Title of Invention

SUBSTITUTED IMIDAZOLE COMPOUNDS AS KSP INHIBITORS

Abstract The present invention relates to new substituted imidazole compounds and pharmaceutically acceptable salts,esters or prodrugs thereof, compositions of the derivatives together with pharmaceutically acceptable carriers, and uses of the compounds.The compounds of the invention have the following general formula (I).
Full Text

SUBSTITUTED IMIDAZOLE COMPOUNDS AS KSP INHIBITORS
BACKGROUND OF THE INVENTION
Grass Reference to Related Applications
[0001] This application claims the benefit under 35 U.S.C. §119(e) of
United States Provisional Application Serial Number 60/706,901, filed August 9, 2005,
which is hereby incorporated by reference in its entirety.
Field of the Invention
[0002] The present invention relates substituted imidazole compounds and
pharrnaceutically acceptable salts, esters or prodrugs thereof, compositions of these
compounds together with pharrnaceutically acceptable carriers, and uses of these compounds.
State of the Art
[0003] Kinesihs are motor proteins that use adenosine triphosphate to bind to
microtubules andgenerate mechanical force. Kinesins are characterized by a motor domain
haylng about 350 amino acid residues. The crystal structures of several kinesin motor
domains have been resolved.
[0004] Currently,about one hundred kinesin-related proteins (KRP) have
been identified. Kinesins are involved in a variety of cell biological processes including transport of organelles and vesicles, and maintenance of the endoplasmic reticulum. Several
KRPs interact with the microtubules of the mitotic spindle or with the chromosomes directly
and appear to play a pivotal role during the mitotic stages of the cell cycle. These mitotic
KRPs are of particular interest for the development of cancer therapeutics.
[0005] Kinesin spindle protein.(KSP) (also known as Eg5, HsEg5, KNSL1,
or K1F11) is one of several kinesin-like motor proteins that are localized to the mitotic
spindle and known to be required for formation and/or function of the bipolar mitotic spindle..
[0006] In 1995, the depletion of KSP using an antibody directed against the
C-terminus of KSP was shown to arrest HeLa cells in mitosis with monoastral microtubule
arrays (Slangy et-al., Cell 83:1159-1169,; 1995). Mutations in bimC and cut7 genes, which
are considered to be homologues of KSP, cause failure in centrosome separation in


Aspergillus nidulans(Enos, A.P., andN.R. Morris, Cell 60:1019-1027, 1990) and
Schizosaccharomyces pombe (Hagan, I., and M; Yanagida, Nature 347:563-566, 1990).
Treatment of cells with either ATRA (all trans-retinoic acid), which reduces KSP expression
on the protein level, or depletion of KSP using antisense oligonucleotides revealed a
significant growth inhibition in DAN-G pancreatic carcinoma cells indicating that KSP might
be involved in the antiproliferative action of all trans-retinoic acid (Kaiser, A., et al., J. Biol.
Chem., 2.74,. 1,8925-18931, 1999). Interestingly, the Xenopus laevis Aurora-related protein
kinase pEg2 was shown to associate and phosphorylate XlEg5 (Giet, R., et al., J. Biol. Chem.
274:15005-15013, 1999). Potential substrates of Aurora-related kinases are of particular
interest for cancer drug development. For example, Aurora 1 and 2 kinases are overexpressed on the protein and RNA level and the genes are amplified in colon cancer
patients.
[0007] The first cell permeable small molecule inhibitor for KSP,
"monastrol," was shown to arrest cells with monopolar spindles without affecting
microtubule polymerization as do conventional chemotherapeutics such as taxanes and vinca
alkaloids (Mayer, T.U., et al, Science 286:971-91'4, 1999). Monastrol was identified as an
inhibitor in phenotype-based screens and it was suggested that this compound may serve as a
lead for the development of anticancer drugs. The inhibition was determined not to be
competitive in respect to adenosine triphosphate and to be rapidly reversible (DeBonis, S.,
et al., Biochemistry, 42:338-349, 2003; Kapoor, T.M., et al., J. Cell Biol., 150:975-988,
2000).
[0008] In light of the importance of improved chemotherapeutics, there is a
need for KSP inhibitors that are effective in vivo inhibitors of KSP and KSP-related proteins.
SUMMARY OF THE INVENTION
[0009] This invention is directed to substituted imidazole compounds
represented by the formula I:



wherein:
R1 is selected from the group consisting of aminoacyl, acylamino, carboxyl, carboxyl
ester, alkyl, and substituted alkyl with the proviso that substituted alkyl is not substituted with
aryl or substituted aryl;
R2 is selected from the group consisting of hydrogen, alkyl, and aryl;
R3 and R4 are independently selected from the group consisting of hydrogen, hydroxy,
alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic provided that only 1 of R3 or
R4 is hydroxy;
or R3 andR4 together, with the nitrogen atom pendent thereto join to form a
heterocyclic or substituted heterocyclic;
R5 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl,
heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, and substituted
heteroaryl;
or R1 and R5 together with the carbon and nitrogen atoms bound respectively thereto
join to form a heterocyclic or substituted heterocyclic group;
or when R1 and R5, together with the carbon and nitrogen atoms bound respectively thereto, do not form a heterocyclic group, then R4 and R5, together with the atoms bound
thereto, form a heterocyclic or substituted heterocyclic group;
R8 is selected from the group consisting of L-A1 , wherein L is selected from the group
consisting of-S(O)q- where q is one or two, and C1 to C5 alkylene optionally substituted with
hydroxy, halo, or acylamino; and

A1 is selected from the group consisting of aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, and substituted
cycloalkyl; and

oneof either R6 or R7 is selected from the group consisting of cycloalkyl,
. heterocyclic, aryl and heteroaryl, all of which may be optionally substituted with -(R9)m.
where R9 is as defined herein and m is an integer from 1 to 4, and
the other of R6 of R7 is selected from the group consisting of hydrogen, halo, and
alkyl;
R9 is selected from the group consisting of cyano, alkyl, substituted alkyl, alkenyl,. .
substituted alkenyl, alkynyl, substituted alkynyl, -CF3, alkoxy, substituted alkoxy, halo, and
hydroxy;
and when m is an integer from 2 to 4, then each R9 may be the same or different;
or pharmaceutically acceptable salts, esters or prodrugs thereof.
DETAILED DESCRIPTION OF THE INVENTION
A. Compounds of the Invention
[0010] As stated above, compounds of the invention include those of
formula I:

wherein:
R1 is selected from the group consisting of aminoacyl, acylamino, carboxyl, carboxyl
ester, alkyl, and substituted alkyl with the proviso that substituted alkyl is not substituted with
aryl or substituted aryl;
R2 is selected from the group consisting of hydrogen, alkyl, and aryl;
R3 and R4 are independently selected from the group consisting of hydrogen, hydroxy,
alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,

substituted heterparyl, heterocyclic, and substituted heterocyclic provided that only 1 of R3 or
R4 is hydroxy;
or R3 and R4 together with the nitrogen atom pendentthereto join to form a
heterocyclic, or substituted heterocyclic;-
R5 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl,
heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, and substituted
heteroaryl;
or R1 and R5, together with the carbon and nitrogen atoms bound respectively thereto
join to form a heterocyclic or substituted heterocyclic group;
or when R1 and R5, together with the carbon and nitrogen atoms bound respectively
thereto, do not form a heterocyclic group, then R4 and R5, together with the atoms bound
thereto, form a heterocyclic or substituted heterocyclic group;
R8 is selected from the group consisting of L-A1, wherein L is selected from the group
consisting of -S(O)q- where q is one or two, and C1 to C5 alkylene optionally substituted with
hydroxy, halo, or acylamino; and
A1 is selected from the group consisting of aryl, substituted aryl, heteroaryl,
substituted heteroaryl heterocyclic, substituted heterocyclic, cycloalkyl, and substituted
cycloalkyl; and
one of either R6 or R7 is selected from the group consisting of cycloalkyl,
heterocyclic, aryl and heteroaryl, all of which may be optionally substituted with -(R 9)m
where R9 is as defined herein and m is an integer from 1 to 4, and
the other of R6 or R7 is selected from the group consisting of hydrogen, halo, and
alkyl;
R9 is selected from the group consisting of cyano, alkyl, substituted alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, -CF3, alkoxy, substituted alkoxy, halo, and
hydroxy;
and when m is an integer from 2 to 4, then each R9 may be the same or different;
or pharmaceutically acceptable salts, esters or prodrugs thereof.
[0011] In one embodiment of the invention, compounds of the invention are
represented by formula II:


wherein:
pis 1, 2 or 3;
p is 0, 1,2, 3, or 4;
R3 and R4 are independently selected from the group consisting of hydrogen, hydroxy,
alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, arid substituted heterocyclic, provided that only 1 of R3
or R4 is "hydroxy;
or-R3 and R4 together with the nitrogen atom pendent thereto join to form a
heterocyclic or substituted heterocyclic;
R10 is selected from the group consisting of hydrogen, alkyl optionally substituted
with a substituent selected from the group consisting of hydroxy, alkoxy, substituted alkoxy,
amino, substituted amino, acylamino, halo, nitrogen-containing heterocycle, substituted
nitrogen-containing heterocycle, nitrogen-containing heteroaryl, and substituted nitrogen-
containing heteroaryl;
R11 is selected from the group consisting of cyano, alkyl, alkenyl, alkynyl, -CF3,
alkoxy, halo, and hydroxy; and when p is 2, 3, or 4, then each R11 may be the same or
different;
R12 is alkyl,
R13 is hydrogen or alkyl,
R14 is selected from the group consisting of hydrogen, halo, arid alkyl;
or R and R12, together with the carbon and nitrogen atoms bound respectively
there to join toform a heterocyclic of substituted heterocyclic group;

or when R10 and R12, together with the carbon and nitrogen atoms bound respectively
thereto, do not form a heterocyclic "group, then R4 and R10, together with the atoms, bound
thereto, form a heterocyclic or substituted heterocyclic group;
A is selected from the group consisting of aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, and substituted
cycloalkyl;
or pharmaceutically acceptable salts, esters or prodrugs thereof
[0012] In one embodiment, R1 is alkyl. In another embodiment R1 is
selected from the group consisting of isopropyl, t-butyl, and propyl.
[0013] . In another embodiment, R2 is hydrogen.
[0014] In one embodiment R3 and/or R4 is selected from the group consisting
of alkyl, substituted "alkyl and cycloalkyl, and is.selected from the group consisting of methyl,
methoxyethyl, furan-2-ylmethyl, 2-hydroxyethyl, cyclopropyl and isopropyl.
[0015] In one embodiment, R3 and/or R4 is aryl or substituted aryl and is
selected from the group consisting of 4-cyanophenyl, 3,4-difluorophenyl, 2,3,5-
trifluorophenyl, 3,5-dinitrophcnyl, and phenyl.
[0016] In-one embodiment, R3 and/or R4 is heteroaryl or substituted
heteroaryl and is selected from the group consisting of thiophen-2-yl, 3,5-dimethylisoxazol-4-
yl, and 2,6-dichloropyridin-4-yl:
[0017] In one embodiment, R3 and/or R4 is a heterocyclic group or
substituted-heterocyclic group and is tetrahydropyran-4-yl or 4-(ethoxycarbonyl)piperidin-4-
yl-
[0018]- In one embodiment, either of R3 or R4 or both of R3 and R4 are
hydrogen. In another embodiment, one of R3 or R4 is hydroxy.
[0019] In one embodiment, R3 and R4 are cyclized with the nitrogen atom
bound thereto to form a heterocyclic or substituted heterocyclic, and is selected from the
group consisting of 1,1-dioxothiamorpholin-N-yl, 1-oxothiamorpholin-1-yl, 2-
(aminbmethylene)pyrrolidin-N-yl, 2-(memoxycarbony!)pyrrolidin-N-yl, 2,6-
dimethylmbrpholin-N-yl, 3-hydroxypiperidin-N-yl, 3-hydroxypyrrolidin-N-yl, 4-
(butylsulfonyl)pipefazin-N-yl, 4-(cyclopropylsulfonyl)piperazin-N-yl, 4-
(dimethylamino)piperidin-N-yl, 4-(ethoxycarbonyl)pjperazin-N-yl, 4-


(ethylsulfonyl)piperazin-N-yl, 4-(isopropylsulfohyl)piperazin-N-yl, 4-
(methylcarbonyl)piperazih-N-yl, 4-(methylsulfonyl)piperidin-N-yl, 4-
(methysulfonyl)piperazin-N-yl, 4-(morpholin-N-yl)piperidin-N-yl, 4-(piperidih-N-
yl)piperidixi-N-yl,. 4-(propylsulfonyl)piperazin-N-yl, 4-cyclohexylpiperazin-N-yl, 4-
hydroxypiperidin-N-yl, 4-isopropylpiperazin-4-yl, 4-methylpiperidin-N-yl, isoxazolidin-2-yl,
morpholin-N-yl, piperazin-N-yl, piperidin-N-yl, 2-(hydrazinocarbonyl)pyrrolidin-N-yl, and
pyrrolidin-N-yl.
[0020] In some embodiments, R5 is substituted alkyl. In one embodiment,
R5 (or R10) is selected from the group consisting of-(CH2)3NH2, -(CH2)2CH(CH2OH)NH2,
TCH2CH(F)CH2NH2, -CH2-[2-(CH2OH)pyrrolidin-3-yl], -CH2-[4-(OH)pyrrolidin-3-yl],
-GH2:G(F)(spiropyrrolidin-3-yl),-(CH2)2CH(CH2F)NH2,-(CH2)2C(CH3)2NH2,
-(CH2)2CH(CH3)NH2,-(CH2)CH(CH2OCH3)NH2)-(CH2)2CH(CH2F)NHC(O)-[(2-
CH3NHC(O))berizene], and-(CH2)2CH(CH2F)-1,3-dioxo-1,3-dihydroisoindole.
[0021] In one embodiment, wherein R1 and R5 and the atoms bound thereto
join to form a heterocyclic or a substituted heterocyclicgroup and the heterocyclic.group is.2-
oxo-tetrahydropyrimidinyl.
[0022] In one embodiment one of R6 or R7 is aryl or substituted aryl-and is-
selacted from the group consisting of phenyl, 3-chlorophenyl, 3-fluorophenyl, 2,5-
difluorophenyl, and 2,3,5-trifluorophenyr.
[0023] In one embodiment, the. other of R6 or R7 (or RM.) is hydrogen.
[0024] In one embodiment, L is alkylene and A (or A ) is aryl or substituted
aryl.
[0025] In one embodiment, L is methylene and A1 (or A2) is selected from
the group consisting of phenyl, 3-fluorophenyl, or 3-hydroxyphenyl.
[0026] In one.embodiment, R1 is t-butyl, R2 is hydrogen, R3 is hydrogen, L
is methylene, A1 is phenyl, R6 is phenyl or substituted phenyl, R is hydrogen. In another
embodiment K1' is t-butyl, R2 and R3 are hydrogen, L is methylene, A1 is phenyl, R6 is phenyl
substituted with 1 to 2 halo substituents, such as chloro or fluoro. In one embodiment Rl is t-
butyl, R2 and R3 are hydrogen, L is methylene, A1 is phenyl, R5 is substituted alkyl, such as
-(CH2)2CH(CH3)NH2, -(CH2)2C(CH3)2NH2 and -(CH222CH(CH2OH)NH2. In some,
embodiments, R1 is t-butyl, R2 is hydrogen, R3 is hydrogen, L is methylene, A1 is phenyl,
- 8

R° is-phehyl or substituted phenyl, R7 is hydrogen^ and R4 is alkyl. In one embodiment, R1 is
t-biityl and R6 is phenyl substituted with fluoro and may be 3-fluorophenyl or difluofopHenyl'.
In other embodiments, Rl is isopropyl and R6 is phenyl substituted with chloro and may be 3-
chlorophenyl.
Representative Compounds of the Invention
Specific corripounds within the scope of this invention are
1, 2, and 3 in the experimental section.
Methods and Compositions of the Invention
[0028] Also provided is a composition comprising a compound of formulas I
and II (including mixtures and/or salts thereof) and a pharmaceutically acceptable excipient
or carrier.
[0029] In another aspect,, the present invention provides methods of .treating
a mammalian patierifsuffering from a disorder mediated, at least in part, by K.SP. Thus, the
present invention provides methods of treating a mammalian patient in need of such,
treatment comprising administering to the patient a therapeutically effective amount of a
con^q.imd of-formulas Iand_II (including mixtures thereof) either alone_or in combination
with other anticancer agents.
B. Definitions and Overview
[0030] As discussed above, the present invention is directed to new
substitutedjmidazole compounds.
[0031) It is to be understood that the terminology used herein is forthe
purpose of describing particular embodiments only and is not intended to limit the scope of
the present invention. It must be noted that as used herein and in the claims, the singular
forms "a," and "the" include plural referents unless the context clearly dictates otherwise. In
this specification and in the claims which follow, reference will be made to a number of
terms which shall be defined to have the following meanings:
[0032] As used herein, "alkyl" refers to monovalent saturated aliphatic
hydrocarbyl groups having from 1 to 6 carbon atoms and more preferably 1 to 3 carbon

atoms. Thisterm is exemp lified by groups such as methyl,-ethyl, n- prppyl, isorpropyl,n-
butyl, t-butyl, h-pentyl and the like.
[0033] "Substituted alkyl" refers to an alkyl group having from 1 to 3, and preferably 1 to 2, substituents selected from the group consisting of alkoxy, substituted,
alkoxy, acyl, acylamino,.acyloxy, amino, substituted amino, aminoacyl, aryl, substituted aryl,
aryloxy, substituted aryloxy, cyano, halogen, hydroxy, nitro, carboxyl, carboxyl ester,
cycloalkyl, substituted cycloalkyl, spirocycloalkyl, heteroaryl, substituted heteroaryl,
heterocyclic, substituted heterocyclic,.-S02-alkyl, and -S02-Substituted alkyl.
[0034] "Alkylene" refers to divalent saturated aliphatic hydrocarbyl groups
• preferably having from 1 to 5 and more preferably 1 to 3 carbon atoms which are either
;Straight^chained or branched. This term is exemplified by groups such as methylene (^CH2-),
ethylene (-CH2CH2-),propylene(-CH2CH2CH2-), iso-prbpylene (-CH2CH(CH3)-) or
(-CH(eK3)CH2-) and the like.
[0035] "Alkoxy" refers to the group "alkyl-O-" which includes, by way of
example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, t-butoxy, sec-butoxy,
n-peritoxy and the like.
[0036] "Substituted alkoxy" refers to the group "substituted alkyl-O-".
[0037] H'Acyl" refers to the groups H-C(O)-, alkyl-C(O)-, substituted
alkyl-C(O)-, alkenyl-C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted
alkynyl-C(O)- cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, aryl-C(O)-, substituted
aryl-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O)-, heterocyclic-C(O)-, and
substituted heteTocyclic-G(O)-, wherein alkyl, substituted alkyl; alkenyl, substituted alkenyl,
alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
heteroaryl,-substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined
herein.
[0038] "Aminoacyl" refers to the group -C(O)NRR where each R is
independently selected from the group consisting of hydrogen, alkyl, substituted alkyl,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl,.cycloalkyl,
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted
heterocyclic and where each R is joined to form together with the nitrogen atom a
heterocyclic or substituted heterocyclic ring.y/hsreiri alkyl, substituted.alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl>


substituted aryl, heteroaryl, substituted heterparyl, heterocyclic and substituted heterocyclic
are as defined herein.
[0039] "Acyloxy" refers to the groups alkyl-C(O)0-, substituted
alkyl-C(O)0-, alkenyl-C(O)0-, substituted alkenyl-C(O)0-, Jdfcytf^£(G)Q-, substituted
alkynyl-C(O)0-, aryl-C(O)0-, substituted aryl-C(O)0-, cycloalkyl-C(O)6-, substituted
cycloalkyKC(6)0 , heteroaryl-C(O)0-, substituted heteroaryl-G(O)0-, heterocyclic-C(O),Ch
and substituted heterocyclic-C(O)0- wherein alky), substituted alkyl, alkenyl, substituted
alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined
herein.
[0040] "Oxyacyl" or "carboxyl ester" refers to the groups -C(O)0-alkyl, .'
-~C(6)0-substituted alkyl, -C(O)0-alkenyl, -C(O)0-substituted-alkenyl, -C(O)0-alkynyl,
-C(O)Q-substituted alkynyl, -C(O)0-aryl, -C(O)0-substituted aryl, -C(O)0-cycloalkyl,
-C(O)0-substituted cycloalkyl, -C(O)0-heteroaryl, -C(O)0-substituted heteroaryl,
-C(O)0-heterocyclic, and -C(O)0-substiruted heterocyclic wherein alkyl, substituted alkyl,
alkenyl,.substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl,
aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted
-heterocyclic are as defined-herein.
[0041] "Alkenyl" refers to alkenyl groups having from 2 to 6 carbon atoms
and preferably 2 to 4 carbon atoms and having at least 1 and preferably from 1 to 2 sites of
alkenyl unsaturation. Such groups are exemplified by vinyl, allyl, but-3-en-1-yl, and the like.
[0042] "Substituted alkenyl" refers'fo alkenyl groups having from 1 to 3
substituents, and preferably 1 to 2 substituents, selected from the group consisting of alkoxy,
substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aryl,
substituted aryl, aryloxy, substituted aryloxy, cyano, halogen, hydroxy, nitro, carboxyl,
carboxyl ester, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl,
heterocyclic, and substituted heterocyclic with the provjsa that any hydroxy substitution is
not attached to.a vinyl (unsaturated) carbon atom.
[0043] "Alkynyl" refers to alkynyl groups having from 2 to 6 carbon atoms
and preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of
alkynyl unsaturation.


