Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
IAP INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2008/014252
Kind Code:
A2
Abstract:
Smac mimetics that inhibit IAPs.

Inventors:
CONDON STEPHEN M (US)
LAPORTE MATTHEW G (US)
DENG YIJUN (US)
RIPPIN SUSAN R (US)
Application Number:
PCT/US2007/074209
Publication Date:
January 31, 2008
Filing Date:
July 24, 2007
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TETRALOGIC PHARMACEUTICALS COR (US)
CONDON STEPHEN M (US)
LAPORTE MATTHEW G (US)
DENG YIJUN (US)
RIPPIN SUSAN R (US)
International Classes:
C07D401/14; A61K31/4035; A61K38/04; C07D403/06; C07D403/14; C07H19/04; C07K5/06
Domestic Patent References:
WO2006020060A22006-02-23
WO2006091972A22006-08-31
WO2007021825A22007-02-22
Foreign References:
US20050234042A12005-10-20
Attorney, Agent or Firm:
SKERPON, Joseph, M. (1100 13th Street, N.W. Suite 120, Washington DC, US)
Download PDF:
Claims:

What is claimed is;

1. The compound of Formula I:

wherein

Z j and Z 2 are each independently CH or N;

R 1 is H or optionally substituted hydroxy, alkyl,

cycloalkyl, heterocycloalkyl, aryl, or heteroaryl;

R 2 and R 2 ' are each independently H or optionally

substituted alkyl, cycloalkyl, or heterocycloalkyl; or when

R 2 ' is H then R 7 and R 1 can together form an aziridine or

azetidine ring;

R 1 and R 4 are each independently H or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl, or

heteroaryl; or, R 1 and R 4 are each carbon and are linked by

a covalent bond or by an optionally-substituted alkylene or

alkenylene group of 1 to 8 carbon atoms where one to three

R 5 and R 6 are each independently H or optionally

substituted hydroxy, alkyl, cycloalkyl, heterocycloalkyl,

aryl , or heteroaryl; or R 5 and R 6 are each carbon and are

linked by a covalent bond or by an optionally-substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, O, S(O) n , or

C=O;

M is a bond or an optionally substituted alkylene

group of 1 to 5 carbon atoms ;

G is a bond, a heteroatom, -(C=O)-, -S(O) n -, -NR 9 -, -

NCOR 0 -, or -NS(O) n R 6 -, where R 3 is lower alkyl, optionally-

substituted lower alkyl or C ^ a cycloalkyl;

R 7 is optionally substituted alkyl, cycloalkyl,

heterocycloalkyl, aryl, or heteroaryl wherein R 7 is

substituted with -Ll-R 10 and is optionally further

substituted;

Ll is a covalent bond or optionally substituted C 1 . β

alkylene;

R 10 is an optionally substituted 5-, 6-, or 7-merabered

heterocycloalkyl with at least one N or 0 atom in the ring

or R 10 is heteroaryl with at least one N atom in the ring,-

each n can be the same or different and is 0, 1, or 2;

or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1 wherein when Z 1 is N and Z 2 is

CH, then at least one of the following is true:

(i) R b and R 6 together are not both carbon atoms linked

by a single covalent bond;

(ii) R b and R 6 are both carbon atoms linked by a single

covalent bond and R 5 , is disubstituted;

(iii) R 5 and R 6 are both carbon atoms linked by a

single covalent bond and R 6 is mono- or disubstituted;

(iv) R b and R 6 are both carbon atoms linked by a single

covalent bond and R 1 and R 4 are both carbon atoms linked by

a covalent bond or by an optionally-substituted alkylene or

alkenylene group of 1 to 8 carbon atoms where one to three

carbon atoms can be replaced by N, O, S(O) n , or C=O.

3. The compound of claim 1 wherein

M is optionally-substituted C 1 -C 5 alkylene;

G is a bond;

R 7 is aryl or heteroaryl;

L 1 is a covalent bond or C 1 -C 4 alkylene, alkenylene, or

alkynylene ;

R 10 is a tetrahydrofuranyl or tetrahydropyranyl moiety

optionally substituted with hydroxy, lower alkyl, lower

alkoxy, or optionally-substituted lower alkoxy selected

from arylalkyloxy, alkylcarbonyloxy, arylcarbonyloxy,

acetyloxy; or, R 10 is an optionally-substituted nitrogen-

containing 5- to 7-membered heteroaryl or heterocycloalkyl

group .

4. The compound of claim 3 wherein

M is C 1 -C 3 alkylene, but not alkenylene or alkynylene,

optionally-substituted with lower alkyl;

L 1 is a single covalent bond;

R 10 is tetrahydrofuranyl or tetrahydropyranyl

substituted with at least one hydroxy or acetyloxy group,-

5. The compound of claim 3 wherein

M is C 1 -C 3 alkylene, but not alkenylene or alkynylene,

optionally-substituted with lower alkyl;

L 1 is a single covalent bond;

R 10 is a 5- to 7-membered heteroaryl or

heterocycloalkyl group having a single nitrogen atom in the

ring and no additional heteroatoms ,

6. The compound of claim 1 wherein R 1 is H, methyl, allyl,

propargyl, ethyl, cycloalkyl, hydroxyethyl or

cycloalkylmethyl .

7. The compound of claim 1 wherein R 2 and R 2 1 are

independently H, methyl, fluoromethyl, difluoromethyl ,

ethyl, hydroxyethyl, fluoroethyl, and cycloalkyl.

8. The compound of claim 1 wherein R, and R 4 are

independently H, methyl, ethyl, isopropyl, isobutyl, sec-

butyl, tert-butyl, cycloalkyl, heterocycloalkyl , aryl, or

heteroaryl, optionally-substituted with hydroxy1 , mercapto,

sulfonyl, alkylsulfonyl, halogen, pseudohalogen, amino,

carboxyl, alkyl, haloalky, pseudohaloalkyl , alkoxy, or

alkylthio, or R 3 and R 4 are carbon atoms and are linked by a

covalent bond or by an optionally- substituted alkylene or

alkenylene group of 1 to 3 carbon atoms of which 1 or more

atoms can be replaced by N, O, S(O) n , or C=O.

9. The compound of claim 1 wherein R 5 and R 6 are

independently optionally substituted lower alkyl or C 3 -C 3

lower alkσxy, or R 5 and R 6 are carbon atoms and are linked

by a covalent bond or by an optionally-substituted alkylene

or alkenylene group of 1 to 3 carbon atoms of which 1 or

more atoms can be replaced by N, 0, S(O) n , or C=O.

10. The compound of claim 1 wherein

R 1 is H, methyl, allyl, propargyl , ethyl, cycloalkyl,

hydroxyethyl or cycloalkylmethyl;

R 2 and R ? ' are independently H, methyl, fluoromethyl,

difluoromethyl , ethyl, hydroxyethyl, fluoroethyl, and

cycloalkyl ;

R, and R 4 are independently optionally substituted

lower alkyl or C 3 -C 6 cycloalkyl wherein the optional

substituents are hydroxy or lower alkoxy, or R 3 and R 4 are

carbon atoms and are linked by a covalent bond or by an

optionally- substituted alkylene or alkenylene group of 1 to

3 carbon atoms of which 1 or more atoms can be replaced by

N, O, S(O) n , or C-O;

R 5 and R 6 are independently optionally substituted

lower alkyl or C^-C 8 cycloalkyl wherein the optional

substituents are hydroxy or lower alkoxy, or R f and R 6 are

carbon atoms and are linked by a covalent bond or by an

optionally-substituted alkylene or alkenylene group of I to

3 carbon atoms of which 1 or more atoms can be replaced by

N, 0, S(O) „, or C=O;

M is C 1 -C j alkylene optionally-substituted with lower

alkyl.

11. The compound of claim 1 wherein

R 1 is H, methyl, allyl, propargyl , ethyl, cycloalkyl,

hydroxy ethyl or cycloalkylmethyl;

R 2 and R 2 ' are independently H, methyl, fluoromethyl,

difluoromethyl, ethyl, hydroxy ethyl, fluoroethyl, and

cycloalkyl ;

R 3 and R 4 are linked by a covalent bond or by an

optionally-substituted alkylene or alkenylene group of 1 to

3 carbon atoms of which 1 or more atoms can be replaced by

N, 0, S(O) 1 ., or C=O;

R t and R 6 are independently H or optionally substituted

lower alkyl or C 3 -C 8 cycloalkyl wherein the optional

substituents are hydroxy or lower alkoxy, or R k and R 6 are

carbon atoms and are linked by a covalent bond or by an

optionally- substituted alkylene or alkenylene group of 1 to

8 carbon atoms where one to three carbon atoms can be

replaced by W, O, S(O) n , or C=O;

M is C 1 -C 3 alkylene optionally- substituted with lower

alkyl.

12. The compound of claim 1 wherein

R 7 is IIb or IIb:

L 1 is a single covalent bond

X is -N-, -C=C(R 16 )-, -N=C- or -C(O)N-;

Y is -C-, -N-, or -N + -; such that,

When Y is -C- then R 9 , R n , R 17 , R 13 , R 1 ,, R 15 , and R 16 are,

independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl , aryl , heteroaryl, hydroxy, alkoxy,

polyalkylether, amino, alkylamino, dialkylamino,

alkoxyalkyl, sulfonate, aryloxy, heteroaryloxy , acyl ,

acetyl, carboxylate, sulfonate, sulfone, imine, or oxime;

provided that when X is -N- or -C(O)-N-, -L 1 -R 10 is bound to

the -N- atom; and, when X is -C=C(R 16 )- or -N=C-, -L 1 -R 10 is

bound to the -C= atom ; and

When Y is -N- or -N + -, then R 11 is absent or -0 " , and

R 9 , R 12 , R 13 , R 14 , R JS , and R 16 are, independently, -H, halogen,

or optionally substituted alkyl, cycloalkyl,

heterocycloalkyl , aryl, heteroaryl, hydroxyl , alkoxy,

polyalkylether, amino, alkylamino, dialkylamino,

alkoxyalkyl, sulfonate, aryloxy, heteroaryloxy, acyl,

acetyl, carboxylate, sulfonate, sulfone, imine, or oxime;

provided that when X is -N- or -C(O)-N-, -L 1 -R 10 is bound to

the -N- atom; and, when X is -C=C(R 16 )- or -N=C-, -L 1 -R 10 is

bound to the -C= atom;

13. The compound of claim 1 having the formula (III) :

Y is -C-, -N-, or -N + -; such that,

A is a single or double bond;

When A is a single bond and Y is -C- then R 9 a, R 9 b, R n ,

R 12 , R 1J , R 14 , and R 17 are, independently, -H, halogen, or

optionally substituted alkyl, cycloalkyl, heterocycloalkyl,

aryl, heteroaryl, hydroxy, alkoxy, polyalkylether, amino,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl , acetyl, carboxylate, sulfonate,

sulfone, imine, or oxime;

When A is a single bond and Y is -N- or -N + -, then R 11

is absent or -O " , and R 9 a, R 9 b, R 12 , R 13 , R 14 , and R 1 , are,

independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,

hydroxy, alkoxy, polyalkylether, amino, alkylamino,

dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroarylσxy, acyl, acetyl, carboxylate, sulfonate,

When A is a double bond and Y is -C- then R,b and R 17

are absent; and R 9 a, R 11 , R 12 , R 13 , and R 14 are, independently,

~H, halogen, or optionally substituted alkyl, cycloalkyl,

heterocycloalkyl, aryl , heteroaryl, hydroxy, alkoxy,

polyalkylether , amino, alkylamino, dialkylamino,

alkoxyalkyl, sulfonate, aryloxy, heteroaryloxy, acyl ,

acetyl, carboxylate, sulfonate, sulfone, imine, or oxime;

When A is a double bond and Y is -N- or -N + -, then R 9 b

and R 17 are absent; and R 11 is absent or -0 " , and R 9 a, R 12 ,

R 13 , and R 14 are, independently, -H, halogen, or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl,

heteroaryl, hydroxyl , alkoxy, polyalkylether, amino,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl, acetyl, or carboxylate, sulfonate,

sulfone, imine, or oxime;

14. The compound of claim 12 wherein

R, is Ha or lib;

X is -N-;

Y is -C-, -N-, or -N'-; such that

When Y is -C-, then R 9 , R 11 , R 17 , R u , R 14 and R 16 are,

independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl, heterocycloalkyl, aryl , heteroaryl,

hydroxyl, alkoxy, polyalkylether, amino, alkylamino,

dialkylamino, alkoxyalkyl, sulfonate, aryloxy or

heteroaryloxy;

When Y is -N-, then R 11 is absent, and R 9 , R 1 ,, R i3 , R 14

and R 1S are, independently, -H, halogen, or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl,

heteroaryl, hydroxyl, alkoxy, polyalkylether, ammo,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy

or heteroaryloxy;

When Y is -N + -, then RIl is -0 , and R 9 , R 12 , R 13 , R 14 and

R 15 are, independently, -H, halogen, or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl,

heteroaryl, hydroxyl, alkoxy, polyalkylether, amino,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy

or heteroaryloxy;

R ^ is methyl, ethyl, isopropyl, isobutyl, sec-butyl,

tert-butyl, cycloalkyl, heterocycloalkyl, aryl, or

heteroaryl, optionally-substituted with hydroxyl, mercapto,

halogen, pseudohalogen, amino, carboxyl, alkyl, haloalkyl ,

pseudohaloalkyl, alkoxy, or alkylthio;

R, is -H, methyl, fluoromethyl , άifluoromethyl, ethyl,

fluoroethyl, and cycloalkyl;

R 2 ' is H;

R 1 is selected from H, methyl, allyl, propargyl, ethyl,

cycloalkyl, or cycloalkylmethyl ;

Z 1 is nitrogen;

Z 2 is -CH-;

R 4 is -H and R 5 and R 6 are both carbon atoms and

together form C 2 -C 4 alkylene; or, R 3 and R 4 are both carbon

atoms and together form C 2 -C 4 alkylene; or, both R 5 and R 6

together and R 9 and R 4 together both form C 2 -C 4 alkylene

groups ;

L 1 is a covalent bond.

15. The compound of claim 14 wherein

R 9 , R 11 , R 12 , R 13 , R 14 and R 18 are, independently, -H, -

halogen, or optionally- substituted lower alkyl;

R, is optionally-substituted lower alkyl, C 3 -C 8

cycloalkyl, or heterocycloalkyl wherein the optional

substituents are hydroxy, lower alkoxy, or lower alkyl;

R 2 is optionally- substituted lower alkyl, C 3 -C 8

cycloalkyl, or heterocycloalkyl wherein the optional

sυbstituents are hydroxy, lower alkoxy, or lower alkyl;

R/ is H;

R 1 is -H or lower alkyl;

R 5 and R b are both carbon atoms and are linked by a

covalent bond;

R 4 is -H;

Z 2 is -CH- ;

R 10 is tetrahydropyran, tetrahydrofuran, D- or L-

fucose, D- or L-xylose, D- or L-galactose, or D- or L-

glucose, pyrrolidine, piperidine, perhydroazapine,

pyridine, pyrimidine, or pyrazine.

16. A compound selected from the group consisting of

Compounds A through U and Compounds HH through SS, as

follows, or a pharmaceutically acceptable salt thereof:

17. A compound of claim 1 having Formula IV:

wherein

Z 1 a, Z 2 a, Z 1 b, and Z 2 b are independently CH or N;

R 1 a and R 1 b are independently H or optionally

substituted hydroxyl, alkyl, cycloalkyl, heterocycloalkyl,

aryl, or heteroaryl; and when R 2 a' is H then R 2 a and R 1 a can

together form an aziridine or azetidine ring and when R 2 b'

is H then R 2 b and R 1 b can together form an aziridine or

azetidine ring;

R 2 a, R 2 a', R 2 b and R 2 b' are independently H or

optionally substituted alkyl, cycloalkyl, or

heterocycloalkyl; or when R 2 a' is H then R 2 a and R,a can

together form an aziridine or azetidine ring and when R 2 b '

is H then R^b and R j b can together form an aziridine or

azetidine ring;

R 3 a, R 3 b, R 4 a and R 4 b are independently H or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl , aryl, or

heteroaryl; or, R 4 a and R 3 a, or R 4 b and R 3 b, or both, are

carbon atoms linked by an optionally- substituted alkylene

or alkenylene group of 1 to 8 carbon atoms where one to

three carbon atoms can be replaced by N, 0, S(O) n , or C=O;

R 5 a, R 6 a, R 5 b, and R 6 b are independently H or optionally

substituted hydroxyl, alkyl, cycloalkyl, heterocycloalkyl ,

aryl, or heteroaryl; or R 5 a and R 6 a or R 5 b and R 6 b, or both,

are carbon atoms linked by an optionally- substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, 0, S(O) n , or

C-O;

n can be the same or different in each usage and is 0,

1, or 2;

Xa is -0- , -H (La-R 10 a)-, -S-, optionally-substituted -

C(La-R 10 a)=CH-, -C(O)-O-, -C (0) -K (La-R I0 a) - , -N=C{La-R 10 a) - ;

Xb is -O-, -N(Lb-R 10 b) -, -S-, optionally-substituted -

C(Lb-R lO b)=CH~, -C(O)-O-, -C{0) -N(Lb-R 10 b) - , -N=C {Lb-R 10 b) - ,

provided that if Xb is -0-, -S-, or -C(O)-O-, then Xa is -

N(La-R, o a}-, optionally-substituted -C (La-R 10 a) -CH-, -C(O)-

N(La-R ;o a) -, or -N=C (La-R 10 a) - , and if Xa is -0- , -S-, or -

C(O)-O-, then Xb is ~N(Lb-R 10 b) - , optionally-substituted -

C(Lb-R 10 b) =CH-, -C(O) -N(Lb~R 10 b) -, or -N=C (Lb-R 10 b) ~ ;

La and Lb are independently a covalent bond or C 1 -C 4

alkylene;

R 10 a and R 10 b are independently an optionally

substituted 5-, 6-, or 7-membered heterocycloalkyl with at

least one N or 0 atom in the ring or heteroaryl with at

least one N atom in the ring provided that one but not both

of R 10 a and R 10 b can be -H or absent;

Wa and Wb are together a Linker.

