Title of Invention

INHIBITORS OF CASPASES

Abstract The present invention relates to novel classes of compounds which are caspase inhibitors, in particular interleukin-1β converting enzyme ("ICE") inhibitors. This invention also relates to pharmaceutical compositions comprising these compounds. The compounds and pharmaceutical compositions of this invention are particularly well suited for inhibiting caspase activity and consequently, may be advantageously used as agents against interleukin-1- ("IL-1"), apoptosis-, interferon-γ inducing factor- (IGIF), or interferon-γ- ("IFN-γ") mediated diseases, including inflammatory diseases, autoimmune diseases, destructive bone disorders, proliferative disorders, infectious diseases, and degenerative diseases. This invention also relates to methods for inhibiting caspase activity and decreasing IGIF production and IFN-γ production and methods for treating interleukin-1, apoptosis-, and interferon-γ- mediated diseases using the compounds and compositions of this invention. This invention also relates to methods of preparing the compounds of this invention.
Full Text TECHNICAL FIELD OF THE INVENTION
The present invention relates to novel
classes of compounds which are caspase inhibitors, in
particular interleukin-1β converting enzyme ("ICE")
inhibitors. This invention also relates to
pharmaceutical compositions comprising these compounds.
The compounds and pharmaceutical compositions of this
invention are particularly well suited for inhibiting
caspase activity and consequently, may be
advantageously used as agents against interleukin-1-
("IL-1"), apoptosis-, interferon-γ inducing factor-
(IGIF), or interferon-γ- ("IFN-γ") mediated diseases,
including inflammatory diseases, autoimmune diseases,
destructive bone disorders, proliferative disorders,
infectious diseases, and degenerative diseases. This
invention also relates to methods for inhibiting
caspase activity and decreasing IGIF production and
IFN-γ production and methods for treating interleukin-
l, apoptosis-, and interferon-γ- mediated diseases using
the compounds and compositions of this invention. This
invention also relates to methods of preparing the
compounds of this invention.
BACKGROUND OF THE INVENTION
Interleukin l ("IL-l") is a major pro-
inflammatory and immunoregulatory protein that
stimulates fibroblast differentiation and
proliferation, the production of prostaglandins,

collagenase and phospholipase by synovial cells and
chondrocytes, basophil and eosinophil degranulation and
neutrophil activation. Oppenheim, J.H. et al,
Immunology Today, 7, pp. 45-56 (1986) . As such, it is
involved in the pathogenesis of chronic and acute
inflammatory and autoimmune diseases. For example, in
rheumatoid arthritis, IL-1 is both a mediator of
inflammatory symptoms and of the destruction of the
cartilage proteoglycan in afflicted joints. Wood, D.D.
et al. , Arthritis Rheum. 26, 975, (1983); Pettipher,
E.J. et al., Proc. Natl. Acad. Sci. USA 71, 295 (1986);
Arend, W.P. and Dayer, J.M., Arthritis Rheum. 38, 151
(1995) . IL-1 is also a highly potent bone resorption
agent. Jandiski, J.J., J. Oral Path 17, 145 (1988);
Dewhirst, F.E. et al., J. Immunol. 8, 2562 1985). It
is alternately referred to as "osteoclast activating
factor" in destructive bone diseases such as
osteoarthritis and multiple myeloma. Bataille, R. et
al., Int. J. Clin. Lab. Res. 21(4), 283 (1992). In
certain proliferative disorders, such as acute
myelogenous leukemia and multiple myeloma, IL-1 can
promote tumor cell'growth and adhesion. Bani, M.R., J.
Natl. Cancer Inst. 83, 123 (1991); Vidal-Vanaclocha,
F., Cancer Res. 54, 2667 (1994). In these disorders,
IL-1 also stimulates production of other cytokines such
as IL-6, which can modulate tumor development (Tartour
et al., Cancer Res. 54, p. 6243 (1994). IL-1 is
predominantly produced by peripheral blood monocytes as
part of the inflammatory response and exists in two
distinct agonist forms, IL-la and IL-lp. Mosely, B.S.
et al., Proc. Nat. Acad. Sci., 84, pp. 4572-4576
(1987); Lonnemann, G. et al. , Eur. J. Immunol. , 19, pp.
1531-1536 (1989) .
IL-ip is synthesized as a biologically
inactive precursor, pIL-1β. pIL-1β lacks a

conventional leader sequence and is not processed by a
signal peptidase. March, C.J., Nature, 315, pp.641-647
(1985) . Instead, pIL-1β is cleaved by interleukin-1β
converting enzyme ("ICE") between Asp-116 and Ala-117
to produce the biologically active C-terminal fragment
found in human serum and synovial fluid. Sleath, P.R.,
et al., J. Biol. Chem., 265, pp.14526-14528 (1992);
A.D. Howard et al. , J. Immunol. , 147, pp.2964-2969
(1991) . ICE is a cysteine protease localized primarily
in monocytes. It converts precursor IL-1β to the
mature form. Black, R.A. et al., FEBS Lett. , 247, pp. -
386-390 (1989); Kostura, M.J. et al. , Proc. Natl. Acad.
Sci. U.S.A., 86, pp.5227-5231 (1989). Processing by
ICE is also necessary for the transport of mature IL-1β
through the cell membrane.
ICE (or caspase-1) is a member of a family of
homologous enzymes called caspases. These homologs
have sequence similarities in the active site regions
of the enzymes. Such homologs (caspases) include TX
(or ICErel-II or ICH-2) (caspase-4) (Faucheu, et al. ,
EMBO J., 14, p. 1914 (1995); Kamens J., et al. , J.
Biol. Chem., 270, p. 15250 (1995); Nicholson et al., J.
Biol. Chem. , 270 15870 (1995)), TY (or ICErel-III)
(caspase-5) (Nicholson et al., J. Biol. Chem. , 270, p.
15870 (1995); ICH-1 (orNedd-2) (caspase-2) (Wang, L.
et al., Cell. 78, p. 739 (1994)), MCH-2 (caspase-6),
(Fernandes-Alnemri, T. et al. , Cancer Res. , 55, p. 2737
(1995), CPP32 (or YAMA or apopain) (caspase-3)
(Fernandes-Alnemri, T. et al. , J. Biol. Chem. , 269, p.
30761 (1994); Nicholson, D.W. et al. , Nature, 376, p.
37 (1995)), CMH-1 (or MCH-3) (caspase-7) (Lippke, et
al., J. Biol. Chem., 271(4), pl825-1828 (1996));
Fernandes-Alnemri, T. et al., Cancer Res. , (1995)),
MchS (caspase-8) (Muzio, M. et.al. , Cell 85(6), 817-
827, (1996)), MCH-6 (caspase-9) (Duan, H. et.al., J.

Biol. Chem., 271(34), p. 16720-16724 (1996)), Mch4
(caspase-10) (Vincenz, C. et.al., J. Biol. Chem., 272,
p. 6578-6583 (1997); Fernandes-Alnemri, T. et.al.,
Proc. Natl. Acad. Sci. 93, p. 7464-7469 (1996)), Ich-3
(caspase-11) (Wang, S. et.al., J. Biol. Chem. , 271, p.
20580-20587 (1996)), mCASP-12 (caspase-12), (Van de
Craen, M. et.al., FEBS Lett. 403, p. 61-69 (1997);
Yuan, Y.and Miura, M. PCT Publication WO95/00160
(1995)), ERICE (caspase-13) , (Humke, E.W., et.al., J.
Biol. Chem., 273(25) p. 15702-15707 (1998)), and MICE
(caspase-14) (Hu, S. et.al., J. Biol. Chem., 273(45) p.
29648-29653 (1998)).
Each of these ICE homologs, as well as ICE
itself, is capable of inducing apoptosis when
overexpressed in transfected cell lines. Inhibition of
one or more of these homologs with the peptidyl ICE
inhibitor Tyr-Val-Ala-Asp-chloromethylketone results in
inhibition of apoptosis in primary cells or cell lines.
Lazebnik et al., Nature, 371, p. 346. (1994).
Caspases also appear to be involved in the
regulation of programmed cell death or apoptosis.
Yuan, J. et al., Cell, 75, pp.641-652 (1993); Miura, M.
et al., Cell, 75, pp. 653-660 (1993); Nett-Fiordalisi,
M.A. et al., J. Cell Biochem., 17B, p. 117 (1993). In
particular, ICE or ICE homologs are thought to be
associated with the regulation of apoptosis in
neurodegenerative diseases, such as Alzheimer's and
Parkinson's disease. Marx, J. and M. Baringa, Science,
259, pp. 760-762 (1993); Gagliardini, V. et al. ,
Science, 263, pp. 826-828 (1994). Therapeutic
applications for inhibition of apoptosis may include
treatment of Alzheimer's disease, Parkinson's disease,
stroke, myocardial infarction, spinal atrophy, and
aging.

ICE has been demonstrated to mediate
apoptosis (programmed cell death) in certain tissue
types. Steller, H., Science, 267, p. 1445 (1995);
Whyte, M. and Evan, G., Nature, 376, p. 17 (1995);
Martin, S.J. and Green, D.R., Cell, 82, p. 349 (1995);
Alnemri, E.S., et al., J. Biol. Chetn. , 270, p. 4312
(1995); Yuan, J. Curr. Opin. Cell Biol. , 7, p. 211
(1995) . A transgenic mouse with a disruption of the
ICE gene is deficient in Fas-mediated apoptosis (Kuida,
K. et al. , Science 267, 2000 (1995)). This activity of
ICE is distinct from its role as the processing enzyme
for pro-IL-1β. It is conceivable that in certain
tissue types, inhibition of ICE may not affect
secretion of mature IL-1β, but may inhibit apoptosis.
Enzymatically active ICE has been previously
described as a heterodimer composed of two subunits,
p20 and p10 (20kDa and 10kDa molecular weight,
respectively) . These subunits are derived from a 45kDa
proenzyme (p4 5) by way of a p3 0 form, through an
activation mechanism that is autocatalytic.
Thornberry, N.A. et al. , Nature, 356, pp.768-774
(1992) . The ICE proenzyme has been divided into
several functional domains: a prodomain (pl4), a
p22/2 0 subunit, a polypeptide linker and a p10 subunit.
Thornberry et al., supra; Casano et al. , Genomics, 20,
pp. 474-481 (1994).
Full length p4 5 has been characterized by its
cDNA and amino acid sequences. PCT patent applications
WO 91/15577 and WO 94/00154. The p20 and plO cDNA and
amino acid sequences are also known. Thornberry et
al. , supra. Murine and rat ICE have also been sequenced
and cloned. They have high amino acid and nucleic acid
sequence homology to human ICE. Miller, D.K. et al. ,
Ann. N.Y. Acad. Sci., 696, pp. 133-148 (1993);
Molineaux, S.M. et al . , Proc. Nat. Acad. Sci., 90, pp.

1809-1813 (1993). The three-dimensional structure of
ICE has been determined at atomic resolution by X-ray
crystallography. Wilson, K.P., et al. , Nature, 370,
pp. 270-275 (1994). The active enzyme exists as a
tetramer of two p20 and two p10 subunits.
Recently, ICE and other members of the
ICE/CED-3 family have been linked to the conversion of
pro-IGIF to IGIF or to the production of IFN-γ in vivo
(PCT application PCT/US96/20843, publication no. WO
97/22619, which is incorporated herein by reference).
IGIF is synthesized in vivo as the precursor protein
"pro-IGIF".
Interferon-gamma inducing factor (IGIF) is an
approximately 18-kDa polypeptide that stimulates T-cell
production of interferon-gamma (IFN-γ). IGIF is
produced by activated Kupffer cells and macrophages in
vivo and is exported out of such cells upon endotoxin
stimulation. Thus, a compound that decreases IGIF
production would be useful as an inhibitor of such T-
cell stimulation which in turn would reduce the levels
of IFN-γ production by those cells.
IFN-γ is a cytokine with immunomodulatory
effects on a variety of immune cells. In particular,
IFN-γ is involved in macrophage activation and Th1 cell
selection (F. Belardelli, APMIS, 103, p. 161 (1995)).
IFN-γ exerts its effects in part by modulating the
expression of genes through the STAT and IRF pathways
(C. Schindler and J.E. Darnell, Ann. Rev. Biochem. , 64,
p. 621 (1995); T. Taniguchi, J. Cancer Res. Clin.
Oncol., 121, p. 516 (1995)).
Mice lacking IFN-γ or its receptor have
multiple defects in immune cell function and are
resistant tc endotoxic shock (S. Huang et al. , Science,
259, p.1742 (1993); D. Dalton et al., Science, 259, p.-
1739 (1993); B.D. Car et al., J. Exp. Med.. 179, p.1437

(1994)). Along with IL-12, IGIF appears to be a potent
inducer of IFN-γ production by T cells (H. Okamura et
al., Infection and Immunity, 63, p.3966 (1995); H.
Okamura et al., Nature, 378, p.88 (1995); S. Ushio et
al., J.Immunol., 156, p.4274 (1996)).
IFN-γ has been shown to contribute to the
pathology associated with a variety of inflammatory,
infectious and autoimmune disorders and diseases.
Thus, compounds capable of decreasing IFN-γ production
would be useful to ameliorate the effects of IFN-γ
related pathologies.
Accordingly, compositions and methods capable
of regulating the conversion of pro-IGIF to IGIF would
be useful for decreasing IGIF and IFN-γ production in
vivo, and thus for ameliorating the detrimental effects
of these proteins which contribute to human disorders
and diseases.
Caspase inhibitors represent a class of
compounds useful for the control of inflammation or
apoptosis or both. Peptide and peptidyl inhibitors of
ICE have been described (PCT patent applications
WO 91/15577, WO 93/05071, WO 93/09135, WO 93/12076,
WO 93/14777, WO 93/16710, WO 95/35308, WO 96/30395,
WO 96/33209 and WO 98/01133; European patent
applications 503 561, 547 699, 618 223, 623 592, and
623 606; and US patent nos. 5,434,248, 5,710,153,
5,716,929, and 5,744,451). Such peptidyl inhibitors of
ICE have been observed to block the production of
mature IL-1β in a mouse model of inflammation (vide
infra) and to suppress growth of leukemia cells in
vitro (Estrov et al. , Blood, 84, 380a (1994)).
However, due to their peptidic nature, such inhibitors
are typically characterized by undesirable
pharmacologic properties, such as poor cellular
penetration and cellular activity, poor oral

absorption, instability and rapid metabolism.
Plattner, J.J. and D.W. Norbeck, in Drug Discovery
Technologies, C.R. Clark and W.H. Moos, Eds. (Ellis
Horwood, Chichester, England, 1990), pp.92-126. These
properties has hampered their development into
effective drugs.
Non-peptidyl compounds have also been
reported to inhibit ICE in vitro. PCT patent
application WO 95/26958; US Patent 5,552,400; Dolle et
al., J. Med. Chem., 39, pp. 2438-2440 (1996).
It is not clear however whether these
compounds have the appropriate pharmacological profiles
to be therapeutically useful.
Accordingly, the need exists for compounds
that can effectively inhibit caspases, and that have
favorable in vivo activity, for use as agents for
preventing and treating chronic and acute forms of IL-
1-, apoptosis-, IGIF-, or IFN-γ-mediated diseases, as
well as inflammatory, autoimmune, destructive bone,
proliferative, infectious, or degenerative diseases.
SUMMARY OF THE INVENTION
The present invention provides novel classes
of compounds, and pharmaceutically acceptable
derivatives thereof, that are useful as caspase
inhibitors, in particular, as ICE inhibitors. These
compounds can be used alone or in combination with
other therapeutic or prophylactic agents, such as
antibiotics, immunomodulators or other anti-
inflammatory agents, for the treatment or prophylaxis
of diseases mediated by IL-1, apoptosis, IGIF, or IFN-
γ. According to a preferred embodiment, the compounds
of this invention are capable of binding to the active
site of a caspase and inhibiting the activity of that
enzyme.

It is a principal object of this invention to
provide novel classes of compounds represented by
formula I, which have favorable in vivo profiles:

wherein the various substituents are described
herein.
It is a further objective of this invention
to provide pharmaceutical compositions, including
multi-component compositions. This invention also
provides methods for using and preparing the compounds
of this invention and related compounds.
DETAILED DESCRIPTION OF THE INVENTION
In order that the invention described herein
may be more fully understood, the following detailed
description is set forth.
The following abbreviations and definitions
are used throughout the application.



The term "caspase" refers to an enzyme that
is a member of the family of enzymes that includes ICE
(see H. Hara, Natl. Acad. Sci., 94, pp. 2007-2012
(1997) ) .
The terms "HBV", "HCV" and "HGV" refer to
hepatitis-B virus, hepatitis-C virus and hepatitis-G
virus, respectively.
The term "Ki" refers to a numerical measure
of the effectiveness of a compound in inhibiting the
activity of a target enzyme such as ICE. Lower values
of Ki. reflect higher effectiveness. The Ki value is a
derived by fitting experimentally determined rate data
to standard enzyme kinetic equations (see I.H. Segel,
Enzyme Kinetics, Wiley-Interscience, 1975).
The term "interferon gamma inducing factor"
or "IGIF" refers to a factor which is capable of
stimulating the endogenous production of IFN-γ.
The term "caspase inhibitor" refer to a
compound which is capable of demonstrating detectable
inhibition of one or more caspases. The term "ICE
inhibitor" refers to a compound which is capable of
demonstrating detectable inhibition of ICE and
optionally one or more additional caspases. Inhibition
of these enzymes may be determined using the methods
described and incorporated by reference herein.
The skilled practitioner realizes that an in
vivo enzyme inhibitor is not necessarily an in vitro

enzyme inhibitor. For example, a prodrug form of a
compound typically demonstrates little or no activity
in in vitro assays. Such prodrug forms may be altered
by metabolic or other biochemical processes in the
patient to provide an in vivo ICE inhibitor.
The term "cytokine" refers to a molecule
which mediates interactions between cells.
The term "condition" refers to any disease,
disorder or effect that produces deleterious biological
consequences in a subject.
The term "subject" refers to an animal, or to
one or more cells derived from an animal. Preferably,
the animal is a mammal, most preferably a human. Cells
may be in any form, including but not limited to cells
retained in tissue, cell clusters, immortalized cells,
transfected or transformed cells, and cells derived
from an animal that have been physically or
phenotypically altered.
The term "patient" as used in this
application refers to any mammal, preferably humans.
The term "alkyl" refers to a straight-chained
or branched, saturated aliphatic hydrocarbon containing
1 to 6 carbon atoms.
The term "alkenyl" refers to a straight-
chained or branched unsaturated hydrocarbon containing
2 to 6 carbon atoms and at least one double bond.
The term "alkynyl" refers to a straight -
chained or branched unsaturated hydrocarbon containing
2 to 6 carbon atoms and at least one triple bond.
The term "cycloalkyl" refers to a mono- or
polycyclic, non-aromatic, hydrocarbon ring system which
may optionally contain unsaturated bonds in the ring
system. Examples include cyclohexyl, adamantyl.
norbornyl, and spirocyclopentyl.


The term "aryl" refers to a mono- or
polycyclic ring system which contains 6, 10, 12 or 14
carbons in which at least one ring of the ring system
is aromatic. The aryl groups of this invention are
optionally singly or multiply substituted with R11.
Examples of aryl ring systems include, phenyl,
naphthyl, and tetrahydronaphthyl.
The term "heteroaryl" refers to a mono- or
polycyclic ring system which contains l to 15 carbon
atoms and 1 to 4 heteroatoms, and in which at least one
ring of the ring system is aromatic. Heteroatoms are
sulfur, nitrogen or oxygen. The heteroaryl groups of
this invention are optionally singly or multiply
substituted with R11.
The term "heterocyclic" refers to a mono- or
polycyclic ring system which contains 1 to 15 carbon
atoms and 1 to 4 heteroatoms, in which the mono- or
polycyclic ring system may optionally contain
unsaturated bonds but is not aromatic. Heteroatoms are
independently sulfur, nitrogen, or oxygen.
The term "alkylaryl" refers to an alkyl
group, wherein a hydrogen atom of the alkyl group is
replaced by an aryl radical.
The term "alkylheteroaryl" refers to an alkyl
group, wherein a hydrogen atom of the alkyl group is
replaced by a heteroaryl radical.
The term "amino acid side chain" refers to
any group attached to the α carbon of a naturally or

non-naturally accorring amino acid.
The term "substitute" refers to the
replacement of a hydrogen atom in a compound with a
substituent group.
The term "straight chain" refers to a
contiguous unbranching string of covalently bound
atoms. The straight chain may be substituted, but

these substituents are not a part of the straight
chain.
In chemical formulas, parenthesis are used
herein to denote connectivity in molecules or groups.
In particular, parentheses are used to indicate: 1)
that more than one atom or group is bonded to a
particular atom; or 2) a branching point (i.e., the
atom immediately before the open parenthesis is bonded
both to the atom or group in the parentheses and the
atom or group immediately after the closed
parenthesis) . An example of the first use is
"-N(alkyl)2", indicating two alkyl groups bond to an N
atom. An example of the second use is "-C(O)NH2",
indicating a carbonyl group and an amino ("NH2") group
both bonded to the indicated carbon atom. A "-C(O)NH2"
group may be represented in other ways, including the
following structure:

Substituents may be represented in various
forms. These various forms are known to the skilled
practitioner and may be used interchangeably. For
example, a methyl substituent on a phenyl ring may be
represented in any of the following forms:

Various forms of substituents such as methyl are used
herein interchangeably.
Other definitions are set forth in the
specification where necessary.

Compounds of this Invention
The compounds of one embodiment A of this
invention are those of formula I:

wherein:
Y is:

provided that when R7 is -OH then Y can also be:

X is -C(R3)2- or -N(R3)-;
m is 0 or 1;
R1 is H, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S (O) 2N (H) -R8 ,
-S(O)N(H)-R8, -C(O)C(O)N{H)R8, -C(O)CH=CHR8,
-C(O)CH2OR8, -C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O) 2N (R8) 2 ,

-S(O)N(R8)2, -C(O)C(O)N(R8)2, -C(O)CH2N (R8)2, -CH2R8,
-CH2-alkenyl-R8, or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9,
or R2 and one R3 together with the atoms to which they
are bound, form a 3 to 7 membered cyclic or
heterocyclic ring system, wherein a hydrogen atom bound
to any -alkyl or -cycloalkyl carbon atom is optionally
replaced by -R10, a hydrogen atom bound to any -aryl or
-heteroaryl carbon atom is optionally replaced by -R11,
a hydrogen atom bound to any nitrogen atom of the ring
system is optionally replaced by -R1;
R4 is -H and each R5 is independently -H, an
amino acid side chain, -R8, -alkenyl-R9, or -alkynyl-
R9, or R4 and one R5 together with the atoms to which
they are bound form a 3 to 7 membered cyclic or
heterocyclic ring system, wherein a hydrogen atom bound
to any -alkyl or -cycloalkyl carbon atom is optionally
replaced by R10, a hydrogen atom bound to any -aryl or
-heteroaryl carbon atom is optionally replaced by R11,
and a hydrogen atom bound to any nitrogen atom of the
ring system is optionally replaced with R1;
R6 is -H;
R7 is -OH, -OR8, or -N(H)0H;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,

wherein a hydrogen atom bound to any -alkyl or
-cycloalkyl carbon atom is optionally replaced by R10,
a hydrogen atom bound to any -aryl or -heteroaryl
carbon atom is optionally replaced by R11, and a
hydrogen atom bound to any nitrogen atom is optionally
replaced by R1;
each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by R11, and a hydrogen atom bound to any
nitrogen atom is optionally replaced by R1;
each R10 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N(H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl, -N(H)C(O)N(alkyl)2,
-S-alkyl, -S-aryl, -S-alkylaryl, -S(O)2alkyl,
-S(O)alkyl, -C(O)alkyl, -CH2NH2, -CH2N(H)alkyl, or
-CH2N(alkyl)2, -alkyl, -cycloalkyl, -aryl, -heteroaryl,
-heterocyclyl, -alkylcycloalkyl -alkylaryl,
-alkylheteroaryl, or -alkylheterocyclyl, wherein a
hydrogen atom bound to any -aryl or -heteroaryl carbon
atom is optionally replaced by R11 and a hydrogen atom
bound to any nitrogen atom is optionally replaced by
R1; and
each R11 is independently -OH, -SH, -F, -CI,
-Br, -I, -NO2, -CN, -NH2, -C02H, -C(O)NH2, -N(H)C(O)H,
-N(K)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -o-
alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,

-N(H)-alkylaryl, -N(alkyl)2, -C(O)N(H)alkyl,
-C(O)N(alkyl)2, -N(H)C(O) alkyl, -N(H)C(O)N(H) alkyl,
-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2.
In an alternative form of embodiment A:
R1 is H, -R8, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S (O) 2N (H)-R8 ,
-S(O)N(H) -R8, -C(O)C(O)N(H)R8, -C (O) CH=CHR8 ,
-C(O)CH2OR8, -C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O) 2N (R8 ) 2 ,
-S(O)N(R8)2, -C(O)C(O)N(R8)2, -C(O)CH2N(R8)2, -CH2R8,
-CH2-alkenyl-R8, or -CH2-alkynyl-R8,-
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9,
or each R3, together with the atom to which they are
bound, form a 3 to 7 membered cyclic or heterocyclic
cyclic ring system, or R2 and one R3 together with the
atoms to which they are bound, form a 3 to 7 membered
cyclic or heterocyclic ring system, wherein a hydrogen
atom bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by -R10, a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by -R11, a hydrogen atom bound to any nitrogen
atom of the ring system is optionally replaced by -R1;
each R10 is independently -OH, -SH, -F, -CI,
-Br, -I, -NO2, -CN, -NH2, -C02H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -0-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N(H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O) alkyl, -N(H)C(O)Oalkyl, -N(H) C(O)0aryl,

-N(H)C(O)Oalkylaryl, -N(H)C(O)Oheteroaryl,
-N(H)C(O)Oalkylheteroaryl, -N(H)C(O)Ocycloalkyl,
-N(H)C(O)N(H)alkyl, -N (H) C (O)N (alkyl) 2 ,
-N(H)C(O)N(H)aryl, -N(H)C(O)N(H)alkylaryl,
-N(H)C(O)N(H)heteroaryl, -N(H)C(O) N (H) alkylheteroaryl,
-N(H)C(O)N(H)cycloalkyl, -S-alkyl, -S-aryl,
-S-alkylaryl, -S(O)2alkyl, -S(O)alkyl, -C(O)alkyl,
-CH2NH2, -CH2N(H)alkyl, or -CH2N(alkyl)2, -alkyl,
-cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by R11 and a hydrogen atom bound to any
nitrogen atom is optionally replaced by R1; and
the other substituents are as defined above.
Preferably, in any of the above embodiments:
m is 0;
R2 is -H; one R3 is -H and the other R3 is -R8,
-alkenyl-R9, or -alkynyl-R9; or
R4 and one R5 together with the atoms to
which they are bound form a 3 to 7 merabered cyclic or
heterocyclic ring system, wherein a hydrogen atom bound
to any -alkyl or -cycloalkyl carbon atom is optionally
replaced by R10, a hydrogen atom bound to any -aryl or
-heteroaryl carbon atom is optionally replaced by R11,
and a hydrogen atom bound to any nitrogen atom of the
ring system is optionally replaced with R1, wherein the
ring system is:


In an alternative preferred embodiment, X is
C(R3)2 or one R3 is an amino acid side chain, -R8,
alkenyl-R9, or alkynyl-R9.
More preferably, one R3 is -H and the other
R3 is -alkyl; or
R4 and one R5 together with the atoms to
which they are bound form a 3 to 7 membered cyclic or
heterocyclic ring system, wherein any hydrogen atom
bound to a carbon atom of the ring system is optionally
replaced by R10 and any hydrogen atom bound to a
nitrogen atom of the ring system is optionally replaced
by R1, selected from:


Most preferably, one R3 is -H and the other
R3 is -C(H)(CH3)2 or -C(CH3)3; and
R4 and one R5 together with the atoms to
which they are bound form a 3 to 7 membered cyclic or
heterocyclic ring system, wherein any hydrogen atom
bound to a carbon atom of the ring system is optionally
replaced by R10 and any hydrogen atom bound to a
nitrogen atom of the ring system is optionally replaced
by R1, selected from:


In an alternative most preferred embodiment,
one R3 is -H and the other R3 is -CH3, -C(H) (CH3)2 or
-C(CH3)3 and R4 and R5 are as defined directly above.
According to another embodiment B, the
present invention provides a compound of formula I,
wherein Y is:

provided that when R6 is not hydrogen, R6 and Y,
together with the nitrogen to which they are bound,
form a ring (g) :


R12 is -C(O)alkyl, -C(O)cycloalkyl,
-C(O)alkyenyl, -C(O)alkylaryl, -C(O)alkylheteroaryl,
-C (O)heterocyclyl, or -C(O)alkylheterocyclyl;
R13 is -H, -alkyl, -aryl, -alkylaryl or
-alkylheteroaryl; and
the other substituents are as described
above.
Preferably, in (c) , (d) , (e)., or (f), R8 is
methyl, ethyl, n-propyl, isopropyl, cyclopentyl,
phenethyl, or benzyl.
Preferred definitions for the other
individual components of embodiment B are the same as
those set forth above for embodiment A.
A preferred embodiment C of this invention
provides compounds of formula I:

wherein:
Y is:


m is 0 or 1;
X is -C(R3)2-
R1 is H, -R8, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S(O)2N(H)-R8,
-S(O)N(H)-R8, -C(O)C(O)N(H)R8, -C (O) CH=CHR8 ,
-C(O)CH20R8, -C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O) 2N (R8) 2 ,
-S(O)N(R8)2, -C(O)C(O)N(R8)2, -C (O) CH2N (R8) 2 , -CH2R8,
-CH2-alkenyl-R8, or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9,
or each R3 together with the atom to which they are
bound, form a 3 to 7 membered cyclic or heterocyclic
ring system, wherein a hydrogen atom bound to any
-alkyl or -cycloalkyl carbon atom is optionally
replaced by -R10, a hydrogen atom bound to any -aryl or
-heteroaryl carbon atom is optionally replaced by -R11,
a hydrogen atom bound to any nitrogen atom of the ring
system is optionally replaced by -R1;

R4 is -H and each R5 is independently -H, an
amino acid side chain, -R8, -alkenyl-R9, or
-alkynyl-R9, or R4 and one R5 together with the atoms
to which they are bound form a 3 to 7 membered cyclic
or heterocyclic ring system, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by R11, and a hydrogen atom bound to any
nitrogen atom of the ring system is optionally replaced
with R1;
R6 is -H;
R7 is -OH, -OR8, -N(H)0H, or -N(H)S (O)2R8;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,
wherein a hydrogen atom bound to any -alkyl or
-cycloalkyl carbon atom is optionally replaced by R10,
a hydrogen atom bound to any -aryl or -heteroaryl
carbon atom is optionally replaced by R11, and a
hydrogen atom bound to any nitrogen atom is optionally
replaced by R1;
each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by R11, and a hydrogen atom bound to any
nitrogen atom is optionally replaced by R1;

each R10 is independently -OH, -SH, -F, -CI,
-Br, -I, -NO2, -CN, -NH2, -C02H, -C(O)NH2/ -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N{H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N(H)alkyl, -C (O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C(O)Oalkyl, -N(H) C (O)Oaryl,
-N (H) C (O) Oalkylaryl, -N (H) C (O) Oheteroaryl,
-N (H) C (O) Oalkylheteroaryl, -N (H) C (O) Ocycloalkyl,
-N(H)C(O)N(H) alkyl, -N(H)C(O)N(alkyl) 2/
-N (H) C (O) N (H) aryl, -N (H) C (O) N(H) alkylaryl,
-N (H) C (O) N (H) heteroaryl, -N (H) C (O)N (H) alkylheteroaryl,
-N(H)C(O)N(H)cycloalkyl, -S-alkyl, -S-aryl,
-S-alkylaryl, -S(O)2alkyl, -S(O)alkyl, -C(O)alkyl,
-CH2NH2, -CH2N(H) alkyl, or -CH2N(alkyl) 2, -alkyl,
-cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to
any -aryl or -heteroaryl carbon atom is optionally
replaced by R11 and a hydrogen atom bound to any
nitrogen atom is optionally replaced by R1; and
each R11 is independently -OH, -SH, -F, -CI,
-Br, -I, -NO2/ -CN, -NH2, -C02H, -C(O)NH2/ -N(H)C(O)H,
-N(H)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -0-
alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,
-N(H)-alkylaryl, -N(alkyl)2, -C(O)N(H)alkyl,
-C(O)N(alkyl)2, -N (H) C (O) alkyl, -N(H) C (O)N(H) alkyl,
-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2;
provided that if one R3 is -H, then the other
R3 is not -H.

Another preferred embodiment D of the present
invention provides a compound of formula I, wherein Y
is:

R12 is -C(O)alkyl, -C(O)cycloalkyl,
-C(O)alkyenyl, -C(O)alkylaryl, -C(O)alkylheteroaryl,
-C(O)heterocyclyl, or -C(O)alkylheterocyclyl; and
the other substituents are as described above
except that both of the R3 groups may be -H.
In any of embodiments A-D, preferred
compounds are those wherein:
R1 is -C(O)R8 or -C(O)C(O)R8;
R2 and one R3 are both -H and the other R3 is
an amino acid side chain, -R8, alkenyl-R9, or
alkynyl-R9; or
R4 and one R5 together with the atoms to
which they are bound form a ring system selected from:



provided that each of the ring systems are optionally-
substituted with one or more R10 groups.

Alternatively, preferred compounds of
embodiments A-D are those wherein R3 is -H and the
other R3 is methyl, isopropyl, tert-butyl, -CH2SR8,
-CH2S02R8, -CH2CH2SR8, -CH2CH2SO2R8.
More preferred compounds of embodiments A-D
are those wherein R4 and one R5 together with the atoms
to which they are bound form the ring system:

and the other R5 is H; or
one R3 is -H and the other R3 is methyl.
Alternatively, more preferred compounds of
embodiments A-D are those wherein R4 and one R5
together with the atoms to which they are bound form
the ring system:

and the other R5 is H.
In the above alternative embodiment, R10 is
preferably, 4-fluoro or 4,4-difluoro.
Most preferred compounds of this invention
are those wherein R3 is methyl; and
R4 and one R5 together with the atoms to
which they are bound form the ring system:


and the other R5 is H.
Alternatively, most preferred compounds of
embodiments A-D are those wherein R3 is methyl; and
R4 and one R5 together with the atoms to which they are
bound form the ring system:





Specific compounds of this invention include,
but are not limited to, Examples 5a-5bd, 7a-7at, 9a-9g,
15a-15f, 16a-16b, 17a-17e, 18a-18f, 20a-20t, 23a-23i,
24a-24e, 25a-25e, 26a-26h, 27a-27n, 28a-28c, 29a-29s,
32a-32e, 34, G1, G2, 41, 42, 45, 46, 51, 52, 56, 57,
60, 61, 64, 65, 68, 69, 72, 73, 76-93, 98a-z, aa-az,
and ba-bb, 101, 102a, 102b, 108a-d, 110, 111, 116a-h,
120a and b, 121, 122 a-v, and 123 a-c.
The compounds of this invention may contain
one or more "asymmetric" carbon atoms and thus may
occur as racemates and racemic mixtures, single
enantiomers, diastereomeric mixtures and individual
diastereomers. Each stereogenic carbon may be of the R
or S configuration. Although specific compounds and
scaffolds exemplified in this application may be
depicted in a particular stereochemical configuration,
compounds and scaffolds having either the opposite
stereochemistry at any given chiral center or mixtures
thereof are also envisioned.
All such isomeric forms of these compounds
are expressly included in the present invention, as
well as pharmaceutically acceptable derivative thereof.

The term "pharmaceutically acceptable
derivative" means any pharmaceutically acceptable salt,
ester, or salt of such ester, of a compound of this
invention or any other compound which, upon
administration to a recipient, is capable of providing
(directly or indirectly) a compound of this invention
or an active metabolite or residue thereof.
Pharmaceutically acceptable salts of the
compounds of this invention include, for example, those
derived from pharmaceutically acceptable inorganic and
organic acids and bases. Examples of suitable acids
include hydrochloric, hydrobromic, sulfuric, nitric,
perchloric, fumaric, maleic, phosphoric, glycolic,
lactic, salicylic, succinic, toluene-p-sulfonic,
tartaric, acetic, citric, methanesulfonic, formic,
benzoic, malonic, naphthalene-2-sulfonic and
benzenesulfonic acids. Other acids, such as oxalic,
while not in themselves pharmaceutically acceptable,
may be employed in the preparation of salts useful as
intermediates in obtaining the compounds of the
invention and their pharmaceutically acceptable acid
addition salts. Salts derived from appropriate bases
include alkali metal (e.g., sodium), alkaline earth
metal (e.g., magnesium), ammonium and N-(C1-4 alkyl)4+
salts.
This invention also envisions the
"quaternization" of any basic nitrogen-containing
groups of the compounds disclosed herein. The basic
nitrogen can be quaternized with any agents known to
those of ordinary skill in the art including, for
example, lower alkyl halides, such as methyl, ethyl,
propyl and butyl chloride, bromides and iodides;
dialkyl sulfates including dimethyl, diethyl, dibutyl
and diamyl sulfates; long chain halides such as decyl,
lauryl, myristyl and stearyl chlorides, bromides and

iodides; and aralkyl halides including benzyl and
phenethyl bromides. Water or oil-soluble or
dispersible products may be obtained by such
guaternization.
When multiply substituted, each substituent
may be picked independently of any other substituent as
long as the combination of substituents results in the
formation of a stable compound.
Combinations of substituents and variables
envisioned by this invention are only those that result
in the formation of stable compounds. The term
"stable", as used herein, refers to compounds which
possess stability sufficient to allow manufacture and
administration to a mammal by methods known in the art.
Typically, such compounds are stable at a temperature
of 4 0 °C or less, in the absence of moisture or other
chemically reactive conditions, for at least a week.
Preferred compounds of this invention may be
readily absorbed by the bloodstream of patients upon
oral administration. This oral availability makes such
compounds excellent agents for orally-administered
treatment and prevention regimens against IL-1-,
apoptosis-, IGIF-, or IFN-γ-mediated diseases.
It should be understood that the compounds of
this invention may exist in various equilibrium forms,
depending on conditions including choice of solvent,
pH, and others known to the practitioner skilled in the
art. All such forms of these compounds are expressly
included in the present invention. In particular, many
of the compounds of this invention, especially those
which contain aldehyde or ketone groups and carboxylic
acid groups in Y, may take hemi-acetal or hydrated
forms. For example, compounds of embodiment A are in a
hemiacetal form when Y is:


Depending on the choice of solvent and other
conditions known to the practitioner skilled in the
art, compounds of this invention may also take
hydrated, acyloxy acetal, acetal, or enol forms. For
example, compounds of this invention are in hydrated
forms when Y is:

acyloxy acetal forms when Y is:

acetal forms when Y is and R8 is other than H:

and enol forms when Y is:


In addition, it should be understood that the
equilibrium forms of the compounds of this invention
may include tautomeric forms. All such forms of these
compounds are expressly included in the present
invention.
The compounds of formula I may be synthesized
using conventional techniques. Advantageously, these
compounds are conveniently synthesized from readily
available starting materials.
Compounds of this invention may be prepared
using the processes described herein. As can be
appreciated by the skilled practitioner, these
processes are not the only means by which the compounds
described and claimed in this application may be
synthesized. Further methods will be evident to those
of ordinary skill in the art. Additionally, the
various synthetic steps described herein may be
performed in an alternate sequence or order to give the
desired compounds.
It should be understood that the compounds of
this invention may be modified by appropriate
functionalities to enhance selective biological
properties. Such modifications are known in the art
and include those which increase biological penetration
into a given biological system (e.g., blood, lymphatic
system, central nervous system), increase oral
availability, increase solubility to allow

administration by injection, alter metabolism and alter
rate of excretion. In addition, the compounds may be
altered to pro-drug form such that the desired compound
is created in the body of the patient as the result of
the action of metabolic or other biochemical processes
on the pro-drug. Such pro-drug forms typically
demonstrate little or no activity in in vitro assays.
Some examples of pro-drug forms include ketal, acetal,
oxime, imine and hydrazone forms of compounds which
contain ketone or aldehyde groups, especially where
they occur in the Y group of the compounds of this
invention. Other examples of pro-drug forms include
the hemi-ketal, hemi-acetal, acyloxy ketal, acyloxy
acetal, ketal, acetal and enol forms that are described
herein.

Compositions and Methods
The compounds of this invention are caspase
inhibitors, and in particular ICE inhibitors.
Accordingly, these compounds are capable of targeting
and inhibiting events in IL-1-, apoptosis-, IGIF-, and
IFN-γ-mediated diseases, and, thus, the ultimate
activity of that protein in inflammatory diseases,
autoimmune diseases, destructive bone, proliferative
disorders, infectious diseases, and degenerative
diseases. For example, the compounds of this invention
inhibit the conversion of precursor IL-1β to mature IL-
1β by inhibiting ICE. Because ICE is essential for the
production of mature IL-1, inhibition of that enzyme
effectively blocks initiation of IL-1-mediated
physiological effects and symptoms, such as
inflammation, by inhibiting the production of mature
IL-1. Thus, by inhibiting IL-1β precursor activity,
the compounds of this invention effectively function as
IL-1 inhibitors.
Compounds of this invention also inhibit
conversion of pro-IGIF into active, mature IGIF by
inhibiting ICE. Because ICE is essential for the
production of mature IGIF, inhibition of ICE
effectively blocks initiation of IGIF-mediated
physiological effects and symptoms, by inhibiting
production of mature IGIF. IGIF is in turn essential
for the production of IFN-γ. ICE therefore effectively
blocks initiation of IFN-γ- mediated physiological
effects and symptoms, by inhibiting production of
mature IGIF and thus production of IFN-γ.
The compounds of this invention are
surprisingly bioavailable when compared with peptidyl
inhibitors, such as those described in, for example,
EP 618 223, EP 623 592, WO 93/09135, WO 93/16710, US
patent no. 5,434,248, WO 95/35308, or WO 96/33209.