[0044] Substituted alkynyl" refers to alkynyl groupsrhaylngfrom l..{p 3
substituents, and preferably 1 to 2'substituents, selected from the group consisting of alkoxy,
substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aryl,
substituted aryl, aryloxy, substituted aryloxy, cyano, halogen, hydroxy, nitro, carboxyl,
• carboxyl ester, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl,
. heterocyclic, and substituted heterocyclic with the proviso that any hydroxy substitution is
not attached to an acetylenic carbon atom.
[0045] "Amino" refers to the group-NH2.
[0046] "Cyano" refers to the group-CN.
[0047] "Substituted amino" refers to the group -NR'R" where R' and R" are
independently selected from the group consisting"5f hydrogen, alkyl, substituted alkyl,
.alkenyl;"substituted alkenyl,^k*ynylisubstimt^d^aikynyl, aryl, substituted aryl, cycloalkyl; l_s
substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted
heterocyclic, -S02-alkyl, -SC^-substituted alkyl, and where R'and R" are joined, together
with.the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic-group
provided that.R' and R" are both not hydrogen. When R' is hydrogen and R" is alkyl, the
substituted amino group is sometimes referred to herein as alkylamino. When R' and R" are
alkyl, the substitutedamino group =is Sometimes'referred to'herein as dialkyl'arnino. Wheri""'"rr
referring to a monosubstituted amino, it is meant that either R' or R" is hydrogen but not both.
When referring to a disubstituted amino, it is meant that neither R' or R" is hydrogen.
[0048] "Acylamino" refers to the groups -NRC(O)alkyl,
-NRC(O)substituted alkyl, -NRC(O)cycloalkyl, -KRC(O)substituted cycloalkyl,
-NRC(O)alkehyl, -NRC(O)substituted alkenyl,.-NRC(O)alkynyl, -NRC(O)substituted
alkynyl, -NRC(O)aryl, -NRC(O)substiruted aryl, -NRC(O)heteroaryl, -NRC(O)substituted
heteroaryl, -NRC(O)heterocyclic, and -NRC(O)substituted heterocyclic where R is hydrogen
or alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted
alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted
heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
[0049] "Nitro" refers to the group -N02.
[0050] "Aryl" or "Ar" refers to a monovalent aromatic carbocyclic group of
from 6 to 14 carbon atoms having asingle ring (e.g., phenyl) or multiple condensed rings
• (e.g., naphthyl or anthryl) in which the condensed rings may or may not be aromatic (e.g., 2-


bejizpxazolinone^H^,^ and the like) provided that the point of
attachment is at an aromatic carbon atom. Preferred aryls include phenyl and naphthyl.
[0051} "Substituted aryl" refers to aryl groups which are substituted with
from 1 to 3 substituents, and preferably 1 to 2 substituents, selected from the group consisting
of hydroxy, acyl, acylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy,
alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amino, substituted amino,
aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, carboxyl, carboxyl ester,
cyano, thiol, alkylthio, substituted alkylthio, arylthio, substituted arylthio, heteroarylthio,
substituted heteroarylthio, cycloalkylthio, substituted cycloalkylthio, heterocyclicthio,
substituted heterocyclicthio, cycloalkyl, substituted cycloalkyl, halo, nitro, heteroaryl,
substituted hejejoaryl^heterocyclic, substituted heterocyclic, heteroaryloxy, substituted
heteroaryloxy, heterocyclyloxy, subsfitutedjjeterocyclyloxy, amino sulfonyl (NH2-SO2-), and
substituted amino sulfonyl.
[0"052Jt "Aryloxy" refers to the group aryl-O-that includes, by way of
example, phenoxy, naphthoxy, and the like.
[0053] "Substituted aryloxy" refers to substituted.aryl-O- groups.
[0054] "Carboxyl" refers to -COOH or salts thereo_f.__
" [0055] "Cycloalkyl" refers to cyclic alkyl groups of frorrrJ to 10 carbon
atoms having single or multiple cyclic rings including, by.way of example, adamantyl,
cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like.
[0056] "Spirocycloalkyl" refers to cyclic groups from 3 to 10 carbon atoms
having a cycloalkyl ring with a spiro union (the union formed by a single atom which is the
only common-member of the rings).as exemplified by the following structure:

[0057] "Substituted cycloalkyl" refers to a cycloalkyl group, having from 1
t thioxo (=S), alkoxy, substituted alkoxy.-acyl^cylSnirie, acyloxy, amino, substituted amino,
aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, cyano, halogenvhydroxy, nitro,

carboxyl, carboxyl ester, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl,
heterocyclic, substituted heterocyclic, -SO2alkyl and -SCVcycloalkyl
[0058] "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo ajid
preferably is fluoro or chloro.
[0059] "Hydroxy" refers to the group -OH.
[0060] "Heteroaryl" refers to an aromatic group of from 1 to 10 carbon
atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and
sulfur within the ring. Such heteroaryl groups can have a single ring (e.g., pyridinyl or furyl)
or multiple condensed rings (e.g., indolizinyl or benzothienyl) wherein the condensed rings
may or may not be aromatic and/or contain a heteroatom provided that the point of.
attachment is through an atom of the aromatic heteroaryl group. In one embodiment, the
. nitrogen and/or the sulfur ring atom(s) of the heteroaryl group are optionally oxidized to
provide for the N;oxide (N-» O), sulfinyl, or sulfonyl moieties. Preferred heteroaryls include
pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl.
[0061] "Substituted heteroaryl" refers to heteroaryl groupgjhat are
substituted with from 1 to 3 substituents selected from the same group of substituents defined
for substituted aryl.
[0062] "Nitrogen-containing heteroaryl" and "nitrogen-containing
substituted heteroaryl" refers to heteroaryl groups and substituted heteroaryl groups
comprising at least one nitrogen ring atom and optionally comprising other non-nitrogen
hetero ring atoms such as sulfur, oxygen and the like.
[0063] "Heteroaryloxy" refers to the group -O-heteroaryl and "substituted
heteroaryloxy" refers to the group -O-substituted heteroaryl wherein heteroaryl and
substituted hcteroaryl are as defined herein.
[0064] "Heterocycle" or "heterocyclic" or "heterocycloalkyl" or
"heterocyclyl" refers to a saturated or unsaturated (but not aromatic) group having a single
ring or multiple condensed rings, including fused bridged and spiro ring systems, from 1 to
10 carbon atoms and from 1 to 4 hetero atoms selected from the group consisting of nitrogen,
sulfur or oxygen within the ring wherein, in fused ring systems, one or more the rings can be
cycloalkyl, aryl or hcteroaryl provided that the point of attachment is through the heterocyclic
ring. In one cmbodimerit.The nitrogen and/or sulfur atom(s) of the heterocyclic group are
optionally oxidized to provide for the N-oxide, sulfinyl, and sulfonyl moieties.


(0065] "Substituted heterocyclic" or "substituted heterocycloalkyl" or
"substituted heterocyclyl" refers to heterocyclyl groups that are substituted with from 1 to 3
of the same substituents as defined for substituted cycloalkyl.
[0066] Examples of heterocyclyls and heteroaryls include, but are not
limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine,
indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline,
qufnoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine,
carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine,
isoxazole, phenoxazine, phenothiazine, imidazolidine,. imidazoline, piperidine, piperazine,
indoline, phthalimide, l,-2,3i4-tetrahydroisoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene,
thiazole, thiazolidine, thiophene, benzo[b]thiophehe, morpholinyl, thiomorpholinyl (also refeired;to a'stWafr]^ piperidinyl, pyrrolidine,
tetrahydrofuranyl, and the like.
[0067] "Nitrogen-containing heterocyclic" and "nitrogen-containing
substituted heterocyclic" refers to heterocyclic groups and substituted heterocyclic groups
comprising at least,.one nitrogen ring atom and optionally comprising other non-nitrogen
heterp.rijig atoms such as sulfur, oxygen and the like.
[OO'SST ""thiol"Tefers to the group -SH.
[0069] "Alkylthio" or "thioalkoxy" refers to the group -S-alkyl.
[0070] "Substituted alkylthio" or "substituted thioalkoxy" refers to the group
-S-substituted alkyl.
[0071 ] "Arylthio" refers to the group -S-aryl, where aryl is defined above.
[0072] . "Substituted arylthio" refers to the group -S-substituted aryl, where
substituted aryl is defined above.
[0073] "Heteroarylthio" refers to the group -S-heteroaryl, where heteroaryl
is defined above.
[0074] "Substituted heteroarylthio" refers to the group -S-substituted
heteroaryl, where substituted heteroaryl is defined above.
[0075] "Heterocyclicthio" refers to the group -S-heterbcyclic and
"substituted heterocyclicthio" refers to the group -S-substituted heterocyclic, where
heterocyclic and substituted heterocyclic are defined above.


[0076] "Heterocyclyloxy" refers to the group Het'erocyclyl-O- and
"substituted heterocyclyloxy refers to the group substituted heterocyclyl-O- where
heterocyclyl and substituted heterocyclyl are defined above.
[0077] "Cycloalkylthio" refers to the group -S-cycloalkyTand "substituted
cycloalkylthio" refers to the group -S-substiruted cycloalkyl, where cycloalkyl and
substituted cycloalkyl are defined above.
[0078) "Biological activity" as used herein refers to an inhibition
concentration when tested in at least one of the assays outlined in any of Examples 13-15.
[0079] As used herein, the term "pharmaceutically acceptable salts" refers to
the nontoxic acid or alkaline earth metal salts of the compounds of formulas I and II. These
salts can be prepared in situ during the final isolation and purification of the compourids-of
formulas I and.II, or by.separately reacting the base or acid functions with a suitable organic
or inorganic acid or base, respectively. Representative salts include, but are not limited to,
the" following: acetate, adipate, alginate, citrate, aspartate, berizoate,,b.eflzenesulfonate,
bisulfate, butyrate, camphorate, c'amphorsulfonate, digluconate, cyclopentanepropionate,
dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemi-sulfate,
heptanoate, hexanoate, fumaratc, hydrochloride, hydrobromidc, hydroioddde,
Z^hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-napth-.
alenesulfonate,.oxalate, pamoate, pectinate, persulfate, 3-phenylproionate, picrate, pivalate,
propionate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate and undecanoate.
Also, the basic nitrogen-containing groups can be quaternized with such agents as alkyl
halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl
sulfates-lika dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl,
lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and
phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby
obtained.
[0080] Examples of acids that may be employed to form pharmaceutically
acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulfuric acid
and phosphoric acid and such organic acids as oxalic acid, maleic acid, methanesulfonic acid,
succinic acid and citric acid. Basic addition salts can be prepared in situ during the final
isolation and purification of the compounds of formulas I and II, or separately by reacting
carboxylic acid moieties with a suitable base such as the hydroxide, carbonate or bicarbonate


of a pharmaceutically acceptable metal cation or with aminonia, or an organic primary,
secondary or tertiary amine. Pharmaceutically acceptable salts include, but are not limited to,_
cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium,
calcium, magnesium,'aluminum salts and the like, as well as aminonium, quaternary
aminonium, and amine cations, including, but notjimited to_aminonium,
_tetramethylaminonium, tetraethylaminoniurh, methylamine; dimethylamine, trimethylamine,
triethylamine, ethylamine, and the like. Other representative organic amines useful for the
formation of base addition salts include diethylamine, ethylenediamine, ethanolamine,
diethanolamine, piperazine and the like.
[0081] As used herein, the term "pharmaceutically acceptable ester" refers to
esters which hydrolyze in vivo and include those that break down in the human body to leave
. th'e,parent compound, a salt thereof, or a pharmaceutically active metabolite. Suitable ester
groups include, for example, those derived from pharmaceutically acceptable aliphatic
carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in
which each alkyl or alkenyTmoiety advantageously has not more than 6 carbon atoms.
Representative examples of particular esters include, but are not limited to, formates,
acetates, pro'pipn.ates, butyrates, acrylates and ethylsuccinates.
[0082] the term "pharmaceutically acceptable prodrug" as used herein
refers to those prodrugs of the compounds of the present invention which are, within the
scope of sound medical judgment, suitable for use in contact with the tissues of humans and
lower animals without undue toxicity, irritation, allergic response, and the like,
commensurate with a reasonable benefit/risk Tatio, and effective for their intended use, as
well as the zwitterionic forms, where possible, of the compounds of the invention. The term
"prodrug" refers to compounds that are rapidly transformed in vivo to yleld the parent
compound or a pharmaceutically active metabolite of the above formula, for example by
hydrolysis in blood. A discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel
Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed.,
Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon
Press, 1987, both of which are incorporated herein by reference.
[0083 J As used Herein "anticancer agents" or "agent for the treatment of
"cancer" refers to agents that include, by way of example only, agents that induce apoptosis;
polynucleotides (e.g., ribozymes); polypeptides (e.g., enzymes); drugs; biological mimetics;


alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum
compounds; monoclonal antibodies conjugated with anticancer drugs, toxins, and/or
radionuclides; biological response modifiers (e.g. interferons and interleukins, etc.); adoptive
immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell
differentiation (e.g. all-trans-retinoic acid, etc.); gene therapy reagents; antisense therapy
reagents and nucleotides; tumor vaccines; inhibitors of ahgiogenesis, and the like. Numerous
other agents are well within the purview of one of skill in the art. ,
[0084] It is understood that in all substituted groups defined above, polymers
arrived at by defining substituents with further substituents to themselves (e.g., substituted
aryl having a substituted aryl group as a substituent which is itself substituted with a
substituted aiyl group, etc.) are not intended for inclusion herein. In such cases, the
maximum, number ofisueji.sub^iiuerrts,i0fcee'*^a^sjto say that each of the above
definitions, is constrained by a limitation that, for example, substituted aryl groups are limited
tp.-substituted ary1-(substituted aryl)-substituted aryl.
[0085] Similarly, it is understood that the~above definitions are not-intended
to include impermissible substitution patterns (e.g., methyl substituted with 5 fluoro groups
or a.hydroxy group alpha to ethenylic or acetylenic unsaturation). Such impermissible
substitution patterns are well known to the skilled artisan.
[0086] Compounds of this invention may exhibit stereoisomerism by virtue
of the presence of one or more asymmetric or chiral centers in the compounds. The present
invention contemplates the various stereoisomers and mixtures thereof. Depiction of the
compounds of Formulas I and II includes the stereoisomers thereof. Certain of the
compounds of the invention comprise asymmetrically substituted carbon atoms. Such
asymmetrically substituted carbon atoms can result in the compounds of the invention
comprising mixtures of stereoisomers at a particular asymmetrically substituted carbon atom
or a.single stereoisomer. As a result, racemic mixtures, mixtures of diastereomers, single
enantiomer, as well as single diastereomers of the compounds of the invention are included in
the present invention. The terms "S" and "R" configuration, as used herein, are as defined by
the iUPAC 1974 "RECOMMENDATIONS FOR SECTION E, FUNDAMENTAL STEREOCHEMISTRY,"
Pure Appl. Chem. 45:13-30, 1976.,Desired enantiomers can be obtained by.chiral synthesis
from commercially available chiral starting materials by methods well known in the art, or


may be obtained from mixtures of the enantiomers by separating the desired enantiomer by
using known techniques.
[0087] Compounds ofthis invention may also exhibit geometrical
isomerism. Geometrical isomers include the cis and trans forms of compounds of the
invention having alkenyl'or alkeliyrenyl moieties. The present invention comprises.the
individual geometrical isomers and stereoisomers and mixtures thereof.
C. Compound Preparation
(0088] The compounds ofthis invention can be prepared from readily
available starting materials using the following general methods and procedures. Unless
otherwise indicated, the starting materials are commercially available and well known in the
art It will be appreciated that where typical or preferred process conditions (i.e., reaction
temperatures, times, mole ratios of reactants, solvents, pressures) are given, other process
jjpnij.ijjiqns can also be used unless otherwise stated. Optimum reaction conditions may vary
with the particular reactants or solvent used, but such conditions can be determined by one
skilled in the art by routine optimization procedures.
[0089] Additionally, as will be apparent to those skilled in the art,
conventional protecting groups may be necessary to prevent certain-functional.groups from
undergoing undesired reactions. Suitable protecting groups for various functional groups as
well as suitable conditions for protecting and deprotecting particular functional groups are
well known in the art. For example, numerous protecting groups are described in
"T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition,
Wiley,J^ew York, 1991, and references cited therein.
[0090] Furthermore, the compounds of this invention may contain one or
more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as
pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-
enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the
s'cope ofthis invention, unless otherwise indicated. Pure stereoisomers (or enriched
mixtures) may be prepared using, for example, optically active starting materials or
stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such
compounds can be separated using, for example, chiral column chromatography, chiral
resolving agents, and the like.


