Title of Invention

IMIDAZOYL BENZAMIDE ANTI CANCER AGENTS

Abstract Compounds useful for treating cellular proliferative diseases and disorders by modulating the activity of one or more mitotic kinesins are disclosed.
Full Text [0001] This application claims the benefit of U.S. Patent Application No. 60/569,510,
filed May 6,2004, which is hereby incorporated by reference.
[0002] This invention relates to chemical entities which are inhibitors of one or more
mitotic kinesins and are useful in the treatment of cellular proliferative diseases, for example
cancer, hyperplasias, restenosis, cardiac hypertrophy, immune disorders, fungal disorders, and
inflammation.
[0003] Among the therapeutic agents used to treat cancer are the taxanes and vinca
alkaloids, which act on microtubules. Microtubules are the primary structural element of the
mitotic spindle. The mitotic spindle is responsible for distribution of replicate copies of the
genome to each of the two daughter cells that result from cell division. It is presumed that
disruption of the mitotic spindle by these drugs results in inhibition of cancer cell division,
and induction of cancer cell death. However, microtubules form other types of cellular
structures, including tracks for intracellular transport in nerve processes. Because these
agents do not specifically target mitotic spindles, they have side effects that limit their
usefulness.
[0004] Improvements in the specificity of agents used to treat cancer is of
considerable interest because of the therapeutic benefits which would be realized if the side
effects associated with the administration of these agents could be reduced. Traditionally,
dramatic improvements in the treatment of cancer are associated with identification of
therapeutic agents acting through novel mechanisms. Examples of this include not only the
taxanes, but also the camptothecin class of tppoisomerase I inhibitors. From both of these
perspectives, mitotic kinesins are attractive targets for new anti-cancer agents.
[0005] Mitotic kinesins are enzymes essential for assembly and function of the mitotic
spindle, but are not generally part of other microtubule structures, such as in nerve processes.
Mitotic kinesins play essential roles during all phases of mitosis. These enzymes are
"molecular motors" that transform energy released by hydrolysis of ATP into mechanical
force which drives the directional movement of cellular cargoes along microtubules. The
catalytic domain sufficient for this task is a compact structure of approximately 340 amino
acids. During mitosis, kinesins organize microtubules into the bipolar structure that is the
mitotic spindle. Kinesins mediate movement of chromosomes along spindle microtubules, as
well as structural changes in the mitotic spindle associated with specific phases of mitosis.

L. perimental perturbation of mitotic kinesin function causes malformation or dysfunction of
the mitotic spindle, frequently resulting in cell cycle arrest and cell death.
[0006] In one aspect, the invention relates to methods for treating cellular
proliferative diseases, and for treating disorders by inhibiting the activity of one or more
mitotic kinesins.
[0007] Provided is at least one chemical entity chosen from compounds of Formula I

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein
R1 is optionally substituted aryl, optionally substituted heterocycloalkyl, or optionally
substituted heteroaryl;
X is-K*I or-SO2-;
R2 is hydrogen or optionally substituted lower alkyl;
W is -CR4-, -CH2CR4-, or N;
R3 is-K*I-R7, hydrogen, optionally substituted alkyl, optionally substituted heterocyclyl,
cyano, optionally substituted sulfonyl, or optionally substituted aryl;
R4 is hydrogen or optionally substituted alkyl;
R5 is hydrogen, hydroxyl, optionally substituted amino, optionally substituted heterocyclyl; or
optionally substituted lower alkyl;
R6 is hydrogen, optionally substituted alkyl, optionally substituted alkoxy, optionally
substituted aryloxy, optionally substituted heteraryloxy, optionally substituted
alkoxycarbonyl-, optionally substituted aminocarbonyl-, optionally substituted aryl,
optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally
substituted aralkyl; and

k7 is optionally substituted lower alkyl, optionally substituted aryl, hydroxyl, optionally
substituted amino, optionally substituted aralkoxy, or optionally substituted alkoxy.
[0008] In some embodiments, if W is N, then R5 is not hydroxyl or optionally
substituted amino, and R6 is not optionally substituted alkoxy, optionally substituted aralkoxy,
optionally substituted heteroaralkoxy, or optionally substituted amino.
[0009] Also provided is at least one chemical entity chosen from compounds of
Formula II

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R5, R6, and W are as described for compounds of
Formula I and wherein
R11 is optionally substituted heterocyclyl, optionally substituted lower alkyl, nitro,
cyano, hydrogen, sulfonyl, or halo;
R12 is hydrogen, halo, optionally substituted alkyl, optionally substituted amino,
optionally substituted sulfanyl, optionally substituted alkoxy, optionally substituted aryloxy,
optionally substituted heterocyclyl, or optionally substituted heteroaryloxy; and
R13 is hydrogen, acyl, optionally substituted alkyl-, optionally substituted alkoxy, halo,
hydroxyl, nitro, cyano, optionally substituted amino, alkylsulfonyl-, alkylsulfonamido-,
alkylsulfonyl-, carboxyalkyl-, aminocarbonyl-, optionally substituted aryl or optionally
substituted heteroaryl-.
[0010] Also provided is at least one chemical entity chosen from compounds of
Formula III


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R6, R11, R12, and R13 are as described for compounds of
Formula II.
[0011] Also provided is at least one chemical entity chosen from compounds of
Formula IV

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 are as described for compounds of
Formula III.
[0012] Also provided is at least one chemical entity chosen from compounds of
Formula.


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R11, R12, and R13 are as described for compounds of
Formula III and wherein
R14 is optionally substituted heteroaryl; and
R15 is chosen from hydrogen, halo, hydroxyl, and lower alkyl.
[0013] Also provided is at least one chemical entity chosen from compounds of
Formula VI

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 are as described for compounds of
Formula III.
[0014] Also provided is at least one chemical entity chosen from compounds of
Formula VII


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 are as described for compounds of
Formula III and wherein
R9 is chosen from optionally substituted alkoxy, optionally substituted cycloalkoxy,
optionally substituted arylalkoxy, optionally substituted amino and optionally
substituted lower alkyl.
[0015] Also provided is composition comprising a pharmaceutical excipient and at
least one chemical entity described herein.
[0016] Also provided is a method of modulating CENP-E kinesin activity which
comprises contacting said kinesin with an effective amount of at least one chemical entity
described herein.
[0017] Also provided is a method of inhibiting CENP-E which comprises contacting
said kinesin with an effective amount of at least one chemical entity described herein.
[0018] Also provided is a method for the treatment of a cellular proliferative disease
comprising administering to a subject in need thereof at least one chemical entity described
herein.
[0019] Also provided is a method for the treatment of a cellular proliferative disease
comprising administering to a subject in need thereof a composition comprising a
pharmaceutical excipient and at least one chemical entity described herein.
[0020] Also provided is the use, in the manufacture of a medicament for treating
cellular proliferative disease, of at least one chemical entity of described herein.
[0021] Also provided is the use of at least one chemical entity described herein for the
manufacture of a medicament for treating a disorder associated with CENP-E kinesin activity.
[0022] As used in the present specification, the following words and phrases are

generally intended to have the meanings as set forth below, except to the extent that the
context in which they are used indicates otherwise. The following abbreviations and terms
have the indicated meanings throughout:
[0023] As used herein, when any variable occurs more than one time in a chemical
formula, its definition on each occurrence is independent of its definition at every other
occurrence.
[0024] The following abbreviations and terms have the indicated^neanings
throughout:
Ac = acetyl
Boc = t-butyloxy carbonyl
Bu = butyl
K*i- = cyclo
CBZ = carbobenzoxy = benzyloxycarbonyl
DCM = dichloromethane = methylene chloride = CH2Cl2
DCE = dichloroethane
DEAD = diethyl azodicarboxylate
DIG = diisopropylcarbodiimide
DIEA = N,N-diisopropylethylamine
DMAP = 4-N,N-dimethylaminop3'ridine
DMF = N,N-dimethylfonnamide
DMSO = dimethyl sulfoxide
Et = ethyl
Fmoc = 9-fluorenylmethoxycarbonyl
GC = gas chromatography
HATU = 0-(7-Azabenzotriazo1-1-yl)-l,l,3,3-tetramethyluronium
hexafluorophosphate
HOAc = acetic acid
HOBt - hydroxybenzotriazole
LAH = lithium aluminum hydride
Me = methyl
mesyl = methanesulfonyl
NCS = N-chlorosuccinimide
Ph = phenyl

Py = pyridine
rt = room temperature
sat'd = saturated
s- = secondary
t- = tertiary
TES = triethylsilyl
TFA = trifluoroacetic acid
THF = tetrahydrofuran
TMS = trimethylsilyl
tosyl = p-toluenesulfonyl
[0025] A dash ("-") that is not between two letters or symbols is used to indicate a
point of attachment for a substituent. For example, -CONH2 is attached through the carbon
atom.
[0026] By "optional" or "optionally" is meant that the subsequently described event or
circumstance may or may not occur, and that the description includes instances where the
event or circumstance occurs and instances in which it does not. For example, "optionally
substituted alkyl" encompasses both "alkyl" and "substituted alkyl" as defined below. It will
be understood by those skilled in the art, with respect to any group containing one or more
substiruents, that such groups are not intended to introduce any substitution or substitution
patterns that are sterically impractical, synthetically non-feasible and/or inherently unstable.
[0027] "Alkyl" encompasses straight chain and branched chain having the indicated
number of carbon atoms, usually from 1 to 20 carbon atoms, for example 1 to 8 carbon atoms,
such as 1 to 6 carbon atoms. For example C1-C6alkyl encompasses both straight and
branched chain alkyl of from 1 to 6 carbon atoms. Examples of alkyl groups include methyl,
ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl,
hexyl, 2-hexyl, 3-hexyl, 3-methylpentyl, and the like. Alkylene is another subset of alkyl,
referring to the same residues as alkyl, but having two points of attachment. Alkylene groups
will usually have from 2 to 20 carbon atoms, for example 2 to 8 carbon atoms, such as from 2
to 6 carbon atoms. For example, C0 alkylene indicates a covalent bond and C1 alkylene is a
methylene group. When an alkyl residue having a specific number of carbons is named, all
geometric isomers having that number of carbons are intended to be encompassed; thus, for
example, "butyl" is meant to include n-butyl, sec-butyl, isobutyl and t-butyl; "propyl" includes
n-propyl and isopropyl. "Lower alkyl" refers to alkyl groups having one to four carbons.

[0028] "Cycloalkyl" indicates a saturated hydrocarbon ring group, having the
specified number of carbon atoms, usually from 3 to 7 ring carbon atoms. Examples of
cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl as well as
bridged and caged saturated ring groups such as norbornane.
[0029] By "alkoxy" is meant an alkyl group of the indicated number of carbon atoms
attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy,
isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyloxy, isopentoxyj neopentoxy,
hexoxy, 2-hexoxy, 3-hexoxy, 3-methylpentoxy, and the like. Alkoxy groups will usually
have from 1 to 6 carbon atoms attached through the oxygen bridge. "Lower alkoxy" refers to
alkoxy groups having one to four carbons.
[0030] "Acyl" refers to the groups (alkyl)-K*I(O)-; (cycloalkyl)-K*I(O)-; (aryl)-K*I(O)-;
(heteroaryl)-K*I(O)-; and (heterocycloalkyl)-K*I(O)-, wherein the group is attached to the parent
structure through the carbonyl functionality and wherein alkyl, cycloalkyl, aryl, heteroaryl,
and heteroeycloalkyl are as described herein. Acyl groups have the indicated number of
carbon atoms, with the carbon of the keto group being included in the numbered carbon
atoms. For example a C2 acyl group is an acetyl group having the formula CH3(K*I=O)-.
[0031] By "alkoxycarbonyl" is meant an ester group of the formula (alkoxy)(K*I=O)-
attached through the carbonyl carbon wherein the alkoxy group has the indicated number of
carbon atoms. Thus a C1-C6alkoxycarbonyl group is an alkoxy group having from 1 to 6
carbon atoms attached through its oxygen to a carbonyl linker.
[0032] By "amino" is meant the group -NH2.
[0033] The term "aminocarbonyl" refers to the group -CONRbRc, where
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl; or
Rb and Rc taken together with the nitrogen to which they are bound, form an optionally
substituted 5- to 7-membered nitrogen-containing heteroeycloalkyl which optionally includes
1 or 2 additional heteroatoms selected from O, N, and S in the heteroeycloalkyl ring;
where each substituted group is independently substituted with one or more substituents
independently selected from C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-,
heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-, -OC1-C4 alkyl, -OC1-C4 alkylphenyl, -
C1-C4 alkyl-OH, -OC1-C4 haloalkyl,. halo, -OH, -NH2, -C1-C4 alkyl-NH2,
-N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl), -N(C1-C4 alkyl)(C1-C4 alkylphenyl),

-NH(C1-C4 alkylphenyl), cyano, nitro, oxo (as a substituted for heteroaryl), -CO2H,
-K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl)(C1-C4 alkyl), -CONH(C1-C4 alkyl), -CONH2,
-NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl), -N(C1-C4 alkyl)K*I(O)(C1-C4 alkyl),
-N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl, -K*I(O)C1-C4 phenyl,
-K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -SO2(phenyl), -
SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -SO2NH(phenyl), -
NHSO2(C1-C4 alkyl),-NHSO2(phenyl), and-NHSO2(C1-C4 haloalkyl).
[0034] "Aryl" encompasses:
5- and 6-membered carbocyclic aromatic rings, for example, benzene;
bicyclic ring systems wherein at least one ring is carbocyclic and aromatic, for
example, naphthalene, indane, and tetralin; and
tricyclic ring systems wherein at least one ring is carbocyclic and aromatic, for
example, fluorene.
For example, aryl includes 5- and 6-membered carbocyclic aromatic rings fused to a 5- to 7-
membered heterocycloalkyl ring containing 1 or more heteroatoms chosen from N, O, and S.
For such fused, bicyclic ring systems wherein only one of the rings is a carbocyclic aromatic
ring, the point of attachment may be at the carbocyclic aromatic ring or the heterocycloalkyl
ring. Bivalent radicals formed from substituted benzene derivatives and having the free
valences at ring atoms are named as substituted phenylene radicals. Bivalent radicals derived
from univalent polycyclic hydrocarbon radicals whose names end in "-yl" by removal of one
hydrogen atom from the carbon atom with the free valence are named by adding "-idene" to
the name of the corresponding univalent radical, e.K*i., a naphthyl group with two points of
attachment is termed naphthylidene. Aryl, however, does not encompass or overlap in any
way with heteroaryl, separately defined below. Hence, if one or more carbocyclic aromatic
rings is fused with a heterocycloalkyl aromatic ring, the resulting ring system is heteroaryl,
not aryl, as defined herein.
[0035] The term "aryloxy" refers to the group -O-aryl.
[0036] The term "aralkyl" refers to a residue in which an aryl moiety is attached to the
parent structure via an alkyl residue. Examples include benzyl-, phenethyl-, phenylvinyl-,
phenylallyl and the like.
[0037] The term "heteroaralkyl" refers to a residue in which a heteroaryl moiety is
attached to the parent structure via an alkyl residue. Examples include furanylmethyl-,
pyridinylmethyl-, pyrimidinyleth} 1 and the like.

[0038] The term "halo" includes fluoro, chloro, bromo, and iodo, and the term
"halogen" includes fluorine, chlorine, bromine, and iodine.
[0039] "Haloalkyl" indicates alkyl as defined above having the specified number of
carbon atoms, substituted with 1 or more halogen atoms, up to the maximum allowable
number of halogen atoms. Examples of haloalkyl include, but are not limited to,
trifluoromethyl, difluoromethyl, 2-fluoroethyl, and penta-fluoroethyl.
[0040] "Heteroaryl",encompasses:
5- to 7-membered aromatic, monocyclic rings containing one or more, for example,
from 1 to 4, or in certain embodiments, from 1 to 3, heteroatoms chosen from
N, O, and S, with the remaining ring atoms being carbon; and
bicyclic heterocycloalkyl rings containing one or more, for example, from 1 to 4, or in
certain embodiments, from 1 to 3, heteroatoms chosen from N, O, and S, with
the remaining ring atoms being carbon and whereinat least one heteroatom is
present in an aromatic ring.
For example, heteroaryl includes a 5- to 7-membered heterocycloalkyl, aromatic ring fused to
a 5- to 7-membered cycloalkyl ring. For such fused, bicyclic heteroaryl ring systems wherein
only one of the rings contains one or more heteroatoms, the point of attachment may be at the
heteroaromatic ring or the cycloalkyl ring. When the total number of S and O atoms in the
heteroaryl group exceeds 1, those heteroatoms are not adjacent to one another. In certain
embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2.
In certain embodiments, the total number of S and O atoms in the aromatic heterocycle is not
more than 1. Examples of heteroaryl groups include, but are not limited to, (as numbered
from the linkage position assigned priority 1), 2-pyridyl, 3-pyridyl, 4-pyridyl, 2,3-pyrazinyl,
3,4-pyrazinyl, 2,4-pyrimidinyl, 3,5-pyrimidinyl, 2,3-pyrazolinyl, 2,4-imidazolinyl,
isoxazolinyl, oxazolinyl, thiazolinyl, thiadiazolinyl, tetrazolyl, thienyl, benzothiophenyl,
furanyl, benzofuranyl, benzoimidazolinyl, indolinyl, pyridizinyl, triazolyl, quinolinyl,
pyrazolyl, imidazopyridinyl, and 5,6,7,8-tetrahydroisoquinoline. Bivalent radicals derived
from univalent heteroaryl radicals whose names end in "-yl" by removal of one hydrogen
atom from the atom with the free valence are named by adding "-idene" to the name of the
corresponding univalent radical, e.K*i., a pyridyl group with two points of attachment is a
pyridylidene. Heteroaryl does not encompass or overlap with aryl as defined above.
[0041] In the term "heteroaralkyl," heteroaryl and alkyl are as defined herein, and the
point of attachment is on the alkyl group. This term encompasses, but is not limited to,

pyridylmethyl, thiophenylmethyl, and (pyrrolyl)1-ethyl.
[0042] A "leaving group" or "atom" is any group or atom that will, under the reaction
conditions, cleave from the starting material, thus promoting reaction at a specified site.
Suitable examples of such groups unless otherwise specified are halogen atoms, mesyloxy, p-
nitrobenzensulphonyloxy and tosyloxy groups.
[0043] "Optional" or "optionally" means that the subsequently described event or
circumstance may or may not occur, and that the description includes instances where said
event or circumstances occurs and instances in which it does not. For example, "optionally
substituted alkyl" includes "alkyl" and "substituted alkyl" as defined herein. It will be
understood by those skilled in the art with respect to any group containing one or more
substituents that such groups are not intended to introduce any substitution or substitution
patterns that are sterically impractical and/or synthetically non-feasible and/or inherently
unstable.
[0044] "Protecting group" has the meaning conventionally associated with it in
organic synthesis, i.e. a group that selectively blocks one or more reactive sites in a
multifunctional compound such that a chemical reaction can be carried out selectively on
another unprotected reactive site and such that the group can readily be removed after the
selective reaction is complete. A variety of protecting groups are disclosed, for example, in
T.H. Greene and P. K*I. M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John
Wiley & Sons, New York (1999), which is incorporated herein by reference in its entirety.
For example, a hydroxyl protected form is where at least one of the hydroxyl groups present
in a compound is protected with a hydroxyl protecting group. Likewise, amines and other
reactive groups may similarly be protected.
[0045] By "heterocycloalkyl" is meant a single aliphatic ring, usually with 3 to 7 ring
atoms, containing at least 2 carbon atoms in addition to 1-3 heteroatoms independently
selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of
the foregoing heteroatoms. Suitable heterocycloalkyl groups include, for example (as
numbered from the linkage position assigned priority 1), 2-pyrrolinyl, 2,4-imidazolidinyl, 2,3-
pyrazolidinyl, 2-piperidyl, 3-piperidyl, 4-piperdyl, and 2,5-piperzinyl. Morpholinyl groups
are also contemplated, including 2-morpholinyl and 3-morpholinyl (numbered wherein the
oxygen is assigned priority 1).
[0046] As used herein, "modulation" refers to a change in CENP-E activity as a direct
or indirect response to the presence at least one chemical entity described herein, relative to

the activity of CENP-E in the absence of the chemical entity. The change may be an increase
in activity or a decrease in activity, and may be due to the direct interaction of the chemical
entity with CENP-E, or due to the interaction of the compound with one or more other factors
that in turn affect CENP-E activity.
[0047] The term "sulfanyl" includes the groups: -S-( optionally substituted (C1-
C6)alkyl), -S-(optionalIy substituted aryl), -S-(optionally substituted heteroaryl), and
-S-(optionally substituted heterocycloalkyi). Hence, sulfanyl includes the group C1-C6
alkylsulfanyl.
{0048] The term "sulfinyl" includes the groups: -S(O)-H, -S(O)-( optionally
substituted (C1-C6)alkyl), -S(O)-optionally substituted aryl), -S(O)-optionally substituted
heteroaryl), -S(O)-(optionally substituted heterocycloalkyi); and -S(O)-(optionally substituted
amino).
[0049] The term "sulfonyl" includes the groups: -S(O2>H,.-S(O2)-( optionally
substituted (C1-C6)alkyl), -S(O2)-optionally substituted aryl), -S(O2)~optionally substituted
heteroaryl), -S(O2)-(optionally substituted heterocycloalkyi) ,-S(O2)-(optionally substituted
alkoxy), -S(O2)-optionally substituted aryloxy), -S(O2)-optionally substituted heteroaryloxy),
-S(O2)-(optionally substituted heterocyclyloxy); and -S(O2)-(optionally substituted amino).
[0050] The term "substituted", as used herein, means that any one or more hydrogens
on the designated atom or group is replaced with a selection from the indicated group,
provided that the designated atom's normal valence is not exceeded. When a substituent is
oxo (i.e., =O) then 2 hydrogens on the atom are replaced. Combinations of substituents
and/or variables are permissible only if such combinations result in stable compounds or
useful synthetic intermediates. A stable compound or stable structure is meant to imply a
compound that'is sufficiently robust to survive isolation from a reaction mixture, and
subsequent formulation as an agent having at least practical utility. Unless otherwise
specified, substituents are named into the core structure. For example, it is to be understood
that when (cycloalkyl)alkyl is listed as a possible substituent, the point of attachment of this
substituent to the core structure is in the alkyl portion.
[0051] The terms "substituted" alkyl, cycloalkyl, aryl, heterocycloalkyi, and
heteroaryl, unless otherwise expressly defined, refer respectively to alkyl, cycloalkyl, aryl,
heterocycloalkyi, and heteroaryl wherein one or more (such as up to 5, for example, up to 3)
hydrogen atoms are replaced by a substituent independently chosen from:
-Ra, -ORb, -O(C1-C2 alkyl)0- (e.K*i., methylenedioxy-), -SRb, guanidine, guanidine

herein one or more of the guanidine hydrogens are replaced with a lower-alkyl group,
-NRbRc, halo, cyano, nitro, -CORb, -CO2Rb, -CONRbRc, -OCORb, -OCO2Ra, -OCONRbRc,
-NR°CORb, -NRcCO2Ra, -NRcCONRbRc, -CO2Rb, -CONRbRc, -NRcCORb, -SORa, -SO2Ra,
-SO2NRbRc, and-NRcSO2Ra,
where Ra is chosen from optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl;
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl;
where each optionally substituted group is unsubstituted or independently substituted
with one or more, such as one, two, or three, substituents independently selected from
C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-, heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-,
-OC1-C4 alkyl, -OC1-C4 alkylphenyl, -C1-C4 alkyl-K*I, -OC1-C4 haloalkyl, halo, -OH, -NH2,
-C1-C4 alkyl-NH2, -N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl),
-N(C1-C4 alkyl)(C1-C4 alkylphenyl), -NH(C1-C4 alkylphenyl), cyano, nitro, oxo (as a
substituted for heteroaiyl), -CO2H, -K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl)(C1-C4 alkyl),
-CONH(C1-C4 alkyl), -CONH2, -NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl),
-N(C1-C4 alkyl)K*I(O)(C1-C4 alkyl), -N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl,
-K*I(O)C1-C4 phenyl, -K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -
SO2(phenyl), -SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -SO2NH(phenyl), -
NHSO2(C1-C4 alkyl), -NHSO2(phenyl), and -NHSO2(C1-C4 haloalkyl).
[0052] The term "substituted acyl" refers to the groups (substituted alkyl)-K*I(O)-;
(substituted cycloalkyl)-K*I(O)-; (substituted aryl)-K*I(O)-; (substituted heteroaryl)-K*I(O)-; and
(substituted heterocycloalkyl)-K*I(O)-, wherein the group is attached to the parent structure
through the carbonyl functionality and wherein substituted alkyl, cycloalkyl, aryl, heteroaryl,
and heterocycloalkyl, refer respectively to alkyl, cycloalkyl, aryl, heteroaryl, and
heterocycloalkyl wherein one or more (such as up to 5, for example, up to 3) hydrogen atoms
are replaced by a substituent independently chosen from:
-Ra, -ORb, -O(C1-C2 alkyl)0- (e.K*i., methylenedioxy-), -SRb, guanidine, guanidine
wherein one or more of the guanidine hydrogens are replaced with a lower-alkyl group,
-NR'ftO, halo, cyano, nitro, -CORb, -CO2Rb, -CONRbRc, -OCORb, -OCO2Ra, -OCONRbRc,
-NRcCORb, -NR°CO2Ra, -NRcCONRbRc, -CO2Rb, -CONRbRc, -NRcCORb, -SORa, -SO2Ra,
-SO2NRbRc, and -NRcSO2Ra,

where Ra is chosen from optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl;
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl;
where each optionally substituted group is unsubstituted or independently substituted
with one or more, such as one, two, or three, substituents independently selected from
C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-, heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-,
-OC1-C4 alkyl, -OC1-C4 alkylphenyl, -C1-C4 alkyl-OH, -OC1-C4 haloalkyl, halo, -OH, -NH2,
-C1-C4 alkyl-NH2, -N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl),
-N(C1-C4 alkyl)(C1-C4 alkylphenyl), -NH(C1-C4 alkylphenyl), cyano, nitro, oxo (as a
substitutent for heteroaryl), -CO2H, -K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl)(C1-C4 alkyl),
-CONH(C1-C4 alkyl), -CONH2, -NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl),
-N(C1-C4 alkyl)K*I(O)(C1-C4 alkyl), -N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl,
-K*I(O)C1-C4 phenyl, -K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -
SO2(phenyl), -SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -SO2NH(phenyl), -
NHSO2(C1-C4 alkyl), -NHSO2(phenyl), and -NHSO2(C1-C4 haloalkyl).
[0053] The term "substituted alkoxy" refers to alkoxy wherein the alkyl constituent is
substituted (i.e., -O-(substituted alkyl)) wherein "substituted alkyl" refers to alkyl wherein
one or more (such as up to 5, for example, up to 3) hydrogen atoms are replaced by a
substituent independently chosen from:
-Ra, -ORb, -O(C1-C2 alkyl)0- (e.K*i., methylenedioxy-), -SRb, guanidine, guanidine
wherein one or more of the guanidine hydrogens are replaced with a lower-alkyl group,
-NRbRc, halo, cyano, nitro, -CORb, -CO2Rb, -CONRbRc, -OCORb, -OCO2Ra, -OCONRbRc,
-NRcCGRb, -NRcCO2Ra, -NRcCONRbRc, -CO2Rb, -CONRbRc, -NRcCORb, -SORa, -SO2Ra,
-SO2NRbRc, and -NRcSO2Ra,
where Ra is chosen from o^jtionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroa ryl;
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl;
where each optionally substituted group is unsubstituted or independently substituted
with one or more, such as one, two, or three, substituents independently selected from

C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-, heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-,
-OC1-C4 alkyl, -OC1-C4 alkylphenyl, -C1-C4 alkyl-OH, -OC1-C4 haloalkyl, halo, -OH, -NH2,
-C1-C4 alkyl-NH2, -N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl),
-N(C1-C4 alkyl)(C1-C4 alkylphenyl), -NH(C1-C4 alkylphenyl), cyano, nitro, oxo (as a
substituted for heteroaryl), -CO2H, -K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl(C1-C4 alkyl),
-CONH(C1-C4 alkyl), -CONH2, -NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl),
-N(C1-C4 alkyl(O)(C1-C4 alkyl), -N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl,
-K*I(O)C1-C4 phenyl, -K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -
SO2(phenyl), -SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -SO2NH(phenyl), -
NHSO2(C1-C4 alkyl), -NHSO2(phenyl), and -NHSO2(C1-C4 haloalkyl). In some
embodiments, a substituted alkoxy group is "polyalkoxy" or -O-(optionally substituted
alkylene)-(optionally substituted alkoxy), and includes groups such as -OCH2CH2OGH3, and
residues of glycol ethers such as polyethyleneglycol, and -O(CH2CH2O)XCH3, where x is an
integer of 2-2O, such as 2-10, and for example, 2-5. Another substituted alkoxy group is
hydroxyalkoxy or -OCH2(CH2)yOH, where y is an integer of 1-10, such as 1-4.
[0054] The term "substituted alkoxycarbonyl" refers to the group (substituted alkyl)-
O-K*I(O)- wherein the group is attached to the parent structure through the carbonyl
functionality and wherein substituted refers to alkyl wherein one or more (such as up to 5, for
example, up to 3) hydrogen atoms are replaced by a substituent independently chosen from:
-Ra, -ORb, -O(C1-C2 alkyl)0- (e.K*i., methylenedioxy-), -SRb, guanidine, guanidine
wherein one or more of the guanidine hydrogens are replaced with a lower-alkyl group,
-NRbRc, halo, cyano, nitro, -CORb, -CO2Rb, -CONRbRc, -OCORb, -OCO2Ra, -OCONRbRc,
-NRcCORb, -NRcCO2Ra, -NRcCONRbRc, -CO2Rb, -CONRbRc, -NR°CORb, -SGRa, -SO2R-SO2NRbRc, and-NRcSO2Ra,
where Ra is chosen from optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl;
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl;
where each optionally substituted group is unsubstituted or independently substituted
with one or more, such as one, two, or three, substituents independently selected from
C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-, heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-,
-OC1-C4 alkyl, -OC1-C4 alkylphenyl, -C1-C4 alkyl-OH, -OC1-C4 haloalkyl, halo, -OH, -NH2,

-C1-C4 alkyl-NHa, -N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl),
-N(C1-C4 alkyl)(C1-C4 alkylphenyl), -NH(Cj~C4 alkylphenyl), cyano, nitro, oxo (as a
substituted for heteroaryl), -CO2H, -K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl)(C1-C4 alkyl),
-CONH(C1-C4 alkyl), -CONH2, -NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl),
-N(C1-C4 alkyl)K*I(O)(C1-C4 alkyl), -N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl,
-K*I(O)C1-C4 phenyl, -K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -
SO2(phenyl),-SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -S(52NH(phenyl), -
NHSO2(C1-C4 alkyl),-NHSO2(phenyl), and-NHSO2(C1-C4 haloalkyl).
(O055] The term "substituted amino" refers to the group -NHRd or -NRdRd where
each Rd is independently chosen from: optionally substituted alkyl, optionally substituted
cycloalkyl, optionally substituted acyl, optionally substituted aryl, optionally substituted
heteroaryl, optionally substituted heterocycloalkyl, alkoxycarbonyl, sulfinyl and sulfonyl,
wherein substituted alkyl, cycloalkyl, aryl, heterocycloalkyl, and heteroaryl refer respectively
to alkyl, cycloalkyl, aryl, heterocycloalkyl, and heteroaryl wherein one or more (such as up to
5, for example, up to 3) hydrogen atoms are replaced by a substituent independently chosen
from:
-Ra, -ORb, -O(C1-C2 alkyl)0- (e.K*i., methylenedioxy-), -SRb, guanidine, guanidine
wherein one or more of the guanidine hydrogens are replaced with a lower-alkyl group,
-NRbRc, halo, cyano, nitro, -CORb, -CG2Rb, -CONRbRc, -OCORb, -OCO2Ra, -OCONRbRc,
-NRcCORb, -NRcCO2Ra, -NRcCONRbRc, -CO2Rb, -CONRbRc, -NRcCORb, -SORa, -SO2Ra,
-SO2NRbRc, and-NRcSO2Ra,
where Ra is chosen from optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl;
Rb is chosen from H, optionally substituted C1-C6 alkyl, optionally substituted aryl,
and optionally substituted heteroaryl; and
Rc is chosen from hydrogen and optionally substituted C1-C4 alkyl;
where each optionally substituted group is unsubstituted or independently substituted
with one or more, such as one, two, or three, substituents independently selected from
C1-C4 alkyl, aryl, heteroaryl, aryl-C1-C4 alkyl-, heteroaryl-C1-C4 alkyl-, C1-C4 haloalkyl-,
-OC1-C4 alkyl, -OC1-C4 alkylphenyl, -C1-C4 alkyl-OH, -OC1-C4 haloalkyl, halo, -OH, -NH2,
-C1-C4 alkyl-NH2, -N(C1-C4 alkyl)(C1-C4 alkyl), -NH(C1-C4 alkyl),
-N(C1-C4 alkyl)(C1-C4 alkylphenyl), -NH(C1-C4 alkylphenyl), cyano, nitro, oxo (as a
substituted for heteroaryl), -CO2H, -K*I(O)OC1-C4 alkyl, -CON(C1-C4 alkyl)(C1-C4 alkyl),

-CONH(C1-C4 alky!), -CONH2, -NHC(O)(C1-C4 alkyl), -NHC(O)(phenyl),
-N(C1-C4 alkyl)K*I(O)(C1-C4 alkyl), -N(C1-C4 alkyl)K*I(O)(phenyl), -K*I(O)C1-C4 alkyl,
-K*I(O)C1-C4 phenyl, -K*I(O)C1-C4 haloalkyl, -OC(O)C1-C4 alkyl, -SO2(C1-C4 alkyl), -
SO2(phenyl), -SO2(C1-C4 haloalkyl), -SO2NH2, -SO2NH(C1-C4 alkyl), -SO2NH(phenyl), -
NHSO2(C1-C4 alkyl), -NHSO2(phenyl), and -NHSO2(C1-C4 haloalkyl); and
wherein optionally substituted acyl, alkoxycarbonyl, sulfinyl and sulfonyl are as
defined herein.
[0056] The tenn "substituted amino" also refers to the group -NReRf wherein Re and
Rf, together with the nitrogen to which they are bound, form an optionally substituted 5- to 7-
membered nitrogen-containing, non-aromatic, heterocycle which optionally contains 1 or 2
additional heteroatoms chosen from nitrogen, oxygen, and sulfur.
[0057] Compounds of Formula I-XIII include, but are not limited to, optical isomers
of compounds of Formula I-XIII, racemates, and other mixtures thereof. In those situations,
the single enantiomers or diastereomers, i.e., optically active forms, can be obtained by
asymmetric synthesis or by resolution of the racemates. Resolution of the racemates can be
accomplished, for example, by conventional methods such as crystallization in the presence
of a resolving agent, or chromatography, using, for example a chiral high-pressure liquid
chromatography (HPLC) column. In addition, compounds of Formula I-XIII include Z- and
E- forms (or cis- and trans- forms) of compounds with carbon-carbon double bonds. Where
compounds of Formula I-XIII exists in various tautomeric forms, chemical entities of the
present invention include all tautomeric forms of the compound. Compounds of Formula I-
XIII also includes crystal forms such as polymorphs and clathrates.
[0058] Chemical entities of the present invention include, but are not limited to
compounds of Formula I-XIII and all pharmaceutically acceptable forms thereof.
Pharmaceutically acceptable forms of the compounds recited herein include pharmaceutically
acceptable salts, solvates, chelates, non-covalent complexes, prodrugs, and mixtures thereof.
In certain embodiments, the compounds described herein are in the form of pharmaceutically
acceptable salts. Hence, the terms "chemical entity" and "chemical entities" also encompass
pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs, and
mixtures.
[0059] "Pharmaceutically acceptable salts" include, but are not limited to salts with
inorganic acids, such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate,
sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate,

fumarate, tartrate, succinate, citrate, acetate, lactate, methanesulfonate, p-toluenesulfonate, 2-
hydroxyethylsulfonate, benzoate, salicylate, stearate, and alkanoate such as acetate, HOOC-
(CH2)n-K*I where n is (M, and like salts. Similarly, pharmaceutically acceptable cations
include, but are not limited to sodium, potassium, calcium, aluminum, lithium, and
ammonium.
[0060] In addition, if the compound of Formula I-XIII is obtained as an acid addition
salt, the free base can be obtained by basifying a solution of the acid salt* Conversely, if the
product is a free base, an addition salt, particularly a pharmaceutically acceptable addition
salt, may be produced by dissolving the free base in a suitable organic solvent and treating the
solution with an acid, in accordance with conventional procedures for preparing acid addition
salts from base compounds. Those skilled in the art will recognize various synthetic
methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition
salts. '
[0061] As noted above, prodrugs also fall within the scope of chemical entities, for
example ester or amide derivatives of the compounds of Formula I-XIII. The term "prodrug"
includes any compound that becomes a compound of Formula I-XIII when administered to a
patient, e.K*i., upon metabolic processing of the prodrug. Examples of prodrugs include, but
are not limited to, acetate, formate, and benzoate and like derivatives of functional groups
(such as alcohol or amine groups) in the compounds of Formula I-XIII. In some
embodiments, the prodrug is a phosphate ester. A thorough discussion of prodrugs is
provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the
A.CS. Symposium Series, in Edward B. Roche, ed., Bioreversible Carriers in Drug Design,
American Pharmaceutical Association and Pergamon Press, 1987, and in Design of Prodrugs,
ed. H. Bundgaard, Elsevier, 1985, each of which are incorporated herein by reference.
[0062 ] The term "solvate" refers to the chemical entity formed by the interaction of a
solvent and a compound. Suitable solvates are pharmaceutically acceptable solvates, such as
hydrates, including monohydrates and hemi-hydrates.
[0063] The term "chelate" refers to the chemical entity formed by the coordination of
a compound to a metal ion at two (or more) points.
[0064] The term "non-covalent complex" refers to the chemical entity formed by the
interaction of a compound and another molecule wherein a covalent bond is not formed
between the compound and the molecule. For example, complexation can occur through van
der Waals interactions, hydrogen bonding, and electrostatic interactions (also called ionic

bonding).
[0065] The term "active agent" is used to indicate a chemical entity which has
biological activity. In certain embodiments, an "active agent" is a compound having
pharmaceutical utility. For example an active agent may be an anti-cancer therapeutic.
[0066] The term "antimitotic" refers to a drug for inhibiting or preventing mitosis, for
example, by causing metaphase arrest. Some antitumour drugs block proliferation and are
considered antimitotics.
[0067] The term "therapeutically effective amount" of a chemical entity of this
invention means an amount effective, when administered to a human or non-human patient,
to provide a therapeutic benefit such as amelioration of symptoms, slowing of disease
progression, or prevention of disease e.K*i., a therapeutically effective amount may be an
amount sufficient to decrease the symptoms of a disease responsive to CENP-E inhibition. In
some embodiments, a therapeutically effective amount is an amount sufficient to reduce
cancer symptoms. In some embodiments a therapeutically effective amount is an amount
sufficient to decrease the number of detectable cancerous cells in an organism, detectably
slow, or stop the growth of a cancerous tumor. In some embodiments, a therapeutically
effective amount is an amount sufficient to shrink a cancerous tumor.
[0068] The term "inhibition" indicates a significant decrease in the baseline activity of
a biological activity or process. "Inhibition of CENP-E activity" refers to a decrease in
CENP-E activity as a direct or indirect response to the presence of at least one chemical entity
described herein, relative to the activity of CENP-E in the absence of the at least one
chemical entity. The decrease in activity may be due to the direct interaction of the chemical
entity with CENP-E, or due to the interaction of the chemical entity(ies) described herein with
one or more other factors that in turn affect CENP-E activity. For example, the presence of
the chemical entity(ies) may decrease CENP-E activity by directly binding to CENP-E, by
causing (directly or indirectly) another factor to decrease CENP-E activity, or by (directly or
indirectly) decreasing the amount of CENP-E present in the cell or organism.
[0069] A "disease responsive to CENP-E inhibition" is a disease in which inhibiting
CENP-E provides a therapeutic benefit such as an amelioration of symptoms, decrease in
disease progression, prevention or delay of disease onset, or inhibition of aberrant activity of
certain cel1-types.
[0070] "Treatment or treating means any treatment of a disease in a patient, including:

a) preventing the disease, that is, causing the clinical symptoms of the disease not
to develop;
b) inhibiting the disease;
c) slowing or arresting the development of clinical symptoms; and/or
d) relieving the disease, that is, causing the regression of clinical symptoms.
[0071] "Patient" refers to an animal, such as a mammal, that has been or will be the
object of treatment, observation or experiment. The methods of the invention can be useful in
both human therapy and veterinary applications. In some embodiments, the patient is a
mammal; in some embodiments the patient is human; and in some embodiments the patient is
chosen from cats and dogs.
[0072] The present invention is directed to a class of novel chemical entities that are
inhibitors of one or more mitotic kinesins. According to some embodiments, the chemical
entities described herein inhibit the mitotic kinesin, CENP-E, particularly human CENP'-E.
CENP-E is a plus end-directed microtubule motor essential for achieving metaphase
chromosome alignment. CENP-E accumulates during interphase and is degraded following
completion of mitosis. Microinjection of antibody directed against CENP-E or
overexpression of a dominant negative mutant of CENP-E causes mitotic arrest with
prometaphase chromosomes scattered on a bipolar spindle. The tail domain of CENP-E
mediates localization to kinetochores and also interacts with the mitotic checkpoint kinase
hBubR1. CENP-E also associates with active forms of MAP kinase. Cloning of human (Yen,
et al., Nature, 359(6395):536-9 (1992)) CENP-E has been reported. In Thrower, et al., EMBO
J., 14:918-26 (1995) partially purified native human CENP-E was reported on. Moreover, the
study reported that CENP-E was a minus end-directed microtubule motor. Wood, et al., Cell,
91:357-66 (1997)) discloses expressed Xenopus CENP-E in E. coli and that XCENP-E has
motility as a plus end directed motor in vitro. CENP-E See, PCT Publication No. WO
99/13061, which is incorporated herein by reference.
[0073] In some embodiments, the chemical entities inhibit the mitotic kinesin, CENP-
E, as well as modulating one or more of the human mitotic kinesins selected from HSET (see,
U.S. Patent No. 6,361,993, which is incorporated herein by reference); MCAK (see, U.S.
Patent No. 6,331,424, which is incorporated herein by reference); RabK-6 (see U.S. Patent
No. 6,544,766, which is incorporated herein by reference); Kif4 (see, U.S. Patent No.
6,44O,684, which is incorporated herein by reference); MKLP1 (see, U.S. Patent No.
6,448,025, which is incorporated herein by reference); Kifl5 (see, U.S. Patent No. 6,355,466,

which is incorporated herein by reference); Kid (see, U.S. Patent No. 6,387,644, which is
incorporated herein by reference); Mppl, CMKrp, KinI-3 (see, U.S. Patent No. 6,461,855,
which is incorporated herein by reference); Kip3a (see, PCT Publication No. WO 01/96593,
which is incorporated herein by reference); Kip3d (see, U.S. Patent No. 6,492,151, which is
incorporated herein by reference); and KSP (see, U.S. Patent No. 6,617,115, which is
incorporated herein by reference).
[0074] The methods of inhibiting a mitotic kinesin comprise contacting an inhibitor of
the invention with one or more mitotic kinesin, particularly a human kinesin; or fragments
and valiants thereof. The inhibition can be of the ATP hydrolysis activity of the mitotic
kinesin and/or the mitotic spindle formation activity, such that the mitotic spindles are disrupted.
[0075] The present invention provides inhibitors of one or more mitotic kinesins, in
particular, one or more human mitotic kinesins, for the treatment of disorders associated with
cell proliferation. The chemical entities compositions and methods described herein can
differ in their selectivity and are used to treat diseases of cellular proliferation, including, but
not limited to cancer, hyperplasias, restenosis, cardiac hypertrophy, immune disorders, fungal
disorders and inflammation.
i
[0076] Accordingly, the present invention provides at least one chemical entity
chosen from compounds of Formula I

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein
R1 is optionally substituted aryl, optionally substituted heterocycloalkyl, or optionally

substituted heteroaryl; •
X is-K*I or-SO2-;
R2 is hydrogen or optionally substituted lower alkyl;
W is — CR4-, -CH2CR4-, or N;
R3 is -K*I-R7, hydrogen, optionally substituted alkyl, optionally substituted heterocyclyl,
cyano, optionally substituted sulfonyl, or optionally substituted aryl;
R4 is hydrogen or optionally substituted alkyl;
R5 is hydrogen, hydroxyl, optionally substituted amino, optionally substituted heterocyclyl; or
optionally substituted lower alkyl;
R6 is hydrogen, optionally substituted alkyl, optionally substituted alkoxy, optionally
substituted aryloxy, optionally substituted heteraryloxy, optionally substituted
alkoxycarbonyl-, optionally substituted aminocarbonyl-, optionally substituted aryl,
optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally
substituted aralkyl; and
R7 is optionally substituted lower alkyl, optionally substituted aryl, hydroxyl, optionally
substituted amino, optionally substituted aralkoxy, or optionally substituted alkoxy;
provided that ifW is N, then R5 is not hydroxyl or optionally substituted amino, and R6 is not
optionally substituted alkoxy, optionally substituted aralkoxy, optionally substituted
heteroaralkoxy, or optionally substituted amino.
[0077] In some embodiments, R1 is optionally substituted aryl, or optionally
substituted heteroaryl. In some embodiments, R1 is optionally substituted aryl. In some
embodiments, R1 is optionally substituted phenyl. In some embodiments, R1 is phenyl
substituted with one, two or three groups independently selected from optionally substituted
heterocyclyl, optionally substituted alkyl, sulfonyl, halo, optionally substituted amino,
optionally substituted sulfanyl, optionally substituted alkoxy, optionally substituted aryloxy,
optionally substituted heteroaryloxy; acyl, hydroxyl, nitro, cyano, optionally substituted aryl,
and optionally substituted heteroaryl-. In some embodiments, R1 is chosen from 3-halo-4-
isopropoxy-phenyl, 3-cyano-4-isopropoxy-phenyl, 3-cyano-4-isopropylamino-phenyl, 3-
chloro-4-isopropylamino-phenyl, 3-cyano-4-trifluoroisopropyloxyphenyl, 3-chloro-4-
trifluoroisopropyloxyphenyl, 3-cyano-4-cylobutyloxyphenyl, 3-chloro-4-cylobutyloxyphenyl,
3-cyano-4-cylopropyloxyphenyl, and 3-chloro-4-cylopropyloxyphenyl. In some
embodiments, R1 is 3-haIo-4-isopropoxy-phenyl or 3-cyano-4-isopropoxy-phenyl.
[0078] In some embodiments, R2 is hydrogen.

't0079] In some embodiments, X is -K*I-.
[0080] In some embodiments,W is—CR4-and R4 is hydrogen.
[0081] In some embodiments, the compounds described herein possess a potentially
chiral center, for example, when W is —CR4-. The invention contemplates the use of pure
enantiomers and mixtures of enantiomers, including racemic mixtures, although the use of a
substantially optically pure enantiomer will generally be preferred. The term "substantially
optically pure" or "enantiomerically pure" means having at least about 95% of the described
enantiomer with no single impurity greater than about 1% and particularly, at least about
97.5% enantiomeric excess. In some embodiments, the stereogenic center at W is as shown
below:

[0082] In some embodiments, R3 is -K*I-R7; hydrogen; optionally substituted lower
alkyl; cyano; optionally substituted sulfonyl; optionally substituted aryl; or optionally
substituted heterocyclyl. In some embodiments, R3 is optionally substituted lower alkyl. In
some embodiments, R3 is lower alkyl that is optionally substituted with a hydroxyl or a
phosphate ester thereof, lower alkyl that is optionally substituted with a lower alkoxy, lower
alkyl that is optionally substituted with an optionally substituted amino group, or lower alkyl
that is optionally substituted with K*I-R8 where R8 is hydroxyl or optionally substituted
amino.
[0083] In some embodiments, R5 is hydrogen, hydroxyl, or optionally substituted
lower alkyl. In some embodiments, R5 is hydrogen.
[0084] In some embodiments, the compounds described herein possess a potentially
chiral center when R5 is not hydrogen. The invention contemplates the use of pure
enantiomers and mixtures of enantiomers, including racemic mixtures, although the use of a
substantially optically pure enantiomer will generally be preferred. The term "substantially
optically pure" or "enantiomerically pure" means having at least about 95% of the described
enantiomer with no single impurity greater than about 1% and particularly, at least about

97.5% enantiomeric excess.
[0085] In some embodiments, R6 is optionally substituted aryl, optionally substituted
heteroaryl, optionally substituted heterocyclyl, or optionally substituted alkyl (such as wherein
the alkyl group is substituted with an optionally substituted amino group or wherein the alkyl
group is optionally substituted cycloalkyl-). In some embodiments, R6 is phenyl substituted
with one or two of the following substituents: optionally substituted heteroaryl, optionally
substituted amino, aralkoxy,-halo, hydroxymethyl-, hydroxy, cyano, alkoxy, phenyl, phenoxy,
methylenedioxy, ethylenedioxy, sulfonyl, aminocarbonyl, carboxy, alkoxycarbonyl, nitro,
heteroaralkoxy, aralkoxy, and optionally substituted heterocyclyl.
[0086] Also provided is at least one chemical entity chosen from compounds of
Formula II

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R5, R6, and W are as described for compounds of
Formula I and wherein
R11 is optionally substituted heterocyclyl, optionally substituted lower alkyl, nitro,
cyano, hydrogen, sulfonyl, or halo;
R12 is hydrogen, halo, optionally substituted alkyl, optionally substituted amino,
optionally substituted sulfanyl, optionally substituted alkoxy, optionally substituted aryloxy,
optionally substituted heterocyclyl, or optionally substituted heteroaryloxy; and
R13 is hydrogen, acyl, optionally substituted alkyl-, optionally substituted alkoxy, halo,
hydroxyl, nitro, cyano, optionally substituted amino, alkylsulfonyl-, alkylsulfonamido-,
alkylsulfonyl-, carboxyalkyl-, aminocarbonyl-, optionally substituted aryl or optionally
substituted heteroaryl-.
[0087] In some embodiments, R11 is hydrogen, cyano, nitro, or halo. In some

embodiments, R11 is chloro or cyano.
[0088] In some embodiments, R12 is optionally substituted lower alkoxy, optionally
substituted lower alkyl, or optionally substituted amino-. In some embodiments, R11 is
chosen from isopropoxy, isopropylamino, trifluoroisopropyloxy, cylobutyloxy, and
cylopropyloxy. In some embodiments, R12 is lower alkoxy (such as propoxy) or 2,2,2-
trifluoro-1-methyl-ethoxy. In some embodiments, R12 is propoxy or 2,2,2-trifluoro-1-methyl-
ethoxy. In some embodiments, R12 is not -O-(GH2)nNH2 or -O-(CH2)4NH(CH3) wherein n is
4 or 5.
[0089] In some embodiments R11 and R12, taken together, form an optionally
substituted carbocyclic or heterocyclic ring. In some embodiments, R1 and R12, taken
together, form a methylenedioxy or ethylenedioxy ring. In some embodiments, R12 and R13,
taken together, form an optionally substituted carbocyclic or heterocyclic ring. In some
embodiments, R11 and R13, taken together, form an optionally substituted carbocyclic or
heterocyclic ring.
[0090] In some embodiments, R13 is hydrogen.
[0091] In some embodiments, R2 and R11, taken together, form an optionally
substituted carbocyclic or heterocyclic ring, i.e., R1, X, N, and R2, taken together, form an

optionally substituted carbocyclic or heterocyclic ring. In certain embodiments, a substituted
2,4-dioxo-l,4-dihydro-2H-quinazolin-3-yl ring is formed, e.K*i.,

wherein the phenyl ring is optionally substituted. In other embodiments, a 4-oxo-4H-
quinazolin-3-yl ring is formed, e.K*i.,


wherein the phenyl ring is optionally substituted. In certain embodiments, a 4-oxo-4H-
pyridopyrimidin-3-yl ring is formed, e.K*i.,

wherein one of R. S, T, and U is nitrogen with the others being -CH and wherein the pyridine
ring is optionally substituted.
[0092] Also provided is at least one chemical entity chosen from compounds of
Formula HI

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R6, R11, R12, and R13 are as described for compounds of
Formula II.
[0093] Also provided is at least one chemical entity chosen from compounds of
Formula IV


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 ai*e as described for compounds'of
Formula in.
[0094] Also provided is at least one chemical entity chosen from compounds of
Formula V

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R3, R11, R12, and R13 are as described for compounds of
Formula III and wherein
R14 is optionally substituted heteroaryl; and
R15 is chosen from hydrogen, halo, hydroxyl, and lower alkyl.
[0095] In some embodiments, R14 is chosen from
7,8-dihydro-imidazo[1,2-K*i][1,3]oxazin-2-yl,
3a,7a-dihydro-l H-benzoimidazo1-2-yl,

imidazo[2,1-b]oxazo1-6-yl,
oxazo1-4-yl,
5,6,7,8-tetrahydro-imidazo[ 1,2-a]pyridin-2-yl,
1H-[1,2,4]triazo1-3-yl,
2,3-dihydro-imidazo1-4-yl,
1H-imidazo1-2-yl,
imidazo[1,2-a]pyridin-2-yl, »
thiazo1-2-yl.
thiazo1-4-yl,
pyrazo1-3-yl, and
1H-imidazo1-4-yl,
each of which is optionally substituted with one, two, or three groups chosen from optionally
substituted lower alkyl, halo, acyl, sulfonyl, cyano, nitro, optionally substituted amino, and
optionally substituted heteroaryl.
[0096] In some embodiments, R14 is chosen from
1H-imidazo1-2-yl,
imidazo[1,2-a]pyridin-2-yl; and
1H-imidazo1-4-yl,
each of which is optionally substituted with one or two groups chosen from optionally
substituted lower alkyl, halo, and acyl.
[0097] In some embodiments, R15 is hydrogen.
[0098] Also provided is at least one chemical entity chosen from compounds of
Formula VI


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 are as described for compounds of
Formula III.
[0099] Also provided is at least one chemical entity chosen from compounds of
Formula VII

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R2, R6, R11, R12, and R13 are as described for compounds of
Formula HI and wherein
R9 is chosen from optionally substituted alkoxy, optionally substituted cycloalkoxy,
optionally substituted arylalkoxy, optionally substituted amino and optionally
substituted lower alkyl.
[00100] In some embodiments, R9 is lower alkyl substituted with hydroxyl or
optionally substituted amino. In some embodiments, R9 is lower alkyl substituted with
hydroxyl, amino, N-methylamino, or N,N-dimethylamino.
[00101] Also provided is at least one chemical entity chosen from compounds of
Formula VIII


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof,
wherein R1, X ,W, R3, R4, R6, and R7 are as defined for compounds of Formula I and wherein
R2 and R5, together with the atoms to which they are bound, form an optionally substituted 5-
7 membered heterocycle which optionally may include one or two additional
heteroatoms.
[00102] In some embodiments,, R2 taken together with R5, form an optionally
substituted pyrrolidinyl ring or optionally substituted piperidinyl ring.
[00103] Also provided is at least one chemical entity chosen from compounds of
Formula DC

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof,
wherein R1, X ,W, R2, R3, R4, and R7 are as defined for compounds of Formula I and wherein
R5 and R6, together with the atoms to which they are bound, form an optionally substituted 5-
7 membered heterocycle which optionally may include one or two heteroatoms.
[00104] In some embodiments, R5 and R6, together with the atoms to which they are
attached, form an optionally substituted 2H-[1,2,3]triazo1-4-yl; an optionally substituted 1H-
benzoimidazo1-2-yl; an optionally substituted piperazinyl ring; an optionally substituted
morpholinyl ring; or an optionally substituted 1H-Imidazo1-4-yl ring; an optionally
substituted isoxazo1-4-yl ring.
[00105] Also provided is at least one chemical entity chosen from compounds of
Formula X


and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R1, X ,W, R4, R5, R6 and R7 are as defined for compounds of
Formula I and wherein
R2 and R3, taken together with the atoms to which they are attached, form an optionally
substituted 3- to 7-membered heterocyclic ring.
[00106] In some embodiments, R2 and R3, taken together with the atoms to which they
are attached, form an optionally substituted 3- to 7-membered heterocyclic ring. -In some
embodiments, they form an aziridinyl ring.
[00107] Also provided is at least one chemical entity chosen from compounds of
Formula XI

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein W, R3, R4, R5, R6 and R7 are as defined for compounds of
Formula I and wherein
R1, X, N, and R2, taken together, fonn a substituted 2,4-dioxo-l,4-dihydro-2H-quinazolin-3-
yl, 4-oxo-4H-quinazolin-3-yl, or 4-oxo-4H-pyridopyrimidin-3-yl ring.
[00108] Also provided is at least one chemical entity chosen from compounds of

Formula XII

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,
and mixtures thereof, wherein R1, W, R4, R5, and R6 are as defined for compounds of
Formula I and wherein
-X-N(R2)-is-K*I=N-;and
X taken together with R3 forms an optionally substituted heterocyclic ring;
in each case, provided that if W is N, then R5 is not hydroxyl or optionally substituted amino,
and R6 is not optionally substituted alkoxy, optionally substituted aralkoxy, optionally
substituted heteroaralkoxy, or optionally substituted amino.
[00109] In certain embodiments, -X-N(R2)- is -K*I=N-; and X'taken together with R3
forms an optionally substituted heterocyclic ring, including but not limited to 3H-
[1,3,4]oxadiazo1-2-one; 4,5-dihydro-oxazole; thiazole; imidazole; 3,5-dihydro-imidazo1-4-
one; or 3H-pyrimidin-4-one, each of which is optionally substituted.
[00110] Also provided is at least one chemical entity chosen from compounds of
Formula XIII

and pharmaceutically acceptable salts, solvates, chelates, non-covalent complexes, prodrugs,

and mixtures thereof,
wherein R1, X ,W, R2, R4, R5, and R7 are as defined for compounds of Formula I and wherein
R3 and R6, together with the atoms to which they are bound, form an optionally substituted 5-
7 membered heterocycle which optionally may include one or two additional
heteroatoms.
[00111] In some embodiments, R3 and R6 together with the atoms to which they are
attached, form an optionally substituted pyrrolidinyl ring, an optionally substituted piperidinyl
ring, or an optionally substituted 1,2,3,4-tetrahydro-quinolin-3-yl ring.
[00112] Also provided is at least one chemical entity chosen from compounds recited
in Table 1,2, 3, 4, 5, or 6, and phamiaceutically acceptable salts, solvates, chelates, nonr
covalent complexes, prodrugs, and mixtures thereof.
[00113] The compounds can be named and numbered using AutoNom version 2.1,
ChemDraw Ultra 6.O, Cambridgesoft, Cambridge, MA; StructName algorithm of
ChemDraw Ultra 9.O, Cambridgesoft, Cambridge, MA or ISIS-DRAW.