Thus, the pharmaceutical compositions and methods of
this invention will be useful for controlling caspase
activity in vivo. The compositions and methods of this
invention will therefore be useful for controlling IL-
1, IGIF, or IFN-γ levels in vivo and for treating or
reducing the advancement, severity or effects of IL-1-,
apoptosis-, IGIF-, or IFN-γ-mediated conditions,
including diseases, disorders or effects.
Pharmaceutical compositions of this invention
comprise a compound of formula I or a pharmaceutically
acceptable salt thereof and a pharmaceutically
acceptable carrier. Such compositions may optionally
comprise an additional therapeutic agent. Such agents
include, but are not limited to, an anti-inflammatory
agent, a matrix metalloprotease inhibitor, a
lipoxygenase inhibitor, a cytokine antagonist, an
immunosuppressant, an anti-cancer agent, an anti-viral
agent, a cytokine, a growth factor, an immunomodulator,
a prostaglandin or an anti-vascular hyperproliferation
compound.
The term "pharmaceutically acceptable
carrier" refers to a non-toxic carrier that may be
administered to a patient, together with a compound of
this invention, and which does not destroy the
pharmacological activity thereof.
Pharmaceutically acceptable carriers that may
be used in the pharmaceutical compositions of this
invention include, but are not limited to, ion
exchangers, alumina, aluminum stearate, lecithin, serum
proteins, such as human serum albumin, buffer
substances such as phosphates, glycine, sorbic acid,
potassium sorbate, partial glyceride mixtures of
saturated vegetable fatty acids, water, salts or
electrolytes, such as protamine sulfate, disodium
hydrogen phosphate, potassium hydrogen phosphate,

sodium chloride, zinc salts, colloidal silica,
magnesium trisilicate, polyvinyl pyrrolidone,
cellulose-based substances, polyethylene glycol, sodium
carboxymethylcellulose, polyacrylates, waxes,
polyethylene-polyoxypropylene-block polymers, wool fat
and self-emulsifying drug delivery systems (SEDDS) such
as a-tocopherol, polyethyleneglycol 1000 succinate, or
other similar polymeric delivery matrices.
In pharmaceutical composition comprising only
a compound of embodiments A-D as the active component,
methods for administering these compositions may
additionally comprise the step of administering to the
subject an additional agent. Such agents include, but
are not limited to, an anti-inflammatory agent, a
matrix metalloprotease inhibitor, a lipoxygenase
inhibitor, a cytokine antagonist, an immunosuppressant,
an anti-cancer agent, an anti-viral agent, a cytokine,
a growth factor, an immunomodulator, a prostaglandin or
an anti-vascular hyperproliferation compound.
The term "pharmaceutically effective amount"
refers to an amount effective in treating or
ameliorating an IL-1-, apoptosis-, IGIF-, or IFN-
y-mediated disease in a patient. The term
"prophylactically effective amount" refers to an amount
effective in preventing or substantially lessening IL-
1-, apoptosis-, IGIF-, or IFN-γ-mediated diseases in a
patient.
The compounds of this invention may be
employed in a conventional manner for controlling IGIF
and IFN-γ levels in vivo and for treating diseases or
reducing the advancement or severity of effects which
are mediated by IL-1, apoptosis, IGIF, or IFN-γ. Such
methods of treatment, their dosage levels and
requirements may be selected by those of ordinary skill
in the art from available methods and techniques.

For example, a compound of this invention may
be combined with a pharmaceutically acceptable adjuvant
for administration to a patient suffering from an
IL-1-, apoptosis-, IGIF-, or IFN-γ-mediated disease in
a pharmaceutically acceptable manner and in an amount
effective to lessen the severity of that disease.
Alternatively, the compounds of this
invention may be used in compositions and methods for
treating or protecting individuals against IL-1,
apoptosis-, IGIF, or IFN-γ mediated diseases over
extended periods of time. The compounds may be
employed in such compositions either alone or together
with other compounds of this invention in a manner
consistent with the conventional utilization of enzyme
inhibitors in pharmaceutical compositions. For
example, a compound of this invention may be combined
with pharmaceutically acceptable adjuvants
conventionally employed in vaccines and administered in
prophylactically effective amounts to protect
individuals over an extended period of time against IL-
1-, apoptosis-, IGIF, or IFN-γ mediated diseases.
The compounds of formula I may also be co-
administered with other caspase or ICE inhibitors to
increase the effect of therapy or prophylaxis against
various IL-1-, apoptosis-, IGIF-, or IFN-γ mediated
diseases.
In addition, the compounds of this invention
may be used in combination either conventional anti-
inflammatory agents or with matrix metalloprotease
inhibitors, lipoxygenase inhibitors and antagonists of
cytokines other than IL-1β.
The compounds of this invention can also be
administered in combination with immunomodulators
(e.g., bropirimine, anti-human alpha-interferon
antibody, IL-2, GM-CSF, methionine enkephalin,

interferon-alpha, diethyldithiocarbamate, tumor
necrosis factor, naltrexone and EPO), with
prostaglandins, or with antiviral agents (e.g., 3TC,
polysulfated polysaccharides, ganiclovir, ribavirin,
acyclovir, alpha interferon, triraethotrexate and
fancyclovir) or prodrugs of these or related compounds
to prevent or combat IL-1-mediated disease symptoms
such as inflammation.
When the compounds of this invention are
administered in combination therapies with other
agents, they may be administered sequentially or
concurrently to the patient. Alternatively,
pharmaceutical or prophylactic compositions according
to this invention comprise a combination of a compound
of formula I and another therapeutic or prophylactic
agent.
The pharmaceutical compositions of this
invention may be administered orally, parenterally, by
inhalation spray, topically, rectally, nasally,
buccally, vaginally or via an implanted reservoir. We
prefer oral administration. The pharmaceutical
compositions of this invention may contain any
conventional non-toxic pharmaceutically-acceptable
carriers, adjuvants or vehicles. In some cases, the pH
of the formulation may be adjusted with
pharmaceutically acceptable acids, bases or buffers to
enhance the stability of the formulated compound or its
delivery form. The term parenteral as used herein
includes subcutaneous, intracutaneous, intravenous,
intramuscular, intra-articular, intrasynovial,
intrasternal, intrathecal, intralesional and
intracranial injection or infusion techniques.
The pharmaceutical compositions may be in the
form of a sterile injectable preparation, for example,
as a sterile injectable aqueous or oleaginous

suspension. This suspension may be formulated
according to techniques known in the art using suitable
dispersing or wetting agents (such as, for example,
Tween 80) and suspending agents. The sterile
injectable preparation may also be a sterile injectable
solution or suspension in a non-toxic parenterally-
acceptable diluent or solvent, for example, as a
solution in 1,3-butanediol. Among the acceptable
vehicles and solvents that may be employed are
mannitol, water, Ringer's solution and isotonic sodium
chloride solution. In addition, sterile, fixed oils
are conventionally employed as a solvent or suspending
medium. For this purpose, any bland fixed oil may be
employed including synthetic mono- or diglycerides.
Fatty acids, such as oleic acid and its glyceride
derivatives are useful in the preparation of
injectables, as are natural pharmaceutically-acceptable
oils, such as olive oil or castor oil, especially in
their polyoxyethylated versions. These oil solutions
or suspensions may also contain a long-chain alcohol
diluent or dispersant, such as those described in
Pharmacopeia Helvetica, or a similar alcohol.
The pharmaceutical compositions of this
invention may be orally administered in any orally
acceptable dosage form including, but not limited to,
capsules, tablets, and aqueous suspensions and
solutions. In the case of tablets for oral use,
carriers which are commonly used include lactose and
corn starch. Lubricating agents, such as magnesium*
stearate, are also typically added. For oral
administration in a capsule form, useful diluents
include lactose and dried corn starch. When aqueous
suspensions and solutions and propylene glycol are
administered orally, the active ingredient is combined
with emulsifying and suspending agents. If desired,

certain sweetening and/or flavoring and/or coloring
agents may be added.
The pharmaceutical compositions of this
invention may also be administered in the form of
suppositories for rectal administration. These
compositions can be prepared by mixing a compound of
this invention with a suitable non-irritating excipient
which is solid at room temperature but liquid at the
rectal temperature and therefore will melt in the
rectum to release the active components. Such
materials include, but are not limited to, cocoa
butter, beeswax and polyethylene glycols.
Topical administration of the pharmaceutical
compositions of this invention is especially useful
when the desired treatment involves areas or organs
readily accessible by topical application. For
application topically to the skin, the pharmaceutical
composition should be formulated with a suitable
ointment containing the active components suspended or
dissolved in a carrier. Carriers for topical
administration of the compounds of this invention
include, but are not limited to, mineral oil, liquid
petroleum, white petroleum, propylene glycol,
polyoxyethylene polyoxypropylene compound, emulsifying
wax and water. Alternatively, the pharmaceutical
composition can be formulated with a suitable lotion or
cream containing the active compound suspended or
dissolved in a carrier. Suitable carriers include, but
are not limited to, mineral oil, sorbitan monostearate,
polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-
octyldodecanol, benzyl alcohol and water. The
pharmaceutical compositions of this invention may also
be topically applied to the lower intestinal tract by
rectal suppository formulation or in a suitable enema

formulation. Topically-administered transdermal
patches are also included in this invention.
The pharmaceutical compositions of this
invention may be administered by nasal aerosol or
inhalation. Such compositions are prepared according
to techniques well-known in the art of pharmaceutical
formulation and may be prepared as solutions in saline,
employing benzyl alcohol or other suitable
preservatives, absorption promoters to enhance
bioavailability, fluorocarbons, and/or other
solubilizing or dispersing agents known in the art.
Dosage levels of between about 0.01 and about
100 mg/kg body weight per day, preferably between 0.5
and about 75 mg/kg body weight per day and most
preferably between about 1 and 50 mg/kg body weight per
day of the active ingredient compound are useful in a
monotherapy for the prevention and treatment of IL-1-,
apoptosis-, IGIF-, and IFN-γ mediated diseases,
including inflammatory diseases, autoimmune diseases,
destructive bone disorders, proliferative disorders,
infectious diseases, degenerative diseases, necrotic
diseases, inflammatory peritonitis, osteoarthritis,
acute pancreatitis, chronic pancreatitis, asthma, adult
respiratory distress syndrome, glomerulonephritis,
rheumatoid arthritis, systemic lupus erythematosus,
scleroderma, chronic thyroiditis, Graves' disease,
autoimmune gastritis, insulin-dependent diabetes
mellitus (Type I) , autoimmune hemolytic anemia,
autoimmune neutropenia, thrombocytopenia, chronic
active hepatitis, myasthenia gravis, inflammatory bowel
disease, Crohn's disease, psoriasis, atopic dermatitis,
graft vs. host disease, osteoporosis, multiple myeloma-
related bone disorder, leukemias and related disorders,
myelodysplastic syndrome, acute myelogenous leukemia,
chronic myelogenous leukemia, metastatic melanoma,

Kaposi's sarcoma, multiple myeloma, sepsis, septic
shock, Shigellosis, Alzheimer's disease, Parkinson's
disease, cerebral ischemia, myocardial ischemia, spinal
muscular atrophy, multiple sclerosis, AIDS-related
encephalitis, HIV-related encephalitis, aging,
alopecia, neurological damage due to stroke, ulcerative
collitis, infectious hepatitis, juvenile diabetes,
lichenplanus, acute dermatomyositis, eczema, primary
cirrhosis, uveitis, Behcet's disease, atopic skin
disease, pure red cell aplasia, aplastic anemia,
amyotrophic lateral sclerosis, nephrotic syndrome and
systemic diseases or diseases with effects localized in
the liver or other organs having an inflammatory or
apoptotic component caused by excess dietary alcohol
intake or viruses, such as HBV, HCV, HGV, yellow fever
virus, dengue fever virus, and Japanese encephalitis
virus.
Typically, the pharmaceutical compositions of
this invention will be administered from about 1 to 5
times per day or alternatively, as a continuous
infusion. Such administration can be used as a chronic
or acute therapy. The amount of active ingredient that
may be combined with the carrier materials to produce a
single dosage form will vary depending upon the host
treated and the particular mode of administration. A
typical preparation will contain from about 5% to about
95% active compound (w/w) . Preferably, such
preparations contain from about 20% to about 80% active
compound.
When the compositions of this invention
comprise a combination of a compound of formula I and
one or more additional therapeutic or prophylactic
agents, both the compound and the additional agent
should be present at dosage levels of between about 10%

to 80% of the dosage normally administered in a
monotherapy regime.
Upon improvement of a patient's condition, a
maintenance dose of a compound, composition or
combination of this invention may be administered, if
necessary. Subsequently, the dosage or frequency of
administration, or both, may be reduced, as a function
of the symptoms, to a level at which the improved
condition is retained when the symptoms have been
alleviated to the desired level, treatment should
cease. Patients may, however, require intermittent
treatment on a long-term basis upon any recurrence or
disease symptoms.
As the skilled artisan will appreciate, lower
or higher doses than those recited above may be
required. Specific dosage and treatment regimens for
any particular patient will depend upon a variety of
factors, including the activity of the specific
compound employed, the age, body weight, general health
status, s-x, diet, time of administration, rate of
excretion, drug combination, the severity and course of
the disease, and the patient's disposition to the
disease and the judgment of the treating physician.
IL-1 or apoptosis mediated diseases which may
be treated or prevented by the compounds of this
invention include, but are not limited to, inflammatory
diseases, autoimmune diseases, proliferative disorders,
infectious diseases, and degenerative diseases. The
apoptosis-mediated diseases which may be treated or
prevented by the compounds of this invention include
degenerative diseases.
IL-1 or apoptosis mediated inflammatory
diseases which may be treated or prevented include, but
are not'limited to osteoarthritis, acute pancreatitis,
chronic pancreatitis, asthma, and adult respiratory

distress syndrome. Preferably the inflammatory disease
is osteoarthritis or acute pancreatitis.
IL-l or apoptosis mediated autoimmune
diseases which may be treated or prevented include, but
are not limited to, glomerulonephritis, rheumatoid
arthritis, systemic lupus erythematosus, scleroderma,
chronic thyroiditis, Graves' disease, autoimmune
gastritis, insulin-dependent diabetes mellitus (Type
I), autoimmune hemolytic anemia, autoimmune
neutropenia, thrombocytopenia, chronic active
hepatitis, myasthenia gravis, multiple sclerosis,
inflammatory bowel disease, Crohn's disease, psoriasis,
atopic dermatitis and graft vs. host disease.
Preferably the autoimmune disease is rheumatoid
arthritis, inflammatory bowel disease, Crohn's disease,
psoriasis, or atopic dermatitis.
IL-l or apoptosis mediated destructive bone
disorders which may be treated or prevented include,
but are not limited to, osteoporosis and multiple
myeloma-related bone disorder.
IL-l or apoptosis mediated proliferative
diseases which may be treated or prevented include, but
are not limited to, leukemias and related disorders,
such as myelodysplastic syndrome, acute myelogenous
leukemia, chronic myelogenous leukemia, metastatic
melanoma, Kaposi's sarcoma, and multiple myeloma.
IL-l or apoptosis mediated infectious
diseases which may be treated or prevented include, but
are not limited to, sepsis, septic shock, and
Shigellosis.
IL-l or apoptosis mediated degenerative or
necrotic diseases which may be treated or prevented by
the compounds of this invention include, but are not
limited to, Alzheimer's disease, Parkinson's disease,
cerebral ischemia, and myocardial ischemia.

Preferably, the degenerative disease is Alzheimer's
disease.
IL-1 or apoptosis-mediated degenerative
diseases which may be treated or prevented by the
compounds of this invention include, but are not
limited to, Alzheimer's disease, Parkinson's disease,
cerebral ischemia, myocardial ischemia, spinal muscular
atrophy, multiple sclerosis, AIDS-related encephalitis,
HIV-related encephalitis, aging, alopecia, and
neurological damage due to stroke.
Other diseases having an inflammatory or
apoptotic component may be treated or prevented by the
compounds of this invention. Such diseases may be
systemic diseases or diseases with effects localized in
the liver or other organs and may be caused by, for
example, excess dietary alcohol intake or viruses, such
as HBV, HCV, HGV, yellow fever virus, dengue fever
virus, and Japanese encephalitis virus.
IGIF- or IFN-γ-mediated diseases which may be
treated or prevented by the compounds of this invention
include, but are not limited to, inflammatory,
infectious, autoimmune, proliferative,
neurodegenerative and necrotic conditions.
IGIF- or IFN-γ-mediated inflammatory diseases
which may be treated or prevented include, but are not
limited to osteoarthritis, acute pancreatitis, chronic
pancreatitis, asthma, rheumatoid arthritis,
inflammatory bowel disease, Crohn's disease, ulcerative
collitis, cerebral ischemia, myocardial ischemia and
adult respiratory distress syndrome. Preferably, the
inflammatory disease is rheumatoid arthritis,
ulcerative collitis, Crohn's disease, hepatitis or
adult respiratory distress syndrome.
IGIF- or IFN-γ-mediated infectious diseases
which may be treated or prevented include, but are not

limited to infectious hepatitis, sepsis, septic shock
and Shigellosis.
IGIF- or IFN-γ-mediated autoimmune diseases
which may be treated or prevented include, but are not
limited to glomerulonephritis, systemic lupus
erythematosus, scleroderma, chronic thyroiditis,
Graves' disease, autoimmune gastritis, insulin-
dependent diabetes mellitus (Type I), juvenile
diabetes, autoimmune hemolytic anemia, autoimmune
neutropenia, thrombocytopenia, myasthenia gravis,
multiple sclerosis, psoriasis, lichenplanus, graft vs.
host disease, acute dermatomyositis, eczema, primary
cirrhosis, hepatitis, uveitis, Behcet's disease, atopic
skin disease, pure red cell aplasia, aplastic anemia,
amyotrophic lateral sclerosis and nephrotic syndrome.
Preferably, the autoimmune disease is
glomerulonephritis, insulin-dependent diabetes mellitus
(Type I), juvenile diabetes, psoriasis, graft vs. host
disease or hepatitis.
More preferred diseases vhich may be treated
or prevented include rheumatoid arthritis, inflammatory
bowel .disease, including Crohn's disease and ulcerative
colitis, inflammatory peritonitis, septic shock,
pancreatitis, traumatic brain injury, organ transplant
rejection, osteoarthritis, asthma, psoriasis,
Alzheimer's disease, atopic dermatitis, or leukemias
and related disorders, such as myelodysplastic syndrome
or multiple myeloma.
Accordingly, one embodiment of this invention
provides a method for treating or preventing an IL-1 or
apoptosis mediated disease in a subject comprising the
step of administering to the subject any compound,
pharmaceutical composition, or combination described
herein and a pharmaceutically acceptable carrier.

Another embodiment of this invention provides
a method for decreasing IGIF production in a subject
comprising the step of administering to the subject any
compound, pharmaceutical composition, or combination
described herein and a pharmaceutically acceptable
carrier.
Yet another embodiment of this invention
provides a method for decreasing IFN-γ production in a
subject comprising the step of administering to the
subject any compound, pharmaceutical composition, or
combination described herein and a pharmaceutically
acceptable carrier.
Although this invention focuses on the use of
the compounds disclosed herein for preventing and
treating IL-1, apoptosis-, IGIF, and IFN-γ-mediated
diseases, the compounds of this invention can also be
used as inhibitory agents for other cysteine proteases.
The compounds of this invention are also
useful as commercial reagents which effectively bind to
caspases or other cysteine proteases including, but not
limited to ICE. As commercial reagents, the compounds
of this invention, and their derivatives, may be used
to block proteolysis of a target peptide in biochemical
or cellular assays for ICE and ICE homologs or may be
derivatized to bind to a stable resin as a tethered
substrate for affinity chromatography applications.
These and other uses which characterize commercial
cysteine protease inhibitors will be evident to those
of ordinary skill in the art.
In order that this invention be more fully
understood, the following examples are set forth.
These examples are for the purpose of illustration only
and are not to be construed as limiting the scope of
the invention in any way.

GENERAL METHODS
Analytical HPLC conditions:
Column: C-18, Particle size: 5 p, Pore size: 100A,
Column size: 4.6 x 15 0 mm
Solvent A: 0.1% TFA / 1% MeCN / 98.9% water
Solvent B: 0.1% TFA / 99.9% MeCN
Gradient: A to B over 2 0 min at a flow rate of l
mL/min
Column: Cyano, Particle size: 5 µ, Pore size: 100A,
Column size: 4.6 x 150 mm
Solvent A: 0.1% TFA / 1% MeCN / 98.9% water
Solvent B: 0.1% TFA / 99.9% MeCN
Gradient: A / B = 99k / 1% to 50% / 50% over 20 min at
a flow rate of 1 mL/min
HPLC Mass Spectral Analysis
Mass Spectral Analysis: All mass spectral
data were collected using a Micromass Quattro II triple
guadrupole mass spectrometer (Beverly, MA) equipped
with a cross-flow electrospray ionization source. The
mass spectrometer was coupled to a HPLC system
manufactured by Hewlett-Packard (HP110 0) . The
autosampler for the system was a Gilson 215 (Middleton,
WI) liquid handler. All of the equipment was
controlled by the MassLynx software package purchased
from Micromass.
Mass spectral analysis was performed by
liquid chromatography-MS to determine purity and
confirm molecular weight simultaneously. In instances

where the sample purity had been determined by other
means, a flow injection analysis (FIA) was used instead
of the full chromatography analysis. In all cases,
both positive and negative ion spectra were collected.
Mass Spectrum Acquisition Conditions: For all
experiments, the mass spectrometer was configured in
electrospray mode with the cross-flow counter
electrode. A flow splitter was used to reduce the flow
from the HPLC to 4 0% of the original flow. The inlet
temperature was set to 14 0 °C and the drying gas flow
was set to maximize signal. The resolution of the mass
spectrometer was adjusted to 0.65 amu FWHM and data was
collected in centroid mode. In positive ion mode, the
cone voltage was set to 25V, the capillary voltage was
3.8 kV. In negative ion mode, the cone voltage was set
to 25 V and the capillary voltage was set to 3.5 kV.
In both, positive and negative ion mode, the time to
acquire a full spectrum was Is with a switching time of
0.25 seconds between scans. The mass range scanned for
molecules with an expected molecular weight of less
than 350 amu was 70-500 m/z while for molecules with a
expected mass of more than 350 amu the mass to charge
ratio scanned was 200-1000 m/z.
Chromatography Conditions: Liquid
chromatography was performed using a YMC AQ C18 column
(150 mm X 3mm with 5 µ m particle and a 120A pore size) .
For all analysis, MeCN with 0.2 % formic acid was
combined with water with 0.2% formic acid to form the
elution gradient. The gradient profile consisted of
starting with 15 % MeCN: water and increasing the
amount of MeCN linearly over ten minutes to 90%. That
concentration was held constant for 2 minutes before
returning to initial conditions. During the entire
analysis the flow rate was 0.9mL/min.

Flow Injection Conditions: A 1:1 mixture of
the water to MeCN (both with 0.2% formic acid added)
was used to acquire the FIA data. The flow rate was
set to 0.3 ml/min.
-H NMR
All 1H NMR spectra were acquired using a
Bruker Instruments AMX-500 NMR spectrometer in the
solvent given.
SYNTHETIC METHODS
General Procedure for the Preparation of Compounds of
Formula I, Embodiment C (Schemes I-VI)

Procedure for the preparation of analogs 5a-5bd.

In Schemes I-VIII, the variable LR refers to the
linker-resin and is defined as shown above in Scheme I.
Step 1: A 6.7 g portion (0.8 mmol/gram loading, 5.36
mmol) of 4-methyl benzhydrylamine hydrochloride salt
resin (Scheme I) was washed with DMF (3 x 50 mL), 10%
DIEA/DMF. (3 x 50 mL) and N-methylpyrrolidinone (NMP)(3

x 50 mL) . To a suspension of the washed resin in 25 mL
of NMP was added successively compound 1 (1.1 eq, 3.5
g, 5.90 mmol) DIEA (3.33 eq, 3.1 mL, 17.70 mmol), 1-
hydroxybenzotriazole hydrate (HOBt) (1.1 eq, 797 mg,
5.90 mmol), and O-benzotriazole-N,N,N,N'-
tetramethyluronium hexafluorophosphate (HBTU) (1.1 eq,
2.24 g, 5.90 mmol). Compound 1 was prepared according
to the literature procedure of A. M. Murphy et al, J.
Am. Chem. Soc., 114, pp. 3156-3157 (1992). The mixture
was rotated at room temperature overnight using a wrist
arm shaker.
The resulting mixture was filtered, and the resin was
rinsed with DMP then treated with 12 mL of a 2 0%
solution of acetic anhydride in DMF for 3 0 minutes at
room temperature. The mixture was filtered, and the
resin was washed successively with DMP (2 x 5 0mL) ,
CH3OH (50mL) , 1:1 DMP/ CH2Cl2 (2 x 50mL) , CH3OH (50mL)
and CH2Cl2 (3 x 50mL), After drying in vacuo, 9.0
grams of resin 2 were obtained (0.48 mmol/gram
loading).
Step 2: To 4.5 g of resin 2 (0.48 mmol/gram, 2.16 mmol)
was added 25 mL of a 20% solution of piperidine in DMF.
The suspension was rotated at room temperature for 5
minutes and drained. The procedure was repeated over
20 minutes. The resin was then washed successively
with DMF (2 X 40 mL) , CH3OH (40 mL) , CH2Cl2 (2 x 40 mL) ,
CH3OH (40 mL) and NMP (40 mL) . To a suspension of
resin in 40 mL of NMP was added successively 2.92 g of
N-Fmoc-proline (4 eq, 8.64 mmol), 3.0 mL of DIEA (8 eq,
17.28 mmol), 1.17 g of HOBt (4 eq, 8.64 mmol) and 3.27
g of HBTU (4 eq, 8.64 mmol). The mixture was rotated
at room temperature overnight and drained. This

coupling procedure was repeated over 3 hours. The
resin was then washed successively with DMF (2 x 40
mL) , CH3OH (40 mL) , 1:1 DMF/ CH2Cl2 (2 x 40 mL), CH3OH
(40 mL) and CH2Cl2 (3 x 40 mL) , and briefly dried in
vacuo to afford resin 3.
Step 3: A suspension of resin 3 in 25 mL of a 20%
solution of piperidine in DMF was rotated at room
temperature for 5 minutes. The suspension was drained.
The procedure was repeated over 20 minutes. The resin
was washed successively with DMF (2 x 40 mL) , CH3OH (40
mL) , CH2Cl2 (2 x 40 mL), CH3OH (40 mL) and NMP (2 x 40
mL) . To a suspension of resin in 4 0 mL of NMP was
added successively 2.93 g of N-Fmoc-valine (4 eq, 8.64
mmol) , 3.0 mL of DIEA (8 eq, 17.28 mmol) , 1.17 g of
HOBt (4 eq, 8.64 mmol) and 3.27 g of HBTU (4 eq, 8.64
mmol) . The mixture was rotated at room temperature
overnight and drained. This coupling procedure was
repeated over 3 hours. The resin was then washed
successively with DMF (2 x 40 mL) , CH3OH (40 mL) , 1:1
DMF/ CH2Cl2 (2 x 40 mL), CH3OH (40 mL) and CH2Cl2 (3 x
4 0 mL) , and dried in vacuo to afford resin 4 (0.45
mmol/gram) .
Step 4: To a 0.05 mmol portion of resin 4 was added 2
mL of a 2 0% solution of piperidine in DMF. The
suspension was rotated at room temperature for 5
minutes, and drained. The procedure was repeated over
2 0 minutes. The resulting resin was washed
successively with DMF (3x5 mL) , CH3OH (5 mL) , and NMP
(3x5 mL) . The desired carboxylic acid was then added
(4 eq, 0.2 mmol), followed by 0.8 mL of a 0.25M
solution of HOBt in NMP, 0.14 mL of DIEA (8 eq, 0.4

mmol) and 0.8 mL of a 0.25M solution of HBTU in NMP.
The mixture was rotated at room temperature overnight
and drained. The resin was washed successively with
DMF (2X5 mL) , CH30H (5 mL) , 1:1 DMF/ CH2Cl2 (2x5
mL) , CH3OH (5 mL) and CH2Cl2 (3x5 mL) , and dried in
vacuo. A 2 mL portion of a 95% solution of TFA in
water was then added to the resin. The mixture was
stirred at room temperature for one hour, and filtered.
The filtrate was evaporated, and the residue was taken
up in acetonitrile-water and purified by preparative
HPLC to afford compounds 5a-5bd.
Product yield, analytical HPLC conditions,
HPLC retention time, product purity, and mass spectral
data obtained for examples 5a-5bd, 7a-7at, 9a-9g, 15a-
15f, 16a-16b, 17a-17e, 18a-18f, 20a-20t, 23a-23i, 24a-
24e, 25a-25e, 26a-26h, 27a-27n, 28a-28c, 29a-29s, 32a-
32e are provided in Table 1 unless noted otherwise.























Analogs of 7a-7at were prepared as described above in
Scheme I only substituting Fmoc-alanine for Fmoc-valine
in Step 3 (Scheme II).

Step 3: A suspension of resin 3 (3.5 g, 1.75 mmol) in
2 0 mL of a 2 0% solution of piperidine in DMF was
rotated at room temperature for 5 minutes. The
suspension was drained. The procedure was repeated over
2 0 minutes. The resin was washed successively with DMF
(2 x 30 mL), CH3OH (30 mL) , CH2Cl2 (2 x 30 mL) , CH3OH
(30 mL) and NMP (2 x 30 mL) . To a suspension of resin
in 30 mL of NMP was added successively 1.44 g of N-
Fmoc-alanine (4 eq, 7.0 mmol), 2.4 mL of DIEA (8 eq,
14.0 mmol), 0.95 g of HOBt (4 eq, 7.0 mmol) and 2.66 g
of HBTU (4 eq, 7.0 mmol). The mixture was rotated at
room temperature overnight and drained. This coupling
procedure was repeated over 3 hours. The resin was
then washed successively with DMF (2 x 30 mL) , CH3OH
(30 mL) , 1:1 DMF/ CH2Cl2 (2 X 30 mL) , CH3OH (30 mL) and
CH2Cl2 (3x30 mL) , and dried in vacuo to afford resin
6 (0.50 mmol/gram).
Step 4: To a 0.125 mmol portion of resin 6 was added 5
mL of a 20% solution of piperidine in DMF. The
suspension was rotated at room temperature for 5
minutes, and drained. The procedure was repeated over
2 0 minutes. The resulting resin was washed
successively with DMF (3x5 mL) , CH30H (5 mL) , and NMP
(3x5 mL) . The desired carboxylic acid was then added
(4 eq, 0.6 mmol), followed by 2.0 mL of a 0.25M
solution of HOBt in NMP, 0.35 mL of DIEA (8 eq, 1.0
mmol) and 2.0 mL of a 0.25M solution of HBTU in NMP.
The mixture was rotated at room temperature overnight
and drained. The resin was washed successively with
DMF (3 X 5 mL) , CH3OH (5 mL) , 1:1 DMF/ CH2Cl2 (2x5
mL) , CH3OH (5 mL) and CH2Cl2 (3x5 mL) , and dried in
vacuo. A 5 mL portion of a 95% solution of TFA in
water was then added to the resin. The mixture was

stirred at room temperature for one hour, and filtered.
The filtrate was evaporated, and the residue was
dissolved in acetonitrile-water and purified by
preparative HPLC to afford compounds 7a-7at.









Procedure for the preparation of analogs 9a-9g.

Step 1: A 10.0 g portion (0.75 mmol/gram loading, 7.5
mmol) AgroPore-aminomethyl resin (catalog number
800047) was washed with DMF (3 x 40 mL) , 10% DIEA/DMF
(3x40 mL) , DMF and NMP (3 x 40 mL) . To above resin
was added successively compound 1 (0.87 eq, 3.88 g,
6.55 mmol), HBTU (1.14 eq, 3.13 g, 8.25 mmol), HOBt

(1.14 eq, 1.26 g, 8.25 mmol), and NMP (40 mL) . The
reagents were then mixed by bubbling nitrogen through
the bottom of the flask for two minutes at room
temperature. N,N-diisopropylethylamine (3.33 eq, 4.35
mL, 25 mmol) was added and the resulting suspension
mixed at room temperature overnight, filtered, then
washed successively with NMP (3x 40 mL) and DMF (3x 40
mL) . The resin was then treated with 50 mL of a 20%
solution of acetic anhydride in DMF for 3 8 minutes at
room temperature. The mixture was filtered, and the
resin was washed successively with NMP (3x 40 mL)
CH2Cl2 (3x 40 mL) , 1:1 CH3OH / CH2Cl2 (3x 40 mL) , and
CH3OH (3x 40 mL). After drying in vacuo, 13.76 grams
of resin 2 were obtained (0.35 mmol/gram loading).
Step 2: Seven reaction vessels were each charged with
181 mg of resin 2 (0.48 mmol/gram, 0.063 mmol) then
washed with CH2Cl2 (3x1 mL) and NMP (3x1 mL) . Then
each vessel was treated with 1 mL of a 25% solution of
piperidine in DMF and mixed (vorte'x) at room
temperature for 15 minutes. This procedure was repeated
in triplicate. Each vessel was then washed three times
with NMP (3x1 mL) . The vessels were then treated
with 500 µl of a solution of 0.4 M (2S,4R) -Fmoc-4-
amino-l-Boc-pyrrolidine-2-carboxylic acid /0.4 M HOBt/
NMP, 500 µl of a solution of 0.4 M HBTU/ NMP, and 250
µl of a solution of 1.6 M DIEA/ NMP and mixed for 3
hours at room temperature. After mixing, the vessels
were drained and the procedure was repeated.
Step 3: The resulting resin was washed with NMP (3 x
lmL) and then treated with 1 mL of a 25% solution of
piperidine in DMF and mixed (vortex) at room
temperature for 15 minutes. The procedure was repeated

in triplicate. The resulting resin was washed with NMP
(3 x 1mL) then treated with either acetic anhydride, or
isopropyl isocyanate, or methanesulfonyl chloride, or
methyl chloroformate. For acetic anhydride: add 3 00 µl
of a 1.6 M DIEA/ NMP solution and 1 mL of a solution of
0.5 M acetic anhydride/0.125 M DIEA/ 0.015 M HOBt in
NMP. For isopropyl isocyanate: add 300 µl of a 1.6 M
DIEA/ NMP solution and 1 mL of a solution of l M
isopropyl isocyanate in NMP. For methanesulfonyl
chloride: add 600 µl of a solution of 1 M pyridine in
CH2Cl2 and 600 µl of a solution of 1M methanesulfonyl
chloride in CH2Cl2 . For methyl chloroformate: add 500
µl of a 1.6 M DIEA/ NMP solution and 1 mL of a solution
of 0.7 M methyl chlorof ormate in CH2Cl2 The resulting
suspensions were mixed for 6 hours at room temperature,
the solvent drained and the coupling procedure
repeated.
Step 4: The resulting resin was washed with NMP (3 x
lmL) then treated with a 1:1 mixture of TFA/ CH2Cl2 at
room temperature for 30 minutes. The resulting resin
was then washed with CH2Cl2 (3 x lmL) and NMP (3 x
lmL) . The resin was then treated with 500 µl of a
solution of 0.4 M Fmoc-valine-carboxylic acid/0.4 M
HOBt/ NMP, 500 µl of a solution of 0.4 M HBTU/ NMP, and
250 µl of a solution of 1.6 M DIEA/ NMP and mixed for 3
hours at room temperature. After mixing, the vessels
were drained and the coupling procedure was repeated.
Step 5: The resulting resin was washed with NMP (3 x
lmL) then treated with 1 mL of a 25% solution of
piperidine in DMF and mixed (vortex) at room
temperature for 15 minutes. This procedure was repeated
in triplicate. The resulting resin was washed with NMP

(3 x 1mL) then treated with either 500 µl of a solution
of 0.4 M 1-isoquinoline carboxylic acid/0.4 M HOBt/ NMP
or 500 µl of a solution of 0.4 M p-anisic acid acid/0.4
M HOBt/ NMP. The resulting mixtures were treated with
500 µl of a solution of 0.4 M HBTU/ NMP and 2 50 µl of a
solution of 1.6 M DIEA/ NMP then mixed for 3 hours at
room temperature, the solvent drained and the procedure
repeated. The resulting resin was treated with 1.5 mL
of a 95% solution of TFA in water and stirred at room
temperature for one hour then filtered. The filtrate
was evaporated, and the residue was taken up in a 2:1:2
mixture of DMF /acetonitrile/water and purified by
preparative HPLC to afford compounds 9a-9g.




Preparation of analogs 15 and 16 (Scheme IV) :
Synthesis of 2- (S) -piperazine-1, 2, 4-tricarboxylic acid
4-tert-butyl ester 1-(9H-fluoren-9-ylmethyl) ester.


To a solution of 2-(S)-piperazine carboxylic acid
(Lonza) (3g, 15 mmol) in 1:1 H2O:dioxane (30 mL) was
added a solution of (Boc)2O in dioxane (3.3g, 15mmol,
in 5mL dioxane) while maintaining the pH at 11 with 1N
NaOH . The pH was maintained over 3 hours at room
temperature. The solution was adjusted to pH9.5 with 1N
HCl, cooled to 0°C and treated with Fmoc-Cl (3.87g,15
mmol). The pH was maintained at 9.5 for 1 hour and the
mixture stirred at room temperature overnight. The
resulting suspension was filtered and the filtrate
treated with 1N KHSO4 to pH 2 then extracted with ethyl
acetate (2 x 75 mL). The organic layer was dried with
brine and MgSO4 filtered, and concentrated to give
colorless oil. The oil was dissolved in ethyl acetate
and added to hexane to give 3.5g (51% yield) of white
solid after isolation. 1H NMR (500 MHz, DMSO-d6) δ 1.55
(s, 9H) 2.80- 3.5 (m, 3H), 3.8-4.9 (m, 5H) , 5.7 (bs,
1H), 7.3 (m, 2 H), 7.3- 7.9 ppm (m, 8H), LC/MS (ES~)
m/e 451.3 (M-H)
Step 1: To 5 g of resin 2 (0.375 mmol/gram 1.82 mmol)
was added 25 mL of a 20% solution of piperidine in DMF.
The suspension was rotated at room temperature for 5
minutes and drained. The procedure was repeated over
20 minutes. The resin was then washed successively
with DMF (2 x 50 mL) , CH3OH (50 mL) , CH2Cl2 (2 x 50 mL) ,
CH3OH (50 mL) and NMP (50 mL). To a suspension of
resin in 25 mL of NMP was added successively 3.5g of N-
Fmoc-Boc piperazine carboxlyic acid (4 eq, 7.48 mmol),
1.0 mL of DIBA (8 eq, 14.36 mmol), 1.01 g of HOBt (4
eq, 7.48 mmol) and 2.83g of HBTU (4 eq, 7.48 mmol).
The mixture was rotated at room temperature overnight
and drained. This coupling procedure was repeated over
3 hours. The resin was then washed successively with

DMF (2 X 50 mL), CH3OH (50 mL) , 1:1 DMF/ CH2Cl2 (2 x 50
mL) , CH3OH (1 x 50 mL) and CH2Cl2 (3 x 50 mL) , and
briefly dried in vacuo to afford resin 10.
Step 2: To 5 g (0.335 mmol/gram loading, 1.675 mmol) of
10 was added 25 mL of a 20% solution of piperidine in
DMF. The suspension was rotated at room temperature
for 5 minutes and drained. The procedure was repeated
over 2 0 minutes. The resin was then washed
successively with DMF (2 x 50 mL) , CH3OH (50 mL) , CH2Cl2
(2 x 50 mL) , CH3OH (50 mL) and NMP (2 x 50 mL) . To a
suspension of resin in 25 mL of NMP was added
successively 2.08g of N-Fmoc-valine or N-Fmoc-alanine
(4 eg, 6.7 mmol), 1.17mL of DIEA (4 eq, 6.7 mmol),
0.905 g of HOBt (4 eq, 6.7mmol) and 1.38 g of HBTU (4
eq, 3.66 mmol) . The mixture was rotated at room
temperature overnight and drained. This coupling
procedure was repeated over 3 hours. The resin was
then washed successively with DMF (2 x 50 mL), CH3OH
(50 mL) , 1:1 DMF/ CH2Cl2 (2 X 50 mL) , CH3OH (50 mL) and
CH2Cl2 (3 x 50 mL) , and dried in vacuo to afford resin
11 or 12 respectively (0.35 mmol/gram, 5g) .
Step 3: To a 1.5g ( 0.165 mmol) portion of resin 11 or
12 was added 2 mL of a 20% solution of piperidine in
DMF. The suspension was rotated at room temperature
for 5 minutes, and drained. The procedure was repeated
over 20 minutes. The resulting resin was washed
successively with DMF (3 x 15 mL), CH3OH (15 mL), and
NMP (3 x 15 mL) . The desired carboxylic acid was then
added (4 eq, 0.66 mmol), followed by 0.25g HOBt
(0.66mmol), 0.12 mL of DIEA (4 eq, 0.66 mmol) and 0.89g
(0.66mmol) HBTU in NMP. The mixture was rotated at
room temperature overnight and drained. The resin was

washed successively with DMF (2 x 15 mL) , CH3OH (15
mL) , 1:1 DMF/ CH2Cl2 (2 X 15 mL) , CH3OH (15 mL) and
CH2Cl2 (3 x 15 mL) , and dried in vacuo to afford 13 or
14.
Step 4 : A 2 mL portion of a 95% solution of TFA in
water was then added to the resin. The mixture was
stirred at room temperature for one hour, and filtered.
The filtrate was evaporated, and the residue was taken
up in acetonitrile-water and purified by preparative
HPLC to afford compounds 15 and 16.


Procedure for the synthesis of analogs 17 and 18 (see
Scheme IV) :
Step 5: Resin 13 or 14 was treated with 2 mL 25%
TFA/CH2Cl2 for 30 min and washed with DMF (2x5 mL) ,
10% DIEA/CH2Cl2 (2 X 5 mL) DMF/CH2Cl2 (2 x 5 mL) , CH3OH
(5 mL) and CH2Cl2 (3x5 mL) and dried for five
minutes. The resulting resin was washed with NMP (3x1
mL) then treated with acetic anhydride, or methoxacetic

acid, or 2-propanesulfonyl chloride, or isopropyl
isocyanate, or methanesulfonyl chloride, or methyl
chloroformate according to the procedure used to
prepare analogs 9 (Scheme III) . Compounds 17 and IB
were obtained as described in Step 4 for compounds 15
and 16.
Compounds 17a and 17b were prepared by reductive
amination using Na(OAc)3BH and ECHO (38% in H20, 0.2
mL) and CH3COOH (0.02 mL) prior to Step 4 and compound
18e was prepared by treatment with phosgene followed by
ammonia prior to Step 4.



Procedure for the preparation of analogs 20.
Compounds 20a-20t were prepared according to the
procedure described for compounds 5 (Scheme I) only
substituting the appropriate Fmoc-amino acid for Fmoc-
Valine in Step 3 (Scheme V).