[0091] Compounds inihe present invention may be better understood by the'
following synthetic Scheme that illustrate methods for the synthesis of compounds of the
invention. Unless otherwise indicated, the reagents used in the following examples are
commercially available and may be purchased from vendors such as Sigma-Aldrich
Company,-tnc: {Milwaukee" WI>; IIS A).
[0092] As discussed above, compounds of the invention have the following
structure:

where R'^.R^.R^R7, andR8 are as defined herein and R5' is-CH2-R5
(providaTthat R5 is not hydrogen) where R5 is as defineqLherein.

PG refers to a suitable nitrogen protecting group suclrSs BOC.
[0093] Specifically, in Step A, an appropriately protected (PG) amino acid
la, is dissolved in a suitable amount of an inert solvent such as methanol or ethanol. It
should be noted that amino.acid.la^is typically commercially available as are disubstituted amino acids (PG-NH-C(R')(R2)-COOH). To that is added a stoichiometric
amount of a monovalent cation, such as cesium carbonate (CS2CO3) or potassium carbonate
(K2CO3), to form the carboxylate salt (not shown). Upon substantial completion of the
reaction, typically about 15 minutes to about 2 hours, excess solvent is removed by
evaporation under reduced pressure. The residual cesium or potassium salt is then re-

dissolved in a suitable solvent, such asDMF and then treated with one-to four equivalents of
the appropriate o-halo-ketone lb (1 eq.), e.g., 2-bromoacetophenone and stirred at RT until
the reaction is substantially complete.
[0094] The product lc is then recovered by conventional methods such as
extraction, filtration, evaporation, and the like. It is generally pure enough to use directly in
the next step.

[0095] To a stirred solution of keto-ester lc from step A in a suitable
amount of inert solvent, such as xylenes, is added an excess of aminonium acetate, typically
from about 2 to about.-20 equivalents and preferably about 5 equivalents. In one embodiment,
a Dean-Stajjk trap is added and the reaction mixture is heated to about 120 °C.to,about 160 °C
uritjjfthe-*eactiori is substantially complete. In another embodiment, the reactants are refluxed
in toluene. Once the reaction is substantially complete, the mixture is allowed to cool to RT.
The product', imidazole Id', i3"then recovered by conventional methods such as extraction,
filtration, evaporation, and the like. It is generally pure enough to use directly in the next
step. •«»s&2*-

[0096] The imidazole Id is then reacted with an appropriate aryl or
hete'roafyl-substituted alkyl halide, such as benzyl bromide. Typically, this can be
accomplished by stirring the imidazole Id with an excess of potassium carbonate and DMF

and then adding at least an equimolar amount>of the aryl or heteroaryl-substftuted alkyl
halide. Once the reaction is substantially complete, the N-aLkyl imidazole le is recovered by
conventional methods such as extraction, filtration, evaporation, recrystallization, and the
like.
[0091], Compounds of.the. invention when R8 is L-A1 and L is -S(O)q- may
be synthesized using a suitable sulfonyl chloride. Descriptions of various sulfonyl chlorides
may be found, for example, U.S. Patent 6,489,300, which is hereby incorporated by
reference.
[0098] In either case, imidazole le is used in the Step D below.

[0099] The. protecting group, PG-is then removed by conventional
tecliniquesjo^rovide amirie^ifiAvhicrTis then optionally purified by conventional methods
such as extraction, filtration, evaporation, and the like. The amine If is used directly in the
next step.

[0100] Amine 1 f then undergoes conventional reductive amination with an
appropriate aldehyde lg to yleld substituted amine lh which is then recovered and optionally
purified by conventional methods such as extraction, filtration, evaporation, chromatography,
and the like.

[0101] In embodiments where R5 is hydrogen, Steps D and E may be
skipped. In these embodiments, imidazole le is used in Step F to make the suitable
carbamate.
I

[0102] The substituted amine lh from step E is then put into a solution with
asuitable solvent such as methylene chloride and an excess of a suitable base, such as
triethylamine. Then a suitabljejjarbamate-forrning agent, such as triphqsgene asishov/rci's -—
- added to form carbamate li. Once the reaction is complete, the carbamate li is recovered by-
conventional methods such as extraction, filtration, evaporation, and the like. The carbamate
li is Used directly in the next step or may be optionally purified by conventional techniques.

[0103] Carbamate li from step-Fis then combined with a slight excess of the
appropriate amine lp to form the urea lj. Once the reaction is complete, the product lj is
recovered by conventional methods such as extraction, filtration, evaporation, and the like.
[0104] It will be well within me skill of me art to further modify the above
preparation to synthesize other compounds of the invention. For example, reaction of


suitableisocyanate with amine lh provides for urea compounds as illustrated, e.g., in •'
Example 3.
D. Pharmaceutical Formulations
[0105] When employed as pharmaceuticals, the compounds of the subject
invention are usually administered in the form of pharmaceutical compositions. These
compositions can be administered by a variety of routes including oral, parenteral,
transdermal, topical, rectal, and intranasal. These compounds are effective, for example, as
both injectable and oral compositions. Such compositions are prepared in a manner well
known in the pharmaceutical art and comprise at least one active compound.
[0106] This invention also includes pharmaceutical compositions which
contain, as the active ingredient, one or more of the compounds of the subject invention
above associated with pharmaceutically acceptable carriers. In making the compositions of
this invention, the active ingredient is usually mixed with an excipient, diluted by an
excipient or enclosed within such a carrier which can be in the form of a capsule, sachet,
paper or other container. The excipient employed is typically an excipient suitable for
administration to human subjects or other mammals. When the excipient serves as a diluent,
it can be a solid, seini-solid, or liquid material, which acts as a vehicle, carrier or medium for
the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders,
lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a
solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the
active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions,
and sterile packaged powders.
[0107] In preparing a formulation, it may be necessary to mill the active
compound to provide the appropriate particle size prior to combining with the other
ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a
particle size of less, than 200 mesh. If the active compound is substantially water soluble, the
particle size is normally adjusted by milling to provide a substantially uniform distribution in
the formulation, e.g., about 40 mesh. •
[0108] Some examples of suitable excipients include lactose, dextrose,
sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth,
gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile


water, syrup, and methyl cellulose. The formulations can additionally include: lubricating
agents such as talc, magnesium stearafe, and mineral oil; wetting agents; emulsifylng -and
suspending agents;, preserving agents suchas methyl- and propylhydroxy-benzoates;
sweetening agents; and flavoring agents. The compositions of the invention can be
formulated so as to provide quick, sustained or delayed release of the active ingredient after
administration to the patient by employlng procedures known in the art.
[0109] , The quantity of active component, that is the compound according to
the subject invention, in the pharmaceutical composition and unit dosage form thereof may be
varied or adjusted widely depending upon the particular application, the potency of the
particular compound and the desired concentration.
(0110) The compositions are preferably formulated in a unit dosage form,
each dosage containing from about 1 to about 500 mg, usually about 5 to about 100 mg,
occasionally about 10 to about 30.mg, of the active ingredient. The term "unit dosage forms
refers to physically discrete units suitable as unitary dosages for human subjects and other
mammals, each unit containing a predetermined quantity of active material calculated to
produce.the desiredjherapeutteeffect, in association with a suitable pharmaceutical
excipient. Preferably, the compound of the subject invention above is employed at no more
than about 20 weight percent of the pharmaceutical composition, more preferably no more
than about 15 weight percent, with the balance being phaimaceutically inert carrier(s).
[0111] The active compound is effective over a wide dosage range and is
generally administered in a pharmaceutically or therapeutically effective amount. It will be
understood, however, that the amount of the compound acrually.administered will be
determined by a physician, in the light of the relevant circumstances, including the condition
to be treated, the severity of the condition being treated, the chosen route of administration,
the actual compound administered, the age, weight, and response of the individual patient, the
severity of the patient's symptoms, and the like.
[0112] In therapeutic use for treating, or combating, cancer in mammals, the
compounds or pharmaceutical compositions thereof will be administered by any appropriate
route, such as orally, topically, transdermally, and/or parenterally at a dosage to obtain and
maintain a concentration, that is, an amount, or blood-level of active component in the
mammal undergoing treatment that will be therapeutically effective. Generally, such
therapeutically effective amount of dosage of active component (i.e., an effective dosage)


will be in the range of about 0.1 to about 100, more preferably about 1.0 to about 50 mg/lcg of
body weight/day.
[0113] For preparing solid compositions such as tablets, the principal active
ingredient is mixed with a pharmaceutical excipient to form a solid preformulation
composition containing a homogeneous mixture of a compound of the present invention.
When referring to these preformulation compositions as homogeneous, it is meant that the
active ingredient is dispersed evenly throughout the composition so that the composition may
be readily subdivided-into equally effective unit dosage forms such as tablets, pills and
capsules. This solid preformulation is then subdivided into unit dosage forms of the type
described above containing from, for example, 0.1 to about-500 mg of the active ingredientof
the present invention.
[0114] The tablets or pills of the present invention may be coated or
otherwise compounded to provide a dosage form affording the advantage of prolonged
action. For example, the tablet or pill can comprise an inner dosage and an outer dosage
component, the latter being in the form of an envelope over the former. The two components
can be separated by an enteric layer which serves to resist disintegration in the stomach and
permit the inner component to pass intact into the duodenum or to be delayed in release. A
variety of materials can be used for such enteric layers or coatings, such materials including a
number of polymeric acids and mixtures of polymeric acids with such materials as shellac,
cetyl alcohol, and cellulose acetate.
[0115] The liquid forms in which the novel compositions of the present
invention maybe incorporated for administration orally or by injection include aqueous
solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with
edible oils such as com oil, cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as
elixirs and similar pharmaceutical vehicles.
.[0116] Compositions for inhalation or insufflation include solutions and
suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof,
and powders. The liquid or solid compositions may contain suitable pharmaceutically
acceptable excipients as described supra. Preferably the compositions are administered by
the oral or nasal respiratory route for local or systemic effect. Compositions in preferably
pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized
solutions may be inhaled directly from the nebulizing device or the nebulizing device may be


attached to a face mask tent, or intermittent positive pressure breathing machine. Solution,
suspension, or powder compositions may be administered, preferably orally or nasally, from
devices which deliver the formulation in an appropriate manner.
[0117] The following formulation examples illustrate representative
pharmaceutical compositions of the present invention.
Formulation Example 1
[0118). Hard gelatin capsules containing the following ingredients are
prepared:
Quantity
Ingredient (mg/capsule)
Active Ingredient 30.0
Starch 305.0
Magnesium stearate 5.0
[0119] The above ingredients are mixed and filled into hard gelatin capsules
in 340.mg quantities.
Formulation Example 2
[0120] A tablet formula is prepared using the ingredients below:
Quantity
Ingredient (mg/tablet)
Active Ingredient 25.0
Cellulose, microcrystalline 200.0
Colloidal silicon dioxide 10.0
Stearic acid 5.0
[0121] The components are blended and compressed to form tablets, each
weighing 240 mg.

Formulation Example 3
[0122]- A dry powder-inhaler formulation is prepared containin]
following components:
Ingredient Weight %
Active ingredient 5
Lactose 95
(0123] . The active ingredient is mixed with the lactose and the mixture is
added to a dry powder inhaling appliance.
Formulation Example 4
[0124] Tablets, each containing 30 mg of active ingredient, are prepared as
follows
Quantity
Ingredient (mg/tablet)
Active Ingredient 30.0 ,mg
Starch 45.0 mg
Microcrystalline cellulose 35.0 mg
Polyvinylpyrrolidone 4.0 mg
(as 10% solution in sterile water)
Sodium carboxymethyl starch 4.5 mg
Magnesium stearate 0.5 mg
Talc 1.0 mg
Total 120 mg
[0125] The active ingredient, starch and cellulose are passed through a No.
20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed
with the resultant powders, which are then passed through a 16 mesh U.S. sieve. The
granules so produced are dried at 50 °C to 60 °.C and passed through a 16 mesh U.S. sieve.
The sodium carboxymethyl starch/magnesium stearate, and talc, previously passed through a

No. 30 mesh U.S..sieve, are then' added to the granules which, after mixing, are compressed
on a tablet machine to yleld tablets each weighing 120 mg.
Formulation Example 5
[0126] Capsules, each containing 40 mg of medicament are made as follows:
Quantity
Ingredient (mg/capsule)
Active Ingredient 40.0 mg
Starch 109.0 mg
Magnesium stearate . l.Omg
Total 150.0 mg
(0127] The active ingredient, starch and magnesium stearate are blended,
passed through a No r20-mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg
quantities.
Formulation Example 6
[0128] Suppositories, each containing 25 mg of active ingredient are made
as follows:
Ingredient Amount
Active Ingredient 25 mg
Saturated fatty acid glycerides to 2,000 mg
[0129] The active ingredient is passed through a No. 60 mesh U.S. sieve and
suspended in the saturated fatty acid glycerides previously melted using the minimum heat
necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and
allowed to cool.
Formulation Example 7
[0130] Suspensions, each containing 50 mg of medicament per 5.0 mL dose
are made as follows:

Ingredient Amount
Active Ingredient 50.0 mg
Xanthangum 4.0 mg '
Sodium carboxymethyl cellulose (11%)
Microcrystalline cellulose (89%)
50.0 mg
Sucrose 1.75 g
Sodium benzoate 10.0 mg
Flavor and Color q.v.
Puri fied water to .5.0 mL
[01-31] The active ingredient, sucrose and xanthan gum are blended, passed
through a No. 1.0 mesh U.S. sieve, and then mixed with a previously made solution of the
microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium
benzoate, flavor, and color are diluted with some of the water andjgdded with stirring,
Sufficient water is then added to produce the required volume.
Formulation Example 8
Quantity
Ingredient (mg/capsule)
Active Ingredient 15.0 mg
Starch 407.0 mg
Magnesium stearate 3.0 mg
Total 425.0 mg
[0132] The active ingredient, starch, and magnesium stearate are blended,
passed through a No. 20-mesh U.S. sieve, and filled into hard gelatin capsules in 425*0 mg
quantities.
Formulation Example 9
[0133] A subcutaneous formulation may be prepared as follows:

Ingredient Quantity
Active Ingredient 5.0 mg
Com Oil. 1.0 mL
Formulation Example 10
[0134] A topical formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 1-10 g
Emulsifylng Wax 30 g
Liquid Paraffin 20 g
White Soft Paraffin to 100 g
[0135] The white soft paraffin is heated until molten. The liquid parjffiru_
arid emulsifylng wax are incorporated and stirred until dissolved. The active ingredient is
added and stirring is continued until dispersed. The mixture is then cooled until solid.
Formulation Example 11
[0136] An intravenous formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 250 mg
Isotonic saline ' 1000 mL
[0137] Another preferred formulation employed in the methods of the
present invention employs transdermal delivery devices ("patches")- Such transdermal
patches may be used to provide continuous or discontinuous infusion of the compounds of the
present invention in controlled amounts. The construction and use of transdermal patches for
the delivery, of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent
5,023,252, issued June 11, 1991, herein incorporated by reference. Such patches may be
constructed for continuous, pulsatile.'or on demand delivery of pharmaceutical agents.
[0138] Frequently, it will be desirable or necessary to introduce the
pharmaceutical composition to the brain, either directly or indirectly. Direct techniques

usually involve placement of a drug delivery catheter into the host's ventricular system to
bypass the blood-brain barrier. One such implantable delivery system used for the transport
of biological factors to specific anatomical regions of the body is described in U.S. Patent
5,011,472 which is herein incorporated by reference.
[0139] Indirect techniques, which are generally preferred, usually involve
formulating the compositions to provide for drug latentiation by the conversion of
•hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved through
blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to
render the drug more lipid soluble and amenable to transportation across the blood-brain
barrier. Alternatively, the delivery of hydrophilic drugs may be enhanced by intra-arterial
infusion of hypertonic solutions which can transiently open the blood-brain barrier.
[0140] Other suitable formulations for use in the present invention can be
found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia,
PA, 17th ed. (1985).
E.- Dosage and Administration
[0141] As noted above, the compounds described herein are suitable for use
in a variety of drug delivery systems described above. Additionally, in order to enhance the
in vivo serum half-life of the administered compound, the compounds may be encapsulated,
introduced into the lumen of liposomes, prepared as a colloid, or other conventional
techniques may be employed which provide an extended serum half-life of the compounds.
A variety of methods are available for preparing liposomes, as described in, e.g., Szoka, et
al.i U.S. Patent Nos. 4,235,871, 4,501,728 and 4,837,028 each of which is incorporated
herein by reference.
[0142] Comppunds of the instant invention are useful for inhibiting or
treating a disorder mediated, at least in part, by the activity of KSP. In one aspect, the
disorder that is mediated, at least in part by KSP, is a cellular proliferative disorder. The term
"cellular proliferative disorder" or "cell proliferative disorder" refers to diseases including,
for example, cancer, tumor, hyperplasia, restenosis, cardiac hypertrophy, immune disorder
and inflammation. The present invention provides methods of treating a human or
mammalian subject in need of such treatment, comprising administering to the subject a


therapeutically effective amount of a compound of formula lor II, dither alone or in
combination with other anticancer agents.
[0143] The compounds of the invention are useful in vitro or in vivo in
inhibiting the growth of cancer cells. The term "cancer" refers to cancer diseases including,
for example, lung and bronchus; prostate; breast; pancreas; colon and rectum; thyroid;
stomach; liver and intrahepatic bile duct; kidney and renal pelvis; urinary bladder; uterine
corpus; uterine cervix; ovary; multiple myeloma; esophagus; acute myelogenous leukemia;
chronic myelognous leukemia; lymphocytic leukemia; myeloid leukemia; brain; oral cavity
and pharynx; larynx; small intestine; non-hodgkin lymphoma; melanoma; and villous colon
adenoma.
[0144] Cancer also includes tumors or neoplasms selected from the group
consisting of carcinomas, adenocarcinomas, sarcomas, and hematological malignancies.
[0145] Additionally, the type of cancer can be selected from the group
consisting of growth of solid tumors/malignancies, myxoid and round cell carcinoma, locally_
advanced tumors, human soft tissue carcinoma, cancer metastases, squamous cell carcinoma,
esophageal squamous cell carcinoma, oral carcinoma, cutaneous T cell lymphoma, Hodgkin's
lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing
'tumor's, nonsmall cell cancers, breast cancer, gastrointestinal cancers, urological cancers,
malignancies of the female genital tract, malignancies of the male genital tract, kidney
cancer, brain cancer, bone cancers, skin cancers, thyroid cancer, retinoblastoma,
neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's
tumors, gall bladder cancer, trophoblastic neoplasms, hemangiopericytoma', and Kaposi's
sarcoma.
[0146] A compound or composition of this invention may be administered to
a mammal by a suitable route, such as orally, intravenously, parenterally, transdermally,
topically, rcctally, or intranasally.
[0147] Mammals include, for example, humans and other primates, pet or
companion animals, such as dogs and cats, laboratory animals, such as rats, mice and rabbits,
and farm animals, such as horses, pigs, sheep, and cattle.
[0148] Tumors or neoplasms include growths of tissue cells in which the
multiplication of the cells is uncontrolled and progressive. Some such growths are benign,
but others are termed "malignant" and can lead to death of the organism. Malignant


