Title of Invention

MODULATORS OF CELLULAR ADHESION

Abstract The present invention provides compounds having formula (I): and pharmaceutically acceptable derivative wherein R1-R4, n, p, A, B, D, E, L and AR1 are as described generally and in classes and subclasses herein, and additionally pharmaceutical compositions thereof, and methods for the use thereof for the treatment of disorders mediated by the CD family of cellular adhesion molecules (e.g., LFA-1).
Full Text WO 2005/044817 PCT/US2004/036942
MODULA TORS OF CELLULAR ADHESION
PRIORITY
[0001] This application claims priority to U.S. Provisional Application Nos.: 60/560,517, filed April 8, 2004; and 60/517,535, filed November 5,2003; the entire contents of which are incorporated herein by reference.
BACKGROUND OF THE INVENTION
[0002] Research conducted over the last decade has helped elucidate the molecular events attending cell-cell interactions in the body, especially those events involved in the movement and activation of cells in the immune system. See generally, Springer, T. Nature, 1990, 346, 425-434. Cell surface proteins, and especially the Cellular Adhesion Molecules ("CAMs") and "leukointegrins", including LFA-1, MAC-1 and gpl50.95 (referred to as CD18/CD1 la, CD18/CD11b, and CD18/CDllc, respectively) have correspondingly been the subject of pharmaceutical research and development having as its goal the intervention in the processes of leukocyte extravasation to sites of injury and leukocyte movement to distinct targets. For example, it is presently believed that prior to the leukocyte extravasation, which is a mandatory component of the inflammatory response, activation of integrins constitutively expressed on leukocytes occurs and is followed by a tight ligand/receptor interaction between integrins {e.g., LFA-1) and one or several distinct intercellular adhesion molecules (ICAMs) designated ICAM-1, ICAM-2, ICAM-3 or ICAM-4 which are expressed on blood vessel endothelial cell surfaces and on other leukocytes. The interaction of the CAMs with the leukointegrins is a vital step in the normal functioning of the immune system. It is believed that immune processes such as antigen presentation, T-cell mediated cytotoxicity and leukocyte extravasation all require cellular adhesion mediated by ICAMs interacting with the leukointegrins. See generally Kishimoto, T. K.; Rothlein; R. R. Adv. Pharmacol. 1994, 25, 117-138 and Diamond, M.; Springer, T. Current Biology, 1994,4, 506-532.
[0003] Clearly, because of the role that the interaction of the CAMs and the leukointegrins plays in the immune response, it would be desirable to modulate these

WO 2005/044817 PCT/US2004/036942
specific interactions to achieve a desired therapeutic result (e.g., inhibition of the interaction in the event of an overactive immune response). Significantly, it has been demonstrated that the antagonism of the interaction between the CAMs and the leukointegrins can be realized by agents directed against either component. Specifically, blocking of the CAMs, such as for example ICAM-1, or the leukointegrins, such as for example LFA-1, by antibodies directed against either or both of these molecules effectively inhibits inflammatory responses. In vitro models of inflammation and immune response inhibited by antibodies to CAMs or leukointegrins include antigen or mitogen-induced lymphocyte proliferation, homotypic aggregation of lymphocytes, T-cell mediated cytolysis and antigen-specific induced tolerance. The relevance of the in vitro studies are supported by in vivo studies with antibodies directed against ICAM-1 or LFA-1. For example, antibodies directed against LFA-1 can prevent thyroid graft rejection and prolong heart allograft survival in mice (Gorski, A.; Immunology Today, 1994,15, 251-255). Of greater significance, antibodies directed against ICAM-1 have shown efficacy in vivo as anti-inflammatory agents in human diseases such as renal allograft rejection and rheumatoid arthritis (Rothlein, R. R.; Scharschmidt, L., in: Adhesion Molecules; Wegner, C. D., Ed.; 1994, 1-38, Cosimi, C. B.; et al, J. Immunol. 1990,144, 4604-4612 and Kavanaugh, A.; et al, Arthritis Rheum. 1994, 37, 992-1004) and antibodies directed against LFA-1 have demonstrated immunosuppressive effects in bone marrow transplantation and in the prevention of early rejection of renal allografts (Fischer, A.; et al, Lancet, 1989, 2, 1058-1060 and Le Mauff, B.; et al, Transplantation, 1991, 52,291-295).
[0004] As described above, the use of anti-LFA-1 or anti-ICAM-1 antibodies to antagonize this interaction has been investigated. Additionally, the use of LFA-1 or ICAM-1 peptides, fragments or peptide antagonists {see, for example, US Patents 5,149,780, 5,288,854, 5,340,800, 5,424,399, 5,470,953, Published PCT applications WO 90/03400, WO90/13316, WO90/10652, WO91/19511, WO92/03473, WO94/11400, WO95/28170, JP4193895, EP314863, EP362526, EP362531), and small molecule antagonists have been investigated. For example, several small molecules have been described in the literature which affect the interaction of CAMs and leukointegrins. A natural product isolated from the root of Trichilia rubra was
2

WO 2005/044817 PCT/US2004/036942
found to be inhibitory in an in vitro cell binding assay (Musza, L. L.; et al., Tetrahedron, 1994, 50, 11369-11378). One series of molecules (Boschelli, D. H.; et al, J. Med. Chem. 1994, 37, 111 and Boschelli, D. H.; et al, J. Med. Chem. 1995, 38, 4597-4614) was found to be orally active in a reverse passive Arthus reaction, an induced model of inflammation that is characterized by neutrophil accumulation (Chang, Y. H.; et al, Eur. J. Pharmacol. 1992, 69, 155-164). Another series of molecules was also found to be orally active in a delayed type hypersensitivity reaction in rats (Sanfilippo, P. J.; etal, J. Med. Chem. 1995, 38, 1057-1059). All of these molecules appear to act nonspecifically, either by inhibiting the transcription of ICAM-1 along with other proteins or act intracellularly to inhibit the activation of the leukointegrins by an unknown mechanism, and none appear to directly antagonize the interaction of the CAMs with the leukointegrins.
[0005] Clearly, although several classes of compounds have been investigated for therapeutic use, there remains a need for the development of novel therapeutics that are capable of modulating interactions between CAMs and leukointegrins. In particular, it would be desirable to develop therapeutics capable of selectively targeting (preferably inhibiting) the interaction between LFA-1 and ICAM-1 that would be useful as a therapeutic agent for immune and/or inflammatory disorders.
SUMMARY OF THE INVENTION
[0006] As discussed above, there remains a need for the development of novel therapeutics that are capable of modulating interactions between CAMs and leukointegrins. The present invention provides novel compounds of general formula
(I),

and pharmaceutical compositions thereof, as described generally and in subclasses herein, which compounds are useful as modulators of the CD11/CD18 family of cellular adhesion molecules. Thus these compounds are useful, for
3

WO 2005/044817 PCT/US2004/036942
example, for the treatment of various LFA-1-related disorders including immune and/or inflammatory disorders.
[0007] In yet another aspect, the present invention provides methods for treating any disorder mediated through the CD 11/CD 18 family of cellular adhesion molecules comprising administering to a subject in need thereof a therapeutically effective amount of a compound of the invention.
DEFINITIONS
[0008] The term "aliphatic", as used herein, includes both saturated and unsaturated, straight chain (i.e., unbranched) or branched aliphatic hydrocarbons, which .ire optionally substituted with one or more functional groups. As will be appreciated by one of ordinary skill in the art, "aliphatic" is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl moieties. Thus, as used herein, the term "alkyl" includes straight and branched alkyl groups. An analogous convention applies to other generic terms such as "alkenyl", "alkynyl" and the like. Furthermore, as used herein, the terms "alkyl", "alkenyl", "alkynyl" and the like encompass both substituted and unsubstituted groups. In certain embodiments, as used herein, "lower alkyl" is used to indicate those alkyl groups (substituted, unsubstituted, branched or unbranched) having about 1-6 carbon atoms. [0009] In certain embodiments, the alkyl, alkenyl and alkynyl groups employed in the invention contain about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-6 aliphatic carbon atoms. In yet other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain about 1-4 carbon atoms. Illustrative aliphatic groups thus include, but are not limited to, for example, methyl, ethyl, n-propyl, isopropyl, allyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, sec-pentyl, isopentyl, tert-pentyl, n-hexyl, sec-hexyl, moieties and the like, which again, may bear one or more substituents. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl,
4

WO 2005/044817 PCT/US2004/036942
butenyl, l-methyl-2-buten-l-yl, and the like. Representative alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl and the like. [0010] The term "alicyclic", as used herein, refers to compounds which combine the properties of aliphatic and cyclic compounds and include but are not limited to monocyclic, or polycyclic aliphatic hydrocarbons and bridged cycloalkyl compounds, which are optionally substituted with one or more functional groups. As will be appreciated by one of ordinary skill in the art, "alicyclic" is intended herein to include, but is not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties, which are optionally substituted with one or more functional groups. Illustrative alicyclic groups thus include, but are not limited to, for example, cyclopropyl, -CEfe-cyclopropyl, cyclobutyl, -CEb-cyclobutyl, cyclopentyl, -CH2-cyclopentyl, cyclohexyl, -CH2-cyclohexyl, cyclohexenylethyl, cyclohexanylethyl, norborbyl moieties and the like, which again, may bear one or more substituents. [0011] The term "alkoxy" or "alkyloxy", as used herein refers to a saturated {i.e., O-alkyl) or unsaturated (i.e., O-alkenyl and O-alkynyl) group attached to the parent molecular moiety through an oxygen atom. In certain embodiments, the alkyl group contains about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl group contains about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl group employed in the invention contains about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl group contains about 1-6 aliphatic carbon atoms. In yet other embodiments, the alkyl group contains about 1-4 aliphatic carbon atoms. Examples of alkoxy, include but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, z-butoxy, sec-butoxy, tert-bvtoxy, neopentoxy, n-hexoxy and the like.
[0012] The term "thioalkyl" as used herein refers to a saturated (i.e., S-alkyl) or unsaturated (i.e., S-alkenyl and S-alkynyl) group attached to the parent molecular moiety through a sulfur atom. In certain embodiments, the alkyl group contains about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl group contains about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl group employed in the invention contains about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl group contains about 1-6 aliphatic carbon atoms. In yet other embodiments, the alkyl group contains about 1-4 aliphatic carbon atoms.
5

WO 2005/044817 PCT/US2004/036942
Examples of thioalkyl include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
[0013] The term "alkylamino" refers to a group having the structure -NHR'wherein R' is alkyl, as defined herein. The term "aminoalkyl" refers to a group having the structure NH2R'-, wherein R' isalkyl, as defined herein. In certain embodiments, the alkyl group contains about 1-20 aliphatic carbon atoms. In certain other embodiments, the alkyl group contains about 1-10 aliphatic carbon atoms. In yet other embodiments, the alkyl group employed in the invention contains about 1-8 aliphatic carbon atoms. In still other embodiments, the alkyl group contains about 1-6 aliphatic carbon atoms. In yet other embodiments, the alkyl group contains about 1-4 aliphatic carbon atoms. Examples of alkylamino include, but are not limited to, methylamino, ethylamino, iso-propylamino and the like. [0014] Some examples of substituents of the above-described aliphatic (and other) moieties of compounds of the invention include, but are not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; aryltbid; heteroalkylthio; heteroarylthio; F; Cl; Br; I; -OH; -NO2; -CN; -CF3; -CH2CF3; -CHC12; -CH2OH; -CH2CH2OH; -CH2NH2; -CH2SO2CH3; -C(O)RX; -CO2(RX); -CON(RX)2; -OC(O)RX; -OCO2RX; -OCON(RX)2; -N(RX)2; -S(O)2RX; -NRX(CO)RX wherein each occurrence of Rx independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic, alkylaryl, or alkylheteroaryl substituents 'described above and herein may be substituted or unsubstituted, branched or unbranched, saturated or unsaturated, and wherein any of the aryl or heteroaryl substituents described above and herein may be substituted or unsubstituted. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
[0015] In general, the term "aromatic moiety", as used herein, refers to a stable mono- or polycyclic, unsaturated moiety having preferably 3-14 carbon atoms, each of which may be substituted or unsubstituted. hi certain embodiments, the term
6

WO 2005/044817 PCT/US2004/036942
"aromatic moiety" refers to a planar ring having p-orbitals perpendicular to the plane
of the ring at each ring atom and satisfying the Huckel rule where the number of pi
electrons in the ring is (4n+2) wherein n is an integer. A mono- or polycyclic,
unsaturated moiety that does not satisfy one or all of these criteria for aromaticity is
defined herein as "non-aromatic", and is encompassed by the term "alicyclic".
[0016] In general, the term "heteroaromatic moiety", as used herein, refers to a
stable mono- or polycyclic, unsaturated moiety having preferably 3-14 carbon
atoms, each of which may be substituted or unsubstituted; and comprising at least
one heteroatom selected from O, S and N within the ring Q.e., in place of a ring
carbon atom). In certain embodiments, the term "heteroaromatic moiety" refers to a
planar ring comprising at least on eheteroatom, having p-orbitals perpendicular to
the plane of the ring at each ring atom, and satisfying the Huckel rule where the
number of pi electrons in the ring is (4n+2) wherein n is an integer.
[0017] It will also be appreciated that aromatic and heteroaromatic moieties, as
defined herein may be attached via an alkyl or heteroalkyl moiety and thus also
include -(alkyl)aromatic, -(heteroalkyl)aromatic, -(heteroalkyl)heteroaromatic, and
—(heteroalkyl)heteroaromatic moieties. Thus, as used herein, the phrases "aromatic
or heteroaromatic moieties" and "aromatic, heteroaromatic, -(aUcyl)aromatic, -
(heteroalkyl)aromatic, -(heteroalkyl)heteroaromatic, and -
(heteroalkyl)heteroaromatic" are interchangeable. Substituents include, but are not
limited to, any of the previously mentioned substituents, i.e., the substituents recited
for aliphatic moieties, or for other moieties as disclosed herein, resulting in the
formation of a stable compound. ,
[0018] The term "aryl", as used herein, does not differ significantly from the common meaning of the term in the art, and refers to an unsaturated cyclic moiety comprising at least one aromatic ring. In certain embodiments, "aryl" refers to a mono- or bicyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, indenyl and the like.
[0019] The term "heteroaryl", as used herein, does not differ significantly from the common meaning of the term in the art, and refers to a cyclic aromatic radical having from five to ten ring atoms of which one ring atom is selected from S, O and
7

WO 2005/044817 PCT/US2004/036942
N; zero, one or two ring atoms are additional heteroatoms independently selected from S, 0 and N; and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms, such as, for example, pyridyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, and the like.
[0020] It will be appreciated that aryl and heteroaryl groups (including bicyclic aryl groups) can be unsubstituted or substituted, wherein substitution includes replacement of one or more of the hydrogen atoms thereon independently with any one or more of the following moieties including, but not limited to: aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; heteroarylthio; F; Cl; Br; I; -OH; -NO2; -CN; -CF3; -CH2CF3; -CHC12; -CH2OH; -CH2CH2OH; -CH2NH2; -CH2SO2CH3; -C(O)RX; -CO2(RX); -CON(RX)2; -OC(O)RX; -OCO2RX; -OCON(RX)2; -N(RX)2; -S(O)RX; -S(O)2RX; -NRX(CO)RX wherein each occurrence of Rx independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic, alkylaryl, or alkylheteroaryl substituents described above and herein may be substituted or unsubstituted, branched or unbranched, saturated or unsaturated, and wherein any of the aromatic, heteroaromatic, aryl, heteroaryl, -(alkyl)aryl or -(alkyl)heteroaryl substituents described above and herein may be substituted or unsubstituted. Additionally, it will be appreciated, that any two adjacent groups taken together may represent a 4, 5, 6, or 7-membered substituted or unsubstituted alicyclic or heterocyclic moiety. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
[0021] The term "cycloalkyl", as used herein, refers specifically to groups having three to seven, preferably three to ten carbon atoms. Suitable cycloalkyls include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like, which, as in the case of aliphatic, alicyclic, heteroaliphatic
8

WO 2005/044817 PCT/US2004/036942
or heterocyclic moieties, may optionally be substituted with substituents including, but not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkyltbio; heteroarylthio; F; Cl; Br; I; -OH; -NO2; -CN; -CF3; -CH2CF3; -CHC12; -CH2OH; -CH2CH2OH; -CH2NH2; -CH2SO2CH3; -C(O)RX; -CO2(RX); -CON(RX)2; -OC(O)RX; -OCO2RX; -0CON(Rx)2; -N(RX)2; -S(O)2RX; -NRX(CO)RX wherein each occurrence of Rx independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic. alkylaryl, or alkylheteroaryl substituents described above and herein may be substituted or unsubstituted, branched or unbranched, saturated or usaturated, and wherein any of the aromatic, heteroaromatic, aryl or heteroaryl substituents described above and herein may be substituted or unsubstituted. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
[0022] The term "heteroaliphatic", as used herein, refers to aliphatic moieties in which one or more carbon atoms in the main chain have been substituted with a heteroatom. Thus, a heteroaliphatic group refers to an aliphatic chain which contains one or more oxygen, sulfur, nitrogen, phosphorus or silicon atoms, e.g., in place of carbon atoms. Heteroaliphatic moieties may be linear or branched, and saturated o runsaturated. In certain embodiments, heteroaliphatic moieties are substituted by independent replacement of one or more of the hydrogen atoms thereon with one or more moieties including, but not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; alkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; heteroarylthio; F; Cl; Br; I; -OH; -NO2; -CN; -CF3; -CH2CF3; -CHC12; -CH2OH; -CH2CH2OH; -CH2NH2; -CH2SO2CH3; -C(O)RX; -CO2(RX); -CON(RX)2; -OC(O)RX; -OCO2RX; -OCON(RX)2; -N(RX)2; -S(O)2RX; -NRX(CO)RX wherein each occurrence of Rx independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic, aryl,
9