Preparation of 3-({1-[2-(4-amino-3-chloro-
benzoylaroino) -3-methylsulfonyl-propionyl] -pyrrolidine-
2-carbonyl}-amino) -4-oxo-butyric acid (20i) .
A suspension of 0.132 mmol of resin 3 in 4 mL of 20%
piperidine in DMF was rotated at room temperature for 5
minutes, and the mixture was drained. The procedure
was repeated over 2 0 minutes. The resin was washed
successively with DMF (twice), CH3OH (once), CH2Cl2
(twice) , CH3OH (once) and NMP (twice) . To a suspension
of the resin in 4 mL of NMP was added successively 18 9
mg of N-Fmoc-methyl cysteine (4 eq, 0.528 mmol), 0.185
mL of DIEA (8 eq, 1.056 mmol), 71 mg of HOBt (4 eq,

0.528 mmol) and 200 mg of HBTU (4 eq, 0.528 mmol). The
mixture was rotated at room temperature overnight and
drained. This coupling procedure was repeated over 3
hours. The resin was then washed successively with DMF
(twice), CH3OH (once), and 1:1 DMF/ CH2Cl2 (twice),
CH3OH (once) and CH2Cl2 (three times), and dried in
vacuo.
A suspension of 100 mg of this resin in 2 mL of 20%
piperidine in DMF was rotated at room temperature for 5
minutes, and drained. The procedure was repeated over
2 0 minutes. The resin was washed successively with DMF
(twice), CH3OH (once), CH2Cl2 (twice) , CH3OH (once) and
NMP (twice). To a suspension of resin in 2 mL of NMP
was added successively 3 8 mg of 4-amino-3-chlorobenzoic
acid (4 eg, 0.2 mmol), 0.14 0 mL of DIEA (8 eg, 0.4
mmol), 27 mg of HOBt (4 eq, 0.2 mmol) and 16 mg of HBTU
(4 eq, 0.4 mmol). The mixture was rotated at room
temperature overnight and drained. The resin was then
washed successively with DMF (twice) , CH3OH (once), and
1:1 DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2 (three
times), and dried in vacuo. The resin was then treated
with 2 mL of 95% TFA in water for lh. The suspension
was filtered, the filtrate was concentrated in vacuo
and purified by preparative HPLC to afford the title
compound (20i).
Preparation of 3-({l-[2-(3,5-dichloro-4-hydroxy-
benzoylamino) -4-methanesulfonyl-butyryl] -pyrrolidine-2-
carbonyl}-amino)-4-oxo-butyric acid (20p).
Compound 20p was prepared according to the procedure
used for the preparation of 20i using N-Fmoc-methionine

as the first component coupled to resin 3, and 3,5-
dichloro-4-hydroxybenzoic acid as the second component.
Preparation of 3-[(l-{2-[(isoquinoline-1-carbonyl)-
amino] -3-methanesulfonyl-propionyl}-pyrrolidine-2-
carbonyl)-amino]-4-oxo-butyric acid (20r) .
N-Fmoc methyl cysteine was oxidized to the
corresponding sulfone using the method of B. M. Trost
and D. P. Curran, Tetrahedron Lett. 22, pp. 1287-190
(1981). To a solution of 0.714 g (2 mraol) of N-Fmoc
methyl cysteine in 24 mL of a 1:1 solution of CH3OH -
water stirred at 0°C was added 3.68 g (3 eq, 6 mmol) of
Oxone™. The mixture was stirred at room temperature
for 48 h, diluted with water, acidified to pH 2 using
6N HCl, and extracted with three 100 mL portions of
ethyl acetate. The combined organic extracts were
dried (MgSO4) and concentrated in vacuo to afford 0.700
g (89% yield) of sulfone: 1H NMR (DMSO-d6, 500 MHz) δ
2.97 (s, 3H) , 3.49-3.59 (m, 2H), 4.25 (m, 1H), 4.30-
4.38 (m, 2H), 4.46 (m, 1H), 7.33 (t, 2H), 7.42 (t,
2H,), 7.70-8.00 (m, 4H) ; exact mass calculated for
Cl9H19NO6S m/e 3 89.09, found m/e 390.2.
A suspension of 0.250 mmol of resin 3 in 10 mL of 20%
piperidine in DMF was rotated at room temperature for 5
minutes, and the mixture was drained. The procedure
was repeated over 2 0 minutes. The resin was washed
successively with DMF (twice) , CH3OH (once) , CH2Cl2
(twice) , CH3OH (once) and NMP (twice). To a suspension
of the resin in 6 mL of NMP was added successively 200
mg of N-Fmoc-methyl cysteine sulfone (4 eq, 0.50 mmol),
0.175 mL of DIEA (8 eq, 1.00 mmol), 70 mg of HOBt (4
eq, 0.50 mmol) and 188 mg of HBTU (4 eq, 0.50 mmol).
The mixture was rotated at room temperature overnight

and drained. This coupling procedure was repeated over
3 hours. The resin was washed successively with DMF
(twice), CH3OH (once), 1:1 DMF/ CH2Cl2 (twice), CH3OH
(once) and CH2Cl2 (three times), and dried in vacuo .
A suspension of 150 mg of this resin in 4 mL of 20%
piperidine in DMF was rotated at room temperature for 5
minutes, and drained. The procedure was repeated over
20 minutes. The resin was washed successively with DMF
(twice), CH3OH (once), CH2Cl2 (twice), CH3OH (once) and
NMP (twice). To a suspension of resin in 3 mL of NMP
was added successively 52 mg of 1-
isoguinolinecarboxylic acid (4 eq, 0.3 mmol), 0.104 mL
of DIEA (8 eq, 0.6 mmol), 37 mg of HOBt (4 eq, 0.3
mmol) and 104 mg of HBTU (4 eq, 0.3 mmol). The mixture
was rotated at room temperature overnight and drained.
The resin was washed successively with DMF (twice),
CH3OH (once), and 1:1 DMF/ CH2Cl2 (twice), CH3OH (once)
and CH2Cl2 (three times), and dried in vacuo. The
resin was then treated with 2 mL of 95% TFA in water
for lhour. The suspension was filtered, the filtrate
was' concentrated in vacuo and purified by preparative
HPLC to afford the title compound (20r).
Preparation of 3-({1-[2-(3,5-dichloro-4-hydroxy-
benzoylamino) -3-methanesulfonyl-propionyl] -pyrrolidine-
2-carbonyl}-amino)-4-oxo-butyric acid (20s).
Compound 20s was prepared according to the procedure
used for the preparation of 20i, using 3,5-dichloro-4-
hydroxybenzoic acid in place of 1-
isoquinolinecarboxylic acid.




Procedure for the preparation of analogs 23.
Compounds 23a-23i were prepared according to the
procedure described for compounds 7 (Scheme II) only-
substituting the appropriate Fmoc-amino acid for Fmoc-
proline in Step 2 (Scheme VI).


Preparation of 3-({2-[2-(4-Amino-3-chloro-
benzoylamino) -propionyl] -4-methyl-3,4-dihydro-2H-
pyrazole-3-carbonyl}-amino) -4-oxo-butyric acid (23g) .
Compound 23g was prepared according to the procedure
described for compounds 7 only substituting 4-methyl-
4 , 5-dihydro-pyrazole-l, 5-dicarboxylic acid 1-(9H-

fluoren-9-ylmethyl) ester for Ftnoc-proline (Scheme II)
in Step 2.
Preparation of 4-methyl-4, 5-dihydro-pyrazole-l, 5-
dicarboxylic acid 1- (9H-fluoren-9-ylmethyl) ester:
To a solution of 65 0 mg (2 mmol) of (10,10-dimethyl-
3,3-dioxo-λ6-thia-4-aza-tricyclo[5.2.1.0°'°]dec-4-yl) -
(4-methyl-3 , 4-dihydro-2H-pyrazol-3-yl) -methanone (J.
Am. Chem. Soc. , 119, pp. 8379-8380 (1997)) in 6 mL of
water and 14 mL of THF stirred at 0°C was added 420 mg
(10 mmol, 5 eq) of lithium hydroxide. The mixture was
stirred at 0°C for 2 hours and at room temperature for
3 0 minutes, diluted with 20 mL of water and washed with
ether (20 mL) . The pH of the solution was then
adjusted to 9, and a solution of 519 mg (2 mmol, 1 eq)
of Fmoc-Cl in 3 mL of dioxane was added. The mixture
was stirred at room temperature overnight, washed with
ether, acidified to pH 2-3 and extracted with 3 40-mL
portions of ethyl acetate. The combined organic
extracts were washed with brine, dried (MgSO4) and
concentrated in vacuo to afford 690 mg (98% yield) of a
colorless foam which was identified as the title
compound. 1H NMR (DMSO-d6, 500 MHz) δ 1.2 (d, 3H) , 3.2
(m, 1H) , 4.2-4.6 (m, 3H) , 7.1 (s, 1H) , 7.2-7.5 (m, 5H),
7.7-8.0 (m, 4H) . Exact mass calculated for C20H18N2O4
m/e 3 50.13, found m/e 351.3
Preparation of 3-({1- [2- (4-amino-3-chloro-
benzoylamino) -propionyl] -4-methoxy-pyrrolidine-2-
carbonyl}-amino) -4-oxo-butyric acid (23i) .

Compound 23i was prepared according to the procedure
described for compounds 7 only substituting N-Fmoc-4-
methoxyproline for Fmoc-proline (Scheme II) in Step 2.
Preparation of N-Fmoc-4-methoxyproline:
To a solution of 735 mg (3 mmol) of N-Boc-4-
hydroxyproline methyl ester in 2 0 mL of THF stirred at
0°C was added 79 mg (1.1 eq, 3.3 mmol) of 6 0% sodium
hydride in mineral oil. The mixture was stirred at 0°C
for 1 hour, and methyl iodide (0.56 mL, 3 eq, 9 mmol)
was added. The mixture was stirred at room temperature
overnight, quenched by addition of saturated aqueous
ammonium chloride, diluted with water, and extracted
with three 80raL portions of ethyl acetate. The
combined organic extracts were washed with brine, dried
(MgSO4) , and concentrated in vacuo to afford a pale
yellow oil. The oil was taken up in 9 mL of CH3OH and
3 mL of water, and 378 mg (3 eq, 9 mmol) of lithium
hydroxide was added. The mixture was stirred at room
temperature overnight, diluted with water, acidified to
pH 3 and extracted with three 80 mL portions of ethyl
acetate. The combined organic extracts were washed with
brine, dried (MgSO4) and concentrated in vacuo. The
residual oil was taken up in 10 mL of TFA and the
solution was stirred at room temperature for 2 hours,
and concentrated in vacuo. The residual oil was
diluted with 6 mL of 10% aqueous sodium carbonate and 3
mL of dioxane, and a solution of 9-fluorenylmethyl
chloroformate (779 mg, leq, 3 mmol) in 5 mL of dioxane
was added. The mixture was stirred at room temperature
overnight, diluted with water, acidified to pH 3 and
extracted with three 8 0 mL portions of ethyl acetate.
The combined organic extracts were washed with brine,
dried (MgSO4) and concentrated in vacuo to afford an

oil, which was purified by column chromatography over
silica gel eluted with CH2Cl2/ CH3OH 20:1, to afford 600
mg (55%) of N-Fmoc-4-methoxyproline: exact mass
calculated for C21H21NO5 m/e 367.14 found m/e 368.4.
To a 0.125 mmol portion of resin 2 was added 4 mL of
20% piperidine in DMF. The mixture was rotated at room
temperature for 5 minutes and drained. The procedure
was repeated over 2 0 minutes. The resin was washed
successively with DMF (twice), CH3OH (once), CH2Cl2
(twice), CH3OH (once) and NMP (twice). To a
suspension of the resin in 4 mL of NMP was added
successively 184 mg of N-Fmoc-4-methoxyproline (4 eq,
0.50 mmol), 0.175 mL of DIEA (8 eq, 1.00 mmol), 70 mg
of HOBt (4 eq, 0.50 mmol) and 188 mg of HBTU (4 eq,
0.50 mmol). The mixture was rotated at room
temperature overnight and drained. This coupling
procedure was repeated over 3 hours. The resin was
washed successively with DMF (twice), CH3OH (once), 1:1
DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2 (three
times), and dried in vacuo.
To the resin was added 4 mL of 20% piperidine in DMF.
The mixture was rotated at room temperature for 5
minutes and drained. The procedure was repeated over
20 minutes. The resin was washed successively with DMF
(twice), CH3OH (once), CH2Cl2 (twice), CH3OH (once) and
NMP (twice). To a suspension of the resin in 4 mL of
NMP was added successively 156 mg of N-Fmoc-alanine (4
eq, 0.5 0 mmol), 0.175 mL of DIEA (8 eq, 1.0 0 mmol), 70
mg of HOBt (4 eq, 0.50 mmol) and 188 mg of HBTU (4 eq,
0.5 0 mmol). The mixture was rotated at room
temperature overnight and drained. This coupling
procedure was repeated over 3 hours. The resin was

washed successively with DMF (twice), CH3OH (once), 1:1
DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2 (three
times), and dried in vacuo.
To the resin was added 4 mL of 2 0% piperidine in DMF.
The mixture was rotated at room temperature for 5
minutes and drained. The procedure was repeated over
20 minutes. The resin was washed successively with DMF
(twice), CH3OE (once), CH2Cl2 (twice), CH3OH (once) and
NMP (twice) . To a suspension of the resin in 4 mL of
NMP was added successively 80 mg of 4-amino-3-
chlorobenzoic acid (4 eq, 0.50 mmol), 0.175 mL of DIEA
(8 eq, 1.00 mmol), 70 mg of HOBt (4 eq, 0.50 mmol) and
188 mg of HBTU (4 eq, 0.50 mmol). The mixture was
rotated at room temperature overnight and drained. The
resin was washed successively with DMF (twice) , CH3OH
(once), 1:1 DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2
(three times) , and dried in vacuo.
The resin was treated with 4 mL of 95% TFA in water for
1 hour. The mixture was filtered. The filtrate was
concentrated in vacuo to afford an oil, which was
purified by HPLC to afford the title compound (23i) .


Procedure for the preparation of analogs 24a-e.
Compounds 24a-24e were prepared according to the
procedure described for compounds 5 (Scheme I) only
substituting either Fmoc-azetidine carboxylic acid or

trans-2-phenyl- Fmoc-azetidine carboxylic acid for
Fmoc-proline in Step 2.

Procedure for the preparation of analogs 25.
Compounds 25a-25e were prepared according to the
procedures described for compounds 5 and 7 (Scheme I
and Scheme II) only substituting Fmoc-2 (S) -pipecolic
acid for Fmoc-proline in Step 2 and coupling either
Fmoc-valine or Fmoc-alanine or Fmoc-tert-leucine in
Step 3 .


Procedure for the preparation of analogs 26a-h.
Compounds 26a- 26h were prepared according to the
procedure described for compounds 23 (Scheme VI) only
substituting Fmoc-valine for Fmoc-alanine in Step 3 .


Procedure for the preparation of analogs 27.

Compounds 27a- 27n were prepared according to the
procedure described for compounds 7 (Scheme II) only-
substituting Fmoc-4,4-difluoroproline for Fmoc-proline
in Step 2.
Preparation of N-Boc 4,4-difluoroproline methyl ester:
To a solution of 9.63 mL (7.2 mmol) of oxalyl chloride
in 10.6 mL of CH2Cl2 stirred at -78 °C was added a
solution of 0.94 mL (13.2 mmol) of methyl sulfoxide in
15 mL of CH2Cl2. The solution was stirred at -78 °C for
3 0 min. A solution of 1.4 7 g (6 mmol) of N-Boc-4-
hydroxyproline methyl ester in 19 mL of CH2Cl2 was then
added dropwise. The mixture was stirred at -78°C for
1.5 h, and 3.34 mL (24 mmol) of triethylamine was
added. The solution was allowed to warm up to room
temperature and stirred overnight. It was then diluted
with 100 mL of CH2Cl2/ washed successively with 100 mL
of water, 100 mL of IN HCl, and 100 mL of brine, dried
(MgSO4) and concentrated in vacuo. The residue was
purified by column chromatography over silica gel
(eluted with ethyl acetate/hexanes, 1:3), to afford
1.294 g (89% yield*) of N-Boc-4-oxo-proline methyl
ester. 1H NMR (500 MHz, CDCl3) δ 1.45 (m, 9H), 2.60 (m,
1H) , 2.95 (m, 1H) , 3.75 (m, 3H) , 3.90 (m, 2H), 4.80 (m,
1H) .
To a solution of 808 mg (3.33 mmol) of N-Boc-4-oxo-
proline methyl ester in 13 mL of CH2Cl2 stirred at 0°C
was added 0.88 mL (7.19 mmol, 2.2 eq) of DAST. The
mixture was stirred at 0°C for 2 hours, at room
temperature for 16 hours, and poured into ice water.
The mixture was stirred at room temperature for 2
hours. The organic phase was separated, washed with
water, dried (MgSO4) and concentrated in vacuo. The

residue was purified by column chromatography over
silica gel (eluted with ethyl acetate-hexanes, 1:8), to
afford 754 mg (79% yield) of difluorinated derivative
as a pale yellow oil. 1H NMR (500 MHz, CDCl3) 6 1.50 (m,
9H), 2.45 (m, 1H), 2.70 (m, 1H) , 3.75 (m, 3H), 3.80 (m,
2H), 4.50 (m, 1H).
Preparation of N-Fmoc-4,4-difluoroproline:
To a solution of 754 mg (2.85 mmol) of N-Boc 4,4-
difluoroproline methyl ester in 5 mL of THF stirred at
0°C was added a solution of 179 mg (4.2 7 mmol) of
lithium hydroxide in 5 mL of water. The solution was
stirred at 0°C for 3 h, at room temperature for 1 hour,
diluted with water, extracted with ether, acidified to
pH 2-3, and extracted with two 3 0 mL portions of ethyl
acetate. The combined organic extracts were washed
with brine, dried (MgSO4) and concentrated in vacuo to
afford 652 mg (91%) of acid as a pale yellow solid.
A solution of 652 mg (2 mmol) of N-Boc-4,4-
difluoroproline in 10 mL of 1;1 TFA/ CH2Cl2 was stirred
at OoC for 45 rain, and concentrated in vacuo. The
residue was taken up in 3 mL of dioxane, and 5 mL of
10% aqueous sodium carbonate was added, followed by a
solution of 675 mg (1 eq) of Fmoc-Cl in 5 mL of
dioxane. The mixture was stirred at room temperature
for 16 h, diluted with 20 mL of water, extracted with 2
20-mL portions of diethyl ether, acidified to pH 2, and
extracted with three 30-mL portions of ethyl acetate.
The combined organic extracts were washed with 50 mL of
brine, dried (MgSO4) and concentrated in vacuo. The
residue was purified by column chromatography over
silica gel (eluted with CH2Cl2/ CH3OH 10:1), to afford
850 mg (88%) of N-Fmoc-4,4-difluoroproline as a

brownish solid. 1H NMR (500 MHz, CDCl3) δ 2.55 (m, 1H) ,
2.95 (m, 1H), 3.80 (m, 2H) , 4.20 (m, 1H), 4.30 (m, 2H),
4.55 (m, 1H), 7.32 (m, 2H), 7.45 (m, 2H) , 7.70 (m, 2H),
7.90 (m, 2H). Exact mass calculated for C20H17F2NO4 m/e
373.11, found m/e 374.4.



Compounds 28a- 28c were prepared according to the
procedure described for compounds 5 and 7 (Scheme I and
Scheme II) only substituting Pmoc-dimethylthioproline
for Fmoc-proline in Step 2.

GENERAL PROCEDURES FOR THE PREPARATION OF COMPOUNDS OF
EMBODIMENT A FORMULA I (SCHEMES VII-VIII)
Compounds of Embodiment A Formula I where R4=H and one
R5=H:
Procedure for the preparation of analogs 29.
Compounds 29a- 29s were prepared according to the
procedure described for compounds 5 (Scheme I) only
substituting Fmoc-alanine for Fmoc-proline in Step 2
and using either Fmoc-valine or Fmoc-alanine or Fmoc-
tert-leucine in Step 3.




Procedure for the preparation of analogs 32.
Compounds 32a-32e were prepared according to the
procedure described for compounds 5 (Scheme I) only
substituting 2- (3-tert-butoxycarbonylamino-2-oxo-
pyrrolidin-1-yl)-4-methyl-pentanoic acid (30)
(Neosystem catalog number BB02101)for Fmoc-proline in
Step 2 followed by Step 4 (Scheme VII).

Compounds of Embodiment A Formula I wherein R2 and R3
together with the atoms to which they are bound form a
5 membered ring.



Compound of Embodiment A Formula I wherein X=N-CH3
i



was washed successively with DMF (twice), CH3OH . (once),
1:1 DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2 (three
times), and dried in vacuo.
To a 0.075 mmol portion of the resin in 3 mL of NMP was
added successively 52 mg of 1-isoquinolinecarboxylic
acid (4 eq, 0.3 mmol), 0.19 mL of DIEA (8 eq, 0.6
mmol), 37mg of HOBt (4 eq, 0.3 mmol) and 104 mg of HBTU
(4 eq, 0.3 mmol) . The mixture was rotated at room
temperature overnight and drained. The resin was
washed successively with DMF (twice), CH3OH (once), 1:1
DMF/ CH2Cl2 (twice), CH3OH (once) and CH2Cl2 (three
times), and dried in vacuo.
The resin was treated with 4 mL of 95% TFA in water for
1 hour. The mixture was filtered. The filtrate was
concentrated in vacuo to afford an oil, which was
purified by. HPLC to afford the title compound (34).
Compounds of Embodiment A Formula I wherein R3= R3= H:

3- ({1-[ (4-Amino-3-chloro-benzoylamino) -acetyl] -
pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric acid (Gl) .
Prepared as described for compounds 7 only substituting
Fmoc-glycine for Fmoc-alanine in Step 3 (Scheme II) to
afford 4.3mg of the title compound. LC-MS (ES+)
m/e=425.2 (M+H)


3-({l-[(4-Amino-3-chloro-benzoylamino)-acetyl]-4,4-
dif luoro -pyrrolidine - 2 - carbony 1} - amino) - 4 - oxo-butyric
acid (G2).
Prepared as described for compounds 7 and 27 only
substituting Fmoc-glycine for Fmoc-alanine in Step 3
(Scheme II) to afford lO.Omg of the title compound. LC-
MS (ES+) m/e=461.2 (M+H)

GENERAL PROCEDURES FOR THE PREPARATION OF COMPOUNDS OF
EMBODIMENT C FORMULA I AND EMBODIMENT D FORMULA I
WHEREIN Y= C (SCHEMES IX-XXII)






5- tert-Butyl-3-[2-(9H-fluoren-9-
ylmethoxycarbonylamino) -3-methyl butyryl] -2, 3-dihydro-
[1,3,4]thiadiazole-2-carboxylic acid ethyl ester (37).
A stirred suspension of polyvinylpyridine (2.63g,
25mmol) in a solution of 5- tert-butyl-2,3-dihydro-
[1,3,4]thiadiazole-2-carboxylic acid ethyl ester (36),
(J. Med. Chem., 34, p. 439 (1991)), (2.16g, lOmmol) in
dry toluene was treated with the dropwise addition of
(i-chlorocarbonyl-2-methyl-propyl) -carbamic acid 9H-
fluoren-9-ylmethyl ester (4.76g, 12.1mmol) in 20mL of
anhydrous toluene. After stirring for 16 hours, the
suspension was filtered and the filtrate was washed
with saturated sodium bicarbonate solution. The
organic layer was separated, washed with water, dried
over anhydrous sodium sulfate, and evaporated to give a
yellow oil. Purification by flash chromatography
eluting with 9/1 hexane/ ethyl acetate gave 2.66 g (49%
yield) of the title compound (37) as a clear, viscous
oil. 1H NMR (500MHz, CD3OD) δ 0.89(d, 1.5H), 0.93 (d,
1.5H), 1.00 (d, 1.5H), 1.06 (d, 1.5H), 1.22 (t, 3H) ,
1.28, (s, 9H), 2.12-2.22 (m, 0.5H), 2.32-2.42 (m, 0.5H),
4.18-4.28 (m, 2H) , 4.31-4.45 (m, 2H) , 4.96-5.01 (m,
0.5H), 5.02-5.10 (m, 0.5H), 5.52 (d, 0.5H), 5.61 (d,
0.5H), 6.10 (s, 0.5H), 6.13 (s, 0.5H), 7.27-7.34 (m,
2H) , 7.35-7.42 (m, 2H) , 7.56-7.64 (m, 2H) , 7.73-7.78
(m, 2H) .
3- (2-Acetylamino-3-methylbutyryl) -5- tert-butyl-2,3-
dihydro- [1,2,4] thiadiazole-2-carboxylic acid ethyl
ester (38).
To a solution of (37) (Scheme IX) (0.508g, 0.94 mmol)in
CH3CN (10 mL) was added diethylamine (1 mL) . The
solution was stirred at room temperature for 2 hours,

the solvent removed in vacuo and the resultant oil
azeotroped with CH2Cl2 (4.x) . The crude oil was
dissolved in CH;Cl2 (5 mL) and triethylamine (0.2 6mL,
1.86mmol) and acetyl chloride (80ul, l.lmmol) were
added. The solution was stirred at room temperature
under an N2 atmosphere for 2 hours. The solvent was
evaporated, and the crude material dissolved in EtOAc
and washed with 0.5N NaHS04 (2x), saturated NaHCO3 (2x)
and brine and was dried over anhydrous Na2SO4, filtered
and evaporated to give a yellow oil. Purification by
flash column chromatography on silica gel using
hexanes/EtOAc {95/5 to 90/10%) yielded the product as a
yellow oil (0.301g, 89% yield). 1H-NMR (500MHz, CDCl3)
δ 0.88 (dd, 3H), 0.99 (dd, 3H), 1.16-1.45 (m, 12H),
2.02 (s, 3H), 2.09-2.19 (m, 0.5H), 2.30-2.40 (m, 0.5H),
4.12-4.29 (m, 2H), 5.20-5.27 (m, 0.5H), 5.30-5.36 (m,
0.5H), 6.60(s, 0.5H), 6.90 (s, 0.5H), 6.20-6.31 (m,
1H) . Analytical HPLC (Cl8 column), (mixture of
diastereomers) 7.77, 7.98min. LC-MS (ES+) m/e=358.3
(M+H) .
3- (2-Acetylamino-3-methylbutyryl) -5- tert-butyl-2 , 3-
dihydro- [1,2,4] thiadiazole-2-carboxylic acid (39).
To a solution of 38 (0.301g, 0.84mmol) in MeOH (10mL)
was added 1N NaOH solution (l.7mL, l.7mmol). The
reaction was stirred at room temperature for 2 hours
and solvent was evaporated. The residue was dissolved
in EtOAc and washed with 0. 5N NaHSO4 (2x) and brine and
was dried over anhydrous Na2SO4, filtered and
evaporated to give the title compound as a yellow solid
(0.277g, quantitative).
Preparation of 2- (Benzyloxy-5-oxo-tetrahydro-furan-3-
yl)-carbamic acid allyl ester (40).

Compound 4 0 was prepared from 3-allyloxycarbonylamino-
4-hydroxy-butyric acid tert-butyl ester by a
modification of the procedure described in Bioorg. Med.
Chem. Lett. Vol. 2, No. 6, pp. 613-618, (1992).
To a solution of DMSO (27.52 g, 352 mmol) in CH2Cl2
(24 0 mL) at -78°C was added oxalyl chloride (24.4 g,
192 mmol) .. After 15 min, a solution of 3-
allyloxycarbonylamino-4-hydroxy-butyric acid tert-butyl
ester (41.44 g, 160 mmol) in CH2Cl2 (100 mL) was slowly
added and the mixture was stirred at -78°C for an
additional 1.5 hours. DIEA (62.0 g, 480 mmol) was
added and the mixture allowed to warm to room
temperature for 15 min. The resulting solution was
diluted with CH2Cl2 (300 mL) , washed with 0.5 N NaHS04
(500 mL x 2), water (300 mL x 2), and brine (400 mL x
2) . The organic layer was dried over anhydrous Na2S04,
filtered and concentrated in vacuo to 200mL volume. To
this solution was added, benzyl alcohol (48 g, 444
mmol), followed by 3A molecular sieves (30 g) and p-
toluenesufonic acid (0.8 g). The reaction mixture was
allowed to stir for 4 days and TFA (96 mL) was added.
The resulting suspension was stirred for one hour then
evaporated in vacuo. Ethyl acetate (500 mL) was added
and the mixture was filtered through Celite. The
filtrate was washed with saturated NaHCO3 (500 mL x 2),
water (400 mL x 2) , and brine (300 mL x 2) . The
organic solution was dried over anhydrous Na2SO4,
filtered and evaporated in vacuo to give 90 g of pale
yellow oil, which was stirred with hexane (400 mL x 2)
to give 31 g of crude product from the lower layer
residue. Chromatography using ethyl acetate/hexane
(4/96 to 22/78) afforded 6.97 g of anti-2- (benzyloxy-5-
oxo-tetrahydro-furan-3-yl)-carbamic acid allyl ester

(higher Rf), 4.53 g of syn diastereomer and 12.97 g of
the mixture of the diastereomers (overall yield 53%) .
1H-NMR (500 MHz, CDCl3) for anti diastereomer: 5 2.41-
2.45 (m, H), 3.02-3.07 (m, H), 4.28 (br, H), 4.50-4.80
(m, 3H), 4.80-5.15 (m, 2H) , 5.24-5.32 (m, 2H), 5.48 (s,
H) , 5.88-6.00 (m, H) , 7.31-7.56 (m, 5H) ; for syn
diastereomer: 82.49-2.53 (m, H) , 2.83-2.89 (m, H),
4.57-4.65 (m, 4H), 4.87-4.90 (m, H) , 5.12-5.30 (m, 3H) ,
5.52-5.53 (d, H) , 5.88-6.00 (m, H) , 7.31-7.39 (m, 5H) ;
retention time on analytical HPLC: 10.49 min for anti
diastereomer and 10.3 7 min for syn diastereomer; LC-MS:
m/z = 292 (M+H+) .
3- (2-Acetylamino-3-methylbutyryl) -5- tert-butyl-2, 3-
dihydro-[1,2,4] thiadiazole-2-carboxylic acid (2-
benzyloxy-5-oxo-tetrahydrofuran-3-yl)-amide (41) .
To a solution of (2-benzyloxy-5-oxo-te.trahydrofuran-3-
yl)-carbamic acid allyl ester (40) (0.385g, 1.32mmol)
in DMF (2ml) and CH2Cl2 (2ml) was added DMBA (0.456g,
2.92mmol) and Pd(PPh3)4 (0.136g, 0.12mmol) and the
solution was stirred at room temperature for 15min. A
solution of (39) in CH2Cl2 (4.5ml) and DMF (0.5ml) was
added, followed by HOBT (0.168g, 1.24mmol) and EDC
(0.256g, 1.33mmol). The reaction was stirred at room
temperature for 18hours under N2. The solvent was
evaporated. The crude material was dissolved in EtOAc
and washed with 0.5N NaHS04 (2x), saturated NaHC03 (2x)
and brine and was dried over anhydrous Na2SO4, filtered
and evaporated to give a yellow solid. Purification by
flash column chromatography gave the title compound
(41) as a mixture of diastereomers (374mg, 88% yield) .
1H-NMR (500MHz, CDCl3) δ 0.75-1.05 (m, 6H) , 1.19-1.34
(m, 9H) , 1.93-2.08 (m, 3H) , 2.19-2.50 (m, 2H) , 2.80-

3.03 (m, 1H), 4.56-4.93 (m, 3H) , 5.02-5.20 (m, 1H) ,
5.46-5.56 (m, 1H) , 5.95-6.16 (m, 2H) , 6.86-6.95 (m,
1H) , 7.20-7.43 (m, 5H) . Analytical HPLC (Cl8
column), (mixture of diastereomers) 8.58min. LC-MS
(ES+) m/e=519.2 (M+H).
Preparation of 3-{ [3-(2-acetylamino-3-methyl-butyryl) -
5- tert-butyl-2, 3-dihydro- [1,3,4] th.iadiazole-2-
carbonyl]-amino}-4-oxo-butyric acid (42).
A 45 mg (0.087mmol) sample of 41 was hydrolyzed
according to method A (see Scheme XXIII) to give 17 mg
(45% yield) of the title compound. Analytical HPLC
(Cl8 column): 5.15min. LC-MS (ES+) m/e=429.3 (M+H).
5-tert-Butyl-3- [2- (4-methoxy-benzoylamino) -3-methyl-
butyryl] -2,3-dihydro- [1,3,4] thiadiazole-2-carboxylic
acid ethyl ester (43) . .
Was prepared by the method reported above for compound
3 8 using anisoyl chloride to give 216mg (50%) of the
title compound as an amorphous solid. 1H NMR (500 MHz,
CDCl3) δ 0.92 (d, 1.5H), 0.98 (d, 1.5H), 1.03 (d, 1.5H),
1.07 (d, 1.5H), 1.21 (t, 3H) , 1.28 (s, (H) , 2.21-2.28
(m, 0.5H), 2.41-2.48' (m, 0.5H), 3.83 (s, 3H) , 4.15-4.28
(m, 2H) , 5.41-5.46 (m, 0.5H), 5.48-5.53 (m, 0.5H), 6.08
(S, 0.5H), 6.13 (s, 0.5H), 6.75 (d, 0.5H), 6.85 (d,
0.5H), 6.91 (d, 2H) , 7.59 (d, 2H) .
5-tert-Butyl-3- [2- (4-methoxy-benzoylamino) -3-methyl-
butyryl] -2,3-dihydro- [1,3,4] thiadiazole-2-carboxylic
acid (44).
Prepared by the procedure described for 3 9 to give
180mg (quantitative) of the title compound as a white
solid. 1H NMR (500 MHz, CDCl3) δ 0.92 (d, 1.5H), 0.96
(d, 1.5H), 1.03 (d, 1.5H), 1.07 (d, 1.5H), 2.22-2.30

(m, 0.5H), 2.37-2.45 (m, 0.5H), 3.83 (s, 1.5H) 3.84 (s,
1.5H), 5.41-5.48 (m, 1H) , 6.14 (s, 0.5H), 6.15 (s,
0.5H), 6.87-6.95 (m, 2H), 7.75-7.83 (m, 3H).
5-tert-Butyl-3- [2- (4-methoxy-benzoylami.no) -3-methyl-
butyryl]-2, 3 -dihydro-[1,3,4]thiadiazole-2-carboxylic
acid (2-benzyloxy-5-oxo-tetrahydro-furan-3-yl)-amide
(45a and 45b).
Was prepared by the procedure reported for compound 41
to give the crude title compound as 4 diastereomers.
The crude material was purified by flash
chromatography, eluting with a gradient of CH2Cl2 to
CH2Cl2/ethyl acetate (6/4) to give 31mg of the higher Rf
component as a single diastereomer (45a). Analytical
HPLC (Microsorb Cl8 column) 19.87 min. 1H NMR (500
MHz, CDCl3) (single diastereomer) δ 1.04 (d, 3H), 1.14 (d,
3H), 1.28 (s, 9H), 2.77 (d, 0.5H), 2.81 (d, 0.5H), 2.90
(d, 0.5H), 2.95 (d, 0.5H), 3.84 (s, 3H), 4.44-4.49 (m,
1H), 4.53 (d, 1H), 4.85 (d, 1H), 5.02-5.08 (m, 1H),
6.37 (s, 1H) , 6.41 (d, 1H) , 6.93 (d, 2H) , 7.26-7.40 (m,
5H), 7.75 (d, 2H), 7.92-7.96 (m, 1H).
The lower Rf fraction contained 185mg of a solid as a
3:1:2 mixture of diastereomers (45b). Analytical HPLC:
Microsorb Cl8 column. 19.00, 19.26, 20.02 mins, 1H NMR
(500 MHz, CDCl3) (3:1:2 mixture of 3 diastereomers)
δ 0.89 (d, 2.25 H), 0.98 (d, 0.75H), 1.02 (d, 0.5H),
1.03 (d, 1.5H), 1.08 (d, 0.25H), 1.10 (d, 0.75H), 1.16
(s, 0.75H), 1.17 (S, 2.25H), 1.23 (s, 0.375H), 1.24 (s,
1.125H), 1.28 (s, 1.125 H), 1.29 (s, 3.375H), 2.12-2.18
(m, 0.33H), 2.32-2.42 (m, 0.67H), 2.43-2.51 (m, 0.5H),
2.61-2.67 (m, 0.5H), 2.84-2.92 (m, 0.5H), 2.96-3.07 (m,
0.5H), 3.85 (s, 3H), 4.58-4.71 (m, 2H), 4.81 (d,

0.16H), 4.86 (d, 0.32H), 4.91 (d, 0.52H), 5.09-5.13 (m,
0.33H), 5.14-5.18 (m, 0.67H), 5.35 (dd, 1H) , 5.46 (s,
0.16H), 5.53 (d, 0.32H), 5.58-5.62 (d, 0.52H), 6.17 (s,
0.52H), 6.20 (s, 0.16H), 6.34 (s, 0.32H), 6.50 (d,
0.32H), 6.62 (d, 0.16H), 6.67 (d, 0.52H), 6.86 (d,
0.33H), 6.91 (d, 0.67H), 6.94 (d, 1.0H), 7.24-7.43
(m, 5H), 7.61 (d, 1H), 7.70 (d; 0.33H), 7.71 (d,
0.67H), 7.76 (d, 1H).
Preparation of 3-({5-tert-butyl-3-[2-(4-methoxy-
benzoylamino) -3-methyl-butyryl] -2,3-dihydro-
[1,3,4]thiadiazole-2-carbonyl}-amino)-4-oxo-butyric
acid (46a) .
A 30mg sample of 45a was hydrolyzed according to method
B (see Scheme XXIII) to give 8mg (30% yield) of the
desired product. Analytical HPLC (Microsorb C-18
column, acetonitrile/ water, with TFA buffer) I2.85min,
1H NMR (500 MHZ, CD3OD) δ 0.98-1.1 (m, 6H), 1.28 (s,
9H), 2.20-2.31 (m, 1H), 2.40-2.48 (m, 1H), 2.6-2.72 (m,
1H), 3.84 (s, 3H), 4.18-4.26 (m, 1H), 4.56-4.62 (m,
1H), 5.25-5.32 (m, 1H) , 6.24-6.28 (m, 1H) , 6.98 (d,
2H), 7.85 (d, 2H).
Preparation of 3-({5-tert-butyl-3-[2-(4-methoxy-
benzoylamino) -3-methyl-butyryl] -2,3-dihydro-
[1,3,4] thiadiazole-2-carbonyl}-amino) -4-oxo-butyric
acid (46b).
A 3 0mg sample of 45b (mixture of 3 diastereomers) was
hydrolyzed according to method B (see Scheme XXIII) to
give 22mg (84% yield) of the desired product as a 3:2
mixture of diastereomers. Analytical HPLC (Microsorb
cyano column) 7.08, 7.78min. 1H NMR (500MHz, CD3OD) δ
0.98-1.08 (m, 4H), 1.09-1.12 (m, 2H), 1.29 & 1.31 (2
singlets, 9H), 2.23-2.30 (m, 0.5H), 2.36-2.55 (m,

1.5H), 2.62-2.72 (m, 1H) , 3.85 (s, 3H) , 4.18-4.27 (m,
1H) , 4.58-4.65 (m, 1H) , 5.27-5.33 (m, 1H) , 6.23-6.27
(m, 1H) , 7.00 (d, 2H), 7.70-7.88 (m, 2H) .

1- (2-Benzyloxycarbonylamino-2-inethyl-propionyl) -
pyrrolidine-2-carboxylic acid tert-butyl ester (49) .
To a solution of proline-tert-butyl ester (47) (2.00g,
12mmol, in CH2Cl2 (15ml) was added N-carbobenzyloxy-2-
methylalanine (3.05g, 13mmol) , HOBT (2.36g, 17ramol) and
EDC (3.43g, l8mmol) and the solution was stirred at
room temperature under N2 for 4 8hours. The solvent was
evaporated, the crude material dissolved in EtOAc and

washed with 0.5N NaHSO4 (2x), saturated NaHC03 (2x) and
brine and was dried over anhydrous Na2SO4, filtered and
evaporated to give a white solid (4.68g, 100%). 1H-NMR
(500MHz, CDCl3) δ 1.20-2.15 (m, 4H) , 1.43 (s, 9H) , 1.59
(d, 6H) , 3.21-3.79 (m, 2H), 4.35 (br s, 1H), 4.82-5.19
(m, 3H), 5.74 (br s, 1H), 7.17-7.49 (m, 5H). Analytical
HPLC (Cl8 column) 10.66min. LC-MS (ES+) tn/e= 391.3
(M+H) .
1- [2- (4-Methoxy-benzoylamino) -2-methyl-propionyl] -
pyrrolidine-2-carboxylic acid tert-butyl ester (50) .
To solution of 4 9 (l.OOg, 2.56mmol) in MeOH (2 0ml) was
added 10% Pd/C (200mg) and the mixture was stirred
under H2 for 2 hours. The mixture was filtered through
a 0.4 5µm PTFE filter and the solvent removed in vacuo
to yield a colorless oil. This oil was dissolved in
CH2Cl2 (25mL) and DIEA (660µl, 3.79mmol) and p-anisoyl
chloride (4 80mg, 2.8mmol) were added. The solution was
stirred at room temperature under N2 for 18hours. The
solvent was removed in vacuo and the oil dissolved in
EtOAc. The organic phase was washed with 0.5N NaHS04
(2x), water, saturated NaHCO3 (2x) and brine. The
organic phase was dried over Na2SO4, filtered and
evaporated to give a white solid which was purified by
flash column chromatography, eluting with CH2Cl2/MeOH
(99/1 to 98/2%) to give the title compound as a white
solid (655mg, 65% yield). 1H-NMR (500MHz, CDCl3) δ 1.47
(S, 9H), 1.68-2.24 (m, 5H)1, 1.80 (d, 6H), 3.55-3.68 (m,
1H), 3.72-3.93 (m, 1H), 3.84 (s, 3H), 4.43-4.55 (m,
1H), 6.90 (d, 2H) , 7.60 (br s, 1H) , 7.77 (d, 2H) .
Analytical HPLC (Cl8 column)8.98min.

1- [2- (4-Methoxy-benzoylamino) -2-methyl-propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (51).
To a solution of 50 (325mg, 0.83mmol) in dioxane (5mL)
was added triethylamine (463ul, 3.32mmol) and TMS-
triflate (642µl, 3.32mmol) and the solution was stirred
at 100°C for 5 hours, then at room temperature for 18
hours. The reaction was diluted with water, adjusted
to pH 8 with saturated NaHCO3 and extracted with Et2O,
dried over Na2SO4, filtered and evaporated to give a
white solid (230mg, 83% yield) which was used directly
in the next step.
To a solution of (2-benzyloxy-5-oxo-tetrahydrofuran-3-
yl)-carbamic acid allyl ester (40) (1.027g, 3.5mmol) in
CH2Cl2 (20ml) was added DMBA (543mg, 3.48mmol) and
Pd(PPh3)4 (280mg, 0.24mmol) and the solution was stirred
at room temperature under N2 for 2 0 minutes. A solution
of l- [2- (4-methoxy-benzoylamino) -2-methyl-propionyl] -
pyrrolidine-2-carboxylic acid (818mg, 2.45mmol) in
CH2Cl2 (5ml) was added, followed by HOBT (0.534g,
3.95mmol) and EDC (738mg, 3.84mmol). The reaction was
stirred at room temperature for 18 hours under N2. The
solvent was evaporated, the crude material dissolved in
EtOAc and washed with 0.5N NaHSO4 (2x) , saturated
NaHCO3 (2x) and brine and was dried over anhydrous
Na2SO4, filtered and evaporated to give a yellow solid.
Purification by flash column chromatography, eluting
with ethyl acetate/hexanes (20/80 to 50/50%), gave the
product as pale yellow solid (760mg, 61% yield) . 1H-
NMR (500MHZ, CD3OD) δ 1.53 (d, 6H) , 1.65-1.93 (m, 3H) ,
1.96-2.14 (m, 1H), 2.60 (dd, 0.1H), 2.77 (dd, 0.85H),
2.94 (dd, 0.85H), 3.04-3.11 (m, 0.2H), 3.42-3.52 (m,
1H), 3.57-3.67 (m, 1H), 3.84 (s, 3H), 4.38-4.76 (m,

3H) , 4.84 (d, 1H) , 5.64-5.70 (m, 1H), 6.96-7.03 (m,
2H) , 7.23-7.43 (m, 5H), 7.78-7.97 (m, 2H) . Analytical
HPLC (Cl8 column) 13.32, 14.37min. LC-MS (ES+)
m/e=524.3 (M+H).
Preparation of 3- ({1- [2- (4-methoxy-benzoylamino) -2-
methyl-propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-
butyric acid (52) .
A 61mg (0.l4mmole) sample of 51 was hydrolyzed
according to method C (see Scheme XXIII) to afford 30mg
(60% yield) of the title compound: Analytical HPLC (Cl8
column) 6.79min. LC-MS (ES+) m/e=434.3 (M+H).