18. The compound of claim 17 wherein when Z x a is N and Z 2 a

is CH, and Z t b is N and Z ? b is CH, then at least one of the

following is true:

(i) R t a and R 6 a are not both carbon atoms linked by a

single covalent bond;

(ii) R 5 a and R 6 a are both carbon atoms linked by a

single covalent bond and R^a is disubstituted;

(iii) R 5 a and R 6 a are both carbon atoms linked by a

single covalent bond and R 6 a is mono- or disubstituted;

(iv) R b a and R 6 a are both carbon atoms linked by a

single covalent bond and R 3 a and R^a are both carbon atoms

linked by a covalent bond or by an optionally-substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, 0, S(O) n , or

C=O.

(v) R b a and R 6 a are both carbon atoms linked by a

single covalent bond and neither R 2 a nor R 2 a' are H.

19. The compound of claim 17 wherein R 3 a, R 4 a, Rjb,

and R 4 b are independently selected from H, methyl, ethyl,

isopropyl, isobutyl, sec-butyl, tert-butyl, cycloalkyl,

heterocycloalkyl, aryl, or heteroaryl, optionally-

substituted with hydroxyl, mercapto, sulfonyl,

alkylsulfonyl, halogen, pseudohalogen, amino, carboxyl,

alkyl, haloalky, pseudohaloalkyl , alkoxy, or alkylthio.

20. The compound of claim 17 wherein R,a and R 2 b

are independently selected from -H, methvl. fluoromethvl

difluoromethyl, ethyl, fluoroethyl, hydroxyethyl , and

cycloalkyl .

21. The compound of claim 17 wherein R 1 a and R 1 b

are independently selected from H, methyl, allyl,

propargyl , ethyl, hydroxyethyl, cycloalkyl, or

cycloalkylmethyl .

22. The compound of claim 17 wherein R 3 a, R 4 a, R 3 b,

and R 4 b are independently optionally substituted lower alkyl

or C 3 -C 8 cycloalkyl wherein the optional substituents are

hydroxy or lower alkoxy.

23. The compound of claim 17 wherein Z 1 a and Z 1 b

are both N and Z 2 a and Z 2 b are both C and wherein R 5 a and

R 6 a, and R 5 b and R 6 b, are each carbon and are linked by a

covalent bond or by an optionally- substituted alkylene or

alkenylene group of 1 to 8 carbon atoms where one to three

carbon atoms can be replaced by N, 0, S(O) n , or C=O.

24. The compound of claim 17 wherein Z 1 a and Z 1 b

are both N and Z 2 a and Z 2 b are both C and wherein R 3 a and

R 4 a, and R 3 b and R 4 b, are each carbon and are linked by a covalent bond or by an optionally-substituted alkaline or

alkenylene group of 1 to 8 carbon atoms where one to three

carbon atoms can be replaced by N, 0, S(O) n , or C=O.

25. The compound of claim 17 wherein Wa and Wb

together are a covalent bond or optionally substituted

alkylene, cyclσalkyl, or aryl, of 2 to 20 carbon atoms

where one or more carbon atoms can be replaced with N, 0,

or S(O) n ; and Xa and Xb are independently -0- , -S-, or -

C(O) -0-.

26. The compound of claim 17 wherein one of R 10 a

and R,,,b is -H or is absent.

27. A compound selected from the group consisting

of Compounds V through Z and AA through GG, as follows, or

a pharmaceutically acceptable salt thereof:

28. The compound of claim 2 having the formula

wherein R 1 , R 2 , and R 3 are independently lower alkyl, lower

alkoxy, lower alkanol, or C 3 -C 6 cycloalkyl; R 18 is H or OH;

R 9 is H or phenyl optionally substituted with halogen; R n ,

R 12 , and R 1 ^ are independently H or halogen and R 10 is an

optionally substituted 5-, 6-, or 7-membered

heterocycloalkyl with at least one N or 0 atom in the ring

or R 10 is heteroaryl with at least one N atom in the ring.

29. The compound of claim 16 having the formula

wherein R 1 , R 2 , and R 3 are independently lower alkyl, lower

alkoxy, lower alkanol, or C 3 -C 6 cycloalkyl; R 18 is H or OH;

R 11 , R 1? , and R 13 are independently H or halogen and R 10 is an

optionally substituted 5-, 6-, or 7-membered

heterocycloalkyl with at least one N or 0 atom in the ring

or R 10 is heteroaryl with at least one N atom in the ring.

30. A method for inducing apoptosis in a cell

comprising contacting the cell with a compound of claim 1

in an amount sufficient to induce apoptosis in the cell.

31. The method of claim 30, wherein said cell is

neoplastic .

32. The method of claim 30, wherein said cell

overexpresses an inhibitor of caspase .

33. The method of claim 30, wherein the inhibitor

inhibits activation or activity of one or more of a caspase

selected from caspase-3, caspase-7 and caspase-9.

34. A method of stimulating apoptosis in a cell

comprising contacting the cell with a compound of claim 1

in an amount sufficient to stimulate apoptosis in the cell.

35. The method of claim 34, wherein said cells are

cancer cells .

36. A method of enhancing apoptosis of pathogenic

cells in vivo in an individual comprising administering to

the individual a therapeutically effective amount of a

compound of claim 1.

37. The method of claim 36 further comprising

administering a second therapy selected from radiation,

chemotherapy, immunotherapy, photodynamic therapy and

combinations thereof .

38. A method of treating a disease associated with

the overexpression of IAP in an individual comprising

administering to said individual an effective amount of a

compound of claim 1.

39. A method of treating cancer comprising administering a

therapeutically effective amount of a compound of claim 1.

40. A pharmaceutical composition comprising: a compound

selected from a compound of claim 1 and a pharmaceutically

acceptable excipient.

41. The composition of claim 40 further comprising a second

chemotherapeutic agent.

42. The composition of claim 41, wherein said second

chemotherapeutic agent is selected from alkylating agents,

plant alkaloids, antitumor antibiotics, antimetabolites,

topoisomerase inhibitors and combinations thereof.

43. The composition of claim 41, wherein said

chemotherapeutic agent is selected from altretamine,

busulfan, carboplatin, carmustine, chlorambucil, cisplatin,

cyclophosphamide, dacarbazine, hexamethylmelamine,

ifosfamide, lomustine, melphalan, mechlorethamine, oxaliplatin, procarbazine, streptozocin, temozolomide,

thiotepa, uramustiπe, docetaxel, etoposide, lπnotecan, paclitaxel, tenisopide, topotecan, vincristine,

vinblastine, vmdesme, vmorelbme, bleomycin, dactmomycm, daunorubicin, epirubicm, hydroxyurea,

idarubicm, mitomycin, mitoxantrone, plicamycm, azathioprme, capecitabme, cladribme, cytarabme,

fludarabme, fluorouracil , floxuridine, gemcitabme, mercaptopurine , methotrexate, nelarabine, pemetrexed,

pentostatm, thioguanme, camptothecan, lπnotecan,

topotecan, BNP 1350, SN 38, 9-ammo-camptothecan, lurtotecan, gimatecan, diflomotecan, an anthracyclme,

anthraqumone, podophyllotoxm, doxorubicin, epirubicm,

idarubicm, nemorubicm, mitoxantrone, loxoxantrone,

etoposide, temposide and combinations thereof.

Description:

IAP INHIBITORS

[0001J This application claims priority to and benefit

of U.S. Provisional Application No. 60/820,141 entitled

"IAP Inhibitors" filed on July 24, 2006; the entire

contents of which is hereby incorporated by reference in

its entirety.

[0002) Apoptosis (programmed cell death) plays a

central role in the development and homeostasis of all

multi-cellular organisms. Apoptosis can be initiated

within a cell from an external factor such as a chemokine

(an extrinsic pathway) or via an intracellular event such a

DNA damage (an intrinsic pathway) . Alterations in

apoptotic pathways have been implicated in many types of

human pathologies, including developmental disorders,

cancer, autoimmune diseases, as well as neuro-degenerative

disorders. One mode of action of chemotherapeutic drugs is

cell death via apoptosis.

J0003] Apoptosis is conserved across species and

executed primarily by activated caspases, a family of

cysteine proteases with aspartate specificity in their

substrates. These cysteine containing aspartate specific

catalytically inactive zymogens and are proteolytically

processed to become active proteases during apoptosis.

Once activated, effector caspases are responsible for

proteolytic cleavage of a broad spectrum of cellular

targets that ultimately lead to cell death. In normal

surviving cells that have not received an apoptotic

stimulus, most caspases remain inactive. If caspases are

aberrantly activated, their proteolytic activity can be

inhibited by a family of evolutionarily conserved proteins

called IAPs (inhibitors of apoptosis proteins}.

[0004] The IAP family of proteins suppresses apoptosis

by preventing the activation of procaspases and inhibiting

the enzymatic activity of mature caspases. Several

distinct mammalian IAPs including XIAP, c-IAPl, C-IAP2, ML-

IAP, NAIP (neuronal apoptosis inhibiting protein) , Bruce,

and survivin, have been identified, and they all exhibit

anti -apoptotic activity in cell culture. IAPs were

originally discovered in baculovirus by their functional

ability to substitute for P35 protein, an anti -apoptotic

gene. IAPs have been described in organisms ranging from

Drosophila to human, and are known to be overexpressed in

many human cancers. Generally speaking, IAPs comprise one

to three Baculovirus IAP repeat (BIR) domains, and most of

them also possess a carboxyl- terminal RING finger motif.

The BIR domain itself is a zinc binding domain of about 70

residues comprising 4 alpha-helices and 3 beta strands,

with cysteine and histidine residues that coordinate the

zinc ion. It is the BIR domain that is believed to cause

the anti-apoptotic effect by inhibiting the caspases and

thus inhibiting apoptosis. XIAP is expressed ubiquitously

in most adult and fetal tissues. Overexpression of XIAP in

tumor cells has been demonstrated to confer protection

against a variety of pro-apoptotic stimuli and promotes

resistance to chemotherapy. Consistent with this, a strong

correlation between XIAP protein levels and survival has

been demonstrated for patients with acute myelogenous

leukemia. Down-regulation of XIAP expression by antisense

oligonucleotides has been shown to sensitize tumor cells to

death induced by a wide range of pro-apoptotic agents, both

in vitro and in vivo. Smac/DIABLO-derived peptides have

also been demonstrated to sensitize a number of different

tumor cell lines to apoptosis induced by a variety of pro-

apoptotic drugs.

|OOθ5] In normal cells signaled to undergo apoptosis,

however, the IAP-mediated inhibitory effect must be

removed, a process at least in part performed by a

mitochondrial protein named Smac (second mitochondrial

activator of caspases) . Smac (or, DIABLO) , is synthesized

as a precursor molecule of 239 amino acids; the N-terminal

55 residues serve as the mitochondria targeting sequence

that is removed after import. The mature form of Smac

contains 184 amino acids and behaves as an oligomer in

solution. Smac and various fragments thereof have been

proposed for use as targets for identification of

therapeutic agents .

[0006] Smac is synthesized in the cytoplasm with an N-

terminal mitochondrial targeting sequence that is

proteolytically removed during maturation to the mature

polypeptide and is then targeted to the inter-membrane

space of mitochondria. At the time of apoptosis induction,

Smac is released from mitochondria into the cytosol,

together with cytochrome c, where it binds to IAPs, and

enables caspase activation, therein eliminating the

inhibitory effect of IAPs on apoptosis. Whereas cytochrome

c induces multimerization of Apaf-1 to activate procaspase-

9 and -3, Smac eliminates the inhibitory effect of multiple

IAPs. Smac interacts with essentially all IAPs that have

been examined to date including XIAP, c-IAPl, C-IAP2, ML-

IAP, and survivin. Thus, Smac appears to be a master

regulator of apoptosis in mammals.

[0007] It has been shown that Smac promotes not only

the proteolytic activation of procaspases, but also the

enzymatic activity of mature caspase, both of which depend

upon its ability to interact physically with IAPs. X-ray

crystallography has shown that the first four amino acids

(AVPI) of mature Smac bind to a portion of IAPs. This N-

terminal sequence is essential for binding IAPs and

blocking their anti-apoptotic effects.

(0008] Current trends in cancer drug design focus on

selective targeting to activate the apoptotic signaling

pathways within tumors while sparing normal cells. The

tumor specific properties of specific chemotherapeutic

agents, such as TRAIL have been reported. The tumor

necrosis factor-related apoptosis- inducing ligand (TRAIL)

is one of several members of the tumor necrosis factor

(TNF) superfamily that induce apoptosis through the

engagement of death receptors. TRAIL interacts with an

unusually complex receptor system, which in humans

comprises two death receptors and three decoy receptors .

TRAIL has been used as an anti -cancer agent alone and in

combination with other agents including ionising radiation.

TRAIL can initiate apoptosis in cells that overexpress the

survival factors BcI-2 and BcI-XL, and may represent a

treatment strategy for tumors that have acquired resistance

to chemotherapeutic drugs. TRAIL binds its cognate

receptors and activates the caspase cascade utilizing

adapter molecules such as TRADD. TRAIL signaling can be

inhibited by overexpression of cIAP-1 or 2, indicating an

important role for these proteins in the signaling pathway.

Currently, five TRAIL receptors have been identified. Two

receptors TRAIL-Rl (DR4) and TRAIL-R2 (DR5) mediate

apoptotic signaling, and three non- functional receptors,

DcRl, DcR2 , and osteoprotegerin (OPG) may act as decoy

receptors. Agents that increase expression of DR4 and DR5

may exhibit synergistic anti- tumor activity when combined with TRAIL.

[0009] The basic biology of how IAP antagonists work

suggests that they may complement or synergize other

chemotherapeutic/anti -neoplastic agents and/or radiation.

Chemotherapeutic/anti -neoplastic agents and radiation would

be expected to induce apoptosis as a result of DNA damage

and/or the disruption of cellular metabolism.

(0010} Inhibition of the ability of a cancer cell to

replicate and/or repair DNA damage will enhance nuclear DNA

fragmentation and thus will promote the cell to enter the

apoptotic pathway. Topoisomerases, a class of enzymes that

reduce supercoiling in DNA by breaking and rejoining one or

both strands of the DNA molecule, are vital to cellular

processes, such as DNA replication and repair. Inhibition

of this class of enzymes impairs the cells ability to

replicate as well as to repair damaged DNA and activates

the intrinsic apoptotic pathway.

[0011} The main pathways leading from topoisomerase-

mediated DNA damage to cell death involve activation of

caspases in the cytoplasm by proapoptotic molecules

released from mitochondria, such as Smac . The engagement

of these apoptotic effector pathways is tightly controlled

by upstream regulatory pathways that respond to DNA

lesions -induced by topoisomerase inhibitors in cells

undergoing apoptosis. Initiation of cellular responses to

DNA lesions- induced by topoisomerase inhibitors is ensured

by the protein kinases which bind to DNA breaks. These

kinases (non- limiting examples of which include Akt, JNK

and P38) commonly called "DMA sensors" mediate DNA repair,

cell cycle arrest and/or apoptosis by phosphorylating a

large number of substrates, including several downstream

kinases.

{0012] Platinum chemotherapy drugs belong to a general

group of DNA modifying agents. DNA modifying agents may be

any highly reactive chemical compound that bonds with

various nucleophilic groups in nucleic acids and proteins

and cause mutagenic, carcinogenic, or cytotoxic effects.

DNA modifying agents work by different mechanisms,

disruption of DNA function and cell death; DNA damage/the

formation of cross-bridges or bonds between atoms in the

DNA; and induction of mispairing of the nucleotides leading

to mutations, to achieve the same end result. Three non-

limiting examples of a platinum containing DNA modifying

agents are cisplatin, carboplatin and oxaliplatin.

(0013} Cisplatin is believed to kill cancer cells by

binding to DNA and interfering with its repair mechanism,

eventually leading to cell death. Carboplatin and

oxaliplatin are cisplatin derivatives that share the same

mechanism of action. Highly reactive platinum complexes

are formed intracellularly and inhibit DNA synthesis by

covalently binding DNA molecules to form intrastrand and

interstrand DNA crosslinks,

[0014] Non-steroidal anti- inflammatory drugs (MSAIDs)

have been shown to induce apoptosis in colorectal cells.

NSAIDs appear to induce apoptosis via the release of Smac

from the mitochondria (PNAS, November 30, 2004, vol.

101:16897-16902). Therefore, the use of NSAIDs in

combination with Smac mimetics would be expected to

increase the activity each drug over the activity of either

drug independently.

[0015] Many naturally occurring compounds isolated from

bacterial, plant, and animals can display potent and

selective biological activity in humans including

anticancer and antineoplastic activities. In fact, many

natural products, or semi -synthetic derivatives thereof,

which possess anticancer activity, are already commonly

used as therapeutic agents,- these include paclitaxel,

etoposide, vincristine, and camptothecin amongst others.

Additionally, there are many other classes of natural

products such as the indolocarbazoles and epothilones that

are undergoing clinical evaluation as anticancer agents.

[0016] A reoccurring structural motif in many natural

products is the attachment of one or more sugar residues

onto an aglycone core structure. In some instances, the

sugar portion of the natural product is critical for making

discrete protein- ligand interactions at its site of action

(i.e., pharmacodynamics) and removal of the sugar residue

results in significant reductions in biological activity.