WO 2005/044817 PCT/US2004/036942
heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic, alkylaryl, or alkylheteroaryl substituents described above and herein may be substituted or unsubstituted, branched or unbranched, saturated or unsaturated, and wherein any of the aromatic, heteroaromatic, aryl or heteroaryl substituents described above and herein may be substituted or unsubstituted. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
[0023] The term "heterocycloalkyl", "heterocycle" or "heterocyclic", as used herein, refers to compounds which combine the properties of heteroaliphatic and cyclic compounds and include, but are not limited to, saturated and unsaturated mono- or polycyclic cyclic ring systems having 5-16 atoms wherein at least one ring atom is a heteroatom selected from O, S and N (wherein the nitrogen and sulfur heteroatoms may be optionally be oxidized), wherein the ring systems are optionally substituted with one or more functional groups, as defined herein. In certain embodiments, the term "heterocycloalkyl", "heterocycle" or "heterocyclic" refers to a non-aromatic 5-, 6- or 7- membered ring or a polycyclic group wherein at least one ring atom is a heteroatom selected from O, S and N (wherein the nitrogen and sulfur heteroatoms may be optionally be oxidized), including, but not limited to, a bi- or tri-cyclic group, comprising fused six-membered rings having between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally be oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl ring. Representative heterocycles include, but are not limited to, heterocycles such as furanyl, thiofuranyl, pyranyl, pyrrolyl, thienyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, dioxazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, triazolyl, thiatriazolyl, oxatriazolyl, thiadiazolyl, oxadiazolyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, dithiazolyl, dithiazolidinyl, tetrahydrofuryl, and benzofused
10

WO 2005/044817 PCT/US2004/036942
derivatives thereof. In certain embodiments, a "substituted heterocycle, or heterocycloalkyl or heterocyclic" group is utilized and as used herein, refers to a heterocycle, or heterocycloalkyl or heterocyclic group, as defined above, substituted by the independent replacement of one, two or three of the hydrogen atoms thereon with but are not limited to aliphatic; alicyclic; heteroaliphatic; heterocyclic; aromatic; heteroaromatic; aryl; heteroaryl; alkylaryl; heteroalkylaryl; alkylheteroaryl; heteroalkylheteroaryl; alkoxy; aryloxy; heteroalkoxy; heteroaryloxy; alkylthio; arylthio; heteroalkylthio; heteroarylthio; F; Cl; Br; I; - OH; -NO2; -CN; -CF3; -CH2CF3; -CHC12; -CH2OH; -CH2CH2OH; -CH2NH2; -CH2SO2CH3; -C(O)RX; -CO2(RX); -CON(RX)2; -OC(O)RX; -OCO2RX; -OCON(RX)2; -N(RX)2; -S(O)2RX; -NRX(CO)RX wherein each occurrence of Rx independently includes, but is not limited to, aliphatic, alicyclic, heteroaliphatic, heterocyclic, \ aromatic, heteroaromatic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl, wherein any of the aliphatic, alicyclic, heteroaliphatic, heterocyclic, alkylaryl, or alkylheteroaryl substituents described above and herein may be substituted or unsubstituted, branched or unbranched, saturated or unsaturated, and wherein any of the aromatic, heteroaromatic, aryl or heteroaryl substitutents described above and herein may be substituted or unsubstituted. Additional examples or generally applicable substituents are illustrated by the specific embodiments shown in the Examples, which are described herein. [0024] Additionally, it will be appreciated that any of the alicyclic or heterocyclic moieties described above and herein may comprise an aryl or heteroaryl moiety fused thereto. Additional examples of generally applicable substituents are illustrated by the specific embodiments shown in the Examples that are described herein.
[0025] The terms "halo" and "halogen" as used herein refer to an atom selected from fluorine, chlorine, bromine and iodine.
[0026] The term "haloalkyl" denotes an alkyl group, as defined above, having one, two, or three halogen atoms attached thereto and is exemplified by such groups as chloromethyl, bromoethyl, trifiuoromethyl, and the like.
[0027] The term "amino", as used herein, refers to a primary (-NH2), secondary (-NHRX), tertiary (-NRxRy) or quaternary (-N+RxRyRz) amine, where Rx, Ry and Rz
11

WO 2005/044817 PCT/US2004/036942
are independently an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or
heteroaromatic moiety, as defined herein. Examples of amino groups include, but
are not limited to, methylamino, dimethylamino, ethylamino, diethylamino,
diethylaminocarbonyl, methylethylamino, iso-propylamino, piperidino,
trimethylarnino, and propylamino.
[0028] The term "acyl", as used herein, refers to a group having the general
formula -C(=O)R, where R is an aliphatic, alicyclic, heteroaliphatic, heterocyclic,
aromatic or heteroaromatic moiety, as defined herein.
[0029] The term "sulfonamido", as used herein, refers to a group of the general
formula -SC^NRxRy, where Rx and Ry are independently hydrogen, or an aliphatic,
alicyclic, heteroaliphatic, heterocyclic, aromatic, heteroaromatic or acyl moiety, as
defined herein.
[0030] The term "benzamido", as used herein, refers to a group of the general
formula PhNRx-, where Rx is hydrogen, or an aliphatic, alicyclic, heteroaliphatic,
heterocyclic, aromatic, heteroaromatic or acyl moiety, as defined herein.
[0031] The term "Ci.6alkylidene", as used herein, refers to a substituted or
unsubstituted, linear or branched saturated divalent radical consisting solely of
carbon and hydrogen atoms, having from one to six carbon atoms, having a free
valence "-" at both ends of the radical.
[0032] The term "C2-6alkenylidene", as used herein, refers to a substituted or
unsubstituted, linear or branched unsaturated divalent radical consisting solely of
carbon and hydrogen atoms, having from two to six carbon atoms, having a free
valence "-" at both ends of the radical, and wherein the unsaturation is present only
as double bonds and wherein a double bond can exist between the first carbon of the
chain and the rest of the molecule.
[0033] As used herein, the terms "aliphatic", "heteroaliphatic", "alkyl",
"alkenyl", "alkynyl", "heteroalkyl", "heteroalkenyl", "heteroalkynyl", and the like
encompass substituted and unsubstituted, saturated and unsaturated, and linear and
branched groups. Similarly, the terms "alicyclic", "heterocyclic",
"heterocycloalkyl", "heterocycle" and the like encompass substituted and
unsubstituted, and saturated and unsaturated groups. Additionally, the terms
"cycloalkyl", "cycloalkenyl", "cycloalkynyl", "heterocycloalkyl",
12

WO 2005/044817 PCT/US2004/036942
"heterocycloalkenyl", "heterocycloalkynyl", "aromatic", "heteroaromatic", "aryl", "heteroaryl" and the like encompass both substituted and unsubstituted groups. [0034] By the term "protecting group", has used herein, it is meant that a particular functional moiety, e.g., O, S, or N, is temporarily blocked so that a reaction can be carried out selectively at another reactive site in a multifunctional compound. In preferred embodiments, a protecting group reacts selectively in good yield to give a protected substrate that is stable to the projected reactions; the protecting group must be selectively removed in good yield by readily available, preferably nontoxic reagents that do not attack the other functional groups; the protecting group forms an easily separable derivative (more preferably without the generation of new stereogenic centers); and the protecting group has a minimum of additional functionality to avoid further sites of reaction. As detailed herein, oxygen, sulfur, nitrogen and carbon protecting groups may be utilized. For example, in certain embodiments, as detailed' herein, certain exemplary oxygen protecting groups are utilized. These oxygen protecting groups include, but are not limited to methyl ethers, substituted methyl ethers (e.g., MOM (methoxymethyl ether), MTM (methylthiomethyl ether), BOM (benzyloxymethyl ether), PMBM or MPM (p-methoxybenzyloxymethyl ether), to name a few), substituted ethyl ethers, substituted benzyl ethers, silyl ethers (e.g., TMS (trimethylsilyl ether), TES (triethylsilylether), TIPS (triisopropylsilyl ether), TBDMS (t-butyldimethylsilyl ether), tribenzyl silyl ether, TBDPS (t-butyldiphenyl silyl ether), to name a few), esters (e.g., formate, acetate, benzoate (Bz), trifluoroacetate, dichloroacetate, to name a few), carbonates, cyclic acetals and ketals. In certain other exemplary embodiments, nitrogen protecting groups are utilized. These nitrogen protecting groups include, but are not limited to, carbamates (including methyl, ethyl and substituted ethyl carbamates (e.g., Troc), to name a few) amides, cyclic imide derivatives, N-Alkyl and N-Aryl amines, imine derivatives, and enamine derivatives, to name a few. Certain other exemplary protecting groups are detailed herein, however, it will be appreciated that the present invention is not intended to be limited to these protecting groups; rather, a variety of additional equivalent protecting groups can be readily identified using the above criteria and utilized in the present invention. Additionally, a variety of protecting groups are described in
13

WO 2005/044817 PCT/US2004/036942
"Protective Groups in Organic Synthesis" Third Ed. Greene, T.W. and Wuts, P.G., Eds., John Wiley & Sons, New York: 1999, the entire contents of which are hereby incorporated by reference.
[0035] The term "natural amino acid" as used herein refers to any one of the common, naturally occurring L-amino acids found in naturally occurring proteins: glycine (Gly), alanine (Ala), valine (Val), leucine (Leu), isoleucine (He), lysine (Lys), arginine (Arg), histidine (His), proline (Pro), serine (Ser), threonine (Thr), phenylalanine (Phe), tyrosine (Tyr), tryptophan (Trp), aspartic acid (Asp), glutamic acid (Glu), asparagine (Asn), glutamine (Gin), cysteine (Cys) and methionine (Met). [0036] The term "unnatural amino acid" as used herein refers to all amino acids which are not natural amino acids. This Includes, for example, a-, p-, D-, L- amino
acid residues, and compounds of the general formula o wherein the
side chain R is other than the amino acid side chains occurring in nature.
[0037] More generally, the term "amino acid", as used herein, encompasses
natural amino acids and unnatural amino acids.
[0038] The term "bioisosteres", as used herein, generally refers to two or more
compounds or moieties that possess similar molecular shapes and/or volumes. In
certain embodiments, bioisosteres have approximately the same distribution of
electrons. In certain other embodiments, bioisosteres exhibit similar biological
properties. In preferred embodiments, bioisosteres possess similar molecular shapes
and volumes; have approximately the same distribution of electrons; and exhibit
similar biological properties.
[0039] As used herein, the term "isolated", when applied to the compounds of
the present invention, refers to such compounds that are (i) separated from at least
some components with which they are associated in nature or when they are made
and/or (ii) produced, prepared or manufactured by the hand of man.
[0040] The term, "pharmaceutically acceptable derivative", as used herein,
denotes any pharmaceutically acceptable salt, ester, or salt of such ester, of such
compound, or any other adduct or derivative which, upon administration to a patient,
is capable of providing (directly or indirectly) a compound as otherwise described
14

WO 2005/044817 PCT/US2004/036942
herein, or a metabolite or residue thereof. Pharmaceutically acceptable derivatives thus include among others pro-drugs. A pro-drug is a derivative of a compound, usually with significantly reduced pharmacological activity, which contains an additional moiety, which is susceptible to removal in vivo yielding the parent molecule as the pharmacologically active species. An example of a pro-drug is an ester, which is cleaved in vivo to yield a compound of interest. Pro-drugs of a variety of compounds, and materials and methods for derivatizing the parent compounds to create the pro-drugs, are known and may be adapted to the present invention. Certain exemplary pharmaceutical compositions and pharmaceutically acceptable derivatives will be discussed in more detail herein below. [0100] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts of amines, carboxylic acids, and other types of compounds, are well known in the art. For example, S.M. Berge, et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977), incorporated herein by reference. The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting a free base or free acid function with a suitable reagent, as described generally below. For example, a free base function can be reacted with a suitable acid. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may, include metal salts such as alkali metal salts, e.g. sodium or potassium salts; and alkaline earth metal salts, e.g. calcium or magnesium salts. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate,
15

WO 2005/044817 PCT/US2004/036942
camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fomarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulforfate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0101] As used herein, the term "pharmaceutically acceptable ester" refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
[0041] As used herein, the term "pharmaceutically acceptable prodrugs" refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the issues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drags as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American
16

WO 2005/044817 PCT/US2004/036942
Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
[0042] The term "LFA-1 mediated disorders", as used herein refers generally to pathological states caused by cell adherence interactions involving the LFA-1 receptor on lymphocytes. Examples of such disorders include, but are not limited to T-cell inflammatory responses such as inflammatory skin diseases including psoriasis; responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); adult respiratory distress syndrome, dermatitis, meningitis, encephalitis, uveitic, allergic conditions such as eczema and asthma and other conditions involving infiltration of T-cells and chronic inflammatory responses, skin hypersensitivJty reactions (including poison ivy and poison oak), atherosclerosis, leukocyte adhesion deficiency, autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus (SLE), diabetes mellitus, multiple sclerosis, Reynaud's syndrome, autoimmune thyroiditis, experimental autoimmune encephalomyelitis, Sjorgen's syndrome, type 1 diabetes, juvenile onset diabetes, and immune responses associated with delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis, and vasculitis, pernicious anemia, diseases involving leukocyte diapedesis, CNS inflammatory disorder, multiple organ injury syndrome secondary to septicaemia or trauma, autoimmune haemolytic anemia, myethamia gravis, antigen-antibody complex mediated diseases, and all types of transplantations, including graft vs. host or host vs. graft disease. [0043] The term "LFA-1 antagonist", as used herein, generally refers to inventive compounds, as described herein, that act as a competitive inhibitors of the CD1 la and/or CD18 interaction with ICAM-1, ICAM-2 or ICAM-3. [0044] The term "treating", as used herein generally means that the compounds of the invention can be used in humans or animals with at least a tentative diagnosis of disease. The compounds of the invention will delay or slow the progression of the disease thereby extending the individual's life span.
[0045] The term "preventing" as used herein generally means that the compounds of the present invention are useful when administered to a patient who has not been diagnosed as possibly having the disease at the time of administration,
17

WO 2005/044817 PCT/US2004/036942
but who would normally be expected to develop the disease or be at increased risk for the disease. In certain embodiments, compounds of the invention slow the development of disease symptoms, delay the onset of disease, or prevent the individual from developing the disease at all.
[0046] As used herein the term "'biological sample " includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from an animal (e.g., mammal) or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. For example, the term "biological sample" refers to any solid or fluid sample obtained from, excreted by or secreted by any living organism, including single-celled micro-organisms (such as bacteria and yeasts) and multiceilular organisms (such as plants and animals, for instance a vertebrate or a mammal, and in particular a healthy or apparently healthy human subject or a human patient affected by a condition or disease to be diagnosed or investigated). The biological sample can be in any form, including a solid material such as a tissue, cells, a cell pellet, a cell extract, cell homogenates, or cell fractions; or a biopsy, or a biological fluid. The biological fluid may be obtained from any site (e.g. blood, saliva (or a mouth wash containing buecal cells), tears, plasma, serum, urine, bile, cerebrospinal fluid, amniotic fluid, peritoneal fluid, and pleura! fluid, or cells therefrom, aqueous or vitreous humor, or any bodily secretion), a transudate, an exudate (e.g. fluid obtained from an abscess or any other site of infection or inflammation), or fluid obtained from a joint (e.g. a normal joint or a joint affected by disease such as rheumatoid arthritis, osteoarthritis, gout or septic arthritis). The biological sample can be obtained from any organ or tissue (including a biopsy or autopsy specimen) or may comprise cells (whether primary cells or cultured cells) or medium conditioned by any cell, tissue or organ. Biological samples may also include sections of tissues such as frozen sections taken for histological purposes. Biological samples also include mixtures of biological molecules including proteins, lipids, carbohydrates and nucleic acids generated by partial or complete fractionation of cell or tissue homogenates. Although the sample is preferably taken from a human subject, biological samples may be from any animal, plant, bacteria, virus, yeast, etc. The term animal, as used herein, refers to humans as well as non-human animals, at any stage of development, including, for example, mammals, birds,
18

WO 2005/044817 PCT/US2004/036942
reptiles, amphibians, fish, worms and single cells. Cell cultures and live tissue samples are considered to be pluralities of animals. In certain exemplary embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). An animal may be a transgenic animal or a human clone. If desired, the biological sample may be subjected to preliminary processing, including preliminary separation techniques.
DETAILED DESCRITPION OF CERTAIN PREFERRED EMBODIMENTS OF THE
INVENTION
[0047] The present invention provides compounds that modulate interactions between intracellular adhesion molecules (e.g., ICAM-1, -2 and -3) and the leukocyte integral family of receptors. In certain embodiments, the inventive compounds are antagonists and are useful for the treatment of CD11/CD18 mediated disorders, hi certain embodiments of special interest, the inventive compounds are useful for the treatment of Mac-1 and LFA-1 mediated disorders. In still other embodiments, the compounds are useful for the treatment of LFA-1 mediated disorders, for example, inflammatory disorders and autoimmune disorders to name a few.
[0048] 1) General Description of Compounds of the Invention
[0049] The compounds of the invention include compounds of the general
formula (I) as further defined below:

and pharmaceutically acceptable derivatives thereof;
wherein R1 and R2 are each independently hydrogen, an amino acid side chain, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or wherein R1 and R2 taken together are an alicyclic or
19

