic,
heterocyclyloxy,
substituted heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio,
nitro,
spirocycloalkyl, S03H, substituted sulfonyl, sulfonyloxy, thioacyl,
thiocyanate, thiol,
alkylthio, and substituted alkylthio, wherein said substituents are as defined
herein.

"Alkylidene" or "alkylene" refers to divalent saturated aliphatic hydrocarbyl
groups having from 1 to 10 carbon atoms and, in some embodiments, from 1 to 6
carbon
atoms. "(Cõ_õ)alkylene" refers to alkylene groups having from u to v carbon
atoms. The
alkylidene or alkylene groups include branched and straight chain hydrocarbyl
groups. For
example "(Ci_6)alkylene" is meant to include methylene, ethylene, propylene, 2-

methypropylene, pentylene, and the like.

The term "aralkyl" refers to the term aryl-alkylene wherein alkylene is as
defined above and aryl is as defined below. Examples of this group include,
but are not
limited to benzyl, phenethyl, and the like.

"Substituted alkylidene" or "substituted alkylene" refers to an alkylidene
group having from 1 to 5 and, in some embodiments, 1 to 3 or 1 to 2
substituents selected
-50-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino,
acyloxy, amino,
substituted amino, aminocarbonyl, aminothiocarbonyl, aminocarbonylamino,
aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy,
aminosulfonylamino, amidino, aryl, substituted aryl, aryloxy, substituted
aryloxy, arylthio,
substituted arylthio, azido, carboxyl, carboxyl ester, (carboxyl ester)amino,
(carboxyl
ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy,
substituted
cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino,
substituted guanidino,
halo, hydroxy, hydroxyamino, alkoxyamino, hydrazino, substituted hydrazino,
heteroaryl,
substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy,
heteroarylthio, substituted
heteroarylthio, heterocyclic, substituted heterocyclic, heterocyclyloxy,
substituted
heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, nitro, oxo,
thione,
spirocycloalkyl, SO3H, substituted sulfonyl, sulfonyloxy, thioacyl,
thiocyanate, thiol,
alkylthio, and substituted alkylthio, wherein said substituents are as defined
herein.

"Alkenyl" refers to a linear or branched hydrocarbyl group having from 2 to
10 carbon atoms and in some embodiments from 2 to 6 carbon atoms or 2 to 4
carbon atoms
and having at least 1 site of vinyl unsaturation (>C=C<). For example, (CX-
Cy)alkenyl
refers to alkenyl groups having from x to y carbon atoms and is meant to
include for
example, ethenyl, propenyl, 1,3-butadienyl, and the like.

"Substituted alkenyl" refers to alkenyl groups having from 1 to 3 substituents
and, in some embodiments, 1 to 2 substituents selected from the group
consisting of alkoxy,
substituted alkoxy, acyl, acylamino, acyloxy, alkyl, substituted alkyl,
alkynyl, substituted
alkynyl, amino, substituted amino, aminocarbonyl, aminothiocarbonyl,
aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl,
aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl,
aryloxy, substituted
aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl
ester)amino,
(carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy,
substituted
cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino,
substituted guanidino,
halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted
heteroaryloxy,
heteroarylthio, substituted heteroarylthio, heterocyclic, substituted
heterocyclic,
heterocyclyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted
heterocyclylthio,
nitro, S03H, substituted sulfonyl, sulfonyloxy, thioacyl, thiol, alkylthio,
and substituted
-51-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
alkylthio, wherein said substituents are defined herein and with the proviso
that any
hydroxy or thiol substitution is not attached to a vinyl (unsaturated) carbon
atom.

"Alkynyl" refers to a linear monovalent hydrocarbon radical or a branched
monovalent hydrocarbon radical containing at least one triple bond. The term
"alkynyl" is
also meant to include those hydrocarbyl groups having one triple bond and one
double
bond. For example, (C2-C6)alkynyl is meant to include ethynyl, propynyl, and
the like.
"Substituted alkynyl" refers to alkynyl groups having from 1 to 3
substituents and, in some embodiments, from 1 to 2 substituents selected from
the group
consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, amino, substituted amino, aminocarbonyl,
aminothiocarbonyl,
aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl,
aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl,
aryloxy, substituted
aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl
ester)amino,
(carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy,
substituted
cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino,
substituted guanidino,
halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted
heteroaryloxy,
heteroarylthio, substituted heteroarylthio, heterocyclic, substituted
heterocyclic,
heterocyclyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted
heterocyclylthio,
nitro, S03H, substituted sulfonyl, sulfonyloxy, thioacyl, thiol, alkylthio,
and substituted
alkylthio, wherein said substituents are as defined herein and with the
proviso that any
hydroxy or thiol substitution is not attached to an acetylenic carbon atom.

"Alkoxy" refers to the group -0-alkyl wherein alkyl is defined herein.
Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-
butoxy,
t-butoxy, sec-butoxy, and n-pentoxy.

"Substituted alkoxy" refers to the group -O-(substituted alkyl) wherein
substituted alkyl is as defined herein.

"Acyl" refers to the groups H-C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-,
alkenyl-C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted alkynyl-
C(O)-,
cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, aryl-C(O)-, substituted aryl-
C(O)-,

-52-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
substituted hydrazino-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O)-,
heterocyclic-C(O)-, and substituted heterocyclic-C(O)-, wherein alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl,
substituted cycloalkyl,
aryl, substituted aryl, substituted hydrazino, heteroaryl, substituted
heteroaryl, heterocyclic,
and substituted heterocyclic are as defined herein. Acyl includes the "acetyl"
group
CH3C(O)-.

"Acylamino" refers to the groups -NR20C(O)alkyl, -NR2OC(O)substituted
alkyl, -NR20C(O)cycloalkyl, -NR20C(O)substituted cycloalkyl, -NR2OC(O)alkenyl,
-NR20C(O)substituted alkenyl, -NR2OC(O)alkynyl, -NR2OC(O)substituted alkynyl,
-NR20C(O)aryl, -NR2OC(O)substituted aryl, -NR2OC(O)heteroaryl, -
NR2OC(O)substituted
heteroaryl, -NR20C(O)heterocyclic, and -NR2OC(O)substituted heterocyclic
wherein R20 is
hydrogen or alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted
alkenyl, alkynyl,
substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted
aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic are as
defined herein.

"Acyloxy" refers to the groups alkyl-C(O)O-, substituted alkyl-C(O)O-,
alkenyl-C(O)O-, substituted alkenyl-C(O)O-, alkynyl-C(O)O-, substituted
alkynyl-C(O)O-,
aryl-C(O)O-, substituted aryl-C(O)O-, cycloalkyl-C(O)O-, substituted
cycloalkyl-C(O)O-,
heteroaryl-C(O)O-, substituted heteroaryl-C(O)O-, heterocyclic-C(O)O-, and
substituted
heterocyclic-C(O)O- wherein alkyl, substituted alkyl, alkenyl, substituted
alkenyl, alkynyl,
substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted
aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic are as
defined herein.
"Amino" refers to the group -NH2.

"Substituted amino" refers to the group -NR2iR22 where R 21 and R22 are
independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic,
substituted
heterocyclic, -S02-alkyl, -S02-substituted alkyl, -S02-alkenyl, -S02-
substituted alkenyl,
-S02-cycloalkyl, -S02-substituted cylcoalkyl, -S02-aryl, -S02-substituted
aryl,
-S02-heteroaryl, -SOz-substituted heteroaryl, -SOz-heterocyclic, and -SOz-
substituted
heterocyclic and wherein R 21 and R22 are optionally joined together with the
nitrogen bound
-53-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
thereto to form a heterocyclic or substituted heterocyclic group, provided
that R21 and R22
are both not hydrogen, and wherein alkyl, substituted alkyl, alkenyl,
substituted alkenyl,
alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl,
substituted aryl,
heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic
are as defined
herein. When R 21 is hydrogen and R22 is alkyl, the substituted amino group is
sometimes
referred to herein as alkylamino. When R21 and R22 are alkyl, the substituted
amino group
is sometimes referred to herein as dialkylamino. When referring to a
monosubstituted
amino, it is meant that either R21 or R22 is hydrogen but not both. When
referring to a
disubstituted amino, it is meant that neither R 21 nor R22 are hydrogen.

"Hydroxyamino" refers to the group -NHOH.

"Alkoxyamino" refers to the group -NHO-alkyl wherein alkyl is defined
herein.

"Aminocarbonyl" refers to the group -C(O)NR23R24 where R23 and R24 are
independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic,
substituted
heterocyclic, hydroxy, alkoxy, substituted alkoxy, amino, substituted amino,
and acylamino,
and where R23 and R24 are optionally joined together with the nitrogen bound
thereto to
form a heterocyclic or substituted heterocyclic group, and wherein alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl,
substituted cycloalkyl,
aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic are as defined herein.

"Aminothiocarbonyl" refers to the group -C(S)NR23R24 where R23 and R24
are independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic and where R23 and R24 are optionally joined together with the
nitrogen bound
thereto to form a heterocyclic or substituted heterocyclic group, and wherein
alkyl,
substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl,
cycloalkyl,

-54-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted
heteroaryl, heterocyclic,
and substituted heterocyclic are as defined herein.

"Aminocarbonylamino" refers to the group -NR20C(O)NR23R24 where R20 is
hydrogen or alkyl and R23 and R24 are independently selected from the group
consisting of
hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl,
substituted alkynyl,
aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl,
substituted heteroaryl,
heterocyclic, and substituted heterocyclic and where R23 and R24 are
optionally joined
together with the nitrogen bound thereto to form a heterocyclic or substituted
heterocyclic
group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl,
alkynyl, substituted
alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
heteroaryl, substituted
heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.
"Aminothiocarbonylamino" refers to the group -NR20C(S)NR23R24 where
R20 is hydrogen or alkyl and R23 and R24 are independently selected from the
group
consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted
alkenyl, alkynyl,
substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted
cycloalkyl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic and where
R23 and R24 are
optionally joined together with the nitrogen bound thereto to form a
heterocyclic or
substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl,
substituted
alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl,
aryl, substituted
aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted
heterocyclic are as
defined herein.

"Aminocarbonyloxy" refers to the group -O-C(O)NR23R24 where R23 and
R24 are independently selected from the group consisting of hydrogen, alkyl,
substituted
alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl,
substituted aryl,
cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl,
heterocyclic, and
substituted heterocyclic and where R23 and R24 are optionally joined together
with the
nitrogen bound thereto to form a heterocyclic or substituted heterocyclic
group, and wherein
alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted
alkynyl,
cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,
substituted heteroaryl,
heterocyclic, and substituted heterocyclic are as defined herein.

-55-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
"Aminosulfonyl" refers to the group -SO2NR23R24 where R23 and R24 are
independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic and where R23 and R24 are optionally joined together with the
nitrogen bound
thereto to form a heterocyclic or substituted heterocyclic group, and wherein
alkyl,
substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl,
cycloalkyl,
substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted
heteroaryl, heterocyclic,
and substituted heterocyclic are as defined herein.

"Aminosulfonyloxy" refers to the group -O-SO2NR23R24 where R23 and R24
are independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic and where R23 and R24 are optionally joined together with the
nitrogen bound
thereto to form a heterocyclic or substituted heterocyclic group, and wherein
alkyl,
substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl,
cycloalkyl,
substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted
heteroaryl, heterocyclic,
and substituted heterocyclic are as defined herein.

"Aminosulfonylamino" refers to the group -NR20-SO2NR23R24 where R20 is
hydrogen or alkyl and R23 and R24 are independently selected from the group
consisting of
hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl,
substituted alkynyl,
aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl,
substituted heteroaryl,
heterocyclic, and substituted heterocyclic and where R23 and R24 are
optionally joined
together with the nitrogen bound thereto to form a heterocyclic or substituted
heterocyclic
group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl,
alkynyl, substituted
alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
heteroaryl, substituted
heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

"Amidino" refers to the group -C(=NR25)NR23R24 where R25, R23, and R24
are independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted
aryl, cycloalkyl,
-56-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic and where R23 and R24 are optionally joined together with the
nitrogen bound
thereto to form a heterocyclic or substituted heterocyclic group, and wherein
alkyl,
substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl,
cycloalkyl,
substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted
heteroaryl, heterocyclic,
and substituted heterocyclic are as defined herein.

"Aryl" or "Ar" refers to an aromatic group of from 6 to 14 carbon atoms and
no ring heteroatoms and having a single ring (e.g., phenyl) or multiple
condensed (fused)
rings (e.g., naphthyl or anthryl). For multiple ring systems, including fused,
bridged, and
spiro ring systems having aromatic and non-aromatic rings that have no ring
heteroatoms,
the term "Aryl" or "Ar" applies when the point of attachment is at an aromatic
carbon atom
(e.g., 5,6,7,8 tetrahydronaphthalene-2-yl is an aryl group as its point of
attachment is at the
2-position of the aromatic phenyl ring).

The term "biaryl" refers to the group aryl-arylene where the term aryl is as
noted above and the term arylene refers to a divalent aryl group. Examples of
this include,
but are not limited to, biphenyl.