N- {1-[4-(S-Bromo-5-methyl-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-( 1-Acetylamino-ethyl)-1-
ethyl-1H-irnidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide
N-(1-{4-[2-acetyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(isopropoxy)-
benzamide


N-{ 1-[4-(8-ethyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide

N-{1-[4-(8-isopropenyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(l -Acetylamino-propyl)-1-
ethyl-1H-irnidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
(isopropoxy)-benzamide
N-(1-{4-[2-(1-Acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-[1-(4-{2-[1-(Acetyl-methyl-amino)-
ethyl]-1-ethyl-1H-imidazo1-4-yl}-
benzyl)-3-hydroxy-propyl]-3-chloro-4-
(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(1-{4-[2-(l -Acetylamino-ethyl)-1-
propyl-1H-imidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide


N-{ 1-[4-(8-chloro-imidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-{1-[4-(8-trifluoromethyl-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-[ 1-(Acetyl-methyl-amino)-
ethyl]-1-ethy]-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N- {1-[4-(8-Bromo-imidazo [ 1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-( 1-Acetylamino-ethyl)-1-
isopropyl-1H-imidazo1-4-yl] -benzyl}-
3-hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-( 1-Acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide


N-(2-(2-amino-2-methyl-
propionylamino)-1- {4-[8-(l -hydroxy-
ethyl)-imidazo[ 1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-[1-(3-methyl-ureido)-
ethyl]-l -ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(2-(2-dimethylamino-aeetylamino)-
1- {4-[8-methyl-imidazo[1,2-a]pyridin-
2-yl]-benzyl}-ethyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-Acetyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-Acetylamino-2-methyl-
propyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N- {1-[4-(8-chloro-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
butyl}-3-cyano-4-isopropoxy-
benzamide


N-{1-[4-(8-Bromo-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-
(isopropoxycarbonylamino)-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide
N- {1-[4-(8-methyl-imidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-3-carbamoyl-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-dimethylaniino-acetylamino)-
1-{4-[8-(1-hydroxy-ethyl)_
imidazo[ 1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-cyano-4-isopropoxy-
benzamide
N-'{ 1-[4-(8-acetyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N- {1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide


N-(2-(2-dimethy]amino-acetylamino)-
1-{4-[8-(1-hydroxy-ethyl)-
imidazo [ 1,2-a]pyridin-2-yl]-benzy 1}-
ethyl)-3-chIoro-4-isopropoxy-
benzamide
N-(i-{4-[2-(1-Acetylamino-ethyl)1-
cyclopropylmethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzamide
N-{ 1-[4-(8-isopropyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-amino-2-methyl-
propionylamino)-1-{4-[8-bromo-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-formylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benz3'l}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{3-fluoro-4-[2-(1-methyl-
1hydroxy-ethyl)-l -ethyl-1H-imidazo1-
4-yl]-benzyl}-3-hydroxy-propyl)-3-
chloro-4-isopropoxy-benzamide


N-(2-(2-dimethylamino-acetylaminp)-
1-{4-[8-bromo-imidazo[1,2-a]pyridin-
2-yl]-benzyl}-ethyl)-3-chloro-4-
isopropoxy-benzamide
N-{1-[2-fluoro-4-(8-methyl-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-(3-
hydroxypropyl)-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzamide
N- {1-[4-(8-acetyl-5-methyl-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(l -{4-[2-(l -Acetylamino-2-methyl-
propyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzamide
N-[1-[4-(2-acetyl-1-ethyl-1H-
imidazo1-4-yl)-benzyl]-2-(2-hydroxy-
acetylamino)-ethyl]-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide


N-(1-{4-[2-t-butyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-dimethylamino-acetylamino)-
1-{4-[8-bromo-imidazo[1,2-a]pyridin-
2-yl]-benzyl}-ethyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-( 1-acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-2-
dimethylcarbamoyl-etliyl)-3-chloro-4-
isopropoxy-benzamide
N- {1-[4-(8-methyl-irnidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
butyl}-3-cyano-4-isopropoxy-
benzamide
N-(l -{2-fluoro-4-[2-t-butyl-l -methyl-
1H-imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-carbamoyl-
propy!)-3-chloro-4-isopropoxy-
benzamide


N-(1-{4-[2-isobutyryl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-(2-hydroxy-ethyl)-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-methyl-1-hydroxy-
ethyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(1-{3-fluoro-4-[2-acetyl-1-methyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-[1-[4-(8-Bromo-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-2-(2-oxo-
tetrahydro-pyrimidin-1-yl)-ethyl]-3-
cyano-4-isopropoxy-benzamide
N-(1-{4-[2-(3-hydroxy-pent-3-yl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide


N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-(2,2,2~trifluoro-1-
methyl-ethoxy)-benzamide
N-{ 1-[4-(8-(l -hydroxy-ethyl)-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-1-methyl-
ethyl)-1-(2,2,2-trifluoroethyl)-1H-
imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-[1-[4-(2-acetyl-1-ethyl-1H-
imidazo1-4-yl)-benzyl]-2-(2-hydroxy-
acetylamino)-ethyl] -3-chloro-4-
isopropoxy-benzamide
N-(1-{4- [2-acetyl-1-(2-methoxyethyl)-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-amino-2-methyl-
propionylamino)-1- {4-[8-( 1-hydroxy-
ethyl)-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-cyano-4-isopropoxy-
benzamide


N-(2-(2-amino~propionylamino)-1- {4-
[8-(1-hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)3-
chloro-4-isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-methyl-l H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-(5,5-dimethyl-758-dihydro-
imidazo[1,2-K*i] [1,3]oxazin-2-yl)-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzatnide
N-(1-{4-[2-isobutyryl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-(8-methyl-5!6,7,8-tetrahydro-
imidazo[1,2-a]pyridin-2-yl)-benzyl}-
3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-l-propyl-1H-
imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide


N-(l -{4-[2-acetyl-l -ethyl-1H-
imidazo1-4-yl]-benzyl}-2-carbamoy]
ethyl)-3-chloro-4-isopropoxy-
benzamide
N-(l-{4-[2-acetyl-l -methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-arnino-propionylamino)-1-{4-
[8-methyl-imidazo[1,2-a]pyridin-2-yl]
benzyl}-ethyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-hydroxy-2-methyl-
propyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(1-{3-fluoro-4-[2-(1-hydroxy-1-
methyl-ethyl)-1-ethyl-1H-imidazo1-4
yl]-benzyl}-3-hydroxy-propyl)-3-
chloro-4-(2,2,2-trifluoro-1-methyl-
ethoxy)-benzamide
N-(1-{4-[2-propionyl-1-methyl-1H-
imidazol-4-yl)-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide


N-(1-{4-[2-(1-hydroxy-1-methyl-
ethyl)-1-(2,2,2-trifluoroethyl)-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
-at
N-(1-{4-[2-(1-formylamino-ethyl)-1-
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(2-(2-hydroxy-acetylamino)-1- {4-
[8-(1-hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
chloro-4-isopropoxy-benzamide
N-(3-fluoro-1-{4-[2-(1-hydroxy-1-
methyl-ethyl)-1-methyl-1H-imidazo1-
4-yl]-benzyl}-3-hydroxy-propyl)-3-
cyano-4-isopropoxy-benzamide
N-( 1-{4-[2-(l -acetylamino-ethyl)-1-
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-{1-[4-(8-chloro-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-isopropoxy-
benzamide


N-(2-(2-amino-2-methyl-
propionylamino)-1- {4-[8-methyl-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-cyano-4-isopropoxy-
benzamide
N-{ 1-[4-(S-methyl-imidazo[1,2-.
a]pyridin-2-}yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-carbamoyl-
butyl)-3-cyano-4-isopropoxy-
benzamide
N-{1-4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-carbamoyl-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-(l -{4-[2-(1-hydroxy-1-methyl-
ethyl)-1-ethyl-1H-imidazo1-4-yl] -
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide


N-(1-{3-fluoro-4-[2-acetyl-1-methyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-carbamoyl-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{3-fluoro-4-[2-acetyl-1-ethyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-propionyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-{ 1-[4-(8-methyl-imidazo[] ,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-(2,2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-(2-(2-hydroxy-acetylamino)-1-{4-
[8-methyl-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-cyano-4-isopropoxy-
benzamide


N-(1-{4-[2-(1-hydroxyimino-ethyl)-1-
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-(3-hydroxy-pent-3-yl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(2-(2-dimethylamino-acetylamino)-
1-{4-[8-methyl-imidazo[1,2-a]pyridin-
2-yl]-benzyl}-ethyl)3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-(2,2,2-trifluoro- '
ethyl)-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(2-(3-hydroxy-propionylamino)-1-
{4-[8-bromo-imidazo[1,2-a]pyridin-2-
yl]-benzyl}-ethyl)-3-chloro-4-
isopropoxy-benzamide
N-(2-(2-hydroxy-acetylamino)-1-{4-
[8-bromo-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide


N-(2-(2-amino-2-methyl-
propionylamino)-1- {4-[8-bromo-
imidazo[ 1,2-a]pyridin-2-yl] -benzyl}-
ethyl)-3-cbJoro-4-isopropoxy-
benzamide
N-[1-[4-(2-t-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl] -2-ureido-
ethyl]-3-cyano-4-isopropoxy-
benzamide
N-{ 1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-carbamoyl-
propyl}-3-chloro~4-isopropoxy-
benzamide
N-(1-{4-[2-(1-hydroxypropyl)-1-ethyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-2-carbamoyl-
ethyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-hydroxy-acetylamino)-1-{4-
[8-methyl-imidazo[ 1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide


N-(H4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2,2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-(1-{4-[2-isobutyryl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-3-bydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-{1-[4-(8-(1-hydroxy-ethyl)- .
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
carbamoyl-propyl}-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-]-metliyl-
ethyl)-1-ethyl-1H-imidazo1-4-)d]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzamide
N- {1-[4-(8-bromo-imidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-3-carbamoyl-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-hydroxy-propionylamino)-1-
{4-[8-bromo-imidazo[ 1,2-a]pyridin-2-
yl] -benzyl}-etliyl)-3-chloro-4-
isopropoxy-benzamide


N-{1-[4-(8-(carbamoyl)-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-{1-[4-(S-(7-hydroxy-ethyl)-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-[1-[4-(2-t-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-
(acetylamino)-ethyl]-3-cyano-4-
isopropoxy-benzamide
N- {1-[2-fluoro-4-(8-methyl-
imidazof 1,2-a]pyridin-2-yl)rbenzy] j-3-
hy droxy-propy]}-3-chloro-4-
isopropoxy-benzamide
N-{1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-(2,2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-(l -{4-[2-(1-hydroxy-1-methyl-
ethyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide


N-( I - {4-[2-( 1-AcetyJamino-ethyl)-1-
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
■»
N-(1-{4-[2-f-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-carbamoyl-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-{1-[4-(4-methyl-3a,7a-dihydro-1H-
benzoimidazo1-2-yl)-benzyl]-3-
hydroxy-propyl}-3-chIoro-4-
isopropoxy-benzamide
N-(1-{2,3..5,6-tetrafluoro-4-[2-t-butyl-
1-methyl-1H-imidazo1-4-yl]-benzyl}-
3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-( 1-fluoro-prop-2-
yloxy)-benzamide
N-(1-{4-[2-r-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide


N-(1-{4-[2-(1-hydroxy-1-methyl-
ethyl)-l -emyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-{1-[4-(8-(3,5-dimethyl-isoxazo1-4-
yl)-imidazo[1,2-a]pyridin-2-yl)-
benzyl]-3-hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-{ 1-[4-(8-(1-hydroxy-ethyl)-
imidazo[1s2-a]pyridin-2-yl)-benzyl]-3-
carbamoyl-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-2-methyl-
propyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-!-methyl-
ethyl)-1-methyl-1H-imidazo1-4-yl]-
benzy 1}-3-hy droxy-propyl)-3-chl oro-
4-isopropoxy-benzamide
N-(1-{4-[2-isopropenyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide


N-(1-{4-[2-acetyl-1-isopropyl-1H-
imidazoI-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-trifluoromethyl-l -methyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3~cyano-4-isopropoxy-
benzamide
N-(2-{4:[2-/~butyl-1-methyl-1H-
imidazo1-4-yl]-phenyl}-1-(1H-
[1,2,4]triazo1-5-yl)-ethyl)-3-cyano-4-
isopropoxy-benzamide
N-{ 1-[3-chloro-4-(S-methyl-
imidazo [ 1,2-a]pyridin-2-y])-benzyl] -3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N- {1-[4-(3-methyl-imidazo[2,1-
b]oxazo1-6-yl)-benzyl]-3-hydroxy-
butyl}-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-hydroxy-propiony]araino)-1-
{4-[8-(l -hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
chloro-4-isopropoxy-benzamide


N-(1-hydroxy-l -{4-[2-f-butyl-1-
methyl-1H-imidazo1-4-yl]-phenyl}-4-
hydroxy-butyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-2-(N,N-
ditnethylcarbamoyl)-ethyl)-3-chloro-4-
isopropoxy-benzamide
N-(2-(2-hydroxy-propionylamino)-1-
{4-[8-methyl-imidazo[1,2-a]pyridin-2-
yl]-benzyl}-ethyl)-3-cyano-4-
isopropoxy-benzamide
N-[ 1-[4-(8-bromo-imidazo[1,2-
a]pyridin-2-yl)-benzylj-2-(3-methyl-
ureido)-ethyl]-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-hydroxy-lmethyl-
ethyl)-1-methyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4- [2-[1-(acetylamino)-propyl]-
1-ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-metIiyl-ethoxy)-benzamide


N-[1-[4-(2-M>utyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-(3-methyl-
ureido)-ethyl]-3-cyano-4-isopropoxy-
benzamide
N-(l -{4-[2-(cyclopropylcarbonyl)-l -
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-cyclobutoxy-
benzamide
N-(l -{4-[2-(l -hydroxy- lmethyl-
ethyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-2-carbamoyl-ethyl)-3-chIoro-
4-isopropoxy-benzamide
N-[1-[4-(2-r-butyl-1-methyl-1H-
imidazoJ-4-yl)-benzyl]-2-( 1-methyl-
ureido)-ethyl]-3-chloro-4-isopropoxy-
benzamide
N- {1-[4-(8-hydroxymethyl-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide


N-(1-{4-[2-r-butyl-1-(2-hydroxyethyl)-
1H-imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-[ 1-[4-(8-methyl-imidazo[ 1,2-
a]pyridin~2-yl)-benzyl]-2-ureido-
ethyl]-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-(methylsulfonyl)-l -methyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-isbpropoxy-
benzamide
N-(1-{3-fluoro-4-[2-acetyl-1-ethyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2,2,2-trifluoro-1-
methyl-etlioxy)-benzamide
N-[1-[4-(8-bromo-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-2-ureido-
ethyl]-3-cyano-4-isopropoxy-
benzamide
N-(2-{4-[2-t-butyl-l -methyi-1H-
imidazo1-4-yl]-phenyl}-l -(5-methyl-
[1,2,4]oxadiazo1-3-yl)-ethyl)-3-cyano-
4-isopropoxy-benzamide


N-{ 1-[2,6-difluoro-4-(8-chloro-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-2,2-dimethyl-
propyl)-1-methyl-1H-imidazo1-4-yl] -
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-forniylamino-ethyl)-1-
methyl-1H-imidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4- [2-( 1-hydroxy-1 methyl-
ethyl)- 1-methyl-1H-imidazo1-4-yl]-
benzyl}-3-carbamoyl-propyl)-3-
chloro-4-isopropoxy-benzamide
N-(2-(2-amino-propionylamino)-1- {4-
[8-(1-hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
cyano-4-isopropoxy-benzamide
N-(l -{4-[2-(l -hydroxy-lmethyl-
ethyl)-1-methyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-tri fluoro-1-methyl-ethoxy)
-benzamide


N-(2-(2-amino-propionylamino)-1- {4-
[8-bromo-imidazo[ 1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chIoro-4-isopropoxy-
benzamide
N-(1-{4-[2-(1-acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-2-
methylcarbamoyl-ethyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-acetyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-2-
dimethylcai-bamoyl-ethyl)-3-chloro-4-
(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(2-(2-hydroxy-propionylamino)-1-
{4-[8-methyl-imidazo[ 1,2-a]pyridin-2-
yl]-benzyl}-ethyl)-3-cyano-4-
isopropoxy-benzamide
N-{1-[4-(8-cyano-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-amino-2-methyl-
propionylamino)-1- {4- [8-methyl-
imidazo [ 1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-chloro-4-isopropoxy-
benzamide


N-{ 1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-2,3-dichhloro-4-isopropoxy-
benzamide
N-(1-{4-[2-a«etyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2)2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-(1-{3-fluoro-4-[2-acetyl-1-methyl-
1H-imidazoI-4-yl]-benzyl}-3-h}'droxy-
propyl)-3-cyano-4-(2,2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-{ 1-[4-(8-(1-hydroxy-ethyl)-
imidazof 1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-chloro-4-
isopropoxy-benzamide
N-[1-[4-(2-t-butyl-1-methyl-1H-
irnidazo1-4-yl)-benzyl]-2-(2-oxo-
tetrahydro-pyrimidin-1-yl)-ethyl] -3-
cyano-4-isopropoxy-benzamide
N-[1-[4-(2-t-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-3-
hydroxycarbamoyl-propyl] -3-cyano-4-
i sopropoxy-benzamide


N-{ 1-[4-(5-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzylJ-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-meth µMH-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-cyano-4-(isopropylamino)-
benzamide
N-[1-[4-(2-r-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-
(fonnylamino)-ethyl]-3-cyano-4-
ispropoxy-benzamide
N-(1-{4-(2-Acetyl-oxazo1-4-yl)-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(2-(2-amino-acetylamino)-1- {4-[8-
bromo-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-hydroxy-2-methyl-
propionylamino)-1- {4-[8-bromo-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-chloro-4-isopropoxy-
benzamide


N-(2-{4-[5-t-buty]-1-methyl-1H-
[1,2,4]triazo1-3-yl]-phenyl}-1-
([1,2,4]oxadiazo1-3-yl)-ethyl)-3-cyajio-
4-isopropoxy-benzamide
N-(l -{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-2-amino-3-chloro-4-
isopropoxy-benzamide
N-( 1- (4-[2-( 1-acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
carbamoyl-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide
N-(2-(2-amino-3-hydroxy-
propionylamino)-1- {4-[8-bromo-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-chloro-4-isopropoxy-
benzamide
N-{ 1-[2,6-difluoro-4-(8-methyl-
imidazo[152-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-chloro-4-
isopropoxy-benzamide
N-{ 1-[4-(S-amino-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide


N-{ 1-[4-(8-acetyl-imidazo[1,2-
a]pyridin-2-yl)-benzylj|-3-hydroxy-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-{1-[4-(8-(1-methyl-1-hydroxy-
ethyl)-imidazo[1,2-a]pyridin-2-yl)-
benzyl]-3-hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-
(raethoxycaibonylamino)-ethyl)-1-
methy]-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N-(1-{4-[2-(1-
(methoxycarbonylamino)-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl-ethoxy)-benzamide
N-(2-(2-hydroxy-acetylamino)-1- {4-
[8-( 1-hydroxy-ethyl)-imidazo [ 1.2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
cyano-4-isopropoxy-benzamide
N-[1-[4-(2-r-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-carbamoyl-
ethyl]-3-chloro-4-ispropoxy-
benzamide


N-(1-{4-[2-( 1-(ethoxycarbonylamino)-
ethyl)-1-ethyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-chloro-
4-(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
•»
N-{ 1-[4-(imidazo[1,2-a]pyridin-2-yl)-
benzyl]-3-hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(2-{4-[2-r-butyl-l -methyl-1H-
imidazo1-4-yl]-phenyl}-1-(1H-
imidazo1-2-yl)-ethyl)-3-cyano-4-
isopropoxy-benzamide
N- {1-[4-(8-methyl-imidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-(isopropylamino)-
benzamide
N-[1-[4-(8-.bromo-imidazo[1,2-
a]pyridin-2-yl)-benzyl] -2-(3-methyl-
ureido)-ethyl]-3-chIoro-4-isopropoxy-
benzamide
N-{ 1-[4-(8-(1-hydroxypropyl)-
imidazo [ 1,2-a]pyridin-2-yl)-benzyl] -3-
hydroxy-propyl}-3-chloro-4-
isopropoxy-benzamide


N-[ I -[4-(2-r-butyl-1-(2-aminoethyl)-
1H-imidazo1-4-yl)-benzyl]-2-(2-
hydroxy-acetylamino)-ethyl]-3-chloro-
4-isopropoxy-benzamide
N-(1-{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-(2-hydroxy-propionylamino)-1-
{4-[8-naethyl-imidazo[1,2-a]pyridin-2-
yl] -benzyl}-ethyl)-3-chloro-4-
isopropoxy-benzamide
N-[1-[4-(2-t-butyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-(3-hydroxy-
ureido)-ethyl]-3-cyano-4-isopropoxy-
benzamide
N-(2-(3-amino-propionylamino)-l~{4-
[8-bromo-imidazo[1,2-a]pyridin-2-ylJ-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide
N-(2-{4-[2-r-butyl-1-methyl-1H-
imidazo1-4-yl]-phenyl}-1-
([1,2,4]oxadiazo1-3-yl)-ethyl)-3-cyano-
4-isopropoxy-benzamide


N-(2-(2-amino-3-hydroxy-
propionylamino)-1- {4-[8-bromo-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-chloro-4-isopropoxy-
benzamide
N- {1-[4-(8-nitro-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-cyano-4-isopropoxy-
benzamide
N- {1-[2,6-difluoro-4-(8-methyl-
5,6,7.,8-tetrahydro-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-ethyl-1H-
imidazo1-4-yl]-benzyl}-2-
methylcarbamoyl-ethyl)-3-chlbro-4-
(2,2,2-trifluoro-1-methyl-ethoxy)-
benzamide
N-(2-(2-amino-propionylamino)-1- {4-
[8-meth)'1-imidazo[ 1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide
N-[1 ~[4-(8-bromo-imidazo[ 1,2-
a]pyridin-2-yl)-benzyl]-2-(2-oxo
imidazolidinyl)-ethyl]-3-chloro-4-
isopropoxy-benzamide


N-[1-[4-(2-(1-hydroxy-1-methyl-
ethyl)-1-methyl-1H-imidazo1-4-yl)-
benzyl]-2-(2-amino-propionylamino)-
ethyl]-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-acetyl-1-butyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-( 1-acetylamino-ethyl)-1-
ethyl-1H-imidazo1-4-yl]-benzyl}-2-
carbamoyl-ethyl)-3-chloro-4~
isopropoxy-benzamide
N-(1-{4-[4-r-butyl-1H-imidazoI-2-yl]-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[2-(2,2-dimethyl-propyl)-1-
methyl-1H-imidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(2-(2-hydroxy-propionylamino)-1-
{4-[S-(1-hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
cyano-4-isopropoxy-benzamide


N-(1-{4- [2-isobutyryl-1-methyl-1H-
imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2,2,2-trifluoro-1-
methyl-ethoxy)-benzamide
N-(1-{3-fluoro-4-[2-(1-hydroxy-1-
methyl-ethyl)-1-methyl-1H-imidazo1-
4-yl] -benzyl}-3-hydroxy-propyl)-3-
chloro-4-isopropoxy-benzamide
N-(1-{4-[2-(1-hydroxy-ethyl)-1-
methyl-1H-imidazo1-4-yl] -benzyl}-3-
hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-(1-{4-[5-r-butyl-1-methyl-1H-
[1,2,4]triazo1-3-yl]-benzyl}-3-
hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-[ 1-[4-(2-t-butyl-1-(2-amino-ethyl)-
1H-imidazo1-4-yl)-benzyl]-2-(2-
dimethylamino-acetylamino)-ethyl] -3-
chloro-4-isopropoxy-benzamide
N-[1-{4-[1-(3-(t-
butoxycarbonylamino)-propyl)-2-t-
butyl-2J3-dihydro-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl]-3-chloro-
4-isopropoxy-benzamide


N-(2-(2-hydroxy-propionylamino)-1-
{4-[8-(1-hydroxy-ethyl)-imidazo[1,2-
a]pyridin-2-yl]-benzyl}-ethyl)-3-
cyano-4-isopropoxy-benzamide
N-(2-(acetylamino)-1- (4-[8-methyl-
imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-t-butyl-l -(2-aminoethyl)-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propy])-3-cyano-4-is6propoxy-
benzamide
N-(1-{4-[2-(1-methoxyimino-ethyl)-1-
metyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-
isopropoxy-benzamide
N- {1-[4-(8-(3-hydroxy-propenyl)-
imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide
N-(2-(2-dimethylamino-acetylamino)-
1-{4-[8-carbamoy]-imidazo[1.,2-
a]pyridin-2ryl]-benzyl}-ethyl)-3-
cyano-4-isopropoxy-benzamide


N-(1-{3-fluoro-4- [2-acetyl-1-methyl-
1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2,2,2-triiluoro-1-
methyl-ethoxy)-benzarnide
3k
N-(2-(2-ammo-acetylarnino)-1-{4-[S-
bromo-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-cyano-4-isopropoxj'-
benzamide
N-(2- {4-[2-acetyl-l -metiiyl-1H-
imidazo1-4-yl]-phenyl}-1-(5-methyl-
[1,2,4]oxadiazo1-3-yl)-ethyl)-3-cyano-
4-isopropoxy-benzamide
N-[1-[4-(2-^utyl-1-methyl-1H-
imidazo1-4-yl)-benzyl]-2-hydroxy-3-
azido-propyl]-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[5-f-butyl-4-methyl-1H-
hriidazo1-2-yl]-benzyl}-3-hydroxy-
propy])-3-cyano-4~isopropoxy-
benzamide
N-{ 1-[4-(8-(l -hydroxy-ethyl)-
imidazof 1,2-a]pyridin-2-yl)-benzyl] -3-
hydroxy-propyl}-3-cyano-4-
isopropoxy-benzamide


N-(1-{4-[5-r-butyl-4-methyl-1H-
imidazo1-2-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-isopropoxy-
benzamide
N-(1-{4-[2-( 1-acetylamino-ethyl)-1-
methyl-1H-imidazo1-4-yl]-benzyl}-3-
hydroxy-propyl)-3-chloro-4-(2,2,2-
trifluoro-1-methyl~ethoxy)-benzamide
N-(1-{4-[2-^butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-2-amino-
butyl)-3-cyano-4-isopropoxy-
benzamide
N-{ 1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
butyl}-3-cyano-4-isopropoxy-
benzamide
N-(1-{4-[2-r-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-2-hydroxy-
propyl)-3-cyano-4-isopropoxy-
benzamide
N-(2-(2-amino-propionylamino)-1- {4-
[8-bromo-imidazo[1,2-a]pyridin-2-yl]-
benzyl}-ethyl)-3-chloro-4-isopropoxy-
benzamide

N-(1-{4-[2-acetyl-1-methyl-1H-
imidazo1-4-yl] -benzyl}-3-hydroxy-
propyl)-3-cbloro-4-isopropylamino-
benzamide
N-(1-{4-[2-r-butyl-1H-imidazo1-4-yl]-
benzyl}-3-hydroxy-propyl)-3-cyano-4-
isopropoxy-benzamide
N-{1-[4-(8-methyl-imidazo[1,2-
a]pyridin-2-yl)-benzyl]-3-hydroxy-
propyl}-2-nitro-4-trifluoromethyl-
benzamide
N-(1-{4-[2-t-butyl-1-methyl-1H-
imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-iodo-4-isopropoxy-
benzamide
(S)-3-chloro-N-(l -(4-(1-eAyl-2-(2-hydroxypropan-2-yl)-1H-imidazo1-4-yl)phenyl)-4-
hydroxybutan-2-yl)-4-isopropoxybenzamide
(S>N-(1-(4-(2-acetyl-1-ethyl-1H-imidazo1-4-yl)phenyl)-4-hydroxybutan-2-yl)-3-chloro-4-
isopropoxybenzamide
N-((S)-1-(4-(2-(l -acetamidoethyl)-1-ethyl-l H-imidazoI-4-yl)phenyl)-4-hydroxybutan-2-yl)-3-
chloro-4-isopropoxybenzamide
3-chloro-N-((S)-1-(4-(1-ethyl-2-(2-hydroxypropan-2-yl)-1H-imidazo1-4-yl)phenyl)-4-
hydroxybutan-2-yl)-4-(l ,1,1-trifluoropropan-2-yloxy)benzamide
N-((S)-1-(4-(2-(1-acetamidoethyl)-1-ethyl-1H-imidazo1-4-y])phenyl)-4-hydroxybutan-2-yl)-3-
chloro-4-( 1,1,1-trifluoropropan-2-yloxy)benzamide
(S)-N-(1-(4-(2-acetyl-1-(2,2,2-trifluoroethyl)-1H-imidazo1-4-y])phenyl)-4-hydroxybutan-2-
yl)-3-chloro-4-isopropoxybenzamide

3-cWoro-N-((S)-4-hydroxy-1-(4-(8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl)phenyl)butan-
2-yl)-4-isopropoxybenzamide
(S)-3-chloro-N-(I-(2-(dimethylamino)acetamido)-3-(4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl)propan-2-yl)-4-isopropoxybenzamide
3-chloro-N-((S)-1-(2-(dimethyiamino)acetamido)-3-(4-(8-(1-hydroxyethyl)imidazo[1,2-
a]pyridin-2-yl)phenyl)propan-2-yl)-4-isopropoxybenzamide
3-Cyano-A^-((l^-3-hydroxy-1-{[4-(8-methylimidazo[1,2-a3pyridin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide;
3-Oiloro-A^-[(liS)-3-hydroxy-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-
yl]phenyl} methyl)propyl]-4-[( 1-methylethyl)oxy]benzamide
3-Chloro-JV-[(l 5)-2-[(AyV-dimethylglycyl)amino]-1-({4-[8-( 1-hydroxyethyl)imidazo[ 1,2-
a]pyridin-2-yl]phenyl}methyl)ethyl]-44J(1-methylethyl)oxy]benzamide
iV-((l^-2-(D-Alanylamino)-1-{[4-(8-bromoimidazo[1,2- 3-chloro-4-[(1-methylethyl)oxy]benzamide
3-Chloro-A^-((l^-2-[(2-methylalanyl)amino]-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]raethyl}ethyl)-4-[(1-methylethyl)oxy]benzamide .
3-Chloro-JV-(( 1 ^-2-[(JV^-dimethylglycyl)amino]-1- {[4-(8-metbylimidazo[ 1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-[(1-methyl6thyl)oxy]benzamide
JV-(( li?)-4-Amino-1- {[4-(8-methylimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-4-oxobutyl)-3-
chloro-4-[( 1-methylethyl)oxy]benzamide
7V:((li?)-1-{[4-(2-ace(yl-1-methyl-liy-imidazo1-4-yl)phenyl]methyl}-4-amino-4-oxobutyl)-3-
. chloro-4-[(l -methylethyl)oxyJbenzamide
3-Cyano-JV-[( 1 S)-3-hydroxy-1-({4-[8-(l -hydroxyethyl)imidazo[ 1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-A'-((l»S)-3-hydroxy-1-{[4-(8-niethylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl} propyl)-4-[(l -methylethyl)oxy]benzamide
3-Cyano-A4(l ^)-2-[(A^V-dime.hylglycyl)amino]-1-( {4-[8-(l -hydroxyethyl)imidazo[ 1,2-
a]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
3-Cyano-A^((l^-2-[(i\yV,-dimethylglycyl)amino]-1-{[4-(8-methylimidazo[1,2-a]pyri
yl)phenyl]methyl} ethyl)-4-[(l -raethylethyl)oxy]benzamide;
N-((l R)-1- {[4-(2-Acetyl-l -methyl-1H-imidazo1-4-yl)phenyl]methyl}-4-amino-4-oxobutyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide

N-[(l R)-4-Amino-1-({4-[2-( 1-hydroxy-1-methylethyl)-1-methyl-1H-imidazo1-4-
yl]phenyl} methyl)-4~oxobxityl] -3-cyano-4-[( 1-methyl ethyl)oxy]benzamide
N-[(lS)-2-(D-Alanylamino)-1-((4-[ 1-(2-aminoethyl)-2-( 1,1-dimethylethyl)-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide
N-((lS)-2-{4-[1-(2-Ammoethyl)-2-(l,1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1-{[(2-
methylalanyl)amino]methyl}ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide
N-[(lS)-2-(D-Alanylamino)-1-({4-[1-(2-aminoethyl)-2-(l,1-dimethyleth^l)-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide
N-((lS)-2-{4-[1-(2-Aminoethyl)-2-(l,1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1-
{[(hydroxyacetyl)amino]methyl}ethyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide
N-((lS)-2- {4-[ 1-(2-Aminoethyl)-2-(l ,1-dimethylethyl)-1H-imidazoM-yl]phenyl}-1-{[(2-
methylalanyl)amino]methyl}ethyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide
N-((l S)-2- {4-[ 1-(2-Aminoethyl)-2-(l ,1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1- {[(N,N-
dimethylglycyl)amino]methyl} ethyl)-3-cyano-4- [(1-methylethyl)oxy]benzamide
3-Chloro-^-[(15)-2-{4-[8-(1-hydroxyethy])imidazo[1,2-a]pyridin-2-yl]phenyl}-1-({[(27?)-2-
hydroxypropanoyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
N-((l S)-2-[(Aminocarbonyl)amino]-1- {[4-(8-bromoimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide
N- {(1 S)-2-[4-(8-Bromoimidazo[ 1,2-a]pyridin-2-yl)phenyl]-1-[(2-oxotetrahydro-l (2H)-
pyrimidinyl)methyl]ethyl}-3-chloro-4-[(1-methylethyl)oxy]benzamide
N- {(1 S)-2-[4-(8-Bromoimidazo[ 1,2-a]pyridin-2-yl)phenyl]-1-[(2-oxohexahydro-1H-1,3-
diazepin-1-yl)methyl] ethyl}-3-chloro-4-[( 1-methyl ethyl)oxy]benzamide
N-((l S)-2-[(Aminocarbonothioyl)amino]-1- {[4-(8-bromoimidazo[ 1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide
2-(4- {(2S)-2-[( {3-Cyano-4-[(l -methylethyl)oxy]phenyl) carbonyl)amino]-3-[( 1,2,3-thiadiazo1-
4-ylcarbony])amino]propyl}phenyl)imidazo[1,2-a]pyridine-S-carboxamide
N-((l S)-2-[(Aminosulfonyl)amino]-1- {[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl} ethyl)-3-cyano-4-[( 1-methylethyl)oxy]benzamide
(3 S)-3-[( {3-Chloro-4-[( 1-methylethyl)oxy]phenyl} carbonyl)amino]-4- {4-[2-(l, 1-
dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}butanoic acid
N-[(l S)-2-[(Aminosulfonyl)amino]-l -({4-[2-(l, 1-dimethylethyl)-1-methyl-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide

r-((l^-1-{[4-(li^-BenzimidazoI-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-chloro-4-[(1-
methylethyl)oxy]benzamide
3-Chloro-JV-[(l,S)-3-hydroxy-]-({4-[5-(trifluoromethyl)-li7-benzimidazo1-2-
yl]phenyl}methyl)propyl]-4-[(1-methy]ethyl)oxy]benzamide
3-Chloro-iV-((l S)-1- {[4-(5,6-dimethyl- l#-benzimidazo1-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxyJbenzamide
3-Chloro-iV-[(15)-3-hydroxy-l -({4-[5-(methyloxy)- 1H-benzimidazo1-2- "
yl]phenyl}methy])propyl]-4-[(1-methylethyl)oxy]benzamide
3-ChIoro-A^-((15)-1-{[4-(5-cMoro-li7-benzimidazo1-2-yl)phenyl]methyl}-3-hydroxypropyl)-
4-[( 1-methyl ethy])oxy]benzamide
3-Chloro-Ar-((15)-3-hydroxy-1-{[4-(4-methyl-l^benzimidazo1-2-yl)phenyl]methy]}propyl)-
4-[(1-methylethyl)oxy]benzamide
3-Chloro-7V-((15)-1- {[4-(6-chloro-lif-imidazo[4,5-&]pyridin-2-yl)phenyl]methyl}-3-
hydrox)'propyl)-4-[(1-methylethyl)oxy]benzamide
Ethyl 2-(4-{(25)-2-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)-liJ-benzimidazole-5-carboxylate
2-(4-{(2.S)-2-[({3-Chloi-o-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)-lif-benzimidazole-5-carboxylicacid
iV-((15)-3-Amino-1-{[4-(li7-benzimidazo1-2-yl)phenyl]methyl}propyl)-3-chloro-4-[(1-
methylethyl)oxy]benzamide
3-Cyano-iV-(( 1S)-1- {[4-(8-cyanoimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide
Ar-((15)-1-{[4-(8-Chloroimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
cyano-4-[( 1-methylethyl)oxy]benzamide
3-Cyano-N-[( 15)-3-hydroxy-1-({4-[8-(trifluoromethyl)imidazo[ 1,2-a]pyridin-2-
yl]phenyl} methyl)propyl]-4-[( 1-methylethyl)oxy]benzamide
S-Cyano-A^CCli^-S-hydroxy-1-l^-CS-hydroxyimidazotl^-a]pyridin^-
yl)phenyl]methyl}propyl)-4-[(l -methylethyl)oxy]benzamide
2-(4-{(2^}-2-[({3-Cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazof1,2-(3]pyridine-7-carboxamide
Ethyl 2-(4-{(2^)-2-[({3-cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[1,2-a]pyridine-7-carboxylate

J-Cyano-//-((15)-3-hydroxy4-{[4-(8-nitroimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}propyl)-
4-[(1-methylethyl)oxy]benzamide
iV"-((l^-1-{[4 cyano-4-[(1-methylethyl)oxy]benzamide
2-(4- {(25)-2-[( {3-Gyano-4-[(l -methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[1,2-a]pyridine-8-carboxamide
3-Cyano-A^4(l,S)-3-hydroxy-lK{4-[8-(hydroxymethyl)imidazo[1,2-a]pyn(lin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide
N-[( IS)-1-({4-[S-(Aminomethyl)imidazo[ 1,2-a]pyridin-2-yl]phenyl}methyl)-3-
hydroxypropyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide
N-((\S)-1- {[4-(8-Acetylimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide
3-Cyano-Ar-[(15)-3-hydroxy4K{4-[8-(1-hydroxy-1-methylethyl)imidazo[1,2-fl]pyridin-2-
yljphenyl} methyl)propyl] -4-[.( 1-methylethyl)oxy]benzamide
3-Cyano-W-[(l1S)-3-hydroxy-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-
yl]phenyl} methyl)propyl]-4-[( 1-methyl ethyl)oxy]benzamide
3-Cyano-Ar-((15)-3-hydroxy-1-{[4-(8-methyl-5,6,7,8-tetrahydroimidazo[1,2-fl]pyridin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide
3-Cyano-A-[( 1S)-1-({4-[2-( 1,1-dimethylethyl)-1-(2-b.ydroxyethyl)- l//-imidazo1-4-
yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide
N-[( lS)-\-( {4-[ 1-[2-(Acetylamino)ethyl]-2-( 1,1-dimethy]ethyl)-lif-imidazo1-4-
yl]phenyl}methyl)-3-hydroxypropyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide
3-Cyano-A7- {(l^-3-hydroxy-1-[(4- {8-[(li?)-l -hydroxyethyl]imidazo[1,2-a]pyridin-2-
yl}phenyl)methyl]propyl}-4-[(1-methylethyl)oxy]benzamide
3-Cyano-^-{(15r)-3-hydroxy-1-[(4-{8-[(15}-1-hydroxyethyl]imidazo[1)2-fl]pyridin-2-
yl}phenyl)methyl]propyl}-4-[(l -methylethyl)oxy]benzamide
3-Chloro-iV-[(l S)-3-hydroxy-l -({4-[8-(l -hydroxypropyl)imidazo[ 1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide
Ar-((l1S)-1-{[4-(8-Bromoimidazo[1,2-«]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
chloro-4-[(1-methylethyl)oxy]benzamide
S-Chloro-AT-ft 1 S)-1-{[4-(8-chloroimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methyletb.yl)oxy]benzamide

^•^^^-^[(l^-S-hydroxy-lKi^CSK1-hydroxy^-methylpropy^imidazoll^-alpyridin^-
yl]phenyl}methyl)propyl]-4-[(l -methylethyl)oxy]benzamide
iV-[(li?)-4-Amino-l -({4-[8-(l -hydroxyethyl)imidazo[1,2-o]pyridin-2-yl]phenyl}methyl)-4-
oxobutyl]-3-chloro-4-[(l -methylethyl)oxy]benzamide
iV-[(li?)^-Amino-1-({4-[8K1-hydroxye%l)imidazo[1,2-fl]pyridin-2-yl]phenyl}meihyl)-4-
oxobutyl]-3-cyano-4-[(1-raethylethyl)oxy]benzamide
S-Oiloro-iV-^l^^-I^CS-fluoro-K*i-methylimidazoCl^-a]pyridin^-yOpb^yllmeihylj-S-
hydroxypropyl)-4-[(l -methylethyl)oxy]benzamide
3-Cyano-AT-((l^-1-{[4-(3-fluoro-8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-iV-(( 1 S)-2-hydioxy-1- {[4-(8-methylimidazo[ 1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide
3-CWoro-4-[(1-methylethyl)oxy]-A^[(15}-2-[4-(8-methylimidazo[1,2-fl]pyridin-2-yl)phenyl]-
1-(4-morpholinylmethyl)ethyl]benzamide
3-Chloro-JV-((15)-2-(4-hydroxy-1-piperidinyl)-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-iV-((l^-2-(3-hydroxy-1-pyiTolidinyl)-1-{[4-(8-me%limidazo[1,2-a]pyridin-2-
yl)phenyl]methyl} ethyl)-4-[(l -methylethyl)oxy]benzamide
3-Chloro-iV-((l^)-2-[(25}-2-(hydroxymethyl)-l -pyrrolidinyl]-!- {[4-(8-methylimidazo[1,2-
a]pyiidin-2-yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-^-((l^-2-[(2i?)-2-(hydroxymethyl)-1-pyrrolidinyl]-1-{[4-(8-methylimidazo[1,2-
3-CMoro-4-[(1-methylethyl)oxy]-^-((lS)-2-[4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]-
1-{[(2,2,2-trifluoroeihyl)amino]methyl}ethyl)benzamide
3-Chloro-iV-((l1S)-2-[(2-hydroxye yl)phenyl]methyl} ethyl)-4-[(l -methylethyl)oxy]benzamide
3-Cyano-^-((l,S)-1-{[4-(8-ethyl-5-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(l -methylethyl)oxy]benzamide
Methyl (3S)-3-[( {3-chloro-4-[(l -methylethyl)oxy]phenyl}carbonyl)amino]-4- {4-
[(phenylcarbonyl)amino]phenyl}butanoate
3-Chloro-A'-[(15)-3-hydroxy-1-({4-[(phenylcarbonyl)amino]phenyl}methyl)propyl]-4-[(1-
metiiylethyl)oxy]benzamide

3-Chloro-JV-{(liS}-1-[(4-{[(4-chlorophenyl)carbonyl]amino}phenyl)methyl]-3-
hydroxypropyl}-4- [(1-methylethyl)oxy]benzamide
Phenylmethyl(4-{(2.S)-2-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl} phenyl)carbamate
3-Chloro-7V-((15)-3-hydroxy-1-{[4-({[2-
(methylamino)phenyl]carbonyl}amino)phenyl]methyl}propyl)-4-[(1-
methylethyl)oxy]benzamide. *
jV-(4-{(2.S)-2-[({3-Chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl} phenyl)-4-pyridinecarboxamide
3-Chloro-iV-[(15)-1-({4-[(cyclohexylcarbonyl)amino]phenyl}methyl)-3-hydroxypropyl]-4-[(1-
methylethyl)oxy]benzamide
3-CMoro-iV-[(ljS)-1-({4-[(3,3-dimethylbutanoyl)amino]phenyl}methyl)-3-hydroxypropyl]-4-
[(1-methylethyl)oxy]benzamide
3-Chloro-JV-[(15)-3-hydxoxy-1-({4-[(phenylacetyl)amino]phenyl}methyl)propyl]-4-[(1-
methylethyl)oxy]benzamide
3-Chloro-iV-{(15,)-3-hydroxy-1-[(4-{[(phenylamino)carbonyl]amino}phenyl)methyl]propyl}-
4-[(1-methylethyl)oxy]benzamide
3-Cyano-7V-((15)-3-hydroxy-1-{[4-(8-methyl-5^xo-5,6-dihydroimidazo[1,2-K*i]pyri)midin-2-
yl)phenyl]methyl} propyl)-4-[( 1-methylethyl)oxy]benzamide
3-Cyano-AT-((15)-3-hydroxy-1-{[4-(1-methyl-3-oxo-2,3-dihydro-lif-imidazo[1,2-a]imidazo1-
6-yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide
3-Cyano-JV-((l S)-3-hydroxy-1-{[4-(8-oxo-7,8-dihydroimidazo[ 1,2-a]pyrazin-2-
yl)phenyl]methyl}propyl)-4-[(l -methylethyl)oxy]benzamide
2,3-DichLoro-iV:-((l'S)-3-hydroxy-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}propyl)-4-[(l -methylethyl)oxy]benzamide
N-((l S)-3-Hydroxy-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}propyl)-4-[(l -
methylethyl)oxy]-3-nitrobenzamide
3-Chloro-A'-[(liS)-2-[(hydroxyacetyl)amino]-1-({4-[8-(1-hydroxyetliyl)imidazo[1,2-a]pyridin-
2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-^-[(15)-244-[8K1-hydroxyethyl)imidazo[152-a]pyridin-2-yl]phenyl}-1-({[(2i?)-2-
hydroxypropanoyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
3-CWoro-Ar-[(l^-2-{4-[8-(1-hydroxyethyl)imidazo[152-a]pyridin-2-yl]phenyl}-1-({[(21S)-2-

liydroxypropanoyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-JV^-[(lS)-2-[(Ar//"-dimethylglycyl)amino]-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-
fl]pyridin-2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamid6
JV-[(l,S)-2-(D-Alanylaiiiino)-1-({4-[8-(1-hydroxyethyl)iinidazo[152-fl]pyridin-2-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide
3-Chloro-iV-[(l>S)-3-hydroxy-1-({4-[8-(1-hydroxyethyl)imidazo[152-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-JV-((15)-2-{4-[S-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}-1-{[(2-
methylalanyl)amino]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide
(35)-3-[({3-Chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-{4-
[(phenylcarbonyl)amino]phenyl} butanoic acid
3-K*I^loro-^/-{(15)-3-hydroxy-1-[(4-imidazo[1,2-a]pyridin-6-ylphenyl)methyl]propyl}-4-[(1-
methylethyl)oxy]benzamide
3-Chloro-N-IX 1S)-1-((4-[2-( 1,1-dimethylethyl)imidazo[1,2-a]pyridin-6-yl]phenyl}methyl)-3-
hydroxypropyl]-4-[(l -methylethyl)oxy]benzamide
3-Chloro-N- {(15)-3-hydroxy-l -[(4-imidazo[1,2-a]pyridin-2-ylphenyl)methyl]propyl}-4-[(l -
methylethyl)oxy]benzamide
3-K*I^loro-7V-{(lS)-3-hydroxy-1-[(4-imidazo[1,2-a]pyrimidin-2-ylpb.enyl)methyl]propyl}-4-
[(1-methylethyl)oxy]benzamide
S-CWoro-iV-CCl^-S-hydroxy-1-l^-CS-methylimidazotl^-a^yridin^-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide
S-Chloro-AHO 5)-3-hydroxy-1-{[4-(7-methylimidazo[ 1,2-a]pyrimidin-2-
yl)phenyl]methyl }propyl)-4-[(1-methylethyl)oxy]benzamide
3-Cyano-Ar-{(15)-3-hydroxy-1-[(4-imidazo[2,1-6][1,3]thiazo1-6-ylphenyl)methyl]butyl}-4-
[(1-methylethyl)oxy]benzamide
3-Cyano-AT-((15)-3-hydroxy-1-{[4-(3-methylimidazo[2,1-6][1,3]thiazo1-6-
yl)phenyl]methyl} butyl)-4-[(1-methylethyl)oxy]benzamide
3-Cyano-iV-((15)-1-{[4-(2,3-dihydroimidazo[2,1-&][1,3]thiazo1-6-yl)phenyl]methyl}-3-
hydroxybutyl)-4-[( 1-methylethyl)oxy]benzamide
3-Cyano-iV-((15)-1-{[4-(l51-dioxido-2,3-dihydroimidazo[2,1-6][1J3]thiazo1-6-
yl)phenyl]methyl}-3-hydroxybutyl)-4-[(1-methylethyl)oxy]benzamide
7V-[(1 S)-l -({4-[ 1-(3-Aminopropyl)-2-(l, 1-dimethylethyl)- lF-imidazo1-4-yl]phenyl}methyl)-