1- [2- (4-Methoxy-benzoylamlno) -3-mathylbutyryl] -
pyrrolidine-2-carboxylic acid tert-butyl ester (54) .
To a suspension of H-val-pro-OtBu-HCl (53) (2.011g,
7.44mmol) in CH2Cl2 (20ml) was added DIEA (3.2ml,
18.4mmol) followed by a solution of 4-methoxy-benzoyl
chloride (1.26g, 7.4mmol) in CH2Cl2 (5ml). The solution
was stirred at room temperature under nitrogen for
lhour then concentrated. The resulting oil was
dissolved in EtOAc and washed with 0.5N KHSO4 (2x),

saturated NaHCO3 (2x) and brine, then concentrated in
vacuo to give the title compound as a white solid
(2.814g, 94% yield). 1H-NMR (500MHz, CDCl3) δ 1.05 (dd,
6H) , 1.46 (s, 9H) , 1.88-2.29 (m, 5H) , 3.65-3.74 (m,
1H) , 3.81-3.92 (m, 1H) , 3.85 (s, 3H) , 4.32-4.42 (m,
1H) , 4.81-4.91 (m, 1H) , 6.79-6.86 (m, 1H) , 6.91 (d,
2H) , 7.78 (d, 2H) . Analytical HPLC (cyano column)
10.I8min.
1- [2- (4-Methoxy-benzoylamino) -3-methylbutyxyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydrofuran-3-yl) amide (56) .
A 1.079g (2.67mmol) sample of 54 was dissolved m 15%
TFA in CH2Cl2 (4 0mL) and stirred at room temperature
for 4hours. The solvent was concentrated in vacuo to
give 55 as a white solid (0.93g, 100%) which was used
in the next step.
To a solution of 4 0 (l.796g, 6.17mmol) in CH2Cl2 (2 0ml)
was added DMBA (1.119g, 7.17mmol) and Pd(PPh3)4 (0.683g,
0.59mmol) and the solution stirred at room temperature
for 20. minutes. A solution of 55 (0.928g, 2.67mmol) in
CH2Cl2 (17ml) and DMF (2ml) was added, followed by HOBT
(0.8llg, 6.01mmol) and EDC (1.16g, 6.04mmol). The
reaction was stirred at room temperature for 18hours
under N2. The solvent was evaporated, the crude
material dissolved in EtOAc and washed with 0. 5N NaHS04
(2x) , saturated NaHCO3 (2x) and brine and was dried
over anhydrous Na2SO4, filtered and evaporated to give
a yellow solid. Purification by flash chromatography
eluting with ethyl acetate/ CH2Cl2 (10/90 to 40/60%)
gave the title compound as pale yellow solid (910mg,
63% yield). 1H-NMR (500MHz, CDCl3) δ 0.96 (dd, 6H) ,
1.84-2.19 (m, 4H) , 2.25-2.38 (m, 1H) , 2.45 (dd, 1H) ,

2.80-2.98 (m, 1H), 3.60-3.72 (m, 1H), 3.82-3.95 (m,
1H) , 3.86 (s, 3H) , 4.26-4.95 (m, 6H) , 5.41(s, 0.2H),
5.53 (d, 0.8H), 6.67-6.77 (m, 1H), 6.88-6.99 (d, 2H),
7.22-7.57 (m, 5H), 7.71-7.82 (d, 2H) .
Analytical HPLC (cyano column)(mixture of 2
diastereomers) 9.2lmin. LC-MS (ES+) m/e= 538.3 (M+H).
3- ({l- [2- (4-Methoxy-benzoylamino) -3-methyl-butyryl] -
pyrrolidine-2-carbonyl}-amino)-4-oxo-butyric acid (57).
A 125mg (0.23mmol) sample of 56 was hydrolyzed
according to method A (see Scheme XXIII) to afford 60mg
(58% yield) of the title compound: Analytical HPLC
5.71min. LC-MS (ES+) m/e=448.2 (M+H).
Preparation of 4-Amino-3-chloro-benzoic acid:
A suspension of 4-amino-3-chlorobenzonitrile (4.82g,
3l.58tnmol) was heated to reflux in 6N HCl (140ml) . The
precipitate dissolved upon heating to give a colorless
solution. Upon further heating the solution became
cloudy. After 9hours the reaction was cooled to room
temperature. The resulting precipitate was filtered,
then'dissolved in THF and the solvent evaporated. The
residue was repeatedly concentrated from toluene to
give a white solid (3.18g, 59% yield). 1H-NMR (500MHz,
CD3OD:CDCl3 1:4) δ 6.80 (d, 1H) , 7.75 (dd, 1H) , 7.94 (d,
1H). Analytical HPLC (cyano column) 8.73min.
1- [2- (4-Amino-3-chloro-benzoylamino) -3-methylbutyryl] -
pyrrolidine - 2-carbbxylic acid tert-butyl ester (58) .
To a suspension of 53 (1.707g, 6.3lmmol) in CH2Cl2
(25ml) at 0°C was added DIEA (3.2ml, 18.4mmol) followed
by a solution of 4-amino-3-chlorobenzoic acid (1.298g,
7.56mmol), HOBT (1.005g, 7.44mmol) and EDC (1.456g,
7.58mmol) . The resulting mixture was stirred at 0'C



Analytical HPLC (cyano column) (mixture of 2
diastereomers) 14.90, I5.20min. LC-MS (ES+) m/e=557.2
(M+H).
3- ({1- [2- (4-Amino-3-chloro-benzoylamino) -3-methyl-
butyryl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (61).
A 4 5mg (0.08mmol) sample of 60 was hydrolyzed according
to method A (see Scheme XXIII) to afford 30mg (80%
yield) of the title compound: 1H NMR (500MHz, CD3OD) δ
1.06 (dd, 6H) , 1.78-2.38 (m, 5H) , 2.38-2.86 (m, 2H) ,
3.62-3.83 (m, 1H), 4.12-4.76 (m, 4H), 7.04-7.21 (m,
1H), 7.58-8.01 (m, 2H) ; Analytical HPLC 8.16min. LC-MS
(ES+) m/e=467.3 (M+H).

1- [2- (4-Hydroxy-3, 5-dimethyl-benzoylamino) -3-methyl-
butyryl]-pyrrolidine-2-carboxylic acid tert-butyl ester
(63).
To a solution of 62 (prepared from 53 and Fmoc-Cl)
(60 0mg, 1.22mmol) in anhydrous DMF (10ml) was added

diethylamine (3ml). The solution was stirred at room
temperature under N2 for 3hours and the solvent was
evaporated. The resulting oil was dissolved in CH2Cl2
(8ml) and 3 , 5-dimethyl-4-hydroxybenzoic acid (0.302g,
1.82mmol), HOBT (338mg, 2.5mmol) and EDC (0.456g,
2.43mmol) were added and the solution stirred at room
temperature under N2 for 18hours. The solvent was
concentrated in vacuo and the resulting oil dissolved
in EtOAc, washed with 0.5N NaHS04 (2x) , saturated
NaHCO3 (2x) and brine to give the crude product as a
white solid (0.80g). Flash chromatography eluting with
MeOH/CH2Cl2 (1/99 to 2/98%) gave 380mg (75% yield) of a
white solid. 1H-NMR (500MHz, CDCl3) δ 1.06 (dd, 6H) ,
1.47 (s, 9H) , 1.90-2.32 (m, 5H) , 2.24 (s, 6H) , 3.65-
3.75 (m, 1H), 3.84-3.92 (m, 1H) , 4.36-4.42 (m, 1H) ,
4.82-4.88 (m, 1H), 5.53-5.61 (m, 1H), 6.77-6.85 (m,
1H) , 7.42 (s, 2H) . Analytical HPLC (cyano column)
17.53min. LC-MS (ES+) m/e=419.3 (M+H).
1- [2- (4-Hydroxy-3,5-dimethyl-benzoylamino) -3-methyl-
butyryl]-pyrrolidine-2-carboxylie acid (2-benzyloxy-5-
oxo-tetrahydro-furan-3-yl)-amide (64).
1
Prepared from 63 and 40 by the method used to prepare
56 to give title compound (64) as a pale yellow solid
(352mg, 72% yield). 1H-NMR (500MHz, CD3OD) δ 0.83-1.28
(m, 6H), 1.66-2.37 (m, 3H), 2.23 (s, 6H), 2.48-2.54 (m,
0.2H), 2.61 (ddd, 0.8H), 2.72 (ddd, 0.9H), 3.01-3.09
(m, 1H), 3.66-3.76 (m, 1H), 3.95-4.07 (m, 1H), 4.48-
4.73 (m, 3H) , 4.75-4.92 (m, 1H) , 5.45-5.48 (m, 0.1H) ,
5.61-5.64 (m, 0.1H), 5.64-5.70 (m, 0.8H), 7.21-7.62 (m,
6H), 7.88-8.04 (m, 1H) . Analytical HPLC (cyano
column)(mixture of 2 diastereomers) 17.73min. LC-MS
(ES+) m/e= 552.3(M+H).

3- ({1- [2- (4-Hydroxy-3, 5-dimethyl-benzoylamino) -3-
methyl-butyryl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-
butyric acid (65).
A 160mg (0.29mmol) sample of 64 was hydrolyzed
according to method A (see Scheme XXIII) to afford
13.1mg (10% yield) of the title compound: Analytical
HPLC (cyano column) 10.28min. LC-MS (ES+) m/e=462.2
(M+H).

1- [2- (2-9H-Fluoren-9-yl-acetylamino) -3 , 3 -dimethy 1-
butyryl] -pyrrolidine-2-carboxylic acid tert-butyl ester
(66) .
To a solution of H-pro-OtBu (53) (l.033g, 6.0mmol, II,
Scheme 5) in CH2Cl2 (20ml) and DMF (5ml) was added
Fmoc-tLeu-OH (2.337g, 6.60mmol, I, Scheme 5), HOBT
=. l.S3g, i2.immol) and EDC (2.30g, 12. Ommol) and the
solution stirred at room temperature under N2 for
. The solvent was removed in vacuo, and the
residue dissolved in EtOAc then washed with 0. 5N NaKSO4
saturated NaHCO3 (2x and brine. The organic

layer was dried over anhydrous Na2SO4 and evaporated to
give a pale yellow solid (3.65g). Flash chromatography
using EtOAc/hexanes (10/90 to 20/80%) give the title
compound (66) (2.25g, 74% yield). 1H-NMR (500MHz,
CDCl3) δ 1.09 (s, 9H) , 1.47 (s, 9H) , 1.79-2.28 (m, 3H) ,
3.62-3.72 (m, 1H), 3.76-3.83 (m, 1H), 4.18-4.43 (m,
4H) , 5.48-5.67 (m, 1H) , 7.28-7.44 (m, 4H) , 7.55-7.64
(m, 2H), 7.72-7.82 (m, 2H) . Analytical HPLC (cyano
column) 11.95min. LC-MS (ES+) m/e=507.3 (M+H).
1- [2- (4-Methoxy-benzoylamino) -3,3-dimethyl-butyryl] -
pyrrolidine-2-carboxylic acid tert-butyl ester (67) .
To a solution of 66 (0.5 03g, 0.99mmol) in DMF (8ml) was
added diethylamine (2.5ml) and the solution stirred at
room temperature for lhour and the solvent evaporated.
The resulting residue was repeatedly concentrated from
CH2Cl2 (3x). The resulting oil was dissolved in CH2Cl2
(9ml) and DIEA (2 60µl, 1.4 9mmol) and 4-methoxy-benzoyl
chloride (190mg, 1.05mmol) was added. The solution was
stirred under N2 for 18hours and the solvent
concentrated in vacuo. The residue was dissolved in
EtOAc and washed with 0.5N NaHSO4 (2x), saturated
NaHCO3 (2x) and brine then dried over anhydrous Na2SO4
and evaporated to give a white solid (0.529g). Flash
chromatography on silica gel using MeOH/CH2Cl2 (1/99 to
2/98%) gave the title compound (2.25g, 74% yield). XH-
NMR (500MHz, CD3OD) 6 1.01 (s, 1.4H), 1.11 (s, 7.6H),
1.73-2.25 (m, 4H) , 2.47-2.77 (m, 1H) , 2.81 (dd, 0.7H),
2.91-3.11 (m, 0.3H), 3.61-4.03 (m, 3H), 3.84 (s, 3H),
4.29-4.49 (m, 1H), 4.49-5.00 (m, 5H), 5.46 (s, 0.15H),
5.58-5.73 (m, 0.85H), 6.94-7.04 (m, 2H), 7.27-7.41 (m,
4H), 7.61-7.73 (m, 1H) , 7.74-7.84 (m, 2H). Analytical
HPLC (cyano column) 13.10min.

1- [2- (4-Methoxy-benzoylamino) -3, 3-dimethyl-butyryl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (68).
To a solution of 67 (0.90g, l.74mmol) in CH2Cl2 (25ml)
was added 2,6-lutidine (2.1ml, I8.0ramol) and TMS-
triflate (2.3ml, ll.9mmol) and the reaction stirred at
room temperature under N2 for 1.5hours. The resulting
mixture was diluted with CH2Cl2, washed with 10% NaHCO3
(2x) and brine then dried over Na2SO4, filtered and
evaporated. The residue was dissolved in CH2Cl2 then
treated with DIEA (0.6ml, 3.5mmol) and 4-methoxy-
benzoyl chloride (0.355g, 2.09mmol) and allowed to stir
under N2 at room temperature for 18hours. The crude
product was purified by flash chromatography, eluting
with CH2Cl2/MeOH (99/1) to yield the title compound
(274mg, 28% yield). ^-NMR (500MHz, CD3OD) 6 1.01 (s,
1.4H), 1.11 (S, 7.6H), 1.73-2.25 (m, 4H), 2.47-2.77 (m,
1H), 2.81 (dd, 0.7H), 2.91-3.11 (m, 0.3H), 3.61-4.03
(m, 3H) , 3.84 (s, 3H) , 4.29-4.49 (m, 1H) , 4.49-5.00 (m,
5H), 5.46 (s, 0.15H), 5.58-5.73 (m, 0.85H), 6.94-7.04
(m, 2H) , 7.27-7.41 (m, 4H) , 7.61-7.73 (m, 1H), 7.74-
7.84 (m, 2H) . Analytical HPLC (cyano column) (mixture
of 2 diastereomers) 17.03, 17.39min. LC-MS (ES+)
m/e=552.3 (M+H).
3- ({l- [2- (4-Methoxy-benzoylamino) -3,3-dimethyl-
butyryl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (69) .
A 117mg (0.2lmmol) sample of 68 was hydrolyzed
according to method C (see Scheme XXIII) to afford 4 0mg
(41% yield) of the title compound: Analytical HPLC
7.l6min. LC-MS (ES+) m/e=462.3 (M+H).


1- (2- tert-Butoxycarbonylamino-3, 3-dimethyl-butyryl) -
pyrrolidine-2-carboxylic acid benzyl ester (70).
To a suspension of H-pro-OBzl-HCl (2.00g, 8.66mmol) in
CH2Cl2 (20ml) was added DIEA (2.25ml, l2.92mmol) to
give a colorless solution. Boc-tLeu-OH (1.95g,
9.52mmol), HOBT (1.769, 13.03mmol) and EDC (2.49g,
12.95mmol) were added and the solution stirred under N2
at room temperature for 18hours. Removed solvent in
vacuo, dissolved in'EtOAc and washed with H2o, 0.5N
NaHSO4 (2x), saturated NaHCO3 (2x) and brine. Dried
over anhydrous Na2SO4 and evaporated to give the title
compound. (3.57g, 99% yield). 1H-NMR (500MHz, CDCl3) δ
0.99 (S, 9H) , 1.40 (s, 9H) , 1.88-2.33 (m, 4H) , 3.58-
3.90 (m, 2H), 4.21-4.35 (d, 1H), 4.S3-4.66 (m, 1H),
5.04-5.38 (m, 3H) , 7.14-7.42 (m, 5H) . LC-MS (ES+)
m/e=419.4 (M+H).
{1- [2- (2-Benzyloxy-5-oxo-tetrahydro-furan-3-
ylcarbomoyl) -pyrrolidine-1-carbonyl] -2,2-dimethyl-
propyl}-carbamic acid text-butyl ester (71) .

An 87lmg (2.08mmol) sample of 70 was dissolved in MeOH
(15mL) and 10% Pd/C (200mg) added. The suspension was
stirred under H2 for lhour then filtered through Celite
and the solvent evaporated. This resulting residue was
reacted with 40 according to the procedure used to
prepare 56 to give 889mg (71% yield) of the title
compound (71). 1H-NMR (500MHz, CDCl3) δ 0.93 (s, 9H) ,
1.44 (s, 9H), 1.78-2.18 (m, 4H) , 2.29-2.49 (m, 2H),
2.76-3.04 (m, 1H), 3.50-3.70 (m, 1H), 3.70-3.85 (m,
1H) , 4.20-4.37 (m, 1H) , 4.49-4.78 (m, 3H) , 4.78-4.98
(m, 1H) , 5.12-5.26 (m, 1H), 5.40-5.59 (m, 1H) , 7.10-
7.78 (m, 5H). Analytical HPLC (cyano column) 11.17min.
LC-MS (ES+) m/e=518.3 (M+H).
1- [2- (4-Amino-3-chloro-benzoylamino)-3,3-dimethy1-
butyryl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-5-
oxo-tetrahydro-furan-3-yl)-amide (72).
A solution of 456mg (0.088mmol) of 71 in CH2Cl2 (20ml)
was treated with anhydrous TFA (5mL) then stirred at
room temperature under N2 for 1 hour and evaporated to
dryness. The residue was repeatedly concentrated from
CH2Cl2 (3x) then dried under vacuum. The resulting
residue was dissolved in CH2Cl2 (20ml) , cooled to 0°C,
then treated with DIEA (1.3ml, 8eq, 2.4 6mmol) followed
by 4-amino-3-chloro-benzoic acid (202mg, l.l7mmol),
HOBT (183mg, 1.35mmol), and EDC (279mg, 1.45mmol). The
resulting mixture was allowed to warm to room
temperature and stir for 18hours. The solvent was
removed in vacuo and the residue dissolved in EtOAc
then washed with distilled water (3x), 0.5N NaHSO4
(2x) , saturated NaHCO3 (2x) and brine. The organic
layer was dried over Na2SO4, filtered and evaporated to
give a residue that was purified by flash
chromatography, eluting with CH2Cl2/MeOH (99/1 to

97/3%), affording 265mg (57% yield) of the title
compound (72) as a yellow solid.
1H-NMR (500MHz, CD3OD) δ 0.91-1.24 (m, 9H) , 1.70-2.27
(m, 4H), 2.47-2.85 (m, 1.5H), 2.99-3.13 (m, 0.5H),
3.39-3.53 (m, 0.5H), 3.60-3.78 (m, 1.5H), 3.85-4.04 (m,
1H), 4.24-4.47 (m, 2H) , 4.53-4.97 (m, 4H) , 5.46 (s,
0.3H), 3.88-4.02 (m, 0.1H), 5.60-5.69 (m, 0.6H), 6.80
(d, 1H), 7.22-7.77 (m, IE). Analytical HPLC (cyano
column)(mixture of 2 diastereomers) 15.90, 16.23min.
LC-MS (ES+) m/e=571.2 (M+H).
3- ({1- [2- (4-Amino-3-chloro-benzoylamino) -3,3-dimethyl-
butyryl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (73).
A 40mg (0.07mmol) sample of 72 was hydrolyzed according
to method A (see Scheme XXIII) to afford 25mg (74%
yield) of the title compound: Analytical HPLC (cyano
column) 10.66min. LC-MS (ES+) m/e=481.3 (M+H).


{2- [2- (2-Benzyloxy-5-oxo-tetrahydro-furan-3-
ylcarbamoyl) -pyrrolidin-1-yl] -l-methyl-2-oxo-ethyl}-
carbamic acid tert-butyl ester (75).
To a solution of 40 (6.69g, 23.0mmol) in anhydrous
CH2Cl2 was added 1,3-dimethylbarbituric acid (DMBA)
(3.97g, 25.4mmol) andPd(PPh3)4 (1.12g, 0.97mmol). The
solution was stirred under N2 at room temperature for
15min, cooled to 0°C, followed by the addition of Boc-
ala-pro-OH (BaChem) (5.087g, l7.8mmol), HOBT (3.60g,
26.7mmol) and EDC (5.l2g, 26.7mmol). The resulting
solution was allowed to warm to room temperature and
stir under N2 for 18hours. The solvent was
concentrated in vacuo and the residue dissolved in
EtOAc, then washed with 0.5N NaHSO4 (2x), saturated
NaHCO3 (2x) and brine. The organic layer was dried
over anhydrous Na2SO4 and evaporated to give an orange
oil (12.23g). Flash column chromatography on silica gel
using CH2Cl2/EtOAc (80/20 to 60/40) gave the title
compound 75 as a yellow solid (7.28g, 86% yield). 1H-
NMR (500MHz, CD3OD) δ 1.19-1.31 (m, 3H) , 1.42 (s, 9H) ,
1.69-2.29 (m, 4H) , 2.45-2.67 (m, 0.9H), 2.71-2.86 (m,
0.5H), 2.99-3.10 (m, 0.6H), 3.49-3.84 (m, 2H) , 4.24-
4.45 (m, 2.5H), 4.57-4.73 (m, 1.5H), 4.76-4.92 (m, 1H) ,
5.45 (s, 0.45H), 5.63-5.68 (m, 0.55H), 7.25-7.40 (m,
5H). Analytical HPLC (cyano column)(mixture of 2
diastereomers) 15.99, 16.33min. LC-MS (ES+) m/e=476.3
(M+H).


[1- [2- (4-Amino-3-chloro-benzoylamino)-propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide] (76) .
A 1.899g (3.99mmol) sample of 75 in CH2Cl2 (20ml) was
treated with anhydrous TFA (5ml) then stirred at room
temperature under N2 for 1 hour and evaporated to
dryness. The residue was repeatedly concentrated from
CH2Cl2 (3x) then dried under vacuum. The resulting
residue was dissolved in CH2Cl2 (20ml), cooled to 0 C,
then treated with DIEA (5.6ml, 8eg, 32.1mmol), 4-amino-
3-chloro-benzoic acid (0.910g, 5.3mmol), HOBT (0.824g,
6.lmmol), and EDC (1.197g, 6.23mmol). The resulting
mixture was warmed to room temperature and stirred for
18hours. The solvent was removed in vacuo and the
residue dissolved in EtOAc then washed with distilled
water (3x), 0. 5N NaHSO4 (2x), saturated NaHCO3 (2x) and
brine. The organic layer was dried over Na2SO4,
filtered and evaporated to give a residue that was
purified by flash chromatography using CH2Cl2/MeOH
(99/1 to 97/3%) . The title compound was obtained as a
white solid (1.221g, 58% yield) . 1H-NMR (500MHz, CD3OD)
δ 1.15 (d, 0.25H),, 1.29-1.60 (m, 2.75H), 2.41-2.54 (m,
0.5H), 2.55-2.70 (m, 0.5H), 2.77 (dd, 0.5H), 3.03 (ddd,
0.5H), 3.59-3.75 (m, 1H), 3.75-3.98 (m, 1H), 4.26-5.01
(m, 5H) , 5.41-5.57 (m, 1H) . 5.60-5.76 (m, 0.5H), 6.70-
6.92 (m, 0.5H), 7.15-7.48 (m, 5H), 7.48-7.68 (m, 1H),
7.68-7.88 (m, 1H) , 8.15-8.34 (m, 1H) . Analytical HPLC
(cyano column)(mixture of 2 diastereomers) 14.44,
14.89min. LC-MS (ES+) m/e=529.3 (M+H) .


[1- [2- (4-Methoxy-3, 5-dimethyl-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide] (77) was
synthesized from 75 and 3 , 5-dimethyl-4-methoxy benzoic
acid according to the procedure used to prepare 76 to
afford the title compound (I.18g, 44% yield). 1H-NMR
(500MHz, CD3OD) 6 1.40 (m, 3H), 1.67-2.41 (m, 4H), 2.28
(s, 6H), 2.48 (ddd, 0.5H), 2.62 (dd, 0.5H), 2.78 (ddd,
0.5H), 3.04 (ddd, 0.5H), 3.62-3.94 (m, 3H), 3.71 (s,
3H) , 4.21-4.51 (m, 2H) , 4.59-4.85 (m, 4H) , 5.46 (s,
0.25H), 5.52 (S, 0.25H), 5.63 (d, 0.4H), 5.67 (d,
0.1H), 7.17-7.45 (m, 5H) , 7.45-7.65 (m, 2H) .
Analytical HPLC (cyano column) (mixture of 2
diastereomers) 15.06, 15.39min. LC-MS (ES+) m/e=538
(M+H)
Preparation of 4-Acetylamino-3-chlorobenzoic acid
To a solution of 4-amino-3-chloro-benzoic acid (10. 0g,
58.3mmol) in anhydrous THF (100mL) was added acetyl
chloride (20.7ml, 291.1mmol) and the solution stirred
at room temperature for 48hours. The solvent was
evaporated and the product precipitated from hexanes
then filtered and dried to give a white solid (11.73g,
94% yield). 1H-NMR (500MHz, CD3OD) δ 2.28 (s, 3H) , 7.92
(dd, 1H) , 7.99-8.16 (m, 2H) . Analytical HPLC (cyano
column) 7.84min.


1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (78) .
Prepared from 75 and 4-acetylamino-3-chloro-benzoic
acid according to the procedure used to prepare 7 6 to
afford the title compound (14 6mg, 19% yield) . 1H-NMR
(500MHz, CD3OD) δ 1.28-1.52 (m, 3H) , 1.68-2.38 (m, 4H) ,
2.20 (s, 3H), 2.41-2.88 (m, 1.5H), 2.96-3.10 (m, 0.5H),
2.96-3.10 (m, 0.5H), 3.43-3.75 (m, 1H) , 3.80-3.96 (m,
1H) , 4.25-5.00 (m, %H) , 5.42-5.54 (m, 0.5H), 5.63-5.78
(m, 0.5H), 7.13-7.48 (m, 05H), 7.79-8.14 (m, 2.5H),
8.56-8.70 (m, 0.5H). Analytical HPLC (cyano column)
(mixture of 2 diastereomers) δ.64min. LC-MS (ES+)
m/e=571.2 (M+H).

1- [2- (3-Isopropoxy-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (79).
Prepared from 75 and 3-isopropoxybenzoic acid according
to the procedure used to prepare 76 to afford the title
compound (120mg, 58% yield). 1H-NMR (500MHz, CD3OD) δ
1.27 (d, 6H), 1.33-1.52 (m, 3H), 1.69-2.31 (m, 4H) ,
2.49 (dd, 0.3H), 2.63 (dd, 0.7H), 2.78 (dd, 0.7H), 3.03
(dd, 0.3H), 3.43-3.73 (m, 1H), 3.78-3.94 (m,1H), 4.27-
4.47 (m, 2H) , 4.47-4.87 (m, 4H) , 5.47 (s, 0.7H) , 5.53
(d, 0.3H), 5.64 (d, 0.8H), 5.72 (d, 0.2H), 6.98-7.12
(m, 1H), 7.19-7.47 (m, 9H). Analytical HPLC (cyano

column) (mixture of 2 diastereomers) 14.54, 14.85min.
LC-MS (ES+) m/e=538 (M+H).

Quinoxaline-2-carboxylic acid {2- [2- (2-benzyloxy-5-oxo-
tetrahydrofuran-3-ylcarbamoyl) -pyrrolidin-1-yl] -1-
methyl-2-oxo-ethyl}-amide (80).
Prepared from 75 and 2-quinoxaline carboxylic acid
according to the procedure used to prepare 76 to afford
the title compound (122mg, 60% yield). 1H-H-NMR (500MHz,
CD3OD) δ 1.12-1.67 (m, 3H) , 1.68-2.34 (m, 4H) , 2.35-2.70
(m, 0.85H), 2.70-2.95 (m, 0.75H), 3.06 (dd, 0.4H),
3.41-3.49 (m, 2H) , 4.18-5.03 (m, 6H), 5.47 (d, 0.5H),
5.55, (d, 2H), 5.67 (dd, 1H), 5.71 (dd, 0.3H), 7.03-
7.53 (m, 5H), 7.80-8.06 (m, 2H), 8.06-8.34 (m, 2H) ,
9.43-9.48 (m, 1H).Analytical HPLC (cyano column)
(mixture of 2 diastereomers) 9.06min. LC-MS (ES+)
m/e=532.3 (M+H).


1- [2- (3-Benzyloxy-4-methoxy-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (81) .
Prepared from 75 and 3-benzyloxy-4-methoxy benzoic acid
according to the procedure used to prepare 76 to afford
the title compound (142mg, 58% yield) . 1H-H-NMR (500MHz,
CD3OD) δ 1.14 (d, 0.3H), 1.27-1.52 (m, 2.7H) , 1.66-2.30
(m, 4H), 2.47 (dd, 0.4H), 2.59 (dd, 0.6H), 2.77 (dd,
0.6H), 3.02 (dd, 0.4H), 3.41-3.72 (m, 1H), 3.72-3.99
(m, 2H), 3.86 (S, 3H), 4.19-4.86 (m, 5H), 4.99-5.15 (m,
2H) , 5.45 (m, 0.8H), 5.65 (m, 1.2H), 6.98 (dd, 1H) ,
7.11-7.63 (m, 12H). Analytical HPLC (cyano
column)(mixture of 2 diastereomers) 12.28, 12.44min.
LC-MS (ES+) m/e=616.3 (M+H).
4-Allyloxy-3 , 5-dimethyl-benzoic acid.
A mixture of 4-hydroxy-3,5-dimethyl-benzoic acid (3.32
g, 20 mmol) , allyl bromide (7.26 g, 60 mmol) ,
benzyltriethylammonium chloride (455 mg, 2 mmol) and
K2CO3. (6.9 g, 50 mmol) in DMF (50 mL) was stirred at
room temperature for 16 hours. The mixture was diluted
with ethyl acetate (200 mL) , washed with water, brine.
The organic layer was dried over Na2SO4, filtered and
evaporated in vacuo to give 5.3 g of the ester as an
oil. The ester was refluxed with NaOH (5 g, 125 mmol)
in water/methanol (50 mL/50 mL) for 6 hours. The
mixture was evaporated in vacuo to remove methanol and
the resulted solution was diluted with water (200 mL) ,
washed with ethyl acetate/hexane (30 mL/70 mL) . The
aqueous layer was acidified at o°C with concentrated
HCl solution to pH 2 . The resulted precipitate was
collected by filtration and washed with water, dried

over high vacuum to afford 3.86 g (yield 94%) of the
title compound. 1H-NMR (500 MHz, CDCl3) : δ 2.33 (s,
6H), 4.35-4.37 (m, 2H), 5.28-5.30 (m, H), 5.42-5.46 (m,
H), 6.07-6.15 (m, H), 7.79 (s, 2H); retention time on
analytical HPLC: 11.28 min; LC-MS: m/z = 205 (M-H+) .

1- [2- (4-Allyloxy-3,5-dichloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (82).
Prepared from 75 and 4-allyloxy-3,5-dichloro-benzoic
acid according to the procedure used to prepare 16 to
afford the title compound (208mg. 47% yield). 1H-NMR
(500MHz, CDCl3) δ 1.05-1.58 (m, 3H), 1.68-3.21 (m, 7H),
3.39-3.90 (m, 3H), 4.05-5.01 (m, 6H), 5.22-5.62 (m,
3H), 6.04-6.25 (m, 1H), 6.94-7.63 (m, 8H) . Analytical
HPLC (cyano column) (mixture of 2 diastereomers) 9.69,
9.89min. LC-MS (ES+) m/e=604.2 (M+H).

1- [2- (3 , 5-Dichloro-4-hydroxy-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (83).
A 14 0 mg sample of 82 (0.23mmol) was dissolved in
CH2Cl2 (4mL) and treated with DMBA (35.4mg, 0.26mmol)
and Pd(PPh3)4 (32mg, 0.028mmol). The solution was

stirred at 0°C for I5mins, warmed to room temperature
for 2hours, then diluted with CH2Cl2 and washed with
water (2x) and brine. The solvent was concentrated in
vacuo and the residue purified by flash chromatography
on silica gel using MeOH/CH2Cl2 (1/99 to 3/97) to give
the title compound (93.2mg, 71% yield). 1H-NMR
(500MHz, CD3OD) δ 1.16 (d, 0.25H), 1.28-1.49 (m, 2.75H),
1.63-2.33 (m, 4H) , 2.48 (dd, 0.4H), 3.39-3.59 (m,
0.2H), 3.60-3.73 (m, 0.8H), 3.73-3.96 (tn, 1H) , 4.24-
4.48 (m, 2H) , 4.57-4.92 (m, 7H) , 5.44 {s, 0.4H), 5.50
(d, 0.4H), 5.64 (d, 0.8H), 5.75 (d, 0.5H), 7.16-7.43
(m, 5H), 7.78-7.89 (m, 1.6H), 8.40-8.63 (m, 0.4H).
Analytical HPLC (cyano column) (mixture of 2
diastereomers) 11.57, 11.82min. LC-MS (ES+) m/e=564.1
(M+H)

1- (2-Benzoylamino-propionyl) -pyrrolidine-2-carboxylic
acid (2-benzyloxy-5-oxo-tetrahydro-furan-3-yl) -amide
(84) .
Prepared from 75 and benzoyl chloride according to the
procedure used to prepare 76 to afford the title
compound as a colorless oil (8mg, 38% yield) . 1H-NMR
(500MHz, CD3OD) δ 1.35-1.54 (m, 3H) , 1.72-2.30 (m, 4H) ,
2.42-2.70 (m, 1.3H), 2.74-2.84 (m, 0.5H), 3.03 (dd,
0.2H), 3.41-3.75 (m, 2H) , 3.81-3.96 (m, 1H) , 4.22-4.86
(m, 4H), 5.46 (s, 0.3H), 5.51-5.54 (m, 0.1H), 5.66 (d,
0.5H), 5.72 (d, 0.1H), 7.20-7.57 (m, 7H), 7.77-7.89 (m,
2H) , 8.42-8.67 (m, 1H) . Analytical HPLC (cyano

column) (mixture of 2 diastereomers) 15.23, 15.67min.
LC-MS (ES+) m/e=481.2 (M+H)

Isoguinoline-1-carboxylic acid {2- (2-benzyloxy-5-oxo-
tetrahydro-furan-3-ylcarbamoyl) -pyrrolidin-1-yl] -1-
methyl-2-oxo-ethyl}-amide (85) .
Prepared from 75 and 1-isoquinolinecarboxylic acid
according to the procedure used to prepare 76 to afford
the title compound (732mg, 53% yield). 1H-NMR (500MHz,
CD3OD) δ 1.22-1.56 (m, 3H) , 1.70-2.34 (m, 4H) , 2.43-2.71
(m, 0.9H), 2.73-2.89 (m, 0.5H), 3.06 (ddd, 0.6H), 3.42-
3.81 (m, 2H), 3.84-4.01 (m, 1H), 4.29-5.00 (m, 5H) ,
5.47 (d, 0.65H), 5.55 (s, 0.3H), 5.67 (d, 0.8H), 5.72
(d, 0.25H), 7.21-7.43 (m, 5H), 7.49-7.83 (m, 2.8H),
7.88-8.04 (m, 1.8H), 8.45-8.54 (m, 0.8H), 8.97-9.06 (m,
0.6H). Analytical HPLC (mixture of 2 diastereomers)
15.71, 16.04min. LC-MS (ES+) m/e=531.2 (M+H).-

1- [2- (4-Amino-5-chloro-2-methoxy-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (86).
Prepared from 75 and 4-amino-5-chloro-2-methoxy benzoic
acid according to the procedure used for 76' to afford

the title compound (330tng, 61% yield) . 1H-NMR (500MHz,
CD3OD) δ 1. 22 (d, 0.25H), 1.29-1.50 (m, 0.75H), 1.68-
2.36 (m, 4H), 2.38-2.89 (m, 1.5H), 2.94-3.14 (m, 0.5H),
3.37-3.98 (m, 6H), 4.27-4.98 (m, 6H), 5.44-5.50 (m,
0.4H), 5.53-5.56 (s, 0.1H), 5.60-5.75 (m, 0.5H), 6.50
(s, 1H) , 7.17-7.45 (m, 4H) , 7.73-7.90 (m, 1H) , 8.49-
8.70 (m, 1H). Analytical HPLC (cyano column)(mixture
of 2 diastereomers) 16.39, 16.82min. LC-MS (ES+) m/e=
559.2 (M+H).

1- [2- (4-Acetylamino-5-chloro-2-methoxy-benzoylamino) -
propionyl] -pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl) -amide (87) .
Prepared from 75 and 4-acetylamino-5-chloro-2-methoxy
benzoic acid according to the procedure used for 76 to
afford the title compound (364mg, 64% yield). 1H-NMR
(500MHz, CD3OD) δ 1:20-1.27 (m, 0.25), 1.35-1.49 (m,
0.75H), 1.72-2.30 (m, 4H) , 2.23 (s, 3H), 2.42-2.58 (m,
0.6H), 2.59-2.68 (m, 0.5H), 2.73-2.86 (m, 0.7H), 2.99-
3.11 (m, 0.7H), 3.41-4.07 (m, 5H), 4.29-4.97 (m, 5H),
4.79-5.56 (m, 0.5H), 5.65-5.73 (m, 0.5H), 7.18-7.44 (m,
4.3H), 7.90-8.09 (m, 2H) , 8.71-8.85 (m, 0.7H).
Analytical HPLC (cyano column) (mixture of 2
diastereomers) 15.61, 16.0lmin. LC-MS (ES+) m/e= 601.1
(M+H).


Pyridine-2-carboxylic acid{2- [2- (2-benzyloxy-5-oxo-
tetrahydro-furan-3-ylcarbamoyl) -pyrrolidin-1-yl] -1-
methyl-2-oxo-ethyl}-amide (88).
Prepared from 75 and pyridine-2-carboxylic acid
according to the procedure used for 76 to afford the
title compound (233mg, 42% yield). 1H-NMR (500MHz,
CD3OD) δ 1.30-1.59 (m, 3H), 1.68-2.36 (m, 4H) , 2.39-2.57
(m, 0.6H), 2.57-2.69 (m, 0.35H), 2.71-2.87 (m, 0.4H),
3.05 (dd, 0.65H), 3.39-3.93 (m, 3H) , 4.24-4.99 (m, 5H) ,
5.49-5.55 (m, 0.8H), 5.63-5.77 (m, 1.2H), 7.17-7.46 (m,
5H) , 7.49-7.60 (m, 1H) , 7.89-7.99 (m, 1H) , 8.03-8.12
(m, 1H) , 8.58-8.67 (m, 1H) . Analytical HPLC (cyano
column) (mixture of 2 diastereomers) 8.63min. LC-MS
(ES+) m/e=481.3 (M+H) .

[1- [2- (4-Amino-3, 5-dich.loro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide] (89) .
Prepared from 75 and 3,5-dichloro-4-aminobenzoic acid
according to the procedure used for 76 to afford the
title compound (I62mg, 70% yield) . 1H-NMR (500MHz,
CD3OD) δ 1.21-1.58 (m, 3H), 1.58-2.37 (m, 4H), 2.37-3.13
(m, 2H) , 3.43-3.74 (m, 1.5H), 3.77-3.94 (m, 1H). , 4.28-

4.51 (m, 1.5H), 4.50-5.01 (m, 3H) , 5.41-5.77 (m, 1H) ,
7.15-7.49 (m, 5H), 7.66-7.88 (m, 2H). Analytical HPLC
(cyano column)(mixture of 2 diastereomers) δ.36min. LC-
MS (ES+) m/e=563.2 (M+H).

1- [2- (4-Methoxy-benzoylamino) -propionyl] -pyrrolidine-2-
carboxylic acid (2-benzyloxy-5-oxo-tetrahydro-furan-3-
yl)-amide (90).
Prepared from 75 and 4-methoxy-benzoylchloride
according to the procedure used for 76 to afford the
title compound (404mg, 50%) . 1H-NMR (500MHz, CD3OD) δ
1.19 (d, 0.3H), 1.29-1.58 (m, 2.7H), 1.58-2.38 (m, 4H),
2.43-2.69 (m, 1H), 2.74-2.86 (m, 0.6H), 2.99-3.11 (m,
0.4H), 3.39-3.75 (m, 1.5H), 3.77-3.94 (m, 1H), 3.84 (s,
3H), 4.29-4.94 (m, 4.5H), 5.45-5.55 (m, 4.5H), 5.63-
5.71 (m, 0.5H), 5.73 (d, 0.1H), 6.85-7.09 (m, 2H),
7.19-7.44 (m, 4H), 7.73-7.92 (m, 2H), 8.26-8.44 (m,
1H). Analytical HPLC (cyano column)(mixture of 2
diastereomers) 15.18, 15.65min. LC-MS (ES+) m/e=510.2
(M+H).


l-{2- [ (9-Oxo-9H-fluorene-4-carbonyl) -amino] -propionyl}-
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (91) .
Prepared from 75 and 9-oxo-9H-fluorene-carboxylic acid
according to the procedure used for 76 to afford the
title compound (403mg, 44% yield) . 1H-NMR (500MHz,
CDCl3) δ 1.38-1.59 (m, 3H) , 1.75-2.37 (m, 4H) , 2.43-2.59
(m, 0.65H), 2.59-2.72 (m, 0.35H), 2.79-2.89 (m, 0.35H),
3.01-3.11 (m, 0.65H), 3.68-3.86 (m, 1H) , 3.92-4.09 (m,
1H) , 4.35-5.03 (tn, 7H) , 5.42-5.90 (m, 1H) , 7.06-8.00
(m, 12H) . Analytical HPLC (cyano column) (mixture of 2
diastereomers) 12.30min. LC-MS (ES+) m/e=582.1 (M+H).