"Substituted aryl" refers to aryl groups which are substituted with 1 to 8
and,
in some embodiments, 1 to 5, 1 to 3, or 1 to 2 substituents selected from the
group
consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl,
substituted

alkynyl, alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino,
substituted amino,
aminocarbonyl, aminothiocarbonyl, aminocarbonylamino, aminothiocarbonylamino,
aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonylamino,
amidino, aryl,
substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted
arylthio, azido, carboxyl,
carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl,
substituted
cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio,
substituted
cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, hydroxyamino,
alkoxyamino, hydrazino, substituted hydrazino, heteroaryl, substituted
heteroaryl,
heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted
heteroarylthio,
heterocyclic, substituted heterocyclic, heterocyclyloxy, substituted
heterocyclyloxy,
heterocyclylthio, substituted heterocyclylthio, nitro, S03H, substituted
sulfonyl,
-57-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
sulfonyloxy, thioacyl, thiocyanate, thiol, alkylthio, and substituted
alkylthio, wherein said
substituents are defined herein.

"Aryloxy" refers to the group -0-aryl, where aryl is as defined herein, that
includes, by way of example, phenoxy and naphthyloxy.

"Substituted aryloxy" refers to the group -O-(substituted aryl) where
substituted aryl is as defined herein.

"Arylthio" refers to the group -S-aryl, where aryl is as defined herein.
"Substituted arylthio" refers to the group -S-(substituted aryl), where
substituted aryl is as defined herein.

"Azido" refers to the group -N3.
"Hydrazino" refers to the group -NHNH2.

"Substituted hydrazino" refers to the group -NR26NR2'R2g where R26, R27,
and R28 are independently selected from the group consisting of hydrogen,
alkyl, substituted
alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl,
substituted aryl,
carboxyl ester, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted
heteroaryl,
heterocyclic, substituted heterocyclic, -SO2-alkyl, -SO2-substituted alkyl, -
SO2-alkenyl,
-SOz-substituted alkenyl, -SOz-cycloalkyl, -SOz-substituted cylcoalkyl, -SOz-
aryl,
-SOz-substituted aryl, -SOz-heteroaryl, -SOz-substituted heteroaryl, -SOz-
heterocyclic, and
-SO2-substituted heterocyclic and wherein R 27 and R 28 are optionally joined,
together with
the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic
group,
provided that R 27 and R28 are both not hydrogen, and wherein alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl,
substituted cycloalkyl,
aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic are as defined herein.

"Cyano" or "carbonitrile" refers to the group -CN.

"Carbonyl" refers to the divalent group -C(O)- which is equivalent to
-C(=0)-.
-58-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
"Carboxyl" or "carboxy" refers to -COOH or salts thereof.

"Carboxyl ester" or "carboxy ester" refers to the groups -C(O)O-alkyl,
-C(O)O-substituted alkyl, -C(O)O-alkenyl, -C(O)O-substituted alkenyl, -C(O)O-
alkynyl,
-C(O)O-substituted alkynyl, -C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-
cycloalkyl,
-C(O)O-substituted cycloalkyl, -C(O)O-heteroaryl, -C(O)O-substituted
heteroaryl,
-C(O)O-heterocyclic, and -C(O)O-substituted heterocyclic wherein alkyl,
substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl,
substituted cycloalkyl,
aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and
substituted
heterocyclic are as defined herein.

"(Carboxyl ester)amino" refers to the group -NR20-C(O)O-alkyl,
-NR20-C(O)O-substituted alkyl, -NR2O-C(O)O-alkenyl, -NR2O-C(O)O-substituted
alkenyl,
-NR20-C(O)O-alkynyl, -NR2O-C(O)O-substituted alkynyl, -NR2O-C(O)O-aryl,
-NR20-C(O)O-substituted aryl, -NR2O-C(O)O-cycloalkyl, -NR2O-C(O)O-substituted
cycloalkyl, -NR20-C(O)O-heteroaryl, -NR2O-C(O)O-substituted heteroaryl,
-NR20-C(O)O-heterocyclic, and -NR2O-C(O)O-substituted heterocyclic wherein R20
is alkyl
or hydrogen, and wherein alkyl, substituted alkyl, alkenyl, substituted
alkenyl, alkynyl,
substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted
aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic are as
defined herein.
"(Carboxyl ester)oxy" refers to the group -O-C(O)O-alkyl,
-O-C(O)O-substituted alkyl, -O-C(O)O-alkenyl, -O-C(O)O-substituted alkenyl,
-O-C(O)O-alkynyl, -O-C(O)O-substituted alkynyl, -O-C(O)O-aryl, -O-C(O)O-
substituted
aryl, -O-C(O)O-cycloalkyl, -O-C(O)O-substituted cycloalkyl, -O-C(O)O-
heteroaryl,
-O-C(O)O-substituted heteroaryl, -O-C(O)O-heterocyclic, and -O-C(O)O-
substituted
heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl,
alkynyl,
substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted
aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic are as
defined herein.
"Cycloalkyl" refers to a saturated or partially saturated cyclic group of from
3 to 14 carbon atoms and no ring heteroatoms and having a single ring or
multiple rings
including fused, bridged, and spiro ring systems. For multiple ring systems
having aromatic
and non-aromatic rings that have no ring heteroatoms, the term "cycloalkyl"
applies when
-59-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
the point of attachment is at a non-aromatic carbon atom (e.g. 5,6,7,8,-
tetrahydronaphthalene-5-yl). The term "Cycloalkyl" includes cycloalkenyl
groups.
Examples of cycloalkyl groups include, for instance, adamantyl, cyclopropyl,
cyclobutyl,
cyclopentyl, cyclooctyl, and cyclohexenyl. "Cõ_õcycloalkyl" refers to
cycloalkyl groups
having u to v carbon atoms.

"Cycloalkenyl" refers to a partially saturated cycloalkyl ring having at least
one site of >C=C< ring unsaturation.

"Substituted cycloalkyl" refers to a cycloalkyl group, as defined herein,
having from 1 to 8, or 1 to 5, or in some embodiments 1 to 3 substituents
selected from the
group consisting of oxo, thione, alkyl, substituted alkyl, alkenyl,
substituted alkenyl,
alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, acyl, acylamino,
acyloxy, amino,
substituted amino, aminocarbonyl, aminothiocarbonyl, aminocarbonylamino,
aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy,
aminosulfonylamino, amidino, aryl, substituted aryl, aryloxy, substituted
aryloxy, arylthio,
substituted arylthio, azido, carboxyl, carboxyl ester, (carboxyl ester)amino,
(carboxyl
ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy,
substituted
cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino,
substituted guanidino,
halo, hydroxy, hydroxyamino, alkoxyamino, hydrazino, substituted hydrazino,
heteroaryl,
substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy,
heteroarylthio, substituted
heteroarylthio, heterocyclic, substituted heterocyclic, heterocyclyloxy,
substituted
heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, nitro, S03H,
substituted
sulfonyl, sulfonyloxy, thioacyl, thiocyanate, thiol, alkylthio, and
substituted alkylthio,
wherein said substituents are as defined herein. The term "substituted
cycloalkyl" includes
substituted cycloalkenyl groups.

"Cycloalkyloxy" refers to -0-cycloalkyl wherein cycloalkyl is as defined
herein.

"Substituted cycloalkyloxy" refers to -O-(substituted cycloalkyl) wherein
substituted cycloalkyl is as defined herein.

-60-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
"Cycloalkylthio" refers to -S-cycloalkyl wherein cycloalkyl is as defined
herein.

"Substituted cycloalkylthio" refers to -S-(substituted cycloalkyl).
"Guanidino" refers to the group -NHC(=NH)NH2.

"Substituted guanidino" refers to -NR29C(=NR29)N(R29)2 where each R29 is
independently selected from the group consisting of hydrogen, alkyl,
substituted alkyl, aryl,
substituted aryl, heteroaryl, substituted heteroaryl, heterocyclyl, and
substituted heterocyclyl
and two R29 groups attached to a common guanidino nitrogen atom are optionally
joined
together with the nitrogen bound thereto to form a heterocyclic or substituted
heterocyclic
group, provided that at least one R29 is not hydrogen, and wherein said
substituents are as
defined herein.

"Halo" or "halogen" refers to fluoro, chloro, bromo, and iodo.
"Haloalkyl" refers to substitution of alkyl groups with 1 to 5 or in some
embodiments 1 to 3 halo groups.

"Haloalkoxy" refers to substitution of alkoxy groups with 1 to 5 or in some
embodiments 1 to 3 halo groups.

"Hydroxy" or "hydroxyl" refers to the group -OH.

"Heteroaryl" refers to an aromatic group of from 1 to 14 carbon atoms and 1
to 6 heteroatoms selected from the group consisting of oxygen, nitrogen, and
sulfur and
includes single ring (e.g. imidazolyl) and multiple ring systems (e.g.
benzimidazol-2-yl and
benzimidazol-6-yl). For multiple ring systems, including fused, bridged, and
spiro ring
systems having aromatic and non-aromatic rings, the term "heteroaryl" applies
if there is at
least one ring heteroatom and the point of attachment is at an atom of an
aromatic ring (e.g.
1,2,3,4-tetrahydroquinolin-6-yl and 5,6,7,8-tetrahydroquinolin-3-yl). In one
embodiment,
the nitrogen and/or the sulfur ring atom(s) of the heteroaryl group are
optionally oxidized to
provide for the N-oxide (N--->O), sulfinyl, or sulfonyl moieties. More
specifically the term
heteroaryl includes, but is not limited to, pyridyl, furanyl, thienyl,
thiazolyl, isothiazolyl,

-61-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
triazolyl, imidazolyl, isoxazolyl, pyrrolyl, pyrazolyl, pyridazinyl,
pyrimidinyl, benzofuranyl,
tetrahydrobenzofuranyl, isobenzofuranyl, benzothiazolyl, benzoisothiazolyl,
benzotriazolyl,
indolyl, isoindolyl, benzoxazolyl, quinolyl, tetrahydroquinolinyl,
isoquinolyl,
quinazolinonyl, benzimidazolyl, benzisoxazolyl, benzothienyl, or oxindolyl.

"Substituted heteroaryl" refers to heteroaryl groups that are substituted with
from 1 to 8 or in some embodiments 1 to 5, or 1 to 3, or 1 to 2 substituents
selected from the
group consisting of the substituents defined for substituted aryl.

"Heteroaryloxy" refers to -0-heteroaryl wherein heteroaryl is as defined
herein.

"Substituted heteroaryloxy refers to the group -O-(substituted heteroaryl)
wherein substituted heteroaryl is as defined herein.

"Heteroarylthio" refers to the group -S-heteroaryl wherein heteroaryl is as
defined herein.

"Substituted heteroarylthio" refers to the group -S-(substituted heteroaryl)
wherein substituted heteroaryl is as defined herein.

"Heterocyclic" or "heterocycle" or "heterocycloalkyl" or "heterocyclyl"
refers to a saturated or partially saturated cyclic group having from 1 to 14
carbon atoms
and from 1 to 6 heteroatoms selected from the group consisting of nitrogen,
sulfur, or
oxygen and includes single ring and multiple ring systems including fused,
bridged, and
spiro ring systems. For multiple ring systems having aromatic and/or non-
aromatic rings,
the terms "heterocyclic", "heterocycle", "heterocycloalkyl", or "heterocyclyl"
apply when
there is at least one ring heteroatom and the point of attachment is at an
atom of a non-
aromatic ring (e.g. 1,2,3,4-tetrahydroquinoline-3-yl, 5,6,7,8-
tetrahydroquinoline-6-yl, and
decahydroquinolin-6-yl). In one embodiment, the nitrogen and/or sulfur atom(s)
of the
heterocyclic group are optionally oxidized to provide for the N-oxide,
sulfinyl, sulfonyl
moieties. More specifically the heterocyclyl includes, but is not limited to,
tetrahydropyranyl, piperidinyl, N-methylpiperidin-3-yl, piperazinyl, N-
methylpyrrolidin-3-
yl, 3-pyrrolidinyl, 2-pyrrolidon-l-yl, morpholinyl, and pyrrolidinyl. A prefix
indicating the

-62-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
number of carbon atoms (e.g., C3-Cio) refers to the total number of carbon
atoms in the
portion of the heterocyclyl group exclusive of the number of heteroatoms.

"Substituted heterocyclic" or "Substituted heterocycle" or "substituted
heterocycloalkyl" or "substituted heterocyclyl" refers to heterocyclic groups,
as defined
herein, that are substituted with from 1 to 5 or in some embodiments 1 to 3 of
the
substituents as defined for substituted cycloalkyl.

"Heterocyclyloxy" refers to the group -0-heterocyclyl wherein heterocyclyl
is as defined herein.

"Substituted heterocyclyloxy" refers to the group -O-(substituted
heterocyclyl) wherein substituted heterocyclyl is as defined herein.

"Heterocyclylthio" refers to the group -S-heterocyclyl wherein heterocyclyl
is as defined herein.

"Substituted heterocyclylthio" refers to the group -S-(substituted
heterocyclyl) wherein substituted heterocyclyl is as defined herein.

Examples of heterocycle and heteroaryl groups include, but are not limited
to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine,
pyridazine,
indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine,
isoquinoline,
quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline,
pteridine,
carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole,
phenazine,
isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine,
piperazine,
indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7-
tetrahydrobenzo[b]thiophene,
thiazole, thiazolidine, thiophene, benzo[b]thiophene, morpholinyl,
thiomorpholinyl (also
referred to as thiamorpholinyl), l,l-dioxothiomorpholinyl, piperidinyl,
pyrrolidine, and
tetrahydrofuranyl.