3-hydroxypropyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide
3-Cyano-4-[(1-methylethyl)oxy]-iV^-[(15)-2-[4K8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]-
1-(5-methyl-1,2,4-oxadiazo1-3-yl)ethyl]benzamide
3-Cyano-iV'-[(1^4-({4-[8-(3J5-dimethyl-4-isoxazolyl)imidazo[132-a]pyridin-2-
yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide
3-Cyano-7V-((15)-3-hydroxy-1-{[4-(8-phenylimidazo[1,2-fl]pyridin-2-
yl)phenyl]methyl}propyl)-4-.[(1-methylethyl)oxy]benzamide
3-Cyano-JV-[(15)-3-hydroxy-1-({4-[8-(liy-pyrazo1-4-yl)imidazo[1,2-a]pyridin-2-
yi]phenyl}methyl)propyl]-4-[(l -methylethyl)oxy]benzamide
3-Cyano-N-[(l S)-3-hydroxy-l -( {4-[8-(4-isoxazolyl)imidazo[ 1,2-a]pyiidin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide
iV-((liS)-1-{[4-(8-Ac^tylimidazo[1,2-fl]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
chloro-4-[(1-methylethyl)oxy]benzamide
Ethyl (2^)-3-[2-(4-{(25)-2-[({3-cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[1,2-a]pyridin-8-yl]-2-propenoate
(2£)-3-[2-(4-{(2iS)-2-[({3-Cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[1,2-a]pyridin-8-yl]-2-propenoic acid
iV-{(16)-1-[(4-{8-[(l£)-3-Amino-3-oxo-1-propen-1-yl]imidazo[1,2-a]pyridin-2-
yl}phenyl)methyl]-3-hydroxypropyl}-3-cyano-4-[(1-methyleth.yl)oxy]benzamide
7V-[(15)-1-({4-[8-(3-Amino-3-oxopropyl)imidazo[1,2-fl]pyridin-2-yl]phenyl}methyl)-3-
hydroxypropyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide
3-Chloro-^K*I(l^-1-{[4-(3-chloro-8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide
iV-((l^)-1-{[4-(3-Chloro-8-methylimidazo[1,2-fl]pyridin-2-yl)phenylimethyl}-3-
hydroxypropyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide
3-Cyano-7^-[(15)-l yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-jV-((l,S}-2-hydroxy-1-{[5-(8-niethylimidazo[1,2-a]pyridin-2-yl)-2-
pyridinyl]methyl} ethyl)-4-[(l -methylethyl)oxy]benzamide
3-Chloro-^-((15}-2-hydroxy-1-{[5-(8-metihylimidazo[1,2-a]pyridin-2-yl)-2-
thienyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-#-[(15)-1-({4-[2-(l,1-dimethylethyl)-1-methyl-lif-imidazo1-4-yl3-2-

iluorophenyl}methyl)-3-hydioxypropyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-^[(15)4-({4-[2Kl,1-dimethylethyl)4-methyl-m-imidazo1-4-yl]-2,6-
difluorophenyl}methyl)-3-hydrox>'propyl]-4-[(1-methylethyl)oxy]benzamide
3-Chloro-iV-[(l S)-1-( (2-chloro-4-[2-(l, 1-dimethylethyl)-1-methyl-liJ-imidazo1-4-
yl]phenyl} methyl)-3-hydroxypropyl]-4-[(l -methylethyl)oxy]benzamide
3-K*I^loro-^-[(15)4-({5-[2Kl,1-dimethyletlvyl)-1-raetliyl-l^-iinidazo1-4-yl]-2-
pyridinyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide
3-CWoro-jV-((15)4-{[2-chloro^^8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide
3-Chloro-7V-(( 15)-1- {[2-chloro-4-(8-chloroimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-7V-((15)-1-{[2,5-difluoro-4-(S-methylimidazo[1,2-fl]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
3-Chloro-iV-((l S)-1-{[3-chloro-4-(8-methylimidazo[1,2-o]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(l -methylethyl)oxy]benzamide
3-Chloro-N-[(lS)-1-({4-[2^1,1-dimethylethyl)4-methyl-1H-imidazo1-4-yl]phenyl}methyl)
3-(methylamino)-3-oxopropyl]-4-[(1-methylethyl)oxy]benzamide
3-Cyano-N-[(lS)-2-{442Kl,1-dimethyiethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-
({[(phenylamino)carbonyl]amino}methyl)ethyl]-4-[(1-methylediyl)oxy]benzamide
3-Cyano-N-[(lS)-2-{4-[2-(l,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-
({[(ethylamino)carbonyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
N-[(l S)-2-(Aminosulfonyl)-l -((4-[2-(l, 1-dimethylethyl)-l -methyl-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide
3-Cyano-N-((lS)-2-{442-(14-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-
{[(methylsulfonyl)amino]methyl}ediyl)-'4-[(1-methylethyl)oxy]benzamide
3-Cyano-N-{(lS)-2-{4-[2-(l,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-[({[(2-
hydroxyethyl)amino]carbonyl}amino)mefliyl]ethyl}-4-[(1-methylethyl)oxy]benzamide
N-[(S)-1-[4-(2-tert-Butyl-1-methyl-1H-imidazo1-4-yl)-benzyl]-2-(2-methoxy-ethanoylamino)-
ethyl]-3-cyano-4-isopropoxy-benzamide
(4R)-4-[( {3-Cyano-4-[(l -methylethyl)oxy]phenyl} carbonyl)amino]-5- {4-[2-(l ,1-
dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}pentanoicacid
3-Cyano-N- {(1 S)-2- (4-[2-(l ,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-[(2-oxo-

1-imidazolidinyl)methyl]ethyl}-4-[(l -methylethyl)oxy]benzamide
N-((lS)-2-AnMno-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}ethyl)-3-cyano-
4-[(l -methylethyl)oxy]benzamide
N-((l S)-2-(Acetylamino)-1- {[4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}ethyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide
3-Chioro-N-((lS)-2-{[(2R)-2-hydroxypropanoyl]amino}-1-{[4-(8-methylimidazo[1,2-
a]pyridin-2-yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide *
3-Chloro-N-[(lS)-2-[(N,N-dimethylglycyl)amino]-1-({4-[2-(1-hydroxy-1-methylethyl)-1-
rnethyl-1H-imidazo1-4-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
3-Cyano-N^(lS)-2-[(N,N-dimethylglycyl)amino]-1-({4-[2-(1-hydroxy-1-methylethyl)-1-
methyl-1H-imidazo1-4-yl]phenyl} methyl)ethyl]-4-[( 1-methylethyl)oxy]benzamide
3-chloro-N-((S)-4-hydroxy-1-(4-(1-methyl-2-((R)-1-(2-oxopyrrolidin-1-yl)ethyl)-1H-
imidazo1-4-yl)phenyl)butan-2-yl)-4-isopropoxybenzamide
3-cWoro-N-((S)-4-hydroxy-1-(4-(1-methyl-2-((R)-1-(2-oxopyrrolidin-1-yl)ethyl)-1H-
irnidazo1-4-yl)phenyl)butan-2-yl)-4-( 1,1,1-trifluoropropan-2-yloxy)benzamide
3-chloro-N-((S)-4-hydroxy-1-(4-(1-methyl-2-((R)-1-(2-oxooxazolidin-3-yl)ethyl)-1H-
imidazo1-4-yl)phenyl)butan-2-yl)-4-isopropoxybenzamide
3-cWoro-N-((S)-4-hydroxy-1-(4-(1-methyl-2-((R)-1-(2-oxooxazolidin-3-yl)ethyl)-1H-
imidazo1-4-yl)phenyl)butan-2-yl)-4-(l,l,1-trifluoropropan-2-yloxy)benzamide
[001141 Particular compounds include those shown in the following tables:









































[00115] In some embodiments, the chemical entity is a prodrug, such as a phosphate
ester, of one of the compounds listed in Table 1,2, 3,4, 5, or 6. In some embodiments, the
chemical entity is chosen from (3>S)-4-[4-(2-acetyl-1-methyl-li7-imidazo1-4-yl)phenyl]-3-
[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]butyl dihydrogen phosphate; and
(3S)-3-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-[4-(8-
methylimidazo[1,2-a]pyridin-2-yl)phenyl]butyl dihydrogen phosphate.
[00116] The chemical entities described herein can be prepared by following the
procedures set forth, for example, in PCT WO 99/13061, U.S. Patent No. 6,42O,561 and PCT
WO 98/56756, each of which is incorporated herein by reference. The starting materials and
other reactants are commercially available, e.K*i., from Aldrich Chemical Company,
Milwaukee, WI, or may be readily prepared by those skilled in the art using commonly
employed synthetic methodology.
[00117] Unless specified otherwise, the terms "solvent", "inert organic solvent" or

"inert solvent" mean a solvent inert under the conditions of the reaction being described in
conjunction therewith, including, for example, benzene, toluene, acetonitrile, tetrahydrofuran
("THF"), dimethylformamide ("DMF"), chloroform, methylene chloride (or
dichloromethane), diethyl ether, methanol, pyridine and the like. Unless specified to the
contrary, the solvents used in the reactions of the present invention are inert organic solvents.
[00118] In general, esters of carboxylic acids may be prepared by conventional
esterification procedures, for example alkyl esters may be prepared by treating the required
carboxylic acid with the appropriate alkanol, generally under acidic conditions. Likewise,
amides may be prepared using conventional amidation procedures, for example amides may
be prepared by treating an activated carboxylic acid with the appropriate amine. Alternatively,
a lower-alkyl ester such as a methyl ester of the acid may be treated with an amine to provide
the required amide, optionally in presence of trimethylalluminium following the procedure
described in Tetrahedron Lett. 48,4171-4173, (1977). Carboxyl groups may be protected as
alkyl esters, for example methyl esters, which esters may be prepared and removed using
conventional procedures, one convenient method for converting carbomethoxy to carboxyl is
to use aqueous lithium hydroxide.
[00119] The salts and solvates mentioned herein may as required be produced by
methods conventional in the art. For example, if an inventive compound is an acid, a desired
base addition salt can be prepared by treatment of the free acid with an inorganic or organic
base, such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth
metal hydroxide; or the like. Illustrative examples of suitable salts include organic salts
derived from amino acids such as glycine and arginine; ammonia; primary, secondary, and
tertiary amines; such as ethylenediamine, and cyclic amines, such as cyclohexylamine,
piperidine, morpholine, and piperazine; as well as inorganic salts derived from sodium,
calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
[00120] If a compound is a base, a desired acid addition salt may be prepared by any
suitable method known in the art, including treatment of the free base with an inorganic acid,
such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and
the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid,
fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl
acid, such as glucuronic acid or galacturonic acid, alpha-hydroxy acid, such as citric acid or
tartaric acid, amino acid, such as aspartic acid or glutamic acid, aromatic acid, such as
benzoic acid or cinnamic acid, sulfonic acid, such as p-toluenesulfonic acid, methanesulfonic

acid, ethanesulfonic acid, or the like.
[00121] Isolation and purification of the chemical entities and intermediates described
herein can be effected, if desired, by any suitable separation or purification procedure such as,
for example, filtration, extraction, crystallization, column chromatography, thin-layer
chromatography or thick-layer chromatography, or a combination of these procedures.
Specific illustrations of suitable separation and isolation procedures can be had by reference
to the examples hereinbelow. However, other equivalent separation or isolation procedures
can, of course, also be used.

[00122] Referring to Reaction Scheme 1, Step 1, to a solution of a compound of
Formula 103 in an inert solvent such as DCM are added an excess (such as about 1.2
equivalents) of pentafluorotrifluoroacetate and a base such as triethylamine at about 0 °K*I.
The reaction mixture is stirred for about 1 h. The product, a compound of Formula 105, is
isolated and purified.
[00123] Referring to Reaction Scheme 1, Step 2, to a solution of a compound of
Formula 105 in a polar, aprotic solvent are added an excess (such as about 1.2 equivalents) of
a compound of formula R7(K*I)-CH(NHR2)-CH(R5)(R6) and a base such as N, N-
diisopropylethylamine. The reaction is monitored by, for example, LC/MS, to yield a

compound of Formula 107 wherein R7 is NH2, which is isolated and optionally purified.

[00124] Referring to Reaction Scheme 2, to a solution of a compound of Formula 201
in a polar, aprotic solvent such as DMF are added an excess (such as about 1.2 equivalents) of
a compound of Formula 105 and abase such as diisopropylethylamine at room temperature.
The reaction mixture is monitored by, for example, LC/MS. After completion, a primary or
secondary amine in an inert solvent such as THF and HBTU is added to the reaction solution.
The reaction mixture is stirred for about 2 days. The product, a compound of Formula 203
wherein R7 is optionally substituted amino, is isolated and purified.
[00125] In certain embodiments, R6 in a compound of Formula 203 is a halide, alkyl
halide, or aryl halide. This halide can be converted to various other substituents using a
variety of reactions using techniques known in the art and further described in the examples
below.
[00126] In other embodiments, R6 in a compound of Formula 203 is an alkyl or aryl
amine. Again, the amine moiety can be alkylated, acylated, converted to the sulfonamide, and
the like using techniques known in the art and further described below.
[00127] In yet other embodiments, R6 in a compound of Formula 203 is an alkyl
alcohol or an aryl alcohol. The hydroxyl moiety can be converted to the corresponding ether
or ester using techniques known in the art.


[00128] Referring to Reaction Scheme 3, to a solution of a compound of Formula 301
in a polar, aprotic solvent such as DMF added glycinamide hydrochloride, a base such as
diisopropylethylamine, and HBTU. The reaction mixture is stirred for about 15 hours. The
product, a compound of Formula 303, is isolated and purified.

[00129] Referring to Reaction Scheme 4, Step 1, to a stirred solution of a compound of
Formula 401 wherein n is O, 1, or 2 in an inert solvent such as THF at about 0°K*I is added an
excess (such as about 2 equivalents) of LAH (such as a 1.0 M solution in THF). After
stirring for about 2 hours, the product, a compound of Formula 403, is isolated and used
without further purification.
[00130] Referring to Reaction Scheme 4, Step 2, the hydroxyl group is converted to a
protected amino group. If the protecting group is phthamide, it can be made as follows. To a
stirred solution of a compound of Formula 403 in an inert solvent such as THF are added an
excess (such as about 1.1 equivalents) of isoindole-1.3-dione and triphenylphosphine. An
excess (such as about 1.1 equivalents) of DEAD is then added dropwise and the reaction is
stirred for about 30 min. The product, a compound of Formula 405, is isolated and purified.
[00131] Referring to Reaction Scheme 4, Step 3, the Boc protecting group is then

removed to form the corresponding free amine. One of skill in the art will appreciate that this
should be accomplished in such a manner as to leave the other protected amine intact. For
example, to a solution of a compound of Formula 405 in a nonpolar, aprotic solvent such as
DCM is added an acid, such as TFA, at room temperature. The reaction mixture is stirred for
about 20 min. The product, a compound of Formula 407, is isolated and used without further
purification.
(O0132] Referring to Reaction Scheme 4. Step 4, to a solution of a compound of
Formula 407 in an inert solvent such as DMF are added a compound of Formula 105 and a
base such as diisopropylethylamine at room temperature. The reaction mixture is stirred
overnight. The product, a compound of Formula 409, is isolated and purified.
[00133] Referring to Reaction Scheme 4, Step 5, the amine protecting group, PG, is
then removed. If the amine protecting group, PG, is a phthalimide, it can be removed is
follows. To a solution of a compound of Formula 409 in a polar, protic solvent such as
methanol is added an excess (such as about 10 equivalents) of hydrazine hydrate. The
reaction mixture is stirred at about 50 °K*I for about 5 h, and then cooled to room temperature.
The product, a compound of Formula 411, is isolated and optionally, purified; Conditions for
removing other protecting groups are known to those of skill in the art.
[00134] The free amine of a compound of Formula 411 can be acylated, alkylated,
reductively alkylated, or sulfonylated using techniques known to those of skill in the art.



(O0136) In certain compounds of the invention, particular stereoconfiguration may be
preferred for the compound of Formula I-XIII. For the sake of brevity in the remaining
description of the synthesis of compounds of Formula I-XIII, it should be understood that
either single isomer or a mixture of isomers can be employed to give the corresponding
product.
[00137] Particular stereoisomers can be obtained from mixtures using techniques
known in the art. For example, some embodiments, a free amine of Formula 605 is dissolved
in an inert organic solvent (such as IP A) and warmed to 60°K*I. In a separate vessel, a
resolving agent (such as dibenzoyl-D-tartaric acid) is dissolved, such as in the same warm
solvent, and then quickly added (with agitation) to the warm amine solution. The reaction
mixture is left to crystallize by cooling to room temperature over 16 hours under continuing
agitation. The desired isomer is isolated and purified in the usual manner.
[00138] In some embodiments, an optically active amine of Formula 607 can be
prepared from the corresponding aryl aldehyde as shown in Reaction Scheme 5.
[00139) Referring to Reaction Scheme 5, Step 1, a solution of a compound of Formula
601 and an excess of ammonium acetate in nitroethane is heated to about reflux for about 8
hours. The product, a compound of Formula 603, is isolated and optionally purified.
[00140] Referring to Reaction Scheme 5, Step 2, to an about 0 °K*I solution of a
reducing agent such as sodium borohydride in an inert solvent such as tetrahydrofuran is
added an excess (such as about 1.2 equivalents) of borane-tetrahydrofuran complex. The
resulting solution is stirred at room temperature for about 15 minutes. A compound of
Formula 603 in an inert solvent such as tetrahydrofuran is added dropwise, and the resulting
solution is refluxed for about 4 hours; The product, a compound of Formula 605, is isolated
and optionally purified.
[00141] The amine of Formula 605 can be then resolved using techniques known in the
art. For example, a 0 °K*I solution of the amine of Formula 605 in an inert solvent such as
ethyl acetate is saturated with hydrochloric acid (gas). The resulting salt is collected by

filtration and dried in vacuo. L-N-Acetylleucine sodium salt is added slowly to a stirred
solution of the aforementioned salt in water. Crystals form overnight and are removed by
filtration, washed with a small amount of cold water, and recrystallized from absolute
methanol. The crystalline salt of Formula 607a is isolated and optionally purified.
[00142] The mother liquors, which were rich in a compound of Formula 607b, are
combined, made strongly alkaline, and washed three times with diethyl ether. The combined
organic layers are washed with water and dried over sodium sulfate. Hydrochloric acid is
passed through the solution until the precipitation of hydrochloride salt is complete. The same
procedure as above can be applied with D-N-acetylleucine salt. The crystalline compound of
Formula 607b is isolated and optionally purified.

[00143J Referring to Reaction Scheme 6, Step 1, to a solution of a compound of
Formula 701 in a polar protic sob 'ent such as methanol is added an excess (such as about 2
equivalents) of SOCl2. After stirring overnight at ambient temperature, the product, a
compound of Formula 703, is isolated and used without further purification.
(O0144] Referring to Reaction Scheme 6, Step 2, to a solution of a compound of
Formula 703 in a polar, protic solvent such as ethanol is added an excess (such as about 5

equivalents) of N2H4H2O. The reaction mixture is heated to reflux and stirred for about 3 h.
Upon cooling, the product, a compound of Formula 705, is isolated and purified.
[00145] Referring to Reaction Scheme 6, Step 3, to a solution of a compound of
Formula 705 in an inert solvent such as THF is added, an excess (such as about 1.1
equivalents) of carbonyldiimidazole. The reaction mixture is heated to reflux and stirred for
1.5 h. Upon cooling, the product, a compound of Formula 707, is isolated and purified.
[00146] Referring to Reaction Scheme 6, Step 4, to a solution of a compound of
Formula 707 in an inert solvent such as acetonitrile is added an excess (such as about 1.1
equivalents) of R5R6CH-Z wherein Z is a leaving group and a base such as K2CO3. The
reaction mixture is heated to about SO °K*I under microwave irradiation for about 30 min
followed by filtration and concentration in vacuo. The product, a compound of Formula 709,
is isolated and optionally purified.
[00147] Referring to Reaction Scheme 6, Step 5. to a compound of Formula 709 is
added an excess of a primary amine in an inert solvent such as THF. The reaction mixture is
heated to about about 100 °K*I under microwave irradiation for about 4 h. The product, a
compound of Formula 711, is isolated and purified.

[00148] Referring to Reaction Scheme 7. Step 1, to a suspension of zinc powder in a
dry degassed polar, aprotic solvent .such as DMF was activated using techniques known in the
art and further described in the example as follows. 1,2-Dibromoethane was added to the zinc
solution under nitrogen. The mixture was heated using a heat gun for about 30 seconds until
gas starts to evolve from the solution, indicating the activation of the zinc. The mixture was
then allowed to cool to room temperature followed by the addition of TMSC1, and allowed to
stir at room temperature for 30 min. A solution of a compound of Formula 701 in a dry
degassed polar, aprotic solvent such as DMF was added to the zinc solution, and the reaction
mixture was stirred for 1 hour at room temperature. The solution of 702 is used for the next

step.
[00149] Referring to Reaction Scheme 7, Step2, to a solution of 702 was a added a
solution of a compound of Formula 703 (where X1 is Br or I) in a dry degassed polar, aprotic
solvent such as DMF, and a palladium catalyst and a ligand such as Pd2(dba3), and tri-o-
tolylphospine. The reaction mixture was stirred for 3 hour. The product, a compound of
Formula 704 is isolated and purified.
[00150] Once made, the chemical entities of the invention find use in a variety of
applications involving alteration of mitosis. As will be appreciated by those skilled in the art,
mitosis may be altered in a variety of ways; that is, one can affect mitosis either by increasing
or decreasing the activity of a component in the mitotic pathway. Stated differently, mitosis
may be affected (e.K*i., disrupted) by disturbing equilibrium, either by inhibiting or activating
certain components. Similar approaches may be used to alter meiosis.
[00151] In some embodiments, the chemical entities of the invention are used to inhibit
mitotic spindle formation, thus causing prolonged cell cycle arrest in mitosis. By "inhibit" in
this context is meant decreasing or interfering with mitotic spindle formation or causing
mitotic spindle dysfunction. By "mitotic spindle formation" herein is meant organization of
microtubules into bipolar structures by mitotic kinesins. By "mitotic spindle dysfunction"
herein is meant mitotic arrest.
[00152] The chemical entities of the invention bind to, and/or inhibit the activity of,
one or more mitotic kinesin. In some embodiments, the mitotic kinesin is human, although
the chemical entities may be used to bind to or inhibit the activity of mitotic kinesins from
other organisms. In this context, "inhibit" means either increasing or decreasing spindle pole
separation, causing malformation, i.e., splaying, of mitotic spindle poles, or otherwise causing
morphological perturbation of the mitotic spindle. Also included within the definition of a
mitotic kinein for these purposes are variants and/or fragments of such protein and more
particularly, the motor domain of such protein.
[00153] The chemical entities of the invention are used to treat cellular proliferation
diseases. Such disease states which can be treated by the chemical entities provided herein
include, but are not limited to, cancer (further discussed below), autoimmune disease, fungal
disorders, arthritis, graft rejection, inflammatory bowel disease, cellular proliferation induced
after medical procedures, including, but not limited to, surgery, angioplasty, and the like.
Treatment includes inhibiting cellular proliferation. It is appreciated that in some cases the
cells may not be in an abnormal state and still require treatment. Thus, in some

embodiments, the invention herein includes application to cells or individuals afflicted or
subject to impending affliction with any one of these disorders or states.
[00154] The chemical entities, pharmaceutical formulations and methods provided
herein are particularly deemed useful for the treatment of cancer including solid tumors such
as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc. More particularly,
cancers that can be treated include, but are not limited to:
• Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma),
myxoma, rhabdomyoma, fibroma, lipoma and teratoma;
•' Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell,
undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial
adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;
• Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma,
leiomyosarcoma, lymphoma), stomach '(carcinoma, lymphoma, leiomyosarcoma),
pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid
tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's
sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel
(adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma);
• Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma],
lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell
carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma,
teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial
cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma);
• Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma,
angiosarcoma, hepatocellular adenoma, hemangioma;
• Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous
histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell
sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma
(osteocartilaginous exostoses), benign chondroma, chondroblastoma,
chondromyxofibroma, osteoid osteoma and giant cell tumors;
• Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis
deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma,
medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma

multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal
cord neurofibroma, meningioma, glioma, sarcoma);
• Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor
cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous
cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-
Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell
carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina
(clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal
rhabdomyosarcoma], fallopian tubes (carcinoma);
• Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic
leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma,
myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant
lymphoma];
• Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's
sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and
• Adrenal glands: neuroblastoma.
As used herein, treatment of cancer includes treatment of cancerous cells, including cells afflicted .
by any one of the above-identified conditions. Thus, the term "cancerous cell" as provided
herein, includes a cell afflicted by any one of the above identified conditions.
[00155] Another useful aspect of the invention is a kit having at least one chemical
entity described herein and a package insert or other labeling including directions treating a
cellular proliferative disease by administering an effective amount of the at least one chemical
entity. The chemical entity in the kits of the invention is particularly provided as one or more
doses for a course of treatment for a cellular proliferative disease, each dose being a
pharmaceutical formulation including a pharmaceutical excipient and at least one chemical
entity described herein.
[00156] For assay of mitotic kinesin-modulating activity, generally either a mitotic
kinesin or at least one chemical entity described herein is non-diffusably bound to an
insoluble support having isolated sample receiving areas (e.K*i., a microtiter plate, an array,
etc.). The insoluble support may be made of any composition to which the sample can be
bound, is readily separated from soluble material, and is otherwise compatible with the
overall method of screening. The surface of such supports may be solid or porous and of any
convenient shape. Examples of suitable insoluble supports include microtiter plates, arrays,

membranes and beads. These are typically made of glass, plastic (e.K*i., polystyrene),
polysaccharides, nylon or nitrocellulose, Teflon™, etc. Microtiter plates and arrays are
especially convenient because a large number of assays can be carried out simultaneously,
using small amounts of reagents and samples. The particular manner of binding of the
sample is not crucial so long as it is compatible with the reagents and overall methods of the
invention, maintains the activity of the sample and is nondiffusable. Particular methods of
binding include the use of antibodies (which do not sterically block either the ligand binding
site or activation sequence when the protein is bound to the support), direct binding to
"sticky" or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the
surface, etc. Following binding of the sample, excess unbound material is removed by
washing. The sample receiving areas may then be blocked through incubation with bovine
serum albumin (BSA), casein or other innocuous protein or other moiety.
[00157] The chemical entities of the invention may be used on their own to inhibit the
activity of a mitotic kinesin. In some embodiments, at least one chemical entity of the
invention is combined with a mitotic kinesin and the activity of the mitotic kinesin is
assayed. Kinesin activity is known in the art and includes one or more of the following: the
ability to affect ATP hydrolysis; microtubule binding; gliding and
polymerization/depolymerization (effects on microtubule dynamics); binding to other proteins
of the spindle; binding to proteins involved in cel1-cycle control; serving as a substrate to
other enzymes, such 3S kinases or proteases; and specific kinesin cellular activities such as
spindle pole separation.
[00158] Methods of performing motility assays are well knoAvn to those of skill in the
art. (See e.K*i., Hall, et al. (1996), Biophys. J., 71: 3467-3476, Turner et al., 1996, AnaL
Biochem. 242 (l):20-5; Gittes et al., 1996, Biophys. J. 7O(1): 418-29; Shirakawa et al., 1995,
J. Exp. BioL 198: 1809-15; Winkelmann et al., 1995, Biophys. J. 68: 2444-53; Winkelmann
et al., 1995, Biophys. J. 68: 72S.)
[00159] Methods known in the art for determining ATPase hydrolysis activity also can
be used. Suitably, solution based assays are utilized. U.S. Patent 6,410,254, hereby
incorporated by reference in its entirety, describes such assays. Alternatively, conventional
methods are used. For example, Pi release from kinesin (and more particularly, the motor
domain of a mitotic kinesin) can be quantified. In some embodiments, the ATPase hydrolysis
activity assay utilizes 0.3 M PCA (perchloric acid) and malachite green reagent (8.27 mM
sodium molybdate II. 0.33 mM malachite green oxalate, and 0.8 mM Triton X-100). To

perform the assay, 10 µL of the reaction mixture is quenched in 90 µL of cold 0.3 M PCA.
Phosphate standards are used so data can be converted to mM inorganic phosphate released.
When all reactions and standards have been quenched in PCA, 100 µL of malachite green
reagent is added to the relevant wells in e.K*i., a microtiter plate. The mixture is developed for
10-15 minutes and the plate is read at an absorbance of 650 nm. If phosphate standards were
used, absorbance readings can be converted to mM Pi and plotted over time. Additionally,
ATPase assays known in the art include the luciferase assay.
[00160] ATPase activity of kinesin motor domains also can be used to monitor the
effects of agents and are well known to those skilled in the art. In some embodiments
ATPase assays of kinesin are performed in the absence of microtubules. In some
embodiments, the ATPase assays are performed in the presence of microtubules. Different
types of agents can be detected in the above assays. In some embodiments, the effect of an
agent is independent of the concentration of microtubules and ATP. In some embodiments,
the effect of the agents on kinesin ATPase can be decreased by increasing the concentrations
of ATP, microtubules or both. In some embodiments, the effect of the agent is increased by
increasing concentrations of ATP, microtubules or both.
[00161] Chemical entities that inhibit the biochemical activity of a mitotic kinesin in
vitro may then be screened in vivo. In vivo screening methods include assays of cell cycle
distribution, cell viability, or the presence, morphology, activity, distribution, or number of
mitotic spindles. Methods for monitoring cell cycle distribution of a cell population, for
example, by flow cytometry, are well known to those skilled in the art, as are methods for
determining cell viability. See for example, U.S. Patent 6,437,115, hereby incorporated by
reference in its entirety. Microscopic methods for monitoring spindle formation and
malformation are well known to those of skill in the art (see, e.K*i., Whitehead and Rattner
(1998), J. Cell Sci. 111:2551-61; Galgio et al, (1996) J. Cell Biol., 135:399-414), each
incorporated herein by reference in its entirety.
[00162] The chemical entities of the invention inhibit one or more mitotic kinesins.
One measure of inhibition is IC50, defined as the concentration of the chemical entity at which
the activity of the mitotic kinesin is decreased by fifty percent relative to a control. In some
embodiments, the at least one chemical entity has an IC50 of less than about 1 mM. In some
embodiments, the at least one chemical entity has an IC50 of less than about 100 µM. In some
embodiments, the at least one chemical entity has an IC50 of less than about 10 µM. In some

- embodiments, the at least one chemical entity has an IC50 of less than about 1 µM. In some
embodiments, the at least one chemical entity has an IC50 of less than about 100 nM. In some
embodiments, the at least one chemical entity has an IC50 of less than about 10 nM.
Measurement of IC50 is done using an ATPase assay such as described herein.
[00163] Another measure of inhibition is Kj. For chemical entities with IC50'S less than
1 µM, the K*i or Kd is defined as the dissociation rate constant for the interaction of the
compounds described herein, with a mitotic kinesin. In some embodiments, the at least one
chemical entity has a K*i of less than about 100 µM. In some embodiments, the at least one
chemical entity has a K*i of less than about 10 µM. In some embodiments, the at least one
chemical entity has a K*i of less than about 1 µM. In some embodiments, the at least one
chemical entity has a K*i of less than about 100 nM. In some embodiments, the at least one
chemical entity has a K*i of less than about 10 nM.
[00164] The K*i for a chemical entity is determined from the IC50 based on three
assumptions and the Michaelis-Menten equation. First, only one compound molecule binds
to the enzyme and there is no cooperativity. Second, the concentrations of active enzyme and
the compound tested are known (i.e., there are no significant amounts of impurities or
inactive forms in the preparations). Third, the enzymatic rate of the enzyme-inhibitor
complex is zero. The rate (i.e., compound concentration) data are fitted to the equation:

where V is the observed rate, Vmax is the rate of the free enzyme, I0 is the inhibitor
concentration, Eo is the enzyme concentration, and Kd is the dissociation constant of the
enzyme-inhibitor complex.
[00165] Another measure of inhibition is GI50, defined as the concentration of the
chemical entity that results in a decrease in the rate of cell growth by fifty percent. In some
embodiments, the at least one chemical entity has a GI50 of less than about 1 mM. In some
embodiments, the at least one chemical entity has a GI50 of less than about 20 µM. In some
embodiments, the at least one chemical entity has a GI50 of less than about 10 µM. In some
embodiments, the at least one chemical entity has a GI50 of less than about 1 µM. In some
embodiments, the at least one chemical entity has a GI50 of less than about 100 nM. In some
embodiments, the at least one chemical entity has a GI50 of less than about 10 nM.

Measurement of GISO is done using a cell proliferation assay such as described herein.
Chemical entities of this class were found to inhibit cell proliferation.
(O0166] In vitro potency of small molecule inhibitors is determined, for example, by
assaying human ovarian cancer cells (SKOV3) for viability following a 72-hour exposure to a
9-point dilution series of compound. Cell viability is determined by measuring the
absorbance of formazon, a product formed by the bioreduction of MTS/PMS, a commercially
available reagent. Each point on the dose-response curve is calculated as a percent of
untreated control cells at 72 hours minus background absorption (complete cell kill).
[00167] Anti-proliferative compounds that have been successfully applied in the clinic
to treatment of cancer (cancer chemotherapeutics) have GIso's that van' greatly. For example,
in A549 cells, paclitaxel GI50 is 4 nM, doxorubicin is 63 nM, 5-fluorouracil is 1 µM, and
hydroxyurea is 500 uM (data provided by National Cancer Institute, Developmental
Therapeutic Program, http://dtp.nci.nih.gov/). Therefore, compounds that inhibit cellular
proliferation, irrespective of the concentration demonstrating inhibition, have potential
clinical usefulness.
[00168] To employ the chemical entities of the invention in a method of screening for
compounds that bind to a mitotic kinesin, the mitotic kinesin is bound to a support, and a
compound of the invention is added to the assay. Alternatively, the chemical entity of the
invention is bound to the support and a mitotic kinesin is added. Classes of compounds
among which novel binding agents may be sought include specific antibodies, non-natural
binding agents identified in screens of chemical libraries, peptide analogs, etc. Of particular
interest are screening assays for candidate agents that have a low toxicity for human cells. A
wide variety of assays may be used for this purpose, including labeled in vitro protein-protein
binding assays, electrophoretic mobility shift assays, immunoassays for protein binding,
functional assays (phosphorylation assays, etc.) and the like.
[00169] The determination of the binding of the chemical entities of the invention to a
mitotic kinesin may be done in a number of ways. In some embodiments, the chemical entity
is labeled, for example, with a fluorescent or radioactive moiety, and binding is determined
directly. For example, this may be done by attaching all or a portion of a mitotic kinesin to a
solid support, adding a labeled test compound (for example a chemical entity of the invention
in which at least one atom has been replaced by a detectable isotope), washing off excess
reagent, and determining whether the amount of the label is that present on the solid support.
[00170] By "labeled" herein is meant that the compound is either directly or indirectly

labeled with a label which provides a detectable signal, e.K*i., radioisotope, fluorescent tag,
enzyme, antibodies, particles such as magnetic particles, chemiluminescent tag, or specific
binding molecules, etc. Specific binding molecules include pairs, such as biotin and
streptavidin, digoxin and antidigoxin etc. For the specific binding members, the
complementary member would normally be labeled with a molecule which provides for
detection, in accordance with known procedures, as outlined above. The label can directly or
indirectly provide a detectable signal.
(O0171] In some embodiments, only one of the components is labeled. For example,
the kinesin proteins may be labeled at tyrosine positions using 125I, or with fluorophores.
Alternatively, more than one component may be labeled with different labels; using l25I for
the proteins, for example, and a fluorophor for the antimitotic agents.
[00172] The chemical entities of the invention may also be used as competitors to
screen for additional drug candidates. "Candidate agent" or "drug candidate" or grammatical
equivalents as used herein describe any molecule, e.K*i., protein, oligopeptide, small organic
molecule, polysaccharide, polynucleotide, etc., to be tested for bioactivity. They may be
capable of directly or indirectly altering the cellular proliferation phenotype or the expression
of a cellular proliferation sequence, including both nucleic acid sequences and protein
sequences. In other cases, alteration of cellular proliferation protein binding and/or activity is
screened. Screens of this sort may be performed either in the presence or absence of
microtubules. In the case where protein binding or activity is screened, particular
embodiments exclude molecules already known to bind to that particular protein, for
example, polymer structures such as microtubules, and energy sources such as ATP.
Particular embodiments of assays herein include candidate agents which do not bind the
cellular proliferation protein in its endogenous native state termed herein as "exogenous"
agents. In some embodiments, exogenous agents further exclude antibodies to the mitotic
kinesin.
[00173] Candidate agents can encompass numerous chemical classes, though typically
they are small organic compounds having a molecular weight of more than 100 and less than
about 2,500 daltons. Candidate agents comprise functional groups necessary for structural
interaction with proteins, particularly hydrogen bonding and lipophilic binding, and typically
include at least an amine, carbonyl-, hydroxyl-, ether, or carboxyl group, generally at least
two of the functional chemical groups. The candidate agents often comprise cyclical carbon
or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or

more of the above functional groups. Candidate agents are also found among biomolecules
including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives,
structural analogs or combinations thereof.
[00174] Candidate agents are obtained from a wide variety of sources including
libraries of synthetic or natural compounds. For example, numerous means are available for
random and directed synthesis of a wide variety of organic compounds and biomolecules,
including expression of randomized oligonucleotides. Alternatively, libraries of natural
compounds in the form of bacterial, fungal, plant and animal extracts are available or readily
produced. Additionally, natural or synthetically produced libraries and compounds are readily
modified through conventional chemical, physical and biochemical means. Known
pharmacological agents.may be subjected to directed or random chemical modifications, such
as acylation, alkylation, esterification, and/or amidification to produce structural analogs.
[00175] Competitive screening assays may be done by combining a mitotic kinesin and
a drug candidate in a first sample. A second sample comprises at least one chemical entity of
the present invention, a mitotic kinesin and a drug candidate. This may be performed in
either the presence or absence of microtubules. The binding of the drug candidate is
determined for both samples, and a change, or difference in binding between the two samples
indicates the presence of a drug candidate capable of binding to a mitotic kinesin and
potentially inhibiting its activity. That is, if the binding of the drug candidate is different in
the second sample relative to the first sample, the drug candidate is capable of binding to a
mitotic kinesin.
[00176] In some embodiments, the binding of the candidate agent to a mitotic kinesin
is determined through the use of competitive binding assays. In some embodiments, the
competitor is a binding moiety known to bind to the mitotic kinesin, such as an antibody,
peptide, binding partner, ligand, etc. Under certain circumstances, there may be competitive
binding as between the candidate agent and the binding moiety, with the binding moiety
displacing the candidate agent.
[00177] In some embodiments, the candidate agent is labeled. Either the candidate
agent, or the competitor, or both, is added first to the mitotic kinesin for a time sufficient to
allow binding, if present. Incubations may be performed at any temperature which facilitates
optimal activity, typically between 4 and 40°K*I.
[00178] Incubation periods are selected for optimum activity, but may also be
optimized to facilitate rapid high tliroughput screening. Typically between 0.1 and 1 hour

will be sufficient. Excess reagent is generally removed or washed away. The second
component is then added., and the presence or absence of the labeled component is followed,
to indicate binding.
[00179] In some embodiments, the competitor is added first, followed by the candidate
agent. Displacement of the competitor is an indication the candidate agent is binding to the
mitotic kinesin and thus is capable of binding to, and potentially inhibiting, the activity of the
mitotic kinesin. In some embodiments, either component can be labeled.Thus, for example,
if the competitor is labeled, the presence of label in the wash solution indicates displacement
by the agent. Alternatively, if the candidate agent is labeled, the presence of the label on the
support indicates displacement.
[001801 In some embodiments, the candidate agent is added first, with incubation and
washing, followed by the competitor. The absence of binding by the competitor may indicate
the candidate agent is bound to the mitotic kinesin with a higher affinity. Thus, if the
candidate agent is labeled, the presence of the label on the support, coupled with a lack of
competitor binding, may indicate the candidate agent is capable of binding to the mitotic
kinesin.
[00181] Inhibition is tested by screening for candidate agents capable of inhibiting the
activity of a mitotic kinesin comprising the steps of combining a candidate agent with a
mitotic kinesin as above, and determining an alteration in the biological activity of the mitotic
kinesin. Thus, in some embodiments, the candidate agent should both bind to the mitotic
kinesin (although this may not be necessary), and alter its biological or biochemical activity
as defined herein. The methods include both in vitro screening methods and in vivo
screening of cells for alterations in cell cycle distribution, cell viability, or for the presence,
morpohology, activity, distribution, or amount of mitotic spindles, as are generally outlined
above.
[00182] Alternatively, differential screening may be used to identify drug candidates
that bind to the native mitotic kinesin but cannot bind to a modified mitotic kinesin.
[00183] Positive controls and negative controls may be used in the assays. Suitably all
control and test samples are performed in at least triplicate to obtain statistically significant
results. Incubation of all samples is for a time sufficient for the binding of the agent to the
protein. Following incubation, all samples are washed free of non-specifically bound material
and the amount of bound, generally labeled agent determined. For example, where a
radiolabel is employed, the samples may be counted in a scintillation counter to determine the

amount of bound compound.
[00184] A variety of other reagents may be included in the screening assays. These
include reagents like salts, neutral proteins, e.K*i., albumin, detergents, etc which may be used
to facilitate optimal protein-protein binding and/or reduce non-specific or background
interactions. Also reagents that otherwise improve the efficiency of the assay, such as
protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may be used. The mixture
of components may be added in any order that provides for the requisite binding.
[00185] Accordingly, the chemical entities of the invention are administered to cells.
By "administered" herein is meant administration of a therapeutically effective dose of at least
one chemical entity of the invention to a cell either in cell culture or in a patient. By
"therapeutically effective dose" herein is meant a dose that produces the effects for which it is
administered. The exact dose will depend on the purpose of the treatment, and will be
ascertainable by one skilled in the art using known techniques. As is known in the art,
adjustments for systemic versus localized delivery, age, body weight, general health, sex, diet,
time of administration, drug interaction and the severity of the condition may be necessary,
and will be ascertainable with routine experimentation by those skilled in the art. By "cells"
herein is meant any cell in which mitosis or meiosis can be altered.
[00186] A "patient" for the purposes of the present invention includes both humans and
other animals, particularly mammals, and other organisms. Thus the methods are applicable
to both human therapy and veterinary applications. In some embodiments, the patient is a
mammal, and more particularly, the patient is human.
[00187] Chemical entities of the invention having the desired pharmacological activity
may be administered, in some embodiments, as a pharmaceutically acceptable composition
comprising an pharmaceutical excipient, to a patient, as described herein. Depending upon
the manner of introduction, the chemical entities may be formulated in a variety of ways as
discussed below. The concentration of the at least one chemical entity in the formulation may
vary from about 0.1-100 wt.%.
[00188] The agents may be administered alone or in combination with other
treatments, i.e., radiation, or other chemotherapeutic agents such as the taxane class of agents
that appear to act on microtubule formation or the camptothecin class of topoisomerase I
inhibitors. When used, other chemotherapeutic agents may be administered before,
concurrently, or after administration of at least one chemical entity of the present invention.
In one aspect of the invention, at least one chemical entity of the present invention is K*i-

administered with one or more other chemotherapeutic agents. By "K*i-administer" it is meant
that the at least one chemical entity is administered to a patient such that the at least one
chemical entity as well as the K*i-administered compound may be found in the patient's
bloodstream at the same time, regardless when the compounds are actually administered,
including simultaneously.
[00189] The administration of the chemical entities of the present invention can be
done in a variety of ways, including, but not limited to, orally, subcutaneSusly, intravenously,
intranasally, transdermally, intraperitoneal!}', intramuscularly, intrapulmonary, vaginally,
rectally, or intraocularly. In some instances, for example, in the treatment of wounds and
inflammation, the compound or composition may be directly applied as a solution or spray.
[00190] Pharmaceutical dosage forms include at least one chemical entity described
herein and one or more pharmaceutical excipients. As is known in the art, pharmaceutical
excipients are secondary ingredients which function to enable or enhance the delivery of a
drug or medicine in a variety of dosage forms (e.K*i.: oral fonns such as tablets, capsules, and
liquids; topical forms such as dermal, opthalmic, and otic forms; suppositories; injectables;
respiratory forms and the like). Pharmaceutical excipients include inert or inactive
ingredients, synergists or chemicals that substantively contribute to the medicinal effects of
the active ingredient. For example, pharmaceutical excipients may function to improve flow
characteristics, product uniformity, stability, taste, or appearance, to ease handling and
administration of dose, for convenience of use, or to control bioavailability. While
pharmaceutical excipients are commonly described as being inert or inactive, it is appreciated
in the art that there is a relationship between the properties of the pharmaceutical excipients
and the dosage forms containing them.
[00191] Pharmaceutical excipients suitable for use as earners or diluents are well
known in the art, and may be used in a variety of formulations. See, e.K*i., Remington's
Pharmaceutical Sciences, 18th Edition, A. R. Gennaro, Editor, Mack Publishing Company
(199O); Remington: The Science and Practice of Pharmacy, 20th Edition, A. R. Gennaro,
Editor, Lippincott Williams & Wilkins (200O); Handbook of Pharmaceutical Excipients, 3rd
Edition, A. H. Kibbe, Editor, American Pharmaceutical Association, and Pharmaceutical
Press (200O); and Handbook of Pharmaceutical Additives, compiled by Michael and Irene
Ash,Gower (1995), each of which is incorporated herein by reference for all purposes.
[00192] Oral solid dosage forms such as tablets will typically comprise one or more
pharmaceutical excipients, which may for example help impart satisfactory processing and

compression characteristics, or provide additional desirable physical characteristics to the
tablet. Such pharmaceutical excipients may be selected from diluents, binders, glidants,
lubricants, disintegrants, colors, flavors, sweetening agents, polymers, waxes or other
solubility-retarding materials.
[00193] Compositions for intravenous administration will generally comprise
intravenous fluids, i.e., sterile solutions of simple chemicals such as sugars, amino acids or
electrolytes, which can be easily carried by the circulatory system and assimilated. Such
fluids are prepared with water for injection USP.
(O0194] Dosage forms for parenteral administration will generally comprise fluids,
particularly intravenous fluids, i.e., sterile solutions of simple chemicals such as sugars,,
amino acids or electrolytes, which can be easily earned by the circulatory system and
assimilated. Such fluids are typically prepared with water for injection USP. Fluids used
commonly for intravenous (TV) use are disclosed in Remington, The Science and Practice of
Pharmacy [full citation previously provided], and include:
• alcohol, e.K*i., 5% alcohol (e.K*i., in dextrose and water ("D/W") or D/W in
normal saline solution ("NSS"), including in 5% dextrose and water ("D5/W"),
orD5/WinNSS);
• synthetic amino acid such as Aminosyn, FreAmine, Travasol, e.K*i., 3.5 or 7;
8.5; 3.5, 5.5 or 8.5 % respectively;
• ammonium chloride e.K*i., 2.14%;
• dextran 4O, in NSS e.K*i., 10% or in D5/W e.K*i., 10%;
• dextran 7O, in NSS e.K*i., 6% or in D5/W e.K*i., 6%;
• dextrose (glucose, D5AV) e.K*i., 2.5-50%;
• dextrose and sodium chloride e.K*i., 5-20% dextrose and 0.22-0.9% NaCl;
• lactated Ringer's (Hr rtmann's) e.K*i., NaCl 0.6%, KC1 0.03%. CaCl2 0.02%;
• lactate 0.3%;
• mannitol e.K*i., 5%, K*i ptionally in combination with dextrose e.K*i., 10% or NaCl
e.K*i., 15 or 20%;
• multiple electrolyte solutions with varying combinations of electrolytes,
dextrose, fructose, invert sugar Ringer's e.K*i., NaCl 0.86%, KC1 0.03%, CaCl2
0.033%;
• sodium bicarbonat • e.K*i., 5%;

• sodium chloride e.K*i., 0.45, 0.9, 3, or 5%;
• sodium lactate e.K*i., 1/6 M; and
• sterile water for injection
The pH of such IV fluids may vary, and will typically be from 3.5 to 8 as known in the art.
[00195] The chemical entityies of the invention can be administered alone or in
combination with other treatments, i.e., radiation, or other therapeutic agents, such as the
taxane class of agents that appear to act on microtubule formation or the*amptothecin class
of topoisomerase I inhibitors. When so-used, other therapeutic agents can be administered
before, concurrently (whether in separate dosage forms or in a combined dosage form), or
after administration of an active agent of the present invention.
[00196] The following examples serve to more fully describe the manner of using the
above-described invention, as well as to set forth the best modes contemplated for carrying
out various aspects of the invention. It is understood that these examples in no way serve to
limit the true scope of this invention, but rather are presented for illustrative purposes. All
publications, including but not limited to patents and patent applications, cited in this
specification are herein incorporated by reference as if each individual publication were
specifically and individually indicated to be incorporated by reference herein as though fully
set forth.

EXAMPLES
[00197] The following examples serve to more fully describe the manner of using the
above-described invention, as well as to set forth the best modes contemplated for carrying
out various aspects of the invention. It is understood that these examples in no way serve to
limit the true scope of this invention, but rather are presented for illustrative purposes. All
references cited herein are incorporated by reference in their entirety.

[00198J To a solution of 4-isopropoxylbenzoic acid 1 (25 K*i, 140 mmol) in DMF
(150mL) was added NCS (24 K*i, 182 mmol). The reaction mixture was stirred overnight. H?O (500mL) was added to the reaction mixture. The precipitate was collected and washed with
water, and dried in vacuo to give 2 (26.4 K*i, 88 %) as a white solid, which was used in the next
step without further purification. LRMS (M+H4) m/z 213.0.
[00199] To a solution of 2 (20 K*i, 93 mmol) in DCM were added
pentafluorotrifluoroacetate (20 mL, 112 mmol) and triethylamine (17 mL, 112 mmol) at 0 °K*I.
The reaction mixture was stirred for 1 h. The solution was concentrated and the mixture
purified by flash column chromatography (100% DCM) to give 3 (35 K*i, quant.) as a white

solid.
[00200] To a solution of 3 in DMF (0.2 M) were added amino acid (1.2 equiv.) and N,
N-diisopropylethylamine (3 equiv.). The reaction was monitored by LC/MS. After
completion, methylamine (2 M in THF, 1.5 equiv.) and HBTU (1.5 equiv.) were added to the
reaction solution. The reaction mixture was stirred for 4 h. The product was purified by either
HPLC or flash column chromatography to give 4.

[00201] To a solution of H-Phe(4-Br)-OH (2, 2.5 K*i, 10 mmol) in DMF (20 mL) were
added 3 (4.7 K*i, 12 mmol) and diisopropylethylamine (5.4 mL, 30 mmol) at room temperature.
The reaction mixture was monitored by LC/MS. After completion, methylamine (2M in
THF, 7.7 mL. 15 mmol) and HBTU (5.8 K*i, 15 mmol) were added to the reaction solution.
The reaction mixture was stirred for 2 days. The mixture was filtered, and the filtrate was
purified on RP-HPLC using a mixture of acetonitrile and H2O to give 4 (2.3 K*i, 50%). LRMS
(M+H+) m/z 455.0.
[00202] To a suspension of 4 (71 mg, 0.16 mmol) in dioxane (1 mL) were added
piperazine (16mg, 0.19 mmol), palladium (II) acetate (4 mg, 0.016 mmol),
dicyclohexylphosphino-2'-(AyV,-dimethylamino)-biphenyl (6 mg, 0.016 mmol), and cesium
carbonate (104 mg, 0.32mmol). The resulting mixture was stirred for 36 hours at 110 °K*I. The
reaction mixture was diluted with EtOAc. The organic layer was washed with saturated

i^aHCO3 (20mL) and brine, dried over Na2SO4, and concentrated. The residue was purified
on RP-HPLC using a mixture of acetonitrile and H2O to give la (6 mg, 8%). LRMS (M+H*)
m/z 459.2.

[00203] To a solution of H-Tyr-NH2 HC1 (2, 830 mg, 3.8 mmol) in DMF (5 mL) were
added 3 (1.8 K*i. 4.5 mmol) and diisopropylethylamine (3.4 mL, 19 mmol) at room
temperature. The reaction was stirred for 20 hours and filtered after adding water. The white
precipitate was recrystallized in DCM and methanol to give 4 as white crystals (1.120 K*i,
78%). LRMS (M+H+) m/z 377.1.
[00204] To a solution of 4 (50 mg, 0.13 mmol) in DMF (1 mL) were added (s)-(+)-3-
bromo-2-methyl-1-propanol (0.083 mL, 0.8 mmol) and potassium carbonate (110 mg, 0.8
mmol). The resulting mixture was stirred for 15 hours at 50 °K*I. The mixture was filtered,
and the filtrate was purified on RP-HPLC using a mixture of acetonitrile and H2O to give lb
(30 mg, 51 %). LRMS (M+H+) m/z 449.1.
Example 4


[00205] To a solution of H-Tyr-OBut (2, 1.9 K*i, 8 mmol) in DMF (50 mL) were added 3
(2.4 K*i, 6.2 mmol) and diisopropylethylamine (3.3 mL, 19 mmol) at room temperature. The
reaction was stirred for 2 hours. The resulting solution was diluted with EtOAc (200 mL) and
washed with saturated NaHCCb (50 mL). The organic layer was separated, washed with
brine, dried over Na2SO.|, and concentrated to give a yellow solid. To a solution of the yellow
solid in dichloromethane (10 mL) was added tfifluoroacetic acid (30 mL). The mixture was
stirred at room temperature for 12 hours and then concentrated under reduced pressure. The
residue was dried in vacuo to give 4 (3.1 K*i), which was used in the next step without further
purification. LRMS (M-H+) m/z 376.1.
[00206] To a solution of 4 (3.1 K*i, 8 mmol) in DMF (25 mL) was added glycinamide
hydrochloride (1.1 K*i, 9.6 mmol), diisopropylethylamine (7 mL, 40 mmol), and HBTU (3.6 K*i,
9.6 mmol). The reaction mixture was stirred for 15 hours, after which solution was diluted
with ethyl acetate and washed with saturated NaHCOa. The organic layer was separated,

washed with brine, dried over Na2SO4, and concentrated. The resulting crude was purified on
RP-HPLC using a mixture of acetonitrile and H2O to give 5 (38 mg, 35%). LRMS (M+rf)
m/z 434.1.
[00207] To a solution of 5 (100 mg, 0.23 mmol) in DMF (1 mL) were added
cyclopropylmethyl bromide (0.1S mL, 1.84 mmol) and potassium carbonate (317 mg, 2.3
mmol). The resulting mixture was stirred for 10 hours at 80 °K*I. The mixture was filtered,
and the filtrate was purified on RP-HPLC using a mixture of acetonitrile and H2O to give lc
(36 mg, 34%). LRMS (M+H*) m/z 488.1.