WO 2005/044817 PCT/US20O4/036942


heterocyclic moiety, or together are R1A; wherein R1A is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety;
R3 is -C(=O)OR3A, -C(O)H, -CH2OR3A, -CH2O-C(=O)-alkyl, -C(=O)NH(R3A) or -CH2X°; wherein each occurrence of R3A is independently hydrogen, a protecting group, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or R3A, taken together with R1 or R2, forms a heterocyclic moiety; wherein X° is a halogen selected from F, Cl, Br or I;
R4, for each occurrence, is independently hydrogen, halogen, -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO^-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRmC(=O)- or -SO2NRG2-, and RG1 and R02 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety;
n is an integer from 0-3;
AR1 is an aromatic, heteroaromatic, alicyclic or heterocyclic moiety;
A, B, D and E are connected by either a single or double bond, as valency permits; wherein each occurrence of A, B, D and E is independently C=O, CR'R", NR1, CR1, N, O, S, S(=O) or SO2; wherein each occurrence of R1 is independently hydrogen, halogen, -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NR^C^O)- or -SO2NR02-, and RG1 and R02 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or any two adjacent occurrences of R1, taken together, represent an alicyclic, heterocyclic, aromatic or heteroaromatic moiety;
p is an integer from 0-4; and
L is absent or is V-W-X-Y-Z, wherein each occurrence of V, W, X, Y and Z is independently absent, C=O, NRL1, -O-, -C(RL1)=, =C(RL1)-, -C(RL1)(RU), C(=N-O-RL1), C(=N-RL1), -N=, S(O)0-2; a substituted or unsubstituted C^alkylidene or C2. 6alkenylidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O), -CO2-, -C(=O)C(=O), -C(=0)NR13-,
20

WO 2005/044817 PCT/US2004/036942
-OC(=O)-, -OC(=O)NRL3-, -NRL3NRL4-, -NRL3NRL4C(=O)-, -NRL3C(=O)-, -NRL3COa-, -NRL3C(=0)NRLA-, -S(=O)-, -SO2-, -NRUSO2-, -SO2NR13-, -NRL3SO2NRL4-, -O-, -S-, or -NRL3-; wherein each occurrence of Ru and Ru is independently hydrogen, alkyl, heteroalkyl, aromatic, heteroaromatic or acyl; or an
5 aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety; and each occurrence of RL1 and Ru is independently hydrogen, hydroxyl, protected hydroxyl, amino, protected amino, thio, protected thio, halogen, cyano, isocyanate, carboxy, carboxyalkyl, formyl, formyloxy, azido, nitro, ureido, thioureido, thiocyanato, alkoxy, aryloxy, mercapto, sulfonamido, benzamido, tosyl, or an
10 aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety, or wherein one or more occurrences of RL1 and Ru, taken together, or taken together with one of V, W, X, Y or Z form an alicyclic or heterocyclic moiety or form an aromatic or heteroaromatic moiety. [0050] In another aspect, the invention provides compounds of formula (II):



wherein AR has one of the following structures:

and pharmaceutically acceptable derivatives thereof; wherein R1, R2, R3, R4, A, B, D, E, n, p are as defined generally above and in classes and subclasses herein; and
Y1, Y2 and Y3 are each independently CR4 or N; -with the proviso that, when AR has the structure:


wherein Y1 is CH or N and p is 0-2, then R4 is not carbocycle, aryl, heteroaryl or heterocyle, and A, B, D and E do not comprise a carbocyclic, aryl, heteroaryl or heterocyclic moiety.
21

WO 2005/044817

PCT/US2004/036942

[0051] In certain embodiments, for compounds of formula (II), AR represents a moiety having one of the following structures:

wherein each occurrence of n is an integer from 0-6; each occurrence of R4 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-,-OC(=O)-, -NRG2C(=O) or -
22

WO 2005/044817 PCT/US2004/036942
SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is 0 RP5 may also be hydrogen; and each occurrence of R4A is 5 independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic moiety or a nitrogen protecting group; wherein any two adjacent occurrences of R4 and R4A, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety. In certain exemplary embodiments, AR has the structure:
In yet other exemplary embodiments, AR has the structure:
10 wherein each occurrence of Xo is independently a halogen selected from
F, Cl, Br and I. In certain embodiments, each occurrence of X° is Cl.
[0052] A number of important subclasses of each of the foregoing classes deserve separate mention; these subclasses include subclasses of the foregoing
15 classes in which:
[0053] i) R1 and R2 are each independently hydrogen, an amino acid side chain, -(CH2)mOH, -(CH2)maryl, -(CH2)mheteroaryl, wherein m is 0-6, -CH(R1A)(OR1B), -CH(R1A)(NHR1B), U-T-Q, or an alkyl, cycloalkyl, heteroalkyl or heterocyclic moiety optionally substituted with U-T-Q, wherein U is absent, -O-, -S(O)0-2-, -SO2N(R1A),
20 -N(R1A)-, -N(R1A)C(=O)-, -N(R1A)C(=O)-O-, -N(R1A)C(=O)-N(R1B)-, -N(R1A)-SO2-, -C(=O)-, -C(=O)-O-, -O-C(=O)-, aryl, heteroaryl, alkylaryl, alkylheteroaryl, -C(=O)-N(RIA)-, -O-C(=O)-N(R1A)-, -C(=N-R1E)-, -C(=N-R1E)-O-, -C(=N-R1E)-N(R1A)-, -O-C(=N-R1E)-N(R1A), -N(R1A)C(=N-RIE), -N(R1A)C(=N-R1E)-O-, N(R1A)C(=N-R1E)-N(R1B)-, -P(=O)(OR1A)-O-, or -P(=O)(R1A)-O-; wherein T is
25 absent, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl moiety, and wherein Q is hydrogen, halogen, cyano, isocyanate, -OR1B, -SR1B; -N(RIB)2, -NHC(=O)OR1B, -NHC(=O)N(R1B)2, -NHC(=O)R1B, -NHSO2RIB, -NHSO2N(R1B)2, NHSO2NHC(=0)OR1B, -NHC(=O)NHSO2R1B, -C(=O)NHC(=O)OR1B,
23

WO 2005/044817 PCT/US2004/036942
C(=O)NHC(=O)R1B, -C(=O)NHC(=O)N(R1B)2, -C(=O)NHSO2R1B C(=O)NHSO2N(R1B)2, -C(=S)N(R1B)2, -SO2R1B, -SO2-O-R1B, -SO2-N(R1B)2, -SO2-NHC(=O)OR1B, -SO2-NHC(=0)-N(R1B)2, -SO2-NHC(=O)R1B, -O-C(=O)N(R1B)2, -O-C(=O) R1B, -O-C(=O)NHC(=O)R1B, -O-C(=O)NH-SO2R1B, -O-SO2R1B, or an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl or heteroaryl moiety, or wherein R1 and R2 taken together are a cycloalkyl or heterocyclic moiety, or together are
wherein each occurrence of R1A and R1B is independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl or heteroaryl moiety, -COR1C, or -CONR1CR1D; wherein each occurrence of R1C and Rir> is independently hydrogen, hydroxyl, or an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl or heteroaryl moiety; and R1E is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, -CN, -OR1C, -NR1CR1D or -SO2R1C;
[0054] ii) R3 is carboxyl, protected carboxyl or a prodrug thereof, wherein R3 is
C(=O)R3A, wherein R3A is hydroxy, alkoxy, cycloalkoxy, aralkoxy, arcycloalkoxy,
aryloxy, alkylcarbonyloxyalkyloxy, alkoxycarbonyloxyalkyloxy,
alkoxycarbonylalkyloxy, cycloalkylcarbonyloxyalkyloxy,
cycloalkoxycarbonyloxyalkyloxy, cycloalkoxycarbonylalkyloxy,
arylcarbonyloxyalkyloxy, aryloxycarbonyloxyalkyloxy, arylcarbonyloxyalkyloxy, alkoxyalkylcarbonyloxyalkyloxy, or one or the structures:
[0055] iii) R3 is -C(=O)OR3A, -C(=O)H, -CH2OR3A, -CH2O-C(=O)-alkyl, -C(=O)NH(R3A), or -CH2X0; wherein each occurrence of R3A is independently
24

WO 2005/044817 PCT/US2004/036942
hydrogen, a protecting group, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl,
heteroaryl, alkylaryl or alkylheteroaryl moiety, or R3A, taken together with R1 or R2,
forms a heterocyclic moiety; wherein X° is a halogen selected from F, Cl, Br or I;
[0056] iv) R3 is -C(=O)OR3A; wherein R3A is hydrogen, a protecting group, an
alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or
alkylheteroaryl moiety, or R3A, taken together with R1 or R2, forms a heterocyclic
moiety;
[0057] v) R3 is -C(=O)OR3A; wherein R3A is d.5alkyl;
[0058] vi) R3 is -C(=O)OR3A; wherein R3A is Ci.3alkyl;
[0059] vii) R3 is -C(=O)OR3A; wherein R3A is ethyl;
[0060] viii) R3 is-C(=O)OR3A; wherein R3A is benzyl;
[0061] ix) R3 is CO2H;
[0062] x) R3 is -C(=O)OR3A, wherein R3A is as defined in any one of subsets ii)-
ix) above, and -C(=O)NHC(R1)(R2)R3 is a moiety having the following structure:

wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and R is hydrogen, alkyl, heteroalkyl, aryl, heteroaryl, or is -G°RG1 wherein G° is -O-, -S- or -NR02-, and RG1 and RG2 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety; [0063] xi) Compounds of subset x) above wherein -C(=O)NHCH(CO2R3A)CH(Rs)Ar2 has the following stereochemistry:

[0064] xii) R3 is -C(=O)0R3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C^CVJNHCCR^CR^R3 is a moiety having the following structure:

25

WO 2005/044817 PCT/US2004/036942
wherein R1A is Ar2> -OR1B, -SR1B or -NR1BR1C; or an alkyl or heteroalkyl moiety; and Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; wherein R1B and R1C are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclic, aryl, heteroaryl, or R1B and R1C, taken together with the nitrogen atom to which they are attached, form a heterocylic or heteroaryl moiety; [0065] xiii) Compounds of subset xii) above wherein -C(=O)NHCH(CO2R3A)CH2NHC(=O)R1A has the following stereochemistry:

[0066] xiv) R3 is -C(=O)OR3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C(=O)NHC(R1)(R2)R3 is a moiety having the following structure:

wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and R2A is hydrogen, d.6alkyl, C2.6alkenyl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; or R2A, taken together with a substituent on Ar2, forms a substituted or unsubstituted heterocyclic or heteroaryl moiety;
[0067] xv) Compounds of subset xiv) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)Ar2 has the following stereochemistry:

[0068] xvi) R3 is -C(=O)OR3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C(=O)NHC(R1)(R2)R3 is a moiety having the following structure:
26

WO 2005/044817 PCT/US2004/036942

wherein R2A is hydrogen, Ci^alkyl, C2-6aIkenyl, aryl, heteroaryl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; or R2A, taken together with R2C or R2D, forms a substituted or
5 unsubstituted heterocyclic or heteroaryl moiety; R2 is hydrogen, CN, -C=NMe, =NO2, =NC(=O)NH2, =NS(O)2R, =NS(0)2NRR\ -SO2R2G, or an aHphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; wherehi R and R' are each independently hydrogen or methyl, and R2G is lower alkyl; and R2D is Ar2, hydrogen, halogen, CN, NO2, an aliphatic, heteroaliphatic,
10 alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -COCONR02-, -OC(=O)-, -NH3CCO)- or -SOaNR02-, and R and R are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; [0069] xvii) Compounds of subset xvi) above wherein -
15 C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following stereochemistry:

[0070] xviii) Compounds of subset xvii) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following structure:

20 wherein R2E and R2F are each independently hydrogen, or an aliphatic,
alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or
27

WO 2005/044817 PCT/US2004/036942
alkylheteroaryl moiety, or R2E and R2F , taken together, form a substituted or unsusbtituted heterocyclic or heteroaryl moiety; [0071] xix) Compounds of subset xvii) above wherein -CC^NHCHCCOz^^CHaNCR^C^NR20)^0 has the following structure:

wherein R2C is hydrogen, CN, -C=NMe, =NO2, =NC(=O)NH2, =NS(O)2R, or =NS(O)2NRR'; wherein R and R' are each independently hydrogen or methyl; [0072] xx) Compounds of subset xvii) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following structure:

wherein R2C is hydrogen, CN, -C=NMe, =NO2, =NC(=O)NH2, =NS(O)2R, or =NS(O)2NRR'; wherein R and R' are each independently hydrogen or methyl; [0073] xxi) Compounds of subset xvii) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following structure:

[0074] xxii) Compounds of subset xvii) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following structure:
28

WO 2005/044817 PCT/US2004/036942
[0075] xxiii) Compounds of subsets xvii) and xviii) above wherein -C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the following structure:

or bioisosteres thereof;
wherein R2^ R2D, R2E and R2F are as defined in xvi) and xviii) above; [0076] xxiv) Compounds of subset xxiii) above wherein the bioiosteres have one of the following structures:

wherein R2C is lower alkyl;
[0077] xxv) Compounds of subset xxiii) above wherein R2D is, or R2E and R2F together with the nitrogen atom to which they are attached form, a moiety having one of the structures:
29

WO 2005/044817 PCT/US2004/036942

wherein s is an integer between 0 and 6; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, ~P(=O)(YRP5)2, an alkyl,
5 cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SOaNR02-, and RG1 and R^ are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is
10 independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O Rp5 may also be hydrogen; and each occurrence of RP2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two
15 adjacent occurrences of Rpl and R1*2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety;
[0078] xxvi) Compounds of subset xxv) above wherein R is, or R and R together with the nitrogen atom to which they are attached form, a moiety having one of the structures:
20
wherein each occurrence of Rpl is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3, or -NCCHaV,
[0079] xxvii) Compounds of subset xxvi) above wherein R2D is, or R2E and R2F together with the nitrogen atom to which they are attached form, a moiety having
25 one of the structures:
30

WO 2005/044817 PCT/US2004/036942

[0080] xxviii) R3 is -C(O)0R3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C(=O)NHC(R1)(R2)R3 is a moiety having the following structure:
wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; [0081] xxix) Compounds of subsets x)-xii), xiv)-xv) and xxviii); and compounds of subset xvi) wherein R2D is A12', wherein Ar2 is one of the following structures:

31

WO 2005/044817 PCT/US2004/036942

32

WO 2005/044817 PCT/US2004/036942

wherein each occurrence of s is an integer from 0-6; w is an integer from 1-6; Xj is CHRP1 or NRP2; X2 and X3 are independently CHRP1, NRM, CHSO2Rra or NSC>2RP3; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, -(aliphatic)aryl or -(aliphatic)heteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR°2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO2NRG2-, and RG1 and R02 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, -(aliphatic)aryl or -(aliphatic)heteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of Rps is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O RP5 may also be hydrogen; and each occurrence of RP2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two adjacent occurrences of RP1 and RP2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and each occurrence of R is independently alkyl, aryl, heteroaryl or -N(RP2)2.
33

WO 2005/044817 PCT/US2004/036942
[0082] xxx) Compounds of subset xxix) above wherein Ax% is one of the following structures:

wherein s, X1, X2 and X3 are as defined in xx) above; X5 is O, S or NR1"2; each occurrence of Rpl is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NR^C^O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O Rp5 may also be hydrogen; each occurrence of R1"2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two adjacent occurrences of RPI and Rp2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and each occurrence of RP3 is independently alkyl, aryl, heteroaryl or -N(R \[0083] xxxi) Compounds of subset xxx) above wherein each occurrence of RP1 is independently hydrogen, halogen, -P(=O)(YRP5)2, lower alkyl or heteroalkyl moiety, or is -GRG1 -wherein G is -O-, -S-, -NRG2- or -SO2-5 and RG1 and RG2 are independently hydrogen, lower alkyl or aryl; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently lower alkyl, or when Y is O RP5 may also be hydrogen; and each occurrence of R**2 is independently hydrogen, lower alkyl or a nitrogen protecting group; wherein any two adjacent occurrences of RP1 and R1"2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety;
34

WO 2005/044817 PCT/US2004/036942
[0084] xxxii) Compounds of subset xxx) above wherein Ar2 has one of the following structures:

wherein Xj is N or CR?1; s is an integer from 0-6; each occurrence of RP1 is independently hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=O)O-, -CKONR02-, -OC(=O>, -NR^CCO)- or -SOaNR02-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and RP3 is independently lower alkyl or aryl; [0085] xxxiii) Compounds of subsets xxix), xxx) and xxxii) wherein s is 0; [0086] xxxiv) Compounds of subsets xxix), xxx) and xxxii) wherein s is 1; [0087] xxxv) Compounds of subsets xxix), xxx) and xxxii) wherein s is 2; [0088] xxxvi) Compounds of subsets x) and xi) above wherein Ar2 is one of the following structures:

wherein s is an integer from 0-2; each occurrence of RPI is independently hydrogen, halogen, CN, isocyanate, NO2, -ORG1, -S RG1, -NRG1RG2-, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of Rps is independently lower alkyl, or when Y is O RP5 may also be hydrogen; each occurrence of RK is independently hydrogen, alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or
35

WO 2005/044817 PCTYUS2004/036942
heteroalkylheteroaryl moiety or a nitrogen protecting group; Rp3 is lower allcyl or -N(RP2)2; and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; [0089] xxxvii) Compounds of subsets x) and xi) above wherein Ar2 is one of the following structures:

[0090] xxxviii) Compounds of subsets x) and xi) above wherein A12 is one of the following structures:

wherein RP3 is lower alkyl; and Rp2 and RG1 are independently hydrogen or lower alkyl;
[0091] xxxix) Compounds of subsets x) and xi) above wherein A12 is one of the following structures:

wherein RP3 is lower alkyl and RG1 is hydrogen or lower alkyl; [0092] xl) Compounds of subsets x) and xi) above wherein R is hydrogen, hydroxyl or lower alkoxy and Ar2 is one of the following structures:

wherein Rp3 is lower alkyl; and RGI is hydrogen or lower alkyl; [0093] xli) Compounds of subsets xii) and xiii) wherein R1A is alkyl or -NR1BR1C; wherein RIB and R1C are independently hydrogen or lower alkyl; [0094] xlii) Compounds of subsets xii) and xiii) wherein R1A is -NH2 or a moiety having the structure:
36

WO 2005/044817 PCT/US2004/036942

wherein RP1 is independently hydrogen, hydroxyl, lower alkyl or lower heteroalkyl; and each occurrence of RK is independently hydrogen or lower alkyl; [0095] xliii) Compounds of subsets xii) and xiii) wherein R1A is -NH2 or a moiety having the structure:
wherein Rpl is hydrogen or lower alkyl;
[0096] xliv) Compounds of subsets xii) and xiii) wherein R1A is cycloalkyl, aryl, or a moiety having one of the structures:

wherein s is an integer between 0 and 6; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SOzNR02-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O RP5 may also be hydrogen; and each occurrence of RP2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two
37

O 2005/044817 PCT/US2004/036942
adjacent occurrences of R ] and RP2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety;
[0097] xiv) Compounds of subset xliv) wherein s is an integer between 0 and 2; each occurrence of Rpl is independently lower alkyl or is -GRG1 wherein G is -O-or -NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and each occurrence of RP2 is independently hydrogen, lower alkyl, aryl or heteroaryl; [0098] xlvi) Compounds of subsets xxi) and xxii) wherein R1A is a moiety having one of the structures:

wherein s is an integer between 0 and 2; X° is halogen; each occurrence of RP1 is independently hydrogen, hydroxyl, lower alkyl or lower heteroalkyl; G is -O-or -NRG2-, and RG1 and R02 are independently hydrogen or lower alkyl; and RP2 is independently hydrogen or lower alkyl;
[0099] xlvii) Compounds of subset xlvi) wherein R1A is a moiety having one of the structures:

wherein G is -O- or -NR02-, and RG1 and R*32 are independently hydrogen or lower alkyl;
38

WO 2005/044817 PCT/US2004/036942
[0100] xlviii) R3 is -C(=O)OR3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C(=O)NHC(R1)(R2)R3 is a moiety having the following structure:
wherein RP3 is alkyl, cycloalkyl, heterocyclic, aryl or heteroaryl moiety; [0101] xlix) compounds of subsets xiv)-xv) above wherein -NH(R2A)Ar2 has one of the following structures:
wherein Xi is N or CRP1; s is an integer from 0-5; and each occurrence of RP1 is independently hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and R1"3 is alkyl, heteroalkyl, aryl or heteroaryl; [0102] 1) compounds of subset xlix) above wherein s is 0; [0103] li) compounds of subset xlix) above wherein RP1 is hydrogen, halogen or lower alkyl;
[0104] lii) compounds of subset li) above wherein RP1 is hydrogen, chloro or methyl;
[0105] liii) compounds of subset xlix)above wherein R1"3 is lower alkyl; [0102] liv) compounds of subset liii) above wherein RP3 is methyl; [0103] lv) compounds of subset xlix) above wherein -NH(R2A)Ar2 has the following structure:
39

WO 2005/044817 PCT/US2004/036942

wherein RP1 is hydrogen, halogen or lower alkyl;
[0104] lvi) compounds of subset xlix) above wherein -NH(R2A)Ar2 has the following structure:
[0105] lvii) Compounds of subsets xvii) having the following structure:

or bioisosteres thereof;
wherein each occurrence of Rpl is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3 or-N(CH3)2; R^ is hydrogen, C^alkyl, C2-6aUcenyl, aryl, heteroaryl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; and q is 1 or 2; [0106] lviii) Compounds of subset lvii) above wherein the bioisosteres have one of the following structures:
40

WO 2005/044817 PCT/US2004/036942

wherein q is 1 or 2; and R2C is lower alkyl;
[0107] lix) Compounds of subset xxviii) wherein -C(=O)NHC(=CHAr2)CO2R3A has one of the following structures:

wherein RP3 is lower alkyl or aryl; Xi and X2 are independently N or CRP1; X3 is O, S or NR1*2; wherein RP1 is hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-5 -NRG2C(=O)- or -SOzNR02-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and RK is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, orheteroalkylheteroaryl moiety;
[0108] lx) Compounds of subset xxviii) wherein -C(=O)NHC(=CHAr2)CO2R3A has the following structure:
41

WO 2005/044817 PCT/US2004/036942

wherein Xi is N or CH;
[0109] Ixi) R3 is -C(=O)OR3A, wherein R3A is as defined in any one of subsets ii)-ix) above, and -C(=O)NHC(RI)(R2)R3 has the structure -C(=O)NHC(=C(Rs)Ar2)CO2R3A wherein R3Aand Rs, taken together, form a substituted or unsubstituted heterocyclic moiety;
[0110] lxii) Compounds of subset Ixi) wherein -C(=O)NHC(=C(Rs)Ar2)CO2R3A has one of the following structures:

wherein Ar2 is as defined in classes and subclasses herein; and Xi is O, S or NH;
[0111] lxiii) Compounds of subset Ixi) wherein -C(=O)NHC(=C(Rs)Ar2)CO2R3A has one of the following structures:
wherein Xi is O, S or NH; and X2 is N or CH;
[0112] lxiv) L is absent, -C(=O), -CH2C(=O)NH-, -CH2NH-C(=O)-, -O-CH2-C(=O)-, -CH2-CH2-C(=O)-, -CH=CH-C(=O)NH-CH2-, -CH(OH)-CH2-O-, -CH(OH)-CH2-N(CH3)-, -CH(OH)-CH2-CH2-, -CH2-CH2-CH(OH)-, -O-CH2-CH(OH)-, -O-CH2-CH(OH)-CH2-, -O-CH2-CH2-CH(OH)-, O-CH2-CH2-0-, -CH2-CH2-CH2-O-, -CH2-CH(OH)-CH2-O, -CH2-CH2-O-, -CH-(CH3)-NH-C(=O)-, -CH2-NH-SO2-, -NH-SO2-CH2-, -CH2-SO2-NH-, -SO2NH-CH2-, -C(=O)-NH-C(=O)-, -NH-C(=O)-NH-, -NH-C(=O)-"NH-CH2-, -CH2-NH-C(=O)-NH-, -C(=O)-NH-CH2-
42

WO 2005/044817 PCT/US2004/036942
C(=O)-NH, -NH-C(=O)-O-, -O-C(=O)-NH-; or a substituted or unsubstiruted Ci. 6alkylidene or C2-6alkenyIidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O)-, -CO2-, -C(=0)C(=O)-, -C(=O)NRU-, -OC(=O)-, -0C(=0)mP-, -NRUNRU-, -NR^NR^CCK))-, -NRUC(=O)-, -NRUCO2-, -NRUC(O)NRW-, -S(=O)-, -SO2-, -NRUSO2-, -SO2NR13-, -NR^SC^NR14-, -O-, -S-, or -NRU-; wherein each occurrence of Ru and Ru is independently hydrogen, alkyl, heteroalkyl, aryl, heteroaryl or acyl; [0113] lxv) L is absent, -C(=O), or a substituted or unsubstituted Ci_6alkylidene or C2-6alkenylidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O>, -CO2-, -C(=O)C(=O)-, -C^CONR13-, -OC(=O)-, -OC(=O)NRU-, -NRUNRM-, -NR°NRWC(=O)-, -NRUC(=O)-, -NRUCO2-, -NRUC(=O)NRW-, -S(=O>, -SO2-, -NRUSO2-, -SO2NR13-, -NR^SCbNR14-, -O-, -S-, or -NRU-; wherein each occurrence of Ru and R14 is independently hydrogen, alkyl, heteroalkyl, aryl, heteroaryl or acyl; [0114] lxvi) L is absent; [0115] Ixvii) L is -C(=O);
[0116] Ixviii) L is absent, -C(=O), -CH2C(=O)NH-, -CH2NH-C(=O)-, -O-CH2-C(=O)-, -CH2-CHa-C(=O>, -CH=CH-C(=O)NH-CH2-5 -CH(OH)-CH2-O-, -CH(OH)-CH2-N(CH3)-, -CH(OH)-CH2-CH2-, -CH2-CH2-CH(OH)-, -O-CH2-CH(OH)-, -O-CH2-CH(OH)-CH2-, -O-CH2-CH2-CH(OH)-, O-CH2-CH2-O-, -CH2-CH2-CH2-O-, -CH2-CH(OH)-CH2-O, -CH2-CH2-O-, -CH-(CH3)-NH-C(=O)-, -CH2-NH-SO2-, -NH-SO2-CH2-, -CH2-SO2-NH-, -SO2NH-CH2-, -C(=O)-NH-C(=O)-, -NH-C(=O)-NH-, -NH-C(=O)-NH-CH2-,' -CH2-NH-C(=O)-NH-, -C(=O)-NH-CH2-C(=O)-NH, -NH-C(=O)-O-, or -O-C(=O)-NH-;
[0117] lxix) L is -{CH2)q- wherein q is 1 -5;
[0118] lxx)Lis-CH2-;
[0119] lxxi)Lis-(CH2)3-;
[0120] lxxii) L is a moiety having the structure:

[0121] lxxii) AR1 is one of the following structures:
43

WO 2005/044817
PCT/US2004/036942

44

WO 2005/044817 PCT/US2004/036942

5
wherein each occurrence of r is an integer from 0-6; Xi, X2, X3 and X4 are each independently N or CRQI; AR3 is a heterocyclic, aryl or heteroaryl moiety; each occurrence of RQ1 is independently hydrogen, ORQ3, OCF3, SRQ3, halogen, CN, isocyanate, NO2, CF3, NRQ3QRQ4, -SO2RQ3, alkyl-NR^R^, alkyl-C(=O)-NRQ3RQ4, alkyl-C(=O)RQ3, or an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety, wherein each occurrence of RQ3 and RQ4 is independently hydrogen, a protecting group, or an aliphatic, heteroaliphatic, aryl or heteroaryl moiety; and RQ2 is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; [0122] lxxiv) AR1 is one of the following structures:

45

WO 2005/044817 PCT/US2004/036942

wherein each occurrence of r is an integer from 0-6; Xi, X2, X3 and X4 is independently N or CRQ1; X5 is O, S or NRQ2; AR3 is a heterocyclic, aryl or heteroaryl moiety; each occurrence of RQ1 is independently hydrogen, ORQ3, OCF3, SRQ3, halogen, CN, isocyanate, NO2, CF3, NR^V34, -SO2RQ3, alkyl-NR^R04, alkyl-C(=O)-NRQ3RQ4, alkyl-C(=O)RQ3, or an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety, wherein each occurrence of RQ3 and R^ is independently hydrogen, a protecting group, or an aliphatic, heteroaliphatic, aryl or heteroaryl moiety; and R^ is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; [0123] Ixxv) AR1 is one of the following structures:

wherein r is as defined above; Xi, X2, X3 and X4 is independently N or CH; X5 is CHRQ1 or NH; each occurrence of RQ1 is independently hydrogen, ORQ3, OCF3, SRQ3, halogen, CN, isocyanate, NO2, CF3, NR^*54, -SO2RQ3, alkyl-NR^R^, alkyl-C(=O)-NRQ3RQ4, alkyl-C(=O)RQ3, or an aliphatic, alicyclic,
46

WO 2005/044817 PCT/US2004/036942
heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety, wherein each occurrence of RQ3 and RQ4 is independently hydrogen, a protecting group, or an aliphatic, heteroaliphatic, aryl or heteroaryl moiety; and RQ2 is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; [0124] lxxvi) AR1 is one of the following structures:

wherein X° is F or Cl; X2 is N or CRQ1; X5 is CH, O, S or NH; RQ1 is hydrogen, methyl, -CF3, -OCH3,- OCF3 or halogen; [0125] Ixxvii) AR1 is one of the following structures:

[0126] Ixxviii) AR1 is one of the following structures:

[0127] lxxix) AR1-!,- is one of the following structures:
47


WO 2005/044817 PCT/US2004/036942
[0128] lxxx) ARJ-L- is one of the following structures:
[0129] lxxxi) R4, for each occurrence, is independently hydrogen, halogen, -CN,
-NO2, an alkyl, alkylenyl; alkynyl; cycloalkyl, heteroalkyl, heterocyclic, aryl,
heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -
NR02-, -CO-, -SO-, -SOr, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O> or -
SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, alkylenyl;
alkynyl; cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or
alkylheteroaryl moiety;
[0130] Ixxxii) R4, for each occurrence, is independently hydrogen, halogen, or
lower alkyl;
[0131] Ixxxiii) R4, for each occurrence, is independently hydrogen or chloro;
[0132] lxxxiv) n is 0;
[0133] [0134] lxxxvi) n is 2 and each occurrence of R4 is a halogen;
[0135] lxxxvii) n is 2 and each occurrence of R4 is Cl;
[0136] lxxxviii) p is 1;
[0137] lxxxix) p is 2; and/or
[0138] xc) Compounds of formula (II) wherein when -C(=O)NHC(RI)(R2)R3
48
hf\

WO 2005/044817 PCT/US2004/036942
wherein Y1 is N or CRQl; X1, X2, X3 and X4 are independently CRQ1; X5 is NRQ1, O or S; r is 0-3; and each occurrence of RQ1 is independently CN, NO2, halogen, CF3, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety or is GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -Co.6alkylS02-, -C0-6alkylSO2NRG2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)- or -NRG2C(=O)-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety.
[0139] It will be appreciated that for each of the classes and subclasses described above and herein, any one or more occurrences of aliphatic or heteroaliphatic may independently be substituted or unsubstituted, cyclic or acyclic, linear or branched and any one or more occurrences of aryl, heteroaryl, cycloaliphatic, cycloheteroaliphatic may be substituted or unsubstituted.
[0140] The reader will also appreciate that all possible combinations of the variables described in i)- through xc) above (e.g., Rl, R2, R3, R4, L, and AR1, among others) are considered part of the invention. Thus, the invention encompasses any and all compounds of formula I or II generated by taking any possible permutation of variables R1, R2, R3, R4, L, AR1, etc. and other variables/substituents (e.g., X1, X2, X3, X4, R1A, R2A, R2C, R2D, etc.) as further defined for R1, R2, R3, R4, L, AR1, etc. described in i)- through xc) above.
[0141] For example, an exemplary combination of variables described in i)-through xc) above includes those compounds of Formula I wherein:
R1 and R2 are each independently hydrogen, an amino acid side chain, -(CH2)mOH, -(CH2)maryl, -(CH2)mheteroaryl, wherein m is 0-6, -CH(R1A)(OR1B), -CH(R1A)(NHR1B), U-T-Q, or an aliphatic, alicyclic, heteroaliphatic or heteroalicyclic moiety optionally substituted with U-T-Q, wherein U is absent, -O-, -S(0)o-2-, -SO2N(R1A), -N(R1A)-, -N(R1A)C(=O)-, -N(R1A)C(=O)-O-, -N(R1A)C(=O)-N(R1B>, -N(R1A)-SO2-, -C(=O)-, -C(=O)-O-, -O-C(=O)-, aryl, heteroaryl, alkylaryl, alkylheteroaryl, -C(=O)-N(R1A)-, -O-C(=O)-N(R1A>, -C(=N-R1E)-, -C(=N-R1E)-O-, -C(=N-R1E)-N(R1A)-, -O-C(=N-R1E)-N(R1A)-, -N(R1A)C(=N-RIE>, -N(R1A)C(=N-R1E)-O-5 N(R1A)C(=N-R1E)-N(R1B)-5 -P(=O)(ORIA)-O-5 or -P(=O)(R1A)-O-; T is absent, an aliphatic, heteroaliphatic, aryl, heteroaryl, alkylaryl or alkylheteroaryl
49

WO 2005/044817 PCT/US2004/036942
moiety; and Q is hydrogen, halogen, cyano, isocyanate, -OR1B, -SR1B; -N(R1B)2, -NHC(=O)OR1B, -NHC(=O)N(R1B)2, -NHC(=O)RIB, -NHSO2RIB, -NHSO2N(R1B)2, -NHSO2NHC(=O)OR1B, -NHC(=O)NHSO2R1B, -C(=O)NHC(=O)ORm, -C(=O)NHC(=O)R1B, -C(=O)NHC(=O)N(R1B)2, -C(=O)NHSO2R1B, C(=O)NHSO2N(R1B)2, -C(=S)N(R1B)2, -SO2R1B -SO2-O-R1B, -SO2-N(R1B)2, -SO2-NHC(=O)ORIB, -SO2-NHC(=O)-N(R1B)2, -SO2-NHC(=O)R1B, -O-C(=O)N(RlB)2, -O-C(=O) R1B, -O-C(=O)NHC(=O)R1B, -O-C(=O)NH-SO2R1B, -O-SO2R1B, or an aliphatic heteroaliphatic, aryl or heteroaryl moiety, or wherein R1 and R2 taken
together are an alicyclic or heterocyclic moiety, or together are wherein
each occurrence of RIA and RIB is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, -CORIC, or -CONR1CR1D; wherein each occurrence of R1C and R1D is independently hydrogen, hydroxyl, or an aliphatic, heteroaliphatic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and R is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, -CN, -OR1C, -NRlcR1Dor-SO2Rlc;
R3 is -C(=O)OR3A, -C(=O)H, -CH2OR3A, -CH2O-C(=O)-alkyl, -C(=O)NH(R3A), - CH2X°; wherein each occurrence of R3A is independently hydrogen, a protecting group, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl moiety, or R3A, taken together with R1 or R2, forms a heterocyclic moiety; wherein X° is a halogen selected from F, Cl, Br or I;
R4, for each occurrence, is independently hydrogen, halogen, -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(O)0-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety;
AR1 is a monocyclic or polycyclic aryl, heteroaryl, alkylaryl, alkylheteroaryl, I alicyclic or heterocyclic moiety;
50