"Nitro" refers to the group -NOz.
"Oxo" refers to the atom (=0).

- 63 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
"Oxide" refers to products resulting from the oxidation of one or more
heteroatoms. Examples include N-oxides, sulfoxides, and sulfones.

"Spirocycloalkyl" refers to a 3 to 10 member cyclic substituent formed by
replacement of two hydrogen atoms at a common carbon atom with an alkylene
group
having 2 to 9 carbon atoms, as exemplified by the following structure wherein
the
methylene group shown here attached to bonds marked with wavy lines is
substituted with a
spirocycloalkyl group:

"Sulfonyl" refers to the divalent group -S(O)2-.

"Substituted sulfonyl" refers to the group -S02-alkyl, -S02-substituted alkyl,
-S02-alkenyl, -S02-substituted alkenyl, -S02-alkynyl, -S02-substituted
alkynyl,
-S02-cycloalkyl, -S02-substituted cylcoalkyl, -S02-aryl, -S02-substituted
aryl,
-S02-heteroaryl, -S02-substituted heteroaryl, -S02-heterocyclic, -S02-
substituted
heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl,
alkynyl,
substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted
aryl, heteroaryl,
substituted heteroaryl, heterocyclic and substituted heterocyclic are as
defined herein.
Substituted sulfonyl includes groups such as methyl-SO2-, phenyl-SO2-, and
4-methylphenyl-SO2-.

"Sulfonyloxy" refers to the group -OSOz-alkyl, -OSOz-substituted alkyl,
-OSOz-alkenyl, -OSOz-substituted alkenyl, -OSOz-cycloalkyl, -OSOz-substituted
cylcoalkyl, -OSO2-aryl, -OSO2-substituted aryl, -OSO2-heteroaryl, -OSO2-
substituted
heteroaryl, -OSOz-heterocyclic, -OSOz-substituted heterocyclic, wherein alkyl,
substituted
alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl,
substituted
cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl,
heterocyclic and
substituted heterocyclic are as defined herein.

"Thioacyl" refers to the groups H-C(S)-, alkyl-C(S)-, substituted
alkyl-C(S)-, alkenyl-C(S)-, substituted alkenyl-C(S)-, alkynyl-C(S)-,
substituted
-64-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
alkynyl-C(S)-, cycloalkyl-C(S)-, substituted cycloalkyl-C(S)-, aryl-C(S)-,
substituted
aryl-C(S)-, heteroaryl-C(S)-, substituted heteroaryl-C(S)-, heterocyclic-C(S)-
, and
substituted heterocyclic-C(S)-, wherein alkyl, substituted alkyl, alkenyl,
substituted alkenyl,
alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl,
substituted aryl,
heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic
are as defined
herein.

"Thiol" refers to the group -SH.

"Alkylthio" refers to the group -S-alkyl wherein alkyl is as defined herein.
"Substituted alkylthio" refers to the group -S-(substituted alkyl) wherein
substituted alkyl is as defined herein.

"Thiocarbonyl" refers to the divalent group -C(S)- which is equivalent to
-C(=S)-.

"Thione" refers to the atom (=S).
"Thiocyanate" refers to the group -SCN.

"Compound" and "compounds" as used herein refers to a compound
encompassed by the generic formulae disclosed herein, any subgenus of those
generic
formulae, and any forms of the compounds within the generic and subgeneric
formulae.
Unless specified otherwise, the term further includes the racemates,
stereoisomers, and
tautomers of the compound or compounds.

"Racemates" refers to a mixture of enantiomers.

"Solvate" or "solvates" of a compound refer to those compounds, where
compounds is as defined above, that are bound to a stoichiometric or non-
stoichiometric
amount of a solvent. Solvates of a compound includes solvates of all forms of
the
compound. Preferred solvents are volatile, non-toxic, and/or acceptable for
administration
to humans in trace amounts. Suitable solvates include water.
- 65 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
"Stereoisomer" or "stereoisomers" refer to compounds that differ in the
chirality of one or more stereocenters. Stereoisomers include enantiomers and
diastereomers.

"Tautomer" refer to alternate forms of a compound that differ in the position
of a proton, such as enol-keto and imine-enamine tautomers, or the tautomeric
forms of
heteroaryl groups containing a ring atom attached to both a ring -NH- moiety
and a ring =N-
moiety such as pyrazoles, imidazoles, benzimidazoles, triazoles, and
tetrazoles.

"Prodrug" refers to any derivative of a compound of the embodiments that is
capable of directly or indirectly providing a compound of the embodiments or
an active
metabolite or residue thereof when administered to a subject. Particularly
favored
derivatives and prodrugs are those that increase the bioavailability of the
compounds of the
embodiments when such compounds are administered to a subject (e.g., by
allowing an
orally administered compound to be more readily absorbed into the blood) or
which
enhance delivery of the parent compound to a biological compartment (e.g., the
brain or
lymphatic system) relative to the parent species. Prodrugs include ester forms
of the
compounds of the invention. Examples of ester prodrugs include formate,
acetate,
propionate, butyrate, acrylate, and ethylsuccinate derivatives. An general
overview of
prodrugs 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 Edward B. Roche, ed.,
Bioreversible
Carriers in Drug Design, American Pharmaceutical Association and Pergamon
Press, 1987,
both of which are incorporated herein by reference.

For example in this invention, the term "prodrug" refers to compounds of
formula I, II, or III that include chemical groups which, in vivo, can be
converted to the
carboxylate group, hydroxyl group, or amine group on the R3 group, the
hydroxyl group or
amine group on the Rigroup of the A ring or the amine of a heteroaryl A ring.
Examples of
such chemical groups which can act as prodrugs for carboxylates and hydroxyl
groups are
esters which can be chemically cleaved or enzymatically cleaved by esterases.
Examples of
such chemical groups which can act as prodrugs for amines are amides which can
be
enzymatically cleaved by proteases and phosphoryloxymethyl carbamates which
can be

-66-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
enzymatically cleaved by alkaline phosphatases (see Safadi et al.
Pharmaceutical Research
1993, 10 (9), 1350-1355).

"Pharmaceutically acceptable salt" refers to pharmaceutically acceptable
salts derived from a variety of organic and inorganic counter ions well known
in the art and
include, by way of example only, sodium, potassium, calcium, magnesium,
ammonium, and
tetraalkylammonium, and when the molecule contains a basic functionality,
salts of organic
or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate,
acetate,
maleate, and oxalate. Suitable salts include those described in P. Heinrich
Stahl, Camille G.
Wermuth (Eds.), Handbook of Pharmaceutical Salts Properties, Selection, and
Use; 2002.

Unless indicated otherwise, the nomenclature of substituents that are not
explicitly defined herein are arrived at by naming the terminal portion of the
functionality
followed by the adjacent functionality toward the point of attachment. For
example, the
substituent "arylalkyloxycabonyl" refers to the group (aryl)-(alkyl)-O-C(O)-.

It is understood that in all substituted groups defined above, polymers
arrived
at by defining substituents with further substituents to themselves (e.g.,
substituted aryl
having a substituted aryl group as a substituent which is itself substituted
with a substituted
aryl group, which is further substituted by a substituted aryl group etc.) are
not intended for
inclusion herein. In such cases, the maximum number of such substitutions is
three. For
example, serial substitutions of substituted aryl groups with two other
substituted aryl
groups are limited to -substituted aryl-(substituted aryl)-substituted aryl.

Similarly, it is understood that the above definitions are not intended to
include impermissible substitution patterns (e.g., methyl substituted with 5
fluoro groups).
Such impermissible substitution patterns are well known to the skilled
artisan.

3. Compound Selection

A preferred method of selecting compounds for their use in the methods of
the invention involves assaying the compounds with Fluorescence Resonance
Energy
Transfer (FRET). FRET has been used to measure, detect, identify, assay,
analyze, and
characterize various interactions and processes in biological systems (see
e.g., Mitra et al.

-67-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
(1996) Gene 173:13-17; De Angelis (1999) Physiol. Genomics. 21:93-99; Latif
and Graves
(2000) Thyroid 10(5):407-412; Rye (2001) Methods 24(3):278-288; Kenworthy
(2001)
Methods 24(3):289-296; Periasamy (2001) J. Biomed. Opt. 6(3):287-291; Truong
and Ikura
(2001) Curr. Opin. Struct. Biol. 11(5):573-578; Zhang et al. (2002) Nat. Rev.
Mol. Cell.
Biol. 3(12):906-918; Sitte and Freissmuth (2003) Eur. J. Pharmacol. 479:229-
236; Milligan
(2004) Eur. J. Pharm. Sci. 21(4):397-405; Herman et al. (2004) Methods Mol.
Biol.
261:351-370; Roda et al. (2004) Trends Biotechnol. 22(6):295-303; Wallrabe and
Periasamy (2005) Curr. Opin. Biotechnol. 16(1):19-27; Milligan and Bouvier
(2005) FEBS
J. 272(12):2914-2915; references in any of the foregoing; and the like).

FRET methods, protocols, techniques, assays, and the like are described
generally and specifically in a number of patents and patent applications,
including: U.S.
Patent No. 6,908,769; U.S. Patent No. 6,824,990; U.S. Patent No. 6,762,280;
U.S. Patent
No. 6,689,574; U.S. Patent No. 6,661,909; U.S. Patent No. 6,642,001; U.S.
Patent No.
6,639,078; U.S. Patent No. 6,472,156; U.S. Patent No. 6,456,734; U.S. Patent
No.
6,376,257; U.S. Patent No. 6,348,322; U.S. Patent No. 6,323,039; U.S. Patent
No.
6,291,201; U.S. Patent No. 6,280,981; U.S. Patent No. 5,914,245; U.S. Patent
No.
5,661,035; references in any of the foregoing; and the like.

Similarly, fluorescence polarization (FP) has also been used to measure,
detect, identify, assay, analyze, and characterize various interactions and
processes in
biological systems (see e.g., Lundblad et al. (1996) Mol. Endocrinol.
10(6):607-612; Nasir
and Jolley (1999) Comb. Chem. High Throughput Screen. 2(4):177-190; Park and
Raines
(2004) Methods Mol. Biol. 261:161-166; references in any of the foregoing; and
the like).

Fluorescence polarization (FP) methods, protocols, techniques, assays, and
the like are described generally and specifically in a number of patents and
patent
applications, including: U.S. Patent No. 6,794,158; U.S. Patent No. 6,632,613;
U.S. Patent
No. 6,630,295; 6,596,546; U.S. Patent No. 6,569,628; 6,555,326; U.S. Patent
No.
6,511,815; U.S. Patent No. 6,448,018; U.S. Patent No. 6,432,632; U.S. Patent
No.
6,331,392; U.S. Patent No. 6,326,142; U.S. Patent No. 6,284,544; U.S. Patent
No.
6,207,397; U.S. Patent No. 6,171,807; U.S. Patent No. 6,066,505; U.S. Patent
No.
5,976,820; U.S. Patent No. 5,804,395; U.S. Patent No. 5,756,292; U.S. Patent
No.
-68-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
5,445,935; U.S. Patent No. 5,427,960; U.S. Patent No. 5,407,834; U.S. Patent
No.
5,391,740; U.S. Patent No. 5,315,015; U.S. Patent No. 5,206,179; U.S. Patent
No.
5,070,025; U.S. Patent No. 5,066,426; U.S. Patent No. 4,952,691; U.S. Patent
No.
4,863,876; U.S. Patent No. 4,751,190; U.S. Patent No. 4,681,859; U.S. Patent
No.
4,668,640; U.S. Patent No. 4,614,823; U.S. Patent No. 4,585,862; U.S. Patent
No.
4,510,251; U.S. Patent No. 4,476,229; U.S. Patent No. 4,429,230; U.S. Patent
No.
4,420,568; U.S. Patent No. 4,203,670; references in any of the foregoing; and
the like.

FRET and FP have been used in the field of amyloid research (see e.g., U.S.
Patent No. 6,927,401; U.S. Patent No. 6,906,104; U.S. Patent No. 6,905,827;
U.S. Patent
No. 6,881,546; U.S. Patent No. 6,864,290; U.S. Patent No. 6,864,103; U.S.
Patent No.
6,858,383; U.S. Patent No. 6,846,813; U.S. Patent No. 6,828,106; U.S. Patent
No. U.S.
Patent No. 6,803,188; U.S. Patent No. 6,770,448; U.S. Patent No. 6,713,276;
U.S. Patent
No. 6,600,017; U.S. Patent No. 6,515,113; U.S. Patent No. 6,495,664; U.S.
Patent No.
6,323,039; U.S. Patent No. 6,294,330; U.S. Patent No. 6,280,981; U.S. Patent
No.
6,197,928; U.S. PatentNo. 5,981,200; Kim and Lee (2004) Biochem. Biophys. Res.
Commun. 316(2):393-397; Bacskai et al. (2003) J. Biomed. Opt. 8(3):368-375;
Gorman,
P.M. et al. (2003) J. Mol. Biol. 325(4):743-757; Garzon-Rodrequez et al.
(1997) J. Biol.
Chem. 272(34):21037-21044; Lindgren et al. (2005) Biophys. J. 88(6):4200-4212;
Lewis et
al. (2004) Neurobiol. Aging 25(9):1175-1185; Leissring et al. (2003) J. Biol.
Chem.
278(39):37314-37320; Taylor et al. (2003) J. Protein Chem. 22(1):31-40; Allsop
et al.
(2001) Biochem. Soc. Symp. 67:1-14; Allsop et al. (2001) Biochem. Biophys.
Res.
Commun. 285(1):58-63; Huang et al. (2000) J. Biol. Chem. 275(46):36436-36440;
references in any of the foregoing; and the like).