[00208] To a solution of 4 (80 mg, 0.2 mmol) in DMF (1 mL) were added (±)-3-
bromo-1-phenyl-2-pyrrolidinone (250 mg, 1 mmol) and potassium carbonate (235 mg, 1.7
mmol). The resulting mixture was stirred for 10 hours at 110 °K*I. The mixture was filtered,
and the filtrate was purified on RP-HPLC using a mixture of acetonitrile and H2O to give 5
(38 mg, 35%). LRMS (M+H4) m/z 536.1.

[00209] To a solution of 4 (70 mg, 0.19 mmol) in DMF (1 mL) were added 3-
(hydroxymethyl) pyridine (0.023 mL, 0.23 mmol), triphenylphosphine (100 mg, 0.38 mmol),
and diisopropylazodicarboxylate (0.055 mL, 0.38 mmol). The resulting mixture was stirred
for 20 hours at room temperature. The reaction solution was concentrated and purified via

flash column chromatography using a mixture of ethyl acetate and hexane as eluent to give If
(22 mg, 25%). LRMS (M+H1) m/z 468.2.

J00210] To a solution of 4 (50 mg, 0.12 mmol) in toluene (2 mL) was added 2-
(fluorophenyl) boronic acid (20 mg, 0.14 mmol), tetrakis(triphenylphosphine)palladium(O)
(42 mg. 0.04 mmol), and 2M sodium carbonate (0.18 mL, 0.36 mrriol). The reaction mixture
was stirred for 90 min at 100 °K*I. The resulting solution was purified on RP-HPLC using a
mixture of acetonitrile and H2O to give 5 (22 mg, 40%). LRMS (M+H+) m/z 469.2.

[00211] To a solution of 4 (45 mg, 0.1 mmol) in DMF (1 mL) were added
bis(pinacolate) diboron (30 mg, 0.12 mmol), l,l'-bis(diphenylphosphino)ferrocene-
palladium(II) dichloride dichloromethane complex (17 mg, 0.02 mmol), and potassium
acetate (39 mg, 0.4 mmol). The reaction mixture was stirred for 1 hour at 80 °K*I. The
resulting mixture was added 4-bromo-3,5-dimethylisoxazole (35 mg, 0.2 mmol) and 2M
sodium carbonate (0.4 mL, 0.8 mmol). The mixture was stirred at 80 °K*I for 90 min. The
resulting residue was filtered, and the filtrate was purified on RP-HPLC using a mixture of
acetonitrile and H2O to give lh (23 mg, 49%). LRMS (M+H*) m/z 470.1.


[00212] To a solution of 4 (62 mg, 0.14 mmol) in DMF (1 mL) was added
bis(pinacolate) diboron (42mg, 0.16 mmol), l,l'-bis(diphenylphosphino)ferrocene-
palladium(II) dichloride dichloromethane complex (34 mg, 0.04 mmol), and potassium
acetate (54 mg, 0.55 mmol). The reaction mixture was stirred for 1 hour at 80 °K*I. The
resulting mixture was added N-methyl-2-bromobenzimidazole (5S mg, 0.27 mmol) and 2M
sodium carbonate (0.54 mL, 1.0S mmol). The mixture was stirred at 80 °K*I for 60 min. The
resulting solution was diluted with ethylacetate (20mL), and washed with saturated NaHCCb
(20 mL). The organic layer was separated, washed with brine, dried over NazSO,}, and
concentrated. The resulting residue was purified on RP-HPLC using a mixture of acetonitrile
and H2O to give li (44 mg, 64%). LRMS (M+H4) m/z 505.1.

[00213] To a solution of 4 (50 mg, 0.13 mmol) in DMF (1 mL) were added
bis(pinacolate) diboron (34 mg, 0.13 mmol), l,r-bis(diphenyiphosphino)ferrocene-
palladium(II) dichloride dichloromethane complex (27 mg, 0.03 mmol), and potassium
acetate (43 mg, 0.44 mmol). The reaction mixture was stirred for 1 hour at 80 °K*I. To the
resulting mixture was added 3-bromothiophene-2-carbonitrile (41 mg, 0.22 mmol) and 2M
sodium carbonate (0.44 mL, 0.88 mmol). The mixture was stirred at 80 °K*I for 90 min. The
resulting residue was filtered, and the filtrate was purified on RP-HPLC using a mixture of

acetonitrile and H2O to give 5 (20 mg, 37%). LRMS (M+H4) m/z 482.1.

[00214] To a solution of 4 (85 mg, 0.2 mmol) in toluene (2 mL) was added 1-t-butyl-
l,3-dihydro-imidazo1-2-one (53 mg, 0.4 mmol), copper(I) iodide (18 mg. 0.1 mmol), trans-
1,2-diamino-cyclohexane (11 mg, 0.1 mmol), and cesium carbonate (124 mg. 0.4 mmol).
The reaction mixture was stirred for 4 hours at 100 °K*I. The mixture was filtered, and the
filtrate was concentrated. The resulting residue was filtered, and the filtrate was purified on
RP-HPLC using a mixture of acetonitrile and H2O to give Ik (27 mg, 28%). LRMS (M+H+)
m/z 513.1.

[00215J To a solution of 4 (100 mg. 0.2 mmol) in dioxane (2 mL) was added
benzimidazole (39 mg, 0.33 mmol), copper (I) iodide (8.4 mg, 0.04 mmol), 1,10-
phenanthroline (16 mg. 0.1 mmol), and cesium carbonate (144 mg, 0.44 mmol). The reaction
mixture was stirred for 15 hours at 100 °K*I. The mixture was filtered, and the filtrate was
concentrated. The resulting residue was filtered, and the filtrate was purified on RP-HPLC
using a mixture of acetonitrile and H2O to give 11 (5.7 mg, 6 %). LRMS (M+H4) m/z 491.1.
Example 13


[00216] To a solution of 4 (60 mg, 0.16 mmol) in toluene (1 mL) v&s added 2-
chlorobenzimidazole (50 mg, 0.32 mmol). copper(I) iodide (9 mg, 0.05), and cesium
carbonate (105 mg, 0.32 mmol). The reaction mixture was stirred for 28 hours at 110 °K*I.
The mixture was filtered, and the filtrate was purified on RP-HPLC using a mixture of
acetonitrile and H2O to give lm (8 mg, 10%). LRMS (M+H+) m/z 493.0.

[00217] To a solution of 4 (50 mg, 0.1 mmol) in toluene (1 mL) was added phenol (21
mg, 0.2 mmol), copper(I) iodide (6 mg, 0.03 mmol), and cesium carbonate (72 mg, 0.2
mmol). The reaction mixture was stirred for 5 hours at 115 °K*I. The mixture was filtered, and
the filtrate was concentrated. The resulting residue was purified via flash column
chromatography using a mixture of ethyl acetate and hexane as eluent to give In (23 mg,
45%). LRMS (M+H+) m/z 467.1.



[00218] To a solution of 1 (2.3 K*i, 12.6 mmol) in DMF (30 mL) were added 2 (4.0 K*i,
10.5 mmol) and N, JV-diisopropylethylamine (5.2 mL, 30 mmol). The reaction was monitored
by LC/MS. The resulting solution was used in the next step without further purification.
■LRMS (M+H4) wfe 377.1.
[00219] To a solution of crude 3 in DMF (6 mL, - 2mmol) were added glycinamide
HC1 (330 mg, 3 mmol), HBTU (1.14 K*i, 3 mmol) and K*I TV-diisopropylethylamine (522 µL, 3
mmol). The reaction was stilted overnight. The resulting crude product was purified via RP-
HPLC using a mixture of acetonitrile and H2O to give 4 (600 mg, 69% from 2). LRMS
(M+lt) m/z 433.1.

[00220] To a solution of crude 3 in DMF (15 mL, ~ 5.25 mmol) were added
methylamine (2 M in THF, 4 mL, 8 mmol), and HBTU (3 K*i, 7.9 mmol). The reaction was

stirred overnight. The mixture was diluted with ethyl acetate (200 mL). The organic layer was
washed with H2O, brine, dried over sodium sulfate, and concentrated. The resulting crude 5
was used in the next step without further purification. LRMS (M+H*) m/z 390.1.
[00221] To a solution of crude 5 (75 mg; ~ 0.2 mmol) in DCM (2 mL) were added
benzoyl chloride (23 µL, 0.2 mmol) and N, TV-diisopropylethylamine (35 µL, 0.2 mmol). The
reaction mixture was stirred overnight. The solution was concentrated and purified on RP-
HPLC using a mixture ofacetonitrile and H2O to give 6 (36 mg, 40 % from 2). LRMS
(M+H+) m/z 494.1

[00222] To a solution of 5 (75 mg, ~ 0.2 mmol) in DCM (2 mL) was added phenyl
isocyanate (26 µL, 0.24 mmol). The reaction mixture was stirred overnight. The resulting
solution was concentrated and purified on RP-HPLC using a mixture ofacetonitrile and H2O
to give 7 (40 mg, 39 % from 2). LRMS (M+H+) m/z 509.1.

[00223] To a solution of 5 (75 mg, 0.19 mmol) in DCM (3 mL) were added isobutyl
chloroformate (38 µL, 0.29 mmol) and TV, jV-diisopropylethylamine (50 µL, 0.29 mmol). The
reaction mixture was stirred overnight. The resulting solution was concentrated and purified
on RP-HPLC using a mixture of acetonitrile and H2O to give 8 (45 mg, 48%). LRMS (M+H4)
wt/z 490.1.


[00224] To a solution'of 5 (75 mg, 0.19 mmol) in DCM (5 mL) were added
dimethylsulfamoyl chloride (30 µL, 0.29 mmol), N, N-diisopropylethylamine (50 µL, 0.29
mmol) and DMAP (50 mg, 0.4 mmol). The reaction mixture was stirred overnight. The
reaction mixture was then heated to 30 °K*I and stirring continued for 8 h. The resulting
solution was concentrated and purified on RP-HPLC using a mixture of acetonitrile and HiO
to give 9 (30 mg, 32%). LRMS (M+H+) m/z A91.1.



100225] To a solution of H-Phe(4-K*I/Bu)-OH (5.S K*i, 22 mmol) in ethyl acetate (60
mL) and water (20 mL) were added platinum (TV) oxide (400 mg, 1.8 mmol) and acetic acid

(50 mL). The reaction mixture was stirred under a stream of H2 (60psi) for 20 lirs. The
catalyst was removed by filtration through a PTFE (0.45 urn) filter and the solvent evaporated
to give 2 (5.9 K*i), which was used in the next step without further purification, LRMS (M+H"1)
m/z 212.1.
[00226J To a solution of 2 (6.9 K*i, 18 mmol) in DMF (30 mL) were added 3 (5.9 K*i, 21.8
mmol) and N, jV-diisopropylethylamine (9.5 mL, 54.3 mmol). The reaction was monitored by
LC/MS. After completion, 2M methylamine in THF (13.6 mL, 27 mmol* HOBt (4 K*i, 27
mmol), and HBTU (10 K*i, 27 mmol) ivere added to the reaction solution. The reaction was
stirred for 4 hours. The mixture was diluted with ethyl acetate (60 mL) and washed with
saturated NaHCC>3 (20 mL). The organic layer was separated, and the aqueous phase was
extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with
brine, dried over sodium sulfate, and concentrated. The resulting crude product was purified
via flash column chromatography using a mixture of ethyl acetate and hexane as eluent to
give 4 (cis isomer 808 mg, 1.68 mmol, trans isomer 300mg, 0.63 mmol). LRMS (M+H+) m/z
481.1.
[00227J To a solution of 4 (290 mg, 0.6 mmol) in dichloromethane (20 mL) was added
trifluoroacetic acid (5 mL). The resulting solution was stirred at room temperature for 1 hour
and then concentrated under reduced pressure. The residue was purified via flash column
chromatography using a mixture of 99% ethyl acetate and 1% acetic acid as eluent to give 5
as a white solid (140 mg, 55%). LRMS (M+H+) m/z 425.1.
[00228] To a solution of 5 (330 mg: 0.7 mmol) in DMSO (5 mL) were added
ammonium chloride (83 mg, 1.5 mmol), diisopropylethylamine (0.27 mL, 1.5 mmol), and
HBTU (580 mg. 1.5 mmol). The resulting solution was stirred at room temperature for 15
hours. Additional ammonium chloride (37 mg, 0.7 mmol), diisopropylethylamine (0.12 mL,
0.7 mmol), and HBTU (266 mg, 0.7 mmol) were added. Stirring was continued for additional
5 hours, and the mixture was diluted with ethyl acetate (50 mL) and washed with saturated
NaHCCb (20 mL). The organic layer was separated, and the aqueous phase was extracted with
ethyl acetate (2 x 50 mL). The combined organic layers were washed with brine, dried over
sodium sulfate, and concentrated to yield slightly yellow crude. The residue was purified on
RP-HPLC using a mixture of acetonitrile and H2O to give 6 (128 mg, 30%). LRMS (M+H*)
m/z 424.1.
[00229] To a solution of 6 (94 mg, 0.2 mmol) in DMF (2 mL) was added cyanuric
chloride (45 mg, 0.2 mmol) at 0 °K*I, and the reaction micxture was stirred under nitrogen.

After 1 hour, the reaction solution was concentrated to give 7 (79 mg), which was used in the
next step without further purification. LRMS (M+Hf) m/z 406.1.
[00230] To a solution of 7 (17 mg, 0.04 mmol) in methanol (2 mL) was stirred under a
stream of HC1 for 15 min. A stream of nitrogen was then bubbled through the reaction
mixture. After 1 hour, the reaction solution was concentrated to give 8, which was used in
the next step without further purification. LRMS (M+H+) m/z 438.1.
[00231] To a solution of crude 8 (17 mg, -0.04 mmol) in acetic acid (2 mL) was added
o-phenylenediamine(50 mg, 0.46 mmol), and the resulting solution was stirred at SO °K*I for 1
h. The reaction mixture was concentrated and purified via preparative thin layer
chromatography using 5% methanol in dichloromethane as eluent to give a white solid.: The
solid was purified on RP-HPLC using a mixture of acetonitrile and H2O to give 9 (9 mg,
45%). LRMS (M+H+) m/z 497.1.

[00232J To a solution of 5 (50 mg, 0.12 mmol) in DMF (1 mL) were added
benzylamine (16 mg, 0.14 mmol) and HATU (57 mg, 0.14 mmol). The reaction mixture was
stirred at room temperature for 3 hours. The resulting solution was filtered and the filtrate
was purified on RP-HPLC using a mixture of acetonitrile and H2O to give 6 (16 mg, 26%).
LRMS (M+H+)77i


[00233] To a solution of H-Phe(4-NHBoc)-OH (4.8 K*i, 17.1 mmol) in ethanol (60 mL),
methanol (20 mL), acetic acid (60 mL), and water (30 mL) was added platinum (IV) oxide
(360 mg, 1.6 mmol). The reaction mixture was stirred under a stream of H2 (45 psi) for 20
hrs. The catalyst was removed by filtration through a PTFE (0.45 um) filter and the solvent
evaporated to give 2 (5 K*i), which was used in the next step without further purification.
LRMS(M+H+)m/z2S7.1.
[00234] To a solution of crude 2 (3.2 K*i, 11.2 mmol) in DMF (20 mL) were added 3
(3.8 K*i, 10 mmol) and N, jV-diisopropylethylamine (5.2 mL, 30 mmol). The reaction was
monitored by LC/MS. After completion, methylamine (2 M in THF, 7.5 mL, 15 mmol), and
HBTU (5.7 K*i, 15 mmol) were added to the reaction solution. The reaction was stirred
overnight. The mixture was diluted with ethyl acetate (60 mL) and washed with saturated
NaHCC>3 (20 mL). The organic layer was separated, and the aqueous phase was extracted with
ethyl acetate (2 x 50 mL). The combined organic layers were washed with brine, dried over

sodium sulfate, and concentrated. The resulting crude was purified via RP-HPLC using a
mixture of acetonitrile and H2O to give 4 (1.0 K*i, 18% from 1). LRMS (M+H4) m/z 496.1.
[00235] To a solution of 4 (1.0 K*i, 2.0 mmol) in dichloromethane (25 mL) was added
trifluoroacetic acid (8 mL). The resulting solution was stirred at room temperature for 4 hous
and then concentrated under reduced pressure. The residue was purified via RP-HPLC using
a mixture of acetonitrile and H2O to give 5 (820 mg, 79%). LRMS (M+H+) m/z 396.1.
[00236] To a solution of 5 (75 mg, 0.16 mmol) in THF (3 mL) were added 4-
fluorobenzoyl chloride (2S µL, 0.23 mmol) and N, iV-diisopropylethylamine (100 µL, 0.57
mmol). The reaction mixture was stirred overnight. The resulting solution was concentrated
and purified on RP-HPLC using a mixture of acetonitrile and H2O to give 6 (65 mg, 7S%).
LRMS (M+H+) 77'^ 518.1.
1 rA-V- — 1 rV^V^- •
XQX^) H O DIEA.THF A0iy H *
CI CI
7
3
1 rA-V- — 1 rV^V^- •
XQX^) H O DIEA.THF A0iy H *
CI CI
7
[00237] To a solution of 5 (75 mg, 0.16 mmol) in THF (3 mL) were added isobutyl
chloroformate (30 µL, 0.23 mmol) and N, N-diisopropylethylamine (100 uL. 0.57 mmol). The
reaction mixture was stirred overnight. The resulting solution was concentrated and purified
on RP-HPLC using a mixture of acetonitrile and H2O to give 7 (62 mg, 78%). LRMS (M+H4)
m/z 496.1.

[00238] To a solution of 5 (75 mg, 0.16 mmol) in THF (3 mL) was added terr-butyl
isocyanate (26 µL, 0.23 mmol) and N, yV-diisopropylethylamine (100 µL, 0.57 mmol). The
reaction mixture was stirred overnight. The resulting solution was concentrated and purified
on RP-HPLC using a mixture of acetonitrile and H2O to give 7 (55 mg, 69%). LRMS (M+H+)
m/z 495.1.


[00239] To a solution of Boc-L-serine-beta-lactone 28 (200mg, 1.07 mmol) in
acetonitrile (5 mL) was added 29 (154 mg, 1.07 mmol). The mixture was stirred at 56 "K*I
overnight. Concentrated under reduced pressure to give 30.
[00240] Crude 30 was redissolved in DMF (1 mL) treated with methylamine (2 M in
THF) (0.54 mL, 1.08 mmol) and HBTU (404 mg, 1.07 mmol). The mixture was stirred for 1
hour, after which it was filtered, and the filtrate purified on reverse phase HPLC (CI 8) using
a mixture of acetonitrile and H2O to give 31 (50.0 K*i, 14%).
[00241] To a solution of 31 (50.0 K*i, 0.145 mmol) in DCM (5 mL) was added TFA (5
mL) at room temperature. The reaction mixture was stirred for 20 min. The solvents were
evaporated under reduced pressure and the residue re-suspended in DMF (100 mL) followed
by the addition of 6 (66.3 mg, 0.174 mmol) and diisopropylethylamine (51 µL, 0.290 mmol)
at room temperature. The reaction mixture was stirred for 1 hour, then concentrated under
reduced pressure, and the residue purified on a flash silica gel soumn (hexane: EtOAc, 1:1) to
give 32 (50.0 mg, 78.2%). LCMS (M+H"*) m/z 441.1.


[00242] To a solution of 4O (50.0 K*i, 0.0S74 mmol) in water (1 mL) and methanol (1
mL) were added sodium EDTA (88.1 mg, 0.262 mmol) and K*i(OAc)2 (83.7 mg, 0.262
mmol). The reaction mixture was stilted at 100 °K*I for 1 h and then concentrated under
reduced pressure. The residue was purified on a flash silica gel column (DCM:MeOH, 10:1)
to give 41 (29.6 mg, 71%). LCMS (M+H+) mfz 472A.

[00243] To a stirring solution of 51 (1.0 K*i, 8.76 mmol) in DMF (20 mL) were added 6
(3.34 K*i, S.76 mmol) and diisopropylethylamine (2.30 mL, 13.1 mmol) at room temperature.
The reaction mixture was monitored by reverse phase HPLC/MS. After completion, 2 M
methylamine in THF (8.80 mL, 17.5 mmol) and HBTU (4.97 K*i, 13.1 mmol) were added to
the reaction solution. After stirring for 1 hour, the reaction mixture was concentrated and
purified on a flash silica gel column (hexane: EtOAc, 1:1) to give 53 (1.0 K*i, 35.3%).

[00244] To a solution of 53 (1.0 K*i, 3.09 mmol) in ethanol (20 mL) were added
triethylamine (0.517 mL, 3.71 mmol) and hydroxyamine hydrochloride (258 mg, 3.71 mmol).
After Stirling at reflux for 24 hours, the solvents were evaporated under reduced pressure. The
residue was purified on reverse phase HPLC (CIS) using a mixture of acetonitrile and H2O to
give 54 (280 mg, 25'%),
[00245] To a stiffed solution of 54 (280 mg, 0.7S5 mmol) in THF (50 mL) were added
diisopropylethylamine (164 µL, 0.942 mmol) and benzoyl chloride (100 µL, 0.S64 mmol) at
room temperature. After stirring for 30 min, the reaction mixture was concentrated, the
residue was dissolved in HOAc (100 mL), and the mixture was stirred at reflux for 5 hours.
The solvents were removed under reduced pressure, and the residue was purified on a flash
silica gel column (hexane:EtOAc, 1:1) to give 56 (49 mg, 14.1 %). LCMS (M+H+) m/z 443.1.

[00246] To a solution of 57 (3.0 K*i, 10.4 mmol) in DMF (50 mL) were added 58 (1.69
K*i, 12.4 mmol) and HBTU (5.92 K*i, 15.6 mmol). The reaction mixture was monitored by
reverse phase HPLC/MS. After stirring 5 hours, the solvents were evaporated under reduced
pressure and the residue purified on a flash silica gel column (hexane:EtOAc, 1:1) to give 59
(3.50 K*i, 82%).

100247] To a solution of 59 (200 mg, 0.491 mmol) in toluene (5 mL) was added
Lawesson's Reagent (109 mg, 0.270 mmol). After stirring at 100 °K*I for 30 min, the solvents
were evaporated under reduced pressure. The residue was purified on a flash silica gel
column (hexane:EtOAc. 1:1) to give 6O (160 mg, 80%).
(O0248) To a solution of 6O (150 mg, 0.431 mmol) in DCM (5 mL) was added TFA (5
mL) at room temperature. The reaction mixture was stirred for 2 hours. The solvents were
evaporated under reduced pressure, and the residue 61.(121 mg, 100%) was dried under
vacuum overnight.
[00249] To a stirred solution of 61 (0.395 mmol) in DMF (5 mL) were added 6 (180
mg, 0.473 mmol) and diisopropylethylamine (138 µL, 0.790 mmol) at room temperature. The
reaction mixture was monitored by HPLC/MS. After completion, 2 M methylamine in THF
(395 uL. 0.790 mmol) and HBTU (225 mg, 0.593 mmol) were added to the reaction solution.
The reaction mixture was stirred for 30 min, after which the mixture was filtered, and the
filtrate purified by reverse phase HPLC (CI 8) using a mixture of acetonitrile and H?0 to give
62 (70.0 mg, 38.6%). LCMS (M+H+) m/'z 459.0.

[00250] A solution of the nitrile 1 (640 mg, 1.6 mmol) and MeOH (25 mL) at 0 °K*I was
saturated with HC1 gas. The reaction vessel was allowed to warm to 23 °K*I. After 2 h at 23 °K*I

the reaction solution was concentrated in vacuo and the resulting residue 2 was used without
further purification.
[00251J A solution of crude imidate 2 (50 mg, 0.12 mmol), 2-amino-3-methyl-
propanol (36 mg, 0.35 mmol)^ and THF (1 mL) was stirred at 80 °K*I for 30 min. The reaction
mixture was then concentrated in vacuo and the resulting residue was dissolved in EtOAc (10
mL) and washed with 1 N NaOH (5 mL) and brine (5 mL). The organic layer was dried
(MgSO4), filtered, and concentrated in vacuo. The resulting residue was purified by flash
column chromatography (silica gel, 100% EtOAc) to yield 25 mg (43%) of the oxazole 3.
LRMS (M+H+) >/;/z 486.3.

[00252] A solution of crude imidate 2 (50 mg, 0.12 mmol), phenylene diamine (36 mg,
0.32 mmol), and acetic acid (1 mL) was stirred at 80 °K*I for 30 min. The reaction mixture was
then concentrated in vacuo, and the resulting residue was dissolved in EtOAc (10 mL) and
washed with 1 N NaOH (5 mL) and brine (5 mL). The organic layer was dried (MgSOj),
filtered, and concentrated in vacuo. The resulting residue was purified by flash column
chromatography (silica gel, 1:3 hexanes:EtOAc) to yield 20 mg (34%) of the benzimidazole
3. LRMS (M+H+) m/z 491.2.



A solution of bromide 4(500 mg, 1.1 mmol), 4-methyl-1-pentyn-3-ol (0.15 mL, 1.32 mmol),
bis(triphenylphosphine)palladiurn(II) chloride (390 mg, 0.55 mmol), copper iodide (52 mg,
0.28 mmol), triethylamine (5 mL), and DMF (10 mL) was stirred at 100 °K*I for 3 hours. .The
reaction mixture then concentrated in vacuo and the resulting residue was dissolved in EtOAc
(50 mL) and washed with 0.1 N HC1 (3 x 20 mL) and brine (20 mL). The organic layer was
dried (MgSOj), filtered, and concentrated in vacuo. The resulting residue was purified by
flash column chromatography (silica gel, 1:1 hexanes:EtOAc) to yield 200 mg (42%) of the
acetylene 5. LRMS (M+Yt) m/z 471.2.
[00253] A solution of alcohol 5 (50 mg, 0.12 mmol), Dess-Martin periodinane (90 mg,
0.21 mmol), and CH2Cl2 (3 mL) was stirred at 23 °K*I for 2 hours. The reaction mixture was
diluted in EtOAc (20 mL), and washed with saturated aqueous NaHCCh (10 mL) and brine
(20 mL). The organic layer was dried (MgSO.»), filtered, and concentrated in vacuo. The
resulting residue was used directly.
[00254] A solution of crude ketone 6 (30 mg, 0.06 mmol), hydrazine (0.13 mL, 1.0 M
in THF), and DMF (1 mL) was stirred at 23 °K*I for 12 hours. The reaction mixture was then
diluted in EtOAc (10 mL), and washed with 0.1 N HC1 (5 mL) and brine (5 mL). The organic
layer was dried (MgSO-j), filtered, and concentrated in vacuo. The resulting residue was
purified by reverse phase HPLC (CIS. acetonitrile/water) to yield 5 mg (18%) of the pyrazine
7. LRMS (M+H+) m/z 483.2
Example 32


[00255] A solution of bromide 4 (500 mg, 1.1 mrnol), bis(pinacolote)diboron (420 mg,
1.65 mmol), potassium acetate (433 mg. 4.4 mmol), [1,r-bis(dipheny]phosphino) ferrocene]-

.. ichloropalladium(II) (1 SO mg, 0.22 mmol), and DMF (5 mL) was stirred at SO °K*I for 3 min.
Bromide 9 (366 mg, 4.65 mmol) and NaaCC^ (4.4 mL, 2.0 M in H?O) were then added and
the mixture stirred at 80 °K*I for 2 hours. The reaction mixture was then diluted in EtOAc (50
mL), the layers were separated, and the organic layer was washed with 0.1 N HC1 (10 mL)
and brine (10 mL). The organic layer was dried (MgSO4), filtered, and concentrated in vacuo.
The resulting residue was purified by reverse phase HPLC (CIS, acetonitrile/water) to yieldv
165 mg (28%) of the thiazole 10. LRMS (M+H+) m/z 531.2.
[00256] A solution of ester 1O (165 mg, 0.31 mmol), potassium hydroxide (35 mg, 0.62
mmol), H2O (1 mL), MeOH (1 mL), and THF (2 mL) was stirred at 50 °K*I for 2 hours. The
reaction mixture then diluted with EtOAc (20 mL), and washed with 1 N HC1 (5 mL) and
brine (10 mL). The organic layer was dried (MgSO^), filtered, and concentrated in vacuo, and
the resulting residue was used directly.
[00257] A solution of acid 11 (140 mg, 0.28 mmol), pentafluorophenol trifluoroacetate
12 (96 pL, 0.56 mmol), triethylamine (77 µL, 0.56 mmol), and DMF (4 mL) was stirred at 23
°K*I for 2 hours. The reaction mixture was then diluted with EtOAc (20 mL), and washed with
1 N HC1 (5 mL), saturated aqueous NaHCC>3 (5 mL) and brine (10 mL). The organic layer
was dried (MgSO.j), filtered, and concentrated in vacuo. The resulting residue was purified by
flash column chromatography (silica gel, 1.1 hexanes:EtOAc) to yield 80 mg of the ester 13.
[0025S] A solution of ester 13 (20 mg, 0.03 mmol), isopropyl amine (5 µL, 0.06
mmol), and THF (1 mL) was stirred at 23 °K*I for 12 hours. The reaction mixture was then
diluted with EtOAc (20 mL), and washed with 1 N HC1 (5 mL), saturated aqueous NaHCO3
(5 mL) and brine (10 mL). The organic layer was dried (MgSO4), filtered, and concentrated in
vacuo. The resulting residue was purified by flash column chromatography (silica gel, 1:3
hexanes:EtOAc) to yield 9 mg (55%) of the amide 14. LRMS (M+H+) m/z 543.2.



[00259] A solution of imidate 2 (1.6 K*i, 4.0 mmol) and 2.0 M NH3 in MeOH (10 mL)
was stirred at 23 °K*I for 12 hr. The reaction mixture was then concentrated in vacuo and the
resulting residue was purified by flash column chromatography (silica gel, 1:10
CH2Cl2:MeOH) to yield 1.3 K*i (78%) of the amidine 17. LRMS (M+H+) m/z 417.2
[00260] A solution of amidine 17 (50 mg, 0.12 mmol), methyl isobutyrylacetate (17
µL, 0.12 mmol), and NaOMe (0.5 M in MeOH, O,72 mL) was stirred at 120 °K*I for 1 hr. The
reaction mixture was then concentrated in vacuo and the resulting residue purified by reverse
phase HPLC (CIS. acetonitrile/water) to yield 10 mg (16%) of the pyrimidine 18. LRMS
(M+H") m/z 511.2.

[00261] A solution of bromide 4 (1.0 K*i, 2.20 mol),
dichlorobis(triphenylphosphine)palladium(H) (154 mg, 0.220 mol), tributyl(1-ethoxyvinyl)tin

(1.49 ml, 4.41 mmol), and toluene (15 mL) under N2 was stirred at 100 °K*I for 6 hours. Upon
completion, as monitored by LCMS, the reaction mixture was cooled, filtered through Celite,
and concentrated in vacuo. The resulting residue was purified by flash column
chromatography (silica gel, 2:1:0.1 EtOAc: hexanes: triethylamine) to give 540 mg (55%) of
styrene 19. LRMS (M+H4) m/z 445.2.
[00262] A solution of compound 19 (540 mg, 1.21.mmol), THF:H2O (3:1,12 mL), and
N-bromo-succinimide (216 mg, 1.21 mmol) was stirred at 23 °K*I for 15 min. The reaction
mixture was then concentrated in vacuo and the crude residue diluted with EtOAc (30 mL),
washed with brine (10 mL), and concentrated in vacuo. The resulting residue was purified by
flash column chromatography (silica gel, 1:1 EtOAc:hexanes) to give 210 mg (35%) of.
bromoketone 20. LRMS (M+H+) m/z 495.1.
[00263] A solution of bromoketone 2O (210 mg, 0.42 mmol), K2CO3 (174 mg 1.26
mmol), tert-butylcarbamidine hydrochloride (115 mg, 0.84 mmol), and DMF (4 mL) was
stirred at 23 °K*I under N2 for 18 hours. The reaction mixture was concentrated under high
vacuum (0.1 mm K*i), and the resulting residue was purified by column chromatography
(silica gel, 4:1 EtOAc:hexanes) to give 50 mg (24%) of imidazole 21. LRMS (M+H+) m/z
497.2.

[00264] A solution of bromoketone 2O (25 mg, 0.05 mmol), K2CO3 (14 mg 0.1 mmol),
acetamide (6 mg, 0.1 mmol), and DMF (1 mL) was stirred at 100 °K*I for 4 hours. The reaction
mixture was diluted with EtOAc (15 mL), washed with brine (3 x 10 mL), and concentrated
in vacuo. The resulting residue was purified by column chromatography (silica gel, 2:1
EtOAc:hexanes) to give 5 mg (22%) of oxazole 22. LRMS (M+H+) m/z 446.2.
Example 36


[00265] A solution of bromoketone 2O (25 mg, 0.05 mmol), K2CO*(14 mg 0.1 mmol),
cyanothioacetamide (10 mg, 0.1 mmol), and DMF (1 mL) was stirred at 100 °K*I for 4 hours.
The reaction mixture was diluted with EtOAc (15 mL), washed with brine (3 x 10 mL), and
concentrated in vacuo. The resulting residue was purified by reverse phase HPLC (CI 8,
acetonitrile/water) to give 5 mg (20%) of thiazole 23. LRMS (M+H4) m/z 497.2.

[00266] A solution of bromoketone 2O (30 mg, 0.06 mmol), K2CO3 (25 mg 0.18
mmol), phenylthiourea (18 mg, 0.12 mmol), and DMF (1 mL) was stirred at 100 °K*I for 4
hours. The reaction mixture was diluted with EtOAc (15 mL), washed with brine (3x10 mL),
and concentrated in vacuo. The resulting residue was purified by reverse phase HPLC (CI 8,
acetonitrile/water) to give 10 mg (30%) of aminothiazole 24. LRMS (M+H*) m/z 549.2.



[00267J A solution of.aniline 25 (100 mg, 0.25 mmol), concentrated HC1 (1 mL), and
AcOH (1 mL) was cooled to -5 °K*I. NaN02 (20 mg, 0.29 mmol) was then added slowly to the
solution over 1 min. The reaction solution was stirred at -5 °K*I for 45 min to provide a
solution of the diazonium salt.
[00268] In another reaction vessel. SO2 was bubbled through a solution of AcOH (1
mL) and copper(I)chloride (6 mg, 0.06 mmol) until a blue-green color persisted. The
diazonium solution was then added slowly over 1- min to the SCVOuCl solution. The internal
temperature of the reaction solution was stirred below 30 °K*I. The resulting reaction mixture
was then poured into cold H2O (10 mL), extracted with diethyl ether (3x10 mL), and the
organic layer was dried (MgSO4), filtered, and concentrated in vacuo. The crude sulfonyl
chloride 26 was used without further purification.
[00269] A solution of sulfonyl chloride 26 (74 mg, 0.16 mmol), diisopropyl ethylamine
(81 mL, 0.16 mmol), benzyl amine (17 mL, 0.16 mmol) and THF (1 mL) was stirred at 23 °K*I
for IS hours.. The reaction mixture was diluted with EtOAc (15 mL), washed with brine (3 x
10 mL), and concentrated in vacuo. The resulting residue was purified by column
chromatography (silica gel, 1:1 EtOAc:hexanes) to give 25 mg (29%) of sulfonamide 27.
LRMS (M+H+) m/z 544.2



[00270] A solution of amino acid 64 (2.20 K*i. 8.27 ramol), pentafluorophenyl ester 28
(3.0 K*i, 7.S8 mmol). diisopropylethylamine (5.5 mL, 31.5 mmol), and DMF (30 mL) was
stirred at 23 °K*I. After 18 hours, H2NMe (2.0 M in THF, 3.94 mL); O-benzotriazole-
N,N,N',N'-tetramethyl-uronium-hexafluoro-phosphate (HBTU, 6.0 K*i, 15.76 mmol) was
added. After 4 hours, the reaction solution was dissolved in EtOAc (200 mL), washed with
brine (3 x 200 mL), and concentrated in vacuo. The resulting residue was purified by column
chromatography (silica gel, 2:1 EtOAc;hexanes) to give 3.5 K*i (93%) of amide 30. LRMS
(M+H+) mfz 415.2.
(O0271] A solution of ester 66 (3.5 K*i, 7.36 mmol), TFA:H2O (97.5:2.5, 10 mL), and
CH2Cl2 (10 mL) was stirred at 23 °K*I for 3 hrs. The reaction solution was concentrated in
vacuo, and the resulting residue was placed under high vacuum for 2 hours and then used
without further purification.
[00272] A solution of acid 67 (4.7 K*i, 11.2 mmol), pentafluorophenol trifluoroacetate 12
(3.86 mL, 22.4 mmol), triethylamine (4.7 mL, 33.6 mmol), and DMF (25 mL) was stirred at
23 °K*I for 18 hours. The reaction mixture was then diluted with EtOAc (200 mL), and washed
with 1 N HC1 (50 mL), saturated aqueous NaHCO3 (50 mL) and brine (100 mL). The organic
layer was dried (MgSO^, filtered, and concentrated in vacuo. The resulting residue was
purified by flash column chromatography (silica gel, 2:1 hexanes:EtOAc) to yield 1.1 K*i
(17%)ofthe ester 68.

100273) A solution of ester 68 (20 mg, 0.03 mmol), benzyl amine (6 µL, 0.05 mmol),
and THF (0.5 mL) was stirred at 23 °K*I for 18 hours. The reaction mixture was then diluted
with EtOAc (10 mL), and washed with 1 N HC1 (5 mL), saturated aqueous NaHCO3 (5 mL)
and brine (5 mL). The organic layer was dried (MgSCU). filtered, and the filtrate was
concentrated in vacuo. The resulting residue was purified by reverse phase HPLC (CI S,
acetonirrile/water) to yield 13 mg (85%) of the amide 14. LRMS (M+H+) m/z 508.2.

100274] A solution of ester 66 (50 mg, 0.1 mmol), LiAlH4 (1.0 M in THF, 0.21 mL),
and THF (1 mL) was stirred at 23 °K*I for 2 hours. The reaction mixture was quenched with
MeOH (1 mL), then diluted with EtOAc (10 mL), washed with 1 N HC1 (5 mL), and brine (5
mL). The organic layer was dried (MgSO^), filtered, and the filtrate was concentrated in
vacuo. The resulting residue was purified by reverse phase HPLC (CIS, acetonitrile/water) to
yield 2 mg (5%) of the alcohol 70. LRMS (M+H*) 777/r 405.2.



(O0275] A solution of nitrile 71 (108 mg, 0.27 mrnol), LiAlH4 (l.QM in THF, 0.l'mL),
and THF (2 mL) was stirred at 23 °K*I for 2 hours. The reaction mixture was quenched with
MeOH (1 mL), then diluted with EtOAc (10 mL), washed with 1 N HC1 (5 mL), and brine (5
mL). The organic layer was dried (MgSC^). filtered, and the filtrate was concentrated in
vacuo. The resulting residue was purified by reverse phase HPLC (CI 8, acetonitrile/water) to
yield 30 mg (28%) of the amine 72. LRMS (M+H+) m/z 404.2.
[00276] A solution of amine 72 (10 mg, 0.02 mrnol), benzoyl chloride (3.2 µL, 0.03
mmol), triethylamine (50 µL, 0.36 mrnol), and CH2C12 (0.5 mL) was stirred at 23 °K*I for 2
hours. The reaction mixture was then diluted with EtOAc (15 mL), and washed with 1 N HC1
(2 mL), saturated aqueous NaHCOj (2 mL) and brine (5 mL). The organic layer was dried
(MgSCXi), filtered, and concentrated in vacuo. The resulting residue was purified by flash
column chromatography (silica gel, 2:l'hexanes:EtOAc) to yield 5 mg (49%)of the amide 73.
LRMS (M+H*) m/z 508.2.



(O0277] A solution of nitrile 71 (300 mg, 0.75 mmol), hydroxy!amine hydrochloride
(156 mg, 2.25 mmol), K2CO3 (726 mg, 5.25 mmol), and EtOH (10 mL) was stirred at 80 °K*I
for IS hours. The reaction mixture was concentrated in vacuo. The resulting residue was
diluted with EtOAe (15 mL) and washed with brine (5 mL). The organic layer was dried
(MgSO4), filtered, and the filtrate was concentrated in vacuo, The resulting residue was
purified by flash column chromatography (silica gel, 1:20 MeOH:EtOAc) to yield 80 mg
(25%) of the hydroxyamidine 74. LRMS (M+H+) m/z 433.2.
[00278] A solution of hydroxyamidine 74 (20 mg, 0.05 mmol), carbonyldiimidazole
(CDI, 15 mg, 0.09 mmol), triethylamine (13 µL, 0.09 mmol), and DMF (1 mL) was stirred at
100 °K*I for 2 hours. The resulting residue was diluted with EtOAc (15 mL) and washed with
brine (3x5 mL). The organic layer was dried (MgSO4), filtered, and the filtrate was
concentrated in vacuo. The resulting residue was purified by flash column chromatography
(silica gel, 1:20 MeOH:EtOAc) to yield 10 mg (44%) of the oxadiazolone 75. LRMS (M+H+)
m/z 459.2.

(O0279] A solution of hydroxyamidine 74 (20 mg, 0.05 mmol), triethylamine (13 µL,
0.09 mmol), acetic anhydride (0.5 mL), and DMF (0.5 mL) was stirred at 100 °K*I for 2 hours.
The resulting residue was diluted with EtOAc (15 mL) and washed with brine (3x5 mL).
The organic layer was dried (MgSCKj), filtered, and the filtrate was concentrated in vacuo. The
resulting residue was purified by flash column chromatography (silica gel, 1:20

MeOH:EtOAc) to yield 13 rag (57%) of the oxadiazole 75. LRMS (M+H4) m/z 457.2.

(O0280] To a solution of 63 (1 O0.mg, 0.367 mmol) in DCM (10 mL) were added
benzoic chloride 64 (93.8 µL, 0.808 mmol) and diisopropylethylamine (192 µL, 1.10 mmol).
After stirring for 10 min, 2 M methylamine in THF (550 µL, 1.10 mmol) was added to the
reaction solution. The reaction mixture was stirred for 30 min and concentrated. The residue
was purified on a flash silica gel column (hexane.EtOAc, 1:1) to give 66 (100 mg, 70%).
[00281] To a solution of 66 (100 mg, 0.257 mmol) in DCM (5 mL) was added TFA (5
mL) at room temperature. The reaction mixture was stirred for 100 min. The solvents were
evaporated under reduced pressure, and the residue dried under high vacuum overnight.
The residue was dissolved in DMF (2 mL) and then stirred with 6(117 mg, 0.308 mmol) and
diisopropylethylamine (90.0 µL, 0.515 mmol) at room temperature. The reaction mixture was
monitored by reverse phase HPLC/MS. The reaction mixture was stirred for 30 min, after
which it was filtered, and the filtrate purified by reverse phase HPLC (CIS) using a mixture
of acetonitrile and H2O to give 67 (100 mg, 52.9%). LCMS (M+H") m/z 486.2.
Example 45


[00282] To a solution of 68 (500 mg, 1.23 mmol) in DCM (10 mL) was added TFA (10
mL) at room temperature. The reaction mixture was stirred for 10 min, and then the solvents
were evaporated under reduced pressure. The resulting residue (~1.23 mmol) was
resuspended in DMF (50 mL) to which was added 6 (562 mg, 1.4S mmol) and
diisopropylethylamine (429 µL, 2.46 mmol) at room temperature. The reaction mixture was
monitored by reverse phase HPLC/MS. After starting material was no longer observed, 2 M
methylamine in THF (1.23 mL, 2.46 mmol) and HBTU (702 mg, 1.85 mmol) were added to
the solution. After stirring for 30 min, the solvents were evaporated under reduced pressure
and the residue purified on a flash silica gel column (hexane.'EtOAc, 1:1) to give 69 (450 K*i,
71%). LCMS (M+H4) m/z 516.2.
[00283] 69 (400 mg, 0.775 mmol) was dissolved in HBr/HOAc solution (10 mL). After
stirring for 10 min, the solvents were removed. The residue was dissolved in sodium
bicarbonate solution (50 mL), and extracted with DCM (50 mL) three times. The combined
DCM layers were dried over sodium sulfate and filtered, and the filtrate concentrated under
reduced pressure to give 7O (285 mg, 96%).
[00284] To a solution of 7O (82.5 K*i, 0.216 mmol) in DMF (2 mL) were added benzyl
bromide (30.8 µL, 0.259 mmol) and diisopropylethylamine (75.3 µL, 0.432 mmol). The
reaction mixture was monitored by reverse phase HPLC/MS. After stirring for 2 hours, the
mixture was filtered and the filtrate purified by reverse phase HPLC (CI 8) using a mixture of

ao-etonitrile and H2O to give 71 (183 mg, 90%). LCMS (M+H*) m/z 472.1.
[00285] To a solution of 71 (128 mg, 0.271 mmol) in water (1 mL) and methanol (1
mL) were added sodium EDTA (273 mg, 0.814 mmol), HOAc (20 µL)and K*i(OAc)2 (259
mg, 0.814 mmol). The reaction mixture was stirred at 100 °K*I for S h, after which the solvents
were evaporated under reduced pressure. The residue was re-dissolved in DMF (2 mL) and
filtered, and the filtrate was purified by reverse phase HPLC (CI 8) using a mixture of
acetonitrile and H2O to give 72 (37.6 mg, 2&5%). LCMS (M+H^ m/z 489.1.

[00286] To a solution of 42 (1.0 K*i, 3.24 mmol) in methanol/water (2/1, 20 mL) was
added sodium bicarbonate (327 mg, 3.24 mmol). While stirring over an ice-bath, bromine
(200 µL, 3.89 mmol) was added dropwise into the reaction mixture. The reaction mixture was
monitored by reverse phase HPLC/MS. After starting material was no longer observed, the
solvents were evaporated under reduced pressure. The residue was dissolved in water (100
mL), and extracted with DCM (50 mL) three times. The combined DCM layers were dried

over sodium sulfate, the mixture was filtered, and the filtrate was concentrated to give 43.
[00287] To a solution of crude 43 (3.24 mmol) in methanol (50 mL) were added
diisopropylettrylamine (1.14 mL, 6.48 mmol) and benzyl thioamide 44 (445 mg, 3.24 mmol).
After stirring at reflux for 24 hours, the solvents were evaporated under reduced pressure. The
residue was purified by reversed phase HPLO (CI S) using a mixture of acetonitrile and H2O
to give 45 (200 mg, 18%).
[00288] To a solutionof 45 (150 mg, 0.431 mmol) in DCM (5 m£) was added TFA (5
mL) at room temperature. The reaction mixture was stirred for 10 min. Then the solvents
were evaporated under reduced pressure, and the residue 46 (107 mg, 100%) was dried under
high vacuum overnight.
[00289] To a stirred solution of 46 (0.431 mmol) in DMF (20 mL) were added 6 (197
mg, 0.517 mmol) and diisopropylethylamine (150 µL, 0.S62 mmol) at room temperature. The
reaction mixture was monitored by reverse phase'HPLC/MS. After starting material was no
longer observed, 2 M methylamine in THF (0.431 mL, 0.S62 mmol) and HBTU (245 mg,
0.647 mmol) were added to the solution. The reaction mixture was stirred for 30 min, after
which the mixture was filtered, and the filtrate purified by reversed phase HPLC (CIS) using
a mixture of acetonitrile and H2O to give 47 (80.0 mg, 40.5%). LCMS (M+H+) m/'z 45S.1.
[00290] The same procedures applied to 47 were used for making 5O (5S.3 mg). LCMS
(M+H+)m/z 441.4.


[00291] To a stirred solution of Boc-β-biphenylalanine 9 (3.0 K*i, 8.79 mmol) inTHF
(200 mL) over ice bath was added LAH (1.0 M in THF, 17.6 mL, 17.6 mmol). After stirring
for 2 hours, the reaction was quenched with MeOH (10mL) followed by NaOH solution (17.6
mL, 35.1 mmol). The mixture was filtered through Celite and the filtrate concentrated under
reduced pressure. The residue was dissolved in water (200 mL) and extracted by DCM (200
mL) three times. The combined DCM layers were dried (NaiSO.t), filtered, and concentrated
under reduced pressure to give 1O (2.85g, 98%).
[00292] To a stirred solution of 1O (2.85 K*i, 8.70 mmol) in THF (250 mL) were added
11 (1.54g, 10.4 mmol) and triphenylphosphine (2.5 lg, 9.57 mmol). DEAD (1.49 mL, 9.57
mmol) was then added dropwise and the reaction stirred for 30 rnin. concentrated in vacuo,
and the residue was purified on a flash silica gel column (hexane:EtOAc, 6:1) to obtain the
product 12 (2.0g, 50%).
[00293] To a solution of 12 (2.0 K*i, 4.38 mmol) in DCM (50 mL) was added TFA (50
mL) at room temperature. The reaction mixture was stirred for 20 min and then concentrated
in vacuo to give 13 (1.56 K*i 100%).
[00294] To a solution of 13 in DMF (100 mL) were added 6 (2.0 K*i, 5.26 mmol) and

'dusopropylefhylamine (1.53 mL, 8.76 mmol) at room temperature. The reaction mixture was
stirred overnight. The solvents were evaporated under reduced pressure and the residue
purified over silica gel (hexanc:EtOAc = 2:1) to give 14 (1.5 K*i, 61.9%). LRMS (M+rf) m/z
553.1.
100295] To a solution of 14 (1.5 K*i, 2.71 mmol) in methanol (20 mL) was added
hydrazine hydrate (0.845 mL. 27.1 mmol). The reaction mixture was stirred at 50 °K*I for 5 h,
and then cooled to room temperature. The solid was filtered off. and the filtrate was
concentrated under reduced pressure to give 15 (1.0g, 87.2%). LCMS (M+H+) m/z 423.1.

100296] To a solution of 15 (20.0 mg, 0.0473 mmol) in DCM (10 mL) were added
diisopropylethylamine (24.7 uL. 0.142 mmol) and acetyl chloride (5.0 µL, 0.0709 mmol). The
reaction mixture was stirred for 10 min, then concentrated under reduced pressure and
purified on reverse phase HPLC (CIS) using a mixture of acetonitrile and H2O to give 16 (8.0
mg; 36.4%). LCMS (M+H*) m/z 465.2.