neoplasms or "cancers" are distihpished from benign growths in that, in addition to
exhibiting aggressive cellular proliferation, they can invade surrounding tissues and
metastasize. Moreover, malignant neoplasms are characterized in that they show a greater
loss of differentiation (greater "dedifferentiation") and organization relative to one another
and to surrounding tissues. This property is called "anaplasia."
[0149] Compounds having the desired biological activity may be modified
as necessary to provide desired properties such as improved pharmacological properties (e.g.,
in vivo stability, bio-availability), or the ability to be detected in diagnostic applications.
Stability can be assayed in a variety of ways such as by measuring the half-life of the
compounds during incubation with peptidases or human plasma or serum.
[0150] For diagnostic purposes, a wide variety of labels may be linked to the
compounds, which-may provide, directry or indirectly, a detectable signal. Thus, the
compounds and/or compositions of the subject invention may be modified in a variety of
ways for a variety of end purposes while still retaining biological activity. In addition,
various reactive sites may be introduced for linking to.particles, solid substrates,
macrtimo]ecures, "and the like.
[0151] . Labeled compounds can be used in a variety of in vivo or in vitro
applications. "A wide variety of labels may be employed, such as radionuclides (e.g., gamma-
emitting radioisotopes such as technetium-99 or indium-111), fluorescers (e.g., fluorescein),
enzymes, enzyme-substrates, enzyme cofactors, enzyme inhibitors, chemiluminescent
compounds, bioluminescent compounds, and the like. Those of ordinary skill in the art will
know of other suitable labels for binding to the complexes, or will be able to ascertain such
using routine" experimentation. The binding of these labels is achieved using standard
techniques common to those of ordinary skill in the art.
[0152] Pharmaceutical compositions of the invention are suitable for use in a
variety of drug delivery systems. Suitable formulations for use in the present invention are
found in Remington's.Pharmaceutical Sciences, Mace Publishing Company, Philadelphia,
Pa.„17thed..(1985).
[0153] The amount administered to the patient will vary depending upon
what is being administered, the purpose of the administration, such as prophylaxis or therapy,
the; state of the patient, the manner of administration, and the like. In therapeutic
applications, compositions are administered to a patient already suffering from a disease in an


amount suTficieht to cure or at least partially arrest the progression or symptoms of the
disease and its complications. An amount adequate to accomplish this is defined as
"therapeutically effective dose." Amounts effective for this use will depend on the disease
condition being treated as well as by the judgment of the attending clinician depending upon
factors such as the severity of the disease, disorder or condition, the age, weight and general
condition of the patient, and the like. .
[0154J The compounds administered to a patient are typically in the form of
pharmaceutical compositions described above. These compositions may be sterilized by
. conventional sterilization techniques, or may be sterile filtered. The resulting aqueous
solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being
combined with a sterile aqueous carrier prior to administration. The pH of the compound
preparations typically will be between about 3 and 11, more preferably from about 5 to 9 and
most preferably from about 7 to 8. It will be understood that use of certain of the foregoing
excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
J0155] The therapeutic dosage of the compounds and/or compositions of the
present invention will vary according to, for example, the particular use for which the
treatment is made, the manner of administration of the compound, the" health and condition of
the patient, and tRe judgment of the prescribing physician. For example, for oral
administration, the dose will typically be in the range of about 5 ug to about 50 mg per
kilogram body weight per day,.preferably about 1 mg to about 10 mg per kilogram body
weight per day. In the alternative, for intravenous administration, the dose will typically be
in the range of about 5 ug to about 50 mg per kilogram body weight, preferably about 500 ug
to about 5000 ug per kilogram body weight. Alternative routes of administration
contemplated include, but are not limited to, intranasal, transdermal, inhaled, subcutaneous
and intramuscular. Effective doses can be extrapolated from dose-response curves derived
from in vitro or animal model test systems.
[0156] In general, the compounds and/or compositions of the subject
invention will be administered in a therapeutically effective amount by any of the accepted
modes of administration for agents that serve similar utilities. Toxicity and therapeutic
efficacy of such compounds can be determined by standard pharmaceutical procedures in cell
cultures or experimental animals, e.g., for determining the LD^ (the dose lethal to 50% of the
population) and the ED5o (the dose therapeutically effective in 50% of the population). The


dose ratio between toxic and therapeutic effects is the therapeutic index' and it can be
expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are
preferred.
[0157] The data obtained from the cell culture assays and animal studies can
be used in formulating a range of dosage for use in humans. The dosage of such compounds
lies preferably within a range of circulating concentrations that include the ED50 with littleor
no toxicity. The dosage may vary within this range depending upon the dosage form
employed.and the route of administration utilized. For any compound and/or composition
used in the method of the invention, the therapeutically effective dose can be estimated
initially from cell culture assays. A dose may be formulated in animal models to achieve a
circulating plasma concentration range which includes the IC50 (the concentration of the test
compound which achieves a half-maximal inhibition of activity) as determined in cell culture.
Such information can be used to more accurately determine useful doses in humans. Levels
in plasma may be measured, for example, by high performance liquid chromatography.
[0158] The following synthetic and biological examples are offered to
illustrate this invention and are not to be construed in any way as limiting the scope of this
invention.
EXAMPLES
[0159] Referring to the examples that follow, compounds of the present
invention were synthesized.using the methods described herein, or other methods, which are
well known in the art.
[0160] The compounds and/or intermediates were characterized by high
performance liquid chromatography (HPLC) using a Waters Millenium chromatography
system with a 2690 Separation Module (Milford, MA). The analytical columns were AUtima
C-18 reversed phase, 4.6 x 250 mm from Alltech (Deerfield, IL). A gradient elution was
used, typically starting with 5% acetonitrile/95% water and progressing to 100% acetonitrile
over a period of 40 minutes. All solvents contained 0.1% trifluoroacetic acid (TFA).
Compounds were detected by ultraviolet light (UV) absorption at either 220 or 254 nm.
HPLC-solvents were from Burdick and Jackson (Muskegan, MI), or Fisher Scientific
(Pittsburgh, PA). In some instances, purity was assessed by thin layer chromatography
(TLC) using glass or plastic backed silica gel plates, such as, for example, Baker-Flex Silica


Gel 1B2-F flexible sheets. TLC results were readily detected visually under ultraviolet light,
or by employlng well known iodine vapor, and' other various staining techniques.
[0161] Mass spectrometric analysis was performed on one of two LC/MS
/instruments: a Waters System (Alliance HT HPLC and a Micromass ZQ mass spectrometer;
Column: Eclipse XDB-C18, 2.1 x 50 mm; solvent system: 5-95% (or 35-95%, or 65-95% or
95-95%) acetonitrile in water with 0.05% TFA; flow rate 0.8 mL/min; molecular weight
range 500-1500; cone Voltage 20 V; column temperature 40 °C) or a Hewlett Packard
System (Series 1100 HPLC; Column: Eclipse XDB-C18, 2.1 x 50 mm; solvent system:
1-95% acetonitrile in water with 0.05% TFA; flow rate 0.4 mL/min; molecular weight range
150-850; cone Voltage 50 V; column temperature 30 °C). All masses were reported as those
of the protonated parent ions.
[0162] GC/MS analysis is performed on a Hewlett Packard instrument
(FIP6890 Series gas chromatograph with a Mass Selective Detector 5973; injector volume: 1
mL; initial column temperature: 50 °C; final column temperature: 250 °C/ramp time: 20
minutes; gas flow rate: 1 mL/min; column: 5% phenyl methyl siloxane, Model
No. HP 190915-443, dimensions: 30.0 m x 25 m x 0.25 m).
[0163] Nuclear magnetic resonance (NMR) analysis was performed on some
of the compounds with a Varian 300 MHz NMR (Palo Alto, CA). The.spectral referenQCjvjis
either TMS or the known chemical shift of the solvent. Some compound samples were run at
elevated temperatures (e.g., 75 °C) to promote increased sample solubility.
[0164] The purity of some of the invention compounds is assessed by
elemental analysis (Desert Analytics, Tucson, AZ).
[0165] Melting points are determined on a Laboratory Devices Mel-Temp
apparatus (Holliston, MA).
[0166] Preparative separations were carried out using a Flash 40
chromatography system and KP-Sil, 60A (Biotage, Charlottesville, VA), or by flash column
chromatography using silica gel (230-400 mesh) packing material, or by HPLC using a C-18
reversed phase column. Typical solvents employed for the Flash 40 Biotage system and flash
column chromatography were dichloromethane, methanol, EtOAc, hexane, acetone, aqueous
hydroxyamine and triethyl amine. Typical solvents employed for the reverse phase HPLC
were varylng concentrations of acetonitrile and water with 0.1% trifluoroacetic acid.


. [0167] Unless otherwise stated all.temperatures are in degrees Celsius.
Also, in these examples and elsewhere, abbreviations have the following meanings:



[0168] A stirred 0.4 M solution of the appropriate N-Boc-acid (1 eq.), e.g.,
tert -butyl leucine 1-1 in EtOH/was treated with Cs2C03 (0.5 eq.). After 45 min, the EtOH
was removed by evaporation under reduced pressure. The residual cesium salt was re-
dissolved in DMF (1.5X volume of DMF used in the reaction) and then treated with the
appropriate a-halo-ketone, e.g., 2-bromoacetophenone (1 eq.) and stirred at RT until the
reaction was complete. The reaction mixture was then partitioned betweer^Ej.QAc and H2Q,
and the organics separated, then washed with H2O (x3), brine (x3), then dried.(Na2S04),

.filtered* and evaporated under reduceSd'pressure to give the keto ester 1-2 which was pure
enough to use,directly in the next step.

[0169] To a stirred 0.1 M solution of keto-ester 1-2 (1 eq.) in xylenes was
added aminonium acetate (5 eq.). A Dean-Stark trap was added and the reaction heated to
140 °C. Once the reaction was complete, the mixture was allowed to cool to RT, then
partitioned between EtOAc and sat. aq. NaHCCh. The organics were separated, then washed
withjat. aq. NaHC03 (x2), H20 (x3), brine (x3), then dried (Na2S04), filtered, and
evaporated under reduced pressure to give the phenyl imidazole 1-3 which was pure enough
to use directly in the next step.

[0170] To a stirred 0.4 M solution/ suspension of imidazole 1-3 (1 eq.)in
DMF was added K2CO3 (2 eq.) and the benzylating agent, e.g., benzyl bromide (1.1 eq.).
Once the reaction was complete, the mixture was partitioned between EtOAc and H2O. The
organic layer was separated and washed with H2O (x3), brine (x3), then dried (NaiSOj),
filtered, and evaporated under reduced pressure to give the crude benzyjated phenyl
imidazole. The crude reaction material was then crystallized (EtOAc, hexanes) to give pure
product 1-4.


[0171] Boc-protected amine 1-4 was treated with 10% TFA in DCM. Once
reaction was complete, the reaction mixture was concentrated in vacuo and then partitioned
between EtOAc and sat. aq. NaHC03. The organics were separated, then washed with sat. aq.
NaHC03 (x2), H20 (x2), brine (x2), then dried (Na2S04), filtered, and evaporated under
reduced pressure.to give the phenyl imidazole free amine 1-5 which was pure enough to use
directly in the next step.

[0172] A mixture of 0.83 M solution of N-Boc-3-formyl pyrrolidine 1-6 (1
eq.) in DMF, TMSC1 (2.5 eq.) and Et3N (5 eq.) was heated for 6 h. The mixture was then
diluted with hexanes and filtered (Celite). The filtrate was then evaporated under reduced
pressure to give the TMS enol ethers 1-7 as a mixture of £ and Z isomers that were used
directly in the next step.


,[6173] To a 0.1 M solution of TMS enol ether 1-7 (1 eq.) in CH3CN was
added SelectFlu6r®(l.l eq., available from Sigma-Aldrich). Once the reaction was complete,
the mixture was evaporated under, reduced pressure and the remaining solid/oil was extracted
with Et20 (x5). The ether extracts were evaporated under reduced pressure to give the crude
aldehyde. Purification by silica gel chromatography afforded the desired aldehyde 1-8.

[0174] To a stirred 0.1 M solution of (R)-amine 1-5 (1 eq.) from step D in
DCM, was added aldehyde 1-8 (1.1 eq.) followed by AcOH (1 eq.) followed by sodium tris-
acetoxyborohydride (1.5 eq.). After 19 h, further sodium tris-acetoxyborohydride (0.5 eq.)
was added. Once the reaction was complete, the mixture was concentrated in vacuo,
partitioned between EtOAc and 1M NaOH. The.organics were separated, then washed with
1M NaOH (x2), H20 (xl), brine (x2), then'dried (Na2S04), filtered, and evaporated'under
reduced pressure to give crude product. Purification by silica gel chromatography afforded
the amine 1-9 as a mixture of (R,R) and (R,S) diastereomers.


[0175] To a 0.08 M solution of (R) amine 1-9 from step F (1 eq'.) in DCM
was added Et3N (4 eq.) followed by triphosgene (1.2 eq.). Once the reaction was complete,
the reaction mixture was concentrated in vacuo, partitioned between EtOAc and sat. aq.
NaHC03. The organics were separated, then washed with sat. aq. NaHCCh (x2), H20 (xl),
brine (x2), then dried (Na^SO-O, filtered, and evaporated under reduced pressure to give crude
trichlorocafbamate 1-10 which was used directly in the next step.

[0176] To a stirred 0.16 M solution of trichlorocarbamate 1-10 from step G
(.1 eq.)'in DCM was added DIPEA (5 eq.) followed by 4-piperidinopiperidine (3 eq.). Once
the reaction was complete, the mixture was concentrated in vacuo, partitioned between
EtOAc and sat. aq. NaHC03. The organics were separated, then washed with sat. aq.

•NaHCGi ;(x2), H20 (x 1), brine (x2), then dried (Na2S04),filtered, and evaporated under
reduced pressure to give crude product as a mixture of 1-11 and l-12.This was purified by
reverse phase prep. HPLC which separated the (RJR.) and the (R,S) diastereomers 1-11 and
12

[0177] Boc-protected a'mine 1-11 was treated with 10% TFA/ DCM. Once
reactigfi^was complete, the reaction mixture was evaporated under reduced pressure to give
the title compound that was purified by reverse phase prep. HPLC to give the pure 76.
Compound 77 (not shown here) was synthesized using the other isomer 1-12 from step H.

Example 2
Prepat ationof N-[(2R)-3-amino-2-fiuoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazoI-2-yl)^2,2-dimethylpropyl]piperazine-1-carboxamide (26)

[0178] The phenyl imidazole 1-5 from step D of Example 1 (1 eq., 2.0 g)
was combined with the aldehyde (1.3 eq., 1.56 g) and sodium triacetoxyborohydride (2 eq.,
.'2;65 g) in 30 mL of methylene chloride. This was followed by acetic acid (2 eq., 0.72 mL)
and the reaction was stirred at R.T under nitrogen overnight. The reaction was worked up with
water, saturated sodium bicarbonate then saturated sodium chloride. The organic layer was
dried over magnesium sulfate, filtered and concentrated. The material was purified on a
column "and gave the resulting product 2-1 as 2.15 g of a white solid.

[0179] The phenyl imidazole amine.2-1 (1 eq., 100 mg) was dissolved in 4
mL.of tetrahydrofuran and cooled down to 0 °C. Triphosgene (1.7 eq., 102 mg) was added to
the; s'olution.followed by triethylamine (6 eq., 169 mL).-The reaction was stirred for 2 h and
allowed to warm up to RT. The solvent was evaporated and the material dissolved in EtOAc

arid worked up. as follows: water, saturated sodium'bicarbpnate and saturated s'odiurri.
chloride. The organic layer was dried over magnesium sulfate, filtered and dried resulting in
133 mg;of the trichlorocarbarnate 2-2.

[0180] The trichlorocarbarnate 2^2 (1 eq., 133 mg) from step B above was
reacted with piperazine (10 eq., 175 mg) by stirring in 4 mL of tetrahydrofuran at RT for 2 h.
The spiverit was evaporated and the material redissolved.in.EtOAc and washed with saturated
sodium bicarbonate and saturated sodium chloride. The organic layer was dried over
magnesium sulfate, filtered and concentrated resulting in 115 mg of the urea as a mixtureof
two isomers. The isomers were separated by purification, only isomer 2-3 is shown here.


[0181] The Boc protected urea 2-3 from step C above (1 eq., 18 mg) was
treated with 1 mL of 20% trifjuoroacetic acid in methylene chloride at RT for 2 h. The
solvent was evaporated to give the final product 26. Compound 25 was synthesized using the
other isomer from step C.
Example 3
Preparation of N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N'-(4-cyandpheny))urea (64)

[0182] The phenyl imidazole amine 2-1 (1 eq., 15 mg) from Step A of
Example 2 was reacted with 4-cyanophenyl isocyariate (10 eq., 44 mg) in 1 m'Lof
tetrahydrofuran at 60 °C overnight. The solvent was evaporated and the material dissolved in
EjOAc and washed with water, saturated sodium bicarbonate and saturated sodium chloride.

The organic layer was dried over magnesium sulfate, filtered and dried. The Boc was
deprotected following step D in Example 2;
Example A.....
Preparation of l-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]tetrahydropyrimidin-2(lH)-one (52)

[0183] To a stirred solution of amine 1-5 from Step D of Example 1 (1.0 eq.)
in DCM was added appropriate aldehyde (1.0 eq.). The mixture was allowed to stir for 5 min
before the addition of sodium tris-acetoxyborohydride (1.0 eq.). Once the reaction was
complete, the mixture was concentrated in vacuo, partitioned between EtOAc and 2M aq.
Na2C03. The organic^ were separated, then washed with 2M aq. Na2C03 (x2), H20 (x2),
brine (x2), then dried (Na2S04), filtered, and evaporated under reduced pressure to give
product 4-1 which was used directly in the next step.

[0184J To a 50 mL flask was added 1.97 mL of 20% phosgene (10 eq.). it
.toluene. To that was added a solution of product 4-1 from Step A (200 mg, 1 eq.) in dry

;E).CM;(3 mL) ahd'triethylamirie (0.517 mL, 10 eq.). The reaction rriixture was stirred at RT
for about 10 min. After the completion of the reaction, the mixture wasdiluted in DCM,
washed with .water, brine, dried (Na2S04), filtered, and evaporated under reduced pressure.
Purification on silica gel to afford 180 mg of carbamoyl chloride 4-2 as white solid.