1- [2- (3, 5-Dichloro-4-methoxy-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (92) .
Prepared from 75 and 3, 5-dichloro-4-methoxy-benzoic
acid according to the procedure used for 76 to afford
the title compound (364mg. 46% yield). 1H-NMR (500MHz,
CD3OD) δ 1.17 (d, 0.25H), 1.28-1.53 (m, 2.75H), 1.64-
.2.33 (m; 4H) , 2.39-2.94 (m, 1.5H), 2.94-3.12 (m, 0.5H) ,
3.41-3.74 (m, 2H) , 3.74-4.00 (m, 1H), 3.91 (s, 3H),
4.26-5.02 (m, 5H) , 5.42-5.81 (m, 1H), 7.08 (d, 0.4H),
7.21-7.43 (m, 4.6H), 7.53-7.69 (m, 0.8H), 7.85-7.97 (m,
1.2H) . Analytical HPLC (cyano column) (mixture of 2
diastereomers) 10.79min. LC-MS (ES+) m/e=578.2 (M+H).


Quinoline-6-carboxylic acid {2-(2-benzyloxy-5-oxo-
tetrahydro-£uran-3-ylcarbamoyl) -pyrrolidin-1-yl] -1-
methyl-2-oxo-ethyl}-amide (93).
Prepared from 75 and 6-quinolinecarboxylic acid
according to the procedure used for 76 to afford the
title compound (344mg, 71% yield) . 1H-NMR (500MHz,
CD3OD) δ 1.11-1.58 (m, 3H), 1.69-2.40 (m, 4H) , 2.42-3.15
(m, 2H) , 3.80-4.01 (m, 1H), 4.29-4.99 (m, 5H), 5.44-
5.54 (m, 0.5H), 5.63-5.73 (d, 0.4H), 5.73-5.79 (d,
0.1H), 7.18-7.43 (m, 5H), 7.56-7.67 (m, 1H), 8.08 (d,
1H) , 8.13-8.25 (m, 1H), 8.40-8.56 (m, 2H), 8.88-8.99
(m, 1H). Analytical HPLC (cyano column)(mixture of 2
diastereomers) 10.27, 10.50min. LC-MS (ES+) m/e=531.2
(M+H).


1- (2-Benzyloxycarbonylamino-propionyl) -pyrrolidine-2-
carboxylic acid tert-butyl ester (95).
Prepared according to the method described in Pierre
Chevallet, Patrick Garrouste, Barbara Malawaska & Jean
Martinez in Tetrahedron Letters, Vol. 34, pp. 7409-
7412, (1993). A mixture of Cbz-ala-pro-OH (10.0 g,
31.2 mmol), tert-butyl bromide (180 g, 1.31 mol) ,
benzyltriethylammonium chloride (7.11 g, 31.2 mmol) and
K2CO3 (180 g, 1.3 0 mol) in N,N-dimethylacetamide (DMA)
(225 mL) was stirred at 55°C for 24 hours. The
reaction mixture was cooled to room temperature and
diluted with one liter of ice-water, extracted with
ethyl acetate (200 mL x 3) . The organic layer was
dried over anhydrous Na2SO4, filtered and evaporated in

vacuo to give 14 g of oil, which was purified by flash
chromatography using hexane/ethyl acetate (95/5 to
50/50) to afford 11.73 g (yield 99.7%) of the title
compound as a clear oil. 1H-NMR (500 MHz, CDCl3): δ
1.25-1.50 (m, 12 H), 1.85-2.25 (m, 4H) , 3.42-3.70 (m,
2H) , 4.25-4.57 (m, 2H) , 5.07-5.11 (m, 2H) , 5.69 (d, H) ,
7.28-7.38 (m, 5H) ; retention time on analytical HPLC:
11.07 min; LC-MS: m/z = 377 (M+H+).

1- [2-(4-Amino-3-chloro-benzoylamino)-propionyl]-
pyrrolidine-2-carboxylic acid tert-butyl ester (96a).
To a solution of 95 (10.50g, 27.9mmol) in MeOH (100ml)
was added a suspension of 10% Pd/C (5.00g) in EtOAc
(50ml). The mixture was stirred under H2 for 48hours,
filtered through celite and the solvent evaporated to
yield a waxy solid. This was dissolved in CH2Cl2
(100ml) and DMF (50ml) and the solution cooled to 0°C.
4-Amino-3-chlorobenzoic acid (5.82g, 27.2mmol), DIEA
(14.58ml, 83.7mmol), HOBT (3.77g, 27.9mmol) and EDC
(6.68g, 34.8mmol) were added and the solution stirred
at 0°C for I5mins then at room temperature for 24hours.
The reaction mixture was diluted with EtOAc, washed
with NaHSO4 (2x) , 10% NaHCO3 (2x) and brine then dried
over MgSO4, filtered and evaporated. The crude product
was purified by flash column chromatography, using
CH2Cl2/MeOH (99/1 to 97/3%) to yield the title compound

as a white solid (7.75g, 70% yield). 1H-NMR (500MHz,
CD3OD) δ 1.27-1.67 (m, 12H), 1.82-2.14 (m, 4H) , 3.48-
3.85 (m, 2H) , 4.26-4.53 (m, 3H) , 4.81-4.98 (m, 1H) ,
6.71 (d, 1H), 7.15 (m, 1H), 7.50 (dd, 1H), 7.75 (d,
1H). Analytical HPLC I0.83min. LC-MS (ES+) m/e=396.3
(MH+) .
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (97a).
Prepared from 96a by treatment with TFA/ CH2Cl2. After
complete reaction, the solvent is removed in vacuo and
the residue repeatedly concentrated from toluene. The
resulting residue was dried under vacuum to a constant
weight.
1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid tert-butyl ester (96b).
Prepared from 95 and 4-acetylamino-3-chlorobenzoic acid
according to the method used for 96a to afford the
title compound as a white solid (9.18g, 77% yield).
1H-NMR (500MHZ, CD3OD) δ 1.30-1.62 (m, 12H), 1.85-2.16
(m, 3H) , 2.16-2.44 (m, 1H), 2.27 (s, 3H) , 3.47-3.83 (m,
2H) , 4.34-4.54 (m, 1H) , 4.8.9 (m, 1H) , 7.27-7.39 (m,
1H) , 7.59-7.71 (m, 2H), 7.83-7.97 (m, 1H) , 8.47 (d,
1H). Analytical HPLC 9.43min.
1- [2- (4-Acetylmino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (97b).
Prepared from 96b by treatment with TFA/ CH2Cl2. After
complete reaction, the solvent is removed in vacuo and
the residue repeatedly concentrated from toluene. The
resulting residue was dried under vacuum to a constant
weight.

4-Acetylamino-5-chloro-2-methoxy-benzoic acid.
4-Acetylamino-5-chloro-2-methoxy-benzoic acid methyl
ester (2.09g, 8.11mmol) was dissolved in MeOH (110ml)
and LiOH solution (25.48mmol in 30ml, 1:1 MeOH:H2O)
added and the solution stirred at room temperature for
6hours. The solvent was concentrated in vacuo, EtOAc
added and the organic phase was washed with 0. 5N HCl
then extracted with saturated NaHCO3 (2x) . The aqueous
phase was acidified with 12N HCl to pH 1 and the
resulting precipitate extracted into CH2Cl2. The
combined extracts were dried over anhydrous Na2SO4,
filtered and evaporated to give the title compound as a
white solid (0.933g, 50% yield). 1H-NMR (500MHz, CDCl3)
δ 2.31 (s, 3H) , 4.10 (s, 3H), 7.78-7.92 (br s, 1H) ,
8.17 (s, 1H) , 8.45 (s, 1H) . Analytical HPLC 5.62min.
1- [2- (4-Acetylamino-5-chloro-2-methoxy-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid tert-butyl
ester (96c) .
To a solution of 95 (1.534g, 4.07mmol) in MeOH (40ml)
was added 10%Pd/C (650mg) and the mixture stirred under
H2 for 2hours. The suspension was filtered through
celite and evaporated to give a yellow oil. This was
allowed to react with 4-acetyl-5-chloro-2-methoxy
benzoic acid following the procedure used for the
preparation of 96a to give the title compound (497mg,
52% yield). 1H-NMR (500MHz, CD3OD) δ 1.46 (d, 3H) , 1.49
(s, 9H) , 1.80-2.01 (m, 3H) , 2.19-2.40 (m, 1H) , 2.22 (s,
3H) , 3.58-3.72 (m, 1H), 3.78-3.89 (m, 1H) , 3.98-4.09
(s, 3H) , 4.31-4.45 (s, 1H) , 4.78-4.95 (m, 1H) , 7.89-
8.10 (m, 2H) . Analytical HPLC 11.31min.

1- [2- (4-Acetylamino-5-chloro-2-methoxy-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (97c).
Prepared from 96c by treatment with TFA/ CH2Cl2 • After
complete reaction, the solvent is removed in vacuo and
the residue repeatedly concentrated from toluene. The
resulting residue was dried under vacuum to a constant
weight

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (5-oxo-2-phenethyloxy-
tetrahydro-furan-3-yl)-amide (98a) .
To a solution of (5-oxo-2-phenethyloxy-tetrahydro-
furan-3-yl)-carbamic acid allyl ester (194mg, 0.54mmol)
(prepared as described for (40) using phenethyl
alcohol) in anhydrous CH2Cl2 (5mL) at 0°C was added DMBA
(196mg, 1.26mmol) and Pd(PPh3)4 (32mg, 0.03mmol). The
solution was stirred for 15min and a solution of 97a
(prepared from 96a by treatment with TFA in
CH2Cl2)(166mg, 0.49mmol) and DIEA (680ul, 3.90mmol) in
CH2Cl2 (2mL) was added followed by HOBT (98mg,
0.73mmol) and EDC (I22mg, 0.63mmol). The solution was
stirred at 0°C for I5min then at room temperature for
18hours. The solvent was removed in vacuo and the
residue dissolved in EtOAc then washed with 0. 5N NaHSO4
(2x) , saturated NaHCO3 (2x) and brine. Dried over
anhydrous Ka2SO4 and evaporated to give an orange solid
which was purified by flash column chromatography,

using CH2Cl2/MeOH (99/1 to 97/3%) to yield the title
compound as a white solid (190mg, 73% yield) . 1H-NMR
(500MHz, CD3OD) δ 1.29 (d, 0.6H), 1.41 (d, 2.4H), 1.78
(m, 1H), 2.08 (m, 3H), 2.56 (m, 1H) , 2.77 (dd, 1H) ,
2.94 (t, 2H) , 3.53 (m, 0.3H), 3.67 (m, 0.8H), 3.85 (in,
2H) , 3.96-4.08 (m, 1H), 4.40 (m, 2H) , 4.62 (m, 1H) ,
4.67-4.79 (m, 1H), 5.57 (d, 0.7H), 5.60 (d, 0.3H), 6.78
(dd, 1H), 7.21 (m, 5H) , 7.58 (m, 1H) , 7.79 (m, 1H) ,
8.26 (d, 1H). Analytical HPLC 14.52min. LC-MS (ES+)
m/e=543.2 (MH+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-£uran-3-yl}-amide (98b) .
Was prepared from the syn diastereomer of (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-carbamic acid allyl ester
(40) and 97a following the method used for 98a. The
title compound was isolated as a pale yellow solid
(720mg, 51% yield). 1H-NMR (500MHz, CD3OD) δ 1.16 (d,
0.5H), 1.40 (d, 2.5H), 1.64-2.25 (m, 4H), 2.61 (dd,
1H) , 2.79 (dd, 1H), 3.37-3.59 (m, 1H) , 3.59-3.74 (m,
1H) , 3.77-3.92 (m, 1H) , 4.29-4.47 (m, 1H) , 4.47-5.02
(m, 4H), 5.48 (S, 0.5H), 5.66 (d, 1H) , 5.68 (d, 0.5H),
6.79 (d, 1H), 7.17-7.52 (m, 5H), 7.48-7.62 (m, 1H),
7.68-7.83 (m, 1H) . Analytical HPLC 15.98min. LC-MS (ES+)
m/e=529.2 (MH+).


1-[2-(4-Amino-3-chloro-benzoylamino)-propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98c) .
Prepared from the anti-(2-benzyloxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester (40) and 97a
following the method used for 98a. The title compound
was isolated as a white solid (186.6mg, 46% yield).
1H-NMR (500MHZ, CD3OD) δ 1.30-1.52 (m, 3H) , 1.76-2.33
(m,4H), 2.41-2.59 (m, 1H), 2.90 (dd, 0.15H), 3.04 (dd,
0.85H), 3.44-3.75 (m, 1.5H), 3.82-3.95 (m, 1H) , 4.27-
4.42 (m, 2H), 4.42-4.56 (m, 0.5H), 4.56-4.86 (m, 4H) ,
5.42-5.55 (m, 1H) , 6.79 (d, 1H), 7.21-7.42 (m, 4.6H),
7.54-7.63 (m, 1.4H), 7.76-7.83 (m, 0.65H), 8.60-8.68
(m, 0.35H). Analytical HPLC 15.19min. LC-MS (ES+)
m/e=529.3 (MH+) .

2- (Ethoxy-5-oxo-tetrahydro-furan-3-yl) -carbamic acid
allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for (40) using

ethanol. Chromatography using hexane/ethyl acetate
(95/5 to 80/20) gave 0.94 g of anti-2-(ethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
(higher Rf), 1.96 g of syn diastereomer (lower Rf) and
8.08 g of the mixture of the diastereomers (total
overall yield 60%) . 1H-NMR (500 MHz, CDCl3) for the
anti diastereomer: δ 1.13-1.31 (m, 3H) , 2.31-2.45 (m,
1H) , 2.92-3.08 (m, 1H) , 3.52-3.72 (m, 1H) , 3.78-3.92
(m, 1H), 4.10-4.25 (m, 1H) , 4.45-4.70 (m, 2H), 5.00
(bs, 1H) , 5.12-5.45 (m, 3H) , 5.80-5.95 (m, 1H) ; for syn
diastereomer 1.13-1.35 (m, 3H) , 2.38-2.50 (m, 1H),
2.75-2.92 (m, 1H) , 3.60-3.73 (m, 1H) , 3.82-3.95 |m,
1H) , 4.40-4.70 (m, 3H) , 5.10-5.52 (m, 4H) , 5.80-5.94
(m, 1H); LC-MS: m/z = 230 (M+H+) for both
diastereomers.
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-ethoxy-5-oxo-
tetrahydro-furan-3-yl) -amide (98d) .
Prepared from (2-ethoxy-5-oxo-tetrahydro-furan-3-yl) -
carbamic acid allyl ester and 97a following the method
used for 98a. The, title compound was isolated as a
white solid (175mg, 77% yield) . 1H-NMR (500MHz, CD3OD)
δ 1.13 (t, 0.5H), 1.23 (t, 2.5H), 1.36 (d, 0.5H), 1.44
(d, 2.5H), 1.75-2.38 (m, 4H) , 2.56 (dd, 1H), 2.76 (dd,
1H) , 3.45-3.97 (m, 5H), 4.47 (dd, 1H), 4.59-4.67 (m,
1H), 4.74 (q, 1H), 5.55 (d, 0.2H), 5.56 (d, 0.8H),
6.75-6.82 (m, 1H) , 7.56 (dd, 1H) , 7.77 (d, 1H) , 8.39
(d, 1H) . Analytical HPLC 8.17min. LC-MS (ES+)
m/e=467.4 (MH+).


(2 - Cyclopentyloxy- 5 - oxo - tetrahydro -furan - 3 -yl) - carbamic
acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using
cyclopentanol to afford the title compound as a mixture
of diastereomers. Flash column chromatography using
hexanes/EtOAc (90/10 to 80/20) afforded the syn
diastereomer of the title compound: syn diastereomer 1H
NMR (500MHz, CDCl3) δ 1.5-2.0 (m, 8H), 2.45 (dd, 1H) ,
2.81 (dd, 0.9H), 3.0 (dd, 0.1H), 4.31 (m, 1H), 4.59 (m,
4H), 5.23 (m, 1H), 5.32 (m, 1H), 5.45 (s, 0.1H), 5.51
(s, 0.9H), 5.92, (m, 1H) ppm; anti diastereomer 1H-NMR
(500 MHz, CDCl3) δ 1.50 (m, 2H), 1.67 (m, 6H), 2.36 (d,
1H), 2.8 (dd, 0.08H), 2.96 (dd, 0.92H), 4.13 (m, 1H),
4.25 (m, 1H) , 4.55 (br, 2H) , 5.20 (d, 1H), 5.30 (m,
2H), 5.43 (s, 0.92H), 5.5 (d, 0.08H), 5.89 (s, 1H) ppm.
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98e) .
Prepared from (2-cyclopentyloxy-5-oxo-tetrahydro-furan-
3-yl)-carbamic acid allyl ester and 97a following the
method used for 98a to give the title compound (2 8 0mg,
51% yield). 1H-NMR (500MHz, CD3OD) δ 1.38 (d, 0.5H),
1.44 (d, 2.5H), 1.49-2.35 (m, 12H), 2.47 (dd, 0.7H),
2.56 (dd, 0.3H), 2.75 (dd, 0.3H), 2.81-2.88 (m, 0.1H),
2.97 (dd, 0.6H), 3.47-3.76 (m, 0.2H), 3.82-3.96 (m,

1H) , 4.10-4.40 (m, 2H) , 4.40-4.46 (m, 1H) , 5.44 (d,
0.5H), 5.50 (d, 0.2H), 5.65 (d, 0.3H), 6.79 (d, 1H) ,
7.54-7.64 (m, 1H) , 7.78 (d, 1H) , 8.21-8.31 (m, 1H) .
Analytical HPLC 15.02, 15.34min. LC-MS (ES+) m/e=507.3
(MH+)

(2 -Cyclohexyloxy- 5 -oxo-1e trahydro- furan-3 -yl)-carbamic
acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using
cyclohexanol to afford the title compound as a mixture
of diastereomers (pale yellow oil) (4.62g, 85% yield).
Flash column chromatography using hexanes/EtOAc (90/10
to 80/20) gave 394 mg (7% yield) of the syn
diastereomer of the title compound. 1H NMR (500MHz,
CDCl3) δ.1.11-2.09 (m, 10H) , 2.35-2.61 (dd, 1H) , 2.72-
2.98 (dd, 1H), 3.60-3.83 (m, 1H), 4.32-4.72 (m, 3H),
5.06-5.43 (m, 2H) , 5.60 (d, 1H), 5.82-6.03 (m, 1H).
1-[2-(4-Acetylamino-3-chloro-benzoylamino)-propionyl]-
pyrrolidine-2-carboxylic acid (2-cyclohexyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98f) .
Prepared from syn-(2-cyclohexyloxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester and 97b following
the method used for 98a to give the title compound
;121mg, 33% yield). 1H-NMR (500MHz, CD3OD) δ 1.06-1.61
(m 9H) , 1.61-2.37 (m, 7H) , 2.22 (s, 3H) , 2.52-2.81 (m,
2H) , 3.45-3.78 (m, 2H) , 3.84-3.97 (m, 1H) , 4.42-4.57

(m, 1H) , 4.57-4.69 (m, 1H) , 5.67-5.81 (m, 1H) , 7.72-
7.89 (m, 1H), 7.89-8.12 (m, 2H). Analytical HPLC
9.84min. LC-MS (ES+) m/e=563.3 (MH+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclohexyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98g) .
Prepared from syn- (2-cyclohexyloxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester and 97a following
the method used for 98a to give the title compound
(153mg, 47% yield). 1H-NMR (500MHz, CD3OD) δ 1.06-2.38
(m, 14H), 1.42 (d, 3H), 2.50-2.66 (m, 1H) , 2.69-2.82
(dd, 1H) , 3.06-3.75 (m, 2H) , 3.80-3.94 (m, 1H) , 4.40-
4.52 (m, 1H) , 4.57-4.65 (tn, 1H) , 4.70-4.80 (m, 1H) ,
5.72 (d, 1H) , 6.71 (m, 1H) , 7.50-7.63 (m, 1H) , 7.78 (d,
0.6H), 8.42 (d, 6.4H). Analytical HPLC 10.30min. LC-MS
(ES+) m/e=521.2 (MH+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl]-
pyrrclidine-2-carboxylic acid (2-ethoxy-5-oxo-
terrahydro-furan-3-yl;-amide (98h).

Prepared from (2-ethoxy-5-oxo-tetrahydro-furan-3-yl) -
carbamic acid allyl ester and 97a following the method
used for 98a. The title compound was isolated as a
white solid (195mg, 82% yield). 1H-NMR (500MHz, CD3OD)
δ 1.32-1.55 (m, 3H) , 1.58-1.77 (m, 3H) , 1.98-2.54 (m,
4H), 2.68-2.76 (d, 0.3H), 2.79-2.89 (m, 0.7H), 2.96-
3.10 (m, 0.7H), 3.18-3.27 (dd, 0.3H), 3.72-4.18 (m,
4H), 4.46-5.12 (m, 3H), 5.60 (s, 0.4H), 5.74-5.84 (m,
0.6H), 7.03 (d, 0.8H), 7.75-7.86 (m, 1H), 8.01(d,
0.7H), 8.35 (d, 0.3H), 8.74 (d, 0.2H). Analytical HPLC
8.31min. LC-MS (ES+) m/e=467.3 (MH+) .

[5-OXO-2- (tricyclo [3 .3 .l.l°'°]dec-2-yloxy) -tetrahydro-
furan-3-yl]-carbamic acid allyl ester.
Prepared from 3-ariyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 4 0 using 2-
adamantanol (6.21g, 5 equivalents) to afford the title
compound as a pale yellow oil (1.52g, 61% yield) . 1H
NMR (500MHz, CDCl3) .δ.1.38-2.22 (m, 14H) , 2.40 (d,
0.2H), 2.53 (dd, 0.7H), 2.87 (dd, 0.7H), 2.87 (dd,
0.8H), 3.00-3.12 (m, 0.3H), 3.84-3.97 (m, 1H), 4.40-
4.71 (m, 3H) , 5.18-5.44 (m, 2H) , 5.53-5.69 (m, 1H) ,
£.82-6.02 (m, 1H).
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid [5-oxo-2-

(tricyclo[3.3.1.1°'°] dec-2-yloxy)-tetrahydro-furan-3-yl]
amide (98i).
Prepared from [5-oxo-2-(tricyclo[3.3.1.1°'°) dec-2-
yloxy)-tetrahydro-furan-3-yl]-carbamic acid allyl ester
and 97a following the method used for 98a. The title
compound was isolated as a white solid (76mg, 13%
yield). 1H-NMR (500MHz, CD3OD) δ 1.38-2.22 (m, 14H) ,
2.40 (d, 0.2H), 2.53 (dd, 0.7H), 2.87 (dd, 0.8H), 3.00-
3.12 (m, 0.3H), 3.84-3.97 (m, 1H), 4.40-4.71 (m, 3H) ,
5.18-5.44 (m, 2H), 5.53-5.69 (m, 1H), 5.82-6.02 (m,
1H). Analytical HPLC. 11.89min. LC-MS (ES+) m/e= 573.2
(MH+).

1- [2- (4-Acetylamino-5-chloro-2-methoxy-benzoylamino.) -
propionyl] -pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (98j) .
Prepared from syn- (2- [2- (2-benzyloxy-5-oxo-tetrahydro-
furan-3-ylcarbamoyl) -pyrrolidin-1-yl] -l-methyl-2-oxo-
ethyl}-carbamic acid tert-butyl ester and 97c following
the procedure used for 98a to afford.the title compound
(222mg, 82% yield). 1H-NMR (500MHz, CD3OD) δ 1.23 (d,
0.6H), 1.42 (d, 2.4H), 1.72-2.27 (m, 4H) , 2.23 (s, 3H) ,
2.63 (dd, 1H), 2.77-2.88 (m, 1H), 3.43-3.52 (m, 0.5H),
2.56-3.71 (m, 1.5H), 3.74-3.85 (m, 1H), 3.98 (s, 3H), (m, 1.5H), 4.51-4.92 (m, 4.5H), 5.63-5.76 (m,
1H . 7.23-7.4C (in, 5H), 7.97 (s, 1H) , 8.45 (d, 1H) ,

8.69-8.80 (m, 1H) Analytical HPLC ll.63min LC-MS (ES+)
m/e= 601.2 (MH+) .

Synthesis of 1-[2-(4-amino-3-chloro-benzoylamino)-
propionyl]-pyrrolidine-2-carboxylic acid (2-ethoxy-5-
oxo-tetrahydro-furan-3-yl)-amide (98k).
Prepared from anti-(2-ethoxy-5-oxo-tetrahydro-furan-3-
yl)-carbamic acid allyl ester and 97a following the
method used for 98a to afford 175 mg of title compound
(59%). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.10-1.28 (m,
3H) , 1.42 (d, 0.6H), 1.46 (d, 2.4H), 1.75-2.45 (m, 4H) ,
2.45-2.70 (m, 1H), 2.80-3.05 (m, 1H), 3.50-3.95 (m,
4H), 4.20-4.75 (m, 3H) , 4.75-4.90 (m, 1H) , 5.32 (s,
0.8H), 5.38 (s, 0.2H), 6.80 (d, 1H) , 7.55-7.84 (m, 2H) .
Analytical HPLC: 10.47 min. LC-MS (ES+): m/e = 467.3
(M+H+).

Synthesis of 1-[2-(4-amino-3, 5-dichloro-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (2-ethoxy-5-
oxo-tetrahydro-furan-3-yl)-amide (981).
Prepared from (2-ethoxy-5-oxo-tetrahydro-furan-3-yl) -
carbamic acid allyl ester and 1-[2-(4-amino-3,5-
dichlcrc-benzoylamino!-propionyl]-pyrrolidine-2-

carboxylic acid tert-butyl ester according to the
method used for 98a to afford 158 mg of title compound
(54% yield). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.08-
1.30 (m, 3H) , 1.32-1.52 (m, 3H) , 1.72-2.44 (m, 4H) ,
2.40-3.05 (m, 2H) , 3.50-3.97 (m, 4H), 4.25-4.70 (m,
3H) , 4.70-4.86 (m, 1H) , 5.33 (s, 0.4H), 5.47 (s, 0.1H),
5.56 (d, 0.4H), 5.62 (d, 0.1H), 7.50 (s, 1H), 7.80 (s,
1H) . Analytical HPLC: 10.84 min. LC-MS (ES+): m/e =
501.2 (M+H+).

1- [2- (4-amino-3-chloro-benzoylaadno) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98m) .
Prepared according to the procedure used to prepare 98a
using Cbz-Ala-D-pro-OH to afford 230 mg of title
compound (69% yield). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD)
δ 'l.30 (d, 1.2H); 1.45 (d, 1.8H), 1.62-2.40 (m, 4H) ,
2.40-3.10 (tn, 2H) , 3.30-3.97 (m, 2H) , 4.33-4.95 (m,
5H) , 5.30 (s, 0.5H), 5.68 (d, 0.5H), 6.80 (d, 1H) ,
7.25-7.95 (m, 7H). Analytical HPLC: 11.56, 11.91 min.
LC-MS (ES+): m/e = 529.2 (M+H+).


1- [2- (4-acetylamino-3-chloro-benzoylamino)-propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98n).
Prepared from 97b and syn-(2-benzyloxy-5-oxo-
tetrahydro-furan-3-yi)-carbamic acid allyl according to
the procedure used to prepare 98a to afford 210 mg of
title compound (64% yield). 1H-NMR (500 MHz, 1:1
CDCl3:CD3OD) δ 1.33 (d, 0.6H), 1.44 (d, 2.4H), 1.68-2.40
(m, 4H), 2.26 (s, 3H), 2.55-3.05 (m, 2H), 3.40-3.90 (m,
2H) , 4.20-4.95 (m, 5H) , 5.68 (d, 0.8H) , 5.84 (d, 0.2H),
7.15-8.30 (m, 8H) . Analytical HPLC: 15.67 tnin. LC-MS
(ES+): m/e = 571.1 (M+H+).

(2-Isopropoxy-5-oxo-tetrahydro-furan-3-yl) -carbamic
acid allyl ester
Prepared as described for compound 40 using isopropanol
to afford 3.80 grams (81% yield) of the title compound
as a colorless oil. 1H-NMR (500 MHz, CDCl3) δ 1.10-1.35
(m, 6H), 2.32-2.60 (m, 1H), 2.82 (dd, 0.5H), 3.02 (dd,
0.5H), 3.82-4.11 (m, 1H), 4.48-4.66 (m, 3H), 5.20-5.36
(m, 2H) , 5.54 (dd, 1H) , 5.82-6.05 (m, 1H) . LC-MS
(ES+): m/e = 244.2 (M+H+).
1- [2-(4-amino-3-chloro-benzoylamino)-propionyl]-
pyrrolidine-2-carboxylic acid (2-isopropoxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98o) .
Prepared from 97a and (2-isopropoxy-5-oxo-tetrahydro-
furar-3-yl -carbamic acid allyl ester according tc the

procedure used to prepare 98a to afford 200 mg of title
compound (66% yield). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.05-1.35 (HI, 6H), 1.35-1.50 (m, 3H) , 1.70-2.45 (m,
4H) , 2.45-3.05 (m, 2H) , 3.55-4.10 Ira, 3H) , 4.15-4.88
(m, 4H), 5.48 (S, 0.4H), 5.58 (s, 0.1H), 5.64 (d,
0.4K), 5.70 (d, 0.1H), 6.78 (d, 1H) , 7.58 (d, 1H) , 7.80
(s, 1H). Analytical HPLC: 12.19, 12.40 min. LC-MS
(ES+) : m/e = 581.2 (M+H+) .

1- [2- (4-acetyleunino-3,5-dichloro-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (98p).
Prepared from 1-[2-(4-acetylamino-3 , 5-dichloro-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester and syn- (2-benzyloxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester according to the
procedure used to prepare 98a to afford 23 0 mg of title
compound (72% yield). 1H-NMR (500 MHz, 1:1 CDCl3 :CD3OD)
δ1.36 (d, 0.6H), 1.47 (d, 2.4H), 1.68-2.47 (m, 4H) ,
2.23 (s, 3H) , 2.60-3.15 (m, 2H), 3.40-3.90 (m, 2H) ,
4.15-4.95 (m, 5H), 5.68 (d, 0.8H), 5.84 (d, 0.2H),
7.20-7.98 (m, 7H). Analytical HPLC: 13.07 min. LC-MS
(ES+): m/e = 605.1 (M+H+).


1- [2- (4-acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98q) .
Prepared from 97b and (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 215 mg of title compound (69% yield) . -"-H-NMR
(500 MHZ, 1:1 CDCl3:CD3OD) δ 1.35-1.90 (m, 11H), 1.90-
2.35 (m, 4H) , 2.24 (s, 3H) , 2.40-3.10 (m, 2H) , 3.50-
3.95 |n, 3H) , 4.15-4.90 (m, 3H) , 5.44 (s, 0.55H), 5.56
(s, 0.15H), 5.64 (d, 0.22H), 5.71 (d, 0.08H), 7.70-8.25
(m, 3H) . Analytical HPLC: 12.13 min. LC-MS (ES+) : m/e
= 54 9.2 (M+H+) .

Synthesis of 1- [2- (4-acetylamino-3-chloro-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
(2-ethoxy-5-oxo-tetrahydro-furan-3-yl)-amide (98r) .
Prepared from 97b and syn- (2-ethoxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester according to the
procedure used to prepare 98a to afford 68 mg of title
compound (24%). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.13
(t, 0.6H), 1.28 (t, 2.4H), 1.38 (d, 0.6H), 1.48 (d,
2.4H), 1.75-2.40 (m, 4H) , 2.22 (s, 3H) , 2.55-2.88 (m,
2K) , 3.50-3.92 (m, 4H) , 4.40-4.90 (m, 3H) , 5.57 (d,
0.8H1, 5.61 (d, 0.2H), 7.60-8.20 (m, 3H). Analytical
HPLC: E.64 mir.. LC-MS (ES*): m/e = 509.2 (M+H+).


Preparation of (2-cyclopentyliaeth.oxy-5-oxo-tetrahydro-
furan-3-yl)-carbamie acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for compound 4 0
using cyclopentylmethanol (6.5 mL, 60 mmol) to afford
2.98 grams (52% total yield) of the title compound as a
mixture of epimers. Purification provided 0.97 grams
(17% yield) of the 4(S), 5(R) as a colorless oil. 1H
NMR (500 MHZ, CDCl3) δ 1.19 (m, 2H), 1.54 (m, 4H) , 1.71
(m. 2H), 2.16 (m, 1H), 2.44 (dd, J=17.2, 10.4Hz, 1H),
2.82 (dd, J=17.2, 8.4Hz, 1H), 3.44 (dd, J=9.3, 7.2Hz,
1H), 3.71 (dd, J=9.3, 7.2Hz, 1H), 4.57 (m, 3H), 5.32
(m, 3H), 5.41 (d, J=S.2Hz, 1H), 5.91 (ddt, J=17.1,
10.4, 5Hz, 1H) ppm. LC-MS. (ES+): m/e=284.
Also isolated was epimer mixture (0.66 gram, 11% yield)
and the 4(S), 5(S) epimer (1.35 gram, 24% yield) as a
waxy solid. 1H-NMR (500MHZ, CDCl3) 6 1.20 (m, 2H) , 1.54
(m. 4H), 1.69 (m, 2H), 2.10 (m, 1H), 2.37 (d, J=8.1Hz,
1H), 2.97 (dd, J=18.0, 7.6Hz, 1H), 3.42 (dd, J=7.3,
1.7Hz, 1H), 3.49 (m, 2H), 3.64 (dd, J=9.0, 7.3Hz, 1H),
4.19 (br, 1H), 4.55 (m, 2H), 5.25 (m, 2H), 5.36 (s,
1H), 5.87 (m, 1H) ppm. LC-MS (ES+); m/e=284 (M+H).
1- [2- (4-amino-3-chloro-benzoylaniino) -propionyl] -
pyrrolidixie-2-carboxylic acid (2-cyclopentylmethoxy-5-
oxo-tetrahyd.ro-furan-3-yl)-amide (98s) -

Prepared from 97a and syn-(2-cyclopentylmethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 195 mg of title compound (51% yield). 1H-NMR
(500 MHz, 1:1 CDCl3:CD3OD) δ 1.15-1.90 (m, 11H) , 1.90-
2.40 (m, 5H), 2.55-2.76 (m, 2H), 3.50-3.90 (m, 4H),
4.38-4.92 (m, 3H), 5.53 (d, 0.8H), 5.57 (d, 0.2H), 6.78
(d, 1H), 7.50-8.15 (m, 2H). Analytical HPLC: 10.48
min. LC-MS (ES+) : m/e = 521.2 (M+H+) .

(5-oxo-2-(3-phenyl-propoxy)-tetrahydro-furan-3-yl)-
carbamic acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for compound 40
using 3-phenylpropanol to afford 1.15 grams (32% yield)
of the title compound as a colorless oil. 1H-NMR (500
MHz,.CDCl3) δ 1.82-2.05 (m, 2H) , 2.38 (dd, 1H) , 2.68 (m,
2H), 2.82 (dd, 1H), 3.55-3.65 (m, 1H), 3.82-3.92 (m,
1H) , 4.48-4.72 (m, 3H) , 5.12-5.59 (m, 3H) , 5.82-6.03
(m, 1H) , 7.11-7.45 (m, 5H) . Analytical HPLC: 9.08 min.
LC-MS (ES+): m/e = 320.2 (M+H+) .
1- [2- (4-amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (5-oxo-2-(3-phenyl-
propoxyl)-tetrahydro-furan-3-yl)-amide (98t).
Prepared from 97b and syn-(5-oxo-2-(3-phenyl-propoxyl) -
tetrahyaro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 200 mg of title compound (57% yield) . 1H-NMR

(500 MHz, 1:1 CDCl3:CD3OD) δ 1.34 (d, 0.6H), 1.44 (d,
2.4H), 1.75-2.40 (m, 6H), 2.50-2.95 (m, 4H), 3.47-3.95
(m, 4H) , 4.38-4.82 (m, 3H), 5.52 (d, 0.8H), 5.56 (d,
0.2H), 6.75-8.25 (m, 8H). Analytical HPLC: 10.79 min.
LC-MS (ES+): m/e = 557.2 (M+H+).

Synthesis of 1- [2- (4-acetylamino-3-chloro-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
(2-cyclopentylmethoxy-5-oxo-tetrahydro-furan-3-yl) -
amide (98u) .
Prepared from 97b and syn- (2-cyclopentylmethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 215 mg of title compound (67% yield) . 1H-NMR
(500 MHZ, 1:1 CDCl3:CD3OD) δ 1.38 (d, 0.6H), 1.47 (d,
2.4H), 1.11-1.88 (m, 8H) , 1.92-2.40 (m, 5H), 2.24 (s,
3H), 2.53-2.86 (m, 2H) , 3.30-3.90 (m, 4H), 4.38-4.89
(m, 3H), 5.53 (d, 0.8H), 5.60 (d, 0.2H), 7.68-8.22 (m,
3H). Analytical HPLC: 9.90 min. LC-MS (ES+): m/e =
563.3 (M+H+) .

1- [2- (4-acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (5-oxo-2-(3-phenyl-
propoxyl)-tetrahydro-furan-3-yl)-amide (98v).

Prepared from 1-[2-(4-acetylamino-3-chloro-
benzoylamino)-propionyl]-pyrrolidine-2-carboxylic acid
tert-butyl ester and syn-(5-oxo-2-(3-phenyl-propoxyl)-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 238 mg of title compound (75% yield). 1H-NMR
(500 MHz, 1:1 CDCl3:CD3OD) δ 1.33 (d, 0.6H), 1.56 (d,
2.4H), 1.78-2.45 (m, 6H) , 2.27 (s, 3H) , 2.53-2.97 (m,
4H) , 3.53-3.94 (m, 4H), 4.47-4.86 (m, 3H), 5.53 (d,
0.8H), 5.62 (d, 0.2H), 7.11-8.26 (m, 8H). Analytical
HPLC: 10.27 min. LC-MS (ES+) : m/e = 599.2 (M+H+) .

1- [2- (4-amino-3-trifluoromethyl-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-£uran-3-yl)-amide (98w) .
Prepared from {2-[2-(2-benzyloxy-5-oxo-tetrahydro-
furan-3-ylcarbmoyl) -pyrrolidin-1-yl] -1-methyl-2-oxo-
ethyl}-carbamic acid tert-butyl ester and 4-amino-3-
trifluoromethyl-benzoic acid according to the procedure
used to prepare 98a to afford 56 mg of title compound
(48% yield). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.20-
1.55 (m, 3H) , 1.75-2.50 (m, 4H), 2.50-3.10 (m, 2H) ,
3.50-4.00 (m, 2H) , 4.30-5.00 (m, 5H) , 5.42 (s, 0.4H),
5.51 (s, 0.2H), 5.62 (d, 0.3H),5.78 (d, 0.1H), 6.84
(d, 1H), 7.20-8.15 (m, 7H). Analytical HPLC: 14.90,
15.20 min. LC-MS (ES+) : m/e = 563.2 (M+H+) .

the procedure used to prepare 98a to afford 106 mg of
title compound (65% yield). 1H-NMR (500 MHz, 1:1
CDCl3:CD3OD) δ 1.10-1.55 (m, 3H) , 1.75-2.30 (m, 4H) ,
2.45-3.15 (m, 2H) , 2.64 (s, 6H) , 3.40-3.95 (m, 2H) ,
4.15-4.95 (m, 5H). 5.47 (s, 0.35H), 5.54 (s, 0.15H),
5.67 (d, 0.4H), 5.77 (d, 0.1H) , 7.20-7.70 (m, 7H) .
Analytical HPLC: 12.21, 12.51 min. LC-MS (ES+): m/e =
559.2 (M+H+).

1- [2- (4-amino-2,3,5,6-tetrafl'uoro-ben2oylamino)-
propionyl]-pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (98z) .
Prepared from (2- [2- (2-benzyloxy-5-oxo-tetrahydro-
furan-3-ylcarbmoyl) -pyrrolidin-l-yl] -l-methyl-2-oxo-
ethyl}-carbamic acid tert-butyl ester and 4-amino-
2, 3,5,6-tetrafluoro-benzoic acid according to the
procedure used to prepare 98a to afford 58 mg of title
compound (73% yield). 1H-NMR (500 MHz, 1:1 CDCl3:CD3OD) δ 1.30-1.50 (m, 3H) , 1.62-2.35 (m, 4H) , 2.45-3.12 (m,
2H) , 3.50-3.90 (m, 2H), 4.20-4.95 (m, 5H) , 5.42 (s,
0.4H), 5.52 (s, 0.1H), 5.64 (d, 0.4H), 5.82 (d, 0.1H),
7.25-7.65 (m, 5H). Analytical HPLC: 16.56, 16.90 min.
LC-MS (ES+) : m/e = 567.2 (M+H+) .


l-{2- [3-chloro-4- (2 , 2 -dimethylpropionylamino) -
benzoylamino] -propionyl}-pyrrolidine-2-carboxylic acid
(2-benzyloxy-5-oxo-tetrahydro-furan-3-yl) -amide (98aa) .
To a suspension of 98b (100 mg, 0.19 mmol) and poly (4-
vinylpyridine) (2 00 mg) was added pivaloyl chloride
(70uL, 0.57 mmol). The resulting suspension was stirred
overnight at room temperature the filtered and diluted
with EtOAc (25 mL) . The organic layer was washed with
10% NaHCO3 (2 x 25 mL) , saturated NaCl (1 x 25 mL) ,
dried (MgSO4), and evaporated to dryness to afford 98
mg of title compound (8 5% yield) after chromatography.
1H-NMR (500 MHZ, 1:1 CDCl3:CD3OD) δ 1.10-1.55 (m, , 3H) ,
1.38 (s, 9H) , 1.65-2.40 (m, 4H) , 2.60-3.10 (m, 2H) ,
3.46-3.88 (m, 2H) , 4.20-4.95 (m, 5H) , 5.62 (d, 0.8H),
5.78 (d, 0.2H), 7.15-8.30 (m, 8H) . Analytical HPLC:
11.82 min. LC-MS (ES+) : m/e = 613.2 (M+H+) .

1- [2- (3-chloro-4-propionylamino) -benzoylamino) -
propionyl] -pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (98ab) .
Prepared from 98b and propionyl chloride according to
the procedure used to prepare 98aa to afford 104 mg of
title compound (95% yield). 1H-NMR (500 MHz, 1:1
CDCl3: CD3OD) δ 1.16 (t, 0.6H), 1.18 (d, 0.6H), 1.27 (t,
2.4H), 1.38 (d, 2.4H), 1.72-2.35 (m, 4H), 2.45-2.58 (m,
2E) , 2.58-3.05 (m, 2H) , 3.45-3.85 (m, 2H) , 4.20-4.88
(m, 5E) , 5.64 (d, 0.8K), 5.76 (d, 0.2H), 7.20-8.35 (m,
Analytical HPLC: 9.89 min. LC-MS (ES+) : m/e =
SEE.2 (M+H).