In other cases, the sugar moiety or moieties are important

for modulating the physical and pharmacokinetic properties

of the molecule. Rebeccamycin and staurosporine are

representative of the sugar- linked indolocarbazole family

of anticancer natural products with demonstrated anti-

kinase and anti-topoisomerase activity.

SUMMARY OF THE INVENTION

[0017] The present invention provides IAP antagonists

that are peptidomimetic compounds that mimic the tertiary

binding structure and activity of the N-terminal four amino

acids of mature Smac to IAPs. The invention also provides

methods of using these mimetics to modulate apoptosis and

further for therapeutic purposes.

One aspect of the present invention is an

antagonist/inhibitor of an IAP that is a peptidomimetic

compound that mimics the tertiary binding structure of the

N-terminal amino acids of mature Sraac to IAPs and that has

either an optionally- substituted 5-, 6-, or 7-membered

heterocycloalkyl group with at least one N or 0 atom in the

ring, such as, for example, D- or L-fucose, xylose,

galactose, glucose, pyrrolidine, piperidine, or

perhydroazapine, or an optionally-substituted heteroaryl

group containing at least one N atom such as, for example,

pyridine, pyrimidine, or pyrazine, at the C- terminus of the

peptidomimetic. Such compounds include but are not limited

to monomers, homodimers, and heterodimers of such Smac

mimetics .

[0018] In one aspect of the present invention, an IAP

antagonist that is a monomeric, homodimeric or

heterodimeric compound having the general formula I or IV,

depicted below, and pharmaceutically acceptable salts

thereof. Solvates including hydrates, stereoisomers

including enantiomers, crystalline forms including

polymorphs, and the like are encompassed within the scope

of the invention.

[0019] Another embodiment of the present invention is

the therapeutic combination of compounds of the present

invention with TRAIL or other chemical or biological agents

which bind to and activate the TRAIL receptor (s) . TRAIL

has received considerable attention recently because of the

finding that many cancer cell types are sensitive to TRAIL-

induced apoptosis, while most normal cells appear to be

resistant to this action of TRAIL. TRAIL-resistant cells

may arise by a variety of different mechanisms including

loss of the receptor, presence of decoy receptors, or

overexpression of FLIP which competes for zymogen caspase-8

binding during DISC formation. In TRAIL resistance, Smac

mimetics increase tumor cell sensitivity to TRAIL leading

to enhanced cell death, the clinical correlations of which

are expected to be increased apoptotic activity in TRAIL

resistant tumors, improved clinical response, increased

response duration, and ultimately, enhanced patient

survival rate. In support of this, reduction in XIAP

levels by in vitro antisense treatment has been shown to

cause sensitization of resistant melanoma cells and renal

carcinoma cells to TRAIL (Chawla-Sarkar, et al . , 2004).

The Smac mimetics disclosed herein bind to IAPs and inhibit

their interaction with caspases, therein potentiating

TRAIL- induced apoptosis.

[0020] Another embodiment of the present invention

provides Smac mimetics which act synergistically with

topoismerase inhibitors to potentiate their apoptotic

inducing effect. Topoisomerase inhibitors inhibit DNA

replication and repair, thereby promoting apoptosis and

have been used as chemothemotherapeutic agents.

Topoisomerase inhibitors promote DNA damage by inhibiting

the enzymes that are required in the DNA repair process.

Therefore, export of Smac from the mitochondria into the

cell cytosol is provoked by the DNA damage caused by

topoisomerase inhibitors.

(0021] Topoisomerase inhibitors of both the Type I

class {camptothecin, topotecan, SN-38 {irinotecan active

metabolite) and the Type II class (etoposide) show potent

synergy with the Smac mimetics of the invention in a multi-

resistant glioblastoma cell line (T98G) , breast cancer line

(MDA~MB~231) , and ovarian cancer line (OVCAR-3) among

others . Further examples of topoisomerase inhibiting

agents that may be used include, but are not limited to,

irinotecan, topotecan, etoposide, amsacrine, exatecan,

gimatecan, etc. Other topoisomeraae inhibitors include,

for example, Aclacinomycin A, camptothecin, daunorubicin,

doxorubicin, ellipticine, epirubicin, and mitaxantrone .

[0016] In another embodiment of the invention, the

chemotherapeutic/anti -neoplastic agent may be a platinum

containing compound. In one embodiment of the invention

the platinum containing compound is cisplatin. Cisplatin

can synergize with a Smac peptidomimetic and potentiate the

inhibition of an IAP, such as but not limited to XIAP,

cIAP- 1, c-IAP-2, ML-IAP, etc. In another embodiment a

platinum containing compound is carboplatin. Carboplatin

can synergize with a Smac peptidomimetic and potentiate the

inhibition of an IAP, including, but not limited to, XIAP,

cIAP- 1, c-IAP-2, ML-IAP, etc. In another embodiment a

platinum containing compound is oxaliplatin. The

oxaliplatin can synergize with a Smac peptidomimetic and

potentiate the inhibition of an IAP, including, but not

limited to, XIAP, cIAP-1, c-IAP-2, ML-IAP, etc.

[0022) In another embodiment of the invention, the

chemotherapeutic/anti -neoplastic agent that synergizes with

a compound according to the present invention is a taxane .

Taxanes are anti-mitotic, mitotic inhibitors or microtubule

polymerization agents. Taxanes include but are not limited

to, dσcetaxel and paclitaxel.

[0023] Taxanes are characterized as compounds that

promote assembly of microtubules by inhibiting tubulin

depolymerization, thereby blocking cell cycle progression

through centrosomal impairment, induction of abnormal

spindles and suppression of spindle microtubule dynamics.

The unique mechanism of action of taxane is in contrast to

other microtubule poisons, such as Vinca alkaloids,

colchicine, and cryptophycines, which inhibit tubulin

polymerization. Microtubules are highly dynamic cellular

polymers made of alpha-beta-tubulin and associated proteins

that play key roles during mitosis by participating in the

organization and function of the spindle, assuring the

integrity of the segregated DNA. Therefore, they represent

an effective target for cancer therapy.

[0024J In another embodiment, any agent that activates

the intrinsic apoptotic pathway and/or causes the release

of Smac or cytochrome c from the mitochondria has the

potential to act synergiεtically with a Smac mimetic .

JO025J A combination of a Smac peptidomimetic and a

chemotherapeutic/anti neoplastic agent and/or radiation

therapy of any type that activates the intrinsic pathway

may provide a more effective approach to destroying tumor

cells. Smac peptidomimetics interact with IAP 1 s, such as

XIAP, cIAP-I, cIAP-2, ML-IAP, etc., and block the IAP

mediated inhibition of apoptosis while

chemotherapeutics/anti neoplastic agents and/or radiation

therapy kills actively dividing cells by activating the

intrinsic apoptotic pathway leading to apoptosis and cell

death. As is described in more detail below, embodiments

of the invention provide combinations of a Smac

pepidomimetc and a chemotherapeutic/anti-neoplastic agent

and/or radiation which provide a synergistic action against

unwanted cell proliferation. This synergistic action

between a Smac peptidomimetic and a chemotherapeutic/anti-

neoplastic agent and/or radiation therapy can improve the

efficiency of the chemotherapeutic/anti-neoplastic agent

and/or radiation therapy. This will allow for an increase

in the effectiveness of current chemotherapeutic/anti-

neoplastic agents or radiation treatment allowing the dose

of the chemotherapeutic/anti -neoplastic agent to be

lowered, therein providing both a more effective dosing

schedule as well as a more tolerable dose of

chemotherapeutic/anti -neoplastic agent and/or radiation

therapy.

fθO26] For simplicity and illustrative purposes, the

principles of the invention are described by referring

mainly to specific illustrative embodiments thereof. In

addition, in the following description, numerous specific

details are set forth in order to provide a thorough

understanding of the invention. It will be apparent

however, to one of ordinary skill in the art, that the

invention may be practiced without limitation to these

specific details. In other instances, well known methods

and structures have not been described in detail so as not

to unnecessarily obscure the invention.

DEFINITIONS

[0027] "Alkyl" and "alkylene" mean a branched or

unbranched, saturated or unsaturated (i. e. alkenyl,

alkenylene, alkynyl, alkynylene) non-cyclic aliphatic

hydrocarbon group, having up to 12 carbon atoms unless

otherwise specified. (However, if alkenylene is specified

but alkynylene is not, then alkynylene is excluded. E.g.,

"alkylene or alkenylene" excludes alkynylene.) When used

as part of another term, for example, "alkylamino" , the

alkyl portion may be a saturated hydrocarbon chain, however

also includes unsaturated hydrocarbon carbon chains such as

"alkenylamino" and "alkynylamino" . Examples of particular

alkyl groups include methyl, ethyl, n-propyl r isopropyl, n-

butyl , iso-butyl, sec-butyl, tert-butyl, n-pentyl, 2-

methylbutyl, 2 , 2 -dimethylpropyl, n-hexyl, 2- methylpentyl ,

2, 2-dimethylbutyl, n-heptyl, 3-heptyl, 2-methylhexyl , and

the like. The terms "lower alkyl", "C 1 -C 4 alkyl" and "alkyl

of 1 to 4 carbon atoms" are synonymous and used

interchangeably to mean methyl, ethyl, 1-propyl, isopropyl,

cyclopropyl, 1-butyl, sec-butyl or t-butyl. Unless

specified, optionally substituted alkyl groups may contain

one, two, three or four substituents which may be the same

or different. Examples of the above substituted alkyl

groups include, but are not limited to; cyanomethyl,

nitromethyl, hydroxymethyl , trityloxymethyl ,

propionyloxymethyl, aminomethyl, carboxymethyl,

carboxyethyl, carboxypropyl , alkyloxycarbonylmethyl,

allyloxycarbonylaminomethyl, carbamoyloxymethyl,

methoxymethyl, ethoxymethyl, t- butoxymethyl,

acetoxymethyl, chloromethyl, bromomethyl, iodomethyl,

trifluoromethyl , 6- hydroxyhexyl , 2,4-dichloro (n-butyl),

The alkyl group may also be substituted with a carbocycle

group. Examples include cyclopropylmethyl ,

cyclobutylmethyl , cyclopentylmethyl, and cyclohexylmethyl

groups, as well as the corresponding-ethyl , -propyl, -

butyl, -pentyl, -hexyl groups, etc. Particular substituted

alkyls are substituted methyl groups. Examples of the

substituted methyl groups include groups such as

hydroxymethyl, protected hydroxymethyl (e.g.,

tetrahydropyranyloxymethyl) , acetoxymethyl,

carbamoyloxymethyl, trifluoromethyl, chloromethyl,

carboxymethyl, bromomethyl and iodomethyl .

"Cycloalkyl" means a saturated or unsaturated cyclic

aliphatic hydrocarbon group, having up to 12 carbon atoms

unless otherwise specified and includes cyclic and

polycyclic, including fused cycloalkyl.

[0028] "Amino" denotes primary (i e., -NH 2 ), secondary

(i.e., -NRH) and tertiary {i.e. -NRR) amines.

[0029] Particular secondary and tertiary amines are

alkylamine, dialkylamine, arylamine, diarylamine,

aralkylamine and diaralkylamine . Particular secondary and

tertiary amines are methylamine, ethylamine, propylamine,

isopropylamine, phenylamine, benzylamine dimethylamine,

diethylamine, dipropylamine and disopropylamine .

[0030] "Aryl" when used alone or as part of another

term means a carbocyclic aromatic group whether or not

fused having the number of carbon atoms designated or if no

number is designated, up to 14 carbon atoms. Particular

aryl groups include phenyl, naphthyl, biphenyl,

phenanthrenyl , naphthacenyl, and the like (see e. g. Lang's

Handbook of Chemistry (Dean, J. A., ed) 13 th ed. Table 7-2

[1985] ). In a particular embodiment an aryl group is

phenyl. Optionally substituted phenyl or optionally

substituted aryl denotes a phenyl group or aryl group that

may be substituted with one, two, three, four or five

substituents chosen, unless otherwise specified, from

halogen (F, Cl, Br, I) , hydroxy, protected hydroxy, cyano,

nitro, alkyl (such as C 1 -C 6 alkyl) , alkoxy {such as C 1 -C 6

alkoxy} , benzyloxy, carboxy, protected carboxy,

carboxymethyl, protected carboxymethyl , hydroxymethyl,

protected hydroxymethyl, aminomethyl, protected

aminomethyl, trifluoromethyl, alkylsulfonylamino,

arylsulfonylamino, heterocyclylsulfonylatnino, heterocyclyl,

aryl, or other groups specified. One or more methyne (CH)

and/or methylene (CH;,) groups in these substituents may in

turn be substituted with a similar group as those denoted

above. Examples of the term "substituted phenyl" includes

but is not limited to a mono-or di (halo) phenyl group such

as 2-chlorophenyl , 2- bromophenyl , 4-chlorophenyl, 2,6-

dichlorophenyl, 2, 5-dichlorophenyl, 3 , 4-dichlorophenyl, 3-

chlorophenyl , 3 -bromophenyl, 4 -bromophenyl, 3,4-

dibromophenyl , 3 -chloro-4- fluorophenyl , 2- fluorophenyl and

the like; a mono-or di (hydroxy) phenyl group such as 4-

hydroxypheny1 , 3 - hydroxyphenyl , 2,4-dihydroxypheny1 , the

protected-hydroxy derivatives thereof and the like; a

nitrophenyl group such as 3 -or 4-nitrophenyl; a cyanophenyl

group, for example, 4 -cyanophenyl ; a mono-or di (lower

alkyl) phenyl group such as 4-methylphenyl, 2,4-

dimethylphenyl, 2- methylphenyl, 4- (iso-propyl) phenyl, 4~

ethylphenyl, 3- (n-propyl) phenyl and the like; a mono or

di (alkoxy) phenyl group, for example, 3 , 4-dimethoxyphenyl,

3~methoxy-4-benzyloxyphenyl, 3- methoxy-4- (1-chloromethyl)

benzyloxy-phenyl, 3-ethoxyphenyl, 4- (isopropoxy) phenyl,

4- (t-butoxy) phenyl, 3 -ethoxy-4 -methoxyphenyl and the

like; 3 -or 4 - trifluoromethylphenyl ; a mono- or

dicarboxyphenyl or (protected carboxy) phenyl group such 4-

(protected hydroxymethyl) phenyl such as 3- (protected

hydroxymethyl) phenyl or 3,4-di (hydroxymethyl) phenyl; a

mono-or di (aminomethyl) phenyl or (protected aminomethyl)

phenyl such as 2- (aminomethyl) phenyl or 2, 4- (protected

aminomethyl} phenyl; or a mono-or di (N-

(methylsulfonylamino) } phenyl such as 3- (N-

methylsulfonylamino) ) phenyl. Also, the term" substituted

phenyl" represents disubstituted phenyl groups where the

substituents are different, for example, 3-methyl-4-

hydroxyphenyl , 3- chloro-4-hydroxyphenyl, 2-methoxy~4 ~

bromophenyl , 4-ethyl-2-hydroxyphenyl , 3 -hydroxy-4-

nitrophenyl, 2-hydroxy-4 -chlorophenyl , and the like, as

well as trisubεtituted phenyl groups where the substituents

are different, for example 3-methoxy-4-benzyloxy-6-methyl

sulfonylamino, 3~ methoxy-4 -benzyloxy- 6 -phenyl

sulfonylamino, and tetrasubstituted phenyl groups where the

substituents are different such as 3 -methoxy-4 -benzyloxy- 5-

methyl -6 -phenyl sulfonylamino. Particular substituted

phenyl groups are 2-chlorophenyl , 2-aminophenyl, 2-

bromophenyl, 3- methoxyphenyl , 3-ethoxy-phenyl , 4-

benzyloxyphenyl, 4 -methoxyphenyl , 3-ethoxy-4-

benzyloxyphenyl , 3 , 4 -diethoxyphenyl , 3 -methoxy-4 -

phenyl, 3-methoxy-4- (1-chloromethyl) benzyloxy-6-methyl

sulfonyl aminopheπyl groups. Fused aryl rings may also be

substituted with the substituents specified herein, for

example with 1, 2 or 3 substituents, in the same manner as

substituted alkyl groups.

[0031] "Heterocyclic group", "heterocyclic",

"heterocycle" , "heterocyclyl" , "heterocycloalkyl" or

"heterocyclo" alone and when used as a moiety in a complex

group are used interchangeably and refer to cycloalkyl

group, i.e., any mono-, bi-, or tricyclic, saturated or

unsaturated, non-aromatic ring systems having the number of

atoms designated, generally from 5 to about 14 atoms, where

the ring atoms are carbon and at least one heteroatom

(nitrogen, sulfur or oxygen) . In a particular embodiment

the group incorporates 1 to 4 heteroatoms. Typically, a 5-

membered ring has 0 to 2 double bonds and 6 -or 7-membered

ring has 0 to 3 double bonds and the nitrogen or sulfur

heteroatoms may optionally be oxidized (e. g. SO, SO 2 ), and

any nitrogen heteroatσm may optionally be quaternized.

Particular non-aromatic heterocycles include morpholinyl

(morpholino) , pyrrolidinyl , oxiranyl, oxetanyl,

tetrahydrofuranyl , 2,3- dihydrofuranyl , 2H-pyranyl,

tetrahydropyranyl , aziridiπyl, azetidinyl, l-methyl-2-

pyrrolyl, piperazinyl and piperidinyl . For the avoidance

of doubt, "heterocycloalkyl includes heterocycloalkyl

alkyl.