[0185] To a stirred solution of carbamoyl chloride 4-2 from step B (1 eq.) in
-DMF was added 3-amino-1,2,4-triazole (2 eq.), Et3N (2 eq.) and DMAP (1 eq.). The reaction
was stirred at RT and progress was monitored by LC/MS. After completion, the solvent was
evaporated under reduced pressure. The crude product was dissolved in ethanol and
hydrazine was added. The reaction was stirred at RT. After completion, the solvent was
evaporated under reduced pressure and portioned between EtOAc and water. The organics
were separated, then washed with 2 M aq. Na2CC>3 (x2), H20 (x2), brine (x2), then dried
(Na2S04), filtered, and evaporated under reduced pressure. Purification by reverse phase
prep. HPLC afforded 52. MS: m/z 403.2

Examples
Preparation of ^
{[(;3R,4R)-4-hydrbxypyrrolidin-3-yl]met^
N-[(1R)^1-(1-benzyl-4-pheny l-lIWmidazol-2-yl)-2,2-dirnethylpropyl]-ft-{ [(3S,4S)-4-
hydrbxypyrrolidin-3-yl]me^

[0186] To a 100 mL round bottom flask was added 5.0 g (29.5 mmol) 2,5-
dihydropyrrolg-rl-carboxylic acid tert-butyl ester, 11.7 g (67.9 mmol) 3-chlorppejpxybenzoic
acid, and 70 mL of DCM. The mixture was stirred at ambient temperature under nitrogen for
20 h. Excess IN NaOH was then added and the reaction mixture was extracted with DCM
(x3). The product was determined by thin layer chromatography (4:1 hexanes: EtOAc) using
nirihydriri stain. The organic layers were combined, dried over MgSCM, and the solvent was
removed-m vacwo ylelding 5.1974 g (28.1 mmol, 95%) of product 5-1 as a yellow oil.

[0187] To a dry 200 mL round bottom flask was added 4.31 g (23.3 mmol)
product 5-1 fronrstep A, 0.21 g (2.33 mmol) copper cyanide, and 50 mL anhydrous THF and
the resulting solution was cooled to -78 °C. To the reaction mixture was then added drdpwise
73.3 mL (73.3 mmol) vinylmagnesium bromide and the resulting solution was allowed to
Warm slowly to ambient temperature under nitrogen over the course of 7 h. The reaction
mixture was quenched with saturated NH4CI and extracted with EtOAc (x3). The product
was determined by thin layer chromatography (2:1 hexanes: EtOAc) using ninhydrin stain.

The organic layers were combined,.dried oyer MgS64,.and thesolvent was removed in vacuo ylelding 4.75 g product 5-2 as a tan oil.

[0188] To a solution of.2.0 g (9.4 mmol) of product 5-2 from step B in 30
mL of TKF and 15 mL of H20, was added 3.5 g (16.4 mmol). of NaI04 and 0.23 mL (0.94
'fflrhol) of Os04. Formation of a white precipitate was observed after approximately 30
minutes. The reaction was monitored by thin layer chromatography^.: 1 hexanes: EtOAc)
uMhg^iihhydrinstain. Stirring continued for an additional 7 h at ambient temperature under
nitrogen and the reaction was then quenched with H2O and extracted with EtOAc (x3). The
organic layers were combined, washed with saturated NaHCCh and brine, dried over MgSCU,
and the solvent was removed in vacuo ylelding 1.35 g (6.3 mmol, 67%) product 5-3 as a tan
foam.

[0189] To a dry 100 mL round bottom flask was added 1.6 g (5.0 mmol) of
phenylimidazole free amine 1-5 from step D of Example 1, 1.35 g (6.26 mmol) of product 5-
3 from step C above, 1.38 g (6!5 rrimol) sodium tris-acetoxyborohydride, 25 mL anhydrous

iJ'Clylf; .arid 0.37 mL (6/5 mmbl)a6etic acid. The resulting solution was stirred at arnbient .
temperature under nitrogen for 2 h. Excess DCM was then added to the reaction mixture.
The organic layer was washed with H20, saturated NaHCCh (x2), and brine (x2). The
combined organic layers were dried over MgSO'4 and the solvent was removed in vacuo. The
resulting crude material was subjected to flash column chromatography and the product was
eluted with a gradient of hexanes, 20% EtOAc in hexanes, 50% EtOAc in hexanes, and 10%
methanol.and 0.3% aminonia in DCM ylelding 1.56 g (3.0 rhmol, 60%) product 5-4 as a tan
foam. Product 5-4 was provided as a mixture of diastereomers.
[0190] MH+ = 519.3

[0191] To a 25 mL round bottom flask was added 0.2 g (0.39 mmol) product
5-4-from step D, 0.04 g (0.56 mmol) imidazole, 0.08 g (0.56 mmol) tert-
butylchlorodimethylsilane, 0.005 g (0.04 mmol) DMAP, and 3 mL DMF. The resulting
solution was stirred at ambient temperature under nitrogen for 24 h. The reaction was
quenched with H2O and extracted with EtOAc (x3). The combined organic layers were
washed with saturated NaHCCh and brine, dried over MgSC>4, and the solvent was removed
in vacuo ylelding 0.28 g (0.44 mmol, 113%) product 5-5 as a crude tan oil. MH+ = 633.3


[0192] To a solution of 0.05 g (0.08 mmol) product 5-5 from step E in 1 mL
of anhydrous THF at 0 °C, was added 0.04 g (0.14 mmol) triphosgene and 0.07 mL (0.48
:mmol) triethylamine. The mixture was stirred at 0 °C for 1 h. The reaction was monitored by
TLC (4:1 hexanes: EtOAc). After the completion of the reaction, H2O was added and it was
extracted with EtOAc (x2). The combined organic layers were washed with saturated
NaHCCb and brine, dried over MgS04, and the solvent was removed in vacuo ylelding 0.08 g
•(0.10 mmol,. 125%) product 5-6 as a crude tan oil.

[0193] To a dry reaction vial was added 0.08 g (0.1 mmol) product 5-6 forrr
step F, 0.17 g (1.0 mmol) 4-piperidinopiperidine, and 1.5 mL anhydrous THF. The resulting
solution was stirred at ambient temperature for 20 h. The reaction was then quenched with
:H2.p and: extracted with EtOAc (x3). The combined; organic4ayers were washed with H20,

saturated NaHCCh and brine, dried over MgS04, and the solvent was removed in vacuo
ylelding 0.06 g (0.07 mmol, 73%) product 5-7 as a crude tan oil. MH+ = 827.5

[0194] To a reaction vial was added 0.028 g (0.03 mrriol) product 5-7 from
step G and 0.5 mL THF and the resulting solution was cooled to 0 °C. To the reaction
mixture was then added 0.05 g (0.17 mmol) tetrabutylamrnonium fluoride and the reaction
was allowed to warm to ambient temperature over 1 h. The reaction was then quenched with
saturated-NH4Cl, extracted with EtOAc (x3), and the combined organic layers were dried
over MgS04, and the solvent was removed in vacuo ylelding 0;05 g (0.07 mmol, 213%)
product 5-8 as a crude tan/yellow foam. MH+,= 713.4


[0195] To a reaction vial was added 0.05 g (0.07 mrnol) product 5-8 from
step H, 1.0 mL DCM,and 0.1 mLTFA and the resulting solution was shaken at ambient
temperature, for 2h. The solvent was then removed in vacuo and the crude reaction material
,was purified by reverse phase HPLC ylelding 3.8 mg (0.006 mrnol, 9%) 83 as a white TFA
•salt and 4.6 mg (0.008 mrnol, 11%) 84 (other diastereomer not shown here) as a white TFA
salt. MH+«of 83^.614^4-and MH+-of-84-^.6.13,3__.„^^_

[0196] To a stirred solution of anhydrous K2C03 (46.53 g, 0.3371 rriolj in
DMF (500 mL), D-serinefnethyl; ester hyd'r6'cnloride-(35.0 g,0.2250 rhol), Kl (18.66 g,

.0.1124 mol) and benzyl-bromide (96.18 g, 0:5 623'mol) were added in one shot. The reaction
mixture was stirred vigorously for 5 h at RT. After completion of the reaction, the contents
were poured into ice water and extracted with EtOAc. The combined organic layer was
washed with water, brine, dried over Na2S04 and concentrated to give a crude product 6-2.
Purification was carried out by column chromatography to yleld pure (61.7 g, 91.7%) as pale
yellow oil.

[0197] To a stirred solution of diethylamine sulphur trifluoride (32.3 mL,
0.2006 mol) in THF (400 mL), was added compound 6-2 during the span of 3 h at RT. After
compietion.of addition, stirringwas continued for further 1 h. The mixture was extracted
with ethylacetate and combined organic phase was washed with saturated solution of
NaHC03rR~ernoval of solvent under vacuum lead to a crude product, which-was purified by
column chromatography using hexane grading to 3% EtOAc in hexane afforded product 6-3
(70.4g, 69.9%) as pale yellow oil.

[0198] To a mechanically stirred solution of LiBJLt (230.8 mL, 0.4651 mol)
in THF (2.0 L), methyl ester (100.0 g, 0.3322 mol) in THF (1.0 L) was added dropwise 6-3
■through addition funnel during the span of 3h at -15 °C under N2. After the completion of
addition, stirring was continued for 4 h at RT. Saturated sojution of NH4G1 (500 mL) was
added dropwise to the above mixture and extracted with EtOAc. The combined organic
phase was washed with water, brine, dried over T^SCU and concentrated under vacuum.

Residual oil was dissolved in IN HC1 (200 mL), extracted with diethylether and pH of the aq.
layer was adjusted to. .10 with the help of NH4OH (50%,-300 mL). The resultant was extracted
with 'EtOAc and combined extracts were concentrated under vacuum to give product 6-4
(S6.2g,:95.0%) as pale brown oil.

[0199] A mixture of alcohol 6-4 (50. g, 0:18315 mol) and Pd(OH)2 on carbon.
(20%, 6.26 g, 0.04395 mol) in absolute ethanol (500 mL) was stirred for 7 h under the
pressure ofhydrogen at 50-60 psi. After the reaction, charcoal was removed by filtration and
residue was concentrated on rota evaporator to provide for product 6-5 (15.8 g, 92.7%) as
pale brown oil.

^[0200] to a stirred mixture of amino alcohol 6-5 (15.0 g, 0.16129 mol) and
K2CO3 (33.39 g, 0.24195. mol) in aq. dioxane (about 25%, 375 mL dioxane in 125 mL water),
(Bbc^O (38.66 g, 0.17733 mol) was added drop wise at 0 °C. The reaction mixture was
stirred ovemight-at RT after the addition. Saturated solution of KHSO4 was added to the
"above mixture to adjust the pH 3-4 and extracted with EtOAc. The organic phase was
concentrated under.vacuum to give pure product 6-6 (27.7 g, 89.0%) as a pale brown oil.


'■■ ; [0201] To a cooled (-78 CC), stirred solution of oxalyl chloride (84 mmol) in
CH2GI2 (180 mL) was added a solution.pf DMSO (168 mmol) in CH2C12 (90 mL). After 1 h,
a solution of alcohol 6-6 (56 mmol) in CH2G2 (90 mL) was added. After 1 h, triethyl amine
(281 mmol) was added and stirred for a furtherhour. Then a solution of saturated aq. NH4CI
was added and allowed to warm to RT. The-organics were separated, washed with H2O (x2),
saturated brine (x2), thendried, filtered and evaporated under reduced pressure to give the .
.crude aldehyde. Purification by column chromatography affored the pure (S)-aldehyde 6-7.
[0202] Starting from the other enantiomer, (Z)-serine methyl ester leads to
the (R) enantiomer (6-8).-



__J02Q3] To a stirred solution of-compound 7-1 (10.0 rnmol) in 20 mLof
DCM was'acTded 10 mLofTFA. The mixture was stirred at RT for 24 h. The reaction
progress was followed by LC/MS. After completion, the solvent and TFA were removed by
evaporation,under reduced pressure and lyophilization to get white solid as TFA salts. The
crude solid was suspended in 50 mL of THF and N-carboethoxy phthalimide (10.5 mmol),
Et3N (10 mmol) were-added. The mixture was refluxed under N2 for 18 h. The reaction was
cooled and the solvents were evaporated. DCM was added and washed with water, brine,
dried over sodium sulfate, filter and concentrated. Purifipation by chromatography on silica
gel column (hexane/EtOAc) to give 2.68 g of colorless oil, compound 7-2..

[0204] To a stirred solution of (S)-3-((benzyloxy)carbonyl)-2-( 1,3-
. dio^pisoihdolin-2-yl)propanoic acid (compound 7-2, 6.07 mmol) in 30 mL of-dry THF at -15
°C were successively added N-methylmorpholine (6.07 mmol),.iso-butylchloroformate (6.07
rnmol): -After stirring for 5 min at -15 °C, a solution'of NaBK, (689 mg, 1.8.21 mmol) in 2.73,.

mL of water were added at once. The reaction was stirred at -15 °C for 2 min, then
hydro lyzed. with water (30 mL). Extracted with EtOAc (x 3), Washed With water (x 3), brine
.'(x-liadned over sodium sulfate, filled, concentrated. Purification by chromatography on
silica gel column (hexane/EtOAc) to give 1.9 g of colorless oil, compound 7-3.

[0205] . To a stirred solution of (S)-benzyl 4-hydroxy-3-(l,3-dioxoisoindolin-
2-.yl)butanoate (7-3, 5.6 mmol) in acetonitrile (28 mL) were added perfluoro-1-butane
sulfohyl fluoride (44.8 mmol), diisopropylethylamine (44.8 mmol), and
diisoprppylethylarninie trihydrofluoride (134 mmol). The mixture was stirred at 50 °C
overnight. The reaction progress was followed by LC/MS. After completion, the reaction
Was cooled'to RT and then evaporated under reduced pressure. The mixture was then
partitioned with DCM, washed with water (x 3), brine (x2), dried over sodium sulfate,
filtered, concentrated. Purification by chromatography on silica gel column (hexane/EtOA.c)
to give light yellow oil, compound 7-4.

[0206] To a stirred solution of (S)-benzyl 4-fluoro-3-(l,3-dioxoisoindolin-2-
-yl)butanoate (compound 7-4, 0.5 mmol) in dry ether (5 mL) was added dropwise to
diifoButylaluminum hydride (1.0 M in toluene, 1.5 mmol) at -78 °C. The reaction was stirred
at -78 °G for approximately 30 min as monitored by LC/MS. After'complction, the reaction
was.quenched by adding water (10 mL) at--78-pG. Extracted with ethyl acetate, washed with
water (x3), brine (x2), dried over sodium sulfate, filtered and concentrated. The crude
product, compound 7-5, was used in the next reaction step.


[0207] To prepare (S)-4-fluoro-3-(l,3-dioxoisoindolin-2-yl)butanoic acid,
compound 7-4 (0.20 mmol) was dissolved in ethanol (5 mL). This solution was purged with
nitrogen for 10 minutes, then 10% palladium on carbon was added, (0.02 mmol of palladium)
under an.atmosphere of nitrogen. Hydrogen was then bubbled rapidly through the solution,
while stirring, for approximately 1 h. The reaction progress was followed with LC/MS;
[0208] The reaction mixture was filtered through celite to remove the
palladium. The celite was rinsed twice with methylene chloride. The filtrate was then
concentrated to give the crude product, compound 7-6. The crude product was used for the
next- reaction step.

[0209] Compound 7-5 (0.20 mmol), .1,3 dicyclohexyl carbodiimide (0.30
mmbl), ethanethiol (0.6 mmol), and 4-dimethylaminopyridine (0.10 mmol) were dissolved in
DMF (5 mL). The mixture was stirred overnight at room temperature. The reaction was-
monitored with LC/MS.
[0210] EtOAc was added to the reaction mixture. This was then washed
with water (2x) and brine (2X). The EtOAc layer was then dried over sodium sulfate,-

filtered, and concentrated. The crude:product, compound 7-7, was then purified using flash
chromatography..

. [0211] Compound 7-7 (0.20 mmol) was dissolved in dry acetone (10 mL).
10% Palladium (0.02 rnrnol) on carbon was then added under an atmosphere of nitrogen.
Triethyl silane (0.5 mmol) was then added. Bubbling occurred after about 10 seconds, and
the reaction was allowed to continue until the bubbling ceased (30 rnin). The reaction was
monitored using LC/MS.
[0212] The reaction mixture was filtered through a celite plug.. The plug
was washed twice with methylene chloride, and the filtrate was then concentrated to give the
crude product, compound 7-5. The crude product was used in the next reaction.
[0213] Starting from the other (R) enantiomer, (R)-3-((benzyloxy)carbonyl)-
2-(l,3-dioxoisbindoiin-2-yl)propanoic acid, leads to the (R) enantiomer (7-8), having the.
following chemical structure:



Step A: Preparation of Compound 7-10
[0214) Methanol (300 mL) was charged to a 1000 mL round bottom flask
and the system wascooled with an ice bath. Ac.etyl chloride (89.3 mL; 1251 mmol) was
added dropwise over a period of 15 minutes. The resulting solution^vyas warmed to ambient
temperature and the (S)-2-amino-4-pentenoic acid (7-9) (6.0 g; 139 mmol) was added in a
•single portion. The reaction mixture was heated at reflux for two hours and was then cooled
to ambient temperature. The mixture was then concentrated in vacuo to provide a pale
yellow oil. The product was dispersed in ethyl acetate (150 mL) and was again concentrated
in vacuo. This sequence was repeated four times. The product 7-10 was an oil that solidified
upon standing under vacuum overnight. !H NMR analysis showed the product to be of
sufficient purity for use without further purification.
[021St TLC:R^1-L(silica;eluant 5:3:1 CHCl3:MeOH:(7:3 H20:-A;c£)H);
visualization with ninhydrin).
[0216] \ 'H NMR (400 MHz, CD3OD): 5.5.84-5,.73.(m,^,^.3^>5,26 (m,
2H), 4.17(dd, 1H, J=7;:0, 1.6 MHz), 3.84 (s, 311), 2.73-2.65 (m, 2H)- (400 MHz, c/6-DMSO):