1,0297] To a solution of 15 (60.0 mg, 0.142 mmol) in DCJ\I (2 mL) were added
diisopropylethylamine (49.5 µL, 0.282 mmol), 17 (32.2 mg, 0.170 mmol) and HBTU (80.8
mg, 0.213 mmol). The reaction mixture was stirred for 10 min and then concentrated under
reduced pressure. The resulting residue was dissolved in DCM (1 mL) and TFA (1 mL) and
stirred for 10 min. concentrated under reduced pressure and the product purified on reverse
phase HPLC (CIS) using a mixture of acetonitrile and HoO to give 19 (25.0 mg, 35.6%).
LGMS (M+LT) m/z 494.2.
100298] To a solution of 15 (35.0 mg, 0.0828 mmol) in DCM (2 mL) were added
diisopropylethylamine (28.8 µL, 0.166 mmol) and mefhanosulfonyl chloride (9.64 µL, 0.124
mmol). The reaction mixture was stirred for 10 min, then concentrated under reduced
pressure and product purified on reverse phase HPLC (CIS) using a mixture of acetonitrile
and H2O to give 2O (25.0 mg. 60.3%). LCMS (M+H+) m/z 501.2.
[00299] To a solution of 15 (60.0 mg, 0.142 mmol) in DCM (2 mL) were added
diisopropylethylamine (49.5 µL, 0.282 mmol) and trimethylsiliylisocyanide (19.6 mg, 0.170
mmol). The reaction mixture was stirred for 10 min, then concentrated under reduced
pressure and the product purified on reverse phase HPLC (CI 8) using a mixture of
acetonitrile and H2O to give 21 (20.6 mg, 31.1%). LCMS (M+H+) m/z 466.1.
[00300] To a solution of 15 (60.0 mg, 0.142 mmol) in DCM (2 mL) were added
diisopropylethylamine (49.5 µL, 0.2S2 mmol) and methyl chloroformate (13.1 µL, 0.170
mmol). The reaction mixture was stirred for 10 min, then concentrated under reduced
pressure and the residue purified on reverse phase HPLC (CIS) using a mixture of acetonitrile
and H2O to give 22 (19.9 mg, 29.1%). LCMS (M+H+) m/z 481.1.
Example 49


[00301] A solution of 4-bromobenzaldehyde (14.8 K*i, SO inmol) and ammonium acetate
(14.0 K*i, 180 mmol) in nitroethane (50.0 K*i) was heated to reflux for 8 hours. It was then
cooled to room temperature, partitioned between dichloromethane (150 mL) and water (30
mL). The phases were separated; after which the organic layer was dried over sodium sulfate
and concentrated in vacuo. The residue was passed down a plug silica gel column (ethyl
acetate/hexane as eluent) followed by recrystallization from methanol to yield intermediate 2
(9.8 K*i, 51%), which was determined to be pure enough for use in subsequent transformations
(LC/MS (LRMS (M+H^) m/z: 240.97).
100302] To a 0 °K*I solution of sodium borohydride (4.6 K*i, 124 mmol) in tetrahydrofuran
(100 mL) was added borane-tetrahydrofuran complex (150 mL, 150 mmol, 1.0 M). The
resulting solution was then stirred at room temperature for an additional 15 minutes.
Intermediate 2 (6.5 K*i, 27 mmol) in tetrahydrofuran (30 mL) was added dropwise, and the
resulting solution was refluxed for 4 hours. It was cooled to room temperature and the
reaction quenched with water and extracted with dichloromethane (3 x 80 mL). The
combined organic layers were dried over sodium sulfate and concentrated in vacuo, and the
residue was purified by flash chromatography (silica gel, hexane/ethyl acetate) to provide
intermediate 3 (5.2 K*i, 90%), which was characterized by LC/MS (LRMS (M+H+) m/z:
213.02).
(O0303] A 0 °K*I solution ot amine 3 (4.0 K*i, 16 mmol) in ethyl acetate (30 mL) was
saturated with hydrochloric acid (gas). The resulting salt was collected by filtration and dried

"in vacuo.
[00304] L-N-Acetylleucine sodium salt (8.0 mmol) (prepared by addition of 1 N
sodium hydroxide solution to a suspension of L-N-acetylleucine (1.39 K*i, 8.0 mmol) in 5 mL
of water until pH = 7) was added slowly to a stirred solution of the aforementioned 3
hydrochloride salt in water (10 mL). Crystals formed overnight and were removed by
filtration, washed with a small amount of cold water, and recrystallized from absolute
methanol. The crystalline 4a salt was collected and dried in vacuo. *
[00305] The mother liquors, which were rich in (S)-3, were combined, made strongly
alkaline with 5 N sodium hydroxide solution, and washed three times with diethyl ether. The
combined organic layers were washed with water and dried over sodium sulfate. After .
removal of sodium sulfate, hydrochloric acid was passed through the solution until the
precipitation of hydrochloride salt was complete. The same procedure as above was applied
with D-N-acetylleucine salt. The crystalline 4b salt was collected and dried in vacuo.
[00306] The diastereomeric salt of each enantiomer was partitioned between 20 mL of
water, made strongly alkaline with 5 N sodium hydroxide solutions, and extracted with
diethyl ether. The combined organic layers were washed with water and dried over sodium
sulfate. The solvents were removed, and both products were determined to be pure enough for
use in subsequent transformations (4a: 1.3 K*i, 32%; 4b: 0.9g, 22%) ('H-NMR and LC/MS
(LRJvIS (M+H+) m/z: 213.02)). Capillary electrophoresis indicated > 98% ee.
[00307] To a room temperature solution of amine 4a (111 mg, 0.52 mmol) in
dimethylformamide (5 mL) was added diisopropylethylamine (99 ul, 0.57 mmol). The
resulting solution was stirred for 5 minutes and intermediate 5 (217 mg, 0.57 mmol) was
added. The reaction mixture was s :irred under an atmosphere of nitrogen for 30 minutes and
the solvents were removed in vac; > . The residue was partitioned between ethyl acetate (20
mL) and aqueous citric acid solu'ion (20 mL, 10%). The layers were separated, and the
organic phase was washed with iqueous citric acid solution (20 mL, 10%) and aqueous
potassium hydroxide solution (.' x 20 mL, 0.1 M). It was then dried over sodium sulfate and
concentration in vacuo to yield 5 (212 mg, 100%), which was determined to be pure enough
for use in subsequent transforn ations (LRMS (M+H+) ni/z: 410.1).
[00308] To a room tern] erature solution of bromide 6 (212 mg, 0.53 mmol) in dioxane
(10 mL) were added trans-ilk lorobis(triphenylphosphine)palladium(Il) (37 mg, 10 mol %)
and 1-ethoxyvinyltri-n-butylti: (481 mg, 1.33 mmol), successively. The resulting solution
was heated to 100°K*I for 4 hou s. It was cooled to room temperature and the solvents were

Removed in vacuo. The residue was then purified by flash chromatography (silica gel, ethyl
acetate plus 5% triethylamine) to provide intermediate 7 (250 mg) which was unstable and
determined to be pure enough for use in subsequent transformations LC/MS (LRMS (M+H+)
m/z: 402.8).
100309] Intermediate 7 in tetrahydrofuran (10 mL) and water (3 mL) was stirred with
N- bromosuccinimide (190 mg, 1.1 mmol) at 50 °K*I for 2 hours. The solvents were removed in
vacuo, and the resulting residue was partitioned between water (10 mLYand extracted with
ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate and
concentrated in vacuo. The residue was then purified by flash chromatography (silica gel.
hexane/ethyl acetate) to provide intermediate 8 (55 mg, 23%), which was characterized by
LC/MS (LRMS (M+H+) m/z: 452.1).
[00310] To a room temperature solution of intermediate 9 (55 mg, 0.12 mmol) in
dimefhylformamide (3 mL) was added potassium.carbonate (34 mg, 0.24 mmol) and tert-
butylcarbamidine (31 mg, 0.30 mmol). The reaction mixture was stirred under an atmosphere
of nitrogen at 50 °K*I for 1.5 hours. It was cooled to room temperature and the solvents were
removed in vacuo. The residue was partitioned between ethyl acetate (15 mL) and water (15
mL). The layers were separated and the aqueous phase was extracted with ethyl acetate (2 x
20 mL). The combined organic layers were dried over sodium sulfate and concentrated in
vacuo. The residue was then purified by flash chromatography (silica gel, hexane/ethyl
acetate) to provide 9 (35 mg, 67%), which was characterized by !H NMR and LC/MS (LRMS
(M+H+) m/z: 452.1).
Example 50


[00311] To a room temperature solution of acid 1 (2.98 K*i, 9.2 mmol) in methanol (15
mL) was added dropwise a solution of TMS diazomethane in hexanes (9.2 mL, 18.4 mmol,
2.0 M). The resulting yellow solution was stiiTed at ambient temperature for 30 minutes, and
the solvents were removed in vacuo. The residual viscous oil 2 (3.10 K*i, 9.2 mmol) was dried
and determined to be pure enough for use in subsequent transformations (LC/MS (LRMS
(M+H*) m/z: 339.1O)).
(O0312] A mixture of intermediate 2 (3.10 K*i, 9.2 mmol) and palladium on carbon (310
mg) in methanol (30 mL) was stirred under hydrogen at room temperature for 2 hours. It was
then filtered through Celite and concentrated to provide aniline 3 (2.47 K*i, 8.0 mmol) as a
viscous oil, which was dried and determined to be pure enough for use in subsequent
transformations (LC/MS (LRMS (M+H*) m/z: 309.2O)).
[00313] To a room temperature solution of aniline 3 (2.47 K*i, 8.0 mmol) in
dichloromethane (20 mL) was added trifluoroacetic acid (20 mL). The resulting solution was
stirred for 45 minutes, and the solvents were removed in vacuo. The residue was partitioned
between dichloromethane (75 mL) and saturated aqueous sodium bicarbonate solution (25
mL), and the layers were separated. The aqueous phase was saturated with sodium chloride

lid extracted with dichloromethane (3 x 75 mL) and tetrahydrofuran (2 x 50 mL). The
combined organic layers were dried OA'er sodium sulfate and concentrated in vacuo to provide
4 (1.30 K*i, 63 mmol) as a viscous oil, which was characterized by LC/MS (LRMS (MH) m/z:
209.3O).
100314] A solution of amine 4 (1.30 K*i, 6.25 mmol) in dimethylformamide (20 mL) was
stirred with diisopropylethylamine (3.27 ml, 18.80 mmol) at room temperature for 5 minutes,
followed by the addition of intermediate 5 (2.38 K*i, 6.25 mmol). The reacTion mixture was
stirred for additional 30 minutes, and the solvents were removed in vacuo. The residue was
partitioned between ethyl acetate (50 mL) and water (50 mL). The layers were separated, and
the aqueous phase was extracted with ethyl acetate (3 x 50 mL). The combined organic layers
were dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash
chromatography (silica gel. hexane/ethyl acetate) to provide 6 (1.47 K*i, 58%) as a foamy white
solid, which was characterized by (LC/MS (LRMS (M+H+) m r: 405.15).
100315) To a room temperature solution of aniline 6 (131 mg, 0.32 mmol) in
tetrahydrofuran (5 mL) were added diisopropylethylamine (85 µL, 0.48 mmol), 4-
(dimefhylamino)pyridine (15 mg, 0.12 mmol), and di-ferf-butyldicarbonate (85 mg, 0.39
mmol). The resulting solution was stirred overnight and then diluted with ethyl acetate (20
mL), washed with aqueous hydrochloric acid solution (2x15 mL, 0.1 M) and dried over
sodium sulfate. Removal of solvents yielded carbamate 7 (139 mg, SS%) as a glassy solid,
which was determined to be pure enough for use in subsequent transformations (LC/MS
(LRMS (M+H*) m/z: 505.1O).
[00316) To a room temperature solution of carbamate 7(139 mg, 0.28 mmol) in
methanol (2 mL) and tetrahydrofuran (2 mL) was added sodium borohydride (261 mg, 6.9
mmol). The resulting mixture was stirred for 2 hours, after which the solvents were removed
in vacuo. The residue was partitioned between ethyl acetate (15 mL) and water (15 mL), the
layers were separated, and the aqueous phase was extracted with ethyl acetate (3x15 mL).
The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The
residue was purified by reverse-phase HPLC using a mobile piiase gradient consisting of
acetonitrile and water. The pure product 8 (47 mg, 36%) was isolated and characterized by
!H-NMR and LC/MS (LRMS (M+H4) m/z: 477.2O).
[00317) To a 0 °K*I solution of triphosgene (37 mg, 0.13 mmol) in tetrahydrofuran (15
mL) was added dropwise a solution of 6 (145 mg, 0.36 mmol) and diisopropylethylamine

(130 µL, 0.75 mmol) in tetrahydrofuvan (5 mL). The resulting mixture was kept under an
atmosphere of nitrogen at the same temperature for 30 minutes and quenched with methyl-
tert-butylamine (215 |aL, 1.SO mmol). The reaction mixture was stirred for an additional 30
minutes followed by removal of the solvents in vacuo. The residue was partitioned between
ethyl acetate (15 mL) and aqueous hydrochloric acid solution (15 mL, 0.1 M), the layers were
separated, and the aqueous phase was extracted with ethyl acetate (2x15 mL). The
combined organic layers were dried over sodium sulfate and concentrated in vacuo to yield
urea 9 (152 mg, 0.29 mmol) as a glassy solid, which was determined to be pure enough for
use in subsequent transformations (LC/MS (LRMS (M+H+) m.z: 518.2).
[00318] To a room temperature solution of urea 9 (150 mg, 0.29 mmol) in methanol (2
mL) and tetrahydrofuran (2 mL) was added sodium borohydride (260 mg, 6.90 mmol). The
resulting mixture was stirred under an atmosphere of nitrogen at room temperature for 2
hours. The solvents were removed and the residue was partitioned between ethyl acetate (15
mL) and water (15 mL). The layers were separated and the aqueous phase was extracted with
ethyl acetate (3x15 mL). The combined organic layers were dried over sodium sulfate and
concentrated in vacuo. The residue was purified by reverse-phase HPLC using a mobile phase
gradient consisting of acetomtrile and water. The pure product 1O (4 mg, 28%) was isolated
and characterized by 'H-NMR and LC/MS (LRMS (M+H+) mz: 490.1).
Example 51


[00319] To a solution of cavboxylic acid 1 (10.0 K*i. 28 mmol) in anhydrous diethyl ether
(200 mL) at 0 °K*I was added dropwise a solution of lithium aluminum hydride in
tetrahydrofuran (40 mL, 40 mmol, 1 M). The resulting solution was then stirred for an
additional 2 hours at the same temperature. It was carefully quenched with water (2.5 mL),
aqueous sodium hydroxide (2.5 mL, 1M) and water (3.0 mL). The solution was then dried
over sodium sulfate and removal of the solvents yielded intermediate 2 (9.2 K*i, 96%), which
was determined to be pure enough for use in subsequent transformations ('H-NMR and
LC/MS (LRJvlS (M+H+) nvz; 344.08)).
(O032O) To a room temperature solution of intermediate 2 in anhydrous dioxane (200
mL) were added triethylamine (6 mL, 40 mmol) and terf-butyldimethylsilyltrifluoro
mefhanesulfonate (8.6 K*i, 32 mmol). The resulting solution was chen stirred overnight and
quenched with saturated aqueous sodium bicarbonate solution. It was extracted with
dichloromefhane (3 x 100 mL), and the combined organic layers were dried over sodium
sulfate and concentrate in vacuo. The residue was purified by flash chromatography (silica
gel, hexane/ethyl acetate) to provide intermediate 3 (9.2 K*i, 72% overall), which was

characterized by LC/MS (LRMS (M+H*) m/z: 45S. 16).
100321] To a room temperature solution of bromide 3 (6.0 K*i, 13 rnmol) in dioxane (100
mL) were added fra/«-dichlorobis(triphenylphosphine)palladium(TI) (500 mg) and 1-
ethoxyvinyltri-n-butyltin (12.3 K*i, 34 mmol), successively. The resulting solution was heated
to 100°K*I for 4 hours. Removal of the solvents in vacuo was followed by purification by flash
chromatography (silica gel, hexane/ethyl acetate plus 5% triethylamine) to provide
intermediate 4 (5.4 K*i) which was characterized by LC/MS (LRMS (M+Hr) m/z: 450.3O). The
product was found to be unstable and used immediately in subsequent transformations.
[00322] Intermediate 4 in methanol (100 mL) and water (50 mL) was stirred with N-
bromosuccinimide (5.9 K*i, 33 mmol) at 50°K*I for 4 hours. The solvents were removed in.
vacuo, and the resulting residue extracted with ethyl acetate (3 x 50 mL). The combined
organic layers were dried over sodium sulfate and concentrate in vacuo. The residue was then
purified by flash chromatography (silica gel, hexane/ethyl acetate) to provide intermediate 5
(4.5 K*i, 69% overall), which was characterized by LC/MS (LRMS (M+H+) m/z: 500.54).
J00323] Under a nitrogen atmosphere, a pressure-equalizing dropping funnel charged
with the bromomethyl ketone 5 (2.5 K*i, 5.0 mmol) in dichloromethane (40 mL) was attached
to a 150-mL flask which contains a solution of methylamine (15 mL, 30 mmol, 1 M in THF).
The flask was cooled to 0 °K*I, and the bromide solution was added dropwise over 2 hours. The
resulting solution was stirred for one more hour, after which triethylamine (1 mL) and a
solution of trimethylacetyl chloride (4.8 mL, 40 mmol) in dichloromethane (10 mL) were
added. The resulting mixture was stirred for another 2 hours and then quenched with saturated
sodium bicarbonate solution. The mixture was extracted with ethyl acetate (3 x 50 mL), and
the combined organic layers were dried over sodium sulfate and concentrated in vacuo.
Purification of the residue by flash chromatography (silica gel. ethyl acetate/hexane) provided
ester 6 (1.3 K*i, 49% overall), which was characterized by 'H-NMR and LC/MS analysis
(LRMS (M+rf) m/z: 535.35).
[00324] A solution of 6 (1.3 K*i, 2.6 mmol) in an excess of ammonium acetate in
formamide (10 mL) was heated to 130 °K*I under a nitrogen atmosphere for 3 hours. The
resulting mixture was cooled to room temperature, partitioned between water and extracted
with dichloromethane (3 x 50 mL). The combined organic layers were dried over sodium
sulfate and concentrated in vacuo. The residue was purified by flash chromatography (silica
gel, ethyl acetate/hexane) providing imidazole 7 (0.8 K*i, 60%), which was characterized by *H-
NMR and LC/MS analysis (LRMS (M+H+) m/z: 516.35).

■, 00325] A solution of 7 (800 mg, 1.55 mmol) in tetrahydrofuran (10 mL) was stirred
with hydrogen chloride in 1,4-dioxane (10 mL, 4.0 M) at room temperature for one hour. The
solvents were removed in vacuo, and the residue was dried under high vacuum overnight to
yield intermediate 8 (600 mg), which was determined to be pure enough for the next
transformation ('H-NMR and LC/MS (LRMS (M+H*) m/z 302.22)).
[00326] To a room temperature solution of amine 8 (60 mg, 0.02 mmol) in
dimethylformamide (3 mL) was added diisopropylethylamine (53 ul, 0.3 resulting solution stirred at room temperature for 5 minutes. Iintermediate 9 (23 mg, 0.06
mmol) was then added, and the reaction mixture was stirred under an atmosphere of nitrogen
for 30 minutes. The solvents were removed in vacuo, and the residue purified by flash
chromatography (silica gel, methanol/dichloromethane) to provide 1O (25 mg, 26%) as a
glassy solid, which was characterized by ]H NMR and LC/MS (LRMS (M+H+) m/z\ 489.23).

]00327] To a room temperature solution of 1 (4.96 K*i, 17 mmol) in methanol (15 mL)
was added dropwise a solution of TMS diazomethane in hexanes (17.0 mL, 34 mmol, 2 M).
The resulting yellow solution was stirred at ambient temperature for 30 minutes. The solvents
were removed in vacuo, and the v-scous oil 2 (5.19 K*i, 17 mmol) was dried under high vacuum
and determined to be pure enough for use in subsequent transformations (LC/MS (LRMS
(M+H*) m/z: 305.3)).

[00328] Intermediate 2 (5.19 K*i, 17 mmol) was stirred with sodium borohydride (3.23 K*i,
85 mmol) in methanol (50 mL) and tetrahydrofuran (50 ml) at room temperature for 2 hours.
The solvents were removed in vacuo, and the residue was partitioned between ethyl acetate
(50 mL) and water (50 mL). The layers were separated and the aqueous phase was extracted
with ethyl acetate (3 x 50 mL). and the combined organic layers were dried over sodium
sulfate and concentrated in vacuo to yield 3 (4.71 K*i, 17 mmol) as a white solid, which was
determined to be pure enough for use in subsequent transformations LC/MS (LRMS (M+H*)
m/z: 277.3). Nitrile 3 (1.92 K*i. 6.9 mmol) was stirred with sodium methoxide in methanol
(27.7 mL, 13.9 mmol, 0.5 M) and hydroxylamine hydrochloride (964 mg, 13.9 mmol) under
an atmosphere of nitrogen at 50 °K*I for 2 hours. It was then cooled to room temperature and
the solvents were removed in vacuo. The residue was partitioned between saturated aqueous
ammonium chloride solution (30 mL) and ethyl acetate (30 mL). The layers were separated
and the aqueous phase was extracted with ethyl acetate (2 x 30 mL). The combined organic
extracts were dried over sodium sulfate and concentrated in vacuo, and the residue was
purified by flash chromatography (silica gel, hexane/ethyl acetate) to yield intermediate 4
(1.08 K*i, 51%), which was characterized by *H NMR and LC/MS (LRMS (M+H+) m/z: 310.2).
100329] To a room temperature solution of 4 (1.08 K*i, 3.5 mmol) in methanol (30 mL)
was added Raney nickel (200 mg) and acetic acid (1 mL). The resulting mixture was stirred
under an atmosphere of hydrogen at room temperature for 2 hours. It was filtered through
Celite and concentrated in vacuo to provide 5 (1.02 K*i, 100%) as a white solid, which was
determined to be pure enough for use in subsequent transformations (LC/MS (LRMS (M+H'1")
m/z: 294.3)).
100330] To a room temperature solution of amidine 5 (304 mg, 1.0 mmol) in
anhydrous ethanol (15 mL) was added l,8-diazabicyclo[5.4.0]undec-7-ene (622 µL, 4.2
mmol) and 3-bromo-l,l,1-trifluoro-2-butanone (424 mg, 2.1 mmol). The resulting mixture
was stirred under an atmosphere of nitrogen at 115 °K*I for 30 minutes. It was then cooled to
room temperature and the solvents removed in vacuo. The residue was purified by reverse-
phase HPLC using a mobile phase gradient consisting of acetonitrile and water. Compound 6
(76 mg, 20%) was isolated and characterized by 'H NMR and LC/MS (LRMS (M+H^) m/z:
400.1).
[00331] A solution of 6 (76 mg, 0.2 mmol) in dichloromethane (2 mL) was stirred with
trifluoroacetic acid (2 mL) at room temperature for 45 minutes. The solvents were removed in

vacuo to provide 7 (57 mg, 100%), which was determined to be pure enough for use in
subsequent transformations (LC/MS (LRMS (M+H4") m/'z: 300.3)).
[00332] To a room temperature solution of amine 7 (25 mg; 0.0S mmol) in
dimethylforrnamide (3 mL) was added diisopropylethylamine (87 ul. 0.50 mmol). The
resulting solution was stirred at room temperature for 5 minutes and intermediate 8 (32 mg,
0.08 mmol) was added. The reaction mixture was stirred under an atmosphere of nitrogen at
room temperature for 30 minutes, and the solvents were removed in vacuo. The residue was
partitioned between ethyl acetate (5 mL) and water (5 mL), after which the layers were
separated and the aqueous phase was extracted with ethyl acetate (3 x 10 mL). The combined
organic layers were dried over sodium sulfate and concentrated in vacuo. The residue was
purified by flash chromatography (silica gel, ethyl acetate) to provide 9 (7 mg, 18%) as a
glassy solid, which was characterized by ]H NMR and LC/MS (LRMS (M+H+) m/z: 496.4).

100333] To a room temperature solution of aniline 1 (500 mg, 1.3 mmol) in
dimethylforrnamide (3 mL) were added potassium carbonate (1.5 K*i) and 1-bromopinacolone
(500 mg. 2.8 mmol). The reaction mixture was stirred at 50 °K*I for 4 hours, and the solvents
were removed in vacuo. The residue was partitioned between ethyl acetate (50 mL) and water
(15 mL), the layers were separated, and the aqueous phase was extracted with ethyl acetate (3
x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated in
vacuo. The residue was purified by flash chromatography (silica gel, ethyl acetate) to provide
2 (320 mg; 51%), which was characterized by 'H NMR and LC/MS (LRMS (M+H+) m/z:
479.74).
100334] To a room temperature solution of 2 (307 mg, 0.64 mmol) in triethyl

,,/thoformate (20 mL) was added concentrated aqueous HC1 (25 fiL). The resulting mixture
was stirred at 90 °K*I for 3 hours and then cooled to room temperature. The solvents were
removed in vacuo and the residue partitioned between water (15 mL) and ethyl acetate (50
mL). The layers were separated and the organic layer washed with water (3 x 20 mL) and
brine (3 x 20 mL), and dried over sodium sulfate. Removal of the solvents yielded
intermediate 3 (326 mg, 100%) as a viscous oil, which was characterized by LC/MS (LRlvlS
{M+H+) m/z: 507.1).
[00335] Intermediate 3 (207 mg, 0.41 mmol) was stirred with ammonium acetate (1.57
K*i, 20.40 mmol) in formamide under an atmosphere of nitrogen at 130 °K*I for 4.5 hours. The
resulting solution was cooled to room temperature and partitioned between ethyl acetate (50
mL) and water (10 mL). The layers were separated, the organic layer was washed with water
(4 x 10 mL) and brine (20 mL) and dried over sodium sulfate. The solvents were removed in
vacuo, and the residue was purified by reverse-phase HPLC using a mobile phase gradient
consisting of acetonitrile and water to yield imidazole 4 (159 mg. SO%), which was' isolated
and characterized by 'H NMR and LC/MS (LRMS (M+H4) m/z: 488.2).
[00336J To a room temperature solution of 4 (159 mg, 0.33 mmol) in dichloromethane
(4 mL) was added trifluoroacetic acid (4 mL), and the resulting solution stirred at room
temperature overnight. The solvents were removed in vacuo provide amine 5 (89 mg) as a
glassy solid, which was determined to be pure enough for use in subsequent transformations
LC/MS (LRMS (M+H+) m. =: 274.1).
[00337] Crude amine 5 (72 mg, 0.26 mmol) was stirred with diisopropylethylamine
(197 ul, 1.1 mmol) in dimethylformamide (3 mL) at room temperature for 5 minutes, after
which intermediate 6 (100 mg, 0.26 mmol) was added. The resulting mixture was stirred for
another 30 minutes and the solvents removed in vacuo. The crude residue was purified by
reverse-phase HPLC using a mobile phase gradient consisting of acetonitrile and water to
give compound 7 (10 mg, 8%) as a glassy solid, which was characterized by 'H NMR and
LC/MS (LRMS (M4-H*) wz: 470.2).


[00338] To a room temperature solution of alcohol 1 (2 59 K*i, 9.4 mmol) in benzene (50
mL) was added 2.2-dimethoxypropane (1.75 mL, 14.1 mmol) and p-toluenesulfonic acid (179
mg, 0.94 mmol). The resulting solution was stirred under an atmosphere of nitrogen at 110 °K*I
for 1.5 hours. The solvents were removed in vacuo, and the residue purified using flash
chromatography (silica gel, ethyl acetate/hexanes) to provide 2 (765 mg, 27%), which was
characterized using LC/MS (LRMS (M+H+) m/z\ 317.4).
[00339] Nitrile 2 ("65 mg, 2.4 mmol) was stirred with sodium methoxide in methanol
(10.0 mL, 5.0 mmol. 0.5 M) and hydroxylamine hydrochloride (336 mg, 4.8 mmol) under an
atmosphere of nitrogen at 50 K*i for 2 hours. It was then cooled to room temperature and the
solvents removed in vacuo. The residue was partitioned between saturated aqueous
ammonium chloride solution (30 mL) and ethyl acetate (30 mL), the layers were separated,
and the aqueous phase extracted with ethyl acetate (2 x 30 mL). The combined organic
extracts were dried over sodium sulfate and concentrated in vacuo. The residue was purified
by flash chromatography (silica gel, hexane/ethyl acetate) to vield intermediate 3 (314 mg,
38%), which was characterized by 'H NMR and LC/MS (LRMS (M+H4) mt\ 350.1).
[00340] To a room temperature solution of 3 (314 mg. 0.9 mmol) in methanol (15 mL)
was added Raney nickel (50 mg) and acetic acid (300 µL). The resulting mixture was stirred
under an atmosphere of hydrogen at room temperature for 2 hours. It was filtered through

Celite and concentrated in vacuo to provide 4 (275 mg, 0.83 mmol) as a white solid, which
was determined to be pure enough for use in subsequent transformations (LC/MS (LRMS
(U+U*)m/z: 414.1)).
100341] To a room temperature solution of amidine 4 (138 mg, 0.4 mmol) in
anhydrous ethanol (15 mL) was added 1.8-diazabicyclo[5.4.0]undec-7-ene (622 µL, 4.2
mmol) and 1-bromopinacolone (84 µL, 0.6 mmol). The resulting mixture was stirred under an
atmosphere of nitrogen at 115 °K*I for 30 minutes. It was then cooled to room temperature and
the solvents removed in vacuo. The residue was purified by reverse-phase HPLC using a
mobile phase gradient consisting of acetonitrile and water to give compound 5 (29 mg, 17%),
which was characterized using 'H NMR and LC/MS (LRMS (M+H") m/z: 414.1).
[00342] To a room temperature solution of imidazole 5 (29 mg, 0.07 mmol) in
anhydrous dimethylformamide (5 mL) were added potassium carbonate (39 mg, 0.28 mmol)
and dimethylsulfate (133 µL, 1.40 mmol). The resulting mixture was stirred under an
atmosphere of nitrogen at 50 °K*I for 24 hours, after which the solvents were removed in
vacuo. The residue was purified using Hash chromatography (silica gel, ethyl acetate/hexanes)
to provide 6 (15 mg, 43%) as a glassy solid, which was characterized by !H "NMR and LC/MS
(LRMS (M+H+) m/z: 428.3).
[00343] A solution of 6(15 mg, 0.04 mmol) in anhydrous methanol (3 mL) and water
(300 µL) was stirred with DOWEX 50WX8-400 ion-exchange resin (100 mg) at room
temperature for 16 hours. The resin was removed by filtration and rinsed with triethylamine
(3 mL). The solvents were removed under high vacuum to provide 7 (12 mg, O,04 mmol),
which was determined to be sufficiently pure for the next transformation (LRMS (M+H*)
m/z: 288.2).
[00344] To a room temperature solution of amine 7 (12 mg, 0.04 mmol) in
dimethylformamide (3 mL) was added diisopropylethylamine (20 m1, 0.10 mmol). The
resulting solution was stirred at room temperature for 5 minutes, after which intermediate 8
(16 mg, 0.04 mmol) was added. The reaction mixture was stirred for another 30 minutes. The
solvents were removed in vacuo and the residue purified by flash chromatography (silica gel,
methanol/ dichloromethane) to provide 9 (10 mg, 50%) as a glassy solid, which was
characterized by !H NMR and LC/MS (LRMS (M+H*) m/z: 484.2).


100345] To a solution of 3oc-L-|3-homotyrosine(OBzl i (5 K*i, 13 mmol) in methanol
(200 mL) was added trimethyls lyldiazomethane (2 M in hexanes, 40 mL, 78 mmol)
dropwisely. The reagent was ct ntinuously added if necessary until bubbling ceased. The
mixture was concentrated to gi\ e 2 (5.5 K*i), which was used in the next step without further
purification. LRMS (M+H4) nv i 300.3.
(O0346) To a solution of 2 (5.5 K*i, 13.76 mmol) in THF (100 mL) was added LAH (1 M
in THF, 13.7 mL, 13.7 mmol); t 0 °K*I. The resulting solution was stirred for 2 hours, and
methanol (~20 mL) was added to quench the reaction. The solvents were then evaporated to

obtain the yellowish solid which was diluted in ethyl acetate and washed in saturated
NaHCCb. The organic layer was washed with brine, dried over Nu2SO4 and concentrated
under reduced pressure. The residue was purified via flush column chromatography using a
mixture of ethyl acetate and hcxanes as eluent to give 3 as a white solid (3.5 K*i, 70%). LRMS
(M+H+) m/z 394.4.
[00347] A solution of 3 (1.9 K*i, 5 mmol) in M _ A (40 m'L» was stirred under a stream
of H2 (50 psi) in the presence of 10% Pd/K*I (200 mg) for 30 h. The catalyst was removed by
filtration through a PTFE (0.45 u-m) filter and the solvent evaporated to give a white solid
(1.5 K*i), which was stirred in the mixture of TFA (1 mL) and DCM (9 mL) for 2 hours. The
resulting solution was concentrated and used in the next step without further purification.
LRMS (M+H+) w/z 182.3.
[00348] To a solution of 4 (926 mg, 5 mmol) in THF (10 mL) were added 5 (950 mg,
2.6 mmol) and N, TV-diisopropylethylamine (4.5 mL, 25.5 mmoli. The reaction was stirred at
room temperature for 10 hours. The mixture was concentrated and dried on high vacuum.
The resulting crude product was purified via flash column chromatography using ethyl
acetate as eluent to give 6(710 mg, 74 %). LRMS (M+H4) mz 370.4.
[00349] To a solution of 6 (70 mg, 0.2 mmol) in DMF (1 mL) was added 4-
fluorobenzyl bromide (0.15 mL, 1.2 mmol) and potassium carbonate (166 mg, 1.2 mmol).
The resulting mixture was stirred for 12 hours at room temperature. The mixture was filtered,
and the filtrate was purified on RP-HPLC using a mixture of acetonitrile and H2O to give le
(35 mg, 37%). LRMS (M+H*) m/z 477.5.

[00350] A solution of imidate 15 (20 mg, 0.05 mmol), pivalic acid hydrazide (9 mg,
0.08 mmol), and acetic acid (1 mL) was stirred at 80 °K*I for 1 hr. The reaction mixture was
then concentrated in vacuo and the resulting residue purified by reverse phase HPLC (CIS,
acetonitrile/water) to yield 10 mg (43%) of the tetrazole 16. LRMS (M+H") m/z 471.2,


(O0351] A solution of pentafluorophenyl ester 28 (1.0g, 2.62 mmol), amine 29 (0.49
mL, 3.15 mmol), and THF (10 mL) was stirred at 23 °K*I for 4 hours. The reaction solution was
concentrated in vacuo, and the resulting residi ie was purified by column chromatography
(silica gel, 1:1 EtOAc:hexanes) to give 1.1 gi8S%) of amide 30. LRMS (M+H+) miz 396.1.
J00352] A solution of bromide 3O (20C mg, 0.51 mol). dichlorobis(triphenylphosphine)
palladium(Il) (35 mg, 0.05 mol), tributyl(1-elhoxyvinyl)tin (0.34 ml, 1.0 mmol), and toluene
(2 mL) under N2 was stirred at 100 °K*I for 4 hours. Upon completion, as monitored by LCMS,
the reaction mixture was cooled, fleered through cotton, and concentrated in vacuo. The
resulting residue was purified by flash column chromatography (silica gel. 1:4:0.1 EtOAc:
hexanes: triefhylamine) to give 1)0 mg (52%) of styrene 31. LRMS (M+H+) mlz 388.2.
100353] A solution of con pound 31 (100 mg, 0.25.mmol), THF:H2O (3:1, 4 mL), and
N-bromo-succinimide (46 mg, 0.15 mmol) was stirred at 23 UC for 15 min. The reaction
mixture was then concentrated ii vacuo, and the crude residue was diluted with EtOAc (30

*mL), washed with brine (10 inL), and concentrated in vacuo. The resulting residue was
purified by flash column chromatography (silica gel, 4:1 EtOAc.hexanes) to give 50 mg
(46%) ofbromoketone 32. LRMS (M+H+) m/z 43S.1.
[00354] A solution ofbromoketone 32 (50 mg, 0.11 mmol), K1CO3 (47 mg 0.34
rrrmol), /en-butylcarbamidine hydrochloride (21 mg, 0.23 mmol)., and DMF (2 mL) was
stirred at 23 °K*I under N2 for 18 hours. The reaction mixture was concentrated in vacuo under
high vacuum (0.1 mm K*i), and the resulting residue was purified by column chromatography
(silica gel 2:1 EtOAc:hexanes) to give 35 mg (72%) of imidazole 34. LRMS (M+H+) m/z
440.2.



[00355] Chloroform (20 mL) was added slowly over 2 hours to a solution of Boc-
tyrosine (20 K*i, 71 mmol) and 10% NaOH in H2O (400 mL) at 85 °K*I. After a total of 4 hours,
the reaction solution was acidified with 3 N HC1 (200 mL) and extracted with EtOAc (3 x
150 mL). The organic layer was dried (MgSO.,), filtered, and concentrated in vacuo. The
resulting residue was purified by flash column chromatography (silica gel, 1:1:0.1
hexanes;EtOAc:AcOH) to yield 6.3 K*i of a mixture of aldehyde 35 and some recovered 34.
[00356] A solution of aldehyde 35 (contaminated with 34, 6:3 K*i, 20 mmol), K2CO3 (5.8
K*i, 42 mmol), benzyl bromide (5.0 mL, 42 mmol), and DMF (100 mL) was stirred at 23 °K*I for
18 hours. The reaction mixture was diluted with EtOAc (200 mL), and washed with 1 N
HCL (3 x 200 mL) and brine (3 x 200 mL). The organic layer was dried (MgSO.,), filtered,
and concentrated in vacuo, The resulting residue was purified by column chromatography
(silica gel, 1:4 EtOAc:hexanes) to give 2.2 K*i (22%) of ester 36. LRMS (M+H+) m/z 490.2.
[00357] A solution of aldyhyde 36 (570 mg, 1.16 mmol), KMnO4 (368 mg, 2.32
mmol), dioxane (3 mL), and H2O (1 mL) was stirred at 23 °K*I for 3 hours. The reaction
mixture was concentrated in vacuo and the resulting residue was purified by column
chromatography (silica gel. 1:1 EtOAc:hexanes) to give 350 mg (60%) of acid 37. LRMS
(M+H+) m/z 506.2.
[00358] A solution of acid 37 (115 mg, 0.23 mmol), dimethyl amine (0.23 mL, 2.0 M
in THF), 1-ethyl-3-(3-dimcthylaminopropyl)carbodiimide hydrochloride (65 mg, 0.34 mmol),
diisopropyl ethyl amine (0.12 mL, 0.68 mmol), and CH2Cl2 (1 mL) was stirred at 23 °K*I for 4
hours. The reaction mixture was then diluted with EtOAc (10 mL), and washed with 1 N HC1
(5 mL) and brine (5 mL). The organic layer was dried (MgSO.;), filtered, and concentrated in
vacuo. The resulting residue was purified by flash column chromatography (silica gel, 1:1
hexanes:EtOAc) to yield 60 mg (49%) of the amide 38. LRMS (M+H+) m/z 533.3.
[00359] A solution of amide 38 (60 mg, 0.11 mmol), TFA:H2O (97.5:2.5, 1 mL) and
CH2Cl2 (1 mL) was stirred at 23 °K*I for 30 min. The reaction solution was concentrated in
vacuo, and the resulting residue was placed under high vacuum for 2 hours and then used

without further purification.
[00360] A solution of amine 39 (69 mg, 0.16 mmol), pentafluorophenol ester 4O (71
mg, 0.19 mmol), diisopropylethylamine (S3 p.L, 0.68 mmol), and DMF (1 mL) was stirred at
23 °K*I for 4 hours. The reaction mixture was then diluted with EtOAc (10 mL), and washed
with 1 N HC1 (5 mL) and brine (5 mL). The organic layer was dried (MgSO4), filtered, and
concentrated in vacuo. The resulting residue was purified by flash column chromatography
(silica gel, 1:1 hexanes:EtOAc) to yield 60 mg (60%) of the ester amide 41. LRJV1S (M+H"1")
m/z 620.3.
[00361] A solution of ester 41 (50 mg, 0.08 mmol), NaBH., (30 mg, 0.81 mmol), THF
(0.5 mL), and MeOH (0.5 mL) was stirred at 23 °K*I for 2 hours. The reaction mixture was then
diluted with EtOAc (10 mL), and washed with 1 N HC1 (5 mL) and brine (5 mL). The organic
layer was dried (MgSO4), filtered, and concentrated in vacuo. The resulting residue was
purified by flash column chromatography (silica gel, 1:50 MeOI-LEtOAc) to yield 31 mg
(75%) of the alcohol 42. LRMS (M+H*) m/z 516.3.

[00362] A solution of acid 43 (300 mg, 1.0 mmol), K2CO3 (276 mg, 2.0 mmol), benzyl

bromide (0.24 mL, 2.0 mmol), and DMF (4 mL) was stirred at 23 °K*I for 18 hours. The
reaction mixture was diluted with EtOAc (30 mL), and washed with 1 N HC1 (10 mL) and
brine (3x15 mL). The organic layer was dried (MgSO4), filtered, and concentrated in vacuo.
The resulting residue was purified by column chromatography (silica gel, 1:3
EtOAc.hexanes) to give 400 mg (83%) of ester 44. LRMS (M-rH*) m/z 480.2.
[00363] A solution of ester 44 (100 mg, 0.21 mmol), NaBH4 (24 mg; 0.63 mmol), THF
(1 mL), and MeOH (1 mL) was stirred at 23 °K*I for 18 hours. The reaction mixture was then
diluted with EtOAc (10 mL). washed with 1 N HC1 (5 mL), and brine (5 mL). The organic
layer was dried (MgSO4), filtered, and concentrated in vacuo. The resulting residue was used
without further purification.
100364] A solution of alcohol 45 (100 mg, 0.27 mmol) and TFA:H2O (97.5:2.5, 1 mL)
was stirred at 23 °K*I for 30 min. The reaction solution was concentrated in vacuo, and the
resulting residue was placed under high vacuum for 2 hours and then used without further
purification.
[00365] A solution of amine 46 (40 mg, 0.15 mmol), pentafluorophenyl ester 4O (43
mg, 0.12 mmol), triefhylamine (51 µL, 0.29 mmol), and DMF (0.6 mL) was stirred at 23 °K*I
for 4 hours. The reaction mixture was then diluted with EtOAc (10 mL), and washed with 1
N HC1 (5 mL) and brine (5 mL). The organic layer was dried (MgSO4), filtered, and
concentrated in vacuo. The resulting residue was purified by flash column chromatography
(silica gel, 1:2 hexanes:EtOAc) to yield 30 mg (43%) of the ester amide 47. LRMS (M+H+)
m/z 463.2.



[00366] A solution of aldehyde 36 (300 mg, 0.6 mmol), dimethyl hydrazine (47 µL, 0.6
mmol), and MeOH (2.5 mL) was stirred at 0 °K*I for 2 hours, then allowed to warm to 23 °K*I
and stirred for an additional 15 hours. The reaction solution was concentrated in vacuo and
the resulting residue was used without further purification.
[00367] To a -5°K*I solution of crude hydrazone 48 (325 mg, 0.6 mmol) and CHC13 (2
mL) was added dropwise a solution of ;;?-chloroperoxybenzok acid (212 mg, 1.23 mmol) and
CHCI3 (2 mL). The reaction solution was allowed to warm to 23 °K*I and was stirred for 2
days. The reaction mixture was then diluted with EtOAc (10 mL), and washed with saturated
aqueous NaHCCh (5 mL) and bnne (5 mL). The organic layer was dried (MgSO-t), filtered,
and concentrated in vacuo. The resulting residue was purified by flash column
chromatography (silica gel, 1:2 hexanes:EtOAc) to yield 100 mg (34%) of the nitrile 49.
LRMS (M+H*) m/z 487.2.
[00368] A solution of nitrile 49 (60 mg, 0.27 mmol) and TFA:H2O (97.5:2.5, 2 mL)
was stirred at 23 °K*I for 30 min. The reaction solution was concentrated in vacuo, and the
resulting residue was placed under high vacuum for 2 hours and then used without further
purification.
[00369] A solution of amine 5O (100 mg, 0.25 mmol), pentafluorophenyl ester 4O (S5
mg, 0.22 mmol), triethylamine (96 p.L, 0.74 mmol), and DMF
4 hours. The reaction mixture then diluted with EtOAc (10 mL). and washed with 1 IN HC1 (5
rnL) and brine (5 mL). The organic layer was dried (MgSO4), filtered, and concentrated in
vacuo. The resulting residue was purified by flash column chromatography (silica gel, 1:1
hexanes:EtOAc) to yield 60 mg (42%) of 51. LRMS (M+H*) r.iz 574.2.
[00370] A solution of ester 51 (60 mg, 0.1 mmol), NaBtU (12 mg, 0.3 mmol), THF (1
mL), and MeOH (1 mL) was maintained at 23 °K*I for 18 hrs. The reaction mixture then
diluted with EtOAc (10 mL). washed with 1. N HC1 (5 mL), and brine (5 mL). The organic
layer was dried (MgSO.4), filtered, and concentrated 'in vacuo. The resulting residue was
purified by flash column chromatography (silica gel, 1:2 hexanes:EtOAc) to yield 30 mg
(64%) of the alcohol 52. LRMS (M+H+) m/z 470.2.

[00371] A solution of amide 55 (1.6 K*i, 4.38 mmol) and diethylaniline 56 (5 mL) was
maintained at 240 K*I for 18 hrs. The reaction solution was cooled to 23 °K*I, diluted with
EtOAc (30 mL), and washed with 1 N HCl (3 x 50 mL) and brine (2 x 50 mL). The organic
layer was dried (MgSO4).. filtered, and the filtrate was concentrated in vacuo. The resulting
residue was purified by flash column chromatography (silica gel, 2:1 hexanes:EtOAc) to yield
1 K*i (63%) of the phenol 56. LRMS (M+H+) m/z 365.2.
[00372] A solution of phenol 56 (700 mg, 1.92 mmol). Cs2CO3 (1.25 mg, 3.84 mmol),

uenzyl bromide (0.46 mL, 3.S4 mmol), and DMF (10 mL) was maintained at 50 °K*I for 2 hrs. .
The reaction mixture was diluted with EtOAc (30 mL), washed with 1 N HCL (20 mL) and
brine (3 x 30 mL). The organic layer was dried (MgSO-i), filtered, and the filtrate was
concentrated in vacuo. The resulting residue was purified by flash column chromatography
(silica gel, 1:3 EtOAc:hexanes) to give 500 mg (57%) of amide 57. LRMS (M+H+) m/z 455.2.
[00373] A solution of amide 57 (150 mg, 0.33 mmol), osmium tetroxide (8 mg, 0.03
mmol), N-mefhylmorpholine-N-oxide (182 mg, 1.55 mmol), pyridine (2.4 µL, 0.03 mmol),
THF (2 mL) and H2O (2 mL) was maintained at 23 °K*I. After 2 hrs, Celite (1 K*i), NaHSO3 (1 K*i)
and EtOAc (20 mL) were added and the resulting mixture was stirred. After 30 mins, the
reaction mixture was filtered and the resulting filtrate was concentrated in vacuo. The .
resulting residue was purified by flach column chromatography i silica gel, 3:1
EtOAc:hexanes) to give 100 mg (62%) of diol 58. LRMS (M+lD m/z 489.2.
[00374] A solution of diol 58 (52 mg, 0.11 mmol), Pb(OAc)4, and CH2C12 (2 mL) was
maintained at 23 K*I for 30 mins. The reaction mixture was then filtered through a plug of
Celite and the filtrate was concentrated to provide the aldehyde as a colorless oil.
[00375] A solution of the crude aldehyde (-50 mg, -0.11 mmol), NaBFL (24 mg, 0.63
mmol), THF (1 mL), and MeOH (1 mL) was maintained at 23 °K*I for 30 nims. The reaction
mixture then diluted with EtOAc (10 mL), washed with 1 N HCi (5 mL), and brine (5 mL).
The organic layer was dried (MgSO.;), filtered, and the filtrate was concentrated in vacuo. The
resulting residue was purified by flash column chromatography (silica gel, 2:1
EtOAc:hexanes) to give 20 mg (40%) of alcohol 59. LRMS (M+H+) m/z 459.2.



(O0376] A solution of styrene 6O (190 mg, 0.54 mmol), borane-THF (1.0 M, 0.54 mL)
was maintained at 23 °K*I for 2 his. An additional amount of borane-THF (0.54 mL) was then
added. After another 2 hrs, an third portion (0.54 mL) was added. The reaction solution was
maintained for 18 hrs, cooled to 0 °K*I, then 3 N NaOH (0.5 mL) and H2O2 (0.5 mL) was
added. After 2 hrs at 23 °K*I, the reaction mixture was diluted with EtOAc (20 mL) and washed
with brine (20 mL). The organic layer was dried (MgSC^), filtered, and the filtrate was
concentrated in vacuo. The resulting residue was purified by f ash column chromatography
(silica gel, 2:1 EtOAc:hexanes) to give 150 mg (75%) of alcohol 61. LRMS (M+Ff) m/z
372.2.
[00377] A solution of alcohol 61 (120 mg, 0.32 mmol), TFA:H:O (97.5:2.5, 4 mL) was
maintained at 23 °K*I for 30 mins. The reaction solution was concentrated in vacuo, and the
resulting residue was placed under high vacuum for 2 hours and then used without further
purification.
{00378] A solution of the above amine 62 (50 mg, 0.1 S mmol), pentafluorophenol ester
4O (82 mg, 0.22 mmol), tnethylamine (96 µL, 0.55 mmol), and DMF (1 mL) was maintained
at 23 °K*I for 2 hrs. The reaction mixture then diluted with EtOAc (10 mL), washed with 1 N
HC1 (5 mL), and brine (5 mL). The organic layer was dried (MgSO4), filtered, and the filtrate
was concentrated in vacuo. The resulting residue was purified by reverse phase HPLC (CIS,
acetonitrile/water) to yield 6 mg (7° >>) of the amide 63. LRMS (M+H+) m/z 459.2.
Example 63


[00379] To a solution of 1O (1.15 K*i, 2.71 mmol) in DCM (.100 mL) was added Dess-
Martin periodinane (2.30 K*i, 5.42 mmol). The reaction mixture was stirred for 1 h, after which
the DCM solution was washed by sodium fhiosulfate solution and sodium bicarbonate
solution, and dried over sodium sulfate. The mixture was filtered and the filtrate concentrated
under reduced pressure to give 23 (l.Og, 87%).
[00380] To a solution of 23 (30.0 mg, 0.0711 mmol) in DCM (2 mL) were added
diisopropylethylamine (37.0 µL, 0.213 mmol), 24 (12.9 µL, 0.213 mmol) and sodium
triacetoxyborohydride (20.0 mg, 0.142 mmol). The reaction mixture was stirred overnight,
and then concentrated under reduced pressure. The residue was purified on reverse phase
HPLC (C18) using a mixture of acetonitrile and H2O to give 25 (5.6 mg; 16.9%). LRMS
(M+H+) m/z 467.4.
[00381] To a solution of 23 (50.0 mg, 0.119 mmol) in methanol (2 mL) were added
diisopropylethylamine (62.0 µL, 0.356 mmol), 26 (31.1 µL, 0.356 mmol) and sodium
cyanoborohydride (22.4 mg, 0.356 mmol). The reaction mixture was stirred for overnight,
then concentrated under reduced pressure and the residue purified on reverse phase HPLC
(CIS) using a mixture of acetonitrile and H2O to give 27 (31.0 mg, 22.9%). LRMS (M+H+)
m/z 518.5.


[00382] To a solution of 1 (1.0 K*i, 4.66 mmol) in MeOH (10.0 mL) was added
SOClo (0.68 mL, 9.32 mmol). After stirring overnight at ambient temperature, the solution
was concentrated in vacuo and taken on without purification.
[00383] To a solution of 2 (~1.065 K*i crude, 4.66 mmofi in EtOH (1.5 mL) was added
N;H4'H2O (1.13 mL, 23.3 mmol). The reaction mixture was heated to reflux and stirred for 3
h. Upon cooling, the solution was treated with HiO, extracted with trice with EtOAc, dried
over MgSO4, filtered, and concentrated. Recrystallization from CHoCL yielded l.Olg 3 as
white crystals; 95% yield, 2 steps.
100384] To a solution of 3 (0.477 K*i, 2.09 mmol) in THF (S.O mL) was added
carbonyldiimidazole (0.379 K*i, 2.29 mmol). The reaction mixture was heated to reflux and
stirred for 1.5 h. Upon cooling, the solution was concentrated in vacuo and purified via flash

column chromatography (10-10% EtOAc/Hex) to yield 0.515 K*i 4 as a white solid; 97% yield.
[00385] To a solution of 4 (1.0 equiv.; typically 0.3-1.0 mmol) in CH3CN (2.0 mL)
was added the electrophile (1.1 equiv.) and K2CO3 (1.1 equiv.). The reaction mixture was
heated to 80 °K*I under microwave irradiation for 30 min followed by filtration and
concentration in vacuo, The product can be taken on without purification or purified via- flash
column chromatography (typically 10-40% EtOAc/Hex) to afford 5 in generally >90% yield.
[00386] To 5 (1.0 equiv.; typically 0.3-1.0 mmol) was added methylamine (2.0 M
solution in THF, 10.0 equiv.). The reaction mixture was heated to 100 °K*I under microwave
irradiation for 4 h followed by concentration in vacuo. The product was purified via flash
column chromatography (typically 40-80% EtOAc/Hex) to afford 6 in generally 70-85%
yield.


100387] To a stirred solution of 2-aminoacetonitrilebisulfate (2.9 K*i, 0.013 mmol) in
DCM (50 mL) was added bcnzophenone (3.48 mL, 0.0208 mmol) followed by D1EA (4.53
mL, 0.026 mmol). After stirring 18 h, the DCM solution was washed with water (50 mL),
dried over sodium sulfate, filtered, and the filtrate concentrated under reduced pressure. The
residue was purified on a flash silica gel column (hexanes:EtOAc, 1:1) to give 3 (2.40g,
82%). * •
[00388] Lithium bis(trimethylsilyl)amide (1 M solution in THF) was slowly added to a
stirred solution of 3 (1.2g, 0.00545 mol) and_p-phenylbenzyl bromide (1.08g, 0.00436 mol) in
THF (50 mL) over an acetone-dry ice bath under a nitrogen atmosphere. After lhour, the
reaction was quenched by adding methanol, and the solvent was evaporated under reduced
pressure. The residue was purified on a flash silica gel column (hexanes:EtOAc, 1:1) to
obtain 4. 4 was re-suspended in EtOAc (100 mL) and treated with concentrated HC1 (5 mL).
After stirring for 1 hour, the solvents were evaporated under reduced pressure, and the
resulting solid 5 was washed with ethyl ether (50 mL) three times and dried under vacuum
(0.39 K*i, 32.1%).
[00389] To a solution of 5 (0.39 K*i, 1.75 mmol) in DMF (10 mL) were added 6 (0.SO1
K*i, 2.11 mmol) and diisopropylethylamine (0.61 mL, 3.50 mmol) at room temperature. The
reaction mixture was stirred overnight. The solvents were then evaporated under reduced
pressure, and the residue purified on a flash silica gel column (hexane;EtOAc, 3:1) to give 7
(0.40g, 54.5% ). LCMS (M+H+) m/z 419.1.
100390] To a solution of 7 (50 mg, 0.119mmol) in DK IF (2 mL) were added sodium
azide (15.5 mg, 0.239 mmol) and ammonium chloride (12.8 mg, 0.238mmol). The mixture
was stirred at 80 °K*I overnight anc then filtered. The filtrate was purified on reverse phase
HPLC (CI 8) using a mixture of a :etonitrile and H2O to give 8 (6.40 mg, 11.6 %). LCMS
(M+H^/M/Z 462.4.


Methyl 3~cyano-4-[(1-mefh\lethyl)oxy]benzoate:

[00391] To a solution of methyl 3-cyano-4-hydroxybenzoate (82 K*i, 463 mmol; J. Med.
Chen:, 2002, 45, 5769) in dimethylformamide (800 mL) was added 2-iodopropane (93 mL,
926 mmol) and potassium carbonate (190 K*i, 1.4 mol). The resulting mixture was heated at 50
°K*I for 16 h, at which time it was allowed to cool to room temperature. The reaction was
filtered and the mother liquor diluted with 0.5 N sodium hydroxide (1 L). The resulting
mixture was extracted with ether (2 x 1 L) and the organics washed with 1 N HC1 (1 L) and
brine (700 mL), dried (MgS(X;) and concentrated to give 100 K*i ('-100%) of methyl 3-cyano-4-
[(1-methylethyl)oxy]benzoate as a yellow solid.
3-Cyano-4-[(1-methylethyl)oxy]benzoic acid: ^ •

[00392] To a cooled (O °K*I) solution of methyl 3-cyano-4-[(1-methylethyl)oxy]benzoate
(100 K*i, 463 mmol) in tetrahydrofuran (500 mL) was added 10% potassium hydroxide (500
mL). The resulting solution was allowed to warm to room temperature and maintained for 16
h, at which time it was concentrated to remove the tetrahydrofuran. The residue was diluted
with water (500 mL) and washed with ether (2 x 500 mL). The aqueous layer was then
acidified with 3 N HC1 and stood for 2 h. The solids were collected by filtration and washed
several times with water, then dissolved in methylene chloride (1 L). The mostly
homogeneous mixture was filtered through Celite and concentrated to a minimal volume of
methylene chloride. Collection of the solids by filtration gave 82 K*i (87%) of 3-cyano-44^(1-
methylethyl)oxy]benzoic acid as a white solid.
Scheme B:


Reagents and Conditions: a) 4N HCl/dioxane, rt; b) HBTU, /-Pr:NEt, DMF, rt; K*i) 1-
ethoxyvinyltri-n-butyltin, PdCLCPPh:,^, dioxane, 100 °K*I; d) NBS, THF/H2O (3:1), rt; e) 2-
amino-3-picoline, NaHCO3. z-PrOH, 80 °K*I .
(3S)-3-Amino-4-(4-bromophenyl)-1-butanol hydrochloride.