WO 2005/044817 PCT/US2004/036942
A, B, D and E are connected by either a single or double bond, as valency permits; wherein each occurrence of A, B, D and E is independently C=O, CR'R", NR1, CR1, N, 0, S, S(=O) or SO2; wherein each occurrence of R1 is independently hydrogen, halogen, -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=0)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO2NRG2-, and RG1 and R02 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or any two adjacent occurrences of R1, taken together, represent an alicyclic, heteroalicyclic, aryl, or heteroaryl moiety; and
L is absent or is V-W-X-Y-Z, wherein each occurrence of V, W, X, Y and Z is independently absent, C=O, NRL1, -O-, -C(RL1)=, =C(RL1)-, -CCR")^), C(=N-O-RL1), C(=N-RL1), -N=, S(O)0-2; a substituted or unsubstituted C^alkylidene or C2. 6alkenylidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O)-, -CO2-, -C(=O)C(=O)-, -C(=O)NRU-, -OC(=O)-, -OC(=O)NRU-, -NRUNRU-, -NRUNRWC(=O)-, -NRUC(=O)-, -NRUCO2-, -NRL3C(=O)NRM-, -S(=O>, -SO2-, -NRUSO2-, -SO2NRU-, -NRL3SO2NRL4-, -O-, -S-, or -NR°-; wherein each occurrence of Ru and Ru is independently hydrogen, alkyl, heteroalkyl, aryl, heteroaryl or acyl; or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and each occurrence of RL1 and Ru is independently hydrogen, hydroxyl, protected hydroxyl, amino, protected amino, thio, protected thio, halogen, cyano, isocyanate, carboxy, carboxyalkyl, formyl, formyloxy, azido, nitro, ureido, thioureido, thiocyanato, alkoxy, aryloxy, mercapto, sulfonamido, benzamido, tosyl, or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or wherein one or more occurrences of RL1 and Ru, taken together, or taken together with one of V, W, X, Y or Z form an alicyclic or heterocyclic moiety or form an aryl or heteroaryl moiety. [0142] Other exemplary combinations are illustrated by compounds of the following subgroups I through XIV:
51

WO 2005/044817 PCT/US2004/036942
[0143] I) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RB1, RB2 and RE are independently hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In yet other embodiments, one of RB1 and RB2 is hydrogen, the other is substituted or unsubstituted lower alkyl. hi certain exemplary embodiments, RB1 and RB2 are each hydrogen, hi certain other exemplary embodiments, RB1 and RB2 are each lower alkyl. In certain exemplary embodiments, RB1 and RB2 are each methyl.' hi other embodiments, RE is hydrogen, hi yet other embodiments, RE is substituted or unsubstituted lower alkyl. hi yet other embodiments, RE is substituted or unsubstituted methyl, ethyl, propyl, z-propyl, w-butyl, sec-butyl, ter/-butyl, w-pentyl, .sec-pentyl, fert-pentyl or n-hexyl. In certain embodiments, R4 and R4B are each Cl; and RB1 and RB2 are each hydrogen.
[0144] II) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl. hi certain embodiments, R4A and R4B are each Cl. hi other embodiments, RE is hydrogen, hi yet other embodiments, RB is substituted or unsubstituted lower alkyl. hi yet other embodiments, RE is substituted or unsubstituted methyl, ethyl, propyl, /-propyl, n-butyl, sec-butyl, fert-butyl, «-pentyl, jec-pentyl, ter/-pentyl or n-hexyl.
52

WO 2005/044817 PCT/US2004/036942
[0145] IH) Compounds having the structure (and pharmaceutically
acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In other embodiments, RE is hydrogen. In yet other embodiments, RE is substituted or unsubstituted lower alkyl. hi yet other embodiments, RE is substituted or unsubstituted methyl, ethyl, propyl, /-propyl, n-butyl, sec-butyl, tert-bntyl, K-pentyl, sec-ipentyl, tert-pentyl or n-hexyl.
[0146] FV) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In other embodiments, RE is hydrogen. In yet other embodiments, RE is substituted or unsubstituted lower alkyl. hi yet other embodiments, RE is substituted or unsubstituted methyl, ethyl, propyl, /-propyl, n-butyl, sec-b\xty\, tert-bntyl, 72-pentyl, ,yec-pentyl, fer/-pentyl or «-hexyl.
[0147] V) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):
53

WO 2005/044817

PCT/US2004/036942



10


wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA is hydrogen, lower alkyl or acyl; and RE is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In other embodiments, RE is hydrogen. In yet other embodiments, RE is substituted or unsubstituted lower alkyl. In yet other embodiments, RE is substituted or unsubstituted methyl, ethyl, propyl, z-propyl, n-butyl, sec-butyl, tert-bvtyl, «-pentyl, se c-pentyl, fert-pentyl or n-hexyl.
[0148] VI) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA1, R^, RB1 and RB2 are independently hydrogen or substituted or unsubstituted lower alkyl. hi certain embodiments, R4A and R4B are each Cl. In certain embodiments, RA1, R*2, RB1 And RB2 are each hydrogen.
[0149] VII) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA and RB are independently hydrogen or substituted or unsubstituted lower

54

WO 2005/044817 PCTAJS2004/036942
alkyl. In certain embodiments, R4A and R4B axe each Cl. In certain embodiments, RA and RB are each hydrogen.
[0150] VIII) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, RA is hydrogen.
[0151] IX)Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RB is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, RB is hydrogen.
[0152] X) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA is hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, RA is hydrogen.
55

WO 2005/044817 PCT/US2004/036942
[0153] XI)Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA, RB and RE are independently hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, RA and RB are each hydrogen. In certain other embodiments, RB is hydrogen. In yet other embodiments, RA, RB and RE are each hydrogen.
[0154] XII) Compounds having the structure (and pharmaceutically
acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA, RB and RE are independently hydrogen or substituted or unsubstituted lower alkyl. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, RA and RB are each hydrogen. In certain other embodiments, RE is hydrogen. In yet other embodiments, RA, RB and RE are each hydrogen.
[0155] XIII) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):
56

WO 2005/044817 PCT/US2004/036942

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and A and B are independently N or CH. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, A is N. In certain embodiments, A is CH. In certain embodiments, B is N. In certain embodiments, A is CH. In certain embodiments, A and B are each N. . In certain embodiments, A is CH. In certain embodiments, A and B are each CH.
[0156] XTV) Compounds having the structure (and pharmaceutically acceptable derivatives thereof):

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and A and B are independently N or CH. In certain embodiments, R4A and R4B are each Cl. In certain embodiments, A is N. In certain embodiments, A is CH. In certain embodiments, B is N. In certain embodiments, A is CH. Li certain embodiments, A and B are each N. In certain embodiments, A is CH. hi certain embodiments, A and B are each CH.
hi certain embodiments, for compounds of classes I-XIV above, AR!-L- is a moiety having one of the following structures:

and -C(=O)NHC(R1)(R2)R3 is a moiety having one of the following structures:

57
or bioisosteres thereof;

WO 2005/044817 PCT/US2004/036942
wherein R2A and R3A are as defined in classes and subclasses herein; and R2D is a moiety having one of the following structures:

wherein s is an integer between 0 and 6; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycioalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C^NR62-, -OC(=O>, -NRG2C(=O)- or -SO2NRG2-, and RG1 and R02 are independently hydrogen, an alkyl, cycioalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O RP5 may also be hydrogen; and each occurrence of R1"2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two adjacent occurrences of RP1 and R?2, taken together, may form a cycioalkyl, heterocyclic, aryl or heteroaryl moiety. In certain embodiments, R2A and R3A are each hydrogen. In certain embodiments, R2D is a moiety having one of the structures:

wherein each occurrence of RP1 is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3, or-N(CH3)2. In certain embodiments, R2D is a moiety having one of the structures:

58

WO 2005/044817 PCT/US2004/036942

[0157] It will also be appreciated that for each of the subgroups I-XIV described above, a variety of other subclasses are of special interest, including, but not limited to those classes described above i)-xc) and classes, subclasses and species of compounds described above and in the examples herein.
[0158] Some of the foregoing compounds can comprise one or more asymmetric centers, and thus can exist in various isomeric forms, e.g., stereoisomers and/or diastereomers. Thus, inventive compounds and pharmaceutical compositions thereof may be in the form of an individual enantiomer, diastereomer or geometric isomer, or may be in the form of a mixture of stereoisomers. In certain embodiments, the compounds of the invention are enantiopure compounds. In certain other embodiments, mixtures of stereoisomers or diastereomers are provided. [0159] Furthermore, certain compounds, as described herein may have one or more double bonds that can exist as either the Z or E isomer, unless otherwise indicated. The invention additionally encompasses the compounds as individual isomers substantially free of other isomers and alternatively, as mixtures of various isomers, e.g., racemic mixtures of stereoisomers. hi addition to the above-mentioned compounds per se, this invention also encompasses pharmaceutically acceptable derivatives of these compounds and compositions comprising one or
more compounds of the invention and one or more pharmaceutically acceptable
I excipients or additives.
[0160] Compounds of the invention may be prepared by crystallization of compound of formula (I) or (II) under different conditions and may exist as one or a combination of polymorphs of compound of general formula (I) or (II) forming part of this invention. For example, different polymorphs may be identified and/or prepared using different solvents, or different mixtures of solvents for recrystallization; by performing crystallizations at different temperatures; or by using various modes of cooling, ranging from very fast to very slow cooling during crystallizations. Polymorphs may also be obtained by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs may be
59

WO 2005/044817 PCT/US2004/036942
determined by solid probe NMR spectroscopy, IR spectroscopy, differential scanning calorimetry, powder X-ray diffractogram and/or other techniques. Thus, the present invention encompasses inventive compounds, their derivatives, their tautomeric forms, their stereoisomers, their polymorphs, their pharmaceuticaUy acceptable salts their pharmaceutically acceptable solvates and pharmaceutically acceptable compositions containing them.
[0161] 2) Pharmaceutical Compositions
[0162] As discussed above this invention provides novel compounds that have biological properties useful for the treatment of Mac-1 and LFA-1 mediated disorders.
[0163] Accordingly, in another aspect of the present invention, pharmaceutical compositions are provided, which comprise any one of the compounds described herein (or a prodrug, pharmaceutically acceptable salt or other pharmaceutically acceptable derivative thereof), and optionally comprise a pharmaceutically acceptable carrier. In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. Alternatively, a compound of this invention may be administered to a patient in need thereof in combination with the administration of one or more other therapeutic agents. For example, additional therapeutic agents for conjoint administration or inclusion in a pharmaceutical composition with a compound of this invention may be an approved anti-inflammatory agent, or it may be any one of a number of agents undergoing approval in the Food and Drug Administration that ultimately obtain approval for the treatment of any disorder mediated by Mac-1 or LFA-1. It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof.
[0164] As described above, the pharmaceutical compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to
60

WO 2005/044817 PCT/US2004/036942
the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other components) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatine; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil; corn oil and soybean oil; glycols; such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogenfree water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
[0165] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as
61

WO 2005/044817 PCT/US2004/036942
wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[0166] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic 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 water, Ringer's solution, U.S.P. 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 can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[0167] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[0168] In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension or crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle, Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include (poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
62

WO 2005/044817 PCT/US2004/036942
[0169] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[0170] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[0171] Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in
63

WO 2005/044817 PCT/US2004/036942
soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like. [0172] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose and starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. [0173] The present invention encompasses pharmaceutically acceptable topical formulations of inventive compounds. The term "pharmaceutically acceptable topical formulation", as used herein, means any formulation which is pharmaceutically acceptable for intradermal administration of a compound of the invention by application of the formulation to the epidermis. In certain embodiments of the invention, the topical formulation comprises a carrier system. Pharmaceutically effective carriers include, but are not limited to, solvents (e.g., alcohols, poly alcohols, water), creams, lotions, ointments, oils, plasters, liposomes, powders, emulsions, microemulsions, and buffered solutions (e.g., hypotonic or buffered saline) or any other carrier known in the art for topically administering Pharmaceuticals. A more complete listing of art-known carriers is provided by reference texts that are standard in the art, for example, Remington's Pharmaceutical Sciences, 16th Edition, 1980 and 17th Edition, 1985, both published by Mack Publishing Company, Easton, Pa., the disclosures of which are incorporated herein by reference in their entireties. In certain other embodiments, the topical formulations of the invention may comprise excipients. Any pharmaceutically
64

WO 2005/044817 PCT/US2004/036942
acceptable excipient known in the art may be used to prepare the inventive pharmaceutically acceptable topical formulations. Examples of excipients that can be included in the topical formulations of the invention include, but are not limited to, preservatives, antioxidants, moisturizers, emollients, buffering agents, solubilizing agents, other penetration agents, skin protectants, surfactants, and propellants, and/or additional therapeutic agents used in combination to the inventive compound. Suitable preservatives include, but are not limited to, alcohols, quaternary amines, organic acids, parabens, and phenols. Suitable antioxidants include, but are not limited to, ascorbic acid and its esters, sodium bisulfite, butylated hydroxytoluene, butylated hydroxyanisole, tocopherols, and chelating agents like EDTA and citric acid. Suitable moisturizers include, but are not limited to, glycerine, sorbitol, polyethylene glycols, urea, and propylene glycol. Suitable buffering agents for use with the invention include, but are not limited to, citric, hydrochloric, and lactic acid buffers. Suitable solubilizing agents include, but are not limited to, quaternary ammonium chlorides, cyclodextrins, benzyl benzoate, lecithin, and polysorbates. Suitable skin protectants that can be used in the topical formulations of the invention include, but are not limited to, vitamin E oil, allatoin, dimethicone, glycerin, petrolatum, and zinc oxide.
[0174] hi certain embodiments, the pharmaceutically acceptable topical formulations of the invention comprise at least a compound of the invention and a penetration enhancing agent. The choice of topical formulation will depend or several factors, including the condition to be treated, the physicochemical characteristics of the inventive compound and other excipients present, their stability in the formulation, available manufacturing equipment, and costs constraints. As used herein the term " penetration enhancing agent " means an agent capable of transporting a pharmacologically active compound through the stratum corneum and into the epidermis or dermis, preferably, with little or no systemic absorption. A wide variety of compounds have been evaluated as to their effectiveness in enhancing the rate of penetration of drugs through the skin. See, for example, Percutaneous Penetration Enhancers, Maibach H. 1. and Smith H. E. (eds.), CRC Press, Inc., Boca Raton, Fla. (1995), which surveys the use and testing of various skin penetration enhancers, and Buyuktimkin et ah, Chemical Means of Transdermal
65

WO 2005/044817 PCT/US2004/036942
Drag Permeation Enhancement in Transdermal and Topical Drag Delivery Systems, Gosh T. K., Pfister W. R., Yum S. I. (Eds.), Interpharm Press Inc., Buffalo Grove, 111. (1997). In certain exemplary embodiments, penetration agents for use with the invention include, but are not limited to, triglycerides (e.g., soybean oil), aloe compositions (e.g., aloe-vera gel), ethyl alcohol, isopropyl alcohol, octolyphenylpolyethylene glycol, oleic acid, polyethylene glycol 400, propylene glycol, N-decylmethylsulfoxide, fatty acid esters (e.g., isopropyl myristate, methyl laurate, glycerol monooleate, and propylene glycol monooleate) and N-methyl pyrrolidone.
[0175] In certain embodiments, the compositions may be in the form of ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. In certain exemplary embodiments, formulations of the compositions according to the invention are creams, which may further contain saturated or unsaturated fatty acids such as stearic acid, palmitic acid, oleic acid, palmito-oleic acid, cetyl or oleyl alcohols, stearic acid being particularly preferred. Creams of the invention may also contain a non-ionic surfactant, for example, polyoxy-40-stearate. In certain embodiments, the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms are made by dissolving or dispensing the compound in the proper medium. As discussed above, penetration enhancing agents can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[0176] It will also be appreciated that the compounds and pharmaceutical compositions of the present invention can be formulated and employed in combination therapies, that is, the compounds and pharmaceutical compositions can be formulated with or administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular
66