1- [2- (3-chloro-4-phenylacetylamino) -benzoylamino) -
propionyl] -pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide (98ac) .
Prepared from 98b and phenylacetyl chloride according
to the procedure used to prepare 98aa to afford 85 mg
of title compound (77% yield). 1H-NMR (500 MHz, 1:1
CDCl3: CD3OD) δ 1.18 {d, 0.6H), 1.40 (d, 2.4H), 1.72-
2.38 (m, 4H) , 2.58-3.05 (m, 2H), 3.46-3.78 (m, 2H),
3.85 (s, 2H) , 4.18-4.92 (tn, 5H) , 5.63 (d, 0.8H), 5.75
(d, 0.2H), 7.15-8.34 (m, 13H). Analytical HPLC: 11.63
min. LC-MS (ES+) : m/e = 647.2 (M+H+) .

1- [2- (3-ch.loro-4-methyl-butyrylamino) -benzoylamino) -
propionyl] -pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-£uran-3-yl)-amide (98ad).
Prepared from 98b and isovaleryl chloride according to
the procedure used to prepare 98aa to afford 60 mg of
title compound (58% yield). 1H-NMR (500 MHz, 1:1
CDCl3:CD3OD) δ 1.07 (d, 5H), 1.15 (d, 0.8H), 1.27 (d,
1H) , 1.45 (d, 2.2H), 1.67-2.30 (m, 5H) , 2.34 (d, 2H) ,
2.58-3.05 (m, 2H) , 3.48-3.88 (m, 2H) , 4.10-4.98 (m,
5H) , 5.68 (d, 0.7H), 5.78 (m, 0.3H), 7.18-8.33 (m, 8H) .

Analytical HPLC: 10.74 min. LC-MS (ES+) : m/e = 613.2
(M+H+).

1- [2- (4-Methoxy-3, 5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-ethoxy-5-oxo-
tetrahydro-furan-3-yl) -amide (98ae) .
Prepared from 1- [2- (4-methoxy-3, 5-dimethyl-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester and syn-(2-ethoxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester according to the
procedure used to prepare 98a to afford 174 mg (81%
yield) of the title compound. 1H-NMR (500 MHz, CDCl3) :
5 1.04 (t, 0.45H), 1.27 (t, 2.55H), 1.34-1.45 (m, 3H),
1.95-2.45 (m, 10H) , 2.78-2.84 (m, H) , 3.60-3.90 (m,
8H), 4.50-4.70 (m, 2H), 4.90-4.94 (m, H), 5.45 (d,
0.85H), 5.61 (d, 0.15H), 6.99 (d, H), 7.15 (d, H) , 7.45
(s, '2H) ; retention, time on analytical HPLC: 10.09 min;
LC-MS: m/z = 476 (M+H+).

1- [2- (4-Methoxy-3 , 5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-ethoxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98a£).
Prepared from 1- [2- (4 -methoxy-3 , 5-dimethyl -
benzoyl amine)-propionyl] -pyrrolidine-2-carboxylic acid

tert-butyl ester and anti- (2-ethoxy-5-oxo-tetrahydro-
furan-3-yl) -carbamic acid allyl ester according to the
procedure used to prepare 98a to afford 168 mg (77%
yield) of the title compound. 1H-NMR (500 MHz, CDCl3) :
6 1.10-1.35 (m, 3H), 1.35-1.60 (m, 3H) , 1.90-2.45 (m,
10H) , 2.60-3.00 (m, H) , 3.55-3.95 (m, 8H) , 4.15-4.60
(m, 2H) , 4.83-5.00 (m, H) , 5.29 (s, H) , 6.95-7.06 (m,
H) , 7.50 (s, 2H) , 7.92 (d, H) ; retention time on
analytical HPLC: 10.14 min; LC-MS: m/z = 476 (M+H+).

1- [2- (4-Methoxy-3, 5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98ag) .
Prepared from 1- [2- (4-methoxy-3, 5-dimethyl-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester and syn- (2-benzyloxy-5-oxo-tetrahydro-
furan-3-yl) -carbamic acid allyl ester (40) according to
the procedure used to prepare 98a to afford 4 06 mg
(yield 71%) of the title compound. 1H-NMR (500 MHz,
CDCl3) : 6 1.09 (d, 0.6H), 1.35 (m, 2.4H), 1.90-2.20 (m,
3H) , 2.22-2.50 (m, 10H), 2.84-2.90 (m, H) , 3.52-3.62
(m, 1.6H), 3.65-3.80 (m, 3.4H), 4.10-4.40 (m, H) , 4.50-
4.75 (m, 3H) , 4.82-4.95 (m, 2H) , 5.54 (d, 0.8H), 5.80
(d, 0.2H), 6.87 (d, H) , 7.10-7.40 (m, 6H) , 7.45 (s,
2E: ; retention time on analytical HPLC: 16.71 min1; LC-
MS: m/z = 538 (M+H+).


1- [2- (4-Allyloxy-3,5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98ah).
Prepared from 1- [2- [4-allyloxy-3, 5-dimethyl-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester and 40 according to the procedure used
to prepare 98a to afford 264 mg (yield 46%) of the
title compound. 1H-NMR (500 MHz, CDCl3) : δ 1.09-1.43
(m, 3H) , 1.90-2.20 (m, 3H), 2.20-2.38 (m, 7H) , 2.38-
2.52 (m, H), 2.80-2.95 (m, H), 3.52-3.67 (m, H), 3.70-
3.80 (m, H) , 4.10-4.40 (m, 2H) , 4.40-4.95 (m, 5H) ,
5.26-5.55 (m, 3H) , 6.00-6.14 (m, H) , 6.87 (d, H) , 7.10-
7.70 (m, 8H); retention time on analytical HPLC: 18.56
and 18.92 min1; LC-MS: m/z = 564 (M+H+)

{2- [1R- (2S-Isopropyl-5R-methyl-cyclohexyloxy) ] -5-oxo-
tetrahydro-furan-3-yl}-carbamic acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using (1R,
2S, 5R)-(-)-menthol to afford 0.32 g of syn
diastereomer (lower Rf) of the title compound and 4.25

g of the mixture of anti/syn diastereomers (overall
yield 67%). 1H-NMR (500 MHz, CDCl3) mixture: δ 0.70-
1.05 (m, 13H), 1.20-1.47 (m, 2H) , 1.60-1.80 (m, 2H),
1.94-2.20 (m, 2H) , 2.35-2.50 (m, H) , 2.82-3.04 (m, H) ,
3.40-3.61 (m, H) , 4.43-4.70 (m, 3H) , 5.15-5.35 (m, 2H) ,
5.48-5.61 (m, H) , 5.90-5.94 (m, H) ; for syn
diastereomer 0.70-1.05 (m, 13H), 1.20-1.47 (m, 2H),
1.60-1.80 (m, 2H), 1.94-2.18 (m, 2H), 2.40-2.50 (m, H),
2.82-2.92 (m, H), 3.54-3.61 (m, H), 4.45-4.70 (m, 3H),
5.18-5.35 (m, 2H) , 5.58-5.61 (m, H) , 5.90-5.93 (m, H) ;
LC-MS: m/z = 340 (M+H+) for the mixture of anti/syn
diastereomers.
4-Benzyloxy-3, 5-dimethyl-benzoic acid.
Prepared by the method used to synthesize 4-allyloxy-
3,5-dimethyl-benzoic acid to afford 2.43 g (yield 56%)
of the title compound. 1H-NMR (500 MHz, CDCl3) : δ 4.87
(s, 2H), 7.36-7.48 (m, 5H), 7.92 (s, 2H); LC-MS: m/z =
255 (M-H+).
1- [2- (4-Benzyloxy-3, 5-dimethyl-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid {2-[lR-(2S-
isopropyl-5R-methyl-cyclohexyloxy) ] -5-oxo-tetrahydro-
furan-3-yl}-amide (98ai).
Prepared from 1- [2- (4-benzyloxy-3, 5-dimethyl-
benzoylamino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester {2-[1R-(2S-Isopropyl-5R-methyl-
cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-carbamic
acid allyl ester according to the procedure used to
prepare 98a to afford 130 mg (yield 39%) of the title
compound. 1H-NMR (500 MHz, CDCl3) : δ 0.45-1.10 (m,
12H) , 1.15-1.90 (m, 8H) , 1.90-2.45 (m, 12H) , 2.80-2.84
(m, H) , 3.50-3.85 (m, 3H) , 4.45-4.70 (m, 2H) , 4.80-4.95
(m, 3H), 5.62 (d, H) , 7.05 (d, H) , 7.17 (d, H) , 7.30-

7.60 (m, 7H) , 1.62-7.75 (m, H); retention time on
analytical HPLC: 15.90 and 16.08 min; LC-MS: m/z = 662
(M+H+).

1- [2- (4-Hydroxy-3,5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid {2- [1R- (2S-isopropyl-5R-
methyl-cyclohexyloxy) ] -5-oxo-tetrahydro-£uran-3-yl}-
amide (98aj).
A solution of 1-[2-(4-benzyloxy-3,5-dimethyl-
benzoylamino)-propionyl]-pyrrolidine-2-carboxylic acid
{2-[1R-(2S-isopropyl-5R-methyl-cyclohexyloxy) ] -5-oxo-
tetrahydro-furan-3-yl}-amide (110 mg, 0.17 mmol) in
ethyl acetate (2 mL) was stirred with 10% palladium on
carbon (20 mg) under hydrogen atmosphere for 24 hours
then filtered through Celite and evaporated in vacuo.
The resulting residue was purified by chromatography
using CH2Cl2/methanol (99/1 to 96/4) to afford 58 mg of
the title compound. 1H-NMR (500 MHz, CDCl3) : δ 0.70-
1.00 (m, 10H), 1.20-1.80 (m, 10H) , 1.90-2.40 (m, 11H),
2.82-2.86 (m, H) , 3.57-3.78 (m, 3H), 4.55-4.67 (m, 2H) ,
4.90-4.94 (m, H), 5.29 (s, H), 5.62 (d, H), 6.90 (d,
H) , 7.14 (d, H) , 7.42 (s, 2H); retention time on
analytical HPLC: 12.84 and 13.05 min; LC-MS: m/z = 572
(M+H+).


1- [2- (4-Hydroxy-3,5-dimethyl-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (98ak) .
A solution of 98ah (230 mg, 0.41 mmol) in CH2Cl2 (10
mL) was treated with DMBA (65 mg, 0.42 mmol) and
Pd(PPh3)4 (50 mg) at room temperature for 20 hours.
The mixture was concentrated to dryness in vacuo and
purified by flash chromatography using CH2Cl2 /methanol
(99.5/0.5 to 97/3) to afford 181 mg of the title
compound. 1H-NMR (500 MHz, CDCl3) : δ 1.08 (d, 0.75H),
1.20-1.35 (m, 2.25H), 1.70-2.50 (m, 12H), 2.80-2.90 (m,
H) , 3.50-3.65 (m, H), 3.70-3.80 (m, H) , 4.10-4.25 (m,
H) , 4.35-4.98 (m, 3H) , 5.53 (d, 0.75H), 5.85 (d,
0.25H), 6.81 (d, H), 7.13-7.60 (m, 8H); retention time
on analytical HPLC: 10.38 and 10.56 min; LC-MS: m/z =
524 (M+H+).

1- [2- (4-Dimethylamino-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98al) .
Prepared from 1- [2- (4-dimethylamino-benzoylamino) -
propionyl] -pyrrolidine-2-carboxylic acid tert-butyl

ester and syn- (2-benzyloxy-5-oxo-tetrahydro-furan-3-
yl)-carbamic acid allyl ester according to the
procedure used to prepare 98a to afford 6 0 mg (45%
yield). 1H-NMR (500 MHz, CDCl3) : δ 1.04 (d, 0.75H),
1.35 (d, 2.25H), 1.80-2.50 (m, 5H) , 2.75-3.20 (m, 8H) ,
3.45-3.75 (m, 2H) , 4.05-4.20 (m, 0.5H), 4.30-4.80 (m,
3.5H), 4.80-4.95 (m, 1.5H), 5.52 (d, H) , 5.75-6.00 (m,
0.5H), 6.60-6.90 (m, 3H) , 7.10-7.50 (m, 4H) , 7.50-7.80
(m, 2H) ; retention time on analytical HPLC: 10.46 min;
LC-MS: m/z = 523 (M+H+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid {2R- [1R- (2S-isopropyl-5R-
methyl-cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-
amide (98am) .
Prepared from 1-[2-(4-amino-3-chloro-benzoylamino) -
propionyl]-pyrrolidine-2-carboxylic acid tert-butyl
ester (97a) and syn-{2-[1R-(2S-isopropyl-5R-methyl-
cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-carbamic
acid allyl ester according to the procedure used to
prepare 98a to afford 103 mg (yield 67%) of the title
compound. 1H-NMR (500 MHz, CDCl3) : δ 0.70-1.10 (m,
12H), 1.20-1.50 (m, 5H) , 1.50-1.85 (m, 2H), 1.90-2.30
(m, 5H), 2.75-2.85 (m, H) , 3.50-3.70 (m, 2H), 3.70-3.82
(m, H), 4.20-4.65 (m, 4H) , 4.80-4.95 (m, H), 5.61 (d,
H), 6.70-6.73 (m, H), 6.95 (d, H), 7.15 (d, H), 7.49-

7.51 (m, H) , 7.73 (s, H) ; retention time on analytical
HPLC: 12.88 min; LC-MS: m/z = 577 (M+H+).

{2- [IS- (2R-Isopropyl-5S-methyl-cyclohexyloxy) ] -5-oxo-
tetrahydro-furan-3-yl}-carbamic acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using (IS,
2R, 5S)-( + )-menthol to afford 855 mg of anti
diastereomer (higher Rf) of the title compound, 503 mg
of syn diastereomer (lower Rf) and 459 mg of the
mixture of anti/syn diastereomers (overall yield 66%) .
1H-NMR (500 MHz, CDCl3) anti diastereomer: δ 0.74-1.00
(m, 12H), 1.20-1.45 (m, 2H), 1.58-1.72 (m, 2H) , 1.98-
2.12 (m, 2H), 2.18-2.40 (m, H), 2.98-3.03 (m, H) , 3.49-
3.54 (m, H), 4.17 (br, H) , 4.59 (br, 2H), 4.97 (br, H),
5.22-5.33 (m, 2H) , 5.58 (s, H) , 5.87-5.93 (in, H) ; for
syn diastereomer 0.75-1.02 (m, 12H), 1.25-1.45 (m, 2H) ,
1.57-1.70 (m, 2H), 2.00-2.16 (m, 2H), 2.40-2.52 (m, H) ,
2.78-2.90 (m, H), 3.40-3.50 (m, H) , 4.58 (br, 2H) ,
5.24-5.35 (m, 2H), 5.51-5.52 (d, H) , 5.85-5.98 (m, H) ;
LC-MS: m/z = 34 0 (M+H+) for both of diastereomers.
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid {2R- [IS- (2R-isopropyl-5S-
methyl-cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-
amide (9 8an).

5.20-5.50 (m, 3H) , 5.89-5.95 (m, H) ; LC-MS: m/z = 298
(M+H+) for both of diastereomers.
1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclohexylmetb.oxy-5-
oxo-tetrahydro-furan-3-yl) -amide (98ao).
Prepared from 97b and syn- (2-cyclohexylmethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 212 mg (64% yield) of the title compound. 1H-
NMR (500 MHz, CDCl3): 8 0.70-1.30 (m, 5H) , 1.30-1.85 (m,
9H) , 1.85-2.60 (m, 8H), 2.75-3.00 (m, H) , 3.10-3.80 (m,
4H) , 4.30-4.95 (m, 3H) , 5.42 (d, 0.85H), 5.62 (d,
0.15H), 6.87 (d, 0.15H), 7.08 (d, 0.85H), 7.25 (d, H) ,
7.60-7.90 (m, 3H), 8.08 (d, 0.15H), 8.50 (d, 0.85H);
retention time on analytical HPLC: 11.81 min; LC-MS:
m/z = 577 (M+H+).

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid {2R- [1S- (2R-isopropyl-5S-
methyl-cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-
amide (98ap).
Prepared from 97b and syn- {2- [1S- (2R-isopropyl-5S-
methyl-cyclohexyloxy) ] -5-oxo- tetrahydro-furan-3 -yl} -
carbamic acid allyl ester according to the procedure
used to prepare 98a to afford 223 mg (63% yield) of the
title compound. 1H-NMR (500 MHz, CDCl3) : δ 0.70-1.15

(m, 12H) , 1.20-1.85 (m, 8H), 1.85-2.60 (m, 9H) , 2.74-
2.88 (m, H) , 3.35-3.85 (m, 3H), 4.40-4.55 (m, H), 4.65-
4.78 (m, H) , 4.88-4.91 (m, H) , 5.53 (d, H) , 7.00-7.25
(m, 2H), 7.60-7.90 (m, 3H), 8.50 (d, H) ; retention time
on analytical HPLC: 13.31 min; LC-MS: m/z = 619 (M+H+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclohexylmethoxy-5-
oxo-tetrahydro-furan-3-yl)-amide (98aq) .
Prepared from 97a and syn- (2-cyclohexylmethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester '
according to the procedure used to prepare 98a to
afford 113 mg (56% yield) of the title compound. 1H-
NMR (500 MHZ, CDCl3): δ 0.70-1.35 (m, 5H) , 1.35-1.90 (m,
8H), 1.90-2.20 (m, 3H), 2.30-2.60 (m, H) , 2.80-3.00 (m,
H) , 3.15-3.80 (m, 4H) , 4.28-4.75 (m, 4H) , 4.89-4.93 (m,
H) , 5.42 (d, H) , 6.74 (d, H) , 6.87 (d, H) , 7.30 (d, H) ,
7.51-7.53 (m, H) , 7.74 (d, H) ; retention time on
analytical HPLC: 12.02 min; LC-MS: m/z = 535 (M+H+).

(2-Butoxy-5-oxo-tetrahydro-£uran-3-yl) -carbamic acid
allyl ester.


(2-Isobutoxy-5-oxo-tetrahydro-furan-3-yl) -carbamic acid
allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using
isobutanol to afford 190 mg (yield 7.3%) of the title
compound as an anti diastereomer and 290 mg (yield 11%)
of the syix diastereomer. 1H-NMR (500 MHz, CDCl3) for
anti diastereomer: δ (higher Rf) 0.85-1.05 (m, 6H) ,
1.82-1.98 (m, H) , 2.37-2.42 (d, H) , 2.98-3.04 (m, H) ,
3.31-3.35 (m, H) , 3.55-3.58 (m, H) , 4.20-4.30 (t, H) ,
4.58 (br, 2H), 5.07 (br, H), 5.22-5.43 (m, 3H) , 5.84-
5.96 (m, H) , for syn diastereomer (lower Rf) 0.85-1.05
(m, 6H) , 1.88-1.95 (m, H), 2.40-2.51 (m, H) , 2.83-2.90
(m, H), 3.33-3.36 (m, H), 3.61-3.65 (m, H) , 3.87-3.88
(d, H), 4.40-4.68 (m, 3H), 5.20-5.40 (m, 2H) , 5.42-5.43
'(d, H) , 5.80-5.97 (m, H) ; LC-MS: m/z = 258 (M+H+) for
both of diastereomers.
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-isobutoxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98as).
Prepared from 97a and syn- (2-isobutoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford 93 mg (38% yield) of the title compound. 1H-NMR
(500 MHz, CDCl3) : δ 0.74-0.76 (t, 0.6H), 0.80-1.00 (m,
5.4H), 1.40-1.50 (m, 3H), 1.90-2.22 (m, 3H) , 2.33-2.45
(m, H) , 2.80-2.90 (m, H), 3.32-3.38 (m, H) , 3.55-3.80

(m, 3H), 4.38 (br, H), 4.50-4.60 (m, H), 4.70-4.80 (m,
H) , 4.90-5.00 (m, H) , 5.42-5.45 (m, H), 6.74-6.76 (d,
H) , 6.86-6.88 (d, H) , 7.31-7.33 (d, H) , 7.51-7.53 (m,
H), 7.74-7.75 (d, H) ; retention time on analytical
HPLC: 9.63 & 9.80 min; LC-MS: m/z = 495 (M+H+).

[2- (indan-2-yl)oxy-5-oxo-tetrahydro-£uran-3-yl] -
carbamic acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester (5.2 gram, 20 mmol) as described
for 40 using 2-indanol (8.05 gram, 60 mmol) to afford
4.10 grams (65% yield) of the title compound as a
mixture of epimers. Purification provided 1.76 gram
(28% yield ) of the 4 (S), 5 (R) epimer as a yellow oil.
1H NMR (500 MHZ, CDCl3) δ 2.42 (dd, J=17.2, 10.5Hz, 1H),
2.79 (dd, J=17.2, -8.4Hz, 1H) , 2.99 (dd, J=16.7, 4.1Hz,
1H), 3.04 (dd, J=16.7, 4.1Hz, 1H), 3.22 (dd, J=17.2,
6.6Hz, 1H) , 3.26 (dd, J=17.2, 6.6Hz, 1H), 4.53 (m, 3H) ,
4.70 (m, 1H) , 5.20 (m, 2H) , 5.60 (d, J=5.3Hz, 1H), 5.87
(m, 1H) , 7.17 (m, 4H) ppm. LC-MS (ES+) : m/e=318 (M+H) .
Analytical HPLC (Cl8 column): 17.094 min.
Also isolated was epimer mixture (0.75 gram, 12%
yield), and the 4(S), 5(S) epimer (1.59 gram, 25%) as a
white solid. 1H-NMR (500MHz, CDCl3) δ 2.38 (d, J=17.9Hz,
IK), 3.0 (m, 3H), 3.22 (m, 2H), 4.13 (m, 1H) , 4.58 (m,
2H), 4.68 (m, 2H), 4.98 (br s, 1H), 5.26 (m, 1H), 5.57
is, 1H), 5.88 (ddt, J=18.0, 11.1, 5.4Hz, 1H), 7.20 (m,

4H) ppm. LC-MS (ES+): m/e=318 (M+H). Analytical HPLC
(Cl8 column):17.025 (5.5%), 17.325 (94.5%) min.
1- 12- (4-amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid [2- (indan-2-yloxy) -5-oxo-
tetrahydro-furan-3-yl]-amide (98at) .
Prepared from 97a and [2-(indanol-2-yl]oxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford the title compound as a 71:29 mixture of epimers
as an off-white solid (0.20g, 58% yield). 1H-NMR (500
MHz, CDCl3) δ 1.0-1.5 (m, 3H) , 1.6-2.3 (m, 4H), 2.42 (m,
1H) , 2.6-3.4 (m, 6H), 3.5-4.1 (m, 3H), 4.2-4.9 (m; 4H) ,
5.65 (d, J=5.0HZ, 0.80H), 5.8 (m, 0.07H), 5.85 (d,
J=5.0Hz, 0.13H), 6.8-7.3 (m, 6H) , 7.4-7.9 (m, 3H) ppm.
Analytical HPLC (Cl8 column) 16.035 (71.4%), 16.476
(28.6%) min. LC-MS (ES+) : m/e=555 (M+H).

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid [2-(indan-2-yloxy)-5-oxo-
tetrahydro-furan-3-yl]-amide (98au) .
Prepared from 97b and [2-(indanol-2-yl] oxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford the title compound as a 76:24 mixture of epimers
as an off-white solid (0.22g, 57% yield) . 1H-NMR (500
MHZ, CDCl3) δ 1.08 (d, J=6.9Hz, 0.4H), 1.26 (d, J=6.9Hz,

0.6H), 1.35 (d, J=6.9HZ, 2H) , 1.8-2.3 (m, 3H) , 2.28 (s,
2H) , 2.29 (s, 1H) , 2.4 (m, 1H), 2.8 (m, 1H), 3.10 (m,
2H) , 3.27 (m, 2H) , 3.58 (m, 2H), 3.69 (m, 1H) , 4.5-4.9
(m, 4H), 5.65 (d, J=5.3HZ, 0.68H), 5.84 (d, J=5.3Hz,
0.18H), 6.38 (br, 0.14H), 6.9-7.7 (m, 6H), 7.6-7.9 (m,
3H) , 8.33 (br d, J=6.8HZ, 0.18H), 8.51 (br d, J=8.0HZ,
0.82H) ppm. Analytical HPLC (Cl8 column) 15.596
(76.2%), 15.932 (23.8%) min. LC-MS (ES+): m/e=597
(M+H).

1- 12- (4 - amino- 3 - chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98av) .
Prepared from 97a and syn- (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester
according to the procedure used to prepare 98a to
afford the title compound as an off-white solid (0.19g,
59% yield). 1H-NMR (500 MHz, CDCl3) δ 1.2-2.4 (m, 15H) ,
2.4-3.1 (m, 2H) , 3.6-3.9 (m, 2H) , 4.2-4.4 (in, 2H) , 4.5-
5.0 (m, 4H) , 5.40 (d, J=5.0HZ, 0.35H), 5.55 (d,
J=5.0Hz, 0.65H), 6.8-8.2 (m, 5H) ppm. Analytical HPLC
(Cl8 column) 14.065 min. LC-MS (ES+): m/e=507 (M+H).


1- [2- (3, 5-Dichloro-4-hydroxy-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (98aw) .
Prepared from 1-[2-(4-allyloxy-3,5-dimethyl-
benzoyl amino) -propionyl] -pyrrolidine-2-carboxylic acid
tert-butyl ester and syn-40 according to the procedure
used to prepare 98a to afford the title compound as a
pale yellow solid (l.087g, 64% yield) . 1H-NMR (500MHz,
CDCl3) δ 1.09 (d, J=6.9H2, 0.6H), 1.33 (d, J=6.9Hz,
2.4H), 1.96 (m, 1H) , 2.03 (m, 1H) , 2.10 (m, 1H) , 2.28
(m, 0.8H), 2.40 (dd, J=17.3, 10.2H2, 0.8H), 2.56 (m,
0.2H), 2.85 (dd, J=17.3, 8.5Hz, 0.8H), 3.09 (dd,
J=17.7, 10.2Hz, 0.2H), 3.57 (m, 1H) , 3.73 (dt, J=9.2,
7.9HZ, 0.8H), 4.09 (m, 0.2H), 4.21 (d, J=7.9Hz, 0.2H),
4.44 (d, J=9.8Hz, 0.2H), 4.55 (dd, J=8.0, 3.0HZ, 0.8H),
4.62 (d, J=11.6Hz, 1H), 4.70 (m, 1H), 4.80 (m, 1H) ,
4.89 (d, J=11.6Hz, 0.8H), 5.52 (d, J=5.2Hz, 0.8H), 5.82
(d, J=5.2Hz, 0.2H), 6.51 (br, 0.2H), 6.62 (br, 0.8H),
7.0-7.4 (m. 7H), 7.43 (s, 0.4H), 7.66 (d, J=1.0Hz,
1.6H) ppm. Analytical HPLC (Cl8 column) 10.135 min. LC-
MS (ES+): m/e= 564, 566 (6:4) (M+H).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-cyclopentyloxy-5-oxo-
tetrahydro-furan-3-yl) -amide (98ax) .
Prepared according to the procedure used to prepare
(98av) using anti-(2-cyclopentyloxy-5-oxo-tetrahydro-
furan-3-yl)-amide to afford the title compound as an

off-white solid (0.24 g, 74% yield). 1H-NMR (500MHz,
CDCl3) δ 1.41 (d, J=6.5Hz, 3H), 1.7 (m, 7H), 1.98 (br,
2H) , 2.13 (br, 2H), 2.27 (m, 1H), 2.69 (m, 1H) , 2.86
(dd, J=18.0, 6.8Hz, 0.7H), 2.98 (dd, J=18.3, 8.2Hz,
0.3H), 3.60 (br, 1.4H), 3.77 (br, 0.6H), 4.1-4.6 (m,
5H), 4.82 (m, 1H), 5.27 (m, 0.65H), 5,51 (d, J=5.3Hz,
0.05H), 5.59 (br s, 0.3H), 6.76 (br, 1H), 7.00 (br,
1H), 7.49 (br, 1H), 7.74 (br, 1H), 7.89 (br, 1H) ppm.
Analytical HPLC (Cl8 column) 9.756 min. LC-MS (ES+):
m/e=507 (M+H).

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid (2-ethoxy-5-oxo-
tetrahydro-furan-3-yl)-amide (98ay).
Prepared from (2-ethoxy-5-oxo-tetrahydro-furan-3-yl)-
carbamic acid allyl ester and 97b following the method
used for 98a. The title compound was isolated as a
white solid (51 mg, 18% yield). 1H-NMR (500MHz, 1:1
CDCl3:CD3OD) δ 1.08-1.35 (m, 3H) , 1.35-1.55 (m, 3H) ,
1.75-2.44 (m, 4H) , 2.26 (s, 3H) , 2.44-3.07 (m, 2H),
3.48-3.97 (m, 2H), 4.18-4.92 (m, 5H), 5.32 (d, 0.4H),
5.47 (d, 0.1H), 5.58 (d, 0.4H), 5.64 (d, 0.1H), 7.70-
8.35 (m, 3H). Analytical HPLC 10.37, 10.54 min. LC-MS
(ES+) m/e= 509 .'2 (M+H+) .


[2-(2-Chloro-ethoxy)-5-oxo-tetrahydro-furan-3-yl]-
carbamic acid allyl ester.
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester (5.2g, 20 mmol) as described for
40 using chloroethanol (4.05mL, 60 mmol) to afford
1.84g (35% yield) of the title compound as a mixture of
epimers. For the anti-diastereomer: 1H-NMR (500 MHz,
CDCl;-) δ 2.42 (dd, J=18.1Hz, 1H) , 3.00 (dd, J=18.1,
7.8Hz, 1H), 3.63 (m, 2H) , 3.85 (m, 1H) , 4.02 (m, 1H) ,
4.23 (m, 1H), 4.57(br s, 2H), 5.17 (br s, 1H), 5.22 (d,
H=11.5Hz, 1H), 5.29 (d, J=16.8Hz, 1H), 5.44 (s, 1H),
5.89 (m, 1H) ppm; LC-MS (ES+) m/e= 264 (M+H). For the
syrj-diastereoroer: 1H-NMR (500 MHz, CDCl3) δ 2.47 (dd,
J=17.3, 10.7Hz, 1H), 2.83 (dd, J=17.3, 8.4Hz, 1H), 3.65
(m, 2H), 3.83 (m, 1H), 4.11 (m, 1H), 4.57(m, 3H), 5.22*
(d, H=10.4Hz, 1H), 5.30 (d, J=17.2Hz, 1H), 5.33, (m,
1H), 5.47 (d, J=5.2Hz, 1H) , 5.89 (ddt, J=17.1, 11.0,
5.4Hz, 1H) ppm; LC-MS (ES+) m/e= 264 (M+H).
[2- (2-Morpholin-4-yl-ethcoey) -5-oxo-tetrahydro-£uran-3-
yl]-carbamic acid allyl ester.
Is prepared from [2-(2-chloro-ethoxy)-5-oxo-tetrahydro-
furan-3-yl]-carbamic acid allyl ester by reaction with
morpholine (2 eq) and KI (1 eq) in DMF.

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid [2-(2-morpholin-4-yl-
ethoxy)-5-oxo-tetrahydro-furan-3-yl]-amide (98az).
Is prepared from 97a and syn-[2-(2-morpholin-4-yl-
ethoxy)-5-oxo-tetrahydro-furan-3-yl]-carbamic acid
allyl ester following the method used for 98a.

[2-(4-Chloro-benzyloxy) -5-oxo-tetrahydro-furan-3-yl] -
carbamic acid allyl ester
Prepared from 3-allyloxycarbonylamino-4-hydroxy-butyric
acid tert-butyl ester as described for 40 using 4-
chlorobenzylalcohol to afford the title compound as a
white solid. Anti-diastereomer: HPLC (Cl8 column)
10.924 min; 1H-NMR (500 MHz, CDCl3) δ 2.41 (d, J=8.0Hz,
1H) , 3.02 (dd, J=18.1, 7.8Hz, 1H), 4.25 (br, 1H) , 4.56
(m, 2H),4.58 (d, J=11.7Hz, 1H) , 4.79 (d, J=11.7Hz, 1H),
4.99 (br, 1H), 5.22 (dd, J=10.4, 1.1Hz, 1H), 5.28 (dd,
J=17.2, 1.3HZ, 1H), 5.44 (s, 1H), 5.86 (m, 1H), 7.25
(d, J=8.4Hz, 2H), 7.32 (d, J=8.4Hz, 2H) ppm; LC-MS
(ES+) m/e=326 (M+H); Syn-diastereomer: HPLC (Cl8
column) 10.780 min; 1H-NMR (500 MHz, CDCl3) δ 2.47 (dd,
J=17.3, 10.5HZ, 1H) , 2.85 (dd, J=17.3, 8.4Hz, 1H) , 4.55
(m, 3H) , 4.58 (d, J=11.7Hz, 1H), 4.84 (d, J=11.7Hz,
1H), 5.23 (dd, H=10.4, 1.1Hz, 1H), 5.30 (d, J=16.6Hz,
1H) , 5.49 (d, J=5.0HZ, 1H), 5.89 (ddt, J=17.1, 11.0,
5.4HZ, 1H), 7.23 (d, J=8.3Hz, 2H), 7.31 (d, J=8.3Hz,
2H) ppm; LC-MS (ES+) m/e= 326 (M+H).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carboxylic acid [2- (4-chloro-benzyloxy) -
5-oxo-tetrahydro-furan-3-yl] -amide (98ba) .
Prepared from 97a and syn~ [2-(4-chloro-benzyloxy)-5~
oxo-tetrahydro-furan-3-yl]-carbamic acid allyl ester
following the method used for 98a to afford 154 mg (65%
yield) of the title compound as a pale pink solid.
HPLC (C18 column) 10.597 min; 1H-NMR (500 MHz, CDCl3) δ
1.14 (d, J=6.8HZ, 0.75H), 1.34, (d, J=6.8Hz, 2.25H),
1.6 (br, 0.25H), 1.91 (m, 1H), 2.03 (m, 1H) , 2.10 (m,
1H), 2.29 (m, 0.75H), 2.40 (dd, J=17.3, 10.3Hz, 0.75H),
2.51 (m, 0.25H), 2.82 (dd, J=17.3, 8.5Hz, 0.75H), 3.08
(dd, J=17.9, 10.9Hz, 0.25H), 3.58 (m, 1H), 3.72 (dd,
J=16.5, 8.7HZ, 0.75H), 4.10 (m, 0.25H), 4.22 (d,
J=8.0Hz, 0.25H), 4.39 (d, J=10.8Hz, 0.25H), 4.54 (dd,
J=9.1, 2.9HZ, 0.75H), 4.60 (d, J=11.9Hz, 0.75H), 4.68
(m, 1H) , 4.85 (d, J=11.7Hz, 0.75H), 4.86 (m, 1H), 5.49
(d, J=5.2Hz, 0.75H), 5.81 (d, J=5.2Hz, 0.25H), 6.2 (br,
0.25H), 6.74 (m, 2H), 7.05 (d, J=8.5Hz, 0.5H), 7.17 (d,
J=8.4Hz, 0.5H), 7.30 (m, 3.25H), 7.48 (dd, J=8.4,
2.0Hz, 0.75H), 7.56 (d, J=1.9Hz, 0.25H), 7.73 (d,
J=1.9Hz, 0.75H), 8.42 (d, J=5.7Hz, 0.25H) ppm; LC-MS
(ES+) m/e=563, 565 (M+H).


1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl ] -
pyrrolidine-2-carboxylic acid [2-(4-chloro-benzyloxy) -
5~oxo-tetrahydro-furan-3-yl]-amide (98bb) .
Prepared from 97b and syn- [2- (4-chloro-benzyloxy) -5-
oxo-tetrahydro-furan-3-yl]-carbamic acid allyl ester
according to the procedure used tc prepare 98a to
afford 165mg (64 yield) of the title compound as pale
yellow solid. HPLC (Cl8 column) 10.491 min; 1H-NMR (500
MHz, CDCl5) δ 1.16 (d, J=6.8Hz, 0.6H), 1.35, (d,
J=6.8Hz, 2.4H), 1.94 (m, 1H), 2.04 (m, 1H), 2.10 (m,
IK), 2.25 (s, 3H), 2.28 (m, 1H) , 2.40 (dd, J=17.3,
10.4Hz, 0.8H), 2.53 (m, 0.2H), 2.84 (dd, J=17.3, 8.5Hz,
0.8H), 3.02 (dd, J=17.5, 10.5Hz, 0.2H), 3.58 (m, 1H) ,
3.72 (ddd, J=17.2, 8.3, 8.3Hz, 0.8H), 4.13 (m, 0.2H),
4.22 (d, J=8.2Hz, 0.2H), 4.40 (d, J=10.9Hz, 0.2H), 4.54
(dd, J=8.1, 3.0Hz, 0.8H), 4.60 (d, J=11.8Hz, 0.8H),
4.69 (m, 1H), 4.85 (d, J=11.8Hz, 0.8H), 4.87 (m, 1H),
5.49 (d, J=5.2Hz, 0.8H), 5.80 (d, J=5.2Hz, 0.2H), 6.47
(br, 0.2H), 6.95 (d, J=8.3Hz, 0.8H), 7.05 (d, J=8.3Hz,
0.4K), 7.18 (d, J=8.3Hz, 0.4H), 7.29 (m, 3.2H), 7.49
(dd, J=8.6, 1.9Hz, 0.2H), 7.63 (dd, J=8.6, 1.9Hz,
C.8H), 7.74 (d, J=1.9Hz, 1H), 7.85 (d, J=1.9Hz, 0.8H),
8.25 (d, J=6.4Hz, 0.2H), 8.51 (m, 0.8H) ppm; LC-MS
(ES+) m/e=605, 607 (M+H) .


2- (2-Benzyloxy-5-oxo-tetrahydro-£uran-3-ylcarbamoyl) -
piperidine-1-carboxylic acid-tert butyl ester (100) .
Prepared from piperidine-l,2-dicarboxylic acid 1-text-
butyl ester 99 and 40 following the method used in the
preparation of 75 to give the title compound as a
yellow solid (2.63g, 57% yield). 1H-NMR (500MHz, CDCl3) δ 1.15-1.79 (m, 15H), 2.12-2.50 (m, 2H), 2.56-2.83 (m,
1H), 2.89 (dd, 0.5H), 3.05 (dd, 0.5H), 3.81-4.15 (br s,
1H) , 4.36-4.97 (m, 3H) , 5.37-5.61 (m, 1H) , 6.42-6.89
(br s, 1H), 7.17-7.51 (m, 5H). LC-MS (ES+) m/e=419.4
(MH+).
{2-[2-(2-Benzyloxy-5-oxo-tetrahydrd-£uran-3-
ylcarbamoyl) -piperidin-1-yl] -l-methyl-2-oxo-ethyl}-
carbamic acid tert-butyl ester (101) .
2- (2-Benzyloxy-5-oxo-tetrahydro-furan-3-ylcarbamoyl) -
piperidine-l-carboxylic acid tert-butyl ester (100) was
dissolved in 20% TFA in CH2Cl2 (25mL) and stirred at

room temperature for 50min. The solvent was evaporated
and the residual acid azeotroped with CH2Cl (4x). The
resulting oil was dissolved in CH2Cl2 (2 0mL) and DMF
(5mL), cooled to 0°C and treated with DIEA (4.7mL,
27.0mmol), Boc-alanine (970mg, S.lmmol), HOBT (924mg,
6.8mmol) and EDC (l.31g, 6.8mmol) and the solution
stirred under N2 for 18hours. The solvent was
concentrated in vacuo then dissolved in EtOAc and
washed with 0.5N NaHSO4 (2x), saturated NaHCO3 (2x) and
brine. The organic layer was dried over anhydrous
Na2SO4 and evaporated to give an orange solid that was
dissolved in CH2Cl2 and added dropwise to diethyl ether
to afford a white precipitate. The title compound as a
white solid (1.21g, 73% yield). 1H-NMR (500MHz, CDCl3) δ
l.10-l.79 (m, 18H) , 1.98-2.19 (m, 0.5H), 2.28-2.88
(m, 3H), 2.89-3.13 (m, 0.5H), 3.78-3.95 (m, 0.5H),
4.21-5.16 (m, 5.5H), 5.38-5.59 (m, 0.3H), 5.66 (d,
0.4H), 5.80 (d, 0.3H), 7.24-7.40 (m, 5H). LC-MS (ES+)
m/e=490.3 (MH+) .
1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
piperidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)amide (102a).
Prepared from{2- [2- (2-benzyloxy-5-oxo-tetrahydro-furan-
3-ylcarbamoyl) -piperidin-1-yl] -l-methyl-2-oxo-ethyl}-
carbamic acid text butyl ester and 4-acetylamino-3-
chlorobenzoic acid by the procedure used in the
preparation of 98a to give the title compound (7lmg,
47% yield). 1H-NMR (500MHz, CDCl3) δ 1.10-1.97 (m,
10H), 2.10-2.68 (m, 5H), 2.73-3.24 (m, 2H), 3.62-3.92
(m, 1H), 4.24-5.27 (m, 5H), 5.48-5.59 (m, 0.5H), 5.75-
5.85 (m, 0.5H), 6.51-6.61 (d, 1H), 7.05-7.45 (m, 4H) ,

7.52-8.12 (m, 4H) . Analytical HPLC 8.30min. LC-MS (ES*)
m/e=585.3 (MH+) .
1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
piperidine-2-carboxylic acid (2-benzyloxy-5-oxo-
tetrahydro-furan-3-yl)-amide (102b) .
Prepared as above for 102a to give the title compound
(0.06g, 27% yield) as a yellow solid. 1H-NMR (500MHz,
CDCl3) δ1.2-1.8 (m, 7H) , 2.1-2.6 (m, 2H) , 2.7-3.2 (m,
4H) , 3.6-4.0 (m, 1H) , 4.3-4.9 (m, 7H) , 5.0-5.8 (m, 2H) ,
6.5-7.0 (m, 2H) , 7.2-7.8 (m, 8H) ppm. Analytical HPLC
(cyano column) 14.559 (39.6%), 15.198 (60.4%). LC-MS
(ES+) : m/e=543 (M+H)


4-Hydroxy-pyrrolidine-l,2-dicarboxylic acid 1-benzyl
ester 2-tert-butyl ester (104).
Compound 104 was prepared according to the procedure
used to prepare compound 95.
A suspension of Cbz-Hyp-OH (4.854g, l8mmol) in DMA
(135ml), benzyltriethylammonium chloride (4.l05g,
18mmol) , K2CO3 (64g, 46mmol) and 2-bromo-2-
methylpropane (99ml, 859mmol) was stirred at 55°C for
18hours. The mixture was diluted with iced water and
extracted with EtOAc (3x). The organic phase washed
with water, 0.5N NaHSO4 solution and brine dried and
the solvent in vacuo to yield the title compound as a
yellow oil (5.368g, 98% yield). 1H-NMR (500MHz, CDCl3) δ 1.33 (S, 5H), 1.47 (S, 4H) , 2.01-2.14 (m, 1H), 2.22-
2.38 (m, 1H), 3.50-3.72 (m, 2H) , 4.34-4.45 (m, 1H) ,
4.45-4.53 (m, 1H) , 5.04-5.20 (m, 2H) , 7.22-7.42 (til,
5H) . Analytical HPLC io.l4min. LC-MS (ES+) m/e=322.2
(MH+) .
4-Fluoro-pyrrolidine-l,2-dicarboxylic acid 1-benzyl
ester 2-tert-butyl ester (105).
A solution of 104 (4.262, 13.96mmol) in CH2Cl2 (100ml)
at -78°C was treated with DAST (1.80ml, 13.6mraol),
stirred for lOmin then warmed to room temperature and
stirred for 60h under N2. The mixture was poured into
iced NaHCO3 (10% solution, 350ml) and extracted with
CH2Cl2 (2x) . The organic phase was washed with water,
brine, dried over anhydrous Na2SO4 and concentrated to
give a brown oil (4.299g) which was purified by flash
column chromatography on silica gel using hexanes/EtOAc
(90/10- to 80/20%) . The title compound was obtained as a

yellow oil (2.805g, 64% yield) . 1H-NMR (500MHz, CDCl3)
6 1.37 (s, 4.5H), 1.45 (s, 4.5H), 2.20-2.55 (m, 2H) ,
3.61-3.93 (m, 2H), 4.41 (d, 0.5H), 4.49 (d, 0.5H),
5.03-5.21 (m, 3H) , 7.23-7.44 (m, 5H) . Analytical HPLC
12.15min. LC-MS (ES+) m/e=324.2 (MH+) .
1- (2-Benzyloxycarbonylamino-propionyl) -4-fluoro-
pyrrolidine-l,2-dicarboxylic acid 1-benzyl ester 2-
tert-butyl ester (106).
A solution of 105 (2.72g, 8.42 mmol) in MeOH (50ml) and
10% Pd/C (1.27g) was stirred under H2 for 2hours then
filtered through celite and the solvent evaporated to
give a yellow oil (1.526g) . This oil was dissolved in
CH2Cl2 (30ml) and treated with DIEA (l.5ml, 8.6mmol),
Cbz-ala-OH (2.34g, 10.5mmol) and EDC (2.32g, l2mmol) at
0°C. The mixture was stirred an additional 10rain at
0°C then allowed to warm to room temperature and stir
for l8hours. The solvent was concentrated In vacuo and
the residue dissolved in EtOAc then washed with 0.5N
NaHSO4 (2x) , saturated NaHCO3 (2x) and brine. The
organic layer was dried over anhydrous Na2SO4 and
evaporated to give a white solid which was purified by
flash column chromatography, eluting using
hexanes/EtOAc (80/20 to 60/40%) . The title compound
was isolated as a white solid (286g, 86% from 4-fluoro-
pyrrolidine-l,2-dicarboxylic acid 1-benzyl ester 2-
tert-butyl ester). 1H-NMR (500MHz, CD3OD) δ1.26-1.59
(m, 12H), 2.20-2.67 (m, 2H) , 3.45-4.13 (m, 2H), 4.25-
4.47 (m, 1H) , 4.5B-4.71 (m, 1H) , 4.96-5.17 (m, 2H) ,
5.19-5.45 (m, 1H), 7.23-7.48 (m, 5H) . Analytical HPLC
16.3 6min. LC-MS (ES+) m/e=395.3 (MH+) .