[0032] "Heteroaryl" alone and when used as a moiety in

a complex group refers to any aryl group, i.e., mono-, bi-,

or tricyclic aromatic ring system having the number of

atoms designated where at least one ring is a 5-, 6-or 7-

membered ring containing from one to four heteroatoms

selected from the group nitrogen, oxygen, and sulfur

(Lang's Handbook of Chemistry, supra} . Included in the

definition are any bicyclic groups where any of the above

heteroaryl rings are fused to a benzene ring. The following

ring systems are examples of the heteroaryl (whether

substituted or unsubstituted) groups denoted by the term

"heteroaryl": thienyl, furyl, imidazolyl, pyrazolyl ,

thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl,

thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl,

oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl,

thiazinyl, oxazinyl, triazinyl, thiadiazinyl , oxadiazinyl,

dithiazinyl , dioxazinyl, oxathiazinyl, tetrazinyl,

thiatriazinyl, oxatriazinyl, dithiadiazinyl, imidazolinyl ,

dihydropyrimidyl , tetrahydropyrimidyl , tetrazolo [1, 5-b]

pyridazinyl and purinyl, as well as benzo- fused

derivatives, for example benzoxazolyl , benzofuryl,

benzothiazolyl, benzothiadiazolyl , benzotriazolyl ,

benzoimidazolyl and indolyl . Particularly"heteroaryls"

include; 1, 3 -thiazol-2-yl , 4- (carboxymethyl) -5-methyl- 1 ,

3- thiazol~2-yl, 4- (carboxymethyl) -5 -methyl- 1, 3-thiazol-

2-yl sodium salt, 1, 2 , 4-thiadiazol-5-yl , 3- methyl- 1, 2,4-

thiadiazol-5-yl, 1, 3 , 4-triazol-5-yl, 2 -methyl- 1, 3,4-

triazol-5-yl, 2 -hydroxy- 1, 3,4- triazol-5-yl, 2-carboxy-4-

methyl-1, 3 , 4 -triazol-5-yl sodium salt, 2-carboxy-4-methyl-

1, 3,4-triazol- 5-yl, 1, 3 -oxazol-2-yl, 1, 3 , 4 -oxadiazol-5-

yl, 2-methyl-l, 3 , 4-oxadiazol-5-yl , 2- (hydroxymethyl) - 1,

3 , 4-oxadiazol-5-yl, 1, 2 , 4 -oxadiazol-5-yl, I 7 3,4-

thiadiazol-5-yl, 2-thiol-l, 3 , 4- thiadiazol-5-yl , 2-

{methylthio) -1, 3 , 4 - thiadiazol-5 -yl, 2 -amino- 1, 3,4-

thiadiazol-5-yl, IH- tetrazol-5-yl , 1-methyl -IH- tetrazol-5-

yl, 1- (1- (dimethylamino) eth-2-yl}»l H- tetrazol-5-yl , 1-

(carboxymethyl) -1 H- tetrazol-5-yl , 1- {carboxymethyl) -IH-

tetrazol-5-yl sodium salt, 1- (methylsulfonic acid) -IH-

tetrazol-5-yl, 1- (methylsulfonic acid) -IH- tetrazol-5-yl

sodium salt, 2 -methyl -IH- tetrazol-5-yl, 1, 2 , 3 - triazol-5-

5-yl, 4-methyl-l, 2 , 3 -triazol-5-yl , pyrid-2-yl N- oxide, 6-

methoxy-2- (n-oxide) ~pyridaz-3 -yl, 6~hydroxypyridaz-3-yl ,

l-methylpyrid-2-yl, 1- methylpyrid-4-yl, 2 -hydroxypyritnid-

4-yl, 1,4, 5 , 6- tetrahydro-5 , 6-dioxo-4-methyl-as-triazin-3-

yl, 1, 4,5, 6- tetrahydro-4 ~ (formylmethyl) -5 , 6-dioxo-as-

triazin-3-yl, 2 , 5-dihydro-5-oxo-6 -hydroxy- astriazin-3 -yl ,

2 , 5-dihydro-5-oxo-6-hydroxy-as-triazin-3-yl sodium salt,

2, 5-dihydro-5-oxo-6- hydroxy-2 -methyl-astriazin- 3 -yl sodium

salt, 2 , 5-dihydro-5-oxo-6-hydroxy-2 -methyl-as- triazin-3 -yl ,

2 , 5 -dihydro- 5 -oxo- 6 -methoxy- 2 -methyl -as-triazin-3 -yl, 2,5-

dihydro-5-oxo-as-triazin-3-yl , 2,5- dihydro™5~oxo-2-methyl-

as-triazin-3 -yl, 2 , 5 -dihydro-5-oxo- 2 , 6 -dimethyl-as-

triazin-3-yl , tetrazolo [1, 5-b] pyridazin-6-yl and 8-

aminotetrazolo [1, 5-b] -pyridazin-6-yl. An alternative

group of "heteroaryl" includes: 4- (carboxymethyl) -5-

πiethyl-1, 3-thiazol-2-yl, 4- (carboxymethyl) -5- methyl- 1,

3 - thiazol-2-yl sodium salt, 1, 3 , 4 -triazol-5-yl , 2-methyl-

1, 3, 4-triazol-5-yl, lH-tetrazol-5-yl , 1-methyl-lH-

tetrazol-5-yl, 1- (1- (dimethylamino) eth-2-yl) -lH-tetrazol-

5-yl, 1- (carboxymethyl} - IH- tetrazol-5-yl, 1-

( carboxymethyl) -IH- tetrazol- 5 -yl sodium salt, 1-

(methylsulfonic acid) -IH- tetrazol-5-yl, 1- (methylsulfonic

1.4, 5,6- tetrahydro-5, 6~dioxo-4 -methyl-as-triazin-3 -yl , 1,

4.5, 6-tetrahydro-4- (2-formylmethyl) -5 , 6-dioxo- as-

triazin~3~yl , 2, 5-dihydro-5-oxo-6-hydroxy-2-methyl-as-

triazin-3-yl sodium salt, 2, 5~dihydro-5- oxo- 6 -hydroxy- 2-

methyl-as-triazin-3-yl, tetrazolo [1, 5-b] pyridazin-6-yl ,

and 8-aminotetrazolo [1, 5- b] pyridazin-6-yl .

|0033] For the avoidance of doubt, aryl includes fused

aryl which includes, for example, naphthyl, indenyl and

also include arylalkyl; cycloalkyl includes fused

cycloalkyl which includes, for example, tetrahydronaphthyl

and indanyl; heteroaryl includes fused heteroaryl which

includes, for example, indoyl, benzofuranyl, benzothienyl

and also includes cycloalkylalkyl ; heterocyclo includes

fused heterocycloalkyl which includes, for example,

indolinyl, isoindolinyl, tetrahydroquinolinyl ,

tetrahydroisoquinolinyl and also includes

heterocycloalkylalkyl .

[0034] "Optionally substituted" means that a H atom can

be, but is not necessarily, replaced by one or more

different atoms. One of skill in the art will readily

know, or can readily ascertain, what atoms or moieties can

be substituted for a hydrogen atom or atoms in a given

position. Typical optional substituents are any one or

more of hydroxy, alkyl, lower alkyl, alkoxy, lower alkoxy,

cycloalkyl, heterocycloalkyl , aryl, heteroaryl, halogen,

pseudohalogen, haloalkyl, pseudohaloalkyl, carbonyl,

carboxyl , tnercapto, amino, nitro, and thiocarbonyl , but

other moieties can also be optional substituents. So, for

example, optionally substituted nitrogen can mean an amide,

sulfonamide, urea, carbamate, alkylamines, dialkylamines,

arylamines, etc; optionally substituted alkyl includes

methyl, ethyl, propyl, isopropyl, t -butyl, etc.;

optionally substituted aryl includes phenyl, benzyl, tolyl,

pyridine, naphthyl, imidazole, etc. Reference to a group

as "optionally substituted" encompasses that group when it

is substituted as described above or, alternatively, when

it is unsubstituted. When "optionally substituted" is used

in front of or at the end of a listing of chemical groups,

all such groups are optionally substituted (unless

otherwise indicated by context.)

10035] A "Linker" is a bond or linking group whereby

two chemical moieties are directly covalently linked one to

the other or are indirectly linked via a chemical moiety

that covalently links the two chemical moieties, in either

case, to form a homo- or heterodimer. A Linker (L) ,

therefore, is a single, double, or triple covalent bond or

is a contiguous chain, branched or unbranched, substituted

or unsubstituted, of 1 to about 100 atoms, typically 1 to

about 20 atoms and typically up to about 500 MW, e.g.,

alkyl, alkylene, alkylyne, alkyloxyalkyl, alkylarylalkyl ,

or optionally-substituted alkyl, alkylene, alkylyne,

alkyloxyalkyl, alkylarylalkyl chain of 1 to 12 atoms.

Illustrative Linkers are described, e.g., in US 20050197403

as well as in US Patent Application Serial Number

11/363,387 filed 2/27/2006, both of which are incorporated

herein by reference as though fully set forth.

[0036] "Pseudohalogens" are binary inorganic compounds

of the general form XY, where X is a cyanide, cyanate,

thiocyanate etc. group and Y is any of X, or a true

halogen. Not all combinations are known to be stable.

Examples include cyanogen, (CN) 2 and iodine cyanide, ICN.

These anions behave as halogens and the presence of the

internal double bonds or triple bonds do not appear to

affect their chemical behavior.

[0037] "Inhibitor" or "antagonists" means a compound

which reduces or prevents the binding of IAP proteins to

caspase proteins or which reduces or prevents the

inhibition of apoptosis by an IAP protein, or which binds

to an IAP BIR domain in a manner similar to the amino

terminal portion of Smac, thereby freeing Smac to inhibit

the action of an IAP,

[0038] "Pharmaceutically acceptable salts" include both

acid and base addition salts. "Pharmaceutically acceptable

acid addition salt" refers to those salts which retain the

biological effectiveness and properties of the free bases

and which are not biologically or otherwise undesirable,

formed with inorganic acids such as hydrochloric acid,

hydrobromic acid, sulfuric acid, nitric acid, carbonic

acid, phosphoric acid and the like, and organic acids may

be selected from aliphatic, cycloaliphatic, aromatic,

araliphatic, heterocyclic, carboxylic, and sulfonic classes

of organic acids such as formic acid, acetic acid,

propionic acid, glycolic acid, gluconic acid, lactic acid,

pyruvic acid, oxalic acid, malic acid, maleic acid,

maloneic acid, succinic acid, fumaric acid, tartaric acid,

citric acid, aspartic acid, ascorbic acid, glutamic acid,

anthranilic acid, benzoic acid, cinnamic acid, mandelic

acid, embonic acid, phenylacetic acid, methanesulfonic

acid, ethanesulfonic acid, p-tolυenesulfonic acid,

salicyclic acid and the like.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

[0039] It must also be noted that as used herein and in

the appended claims, the singular forms "a", "an", and

"the" include plural reference 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. Although any methods similar or equivalent to

those described herein can be used in the practice or

testing of embodiments of the present invention, the

preferred methods are now described. All publications and

references mentioned herein are incorporated by reference.

Nothing herein is to be construed as an admission that the

invention is not entitled to antedate such disclosure by

virtue of prior invention,

[0040] The terms "mimetic, " "peptide mimetic" and

"peptidomimetic" are used interchangeably herein, and

generally refer to a peptide, partial peptide or non-

peptide molecule that mimics the tertiary binding structure

or activity of a selected native peptide or protein

functional domain (e.g., binding motif or active site) .

These peptide mimetics include recombinantIy or chemically

modified peptides, as well as non-peptide agents such as

small molecule drug mimetics, as further described below.

[0041] As used herein, the terms "pharmaceutically

acceptable", "physiologically tolerable" and grammatical

variations thereof, as they refer to compositions,

carriers, diluents and reagents, are used interchangeably

and represent that the materials can be administered to a

human being .

[0042] As used herein "subject" or "patient" refers to

an animal or mammal including, but not limited to, human,

dog, cat, horse, cow, pig, sheep, goat, chicken, monkey,

rabbit, rat, mouse, etc.

[0043) As used herein, the term "therapeutic" means an

agent utilized to treat, combat, ameliorate, prevent or

improve an unwanted condition or disease of a patient.

Embodiments of the present invention are directed to

promote apoptosis, and thus cell death.

[0044] The terms "therapeutically effective amount" or

"effective amount", as used herein, may be used

interchangeably and refer to an amount of a therapeutic

compound component of the present invention. For example,

a therapeutically effective amount of a therapeutic

compound is a predetermined amount calculated to achieve

the desired effect, i.e., to effectively promote apoptosis,

preferably by eliminating an IAP inhibition of apoptosis,

more preferably by inhibiting an IAP binding to a caspase .

[0045] It has been demonstrated in accordance with the

present invention that the IAP-binding compounds of the

present invention are capable of potentiating apoptosis of

cells .

[0046] Optionally substituted 5-, 6-, or 7-merabered

heterocycloalkyl groups with at least one N or O atom in

the ring that are useful in the practice of the invention

include, for example, pyrrolidine, piperidine,

perhydroazapine rings, or monosaccharides or disaccharides,

each unit comprising three to six carbon atoms, although

longer chain polysaccharides can also be employed. These

include, for example, trioses, tetroses, pentoses, and

hexoses, such as glucose, mannoεe, fructose, xylose,

erythrose, fucose, galactose, etc. The sugars can be

naturally-occurring (including D- and L-sugars} or non-

naturally-occurring sugars or derivatives thereof and can

be the alpha or beta anomers . Heteroaryl groups with at

least one N atom in the ring that are useful in the

practice of the invention include, for example, pyridine,

pyrimidine, or pyrazine.

[0047] Chemical procedures for synthesizing or

derivatizing, or modifying, Smac mimetics by binding an

optionally substituted 5-, 6-, or 7-membered

heterocycloalkyl group with at least one N or O atom in the

ring or a heteroaryl group with at least one M atom in the

ring thereto are known to person of skill in the art or can

be determined without undue experimentation.

[0048) Monomeric IAP antagonists of the invention

include compounds of formula I :

] [0049] wherein

[0050] Z 1 and Z 2 are each independently CH or N;

[0051] R 1 is H or optionally substituted hydroxy, alkyl,

cycloalkyl, heterocycloalkyl, aryl, or heteroaryl; and when

R/ is H then R 2 and R 1 can together form an aziridine or

azetidine ring;

[0052] R 2 and R 2 1 are each independently H or optionally-

substituted alkyl, cycloalkyl, or heterocycloalkyl; or when

R 2 ' is H then R 2 and R 1 can together form an aziridine or

azetidine ring;

(0053} R 3 and R 4 are each independently H or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl, or

heteroaryl; or, R 3 and R 4 are each carbon and are linked by

a covalent bond or by an optionally-substituted alkylene or

alkenylene group of 1 to 8 carbon atoms where one to three

carbon atoms can be replaced by N, 0, S(O) n , or C=O;

[0054] R 5 and R 6 are each independently H or optionally

substituted hydroxy, alkyl, cycloalkyl, heterocycloalkyl,

aryl , or heteroaryl ; or R 5 and R 6 are each carbon and are

linked by a covalent bond or by an optionally-substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, 0, S(O) n , or

OO ;

[0055) M is a bond or an optionally substituted

alkylene group of 1 to 5 carbon atoms;

[0056] G is a bond, a heteroatom, -(C=O)-, -S(O) n -, -

NR 8 -, -NCOR 6 -, or -NS(O) n R 8 -, where R 8 is lower alkyl,

optionally-substituted lower alkyl or C 3 . a cycloalkyl;

[0057] R 7 is optionally substituted alkyl, cycloalkyl,

heterocycloalkyl , aryl , or heteroaryl, wherein R 7 is

substituted with -L 1 -R 10 and is optionally further

substituted;

[0058] L 1 is a covalent bond or optionally substituted

C 1 X 6 alkylene;

[0059J R 10 is an optionally substituted 5-, 6-, or 7-

membered heterocycloalkyl group with at least one N or 0

atom in the ring or R 10 is a heteroaryl group with at least

one N atom in the ring;

[0060] n can be the same or different in each usage and

is 0, 1, or 2;

(0061) and pharmaceutically acceptable salts and

solvates thereof .

[0062] In illustrative embodiments of compounds of

Formula I, when Z 1 is M and Z 2 is CH, then at least one of

the following is true :

[0063) (i) R 5 and R 6 together are not both carbon atoms

1 -i ^ V,

[0064] (ii) R 5 and R 6 are both carbon atoms linked by a

single covalent bond and R 5 is disubstituted;

|0065] (iii) R b and R 6 are both carbon atoms linked by a

single covalent bond and R 6 is mono- or disubstituted;

[0066] (iv) R 5 and R 6 are both carbon atoms linked by a

single covalent bond and R 3 and R 4 are both carbon atoms

linked by a covalent bond or by an optionally-substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, O, S(O) n , or

C=O.