WO 2005/044817 PCT/US2004/036942
combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder (for example, an inventive compound may be administered concurrently with another anti-inflammatory agent), or they may achieve different effects (e.g., control of any adverse effects).
[0177] In certain embodiments, the pharmaceutical compositions of the present invention further comprise one or more additional therapeutically active ingredients (e.g., anti-inflammatory and/or palliative). For purposes of the invention, the term "Palliative" refers to treatment that is focused on the relief of symptoms of a disease and/or side effects of a therapeutic regimen, but is not curative. For example, palliative treatment encompasses painkillers, antinausea medications and anti-sickness drugs.
[0178] 3) Research Uses, Pharmaceutical Uses and Methods of Treatment
[0179] Research Uses
[0180] According to the present invention, the inventive compounds may be
assayed in any of the available assays known in the art for identifying compounds
having the ability to modulate adhesion between intracellular adhesion molecules
and the leukocyte integrin family of receptors; to antagonize CD11/CD18 receptors
associated with leukocytes and/or to antagonize Mac-1 and/or LFA-1. For example,
the assay may be cellular or non-cellular, in vivo or in vitro, high- or low-throughput
format, etc.
[0181] Thus, in one aspect, compounds of this invention which are of particular
interest include those which:
• modulate adhesion between intracellular adhesion molecules (e.g., ICAM-1, -2
and -3) and the leukocyte integrin family of receptors;
• exhibit the ability to antagonize CD 11 /CD 18 receptors associated with
leukocytes;
• exhibit the ability to antagonize Mac-1 and/or LFA-1; and
• are useful for the treatment of LFA-1 mediated disorders.
67

WO 2005/044817 PCT/US2004/036942
[0182] As detailed in the exemplification herein, in assays to determine the ability of compounds to modulate T-cell adhesion to 5dICAM-Ig {e.g., cell attachment assay), certain inventive compounds exhibited IC50 values [0184] As discussed above, certain of the compounds as described herein exhibit activity generally as modulators of adhesion between intracellular adhesion molecules. More specifically, compounds of the invention demonstrate the ability to antagonize CD11/CD18 receptors associated with leukocytes and in certain embodiments exhibit the ability to antagonize LFA-1 interactions. Thus, in certain embodiments, compounds of the invention are useful for the treatment of LFA-1 mediated disorders.
[0185] Accordingly, in another aspect of the invention, methods for treating (or preventing) of LFA-1 mediated disorders are provided comprising administering a
68

WO 2005/044817 PCT/US2004/036942
therapeutically effective amount of a compound of formula (I) or (II) as described herein, to a subject in need thereof. In certain embodiments, a method for the treatment of LFA-1 mediated disorders is provided comprising administering a therapeutically effective amount of an inventive compound, or a pharmaceutical composition, comprising an inventive compound to a subject in need thereof, in such amounts and for such time as is necessary to achieve the desired result. [0186] In certain embodiments, the method involves the administration of a therapeutically effective amount of the compound or a pharmaceutically acceptable derivative thereof to a subject (including, but not limited to a human or animal) in need of it.
[0187] As discussed above this invention provides novel compounds that have biological properties useful for the treatment of Mac-1 and/or LFA-1 mediated disorders, hi certain embodiments, the inventive compounds as useful for the treatment of psoriasis, responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), dermatitis, meningitis, encephalitis, uveitis, allergic conditions such as eczema and asthma, conditions involving infiltration of T-cells and chronic inflammatory responses, skin hypersensitivity reactions (including poison ivy and poison oak), artherosclerosis, autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus (SLE), diabetes mellitus, multiple sclerosis, Reynaud's syndrome, autoimmune thyroiditis, experimental autoimmune encephalomyelitis, Sjorgen's syndrome, juvenile onset diabetes and immune responses associated with delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis, pernicious anemia, diseases involving leukocyte diapedeses, CNS inflammatory disorder, multiple organ injury syndrome secondary to septicaemia or trauma, autoimune hemolytic anemia, myasthemia gravis, antigen-antibody complex mediated diseases, all types of transplantations, including graft versus host or host versus graft disease, HIV and rhinovirus infection, and pulmonary fibrosis to name a few.
[0188] As described in more detail herein, in general, compounds of the invention are useful as antagonists of the interaction between intracellular adhesion
69

WO 2005/044817 PCT/US2004/036942
molecules {e.g., ICAM-1, 2 or 3) and the leukocyte integrin family of receptors. Thus, in certain embodiments, the present invention provides compounds useful for the treatment of disorders mediated by the CD11/CD18 family of cellular adhesion molecules. In certain embodiments of special interest, the present invention provides compounds useful for the treatment of disorders mediated by Mac-1 and/or LFA-1. For example, compounds of the invention are particularly useful for the treatment inflammatory disorders, organ graft rejection and autoimmune disorders, to name a few.
[0189] Thus, as described above, in another aspect of the invention, a method for the treatment of disorders mediated by the CD11/CD18 family of cellular adhesion molecules is provided comprising administering a therapeutically effective amount of a compound of formula (I) or (II) as described herein, to a subject in need thereof. In certain embodiments of special interest the inventive method is used for the treatment of disorders mediated by Mac-1 or LFA-1. It will be appreciated that the compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for the treatment of disorders mediated by the CD11/CD18 family of cellular adhesion molecules. For example, in certain exemplary embodiments, compounds of the invention are useful as antagonists of the interaction between Mac-1 or LFA-1 and intracellular adhesion molecules (e.g., ICAM-1) and thus the compounds are useful for the treatment of LFA-1 mediated disorders including, but not limited to, psoriasis, responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), dermatitis, meningitis, encephalitis, uveitis, allergic conditions such as eczema and asthma, conditions involving infiltration of T-cells and chronic inflammatory responses, skin hypersensitivity reactions (including poison ivy and poison oak), atherosclerosis, autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus (SLE), diabetes mellitus, multiple sclerosis, Reynaud's syndrome, autoimmune thyroiditis, experimental autoimmune encephalomyelitis, Sjorgen's syndrome, juvenile onset diabetes, and immune responses associated with delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis, pernicious anemia, diseases involving leukocyte
70

WO 2005/044817 PCT/US2004/036942
diapedesis, CNS inflammatory (disorder, multiple organ injury syndrome secondary to septicaemia or trauma, autoimmune hemolytic anemia, myasthemia gravis, antigen-antibody complex mediated diseases, all types of transplantations, including graft versus host or host versus graft disease, HIV and rhinovirus infection, pulmonary fibrosis and the like, to name a few. Thus, the expression "effective amount" as used herein, refers to a sufficient amount of agent to antagonize the interaction between intracellular adhesion molecules (e.g., ICAM) and the leukocyte integrin family of receptors, and to exhibit a therapeutic effect. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular therapeutic agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of therapeutic agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts (see, for example, Goodman and Gilman's, "The Pharmacological Basis of Therapeutics", Tenth Edition, A. Gilman, J.Hardman and L. Limbird, eds., McGraw-Hill Press, 155-173, 2001, which is incorporated herein by reference in its entirety).
[0190] Another aspect of the invention relates to a method for inhibiting the interaction between LFA-1 and ICAM-1 in a biological sample or a patient, which method comprises administering to the patient, or contacting said biological sample with a compound of formula I or II or a composition comprising said compound.
71

WO 2005/044817 PCT/US2004/036942
[0191] Another aspect of the invention relates to a method for inhibiting the CDlla and/or CD18 interaction with ICAM-1, ICAM-2 or ICAM-3 in a biological sample or a patient, which method comprises administering to the patient, or contacting said biological sample with a compound of formula I or II or a composition comprising said compound.
[0192] Furthermore, after formulation with an appropriate pharmaceutically acceptable carrier in a desired dosage, the pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention'may ,be administered at dosage levels of about 0.001 mg/kg to about 50 mg/kg, from about 0.01 mg/kg to about 25 mg/kg, or from about 0.1 mg/kg to about 10 mg/kg of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. It will also be appreciated that dosages smaller than 0.001 mg/kg or greater than 50 mg/kg (for example 50-100 mg/kg) can be administered to a subject. In certain embodiments, compounds are administered orally or parenterally.
TREATMENT KIT
[0193] In other embodiments, the present invention relates to a kit for conveniently and effectively carrying out the methods in accordance with the present invention, hi general, the pharmaceutical pack or kit comprises one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Such kits are especially suited for the delivery of solid oral forms such as tablets or capsules. Such a kit preferably includes a number of unit dosages, and may also include a card having the dosages oriented in the order of their intended use. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered. Alternatively, placebo dosages, or calcium dietary supplements, either in a form similar to or distinct from the dosages of the pharmaceutical compositions, can be
72

WO 2005/044817 PCT/US2004/036942
included to provide a kit in which a dosage is taken every day. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
EQUIVALENTS
[0194] The representative examples that follow are intended to help illustrate the invention, and are not intended to, nor should they be construed to, limit the scope of the invention. Indeed, various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including the examples which follow and the references to the scientific and patent literature cited herein. It should further be appreciated that the contents of those cited references are incorporated herein by reference to help illustrate the state of the art. [0195] The following examples contain important additional information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and the equivalents thereof.
EXEMPLIFICATION
[0196] The compounds of this invention and their preparation can be understood further by the examples that illustrate some of the processes by which these compounds are prepared or used. It will be appreciated, however, that these examples do not limit the invention. Variations of the invention, now known or further developed, are considered to fall within the scope of the present invention as described herein and as hereinafter claimed. [0197] 1) General Description of Synthetic Methods:
[0198] The practitioner has a a well-established literature of macrolide chemistry to draw upon, in combination with the information contained herein, for guidance on synthetic strategies, protecting groups, and other materials and methods useful for the synthesis of the compounds of this invention.
73

WO 2005/044817 PCT/US2004/036942
[0199] The various references cited herein provide helpful background information on preparing compounds similar to the inventive compounds described herein or relevant intermediates, as well as information on formulation, uses, and administration of such compounds which may be of interest. [0200] Moreover, the practitioner is directed to the specific guidance and examples provided in this document relating to various exemplary compounds and intermediates thereof.
[0201] The compounds of this invention and their preparation can be understood further by the examples that illustrate some of the processes by which these compounds are prepared or used. It will be appreciated, however, that these examples do not limit the invention. Variations of the invention, now known or further developed, are considered to fall within the scope of the present invention as described herein and as hereinafter claimed.
[0202] According to the present invention, any available techniques can be used to make or prepare the inventive compounds or compositions including them. For example, a variety of solution phase synthetic methods such as those discussed in detail below may be used. Alternatively or additionally, the inventive compounds may be prepared using any of a variety combinatorial techniques, parallel synthesis and/or solid phase synthetic methods known in the art.
[0203] It will be appreciated as described below, that a variety of inventive compounds can be synthesized according to the methods described herein. The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Company (Milwaukee, WI), Bachem (Torrance, CA), Sigma (St. Louis, MO), or are prepared by methods well known to a person of ordinary skill in the art following procedures described in such references as Fieser and Fieser 1991, "Reagents for Organic Synthesis", vols 1-17, John Wiley and Sons, New York, NY, 1991; Rodd 1989 "Chemistry of Carbon Compounds", vols. 1-5 and supps, Elsevier Science Publishers, 1989; "Organic Reactions", vols 1-40, John Wiley and Sons, New York, NY, 1991; March 2001, "Advanced Organic Chemistry", 5th ed. John Wiley and Sons, New York, NY; and Larock 1990, "Comprehensive Organic Transformations: A Guide to Functional Group Preparations", 2nd ed. VCH Publishers. These schemes
74

WO 2005/044817 PCT/US2004/036942
are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to a person of ordinary skill in the art having regard to this disclosure.
[0204] The starting materials, intermediates, and compounds of this invention may be isolated and purified using conventional techniques, including filtration, distillation, crystallization, chromatography, and the like. They may be characterized using conventional methods, including physical constants and spectral data.
[0205] General Reaction Procedures:
[0206] Unless mentioned specifically, reaction mixtures were stirred using a magnetically driven stirrer bar. An inert atmosphere refers to either dry argon or dry nitrogen. Reactions were monitored either by thin layer chromatography, by proton nuclear magnetic resonance (NMR) or by high-pressure liquid chromatography (HPLC), of a suitably worked up sample of the reaction mixture. [0207] General Work Up Procedures:
[0208] Unless mentioned specifically, reaction mixtures were cooled to room temperature or below then quenched, when necessary, with either water or a saturated aqueous solution of ammonium chloride. Desired products were extracted by partitioning between water and a suitable water-immiscible solvent (e.g. ethyl acetate, dichloromethane, diethyl ether). The desired product containing extracts were washed appropriately with water followed by a saturated solution of brine. On occasions where the product containing extract was deemed to contain residual oxidants, the extract was washed with a 10% solution of sodium sulphite in saturated aqueous sodium bicarbonate solution, prior to the aforementioned washing procedure. On occasions where the product containing extract was deemed to contain residual acids, the extract was washed with saturated aqueous sodium bicarbonate solution, prior to the aforementioned washing procedure (except in those cases where the desired product itself had acidic character). On occasions where the product containing extract was deemed to contain residual bases, the extract was washed with 10% aqueous citric acid solution, prior to the aforementioned washing procedure (except in those cases where the desired product itself had basic
75

WO 2005/044817 PCT/US2004/036942
character). Post washing, the desired product containing extracts were dried over
anhydrous magnesium sulphate, and then filtered. The crude products were then
isolated by removal of solvent(s) by rotary evaporation under reduced pressure, at an
appropriate temperature (generally less than 45°C).
[0209] General Purification Procedures:
[0210] Unless mentioned specifically, chromatographic purification refers to
i
flash column chromatography on silica, using a single solvent or mixed solvent as eluent. Suitably purified desired product containing elutes were combined and concentrated under reduced pressure at an appropriate temperature (generally less than 45°C) to constant mass. Final compounds were dissolved in 50% aqueous acetonitrile, filtered and transferred to vials, then freeze-dried under high vacuum before submission for biological testing.
[0211] 1) Synthesis of Exemplary Compounds:
[0212] Unless otherwise indicated, starting materials are either commercially
available or readily accessibly through laboratory synthesis by anyone reasonably
familiar with the art. Described generally below, are procedures and general
guidance for the synthesis of compounds as described generally and in subclasses
and species herein. In addition, synthetic guidance can be found in published PCT
applications WO 99/49856 and WO 02/059114, the entire contents of which are
hereby incorporated by reference.
[0213] EXAMPLE 1
[0214] This example describes the synthesis of

which was prepared according to Scheme 1A and the procedure below.
76

WO 2005/044817 PCT/US2004/036942
[0215] SCHEME 1A

[0216] a) A solution of 3-methoxyphenylethylamine (0.2 mol) and formaldehyde (0.22 mol) in aqueous HC1 (20%, 500 mL) was heated at 80 °C for 4 hours. The reaction was then concentrated to dryness, and the residue was dissolved in hydrobromic acid (40% aqueous, 500 mL), and refluxed for 24 hours. The reaction was concentrated to give a brownish solid, which was used without purification. To the residue was added water (200 mL) and tetrahydrofuran ("THF") (300 mL), and to the resulting mixture was very carefully added with sodium carbonate (solid, 0.5 mol), followed by di-tert-butyl dicarbonate (0.3 mol). After 15 hours at room temperature, the reaction was extracted with ethyl acetate (1 L), and the organic extract was washed with saturated potassium dihydrophosphate and brine, dried over anhydrous magnesium sulfate and filtered.
[0217] The residue after concentration of the filtrate was dissolved in dichloromethane ("DCM"; 100 mL), and to it was slowly added acetic acid (500 mL) and surfuryl chloride (0.6 mol). After the reaction mixture was stirred at room temperature for 24 hours, the reaction was concentrated to dryness, and further dried under high vacuum for 2 hours. The crude product was used without further purification for next step. The crude product was dissolved in water/THF (200 mL/400 mL), and to it was added carefully and slowly sodium carbonate (0.5 mol) with good stirring, followed by di-tert-butyl dicarbonate (0.3 mol). After the
77

WO 2005/044817 PCT/US2004/036942
reaction mixture was stirred for 12 hours, the reaction was carefully neutralized with phosphoric acid (2 M) to pH about 7. The resulting mixture was extracted with ethyl acetate (500 mL x2), and the combined extracts were washed with water and brine, dried over anhydrous magnesium sulfate, filtered and concentrated. The crude solid was recrystalized from ethyl acetate and hexane (about 1:2 ratio) to yield a white solid. The mother liquid was concentrated and purified by column, eluting with 0-10% ethyl acetate in 4:1 hexane:methylene chloride. The combined yield is 14.5 g (23% from commercial 3-memoxyphenemylamine). MS (API-ES*) m/z: 262, 264,266 (M+H-terf-butyl*).
[0218] The product obtained above was dissolved in DCM (100 mL) and pyridine (50 mL). The resulting solution was cooled to -40 °C, and to it was added triflic anhydride (51 mmol) slowly. After the reaction mixture was gradually warmed to room temperature over 4 hours, the reaction mixture was partitioned between ethyl acetate (500 mL) and water (100 mL), and the organic layer was washed with water (100 mL, twice) and brine (50 mL), dried over anhydrous magnesium sulfate, filtered and concentrated. The residue was purified by column, eluting with 0-5% ethyl acetate in 5:1 hexane :DCM to give the corresponding triflate (9.73 g, 48% yield).
[0219] A mixture of lOmmol of the triflate, l.Ommol of 1,3-diphenylphosinepropane ("dppp") and 40mmol of di-isopropylethylamirie ("DIEA") in lOOmL of dry dimethylformamide ("DMF") and 50mL of anhydrous CH3OH was flushed with CO for 15 min, and then l.Ommol of Pd(OAc)2 was added under the atmosphere of CO. Subsequently, the resulting mixture was stirred at 70 °C overnight under an atmosphere of CO. The solvent was removed and the residue was purified by column chromatography using EtOAc/hexane = lA (v/v) as the eluent to give, compound 1.1 with a 56% yield. ESI-MS (m/z): (M^+Na 382.1; ]H NMR (CD3OD, 400MHz): 6 7.32 (s, 1H), 4.60 (s, 2H), 3.95 (s, 3H), 3.69 (m, 2H), 2.84 (m, 2H), 1.50 (s, 9H) ppm.
[0220] b) A mixture of 1.1 (5 mmol) and 30mmol of Lil in 20mL of pyridine was reflux overnight. The solvent was removed and the residue was dissolved in EtOAc. The resulting solution was then washed with saturated aqueous NH4CI and dried with anhydrous Na2SO4. The solvent was removed and the residue was dried
78