S8.7 (br s, 3H), 5:81^/73 (m, 1H),:5.21-5.14 (m, 2H), 4.11 (t, \R, .7=6.1 Hz), 3.72.(s, 3H),
2:60 (dd,2H, .7=7.1, 0.9 Hz).
[0217] 13CNMR(101 MHz, c/6-DMSO): 5 169.33, 131,37,119.88, 52.65,:.
51.65, 34-.S2-.
StepB: Preparation of Compound 7-11
[0218] The crude (S)-methyl-2-arnino-4-pentenoate hydrochloride (7-10)
from the previous step was dissolved in THF (190 mL) with gentle warming. The resulting
solution was added dropwise to a solution of L1AIH4 in THF (280 mL of a 1.0 M solution) at
a rate such that the internal temperature remained at approximately 5 °C. Periodically, slight
heating was-used to warm the addition funnel containing the (S)-methyl-2-amino-4-
peritenbate hydrochloride solution to redissolve crystallized amino ester. Upon completion of
addition, the addition runnel was rinsed with an additional. 20 mL portion of THF. The
•mixture was then diluted with diethyl ether (500 mL) and the excess LiAffiU.was destroyed
by the sequential addition of H20 (11 mL), 15% (w/v) aqueous NaOH (11 mL) and H20 (33
mL) added at a rate such that the internal temperature remained below 10 °C. The mixture
_Was filtered..and the filter cake was washed with additional diethyl ether. The filtrate was
dried over Na2S04, filtered, and concentrated in vacuo to provide a yellow liquid (7-11; 13.4
g; 95% mass recovery based upon 139.0 mmol of (S)-2- amino-4-pentenoic acid). The amino
alcohol (7-11) may be purified by distillation (110 °C; 20 torr). However, minimal
improvement was observed in the subsequent step so the crude material was generally used
without further purification.
[0219]" 'HNMR (400 MHz, J6-DMSO): 5 5.87-5.77 (m, 1H), 5.05-4.97 (m,
2H),.3\26 (dd, 1H, J=10.3, 5.1 Hz), 3.14 (dd, 1H, J=10.3, 6.7 Hz), 2.69-2.63 (m, 1H),.2.15-
2.09 (m, 1H), 1.92-1.86 (m, 1H).
[0220] 13CNMR(101 MHz, d6-i>MSO): 5 136.49,116.31, 66.13, 52.53,
38.5L -
Step C: Preparation of Compound 7-12
[0221] (S)-2-Amino-4-pentenpl (7-11; 13.4 g; 132.5 mmol) and Na2COT
(70.8 g; 668.0 mmol) were dissolved in H20 (400 mL). CH3CN (700 mL) and methyl-2-
[(succihimidooxy)carbonyl]benzoate (33.1 g; 119.4 mmol) were added'and'the resulting

mixture was vigorously stirred at ambient temperature. After 2 hours', TLC analysis showed
the consumption of methyl-2-[(succimimidooxy)'carbonyl]benzoate.. The majority of the
CH3CN was removed on a rotary evaporator and the remaining material was transferred to a
separatory funnel and extracted with EtOAc (3 x 100 mL). - The combined EtOAc extracts
were washed with.0.5 M HCl (2 x 250 mL) and brine (250 mL). The EtOAc phase was dried
over;-Na2S04, filtered and concentrated in vacuo to provide a yellow oil (7-12; 19.3g; 70%)
that was used in the next step without further purification.
[0222] 'HNMR (4.00 MHz, ^6-DMSO): 5 7.90-7.83 (m, 4H), 5.74-5.64 (m,
1H), 4.99,4.91 (rh; 3H), 4.27-4.20 (m.'lH), 3.90-3.84 (m, 1H), 3.63-3.58 (in, 1H), 2.64-2.44
(m,2H).
[0223] 13CNMR(101 MHz, J6-DMSO): 5 168.25, 134.86, 134.42, 131.37,
122.94, 117.41, 60.63, 53.47,32.59.
Step D: Preparation of Compound 7-13
[0224] N,N-Diisopropylethylamine (215 mL; 1240 mmol), triethylamine
trihydrofluoride (81 mL; 496 mmol) and per-fluoro-1-butanesulfonyl fluoride (15.0 mL; 83.5
mmol) were added to a solution of 7-12 (19.1 g; 82.7 mmol) in PhCF3 (310 mL) and the
resulting mixture was stirred at ambient temperature. Additional perfluoro-1-butanesulfonyl
fluoride (7.5 mL; 41.8 mmol) was added after each of 60, 90, 120, 150, and 180 minutes.
After a total of 18 hours, the reaction mixture was transferred to a separatory funnel and was
washed twice with 1.0 N HCl, twice with saturated aqueous NaHC03 and once with H20.
The organic phase was dried over Na2S04, filtered, and concentrated to provide an orange oil.
The crude material was loaded onto a pad of silica and eluted with 4:1 hexane:EtOAc to
provide the product (7-13) as a yellow oil (15.4 g; 80%).
[0225] 'H NMR (400 MHz, d6-DMSO): 5 7.88-7.81 (m, 4H), 5.77-5.66 (m,
1H), 5.04-4.88 (m, 2.5H), 4.80-4.73 (mrtH), 4.65-4.61 (m, 0.5H), 4.60-4.49 (m, 1H), 2.68-
2,47 (m,2H).
[0226] ,3CNMR(101 MHz, J6-DMSO): 5 167.87, 134.79, 133.77, 130.94,
123.2.6,118.21, 81.82 (d,.^=170 Hz)r50:47 (d,y^l-9-Hz), 31.40 (d,J=6 Hz).


Step E: PreparationofCompound 7-8
[0227] Compound 743 (15.3 mmol) was dissolved in 2:1 CH30H:H20
(1500 mL) and a solution of OSCM in H20 (29.3 mL of a 4% w/v solution) was added. NaI04
•(42. ,2 g; .197.2 mmol) was then added in a single portion and the resulting mixture was stirred
at ambient temperature. After 3 hours, the mixture was filtered to remove precipitated solids
and the filter cake was washed with EtOAc. The filtrate was concentrated in vacuo to
remove the majority of the organic solvents. The residue was extracted with three portions of
EtOAc and the combined EtOAc extracts were dried over Na2SC>4, filtered, and concentrated.
The residue.was dissolved in CH2CI2, loaded onto a pad of silica gel and sequentially eluted
with 20%, 30%, 40%, 50%, and 100% EtOAc in hexane. Compound 7-8 was present in the
3O%-50% fractions but contaminated with a more-polar impurity. The fractions were
combined and concentrated and the residue was applied to a second pad of silica and eluted
with 30% EtOAc in hexane to provide Compound 7-8 as a light yellow solid (11.1 g; 72%)
[0228] 'H NMR (400 MHz, c?6-DMSO): 5 9.61 (s, 1H), 7.91-7.83 (m, 4H),
4.97-4.94 (m, 1H), 4.78 (t, 0.5H, ./=9.3Hz), 4.69-4.64 (m, 1H), 4.57-4.53 (m, 0.5H), 3.28-
3:02(m,'2H).
[0229] 13CNMR(101 MHz, 6-DMSO): 8 200.14, 167.65, 134.73, .13.1.15,
123.24, 81.80 (d,J=171Hz), 44.81 (d,y=21Hz), 40.64 (d, 7=6Hz).

[0230] Compound^7-9 was refluxed with 2.2 equivalents of phthalic
anhydride in the presence of 2.2 equivalents triethylamine in ethyl acetate until the reaction

was complete. The solvent was removed under pressure. The residual was dissolved in
water with a pH of 4 and then extracted with ethyl acetate. The combined organic layers
were washed twice with water having a pH of 4. Then, the organic phase was dried with;
sodium sulfate. The solvent was removed providing 7-14 as a white solid.
Step B: Preparation of Compound 7-12
[0231] Compound 7-14 and 1.2 equivalents of DIEA and 1.1 equivalent of
BOP in THF was stirred at room temperature until a clear solution formed. The solution was
cooled to 0 °C, and then 1.0 equivalent of NaBKU was added. The reaction mixture was
stirred at 0 °C under ~N2 until reaction completion. The solvent to changed to DCM and the
reaction was washed once with water. The DCM phase was loaded onto a silica gel plug, and
flushed with 15% EtOAc in hexanes to give Compound 7-12 as a colorless oil.

[0232] Azeotropic mixture of (S)-ethyl 3-(tert-butoxycarbonylamino)
butanoate 8-1 (1 eq.) and toluene (x=3) was dissolved in dichloromethane and cooled to -
■78°C. Then I'M solution of DIB AL in toluene (2 eq.) was added dropwise under N2
atmosphere and stirred at -78 °C for 2 h.
[0233] The reaction was quenched with methanol and concentrated. To the
concentrated residue, was added 2 M potassium sodium tartrate solution at 0°C and stirred
vigorouslyltroorn temperature for 30 min. The reaction mixture was partitioned between
ethyl acetate and water. The organic layer was washed with brine, dried over sodium sulfate,
filtered, evaporated and dried under reduced pressure to provide compound 8-2 as a light
yellow viscous liquid.
[0234] \MS:MH+ =188.2


[0235] To (R) -benzyl 3-aminobutyrate sulfate salt 9-1 (1 eq.) in THF was
added Boc-anhydride (2 eq.) and diisopropylethylamine (4 eq.). The reaction mixture was
stirred at room temperature for 72 h. The reaction mixture was concentrated and partitioned
between ethyl acetate and water. The organic layer was separated, washed with water and
brine, dried over sodium sulfate, filtered, evaporated and dried under reduced pressure to
provide compound 9-2 as a white solid.
[0236] MS:MH+ =294.0

[0237] Azeotropic mixture of (R)-((benzyl) 3-(tert-butoxycarbonylamino)
butanoate 9-2 (1 eq.) and toluene (x=3) was dissolved in dichloromethane and cooled to -
78°C. A 1 M solution of DIBAL in toluene (2 eq.) was added dropwise under N2 atmosphere
and stirred at -78°C for 2 h. The reaction was quenched with methanol and then
concentrated. To the concentrated residue was added 2 M potassium sodium tartrate solution
at 0 °.Q and.stirred vigorously at room temperature for 30 min. The reaction mixture was
partitioned between ethyl acetate and water. The organic layer was washed with brine, dried
over sodium sulfate, filtered, evaporated and dried under reduced pressure to provide
compound 9-3 as a colorless viscous liquid.


[0239] To 3-amino-3-methyl-butyric acid 10-1(1 eq.) in methanol at 0°C
"was added 2 eq. of thionyl chloride. The reaction mixture was warmed to room temperature
and stirred overnight. The solvent was evaporated-to give azeotropic mixture of 10-2 and
toluene (x=3) which was used for Step B.
; [0240] MS:MH+ =132.1

[0241] To methyl 3-amino-3-methylbutanoate HC1 salt 10-2 (1 eq.) in THF
was addedB.oc-anhydride (2 eq.) and diisopropylethylamine (4 eq.). The reaction mixture
was stirred at room temperature for 48 h. The reaction mixture was concentrated and
partitioned between ethyl acetate and water. The organic layer was separated, washed with
water and.brine, dried over sodium sulfate, filtered, evaporated and dried under reduced
pressure to provide product 10-3 as a white solid.
[0242] MS:MH+ =232,1


[0243] Azeo'tropic mixture of methyl 3-tert-butoxycarbonylarhino)-3-
methylbutanoate 10-3 (1 eq.) and toluene (x=3) was dissolved in dichlorbmethane and cooled
to -78°C. To this was added dropwise 1 M solution of DIBAL in toluene (2 eq.) under N2
atmosphere and stirred at -78°C for 2 h. The reaction was quenched with methanol and
concentrated. To concentrated residue was added 2 M potassium sodium tartrate solution at
0°C and stirred vigorously at room temperature for 30 mih. The mixture was partitioned
between ethyl acetate and water. The organic layer was washed with brine, dried over
sodium sulfate, filtered, evaporated and dried under reduced pressure to provide product 10-4
as a colorless viscous liquid.
[0244] MS: MH+ =202.1


[0245] . To a 5-necked flask containing 1 eq. of Compound 11-1 was added
0.7 eq. of K2CO3 to give a 0.25M solution of K2CO3 in acetone. After being stirred under N2
for 45 minutes, 1.0 eq. of Compound 11-2 in 1 M of acetone was added followed by addition
• of 0.2 eq. of KI in 5 M acetone. When the reaction is completed (about 3 hours), the reaction
mixture was cooled with an ice bath. Ice water (equal to approximately 2.5 X volume of
acetone used in reaction) was added via addition funnel at such a speed that the temperature
- did not exceed 15 °C. After being stirred in an ice bath for one hour, the product was
collected by vacuum filtration. The filter cake was.washed 3 times with 20% acetone and 3
. .times, with water. The filter cake was air-dried and further dried in an oven, at 50°C/5 torr
until a consistent weight is reached. yleld 96%;. HPLC purity; 99%;

StepB: Preparation of Compound 11-4
[0246] To a 0.19 M toluene solution of 11-3 in a reaction flask was added 20
eq. of NKjOAc. The rhixture was stirred under reflux until the reaction was completed'
(about §_h). It was cooled to RT and then water (equal to approximately one fourth of the
volume of toluene used in the reaction) was added, the organic phase was separated and
washed with water, sat. NaHGCh, and dried overMgSO^ The solvent was removed in vacuo
to give 11-4. yleld 99.6%. HPLC purity: 91.4%.
Step C: Preparation of Compound 11-5
[0247] A flask containing a 0.5 M DMF and K2C03 solution of 11-4 was
stirred under N2 for 30 min at 6-5 °C, and then 1.1 eq. of PhCH2Br was added to it. The .
mixture was then stirred at RT overnight. It was then stirred in an ice bath during which ice
water (approximately equal to the volume of DMF used in the reaction) was added dropwise.
The product was collected by vacuum filtration, washed twice with 50% DMF, twice with
25% DMF and three times with water. The solid was.drigdjn an oven at_5P..0C / 5 torr. yleld
95%. HPLC:purity: 94%.
Step D: Preparation of Compound 11-6
[0248] MeOH was added to a flask and placed in an ice b,ath._To.this was. -
added 9.85 eq. of CH3COCl dropwi'se over 30 min. followed by 1 eq. of 11-5 to form.a
0.25M solution of 11-5 in MeOH. The mixture was stirred at RT until the reaction was
completed (about 12 h). After removing the solvent under-reduced pressure, the obtained
solid was suspended in MeOH (equal to approximately one half of the volume of MeOH
used in the reaction) and"stirred afO-5 °C. To this mixture were added 2.5 M NaOH / MeOH
solution dropwise until the pH reached about-10 and water was then added. After being
stirred at 0-5 °C for 1 h,lhe product was collected by filtration. It was dried in an oven at 50
°C7 5 torr. yleld 90.5%. HPLC purity: 97.0%. Optical purity was determined to be >99%
(enantiomeric excess (ee)).


[0249] To a 500 mL round bottom flask was added Compound.l2jJ C7.95;g;__
25.7 mmol), 16.0 g of 2,6-di-tert-butyl-4-methyl pyridine (16.0 g; 77.9 mmol) and 100 mL
anhydrous DCM. Subsequently^ 11.36 g of methyl-trifluoromethane sulfonate was added.
The*eaction mixture was stirred at room temperature under N2 and monitored by HPLC.
HPLC monitoring showed att^-17 h that there was 76.0% Compound 12-2, 9.9% Compound
12-1, 14.1% byproduct; at t=20 h, there was 76.4% Compound 12-2, 16.2% byproduct, 7.4%
Compound 12-1. The reaction was stopped by filtering out the solids. The solids were
washed with CH2C12. The combined filtrate was washed with 0.5 N HC1 (2 x 100 mL) and
dried with Na2SQ4 arid concentrated. Flash column chromatography (200 g silica gel, 20%
EtOAc in hexanes) gave 5.28g of tan oilas 12-2. HPLC purity=93.9% and used for the next
step.
Step B: Preparation of Compound 12-3
10250] A solution of Compound 12-2 (1.1 Og, 3.6 mrnol)in anhydrous DCM
(22 mL) was cooled to -78 °C. DIBAL (7.12 mL of1.0M solution in CH2Cl2)was added.
The reaction mixture was stirred at -120 °C ±3 °C (external temperature) and monitored by
HPLC. An in process control (IPC) sample was instantly quenched in pre-chilled MeOH'St-
120 °C and then prepared for HPLC for t=0 reading, the mtemal. temperature was

maintained at -118 °C + 3 °C. AUEPC samples :(t=0,,t=l h, t=2 h) indicated, the absence of
Compound 12-2 in the reaction mixture. The reaction was deemed complete at t=3 h and
quenched with methanol. A temperature of-120 °C + 2 °C was maintained during the entire
quenching process. HPLC indicated reaction mixture contained an aldehyde: alcohol ratio of
95.1:4.9. The reaction mixture was concentrated to dryness then redissolved inCHkCla,.
washed in Na2C03 (2 x 50 mL) and brine. (2 x 50 mL), dried with Na2S04 and concentrated
to a pale yellow oil. HPLC indicated 82% pure for Compound 12-3.
[0251] The compounds in the table below were prepared using the
•methodology described in the previous Examples and Methods. The following tables also
include compounds described in the experimentals. The starting materials used in the
synthesis are recognizable to one of skill in the art and are commercially available or may be
prepared using known methods. The compounds in Table 1 were named using ACD/Name
Batch Version 5.04 (Advanced Chemistry Development Inc.; Xojonto, Ontario;
www.acdlabs.com). The compounds in Table 2 and Table 3 were named using AutoNom
2000 (A_ylpmatic Nomenclature) for ISIS/Base, implementing IUPAC standardized
-nomenclature. In one embodiment, provided is a stereoisomer of any one of the compounds
in Tables 1, 2, or 3. In one aspect, the stereoisomer is an enantiomer. In another aspect, the
stereoisomer is a diastereomer.































































[0252] This example provides a representative in vitro assay for determining
CSP activity in vitro. Purified microtubules obtained from bovine brain were purchased from

Cytoskeleton Inc. (Denver, Colorado, USA). The motor domain of human KSP (Eg 5,
KNSL1) was cloned, expressed, and purified to greater than 95% homogeneity. Biomol
Green was purchased from Affinity Research Products Ltd. (Matford Court, Exeter, Devon,
.United Kingdom). Microtubules and KSP motor protein (i.e., the KSP motor domain) were
diluted in assay buffer (20 mM Tris-HCl (pH-7.-5), 1 rnM MgCl2,10 mM DTT and 0.25
mg/mL BSA) to a final concentration of 35 /xg/mL microtubules and 45 nM KSP. The
microtubule/KSP mixture was then pre-incubated at 37 °C for 10 min to promote the binding
of KSP to microtubules.
[0253] To each well of the testing plate (384-well plate) containing 1.25 /xL
of inhibitor or test compound in DMSO (or DMSO only in the case of controls) were added
25 /xL of ATP solution (ATP diluted to a concentration of 300 JJM in assay buffer) and 25 /xL
of the above-described microtubule/KSP solution. The plates were incubated at RT for 1
hour. Following incubation, 65 ilL of Biomol Green (a malachite green-based dye that
detects the release of inorganic phosphate) was added to each well. The plates were
incubated for an additional 5-10 minutes then the absorbance at 630 nm was determined
' using a Victor II plate reader. The amount of absorbance at 630 nm corresponded to the
amount of KSP activity in'the samples." The IC50 of each inhibitor or test compound was then
determined based on the decrease in absorbance at 630 nm at each concentration, via
nonlinear regression using either XLFit for Excel or Prism data analysis software by
GraphPad Software Inc.
[0254.] Preferred compounds of the invention have a biological activity as
measured by an IC50 of less than about 1 mM in assay protocols described in Example 13,
with preferred embodiments having biological activity of less than about 25 uM, wjth
particularly preferred embodiments having biological activity of less than about 1000 nM,
and with the most preferred embodiments having biological activity of less than about 100
nM,.
Example 14
Inhibition of Cellular Proliferation in Tumor Cell Lines Treated with KSP Inhibitors
[0255] Cells are. plated in 96-well plates at densities of about 500 cells per
well of a 96-well plate and are allowed to grow for 24 hours. The cells are then treated with
various concentrations of compounds for 72 hours. Then, 100 /xl of CellTitcr Glo is added.