[0067] In illustrative embodiments of compounds of

Formula I, one or any two or more of the following

limitations apply to compounds in which the preceding

limitations on R 5 and R 6 apply or in which the preceding

limitations on R 5 and R 6 don't apply:

[0068] (1) M is optionally- substituted C 1 -C j , alkylene,

alkenylene, or alkynylene ; or M is C 1 -C 2 alkylene

optionally-substituted with lower alkyl; or M is C 1 -C 3

alkylene {excluding alkenylene and alkynylene) optionally-

substituted with lower alkyl;

[0069] (2) G is a bond;

[0071| (4) L 1 is a covalent bond or C 1 -C 4 alkylene ; or L 1

is a single covalent bond;

[θ072J (5) R 10 is a tetrahydrofuranyl or

tetrahydropyranyl moiety optionally substituted with

hydroxy, lower alkyl, lower alkoxy, or optionally-

substituted lower alkoxy selected from arylalkyloxy,

alkylcarbonyloxy, arylcarbonyloxy, acetyloxy; or, R 10 is an

optionally-substituted nitrogen-containing 5- to 7-membered

heteroaryl or heterocycloalkyl group; or R 10 is

tetrahydrofuranyl or tetrahydropyranyl substituted with at

least one hydroxy or acetyloxy group; or R 10 is a 5- to 7-

membered heteroaryl or heterocycloalkyl group having a

single nitrogen atom in the ring and no additional

heteroatoms

[0073] (6) R 1 is H, methyl, allyl, propargyl, ethyl,

cycloalkyl, hydroxyethyl or cycloalkylmethyl ; or R 1 is H,

methyl, allyl, propargyl, ethyl, cycloalkyl, hydroxy ethyl

or cycloalkylmethyl ;

(0074] (7) R 2 and R 2 1 are independently H, methyl,

fluoromethyl , difluoromethyl, ethyl, hydroxyethyl,

fluoroethyl, and cycloalkyl; or R 2 and R 2 ' are independently

H, methyl, fluoromethyl , difluoromethyl , ethyl,

hydroxyethyl , fluoroethyl, and cycloalkyl;

[0075] (8} R 3 and R 4 are independently H, methyl, ethyl,

isopropyl, isobutyl, sec-butyl, tert -butyl, cycloalkyl,

heterocycloalkyl , aryl , or heteroaryl, optionally-

substituted with hydroxyl, mercapto, sulfonyl,

alkylsulfonyl , halogen, pseudohalogen, amino, carboxyl,

alkyl , haloalkyl, pseudohaloalkyl, alkoxy, or alkylthio, or

R 3 and R 4 are carbon atoms and are linked by a covalent bond

or by an optionally-substituted alkylene or alkenylene

group of 1 to 3 carbon atoms of which 1 or more atoms can

be replaced by N, O, S(O) n , or C-O; or R 3 and R 4 are linked

by a covalent bond or by an optionally-substituted alkylene

or alkenylene group of 1 to 3 carbon atoms of which 1 or

more atoms can be replaced by N, 0, S(O) n , or C=O;

[0076| (9) R 5 and R 6 are independently optionally

substituted lower alkyl or C 3 -C 8 cycloalkyl wherein the

optional substituents are hydroxy or lower alkoxy, or R 5 and

R 6 are carbon atoms and are linked by a covalent bond or by

an optionally-substituted alkylene or alkenylene group of 1

to 3 carbon atoms of which 1 or more atoms can be replaced

by N, 0, S(O) n , or C-O;

[0077] (10) R 7 is Ha or lib:

[0078] wherein Ll is a single covalent bond

X is -N-, -C=C(R 16 )-, -N=C- or -C(O)N-;

Y is -C-, -N-, or -N + -; such that,

[0079] When Y is -C- then R,, R 11 , R 12 , R 13 , R 14 , R 15 , and

R 16 are, independently, -H, halogen, or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl,

heteroaryl, hydroxy, alkoxy, polyalkylether, amino,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl , acetyl, carboxylate, sulfonate,

sulfoπe, imine, or oxime; provided that when X is -N- or -

C(O)-N-, -Ll-R 10 is bound to the -N- atom; and, when X is -

C=C(R 16 )- or -N=C-, -Ll-R 10 is bound to the -C= atom,- and

[0080} When Y is -N- or -N"-, then RIl is absent or -O ,

and R 9 , R 12 , R 13 , R 4 , R 15 , and R 16 are, independently, -H,

halogen, or optionally substituted alkyl, cycloalkyl,

heterocycloalkyl, aryl , heteroaryl, hydroxyl, alkoxy,

polyalkylether, amino, alkylammo, dialkylamino,

alkoxyalkyl, sulfonate, aryloxy, heteroaryloxy, acyl,

acetyl, carboxylate, sulfonate, sulfone, imine, or oxime;

provided that when X is -N- or -C(O)-N-, -L 1 -R 10 is bound to

the -N- atom; and, when X is -C=C(R 16 )- or -N=C-, -Ll-R 10 is

bound to the -C= atom; or

[0081] ( 11 ) R 7 is Ha or lib ;

|0082] X is -N- ;

[0083J Y is -C-, -N-, or -N + -; such that

(0084] When Y is -C-, then R 9 , R 11 , R^, R 13 , R 14 and R 15

are, independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,

hydroxyl, alkoxy, polyalkylether, ammo, alkylammo,

dialkylamino, alkoxyalkyl, sulfonate, aryloxy or

heteroaryloxy;

[00851 When Y is -N-, then R 11 is absent, and R 3 , R i2 ,

R 1J , R 14 and R 15 are, independently, -H, halogen, or

optionally substituted alkyl , cycloalkyl, heterocycloalkyl ,

aryl, heteroaryl, hydroxyl, alkoxy, polyalkylether, ammo,

alkylammo, dialkylammo, alkoxyalkyl, sulfonate, aryloxy

or heteroaryloxy;

[0086] When Y is -N + -, then R 11 is -O , and R 9 , R 12 , R 13 ,

R 14 and R 1 b are, independently, -H, halogen, or optionally

substituted alkyl, cycloalkyl, heterocycloalkyl, aryl,

heteroaryl, hydroxyl, alkoxy, polyalkylether, amino,

alkylamiπo, dialkylammo, alkoxyalkyl, sulfonate, aryloxy

or heteroaryloxy;

(0087J In illustrative embodiments, the compound of

formula 1 has the formula (III) :

wherein Y is -C-, -N-, or -N + -; such that,

A is a single or double bond;

[0088J When A is a single bond and Y is -C- then R 3 a, R 9 b, R 11 , R 171 R 1 .,, R 14 , and R 17 are, independently, -H,

halogen, or optionally substituted alkyl, cycloalkyl,

heterocycloalkyl , aryl, heteroaryl, hydroxy, alkoxy,

polyalkylether, amino, alkylamino, dialkylamino,

alkoxyalkyl, sulfonate, aryloxy, heteroaryloxy, acyl,

acetyl, carboxylate, sulfonate, sulfone, imine, or oxime;

[0089] When A is a single bond and Y is -N- or -N + -,

then R 11 is absent or -0 ' , and R 9 a, R 5 b, R 12 , R 13 , R 14 , and R 17

are, independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,

hydroxy, alkoxy, polyalkylether, amino, alkylamino,

dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl, acetyl, carboxylate, sulfonate,

sulfone, imine, or oxime;

[0090] When A is a double bond and Y is -C- then R 9 b and

R 17 are absent; and R^a, R 11 , R 12 , R 13 , and R 14 are,

independently, -H, halogen, or optionally substituted

alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,

hydroxy, alkoxy, polyalkylether, amino, alkylamino,

dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl, acetyl, carboxylate, sulfonate,

sulfone, imine, or oxime,-,

(0091] When A is a double bond and Y is -N- or -N + -,

then R 9 b and R 17 are absent; and R n is absent or -O , and

R g a, R ]? , R 13 , and R 14 are, independently, -H, halogen, or

optionally substituted alkyl, cycloalkyl, heterocycloalkyl ,

aryl, heteroaryl, hydroxyl, alkoxy, polyalkylether, amino,

alkylamino, dialkylamino, alkoxyalkyl, sulfonate, aryloxy,

heteroaryloxy, acyl, acetyl, or carboxylate, sulfonate,

sulfone, imine, or oxime;

[0092J Specific illustrative compounds of formula 1

include those shown below as compounds A through U and HH through SS .

J0093J Dimeric compounds of the invention include compounds of formula IV:

[0094 J wherein

Z 1 SL 1 Z 2 a, Z x b, and Z 2 b are independently CH or N;

[0095] R 1 B. and R α b are independently H or optionally

substituted hydroxyl, alkyl, cycloalkyl, heterocycloalkyl ,

aryl , or heteroaryl; and when R ? a' is H then R 2 a and R 1 S. can

together form an aziridine or azetidine ring and when R 2 b •

is H then R 2 b and R^b can together form an aziridine or

azetidine ring;

[0096J R ? a, R 2 a', R 2 b and R ? b ' are independently H or

optionally substituted alkyl, cycloalkyl, or

heterocycloalkyl; or when R 2 a' is H then R 2 a and R 1 a can

together form an aziridine or azetidine ring and when R 2 b '

is H then R 2 b and R τ b can together form an aziridine or

[0097} R,a, R,b, R^a and R 4 b are independently H or

optionally substituted alkyl, cycloalkyl, heterocycloalkyl ,

aryl, or heteroaryl; or, R 4 a and R 3 a, or R 4 b and R 3 b, or

both, are carbon atoms linked by an optionally-substituted

alkylene or alkenylene group of 1 to 8 carbon atoms where

one to three carbon atoms can be replaced by N, 0, S(O) n , or

C=O;

[0098] R 5 a, R 6 a, R 5 b, and R 6 b are independently H or

optionally substituted hydroxyl, alkyl, cycloalkyl,

heterocycloalkyl, aryl, or heteroaryl; or R s a and R β a or R sb

and R 6 b, or both, are carbon atoms linked by an optionally-

substituted alkylene or alkenylene group of 1 to 8 carbon

atoms where one to three carbon atoms can be replaced by N,

0, S(O) n , or C=O;

[0099] n can be the same or different in each usage and

is 0 , 1 , or 2 ;

[0010O] Xa is -0- , -N{La-R xo a) -, -S-, optionally-

substituted -C{La-R, o a) =CH-, -C(O)-O-, -C(O) -N (La-R^a) -, -

N=C(La-R 10 a)-;

JOOlOl] Xb is -0-, -N(Lb-R lc b)-, -S-, optionally-

substituted -C(Lb-R lo b)=CH-, -C(O)-O-, -C(O) -N(Lb-R. o b) -, -

N=C(Lb-R lo b) -, provided that if Xb is -0- , -S-, or -C(O)-O-,

then Xa is -N (La-R :o a) - , optionally-substituted -C(La-

R, o a)=CH-, -C(O) -N(La-R 10 a)-, or -N=C (La-R 10 a) -, and if Xa is

-0-, -S-, or -C(O)-O-, then Xb is -N (Lb-R 10 b) - , optionally-

substituted -C(Lb-R ιo b) =CH~, -C(O) -N(Lb-R ιo b)-, or -N=C(Lb-

R lo b)- ;

[00102] La and Lb are independently a covalent bond or

C 1 -C 4 alkylene;

[00103] R 10 a and R iO b are independently an optionally

substituted 5-, 6-, or 7-membered heterocycloalkyl with at

least one N or 0 atom in the ring or heteroaryl with at

least one N atom in the ring, provided that one but not

both of R 10 a and R ]o b can optionally be -H;

|00104] Wa and Wb are together a Linker.

[00105] In illustrative embodiments, Xa is -N (La-R 10 a) -,

-C(La-R 10 a} -, or -N=C (La-R 10 a) -, Xb is -N-, La is a bond, -

R 10 a is an optionally substituted S-, 6-, or 7-membered

heterocycloalkyl with at least one N or O atom in the ring

or heteroaryl with at least one N atom in the ring, and Lb

is a bond, and -R 10 b is H.

[00106] Any one or any two or more of the above

limitations can also apply to compounds having formula IV.

Other limitations that can apply to dimeric IAP antagonists

of the invention include:

[00107] Wa and Wb together are a covalent bond or

optionally substituted alkylene, cycloalkyl, or aryl , of 2

to 20 carbon atoms where one or more carbon atoms can be

replaced with N, O, or S(O) n ; and Xa and Xb are

independently -0-, -S-, or -C(O)-O-; or Wa and Wb together

form a single covalent bond; and/or

one of R 10 a and R 10 b is -H or, if Xa or Xb is -0- , -S-, or - C(O)-O-, then R lo a or R 10 b, respectively, is absent.

[00108] Illustrative embodiments have the following

formulae :

and

{00109] wherein R 1 , R,, and R 3 are independently lower

alkyl, lower alkoxy, lower alkanol, or C 3 -C 6 cycloalkyl; R 13

is H or OH; R 11 , R 12 , and R 13 are independently H or halogen

and R 10 is an optionally substituted 5-, S-, or 7-membered

heterocycloalkyl with at least one N or 0 atom m the ring

or R 10 is heteroaryl with at least one N atom m the ring.

[00110] Specific illustrative compounds of formula IV

include those shown below as compounds V through GG.

[00111} Following are illustrative schemes illustrating

preparation of modified monomers and dimers. Using similar

synthetic techniques, the sugar-modified Smac mimetics

shown m Tables 1 and 2, below, and the piperdine-

substituted Smac mimetics shown m Table 3, below, were prepared

[00112] The binding affinity of illustrative compounds

of the present invention to an IAP was determined

substantially as described by Nikolovska-Coleska, Z. et.al.

(Analytical Biochemistry (2004), vol. 332:261-273) using a

variety of fluorogenic substrates and is reported as a Kd

value. Briefly, various concentrations of IAP antagonists

were mixed with 5 nM fluorescently labeled peptide (AbuRPF-

K(5-Fam) -NH 2 ) and 40 nM of an IAP-BIR3 for 15 min at RT in

100 mL of 0.1M Potassium Phosphate buffer, pH 7.5

containing 100 mg/ml bovine g-globulin. Following

incubation, the polarization values (mP) were measured on a

Victor2V using a 485nm excitation filter and a 520 nm

emission filter. IC50 values were determined from the plot

using nonlinear least -squares analysis using GraphPad

Prism. The compounds described herein afford Kd values in

the ranges of: Kd <0.1 μM (A), Kd = 0.1-1 μ M (B), Kd = 1-

10 μM (C) , and Kd >10 μM (D) .

[00113] Abbreviations used in the following

preparations, which are illustrative of synthesis of

compounds of the invention generally, are: Cbz :

Benzyloxycarbonyl ; Boc: tert-butyloxycarbonyl ; THF:

tetrahydrofuran; DCM: dichloromethane; DDQ: 2 , 3-dichloro-

5 , 6 -dicyano-1 , 4 -benzoqumone ; NMP: N~methylpyrrolidmone;

DMF: dimethylformamide ; TFA: trifluoroacetic acid; HOAc or

AcOH: acetic acid; Hex: hexanes,- HPLC: high performance

liquid chromatography; TLC: thin layer chromatography;

EtOAc: ethyl acetate,- DIPEA: dnsopropylethylamine; TEA:

triethylamme; HATU; 2- (7-Aza-lH-benzotriazole-l-yl) -

1,1,3,3- tetramethyluronium hexafluorophosphate .

[00114] Preparation of Monomeric IAP Antagonists.

(Formula 13, below. Referred to as Compound A m Table 1,

below. )

Scheme I

HATU

[O0ϊI5J Indoline (2) :A round bottom flask containing

trifluoroacetic acid (60 mL) was cooled to 0 C under

nitrogen and triethylsilane (1.16 g, 1.6 mL, 9.96 mmol) was

added followed by the dropwise addition of indole 1 (1.17

g, 3.32 mmol ; prepared using a modification of the

procedure reported by Macor, et al . J. Med. Chem. 1992, 35,

4503-4505) in 9 mL dry dichloromethane, added over 1 hour.

Following complete addition, the solution was stirred for

10 min. Thin layer chromatography (2/1 Hex/EA) indicated

no remaining 1. The solvent was removed in vacuo and the

residue was dissolved in EtOAc. The organic layer was

washed with saturated NaHCO 3 (2X), and brine. The EtOAc

layer was dried over Na 2 SO 4 , filtered, and concentrated.

Purification by column chromatography on silica gel {2/1

hexane/ethyl acetate) afforded 2 as a yellow oil (0.79 g,

67%). 1 H NMR (CDCl j , 300 MHz) δ7.34-7.31 <m, 5H), 6.99-6.85

(m, IH), 6.34-6.27 (m, 2H), 5.12 (s, 2H), 4.13-3.71 (m,

2H), 3.58-3.16 (m, 5H), 2.05-1.87 (m, 4H), 1.73-1.57 (m,

2H) ppm.

[00116) N- substituted indoline { 3 U

A mixture of compound 2 (0,79 g, 2.23 mmol), D- (+) -Xylose

(1.0 g, 6.69 mmol), and ammonium sulfate {0.88 g, 6.69

mmol) in ethanol (50 mL) was heated at 75 C overnight.

Thin layer chromatography (10% MeOH/CH ? Cl 2 ) indicated no

remaining 2. The reaction mixture was preabsorbed on to

silica gel and purified by column chromatography on silica

gel (2% MeOH/CH 2 Cl 2 to 5% MeOH/CH 2 Cl 2 ) to afford compound 3

as a yellow solid (0.98 g, 90%). 1 H NMR (CDCl 3 , 300 MHz)

67.33-7.31 (m, 5H), 6.91 (t, J = 2.4 Hz, IH), 6.39-6.27 (m,

2H), 5.12-4.99 (m, 3H), 4.62-4.45 (m, IH), 4,05-3.87 (m,

3H), 3.79-3.56 (m, 4H), 3.48-3.29 (m, 6H), 2.29-1.92 (m,

4H), 1.77-1.28 (m, 2H) ppm.

|θO117J Substituted indole (4) :

To a solution of 3 (0.98 g, 2.01 mmol) in anhydrous 1,4-

dioxane (30 mL) was added 2 , 3~dichlαro-5 , 6-dicyano-l, 4 -

benzoquinone (0.55 g, 2.42 mmol} neat in one portion. The

reaction was stirred at room temperature for 30 min.

Thin layer chromatography (10% MeOH/CH 5 Cl 2 ) indicated no

remaining 3. The reaction mixture was filtered and the

solid washed with EtOAc. The filtrate was washed with

saturated MaHCO 3 (4X), brine, dried over Na 3 SO,,, filtered,

and concentrated. Purification by column chromatography on

silica gel (2% MeOH/CH 2 Cl 7 to 5% MeOH/CH 2 Cl 2 ) afforded 4 as

a white solid (0.792 g, 81%). '1 H NMR (CDCl 3 , 300 MHz)

δ7.49-7.44 (m, IH) , 7.34-7.29 (m, 5H) , 7.18-7.02 (m, IH) ,

6.86 (t, J = 3.0 Hz, IH) , 6.73-6.67 (m, IH) , 5.10-5.01 (m,

2H) , 4.90-4.88 (m, 2H) , 4.33-4.18 (m, 2H) , 4.06-3.59 (m,

6H) , 3.41-3.06 (m, 3H) , 2.92-2.55 (m, 3H) , 1.94-1.25 (m,

2H) ppm.