4. Applications of FRET for Compound Discovery

Further compounds useful in the methods of the invention may be evaluated
by the FRET assay described in Example 16. This assay was used on a variety of
commercially available compound libraries to assess the specific inhibition
formation of
soluble amyloid 01_42 oligomers of each compound. Lead compounds were
identified and
further compound libraries were obtained. In addition, structure activity
relationships
around lead compounds were conducted leading to still further enhancements in
activity.
-69-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
D. Synthesis of Compounds of the Invention

The compounds of this invention can be prepared from readily available
starting materials using the following general methods and procedures. It will
be
appreciated that where typical or preferred process conditions (i.e., reaction
temperatures,
times, mole ratios of reactants, solvents, pressures, etc.) are given, other
process conditions
can also be used unless otherwise stated. Optimum reaction conditions may vary
with the
particular reactants or solvent used, but such conditions can be determined by
one skilled in
the art by routine optimization procedures.

Additionally, as will be apparent to those skilled in the art, conventional
protecting groups may be necessary to prevent certain functional groups from
undergoing
undesired reactions. Suitable protecting groups for various functional groups
as well as
suitable conditions for protecting and deprotecting particular functional
groups are well
known in the art. For example, numerous protecting groups are described in T.
W. Greene
and G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley,
New
York, 1991, and references cited therein.

Furthermore, the compounds of this invention may contain one or more
chiral centers. Accordingly, if desired, such compounds can be prepared or
isolated as pure
stereoisomers, i.e., as individual enantiomers or diastereomers, or as
stereoisomer-enriched
mixtures. All such stereoisomers (and enriched mixtures) are included within
the scope of
this invention, unless otherwise indicated. Pure stereoisomers (or enriched
mixtures) may
be prepared using, for example, optically active starting materials or
stereoselective reagents
well-known in the art. Alternatively, racemic mixtures of such compounds can
be separated
using, for example, chiral column chromatography, chiral resolving agents and
the like.

Compounds of the invention were synthesized according to Examples
provided below.

D. Testing and Administration

Effective doses of the compositions of the present invention, for the
treatment of the above described diseases, vary depending upon may different
factors,
including means of administration, physiological state of the patient, whether
the patient is
-70-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
human or an animal, other medications administered, and whether treatment is
prophylactic
or therapeutic. Usually, the patient is a human, but in certain embodiments, a
patient is an
animal, particularly an animal selected from a mammalian species including
rat, rabbit,
bovine, ovine, porcine, canine, feline, murine, equine, and primate.

The compounds can be administered on multiple occasions, wherein
intervals between single dosages can be daily, weekly, monthly, or yearly.
Intervals can
also be irregular as indicated by measuring blood levels of A(3i_42 protein or
ADDLs, or
ADDL complexes in the patient. Alternatively, one or more of the compounds of
the
invention can be administered as a sustained-release formulation, in which
case less
frequent administration is required. Dosage and frequency may vary depending
on the half-
life of the compounds of the invention. In therapeutic applications, a
relatively high dosage
at relatively short intervals is sometimes required until progression of the
disease is reduced
or terminated, and sometimes until the patient shows partial or complete
amelioration of
symptoms of the disease. Thereafter, the patient can be administered a
prophylactic regime.

Administration of a pharmaceutical composition of on the compounds
described herein can be carried out via a variety of routes including, but are
not limited to,
oral, topical, pulmonary, rectal, subcutaneous, intradermal, intranasal,
intracranial,
intramuscular, intraocular, or intra-articular injection and the like. The
most typical route of
administration is oral, although other routes can be equally effective.

One or more compounds described herein can optionally be administered in
combination with other biological or chemical agents that are at least partly
effective in
treatment of a A(31_42 associated disease. An example of such an agent is, but
are not limited
to, A(3i_42 targeted antibodies as described in International Application
Nos.: WO
2003/253673; WO 2006/014478, US Patent No. 2,489,195, US Publication No. 2007-
0048312, and US Application No. 11/571,532, which are incorporated herein by
reference.
The compounds described herein may be administered to a patient in an
amount sufficient to inhibit, regulate and/or modulate the formation of
neurotoxic ADDLs
or the activity of such ligands in said patient. A skilled clinician would be
able to readily
ascertain appropriate amounts of the compounds described here to effectively
inhibit,
regulate and/or modulate the formation of neurotoxic ADDLs or the activity of
such ligands
-71-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
in said patient. Contemplated amounts of the compounds described herein
include for
example, but are not limited to, from about 0.05 to 2000 mg/m2 /day of one
compound or
more than one compound.

As noted above, the compounds described herein may be administered for
example, but are not limited to, orally, topically, pulmonaryly, rectally,
subcutaneously,
intradermally, intranasally, intracranially, intramuscularly, intraocularly,
or intra-arterially
and the like. The carrier or excipient or excipient mixture can be a solvent
or a dispersive
medium containing for example, but are not limited to, various polar or non-
polar solvents,
suitable mixtures thereof, or oils. As used herein "carrier" or "excipient"
means a
pharmaceutically acceptable carrier or excipient and includes any and all
solvents,
dispersive agents or media, coating(s), antimicrobial agents,
iso/hypo/hypertonic agents,
absorption-modifying agents, and the like. The use of such substances and the
agents for
pharmaceutically active substances is well known in the art. Moreover, other
or
supplementary active ingredients can also be incorporated into the final
composition.
Diseases which are treated by the methods described herein include
Alzheimer's disease, Down's syndrome, stroke, mild cognitive impairment, focal
ischemia
associated dementia and neuronal degeneration.

E. Pharmaceutical Formulations and Routes of Administration
When employed as pharmaceuticals, the compounds of this invention are
usually administered in the form of pharmaceutical compositions. These
compounds can be
administered by a variety of routes including oral, topical, pulmonary,
rectal, subcutaneous,
intradermal, intranasal, intracranial, intramuscular, intraocular, or intra-
articular injection.
These compounds are effective as both injectable and oral compositions. Such
compositions are prepared in a manner well known in the pharmaceutical art and
comprise
at least one active compound.

This invention also includes pharmaceutical compositions which contain, as
the active ingredient, one or more of the compounds described herein
associated with
pharmaceutically acceptable carriers. In making the compositions of this
invention, the
active ingredient is usually mixed with an excipient, diluted by an excipient
or enclosed

-72-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
within such a carrier which can be in the form of a capsule, sachet, paper or
other container.
The excipient employed is typically an excipient suitable for administration
to patient.
When the excipient serves as a diluent, it can be a solid, semi-solid, or
liquid material,
which acts as a vehicle, carrier or medium for the active ingredient. Thus,
the compositions
can be in the form of tablets, pills, powders, lozenges, sachets, cachets,
elixirs, suspensions,
emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium),
ointments
containing, for example, up to 10% by weight of the active compound, soft and
hard gelatin
capsules, suppositories, sterile injectable solutions, and sterile packaged
powders.

In preparing a formulation, it may be necessary to mill the active compound
to provide the appropriate particle size prior to combining with the other
ingredients. If the
active compound is substantially insoluble, it ordinarily is milled to a
particle size of less
than 200 mesh. If the active compound is substantially water soluble, the
particle size is
normally adjusted by milling to provide a substantially uniform distribution
in the
formulation, e.g. about 40 mesh.

Some examples of suitable excipients include lactose, dextrose, sucrose,
sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates,
tragacanth, gelatin,
calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose,
water, syrup,
and methyl cellulose. The formulations can additionally include: lubricating
agents such as
talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and
suspending
agents; preserving agents such as methyl- and propylhydroxy-benzoates;
sweetening agents;
and flavoring agents. The compositions of the invention can be formulated so
as to provide
quick, sustained or delayed release of the active ingredient after
administration to the patient
by employing procedures known in the art.

Administration of therapeutic agents by intravenous formulation is well
known in the pharmaceutical industry. An intravenous formulation should
possess certain
qualities aside from being just a composition in which the therapeutic agent
is soluble. For
example, the formulation should promote the overall stability of the active
ingredient(s),
also, the manufacture of the formulation should be cost effective. All of
these factors
ultimately determine the overall success and usefulness of an intravenous
formulation.

-73-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Other accessory additives that may be included in pharmaceutical
formulations of compounds of the present invention as follow: solvents:
ethanol, glycerol,
propylene glycol; stabilizers: ethylene diamine tetraacetic acid (EDTA),
citric acid;
antimicrobial preservatives: benzyl alcohol, methyl paraben, propyl paraben;
buffering
agents: citric acid/sodium citrate, potassium hydrogen tartrate, sodium
hydrogen tartrate,
acetic acid/sodium acetate, maleic acid/sodium maleate, sodium hydrogen
phthalate,
phosphoric acid/potassium dihydrogen phosphate, phosphoric acid/disodium
hydrogen
phosphate; and tonicity modifiers: sodium chloride, mannitol, dextrose.

The presence of a buffer may be necessary to maintain the aqueous pH in the
range of from about 4 to about 8 and more preferably in a range of from about
4 to about 6.
The buffer system is generally a mixture of a weak acid and a soluble salt
thereof, e.g.,
sodium citrate/citric acid; or the monocation or dication salt of a dibasic
acid, e.g.,
potassium hydrogen tartrate; sodium hydrogen tartrate, phosphoric
acid/potassium
dihydrogen phosphate, and phosphoric acid/disodium hydrogen phosphate.

The amount of buffer system used is dependent on (1) the desired pH; and
(2) the amount of drug. Generally, the amount of buffer used is in a 0.5:1 to
50:1 mole ratio
of buffer:drug (where the moles of buffer are taken as the combined moles of
the buffer
ingredients, e.g., sodium citrate and citric acid) of formulation to maintain
a pH in the range
of 4 to 8 and generally, a 1:1 to 10:1 mole ratio of buffer (combined) to drug
present is used.

One useful buffer in the invention is sodium citrate/citric acid in the range
of
5 to 50 mg per mL of sodium citrate to 1 to 15 mg per mL of citric acid,
sufficient to
maintain an aqueous pH of 4-6 of the composition.

The buffer agent may also be present to prevent the precipitation of the drug
through soluble metal complex formation with dissolved metal ions, e.g., Ca,
Mg, Fe, Al,
Ba, which may leach out of glass containers or rubber stoppers or be present
in ordinary tap
water. The agent may act as a competitive complexing agent with the drug and
produce a
soluble metal complex leading to the presence of undesirable particulates.

In addition, the presence of an agent, e.g., sodium chloride in an amount of
about of 1-8 mg/mL, to adjust the tonicity to the same value of human blood
may be

-74-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
required to avoid the swelling or shrinkage of erythrocytes upon
administration of the
intravenous formulation leading to undesirable side effects such as nausea or
diarrhea and
possibly to associated blood disorders. In general, the tonicity of the
formulation matches
that of human blood which is in the range of 282 to 288 mOsm/kg, and in
general is 285
mOsm/kg , which is equivalent to the osmotic pressure corresponding to a 0.9%
solution of
sodium chloride.

The intravenous formulation can be administered by direct intravenous
injection, i.v. bolus, or can be administered by infusion by addition to an
appropriate
infusion solution such as 0.9% sodium chloride injection or other compatible
infusion
solution.

The compositions can be formulated in an oral unit dosage form. The term
"unit dosage forms" refers to physically discrete units suitable as unitary
dosages for a
patient, each unit containing a predetermined quantity of active material
calculated to
produce the desired therapeutic effect, in association with a suitable
pharmaceutical
excipient.

The active compound is effective over a wide dosage range and is generally
administered in a pharmaceutically effective amount. It, will be understood,
however, that
the amount of the compound actually administered will be determined by a
physician, in the
light of the relevant circumstances, including the condition to be treated,
the chosen route of
administration, the actual compound administered, the age, weight, and
response of the
individual patient, the severity of the patient's symptoms, and the like. One
of skill in the
art, based on Example 17, would be able to readily assess the appropriate
concentration of
active compound to provide the necessary cognitive restoration.

For preparing solid compositions such as tablets, the principal active
ingredient is mixed with a pharmaceutical excipient to form a solid
preformulation
composition containing a homogeneous mixture of a compound of the present
invention.
When referring to these preformulation compositions as homogeneous, it is
meant that the
active ingredient is dispersed evenly throughout the composition so that the
composition
may be readily subdivided into equally effective unit dosage forms such as
tablets, pills and
capsules. This solid preformulation is then subdivided into unit dosage forms
of the type
-75-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
described above containing from, for example, 0.05 to about 2000 mg of the
active
ingredient of the present invention.

The tablets or pills of the present invention may be coated or otherwise
compounded to provide a dosage form affording the advantage of prolonged
action. For
example, the tablet or pill can comprise an inner dosage and an outer dosage
component, the
latter being in the form of an envelope over the former. The two components
can be
separated by an enteric layer which serves to resist disintegration in the
stomach and permit
the inner component to pass intact into the duodenum or to be delayed in
release. A variety
of materials can be used for such enteric layers or coatings, such materials
including a
number of polymeric acids and mixtures of polymeric acids with such materials
as shellac,
cetyl alcohol, and cellulose acetate.