WO 2005/044817 PCT/US2004/036942
in vacuo to give a quantitative yield of compound 1.3. The crude product was carried on the next step without further purification. ESI-MS (m/z): (M - flBu +1), 290.
[0221] c) A solution of Boc-Dap-OH (10 mmol) in methanol (30 mL) was treated with trimethylsilyldiazomethane until the color remained light yellow for 10 seconds. The mixture was concentrated, and the residue was dissolved in DCM (30 mL). To the solution was added triethylamine (20 mmotyand followed by 3-thienylcarboxyl chloride (11 mmol). After 0.5 hour at room temperature, the reaction was filtered through silica gel, and concentrated. The residue was purified by column with 10-50% ethyl acetate in hexane. The product obtained this way was dissolved in DCM (10 mL) and treated with HC1 (4 M in dioxane, 10 mL). After 5 hours, the reaction was concentrated to give the title compound (60-80% overall yield).
[0222] d) A mixture of 1.2 (4 mmol), 1.3 (4.4 mmol), 5.0 mmol of o-(7-azabenzotriazol-l-yl)-iV,iV;iV"'iV-tetramethyluronium fluorophosphate ("HATU") and 20mmol of Et3N in 20mL of DMF was stirred at room temperature overnight. The solvent was removed and the residue was purified by column chromatography using CH2Cl2/Et0Ac = 6/4 (v/v) as eluent to give compound 1.4 with a 60% yield. ESI-MS (m/z): (M+l) 556.1.
[0223] e) A solution of 2mmol of 1.4 in 9mL of TFA and 3mL of CH2C12 was stirred at room temperature for 6 hours. The solvent was then removed and the residue was diluted with saturated aqueous NaHCC>3. The mixture was extracted with EtOAc for 3 times. The extracts were then dried with anhydrous Na2SC>4. The solvent was removed and the residue was dried in vacuo to give compound 1.5 which was used without further purification. ESI-MS (m/z): (M+l) 456.1. [0224] f) Intermediate compound 1.11 was prepared according to Scheme IB and the procedure below. [0225] SCHEME IB
79

WO 2005/044817 PCT/US2004/036942

[0226] To a solution of 100 mmol of commercially available 6-hydroxy-[2H]-benzofuran-3-one (compound 1.8) and 150 mmol of imidazole in 300 mL of dry DMF was added 110 mmol of tert-butyldimethylsilylchloride ("TBDMSC1") at room temperature, the resulting mixture was stirred at room temperature overnight. The solvent was removed, and the residue was diluted with 100 mL of EtOAc, washed with saturated aqueous NH4CL and dried with anhydrous NaaSO,*. The solvent was removed, and the residue was purified to give corresponding intermediate in 70% yield. ESI-MS (m/z): (M+H4) 265.1.
[0227] The intermediate is dissolved in 100 mL of CH3OH was added 20 mmol of NaBHt at room temperature. After stirring at room temperature for 12 hours, the reaction mixture was treated with 10 mL of acetone. Subsequently, 60 mL of 4.0 N HC1 were added to the mixture, and the mixture was stirred at room temperature overnight. The organic solvent was removed, and the residue was extracted with EtOAc for several times. The extract was then washed with brine and dried with anhydrous Na2SC>4. The solvent was removed and the residue was dissolved in 100 mmol of Et3N and 180 mL of dry CH2C12 was added 66 mmol of PhNTf2 at 0°C, the resulting mixture was stirred at room temperature overnight. The solvent was removed, and the residue was purified to give compound 1.9 in 90% yield. *H NMR (400 MHz, CD3CI): 8 7.75 (d, J=l.9 Hz, 1H), 7.66 (d, J=8.5 Hz, 1H), 7.50 (s, 1H), 7.21 (d, J=8.5 Hz, 1H), 6.85 (d, J=l.9 Hz, 1H) ppm.
[0228] A mixture of 50 mmol of compound 1.9, 2.5 mmol of dppp (diphenylphosphine-l,3-propane) and 2.5 mmol of Pd(OAc)2 in 100 mmol of DIEA, 125 mL of dry DMF, and 125 mL of anhydrous MeOH was stirred at 65°C under an atmosphere of CO overnight. The solvent was removed and the residue was purified by column chromatography to give compound 1.10 in 65% yield. lH NMR (400 MHz, CDCI3): 5 8.23 (s, 1H), 7.99 (d, /= 8.3 Hz, 1H), 7.78 (s, 1H), 7.65 (d, J= 8.3 Hz, 1H), 6.85 (s, 1H), 3.97 (s, 3H) ppm; ESI-MS (m/z): (M+l) 177.10. [0229] A mixture of 20 mmol of compound 1.10 and 80 mmol of LiOHeH2O in I 60 mL of THF and 15 mL of H2O was stirred at room temperature for 1 hour,
80

WO 2005/044817 PCT/US2004/036942
followed by adding 80 mL of 1 .ON aq. HC1. The organic solvent was removed and the residue was diluted with 50 mL of brine. The mixture was then extracted with EtOAc, and the extract was dried with anyhydrous Na2SO4. The solvent was removed and the residue was dried in vacuo to give a quantitative yield of compound 1.11. !H NMR (400 MHz, CD3OD): 8 8.14 (s, 1H), 7.92 (m,2H), 7.67 (d, J=8.5 Hz, 1H), 6.92 (s, 1H) ppm; ESI-MS (m/z): (M+H4) 163.1. [0230] g) A mixture of 0.25 mmol of compound 1.11 and 0.26 mmol of HATU in 1 mmol of DIEA and 2 mL of DMF was stirred at room temperature for 30 min, followed by adding a solution of 0.22 mmol of compound 1.5 in 1 mL of DMF. The resulting mixture was stirred 45°C for 12 hours. The solvent was removed, and the residue was purified to give compound 1.6 in 50-65% yield. Subsequently, compound 1.6 was hydrolyzed with LiOH (1.0 M aqueous, 0.5 mL) in THF (3 mL) for 2 hours. The rexaction mixture was then acidified with HC1 (aqueous), extracted with ethyl acetate (50 mL), dried over anhydrous magnesium sulfate and concentrated to give compound 1 in quantitative yield. *H NMR (400 MHz, CD3OD): 5 7.91 (s, 1H), 7.75 (d, J= 8.0 Hz, IE), 7.67 (s, 3H), 7.36 (d, J= 8.0 HZ, 1H), 7.13 (s, 1H), 6.96 (s, 1H), 5.01 (t, J= 6.8 Hz, 1H), 4.68 and 4.89 (m, 2H), 3.85 (d, J= 6.8 Hz, 2H), 3.70 and 4.02 (m, 2H), 2.93 (m, 2H) ppm; ESI-MS (m/z): (M+l) 586.10.
[0231] EXAMPLE 2 [0232] This example describes the synthesis of

which was prepared according to the procedure of Example lg except that 4-chlorobenzoic acid was used instead of compound 1.11. *H NMR (400 MHz, CD3OD): 5 7.64 (m, 2H), 7.35-7.49 (m, 5H), 7.11 (s, 1H), 4.98 (t, J= 8.0 Hz, 1H), 4.63 and 4.88 (m, 2H), 3.83 (d, J= 8.0 Hz, 2H), 3.68 and 3.98 (m, 2H), 2.89 (m, 2H) ppm; ESI-MS (m/z): (M+l) 579.90. [0233] EXAMPLE 3
81

WO 2005/044817 PCT/US2004/036942
[0234] This example describes the synthesis of

which was prepared according to Scheme 2 and the procedure below. [0235] SCHEME 2

[0236] a) To a solution of 10 mmol of commercially available 3.1 in 20 mL of MeOH and 20 mL of CH2C12 was slowly added 20 mmol of 2.0M TMSCHN2 in hexanes at 0 °C, and the resulting mixture was stirred at room temperature for 30 minutes. The solvent was removed and the residue was dried in vacuo to give crude 3.2.
[0237] b) Compound 3.2 was then stirred with 15 mmol of isopropylazide in the presence of 0.2 mmol of Cul, 0.2 mmolf of Et3N in 50 mL of CH3CN at room temperature overnight. The solvent was removed and the residue was purified by column chromatography to give compound 3.3 in 55% yield. ESI-MS (m/z): (M+l) 313.20.
82

WO 2005/044817 PCT/US2004/036942
[0238] c) A mixture of 2 mmol of compound 3.3 in 10 mL of 4.0 N HC1 in
dioxane was stirred at room for 12 hours. The solvent was removed and the residue
was dried in vacuo to give compound 3.4 in quantitative yield. ESI-MS (m/z):
(M+l) 213.10.
[0239] d) A solution of 1.1 (3.60 g, 10 mmol) in DCM (20 mL) was treated
with HC1 in 1,4-dioxane (4.0 M, 10 mL) at room temperature. After 2 hours, the
reaction was concentrated to give compound 3.5 in quantitative yield.
[0240] e) Example 3.5 (10 mmol) was mixed with EDC (2.11 g, 11 mmol),
A^iV-dimethylaminopyridine ("DMAP", 0.1 g), triethylamine (2.02 g) and Example
1.11 (1.62g, 10 mmol) in anhydrous DMF (50 mL). After 15 hours at room
temperature, the reaction mixture was diluted with ethyl acetate (200 mL), washed
with water (30 mL, 3 times), dried with anhydrous magnesium sulfate and filtered.
The residue after concentration of the filtrate was purified by column eluting with
10-30% ethyl acetate in hexane to give the title compound (3.7 g, 92%): ESI-MS
(m/z): (M+l) 213.1.
[0241] f) Compound 3.7 was made according to Example lb except that
compound 3.6 was used instead of compound 1.1.
[0242] g) A mixture of 0.25 mmol of compound 3.7 and 0.26 mmol of HATU
in 1 mmol of DIEA and 2 mL of DMF was stirred at room temperature for 30
minutes, followed by adding a solution of 0.22 mmol of compound 3.4 in 1 mL of
DMF. The resulting mixture was stirred 45°C for 4 hours. The solvent was
removed, and the residue was purified to give intermediate ester, which was
subsequently treated with LiOH in THF and water to give the desired compound 3 in
quantitative yield. JH NMR (400 MHz, CD3OD) 5 7.90 (s, 2H), 7.74 (m, 1H),'7.64
(s, 1H), 7.34 (m, 1H), 6.93 (s, 1 H), 5.03 (m, 1H), 4.82 (m, 1 H), 4.65 and 4.88 (m,
2H), 3.72 and 3.97 (m, 2H), 3.40 (m, 1H), 3.18 (m, 1H), 2.90 (m, 2H), 1.55 (m, 6H)
ppm; ESI-MS (m/z): (M+l) 570.1.
[0243] EXAMPLE 4
[0244] This example describes the synthesis of
83

WO 2005/044817 PCT/US2004/036942

which was prepared according to Scheme 3 and the procedure below. [0245] SCHEME 3

[0246] a) Boc-DAP-OH (0.2 g, 1.0 mmol), 2,4-dichloropyrimidine (0.29g, 2.0 mmol), and diisopropylethylamine (0.51 mL, 2.9 mmol) in ethanol (5 mL) were heated to 75 °C for 14 hours. The reaction mixture was cooled to room temperature and the solvent removed under reduced pressure. The resulting crude product 4.1 was pure enough to carry forward to the next chemical transformation. [0247] b) Crude residue 4.1 (0.31 g, 1.0 mmol) was dissolved in 9:1 benzene: methanol (5 mL). Trimethylsilyldiazomethane (1.0 mL, 2.0M in hexanes) was added slowly to the stirring reaction mixture and stirred for an additional 1 hour. The solvents were removed under reduced pressure to obtain oily crude residue. .Purification by silica gel column chromatography using 50% ethyl acetate in hexanes was performed to afford pure 42 (0.21 g, 65%).
[0248] c) Compound 4.2 (0.2 lg, 0.6 mmol) was dissolved in dichloromethane (5 mL). Trifluoroacetic acid (2.5 mL) was added and the reaction was stirred for 1 hour. The resulting reaction mixture was concentrated to remove any excess trifluoroacetic acid to afford amine 4.3 in quantitative yield.
84

WO 2005/044817 PCT/US2004/036942
[0249] d) Compound 4.4 was made according to Example 3d-f except that 4-
chlorobenzoic acid was used instead of compound 1.11.
[0250] e) Compound 4 was made according to Example 3g except that
compound 4.4 was used instead of compound 3.7 and compound 4.3 was used
instead of compound 3.4.
[0251] EXAMPLE 5
[0252] This example describes the synthesis of
which was prepared according to Scheme 4 and the procedure below. [0253] SCHEME 4

[0254] a) To a solution of methane sulfonamide (1.01 g, 10.7 mmol) in 15 mL DMF was added 20 M aqueous NaOH (0.68 mL, 13.6 mmol), resulting in a white precipitate. The solution was cooled to 0°C, carbon disulfide (0.4 mL, 6.63 mmol) slowly added, and stirred at 0°C for 15 minutes. Additional 20M aqueous NaOH (0.32mL, 6.4 mmol) and carbon disulfide (0.2mL, 3.31 mmol) were added and the reaction stirred at 0°C for 20 minutes, then raised to room temperature. At 30 minutes all precipitate had returned to solution, and the reaction mixture was cooled to 0°C. Methyl iodide (1.33 mL, 21.364 mmol) was added and the reaction stirred at 0°C for 20 minutes and at room temperature for 1.5 hours. 20 mL water was added to the reaction and extracted five times with ethyl acetate. The combined organic
85

WO 2005/044817 PCT/US2004/036942
extracts were dried over MgSO4 and concentrated to dryness. Recrystallization from hot ethyl acetate and hexanes afforded 1.44 g compound 5.1. ESI-MS (m/z): (M+H+) 200.0.
[0255] b) To a solution of Boc-Dap-OH (109 mg, 0.53 mmol) and compound 5.1 (125.7 mg, 0.632 mmol) in 5mL ethanol was added 1.0M aqueous NaOH (0.8 mL, 0.8 mmol). The reaction was stirred until conversion was complete, and then concentrated to dryness. The residue was dissolved in water and washed three times with ether. The aqueous layer was acidified to pH 1 with 2.0M phosphoric acid, and extracted four times with ethyl acetate. The combined ethyl acetate extracts were dried over MgSC>4 and concentrated to dryness to afford 160.4 mg of compound 5.2. ESI-MS (m/z): (M+Na+) 378.0.
[0256] c) To a solution of compound 5.2 (160.4 mg, 0.452 mmol) in 1:1 dichloromethane:methanol was added trimethylsilyldiazomethane as a 2.0 M solution in ether (0.4 mL, 0.8 mmol). The reaction was stirred at room temperature until conversion to the methyl ester was complete, and then concentrated to dryness. The product was purified via flash chromatography to afford 146.6mg of compound 5.3. ESI-MS.(m/z): (M+Na4) 270.0; !H NMR (400 MHz, chloroform-d) 5 1.46 (s, 9H), 2.42 (s, 3H), 3.02 (s, 3H), 3.63 (m, 1H), 3.75 (m, 1H), 3.82 (s, 3H), 4.51 (m, 1H).
[0257] d) To a solution of compound 5.3 (146.6mg, 0.40mmol) in 2mL methanol was added ammonia, 7N in methanol (0.6 mL, 4.2 mmol). This mixture was cooled to 0 °C, and a solution of silver nitrate (75.5 mg, 0.444 mmol) in 0.4 mL acetonitrile was dropwise added. The reaction was stirred and allowed to reach room temperature. At three hours solvent was removed, the residue suspended in ethyl acetate, and filtered through celite. The filtrate was concentrated to dryness and redissolved in dichloromethane, to which was added HC1, 4.0 M in dioxane (0.5mL, 2.0 mmol). This solution was stirred at room temperature for 8 hours and concentrated to dryness to afford compound 5.4 as an HC1 salt. ESI-MS {m/z): (M+H4) 239.
[0258] e) Compound 5 was made according to Example 3g except that compound 4.1 was used instead of 3.7 and compound 5.4 was used instead of compound 3.4. ESI-MS (m/z): (M+H+) 590.0.
86

WO 2005/044817 PCT/US2004/036942
[0259] EXAMPLE 6
[0260] This example describes the synthesis of

which was prepared according to Scheme 5 and the procedure below. [0261] SCHEME 5

[0262] a) A solution of (L)-Boc-Dap-OH (10 mmol), dimethyl N-cyanodithioimniocarbonate (10 mmol) and DIEA (30 mmol) in ethanol was stirred at room temperature for 1 hour. Then, pyrrolidine (20 mmol) was added, and the reaction was heated at 65 °C for 10 hours. The reaction mixture was then diluted with ethyl acetate (150 mL), and was extracted with saturated NaH2PO4 (50 mL, the queous layer pH is between 4-6), water (50 ml) and brine. The organic layer was dried with anhydrous magnesium sulfate, filtered and concentrated. The crude product was dissolved in 4:1 DCM:MeOH, cooled at 0°C and to it was added trimthylsilyldiazomethane until it stays yellow for 30 seconds. The solution is concentrated and the residue was purified by column with 30-100% ethyl acetate in hexane to give the title compound in 20-30% yield. MS (m/z): 240(M-99). [0263] b) Compound 6.1 (72 mg, 0.20 mmol) in DCM (1 mL) was treated with HC1 (4 M in dioxane) for 2 hours. The reaction mixture was concentrated to give compound 6.2.
[0264] c) Compound 6 was made according to Example 3g except that compound 6.2 was used instead of compound 3.4. JH NMR (400 MHz, CDCI3) 8 7.77 (s, 1 H), 7.63 (m, 3 H), 7.37 (d, 1 H), 6.85 (s, 1 H), 6.32 (d, 1 H), 4.68 (s, 2 H),
87