CellTiter Gib is a tetfaiolium-based'assay using the reagent 3-(4,5-dimeiJiylthiaz6l-2-yl) 5-
(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) (U.S. Patent No.
5,185,450) (seePromega product catalog #G3580, CellTiter 96 Aqueous One Solution Cell
Proliferation Assay). The cells are then incubated in the dark for 30 minutes. The amount of
luminescence is determined for each well using a Walloc Trilux plate reader, which correlates
with the number of cells per well. The number of viable cells in the wells that receive only
DMSO (0.5%) serve as an indication of 0% inhibition, while wells without cells serve as
100% inhibition of cell growth. The compound concentration'that results in a 50% growth
inhibition (GI50) is determined graphically from sigmoidal dose-response curves of log-
transformed dose values versus cell counts (percent .of control) at 72 hours of continuous
compound exposure,
[0256] The cell lines used are listed below
[0257] The cell proliferation assay is performed as described above.
Cancer Cell Lines
Colo 205 - colon carcinoma.
RPMI1640 +10%FBS+1 % L-glutamine +1 % P/S +1 %NaPyr.+
Hepes
+4.5g/L Glucose +l%NaBicarb.
MDA 435- breast cancer- high met
EMEM + 10% FBS + 1%P/S + 1%L-Glutamine+1%NEAA
+l%NaPyr+l%vitamins
HCT-15 and HCT116 -colon carcinoma
RPMI 1640 +10%FBS +1% L-glutamine +1% P/S
Drug Resistant Cell Lines
KB3.1- colon epidermal carcinoma; parental cell line
Iscove's +10%FBS +1% L-glutamine +1% P/S
KBV1-p-glycoprotein associated multi-drug resistant cell line
RPMI 1640+10%FBS+1% L-glutamine+1% P/S+0.2ug/mL
Vinblastine
KB85 - p-glycoprotein associated multi-drug resistant cell line
DMEM +10%FBS +1% L-glutamine +1% PAS + 1 One/mL Colchicine
[0258] Preferred compounds of the invention have a biological activity as
measured by an GIjo of less than about 1 mM in assay protocols described with some
embodiments having biological activity of less than about 25 uM, with otherembpdiments


hayln^vbidlogicd Activity of less than about 1000 nM;-arid with still other embodiment
having, a GI50 of less than about 100 nM.
Example 15
Clonogenic Softagar Assay Protocol
[0259] Human cancer cells are plated at a density of 3xl05 cells per well in a
6-v/ell plate. The next day, a corhpound of interest at a certain concentration is added to each
well. After 24 and 48 hours of incubation, the cells are harvested, washed and counted. The
following steps are performed using the Multimek 96 robot. Then, 500 viable cells per well
are plated in a 96-well plate that is coated with PolyHema to prevent attachment of the cells
to the bottom of the well. Agarose (3% stock) is melted, diluted in warmed media and added
to the cells to a final concentration of 0.5%. After the soft agar solidified, the plates are
incubated at 37 °C for 6 days. Alamar blue dye is added to cells and plates are incubated for
an additional 6 hours. The optical density change is measured on a Tecan plate reader and is
considered to correlate with the number of colonies formed in soft agar. A cancerous cell is
able to grow on the agar and thus will show an-increase in optical density. A reading of
decreased,optical density means that the cancer cells are being inhibited. It is contemplated
that compounds of this invention will exhibit a decrease in optical density.

WE CLAIM:
1. A compound of formula I:

wherein:
R1 is selected from the group consisting of arninoacyl, acylamino, carboxyl, carboxyl
ester, alkyl, and substituted alkyl, with the proviso that substituted alkyl is not substituted
with aryl or substituted aryl;
R2 is selected from the group consisting of hydrogen, alkyl, and aryl;
R3 and R4 are independently selected from the group consisting of hydrogen, hydroxy,
alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic, provided that only 1 of R3
or R4 is hydroxy,
or R3 and R4 together with the nitrogen atom pendent thereto join to form a
heterocyclic or substituted heterocyclic;
R5 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl,
heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, and substituted
heteroaryl;
or R1 and R5, together with the carbon and nitrogen atoms bound respectively thereto
join to form a heterocyclic or substituted heterocyclic group;
or when R1 and R5, together with the carbon and nitrogen atoms bound respectively
thereto, do not form a heterocyclic group, then R4 and R5, together with the atoms bound
thereto, form a heterocyclic or substituted heterocyclic group;
R8 is selected from the group consisting of L-A1, wherein L is selected from the group
consisting of -S(O)q- where q is one or two, and C1 to C5 alkylene optionally substituted with
hydroxy, halo, or acylamino; and

A1 is selected from the group consisting of aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, and substituted
cycloallcyl; and
one of either R6 or R7 is selected from the group consisting of cycloalkyl,
heterocyclic, aryl and heteroaryl, all of which may be optionally substituted with -(R9)m
where R9 is as defined herein and m is an integer from 1 to 4, and
the other of R6 or R7 is selected from the group consisting of hydrogen, halo, and
alkyl;
R9 is selected from the group consisting of cyano, alkyl, substituted alkyl, alkenyl,
substituted alkenyl, alkynyl, substituted alkynyl, -CF3, alkoxy, substituted alkoxy, halo, and
hydroxy;
and when m is an integer from 2 to 4, then each R9 may be the same or different;
or pharmaceutically acceptable salts or esters thereof.
2. The compound as claimed in claim 1, wherein the compound is of formula II:

wherein:
n is 1,2, or 3;
p is 0, 1,2, 3, or 4;
R3 and R4 are independently selected from the group consisting of hydrogen, hydroxy,
alkyl, substituted alkyl, cycloallcyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, and substituted heterocyclic, provided that only 1 of R3
or R4 is hydroxy;
or R3 and R4 together with the nitrogen atom pendent thereto join to form a
heterocyclic or substituted heterocyclic;

R10 is selected from the group consisting of hydrogen, alkyl optionally substituted
with a substituent selected from the group consisting of hydroxy, alkoxy, substituted alkoxy,
amino, substituted amino, acylamino, halo, nitrogen-containing heterocycle, substituted nitrogen-containing heterocycle, nitrogen-containing heteroaryl, and substituted nitrogen-
containing heteroaryl;
R11 is selected from the group consisting of cyano, alkyl, allcenyl, alkynyl, -CF3,
alkoxy, halo, and hydroxy; provided that when p is 2 - 4, then each R11 may be the same or '
different;
R12 is alkyl;
R13 is hydrogen or alkyl,
R14 isselected from the group consisting of hydrogen, halo, and alkyl; or R10 and R12, together with the carbon and nitrogen atoms bound respectively
thereto join to form a heterocyclic or substituted heterocyclic group;
or when R10 and R12, together'with the carbon and nitrogen atoms bound respectively
thereto, do not form a heterocyclic group, then R4 and R10, together with the atoms bound
thereto, form a heterocyclic or substituted heterocyclic group;
A2 is selected from the group consisting of aryl, substituted aryl, heteroaryl,
substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkyl, and substituted
cycloalkyl;
or pharmaceutically acceptable salts or esters thereof.
3. The compound as claimed in claim 1, wherein R1 is alkyl.
4. The compound as claimed in claim 3, wherein R1 is selected from the group consisting of
isopropyl, t-butyl, and propyl.
5. The compound as claimed in claim 1, wherein R2 is hydrogen.
6. The compound as claimed in claim 1, wherein R3 and/or R4 is selected from the group consisting
of alkyl, substituted alkyl, and cycloalkyl.

7. The compound as claimed in claim 6, wherein the alkyl, substituted allcyl or cycloalkyl is
selected from the group consisting of methyl, methoxyethyl, furan-2-ylmethyl, 2-
hydroxyethyl, cyclopropyl and isopropyl.
8. The compound as claimed in claim 1„ wherein R3 and/or R4 is aryl or substituted aryl.
9. The compound as claimed in claim 8, wherein the aryl or substituted aryl is selected from the
group consisting of 4-cyanophenyl, 3,4-difluorophenyl, 2,3,5-trifluorophenyl, 3,5-
dinitrophenyl, and phenyl.
10. The compound as claimed in claim 1, wherein R3 and/or R4 is heteroaryl or substituted
heteroaryl.
11. The compound as claimed in claim 10, wherein the heteroaryl or substituted heteroaryl is
selected from the group consisting of thiophen-2-yl, 3,5-dirnethylisoxazol-4-yl, and 2,6-
dichloropyridin-4-yl.
12. The compound as claimed in claim 1, wherein R3 and/or R4 is a heterocyclic group or substituted
heterocyclic.
13. The compound as claimed in claim 12, wherein the heterocyclic or substituted heterocyclic group
is tetrahydropyran-4-yl or 4-(ethoxycarbonyl)piperidin-4-yl.
14. The compound as claimed in claim 1, wherein either of R3 or R4 or both of R3 and R4 are
hydrogen.
15. The compound as claimed in claim 1, wherein one of R3 or R4 is hydroxy.
16. The compound as claimed in claim 1, wherein R3 and R4 are cyclized with the nitrogen atom
bound thereto to form a heterocyclic or substituted heterocyclic.

17. The compound as claimed in claim 16, wherein the heterocyclic, substituted heterocyclic,
heteroaryl or substituted heteroaryl is selected from the group consisting of 1,1-
dioxothiamorpholin-N-y1,1-oxothiamorpholin-1-yl, 2-(aminomethylene)pyrrolidin-N-yl,2-
(methoxycarbonyl)pyrrolidin-N-yl, 2,6-dimethylmorpholin-N-yl, 3-hydroxypiperidin-N-yl, 3-
hydroxypyrrolidin-N-yl, 4-(butylsulfonyl)piperazin-N-yl, 4-(cyclopropylsulfonyl)piperazin-
N-yl, 4-(dimethylarnino)piperidin-N-yl, 4-(ethoxycarbonyl)piperazin-N-yl, 4-
(ethylsulfonyl)piperazin-N-yl, 4-(isopropylsulfonyl)piperazin-N-yl, 4-
(methylcarbonyl)piperazin-N-yl, 4-(methylsulfonyl)piperidin-N-yl, 4-
(methysulfonyl)piperazm-N-yl, 4-(morpholin-N-yl)piperidin-N-yl, 4-(piperidin-N-
yl)piperidin-N-yl, 4-(propylsulfonyl)piperazin-N-yl, 4-cyclohexylpiperazin-N-yl, 4-
hydroxypiperidin-N-yl, 4-isopropylpiperazin-4-yl, 4-methylpiperidin-N-yl, isoxazoIidin-2-yl,
morpholin-N-yl, piperazin-N-yl, piperidin-N-yl, 2-(hydrazinocarbonyl)pyrrolidin-N-yl and
pyrrolidin-N-yl.
18. The compound as claimed in claim 1, wherein R5 is selected from the group consisting of
-(CH2)3NH2, -(CH2)2CH(CH2OH)NH2, -CH2CH(F)CH2NH2, -CH2-[2-(CH2OH)pyrrolidin-3-
yl], -CH2-[4-(OH)pyrrolidin-3-yl], -CH2-C(F)(spiropyrrolidin-3-yl), -(CH2)2CH(CH2F)NH2,
-(CH2)2C(CH3)2NH2, -(CH2)2CH(CH3)NH2,-(CH2)2CH(CH2OCH3)NH2,
-(CH2)2CH(CH2F)NHC(O)-[(2-CH3NHC(O))benzene], and -(CH2)2CH(CH2F)-1,3-dioxo-
1,3-dihydroisoindole.
19. The compound as claimed in claim 1, wherein R1 and R5 and the atoms bound thereto join to
form a heterocyclic or a substituted heterocyclic group.
20. The compound as claimed in claim 19, wherein the substituted heterocyclic group is 2-oxo-
tetrahydropyrimidinyl.
21. The compound as claimed in claim 1, wherein one of R6 or R7 is aryl or substituted aryl.
22. The compound as claimed in claim 21, wherein the aryl or substituted aryl is selected from the
group consisting of phenyl, 3-chlorophenyl, 3-fluorophenyl, 2,4-diflurophenyl, 2,5-
difluorophenyl, and 2,3,5-trifluorophenyl.

23. The compound as claimed in claim 1, wherein the other of R6 or R7 is hydrogen.
24. The compound as claimed in claim 1, wherein L is alkylene and A1 is aryl or substituted aryl.
25. The compound as claimed in claim 24, wherein L is methylene and A1 is selected from the group
consisting of phenyl, 3-fIuorophenyl and 3-hydroxyphenyl.
26. A compound selected from the group consisting of:
N-(3-aminopropyl)-N-{(1R)4-[1-benzyl-4-(3-chlorophenyl)-1H-imidazol-2-
yl]-2-methylpropyl}morpholine-4-carboxamide;
N-(3-aminopropyl)-N-{(1R)-1-[1-benzyl-4-(3-chlorophenyl)-1H-imidazol-2-
yl]-2-methylpropyl}piperidine-1-carboxamide;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]piperidine-1-carboxamide;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dirnethylpropyl]-N',N'-dimethylurea;
N-(3-aminopropyl)-N-[(1R)4-(1-benzyl-4-phenyl-1H-rmidazol-2-yl)-2,2-
dimethylpropyl]-N'-(2-methoxyethyl)urea;
methyl (2S)-1-({(3-aminopropyl)[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]amino}carbonyl)pyrrolidine-2-carboxylate;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-hydroxyurea;
N-(3-arrunopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-irnidazol-2-yl)-2,2-
dirnethylpropyl]pyrrolidine-1 -carboxamide;
(2R)-2-(aminomethl)-N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
irmdazol-2-yl)-2,2-dimethylpropyl]pyrrolidine-1-carboxamide;
(3S)-N-(3-amimopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-
2,2-dimethylpropyl]-3-hydroxypyrrolidine-1-carboxamide;
(3R)-N-(3-arninopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-
2,2-dimethylpropyl]-3-hydroxypyrrolidine-1-carboxamide;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-methylurea;
N-(3-aminopropyl)-N4(1R)-1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]piperazine-1-carboxamide;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-irmdazol-2-yl)-2,2-
dimethylpropyl]-1,4'-bipiperidine-1-carboxamide;
N-(3-aminopropyl)-N-[(1R)-1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-thien-2-ylurea;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N-thien-3-ylurea;

N-(3-arninopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-(3,5-dimethylisoxazol-4-yl)urea;
N-(3-aminopropyl)-N-[(1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl3-N'-(2,6-dichloropyridin-4-yl)urea;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-(2-furylmethyl)urea;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-(4-cyanophenyl)urea;
N-(3 -aminopropyl)-N-[(l R)-1 -(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-(3,4-difluorophenyl)urea;
N-(3-aminopropyl)-N4(1R)-1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N-(3,5-dinitrophenyl)urea;
N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]-N'-phenylurea;
(3R)-N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-3-hydroxypyrrolidine-1-carboxamide;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]piperazine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]piperazine-1 -carboxamide;
(3R)-N-(3-aminopropyl)-N-[(1R)4-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-
2,2-dimethylpropyl]-3-3-hydroxypiperidine-1-carboxamide;
(3S)-N-(3-aminopropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-
2,2-dimethylpropyl]-3-hydroxypiperidine-1-carboxamide;
N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N',N'-dimethylurea;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N'-phenylurea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N',N'-dimethylurea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N'-phenylurea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N'-(2-methoxyethyl)urea;
4-acetyl-N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]piperazine-1 -carboxamide;
(4R)-N4(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imdazol-2-yl)-2,2-dimethylpropyl]-4-hydroxylsoxazolidine-2-carboxamide;

(3R)-N-(3-amino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-3-hydroxypiperidine-1 -carboxamide;
N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N'-(2-methoxyethyl)urea;
(3R)-N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)2,2-dimethylpropyl]-hydroxypyrrolidine-1-carboxamide;
(3S)-N-[(2k)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-3-hydroxypyrrolidine-1-carboxamide;
(3S)-N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-3-hydroxypiperidine-1-carboxamide;
ethyl 4-({[(2S)-3-amino-2-fluoropropyl][(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]amino}carbonyl)piperazine-1-carboxylate;
ethyl 4-({[(2R)-3-amino-2-fluoropropyl][(1R)-1-(l -benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]amino} carbonyl)piperazrne-l -carboxylate;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(l R)-1 -(1 -benzyl-4-phenyl-1H-imidazol- 2-yl)-2,2-dimethylpropyl] -4-(ethylsulfonyl)piperazine-1 -carboxamide;
N-[(2S)-3-amino-2-fluoropropyl] -N-[( 1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-hydroxypiperidine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-hydroxypiperidirie-1-carboxamide;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-1,4-bipiperidine-1'-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimetbylpropyl]-1,4'-bipiperidine-1'-carboxamide;
N-(3-aTnino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-4-methylpiperazine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-methylpiperazine-1-carboxamide;
N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N'-(2-hydroxyethyl)-N'-methylurea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-irnidazol-
2-yl)-2,2-dimethylpropyl] -N'-(2-hydroxyethyl)-N'-methylurea;
l-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-
dimethylpropyl]tetrahydropyrimidin-2(1H)-one;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N,-(2-hydroxyethyl)-N'-methylurea;
N~[(2R)-3-amino-2-fluoropropyl]-N-[(l R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N'-(3,4-difluorophenyl)urea;

N-[(2S)-3-amino-2-f[uoropropyl]-N-[(1R)4-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N,-thien-3-ylurea;
N-(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N,-thien-3-ylurea;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-morpholin-4-ylpiperidine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-motpholin-4-ylpiperidine-1-carboxamide;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]morpholine-4-carboxamide;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-4-(methylsulfonyl)piperazine-1 -carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl] -4:(methylsulfonyl)piperazine-1 -carboxamide;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-4-cyclohexylpiperazine-1 -carboxamide;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl] -N'-(4-cyanophenyl)urea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-N'-(4-cyanophenyl)urea;
N-[(2S)-3-amino-2-fluoropropylJ-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-(propylsulfonyl)piperazine-1 -carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropylJ-4-(propylsulfonyl)piperazme-1-carboxamide;
N-[(2S)-3-arnino.2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-(isopropylsulforiyl)piperazine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2;2-dimethylpropyl]-4-(isopropylsulfonyl)piperazme-1-carboxamide;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-(cyclopropylsulfonyl)piperazine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-rmidazol-
2-yl)-2,2-dimemylpropyl]-4-(cyclopropylsulfonyl)piperazine-1-carboxamide;
N-[(2S)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-irnidazol-
2-yl)-2,2-dimethylpropyl]-4-(butylsulfonyl)piperazine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-(butylsulfonyl)piperazine-1 -carboxamide;
N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl-)-2,2-dimethylpropyl]-1,4'-bipiperidine-1 '-carboxamide;