The liquid forms in which the novel compositions of the present invention
may be incorporated for administration orally or by injection include aqueous
solutions
suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions
with edible
oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well
as elixirs and
similar pharmaceutical vehicles.

Compositions for inhalation or insufflation include solutions and suspensions
in pharmaceutically acceptable, aqueous or organic solvents, or mixtures
thereof, and
powders. The liquid or solid compositions may contain suitable
pharmaceutically
acceptable excipients as described supra. Preferably the compositions are
administered by
the oral or nasal respiratory route for local or systemic effect. Compositions
in preferably
pharmaceutically acceptable solvents may be nebulized by use of inert gases.
Nebulized
solutions may be breathed directly from the nebulizing device or the
nebulizing device may
be attached to a face masks tent, or intermittent positive pressure breathing
machine.
Solution, suspension, or powder compositions may be administered, preferably
orally or
nasally, from devices which deliver the formulation in an appropriate manner.

The following formulation examples illustrate the contemplated
pharmaceutical compositions of the present invention.

-76-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Formulation Example 1

Hard gelatin capsules containing the following ingredients are prepared:
Ingredient Quantity (mg/capsule)
Active Ingredient 30.0
Starch 305.0
Magnesium stearate 5.0

The above ingredients are mixed and filled into hard gelatin capsules in 340
mg quantities.

Formulation Example 2

A tablet formula is prepared using the ingredients below:
Ingredient Quantity (mg/tablet)
Active Ingredient 25.0
Cellulose, microcrystalline 200.0
Colloidal silicon dioxide 10.0
Stearic acid 5.0

The components are blended and compressed to form tablets, each weighing
240 mg.

Formulation Example 3

A dry powder inhaler formulation is prepared containing the following
components:

Ingredient Weight %
Active Ingredient 5
Lactose 95
The active ingredient is mixed with the lactose and the mixture is added to a
dry powder inhaling appliance.

Formulation Example 4

Tablets, each containing 30 mg of active ingredient, are prepared as follows:
-77-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Ingredient Quantity (mg/tablet)
Active Ingredient 30.0 mg
Starch 45.0 mg
Microcrystalline cellulose 35.0 mg
Polyvinylpyrrolidone 4.0 mg
(as 10% solution in sterile water)
Sodium carboxymethyl starch 4.5 mg
Magnesium stearate 0.5 mg
Talc 1.0 mg

The active ingredient, starch, and cellulose are passed through a No. 20 mesh
U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed
with the
resultant powders, which are then passed through a 16 mesh U.S. sieve. The
granules so

produced are dried at 50 C to 60 C and passed through a 16 mesh U.S. sieve.
The sodium
carboxymethyl starch, magnesium stearate, and talc, previously passed through
a No. 30
mesh U.S. sieve, are then added to the granules which, after mixing, are
compressed on a
tablet machine to yield tablets each weighing 120 mg.

Formulation Example 5

Capsules, each containing 40 mg of medicament are made as follows:
Ingredient Quantity (mg/capsule)
Active Ingredient 40.0 mg
Starch 109.0 mg
Magnesium stearate 1.0 mg
Total 150.0 mg

The active ingredient, starch and magnesium stearate are blended, passed
through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150
mg quantities.
Formulation Example 6

Suppositories, each containing 25 mg of active ingredient are made as
follows:

Ingredient Amount
Active Ingredient 25 mg
Saturated fatty acid glycerides 2,000 mg
-78-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The active ingredient is passed through a No. 60 mesh U.S. sieve and
suspended in the saturated fatty acid glycerides previously melted using the
minimum heat
necessary. The mixture is then poured into a suppository mold of nomina12.0 g
capacity
and allowed to cool.

Formulation Example 7

Suspensions, each containing 50 mg of medicament per 5.0 ml dose are
made as follows:

Ingredient Amount
Active Ingredient 50.0 mg
Xanthan gum 4.0 mg
Sodium carboxymethyl cellulose
(11%)
Microcrystalline cellulose (89%) 50.0 mg
Sucrose 1.75 g
Sodium benzoate 10.0 mg
Flavor and Color q.s.
Purified water to 5.0 mL

The active ingredient, sucrose and xanthan gum are blended, passed through
a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of
the
microcrystalline cellulose and sodium carboxymethyl cellulose in water. The
sodium
benzoate, flavor, and color are diluted with some of the water and added with
stirring.
Sufficient water is then added to produce the required volume.

Formulation Example 8

Ingredient Quantity
(mg/capsule)
Active Ingredient 15.0 mg
Starch 407.0 mg
Magnesium stearate 3.0 mg
Total 425.0 mg
The active ingredient, starch, and magnesium stearate are blended, passed
through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in
425.0 mg
quantities.

-79-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Formulation Example 9

A subcutaneous formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 5.0 mg
Corn Oil 1.0 mL
Formulation Example 10

An intravenous formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 250 mg
Isotonic saline 1000 mL

Another formulation employed in the methods of the present invention
employs transdermal delivery devices ("patches"). Such transdermal patches may
be used
to provide continuous or discontinuous infusion of the compounds of the
present invention
in controlled amounts. The construction and use of transdermal patches for the
delivery of
pharmaceutical agents is well known in the art. See, e.g., U.S. Patent
5,023,252, issued
June 11, 1991, herein incorporated by reference. Such patches may be
constructed for
continuous, pulsatile, or on demand delivery of pharmaceutical agents.

Frequently, it will be desirable or necessary to introduce the pharmaceutical
composition to the brain, either directly or indirectly. Direct techniques
usually involve
placement of a drug delivery catheter into the host's ventricular system to
bypass the
blood-brain barrier. One such implantable delivery system used for the
transport of
biological factors to specific anatomical regions of the body is described in
U.S. Patent
5,011,472, which is herein incorporated by reference.

Indirect techniques, usually involve formulating the compositions to provide
for drug latentiation by the conversion of hydrophilic drugs into lipid-
soluble drugs.
Latentiation is generally achieved through blocking of the hydroxyl, carbonyl,
sulfate, and
primary amine groups present on the drug to render the drug more lipid soluble
and
amenable to transportation across the blood-brain barrier. Alternatively, the
delivery of

-80-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic
solutions, which
can transiently open the blood-brain barrier.

Other suitable formulations for use in the present invention can be found in
Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia,
PA, 17th
ed. (1985).

As noted above, the compounds described herein are suitable for use in a
variety of drug delivery systems described above. Additionally, in order to
enhance the in
vivo serum half-life of the administered compound, the compounds may be
encapsulated,
introduced into the lumen of liposomes, prepared as a colloid, or other
conventional
techniques may be employed which provide an extended serum half-life of the
compounds.
A variety of methods are available for preparing liposomes, as described in,
e.g., Szoka, et
al., U.S. Patent Nos. 4,235,871, 4,501,728 and 4,837,028 each of which is
incorporated
herein by reference.

In prophylactic applications, compositions are administered to a patient at
risk of developing AD (determined for example by genetic screening or familial
trait) in an
amount sufficient to inhibit the onset of symptoms of the disease. An amount
adequate to
accomplish this is defined as "prophylactically effective dose." Amounts
effective for this
use will depend on the judgment of the attending clinician depending upon
factors such as
the age, weight and general condition of the patient, and the like.

As noted above, the compounds administered to a patient are in the form of
pharmaceutical compositions described above. These compositions may be
sterilized by
conventional sterilization techniques, or may be sterile filtered. The
resulting aqueous
solutions may be packaged for use as is, or lyophilized, the lyophilized
preparation being
combined with a sterile aqueous carrier prior to administration. The pH of the
compound
preparations typically will be between 3 and 11, more preferably from 5 to 9
and most
preferably from 7 and 8. It will be understood that use of certain of the
foregoing excipients,
carriers, or stabilizers will result in the formation of pharmaceutical salts.

-81-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
The following examples are offered to illustrate this invention and are not to
be construed in any way as limiting the scope of this invention. Unless
otherwise stated, all
temperatures are in degrees Celsius.

EXAMPLES
The invention is further understood by reference to the following examples,
which are intended to be purely exemplary of the invention. The present
invention is not
limited in scope by the exemplified embodiments, which are intended as
illustrations of
single aspects of the invention only. Any methods that are functionally
equivalent are
within the scope of the invention. Various modifications of the invention in
addition to
those described herein will become apparent to those skilled in the art from
the foregoing
description and accompanying figures. Such modifications fall within the scope
of the
appended claims.

In these examples and elsewhere, abbreviations have the following
meanings:

L or 1 = microliter
m = micrometer
AcOH = acetic acid
DCM = dichloromethane
DME ethylene glycol dimethyl ether
DMSO = dimethylsulfoxide
EtOH = ethanol
eq. = equivalents
g = gram
h = hour
HC1 = hydrochloric acid
Hz = Hertz
LC = liquid chromatography
LiHMDS lithium hexamethyldisilazide
M = molar
MeOH = methanol
mg = milligram
MgS04 = magnesium sulfate
min = minute
mL = milliliter
mM = millimolar
mm = millimeter
mmol = millimolar
-82-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
mol = moles
MS = mass Spectroscopy
N = normal
nm = nanometer
NMR = nuclear magnetic resonance
M = micromolar
Si02 = silicon dioxide
TLC = thin layer chromatography
TMS = trimethylsilyl
THF = tetrahydrofuran
RT = room temperature
OMe = methoxy
Me = methyl
Pd(dppf)C12 = l,l'-
Bis(diphenylphosphino)ferrocene] dichloropalladium(II)
Compounds of the invention are characterized by assessing melting points,
mass spectroscopy, and/or nuclear magnetic resonance. Melting points are
measured on a
Fisher-Johns Melting point apparatus (Fisher cat 12-144) and are uncorrected.
Mass
spectrometric analysis is performed on an Agilent LC/MSD Multimode Instrument
using
the following parameters:

Source: ESI or APCI positive or negative mode, as noted for individual
samples.
Sample Prep: Typically 1 mg/mL in 1:4 DMSO/MeOH

Chromatography: Injection size 5 L
Column: Diazem-Pfarma C 18-A
5 m,4.6x50mm

Solvents: A = H20 with 0.1 % Formic Acid modifier; B = Acetonitrile with 0.1 %
Formic Acid modifier

Flow rate: 1.0 mL/min
Gradient:

-83-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Time min Solvent Composition
Oto0.3 90%A/10%B
0.3 to 2.5 90%A/10%Bto10%A/90%B
2.5to4.5 10%A/90%B
4.5to4.7 10% A/ 90 % B to 90% A /10 % B
5.7to6.0 90%A/10%B

Nuclear Magnetic resonance (NMR) analysis is performed with a Varian 400
MHz machine. The spectral reference is either TMS or the known chemical shift
of the
solvent. Some compounds are run at elevated temperature (i.e., 75 C) to
promote increased
sample solubility.

Example 1

Preparation of hydrazide starting materials
AuOCH
II 3 + NH2NH2.H2O Ay NHNH2
0 0
1-1 1-2 1-3

General Procedure: An ester 1-1 (l0mmol) was treated with hydrazine
hydrate 1-2 (50 mmol) in refluxing 2-methoxyethanol until the reaction was
completed as
determined by TLC. The reaction mixture was cooled and water was added. The
solution
was concentrated until a precipitate formed. The precipitate was collected by
vacuum
filtration and washed with water, hexane, and ether-ethano19:1 and air dried
to afford the
Compound 1-3.

2-Hydroxy-5-nitrobenzohydrazide (1-4)

In analogy to the general procedure of Example 1, methyl 2-hydroxy-5-
nitrobenzoate (10 mmol) was treated with hydrazine hydrate (50 mmol) in
refluxing 2-
methoxyethanol for 30 minutes. The reaction mixture was cooled whereupon a
precipitate
formed. The precipitate was collected by vacuum filtration and washed with
ether-ethanol
-84-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
9:1, water, and ether and air dried to afford Compound 1-4. LC-MS (APCI neg):
196 (M-
H ).

2-Hydroxy-5-methoxybenzohydrazide (1-5)

Prepared from methyl 2-hydroxy-5-methoxy-benzoate in analogy to the
general procedure of Example 1. LC-MS (APCI neg): 196 (M-H+).
2-Fluoro-6-hydroxybenzohydrazide (1-6)

Prepared from methyl 2-fluoro-6-hydroxy -benzoate in analogy to the
general procedure of Example 1. Structure was confirmed by iH NMR (DMSO-d6).
2-Hydroxy-4-(1H-pyrrol-l-yl)benzohydrazide (1-7)

Prepared from methyl2-hydroxy-4-(1H-pyrrol-l-yl)benzoate in analogy to
the general procedure of Example 1. LC-MS (APCI pos): 218.1 (M+H+).
3-Fluoro-6-hydroxybenzohydrazide (1-8)

Prepared from methyl 5-fluoro-2-hydroxybenzoate in analogy to the general
procedure of Example 1. LCMS (APCI neg): 169 (M-H+).

4-Fluoro-6-hydroxybenzohydrazide (1-9)

Prepared from methyl 4-fluoro-2-hydroxybenzoate in analogy to the general
procedure of Example 1. LCMS (APCI neg): 169 (M-H+).
2-Hydroxy-5-methylbenzohydrazide (1-10)

Prepared from methyl 5-fluoro-2-hydroxybenzoate in analogy to the general
procedure of Example 1. LCMS (APCI pos): 167 (M+H+).
2-Oxindoline-7-carbohydrazide (1-11)

Prepared from methyl oxindole-7-carboxylate in analogy to the general
procedure of Example 1. LC-MS (APCI neg): 190 (M-H+).