[00118] Peracetylated intermediate (5) :

To a solution of 4 (0.79 g, 1.63 mmol) in pyridine (8 rtiL)

was added acetic anhydride (1.66 g, 1.54 πiL, 16.3 mmol) and

the reaction was stirred at room temperature for 6 hours.

Thin layer chromatography (1/1 Hex/EA) indicated no

remaining 4. The reaction was diluted with ethyl acetate

and washed with IM HCl (3X) , water, saturated NaHCO j , brine,

dried over Na 2 SO 4 , filtered, and concentrated. Purification

by column chromatography on silica gel (1/1 Hex/EtOAc)

afforded 5 as a foamy solid (0.86 g, 86%). 1 H NMR (CDCl 3 ,

300 MHz) δ7.72 (t, J = 2.5 Hz, IH), 7.45-7.36 (m, 5H),

7.11-6.86 (m, 2H), 6.61 (t, J = 3.0 Hz, IH), 5.42-5.36 (m,

3H), 5.25-5.17 (m, 3H), 4.29-4.23 (m, IH), 4.16-4.09 (m,

IH), 3.59-3.10 (m, 4H), 2.66-2.49 (m, IH), 2.08 (s, 3H),

2.05 (s, 3H), 1.88-1.61 (m, 6H) ppm.

[00119] Bromo derivative (6) :

[00120] A mixture of 5 (0.20 g, 0.327 mmol) and

potassium acetate (0.096 g, 0.981 mmol) in chloroform (10

mL) was cooled to 0 C and a solution of bromine (0.063g,

0.02mL, 0.393 mmol) in chloroform (1 mL) was added dropwise

via syringe. The reaction was stirred at 0 C for 20 min.

Thin layer chromatography (1/1 Hex/EA) indicated no

remaining 5 . The reaction was diluted with brine and

dichloromethane . The layers were separated and the

organics were washed with saturated Na 2 S 2 O 3 , brine, dried

over Na 2 SO 4 , filtered, and concentrated. Purification by

column chromatography on silica gel {1/1 Hex/EtOAc)

afforded 6 as a foamy solid (0.206 g, 91%). 1 H NMR (CDCl 3 ,

300 MHz) 57.69 (m, IH), 7.46-7.35 (m, 5H), 6.91 (t, J =

3.0 Hz, IH), 6.61 (t, J = 3.0 Hz, IH), 5.58-5.39 (m, 3H),

5.26-5.08 (m, 3H), 4.37-4.32 (m, IH), 4.19-4.09 (m, IH),

3.61-3.07 (m, 4H), 2.71-2.55 (m, IH), 2.09 (s, 3H), 2.05

(s, 3H), 1.96-1.51 (m, 6H) ppm.

(00121] 2 -Arylindole intermediate (7) :

[00122] A mixture of 6 (0.21 g, 0.297 mmol}, potassium

carbonate (0.14 g, 1.04 mmol), and 4- fluorobenzeneboronic

acid (0.054 g, 0.386 mmol) in toluene (12 mL) and ethanol

(6 mL) was degassed by pulling a vacuum until bubbling

occurred. Added next was tetrakis-triphenylphosphine

palladium (0) (0.017g, 0.015 mmol) and the mixture was

degassed again, placed in an oil bath preheated at 90 C,

and stirred for 3 hours. Thin layer chromatography (2/1

Hex/EA) indicated no remaining 6. The reaction was cooled

to room temperature, diluted with ethyl acetate and washed

with 1 M HCl, brine, dried over Na 2 SO 4 , filtered, and

concentrated. Purification by column chromatography on

silica gel (2/1 Hex/EtOAc) afforded 7 as a yellow solid

(0.048 g, 23%). 1 H NMR (CDCI j , 300 MHz) 57.88-7.75 (m,

IH), 7.40-7.19 (m, 9H), 7.01-6.91 (m, IH), 6.65 (t, J =

3.1 Hz, IH), 5.61-5.41 (m, 3H} , 5.29-4.93 (m, 3H), 4.37-

4.32 (m, IH), 4.13-4.05 (m, IH), 3.56-3.35 (m, 4H), 2.77-

2.49 (m, IH), 2.09 (s, 3H), 2.04 (s, 3H} , 1.76-1.39 (m, 6H)

ppm.

|00123] Unprotected pyrrolidine (8) :

[00124} A mixture of 7 (0.09Og, 0.128 mmol) and 10%

palladium on activated carbon (0.020 mg, 20 wt %) in

methanol {8 mL) was shaken under a hydrogen atmosphere at

45 psi on a Parr hydrogenator for 2 hours. Thin layer

chromatography {10% MeOH/CH ? Cl 2 ) indicated no remaining 7.

The mixture was filtered through a 0.45 uM filtering disk

and. washed with MeOH. The filtrate was concentrated and

dried under high vacuum to give 8 as a white solid {0.049

g, 67%}. 1 H NMR (CDCl 3 , 300 MHz) 57.55-7.47 (m, IH), 7.32-

7.27 (m, 2H), 7.23-7.17 (m, 3H), 6.94 (t, J = 3.0 Hz, IH),

5.66 (m, IH), 5.17-5.11 (m, 2H), 4.27 (m, IH), 3.39-3.22

(m, 2H), 3.01-2.87 (m, IH), 2.76-2.65 {m, IH), 2.05 (s,

3H), 1.99 (S, 3H), 1.72-1.47 (m, 6H) ppm.

[00125] N-Acylated intermediate (9) :

[00126] A solution of CBZ-L-tert-leucine

dicyclohexylamine salt (0.057g, 0.129 mmol) and HATU (0.049

g, 0.129 mmol) in dry 1 -methyl- 2 -pyrrolidinone (NMP) (2 mL)

was cooled to 0 C and diisopropylethyl amine (0.022 g,

0.03 mL, 0.172 mmol) was added. After stirring for 15 min

a solution of 8 (0.049 g, 0.086 mmol) in NMP (2 mL) was

added and the reaction was stirred at 0 C for 2 hours

followed by room temperature for 1 hour. Thin layer

chromatography (2/1 Hex/EA) indicated no remaining 8. The

reaction was diluted with ether and washed with 1 M HCl,

water, saturated NaHCO 3 , brine, dried over Na 2 SO 4 ,

filtered, and concentrated. Purification by column

chromatography on silica gel (2/1 Hex/EtOAc) afforded 9 as

a foamy solid (0.041 g, 58%). 1 H NMR (CDC1 J7 300 MHz) δ8.74

(d, J = 1.3 Hz, IH) , 8.43 (dd, J - 0.04, 1.8 Hz, IH) , 8.03-

7.98 {m, IH) , 7.47-7.43 (m, IH) , 7.39-7.35 (m, 8H) , 7.02-

6.97 (m, IH) , 5.62-5.59 (m, IH) , 5.42-5.36 (m, IH) , 5.20-

5.06 (m 4H) , 4.33-4.29 (m, IH) , 3.61-3.29 {m, 2H) , 2.09-

1.98 (m, 3H) , 1.75-1.69 (m, 2H) , 1.56 (m, 6H) , 1.25-1.21

(m, 6H) , 1.04-0.97 (m, 6H) ppm.

[00127] Free amine (10) :

[00128] A mixture of 9 (0.041g, 0.050 mmol) and 10%

palladium on activated carbon (0.010 mg, 20 wt %) in

methanol (8 mL) was shaken under a hydrogen atmosphere at

45 psi on a Parr hydrogenation apparatus for 2 hours. Thin

layer chromatography (10% MeOH/CH 2 Cl 2 ) indicated no

remaining 9. The mixture was filtered through a 0.45 M

filtering disk and washed with MeOH. The filtrate was

concentrated and dried under high vacuum to give 10 as a

white solid (0.034 g, 99%). Mass spectrum, m/z- 684

[M+H] +.

[00129] Cbz-protected dipeptide (11) :

[00130] A solution of Cbz-N-methyl-L-alanine (O.OlSg,

0.074 mmol) and HATU (0.028 g, 0.074 mmol) in dry 1 -methyl-

2 -pyrrolidinone (NMP) (1 mL) was cooled to 0 C and

diisopropylethyl amine (0.012 g, 0.02 mL, 0.099 mmol) was

added. After stirring for 15 min, a .solution of 10 (0,034

g, 0.0497 mraol) in NMP (2 mL) was added and the reaction

was stirred at 0 C for 2 hours followed by room

temperature for 1 hour. Thin layer chromatography (1/1

Hex/EA) indicated no remaining 10. The reaction was

diluted with ether and washed with 1 M HCl, water,

saturated NaHCO 3 , brine, dried over Na 2 SO 4 , filtered, and

concentrated to afford 11 as a foamy solid (0.044 g, 98%

crude). 1 H NMR (CDCl 3 , 300 MHz) 68.06-7.97 (m, IH), 7.39-

7.33 (m, 9H), 7.27-7.22 (m, IH), 7.01-6.96 (m, IH), 5.65

(m, IH), 5.20-5.10 (m 4H), 5.05-5.00 (m, 2H), 3.94-3.89 (m,

IH), 3.65-3.59 (m, IH), 2.88-2.80 (m, 6H), 2.08-1.98 (m,

6H), 1.37-1.23 (m, 6H), 1.04-0.97 (m, 6H), 1.05-0.90 (m,

IH) ppm.

[00131] Hydroxylated intermediate (12) :

[00132] A solution of 11 (0.044 g, 0.049 mmol) in

methanol (2 mL) was cooled to 0 C and IM sodium hydroxide

(0.X6 mL, 0.16 mmol) was added. The reaction was stirred

for 45 minutes. Thin layer chromatography (10% MeOH/CH 2 Cl 2 )

indicated no remaining 11. The reaction was diluted with

brine and saturated ammonium chloride solution and

extracted with ethyl acetate (3X) . The organic phase was

dried over Na 2 SO 4 , filtered, and concentrated. The crude

product 12 (0.037 g) was taken on without further

purification. Mass spectrum, m/z = 777.8 [M+H3+.

|00133j Final dipeptide (13) :

A mixture of 12 (Q.037g, 0.049 mmol) and 10% palladium on

activated carbon (0.010 rag, 20 wt %) in methanol {8 mL) was

shaken under a hydrogen atmosphere at 45 psi on a Parr

hydrogenation apparatus for 1.5 h. Thin layer

chromatography {10% MeOH/CH 2 CI 2 ) indicated no remaining 12.

The mixture was filtered through a 0.45 M filtering disk

and washed with MeOH. Purification by reverse phase HPLC

and lyopholization gave 13 as the monoacetate salt (0.0107

g, 34%). 1 H NMR (CDCl 3 , 300 MHz) 57.99-7.95 (m, IH), 7.43-

7.31 (m, 3H), 7.24-7.18 (m, IH), 7.01-6.94 {m, IH), 4.85-

4.82 (m, IH), 4.45 (m IH), 4.12 (m, 2H), 3.39-3.37 (m, 3H),

3.32-3.20 (m, 4H), 2.41-2.28 (m, 4H), 2.03 (br S, 4H),

1.70 (m, IH), 1.51 (m, 2 H), 1.37-1.27 (m, 3H), 1.05-0.98

(m, 9H) ppm. Mass spectrum, m/z = 643.6 [M+H]+.

(00134) Table 1: Binding of Monomeric IAP Antagonists to XIAP BIR3.

S-

Me Me tβrt -Butyl 4 - F-pϊienyl Me H H L- rucoβe B

OH

R Me Me JR- (Me) CHOMe H 4-F-phenyl Me H H L- fucose B S Me Me tert -Butyl H 4-F-phenyl Me H H L-fucose B

S-

Me Me tert -Butyl 4 - F-pb.enyl H F L- fucose A

OH

S-

O Me Me B - (Me) CHOMe 4 - F-phenyl H F L- fucose A

OH

[00135] Preparation of a Dimeric IAP Antagonist (Formula

23, below. Referred to as Compound ["V,"] in Table 2, below.

II

100137} Indoylindoline (14) : Fluoroindole 1 {3.55 g,

10.0 mmol) was dissolved in trifluoroacetic acid (15 mL) at

0 0 C. After 3 h, the solvent was removed in vacuo and the

residue was dissolved in EtOAc. The EtOAc solution was

washed twice with saturated aqueous NaHCO 3 and once with

brine. The combined aqueous washes were twice back-

dried over anhydrous Na 2 SO 4 , filtered, and concentrated.

The crude product was purified by flash silica gel

chromatography (hexane/EtOAc , 2:1) to afford 2.48 g of 2 as

a foamy solid. Mass spectrum, m/z 705.1 [M + H]+.

[00138] Scheme III

[00139] Carbohydrate -linked indoylindoline (15) : A

mixture of 14 (1.24 g, 1.76 mmol) , L- ( - ) - fucose (0.87 g,

5.27 mmol), and powdered (NHJ 2 SO, (0.7O g, 5.27 mmol) in

absolute EtOH (25 mL) was heated at 75 0 C for 24 h. The

reaction mixture was absorbed onto silica gel and the

product was eluted using 2-5% MeOH in DCM. The fractions

containing the two diastereomeric products [TLC analysis:

10% MeOH/DCM, R r (14} = 0.8; R f (15) - 0.4 and 0.5] were

combined and concentrated to provide 0.89 g of 15 which was

used directly in the next reaction. Mass spectrum, m/z

851.2 [M + H] +.

me IV

(00141] Carbohydrate-linked biindole (16) : At ambient

temperature, DDQ (0,28 g, 1.25 mmol) was added to a

solution of 15 (0.89 g, 1.04 mmol) in 1,4-dioxane (10 mL) .

After 1 h, the reaction mixture was diluted with EtOAc and

washed three times with 0.5 M NaOH, once with brine, dried

over anhydrous Na ? SO 4 , filtered, and concentrated to afford

crude 16 which was used without further purification. Mass

spectrum, m/ z 849.2 [M + H] + .

Scheme V

|00143] Peracetylated intermediate (17) : To a solution

of crude 16 (1.04 mmol) in anhydrous pyridine (10 mL) was

added Ac,Q (1.07 g, 10.4 mmol) at ambient temperature.

After 16 h, the reaction mixture was diluted with EtOAc and

washed three times with IN HCl, once with brine, dried over

anhydrous Na 2 SO 4 , filtered, and concentrated. The crude

product was purified by flash silica gel chromatography

(EtOAc/hexane, 1:1) to afford 0.82 g of 17 as a solid.

Mass spectrum, m/z 975.2 [M]+.

[00144] Scheme VI

Bis-pyrrolidine (18) : A mixture of 17 (0.82 g, 0.84 mmol )

and 10% Pd-on-C (0.16 g, 20 wt %} in MeOH (20 mL} was

placed on a Parr apparatus and shaken under 50 PSI H ^

atmosphere. After 2 h, the reaction mixture was filtered

using a 0.45 μ filter disc which was subsequently washed

with excess MeOH. The clarified filtrate was concentrated

in vacuo to yield 0.57 g of crude 18 which was used without

further purification. Mass spectrum, m/z 354.6 [(M -f 2H) /2]+.

[00145| Scheme VII

[00146] Cbz -Valine-linked intermediate (19) : A solution

containing Cbz-L-VaI -OH (0.47 g, 1.87 mmol) and HATU (0.67

g, 1.77 mmol} in anhydrous NMP (5 mL} was cooled to 0 0 C.

DIPEA (0.28 g, 2.18 mmol) was added via syringe followed by

the addition of 18 (0.57 g, 0.81 mmol) in NMP (5 mL) . The

reaction mixture was slowly warmed to ambient temperature

and the reaction was maintained for -16 h. The reaction

mixture was diluted with diethyl ether and washed

successively with IN HCl, water (excess) , saturated aqueous

NaHCO 3 , and brine. The organic phase was dried over

anhydrous Na 2 SO 4 , filtered, and concentrated. The crude

product was purified by flash silica gel chromatography {2%

MeOH/DCM) to provide 0.76 g of 19 as a foamy solid. Mass

spectrum, m/ z 1174.4 (M + H)+.

[00147] Scheme VIII

[001481 Diamine (20) : A mixture of 19 (0.76 g, 0.65

mmol) and 10% Pd-on-C (0.15 g, 20 wt %) in MeOH (15 mL) was

placed on a Parr apparatus and shaken under 50 PSI H 2

atmosphere. After 3 h, the reaction mixture was filtered

using a 0.45 μ filter disc which was subsequently washed

with excess MeOH. The clarified filtrate was concentrated

in vacuo to yield 0.57 g of crude 20 which was used without

further purification. Mass spectrum, m/z 452.8 [M +

2H/2] +.

|00149] Scheme IX

BIs-[CbZ-N(Me)AIa] intermediate (21): A solution

containing CbZ-L-N(Me)AIa-OH (0.28 g, 1.18 tnmol) and HATU

(0.43 g, 1.13 rnmol) in anhydrous NMP (4 mL) was cooled to 0

0 C. DIPEA (0.18 g, 1.38 mmol) was added via syringe

followed by the addition of 20 (0.47 g, 0.51 mmol) in NMP

(4 mL) . The reaction mixture was slowly warmed to ambient

temperature and the reaction was maintained for about 16 h.

The reaction mixture was diluted with diethyl ether and

washed successively with IN HCl, water (excess), saturated

aqueous NaHCO 3 , and brine. The organic phase was dried over

anhydrous Na 2 SO 4 , filtered, and concentrated to provide 0.69

g of crude 21 as a solid. Mass spectrum, m/z 1343.4 (M)+.