N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N'-(4-cyanophenyl)urea;
N-[(3S)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2,2-dimethylpropyl]-N'-(354-difluorophenyl)urea;
N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl]-N-
{[(3S)-3-fluoropyrrolidin-3 -yl]methyl} -1,4'-bipiperidine-1 '-carboxamide;
N-[(1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl] -N-
{[(3R)-3-fluoropyrrolidin-3-yl]methyl} -1 ,4'-bipiperidine-1 '-carboxamide;
N-[(1R)4-(1-benzyl-4-phenyl-1H-imddazol-2-yl)-2,2-dimethylpropyl]-N-
{[(2S ,3S)-2-(hydroxymethyl)pyrroIidin-3-yl]methyl} -1,4'-bipiperidine-1 '-carboxamide;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-N-tetrahydro-2H-pyran-4-ylurea;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-irnidazol-
2-yl)-2,2-dimethylpropyl]-N,-tetxahydro-2H-pyran-4-ylurea;
N-(3-amino-2-fluoropropyl)-N-[(1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-2-
yl)-2,2-dimethylpropyl]-4-(dimethylamino)piperidine-1-carboxamide;
N-[(2R)-3-amino-2-fluoropropyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-
2-yl)-2,2-dimethylpropyl]-4-(dimethylamino)piperidine-1-carboxamide;
N-[(1R)-1 -(1 -benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl]-N-
{[(3R,4R)-4-hydroxypyrroiidin-3-yl]methyl} -1,4'-bipiperidine-1 '-carboxamide;
N-[(1R)-1 -(1 -benzyl-4-phenyl-1 H-imidazol-2-yl)-2,2-dimethylpropyl]-N-
{[(3S,4S)-4-hydroxypyrrolidin-3-yl]methyl} -1,4-bipiperidine-1 '-carboxamide;
N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl]-N-[(3-
fluoropyrrolidin-3 -yl)methyl] -4-(methylsulfonyl)piperazine-1 -carboxamide;
N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl]-N-
{[(3S)-3-fluoropyrrolidin-3-yl]methyl}-4-(methylsulfonyl)piperazine-1-carboxamide;
N-[(1R)-1-(1-benzyl-4-phenyl-1H-imidazol-2-yl)-2,2-dimethylpropyl]-N-
{[(3R)-3-fluoropyrrolidin-3-yl]methyl} -4-(methylsuIfonyl)piperazine- 1-carboxamide;
N-[(3R)-3-amino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidazol-2-yl)-2;2-dimethylpropyl]-4-(methylsulfonyl)piperazine-1-carboxamide;
N-[(3S)-3-arnino-4-hydroxybutyl]-N-[(1R)-1-(1-benzyl-4-phenyl-1H-
imidaazol-2-yl)-2,2-dimethylpropyl]-4-(methylsulfonyl)piperazine-1-carboxamide;
1-((R)-3-amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(3-fluoro-pbenyl)-1H-
imidazol-2-yl]-2,2-dimethyl-propyl}-3,3-dimethyl-urea;
1-((S)-3-amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-
imidazol-2-yl]-2,2-dimethyl-propyl}-3,3-dimethyl-urea;
1,1 -Dioxo-1 -thiomorpholine-4-carboxylic acid ((R)-3-amino-4-fluoro-butyl)-
{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;

Morpholine-4-carboxylic acid ((R)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-Methanesulfonyl-piperidine-1 -carboxylic acid ((S)-3 -amino-4-fluoro-bixtyl)-
{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1 -((S)-3-Amino-4-fluoro-butyl)-l - {(R)-l -[ 1 -(3-fluoro-benzyl)-4-(3 -fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
1-(R)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-(3-fluoro-benzyl)-4-(3-fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
1,1-Dioxo-1-thiaomorpholine-4-carboxylic acid ((S)-3-amino-4-fIuoro-butyl)-
{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-Methanesulfonyl-piperidine-1-carboxylicacid((R)-3-amino-4-fluoro-butyl)-
{(R)-1 -[ 1 -benzyl-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
1-((R)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-(3-fiuoro-benzyl)-4-(3-fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-urea;
1-(3-Amino-3-methyl-butyl)-1-{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-
imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
1-(3-Amino-3-methyl-butyl)-1-{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H- imidazol-2-yl]-2,2-dimethyl-propyl}-3,3-dimethyl-urea;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-(3-fluoro-benzyl)-4-(3-fluoro-
phenyl)-1H-irmdazol-2-yl]-2;2-dimethyl-propyl}-urea;
4-Methyl-piperazine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-
[1-(3-fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-Methyl-piperazine-1 -carboxylic acid ((R)-3-amino-4-fluoro-butyl)- {(R)-1 -
[1-(3-fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-irddazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-(3-Amino-3-methyl-butyl)-1-{(R)-1-[1-benzyl-4-(3-fluoro-phenyl)-1H-
imidazol-2-yl]-2,2-dimethyl-propyl}-urea;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-irnidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((R)-3-amino-4-fluoro-butyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1 -((S)-3-Amino-4-fluoro-butyl)-1 - {(R)-1 -[ 1 -(3 -fluoro-benzyl)-4-(3 -fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3,3-dirnethyl-urea;
1-((R)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-(3-fluoro-benzyl)-4-(3-fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3,3-dimethyl-urea;
Pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fliioro-butyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;

Pyrrolidine- 1-carboxylic acid ((R)-3-amino-4-fluoro-butyl)-{(R)-1-[1-(3-
fluoro-benzyl-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((S)-3-amino-2-fIuoro-propyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)4H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Pyrrolidine-1 -carboxylic acid ((S)-3-amino-2-fluoro-propyl)- {(R)-1 -[ 1 -(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1 -((S)-3-Amino-4-methoxy-butyl)-1 - {(R)-1 -[1 -(3-fluoro-benzyl)-4-(3-fluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-rnethyl-urea;
Pyrrolidine-1-carboxylic acid ((S)-3-amino-4-methoxy-butyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl] -2,2-dimethyl-propyl} -amide;
Morpholine-4-carboxylic acid ((S)-3-amino-4-methoxy-butyl)-{(R)-1-[1-(3-
fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1,1 -Dioxo-1 -thiomorpholine-4-carboxylic acid ((S)-3-amino-2-fluoro-propyl)-
{(R)-1-[1-(3-fluoro-benzyl)-4-(3-fluoro-phenyl)-1H-irrnclazol-2-yl]-2,2-dimethyl-propyl}-
amide;
1-((S)-3-Arnino-2-fluoro-propyl)-1-{(R)-1-[1-(3-fluoro-benzyl)-4-(3-fluoro-
phenyl)-1 H-imidazol-2-yl] -2,2-dimethyl-propyl} -urea;
1-(3-Amino-propyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-
2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
1 -(3 -Amino-propyl)-1- {(R)-1 - [ 1 -benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-
2-yl]-2,2-dimethyl-propyl} -3,3 -dimethyl-urea;
1 -(3-amino-propyl)-1 - {(R)-1 - [ 1 -benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-
2-yl]-2,2-dimethyl-propyl}-3-(3,5-dimethyl-isoxazol-4-yl)-urea;
Pyrrolidine-1-carboxylic acid (3-amino-propyl)-{(R)-1-[1-benzyl-4-(2,5-
diiluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Isoxazolidine-2-carboxylic acid (3-amino-propyl)-{(R)-1-[1-benzyl-4-(2,5-
difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-Methanesulfonyl-piperazine-1 -carboxylic acid (3-amino-propyl)-{(R)-1-[ 1 -
benzyl-4-(2,5-difluoro-phenyl)-1H-irrndazol-2-yl]-2,2-dimethyl-propyl}-arnide;
4-Methanesulfonyl-piperidine- 1-carboxylic acid (3-amino-propyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid (3-amino-propyl)-{(R)-1-[1-benzyl-4-(2,5-
difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((S)-3-arnino-4-metlioxy-butyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
Morpholine-4-carboxylic acid ((S)-3 -amino-4-fluoro-butyl)- {(R)-1 -[ 1 -benzyl-
4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;

1,1-Dioxo-1-thiomorplioline-4-carboxylic acid (3-amino-propyl)-{(R)-1-[ 1 -
benzyl-4-(2,5-difluoro-phenyl)4H-iirudazol-2-yl]-2,2-dimethyl-propyl}-amide;
Pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-(S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-33-dimethyl-urea;
4-Methanesulfonyl-piperazine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)-1-[ 1 -benzyl-4-(2,5-difluoro-phenyl)-1 H-imidazol-2-yl] -2,2-dimethyl-propyl} -amide;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-(3,5-di-methyl-isoxazol-4-yl)-urea;
1,1 -Dioxo-1-thiomorpholine-4-carboxylic acid ((S)-3 -amino-2-fluoro-propyl)-
{(R)-1-[ 1 -benzyl-4-(2J5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
Isoxazolidine-2-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Thiomorpholine-4-carboxylic acid (3-amino-propyl)-{(R)-1-[1-benzyl-4-(2,5-
difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
Isoxazolidine-2-carboxylic acid ((S)-3-amino-2-fluoro-propyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
Pyrrolidine-1-carboxylic acid ((S)-3-amino-2-fluoro-propyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((S)-3-amino-2-fluoro-propyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl] -2,2-dimethyl-propyl} -amide;
1,1-Dioxo-1-thiomorpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
l-Oxo-1-thiomorpholine-4-carboxylic acid (3-amino-propyl)-{(R)-1-[1-
benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid((S)-3-amino-butyl)-{(R)-1-[1-benzyl-4-(2,5-
difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -amide;
4-Methanesulfonyl-piperazine-1-carboxylic acid ((S)-3-amino-2-fluoro-
propyl)-{(R)-1-[1-bethyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
Morpholine-4-carboxylic acid((R)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(2,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(2,3,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
Morpholine-4-carboxylic acid ((R)-3-amino-butyl)-{(R)-1-[1-benzyl-4-(2,5-
difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;

(2R,6S)-2,6-Dimethyl-morphoIine-4-carboxylic acid ((S)-3-amino-4-fluoro-
butyl)-{(R)-1-[1-ber^l-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
4-Isopropyl-piperazine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)- {(R)-1-
[1-benzyl-(2,5-difIuoro-phenyl)-IH-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-isopropyl-urea;
1 -((S)-3 -Amino-4-fluoro-butyl)-1- {(R)-1-[ 1 -benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl} -3 -cyclopropylmethyl-urea;
1 -((S)-3 -Amino-4-fluoro-butyl)-1- {(R)-1-[ 1 -benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-(tetrahydro-pyran-4-yl)-urea;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(S)-1-[1-benzyl-
4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-(3-((S)-3-Amino-4-fluoro-butyl)-3-{(R)-1-[1-benzyl-4-(2J5-difluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-ureido)-piperidine-1-carboxylic acid ethyl
ester;
(2S,6R)-2,6-Dimethyl-morpholine-4-carboxylic acid ((R)-3-amino-4-fluoro-
butyl)-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)4H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
(S)-3-Hydroxy-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)4-[1-benzyl-4-(2,5-difluoro-phenyl)4H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
(R)-3-Hydroxy-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)4H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-((S)-3-Amino-4-fluoro-butyl)-l.-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-cyclopropyl-urea;
(S)-2-Hydrazinocarbonyl-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-
butyl)-{(R)-1-[1-benzyl-4-(2,5-dfluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
(S)-1-((3-Amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-irnidazol-2-yl]-2,2-dimethyl-propyl}-carbamoyl)-pyrrolidine-2-carboxylic acid methyl
ester;
1 -((R)-3 -Amino-4-fluoro-butyl)-1- {(R)-1-[ 1 -benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
l-((S)-3-Amino-4-methoxy-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-
phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-(3-
hydroxy-benzyl)-4-(2,3,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
(2S,6R)-2,6-Dimethyl-morpholine-4-carboxylic acid((S)-3-amino-4-fIuoro-
butyl)- {(R)-1-[1-(3-hydroxy-benzyl)-4-(2,3,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-
dimethyl-propyl}-amide;

(2R,6S)-2,6-Dimethyl-morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-
butyl)-{(R)-1-[1-benzy-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
4-Isopropyl-piperazine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)- {(R)-1
[1-benzyM-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-isopropyl-urea;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-cycIopropylmethyl-urea;
l-(S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-(tetrahydro-pyran-4-yl)-urea;
Morpholine-4-carboxylic acid((S)-3-amino-4-fluoro-butyl)-{(S)-1-[1-benzyl-
4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
4-(3-((S)-3-Amino-4-fluoro-butyl)-3-{(R)-1-[1-benzyl-4-(2,5-difluoro-
phenyl)-1H-imidazol-2-yl]-2;2-dimethyl-propyl}-ureido)-piperidine-1-carboxylic acid ethyl
ester;
(2S,6R)-2,6-Dimethyl-morpholine-4-carboxylic acid ((R)-3-amino-4-fluoro-
butyl)- {(R)-1-[1 -benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl} -
amide;
(S)-3-Hydroxy-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)4H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
(R)-3-Hydroxy-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-
{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
1-((S)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-cyclopropyl-urea;
(S)-2-Hydrazmocarbonyl-pyrrolidine-1-carboxylic acid ((S)-3-amino-4-fluoro-
butyl)-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-
amide;
(S)-1-((3-Amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-irnidazol-2-yl]-2,2-dimethyl-propyl}-carbamoyl)-pyrrolidine-2-carboxylic acid methyl
ester;
1-((R)-3-Amino-4-fluoro-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-phenyl)-
1H-imidazol-2-yl]-2,2-dimethyl-propyl}-3-methyl-urea;
1-((S)-3-Arnino-4-methoxy-butyl)-1-{(R)-1-[1-benzyl-4-(2,5-difluoro-
pheny)-1H-imidazol-2-yl]-dimethyl-propyl}-3-methyl-urea;
Morpholine-4-carboxylic acid ((S)-3-amino-4-fluoro-butyl)-{(R)-1-[1-(3-
hydroxy-benzyl)-4-(2,3,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
(2S,6R)-2,6-Dimethyl-moroholine-4-carboxylic acid ((S)-3-amino-4-fluoro-
buryl)-{(R)-1-[1-(3-hydroxy-benzyl)-4-(2,3,5-trifluoro-phenyl)-1H-imidazol-2-yl]-2,2-
dimethyl-propyl}-amide;

Pyrrolidine-1-carboxylic acid ((R)-3-amino-4-fluoro-butyl)-{(R)-1-[1-benzyl-
4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-propyl}-amide;
N-(3S)-3-amino-4-fluorobutyl]-N-{(1R)-1-[1-benzyl-4-(2,5-difluorophenyl)-
1H-imidazol-2-yl]-2,2-diraethylpropyl}-N'-hydroxyurea;
N-[(3R)-3-amino-4-fluorobutyl]-N-{(1R)-1-[1-benzyl-4-(2,5--difluorophenyl)-
1H-imidazol-2-yl]-2,2-dimethylpropyl}-N'-hydroxyurea;
l-{(R)4-[1-Benzyl4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-dimethyl-
propyl}-1-[(S)-3-(13-dioxo-1,3-dihydro-isoindol-2-yl)-4-fluoro-butyl]-3-methyl-urea; and-
N-[(S)-3-(1-{(R)-1-[1-Benzyl-4-(2,5-difluoro-phenyl)-1H-imidazol-2-yl]-2,2-
dimethyl-propyl}-3-methyl-ureido)-1-fluoromethyl-propyl]-N'-methyl-phthalamide.





(54) Title:- SUBSTITUTED IMIDAZOLE COMPOUNDS AS KSP INHIBITORS


(57) Abstract: The present invention relates to new substituted imidazole compounds and pharmaceutically acceptable salts,esters or
prodrugs thereof, compositions of the derivatives together with pharmaceutically acceptable carriers, and uses of the compounds.The
compounds of the invention have the following general formula (I).

Documents:

00119-kolnp-2008-abstract.pdf

00119-kolnp-2008-claims.pdf

00119-kolnp-2008-correspondence others.pdf

00119-kolnp-2008-description complete.pdf

00119-kolnp-2008-form 1.pdf

00119-kolnp-2008-form 3.pdf

00119-kolnp-2008-form 5.pdf

00119-kolnp-2008-gpa.pdf

00119-kolnp-2008-international publication.pdf

00119-kolnp-2008-international search report.pdf

00119-kolnp-2008-pct priority document notification.pdf

00119-kolnp-2008-pct request form.pdf

0119-KOLNP-2008-ASSIGNMENT.pdf

0119-KOLNP-2008-CORRESPONDENCE OTHERS 1.1.pdf

119-KOLNP-2008-(03-12-2012)-AMANDED CLAIMS.pdf

119-KOLNP-2008-(03-12-2012)-CORRESPONDENCE.pdf

119-KOLNP-2008-(03-12-2012)-FORM 13.pdf

119-KOLNP-2008-(03-12-2012)-FORM-2.pdf

119-KOLNP-2008-(03-12-2012)-OTHERS -1.pdf

119-KOLNP-2008-(03-12-2012)-OTHERS.pdf

119-KOLNP-2008-(03-12-2012)-REPLY TO EXAMINATION REPORT.pdf

119-KOLNP-2008-(08-01-2013)-CORRESPONDENCE.pdf

119-KOLNP-2008-(08-01-2013)-FORM 3.pdf

119-KOLNP-2008-(12-07-2012)-EXAMINATION REPORT.pdf

119-KOLNP-2008-ASSIGNMENT.pdf

119-KOLNP-2008-CORRESPONDENCE OTHERS 1.2.pdf

119-KOLNP-2008-CORRESPONDENCE.pdf

119-KOLNP-2008-FORM 18.pdf

119-KOLNP-2008-FORM 3-1.1.pdf

119-KOLNP-2008-GPA.pdf

119-KOLNP-2008-GRANTED-ABSTRACT.pdf

119-KOLNP-2008-GRANTED-CLAIMS.pdf

119-KOLNP-2008-GRANTED-DESCRIPTION (COMPLETE).pdf

119-KOLNP-2008-GRANTED-FORM 1.pdf

119-KOLNP-2008-GRANTED-FORM 2.pdf

119-KOLNP-2008-GRANTED-FORM 3.pdf

119-KOLNP-2008-GRANTED-FORM 5.pdf

119-KOLNP-2008-GRANTED-SPECIFICATION-COMPLETE.pdf

119-KOLNP-2008-TRANSLATED COPY OF PRIORITY DOCUMENT.pdf

abstract-00119-kolnp-2008.jpg


Patent Number 255840
Indian Patent Application Number 119/KOLNP/2008
PG Journal Number 13/2013
Publication Date 29-Mar-2013
Grant Date 26-Mar-2013
Date of Filing 08-Jan-2008
Name of Patentee NOVARTIS AG
Applicant Address LICHSTRASSE 35 CH-4056 BASEL
Inventors:
# Inventor's Name Inventor's Address
1 BARSANTI PAUL A C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
2 WANG WEIBO C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
3 MENDENHALL KRIS G C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
4 LAGNITON LIANA M C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
5 RAMURTHY SAVITHRI C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
6 PHILLIPS MEGAN C C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
7 SUBRAMANIAN SHARADHA C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
8 BOYCE RUSTUM C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
9 BRAMMEIER NATHAN M C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
10 CONSTANTINE RYAN C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
11 DUHL DAVID C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
12 WALTER ANNETTE O C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
13 ABRAMS TINYA J C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
14 RENHOWE PAUL A C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
15 XIA YI C/O NOVARTIS VACCINES AND DIAGNOSTICS, INC P.O. BOX 8097, EMERYVILLE, CA 94662-8097
PCT International Classification Number C07D 401/14
PCT International Application Number PCT/US2006/031129
PCT International Filing date 2006-08-09
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/706901 2005-08-09 U.S.A.