-85-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
1-Hydroxy-2-naphthohydrazide (1-12)

Prepared from methyl 1-hydroxy-2-naphthoate in analogy to the general
procedure of Example 1. Structure was confirmed by iH NMR (DMSO-d6).
3-Hydroxy-2-naphthohydrazide (1-13)

Prepared from methyl 3-hydroxy-2-naphthoate in analogy to the general
procedure of Example 1. Structure was confirmed by iH NMR (DMSO-d6).
2-Hydroxy-l-naphthohydrazide (1-14)

Prepared from methyl2-hydroxy-l-naphthoate in analogy to the general
procedure of Example 1. LC-MS (APCI neg): 201.0 (M-H+).

Example 2
N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)methanesulfonamide (5)

,~NHSO2Me
H
/ N, N 0
o I S
F3C ~ ~

Step A - Preparation of Methyl 2-(methylsulfonamido)benzoate (2-3)
N H 2 S02Me
Uly OMe + MeSO2Cl (:~;O
Me
O
O
2-1 2-2 2-3
A solution of methane sulfonyl chloride, 2-2, (0.27 mL, 2.2 mmol) in
chloroform (1.2 mL) was added dropwise over three minutes to a solution of
inethyl2-
aminobenzoate, 2-1, (0.26 mL, 2 mmol) in pyridine (0.29 mL) and chloroform
(2.6 mL).
After stirring at RT for 30 minutes, the mixture was concentrated in vacuo to
provide a solid

-86-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
which was washed with hexane to give the product, 2-3, as a rose colored solid
(393 mg,
86%). LC-MS (APCI neg): 228.0 (M-H+).

Step B -Preparation of N-(2-(hydrazinecarbonyl)phenyl)methanesulfonamide (9)
I ~ NHS02Me C(; HS O2Me
/ OMe + H2NNH2 01 NHNH2
O O
2-3 2-4 2-5
A solution of 1-3 (380 mg, 1.66 mmol) and hydrazine hydrate (0.26 mL, 6.3
mmol) in 2-methoxy ethanol (1 mL) was heated at reflux. After 30 minutes, the
mixture
was cooled and concentrated in vacuo to afford a transparent oil. The compound
was
crystallized from ethyl acetate. The solid was filtered and washed with ethyl
acetate and
ether and air dried to affored the product, 2-5, as a white solid (259 mg,
68%). LC-MS
(APCI neg): 228.0 (M-H+).

Step C -Preparation of N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-

ylidene)hydrazinecarbonyl)phenyl)methanesulfonamide (5)

NHSO2Me O O ~_IINHSO2Me
+ H
I/ NHNH2 F3C N.
UO
O ~ S
O F3C

2-5 2-6 5
The title compound was prepared from 2-5 (23 mg, 0.10 mmol) and thenoyl
trifluoroacetone, 2-6, (Aldrich, 24.4 mg, 0.11 mmol) in DMSO (0.3 mL) and AcOH
(1

drop) at 60 C for 8 hours. The reaction mixture was cooled to room
temperature, and
water was added dropwise over 1-2 minutes whereupon a thick precipitate
formed. The
precipitates were collected by vacuum filtration, washed with water, EtOH, and
ether and
dried in vacuo to afford Compound 5 (30 mg, 70%) as a yellow solid. LC-MS
(APCI neg):
432.0 (M-H+).

-87-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Example 3
2-Hydroxy-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide (1)
/ OH
H
N,
O
O O
F F F I /

The title compound was prepared from 2-hydroxybenzhydrazide (Alfa, 173
mg, 0.84 mmol), 4,4,4-trifluoro-l-(2-furyl)-1,3-butanedione (Aldrich, 122 mg,
0.8 mmol)
and ethanol (1.6 mL) at 90 C for 8 hours. The reaction mixture was cooled to
RT, EtOH
(0.5 mL) was added, followed by filtration, triturating with EtOH (3x0.5 mL)
and air-drying
to afford a residue which was dissolved in DCM (3 mL), loaded on Si02 (Flash
grade),
eluted with 15% ethyl acetate in hexane to afford Compound 1 (20 mg, 7%) as an
off-white
solid. LC-MS (APCI neg): 339.0 (M-H+), 92% purity.
-88-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Example 4

N'-(4-(benzo [b] thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide (8)

OH
H
N" N O
O I S
F F F

Step A - Preparation of 1-Benzo[b]thiophen-2-yl-4,4,4-trifluoro-butane-1,3-
dione (4-3)
O
"ZZ S O F O O
~ + N~ F S
/ F F L ,N F F \

4-1 4-2 4-3
A 50 mL one-neck flask, equipped with magnetic stir bar was charged with
2-acetylbenzo[b]thiophene, 4-1 (Alfa, 2 mmol, 352 mg) and anhydrous THF
(Aldrich, 10
mL). The solution was cooled under nitrogen to -78 C (dry ice-acetone).
LiHMDS

(Aldrich, 1 M solution in THF, 2.2 mmol, 2.2 mL, 1.1 equiv) was added dropwise
over 2
min and the solution was stirred for 30 min at -78 C. N-
Trifluoroacetylimidazole, 4-2
(Alfa, 2.6 mmol, 0.3 mL, 1.3 equiv) was added over -30 sec and the reaction
was further
stirred at -78 C for 30 min and then 30 min at 0 C. 2% HC1(-10 mL) was added
to the
reaction mixture until pH = 5. The resulting mixture was poured to a solution
of ether-
water (30 mL, 1:1). The organic layer was separated, washed with brine (15
mL), dried
(MgSO4) to afford a slowly crystallizing orange oil. This oil was dissolved in
DCM-hexane,
1:1 (2 mL), loaded on Si02 (Flash, wet in EA-hexane, 1:1), eluted with EA-
hexane, 1:6 to
afford 4-3 (102 mg, 19%) as a brown solid.

-89-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Step B - N'-(4-(benzo[b]thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-

hydroxybenzohydrazide (8)

The title compound was prepared in analogy to Example 3 from 2-
hydroxybenzhydrazide and 4-3 to afford Compound 8 (19 mg, 31%) as a greenish
solid.
LC-MS (APCI pos): 407.0 (M+H+).

Example 5
2-Hydroxy-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide
(2)

The title non-proprietary compound was prepared in analogy to Example 3.
LC-MS (APCI pos): 357.0 (M+H+).

2-Hydroxy-N'-(1,1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-ylidene)benzohydrazide
(7)
The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 329.1 (M-H+).

2-Hydroxy-N'-(1,1,1-trifluoro-4-(1-methyl-lH-pyrazol-5-yl)-4-oxobutan-2-
ylidene)benzohydrazide (9)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
pos): 353.5 (M-H+).

3-Chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(5-methylthiophen-2-yl)-4-oxobutan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide (10)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
pos): 463 (M+H+).

4-Methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1,2,3-
thiadiazole-
5-carbohydrazide (11)

The title commercially available compound was prepared in analogy to
Example 3. LC-MS (APCI pos): 361 (M+H+).

-90-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
3-Chloro-6-fluoro-N'-(1,1,1-trifluoro-5,5-dimethyl-4-oxohexan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide (12)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 421 (M-H+).

2-Hydroxy-N'-(1,1,1-trifluoro-4-(5-methylthiophen-2-yl)-4-oxobutan-2-
ylidene)benzohydrazide (13)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
pos): 369 (M-H+).

3-Chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo[b]thiophene-2-carbohydrazide (14)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 428.9 (M-H+).

3-Chloro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-ylidene)benzo [b]
thiophene-2-
carbohydrazide (15)

The title compound was prepared in analogy to Example 3. LC-MS (ESI
neg): 413.5 (M-H+).

4-Chloro-N-(2-(2-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)hydrazinecarbonyl)phenyl)benzenesulfonamide (16)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 528.0 (M-H+).

N'-(4-(benzo [b] thiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-3-chloro-
6-
fluorobenzo[b]thiophene-2-carbohydrazide (17)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
pos): 499 (M+H+).

-91-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(4-(5-chlorothiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide(19)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
pos): 389 (M-H+).

5-Chloro-2-hydroxy-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide (20)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 389 (M-H+).

3-Chloro-4-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)thiophene-2-carbohydrazide (21)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 392.9 (M-H+).

N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1H-indole-7-
carbohydrazide (22)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 378 (M-H+).

3-Chloro-N'-(4-(5-chlorothiophen-2-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-6-

fluorobenzo[b]thiophene-2-carbohydrazide (23)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 480.9 (M-H+).

3-Chloro-6-fluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzo [b] thiophene-2-carbohydrazide (24)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 446.7/448.7 (M-H+).

2-Hydroxy-3-methyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide (25)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 369 (M-H+).

-92-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
4-Nitro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide
(26)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 384.0 (M).

4-Bromo-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide
(27)
The title commerically available compound was prepared in analogy to
Example 3. LC-MS (ESI neg): 419.2 (M-H+).
N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)benzo [b] thiophene-
2-
carbohydrazide (28)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg) 395 (M-H+).

3-Chloro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-1H-indole-
2-
carbohydrazide (30)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 412.0 (M-H+).

3-Chloro-6-fluoro-N'-(1,1,1-trifluoro-4-(furan-2-yl)-4-oxobutan-2-
ylidene)benzo [b] thiophene-2-carbohydrazide (31)

The title compound was prepared in analogy to Example 3. LC-MS (ESI
neg): 431.4 (M-H+).

3-(2-(2-Hydroxybenzoyl)hydrazono)butanamide (32)

The title compound was prepared in analogy to Example 3 with the
modification of using refluxing isopropyl alcohol instead of EtOH. LC-MS (ESI
neg):
234.1 (M-H+).

1-Phenyl-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-ylidene)-5-
(trifluoromethyl)-1H-pyrazole-4-carbohydrazide (34)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 473 (M-H+).
- 93 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2,6-Difluoro-N'-(1,1,1-trifluoro-4-oxo-4-(thiophen-2-yl)butan-2-
ylidene)benzohydrazide (35)

The title compound was prepared in analogy to Example 3. LC-MS (APCI
neg): 375 (M-H+).

Example 6

3-C hloro-6-fluoro-N'-(4-(furan-2-yl)-4-oxobutan-2-ylidene)benzo [b] thiophene-
2-
carbohydrazide (4)

ci
F \/ I CI 0 0 F Q I H
S NHNH2 + \ 0/ - S N.N O

p 0 101
6-1 6-2 4
A mixture of the 1-(2-furyl)-1,3-butanedione 6-2 (Alfa, 33 mg, 0.22 mmol),
2 3-chloro-6-fluorobenzo[b]thiophene-2-carbohydrazide 6-1 (Oakwood, 49 mg, 0.2
mmol)
in THF (1 to 2 mL) was shaken at 25 oC for 48 hours. The compound was
precipitated with
hexanes to obtain Compound 4 as a yellow solid (41 mg, 54%). LC-MS (APCI pos):
379.0
(M+H+).

Example 7

1-(4-Benzylpiperazin-1-yl)-4,4,4-trifluorobutane-1,3-dione (7-3)
F 0 0 H N 0 0
O
F F ON
7-1 7-2 7-3

A mixture of ethy14,4,4-trifluoroacetate, 7-1, (Aldrich, 2.1 g, 11.3 mmol)
and 1-benzylpiperazine, 7-2, (Aldrich, 2 g, 11.3 mmol) in toluene (2.5 mL) was
shaken for 8
hours at 90 C and 105 C for 8 hours. The solvent was removed in vacuo to
obtain the
o o
crude mixture. The crude mixture was purified by silica gel flash
chromatography to obtain
-94-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
the product (7-3, 2.25 g, 63%). LC-MS (APCI pos): 315 (M + H). Analogous
ketoamides
may be prepared in a similar fashion.

Example 8
N'-(4-(4-benzylpiperazin-1-yl)-1,1,1-trifluoro-4-oxobutan-2-ylidene)-2-
hydroxybenzohydrazide (3)

OH
H
OH O O N'N O
N. + FO O
NH2
2 F F I`F
O F N
8-1 7-3 3

A mixture of dione 7-3 (100 mg, 0.32 mmol) and 2-hydroxybenzhydrazide
(8-1, 48 mg, 0.32 mmol) in DMSO (lmL) and AcOH (15 mg) was shaken at 90 ~C for
overnight. The crude mixture was purified by silica gel flash chromatography
to obtain the
Compound 3 (18 mg, 13%): LC-MS (APCI pos): 449.1 (M+H+).
Example 9
2-Hydroxy-N'-(1,1,1-trifluoro-4-(4-methylpiperazin-1-yl)-4-oxobutan-2-
ylidene)benzohydrazide (6)

The title compound was prepared in analogy to Example 8. LC-MS (APCI
pos): 373.3 (M+H+).

2-Hydroxy-N'-(1,1,1-trifluoro-4-morpholino-4-oxobutan-2-ylidene)benzohydrazide
(18)
The title compound was prepared in analogy to Example 8. LC-MS (APCI
pos): 360.1 (M+H+).