[00393] U-Dimethylethyl {(15)-1-[(4-bromophenyl)methyl]-3-
hydroxypropyl) carbamate (4.4 K*i, 12.8 mmol) was dissolved in 4IM HCl/dioxane (20 mL).
After 2 h, the reaction mixture was concentrated in vacuo to gi\ e 3.69 K*i (94%) of the title
compound as a white solid. LC/MS (ES) m/'e 244.O (M + H)+.
Ar-{(15}-1-[(4-Bromophenyl)methyl]-3-hydroxypropyl}-3-cyano-4-[(1-
methylethyl)oxy]benzamidc.

[00394] To a suspension of (35)-3-Amino-4-(4-bromophenyl)-1-butanol hydrochloride
(1.80 K*i, 6.42 mmol) in dry DMF (32 mL) was added JV,N-diisopropylethyl amine (2.49 K*i, 19.3
mmol) and the resultant clear solution was stirred for 3 min. ?-Cyano-4-[(1-
methylethyl)oxy]benzoic acid (1.45 K*i, 7.06 mmol) and HBTU (2.68 K*i, 7.06 mmol) were
added and the reaction was stirred at rt under nitrogen. After 1.5 h. the reaction mixture was
quenched with water (50 mL) and extracted with EtOAc (3X30 mL). The extracts were

tided (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified
by silica gel chromatography (75% EtOAc/hexanes) to give 2.IS K*i (78%) of the title
compound as a white solid. I.K*I/MS (ES) m/e 431.2 (M + H)+.
JV-((15)-1-([4-(Bromoacetyl)phenyl]methyl}-3-hydroxypropyl)-3-cyano-4-[(1-
methylethyl)oxy]benzamide.

J00395] A flask, dried with a heat gun under argon purge, was charged with N-{(lS)-\-
[(4-bromophenyl)methyl]-3-hydroxypropyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide (1.0
K*i. 2.32 mol). dichlorobis(triphenylphosphine)-palladium(Il) (81 mg, 0.116 mol), tributyl(1-
ethoxyvinyl)tin (1.68 K*i, 4.64 mmol), and 1.4-dioxane (15 mL). The mixture was stirred at
100 °K*I for 2 hours under argon. Upon completion, as monitored by LCMS, the reaction was
concentrated under reduced pressure and the residue was purified immediately on deactivated
silica gel (65% EtOAc/hexanes with 5% triethylamine) to give "20 mg (1.70 mmol) of enol
ether intermediate as a colorless foam which was immediately dissolved in THFiHbO (3:1, IS
mL) and treated with TV-bromosuccinimide (318 mg, 1.79 mmol). After 15 min at rt, the
reaction mixture was concentrated under reduced pressure and the crude residue was diluted
with EtOAc (30 mL), washed with brine (10 mL) and water (10 mL) and concentrated under
reduced pressure. The residue was purified by silica gel chromatography (80%
EtOAc/hexanes) to give 651 mg (59%) of iV-((15)-1-{[4-(bromoacetyl)phenyl]methyl}-3-
hydrox\propyl)-3-cyano-44L(1-methylethyl)oxy]benzamide as a white tacky solid. LC/MS
(ES)mv'e 473.2 (M + H)+,
3-Cyano-yV-((lc7)-3-hydrox)'-1-{[4-(8-methylimidazo[1,2-a]p\Tidin-2-
yl)phenyl]methyl}propyl)-4-[(1-mef iylethyl)oxy]benzamide.

[00396] To a solution of N-( ,15)-1-{[4-(bromoacetyl)phenyl]methyl)-3-

nydroxypropyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide (300 mg, 0.634 mmol) in i-PrOH
(6 mL) was added 2-amino-?-picoline (Aldrich, 69 mg, 0.634 mmol) followed by solid
NaHCO3 (64 mg, 0.761 mmol). The resultant suspension was heated to 80 °K*I, After 7 h, a
majority of the i-PrOH was removed under reduced pressure and the residue was dissolved in
3% MeOH/EtOAc (30 mL) and washed with water (10 mL) and brine (10 mL). The
combined aqueous layers were extracted with 3% MeOH/EtOAc (30 mL) and the combined
extracts were dried (NaiSCXt). filtered and concentrated under reduced pressure. The residue
was purified by reverse phase HPLC (MeCN/LLO with 0.1 % TFA) and the clean fractions
were adjusted to pH ~8 with saturated aqueous NaHCOs and exiracted with 3%
MeOH/EtOAc (3 X 30 mL). The extracts were dried (Na2SO4K filtered and concentrated
under reduced pressure to give 215 mg (70%) of the title compound as a pale yellow solid.
LC/MS (ES) m/e 4S3.2 (M + H)+.

1-(2-amino-3-pyridinyl)efhanol:

[00397] To a dry flask (dried with a heat gun under argon purge) was added dry THF
(400 mL) and MeLi-LiBr (137 mL of a 1.5M solution in Et2O, 204.9 mmol) via cannula.
This solution was cooled to -78 °K*I when a solution of 2-aminopyridine-3-carboxaldehyde
(10.0 K*i, 82.0 mmol) in THF (150 mL) was added dropwise via a pressure equalizing addition
funnel over -45 with vigorous stirring (exofherm observed, orange color persisted). Upon
complete addition, the solution was allowed to stir for 1 hour at -7S °K*I, at which time TLC
(KMnO,) stain with heat) indicated that most of the starting material had been converted to
product. The reaction was quenched very carefully with water (200 mL; dropwise initially),
diluted with EtOAc (200 mL) and allowed to warm to rt. The layers were separated and the

aqueous layer was extracted with 3% MeOH in EtOAc. The combined extracts were dried
over sodium sulfate, filtered and concentrated under reduced pressure. The residue was
purified by silica gel chromatography (Analogix; 0 to 5% MeOH in EtOAc) to give 7.78 K*i
(68%) of the desired racemic product as a yellow oil that solidified under high vac over
several days. This material was separated into its respective enantiomers (>9S% ee) by SFC
with a chiralcel OD-H (20x250mm) column (10% EtOH/0.1% isopropylamine in
heptane/0.1% isopropylamine).

1,1-Dimethylethyl [(15)-1-({4-[1-(ethyloxy)ethenyl]phenyl}meihyl)-3-
hydroxypropyljcarbamate:

[00398) To a solution of L1-dimethylethyl {(15)-1-[(4-bromophenyl)methyl]-3-
hydroxypropyl}carbamate (20 K*i, 58 mmol) in dioxane (500 mL) was added tributyl[1-
(ethyloxy)ethenyl]stannane (39 mL, 116 mmol) and PdC^PPhjK The resulting solution was
heated at 100 °K*I for 5 h. The reaction was then concentrated and the residue purified by flash
chromatography (47.5% EtOAc, 47.5% hexanes, 5% triethylaminc) to give 15 K*i (77%) of 1,1-
dimethylethyl [(15)-1-({4-[1-(ethyloxy)ethenyl]phenyl}methyl)-3-hydrox>'propyl]carbamate
as a brown solid.
1,1-Dimethylethyl ((15}-1-{[4-(T»romoacetyl)phenyl]me1:hYl}-3-h;--droxypropyl)carbamate:


100399] To a cooled (O °K*I) solution of 1,1-dimethylethyl [(15)-1-({4-[1-
(ethyloxy)ethenyl]phenyl}methyl)-3-hydrox>'propyl]carbamate (15 K*i, 44 mmol) in
tetrahydrofuran (450 mL) and water (150 mL) was added jV-bromosuccinamide. The
resulting solution was allowed to warm to room temperature and maintained for 90 minutes.
The reaction was then concentrated and diluted with ethyl acetate (1 L). The resulting
solution was washed with water (1 L) and bnne (500 mL), dried (MgSO^) and concentrated to
give 19.5 K*i (-100%) of 1,1-dimethylethyl ((15)-1-{[4-(bromo:tcetyl)phenyl]methyl}-3-
hydroxypropyl)carbamate as a slightly yellow solid. ESMS [IvH-H]+: 386.2.
1.1-Dimethylethyl [(lS)-3-hydTOxy-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-n]pyiidin-2-
yl]phenyl} methyl)propyl] carbamate

[00400] A mixture of 1,1-dimethylethyl ((lS)-1-{[4-(bromoacetyl)phenyl]methyl}-3-
hydroxypropyl)carbamate (1.00 K*i, 2.59 mmol), 1-(2-amino-3-pyridinyl)ethanol (0.358 K*i, 2.59
mmol), and solid sodium bicarbonate (0.272 K*i, 3.24 mmol) in isopropanol (25 mL) was
heated at reflux for 3.5 h. and concentrated in vacuo. The residue was dissolved in ethyl
acetate, washed with water and brine, dried (NajSO^, and concentrated. The resulting pale
yellow solid was used in the next reaction without further purification. MS(ES+) m/e 426
[M+H]+.
3-Chloro-Ar-[(15)-3-hydroxy-1-({4-[8-(1-hydroxyethyl)imidazo[1J2-a]pyridin-2-
yl]phenyl)methyl)propyl]-4-[(1-methylethyl)oxy]benzamide


[00401) A mixture of L1-dimethylethyl [(15)-3-hydroxy-1-({4-[8-t1-
hydroxyethyl)imidazo[1,2-a]p>iidin-2-yl]phenyl}methyl)propyl]carbamate (1,08 K*i. 2.54
mmol) and 4M HC1 in 1,4-dioxane (8.0 mL, 32 mmol) was stirred at room temperature for
30 minutes. The reaction was concentrated to dryness ,redissolved in DMF (25 mL), and to
this solution was added AW-diisopropylethylamine (1.64 K*i, 12.7 mmol) and
pentafluorophenyl 3-chloro-4 [(1-methylethyl)oxy]benzoate (0.963 K*i, 2.54 mmol). The
mixture was stirred for 3.0 h at room temperature, diluted with water, and extracted into ethyl
acetate. The extracts were washed with water and saturated sodium chloride, dried (Na2SO4),
and concentrated in vacuo. The residue was purified by flash chromatography on silica gel
(2% MeOH:EtOAc) to give the title compound (0.7 K*i, 53%) as a pale yellow powder.
MS(ES+) m/e 522 [M+H]+.
Scheme E:


1,1-Dimethylethyl [(1 S)-2-(4-bromophenyl)-1-(hydroxymethyl)ethyl]carbamate:

100402) To a solution of 4-^01x10-/^- {[(1T -dimethylethyl)oxy]carbonyl}-L-
phenylalanine (72.6 mmol), in anhydrous diethyl ether (550 mL) at 0 °K*I was added slowly
lithium aluminum hydride, 95% (108.9 mmol). The resulting solution was stirred for an
additional 2 h at 0 °K*I. The reaction was then carefully quenched with a saturated aqueous
solution of sodium bicarbonate (73 mL) which stirred at RT for half an hour. Lithium
aluminium salts crashed out of solution which were removed by filtration. The filtrate was
concentrated and vacuum pumped for 24 h to afford the title product as a white solid (97%).
ESMS [M+H]+: 331.2.
1,1-Dimethylethyl {(lS)-2-(4-bromophenyl)-1-[(l,3-dioxo-l,3-dihydro-2H-isoindo1-2-
yl)rnethyl]ethyl}carbamate:


[00403] To a solution of 1,1-dimethylethyl [(l.S)-2-(4-bromophenvT)-1-
(hydrox>Tnethyl)ethyl]carbamate (70.6 mmol), tripheylphosphine (84.7 mmol), and
phthalimide (84.7 mmol) in anhydrous tetrahydrofuran (550 mL) at 0 °K*I was added dropwise
diisopropyl azodicarboxylate (84.7 mmol) over 10 minutes. The reaction continued to stir
allowing to warm to RT over 5h. The reaction was then concentrated in vacuo and product
was tritarated out of solution usingl acetate (500 mL). The precipitate was filtered, washed
with ethyl acetate (3 x 100 mL), and dried to afford the title product as a white solid (57%).
ESMS [M+H]+: 460.4.
1,1-Dimethylethyl {(15)-2-[4-(bromoacetyl)phenyl]-1-[(l,3-dioxo-l,3-dihydro-2//-isoindo1-
2-yl)methyl]ethyl} carbamate:

[00404] A solution of L1-dimethylethyl {(lS)-2-(4-bromophenyl)-1-[(l,3-dioxo-l,3-
dihydro-2//-isoindo1-2-yl)methyl]ethyl) carbamate (21.7 mmol), 1-ethoxyvinyltri-n-butylin
(43.5 mmol), and /r«/?s-dichlorobis(triphenylphospine)palladium(ll) (5 mol%) were stirred in
anhydrous dioxane (300 mL) at 100 °K*I for 3h. The reaction was then concentrated in vacuo
and redissolved in a solution of tetrahydrofuran and water (3:1, 400mL) and treated with N-
bromosuccinimide (108.8 mmol) and stirred at RT for half an hour. The reaction solution
was then concentrated to dryness and redissolved in ethyl acetate (150 mL) and precipate
formed upon addition of hexanes (350 mL). The precipitate was filtered and dried to afford
the title product as yellow solid (71%). ESMS [M+H]+: 502.4.

l,1-Dimethylethyl[(15)-2-(l,3-dioxo-l,3-dihydro-2/f-isoindo1-2-yl)-1-({4-[S-(1-
hydroxyethyl)imidazo[1,2-a]p}Tidin-2-yl]phenyl}methyl)ethyl]carbamate:

[00405J A mixture of L1-dimethylethyl {(1 S)-2- {4-(bromoacetyl)phenyl]-l -[(1,3-
dioxo-l,3-dihydro-2//-isoindo1-2-yl)methyl]ethyl}carbamate(1.90 K*i, 3.79 mmol), 1-(2-
amino-3-pyridinyl)ethanol (0.523 K*i, 3.79 mmol),'and solid sodium bicarbonate (0.398 K*i, 4.72
mmol) in isopropanol (24 mL) was refluxed for 3.0 h. and concentrated in vacuo. The residue
was dissolved in ethyl acetate, washed with water and saturated sodium chloride, dried
(NaiSOa), and concentrated to give the title compound (1.79 K*i, 87%) as a light pink solid.
MS(ES+) m/e 541 [M+H]+.
3-Chloro-iV-[(15)-2-(l,3-dioxo-l)3-dihydro-2H-isoindo1-2-yl)-1-({4.[8-(1-
hydroxyethyl)imidazo[1,2-fl]pyridin-2-yl]phenyl}methyl)ethyl]-44^(1-
methylethyl)oxy]benzamide:

[00406] A mixture of 1,1-dimethylethyl [(lS)-2-(l,3-dioxo-l,3-dihydro-2H-isoindo1-2-
yl)-1-({4-[8-(1-hydroxyeth}'l)imidazo[1,2-fl]pyridin-2-yl]phenyl}methyl)ethyl]carbamate
(1.79 K*i, 3.31 mmol) and 4M HC1 in 1,4-dioxane (20 mL, 80 mmol) was stirred at room
temperature for 45 minutes. The reaction was concentrated to dryness ,redissolved in DMF
(30 mL), and to this solution was added yV.A'-diisopropylethylamine (2.14 K*i, 16.55 mmol) and
pentafluorophenyl 3-chloro-4 [(1-methylethyl)oxy]benzoate (1.36 K*i, 3.31 nunol). The

mixture was stirred overnight at room temperature, diluted with water, and extracted into
ethyl acetate. The extracts were washed with water, dried (NaaSO^), and concentrated in
vacuo to give the title compound (2.10 K*i, 100%) as a tan solid. MS(ES+) m/e 637 [M+H]+.
Ar-[(15)-2-Amino-1-({4-[8-(1-hydroxyethyl)imidazo[1,2-a]p>Tidin-2-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide:

[00407] A mixture of 3-chloro-Ar-[(15)-2-(l,3-dioxo-l ,3-dihydro-2//-isoindo1-2-yl)-1-
({4-[8-(1-hydroxyethyl)imidazo[1,2- methylethyl)oxy]benzamide (2.10 K*i, 3.30 mmol) and hydrazine monohydrate (0.83 K*i, 16.5
mmol) in ethanol (30 mL) was heated at 57°K*I overnight. The reaction was cooled, diluted
with ethanol, filtered, and concentrated to give the title compound(1.67 K*i, 100%) as a pale
yellow powder. MS(ES+) nve 507 [M+H]+.
3-Chloro-Ar-[(15)-2-[(Ar^-dimethylglycyl)amino]-1-({4-[8-(1-h}droxyethyl)imidazo[1,2-
«]p>Tidin-2-yl]phenyl}methy])ethyl]-4-[(1-methylethyl)oxy]benzamide:

[00408] A mixture of jV-[(15)-2-amino-1-((4-[8-(1-hydroxyethyl)imidazo[1,2-
a]p>Tidin-2-yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide (0.912 K*i,
1.80 mmol), EDCI (0.69 K*i, 3.6 mmol), A^jV-diisopropylethylamine (0.466 K*i, 3.6 mmol), and
JV,Ar-dimethylglycine (0.372 K*i. 3.6 mmol) in methylene chloride (17 mL) was stirred
overnight at room temperature. The reaction was diluted with water, washed with brine, dried
(Na2SO4), and concentrated. The residue was purified by flash chromatography on silica gel

(8%-l 0% MeOH:CH2Cl2) to give the title compound (0.515 K*i, 48%) as a pale yellow solid.
MS(ES+) m/e 592 [M+Hf.
1,1-Dimethylethyl {(lS)-2-[4-(S-bromoimidazo[1f2-a]pyridin-2-yl)phenyl]-1-[(l,3-dioxo-l,3-
dihydro-2H-isoindo1-2yl)methyl]ethyl) carbamate:

100409] A solution of 1,1-dimethylethyl {(lS)-2-[4-(brornoacetyl)phenyl]-1-[(l ,3-
dioxo-l,3-dihydro-2H-isomdo1-2-yl)methyl]ethyl}carbamate (6.9 mmol), 3-bromo-2-
pyridinamine (S.4 mmol), and sodium bicarbonate (10.4 mmol) in isopropanol (70 mL) were
stirred at 80 °K*I for 18 h. The reaction was then cooled to RT and a precipitate formed which
was filtered, washed with cold hexanes (3 x 100 mL), and dried to afford the title compound
as light gray solid (72%). ESMS [M+H]+: 576.2.
l,1-Dimethylethyl((15)-2-(L3-dioxo-l,3-dihydro-2//-isoindo1-2-yl)-1-{[4-(8-
methylimidazo[1,2-a]pyiidin-2-yl)phenyl]methyl}ethyl)carbamate:

[00410] Following the procedure described above with 3-methyl-2-pyridinamine,
instead of 3-bromo-2-pyridinamine, provided the title product as a light pink solid. ESMS
[M+H]+: 511.0.
N'-{(15)-2-[4-(8-Bromoumdazo[1,2-a]p>Tidin-2-yl)phenyl]-1-[(l,3-dioxo-l,3-dihydro-2H-
isoindo1-2-yl)methyl]ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide:


[00411] A solution of 1,1-dimethylethyl {(15)-2-[4-(8-bromoimidazo[1,2-fl]pyridin-2-
yl)phenyl]-1-[(l ,3-dioxo-l,3-dihydro-2//-isoindo1-2 y])methyl]ethyl}carbamate (3.5 mmol)
and hydrogen chloride in 1,4- dioxane (20 mL, 4.0M) stirred for lh at RT. The reaction was
concentrated to dryness and redissolved in A^N-dimethylformamide (35 mL). Added to-the
solution was diisopropylethylamine (10.5 mmol) and pentafluorophenyl 3-chloro-4-[(1-
methylethyl)oxy]benzoate (3.8 mmol), which was stirred at RT for half an hour. The reaction
was dissolved in ethyl acetate (SO mL) and washed with water (3 x 50 mL) and brine (1 x 50
mL). To the separated organic layer was added hexanes (150 mL) upon which a.precipitate
was formed, filtered, and dried to afford the title compound as an off white solid (65%).
ESMS [M+H]+: 672.2.
3-Chloro-Ar-((l1S)-2-(l,3-dioxo-l,3-dihydro-2^-isoindo1-2-yl)-1-{[4-(8-methylimidazo[1,2-
a]pyiidin-2-yl)phenyl]methyl} ethyl )-4-[(1-methylethyl)oxy]benzamide:

[00412J Following the procedure described above with 1,1-dimethylethyl ((15)-2-(l,3-
dioxo-l;3-dihydro-2//-isoindo1-2-yl)-1-{[4-(S-methylimidazo[1,2-a]p)Tidin-2-
yl)phenyl]methyl}ethyl)carbarnate provided the title product as an off white solid. ESMS
[M+H]+: 608.2.
Ar-((l^-2-Amino-1-{[4-(S-bromoimidazo[1,2-fl]pyridin-2-yl)phenyl]methyl}ethyl)-3-chloro-
4-[( 1-methylethyl)oxy]benzamide:


[00413] To a solution of N- {(15)-2-[4-(8-bromoimidazo[1 .2-a]pywdin-2-yl)phenyl]-1-
[(13-dioxo-l,3-dihydvo-27f-isoindo1-2-yl)methyl]ethyl}-3-chloro-4-[(1-
methylethyl)oxy]benzamide (1.5 mmol) in ethanol (10 mL) was added hydrazine
monohydrate (7.6 mmol). The reaction stirred for 18h at 50 °K*I upon which a white
precipitate formed and filtered. The filtrate was concentrated in vacuo. The resultant light
yellow solid was used directly in the next reaction without further purification. ESMS
[M+H]+: 533.2
Ar-((1 S)-2- Amino-1- {[4-(S-methylimidazo[1 .2-«]pyridin-2-yl)phenyl]methyl) ethyl)-3-chloro-
4-[(1-methylethyl)oxy]benzamide:

[00414] Following the procedure described above with 3-chloro-JV-((1 S)-2-(l .3-dioxo-
l,3-dihydro-2H-isoindo1-2-yl)-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-f(1-methylethyl)oxy]benzamide provided the title product as an off
white solid. ESMS [M+Hf: 478.2.
Ar-((15r)-2-(D-Alanylamino)-1-{[4-(8-bromoimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}ethyl)-
3-chloro-4-[(1-methylethyl)oxy]benzamide:


[00415] A solution of Ar-((l1S)-2-amino-1-{[4-(8-bromoimidazo[1^-a]p}'ridin-2-
y^pheny^methylJethyO-S-chloro^-tC1-methylethyOoxyJbenzamide (0.28 mmol), JV-{[(1,1-
dimethylethyl)oxy]carbonyl}-D-alanine (0.56 mmol), EDCI (0.56 mmol), and TEA (1.12
mmol) stirred in methylene chloride (2 mL) at RT for 18 h. The reaction was then treated
with 4 M HC1 in 1,4-dioxane (2mL) and stirred at RT for 1 h and concentrated in vacuo;
redissolved in ethyl acetate (25 mL) and washed with saturated aqueous sodium bicarbonate
solution (1 x 10 mL). The organic layer was concentrated in vacuo. Purification of the
residue by Gilson reverse phase HPLC afforded the title product as a white solid (25%).
ESMS [M+H]+: 613.2.
3-CWoro-Ar-((15)-2-[(2-methylalanyl)amino]-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl} ethyl)-4-[( 1-methylethyl)oxy]benzamide;

[00416] Following the procedure described above with -V-((15)-2-amino-1-{[4-(8-
methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl)ethyl)-3-chloro-4-[(t-
methylethyl)oxy]benzamideandA-{[(l>1-dimethylethyl)oxy]carbonyl}-2-methylalanine
provided the title product as a white solid. ESMS [M+H]+: 563.2.
3-Chloro-Ar-((l 5)-2-[(Ar^-dimethylglycyl)amino]-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide:


100417] Following the procedure described above with N-((15)-2*amino-1-{[4-(8-
methylimidazo[1,2-o]p>Tidin-2-yl)phenyl]methyl} ethyl)-3-chloro-4-[(l -
methylethyl)oxy]benzamide and N,N-dimethylglycine provided the title product as a white
solid. ESMS[M+Hf: 563.2.

2-Bromo-1-(4-iodophenyl)ethanone:

[00418] A solution of 1-(4-iodophenyl)ethanone (55.9 mmol) in dioxane (160 mL) was
cooled to 10 °K*I, Bromine (1.1 equiv, 61.6 mmol) was added dropwise to the reaction
mixture. After 10 min, the cooling bath was removed and the reaction mixture was stirred at
room temperature. After 1.5 h, the reaction mixture was concentrated in vacuo, poured into
water (100 mL). and extracted with (3 x 100 mL) ethyl acetate. The combined organic layers
were dried over sodium sulfate and concentrated in vacuo to a tan solid (18.2 K*i) which was
used directly in the next step.
2-(4-Iodophenyl)-8-methylimidazo[1,2-fl]pyridine:


[00419] A mixture of crude 2-bromo-1-(4-iodophenyl)ethanone (18.2 K*i), 2-amino-3-
picoline (1.1 equiv, 61.6 mmol), and sodium bicarbonate (1.3 equiv, 72.8 mmol) in
isopropanol (160 mL) was heated at SO °K*I for 16 h. After concentrating the reaction mixture
in vacuo, water (100 mL) was added and the resultant tan slurry was filtered, rinsing with
water (2 x 50 mL). The brown solid was recrystallized from hot isopropanol and further dried
in vacuo to provide the title product as a brown solid (13.2 K*i, 71%). ESMS [M+H)+: 335.0.

4-(4-Bromophenyl)-jV, 1-dimethyl-N-(methyloxy)-1 #-imidazole-2-carboxamide:

[00420] To a solution of ethyl 4-(4-bromophenyl)-1-methyl-l#-imidazole-2-
carboxylate (1.66 K*i, 5.37 mmol) in MeOH (38 mL) was added IN NaOH solution (19 mL).
The reaction turned cloudy white and was stirred at room temperature for 30 minutes. The
reaction mixture was concentrated in vacuo and pumped under high vacuum overnight to give
the sodium salt of 4-(4-bromophenyl)-1-methyl-l#-imidazole-2-carboxylic acid as a white
solid. The sodium salt of 4-(4-bromophenyl)-1-methyl-l//-imidazole-2-carboxylic acid was
dissolved in anhydrous CH2Cl2 (40 mL) under nitrogen at -15°K*I (ice/methanol bath) and N-
mefhylmorpholine (1.1 equiv, 5.91 mmol) was added followed by isobutyl chloroformate (1.1
equiv, 5.91 mmol). The reaction mixture was stirred at -15°K*I for 15 minutes and then N,0-
dimethylhydroxylamine hydrochloride (1.0 equiv, 5.37 mmol) was added. The reaction was
allowed to warm to room temperature and was stirred for 17 hours. The reaction was
quenched with H;O (10mL). The product was extracted using EtOAc (3 x 30 mL) and the
combined organic layers were washed with brine (20 mL), dried over MgSCt, and
concentrated in vacuo. Purification by silica gel chromatography (Analogix IF28O,20-100%
EtOAc/hexanes) afforded the title compound as a tan solid (32%). ESMS [M+H]+: 324.2.


14-Dimethylethyl(4J?)-4-({[(lJ-dimethylethyl)oxy]carbonyUamino)-5-hydvoxypentanoate:

100421] Triethylamine (11.49 mL, 82.4 mmol) and ethyl chloroformate (8.27 mL, 86.5
mmol) were added successively by syringe to JV-t-BOC-D-glutamic acid 5-iert-butyl ester (25
K*i, 82.4 mmol) in THF (588 mL) at min. solids were filtered and were washed with THF (150 mL). The filtrate was transferred to
a 250-mL addition funnel and added to a solution of sodium borohydride (8.42 K*i, 222.5
mmol) in H^O (114 mL) at 0 °K*I over 1 hour. The reaction mixture was maintained at 0 °K*I for
1.5 h and then stirred for 16 h (O°K*I to room temperature). After the bulk of solvents were
removed by rotary evaporation, the concentrate was quenched with ice water (50 mL) and 1 N
HC1 (50 mL). After extraction with EtOAc (4 x 100 mL), the extracts were washed with
lOOmL: 0.5 M citric acid, saturated NaHCC>3, ILO, and brine and concentrated in vacuo to
give the title compound, which was used directly in the next step. ESMS [M+H]+ = 290.4,

[2M+H]+ = 579.4. (Literature prep: J. Med. Chem, 1999, 42(1), 95-108 for other isomer).
1,1-dimethylethyl (4i?)-4-(([(l .1-dimethylethyl)oxy]carbonyl}amino)-5-iodopentanoate:

[00422] To a solution of crude 1,1-dimethylethyl (4i?)-4-({[(l,1-
dimethylethyl)oxy]carbonyl}amino)-5-hydroxypentanoate (23.8 K*i, 82.4 mmol),
triphenylphosphine (32.42 K*i, 123.6 mmol) and imidazole (8.41 K*i, 123.6 mmol) in 515 mL
anhydrous CH;Cl2 under Ni at 0°K*I was added iodine over 15 min portionwise. The ice bath
was removed and the reaction was allowed to warm to room temperature and stirred over 30
minutes. The reaction was quenched with 200 mL H^0. The aqueous layer was extracted with
diethyl ether (2 x 150 mL). The combined organic layers were washed with sat. aq. Na2SOj
solution (2 x 25 mL) and brine (25 mL), dried over MgSCXi, and concentrated in vacuo.
Purification of the residue by silica gel chromatography (Analogix IF28O, 5% - 50%
EtOAc/Hex) afforded the title compound as a white solid (25.34 K*i, 77%). ESMS [M+H]+ =
400.4.
1,1-dimethylethyl (4i?)-4-({[(l, 1-dimethylethyl)oxy]carbonyl} amino)-5-[4-(8-
methylimidazo[ 1,2-a ]pyridin-2-yl)phenyl]pentanoate:

100423] A flask containing zinc dust (6.0 equiv, 325 mesh, Strem) was heated with a
heat gun while evacuating and filling with nitrogen (3 times). Under nitrogen, degassed DMF
(14 mL) was added via syringe followed by 1,2-dibromoethane (0.35 equiv). The grey
reaction mixture was stirred in an oil bath at 100°K*I for 15 minutes and then cooled to room
temperature. Chlorotrimethylsilane (0.25 equiv) was added to the mixture via syringe and
the reaction was stirred at room temperature for 30 minutes. A solution of 1,1-dimethylethyl

(4i?)-4-({[(l,1-dimethylethyl)ox.y]carbonyl}amino)-5-iodopentanoate (2.0 K*i, 1.2 equiv) in
degassed DMF (14 mL) was added to the reaction mixture via cannula. The flask containing
the solution was rinsed with degassed DMF (4 mL) and cannulated into the reaction mixture.
The reaction was stirred at room temperature for 1 hour. Then,
tris(dibenzylideneacetone)dipalladiurn (O) (2.5 mol%), tri-o-totylphosphine (10 mol%) and 2-
(4-iodophenyl)-S-methylimidazo[1,2-a]pyridine (1.4 K*i, 1.0 equiv) were added through the top
all at once. The reaction mixture was stirred at room temperature for l7V>urs. The reaction
was diluted with EtOAc (40 mL) and filtered through Celite®. The filtrate was washed with
H2O (20 mL) and brine (20 mL), and the organic layer was dried over MgS(>4 and
concentrated in vacuo. Purification by silica gel chromatography (Analogix 1F28O, 5-90%
EtOAc/hexanes) afforded the title compound as a white solid (90%). ESMS [M+H]+ = 480.4.
1,1-dimethylethyl (4#)-4-( {[(1,1-dimethylethyl)oxy]carbonyl} amino)-5-[4-(l -methyl-2-
([methyl(methyloxy)amino]carbonyl}-lH'-imidazo1-4-yl)phenyl]pentanoate:

[00424) Following the procedure described above using 4-(4-bromophenyl)-jV,1-
dimethyl-A'-(methyloxy)-1H-imidazole-2-carboxamide provided the title compound as a solid
(82%). ESMS [M+H]+ = 517.2.
(4J?)-4-[( {3-chloro-4-[(l -methylethyl)oxy]phenyl) carbonyl)amino]-5-[4-(S-
methylimidazo[1 .2-a)pyridin-2-yl)phenyl)pentanoic acid:

[00425) To a solution of 1,1-dimethylethyl (4i?)-4-({[(l ,1-
dimethylethyl)oxy)carbonyl}amino)-5-[4-(8-methylimidazo[1,2-a)pyridin-2-
yl)phenyl]pentanoate (1.35 K*i, 2.82 mmol) in CH2Cl2 (14 mL) was added trifluoroacetic acid
(10 mL) followed by triethylsilane (2.5 equiv, 7.04 mmol). The reaction was stirred for 45

minutes at room temperature and then concentrated in vacuo. DMF (35 mL) was added to the
residue followed by diisopropylamine (14.7 mL, 84.51 mmol) under nitrogen. The reaction
was stirred for 5 minutes and pentafluorophenyl 3-chloro-4-[(1-methylethyl)oxy]benzoate
(1.1 equiv, 3.10 mmol) was added. The reaction was stirred for 45 minutes and then
concentrated in vacuo. Ethyl acetate (50mL) was added to the residue and it was washed with
H2O (30 mL). The aqueous layer was extracted with EtOAc (20 mL) and the combined
organic layers were dried over MgSOa and concentrated in vacuo. Purification by silica gel
chromatography (Analogix 1F28O,25-100% EtOAc/hexanes) provided the title compound as
a white foamy solid (61%). ESMS [M+H]+ = 520.2.
(4i?)-4-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-5-[4-(1-methyl-2-
{[methyl(methyloxy)amino]carbonyl}-1H-imidazo1-4-yl)phenyl]pentanoicacid:

[00426] Following the procedure described above with 1,1-dimethylethyl (4i?)-4-
({[(1,1-dimethylethyl)oxy]carbonyl} amino)-5-[4-(l -methyl-2-
{[methyl(methyloxy)amino]carbonyl)-1H-imidazo1-4-yl)phenyl]pentanoate and foregoing
purification provided the title compound as a solid. ESMS |>4+H]+ = 557.2.
(4/?)-5-[4-(2-acetyl-l -methyl-ltf-imidazo1-4-yl)phenyl]-4-[( {3-chloro-4-[(l -
methylethyl)oxy]phenyl}carbonyl)arnino]pentanoicacid:

(O0427J To a solution of crude (4i?)-4-[({3-chloro-4-[(l -
methylethyl)oxy]phenyl} carbonyl)amino]-5-[4-(l -methyl-2-
{[methyl(methyloxy)amino]carbonyl}-1H-imidazo1-4-yl)phenyl]pentanoic acid (3.18 mmol)
in anhydrous THF (16 mL) under nitrogen at 0°K*I was added methylmagnesium bromide (10.6

mL, 10 equiv, 3.0 M in ether) dropwise by syringe. The reaction was stirred for 30 minutes at
0°K*I and then carefully quenched with sat. aq. NH4Cl solution (10 mL), followed by IN HC1
solution (60 mL) such that the pH of the aqueous layer -5.5. The product was extracted with
EtOAc (4 x 40 mL) and the combined organic layers were dried over MgSO4 and
concentrated in vacuo to give the title compound, which was used directly in the next
reaction. ESMS [M+Hf = 512.4.
AHO K*I)-4-Amino-1-{[4-(S-methylimidazo[1,2-a]p>Tidin-2-yl)phenyl]methyl) -4-oxobutyl)-3-
chloro-4-[(1-methylethyl)oxy]benzamide:

(O0428) To a solution of _(4/?)-4-[((3-chloro-4-[(1-
methylethyl)oxy]phenyl} carbonyl)amino]-5-[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]pentanoic acid (900 mg, 1.73 mmol) in anhydrous THF (12.4 mL) at 0°K*I under
nitrogen was added triethylamine (242 µL, 1.73 mmol) followed by ethyl chloroformate (174
µL, 1.82 mmol). The reaction was stirred for 40 mins at 0°K*I and then the solids were filtered
and washed with 5 mL THF. The filtrate was added to a flask containing NH4OH (5 mL) at
room temperature and the reaction mixture was stirred for 1 hour. The product was extracted
from the reaction mixture with EtOAc (50mL). The aqueous layer was extracted with EtOAc
(20 mL) and then acidified with IN HC1 solution (30 mL) and re-extracted with EtOAc (10
mL). The combined organic layers were dried over MgSO4 and concentrated in vacuo to give
a white solid. Purification by recrystallization from hot isopropanol afforded the title
compound as a white solid (90%). ESMS [M+Hf = 519.4.
Ar-((li?)-1-{[4-(2-acetyl-1-methyl-1H-imidazo1-4-yl)phenyl]methyl}-4-amino-4-oxobutyl)-3-
chloro-4-[(l -methylethyl)oxy]benzamide:


[00429] Following the procedure described in above with (4fJ)-5-[4-(2-acetyl-1-
methyl- l#-imidazo1-4-yl)phenyl]-4-[( {3-chloro-4-[(1- *
methylethyl)oxy]phenyl}carbonyl)amino]pentanoic acid and purification by Gilsin reverse
phase HPLC provided the title compound as a white solid. ESMS [M+H]+ = 511.2.

(35)-4-[4-(2-acetyl-1-methyl-lf/-imidazo1-4-yl)phenyl]-3-[({3-chloro-4-[(1-
methylethyl)oxy]phenyl}carbonyl)amino]butyl dimethyl phosphate:

[00430] To a solution of N-((\S)-\-{[4-(2-acetyl-1-methyl-l#-imidazo1-4-
yl)phenyl]methyl}-3-hydroxypropyl)-3-chloro-4-[( 1-methylethyl)oxy]benzamide (500 mg,
1.04 mmol) in dry CH2Cl2 (10 mL) under N2, was added dimethyl chlorophosphate (748 mg,
5.18 mmol) followed by DMAP (660 mg, 5.41 mmol) at it. After 30 min, TLC (95:5
EtOAc/MeOH) showed ~50% conversion, so an additional portion of dimethyl
chlorophosphate (748 mg, 5.18 mmol) and DMAP (660 mg, 5.41 mmol) were added. After

an additional 30 min, the reaction was quenched with saturated aqueous NH4Cl and diluted
with CH2Cl2. The aqueous layer was back-extracted with CH2Cl2 and the combined organics
were dried (Na^SO,}), filtered and concentrated under reduced pressure. The residue was
purified by silica gel chromatography (100% EtOAc; isocratic on Analogix) to give 475 mg
(77%) of the title compound as a pale yellow oil. LC/MS (ES) m/e 592.4 (M + H)+. Note
that the product was contaminated with ~leq of the starting dimethyl
chlorophosphate/dimethyl hydrogenphosphate reagent and carried on as^.
(31S)-4-[4-(2-acetyl-l -methyl-1H-imidazo1-4-yl)phenyl]-3-[( (3-chloro-4-[(l -
memylethyl)oxy]phenyl}carbonyl)amino]butyl dihydrogen phosphate:

100431] A yellow solution of (35)-4-[4-(2-acetyl-1-methyl-1H-imidazo1-4-yl)phenyl]-
3-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]butyl dimethyl phosphate (475
mg, 0.804 mmol) in 30% HBr in AcOH was placed in a pre-heated (60 ° K*I) bath for 10 min,
then immediately allowed to cool to rt. The reaction mixture was concentrated under reduced
pressure and the residue was dissolved in DMSO (6 mL), filtered and purified by Gilson
reverse phase HPLC (MeCN/H2O with 0.1% TFA). The MeCN of the clean fractions was
removed under reduced pressure and the remaining aqueous solution was frozen and
lyophilyzed overnight to give 84 mg (19%) of the title compound as a yellow powder. LC/MS
(ES) m/e 564.2 (M + H)+.
(3S)-3-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-[4-(8-
methylimidazo[1,2-fl]pyridin-2-yl)phenyl]butyl dihydrogen phosphate:

[00432] Following the procedures described above, except substituting 3-chloro-iV-
((lS)-3-hydroxy-1-([4-(8-methylhnidazo[1,2-a]pyridin-2-yl)phenyl]methyl}propyl)-4-[(1-

methylethyl)oxy]benzamideforA-((15)-1-{[4-(2-acet>'1-1-methyl-1H-imidazo1-4-
yl)phenyl]methyl}-3-hydroxypropyl)-3-chloro-4-[(1-methylethyl)oxy]benzamideand
potassium fe/t-butoxide for DMAP, the title compound was prepared as a white powder (35%
yield). LC/MS (ES) m/e 563 (M + H)+.

3-Cyano-7^-[(15)-1-({4-[S-(3,5-dimethyl-4-isoxazolyl)imidazo[1,2-a]pyridin-2-
yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide:

[00433) To a solution of 7V-((15)-1-{[4-(8-bromoimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}-3-hydroxypropyl)-3-cyano-4-[(l -methylethyl)oxy]benzamide (200 mg,
0.366 mmol) in dry DMF (2 mL) were added 3,5-dimethyl-isoxazole-4-boronic acid (63 mg,
0.439 mmol). tetrakistriphenylphosphine palladium(O) (21 mg, 0.018 mmol) and 2.0M
aqueous K*I.2CO3 (0.46 mL) successively at rt. The reaction mixture was purged with argon
and heated to 100 °K*I, stirred for 22 h, cooled to rt. filtered and purified directly by reverse
phase HPLC (MeCN/H2O with 0.1 % TFA). The clean fractions were adjusted to pH ~8 with
saturated aqueous NaHCCh and extracted with 3% MeOH/EtOAc (3 X 30 mL). The extracts
were dried (Na2SO.»)} filtered and concentrated under reduced pressure to give 45 mg (22%)
of the title compound as an off-white solid. LC/MS (ES) m/e 564.2 (M + H)+.
Scheme K*I:


3-chloro-AH(l^-1-{[3-chloro-4 hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide:

(O0434] Following the procedures described in the literature (J. Org. Chem. 2003, 68,
4215; J, Org. Chem. 2002, .67,1738; J. Am. Chem. Soc. 1972,94, 6203), as well as the
procedures above, the title compound was prepared as a white solid. LC/MS (ES) m/e 526
(M + H)+.
The following compounds were prepared using the procedures described above:
Structure
Name
(M + H)+


N-((l R)-1- {[4-(2- Acetyl-1-methyl-1H-imidazo1-4-yl)phenyl]methyl}-4-amino-4-oxobutyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide

N-[(l R)-4-Amino-1-({4-[2-(l -hydroxy-1-methyl ethyl)-1-methyl-1H-imidazo1-4-
yl]phenyl}methyl)-4-oxobutyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide

N-[(l S)-2-(D-Alanylamino)-1-({4-[ 1-(2-aminoethyl)-2-( 1,1-dimethylethyl)-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(r-methylethyl)oxy]benzamide

N-((l S)-2- {4-[ 1-(2-Aminoethyl)-2-(l, 1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1- {[(2-
methylalanyl)amino]methyl}ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide


N-l(l S)-2-(D-Atanylamino)-l -((4-[1-(2-aminoethyl)-2-(l ,1-dimethylethyl)-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide

N-((l S)-2- (4-[ 1-(2- Aminoethyt)-2-(l, 1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1-
{[(hydroxyacetyl)amino]methyl}ethyl)-3-cyano-4-l(1-methylethyl)oxy]benzatnide

N-((l S)-2- (4-[ 1-(2-Aminoethyl)-2-(l ,1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1- {[(2-
methylalanyl)amino]methyl}ethyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide

N-((l S)-2- (4-[ 1-(2-Aminoethyl)-2-(l ,1-dimethylethyl)-1H-imidazo1-4-yl]phenyl}-1- {[(N,N-
dimethylglycyl)amino]methyl}ethyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide
588.4


3-Cyano-Ar-[(15)-2-{4-[8-(1-hydroxyethyl)imidazo[1,2-a]p>'ridin-2-yl]phenyl}-1-({[(2i?)-2-
hydroxypropanoyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzami3e

N-{(1 S)-2-[(Aminocarbonyl)amino]-l -{[4-(8-bromoimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-3-chloro-4-[(1-methylethyl)oxy]benzamide

N-{(lS)-2-[4-(S-Bromoimidazo[1,2-a]pyridin-2-yl)phenyl]-1-[(2-oxotetrahydro-l(2H)-
pyiimidiny])methyl]ethyl}-3-chloro-4-[(1-methylethyl)oxy]benzamide

N-{(1 S)-2-[4-(8-Bromoimidazo[1,2-a]pyridin-2-yl)phenyl]-1-[(2-oxohexahydro-1H-l ,3-
diazepin-1-yl)methyl]ethyl) -3-chloro-4-[(l -methylethyl)oxy]benzamide
639.2


N-((l S)-2-[(Aminocarbonothioyl)amino]-1-{[4-(8-bromoimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl} ethyl)-3-chloro-4-[(l -methylethyl)oxy]benzamide »

2-(4- {(2S)-2-[( {3-Cyano-4-l(l -methylethyl)oxy]phenyl) carbonyl)amino]-3-[(l ,2,3-
thiadiazo1-4-ylcarbonyl)amino]propyl}phenyl)imidazo[1.2-a]pyridine-8-carboxamide

N-((l S)-2-[(Aminosulfonyl)amino]-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide
547.2
x^^ K*I.M
547.2
x^^ K*I.M
(3S)-3-[({3-Chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-{4-[2-(l,1-
dimethykthyl)-l -methyl-1H-imidazo1-4-yl]phenyl}butanoic acid
512.4


N-[( 1 S)-2-[(Aminosulfonyl)amino]-l -({4-[2-( 1,1-dimethylethyl)-1-methyl-1H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide »

N-((15)-1-{[4-(l//-Benzimidazo1-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-chloro-4-[(1-
methylethyl)oxy]benzamide

3-Chloro-A'-[(15)-3-hydroxy-1-({4-[5-(trifluoromethyl)-lif-benzimidazo1-2-
yl]phenyl} methyl)propyl]-4-[(l -methylethyl)oxy]benzamide

3-Chloro-jV-(( 1 S> 1- {[4-(5,6-dimethyl-1 #-benzimidazo1-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methyl ethyl)oxy]benzamide
506.2


3-Chloro-Ar-[(15)-3-hydroxy-1-({4-[5-(methybxy)-m-benzimidazo1-2-
yl]phenyl}methyl)propyl]-4-.[(1-methylethyl)oxy]benzamide

3-Chloro-N-((lS)-1-{[4-(5-chloro-m-benzimidazo1-2-yl)phenyl]methyl}-3-hydroxypropyl)-
4-[(1-methylethyl)oxy]benzamide

3-Chloro-A/-((l S)-3-hydroxy-1- {[4-(4-methyl-1 /f-benzimidazo1-2-yl)phenyl]methyl}propyl)-
4-[( 1-methylethyl)oxy]benzamide

3-Chloro-AT-((15)-1-{l4-(6-chloro-1H-imidazo[4,5-b]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
513.2


Ethyl 2-(4-{(-5)-2-[({3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl} phenyl)-1H-benzimidazole-5-carboxylate ^

2-(4- \(2S)-2-[( {3-Chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl}phenyl)-1 #-benzimidazole-5-carboxylic acid

A^-((lS)0-Amino-1-{[4Kl^-benzimidazo1-2-yl)phenyl]methyl}propyl)-3-chloro-4-[(1-
methylethyl)oxy]benzamide

3-Cymo-N-((\S)-1- {[4-(S-cyanoimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(l -methylethyl)oxy]benzamide

Ar-((l^-1-{[4-(8-Chloroimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-hydrox>propyl)-3-

cyano-4-[(1-methylethyl)oxy]benzamide

3-Cyano-7V-[(lS)-3-hydroxy-l ■»
yl]phenyl}methyl)propyl]-4-[0 -methylethyl)oxy]benzamide

3-Cyano-W-((15)-3-hydroxy-1-{[4-(8-hydroxyimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide

2-(4- {(25)-2-[( {3-Cyano-4-[(l -methylethyl)oxy]phenyl} carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[1,2-i}]pyridine-7-carboxamide

Ethyl 2-(4-{(25)-2-[({3-cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl} phenyl)imidazo[1,2-a]pyridine-7-carboxylate

3-Cyano-N-((lS)-3-hydroxy-1-{[4-(8-nitroimidazo[1,2-a]pyTidin-2-yl)phenyl]methyl}propyl)-
4-[(1-methylethyl)oxy]benzamide


N-(( 1S)-1- {[4-(8-Aminoimidazo[ 1,2-«]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide

2-(4-{(25)-2-[({3-Cyano-4-[(1-raethylethyl)oxy]phenyl}carbonyl)amino]-4-
hydroxybutyl} phenyl)imidazo[ 1,2-fl]pyridine-8-carboxamide

3-Cyano-Ar-[(15)-3-hydroxy-1-({4-[8-(hydiTOymethyl)imidazo[1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

A'-[(15)-1-({4-[8-0'\minomethyl)imidazo[1.2-a]pyridin-2-yl]phenyl}methyl)-3-
hydroxypropyl]-3-cyano-4-[(l *methylethyl)oxy]benzamide
t
N-(( 15)-1-{[4-(8-Acetylimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-hydroxypropyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide
511.2


3-Cyano-.AH0 S)-3-hydroxy-1-({4-[8-(l -hydroxy-1-methylethyl)imidazo[1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

3-Cyano-Ar-t(l £)-3-hydroxy-1-((4-[S-(l -hydroxyethyl)imidazo[ 1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

3-Cyano-JV-((15)-3-hydroxy-1-{[4-(8-methyl-5,6,7,8-tetrahydvoimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide

3-Cyano-7V-[(lS)-1-({4-[2-(U1-dimethylethyl)-1-(2-hydroxyethyl)-17f-imidazo1-4-
yl]phenyl} methyl)-3-hydroxypropyl]-4-[(l -methylethyl)oxy]benzamide

N~[(\ S)-l -({4-[1-[2-(Acetylamino)ethyl1-2-(l > 1-dimethylethyl)-1H-imidazo1-4-

yl]phenyl} methyl)-3-hydroxypropyl]-3-cyano-4-[( 1-methylethyl)oxy]benzamide

S-Cyano-^-tCl^-S-hydroxy-1-^^S-^l^-1-hydroxyethyllimidazotl^-alpyridin^-
yl}phenyl)methyl]propyl}-4-[(1-methylethyl)oxy]benzamide

3-Cyano-Ar-{(15)-3-hydroxy-1-[(4-{8-[(lS)-1-hydroxyethyl]imidazo[1,2-a]p>Tidin-2-
yl} phenyl )methyl]propyl}-4-[(l -methylethyl)oxy]benzamide

3-Chloro-^-[(15)-3-hydroxy-1-({4-[8-(1-hydroxypropyl)imidazo[1,2-a]pyridin-2-
yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

A/-((15)-1-{[4-(8-Bromoimidazo[1,2-a]pyTidin-2-yl)phenyl]methyl}-3-hydrox>propyl)-3-
chloro-4-[( 1-methylethyl)oxy]benzamide

3-Chloro-//-((15)-1-{[4-(8-chloroimidazo[1,2-a]p>Tidin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide
512.4


3-Chloro-Af4(lS)-3-hydroxy4 yl]phenyl}methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

N-[(\ R)-4- Amino-1-((4-[8-(l -hydvoxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl} methyl)-4-
oxobutyl]-3-chlovo-4-[(1-methylethyl)oxy]benzamide

JV-[(1 R)-A- Amino-1-( {4-[S-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl} methyl)-4-
oxobutyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide

3-Ch1oro-.Nr-((lS,)-1-{[4-(3-fluoro-8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide

3-Cyano-N-((l S)-\ - {[4-(3-fluoro-8-methylimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
501.2


3-Chloro-Ar-((15)-2-hydroxy-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-
yl)phenyl]methyl}ethyl)-4-[(l -methylethyl)oxy]ber»2aniide »

3-Ch]oro-4-[(1-methylethyl)oxy]-7^-[(lS)-2-[4K8-methylimidazo[1.2-a]pyridin-2-yl)phenyl]-
1-(4-morpholinylmethyl)ethyl]benzamide

3-Chloro-7^-(( 1,S)-2-(4-hydroxy-1-piperidinyl)-1- {[4-(8-methylimidazo[ 1,2- yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide

3-Chloro-A^(l S)-2-(3-hydroxy-l -pyrrolidinyl)-1-{[4-(S-methylimidazo[ 1,2-a]pyridin-2-
yl)phenyl]methyl} ethyl)-4-[(l -methylethyl)oxy]benzamide


3-Chloro-N-((l S)-2-[(25)-2-(hydroxymethyl)-l -p>Trolidinyl]-l -{[4-(8-methylimidazo[1,2-
a]pyridin-2-yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide

3-Chloro-N-(O S)-2-[(2R)-2-(hydroxymethyl)1-pyrrolidinyl]-1-{[4-(8-methylimidazo[1,2-
a]pyridin-2-yl)phenyl]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamide

3-Chloro-4-[(l -methylethyl)oxy]-JV-(O S)-2-[4-(S-methylimidazo[ 1,2-a]pyridin-2-yl)phenyl]-
1- {[(2,2,2 -trifhioroethyl)amino]methyl) ethyl)benzamide

3-Chloro-JV-((lS)-24(2-hydroxyethyl)amino]4-{[4-(8-methylimidazo[1)2-a]pyridin-2-
yl)phenyl]methyl) ethyl)-4-[(l -methylethyl)oxy]benzamide


3-Cyano-iV-(( 15)-1- {[4-(8-ethyl-5-methylimidazo[ 1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide

Methyl (3S)-3-[( {3-chloro-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-4-{4-
[(phenylcarbonyl)amino]phenyl}butanoate