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carboxylic acid-tert-butyl ester
(107) .
A suspension of 106 (2.65g, 6.72mmol) in MeOH (40ml)
and 10% Pd/C (1.32g) was stirred under an atmosphere of
H2 for 1.5hours, filtered through celite and the
concentrated to give a waxy solid (1.694g). The solid
was dissolved in CH2Cl2 (25ml) and treated with DIEA
(3.4ml, l9.5mmol), 4-amino-3-chlorobenzoic acid
(1.362g, 7.9mmol), HOBT (1.164g, 8.62mmol) and EDC
(1.645g, 8.57mmol) at o'c under N2. The mixture was
allowed to warm to room temperature and stirred for
18hours. The solvent was concentrated in vacuo. The
residue was dissolved in EtOAc, washed with water (4x),
0.5N NaHSO4 (2x), saturated NaHCO3 (2x) and brine. The
organic layer was dried over anhydrous Na2SO4 and
evaporated to give a white solid which was purified by
flash column chromatography, using CH2Cl2/MeOH (99/1 to
98/2%). The product obtained as a white solid (2.705g,
97%). 1H-NMR (500MHZ, CD3OD) δ1.33 (s, 9H) , 1.48 (d,
3H), 2.31-2.55 (m, 2H), 3.93 (d,d, 1H), 4.02-4.21 (m,
1H), 4.59-4.76 (m, 1H), 5.31 (br s, 0.5H), 5.41 (br s,
0.5H), 6.78 (d, 1H), 7.57 (d,d, 1H) , 7.78 (s, 1H), 8.31
(d, 1H) . Analytical HPLC 14.i4min. LC-MS (ES+)
m/e=414.2 (MH+) .


1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carboxylic acid (2-benzyloxy-5-
oxo-tetrahydro-furan-3-yl)-amide (108a).
Prepared from syn- (2-benzyloxy-5-oxo-tetrahydro-furan-
3-yl)-carbamic acid allyl ester and 107a following the
method used for the synthesis of 98a. The title
compound was isolated as a white solid (4lmg, 15%
yield). 1H-NMR (500MHz, CD3OD) δ 0.94 (d, 0.3H), 1.07
(d, 1H), 1.40 (m, 1.7H), 2.21-2.65 (m, 2.2H), 2.70-2.85
(m, 1.4H), 2.96-3.08 (m, 1.4H), 2.96-3.08 (dd, 0.4H),
3.57-4.24 (m,3H), 4.41-4.93 (m, 4H) , 5.14-5.45 (m, 1H) ,
5.60-5.67 (m, 0.6H), 5.77 (d, 0.4H), 6.77 (dd, 1H),
7.15-7.41 (m, 5H) , 7.51-7.62 (m, 1H) , 7.77 (dd, 1H).
Analytical HPLC 12.83min. LC-MS (ES*) m/e= 547.1 (MH+).

1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carboxylic acid (2-benzyloxy-5-
oxo-tetrahydro-ftiran-3-yl)-amide (108b) .
Prepared from (2-benzyloxy-5-oxo-tetrahydro-furan-3-
yl)-carbamic acid allyl ester 107a following the method
used for the synthesis of 98a. The title compound was
isolated as a white solid (654mg, 54% yield) . aH-NMR
(500MHz, CD3OD) δ 1.07 (d, 0.5H), 1.25-1.56 (m, 2.5H),
2.21-2.65 (m, 2.3H), 2.68-2.89 (m, 1H), 2.91-3.10 (m,
0.7H), 3.57-4.23 (m, 2H), 4.32-4.95 (m, 5H), 5.16-5.52
(rc, 1H), 5.45-5.50 (m, 0.3H), 5.54-5.58 (m, 0.2H),
5.61-5.67 (m, 0.3H), 5.77 (d, 0.2H), 6.72-6.84 (m, 1H),

7.16-7.41 (m, 5H), 7.50-7.65 (m, 1H) , 7.71-7.87 (m,
1H) . Analytical HPLC I2.83min. LC-MS (ES+) m/e=547.1
(MH+) .

1- [2- (4-Amino-3-ehloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carboxylic acid (2-ethoxy-5-oxo-
tetrahydro-furan-3-yl)-amide (108c).
Prepared from syn- (2-ethoxy-5-oxo-tetrahydro-furan-3-
yl)-carbamic acid allyl ester and 107a following the
method used for the synthesis of 98a to give the title
compound (100.3mg, 38% yield). 1H-NMR (500MHz, CD3OD) δ
1.09 (t, 1.2H), 1.25 (t, 1.8H), 1.40 (d, 1H), 1.49 (d,
2H) , 2.33-2.61 (m, 2H) , 2.65-2.95 (m, 2H) , 3.44-4.30
(m, 4H) , 4.47-4.79 (m, 3H) , 5.18-5.25 (m, 0.2H), 5.27-
5.36 (m, 0.5H), 5.39-5.46 (m, 0.3H), 5.56 (m, 1H),
6.72-6.94 (m, 0.8H), 7.54-7.69 (m, 0.8H), 7.79 (d,
0.55H), 8.06 (d, 0.55H), 9.00 (d, 0.3H). Analytical
HPLC 8.46min. LC-MS (ES+) m/e= 485.2 (MH+).


1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carboxylic acid [2-(2-isopropyl-5-
methyl-cyclohexyl) -5-oxo-tetrahydro-furan-3-yl] -amide
(108d).
Prepared from {2- [1R- (2S-Isopropyl-5R-methyl-
cyclohexyloxy) ] -5-oxo-tetrahydro-furan-3-yl}-carbamic
acid allyl ester and 107a following the method used for
the synthesis of 98a to give the title compound (95mg,
31% yield). 1H-NMR (500MHz, CD3OD) δ 0.42 (d, 2H) ,
0.57 (d, 2H), 0.60-1.10 (m, 10H), 1.22-1.76 (m, 6H),
1.96-2.17 (m, 1H), 2.29-2.60 (m, 2H), 2.61-2.88 (m,
1.5H), 3.02-3.23 (dd, 0.5H), 3.37-3.47 (m, 0.5H), 3.50-
3.61 (m, 0.5H), 3.63-4.24 (m, 2H) , 4.48-4.62 (m, 3H) ,
5.18-5.48 (m, 1H), 5.72 (d, 0.4H), 5.82 (d, 0.6H),
6.77-6.84 (m, 1H), 7.53-7.67 (m, 1H), 7.78 (d, 0.4H),
7.84 (d, 1H) Analytical HPLC 8.34min. LC-MS (ES+) m/e=
595 (MH+).




To a solution of 110 (194mg, 0.36mmol) in CH2Cl2 (5ml)
was added DMBA (70.7mg, 0.4 5mmol) and Pd(PPh3)4
(50.3mg, 0.044mmol) at 0°C. The solution was warmed to
room temperature after 15mins, stirred for 2hours,
diluted with CH2Cl2 then washed with water (2x) and
brine. The organic layer was dried over anhydrous
Na2SO4 and evaporated to give the crude product. Flash
chromatography using CH2Cl2/MeOH (99/1 to 95/5%)
afforded the title compound (138.6mg, 77% yield). 1H-
NMR (500MHz, CD3OD) δ 1.13-1.31 (m, 3H) , 1.35-1.49 (m,
3H), 1.84-2.35 (m, 4H), 2.43-3.05 (m, 2H), 3.48-3.93
(m, 4H), 4.22-4.80 (m, 3H), 5.38 (d, 0.4H), 5.46 (s,
0.1H), 5.55-5.61 (m, 0.5H), 7.76-7.94 (m, 2H) .
Analytical HPLC 8.70min. LC-MS (ES+) m/e=502.2 (MH+).


Compounds 116a- 116h were prepared as described above
for compounds 98 only substituting l-(2-
benzyloxycarbonylamino-propionyl) -4, 4-difluoro-
pyrrolidine-2-carboxylic acid tert-butyl ester (114)
for 1- (2-benzyloxycarbonylamino-propionyl)-pyrrolidine-
2-carboxylic acid tert-butyl ester (95) .
Preparation of 1- (2-benzyloxycarbonylamino-propionyl) -
4,4-difluoro-pyrrolidine-2-carboxylic acid tert-butyl
ester (114).
A solution of 4, 4-di£luoro-pyrrolidine-l,2-dicarboxylic
acid-l-benzyl ester-2-tert-butyl ester (113)

(Karanewsky, et.al., J. Med. Chem. 33, pp. 1459-1469
(1990)) (0.42 g, 1.23 mmol) and 10% palladium on carbon
(0.22g) in methanol (6 mL) was stirred at l atm
hydrogen pressure for 3h. The mixture was filtered
through Celite and evaporated . The residue was
dissolved in CH2Cl2 (4 mL) and DMF (2 mL) and cooled to
0°C. 2-Benzyloxycarbonylamino-propionic acid (0.30 g,
1.35 mmol), EDC (0.30, 1.54 mmol), DIEA (0.65 mL) and
HOBt (0.17 g, 1.23 mmol) was added and the reaction was
stirred 0.5h at 0°C, then 16h at room temperature under
nitrogen. The solvent was removed in vacuo and the
residue was dissolved in ethyl acetate, then was washed
with 10% sodium bisulfate, saturated sodium
bicarbonate, water and brine, was dried over sodium
sulfate and was evaporated. Purification by flash
chromatography on silica, eluted with 25:75 ethyl
acetate: hexanes provided 1-(2-benzyloxycarbonylamino-
propionyl) -4, 4-dif luoro-pyrrolidine-2-carboxylic acid
text-butyl ester (0.39 g, 77% yield) as a colorless
oil.
1H-NMR (500 MHZ, CDCl3) δ 1.3-1.6 (m, 12H) , 2.5 (m,
0.8H), 2.7 (m, 1.2H), 3.9 (m, 1H) , 4.1 (m, 1H), 4.4 (m,
1H), 4.7 (m, 1H), 5.1 (m, 2H) , 5.59 (brd, J=7.7Hz,
0.8H), 5.7 (brd, J=7.7Hz, 0.2H), 7.35 (m, 5H) ppm.
Analytical HPLC (cyano column) 17.069 min. LC-MS (ES+):
m/e=413 (M+H) , 357 (M+H-tert-butyl), 313 [M+H-(CO2tert-
butyl)].


1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4,4-
difluoro-pyxrolidine-2-carboxylic acid (2-benzyloxy-5-
oxo-tetrahydro-furan-3-yl)-amide (116a).
Prepared from 115a and syn-40 to afford the title
compound as an off-white solid (0.l4g, 73% yield). 1H-
NMR (500 MHz, CD3OD) δ 1.0-1.5 (m, 3H) , 2.0-3.5 (m,
4H+CH3OH), 3.5-5.5 (M, 6H+H20), 5.6-5.8 (m, 1H) , 6.7-6.8
(m, 1H) , 7.1-7.8 (m, 8H) , 8.2-8.6 (m, 1H) ppm.
Analytical HPLC (cyano column) 13.744 min. LC-MS (ES+):
m/e=565 (M+H).

1- [2- (4-AcetylamiTIP-3-chloro-benzoylamino) -propionyl] -
4,4-difluoro-pyrrolidine-2-carboxylic acid (2-
benzyloxy-5-oxo-tetrahydro-£uran-3-yl)-amide (116b) .
Prepared from 115b and syn-40 to afford the title
compound as an off-white solid (0.08 g, 38% yield).
2H-NMR (500 MHz, CDCl3) 5 1.03 (d, J=6.9Hz, 0.4H), 1.30
(d, J=6.9Hz, 0.6H), 2.25 (d, J=2.9Hz, 3H), 2.4-3.2 (m,
4H),"3.6-4.4 (m, 4H), 4.6-4.9 (m 3H) , 5.52 (d, J=5.2Hz,
0.6H), 5.78 (d, J=5.2HZ, 0.4H), 6.6 (br S, 1H), 6.9-7.9
(m, 8H) , 8.39 (d, J=8.1 HZ, 0.4H), 8.44 (d, J= 8.3Hz,
0.6H), 8.74 (d, J=6.8Hz/ 1H) ppm. Analytical HPLC
(cyano column) 11.830 min. LC-MS (ES+): m/e= 607 (M+H).


1- [2- (4-Acetylaonino-5-chloro-2-methoxy-benzoylamino) -
propionyl] -4,4-di£luoro-pyrrolidine-2-carboxylic acid
(2-benzyloxy-5-oxo-tetrahydro-furan-3-yl)-amide (116c) .
Prepared from 115c and syn-40 to afford the title
compound as an off-white (0.07g, 29% yield). 1H-NMR
(500 MHz, CDCl3) δ 0.99 (d, J=6.9Hz, 1.35H), 1.32 (d,
J=6.9Hz, 1.65H), 2.25 (S, 1.5H), 2.26 (s, 1.5H), 2.3-
3.2 (m, 4H), 3.95 (s, 0.55H), 3.98 (s, 0.45H), 3.7-4.1
(m, 2.5H), 4.2-4.5 (m, 1.5H), 4.6-4.9 (m, 3H) , 5.52 (d,
J=5.3Hz, 0.55H), 5.80 (d, J=5.3Hz, 0.45H), 7.0-7.4 (m,
4H) , 7.7-7.9 (m, 2H) , 8.0-8.4 (TO, 2H), 8.49 (d,
J=6.5Hz, 1H) , 8.93 (d, J=6.7Hz, 1H) ppm. Analytical
HPLC (cyano column) 12.959 min. LC-MS (ES+) : m/e=637
IMxfll

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
4,4-difluoro-pyrrolidine-2-carboxylic acid (2-ethoxy-5-
oxo-tetrahydro-furan-3-yl)-amide (116d) .
Prepared from 115b and syn-(2-ethoxy-5-oxo-tetrahydro-
furan-3-yl)-carbamic acid allyl ester to afford the
title compound as a 92:8 mixture of epimers. Off-white
solid. (0.27g, 66% yield). 1H-NMR (500 MHz, CDCl3) δ 1.0-

1.5 (m, 6H) , 2.25 (S, 1.8H), 2.26 (s, 1.2H), 2.3-3.1
(m, 4H) , 3.3-4.3 (m, 4H) , 4.5-4.9 (m, 3H) , 5.45 (d,
J=5.3HZ, 0.75H), 5.59 (d, J=5.2Hz, 0.25H), 6.7-7.1 (m,
2H) , 7.62 (dd, J=8.7, 2.0Hz, 1H) , 7.76 (m, 1H) , 7.85
(d, J=2.0Hz, 1H) , 8.48 (m, 1H) ppm. Analytical HPLC
(Cl8 column) 13.300(91.8%), 14.046 (8.2%) min. LC-MS
(ES+) : m/e 545 (M+H) .

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
4,4-difluoro-pyrrolidine-2-carboxylic acid (2-
cyclohexyloxy- 5 - oxo - te trahydro - furan- 3 -yl) - amide
(116e) .
Prepared from 115b and syn- (2-cyclohexyloxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester to
afford the title compound as a 93:7 mixture of epimers.
1H-NMR (500 MHZ, CDCl3) δ 1.0-2.0 (m, 13H) , 2.25 (s,
2H) , 2.26 (s, 1H) , 2.40 (dd, J=17.3, 10.1Hz, 1H), 2.84
(dd, J=17.3, 8.5Hz, 1H), 2.5-3.0 (m, 2H), 3.5-4.3 (m.
3.5H), 4.5-4.9 (m. 2.5H), 5.59 (d, J=5.3Hz, 0.75H),
5.76 (d, J=5.2Hz, 0.25 H), 6.74 (br d, J= 5.7Hz,
0.25H), 6.93 (br d, J=7.1HZ, 1H) , 7.06 (br d, J=7.8HZ, .
0.75H), 7.62 (dd, J=8.6, 2.0Hz, 1H) , 7.78 (m, 1H) , 7.85
(d, J=2.0HZ, 1H), 8.35 (br d, J=6.6Hz, 0.25H), 8.50 (br
d, J=8.2Hz, 0.75H) ppm. Analytical HPLC (Cl8 column)
17.112 (93%), 17.433 (7%) min. LC-MS (ES+) : m/e=599
(M+H).


1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
4,4-difluoro-pyxrolidine-2-carboxylic acid [2- (indanol-
2-yl)oxy-5-oxo-tetrahydro-£uran-3-yl] -amide (116f) .
Prepared from 115b and [2-(indanol-2-yl] oxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester to
afford the title compound as a 62:38 mixture of
epimers. Off-white solid (0.34g, 71% yield). 1H-NMR
(500 MHz, CDCl3) δ 1.09 (d, J=6.9Hz, 0. 6H) , 1.21 (d,
J=6.9HZ, 0.9H), 1.33 (d, J=6.9Hz, 0.9H), 1.42 (d,
J=6.9HZ, 0.6H), 2.28 (s, 2H) , 2.29 (s, 1H) , 2.40 (dd,
J=17.4, 10.3HZ, 1H), 2.4-3.3 (m, 7H), 3.6-4.2 (m, 2H) ,
4.5-4.8 (m, 4H) , 5.66 (m, 0.6H), 5.84 (d, J=4.3Hz,
0.2H), 6.22 (m, 0.2H), 6.7-7.0 (m, 2H), 7.2-7.3 (m,
4H), 7.5-7.7 (m, 1H), 7.8-8.0 (m, 2H) , 8.52 (m, 0.6H),
8.62 (br d, J=6.5Hz, 0.4H) ppm. Analytical HPLC (Cl8
column) 16.556 (62.0%), 16.824 (38.0%) min. LC-MS
(ES+) : m/e=633 (M+H).

1- [2- (4-Acetylamino-3-ch.loro-benzoylamino) -propionyl] -
4,4-difluoro-pyrrolidine-2-carboxylic acid (2-

cyclopentylmethoxy-5-oxo-tetrahydro-£uran-3-yl) -amide
(116g).
Prepared from 115b and syn- (2-cyclopentylmethoxy-5-oxo-
tetrahydro-furan-3-yl)-carbamic acid allyl ester to
afford the title compound as an off-white solid (0.20g,
44% yield). 1H-NMR (500 MHz, CDCl3) δ 1.0-1.8 (m, 11H) ,
1.9-3.0 (m, 5H) , 2.26 (s, 3H) , 3.29 (m, 0.25H), 3.47
(m, 0.75H), 3.58 (m, 0.25H), 3.74 (m, 0.75H), 3.8 (m,
0.75H), 4.1 (m, 0.25H), 4.25 (m, 1H) , 4.4-4.8 (m, 3H) ,
5.44 (d, J=5.2Hz, 0.75H), 5.62 (d, J=5.2Hz, 0.25H), 6.7
(br, 0.25H), 6.91 (d, J=7.1Hz, 1H) , 7.1 (m, 0.75H),
7.59 (d, J=8.5HZ, 0.25H), 7.63 (dd, J=8.5, 2.5Hz,
0.75H), 7.75 (m, 1H), 7.86 (d, J=1.8Hz, 1H), 8.33 (br
d, J=6.5Hz, 0.25H), 8.49 (br d, J=8.4Hz, 0.75H) ppm.
Analytical HPLC ( Cl8 column) 17.705 min. LC-MS (ES+) :
m/e=599 (M+H).

1- [2- (4-Acetylamino-3-chloro-benzoylamino) -propionyl] -
4,4-di£luoro-pyrrolidine-2-carboxylic acid (2-
phenylethoxy-5-oxo-tetrahydro-furan-3-yl)-amide (116h) .
Was prepared from 115b and syn- (5-oxo-2-phenethyloxy-
tetrahydro-furan-3-yl)-carbamic acid allyl ester to
afford the title compound as an off-white solid (0.15g,
24% yield). 1H-NMR (500MHz, CDCl3) δ 1.29 (d, J=6.9Hz,
0.75H), 1.40 (d, J=6.9HZ, 2.25H), 2.25 (s, 2.25H), 2.26
(s, 0.75H), 2.3-3.0 (m, 6H) , 3.7-4.8 (m, 7H) , 5.38 (d,
J=5.3Hz, 0.75H), 5.67 (d, J=5.1HZ, 0.25H), 6.65 (m,

1H) , 6.90 (d, J=7.0Hz, 0.75H), 7.06 (d, J=7.6Hz,
0.25H), 7.1-7.3 (m, 5H), 7.57 (d, J=8.6Hz, 0.25H), 7.63
(d, J=8.6Hz, 0.75H), 7.75 (m, IH) , 7.86 (d, J=1.8Hz,
1H) , 8.35 (d, J=6.2 Hz, 0.25H), 8.49 (d, J=8.3Hz,
0.75H) ppm. Analytical HPLC (Cl8 column) 17.265 min.
LC-MS (ES+) : m/e=621 (M+H) .

2- (2-Benzyloxy-5-oxo-tetrahydro-£uran-3-ylcarbamoyl) -
pyrrolidine-1-carboxylic acid tert-butyl ester (118) .
Prepared from 40 (1.16 g, 4.0 tnmol) and Boc-Pro-OH
according to the procedure used to prepare 100 (Scheme
XVIII) to afford 1.53 g (94% yield) of the title
compound as a white solid. 1H-NMR (500 MHz, CDCl3) : δ
1.61 (br, 9H), 1.88 (br, 2H), 2.00-2.50 (m, 3H), 2.80-
3.10 (m, H) , 3.20-3.60 (m, 2H) , 4.05-4.45 (m, 1.5H),
4.58-4.80 (m, 1.5H), 4.83-4.98 (m, H) , 5.43-5.53 (m,
H) , 7.26-7.45 (m, 5H), 7.60-7.80 (d, H) ; Analytical
HPLC: 11.32 min; LC-MS: m/e = 405 (M+H+) .


2-Phenylaminopropionic acid (119) .
A mixture of alanine (356 mg, 4.0 mmol) , iodobenzene
(816 mg, 4.0 mmol), trans-dichlorobis(tri-o-
tolylphosphine) palladium (II) (Pd[P(o-Tol)3]2Cl2} (160
mg. 0.2 mmol), copper (I) iodide (40 mg, 0.2 mmol),
K2CO3 (552 mg, 4.0 mmol), benzyltriethylammonium
chloride (160 mg, 0.8 mmol), triethylamine (1.6 mL) and
water (0.8 mL) in DMF (8 mL) was stirred under nitrogen
atmosphere at 100oC for 20 hours. The mixture was
cooled to room temperature, diluted with ethyl acetate
(50 mL) and water (50 mL) , acidified with 6N HCl to the
pH = 2 to 3. The aqueous layer was extracted with
ethyl acetate (50 mL x 4) . The combined organic layers
were washed with water, brine, dried over anhydrous
Na2SO4/ filtered and evaporated in vacuo to give a red
oil. Plash chromatography using hexane/ethyl
acetate/acetic acid (95/5/0.5 to 80/20/0.5) to afford
300 mg (45% yield) of the title compound as a pink
solid. 1H-NMR (500 MHz, CDCl3/CD3OD = 0.5 ml/3 drops):
δ 1.45 (d, 3H), 4.02-4.15 (m, H), 6.57-6.70 (m, 3H) ,
7.11-7.25 (m, 2H); Analytical HPLC: 6.10 min. LC-MS;
m/e =,166 (M+H+) .
1- (2-Phenylaraino-propionyl) -pyrrolidine-2-earboxylic
acid (2-benzyloxy-5-oxo-tetrahydro-furan-3-yl) -amide
(120a and 120b).
A solution of 118 (405 mg, 1.0 mmol) was treated with
TFA (2 mL) in CH2Cl2 (2 mL) for one hour. The reaction
solution was evaporated in vacuo and azeotroped with
CH2Cl2 four times to give pyrrolidine-2-carboxylic acid
(2-benzyloxy-5-oxo-tetrahydro-furan-3-yl) -amide as a
pale yellow solid. 1H-NMR (500 MHz, CDCl3): 5 1,87-2.15
(m, 4H) , 2.30-2.70 (m, 2H) , 2.80-3.08 (m, H), 3.45 (br,

2H) , 4.35-4.98 (m, 3H) , 5.30-5.56 (m, H), 7.10-7.60 (m,
5H) ; Analytical HPLC: 7.78 / 8.20 min.; LC-MS: m/e =
305 (M+H+).
2-Phenylaminopropionic acid (119) (3 00 mg, 1.8 mmol) in
CH2Cl2 (10 mL) was treated with HOBT (270 mg, 2.0 mmol)
and EDC (2.1 g, 11 mmol) at 0°C for 10 min.
Diisopropylethylamine (2 mL) was added followed by a
solution of pyrrolidine-2-carboxylic acid (2-benzyloxy-
5-oxo-tetrahydro-furan-3-yl)-amide in CH2Cl2 do mL) .
The mixture was stirred at room temperature for 4
hours, diluted with CH2Cl2 (4 0 mL) , washed with water
then brine. The organic layer was dried over anhydrous
Na2SO4, filtered and evaporated in vacuo to give a pale
yellow solid. Flash chromatography using CH2Cl2
/methanol (99/1 to 98/2) afforded 151 mg (33% yield) of
anti diastereomer of the title compound (120a) and 129
mg (29% yield) of syn diastereomer (120b) as a white
solid. 1H-NMR (500 MHz, CDCl3) for the anti
diastereomer: δ 1.37-1.41 (m, 3H) , 1.50-2.45 (m, 4H) ,
2.60-2.70 (m, 0.3H), 2.89-2.94 (m, 0.7H), 3.40-3.80 (m,
2H), 4.10-4.50 (m, 3H) , 4.50-4.90 (m, 3H) , 5.26 (s,
0.3H), 5.38 (s, 0.7H), 6.45-6.60 (m, 2.3H), 6.65-6.80
(m, H) , 7.10-7.20 (m, 2.5H), 7.25-7.50 (m, 4.5H), 7.53-
7.70 (m, 0.7H), 7.82 (d, H) . For the syn diastereomer:
6 0.86-0.89 (m, H) , 1.20-1.40 (m, 4H) , 1.80-2.45 (m,
4H) , 2.80-2.86 (m, H) , 3.58-3.65 (m, 2H) , 4.20-4.40 (m,
H), 4.50-4.75 (m, 2H), 4.90 (d, H), 5.52 (d, H), 6.45-
6.70 (m, 3H), 6.75-6.85 (m, H) , 7.10-7.20 (m, 2.3H),
7.30-7.50 (m, 5.7H); Analytical HPLC: 10.55 min for
anti diastereomer and 10.62 min for syn diastereomer;
LC/MS: m/e = 452 (M+H+) for both diastereomers.

4-Oxo-3-{ [1- (2-phenylamino-propionyl) -pyrrolidine-2-
carbonyl] -amino}-butyric acid (121) .
Prepared from 120 (151 mg, 0.33 mmol) using hydrolysis
method A to afford 101 mg (83% yield) of the title
compound as a white solid. 1H-NMR (500 MHz, CDCl3/CD30D
= 1/1): 6 1.20-1.65 (m, 2H) , 1.65-2.35 (m, 3H), 2.40-
3.00 (m, H), 3.20-3.80 (m, 2H), 3.90-4.90 (m, 7H),
7.25-7.80 (m, 5H) ; Analytical HPLC: 6.38 min.; LC-MS:
m/e = 362 (M+H+) .
GENERAL PROCEDURES FOR THE PREPARATION OF COMPOUNDS OF
EMBODIMENT C FORMULA I (SCHEMES XXIII-XXV)

Hydrolysis Method A:
A 0.005-50 mmole sample of the alkylhemiacetal was
dissolved in 2.5 N HCl/ CH3CN (10/ 1) and stirred at
room temperature until the reaction was complete. The
resulting aqueous layer was washed with diethyl ether
(2 x 20 mL) and lyophilized to afford the product.
Hydrolysis Method B:

A 0.005-50 mmole sample of alkylheraiacetal was taken
into neat formic acid and stirred overnight at room
temperature. The mixture was triturated with a 3: l
mixture of hexane/diethyl ether to give a precipitate.
The solvent was decanted and the precipitate washed
with diethyl ether to afford the product.
Hydrolysis Method C:
A 0.005-50 mmole sample of the alkylhemiacetal was
dissolved in CH3OH and 20% Pd(OH)2/C and stirred under
H2 until the reaction was complete. The resulting
suspension was filtered and the solution concentrated
in vacuo, then triturated with a 3:1 mixture of
hexane/diethyl ether to give a precipitate. The
solvent was decanted and the precipitate washed with
diethyl ether to afford the product.
Hydrolysis Method D:
A 0.005-50 mmole sample of the alkylhemiacetal in
CH3CN/ water (1/ 2) was shaken with acidic resin (Dowex
50w x 2, H+ type) until the reaction was complete. The
solution was filtered and the resin washed with
CH3CN/water (1/4). The resulting water layer was washed
with diethyl ether, concentrated to a smaller volume in
vacuo then lyophilized to afford the product.



4-OXO-3- [(l-{2- [9-oxo-9H-fluorene-4-carbonyl) -amino] -
propionyl}-pyrrolidine-2-carbonyl) -amino] -butyric acid
(122a).
A 109.0tng (0.l9mmol) sample of 91 was hydrolyzed
according to method A to afford 88mg (96% yield) of the
title compound: Analytical HPLC 7.l5min. LC-MS (ES+)
tn/e=492.2 (M+H) .

3- ({1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine-2-carbnyl}-amino) -4-oxo-butyric acid
(122b).
A 5l.0mg (0.096mmole) sample of 76 was hydrolyzed
according to method A to afford 43.0mg (100% yield) of
the title compound: 1H-NMR (500 MHz, CD3OD/D2O: 0.5
mL/10 drops): δ 1.37-1.52 (m, 3H) , 1.80-2.20 (m, 3H) ,
2.20-2.37 (m, H) , 2.49-2.60 (m, H) , 2.60-2.75 (m, H) ,
3.70-3.80 (m, H) , 3.80-3.95 (m, H) , 4.20-4.35 (m, H) ,
4.40-4.50 (m, H) , 4.50-4.70 (n, H) , 4.70-4.85 (m, H) ,
6.85-6.87 (d, H) , 7.58-7.60 (m, H) , 7.77 (s, H) ;
retention time on analytical HPLC: 6.54 min; LC-MS: m/z
= 439 (M+H+) .


3- ({1- [2- {3,5-Dichloro-4-methoxy-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122c).
A 51.omg (0.088mmole) sample of 92 was hydrolyzed
according to method A to afford 24.Omg (56% yield) of
the title compound: Analytical HPLC 6.4imin. LC-MS
(ES+) m/e=488.3 (M+H).

3- ({l- [2- (4-Methoxy-3,5-dimethyl-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122d).
A 55.Omg (0.102mmole) sample of 77 was hydrolyzed
according to method A to afford 44.Omg (96% yield) of
the title compound: Analytical HPLC (Cl8) δ.70min, 1H-
NMR (CDCl3/ 500Mhz) : 5 1.23-1.70 (m, 3H) , 1.80-2.70 (m,
10H), 2.70-3.15 (m, 2H), 3.58-4.20 (m, 5H), 4.32-5.50
(m, 3H) , 5.60-6.00 (m, H), 6.80-7.90 (m, 4H); LC-MS
(ES+) m/e=448.2 (M+H).


4-Oxo-3- [ (l-{2- [pyridine-2-carbonyl) -amino] -propionyl}-
pyrrolidine-2-carbonyl)-amino]-butyric acid (122e) .
A 55.0mg (0.114mmole) sample of 88 was hydrolyzed
according to method A to afford 30.0mg (67% yield) of
the title compound: Analytical HPLC 4.60min. LC-MS
(ES+) m/e=391.3 (M+H).

3- ({1- [2- (4-Acetylamino-3-chloro-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122£) .
A 52mg (0.09lmmole) sample of 78 was hydrolyzed
according to method A to afford 40mg (91% yield) of the
title compound: 1H NMR (500MHz, CD3OD) δ 1.08-1.61 (m,
3H) , 1.77-2.41 (m, 3H) , 2.21 (s, 3H) , 2.41-2.77 (m,
2H) , 3.43-3.63 (m, 0.3H), 3.65-3.76 (m, 1H) , 3.81-3.94
(m, 1H), 4.18-4.34 (m, 1H), 4.42-4.64 (m, 1.7H), 4.77
(q, 1H) , 7.79 (dd, 1H) ; Analytical HPLC 4.97min. LC-
MS (ES+) m/e=481.3 (M+H).


3- ({1- [2- (4-Amino-3, 5-dichloro-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122g).
A 44.3mg (0.079mmole) sample of 89 was hydrolyzed
according to method A to afford 30mg (81% yield) of the
title compound: Analytical HPLC 5.40min. LC-MS (ES+)
m/e=473.2 (M+H).

J
3- ({1- [2- (3-Isopropoxy-benzoylamino) -propionyl] -
pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric acid
(122h).
A 52.0mg (0.097mmole) sample of 79 was hydrolyzed
according to method A to afford 30.0mg (69% yield) of
the title compound: Analytical HPLC 8.92min. LC-MS
(ES+) m/e=448.3 (M+H).


3- ({1- [2- (3-benzyloxy-4-methoxy-benzoylamino) -
propionyl] - pyrrolidine-2-carbonyl} -amino) -4-oxo-butyric
acid (122i).
A 50.8mg (0.082mmole) sample of 81 was hydrolyzed
according to method A to afford 22.4mg (52% yield) of
the title compound: Analytical HPLC 6.72min. LC-MS
(ES+) m/e=526.3 (M+H).

4-Oxo-3- [(l-{2- [ (quinoxaline-2-carbonyl) -amino] -
propionyl}-pyrrolidine-2-carbonyl) -amino] -butyric acid
(122j).
A 38.0mg (0.072mmole) sample of 80 was hydrolyzed
according to method A to afford 32.0mg (100% yield) of
the title compound: Analytical HPLC 5.95min. LC-MS
(ES+) m/e=442.3 (M+H).


3- {{1- [2-(3,5-Dichloro-4-hydroxy-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122k).
A 35mg (0.060mmole) sample of 83 was hydrolyzed
according to method A to afford 2 9.4mg (75% yield) of
the title compound: Analytical HPLC 7.9imin. 1H-NMR
(500 MHz, CD3OD) δ 1.47 (m, 3H) . 1.8-2.3 (m, 4H) , 2.49
(m, 1H) , 2.61 (m, 1H) , 3.5 (br m, 0.2H), 3.69 (br m,
0.9H), 3.84 (br m, 0.9H), 4.27 (m, 1H), 4.46 (m, 1H) ,
4.57 (m, 1H) , 4.73 (m, 1H) , 7.83 (m, 2H) ppm, LC-MS
(ES+) m/e=474.1 (M+H).

3- ({l- [2- (4-Amino-3-trifluoromethyl-benzoylamino) -
propionyl ] -pyrrolidine - 2 - carbonyl} - amino) - 4 - oxo -butyric
acid (1221) .
A lOmg (0.021mmole) sample of 98w was hydrolyzed
according to method A to afford 7.9mg (94% yield) of
the title compound: Analytical HPLC 6.64min. LC-MS
(ES+) m/e=473.3 (M+H).


3- ({l- [2- (3-Chloro-4-dimethylamino-benzoylamino) -
propionyl) -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122m).
A 10.Omg (0.02lmmole) sample of 98x was hydrolyzed
according to method A to afford 7.Omg (84% yield) of
the title compound: Analytical HPLC 5.l5min. LC-MS
(ES+) m/e=467.3 (M+H).

3- ({l- [2- (4-Dimethylamino-3,5-difluoro-ben2oylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122n).
A 20. Omg (0.043mmole) sample of 98y was hydrolyzed
according to method A to afford 16.8mg (100% yield) of
the title compound: Analytical HPLC 5.86min. LC-MS
(ES+) m/e=469.3 (M+H).

3- ({l- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -
pyrrolidine - 2-carbonyl}-amino) -4-oxo-butyric acid
(122o).
A 20.0mg (0.046mmole) sample of 98m was hydrolyzed
according to method A to afford 16.7mg (100% yield) of

the title compound: Analytical HPLC 8.47min. LC-MS
(ES+) m/e=439.2 (M+H).

3- ({1-[2-(4-Amino-2,3,5,6-tetrafluoro-benzoylamino)-
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122p).
A 20.0mg (0.042mmole) sample of 98z was hydrolyzed
according to method A to afford 15.3mg (91% yield) of
the title compound: Analytical HPLC 7.90min. LC-MS
(ES+) m/e=477.2 (M+H).

4-OXO-3-[(l-{2-[(quinoline-6-carbonyl)-amino]-
propionyl}-pyrrolidine-2-carbonyl) -amino] -butyric acid
(122q).
A 44mg (0.080mmole) sample of 93 was hydrolyzed
according to method A to afford 41mg (100% yield) of
the title compound: 1H NMR (500MHz, CD3OD) δ 1.24-1.69
(n, 3H) , 1.75-2.37 (m, 4H) , 2.39-2.87 (m, 2H) , 3.46-
4.04 (m, 2H), 4.11-4.77 (m, 3H), 8.19 (dd, 1H), 8.33
(d, 1H), 8.56-8.58 (m, 1H), 8.85 (s, 1H), 9.27-9.39 (m,

2H); Analytical HPLC4.91min. LC-MS (ES+) m/e=441.2
(M+H).

3- ({1- [2- (4-Acetylamino-5-chloro-2-methoxy-
benzoylamino) -propionyl] -pyrrolidine-2-carbonyl}-
amino)-4-oxo-butyric acid (122r) .
A 44.5mg (0.074mmole) sample of 87 was hydrolyzed
according to method A to afford 34.5mg (91% yield) of
the title compound: Analytical HPLC 6.88min. LC-MS
(ES+) m/e=511.2 (M+H).

3-1(1-{2-[3-Chloro-4-(2,2-dimethyl-propionylamino)-
benzoylamino] -propionyl}-pyrrolidine-2-carbonyl) -
amino] -4-oxo-butyric acid (122s).
A 19.0mg (0.036mmole) sample of 98aa was hydrolyzed
according to method A to afford I4.5mg (90% yield) of
the title compound: Analytical HPLC 7.28min. LC-MS
(ES+) m/e=523.3 (M+H).


3- ({1- [2- (3-Chloro-4-propionylamino-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122t).
A 21.0mg (0.042mmole) sample of 98ab was hydrolyzed
according to method A to afford 17.5mg (97% yield) of
the title compound: Analytical HPLC 5.72min. LC-MS
(ES+) m/e=495.2 (M+H).

3- ({1- [2- (3-Chloro-4-phenylacetylamino-benzoylamino) -
propionyl] -pyrrolidine-2-carbonyl}-amino) -4-oxo-butyric
acid (122u).
A 10.0mg (0.017mmole) sample of 98ac was hydrolyzed
according to method A to afford 7.9mg (85% yield) of
the title compound: Analytical HPLC 7.52min. LC-MS
(ES+) m/e=557.2 (M+H).


3- [(1-{2- [3-Chloro-4- (3-methyl-butyrylamino) -
benzoylamino] -propionyl}-pyrrolidine-2-carbonyl}-
amino]-4-oxo-butyric acid (122v) .
An 8.0mg (0.0l5mmole) sample of 98ad was hydrolyzed
according to method A to afford 6.5mg (96% yield) of
the title compound: Analytical HPLC 6.92min. LC-MS
(ES+) m/e=523.2 (M+H) .