2-Hydroxy-N'-(4-oxo-4-(4-phenylpiperidin-1-yl)butan-2-ylidene)benzohydrazide
(29)
The title compound was prepared in analogy to Example 8. LC-MS (APCI
neg): 378.1 (M-H+).

- 95 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Example 10

A NH2NH2 O Ay N,

0 + RR3 0 R2ll R3
10-1 10-2 10-3
General procedure A. A vial (8-20 mL) equipped with magnetic stir bar,
was charged with hydrazides 10-1 (0.30-4.5 mmol, 1 eq.), aldehydes or ketones
10-2 (1.05-
1.1 eq.) and solvent (2.3 vol % of AcOH in DMSO; 2 mL/1 mmol of 10-1). The
vials were
sealed and reaction mixtures were magnetically stirred at RT until TLC (EA-
hexane, 1:2)
indicated the complete disappearance of starting materials 10-1. Brine was
added dropwise
over 1-2 min and the reaction mixtures were stirred for 5-10 min. The
resulting precipitates
were collected by vacuum filtration, washed with water, hexane, hexane-ether,
1:1, ether
and air-dried to afford Compounds 10-3.

General procedure B. A vial (8 mL) equipped with magnetic stir bar, was
charged with hydrazides 10-1 (0.40 mmol, 1 eq.), aldehydes or ketones 10-2
(1.05 eq.) and
solvent (2.3 vol % of AcOH in DMSO or DMSO; 2 mL/1 mmol of 2-1). The vials
were
sealed and reaction mixtures were magnetically stirred at RT until TLC (EA-
hexane, 1:2)
indicated the complete disappearance of starting materials 10-1. The reaction
was
quenched by dropwise addition of a few drops of sat NaHCO3 or water over 1-2
min. After
reaction mixtures were stirred for 5-15 min, the resulting precipitates were
collected by
vacuum filtration, washed with water, hexane, hexane-ether, 1:1 or ether and
air-dried to
afford Compounds 10-3.

-96-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Example 11
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-4-(1H-pyrrol-l-yl)benzohydrazide
(36)
O\N O\N OH O OH OH
H
NHNH2 I \ \ N.
OH
O O
OMe
1-7 11-1 36 OMe

In analogy to general procedure A of Example 10, the title compound was
prepared from 2-hydroxybenzhydrazide 1-7 (65 mg, 0.30 mmol), 2-hydroxy-5-
methoxybenzaldehyde, 3-2 (Aldrich, 50 mg, 0.33 mmol) and solvent (2.3 vol % of
AcOH in
DMSO, 0.6 mL). The vial was sealed and reaction mixture was magnetically
stirred at RT
for 1.5 h until TLC (EA-hexane, 1:2) indicated the complete disappearance of
starting
material 3-1. Brine was added dropwise over 1-2 min and the reaction mixture
was stirred
for 10 min. The resulting precipitate was collected by vacuum filtration,
washed with
water, hexane, ether and air-dried to afford Compound 36 (96 mg, 91%). LC-MS
(APCI
pos): 352.1 (M+H+).

Example 12
3-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-2-naphthohydrazide (37)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 337.1 (M+H+).
1-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-2-naphthohydrazide (38)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 337.1 (M+H+).

-97-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(3-chloro-5-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (40)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 309 (M+H+).

N'-(2,6-dihydroxybenzylidene)-2-hydroxybenzohydrazide (41)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 273.0 (M+H+).
N'-(3,5-dibromo-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (42)

The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 414.9 (M+H+).

3-Chloro-N'-(2-hydroxy-5-methoxybenzylidene)-1H-indole-2-carbohydrazide (44)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 344.0 (M+H+).

N'-(2-bromo-6-hydroxybenzylidene)-2-hydroxybenzohydrazide (45)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 335.2 (M+H+).

N'-(3-chloro-5-cyclohexyl-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (46)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 371.1 (M-H+).

N'-(5-tert-butyl-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (47)
The title compound was prepared in analogy to general procedure A of
Example 10 with additional heating at 60 C for 24 h. LC-MS (APCI pos): 313.1
(M+H+).

N'-(5-bromo-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (50)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 337.0/338.0 (M+H+).
-98-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
4-Chloro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (51)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 319 (M-H+).
N'-(2-hydroxy-5-methoxybenzylidene)-2-oxoindoline-7-carbohydrazide (52)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 324.1 (M-H+).

2-Hydroxy-N'-(2-hydroxy-6-methoxybenzylidene)benzohydrazide (55)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 287.1 (M+H+).

2-Hydroxy-N'-(2-hydroxy-5-(trifluoromethoxy)benzylidene)benzohydrazide (57)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 339 (M-H+).

N'-(2-hydroxy-5-methoxybenzylidene)-1H-indole-7-carbohydrazide (58)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 310.1 (M+H+).

N'-(5-chloro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (59)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 291.0 (M+H+).

3-Chloro-6-fluoro-N'-((2-hydroxynaphthalen-1-yl)methylene)benzo [b] thiophene-
2-
carbohydrazide (61)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 399 (M+H+).

-99-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-3-methylbenzohydrazide (63)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 301.1 (M+H+).
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide (66)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 337.1 (M+H+).

4-Fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (67)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 305 (M+H+).

2-Hydroxy-N'-(2-hydroxy-5-methylbenzylidene)benzohydrazide (68)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 271.1 (M+H+).

N'-(3,5-dichloro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (69)
The title commercially available compound was prepared in analogy to
general procedure A of Example 10. LC-MS (APCI pos): 325.0 (M+H+).

3-Chloro-N'-(2-hydroxy-5-methoxybenzylidene)-4-methylthiophene-2-
carbohydrazide
(70)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos) 325.0 (M+H+).

2-Hydroxy-N'-((1-hydroxynaphthalen-2-yl)methylene)benzohydrazide (72)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos) 307.1 (M+H+).

- 100 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-methylbenzohydrazide (73)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 301.1 (M+H+).
N'-(3-fluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (74)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 275.1 (M+H+).
5-Fluoro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (75)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 305 (M+H+).

2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-4-methoxybenzohydrazide (76)
The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI neg) 315.1 (M-H+).

Ethy16-bromo-5-hydroxy-4-((2-(2-hydroxybenzoyl)hydrazono)methyl)-2-
methylbenzofuran-3-carboxylate (77)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 461/463 (M+H+).

3-(Benzo [d] [ 1,3 ] dioxol-5-yl)-N'-(2-hydroxy-5-methoxybenzylidene)-1H-
pyrazole-5-
carbohydrazide (78)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 381.1 (M+H+).

2-Hydroxy-N'-(2-hydroxy-4-methoxybenzylidene)benzohydrazide (81)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 287.1 (M+H+).

-101-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-methoxybenzohydrazide (82)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 317.1(M+H+).
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (83)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 287.1 (M+H+).
5-Chloro-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (84)

The title non-proprietary compound was prepared in analogy to general
procedure A of Example 10. LC-MS (APCI pos): 321.1 (M+H+).

N'-(5-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (85)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 299.1 (M-H+).

N'-(2,3-dihydroxybenzylidene)-2-hydroxybenzohydrazide (86)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 273.1 (M+H+).

2-Hydroxy-N'-((3-hydroxy-5-nitrobenzofuran-2-yl)methylene)benzohydrazide (88)
The title compound was prepared in analogy to general procedure A of
Example 10.

2-Hydroxy-N'-((2-hydroxynaphthalen-1-yl)methylene)benzohydrazide (89)
The title commercially available compound was prepared in analogy to
general procedure A of Example 10. LC-MS (APCI pos): 307.1 (M+H+).

N'-(2,4-dihydroxybenzylidene)-2-hydroxybenzohydrazide (90)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 273.0(M+H+).
- 102 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(5-chloro-2-hydroxy-3-methoxybenzylidene)-2-hydroxybenzohydrazide (91)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 321 (M+H+).
3-Chloro-N'-(5-chloro-2-hydroxybenzylidene)-4-methylthiophene-2-carbohydrazide
(92)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 328.9 (M+H+).

2-Amino-N'-(2-amino-5-chlorobenzylidene)benzohydrazide (93)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 289.0 (M+H+).

2-Hydroxy-N'-(2-hydroxy-3-methoxybenzylidene)benzohydrazide (94)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 287.1 (M+H+).

2-Fluoro-6-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (95)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 305.1 (M+H+).

2-Hydroxy-N'-(2-hydroxy-3-methylbenzylidene)benzohydrazide (96)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 271.1 (M+H+).

N'-((2-hydroxynaphthalen-1-yl)methylene)-3-methyl-lH-pyrazole-5-carbohydrazide
(97)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 295.1 (M+H+).
-103-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
5-Bromo-2-hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (98)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 365 (M+H+).
2-Hydroxy-N'-(2-hydroxy-5-nitrobenzylidene)benzohydrazide (99)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 300 (M-H+).

N-(2-(2-(2-hydroxy-5-
methoxybenzylidene)hydrazinecarbonyl)phenyl)methanesulfonamide (100)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 364.0 (M+H+).

N'-(3,5-difluoro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (101)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 293 (M+H+).

N'-(1-(5-chloro-2-hydroxyphenyl)-2,2,2-trifluoroethylidene)-2-
hydroxybenzohydrazide
(102)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 357.0 (M-H+).
2-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-5-nitrobenzohydrazide (104)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 330 (M-H+).
8-Hydroxy-N'-(2-hydroxy-5-methoxybenzylidene)-1-naphthohydrazide (105)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 335.1 (M-H+).

- 104 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
N'-(3-ethoxy-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (106)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 301.1 (M+H+).

3-(5-Chlorothiophen-2-yl)-N'-(2-hydroxy-5-methoxybenzylidene)-1H-pyrazole-5-
carbohydrazide (107)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 377.0 (M+H+).

N'-(3-bromo-5-chloro-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (108)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 369.9 (M+H+).

N'-(3-bromo-2-hydroxy-5-methoxybenzylidene)-2-hydroxybenzohydrazide (109)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 366.9 (M+H+).

2-Amino-N'-(2-hydroxy-5-methoxybenzylidene)benzohydrazide (110)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI pos): 286.1 (M+H+).
N'-(5-Chloro-2-hydroxybenzylidene)-4-methyl-1,2,3-thiadiazole-5-carbohydrazide
(111)
The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 295 (M-H+).

N-(4-chloro-2-((2-(2-hydroxybenzoyl)hydrazono)methyl)phenyl)-2,4-
difluorobenzenesulfonamide (114)

The title compound was prepared in analogy to general procedure A of
Example 10. LC-MS (APCI neg): 464 (M-H+).

-105-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Example 13
2-Hydroxy-N'-((4-hydroxy-3'-methoxybiphenyl-3-yl)methylene)benzohydrazide (48)
OH
H
N.
N OH
O I \

OMe
Step A - Preparation of 4-Hhydroxy-3'-methoxybiphenyl-3-carbaldehyde (13-3)
0 OH
0 OH B(OH)2
+ I
OMe
Br
OMe
13-1 13-2 13-3

A mixture of 5-bromosalicylaldehyde, 13-1, (Aldrich, 568 mg, 2.8 mmol), 3-
methoxyphenyl boronic acid, 13-2, (Aldrich, 472 mg, 3.1 mmol) and a 3:1
mixture of DME
and water (4.8 mL) was bubbled with nitrogen for 10 minutes. Sodium carbonate
(448 mg,
4.2 mmol) was added to the mixture, followed by Pd(dppf)C12 (Aldrich, 115 mg,
0.14
mmol). Nitrogen was bubbled through the mixture for 5 minutes and the reaction
was
heated at reflux for 19 hours. (LC-MS (APCI neg) 227.0 (M-H+). Water was added
and the
mixture was shaken and extracted with DCM. The organic fraction was washed
with water
and brine, dried over MgS04, filtered and concentrated. The material was
purifed by silica
gel chromatography to afford 13-3 (189 mg, 29%) as a yellow oil. LC-MS (APCI
neg)
227.0 (M-H+).

- 106 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Step B - 2-Hydroxy-N'-((4-hydroxy-3'-methoxybiphenyl-3-
yl)methylene)benzohydrazide (48)

/ OH
0 OH \ I H
OH N OH
O
NHNH2

O
\ ~ I
OMe
OMe
13-4 13-3 48

In analogy to general procedure B of Example 10, the title compound was
prepared from 2-hydroxybenzhydrazide 13-4 (30 mg, 0.20 mmol), aldehyde, 13-3
(53 mg,
0.21 mmol) and solvent. The vial was sealed and the reaction mixture was
magnetically
stirred at RT for 30 minutes whereupon a precipitate had 1-2 minutes and the
mixture was
stirred for 15 minutes. The resulting precipitate was collected by vacuum
filtration, washed
with water, hexane, hexane-ether, 1:1 and air-dried to afford Compound 48 (69
mg, 95%)
as a white solid. LC- MS (APCI pos): 363.1 (M+H+)
Example 14
2-((2-(2-Hydroxybenzoyl)hydrazono)methyl)benzamide (64)

The title compound was prepared in analogy to general procedure B of
Example 10. LC- MS (APCI neg): 282.1 (M-H+).

N'-(2-amino-5-chlorobenzylidene)-2-hydroxybenzohydrazide (65)

The title compound was prepared in analogy to general procedure B of
Example 10. LC-MS (APCI pos): 290.0 (M+H+).

N'-(4-(diethylamino)-2-hydroxybenzylidene)-2-hydroxybenzohydrazide (80)
The title compound was prepared in analogy to general procedure B of
Example 10. LC-MS (APCI neg): 326.1 (M-H+).