3-Chloio-A'-[(15)-3-hydroxy-1-({4-[(phenylcarbonyl)amino]phenyl}methyl)propyl]-4-[(1-
methylethyl)oxy]benzamide

3-Chloro-Af- {(1 S)-l -[(4- {[(4-chlorophenyl)carbonyl]amino}phenyl)methyl]-3-
hydroxypropyl}-4-[( 1-methylethyl)oxy]benzamide
515


Phenylmethyl (4- {(2S)-2-[({3-chloro-4-[( 1-methylethyl)oxy]phenyl} carbonyl)amino>4-
hydroxybutyl}phenyl)carbamate ^

3-Chloro-N-((l 5)-3-hydroxy-1- {[4-( {[2-
(methylamino)phenyl]carbonyl}amino)phenyl]methyl}propyl)-4-[(1-
methylethyl)oxy|benzamide

N-{4- {(2S)-2-[( (3-Chloro-4-[(l -methylethyl)oxy]phenyl} carbonyl)amino]-4-
hydroxybutyl}phenyl)-4-pyridinecarboxamide

3-Chlovo-Ar.[(l1S)-1-({4-[(cyclohexylcarbonyl)amino]phenyl}methyl)-3-hydroxy'propyl3-4-
[(1-methyl ethyl)oxy]benzamide
487


3-Chloro-7V-[(l,S)-1-({4-[(3,3-dimethylbutanoyl)amino]phenyl}methyl)-3-hydroxypropyl]-4-
[(1-methyl ethyl)oxy]benzamide

3-Chloro-Ar-[(15}-3-hydvoxy-1-({4-[(phenylacetyl)amino]phenyl}methyl)propyl]-4-[(1-
methylethyl)oxy]benzamide

3-Chloro-A- {(1 £)-3-hydroxy-1- [(4- {[(phenyl amino)carbony! ] amino} phenyl )methyl ]propyl}-
4-[(1-methylethyl)oxy]benzamide

3-Cyano-//-((15}-3-hydroxy-1-{[4-(8-methyl-5-oxo-5,6-dihydroimidazo[1,2-K*i]p>Timidin-2-
yl)phenyl]methyl} propyl)-4-[( 1-methylethyl)oxy]benzamide
K*I


3-Cyano-iV-((l 5)-3-hydroxy-1- {[4-(l -methyl-3-oxo-2>dihydro-l#-imi*lazo[1,2-a]imidazo1-
6-yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]ben2amide

3-Cyano-7^-((15)-3-hydroxy-1-{[4-(8-oxo-7,8-dihydroimidazo[1,2-a]p>Tazin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide

2,3-Dichloro-yV-((15)-3-hydroxy-1-{[4-(8-methylimidazo[1,2-fl3pyridin-2-
yl)phenyl]methyl} propyl)-4-[( 1-methylethyl)oxy]benzamide

^((lS)-3-Hydroxy-1-{[4-(8-methylimidazo[1;2-a]pyridin-2-yl)phenyl]methyl}propyl)-4-[(1-
methylethyl)oxy] -3-nitrobenzamide


3-Chloro-Ar-[( 1 ^-[(hydroxyacetyOamino]-1-({4-[8-( 1-hydioxyethyl)iiftidazo[ 1,2-a]pyridin-
2-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

3-Chloro-A-[(15r>-2-{4-[8-(1-hydroxyethyl)imidazo[152-a]pyridin-2-yl]phenyl}-1-({[(2/?)-2-
hydroxypropanoyl]amino}methyl)ethyl]-4-[(l -methylethyl)oxy]benzamide

3-Chloro-A-[( 1 S)-2- {4-[8-(l -hydroxyethyl)imidazo[ 1,2-a]pyridin-2-yl]phenyl}-1-({[(25)-2-
hydroxypropanoyl]amino}methy])ethyl]-4-[(1-methylethyl)oxy]benzamide

S-Chloro-Ar-ft 15)-2-[(AyV-dimethylg]ycyl)amino]-l -({4-[8-(l -hydroxyethyl)imidazo[1,2-
a]pyridin-2-yl]pheny]) methyl)ethyl]-4-[( 1-methylethy])oxy]benzamide
592


N-[(\ S)-2-(D-Alanylamino)-l -((4-[8-(l -bydroxyethyl)imidazo[1,2-a]p>Tidin-2-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide

S-Chloro-iV-^l^-S-hydroxy-1-Cl^tS-C1-hydroxyethylMmidazotl^-alpyridin^-
yVJphenyl} methyl)propyl]-4-[(l -methyl ethyl)oxy]benzamide

3-Chlovo-AH(l S)-2- {4-[8-(1-hydroxyethyl)imidazo[1,2-a]pyridin-2-yl]phenyl}-1-{[(2-
methylalanyl)amino]iTiethyl}ethyl)-4-[(l -methylethyl)oxy]benzamide

(3S)-3-[( (3-Chloro-4-[(l -methylethyl)oxy]phenyl} carbonyl)amino>4- {4-
[(phenylcarbonyl)amino]phenyl}butanoicacid
495


3-Chloro-Ar-{(15)-3-hydroxy4-[(4-imidazo[1,2-a]p>Tidin-6-ylphenyl)m»thyl]propyl}-4-[(1-
methylethyl)oxy]benzamide

3-Chloro-Ar-[(l^-lK{4-[2Hl,1-dimethylethyl)imidazo[1,2-a]p>'ridin-6-yl]pheny]}methyl)-3-
hydroxypropyl]-4-[(l -methylethyl)oxy]benzarnide

3-Chloro-Ar-{(15)-3-hydroxy-1-[(4-imidazo[1,2-a]pyridin-2-ylphenyl)methyl]propyl}-4-[(1-
methylethyl)oxy]benzamide

3-Chloro-A7- {(1 S)-3-hydroxy-1-[(4-imidazo[ 1,2-a]pyrimidin-2-ylphenyl)methyl]propyl}-4-

[(1-methyl ethyl)oxy]benzamide

3-Chloro-Ar-((15)-3-hydroxy-1-{[4-(5-methylimidazo[1,2-a]p}Tidin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide

3-Chloro-Af-((lS)-3-hydroxy-1-{[4-(7-methylimidazo[1,2-a]p>iimidin-2-
yl)phenyl]methyl}propyl)-4-[(1-methylethyl)oxy]benzamide

3-Cyano-A^{(15)-3-hydroxy-1-[(4-imidazo[2,1-6][1,3]thiazo1-6-ylphenyl)iTiethyl]butyl}-4-
[(1-methylethyl)oxy]benzamide


3-Cyano-/^-((15)-3-hydroxy-1-{[4-(3-methylimidazo[2,1-6][1,3]thiazo1-6-
yl)phenyl]methyl} butyl)-4-[( 1-methylethyl)oxy]benzamide

3-Cyano-^-((15)-1-{[4-(2,3-dihydroimidazo[2,1-Z)][1,3]thiazo1-6-yl)phenyl]methyl}-3-
hydroxybutyl)-4-[( 1-methylethyl)oxy]benzamide

3-Cyano-JV-(( 1S)-1- {[4-(l, 1-dioxido-2,3-dihydroimidazo[2,1-b}[ 1,3]thiazo1-6-
yl)phenyl]methyl}-3-hydroxybutyl)-4-[(1-methylethyl)oxy]benzamide

Ar-[(15)-lK{4-[1-(3-Aminopropyl)-2-(l,1-dimethylethyl)-l//'-imidazo1-4-yl]pheny]}niethyl)-
3-hydroxypropyl]-3-cyano-4-[(1-methylethyl)oxy]benzamide

3-Cyano-4-[(1-methylethyl)oxy]-yV-[(l1S}-2-[4-(8-methylimidazo[152-a]p>Tidin-2-yl)phenyl]-

1-(5-methyl-1,2,4-oxadiazo1-3-yl)ethyl]benzamide

3-Cyano-jV-[(15)-1-({4-[8-(3,5-dimethyl-4-isoxazolyl)imidazo[1,2-a]p>Tidin-2-
yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide

3-Cyano-A/-((15)-3-hydroxy-1-{[4-(8-phenylimidazo[1,2-a]p\ridin-2-
yl)phenyl]methyl}propyl )-4-[(l -methylethyl)oxy]benzamide

3-Cyano-A/,-[(15)-3-hydroxy-1-({4-[S-(l/f-pyrazo1-4-yl)imidazo[1,2-a]pyridin-2-
yl]phenyl)methyl)propyl]-4-[(1-methylethyl)oxy]benzamide

3-Cyano-A^[(15)-3-hydroxy-1-({4-[8-(4-isoxazolyl)imidazo[1,2-fl]pyridin-2-.
yl]phenyl}methyl)prop>l]-4-[(1-methylethyl)oxy]benzamide
536.2


AH(15)-1-{[4 chloro-4-[(l -methylethyl »oxy]benzamide

Ethyl (2£)-3-[2-(4-{(25)-2-[({3-cyano-4-[(1-methylethyl)ox}']phenyl}carbonyl)amino]-4-
hydioxybutyl}phenyl)imidazo[1,2-fl]pyridin-S-yl]-2-pi'openoate

(2F)-3-[2-(4-{(25)-2-[({3-Cyano-4-[(1-methylethy])oxy]phen}l}carbonyl)amino]-4-
hydroxybutyl}phenyl)imidazo[ 1,2-fl]pyridin-S-yl]-2-propenoic acid

7V-{(15)-1-[(4-{8-[(l£')-3-^jnino-3-oxo-1-propen-1-yl]imidazo[i,2-a]pyridin-2-
yl}phenyl)methylJ-3-hydrox>propyl}-3-cyano-4-[(1-methyleth)'])oxy]benzamide

A-[(15)-1-({4-[8-(3-Amino-3-oxopropyl)imidazo[1,2-a]pyridin-2-yl]phenyl}methyl)-3-
hydroxypropyl]-3-cyano-4-[(]-methylethyl)oxy]benzamide


3-Chloro-yV-((15)-1-{[4-(3-chloio-8-methylimidazo[1,2-fl]p>Tidin-2-yl)phenyl]methyl}-3-
hydrox)'propyl)-4-[(1-methylethyl)oxy]benzamide

Ar-((15)-1-{[4-(3-Chloro-8-methyhmidazo[1,2-fl]'p>Tidin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-3-cyano-4-[(1-methylethyl)oxy]benzamide

3-Cyano-yV-[(16)-1-({3-fluoro-4-[2-(l%droxy-1-methylethyl)-1-methyl-l/f-imidazo1-4-
yl]phenyl}methyl)-3-hydroxypropyl]-4-[(1-methylethyl)oxy]benzamide

3-Chloro-A^-((l5)-2-hydroxy-1-{[5-(8-methylimidazo[1,2-fl]pyiridin-2-yl)-2-
pyridinyl]methyl) ethyl)-4-[( 1-methylethyl)oxy]benzamide
479


3-Chloro-Ar-((l^)-2-hydroxy-1-{[5-(8-methy]imidazo[1,2-fl]p>Tidin-2-yl)2-
thienyl]methyl}ethyl)-4-[( ] -methylethyl)oxy]benzamide

3-Ch]oro-i\-[(15)-1-({4-[2-(l,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]-2-
fluoroplKnyl)methyl)-3-hydrox>'propyl]-4-[(1-methylethyl)oxy]benzamide

3-Chloro-A-[( 15)-1-({4-[2-( 1.1-dimethylethyl)-1-metliyl-l//-imidazo1-4-yl]-2,6-
difluorophen\])methyl)-3-hydrox>propyl]-4-[(1-methylethyl)oxy]benzamide

3-Chloro-.V-[(15)-1-({2-chloro-4-[2-(l,1-dimethylethyl)-1-methyl-l//-imidazo1-4-
yl]pheny)}methyl)-3-hydrox)propyl]-4-[(1-methylethyl)oxy]benzamide
532


3-Chloro-Ar-[(15)-1-({5-[2-(l,1-dimethylethyl)-1-methyl-lif-imidazo1-4-yl]-2-
pyridinyl)methyl)-3-hydrox\propyl]-4-[(1-methylethyl)oxy]benzamide

3-Ch]oro-A;-((lS)4-{[2-chloro-4-(8-methylimidazo[1,2-a]pyiidin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[( 1-methylethyl)oxy]benzamide

3-Chloro-A^-((15)-1-{[2-chloro-4-(8-chloroimidazo[1,2-fl]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methyl ethyl)oxy]benzamide

3-Chloro-JV'-((l^-1-{[2,5-difluoro-4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}-3-
hydroxypropyl)-4-[(1-methylethyl)oxy]benzamide
528


3-Chloro-N-[( 1S)-1-({4-[2-(l ,1-dimethylethyl)-1-methyl- 1H-imidazo1-4-yl]phenyl Jmethyl}
3-(methylamino)-3-oxopiopyl]-4-[(1-methylethyl)oxy]benzamide »

3-Cyano-N-[(lS)-2-{4-[2-(l.1-dimethylethyl)-1-methyl-1H-imidazo]-4-yl]phenyl}-1-
({[(phenylamino)carbonyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

3-Cyano-N-[(lS)-2-{4-[2-(l,1-dimethylethyl)-I-methyl-1H-imidazo1-4-yl]phenyl}-1-
({[(ethylamino)carbonyl]amino}methyl)ethyl]-4-[(1-methylethyl)oxy]benzamide

N-[( 1 S)-2-(Aminosulfonyl)-1-((4-[2-(l, 1-dimethylethyl)-1-methyl- ] H-imidazo1-4-
yl]phenyl}methyl)ethyl]-3-chloro-4-[(1-methylethyl)oxy]benzamide


3-Cyano-N-(( 1 S)-2- {4-[2-( 1,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-
{[(methylsulfonyl)amino]methyl}ethyl)-4-[(1-methylethyl)oxy]benzamMe

3-Cyano-N- {(1 S)-2- {4-[2-( 1,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-[({[(2-
hydroxyethyl)amino]carbonyl}amino)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide

N-[(S)-1-[4-(2-tert-Butyl-1-methyl-1H-imidazo1-4-yl)-benzyl]-2-(2-methoxy-ethanoylamino)-
ethyl]-3-cyano-4-isopropoxy-benzamide

(4R)-4-[({3-Cyano-4-[(1-methylethyl)oxy]phenyl}carbonyl)amino]-5-{4-[2-(l,1-
dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}pentanoic acid
517.4


3-Cyano-N- {(1 S)-2- {4-[2-( 1,1-dimethylethyl)-1-methyl-1H-imidazo1-4-yl]phenyl}-1-[(2-oxo-
1-imidazolidinyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benz amide

N-((lS)-2-Amino-1-{[4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl]methyl}ethyl)-3-cyano-
4-[(1-methylethy])oxy]benzamide

N-((lS)-2-(Acetylamino)-1-{[4-(S-methylimidazo[1,2-a]pyi-idin-2-yl)phenyl]methyl}ethyl)-3-
cyano-4-[(1-methylethyl)oxy]benzamide

3-Ch]oro-N-((lS)-2-{[(2R)-2-hydroxypropanoyl]amino}-1-{[4-(8-methylimidazo[1,2-
a]p}Tidin-2-y])phenyl]meth>]}ethyl)-4-[(1-methy]ethyl)oxy]benzamide
549.2


3-Chloro-N4(lS)-24(N,K1-dimethylglycyl)amino]-lK{4-[2-(1-hydroxy-1-methylethyl)-1-
methyl-1H-imidazo1-4-yl]phenyl}methyl)ethyl]-4-[(1-methylethyl)oxy]Bfenzarnide

3-Cyano-N-[(lS)-2-[(N,N-dimethylg1ycyl)amino]'-1-({4-[2-(1-hydroxy-1-methylethyl)1-
methyl-1H-imidazo1-4-yl]phenyl}rnethyl)ethyl]-4-[(1-methylethyl)oxy]benzamide
561.4

[00435) To a 0° K*I solution of compound 1 (10.7 K*i, 61.37 mmol), (i?)-U,1-
trifluoropropanol (3.5 K*i, 30.68 mmol) in dimethylformamide (200 mL) was added sodium
hydride( 3.7g, 92.05 mniol) portion wise over 5 minutes. After 10 min, the ice bath was
removed and the reaction mixture was stirred while wanning to room temperature. The
reaction mixture was heated to SO° K*I and stirred overnight. The reaction was monitored by
LC/MS. After the reaction was done it was cooled to room temperature. The reaction
mixture was quenched with HCl (0.5N, 200 mL) and extracted with ethyl acetate (3 x 250
mL). The organic layer was dried over sodium sulfate, filtered, and the filtrate was

concentrated in vacuo giving crude compound 2 (8.2 K*i) which was used directly in the next
step without further purification.
[00436] To a 0 °K*I crude solution of compound 2 ( 4.1 K*i , 15.34 mmol).
triefhylamine(6.4mL, 46.02mmol) in dicholoromethane (200ml) was added pentafluorophenyl
trifluoroacetate(6.35mL , 36.82 mmol) via syringe over 3 mins . After 5 mins the ice bath
was removed and the reaction mixture was stirred while wanning to room temperature for
another 2 hours. The reaction mixture was concentrated in vacuo. The resulting residue was
purified by flash chromatography silica gel, hexanes/ethyl acetate = 1:0 , 50:1) to give
compound 3 (3.5 K*i, 50% yield).

[00437] Methyl 4-hydroxy-3-iodobenzoate 2: Methyl 4-hydroxybenzoate (35.5g, 0.233
mol) was dissolved in 200 inL of acetic acid, and the stirred mixture was warmed to 65°K*I. A
solution of IC1 (37.8 K*i, 0.233 mol) in 50 mL of AcOH was added dropwise over 40 min. The
mixture was stirred at 65 K*I for 5 h and then stirred an additional 16 h at room temperature.
The precipitated product was isolated via filtration, washed with water and dried under
vacuum to give 27.5 K*i (99% pure by LCMS and HNMR)) of desired product. The mother
liquors were evaporated and resulting residue was washed with water and dried under vacuum
to give another 31 K*i (95% pure by LCMS and NMR) of desired product. The combined yield
of methyl 4-hydroxy-3-iodobenzoate was 58.5 K*i (90.3% yield). LCMS m/z = .
[00438] Methyl 3-cyano-4-hydroxybenzoate 3: 28 K*i (0.1 mol) of methyl 4-hydroxy-3-

iodobenzoate 2 dissolved in 100 mL of DMF was treated with 9.92 K*i (0.11 mol) of CuCN and
0.49 K*i (0.11 mol) of NaCN. The system was flushed with nitrogen after which the mixture
warmed to 105°K*I and stirred to 1S h. The mixture was allowed to cool to room temperature,
and any precipitates were removed via filtration and washed with EtOAc. The combined
organics were diluted with 200 mL of water and then extracted with EtOAc (2x200 mL). The
combined layers were dried over sodium sulfate, filtered and evaporated to dryness. After
drying under vacuum, the resulting 18 K*i (100% yield) of 3 was characterized by LCMS and
HNMR.
[00439) Methyl 3-cyano-4-isopropoxybenzoate 4: Methyl 3-cyano-4-hydroxybenzoate
3 (18 K*i, 0.1 mol) was dissolved in 100 mL of DMF and treated with 14.2 mL (0.15 mol) of 2-
bromopropane and 41.9 K*i (0.3 mol) of anhydrous potassium carbonate. The system was
flushed with nitrogen, and the mixture was heated to 90°K*I and stirred overnight. After
cooling to room temperature, the mixture was diluted with 200 mL of water and extracted
with CH2Cl2 (2x200 mL). The combined organic layers were dried over sodium sulfate,
filtered and evaporated to diyness to give 20.5 K*i (99% yield) of 4 as an oil that was
characterized by LCMS and HNMR.
[00440] Perfluorophenyl 3-cyano-4-isopropoxybenzoate 6: 20.5 K*i (0.093 mol) of
methyl 3-cyano-4-isopropoxybenzoate 4 was dissolved in 200 mL of a 6:4 mixture of
methanol and water. To this was added 5.61 K*i (0.14 mol) of NaOH, and the mixture was
stirred for 2 hours at room temperature. The solution was then filtered through a silica gel
plug and the solvents removed under vacuum. The resulting solid was re-dissolved in 200
mL of CH2CI? and treated with 19.3 mL (0.11 mol) of perfluorophenyl 2,2,2-trifluoroacetate 5
and 19.5 mL (0.14 mol) of triefhylamine. After stirring overnight, the solution was filtered
and any solids rinsed with CH2Cl2. The combined organic mixtures were run through a short
silica gel column and then evaporated to dryness to give 29 K*i (83.5% yield) of 6 which was
characterized by LCMS and HNMR.
Example 69


[00441} To a solution of compound 1 (200 mg, 1.077 mmol) and^1-iodopropane,
(322µL, 3.23 mmol) in DMF (10 mL) was added DIEA (750µL, 4.31 mmol). The reaction
mixture was heated to 80°K*I and stirred overnight. The reaction was monitored by LC/MS.
After the reaction was done it was cooled to room temperature. The reaction mixture was
quenched with HC1 (0.5N, 30 mL) and extracted with ethyl acetate (50 mL x 3). The organic
layer was dried over sodium sulfate and concentrated and dried under high vacuum. The
resulting residue was purified by reverse phase chromatography (using a mixture of
acetonitrile and water) to give compound 2 (50 mg, 20%).

[00442J To a solution of compound 2 (50mg, 0.22 mmol), in MEOH (1.0 mL) was
added NaOH in water (1,0M, 330µL, 0.330minol). The reaction mixture was stirred at
ambient temperature for 2 hours. The reaction was monitored by LC/MS. The reaction
mixture was quenched with HC1 (0.5N, 5 mL) and extracted with ethyl acetate (10 mL x 3).
The organic layer was dried over sodium sulfate, and concentrated to give 2 (45 mg). LRMS
(M-H+)w/z 212.0
Example 70


[00443] To a solution of compound 1 (200 mg, 1.077 mmol) and 2-iodopropane, (322
µL, 3.23 mmol) in DMF (10 mL) was added DIEA (750 µL, 4.31 mmol)! The reaction
mixture was heated to 80° K*I and stirred overnight. The reaction was monitored by LC/MS.
After the reaction was done it was cooled to room temperature. The reaction mixture was
quenched with HC1 (0.5 K*I 30 mL) and extracted with ethyl acetate (50 mL x 3). The organic
layer was dried over sodium sulfate and concentrated and dried under high vacuum. The
resulting residue was purified by reverse phase chromatography using a mixture of
acetonitrile and water to give compound 2 (50 mg, 20%).

To a solution of compound 2 (50 mg, 0.22 mmol), in MEOH (1.0 mL) was added NaOH in
water (1.0 M, 330 µL, 0.330 mmol). The reaction mixture was stirred at ambient temperature
for 2 hours. The reaction was monitored by LC/MS. The reaction mixture was quenched with
HC1 (0.5 N, 5 mL) and extracted with ethyl acetate (10 mL x 3). The organic layer was dried
over sodium sulfate, and concentrated to give 2 (45 mg). LRMS (M-H+) mi'z 212.0

[00444] 4-bromo-2-chlorophenol (5.04 K*i, 24.3 mmol) was dissolved in DMF (30 mL)
and K2CO3 (10.10 K*i, 72.9 mmol) was added, followed by 2-chloroethyl-/?-toluenesulfonate


(4.86 mL, 26.7 mmol). The resulting mixture was heated to 60°K*I for 3 hours and then cooled
to room temperature. The reaction was diluted with EtOAc (350 mL) and washed with water
(5 x 150 mL). The organic phase was dried (Na2SO4.) and concentrated to a viscous oil which
solidified to a white solid while under high ■vacuum. Compound 1 (6.46 K*i, 24.1 mmol,
quantitative yield) was characterized using *H NMR and used in the following step without
further purification.

(O0445] Compound 1 (6.46 K*i, 24.1 mmol) was dissolved in DMF (30 mL) and
sodium hydride (1.94 K*i of 60% dispersion in mineral oil, 48.6 mmol) was added. The
resulting mixture was stirred at room temperature for 16 hours. The reaction was diluted with
water (100 mL) and EtOAc (350 mL). The layers were separated, and the organic layer was
washed with water (4 x 150 mL). The organic phase was dried (Na2SO.j) and concentrated to
a white solid. Compound 2 (5.56 K*i, 24.0 mmol. quantitative yield) was dried under high
vacuum and characterized using 'H NMR. It was used in the following step without further
purification.

[00446] Compound 2 (5.56 K*i, 24.0 mmol) was combined with
chloroiodomethane (5.59 mL, 76.8 mmol) and dissolved in 1,2-dichloroethane (35 mL) under
an atmosphere of nitrogen. The solution was cooled to 0°K*I with an ice bath and diethyl zinc
(38.4 mL, 1.0 M in hexanes, 38.4 mmol) was added over 10 minutes. The resulting mixture
was stirred for 30 minutes and allowed to warm to room temperature. It was cooled to 0°K*I
with an ice bath, and saturated aqueous NH4C1 (150 mL) was added, followed by
concentrated aqueous NFLOH (25 mL) and EtOAc (200 mL). The layers were separated and
the aqueous phase was extrated with additional EtOAc (2 x 100 mL). The organic phases
were combined, dried (NaiSO.}) and concentrated to a crude oil which was purified using
silica gel (100 % hexanes). Compound 3 (1.76 K*i, 7.2 mmol, 30 % yield) was a colorless oil
which was characterized using 'H NMR.

f

[00447] In a high-pressure reactor, compound 3 (1.76 K*i. 7.2 mmol) was dissolved in
EtOH (40 mL). Triethylamine (5.0 mL 35.8 mmol) was added, followed by [1.1-
bis(diphenylphosphino)ferrocene]dichloropalladium (II) (188 mg, 0.36 mmol). The reaction
vessel was pressurized with carbon monoxide (100 psi), evacuated and repressurized with
carbon monoxide (100 psi). The vessel was evacuated and then pressurized again with
carbon monoxide (350 psi). The reaction was heated to 90°K*I and stirred for 16 h. It was
cooled to room temperature, depressurized and filtered through celite. The solvents were
evaporated, and the remaining residue was partitioned between dichloromethane (150 mL)
and 1 M aqueous KHSO4 (75 mL). The layers were separated and the organic phase was
washed with additional 1 M aqueous KHSO4 (1 x 75 mL). The organic phase was dried
(NaiSO.}) and concentrated to an oil which was purified using silica gel (EtOAc/Hexanes).
providing compound 4 (648 mg, 2.70 mmol, 38% yield) as a white solid, characterized using
'HNMR.

[00448] To a solution of compound 4 (648 mg. 2.70 mmol) in dichloromethane (3 mL)
and EtOH (15 mL) was added 1 M aqueous K*I (7 mL, 7 mmol). The resulting cloudy
mixture was heated to 60°K*I for 1 h. The dichloromethane and EtOH were evaporated under
reduced pressure, and the remaining aqueous solution was acidified using concentrated HC1.
The resulting precipitate was filtered. The filtered, white solid was pure compound 5 (506
mg, 2.39 mmol, 88% yield), characterized using LC/MS (LRMS (M-H) 211.1 m/z).
Example 72


[00449] To a solution of the amine (580 mg, 1.7 mmol) and triethylamine (449
ul 3.4 mmol, 2 eq.) in THF (8.5 ml, 0.2 M), was added chloroethyl chloroformate (278 pi,
2.6 mmol, 1.5 eq). The mixture was stirred for 30 min. at room temperature. Then, it was
diluted in ethyl acetate, washed with 1 N HC1 and brine. The organic layer was dried,
filtered, and concentrated in vacuo to yield a crude material as a yellow oil (900 mg). To a
solution of the crude material in DMF (10 ml), NaH (272 mg, 6.S mmol, 4 eq) was added and
stirred at room temperature for 16 hrs. The mixture was diluted in ethyl acetate (100 ml) and
washed with brine (5 x 50 ml). The organic layer was dried, filtered, and concentrated in
vacuo to yield a crude material as an oil. Purification by columr chromatography (1:1 Ethyl
acetate.Hexanes) gave SO0 mg (24 %) of the desired product, m/z (+1) = 398.0.

[00450J (R)-4-chloro-N-(1-(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-
y])ethyl)butanamide. To a 100 mL round bottom flask was added (R)-benzyl 1-(4-(4-
iodophenyl)-!-methyl-1H-imidazo1-2-yl)ethylcarbamate (1.50 K*i. 3.27 mmol, 1.0 equiv),
CH3CN (20 mL), and TMSI (900 µL, 6.3 mmol, 1.9 equiv). The reaction mixture was capped
and stirred for 2 hours. Methanol (40 mL) was then added to the flask and the mixture was
concentrated, dissolved in EtOAc (100 mL), and washed with wa:er. The organic layer was
dried over NaaSCXt, filtered, and concentrated. The residue was dissolved in DCM and
purified by silica gel chromatography (35-60% CH3CN/CH2C12; then 20% MeOH/ CH2C12) to
afford 950 mg (90%)of the desired primary amine as an oil (M+H (m/z) = 328).
To this amine was added QTCl2 (20 mL) and pyridine (260 uL; 1.1 equiv), followed by 4-
chlorobutyryl chloride (344 µL, 1.05 equiv) in a dropwise fashion The reaction was stirred
for 15 min, followed by the addition of EtOAc (50 mL) and water (10 mL). The organic layer
was separated, dried over Na^SCU, filtered, and concentrated. The residue was dissolved in

DCM and purified by silica gel chromatography (5-35% CH3CN/CH2Cl2) to afford 747 mg
(60%)of (R)-4-chloro-N-(l -(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-yl)ethyl)butanamide
as an off-white solid (M+H (mJz) = 432).

[00451] (R)-1-(1-(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-yl)ethyl)pyrrolidin-2-one.
To a 20-dram vial was added (R)-4-chloro-N-(1-(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-
yl)ethyl)butanamide and THF (10 mL). The vial was then cooled to 0 °K*I under a nitrogen
atmosphere and potassium r-butoxide (214 mg, 1.91 mmol) was added. The reaction was
stirred for 1.5 h. To the reaction mixture was added EtOAc (50 mL) and water (10 mL). The
organic layer was separated, dried over Na2SO4, filtered, and concentrated, The residue was
then dissolved in DCM and purified by silica gel chromatography (5-50% CH3CN/CH2Cl2) to
afford 593 mg (86%)of (R)-1-(1-(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-
yl)ethyl)pyrrolidin-2-one as a white solid (M+H (m/r) = 396).
Example 75


[00452] To a solution of 1 (10 K*i, 45.7 mmol) in DMF (150 mL) were added rffiTU (26
K*i, 6S.5 mmol), dimethylhydroxylamine HC1 salt (5.35 K*i, 54.8 mmol) and DIEA (9,6 mL,
55.0 mmol) at 0°K*I. After stirring for 2h, the mixture was allowed to warm up to room
temperature. Stirring continued for 2 days. The reaction mixture was partitioned between
EtOAc (500 mL) and H2O (200 mL). The organic layer was washed with NaOH (2N, 200
mL). HC1 (2N, 200 mL), H2O, brine, dried over Na2SO4, and concentrated to give 2 (9.6 K*i),
which was used without further purification. LRMS (M+H+) m'z 262.0.
[00453] To a solution of 2 (9.6 K*i, -36.S mmol) in Et:O (100 mL) was added MeMgBr
(3 M in Et:O, 27 ml) at 0°K*I. The resulting mixture was stirred for 4 h while it was allowed to
warm up to room temperature. The reaction mixture was quenched with saturated NH4Cl (100
mL). The organic layer was washed with H2O., brine, dried over Na?SO4, and concentrated to
give 3 (7 K*i, 71 % from 1), which was characterized by NMR.
[00454] To a solution of 3 (6.5 K*i, 30 mmol) in DCM (200 mL) and MeOH ( 100 mL)
was added tetrabutylammonium tribromide (14.5 K*i, 30 mmol). The reaction mixture was
stilted for 14 h. The mixture was concentrated, and dried under high vacuum to give 5
(characterized by NMR), which was used in the next step without further purification.
[00455] To a solution of 4 (5 K*i, -16.9 mmol) in DCM (50 mL) was added
hexamethylenetetramine (2.6 K*i, 18.5 mmol). The reaction mixture was stirred for 2 h. The
mixture was diluted with DCM (500 mL). The precipitate was collected, washed with DCM

(500 mL x 2), and dried under high vacuum. To the resulting residue was added EtOH ( 60
mL) and concentrated HC1 (30 mL). The reaction mixture was stirred for 2 h. The mixture
was concentrated, dried to give 5, which was used without further purification. LRMS
(M+H")/7J/r 231.9.
[00456) To a solution of crude 5 (-16.9 mmol) in dioxane (50 mL) were added NaOAc
(6.93 K*i, 84.5 mmol), HOAc (4.8 mL, 84.5 mmol), and 5.1. (5.93 K*i, 84.5 mmol). After 1 h: the
reaction mixture was warmed up to SO °K*I and stirred for 3 h. The reaction mixture was
partitioned between EtOAc (500 mL) and saturated NaHCC>3 (200 mL). The aqueous layer
was extracted with EtOAc (300 mL x 2). The combined organic layers were washed with
brine, dried over Na2SO4, and concentrated. The resulting residue was purified on silica gel
(hexane/EtOAc, 1:O, 1:2, 1.1, 0:1) to give 6 (1.2 K*i, 23% from 4). LRMS (M+H+) m/z 312.9.

[00457] To a solution of ethyl thiooxamate (10.0 K*i, 75 mmol) in dichloromethane (400
mL) was slowly added trimethyloxonium tetrafluoroborate (13.1 K*i, 89 mmol) at 0 °K*I. After
10 min the ice bath was removed, and the reaction mixture was stirred overnight. The solvent
was removed to give 1S.0 K*i of product 2 as white solid, which was used without further
purification.

[00458] A mixture of 2-amino-4'-bromoacetophene hydrochloride (10.0 K*i, 40 mmol),
sodium acetate (16.4 K*i, 200 mmol), acetic acid (11.5 mL, 200 mmol) and compound 2 (19.2
K*i, 80 mmol) in dioxane (70 mL) was stirred at 65 ° K*I until TLC showed no compound 2 left
(about 2 h). The reaction mixture was carefully neutralized with saturated NaHCCh solution
and extracted with ethyl acetate. The organic solution was dried over NaiSCi and
concentrated. Purification with flash column chromatography (EtOAc:Hexs 1:1) gave product
3 (9.11 K*i, 79 %) as a white solid.

r

[00459J In a round-bottom flask., product 3 (2.00 K*i, 6.8 mmol) was dissolved in DMF
(20 mL), followed by the addition of lodomethane (5.1 mL, 10.1 mmol), andK^COs (1.4 K*i,
10.1 mmol). The mixture was allowed to stir at 60CC for 3 hours until complete by TLC. The
solution was quenched with brine, extracted three times with EtOAc, dried over sodium
sulfate and concentrated. Purification via column chromatography using EtAc:Hex 1:1. gave
1.3S1 K*i (66 % yield) of product 4.

[00460] To a solution of compound 3 (3.174 K*i, 10.S mmol) in DMF (15 mL) was
added K2CO3 (4.478 K*i, 32.4 mmol) and (2-bromoetho.\y)-/er/-butyldimethylsilane (2.780 mL,
13.0 mmol). The resulting mixture was stirred at 55 °K*I overnight. The solution was
concentrated, diluted with water and extracted with EtOAc (3 x 50 mL). The organic layers
were combined and dried over Na2SO4. The solvent was removed to give a viscous oil (4.805
K*i, 10.6 mmol, 98.4%), which was used in the subsequent step without further purification.

[00461] To a solution of compound 4 (2.174 K*i, 4.S mmol) in anhydrous THF (25 mL)
was added dropwise methylmagnesium bromide (4.S mL, 3 M in diethyl ether, 14.4 mmol)
under nitrogen at 0 °K*I. The reaction was stirred at 0 °K*I for 15 minutes. The reaction was
carefully quenched with saturated ammonium chloride solution (5 mL) and water (30 mL)
and extracted with EtOAc (3 x 50 mL). The organic layers were combined, dried over

Na2SO4 and concentrated to a crude oil. Purification with flash column chromatography (15
% EtOAc/Hexanes) gave the desired product 5 (1.371 K*i, 65%) as a white amorphous solid.

(O0462) To a solution of compound 5 (1.371 K*i, 3.1 mmol) in THF (5 mL) was added
35 mL of HC1 (4 M in 1,4-dioxane). The resulting solution was stirred at room temperature
overnight. The solvents were removed to give the product 6 (1.0 K*i, 99%) as white solid.

(O0463] A mixture of compound 6 (0.5 K*i, 1.54 mmol) and 1 mL of TFA in toluene (60
mL) was refluxed overnight. The solid 6 did not dissolve until around the boiling point of
toluene. The solvent was removed. The residue was diluted with EtOAc, washed with
NaHCOj aqueous solution, dried over NaiSC^, and concentrated. Purification with flash
column chromatography (EtOAc:Hexanes 1:1) gave the product 7 (0.348 K*i, 74%) as a white
solid.


[00464] (R)-methvl 1-(4-(4-iodophenvl VI -methyl- 1H-imidazo1-2-
yl)ethyl(methvl)caibamate. To a 250 rtiL round bottom flask was added (R)-1-(4-(4-
iodophenyl)-]-methyl-1H-imidazo1-2-yl)-N-methylethanamine(3.1 K*i, 9.1 mmol), methyl
chloroformate (0.84 mL, 10.9 mmol), Na2CO3 (1.15 K*i? 10.9 mmol), and THF (100 mL). The
reaction was stirred for 2 hours, followed by the addition of EtOAc (50 mL) and water (10
mL). The organic layer was dried over Na2SO4. filtered, and concentrated to give 1.50 K*i
(41 %) of (7?)-methyl 1-(4-(4-iodophenyl)-1-methyl-1H-imidazo1-2-yl)ethyl(methyl)carbamate
as an off-white solid (M+H (m/z) = 40O).

Ref: J. Med. Chem. 2001, 44, 2990-3000
[00465J To a stirring solution of/?-iodoacetophenone 1 (30.0 K*i, 122 mmol) in dioxane
(200 mL) over an ice-bath was added bromine (6.56 mL, 128 mmol) dropwise. The reaction
mixture was stirred at room temperature and monitored by LC/MS. After completion (about
1 hour), the solvent was evaporated by rotovap, and the residue was dried under vacuum to
give solid 2 (40g, 100%).
[00466] (Based on J. Med. Chem. 2001, 44, 2990-300O) To a solution of Cbz-D-Ala-
OH 3 (5.0 K*i, 22.4 mmol) in NMP (100 mL) was added cesium carbonate (3.72 K*i, 11.4 mmol).
After stirring at RT for 1 h, 2 (7.60 K*i, 22.4 mmol) was added. The reaction mixture was

stirred at room temperature and monitored by LC/'MS to form 4. The reaction solution was
diluted with xylene (100 mL) and ammonium acetate (9.25g, 120 mmol) and then stirred at
120°K*I for 4 hours. Up to 50eq of additional ammonium acetate may be needed depending on
the reaction progress. The K*i is to see solid in the flask at all times. After cooling to room
temperature, the reaction mixture was diluted with ethyl acetate (200 mL). The EtOAc
solution was washed with saturated sodium bicarbonate solution (200 mL) twice, and dried
by sodium sulfate, then filtered, and the filtrate was concentrated under feduced pressure. The
residue was dissolved in DCM (100 mL) and stirred for 1 h to give a precipitate, and the solid
5 (4.0g) was filtered off and dried under vacuum. The mother solution was concentrated by
rotovap, the residue was purified on Bio-tage to give 5 (Hexane: EtOAc = 1:1 to EtOAc
100%). The two products were combined and dried under vacuum to give 5 (5.8 K*i, 58%).

100467] (i?)-Benzvl 1-(4-(4-iodophenyl)-l -methyl- 1H-imidazo1-2-
yl)ethyl(mefhyl)carbamate 2: A stirred mixture of (/?)-benzyl 1-(4-(4-iodophenyl)-1H-
imidazo1-2-yl)ethylcarbamate 1 (5 K*i, 11 mmol) in 55 mL of DMF was cooled to 0°K*I and
treated with NaH (1.33 K*i, 60% dispersion in oil, 33 mmol) in small portions to avoid
foaming. When bubbling from the last portion ceased, Mel (2.1 mL, 34 mmol) was added all
at once and the mixture stirred an additional 30 min. The solvents were removed under
vacuum and the residue dissolved in 200 mL of EtOAc. The solution was washed with
saturated NH4C1 (4x100 mL) and saturated NaCl (4x100 mL), and then filtered and
evaporated to dryness. The crude residue was purified via flash column chromatography over
silica gel (60:4O, EtOAc liexanes) to give 5.13 K*i (97% yield) of 2 which was characterized by
LCMS.
Example 80


[00468] Acetyl chloride (54.6 mL, 0.75 mol) was added drop-wise into ethanol (316
mL) at 0-5 K*I. When the addition was completed, the ice bath was removed and the solution
was allowed to stir while warming to room temperature for another 30 mins. D-aspartic acid
1 (25 K*i, 0.188 mol) was then added. The reaction mixture was refluxed for 2 hours. The
reaction solution was then concentrated in vacuo and placed under high vacuum (0.4 mm K*i)
overnight. Compound 2 was obtained as a white solid (42g, 99%) and used directly in the
next step.
[00469] (BOC)2O (44.7 K*i, 0.21 mol) was added portion-wise over 10 mins to a 0 K*I
solution of compound 1 (42 K*i. 0.19 mol), trimethyl amine (51.9 ml, 0.37 mol), dioxane (140
mL) and water (56 mL). After another 10 min, the ice bath was removed and the reaction
mixture was stirred while warming to room temperature for another 2 hours. The reaction
mixture was diluted in ethyl acetate (150 mL) and washed with 0.5 N HC1 (200 mL x 3). The
organic layer was dried over magnesium sulfate, filtered, and the filtrate was concentrated in
vacuo giving compound 3 (52 K*i, yield 97%) which was used directly in the next step.
[00470] NaBH.) (54.4 K*i, 1,44 mol) was added portion-wise over 30 mins to a 0 K*I
solution of compound 3 (52 K*i, 86.4 mmol) and ethanol (600 mL). The reaction mixture was
extremely exothermic and great care was exercised during the addition of reducing agent.
After the addition was complete, the reaction mixture was heated to reflux for 1 hour. The
solution was cooled to ambient temperature and the reaction mixture solidified. The solid was
broken-up to a slurry, which was then poured into brine (250 mL). The resulting mixture was
filtered and the filtrate was concentrated in vacuo. The resulting residue was vigorously
stirred with ether (200 mL x 5). The ether layers were successively decanted from the
residue. The combined ether extracts were dried over magnesium sulfate, filtered, and the

filtrate was concentrated in vacuo giving compound 4 as white solid (25.2g, yield 6S%).
[Note: Yield was S9% when performed on a 25 K*i (compound 3) scale.]
[00471] r-Butyldiphenylchlorosilane (31.9 mL, 0.123 mol) was added to a solution of
compound 4 (25.2 K*i, 0.123 mol), diisopropylethylamine (42.8 mL, 0.245 mol), and CH2C12
(500 mL). The reaction solution was stirred at ambient temperature for 24 hrs. The reaction
solution was then washed with 0.5 N HC1 (150 mL x 3) and brine (150 mL). The organic
layer was dried over magnesium sulfate, filtered, and the filtrate was concentrated /;; vacuo.
The resulting residue was purified by flash chromatography (silica gel, 4:1 hexanes:EtOAc) to
give compound 5 (42g, yield 77%). [Note: Yield was 85% when performed on 15 K*i
(compound 4) scale.]
[00472] Iodine (24 K*i, 94.7 mmol) was added portion-wise over 15 mins to a 0 K*I
solution of compound 5 (28 K*i, 63.1 mmol), PrrjP (24.8 K*i, 94.7 mmol), imidazole (6.4g, 94.7
mmol). diethyl ether (450 mL) and acetonitrile (150mL). The ice bath was removed and the
reaction solution was allowed to warm to ambient temperature over 30 mins. The reaction
was judged complete by TLC analysis (4:1 hexanes:EtOAc). The reaction was quenched with
water (400 mL). The layers were separated and the aqueous layer was extracted by diethyl
ether (100 mL). The combined organic layers were washed with saturated aqueous Na:SO3
(100 x 2) and brine (100 mL). The organic layer was dried over magnesium sulfate, filtered,
and the filtrate was concentrated in vacuo. The resulting residue was purified by flash
column chromatography (silica gel, 4:1 hexanes:EtOAc) to give compound 6 (32 K*i. 92%).
Example 81


f00473] Under ice-bath, to a solution of D-Aspartic acid 1 (59g, 0.376 mol) in
methanol (200 mL) was bubbled HC1 gas for 10 minutes. After the reacting solution was
stirred at RT overnight, the solvent was'evaporated. The resulting residue was dried under
Vacuum to have product 2 as HC1 salt (0.376 mol)i
[00474] To a stirred solution of 2 (0.376mol); DIEA (196 mL, 1.13 mol) and THF (200
mL), benzyl chloroform ate (59,0 mL, 0.414 mol) was dropped in After the reaction solution
was stiiTed at room temperature for 1 hi, the solution was concentrated on rot-vap. Then the
residue was dissolved in NaHCO3 solution (300 mL) and extracted with DCM (100 mL X 3).
The combined DCM solution was dried over sodium sulfate, filtered, and the filtrate was
concentrated in vacuum to have the product 3 (0.376 mol).
[00475] To a solution of 3 (0.376 mol), THF (200 mL) and Water (100 mL) was added
Lithium hydroxide (31.6g, 0.752 mol) and stirred for 2 hours. The reaction mixture was
filtered through a silic gel plug (the pH of the filtrate was about 7) and concentrated. The
residue was dried under Vacuum to give 4 (0.376 mol).
[00476] After a solution of 4 (0.376 mol) and acetic anhydrate (200 mL) was stirred for
1 hour, the reaction mixture was concentrated. The residue was dried under Vacuum to give 5
(0.376 mol).
[00477] Under ice-bath, to a solution of 5 (0.376 mol) and THF (1000 mL) was added

Sodium borohydride (14.2g; 0.376 mol) during 30 minutes and stirred for 3 hrs. Then HC1
solution (4N) was dropped into the reaction solution until the pH was about 2, The solution
was concentrated to about a quarter left, diluted with water (300 mL) and extracted by ethyl
ether (200 mL X 3). The combined ether solution was dried over sodium sulfate, filtered, and
the filtrate was concentrated in vacuum. The resulting residue was dissolved in benzene (300
mL) and TsOH (500 mg) was added. Then the reaction mixture was stirred at reflux 3 hrs.
The solution was concentrated to about 100 ml and ether (200 mL) was added to form
precipitate. The white solid 6 (57.5g, 65% from 1) was filtered out, washed with some ether
and dried under Vacuum.
[00478] To a solution of 6 (30.0g, 0.128 mol) and methanol (200 mL) was added
triethylamine (142 mL, 1.02 mol) and stirred overnight. The reaction mixture was
concentrated. The residue was dried under Vacuum to give 7 (LC-MS showed about 20%mol
6 was left) which was directly used in the next step.
[00479] Under ice-bath, to a solution of compound 7 (0.128 mol), Ph3P (50.4 K*i, 0.192
mol), imidazole (13.1g, 0.192 mol) and DCM (300 mL) was stirred for 10 min. Iodine (48.7
K*i, 0.192 mol) was added portion-wise over 15 minutes. The ice bath was removed and the
reaction solution was stirred at room temperature over 1 hour. The solid was filtered out.
The filtrate was washed with saturated aqueous Na2SC>3 (200 mL x 2) and brine (200 mL).
The organic layer was dried over sodium sulfate, filtered, and the filtrate was concentrated.
The resulting residue was purified by flash silica gel column chromatography
(hexanes:EtOAc 4:lto 1:1) to give compound 8 (27.5 K*i, 59.2% from 6) as a white solid.
[00480] To a mixture of zinc powder (Strem, 10.1 K*i, 0.154 mol) and DMF (15 mL)
was purged under Nitrogen for 10 minutes and added 1,2 dibromoethane (0.758 mL, 8.80
mmol). The mixture was heated with a heat gun for ~2 minutes, cooled down for 5 minutes
and heated with a heat gun again, then cooled to room temperature. TMSC1 (281 i^L, 2.20
mmol) was added to the mixture. After the mixture was stirred for 30 minutes, 8 (9.98 K*i, 26.5
mmol) was added. After 1 hour, LCMS showed complete consumption of 8. To above
reaction solution was added awl iodide 9 (7.50 K*i, 22,0 mmol), Pd2(dba)3 (50.4 mg, 0.55
mmol) and tri-o-tolylphosphine (670 mg, 2.20 mmol). The reaction mixture was maintained
at 50 K*I for 1 hour (monitered on LC-MS analysis). The reaction mixture was directly loaded
to a silica gel plug and washed with hexanes:EtOAc (3:1 to 1:1) to give compound 1O (5.0 K*i,
49%).


[00481] To an oven dried round-bottom flask, zinc powder (1753 mg, 27 mmol) was
added followed by DMF (15 ml). The flask was capped and purged with nitrogen for 10
min. To the solution Avas added 1,2 dibromoethane (0.139 mL. 1.6 mmol). The mixture was
then heated using a heat gun for about 30 seconds until gas began to evolve indicating the
activation of the zinc. The mixture was allowed to cool while slirring for 1 min before it was
heated again using a heat gun until gas evolved. The mixture was then allowed to cool to
room temperature, followed by the addition of TMSC1 (0.042 mL. 0.33 mmol) and stirring for
30 min. Reagent A was then dissolved in DMF, bubbled with nitrogen, added to the zinc
solution, and allowed to stir for 1 hour at room temperature. Bromide 4 (1.381 K*i, 4.5 mmol)
was dissolved in DMF, bubbled with nitrogen and then injected into the solution, followed by
the addition of Pd2(dba3) (102 mg, 0.11. mmol) and Tri-o-tolylphospine (136 mg, 0.44 mmol).
The solution mixture was bubbled with nitrogen and held under nitrogen while stirring for
one hour at room temperature. After 1 hour, the stirring solution was heated to 40°K*I for 2
hours until complete by TLC and LC/MS. The solution was quenched using brine and
extracted five times using EtOAc. The combined organic layers were dried over sodium
sulfate and concentrated. The crude product 5 was purified via column chromatography using
EtOAcHex 1.1 to obtain 2.0 K*i (70 % yield) of pure product 5.

[004S2] To a solution of compound 6 (940 mg, 1.78 mmol) in THF (5 mL) was added

4 M HC1 in 1,4-dioxane (20 mL, 80 mmol). The resulting mixture was stirred at room
temperature for 1 hour. The solvents were evaporated, and the remaining compound 7 (726
mg, 1.78 mmol, quantitative yield) was thoroughly dried under high vacuum. Compound 7
was characterized using LCiMS (LRMS (MH) 373 m/z) and used in the following step without
further purification.

To a solution of compound 7 (662 mg, 1.62 mmql) in DMF (5 mL) was added DIE A (930 µL,
5.34 mmol) and compound 8 (67S mg, 1.78 mmol). The resulting solution was stirred at
room temperature for 1 hour. The DMF was evaporated and the crude residue was directly
purified using preparative HPLC to provide compound 9 (435 mg, 0.77 mmol, 48% yield),
which was characterized by 'H NMR and LC/MS (LRMS (MH) 569 m/z.)

[00483) Compound 9 (100 mg, 0.176 mmol) was combined with HATU (134 mg,
0.352 mmol), HOAT (48 mg. 0.352 mmol) and NFLjCl (50 mg, 0.944 mmol). The solids
were dissolved in N-methylpyrrolidinone (5 mL) and DIEA was added (93 µL, 0.528 mmol).
The resulting mixture was stirred at room temperature for 2 hours and then directly purified
with preparative HPLC to provide compound 1O (16 mg, 0.028 mmol, 16% yield) as a glassy
solid which was characterized by !H NMR and LC/MS (LRMS (MH) 568 m/z.)


[00484] Compound 9 (91 mg, 0.160 mmol) was combined with HBTU (121
mg, 0.320 mmol) and HOBt (43 mg, 0.320 mmol). The solids were dissolved in DMF (3
mL), and dimethylamine (400 µL, 0.800 mmol, 2 M in THF) and DIEA (93 pL, 0.528 mmol)
were added. The resulting solution was stirred at room temperature for 2 hours. The crude
product was directly purified using preparative HPLC to provide compound 11 (25 mg, 0.042
mmol. 26% yield) as a glassy solid which was characterized by !H NMR and LC/MS (LRMS
(MH) 596 m/:.)