[001501 Scheme X

Hydroxylated pyranose (22) : To a solution of 21 (0.69 g,

0.51 mmol) in MeOH (20 mL} was added IN NaOH (1.7 mL, 1.7

mmol) at 0 0 C. After 2 h, the reaction mixture was diluted

with saturated aqueous NH 4 Cl and brine and the product was

extracted with EtOAc. The combined organic extracts were

dried over anhydrous Na 2 SO 4 , filtered, and concentrated to

afford 0.62 g of crude 22 which was used without further

purification .

[00151] Scheme XI

Dimeric IAP Antagonist (23) : A mixture of 22 (0.62 g, 0.51

mmol) and 10% Pd-on-C (0.15 g, 20 wt %) in MeOH (20 mL) was

atmosphere. After 4 h, the reaction mixture was filtered

using a 0.45 μ filter disc which was subsequently washed

with excess MeOH. The clarified filtrate was concentrated

in vacuo. The crude product was purified by reverse-phase

HPLC (2" Dynamax Cl8 column; Flow rate: 40 mL/min;

Detector; 254 nm; Method: 10-50% ACN/water containing 0.1%

HOAc over 25 min) . The product-containing fractions were

combined and concentrated in vacuo to remove excess ACN

then lyophilized to dryness to provide 0.23 g of 23-2HOAc

as a flocculent white solid. Mass spectrum, m/ z 475.8 [(M +

2H)/2] +.

[00152] Table 2: Binding of Dimeric IAP Antagonists to XIAP BIR3

[00153] Preparation of Smac Mimetic (Piperidine-

substituted Monomer, Formula 30, below. Referred to as

Compound "[HH]" in Table 3, below.)

cheme XII

[00155) N-Alkylated indoline (24) : To a solution

containing indoline 2 (4.6 g, 13.0 tnmol) in glacial HOAc

{40 mL) was added 4-BOC-piperidone (2.85 g, 14.3 mmol) .

After 10 min, Na(AcO) 3 BH {4.13 g, 19.5 mmol) was added in

small portions over 40 min maintaining the temperature

below 30 0 C. After 1 h, the reaction mixture was diluted

with water and EtOAc. Aqueous NaOH (IM) was added and the

layers were separated. The organic phase was washed with

IM NaOH until pH = 12, then washed with brine, dried over

anhydrous Na 2 SO 4 , filtered and concentrated to afford crude

24 (quant.) as an oil which was used without further

purification. 1 H NMR (300 MHz, CDCl 3 ): 7.35-7.35 (m, 5H),

6.92-6.78 (m, IH), 6.25-6.18 (m, IH), 6.09-6.06 {m, IH),

5.12-5.09 {m, 2H), 4.22 (br, 2H), 3.87-3.70 (m, IH), 3.55-

3.41 (m, 3H) , 3.21-3.02 (m, 3H) , 2.75-2.72 (br, 2H} , 1.98-

1.60 (m, 8H) , 1.49-1.46 (m, 9H) ppm.

XIII

[00157] Indole (25) : To a solution containing crude

indoline 24 (7.46 g, 13 mmol) in anhydrous 1,4-dioxane (75

TTiL) was added 2 , 3 -dichloro-5 , 6~dicyanobenzoquinone (3.78 g,

16.6 mraol) in small portions. After 2 h, the reaction

mixture was diluted with EtOAc, and filtered through

Celite ® . The pad was washed with EtOAc and the filtrate

was washed with saturated aqueous NaHCO 3 , brine, dried over

anhydrous Na 2 SO 4 , filtered and concentrated. The product

was purified by flash silica gel column chromatography [2:1

hexane/EtOAc] to afford 3.81 g (51%) indole 25. 1 H NMR (300

MHz, CDClj) : -1:1 mixture of carbamate rotomers, 7.71-7.67

(m, 0.5H), 7.43-7.38 (m, 5H), 7.18-7.14 (m, 0.5H), 6.99-

6.82 (m, 2.5H), 6.61 (t, J = 2.9 Hz, 0.5H), 5.19 (s, 2H),

4.30-4.08 (m, 2H), 3.74-3.70 (m, 0.5H), 3.46-3.39 (m, 2H),

2.88 (br m, 2H) , 2.72-2.54 (m, IH) , 2.44 (t, J = 2.0 Uz,

0.5H) , 2.00 (br, IH) , 1.85-1.67 (m, 6H) , 1.49 (s, 9H) ppm.

[00158J Scheme XIV

[00159] Bromoindole (26) : A solution containing indole

25 {3.81 g, 7.12 mmol) in CHCl, (100 raL) was cooled to 0 0 C

and KOAc (2.1 g, 21.4 πirool) was added followed by the

dropwise addition of a solution of bromine (1.35 g, 8.54

mmol) in CHCl 3 (5 mL) . After 1 h, the reaction mixture was

diluted with brine and DCM. The layers were separated and

the organic phase was washed with saturated aqueous Na 2 S 2 O 4 ,

brine, dried over anhydrous Na 2 SO 4 , filtered and

concentrated. The product was purified by flash silica gel

column chromatography [2:1 hexane/EtOAc] to afford 2.78 g

(64%) of bromoindole 26 as a yellow solid. 3 H NMR (300 MHz,

CDCl 3 ): -1:1 mixture of carbamate rotomers, 7.76-7.71 (m,

0.5H), 7.45-7.32 (m, 5H), 7.13-7.06 (m, 1.5 H), 6.86 (t, J

= 2.6 Hz, 0.5H), 6.56 (t, J = 2.9 Hz), 0.5H), 5.22-5.18 <m,

2H), 4.59-4.55 (br, IH), 4.32-4.23 (br, 2H), 3.59-3.34 (m,

2H) , 3.29 (eld, J = 4.6, 1.0 Hz, 0.5H) , 3.07 (dd, J = 4.6,

1.2 Hz, 0.5H) , 2.86 (br, 2H) , 2.74-2.61 (m, IH) , 2.41 (br,

2H) , 1.76-1.65 (m, 5H) , 1.52 {s, 9H) ppm .

[00161] 2 -Substituted indole (27) : A mixture containing

bromoindole 26 (2.78 g, 4.52 mmol) , K 2 CO 3 (2.19 g, 11.3

mmol) and 4 -fluorobenzeneboronic acid {0.82 g, 5.88 mmol)

in toluene (21 mL) and EtOH (7 mL) was degassed under

vacuum. After the addition of (Ph 3 P) 1 Pd(O) (0.26 g, 0.23

mmol) , the mixture was degassed again and placed in an oil

bath preheated at 90 0 C. After 2.5 h, the reaction mixture

was cooled to ambient temperature and diluted with EtOAc,

washed with IM HCl, brine, dried over anhydrous Na 2 SO,,

filtered and concentrated. The product was purified by

flash silica gel column chromatography [2:1 hexane/EtOAc]

to afford 2.46 g (86%) of indole 27 as a yellow solid. 1 H

NMR (300 MHz, CDCl 3 ); -1:1 mixture of carbamate rotomers,

7.88-7.84 (πi, 0.5H), 7.38-7.14 {m, 10.5H), 6.94-6.87 (m, n cu \

(m, 2H) , 3.30-3.25 (m, 2H) , 3.03 (dd, J = 4.6 Hz, 1.3 Hz,

0.5H) , 2.82 (br, 0.5H) , 2.61-2.57 {m, 2H) , 2.44-2.28 (m,

4H) , 1.83-1.74 (m, IH) , 1.62-1.53 (m, 2H) , 1.52-1.46 {m,

9H) , 1.41-1.37 (m, 2H) ppm.

[0θ162J Scheme XVI

[00163] Cbz-protected amine (28) : A mixture containing

indole 27 (2,46 g, 3.91 mmol) and 10% Pd/C (480 mg, 20 wt

%) in MeOH (25 mL) was shaken under a hydrogen atmosphere

(50 psi) using a Parr apparatus. After 5 h, the reaction

mixture was filtered through Celite ® and the pad was washed

with MeOH. The filtrate was concentrated and the residue

was purified by flash silica gel column chromatography [2%

to 20% MeOH/DCM] to afford 550 mg of intermediate amine.

Mass spectrum, m/z - 495.6 [M+] .

[0θ164J A solution containing Cbz-L~ tert-leucine

dicyclohexylamine salt (644 mg, 1.44 mmol) and HATU (548

mg, 1.44 mmol) in NMP (7 mL) was cooled to 0 0 C and DIPEA

(0.29 g, 2.22 mmol) was added. After 15 min, a solution

containing the previously-prepared intermediate amine (550

ττig, 1.11 mmol) in NMP (5 mL) was added. The reaction

mixture was stirred to ambient temperature. After 16 h,

the reaction mixture was diluted with diethyl ether, washed

with IM HCl, water, saturated aqueous NaHCO 3 , water, and

brine. The organic phase was dried over anhydrous Na 2 SO 11 ,

filtered and concentrated. The product was purified by

flash silica gel column chromatography [1:1 hexane/EtOAc]

to afford 111 mg {13%) of amide 28 as a foam. 1 H NMR (300

MHz, CDCl 3 ): 8.03-7.99 (m, IH), 7.34-7.12 (m, 10H), 7.02-

6.91 (m, IH), 5.62-5.58 <m, IH), 5.16-5.02 (m, 2H), 4.49-

4.47 {m, IH), 4.32-4.08 (m, 2H), 3.99-3.94 (m, IH), 3.61-

3.43 (m, IH), 3.31-3.27 (m, IH), 2.59 (br, IH), 2.41-2.17

(m, 2H), 1.88-1.84 {m, IH), 1.57 (m, 5H), 1.49 (s, 9H),

1.31-1.19 (m, 4H), 1.01-0.95 (m, 9H) ppm. Mass spectrum,

m/z = 743.7 [M + H] +.

10 me XVII

(00166J Boc-protected peptide (29 ) : A mixture of amide

MeOH (10 mL) was shaken under a hydrogen atmosphere at (45

psi) using a Parr apparatus. After 2 h, the reaction

mixture was filtered through a 0.45 mM filter disc which

was rinsed with excess MeOH. The filtrate was concentrated

to afford 69 mg of intermediate amine. Mass spectrum, m/z = 608.7 [M+] .

[00167] A solution containing Boc-N (Me) Ala-OH (35 mg,

0.17 mmol) and HATU (65 mg, 0.17 mmol} in NMP (2 mL) was

cooled to 0 0 C and DIPEA (0.029 g, 0.23 mmol) was added.

After 15 min, a solution containing the previously-prepared

intermediate amine (69 mg, 0.11 mmol) in NMP {3 mL) was

added. The reaction was stirred to ambient temperature

over 2 h then diluted with diethyl ether and washed

successively with IM HCl, water, saturated aqueous NaHCO^,

water, and brine. The organic phase was dried over

anhydrous Na 2 SO 4 , filtered and concentrated to afford 105 mg

of crude Boc-peptide 29 as a foam which was used directly

without further purification. Mass spectrum, m/z = 793.9

[M + ] .

[00168) Scheme XVIII

[00169J Dipeptide (30) : To a solution containing Boc-

peptide 29 (100 mg, 0.13 mmol) in DCM (10 tnL) was added TFA

(2 mL) at 0 0 C. After 1 h, the solvent was removed in vacuo

and the residue was dissolved in EtOAc, washed

successively with saturated aqueous NaHCO 3 , brine, dried

over anhydrous Na^SO 4 , filtered and concentrated. The crude

product was purified by C18 reverse-phase HPLC [10% to 70%

ACN/water containing 0.1% v/v HOAc]. Lyophilization of the

product-containing fractions afforded 28 mg (38%) of

dipeptide 30 as a white solid. 1 H NMR (300 MHz, CDCl 3 ) :

8.03-7.98 (m, IH), 7.86-7.69 (m, 2H), 7.59-7.46 (m, IH),

7.28-7.17 (m, 5H), 7.05-6.91 (m, IH), 4.94-4.77 (br m, 5H),

4.59 (d, J = 3.2 Uz 1 IH), 4.49-4.46 (m, IH), 3.97 (br m,

2H), 3.72-3.20 (m, 3H), 2.13-3.05 (m, IH), 2.89-2.62 (m,

3H), 2.40-2.18 (m, 2H), 2.08-1.94 (m, 3H), 1.08-0.95 (m,

10H) ppiru Mass spectrum, m/z = 593.7 [M+] .

|00170] Table 3. Binding of Monomeric IAP Antagonists to

[00171) in mammalian cells, activation of the caspases

is achieved through at least two independent mechanisms

which are initiated by distinct caspases, but result in the

activation of common executioner (effector) caspases. In

addition to the cytochrome c activated mechanism (somphimpQ

referred to as the 'intrinsic death pathway'} is a

mechanism by which the caspase cascade is activated via

activation of a death receptor located on the cell membrane

(sometimes referred to as the 'extrinsic death pathway 1 }.

Examples of death receptors include CD- 95 and TNF-Rl {as

well as other members of the TNF group of cytokine

receptors) . The corresponding ligands are CD-95L and TNF-

alpha, respectfully. Binding of pro-caspase-8 to the death

receptor induces auto-activation wherein the inhibitory

pro-domain of pro-caspase-8 is cleaved and removed.

Caspase-8 is released from the receptor and can then

activate effector caspases (caspase-3, -6, -7), and, as in

the caspase- 9 initiated pathway, the result is the

proteolytic cleavage of cellular targets by the effector

caspases and the induction of apoptosis.

[00172J The present invention is directed generally to

Smac peptidomimetics and the uses of Smac peptidomimetics .

In one embodiment the Smac peptidomimetics act as

chemopotentiating agents. The term "chemopotentiating

agent" refers to an agent that acts to increase the

sensitivity of an organism, tissue, or cell to a chemical

compound, or treatment namely "chemotherapeutic agents" or

"chemo drugs 11 or radiation treatment. One embodiment of

the invention is the therapeutic composition of a Smac

peptidomimetic . A further embodiment of the invention is

the therapeutic composition of a Smac peptidomimetic, which

can act as a chemopotentiating agent (herein referred to as

Smac mimetic) , and a biological or chemotherapeutic agent

or radiation. Another embodiment of the invention is a

method of inhibiting tumor growth in vivo by administering

a Smac peptidomimetic. Another embodiment of the invention

is a method of inhibiting tumor growth in vivo by

administering a Smac mimetic and a biologic or

chemotherapeutic agent or chemoradiation. Another

embodiment of the invention is a method of treating a

patient with a cancer by administering Smac mimetics of the

present invention alone or in combination with a

chemotherapeutic agent or chemoradiation.

[00173} In an embodiment of the present invention, the

cells are in situ, in an individual, and the contacting

step is effected by administering a pharmaceutical

composition comprising a therapeutically effective amount

of the Smac mimetic wherein the individual may be subject

to concurrent or antecedent radiation or chemotherapy for

treatment of a neoproliterative pathology. The pathogenic

cells are of a tumor such as, but not limited to, bladder

cancer, breast cancer, prostate cancer, lung cancer,

pancreatic cancer, gastric cancer, colon cancer, ovarian

cancer, renal cancer, hepatoma, melanoma, lymphoma,

sarcoma, and combinations thereof.

|00174) As described in US 7,244,851, IAP antagonists

can be used for the treatment of all cancer types which

fail to undergo apoptosis. Examples of such cancer types

include neuroblastoma, intestine carcinoma such as rectum

carcinoma, colon carcinoma, familiary adenomatous polyposis

carcinoma and hereditary non-polyposis colorectal cancer,

esophageal carcinoma, labial carcinoma, larynx carcinoma,

hypopharynx carcinoma, tong carcinoma, salivary gland

carcinoma, gastric carcinoma, adenocarcinoma, medullary

thyroidea carcinoma, papillary thyroidea carcinoma, renal

carcinoma, kidney parenchym carcinoma, ovarian carcinoma,

cervix carcinoma, uterine corpus carcinoma, endometrium

carcinoma, chorion carcinoma, pancreatic carcinoma,

prostate carcinoma, testis carcinoma, breast carcinoma,

urinary carcinoma, melanoma, brain tumors such as

glioblastoma, astrocytoma, meningioma, medulloblastoma and

peripheral neuroectodermal tumors, Hodgkin lymphoma, non-

Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic

leukemia (ALL) , chronic lymphatic leukemia (CLL) , acute

myeloid leukemia (AML) , chronic myeloid leukemia (CML) ,

adult T~cell leukemia lymphoma, hepatocellular carcinoma,

gall bladder carcinoma, bronchial carcinoma, small cell

lung carcinoma, non-small cell lung carcinoma, multiple

myeloma, basalioma, teratoma, retinoblastoma, choroidea

melanoma, seminoma, rhabdomyo sarcoma, craniopharyngeoma,

osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma,

fibrosarcoma, Ewing sarcoma and plasmocytoma.

[00175] In addition to apoptosis defects found in

tumors, defects in the ability to eliminate self -reactive

cells of the immune system due to apoptosis resistance are

considered to play a key role in the pathogenesis of

autoimmune diseases . Autoimmune diseases are characterized

in that the cells of the immune system produce antibodies

against its own organs and molecules or directly attack

tissues resulting in the destruction of the latter. A

failure of those self -reactive cells to undergo apoptosis

leads to the manifestation of the disease. Defects in

apoptosis regulation have been identified in autoimmune

diseases such as systemic lupus erthematosus or rheumatoid

arthritis

[00176] In an embodiment the pathogenic cells are those

of any autoimmune disease or diseases which are resistant

to apoptosis due to the expression of IAPs or members of

the BcI -2 family Examples of such autoimmune diseases are

collagen diseases such as rheumatoid arthritis, systemic

lupus erythematosus, Sharp's syndrome, CREST syndrome

(calcinosis, Raynaud's syndrome, esophageal dysmotility,

telangiectasia), dermatomyositis, vasculitis (Morbus

Wegener's) and Sjogren's syndrome, renal diseases such as

Goodpasture's syndrome, rapidly-progressing

glomerulonephritis and membrano-proliferative

glomerulonephritis type II, endocrine diseases such as

type- I diabetes, autoimmune polyendocπnopathy-candidiasis-

ectodermal dystrophy (APECED) , autoimmune parathyroidism,

pernicious anemia, gonad insufficiency, idiopathic Morbus

Addison's, hyperthyreosis, Hashimoto's thyroiditis and

primary myxedema, skxn diseases such as pemphigus vulgaris,

bullous pemphigoid, herpes gestatioms, epidermolysis

bullosa and erythema multiforme major, liver diseases such

as primary biliary cirrhosis, autoimmune cholangitis,

autoimmune hepatitis type-1, autoimmune hepatitis type- 2,

primary sclerosing cholangitis, neuronal diseases such as

multiple sclerosis, myasthenia gravis, myasthenic Lambert -

Eaton syndrome, acquired neuromyotony, Guillain-Barre

syndrome (Mύller-Fischer syndrome}, stiff-man syndrome,

cerebellar degeneration, ataxia, opsoklonus, sensoric

neuropathy and achalasia, blood diseases such as autoimmune

hemolytic anemia, idiopathic thrombocytopenic purpura

(Morbus Werlhof) , infectious diseases with associated

autoimmune reactions such as AIDS, Malaria and Chagas

disease .