- 107 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
2-Hydroxy-N'-(2-hydroxy-4-morpholinobenzylidene)benzohydrazide (87)
The title compound was prepared in analogy to general procedure B of
Example 10. LC-MS (APCI pos): 342.1 (M+H+).

N-(2-(2-(2-acetamido-5-chlorobenzylidene)hydrazinecarbonyl)phenyl)acetamide
(113)
The title compound was prepared in analogy to general procedure B of
Example 10. LC-MS (APCI pos): 373.6 (M+H+).

Example 15

The following compounds were obtained from commercial sources.
No. Source
11 TOSLab (Ekaterinburg, Russia)
27 ChemDiv (San Diego, CA)
33 ChemDiv (San Diego, CA)
39 Chembridge (San Diego, CA)
43 Aldrich (Milwaukee, WI)
49 Timtec (Newark, DE)
53 AnaLogix (Burlington, WI)
54 AnaLogix (Burlington, WI)
56 Timtec (Newark, DE)
60 Timtec (Newark, DE)
62 Aldrich (Milwaukee, WI)
69 ChemDiv (San Diego, CA)
71 Aldrich (Milwaukee, WI)
79 Chembridge (San Diego, CA)
89 Chembridge (San Diego, CA)
103 Chembridge (San Diego, CA)
112 Chembridge (San Diego, CA)
115 Aldrich (Milwaukee, WI)
116 AnaLogix (Burlington, WI)
Example 16
FRET Assay

Fluorescence (or F6rster) Resonance Energy Transfer (FRET) is a distance-
dependent, non-radiative transfer of energy in which the de-excitation of one
fluorophore
(donor) is coupled to excitation of another fluorophore (acceptor). FRET
occurs if (1) the
quantum of energy emitted by a donor fluorophore corresponds to an acceptor
fluorophore's

-108-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
excitation energy, (2) the orientations of donor and acceptor transition
dipoles are nearly
parallel and (3) the donor fluorescent emission spectrum overlaps the acceptor
absorption
spectrum.

In this assay, assembly of A(3i_42 oligomer formation is monitored by FRET
using N-terminal conjugates of fluorescein-A(31_42 as both the donor and
acceptor
fluorophore (fluorescein-fluorescein R - 45 k). A(31_42 monomers assembling
into
oligomeric species results in a decrease of fluorescence as the fluorescein
labeled A(3i_42
peptides become proximal to each other and FRET efficiency increases.
Inhibition of A(3i_42
assembly is observed as the absence or attenuation of fluorescein quenching.

FRET and FP assays are performed in 384-well Coming Non-Binding
Surface, black, opaque microtiter plates, and the assay buffer consists of 25
mM MOPS-Tris
(pH 8.0) with 100 mM MgC12. The assay volume, containing 0.2 M FITC-A(3(1-42)
and
0.8 M A(3(1-42), is 50 1 and the temperature is 37 C. ADDL assembly is
monitored on a
Tecan GENios Pro plate reader, exciting at a wavelength of 485 nm and
detecting emission
at a wavelength of 515 nm. Kinetic traces are collected by recording
fluorescence intensity
and polarization readings every five minutes over about a three hour time
course. Negative
control reactions, which do not appreciably assemble into ADDLs during this
time, lack
MgC1z but contain all other buffer and peptide components. Positive control
reactions
contain all buffer components in the absence of added small molecule or
antibody reagents.
To test for ADDL assembly inhibition, the compound was incubated with the
peptide
mixture at six concentrations from about 10 M decreasing to about 0.03 M.

Results for the compounds of the invention that were tested in this assay are
shown in Table 2 below.

Table 2
No. IC50
1 4.2
2 4.5

-109-


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. IC50

3 5.2
4 5.9
6
6 7.1
7 7.4
8 7.7
9 8.2
8.3
11 8.4
12 8.6
13 9.1
14 9.5
9.5
16 9.7
17 10.2
18 10.4
19 11.5
11.9
21 12.5
22 13.5
23 16
24 16.1
16.3
26 16.5
27 18.8
28 19.4
29 26.7
28.3
31 30.2
32 35
33 40.8
34 40.9
41.2
36 1
37 1.2
38 1.2
39 1.6
2.1
41 2.3
42 2.8
43 3.2
44 3.2

- 110 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. IC50

45 3.3
46 3.3
47 3.5
48 3.7
49 3.8
50 4.1
51 4.2
52 4.2
53 4.6
54 5
55 5.3
56 6.1
57 6.3
58 6.4
59 6.5
60 6.5
61 6.7
62 6.7
63 7
64 7.1
65 7.5
66 7.7
67 8.3
68 9.2
69 9.2
70 9.4
71 9.7
72 10.4
73 10.4
74 10.9
75 10.9
76 11.1
77 11.6
78 11.6
79 13.3
80 14.4
81 15
82 15.1
83 15.4
84 15.5
85 16
86 17.4

- 111 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
No. IC50

87 18.3
88 20.4
89 20.6
90 21.4
91 22.8
92 23.7
93 23.9
94 24.9
95 26.2
96 28.6
97 30.6
98 31.9
99 32.5
100 33.2
101 33.4
102 34
103 34.5
104 35.1
105 36.5
106 39.2
107 40.1
108 41.7
109 42
110 42.3
111 42.9
112 45.4
113 46
114 46.7
115 47.6
116 50
Example 17

Alternating Lever Cyclic Ratio Rat Model

Preparations of A(3i_42 ADDLs and a potential therapeutic compound under
the Alternating Lever Cyclic Ratio (ALCR) rat model of AD were tested to show
in vivo
efficacy. This highly sensitive model has been able to detect cognitive
deficits due to direct
injection of cell-derived A(3 oligomers into rat brain. Using this technique,
a direct injection

- 112 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
of ADDLs made from synthetic A(3i_42 and a putative therapeutic compound under
the
ALCR procedure were tested.

In this task, rats learned a complex sequence of lever-pressing requirements
in order to earn food reinforcement in a two-lever experimental chamber.
Subjects
alternated between two levers by switching to the other lever after pressing
the first lever
enough to get food reward. The exact number of presses required for each food
reward
changed, first increasing from 2 responses per food pellet up to 56 presses
per food pellet,
then decreasing back to 2 responses per pellet. Intermediate values were based
on the
quadratic function, x2 - x. One cycle was an entire ascending and descending
sequence of
these lever press requirements (e.g., 2, 6, 12, 20, 30, 42, 56, 56, 42, 30,
20, 12, 6, and 2
presses per food reward). Six such full cycles were presented during each
daily session.
Errors were scored when the subject perseveres on a lever after pressing
enough to get the
food reward, i.e., did not alternate (a Perseveration Error), or when a
subject switched levers
before completing the response requirement on that lever (an Approach Error).

Materials and Methods

Synthetic A(3i_42 powder was dissolved in 1, 1, 1,3,3,3 -hexafluorisopropanol
(HFIP) to afford a solution of A(31_42 in HFIP of about 1 mM and allowed to
incubate at
ambient temperature for about 1 h. The resulting solution was chilled on ice
for about 5-10
min, then aliquoted into eppendorf tubes to provide about 50 L of solution
per tube. The
tubes were then placed in a chemical fume hood and allowed to stand overnight
to allow the
HFIP to evaporate under a slow stream of nitrogen. To remove final traces of
HFIP, the
tubes were subjected to two SpeedVac cycles of 15 min at room temperature and
about 15
to 25 mm Hg of vacuum. The resulting films of monomerized A(31_42 peptide were
stored
over desiccant at -80 C until used.

A tube of monomerized A(3i_42 peptide was warmed to room temperature and
the A(31_42 peptide was dissolved in anhydrous DMSO to afford a peptide stock
DMSO
solution containing about 10 M to about 100 M A(31_42 peptide in DMSO.

2 L of a 20 mM stock solution of test compound (Compound 2 and
Compound 89 from Table 1) in anhydrous DMSO was added to an appropriate amount
of
- 113 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
neural basal media (phenol red free, Gibco 12348-017) to provide the requisite
and noted
concentration of the test compound in neural basal media. The concentration of
the test
compound is listed below in Table 3.

For the A(3i_42 peptide only treatment, peptide stock DMSO solution was
added to 37 C neural basal to obtain the requisite A(31_42 peptide monomer
concentration,
provided that the maximum concentration of DMSO is 1% or less, and the tube
was
vortexed for 30 to 60 seconds, spun down briefly in a microfuge and incubated
at 37 C for
min prior to the start of injections.

For the A(31_42 peptide plus test compound treatment, peptide stock DMSO
10 solution was added to 37 C compound neural basal media solution to obtain
the requisite
A(3i_42 peptide monomer concentration, provided that the maximum concentration
of DMSO
is 1% or less, and the tube was vortexed for 30 to 36 seconds, spun down
briefly in a
microfuge and incubated at 37 C for 15 min prior to the start of injections.

For control injections, compound neural basal media solution was incubated
15 at 37 C for 15 min prior to the start of injections.

Rats: Rats are trained under ALCR until their error rates are stable. Once
the rats are placed upon the final ALCR procedure, training sessions are
conducted 7 days
each week until the end of the study.

Surgery: All rats received a single 28 gauge cannula, which was
permanently affixed to the skull, and aimed at the lateral ventricle. Half of
the rats received
cannula in the right ventricle and half receive cannula in the left ventricle.
Rats were
allowed 5 days to recover from surgery before training resumed.

Injection of Test Material and ALCR Testing: Test were conducted about
every fourth day. Animals received a 10-20 L injection of control, peptide,
or peptide plus
compound solutions via the implanted cannula over about 3 to 4 minutes.
Animals were
tested about 3 hours following injection.

- 114 -


CA 02692801 2010-01-07
WO 2009/009768 PCT/US2008/069878
Error Rate Analysis: All error rates were compared to baseline error rates
consisting of at least 3 non-treatment days temporally contiguous to the
injection. Student's
T test of statistical inference was used for analysis of effects.

Results
The preservation errors and approach errors are found in Table 3 below.
Table 3

1nM 1nM
ADDLs ADDLs 1 nM
27 nM + 2 gM + 2 gM ADDLs 2 gM
A(3142 1 nM Cmpd 1 nM Cmpd + 2 gM Cmpd 2 gM
Vehicle Monomer ADDLs 89 ADDLs 89 Cmpd 2 89 Cmpd 2
Perseveration 106% 117% 133% 92% 127% 107% 105% 112% 85%
Errors

Approach 105% 105% 102% 100% 142% 111% 115% 101% 92%
Errors

As can be seen from Table 3, the presence of ADDLs compared to presence
of vehicle or AP1_42 monomer alone, increased the Perseveration error. When
ADDLs were
injected along with either Compound 2 or Compound 89, the Perseveration error
decreased
significantly. It is contemplated that addition of one or more of the
compounds will not
increase or decrease error rates when given alone, but when combined with
ADDLs, will
eliminate the increase in Perseveration Error. Thus, it is contemplated that
at least one or
more of the compounds described herein will reduce errors produced by A(31_42
ADDLs.

From the foregoing description, various modifications and changes in the
compositions and methods will occur to those skilled in the art. All such
modifications
coming within the scope of the appended claims are intended to be included
therein.

- 115 -

Representative Drawing

Sorry, the representative drawing for patent document number 2692801 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2008-07-11
(87) PCT Publication Date 2009-01-15
(85) National Entry 2010-01-07
Examination Requested 2013-06-03
Dead Application 2016-04-07

Abandonment History

Abandonment Date Reason Reinstatement Date
2015-04-07 R30(2) - Failure to Respond
2015-07-13 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2010-01-07
Maintenance Fee - Application - New Act 2 2010-07-12 $100.00 2010-06-11
Maintenance Fee - Application - New Act 3 2011-07-11 $100.00 2011-06-21
Maintenance Fee - Application - New Act 4 2012-07-11 $100.00 2012-06-29
Request for Examination $800.00 2013-06-03
Maintenance Fee - Application - New Act 5 2013-07-11 $200.00 2013-06-25
Maintenance Fee - Application - New Act 6 2014-07-11 $200.00 2014-06-24
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
ACUMEN PHARMACEUTICALS, INC.
Past Owners on Record
BHAGAT, NIKHIL
GOURE, WILLIAM F.
KRAFFT, GRANT A.
LOOK, GARY CHARLES
POLOZOV, ALEXANDRE MIKHAYLOVICH
PRAY, TODD
SCHULTZ, LAURI
WANG, JIAN
ZEMBOWER, DAVID E.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Cover Page 2010-03-30 2 38
Abstract 2010-01-07 1 68
Description 2010-01-07 115 4,448
Claims 2010-01-07 19 735
Abstract 2014-07-24 1 11
Claims 2014-07-24 37 1,438
Description 2014-07-24 115 4,412
Description 2014-09-12 115 4,408
Assignment 2010-01-07 5 145
PCT 2010-01-07 7 271
Correspondence 2010-03-29 1 20
Correspondence 2010-04-07 2 74
Fees 2010-06-11 1 37
Prosecution-Amendment 2013-06-03 2 78
Prosecution-Amendment 2013-07-30 2 80
Prosecution-Amendment 2014-10-07 4 202
Prosecution-Amendment 2014-01-24 6 264
Prosecution-Amendment 2014-07-24 61 2,442
Prosecution-Amendment 2014-09-12 4 118