3- ({1- [2- (4-Amino-3-chloro-benzoylamino) -propionyl] -4-
fluoro-pyrrolidine-2-carbonyl} -amino) -4-oxo-butyric
acid (123a).
A 12.4mg (0.022mmole) sample of 108b was hydrolyzed
according to method A to afford 9.6mg (93% yield) of
the title compound: Analytical HPLC 6.99min. LC-MS
(ES+) m/e=473.2 (M+H).
3-({1- [2- (4-Acetylamino-3-chloro-benzoylamino) -
propionyl] -4,4-difluoro-pyrrolidine-2-carbonyl}-amino) -
4-oxo-butyric acid (123b) .
A 26.2mg (0.043mmole) sample of 116b was hydrolyzed
according to method A to afford 10.8mg (4 9% yield) of
the title compound: Analytical HPLC 9.89min. LC-MS
(ES+) m/e=517.2

3- ({1- [2- (4-Acetylamino-3-chloro-2-:metlioxy-
benzoylamino)-propionyl]-4,4-difluoro-pyrrolidine-2-
carbonyl}-amino) -4-osco-butyric acid (123c).
A 23.1mg (0.036mmole) sample of ll£c was hydrolyzed
according to method A to afford 1.8mg (9% yield) of the
title compound: Analytical HPLC li.87min. LC-MS (ES+)
m/e=547.l (M+H).
BIOLOGICAL METHODS
Me obtained in vitro, ex vivo, and in vivo
data for selected compounds of this invention using the
methods described below. The results are shown in the
Tables 2-8. The designation "ND" indicates that the
compound was not tested in the described assay.
In the ICE Caspase assays, category "A"
indicates 10-1000 nM inhibition. Category "C" indicates >1000 nM
inhibition. See Tables 2 and 3.
In the PBMC assay, category "A" indicates
inhibition. Category "C" indicates 1001-2000 nM
inhibition. Category "D" indicates >2000 nM
inhibition. See Table 4.
In the whole blood assay, category "A"
indicates 2500-7500 nM inhibition. Category "C" indicates
>7500 nM. See Table 5
In the in situ metabolism assay, values of
[£{g) X f(h)] are disclosed as follows: category "A"
indicates Category "C" indicates 0.5-0.75. Category "D"
indicates >0.75. In the biliary excretion measurement.

category "A" indicates 10%. Category "C" indicates >10%. See Table 6.
In the i.v. clearance assay, values are
reported as follows: category "A" indicates Category "B" indicates 50-80. Category "C" indicates
>80. See Table 7.
In the bioavailability assay, the Cmax values
(^g/ml) are disclosed as follows: category "A"
indicates Category "C" indicates >5.0. The AUC values (^g x
hr/ml) are disclosed as follows: category "A"
indicates Category "C" indicates >5.0. Half-life (hrs) ranges
are disclosed as follows: category "A" indicates Category "B" indicates 1.5-2.0. Category "C" indicates
>2.0. The F values (%) are disclosed as follows:
category "A" indicates 67. Category "C" indicates >67. See Table 8.
ID Vitro Assays
Enzyme Inhibition
Ki values for test compounds with the various
caspases were obtained by the method of Margolin et al.
(J. Biol. Chem.. 272 pp. 7223-7228 (1997)). Assays
were performed in 10 mM Tris (Sigma Corp, St Louis MO)
pH 7.5, 1 mM Dithiothreitol (DTT, Research Organic INC,
Cleveland, OH) and 0.1% CHAPS (Pierce, Rockford ID at
37 °C. For caspase-3, a solution of 8% glycerol was
added to the assay buffer to improve enzyme stability.
A 65 µL aliquot of the assay buffer and 5 µL aliquot of
the appropriate dilutions of inhibitor in DMSO where
pipetted into a 96 well plate, treated with 10 µL of
caspase, then diluted in assay buffer (0.5-40 nM active
protein by active site titration). A control

containing DMSO but no compound was included for each
determination. The plates were then incubated for 15
minutes at 37 °C, before addition of the appropriate
substrate (2 0 µL, final concentration 1-4 X KM, final
assay volume 100 µL) to initiate the reaction.
Reaction rates were measured at 37°C either by-
following the time dependant increase in absorbance at
405 nM (for the pNA substrates) or in fluorescence (Ex
3 90, Em 460) (for the AMC substrates). The rates
obtained were plotted against inhibitor concentration
and the data fit to the Morrison tight-binding equation
for competitive inhibitors (Morrison, J.F., Biochem.
Bionhvs. Acta, 185 pp. 269-286 (1969)). The substrates
used for the individual assays were as follows:
Caspase-1 Suc-YVAD-pNA (Bachem, King of
Prussia, PA) (final concentration in the assay 80 µM) ,
Caspase-3 Ac-DEVD-pNA (Bachem, King of Prussia,
PA) (final concentration in assay, 60 µM)
Caspase-4 Ac-WEHD-AMC (Synpep, Dublin, CA) (final
concentration in Assay 20 µM),
Caspase-7 Ac-DEVD-AMC (Bachem, King of Prussia,
PA) (final concentration in assay 50 µM),
Caspase-8 Ac-DEVD-pNA (Bachem, King of Prussia,
PA) (final concentration in assay 80 µM) .



















PBMC Cell Assay
IL-1β Assay with a Mixed Population of Human Peripheral
Blood Mononuclear Cells (PBMC) or Enriched Adherent
Mononuclear Cells
Processing of pre-IL-1β by ICE can be
measured in cell culture using a variety of cell
sources. Human PBMC obtained from healthy donors
provides a mixed population of lymphocyte subtypes and
mononuclear cells that produce a spectrum of
interleukins and cytokines in response to many classes
of physiological stimulators. Adherent mononuclear
cells from PBMC provides an enriched source of normal
monocytes for selective studies of cytokine production
by activated cells.

Experimental Procedure:
An initial dilution series of test compound
in DMSO or ethanol is prepared, with a subsequent
dilution into RPMI-10% FBS media (containing 2 mM L-
glutamine, 10 mM HEPES, 50 U and 50 ug/ml pen/strep)
respectively to yield drugs at 4x the final test
concentration containing 0.4% DMSO or 0.4% ethanol.
The final concentration of DMSO is 0.1% for all drug
dilutions, A concentration titration which brackets
the apparent Ki for a test compound determined in an
ICE inhibition assay is generally used for the primary
compound screen.
Generally 5-6 compound dilutions are tested
and the cellular component of the assay is performed in
duplicate, with duplicate ELISA determinations on each
cell culture supernatant.
PBMC isolation and IL-1 Assay:
Buffy coat cells isolated from one pint human
blood (yielding 40-45 ml final volume plasma plus
cells) are diluted with media to 80 ml and LeukoPREP
separation tubes (Becton Dickinson) are each overlaid
with 10 ml of cell suspension. After 15 min
centrifugation at 1500-1800 xg, the plasma/media layer
is aspirated and then the mononuclear cell layer is
collected with a Pasteur pipette and transferred to a
15 ml conical centrifuge tube (Corning). Media is
added to bring the volume to 15 ml, gently mix the
cells by inversion and centrifuge at 3 00 xg for 15 min.
The PBMC pellet is resuspended in a small volume of
media, the cells are counted and adjusted to 6 x 106
cells/ml.

For the cellular assay, 1.0 ml of the cell
suspension is added to each well of a 24-well flat
bottom tissue culture plate (Corning), 0.5 ml test
compound dilution and 0.5 ml LPS solution (Sigma #L-
3012; 20 ng/ml solution prepared in complete RPMI
media; final LPS concentration 5 ng/ml). The 0.5 ml
additions of test compound and LPS are usually-
sufficient to mix the contents of the wells. Three
control mixtures are run per experiment, with either
LPS alone, solvent vehicle control, and/or additional
media to adjust the final culture volume to 2.0 ml.
The cell cultures are incubated for 16-18 hr at 37 °C
in the presence of 5% CO2.
At the end of the incubation period, cells
are harvested and transferred to 15 ml conical
centrifuge tubes. After centrifugation for 10 min at
2 00 xg, supernatants are harvested and transferred to
1.5 ml Eppendorf tubes. It may be noted that the cell
pellet may be utilized for a biochemical evaluation of
pre-IL-1β and/or mature IL-1β content in cytosol
extracts by Western blotting or ELISA with pre-IL-1β
specific antisera.
Isolation of Adherent Mononuclear cells:
PBMC are isolated and prepared as described
above. Media (1.0 ml) is first added to wells followed
by 0.5 ml of the PBMC suspension. After a one hour
incubation, plates are gently shaken and nonadherent
cells aspirated from each well. Wells are then gently
washed three times with 1.0 ml of media and final
resuspended in 1.0 ml media. The enrichment for
adherent cells generally yields 2.5-3.0 x 105 cells per
well. The addition of test compounds, LPS, cell

incubation conditions and processing of supernatants
proceeds as described above.
ELISA:
Quantikine kits (R&D Systems) may be used for
the measurement of mature IL-1β. Assays are performed
according to the manufacturer's directions. Mature IL-
1β levels of about 1-3 ng/ml in both PBMC and adherent
mononuclear cell positive controls are observed. ELISA
assays are performed on 1:5, 1:10 and 1:20 dilutions of
supernatants from LPS-positive controls to select the
optimal dilution for supernatants in the test panel.
The inhibitory potency of the compounds can
be represented by an IC50 value, which is the
concentration of inhibitor at which 50% of mature IL-1β
is detected in the supernatant as compared to the
positive controls.
The skilled practitioner realizes that values
obtained in cell assays may depend on multiple factors.
The values may not necessarily represent fine
quantitative results.













Whole Blood Assay for IL-1β Production
Whole blood assay IC50 values for compounds
of this invention were obtained using the method
described below:
Purpose:
The whole blood assay is a simple method for
measuring the production of IL-1β (or other cytokines)
and the activity of potential inhibitors. The
complexity of this assay system, with its full
complement of lymphoid and inflammatory cell types,
spectrum of plasma proteins and red blood cells is an
ideal in vitro representation of human in vivo
physiologic conditions.
Materials:
Pyrogen-free syringes (~ 3 0 cc)
Pyrogen-free sterile vacuum tubes containing
lyophilized Na2EDTA (4.5 mg/10 ml tube)


Human whole blood sample (" 30-50 cc)
1.5 ml Eppendorf tubes
Test compound stock solutions (" 25mM in DMSO or other
solvent)
Endotoxin -free sodium chloride solution (0.9%) and
HBSS
Lipopolysaccharide (Sigma; Cat. # L-3012) stock
solution at 1mg/ml in HBSS
IL-1β ELISA Kit (R&D Systems; Cat # DLB50)
TNFa ELISA Kit (R&D Systems; Cat # DTA50)
Water bath or incubator
Whole Blood Assay Experimental Procedure:
Set incubator or water bath at 3 0 °C.
Aliquot 0.25ml of blood into 1.5 ml Eppendorf tubes.
Note: be sure to invert the whole blood sample tubes
after every two aliquots. Differences in replicates
may result if the cells sediment and are not uniformly
suspended. Use of a positive displacement pipette will
also minimize differences between replicate aliquots.
Prepare drug dilutions in sterile pyrogen-
free saline by serial dilution. A dilution series
which brackets the apparent Ki for a test compound
determined in an ICE inhibition assay is generally used
for the primary compound screen. For extremely
hydrophobic compounds, prepare compound dilutions in
fresh plasma obtained from the same blood donor or in
PBS-containing 5% DMSO to enhance solubility.
Add 25 µl test compound dilution or vehicle
control and gently mix the sample. Then add 5.0 µl LPS
solution (250 ng/ml stocked prepared fresh: 5.0 ng/ml
final concentration LPS), and mix again. Incubate the
tubes at 30 °C in a water bath for 16-18 hr with
occasional mixing. Alternatively, the tubes can be

placed in a rotator set at 4 rpm for the same
incubation period. This assay should be set up in
duplicate or triplicate with the following controls:
negative control - no LPS; positive control - no test
inhibitor; vehicle control - the highest concentration
of DMSO or compound solvent used in the experiment.
Additional saline is added to all control tubes to
normalize volumes for both control and experimental
whole blood test samples.
After the incubation period, whole blood
samples are centrifuged for 10 minutes at ~ 2000 rpm in
the microfuge, plasma is transferred to a fresh
microfuge tube and centrifuged at 1000 x g to pellet
residual platelets if necessary. Plasma samples may be
stored frozen at -70 °C prior to assay for cytokine
levels by ELISA.
ELISA:
R&D Systems (614 McKinley Place N.E.
Minneapolis, MN 55413) Quantikine kits may be used for
measurement of IL-1β and TNF-ot. The assays are
performed according to the manufacturer's directions.
IL-1β levels of ~ 1-5 ng/ml in positive controls among
a range of individuals may be observed. A 1:200
dilution of plasma for all samples is usually
sufficient for experiments for ELISA results to fall on
the linear range of the ELISA standard curves. It may
be necessary to optimize standard dilutions if you
observe differences in the whole blood assay. Nerad,
J.L. et al., J. Leukocyte Biol., 52, pp. 687-692
(1992) .













Ex Vivo Assays
Metabolism and Excretion
Single pass perfusion studies in rat were
performed to assess gastrointestinal (GI) wall
metabolism (f(g)), liver metabolism (f(h)), and biliary-
excretion. The method used has been described in Pang,
C.S., J. Pharmacol. Exp. Therapeutics, 333, pp. 788-798
(1984) .



In Vivo Assays
In vivo Rat Clearance Assay - Clearance Rates
The rate of clearance in the rat (ml/min/kg)
for compounds of this invention may be obtained using
the method described below:
Representative Procedure
Cannulations of the jugular and carotid
vessels of rats under anesthesia are performed one day
prior to the pharmacokinetic study. M.J. Free, R.A.
Jaffee; 'Cannulation techniques for the collection
blood and other bodily fluids'; in: Animal Models;
p. 480-495; N.J. Alexander, Ed.; Academic Press;
(1978). Drug (10mg/mL) is administered via the jugular
vein in a vehicle usually consisting of: propylene
glycol/saline, containing 100mM sodium bicarbonate in a
1:1 ratio. Animals are dosed with 10-2 0 mg drug/kg and

blood samples are drawn at 0, 2, 5, 7, 10, 15, 20, 30,
60, and 90 minutes from an indwelling carotid catheter.
The blood is centrifuged to plasma and stored at -2 0 °C
until analysis. Pharmacokinetic analysis of data is
performed by non-linear regression using standard
software such as RStrip (MicroMath Software, UT) and/or
Pcnonlin (SCI Software, NC) to obtain clearance values.
Representative Analytical:
Rat plasma is extracted with an equal
volume of acetonitrile (containing 0.1% TFA) . Samples
are then centrifuged at approximately 1,000 x g and the
supernatant analyzed by gradient HPLC. A typical assay
procedure is described below.
200 µL of plasma is precipitated with 200
µL of 0.1% trifluoroacetic acid (TFA) in acetonitrile
and 10 µL of a 50% aqueous zinc chloride solution,
vortexed then centrifuged at "1000 x g and the
supernatant collected and analyzed by HPLC.
HPLC procedure:
Column: Zorbax SB-CN (4.6 x 150 mm)
(5µ particle size)
Column temperature: 50 °c
Flow rate: 1.0 mL/min
Injection volume: 75 µL.
Mobile phase: A=0.1% TFA in water and B=100%
acetonitrile
Gradient employed: 100% A to 30% A in 15.5 min
0% A at 16 min
100% A at 19.2 min
Wavelength: 214 nm

A standard curve is run at 20, 10, 5, 2 and
1 µg/mL concentrations.

Bioavailability
Oral pharmacokinetic studies
Male Sprague-Dawley rats (Harlan, Indianapolis, IN,
3 00-350 g) were anesthetized by an intramuscular
injection of ketamine/rompun mixture. A PE-50 cannula
was inserted in the right carotid artery for arterial
blood sampling. The rats were allowed to recover from
surgery overnight (≥16 hours) prior to being used in
the study. Test compounds were administered orally in
25% Cremophor EL/water (w/w) or 100% propylene glycol
(PG) in a dose volume of 10 mL/kg. Blood samples
(~0.30 mL) were removed at 0.25, 0.50, 1.0, 1.5, 2, 3,
4, 6, and 8 hours post-dose, plasma separated by
centrifugation and stored at -20°C pending analysis.
Quantitation of the plasma samples was conducted using
a gradient HPLC/MS/MS or enzymatic method detailed
below:

HPLC/MS/MS Method for the quantitation of ICE
inhibitors in rat plasma
Sample Preparation
• 50µl of plasma are aliquotted into Eppendorf
centrifuge vials.
• An equal volume of Acetonitrile is added to the
plasma to precipitate plasma proteins.
• Samples are vortexed for 5 minutes, and centrifuged
at 14,000 rpms for 5 minutes.
• 75µl of the supernatant is loaded into 12mm HPLC
liquid sampler vials.
• 50µl of sample is injected for analysis via the mass
spectrometer.
HPLC Instrumental Parameters
HPLC: Hewlett Packard HP1100 Binary Solvent
Delivery System.
HPLC Gradient Conditions
A = H2O 0.2% Formic Acid
B = Acetonitrile 0.2% Formic Acid-
Mobile Phase
Time %A %B
o 100 0
2 100 0
5 0 100
11 0 100
11.5 100 0
17 100 0
HPLC Analytical Column: Keystone Phenyl -2 Hypersil
2.0x100mm, Sµ 120A pore size, P/N# 105-39-2

Injection Volume: 50µl
Flow Rate: 0.20 mL/min.
Mass Spectrometry Instrumental Parameters
Instrument: P E Sciex API-365 Tandem Mass Spectrometer
Ionization Technique: Turbo-Ionspray (ESI)
Polarity: Positive
Dwell Time: 300msec
Pause Time: 5msec
Scan time: 0.9sec
Scan Mode: MRM (Multiple Reaction
Monitoring)
ICE ENZYMATIC ASSAY FOR THE QUANTITATION OF ICE
INHIBITORS IN RAT PLASMA
50 µL of plasma was extracted with 150 µL acetonitrile,
sonicated, vortexed, centrifuged at 10,000xg and 180 µL
of the supernatant dried in a Sorvall vortex evaporator
at room temperature. Samples were reconstituted in
100 µL buffer (10 mM tris-HCl, pH 7.5 with 0.1% CHAPS,
1 mM DTT) with sonication. 10 µL of each sample was
mixed with 10 µL ICE (1.1 mg/mL) in a microtitre plate
with 60 µL buffer. Samples were incubated for 15 min.
at room temperature then 20 µL Succ YVAD-pNA (400 µM,
prewarmed to 37°C) added, and the plate monitored at
405 nm for 20 min. at 3 7°C used a SpectraMax reader.
The data were fitted using a 4 parameter fit with the
SpectraMax software using an extracted standard curve.
The assay was linear from 0.15 to 2.0-3.0 µg/mL
aldehyde.


Antiviral Assays
The efficacy of the compounds of this
invention at treating or preventing antiviral related
diseases, disorders, or conditions may be evaluated in
various in vitro and in vivo assays. For example,
assays may be preformed to determine the ability of
these compounds to inhibit inflammatory responses
associated with viral infections. In vitro assays may
employ whole cells or isolated cellular components. In
vivo assays include animal models for viral diseases.

Examples of such animal models include, but are not
limited to, rodent models for HBV or HCV infection, the
Woodchuck model for HBV infection, and chimpanzee model
for HCV infection.
Compounds of this invention may also be
evaluated in animal models for dietary alcohol-induced
disease.
Other assays that may be used to evaluate the
compounds of this invention are disclosed in PCT
application PCT/US96/20843, published June 26, 1997,
under publication no. WO 97/22619. Such assays include
in vivo pharmacokinetic studies in the mouse,
inhibition of ICE homologs, inhibition of apoptosis, in
vivo acute assay for anti-inflammatory efficacy,
measurement of blood levels of drugs, IGIF assays,
mouse carrageenan peritoneal inflammation assay, and
type II collagen-induced arthritis.
Insofar as the compounds of this invention
are able to inhibit caspases, particularly ICE, in
vitro and furthermore, may be delivered orally to
mammals, they are of evident clinical utility for the
treatment of IL-1-, apoptosis-, IGIF-, and IFN-γ-
mediated diseases.
While we have described a number of
embodiments of this invention, it is apparent that our
basic constructions may be altered to provide other
embodiments which utilize the products and processes of
this invention.

We claim:
1. A compound represented by formula I: .

r
wherein:
Y is:

provided that when R7 is -OH then Y can also be:

X is -C(R3)2-;

m is 0;
R1 is -H, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S (O)2N (H)-R8,
-S(O)N(H)-R8, -C(O)C(O)N(H)R8, -C(O)CH=CHR8, -C(O)CH2OR8,
-C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O) 2N (R8)2, -S(O)N(R8)2,
-C(O)C(O)N(R8)2, -C(O)CH2N(R8)2, -CH2R8, -CH2-alkenyl-R8,
or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9, or
R2 and one R3 together with the atoms to which they are
bound, form a 3 to 7 membered cyclic or heterocyclic ring
system, wherein the heteroatoms are independently sulfur,
nitrogen, or oxygen, wherein a hydrogen atom bound to any
-alkyl or -cycloalkyl carbon atom is optionally replaced
by -R10, a hydrogen atom bound to any -aryl or -heteroaryl
carbon atom is optionally replaced by -R11, a hydrogen
atom bound to any nitrogen atom of the ring system is
optionally replaced by -R1;
R4 and one R5 together with the atoms to which
they are bound form a ring system selected from:



and the other R5 is H, wherein a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced with R1, or R4 and one R5 together with the atoms
to which they are bound form a ring system:
and the other R5 is H;
R6 is -H;
R7 is -OH, -OR8, or -N(H)OH;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl,
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,
wherein a hydrogen atom bound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by R10, a hydrogen atom
bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by R11, and a hydrogen atom bound to
any nitrogen atom is optionally replaced by R1;

each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11, and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R10 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N(H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl, -N(H)C(O)N(alkyl)2,
-S-alkyl, -S-aryl, -S-alkylaryl, -S(O)2alkyl, -S(O)alkyl,
-C(O)alkyl, -CH2NH2, -CH2N(H)alkyl, -CH2N (alkyl)2, -alkyl,
-cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl, -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11 and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1; and
each R11 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -O-
alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,
-N(H)-alkylaryl, -N(alkyl)2, -C(O)N(H)alkyl,
-C(O)N(alkyl)2, -N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl,

-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2; wherein
each alkyl is a straight-chained or branched,
saturated aliphatic hydrocarbon containing 1 to 6 carbon
atoms;
each alkenyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one double bond;
each alkynyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one triple bond;
each cycloalkyl is a mono- or polycyclic, non-
aromatic, hydrocarbon ring system containing 5 to 10
carbon atoms, which may optionally contain unsaturated
bonds in the ring system;
each aryl is a mono- or polycyclic ring system
which contains 6, 10, 12 or 14 carbons in which at least
one ring of the ring system is aromatic;
each heteroaryl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, and in which at least one ring of the ring
system is aromatic;
each heterocyclyl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, in which the mono- or polycyclic ring system
may optionally contain unsaturated bonds but is not
aromatic-
each heteroatom is sulfur, nitrogen or oxygen;
and

each amino acid side chain is any group attached
to the a carbon of a naturally or non-naturally occuring
amino acid.
2. A compound represented by formula I:


wherein:
Y is:

R6 is H or R6 and Y, together with the nitrogen
to which they are bound, form a ring (g):


(g)
X is -C(R3)2-;
m is 0;
R1 is -H, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S (O)2N (H)-R8,
-S(O)N(H)-R8, -C(O)C(O)N(H)R8, -C(O)CH=CHR8, -C(O)CH2OR8,
-C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O)2N (R8)2, -S(O)N(R8)2,
-C(O)C(O)N(R8)2, -C(O)CH2N(R8)2, -CH2R8, -CH2-alkenyl-R8,
or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or allcynyl-R9, or
R2 and one R3 together with the atoms to which they are
bound, form a 3 to 7 membered cyclic or heterocyclic ring
system, wherein the heteroatoms are independently sulfur,
nitrogen, or oxygen, wherein a hydrogen atom bound to any
-alkyl or -cycloalkyl carbon atom is optionally replaced
by -R10, a hydrogen atom bound to any -aryl or -heteroaryl
carbon atom is optionally replaced by -R11, a hydrogen
atom bound to any nitrogen atom of the ring system is
optionally replaced by -R1;

R4 and one R5 together with the atoms to which
they are bound form a ring system selected from:

and the other R5 is H, wherein a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced with R1, or R4 and one R5 together with the atoms
to which they are bound form a ring system:
and the other R5 is H;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl,
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,
wherein a hydrogen atom bound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by R10, a hydrogen atom


bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by R11, and a hydrogen atom bound to
any nitrogen atom is optionally replaced by R1;
each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11, and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R10 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N (H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C (O)N(H)alkyl, -N(H)C(O)N(alkyl)2,
-S-alkyl, -S-aryl, -S-alkylaryl, -S(O)2alkyl, -S(O)alkyl,
-C(O)alkyl, -CH2NH2, -CH2N(H)alkyl, or -CH2N(alkyl)2,
-alkyl, -cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl, -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11 and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R11 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,


-N(H)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -O-
alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,
-N(H)-alkylaryl, -N(alkyl)2, -C(O)N(H)alkyl,
-C(O)N(alkyl)2, -N (H)C (O)alkyl, -N(H)C(O)N(H)alkyl,
-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2; and
R12 is -C(O)alkyl, -C(O)cycloalkyl,
-C(O)alkylaryl, -C(O)alkylheteroaryl, -C(O)heterocyclyl,
or -C(O)alkylheterocyclyl;
R13 iS -H -alkyl, -aryl, -alkylaryl or -
alkylheteroaryl; wherein
each alkyl is a straight-chained or branched,
saturated aliphatic hydrocarbon containing 1 to 6 carbon
atoms;
each alkenyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one double bond;
each alkynyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one triple bond;
each cycloalkyl is a mono- or polycyclic, non-
aromatic, hydrocarbon ring system containing 5 to 10
carbon atoms, which may optionally contain unsaturated
bonds in the ring system;
each aryl is a mono- or polycyclic ring system
which contains 6, 10, 12 or 14 carbons in which at least
one ring of the ring system is aromatic;

each heteroaryl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, and in which at least one ring of the ring
system is aromatic;
each heterocyclyl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, in which the mono- or polycyclic ring system
may optionally contain unsaturated bonds but is not
aromatic-
each heteroatom is sulfur, nitrogen or oxygen;
and
each amino acid side chain is any group attached
to the a carbon of a naturally or non-naturally occuring
amino acid.
3. A compound represented by formula I:

wherein:
Y is:
(a) or (b)


X is -C(R3)2-;
m is 0;
R1 is -H, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S (O)2N (H)-R8 ,
-S(O)N(H)-R8, -C(O)C(O)N(H)R8, -C(O)CH=CHR8, -C(O)CH2OR8,
-C(O)CH2N(H)R8, -C(O)N(R8)2, -S(O)2N(R8)2, -S(O)N(R8)2,
-C(O)C(O)N(R8)2, -C(O)CH2N(R8)2, -CH2R8, -CH2-alkenyl-R8,
or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9
wherein a hydrogen atom bound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by -R10, a hydrogen
atom bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by -R11, and a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced by -R1;
R4 and one R5 together with the atoms to which
they are bound form a ring system selected from:


and the other R5 is H, wherein a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced with R1, or R4 and one R5 together with the atoms
to which they are bound form a ring system:
and the other R5 is H;
R6 is -H;
R7 is -OH, -OR8 or -N(H)OH;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl,
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,
wherein a hydrogen atom bound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by R10, a hydrogen atom


bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by R11, and a hydrogen atom bound to
any nitrogen atom is optionally replaced by R1;
each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11, and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R10 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N{alkyl)2, -C(O)N(H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl, -N(H)C(O)N(alkyl)2,
-S-alkyl, -S-aryl, -S-alkylaryl, -S(O)2alkyl, -S(O)alkyl,
-C(O)alkyl, -CH2NH2, -CH2N(H)alkyl, -CH2N(alkyl)2, -alkyl,
-cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl, -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11 and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1; and
each R11 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -O-


alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,
-N(H)-alkylaryl, -N(alkyl) 2, -C (O)N(H)alkyl,
-C(O)N(alkyl)2, -N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl,
-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2;
provided that if one R3 is -H, then the other R3
is not -H; wherein
each alkyl is a straight-chained or branched,
saturated aliphatic hydrocarbon containing 1 to 6 carbon
atoms;
each alkenyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one double bond;
each alkynyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one triple bond;
each cycloalkyl is a mono- or polycyclic, non-
aromatic, hydrocarbon ring system containing 5 to 10
carbon atoms, which may optionally contain unsaturated
bonds in the ring system;
each aryl is a mono- or polycyclic ring system
which contains 6, 10, 12 or 14 carbons in which at least
one ring of the ring system is aromatic-
each heteroaryl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, and in which at least one ring of the ring
system is aromatic-
each heterocyclyl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, in which the mono- or polycyclic ring system

may optionally contain unsaturated bonds but is not
aromatic;
each heteroatom is sulfur, nitrogen or oxygen;
and
each amino acid side chain is any group attached
to the a carbon of a naturally or non-naturally occuring
amino acid.
4. A compound represented by formula I:

wherein:
Y is:

X is -C(R3)2-;
m is 0;
R1 is H, -C(O)R8, -C(O)C(O)R8, -S(O)2R8,
-S(O)R8, -C(O)OR8, -C(O)N(H)R8, -S(O)2N (H)-R8 ,
-S(O)N(H)-R8, -C(O)C(O)N(H)R8, -C(O)CH=CHR8, -C(O)CH2OR8,

-C(O)CH2N(H)R8, -C(O)N(R8)2, -S (O)2N (R8)2, -S(O)N(R8)2,
-C(O)C(O)N(R8)2, -C(O)CH2N(R8)2, -CH2R8, -CH2-alkenyl-R8,
or -CH2-alkynyl-R8;
R2 is -H and each R3 is independently -H, an
amino acid side chain, -R8, alkenyl-R9, or alkynyl-R9,
wherein a hydrogen atom hound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by -R10, a hydrogen
atom bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by -R11, and a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced by -R1;
R4 and one R5 together with the atoms to which
they are bound form a ring system selected from:


and the other R5 is H, wherein a hydrogen atom bound to
any nitrogen atom of the ring system is optionally
replaced with R1, or R4 and one R5 together with the atoms
to which they are bound form a ring system:
and the other R5 is H;
R6 is -H;
each R8 is independently -alkyl, -cycloalkyl,
-aryl, -heteroaryl, -heterocyclyl, -alkylcycloalkyl,
-alkylaryl, -alkylheteroaryl, or -alkylheterocyclyl,
wherein a hydrogen atom bound to any -alkyl or -cycloalkyl
carbon atom is optionally replaced by R10, a hydrogen atom
bound to any -aryl or -heteroaryl carbon atom is
optionally replaced by R11, and a hydrogen atom bound to
any nitrogen atom is optionally replaced by R1;
each R9 is independently -aryl, -heteroaryl,
cycloalkyl, or -heterocyclyl, wherein a hydrogen atom
bound to any -alkyl or -cycloalkyl carbon atom is
optionally replaced by R10, a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11, and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R10 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,


-N(H)C(O)NH2, -perfluoroalkyl, -O-alkyl, -O-aryl,
-O-alkylaryl, -N(H)alkyl, -N(H)aryl, -N(H)-alkylaryl,
-N(alkyl)2, -C(O)N(H)alkyl, -C(O)N(alkyl)2,
-N(H)C(O)alkyl, -N(H)C(O)N(H)alkyl, -N(H)C(O)N(alkyl)2,
-S-alkyl, -S-aryl, -S-alkylaryl, -S(O)2alkyl, -S(O)alkyl,
-C(O)alkyl, -CH2NH2, -CH2N(H)alkyl, -CH2N (alkyl)2, -alkyl,
-cycloalkyl, -aryl, -heteroaryl, -heterocyclyl,
-alkylcycloalkyl, -alkylaryl, -alkylheteroaryl, or
-alkylheterocyclyl, wherein a hydrogen atom bound to any
-aryl or -heteroaryl carbon atom is optionally replaced by
R11 and a hydrogen atom bound to any nitrogen atom is
optionally replaced by R1;
each R11 is independently -OH, -SH, -F, -Cl,
-Br, -I, -NO2, -CN, -NH2, -CO2H, -C(O)NH2, -N(H)C(O)H,
-N(H)C(O)NH2, -alkyl, -cycloalkyl, -perfluoroalkyl, -O-
alkyl, -O-aryl, -O-alkylaryl, -N(H)alkyl, -N(H)aryl,
-N(H)-alkylaryl, -N(alkyl)2, -C(O)N(H)alkyl,
-C(O)N(alkyl)2, -N (H) C (O) alkyl, -N (H) C (O) N (H) alkyl,
-N(H)C(O)N(alkyl)2, -S-alkyl, -S-aryl, -S-alkylaryl,
-S(O)2alkyl, -S(O)alkyl, -C(O)alkyl, -CH2NH2,
-CH2N(H)alkyl, or -CH2N(alkyl)2; and
R12 is -C(O)alkyl, -C(O)cycloalkyl,
-C(O)alkylaryl, -C (O)alkylheteroaryl, -C(O)heterocyclyl,
or -C(O)alkylheterocyclyl; wherein
each alkyl is a straight-chained or branched,
saturated aliphatic hydrocarbon containing 1 to 6 carbon
atoms;


each alkenyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one double bond;
each alkynyl is a straight-chained or branched
unsaturated hydrocarbon containing 2 to 6 carbon atoms and
at least one triple bond;
each cycloalkyl is a mono- or polycyclic, non-
aromatic, hydrocarbon ring system containing 5 to 10
carbon atoms, which may optionally contain unsaturated
bonds in the ring system;
each aryl is a mono- or polycyclic ring system
which contains 6, 10, 12 or 14 carbons in which at least
one ring of the ring system is aromatic-
each heteroaryl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, and in which at least one ring of the ring
system is aromatic-
each heterocyclyl is a mono- or polycyclic ring
system which contains 1 to 15 carbon atoms and 1 to 4
heteroatoms, in which the mono- or polycyclic ring system
may optionally contain unsaturated bonds but is not
aromatic;
each heteroatom is sulfur, nitrogen or oxygen;
and
each amino acid side chain is any group attached
to the a carbon of a naturally or non-naturally occuring
amino acid.
5. The compound as claimed in claim 2 or claim
4,wherein Y is:


and V is: CH3O,





6. The compound as claimed in any one of
claims 1-4, wherein one R3 is -H and the other R3 is
methyl, isopropyl, tert-butyl, CH2Salkyl, CH2SO2alkyl,
CH2CH2Salkyl, or CH2CH2SO2alkyl.
7. The compound as claimed in claim 6,wherein
one R3 is -H and the other R3 is methyl.
8. The compound as claimed in claim 7, wherein
R1 is -C(O)R8 or -C(O)C(O)R8.
9. The compound as claimed in any one of
claims 1-4,wherein R4 and one R5 together with the atoms
to which they are bound form a ring system selected from:


and the other R5 is H.
10. The compound as claimed in claim 9, wherein
one R3 is -H and the other R3 is methyl, isopropyl, tert-
butyl, CH2Salkyl, CH2SO2alkyl, CH2CH2Salkyl, or
CH2CH2SO2alkyl.
11. The compound as claimed in claim 10, wherein
one R3 is -H and the other R3 is methyl.
12. The compound as claimed in claim 11, wherein
R1 is -C(O)R8 or -C(O)C(O)R8.
13. The compound as claimed in any one of
claims 1-4,wherein one R4 and one R5 together with the
atoms to which they are bound form a ring system:
, and the other R5 is H.
14. The compound as claimed in claim 13,wherein
one R3 is -H and the other R3 is methyl, isopropyl, tert-
butyl, CH2Salkyl, CH2SO2alkyl, CH2CH2Salkyl, or
CH2CH2SO2alkyl.
15. The compound as claimed in claim 14,wherein
one R3 is -H and the other R3 is methyl.

16. The compound as claimed in claim 15, wherein
R1 is -C(O)R8 or -C(O)C(O)R8.
17. The compound as claimed in claim 1 or claim
3,selected from the group consisting of: 5a-5bd, 7a-7at,
20a-20t, 24d-24e, 52, 57, 61, 65, 69, 73, 121, and 122a-v:






























18. The compound as claimed in claim 4 selected
from the group consisting of: 51, 56, 60, 64, 68, 72, 76-
93, 98a-z, 98aa-az, 98ba, 98bb, 101, 102a, 102b, 110, and
111:














19. A compound selected from the group
consisting of: 71, 75, 109, 120a and 120b:



20. A pharmaceutical composition comprising: a)
a compound as claimed in any one of claims 1-19; and b) a
pharmaceutically acceptable carrier, adjuvant or vehicle.
21. A pharmaceutical composition as claimed in
claim 20, capable of being used in the treatment of a
disease selected from an IL-1 mediated disease, an
apoptosis mediated disease, an inflammatory disease, an
autoimmune disease, a destructive bone disorder, a
proliferative disorder, an infectious disease, a
degenerative disease, a necrotic disease, an excess
dietary alcohol intake disease, a viral mediated disease,
inflammatory peritonitis, osteoarthritis, pancreatitis,
asthma, adult respiratory distress syndrome,
glomerulonephritis, rheumatoid arthritis, systemic lupus
erythematosus, scleroderma, chronic thyroiditis, Grave's
disease, autoimmune gastritis, insulin-dependent diabetes

mellitus (Type I), autoimmune hemolytic anemia, autoimmune
neutropenia, thrombocytopenia, chronic active hepatitis,
myasthenia gravis, inflammatory bowel disease, Crohn's
disease, psoriasis, atopic dermatitis, graft vs host
disease, osteoporosis, leukemias and related disorders,
myelodysplastic syndrome, multiple myeloma-related bone
disorder, acute myelogenous leukemia, chronic myelogenous
leukemia, metastatic melanoma, Kaposi's sarcoma, multiple
myeloma, sepsis, septic shock, Shigellosis, Alzheimer's
disease, Parkinson's disease, cerebral ischemia,
myocardial ischemia, spinal muscular atrophy, multiple
sclerosis, AIDS-related encephalitis, HIV-related
encephalitis, aging, alopecia, neurological damage due to
stroke, ulcerative colitis, traumatic brain injury, organ
transplant rejection, hepatitis-B, hepatitis-C,
hepatitis-G, yellow fever, dengue fever, or Japanese
encephalitis, in a patient.
22. The compound as claimed in any one of
claims 1-4 or 13,wherein R1 is -C(O)R8 or -C(O)C(O)R8.


The present invention relates to novel classes of compounds which are caspase inhibitors, in particular interleukin-1β converting
enzyme ("ICE") inhibitors. This invention also relates to pharmaceutical compositions comprising these compounds. The compounds
and pharmaceutical compositions of this invention are particularly well suited for inhibiting caspase activity and consequently, may be
advantageously used as agents against interleukin-1- ("IL-1"), apoptosis-, interferon-γ inducing factor- (IGIF), or interferon-γ- ("IFN-γ")
mediated diseases, including inflammatory diseases, autoimmune diseases, destructive bone disorders, proliferative disorders, infectious
diseases, and degenerative diseases. This invention also relates to methods for inhibiting caspase activity and decreasing IGIF production
and IFN-γ production and methods for treating interleukin-1, apoptosis-, and interferon-γ- mediated diseases using the compounds and
compositions of this invention. This invention also relates to methods of preparing the compounds of this invention.

Documents:

in-pct-2000-322-kol-abstract-1.1.pdf

in-pct-2000-322-kol-abstract.pdf

in-pct-2000-322-kol-amanded claims.pdf

in-pct-2000-322-kol-amanded pages of specification.pdf

in-pct-2000-322-kol-assignment.pdf

in-pct-2000-322-kol-assignment1.1.pdf

in-pct-2000-322-kol-claims.pdf

in-pct-2000-322-kol-correspondence.pdf

in-pct-2000-322-kol-correspondence1.1.pdf

in-pct-2000-322-kol-correspondence1.2.pdf

in-pct-2000-322-kol-description (complete)-1.1.pdf

in-pct-2000-322-kol-description (complete).pdf

in-pct-2000-322-kol-examination report.pdf

in-pct-2000-322-kol-examination report1.1.pdf

in-pct-2000-322-kol-examination report1.2.pdf

in-pct-2000-322-kol-form 1-1.1.pdf

in-pct-2000-322-kol-form 1.pdf

in-pct-2000-322-kol-form 13-1.1.pdf

in-pct-2000-322-kol-form 13.1.pdf

in-pct-2000-322-kol-form 13.pdf

in-pct-2000-322-kol-form 18.1.pdf

in-pct-2000-322-kol-form 18.pdf

in-pct-2000-322-kol-form 2-1.1.pdf

in-pct-2000-322-kol-form 2.pdf

in-pct-2000-322-kol-form 3-1.1.pdf

in-pct-2000-322-kol-form 3.1.pdf

in-pct-2000-322-kol-form 3.pdf

in-pct-2000-322-kol-form 5.pdf

IN-PCT-2000-322-KOL-FORM-27.pdf

in-pct-2000-322-kol-gpa.pdf

in-pct-2000-322-kol-gpa1.1.pdf

in-pct-2000-322-kol-granted-abstract.pdf

in-pct-2000-322-kol-granted-claims.pdf

in-pct-2000-322-kol-granted-description (complete).pdf

in-pct-2000-322-kol-granted-form 1.pdf

in-pct-2000-322-kol-granted-form 2.pdf

in-pct-2000-322-kol-granted-specification.pdf

in-pct-2000-322-kol-others-1.1.pdf

in-pct-2000-322-kol-others.pdf

in-pct-2000-322-kol-pa.pdf

in-pct-2000-322-kol-petition under rule 137.pdf

in-pct-2000-322-kol-reply to examination report.pdf

in-pct-2000-322-kol-reply to examination report1.1.pdf

in-pct-2000-322-kol-specification.pdf

in-pct-2000-322-kol-translated copy of priority document.pdf


Patent Number 249892
Indian Patent Application Number IN/PCT/2000/322/KOL
PG Journal Number 47/2011
Publication Date 25-Nov-2011
Grant Date 21-Nov-2011
Date of Filing 18-Sep-2000
Name of Patentee VERTEX PHARMACEUTICALS INCORPORATED
Applicant Address 130, WAVERLY STREET, CAMBRIDGE, MA
Inventors:
# Inventor's Name Inventor's Address
1 BEMIS GUY W 256, APPLETON STREET ARLINGTON, MA 01572
2 WANNAMAKER MARION W 375, HARVARD ROAD, STOW MA 01775
3 CHARIFSON PAUL S 7, DARTMOUTH DRIVE FRAMINGHAM, MA 01701
4 LAUFFER DAVID J 254, TAYLOR ROAD, STOW MA 01775
5 MULLICAN MICHAEL D 110, PARKER ROAD, NEEDHAM MA 02194
6 MURCKO MARK A 520, MARSHALL STREET HOLLISTON, MA 01746
7 WILSON KEITH P 6, LONGWOOD DRIVE, HOPKINTON MA 01748
8 JANETKA JAMES W 3612, STEARNS HILL ROAD WALTHAM, MA 02451
9 DAVIES ROBERT J APARTMENT 2K, 225 WELDON STREET, CAMBRIDGE, MA 02109
10 GRILLOT ANNE-LAURE APARTMENT #3R, 31 REGENT STREET, CAMBRIDGE, MA 02140
11 SHI ZHAN APARTMENT 7, 15 SHERIDAN DRIVE, SHREWSBURY, MA 01545
12 FORSTER CORNELIA J 8, NANCY AVENUE, PELHAM NH 03076
PCT International Classification Number C07K 5/023
PCT International Application Number PCT/US1999/05919
PCT International Filing date 1999-03-19
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/078,770 1998-03-19 U.S.A.