[00485] To a 0 DC solution of 1 (40.2 K*i. 117 mmol) in THF (250 mL) and DIEA (11.4
mL, 175 mmol) was added isobutyl chloroformate (21.2 mL, 163 mmol). The resulting
mixture was stirred at room temperature for 3 hours. The reaction was purged with gaseous
ammonia for 1 hour and then stirred at room temperature for 16 hours. It was diluted with
water (200 mL), ethyl acetate (200 mL) and filtered. The white, filtered solid was the desired

product. Additional product was obtained by transferring the biphasic filtrate to a separator}'
funnel and separating the layers. The aqueous phase was extracted with additional ethyl
acetate (3 x 150 mL). The organic phases were combined, dried (Na2SO4) and concentrated
to a white solid, which was recrystallized from ethyl acetate to afford the desired product.
The pure product 2 (20.6 K*i, 60 mmol) was characterized by ' H-NMR and LC/MS (LRMS
(MH)w/*: 343.1).
[00486] Amide 2 (18.1 K*i, 53 mmol) was suspended in 1,4-dioxane (200 mL) and
pyridine (10.7 mL, 132 mmol). Trifluoroacetic anhydride (22.0 mL, 158 mmol) was added,
and the white, suspended solid immediately dissolved. The homogeneous solution was
stirred at room temperature for 30 minutes. The solvents were removed under reduced
pressure, and the remaining residue was dissolved in ethyl acetate (200 mL) and washed with
1 M aqueous KHSQ4 (2 x 100 mL) and saturated aqueous NaHCO3 (2 x 100 mL). The
organic phase was dried over Na2SO4 and concentrated in vacuo. The remaining, desired
product 3 (14.9 K*i, 46 mmol) was determined to be sufficiently pure for the next
transformation (LC/MS (LRMS (MH) m/z: 198.O)).
[004871 Nitrile 3(14.9 K*i, 46 mmol) was dissolved in 1,4-dioxane (100 mL) and tributyl
(1-ethoxyvinyl)tin (23.3 mL, 69 mmol) was added, followed by Pd(PPh3)2Cl2 (1.6 K*i, 5 mol
%). The resulting mixture was heated to 90 DC and stirred for 4 hours. It was cooled to room
temperature and the solvent was removed under reduced pressure. The remaining residue was
directly purified using silica gel, prepared in a slurry using 95% hexane/triethylamine.
Elution was stepwise, beginning with 95% hexane/triethylamine and changing to 50% ethyl
acetate/hexane 5% tnethylamine. The desired product 4 eluted with the latter mobile phase
and was a viscous yellow oil, characterized by LC/MS (LRMS (MH) m/z: 317.1). The
product was used immediately in the next transformation.
[00488] To a solution of vinyl ether 4 (14.5 K*i, 46 mmol) in THF (60 mL) and water (20
mL) was added N-bromosuccinimide (12.3 K*i, 69 mmol). The resulting mixture was stirred at
50 DC for 30 minutes. It was cooled to room temperature and diluted with 2 M aqueous
Na2CC>3. The mixture was extracted with ethyl acetate (2 x 150 mL), the organic extracts
were combined, dried over Na2SO4 and concentrated to an amorphous solid which was
purified using silica gel (dichloromethane/ethyl acetate). The desired product 5 (10.6 K*i, 29
mmol) was a yellow solid, characterized by ' H-NMR and LC/MS (LRMS (MH) m/z: 239.9).
Example 85


[00489] A solution of ketone 5 (10.6 K*i, 29 mmol) in 1,4-dioxane (50 mL) was dripped
into a solution of methylamine (72 mL, 144 mmol, 2 M in THF) over 45 minutes at 0 DC.
The resulting cloudy solution was stirred for an additional 15 minutes at room temperature.
The THF and methylamine were evaporated under reduced pressure, and care was taken not
to evaporate 1,4-dioxane. To the resulting mixture at room temperature was added
triethylamine (12 mL. 87 mmol), followed by trimethylacetyl chloride (15 mL, 144 mmol).
The resulting suspension was stirred at toom temperature for 30 minutes. It was diluted with
water (125 mL) and extracted with ethyl acetate (3 x 100 mL). The organic phases were
combined, dried over Na2SC'4 and concentrated in vacuo. The crude product 6 was
characterized by LC/MS (LRMS (MH) m/z: 402.1) and carried forward without further
purification.
[00490] Amide 6 (11.6 K*i. 29 mmol) was combined with ammonium acetate (55 K*i, 723
mmol) and formamide (150 mL). The resulting mixture was heated to 100 DC and stirred for
3 hours. It was cooled to room temperature, diluted with ethyl acetate (500 mL) and washed
with water (3 x 200 mL). The organic phase was dried over Na2SO4 and concentrated under

reduced pressure. The remaining crude residue was purified using silica gel (diethyl
ether/hexane) to provide pure 7 (6.1 K*i, 16 mmol) as a foamy yellow solid, characterized by
'H-NMR and LC/MS (LRMS (MH) m/z: 383.2).
[00491] Imidazole 7 (1.316 K*i, 3.4 mmol) was combined with hydroxylamine
hydrochloride (478 mg, 6.9 mmol) and dissolved in a solution of sodium methoxide in
methanol (14 mL, 6.9 mmol, 0.5 M). The resulting solution was stirred at 50 DC for 4 hours.
It was concentrated under reduced pressure and directly purified using silica gel (5 %
methanol/dichloromethane) to provide the desired amidoxime 8 (913 mg, 2.2 mmol) as a
white solid, characterized by LC/MS (LRMS (MH) m/z: 416.1).
[00492) To a solution of amidoxime 8 (652 mg, 1.6 mmol) in methanol (10 mL) was
added Raney nickel (50 mg) and acetic acid (250 µL). The mixture was stirred at room
temperature under 60 psi H; for 3 hours and then filtered through abed of celite.
Concentration under reduced pressure provided pure amidine 9 as a white solid (638 mg, 1.6
mmol), characterized using LC/MS (LRMS (MH) m/z: 400.2).
[00493] Chloroacetaldehyde (360 mL, 5.7 mmol) was added to a solution of amidine 9
(283 mg, 0.71 mmol) in DMF (4 mL) and K2CO3 (195 mg, 1.4 mmol). The mixture was
heated to 50 DC and stirred for 4 hours. The reaction was filtered and directly purified by
reverse-phase HPLC using a mobile phase gradient consisting of acetonitrile and water. The
pure product 1O (25 mg, 0.06 mmol)* was a glassy solid characterized by 'H-NMR and
LC/MS (LRMS (MH) m/z: 424.1).

[00494] To a solution of amidoxime 8 (148 mg, 0.35 mmol) in
trimethylorthoacetate (5 ml.) was added glacial acetic acid (100 µL). The resulting solution
was stirred at 65 DC for 16 hours. The solvents were evaporated and the residue was directly
purified through silica gel (5% methanol/dichloromethane) to provide the desired product 9 as
a glassy solid, characterized by LC/MS (LRMS (MH) m/z: 440.1.


[00495J To a solution of 1 (200 mg, 0.5 mmol) in THF (3 mL) were added Bu3P (150
µL, 0.6 mmol) and 2-nitrophenylselenocyanate (136 mg, 0.6 mmol) at room temperature. The
reaction mixture was stirred for 14 h. The mixture was partitioned between EtOAc (200 mL)
and FLO (50 mL). The organic layer was washed with brine, dried over NajSCu, and
concentrated. The resulting residue was used without further purification.
LRMS (M+H*);?^ 587.1.
[00496J To a solution of 2 (-0.5 mmol) in DCM (5 mL) were added aqueous KH2P04
(2 M, 1 ml) and MCPBA (77%, 135 mg, 0.6 mmol). The resulting mixture was stirred for 6
h. The reaction mixture was quenched with saturated Na2S2C>3 (10 mL). The organic layer
was washed with saturated NaHCOj, FLO, brine, dried over Na:SO.i, and concentrated. The
residue was purified on RP-HPLC using a mixture of acetonitrile and H2O to give 3 (150 mg,
65% from 1). LRMS (M+H+) m/z 3S4.2.
[00497J To a solution of 3 (150 mg, 0.39 mmol) in DCM (S mL) was added TFA (1
mL). The reaction mixture was stirred for 4h. The mixture was concentrated, and dried under
high vacuum. To the resulting residue (90 mg, 0.32 mmol) in THF (4 mL) were added DIEA
(165 µL, 0.95 mmol) and 4 (140 mg, 0.38 mmol). The resulting mixture was stirred for 14 h.
The reaction mixture was concentrated. The residue was purified on RP-HPLC using a
mixture of acetonitrile and FLO to give 5 (120 mg, 65%). LRMS (M+Ff) m/z AIM.
[00498) To a solution of 5 (90 mg, 0.19 mmol) in THF/PLO (2mL/2mL) were added
Os04 (4.S mg, 0.019 mmol), NMO (117 mg, 0.95 mmol) and pyridine (1.5 µL, 0.019 mmol).
The resulting mixture was stirred for 6 h. NaHSO] (300 mg) was added. The reaction mixture

was concentrated. The resulting solid was washed with EtOAc (100 mL x 3). The filtrate was
concentrated. The resulting residue was purified on preparative TLC plate (silica gel, 5:1
EtOAc/MeOH) to give diasteroisomers 6a (23 mg, 24%) and 6b (2 mg, 2%). LRMS (M+H4)
m/z 505.2.

[00499] To a solution of amine 1 (150 mg, 0.309 mmol), DCM (2 mL) and DIEA
(53.8 µL, 0.309 mmol) was added acetyl chloride (53.8 µL, 0.309 mmol). The resulting
solution was stirred at room temperature for 10 minutes. The solvent was evaporated, and the
remaining residue was purified on reverse phase Prep-HPLC (AcetonitrileAVater) to provide
2 (43.7 mg, 26.8%). MS (MW+1): 527.2

[00500] To a solution of amine 1 (150 mg, 0.309 mmol), DCM (2 mL) and DIEA
(53.8 µL, 0.309 mmol) was added trimethylsilyl isocyanate (35.6 µL, 0.309 mmol). The
resulting solution was stirred at room temperature overnight. The solvent was evaporated,

and the remaining residue was purified on reverse phase Prep-HPLC (Acetonitrile/Water) to
provide 3 (30.3 mg, 18.6%). A/S(MW+1): 528,2

[ 00501]
To a solution of amine 1 (150 mg, 0.309 mmol), DCM (2 mL) and DIEA (53.8 µL, 0.309
mmol) was added methanesulfonyl chloride (24 µL, 0.309 mmol). The resulting solution was
stirred at room temperature for 30 minutes. The solvent was evaporated, and the remaining
residue was purified on Prep-HPLC (Acetonitrile/Water) to provide 4 (18.4 mg, 10.6%). MS
(MW+1): 563.1

(O0502] To a solution of amine 1 (150 mg, 0.309 mmol), DCM (2 mL) and DIEA
(53.8 µL, 0.309 mmol) was added methyl chloroformate (24 µL, 0.309 mmol). The resulting
solution was stirred at room temperature for 30 minutes. The solvent was evaporated, and the

remaining residue was purified on reverse phase Prep-HPLC (Acetonitrile/Water) to provide
5 (CK1828648) (25.7 mg, 15.3%). MS(MW+1): 543.1

(S)-3-chloro-N-(4-hydroxy-1-(4-(2-(1-(methoxyimino)ethyl)-l -methyl- 1H-imidazo1-4-
yl)phenyl)butan-2-yl)-4-isopropoxybenzamide 2:
[00503] 80 mg (0.031 mmol) of (S)-N-(l -(4-(2-acetyl-1-methyl- 1H-imidazo1-4-
yl)phenyl)-4-hydroxybutan-2-yl)-3-chloro-4-isopropoxybenzamide in 2 mL of pyridine was
treated with 27.6 mg (0.033 mmol) of hydroxylamine methyl ether hydrochloride. The
reaction was stirred overnight after which the solvents were evaporated and the residue
purified via reverse phase HPLC (acetonitrile/water). 11.2 mg (70% yield) of 2 was obtained
and characterized by LCMS and HNMR.

(S)-3-chloro-N-(4-hydroxy-1-(4-(2-( 1-(hydroxyimino)ethyl)-1-methyl-1H-imidazo1-4-
yl)phenyl)butan-2-yl)-4-isopropoxybenzamide 3:
[005041 100 mg(0.21 mmol) of (S)-N-(1-(4-(2-aceryl-l -methyl- 1H-imidazo1-4-
yl)phenyl)-4-hydroxybutan-2-yl)-3-chloro-4-isopropoxybenzamide in 2 mL of pyridine was

treated with 71.8 mg (1.0 mmol) of hydroxylamine hydrochloride. The reaction was stirred
overnight after which the solvents were evaporated and the residue purified via reverse phase
HPLC (acetonitrile/water). 69.7 mg (67% yield) of 3 was obtained and characterized by
LCMS and HNMR.

(S)-3-chloro-N-(4-hydroxy-1-(4-(1-methyl-2-(2-methyl-l,3-dioxolan-2-yl)-1H-imidazo1-4-
yl)phenyl)butan-2-yl)-4-isopropoxybenzamide 4:
[00505] 150 mg (0.31 mmol) of (S)-N-(l -(4-(2-acetyl-l -methyl- 1H-imidazo1-4-
yl)phenyl)-4-hydroxybutan-2-yl)-3-chloro-4-isopropoxybenzamide in 2 mL of benzene was
treated with 34.6 µL (0.62 mmol) of ethane-1,2-diol and 59 mg (0.31 mmol) of p-
toluenesulfonic acid monohydrate. The reaction was stirred at 70°K*I for 2 h after which the
solvents were evaporated and the residue purified via reverse phase HPLC
(acetonitrile/water). 25.5 mg (16% yield) of 4 was obtained and characterized by LCMS and
HNMR.

[00506] To a solution of 1 (1.5 K*i, 2.29 mmol), in ethanol (5.0 mL) was added

NaOH in water (1.0 M, 3.7 mL, 2.80 mmol). The reaction mixture was stirred at ambient
temperature for 2 hours. After the reaction was done it was concentrated to give 2 (1.49 K*i)
which was used directly in the next step without further purification,

[00507] To a solution of 2 (1.49 K*i, 2.29 mmol), HBTU (1.3 K*i, 3.44 mmol),
HOBt (530 mg, 3.44 mmol), and N, O-dimethylhydroxylamine HC1 salt (340 mg, 3.44 mmol)
in DMF (20 mL) was added DIEA (785 µL, 4.58 mmol). The resulting mixture was stirred at
room temperature for 2 hours. The reaction mixture was concentrated. The resulting residue
was purified using silica gel (Hexanes/ EtOAc = 1:3) to give pure compound 3 (1.20 K*i, 78%)
as an off-white, foamy solid.

[00508] To a 0° K*I solution of 3 (1.20g, 1.79 mmol) in anhydrous THF (20 mL) was
added Methylmagnesium bromide (3 M in Et2O,2.38 mL). The reaction mixture was stirred
at 0° K*I for 1 hour. The reaction mixture was quenched with saturated NH4Cl (5 mL) and
water (20 mL). EtOAc (50 mL) was added, and the layers were separated. The aqueous
phase was extracted with additional EtOAc (50 mL x 2). The organic phases were combined,
dried (Na2SO4) and concentrated to a crude oil which was purified using silica gel (50 %
EtOAc/Hexanes). The desired compound 4 (0.82 K*i, 73%) was a viscous oil which became a
white foamy solid while drying under high vacuum.


[00509] To a solution of 4 (0.82 K*i. 1.31 mmol) in Methanol (10 mL) was added
HC1 (4 M in 1,4-dioxane, 20 mL). The reaction was stirred at room temperature overnight.
The mixture was concentrated, and dried under high vacuum to give 5, which was used in the
following step without further purification.

[00510] To a solution of 5 (350 mg, 1.09 mmol) in DMF (5 mL) was added DIEA (280
µL, 1.63 mmol) and ester H (472 mg, 1.09 mmol). The resulting solution was stirred at room
temperature for 1 hour. The crude solution was filtered and then purified by reverse phase
chromatography (using a mixture of acetonitrile and water) to provide CK1904250 as a foamy
white solid (400mg, 68%). LRMS (M+H+)m/z 538.1.

[00511] Ester D (10.2 K*i, 24.5 mmol) was dissolved in EtOH (150 mL) and
water (50 mL). Postassium hydroxide (4.1 K*i, 73.5 mmol) was added, and the reaction was
stirred at room temperature overnight. The reaction mixture was cooled to 0 K*I and
neutralized with concentrated HC1. Great care was taken to not allow the pH to become during the neutralization. The solvents were evaporated in vauco, and the residue was dried
under high vacuum. Acid E (9.5 K*i, 24.5 mmol) was used in the next step without further

purification.

[005121 Acid E (9.5 K*i, 24.5 mmol) was combined with HSTU (18.5 K*i, 48.7
mmol), HOBt (6.6 K*i, 48.7 mmol), and N.O-dimethylhydroxylamine HC1 (4.8 K*i, 48.7 mmol).
To the solids were added DMF (150 mL) and DIEA (12.7 mL, 73.1 mmol). The resulting
mixture was stirred at room temperature for 4 hours. Most of the DMF was evaporated, and
the remaining residue was diluted with ethyl acetate (300 mL) and water (300 mL). The
layers were separated, and the aqueous phase was extracted with EtOAc (1 x 200 mL). The
organic phases were combined, washed with saturated aqueous sodium bicarbonate (2 x 250
mL), and dried over Na2SO4. Concentration under reduced pressure provided crude amide E
which was purified using silica gel (3 % MeOH/DCM) to give pure amide E (6.73 K*i, 17.4
mmol) as an off-white, foamy solid.

[00513] A solution of amide E (6.73 K*i, 17.4 mmol) in anhydrous THF (250 mL) was
cooled to 0 K*I with an ice bath. Methylmagnesium bromide (3 M in diethyl ether, 52.2 mL,
156.6 mmol) was added, and the reaction was stirred at 0 K*I for 15 minutes. The reaction was
carefully quenched with saturated ammonium chloride solution (20 mL) and water (100 mL).
EtOAc (200 mL) was added, and the layers were separated. The aqueous phase was
extracted with additional EtOAc (2 x 200 mL). The organic phases were combined, dried
(Na2SO4) and concentrated to a crude oil which was purified using silica gel (50 %
EtOAc/Hexanes). The desired ketone F (4.45 K*i, 11.5 mmol) was a viscous oil which became
a white foamy solid while drying under high vacuum.


[00514] Ketone F (4.45 K*i, 11.5 mmol) was dissolved in THF (25 mL) and 4 M HC1 in
1,4-dioxane was added (75 mL). The reaction was stirred at room temperature for 1.5 hours.
The solvents were evaporated in vacuo, and the residue was thoroughly dried under high
vacuum to provide amine K*I. Amine K*I was used in the following step without further
purification.

(O0515] To a solution of amine K*I (3.30 K*i, 11.5 mmol) in DMF (50 mL) was added
DIEA (8.0 mL. 46.0 mmol) and ester H (5.25 K*i, 13.S mmol). The resulting solution was
stirred at room temperature for 1 hour. Most of the DMF was evaporated, and the remaining
residue was diluted with EtOAc (250 mL) and water (200 mL). The layers were separated,
and the organic phase was washed with additional water (2 x 150 mL) and brine (2 x 150
mL). The organic phase was dried (Na2SO4) and concentrated. The remaining crude, viscous
oil was purified using silica gel (100% EtOAc) to provide CK1317644 as a foamy white solid
(2.98 K*i, 6.2 mmol).

[00516] To a solution of ethyl thiooxamate (10.0 K*i, 75 mmol) in dichloromethane (400
mL) was slowly added trimethyloxonium tetrafluoroborate (13.1 K*i, 89 mmol) at 0 °K*I. After

10 min the ice bath was removed, and the reaction mixture was stirred overnight. The solvent
was removed to give 18.0 K*i of product 2 as white solid, which was used without further
purification.

[00517] A mixture of 2-amino-4'-bromoacetophene hydrochloride (10.0 K*i, 40 mmol),
sodium acetate (16.4 K*i, 200 mmol). acetic acid (11.5 mL, 200 mmol) and compound 2 (19.2
K*i, 80 mmol) in dioxane (70 mL) was stirred at 65 ° K*I until TLC showed no compound 2 left
(about 2 h). The reaction mixture was carefully neutralized with saturated NaHCO3 solution
and extracted with ethyl acetate. The organic solution was dried over Na2SO4 and
concentrated. Purification with flash column chromatography (EtOAc:Hexs 1:1) gave product
3 (9.11 K*i, 79 %) as a white solid.

[00518] To a solution of compound 3 (3.174 K*i, 10.8 mmol) in DMF (15 mL) was
added K2CO3 (4.478 K*i. 32.4 mmol) and (2-bromoethoxy)-tert-butyldimethylsilane (2.780 mL.
13.0 mmol). The resulting mixture was stirred at 55 °K*I overnight. The solution was
concentrated, diluted with water and extracted with EtOAc (3 x 50 mL). The organic layers
were combined and dried over Na2SO4. The solvent was removed to give a viscous oil (4.805
K*i, 10.6 mmol, 98.4%), which was used in the subsequent step without further purification.

[00519] To a solution of compound 4 (2.174 K*i, 4.8 mmol) in anhydrous THF (25 mL)

was added dropwise methylmagnesium bromide (4.S mL, 3 M in diethyl ether, 14.4 mmol)
under nitrogen at 0 °K*I. The reaction was stirred at 0 °K*I for 15 minutes. The reaction was
carefully quenched with saturated ammonium chloride solution (5 mL) and water (30 mL)
and extracted with EtOAc (3 x 50 mL). The organic layers were combined, dried over
Na2SO4 and concentrated to a crude oil. Purification with flash column chromatography (15
% EtOAc/Hexanes) gave the desired product 5 (1.371 K*i, 65%) as a white amorphous solid.

[00520] To a solution of compound 5 (1.371 K*i, 3.1 mmol) in THF (5 mL) was added
35 mL of HC1 (4 M in 1,4-dioxane). The resulting solution was stirred at room temperature
overnight. The solvents were removed to give the product 6 (1.0 K*i, 99%) as white solid.

[00521] A mixture of compound 6 (0.5 K*i, 1.54 mmol) and 1 mL of TFA in toluene (60
mL) was refluxed overnight. The solid 6 did not dissolve until around the boiling point of
toluene. The solvent was removed. The residue was diluted with EtOAc, washed with
NaHCO3 aqueous solution, dried over Na2SO4, and concentrated. Purification with flash
column chromatography (EtOAc:Hexanes 1:1) gave the product 7 (0.348 K*i, 74%) as a white
solid.

[00522] To a suspension of zinc powder (255 mg, 3.9 mmol) in dry degassed DMF (15
mL) was added 1,2 dibromoethane (.020 mL, 0.23 mmol) under nitrogen. The mixture was

heated using a heat gun for about 30 seconds until gas starts to evolve from the solution,
indicating the activation of the zinc. The mixture was then allowed to cool to room
temperature followed by the addition of TMSC1 (6 µL, 0.05 mmol), and allowed to stir at
room temperature for 30 min. A solution of iodo compound A in degassed DMF was added to
the zinc solution, and the reaction mixture was stilted for 1 hour at room temperature. Then a
solution of compound 7 (200 mg, 0.65 mmol) in degassed DMF was added via a syringe,
followed by the addition of Pd2(dba3) (14.9 mg, 0.016 mmol) and tri-o-tolylphospine (19.8
mg, 0.065 mmol). The reaction mixture was stirred for one hour at room temperature, then at
40 ° K*I for 2 hours. The reaction was complete as shown on TLC. The solution was quenched
with brine and extracted with EtOAc (5 x 50 mL). The combined organic layers were dried
over sodium sulfate and concentrated. Purification with flash column chromatography
(EtOAc:Hex 1:1) gave the product 8 (373 mg, 88 %) as a colorless oil.

[00523| To a solution of compound 8 (373 mg, 0.57 mmol) in MeOH (10 mL) was
added 2 mL of HC1 (4.0 M in Dioxane). The solution was allowed to stir at room temperature
for 2 hours. The solvent was removed to give the crude product 9 (180 mg, 99%), which was
used without further purification.

[00524] A mixture of compound 9 (180 mg, 0.57 mmol) and ester reagent B (260 mg,
0.68 mmol) in DMF (10 mL) containing triethylamine (0.24 mL, 1.71 mmol) was stirred at

room temperature overnight. The reaction solution was diluted with brine and extracted with
EtOAc (3 x 50 mL). The combined organic layers were dried over sodium sulfate and
concentrated. Purification with HPLC (C18 column) gave the product 1O (141 mg, 50%) as a
white solid.

(O0525] To a suspension of NaH (0.39 K*i, 9.3 mmol) in DMF (15 mL) was added a
solution of 3 (1.9 K*i, 6.5 mmol) in DMF (10 mL) at 0 °K*I under nitrogen. The reaction was
stirred for 1.5 hour, and then (2-bromoethoxy)-tert-butyldimethylsilane (2.09 mL, 9.7 mmol)
was added. The reaction mixture was stirred overnight, diluted with EtOAc, quenched with
aqueous ammonium chloride solution, and extracted with EtOAc (3 x 50 mL). The organic
layers were combined and dried over Na2SO4. Purification with biotage (EtOAc) gave the
product 4 (1.2 K*i, 41 %) as light yellow solid.

[00526] To a suspension of zinc powder (1.2 K*i, 18.4 mmol) in dry degassed DMF (15
mL) was added 1,2 dibromoethane (0.13 mL, 1.5 mmol) under nitrogen. The mixture was
heated using a heat gun for about 30 seconds until gas starts to evolve from the solution,
indicating the activation of the zinc. The mixture was then allowed to cool to room
temperature followed by the addition of TMSC1 (100 µL), and allowed to stir at room
temperature for 30 min. A solution of iodo compound A (1.71 K*i, 3.1 mmol) in degassed DMF
was added to the zinc solution, and the reaction mixture was stirred for 1 hour at room
temperature. Then a solution of compound 4 (1.0 K*i, 2.2 mmol) in degassed DMF was added

via a syringer, followed by the addition of Pd2(dba3) (0.14 K*i, 0.015 mmol) and Tri-o-
tolylphospine (0.18 K*i, 0.06 mmol). The reaction mixture was stirred for one hour at room
temperature, then at 60 ° K*I overnight. The solution was quenched with brine and extracted
EtOAc (5 x 50 mL). The combined organic layers were dried over sodium sulfate and
concentrated. Purification with flash column chromatography (EtOAc:Hex 1:1) gave the
product 5 (346 mg, 20%) as colorless oil.

(O0527] To a solution was compound 7 (346 mg) in MeOH (10 mL) was added 2 mL
of HC1 (4.0 M in Dioxane). The solution was allowed to stir at room temperature for 2 hour.
The solvent was removed to give the crude product 7, which was used for the next step
without further purification.

[00528] A mixture of compound 7. ester reagent B (200 mg, 0.52 mmol) in DMF (10
mL) containing triethylamine (0.15 mL, 1.08 mmol) was stirred at room temperature over
night. The reaction solution was diluted with brine, extracted with EtOAc (3 x 50 mL). The
combined organic layers were dried over sodium sulfate and concentrated. Purification with
HPLC (CI 8 column) gave the product S (0.2 K*i, 87%) as white solid, and the lactone product 9
(15.4 mg, 7.3%) as white solid. LC-MS (CI) m/z 489.1 (MH+)
Example 96


[00529) To a solution of ethyl thiooxamate (10.0 K*i, 75 mmol) in dichloromethane (400
mL) was slowly added trimethyloxonium tetrafluoroborate (13.1 K*i, 89 mmol) at 0 °K*I. After
10 min the ice bath was removed, and the reaction mixture was stirred overnight. The solvent
was removed to give 18.0 K*i of product 2 as white solid, which was used without further
purification.

[00530] A mixture of 2-amono-4'-bromoacetophene hydrochloride (10.0 K*i, 40 mmol),
sodium acetate (16.4 K*i, 200 mmol), acetic acid (11.5 mL, 200 mmol) and compound 2 (19.2
K*i, 80 mmol) in dioxane (70 mL) was stirred at 65 ° K*I until TLC show no compound 2 left
(about 2 h). The reaction mixture was carefully neutralized with saturated NaHCO3 solution,
and extracted with ethyl acetate. The organic solution was dried over Na2SO4 and
concentrated. Purification with flash column chromatography (EtOAc:Hexs 1:1) gave product
3 (9.11 K*i, 79 %) as white solid.

[00531) To a solution of compound 3 (5.307 K*i, 18 mmol) in DMF (15 mL) was added
K2CO3 (3.73 K*i, 27 mmol) and iodoethane (3.5 mL, 43.2 mmol). The resulting mixture was
stirred at 60 °K*I for three hours. The mixture was diluted with water and extracted with
EtOAc (3 x 50 mL). The organic layers were combined, dried over Na2SO4 and concentrated.
Purification with column chromatography (Hexanes/EtOAc 50:50) gave the product 4 (3.2 K*i,
55 %) as white solid.


[00532] To a suspension of zinc powder (3.90 K*i, 59.6 mmol) in dry degassed DMF (10
mL) was added 1,2 dibromoethane (308 µL, 3.5S mmol) under nitrogen. The mixture was
heated using a heat gun for about 30 seconds until gas starts to evolve from the solution,
indicating the activation of the zinc. The mixture was then allowed to cool to room
temperature followed by the addition of TMSC1 (92 uL. 0.735 mmol), and allowed to stir at
room temperature for 30 min. A solution of iodo compound A (6.6 K*i, 11.9 mmol) in degassed
DMF was added to the zinc solution, and the reaction mixture was stirred for 1 hour at room
temperature. Then a solution of compound 4 (3.2 K*i, 9.93 mmol) in degassed DMF was added
via a syringe, followed by the addition of Pd2(dba3) (223 mg. 0.244 mmol) and Tri-o-
tolylphospine (302 mg, 0.992 mmol). The reaction mixture was stirred for one hour at room
temperature, then at 60 ° K*I for 2 hours. The reaction was complete as shown on TLC. The
solution was quenched with brine and extracted EtOAc (3 x 80 mL). The combined organic
layers were dried over sodium sulfate and concentrated. Purification with flash column
chromatography (EtOAc:Hex 1:1) gave the product 5 (5.43 mg, 82 %) as colorless oil.

[00533] To a solution of compound 5 (5.43 K*i, 8.1 mmol) in THF (50 mL) was added
dropwise a solution of MeMgBr bromide in ether (9.0 mL, 27 mmol) at 0 ° K*I under nitrogen.
The reaction was complete in 10 min via TLC. The solution was quenched by aqueous
ammonium chloride solution while in ice, extracted with EtOAc (3 x 60 mL). The combined
organic layers were dried over sodium sulfate and concentrated. Purification with column
chromatography (Hexanes/EtOAc 1:1) gave the product 6 (4.86 K*i, 91 %) as colorless oil.


[00534] A mixture of compound 6 (4.86 K*i, 7.4 mmol), and 18 me of HC1 (4M in
Dioxane) in MeOH (10.0 mL) was stirred at room temperature for 1 hour, followed by
heating at 60 ° K*I for 30 min. The reaction was complete via TLC and LC/MS. The solvent
was removed to give the product 7, which was directly used for the next step.

[00535] A mixture of acid B (677 mg, 2.51 mmol), HBTU (3.6 K*i, 9.49 mmol), HOBT
(1.45 K*i, 9.46 mmol) and DIEA (2.20 mL, 12.6 mmol) in DMF (40 mL) was stirr at room
temperature for 1 min followed by the addition of 7 (1.0 K*i. 3.14 mmol). The reaction was
complete in one hour via TLC and LC/MS. The solution was partitioned in EtOAc and brine,
and extracted with EtOAc. The combined organic layers were dried over sodium sulfate and
concentrated down. Purification with HPLC gave product 8 (390 mg) as white solid.

[00536] To a solution of compound 3 (2.66 K*i; 7.27 mmol) in DMF (15 mL) was added
K2CO3 (2.00 K*i, 15 mmol) and ethyl bromoacetate (1.61 mL, 14.5 mmol). The resulting
mixture was stirred at 60 °K*I for three hours. The mixture was diluted with water and
extracted with EtOAc (3 x 50 mL). The organic layers were combined, dried over Na2SO4,

and concentrated. Purification with column chromatography (Hexanes/EtOAc 50:50) gave the
. product 4 (3.02 K*i, 91 %).

[00537] To a solution of compound 4 (3.02 K*i, 6.7 mmol) in MeOH (20 mL) was added
HC1 (4.0 M) in Dioxane (7.0 mL) and stirred at 60 °K*I for one hours. The mixture was
concentrated and no purification was done. The resulting oil was dissolved in DMF (15 mL),
added K2CO3 (2.0 K*i, 14.7 mmol) was added, and stirred at 60 °K*I for overnight. The mixture
was diluted with water and extracted with EtOAc' (3 x 50 mL). The organic layers were
combined, dried over Na2SO4, and concentrated. Purification with column chromatography
(Hexanes/EtOAc 50:50) gave the product 5 (1.80 K*i, 88 %).

[00538] To a solution of compound 3 (5.000 K*i, 17 mmol) in DMF (15 mL) was added
K2CO3 (3.51 K*i, 26 mmol) and Boc-2-amino ethyl bromide (4.56 K*i, 20.35 mmol). The
resulting mixture was stirred at 60 °K*I for three hours. The mixture was diluted with water
and extracted with EtOAc (3 x 50 mL). The organic layers were combined, dried over
Na2SO3, and concentrated. Purification with column chromatography (Hexanes/EtOAc 50:50)
gave the product 4 (4.08 K*i, 55 %) as white solid.



[00539] To a solution of 1 (10.7 K*i, 34.6 mmol) in MeOH / H2O (60 mL / 20 mL) was
added NaOH (2N, 20.8 ml, 41.6 mmol). After the mixture was stirred at 50 °K*I for 2 h, the
solution then was concentrated and under high vacuum to yield 10.3 K*i of light yellow solid
(LRMS (M-H+) m/z 278.9), which was used for the next step without further purification. To
a solution of the crude mixture in DMF (50 mL) were successively added N,0-
dimethylhydroxylamine hydrochloride (4.0 K*i, 40.7 mmol), HBTU (4.0 K*i, 40.7 mmol), HOBT
(6.2 K*i, 40.7 mmol) and DIEA (6.0 ml, 40.7 mmol). The mixture was stirred at rt overnight.
The solution then was partitioned between EtOAc and H2O. The organic layer was washed
with NaOH (1 N), brine, dried over Na2SO4, filtered and concentrated. The residue was
purified by flash column chromatography using a mixture of hexanes and EtOAc to give 2 (8
K*i, 72%). LRMS (M+H+) m/z 324.0.

[00540] To a solution of 2 (3.7 K*i, 11.4 mmol) in THF (40 mL) was added dropwise
MeMgBr in Et2O (3M, 11.4 ml, 34.2 mmol) at 0 °K*I. The mixture was stirred at 0 °K*I for 30
min. The solution was quenched by saturated NH4Cl at 0 °K*I and diluted between EtOAc and
H2O. The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated
to give 3 (3.0 K*i, 94%) without further purification. LRMS (M+H+) m/z 279.0.


[00541] To a solution of 3 (3.0 K*i, 10.8 mmol) in THF / MeOH (10mL /10 mL) was
slowly added NaBH4 (407 mg. 10.8 mmol). The mixture was stirred for 10 min, quenched by
saturated NH4Cl and partitioned between EtOAc and H2O. The organic layer was washed
with SatNaHCOs, brine, dried over Na2SO4, filtered and concentrated to give 4 (3.0 K*i, 99 %),
which was used without further purification. LRMS (M+H+) m/'z 281.0.

[00542] To a solution of 4 (3.0 K*i, 10.7 mmol) in DMF (20 mL) was added TBDMSC1
(1.6 K*i, 10.7 mmol), imidazole (726 mg, 10.7 mmol) and DMAP (271 mg, 21.3 mmol). The
mixture was stirred at rt overnight. The solution was partitioned between EtOAc and H2O.
The organic layer was washed with Sat NaHCO3, H2O, brine, dried over Na2SO4, filtered and
concentrated. The residue was purified by column chromatography using a mixture of
hexanes and EtOAc to give 5 (3.5 K*i, 83%). LRMS (M+H+) mtz 395.1.

[00543] To a suspension of Zn (4.S K*i, 74.4 mmol) in DMF (20 mL) was added
BrCH2CH2Br (320 µL, 3.7 mmol). The mixture was heated by heat gun for 4 min. After the
solution was cooled down, trimethylchlorosilane (95 µL, 0.74 mmol) was added. After 30

min, BOC-β-iodo-Ala-OMe (5.2 K*i, 16.0 mmol) was added, and reaction mixture was stirred
at rt for lh. To this mixture then were added Pd2(dba)3 (243 mg, 0.27 mmol), (O-Tol)3P (269
mg, 0.88 nimol) and 5 (3.5 K*i. 8.9 mmol). The mixture was heated at 50 °K*I for 2h. cooled
down and filtered through celite. The solution was partitioned between EtOAc and H2O. The
organic layer was washed with brine, dried over Na2SO4, filtered and concentrated. The
residue was purified by column chromatography using a mixture of hexanes and EtOAc to
give 6 (3.3 K*i, 72%). LRMS (M+H+) m/z 518.2.

[00544J To a solution of 6 (3.3 K*i, 6.4 mmol) in THF ( 20 ml) was slowly added LAH
(1 M, 6.4 mL, 6.4 mmol) at 0 °K*I. The mixture was stirred at 0 °K*I in 20 min, quenched by
H2O (240 µL), NaOH (3N, 240 µL), H2O (720 µL), filtered. The organic layer was dried over
Na2SO4, filtered and concentrated. The residue was purified by column chromatography using
a mixture of hexanes and EtOAc to give 7 (1.57 K*i, 50%). LRMS (M+H+) m/z 490.2.

[00545] To a solution of 7 (1.57g, 3.2 mmol) in THF (20 mL) were added PPh3 (1.0 K*i,
3.9 mmol), DIAD (746 µl, 3.9 mmol) and phthalimide (567 mg, 3.9 mmol). After the
solution was stirred at rt for 4 hours, which was monitored by LCMS, the reaction was
partitioned between EtOAc and H2O. The organic layer was washed with brine, dried over
Na2SO4, filtered and concentrated. The residue was purified by column chromatography
using a mixture of hexanes and EtOAc to give 8 (2.0 K*i, 99%). LRMS (M+H+) m/z 619.2.


100546] To a solution of 7 (2g, 3.2mmol) in MeOH (15 ml) was added NH2NH2
(1.01ml, 32.3 mmol). After the reaction was stirred at room temperature for about 4 h. the
solution was precipitated, filtered, washed with CH2Cl2, MeOH. The organic layer was
concentrated to give 8 (2.5 K*i), which was used without further purification. LRMS (M+H+)
m/z 489.2.

[00547] To a solution of 8 (1.5g, 3.1 mmol) in CH2C12 / CH3CN (15 ml /15 ml) were
added DIEA (588 µL, 3.4 mmol) and chloroacetyl chloride (269 µL, 3.4 mmol). After the
reaction was stirred at rt for 10 min, azetidine (2 ml, 30.7 mmol) and DIEA (2.7 ml, 15.3
mmol) were added. The reaction mixture was stirred overnight. The solution was
concentrated and diluted between EtOAc and H2O. The organic layer was washed with brine,
dried over Na2SO4, filtered and concentrated. The residue was purified by column
chromatography using a mixture of hexanes and EtOAc to give 9 (900 mg, 51%) LRMS
(M+H+)m/~ 586.3.


[00548] To a solution of 9 (900 mg, 1.53 mmol) in MeOH (1 mL) were added HC1 in
dioxane (4 N, 2 ml) and HC1 in H2O (2 N. 1ml). The solution was stirred at rt overnight,
concentrated to give white solid and directed for the next coupling step, To a DMF (10ml)
solution of the crude compound were added 9.1 (665 mg, 1,53 mmol) and DIEA (800 ul, 4.59
mmol). The mixture was stirred at rt for 1 h and partitioned between EtOAc and H2O. The
organic layer was washed with brine, dried over Na2SO4, filtered and concentrated. The
residue was purified by reverse-phase HPLC to give 1O (600 mg, 63 %). LRMS (M+H+) m/z
622.2.

(O0549] To a solution of 1O (160 mg, 0.24 mmol) in DCM (10 mL) was added MnO2
(416 mg, 4.8 mmol). The suspension was stirred for 14 h. The reaction mixture was filtered,
and the filtrate was concentrated and purified on reverse-phase HPLC using a mixture of
acetonitrile and H2O to give 11 (90 mg, 60%). LRMS (M+H4) m/z 620.1.
Example 98
[00550] The following compounds were prepared using the procedures described
above:















Example 99
Inhibition of Cellular Viability in Tumor Cell Lines Treated with Mitotic Kinesin
Inhibitors
(O0551] Materials and Solutions:
• Cells: SKOV3, Ovarian Cancer (human).
• Media: Phenol Red Free RPMI + 5% Fetal Bovine Serum + 2mM L-glutamine.
• Colorimetric Agent for Determining Cell Viability: Promega MTS tetrazolium
compound.

• Control Compound for max cell kill: Topotecan, 1 µM.
Procedure: Day 1- Cell Plating:
[00552] Adherent SKOV3 cells are washed with 10mLs of PBS followed by the
addition of 2mLs of 0.25% trypsin and incubation for 5 minutes at 37°K*I. The cells are rinsed
from the flask using 8 mL of media (phenol red-free RPMI+ 5%FBS) and transferred to fresh
flask. Cell concentration is determined using a Coulter counter and the appropriate volume of
cells to achieve 1000 cells/100µL is calculated. 100 µL of media cell suspension (adjusted to
1000 cells/100 µL) is added to all wells of 96-well plates, followed by incubation for 18 to 24
hours at 37°K*I, 100% humidity, and 5% CO2, allowing the cells to adhere to the plates.
Procedure: Day 2 - Compound Addition:
(O0553] To one column of the wells of an autoclaved assay block are added an initial
2.5 µL of test compound(s) at 400X the highest desired concentration. 1.25 µL of 400X
(400|uM) Topotecan is added to other wells (ODs from these wells are used to subtract out for
background absorbance of dead cells and vehicle). 500 µL of media without DMSO are
added to the wells containing test compound, and 250 µL to the Topotecan wells. 250 µL of
media + 0.5% DMSO is added to all remaining wells, into which the test compound(s) are
serially diluted. By row, compound-containing media is replica plated (in duplicate) from the
assay block to the corresponding cell plates. The cell plates are incubated for 72hours at
37°K*I, 100% humidity, and 5% CO2.
Procedure: Day 4 - MTS Addition and OD Reading:
[00554] The plates are removed from the incubator and 40 µl MTS / PMS is added to
each well. Plates are then incubated for 120 minutes at 37°K*I, 100% humidity, 5%CO2,
followed by reading the ODs at 490nm after a 5 second shaking cycle in a ninety-six well
spectrophotometer.
Data Analysis
[00555] The normalized % of control (absorbance- background) is calculated and an
XLfit is used to generate a dose-response curve from which the concentration of compound
required to inhibit viability by 50% is determined. The compounds of the present invention

show activity when tested by this method.
Example 100
Application of a Mitotic Kinesin Inhibitor
[00556] Human tumor cells Skov-3 (ovarian) were plated in 96-well plates at densities
of 4,000 cells per well, allowed to adhere for 24 hours, and treated with various
concentrations of the test compounds for 24 hours. Cells were fixed in 4% formaldehyde and
stained with antitubulin antibodies (subsequently recognized using fluorescently-labeled
secondary antibody) and Hoechst dye (which stains DNA).
(O0557) Visual inspection revealed that the compounds caused cell cycle arrest.
Example 101
Inhibition of Cellular Proliferation in Tumor Cell Lines Treated with Mitotic Kinesin
Inhibitors.
[00558] Cells were plated in 96-well plates at densities from 1000-2500 cells/well of a
96-well plate and allowed to adhere/grow for 24 hours. They were then treated with various
concentrations of ding for 4S hours. The time at which compounds are added is considered
To. A tetrazolium-based assay using the reagent 3-(4,5-dimethylthiazo1-2-yl)-5-(3-
carbox\'methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) (I.S> Patent No.
5,185,450) (see Promega product catalog #G358O, CellTiter 96® AQueous One Solution Cell
Proliferation Assay) was used to determine the number of viable cells at To and the number of
cells remaining after 48 hours compound exposure. The number of cells remaining after 48
hours was compared to the number of viable cells at the time of drug addition, allowing for
calculation of growth inhibition.
[00559] The growth over 48 hours of cells in control wells that had been treated with
vehicle only (0.25% DMSO) is considered 100% growth and the growth of cells in wells with
compounds is compared to this. Mitotic kinesin inhibitors inhibited cell proliferation in
human ovarian tumor cell lines (SKOV-3).
[00560] A Gi50 was calculated by plotting the concentration of compound in uM vs the
percentage of cell growth of cell growth in treated wells. The Gi50 calculated for the
compounds is the estimated concentration at which growth is inhibited by 50% compared to
control, i.e., the concentration at which:


[00561] All concentrations of compounds are tested in duplicate and controls are
averaged over 12 wells. A very similar 96-well plate layout and Gi50 calculation scheme is
used by the National Cancer Institute (see Monks, et al., J. Natl. Cancer Inst. 83:757-766
(1991)). However, the method by which the National Cancer Institute quantitates cell number
does not use MTS, but instead employs alternative methods.
Example 102
Calculation of IC50:
Measurement of a composition's IC50 uses an ATPase assay. The following solutions are
used: Solution 1 consists of 3 mM phosphoenolpyruvate potassium salt (Sigma P-7127), 2
mM ATP (Sigma A-3377), 1 mM IDTT (Sigma D-9779), 5 µM paclitaxel (Sigma T-7402),
10 ppm antifoam 289 (Sigma A-8436), 25 mM Pipes/K*I pH 6.8 (Sigma P6757), 2 mM
MgC12 (WVR JT400301), and 1 mM EGTA (Sigma E3889). Solution 2 consists of 1 mM
NADH (Sigma NS129), 0.2 mg/ml BSA (Sigma A7906), pyruvate kinase 7U/ml, L-lactate
dehydrogenase 10 U/ml (Sigma P0294), 100 nM motor domain of a mitotic kinesin. 50 ng/ml
microtubules; 1 mM DTT (Sigma D9779), 5 uM paclitaxel (Sigma T-7402), 10 ppm antifoam
289 (Sigma A-8436), 25 mM Pipes/K*I pH 6.S (Sigma P6757), 2 mM MgC12 (VWR
JT4003-01), and 1 mM EGTA (Sigma E3889). Serial dilutions (8-12 two-fold dilutions) of
the composition are made in a 96-well microliter plate (Corning Costar 3695) using Solution
1. Following serial dilution each well has 50 µl of Solution 1. The reaction is started by
adding 50 pi of solution 2 to each well. This may be done with a multichannel pipettor either
manually or with automated liquid handling devices. The microliter plate is then transferred
to a microplate absorbance reader and multiple absorbance readings at 340 nm are taken for
each well in a kinetic mode. The observed rate of change, which is proportional to the
ATPase rate, is then plotted as a function of the compound concentration. For a standard IC50
determination the data acquired is fit by the following four parameter equation using a
nonlinear fitting program (e.K*i., Grafit 4):

where y is the observed rate and x the compound concentration.
[00562] Other chemical entities of this class were found to inhibit cell proliferation,

although GI50 values varied. GI50 values for the chemical entities tested ranged from 200 nM
to greater than the highest concentration tested. By this we mean that although most of the
chemical entities that inhibited mitotic kinesin activity biochemically did inhibit cell
proliferation, for some, at the highest concentration tested (generally about 20 µM), cell
growth was inhibited less than 50%. Many of the chemical entities have GI50 values less than
10 µM, and several have GI50 values less than 1 µM. Anti-proliferative compounds that have
been successfully applied in the clinic to treatment of cancer (cancer chemotherapeutics) have
GI50's that vary greatly. For example, in A549 cells, paclitaxel GI50 is 4 nM, doxorubicin is
63 nM, 5-fiuorouracil is 1 µM, and hydroxyurea is 500 µM (data provided by National Cancer
Institute, Developmental Therapeutic Program, http://dtp.nci.nih.gov/). Therefore,
compounds that inhibit cellular proliferation at virtually any concentration may be useful.

WE CLAIM:
1. A compound selected from:
N-( 1-{4-[2-( 1-acetylamino-ethyl)-1-ethyl-1H-imidazo1-4-yl]-benzyl}-3-hydroxypropyl)-3-
chloro-4-(2,2,2-trifluoro-1-methyl-ethoxy)-benzamide,
N-(2-(2-dimethylamino-acetylamino)-1- {4-[8-( 1-hydroxy-ethyl)-imidazo[ 1,2-a]pyridin-2-yl]- benzyl}-ethyl)-3-chloro-4-isopropoxy-benzamide,
N-(1-{4-[2-( 1-methyl-1-hydroxy-ethyl)-1-ethyl-1H-imidazo1-4-yl]-benzyl}-3-hydroxy-propyl)-
3-chloro-4-(2,2,2-trifluoro-1-methyl-ethoxy)-benzamide,
N-(2-(2-dimethylamino-acetylamino)-1-{4-[8-methyl-imidazo[1,2-a]pyridin-2-yl]-benzyl}-
ethyl)-3-chloro-4-isopropoxy-benzamide,
N-(1-{4-[2-( 1-hydroxy-1-methyl-ethyl)-1-methyl-1H-imidazo1-4-yl]-benzyl}-3-hydroxy-
propyl)-3-chloro-4-(2,2,2-trifluoro-1-methyl-ethoxy)-benzamide,
N-(2-(2-amino-2-methyl-propionylamino)-1-{4-[8-methyl-imidazo[1,2-a]pyridin-2-yl]-benzyl}- ethyl)-3-chloro-4-isopropoxy-benzamide, and
N-{1-[4-(8-(1-hydroxy-ethyl)-imidazo[1,2-a]pyridin-2-yl)-benzyl]-3-hydroxy-propyl}-3-chloro-
4-isopropoxy-benzamide,
or a pharmaceutically acceptable salt thereof.
2. A compound selected from:





or a pharmaceutically acceptable salt thereof.
3. A pharmaceutical composition comprising at least one pharmaceutical
excipient and at least one compound as claimed in claim 1 or 2, or a pharmaceutically
acceptable salt thereof.
4. The pharmaceutical composition as claimed in claim 3, comprising a taxane,
a vinca alkaloid, or a topoisomerase I inhibitor.


IMIDAZOYL BENZAMIDE ANTI CANCER AGENTS
(57) Abstract: Compounds useful for treating cellular proliferative diseases and disorders by modulating the activity of one or more
mitotic kinesins are disclosed.

Documents:

03220-kolnp-2006 abstract.pdf

03220-kolnp-2006 claims.pdf

03220-kolnp-2006 correspondence others.pdf

03220-kolnp-2006 description(complete).pdf

03220-kolnp-2006 form-1.pdf

03220-kolnp-2006 form-3.pdf

03220-kolnp-2006 form-5.pdf

03220-kolnp-2006 gpa.pdf

03220-kolnp-2006 international publication.pdf

03220-kolnp-2006 international search authority report.pdf

03220-kolnp-2006 pct others.pdf

03220-kolnp-2006 priority document.pdf

03220-kolnp-2006-correspondence others-1.1.pdf

03220-kolnp-2006-correspondence-1.2.pdf

03220-kolnp-2006-form-3-1.1.pdf

03220-kolnp-2006-gpa-1.1.pdf

03220-kolnp-2006-priority document-1.1.pdf

3220-KOLNP-2006-ABSTRACT-1.1.pdf

3220-KOLNP-2006-AMANDED CLAIMS.pdf

3220-KOLNP-2006-ASSIGNMENT-1.1.pdf

3220-KOLNP-2006-ASSIGNMENT.pdf

3220-KOLNP-2006-CORRESPONDENCE-1.1.pdf

3220-KOLNP-2006-CORRESPONDENCE-1.2.pdf

3220-KOLNP-2006-EXAMINATION REPORT.pdf

3220-KOLNP-2006-FORM 1-1.1.pdf

3220-KOLNP-2006-FORM 13-1.1.pdf

3220-KOLNP-2006-FORM 13-1.2.pdf

3220-KOLNP-2006-FORM 13.pdf

3220-KOLNP-2006-FORM 18-1.1.pdf

3220-kolnp-2006-form 18.pdf

3220-KOLNP-2006-FORM 2.pdf

3220-KOLNP-2006-FORM 3-1.1.pdf

3220-KOLNP-2006-FORM 5-1.1.pdf

3220-KOLNP-2006-FORM 6-1.1.pdf

3220-KOLNP-2006-FORM 6.pdf

3220-KOLNP-2006-GPA.pdf

3220-KOLNP-2006-GRANTED-ABSTRACT.pdf

3220-KOLNP-2006-GRANTED-CLAIMS.pdf

3220-KOLNP-2006-GRANTED-DESCRIPTION (COMPLETE).pdf

3220-KOLNP-2006-GRANTED-FORM 1.pdf

3220-KOLNP-2006-GRANTED-FORM 2.pdf

3220-KOLNP-2006-GRANTED-SPECIFICATION.pdf

3220-KOLNP-2006-OTHERS-1.1.pdf

3220-KOLNP-2006-OTHERS.pdf

3220-KOLNP-2006-PETITION UNDER RULE 137-1.1.pdf

3220-KOLNP-2006-PETITION UNDER RULE 137.pdf

3220-KOLNP-2006-REPLY TO EXAMINATION REPORT.pdf


Patent Number 258632
Indian Patent Application Number 3220/KOLNP/2006
PG Journal Number 05/2014
Publication Date 31-Jan-2014
Grant Date 24-Jan-2014
Date of Filing 03-Nov-2006
Name of Patentee CYTOKINETICS, INC.
Applicant Address 280 EAST GRAND AVENUE SOUTH SAN FRANCISCO,CA 94080, UNITED STATES OF AMERICA
Inventors:
# Inventor's Name Inventor's Address
1 QIAN XIANGPING 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080 UNITED STATES OF AMERICA
2 ZHOU HAN-JIE 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
3 ASHCRAFT LUKE W 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
4 YAO BING 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
5 JIANG HONG 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
6 HUANG JENNIFER KUO CHEN 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
7 WANG JIANCHAO 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
8 MORGANS DAVID J,JR. 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
9 MORGANS BRADLEY P 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
10 BERGNES GUSTAVE 280 EAST GRAND AVENUE, SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
11 DHANAK DASHYANT 1250 S.COLLEGEVILLE ROAD, COLLEGEVILLE,PA19426, UNITED STATES OF AMERICA
12 KNIGHT STEVEN D 1250 S.COLLEGEVILLE ROAD, COLLEGEVILLE,PA19426, UNITED STATES OF AMERICA
13 ADAMS NICHOLAS D 1250 S.COLLEGEVILLE ROAD, COLLEGEVILLE,PA 19426, UNITED STATES OF AMERICA
14 PARRISH CYNTHIA A 1250 S.COLLEGEVILLE ROAD, COLLEGEVILLE,PA 19426, UNITED STATES OF AMERICA
15 DUFFY KEVIN ONE FRANKLIN PLAZA,PHILADELPHIA PA 19103,UNITED STATES OF AMERICA
16 FITCH DUKE ONE FRANKLIN PLAZA,PHILADELPHIA PA 19103,UNITED STATES OF AMERICA
17 TEDESCO ROSANNA ONE FRANKLIN PLAZA,PHILADELPHIA, PA 19103,UNITED STATES OF AMERICA
18 MCDONALD ANDREW I 280 EAST GRAND AVENUE SOUTH SAN FRANSISCO,CA 94080, UNITED STATES OF AMERICA
PCT International Classification Number C07C 233/65,A61K 31/
PCT International Application Number PCT/US2005/015666
PCT International Filing date 2005-05-06
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/569,510 2004-05-06 U.S.A.
2 11/121,709 2005-05-03 U.S.A.