[00177) The subject compositions encompass

pharmaceutical compositions comprising a therapeutically

effective amount of a Smac mimetic in dosage form and a

pharmaceutically acceptable carrier, wherein the Smac

mimetic inhibits the activity of an Inhibitor of Apoptosis

protein (IAP), thus promoting apoptosis. Another

embodiment of the present invention are compositions

comprising a therapeutically effective amount of a Smac

mimetic in dosage form and a pharmaceutically acceptable

carrier, in combination with a chemotherapeutic and/or

radiotherapy, wherein the Smac mimetic inhibits the

activity of an Inhibitor of Apoptosis protein (IAP), thus

promoting apoptosis and enhancing the effectiveness of the

chetnotherapeutic and/or radiotherapy.

[00178] In an embodiment of the invention a therapeutic

composition for promoting apoptosis can be a

therapeutically effective amount of a Smac peptidomimetic

which binds to at least one IAP. In one embodiment the IAP

can be XIAP. In another embodiment the IAP can be ML-IAP.

In another embodiment the IAP can cIAP-1 or cIAP-2. In a

further embodiment the IAP can be multiple IAP types.

{00179) Embodiments of the invention also include a

method of treating a patient with a condition in need

thereof wherein administration of a therapeutically

effective amount of a Smac peptidomimetic is delivered to

the patient, and the Smac peptidomimetic binds to at least

one IAP. In one embodiment the IAP can be XIAP. In

another embodiment the IAP can be ML-IAP. In another

embodiment the IAP can cIAP-l or cIAP-2. In a further

embodiment the IAP can be multiple IAP types.

The method may further include the concurrent

administration of another chemotherapeutic agent . The

chemotherapeutic agent can be, but is not limited to,

alkylating agents, antimetabolites, anti- tumor antibiotics,

taxanes, hormonal agents, monoclonal antibodies,

glucocorticoids, mitotic inhibitors, topoisomerase I

inhibitors, topoisomerase II inhibitors, immunomodulating

agents, cellular growth factors, cytokines, and

nonsteroidal anti- inflammatory compounds.

[00180] Administration of Smac peptidomimetics The Smac

peptidomimetics can be administered in effective amounts.

An effective amount is that amount of a preparation that

alone, or together with further doses, produces the desired

response. This may involve only slowing the progression of

the disease temporarily, although preferably, it involves

halting the progression of the disease permanently or

delaying the onset of or preventing the disease or

condition from occurring. This can be monitored by routine

methods. Generally, doses of active compounds would be

from about 0.01 mg/kg per day to 1000 mg/kg per day. It is

expected that doses ranging from 50-500 mg/kg will be

suitable, preferably intravenously, intramuscularly, or

intradermalIy, and in one or several administrations per

day. The administration of the Smac peptidomimetic can

occur simultaneous with, subsequent to, or prior to

chemotherapy or radiation so long as the chemotherapeutic

agent or radiation sensitizes the system to the Smac

peptidomimetic .

[00181] In general, routine experimentation in clinical

trials will determine specific ranges for optimal

therapeutic effect for each therapeutic agent and each

administrative protocol, and administration to specific

patients will be adjusted to within effective and safe

ranges depending on the patient condition and

responsiveness to initial administrations. However, the

ultimate administration protocol will be regulated

according to the judgment of the attending clinician

considering such factors as age, condition and size of the

patient, the Smac peptidomimetic potencies, the duration of

the treatment and the severity of the disease being

treated. For example, a dosage regimen of the Smac

peptidomimetic can be oral administration of from 1 mg to

2000 mg/day, preferably 1 to 1000 mg/day, more preferably

50 to 600 mg/day, in two to four (preferably two) divided

doses, to reduce tumor growth. Intermittent therapy (e.g.,

one week out of three weeks or three out of four weeks) may

also be used.

|00182] In the event that a response in a subject is

insufficient at the initial doses applied, higher doses (or

effectively higher doses by a different, more localized

delivery route) may be employed to the extent that the

patient tolerance permits . Multiple doses per day are

contemplated to achieve appropriate systemic levels of

compounds. Generally, a maximum dose is used, that is, the

highest safe dose according to sound medical judgment.

Those of ordinary skill in the art will understand,

however, that a patient may insist upon a lower dose or

tolerable dose for medical reasons, psychological reasons

or for virtually any other reason.

[00183] Embodiments of the invention also include a

method of treating a patient with cancer by promoting

apoptosis wherein administration of a therapeutically

effective amount of a Smac peptidomimetic, and the Smac

peptidomirnetic binds to at least one IAP, In one

embodiment the IAP can be XIAP . In another embodiment the

IAP can be ML-IAP, In another embodiment the IAP can cIAP-

1 or cIAP- 2. In a further embodiment the IAP can be

multiple IAP types. The method may further include

concurrent administration of a chemotherapeutic agent. The

chemotherapeutic agent can be, but is not limited to,

alkylating agents, antimetabolites, anti-tumor antibiotics,

taxanes, hormonal agents, monoclonal antibodies,

glucocorticoids, mitotic inhibitors, topoisomerase I

inhibitors, topoisomerase II inhibitors, immunomodulating

agents, cellular growth factors, cytokines, and

nonsteroidal anti -inflammatory compounds.

|00184] Routes of administration A variety of

administration routes are available. The particular mode

selected will depend, of course, upon the particular

chemotherapeutic drug selected, the severity of the

condition being treated and the dosage required for

therapeutic efficacy. The methods of the invention,

generally speaking, may be practiced using any mode of

administration that is medically acceptable, meaning any

mode that produces effective levels of the active compounds

without causing clinically unacceptable adverse effects.

Such modes of administration include, but are not limited

to, oral, rectal, topical, nasal, intradermal, inhalation,

intra-peritoneal , or parenteral routes. The term

"parenteral" includes subcutaneous, intravenous,

intramuscular, or infusion. Intravenous or intramuscular

routes are particularly suitable for purposes of the

present invention.

[00185] In one aspect of the invention, a Smac

peptidomimetic as described herein, with or without

additional biological or chemotherapeutic agents or

radiotherapy, does not adversely affect normal tissues,

while sensitizing tumor cells to the additional

chemotherapeutic/radiation protocols . While not wishing to

be bound by theory, it would appear that because of this

tumor specific induced apoptosis, marked and adverse side

effects such as inappropriate vasodilation or shock are

minimized. Preferably, the composition or method is

designed to allow sensitization of the cell or tumor to the

chemotherapeutic or radiation therapy by administering at

least a portion of the Smac peptidomimetic prior to

chemotherapeutic or radiation therapy. The radiation

therapy, and/or inclusion of chemotherapeutic agents, may

be included as part of the therapeutic regimen to further

potentiate the tumor cell killing by the Smac

peptidomimetic .

[00186] Pharmaceutical compositions In one embodiment

of the invention, an additional chemotherapeutic agent

(infra) or radiation may be added prior to, along with, or

following the Smac peptidomimetic . The term

"pharmaceutically-acceptable carrier" as used herein means

one or more compatible solid or liquid fillers, diluents or

encapsulating substances which are suitable for

administration into a human. The term "carrier" denotes an

organic or inorganic ingredient, natural or synthetic, with

which the active ingredient is combined to facilitate the

application. The components of the pharmaceutical

compositions also are capable of being co-mingled with the

molecules of the present invention, and with each other, in

a manner such that there is no interaction which would

substantially impair the desired pharmaceutical efficacy.

[00187] The delivery systems of the invention are

designed to include time-released, delayed release or

sustained release delivery systems such that the delivering

of the Smac peptidomimetic occurs prior to, and with

sufficient time, to cause sensitization of the site to be

treated. A Smac peptidomimetic may be used in conjunction

with radiation and/or additional anti-cancer chemical

agents (infra) . Such systems can avoid repeated

administrations of the Smac peptidomimetc compound,

increasing convenience to the subject and the physician,

and may be particularly suitable for certain compositions

of the present invention.

|00188j Many types of release delivery systems are

available and known to those of ordinary skill in the art.

They include polymer base systems such as poly (lactide-

glycolide) , copolyoxalates, polycaprolactones,

polyesteramides, polyorthoesters , polyhydroxybutyric acid,

and polyanhydrides . Microcapsules of the foregoing

polymers containing drugs are described in, for example,

U.S. Pat. No. 5,075,109. Delivery systems also include

non-polymer systems that are: lipids including sterols such

as cholesterol, cholesterol esters and fatty acids or

neutral fats such as mono-di-and tri-glycerides; hydrogel

release systems; sylastic systems; peptide based systems;

wax coatings; compressed tablets using conventional binders

and excipients; partially fused implants; and the like.

Specific examples include, but are not limited to: (a)

erosional systems in which the active compound is contained

in a form within a matrix such as those described in U.S.

Pat. Nos, 4,452,775, 4,667,014, 4,748,034 and 5,239,660

and (b) diffusional systems in which an active component

permeates at a controlled rate from a polymer such as

described in U.S. Pat. Nos . 3,832,253, and 3,854,480. In

addition, pump-based hardware delivery systems can be used,

some of which are adapted for implantation.

[00189] use of a long-term sustained release implant may

be desirable. Long-term release, are used herein, means

that the implant is constructed and arranged to deliver

therapeutic levels of the active ingredient for at least 30

days, and preferably 60 days. Long-term sustained release

implants are well-known to those of ordinary skill in the

art and include some of the release systems described

above .

[00190] The pharmaceutical compositions may conveniently

be presented in unit dosage form and may be prepared by any

of the methods well known in the art of pharmacy. All

methods include the step of bringing the active agent into

association with a carrier that constitutes one or more

accessory ingredients. In general, the compositions are

prepared by uniformly and intimately bringing the active

compound into association with a liquid carrier, a finely

divided solid carrier, or both, and then, if necessary,

shaping the product .

(00191] Compositions suitable for parenteral

administration conveniently comprise a sterile aqueous

preparation of a chemopotentiatmg agent (e.g. Smac

peptidomimetic) , which is preferably isotonic with the

blood of the recipient. This aqueous preparation may be

formulated according to known methods using suitable

dispersing or wetting agents and suspending agents. The

sterile injectable preparation also may be a sterile

injectable solution or suspension in a non-toxic

parenterally-acceptable diluent or solvent, for example, as

a solution in 1, 3 -butane diol . Among the acceptable

vehicles and solvents that may be employed are water,

Ringer's solution, and isotonic sodium chloride solution.

In addition, sterile, fixed oils are conventionally

employed as a solvent or suspending medium. For this

purpose any bland fixed oil may be employed including

synthetic mono-or di-glycerxdes . In addition, fatty acids

such as oleic acid may be used in the preparation of

injectables Carrier formulation suitable for oral,

subcutaneous, intravenous, intramuscular, etc

administrations can be found in. Remington's Pharmaceutical

Sciences, Mack Publishing Co., Easton, PA which is

incorporated herein in its entirety by reference thereto,

[00192J Addi tiona.1 chemotherapeutic agents

Cheπrotherapeutic agents suitable, include but are not

limited to the chemotherapeutic agents described in "Modern

Pharmacology with Clinical Applications", Sixth Edition,

Craig & Stitzel, Chpt . 56, pg 639-656 (2004), herein

incorporated by reference. This reference describes

chemotherapeutic drugs to include alkylating agents,

antimetabolites, anti- tumor antibiotics, plant-derived

products such as taxanes, enzymes, hormonal agents such as

glucocorticoids, miscellaneous agents such as cisplatin,

monoclonal antibodies, immunomodulating agents such as

interferons, and cellular growth factors. Other suitable

classifications for chemotherapeutic agents include mitotic

inhibitors and nonsteroidal anti -estrogenic analogs. Other

suitable chemotherapeutic agents include toposiomerase I

and II inhibitors and kinase inhibitors.

[00193} Specific examples of suitable biological and

chemotherapeutic agents include, but are not limited to,

cisplatin, carmustine (BCNU), 5 -flourouracil (5-FU),

cytarabine (Ara-C) , gemcitabine, methotrexate,

daunorubicln, doxorubicin, dexamethasone, topotecan,

etoposide, paclitaxel, vincristine, tamoxifen, TNF-alpha,

TRAIL, interferon (in both its alpha and beta forms) ,

thalidomide, and melphalan. Other specific examples of

suitable chemotherapeutic agents include nitrogen mustards

such as cyclophosphamide, alkyl sulfonates, nitrosoureas,

ethylenimines , triazenes, folate antagonists, purine

analogs, pyrimidine analogs, anthracyclines, bleomycins,

mitomycins, dactinomycins , plicamycin, vinca alkaloids,

epipodophyllotoxins, taxanes, glucocorticoids, L-

asparaginase, estrogens, androgens, progestins, luteinizing

hormones, octreotide actetate, hydroxyurea, procarbazine,

mitotane, hexamethylmelamine, carboplatin, mitoxantrone ,

monoclonal antibodies, levamisole, interferons,

interleukins , filgrastim and sargramostim.

Chemotherapeutic compositions also comprise other members,

i.e., other than TRAIL, of the TNF superfarnily of

compounds .

|00194) Radiotherapy protocols Additionally, in several

method embodiments of the present invention the Smac

peptidomimetic therapy may be used in connection with

chemo-radiation or other cancer treatment protocols used to

inhibit tumor cell growth.

|00I95J For example, but not limited to, radiation

therapy (or radiotherapy) is the medical use of ionizing

radiation as part of cancer treatment to control malignant

cells is suitable for use in embodiments of the present

invention. Although radiotherapy is often used as part of

curative therapy, it is occasionally used as a palliative

treatment, where cure is not possible and the aim is for

symptomatic relief . Radiotherapy is commonly used for the

treatment of tumors . It may be used as the primary

therapy. It is also common to combine radiotherapy with

surgery and/or chemotherapy. The most common tumors

treated with radiotherapy are breast cancer, prostate

cancer, rectal cancer, head & neck cancers, gynecological

tumors, bladder cancer and lymphoma. Radiation therapy is

commonly applied just to the localized area involved with

the tumor. Often the radiation fields also include the

draining lymph nodes. It is possible but uncommon to give

radiotherapy to the whole body, or entire skin surface.

Radiation therapy is usually given daily for up to 35-38

fractions {a daily dose is a fraction) . These small

frequent doses allow healthy cells time to grow back,

repairing damage inflicted by the radiation. Three main

divisions of radiotherapy are external beam radiotherapy or

teletherapy, brachytherapy or sealed source radiotherapy

and unsealed source radiotherapy, which are all suitable

examples of treatment protocol in the present invention.

The differences relate to the position of the radiation

source; external is outside the body, while sealed and

unsealed source radiotherapy has radioactive material

delivered internally. Brachytherapy sealed sources are

usually extracted later, while unsealed sources are

injected into the body. Administration of the Smac

peptidomiinetic may occur prior to, concurrently with the

treatment protocol. Annexin V/Propidium Iodide Staining-To

show the ability of Smac mimetics to induce apoptosis,

Annexin V- fluorescein isothiocyanate staining was performed

as per manufacturer's protocol {Invitrogen, Carlsbad, CA).

Briefly, cells were exposed to various concentrations of

Smac mimetics for 18-24 hrs . and then removed from the

assay plate by trypsinization . Cells were then pelleted

and resuspended m assay buffer (supplied by manufacturer) ,

Annexin V and propidium iodide were added to the cell

temperature . Following the incubation additional buffer

(200 μl) was then added to each tube, and the samples were

analyzed immediately by flow cytometry. In the presence of

Smac mimetics apoptosis was strongly promoted, as assessed

by annexin/PI staining and analyzed by flow cytometry. The

amplification in the number of apoptotic cells (Annexin V

positive/propidium iodide negative - lower right quadrant)

by IAP antagonists as compared to control was dose

dependent and due to the induction of apoptosis and not via

increasing the proportion of necrotic cells.

[00196) Biological and chemotherapeutics/anti -neoplastic

agents and radiation induce apoptosis by activating the

extrinsic or intrinsic apoptotic pathways, and, since Smac

mimetics relieve inhibitors of apoptotic proteins (IAPs)

and, thus, remove the block in apoptosis, the combination

of chemotherapeutics/anti-neoplastic agents and radiation

with Smac mimetics should work synergistically to

facilitate apoptosis.

|00197] The relevance of this potent synergy is that it

makes possible the use of the Smac peptidomimetics, which

are IAP antagonists, to improve the efficacy of the

marketed platinum containing compounds (cisplatin and

carboplatin) . This may be accomplished by lowering the

required dose of the poorly tolerated platinum containing

compounds and/or by improving the response rate at the

marketed dose,

[00198] The present invention is not limited to the

embodiments described and exemplified above, but is capable

of variation and modification within the scope of the

appended claims ,