WO 2005/044817 PCT/US2004/036942
4.31 (m, 1 H), 3.85 (m, 2 H), 3.60 (m, 4 H), 2.88 (m, 2 H), 1.89 (m, 4 H) ppm; ESI-
MS (m/z): (M+l) 597.1.
10265] EXAMPLE 7
[0266] This example describes the synthesis of

which was prepared according to Scheme 6 and the procedure below. [0267] SCHEME 6

[0268] a) To a solution of H-2-Mercapto-His-OH (598.8 mg, 3.2 mmol) in 5mL methanol was added 0.26mL sulfuric acid. This mixture was stirred at room temperature for 18 hours. Additional sulfuric acid (0.1 mL, 0.6 mmol) was added and the reaction stirred at 50 °C for 4.5 hours. Sodium carbonate (850.3 mg, 8.02 mmol) was slowly added, and the reaction concentrated via rotary evaporation to remove most of the methanol. 6 mL THF and 3 mL water were added with sodium carbonate (0.851 g, 8.03 mmol) and Boc2O (696.5 mg, 3.19 mmol). The reaction was stirred at room temperature for 1 hour, diluted with ethyl acetate, and washed with water and brine. The organic layer was dried over MgSO4 and concentrated to dryness. Purification via flash chromatography afforded 283.5mg of compound 7.1. ESI-MS (m/z): (M+JT1) 316.1.
88

WO 2005/044817 PCT/US2004/036942
[0269] b) Compound 7.1 (283.5 mg, 0.90 mmol) and 3-chloroperoxybenzoic acid (568.4 mg, 2.53 mmol) were dissolved in 6 mL dichloromethane and stirred at room temperature for 1.5 hours. The reaction was diluted with dichloromethane and poured into 1.0 M potassium carbonate. The aqueous layer was adjusted to neutral pH with 2.0 M phosphoric acid and extracted three times with dichloromethane. The combined organic extracts were dried over MgSC>4 and concentrated to dryness. Purification via flash chromatography afforded 240.8 mg of compound 7.2. ESI-MS (m/z): 370.1(M+Na+), 248.1 (M-Boc+H4); *H NMR (400 MHz, CDC13) 5 1.43(s, 9H), 3.14(m, 2H), 3.26(s, 3H), 3.74(s, 3H), 4.61(m, 1H), 7.02(s, 1H). [0270] c) To a solution of compound 7.2 (240.8 mg, 0.6931 mmol) in dichloromethane was added HC1, 4.0 M in dioxane (1.0 mL, 4.0 mmol). The reaction was stirred at room temperature for 1.5 hours and then concentrated to dryness to afford compound 7.3 as an HC1 salt. ESI-MS {m/z): (M+H4) 248. [0271] d) Compound 7 was made according to Example 3g except that compound 4.1 was used instead of compound 3.7 and compound 7.3 was used instead of compound 3.4. ESI-MS (m/z): (M+H4) 599.0; *H NMR (400 MHz, CDCI3) 8 2.88(s, 2H), 3.10(m, 1H), 3.21(s, 3H), 3.34(m, 1H), 3.67QH), 3.97(1H), 4.63(1H), 4.85(1H), 5.04(dd, 1H), 7.19 (s, 1H), 7.49(m, 5H). [0272] EXAMPLE 8 [0273] This example describes the synthesis of
which was prepared according to Scheme 7A and the procedure below.
89

WO 2005/044817 PCT/US2004/036942
[0274] SCHEME 7A

[0275] a) A three-neck flask containing 32 mmol of Li Cl was flamed with a gas lamp in vacuo, followed by flushing with N2. This sequence was repeated for 3 times in order to get dry LiCl. To the flask was added 10 mmol of (R, R)-(-)-pseudoephedrine glycinamide hydrate (A. G. Myers et al J. Org. Chem. 64: 3322-3327 (1999)) and 30 mL of dry THF at room temperature under an atmosphere of N2. The suspension was then treated with 31 mmol of LiHMDS (1.0 M in THF) at 0°C with stirring for lhour, followed by adding a solution of 10 mmol of 3-methylthiobenzyl chloride (S. LauferetalJ. Med. Chem. 45:2733-2740 (2002)) in 5 mL of dry THF. The resulting mixture was stirred at 0°C overnight and quenched by adding 10 mL of water. The solvent was removed, and the residue was diluted with 50 mL of water. The mixture was extracted with CH2CI2 for 3 times. The organic extract was combined and dried with anhydrous Na2SO4. Subsequently, the solvent was removed, and the residue was purified to give the desired alkylated intermediate in 70% yield. ESI-MS (m/z): (M+H+) 359.2.
[0276] A mixture of 5 mmol of the alkylated intermediate in 12 mL of 1.0 N NaOH was refluxed until the starting material was consumed. The mixture was diluted with 20 mL, of water and extracted with CH2CI2 for 3 times. The aqueous phase was then stirred with 6 mmol of (Boc)2O and 12 mmol of NaHCO3 in 30 mL of 1,4-dioxane for 15 hours. The organic solvent was removed, and the residue was diluted with 30 mL of water and extracted with CH2CI2. The aqueous phase was subsequently treated with solid citric acid to adjust the pH value to 4.0, flowed by extracting with EtOAc for 3 times. The organic extract was dried with anyhydrous Na2SO4. The solvent was removed, and the residue was dried in vacuo to give compound 8.2 in quantitative yield. ESI-MS {m/z): (M+H4) 334.10.
90

WO 2005/044817 PCT/US2004/036942
[0277] b) A mixture of 5 mmol of compound 8.2 in 10 mL of CH3OH and 10 mL of CH2CI2 was added 10 mmol of (trimethylsilyl)diazomethane (2.0 M in hexanes) at 0 °C, the resulting mixture was stirred at room temperature for 30 minutes. The solvent was removed, and the residue was dried in vacuo to give the desired ester in quantitative yield. ESI-MS (m/z): (M+H1) 348.10. [0278] To a solution of the ester in 20 mL of CH3OH and 2 mL of water was added 12 mmol of Oxone® at room temperature, and the resulting suspension was stirred at room temperature for 15 hours. The reaction mixture was concentrated and diluted with 50 mL of EtOAc, washed with water, and dried with anhydrous Na2SO4. The solvent was removed and the residue was dried in vacuo to give compound 8.3 in quantitative yield. 1H NMR (400 MHz, CD3CI): 5 7.82 (m, 1H), 7.69 (s, 1H), 7.50 (m, 1H), 7.43 (m, 1H), 5.03 (m, 1H), 4.62 (m, 1H), 3.74 (s, 3H), 3.26 (m, 1H), 3.09 (m, 1H), 3.03 (s, 3H), 1.39 (s, 9H) ppm; ESI-MS (m/z): (M-fBoc+H4) 258.1.
[0279] c) A mixture of 2 mmol of compound 8.3 in 10 mL of 4.0 N HC1 in dioxane was stirred at room temperature for 15 hours. The solvent was removed, and the residue was dried in vacuo to give compound 8.4 in quantitative yield as an HC1 salt. 1H NMR (400 MHz, CD3OD): 5 7.95 (m, 1H), 7.87 (s, 1H), 7.64 (m, 2H), 4.44 (t, J= 6.85 Hz, 1H), 3.82 (s, 3H), 3.41 (m, 1H), 3.29 (m, 1H), 3.13 (s, 3H) ppm; ESI-MS (m/z): (M+H4) 258.10.
[0280] d) Compound 8 was made according to Example 3g except that compound 4.1 was used instead of compound 3.7 and compound 8.4 was used instead of compound 3.4. XH NMR (400 MHz, CD3OD): 8 7.92 (s, 1H), 7.81 (d, J=7.92 Hz, 1H), 7.68 (d, J= 7.83 Hz), 7.56 (t, J= 7.83 Hz, 1H), 7.30-7.49 (m, 5H), 5.06 (m, 1H), 4.60 and 4.83 (m, 2H), 3.95 and 3.66 (m, 2H), 3.44 (d, J=13.44 Hz, 1H), 3.14 (m, 1H), 3.08 (s, 3H), 2.85 (m, 2H) ppm; ESI-MS (m/z): (M+tf) 609.05. [0281] e) Compound 8.4 can also be prepared according to Scheme 7B and the procedure below.
[0282] SCHEME 7B
91

WO 2005/044817 PCT/US2004/036942

[0283] A mixture of 8.5 (1.0 mmoL), methyl iodide (1.2 mmoL) and potassium carbonate (2 mmoL) in 20 mL of acetone was heated at 50°C for 3 hours. The solvent of the reaction mixture was removed under reduced pressure. The residue was partitioned between EtOAc and water. The aqueous solution was extracted with EtOAc, and combined organic solution was washed with brine, dried over Na2SC>4, filtered, concentrated. The crude product as a white solid (yield 98%) was used for next step without purification.
[0284] To a solution of the compound (1 mmoL) made above in 4 mL of THF at 0 °C was added LiAIH4 (1.1 mmoL) slowly. The reaction mixture was allowed to warm to room temperature and stirred for 1 hour. The reaction was added consecutively with water, 15% aqueous NaOH and water with strong stirring. The filtration and evaporation of filtrate provided the crude product S.6 (yield 92%). No purification was needed. 1H NMR (400 MHz, CDCI3) 5 3.05 (s, 3 H), 4.75 (s, 2 H), 7.53 (t, J=7.58 Hz, 1 H), 7.62 (d, J=7.34 Hz, 1 H), 7.81 (d, J=7.83 Hz, 1 H), 7.93 (s, 1H).
[0285] Solid tetrapropylammonium perruthenate ("TPAP", 0.05 mmol) was added in one portion to a stirred mixture of compound 8.6 (1 mmoL), 4-methylmorpholine N-oxide ("NMO"; 1.5 mmoL) and powdered 4A molecular sieve (equal weight to that of NMO) in 5 mL of DCM at room temperature under N2. The reaction mixture was stirred at room temperature, for 1 hour, and then filtered through a short pad of silica gel, eluting with mixture of DCM and AcOEt (1:1). The filtrate was concentrated and the residue was purified with chromatography (SiO2, AcOEt/hexane 2:1) to afford compound 8.7 (yield 72%). 1H NMR (400
92

WO 2005/044817 PCT/US2004/036942
MHz, CDCI3) 8 3.14 (s, 3 H), 7.81 (t, J=7.58 Hz, 1 H), 8.21 (t, J=9.05 Hz, 2 H), 8.46 (s, 1H), 10.12 (s, lH)ppm.
[0286] The JV, TV, N', iV',-tetramethylguanidine ("TMG"; 1.05 mmole) was added slowly to a solution of (d,l)-Cbz-a-phosphonoglycine trimethylester (1.1 mmole) in 4 ml of DCM at room temperature. After 15 minutes, the mixture was cooled to -30°C and compound 8.7 (1 mmole) added dropwise. The mixture was kept at -30°C for 20 minutes and slowly allowed to warm to 0°C. The solution was diluted with AcOEt and washed consecutively with 1 N NaHSC>4 and brine. The solution was dried (Na2SO4), and solvent evaporated to provide crude product. Purification of thecrude product on chromatography (SiO2, AcOEt/hexanes/DCM 3:3:1) to give nroduct 8.8 (yield 72%). *H NMR (400 MHz, CDCI3) 5.2.97 (s, 3 H), 3.86 (s, 3 H), 5.08 (s, 2 H), 6.78 (s, 1 H), 7.34 (d, J=6.36 Hz, 5 H), 7.50 (t, J=7.83 Hz, 1 H), 7.72 (d, J=7.34 Hz, 1 H), 7.85 (d, J=7.34 Hz, 1 H), 8.04 (s, 1 H). Olefinic proton in the minor trans isomer at 7.19 ppm (s, 1H). ESI-MS (m/z): (M+H4) 346. [0287] To the solution of 8.8 (1 mmole) in MeOH (20 mL, presparged with nitrogen gas) in a glass pressure vessel was added chiral catalyst (+)-bis((2S,5S)-2,5-dimethylphospholano)benzene(cyclooctadiene)rhodium(T)tetrafluoroborate (0.01 mmole). The reactor was then pressurized with H2 to 40 psi and shaking was continued at room temperature for 17 hours. The solvent was evaporated. The residue was dissolved in AcOEt and filtered through a plug of SiO2 with AcOEt. The filtrate was evaporated to provide the crude product 8.9. (yield, 72%). 1H NMR (400 MHz, CDCI3) 5 2.98 (s, 3 H), 3.13 (dd, J=13.69, 6.36 Hz, 1 H), 3.29 (m, 1 H), 3.76 (s, 3 H), 4.69 (m, 1 H), 5.06 (m, 2 H), 5.44 (d, J=6.85 Hz, 1 H), 7.31 (m, 5 H), 7.41 (d, J=7.34 Hz, 1 H, 7.47 (t, J=7.83 Hz, 1 H), 7.72 (s, 1 H), 7.82 (d, J=7.34 Hz, 1 H). ESI-MS (m/z): (M+H4) 348.
[0288] The compound 8.9 was hydrogenated (Pd/C, MeOH,) with a hydrogen balloon to afford compound 8.4 (yield 98%). ESI-MS {m/z): (M+H4) 258. [0289] EXAMPLE 9 [0290] This example describes the synthesis of
93

WO 2005/044817 PCT/US2004/036942

which was prepared according to Scheme 8 and the procedure below. [0291] SCHEME 8

[0292] a) Compound 9.1 was made according to Example ld-e except that compound 8.4 was used instead of compound 1.3.
[0293] b) Compound 9 was made according to Example 3g except that compound 9.1 was used instead of compound 3.7 and 2-hydroxycinnamic acid was used instead of compound 3.4. 1H NMR (400 MHz, CD3OD) 5 7.92 (m, 2H), 7.81 (m, 1 H), 7.68 (s, 1 H), 7.58 (m, 2 H), 7.28 (m, 2 H), 7.18 (m, 1 H), 6.84 (m, 2 H),
5.7 (m, 1 H), 4.80 and 4.88 (m, 2H), 3.96 (m, 2H), 3.42 (m, 1H), 3.15 (m, 1H),
3.7 (s, 3H), 2.71-2.91 (m, 2H) ppm; ESI-MS (m/z): (M+H4) 617.10.
[0294] EXAMPLE 10
[0295] This example describes the synthesis of

which was prepared according to Example 3g except that compound 9.1 was used instead of compound 3.7 and 2-fmorocinnamic acid was used instead of compound 3.4. 1H NMR (400 MHz, CD3OD) 5 7.92 (s, 1 H), 7.81 (m, 3H)5 7.68 (m, 1H), 7.57 (m, 1H), 7.42 (m, 1H), 7.29 (m, 2H), 7.23 (m, 1H), 7.16 (m, 1H), 5.06 (m, 1H), 4.81
94

WO 2005/044817 PCT/US2004/036942
and 4.88 (m, 2H), 3.82 and 3.97 (m, 2H), 3.44 (m, 1 H), 3.16 (m, 1 H), 3.08 (s, 3 H),
2.86 and 2.93 (m, 2H) ppm; ESI-MS (m/z): (M+H4) 619.10.
[0296] EXAMPLE 11
[0297] This example describes the synthesis of

which was prepared according to Example 3g except that compound 9.1 was used instead of compound 3.7 and 6-indazolecarboxylic acid was used instead of compound 3.4. JH NMR (400 MHz, CD3OD) 5 8.13 (s, 1H), 7.93 (m, 3H), 7.83 (m, 1H), 7.70 (m, 1H), 7.60 (m, 1H), 7.20 and 7.34 (m, 1H), 5.06 (m, 1H), 4.64 and 4.88 (m, 2H), 3.69 and 3.98 (m, 2H), 3.40 (m, 1H), 3.15 (m, 1H), 3.08 (s, 3H)5 2.88 (m, 2H)ppm; ESI-MS (m/z): (M+H1) 615.15.
[0298] EXAMPLE 12
[0299] This example describes the synthesis of

[0300] which was prepared according to Example 3g except that-compound 9.1 was used instead of compound 3.7 and 2-indolecarboxylic acid was used instead of compound 3.4. !H NMR (400 MHz, CD3OD) 5 7.93 (s, 1H), 7.83 (m, 2H), 7.72 (m, 1H), 7.83 (m, 1H), 7.57 (mj 1H), 7.45 (m, 1H), 7.27 (s, 1H), 7.20 (m, 1H), 7.07 (m, 1H), 6.93 (s, 1H), 5.08 (m, 1H), 4.88 (m, 2H), 4.07 (m, 2H), 3.46 (m, 1H), 3.15 (m, 1H), 3.08 (s, 3H), 2.96 (m, 2H) ppm; ESI-MS (m/z): (M+H4) 614.10.
[0301] EXAMPLE 13
[0302] This example describes the synthesis of
95

WO 2005/044817 PCT/US2004/036942

[0303] which was prepared according to Example 3g except that compound 9.1 was used instead of compound 3.7 and 2-quinolinecarboxylic acid was used instead of compound 3.4. 1H NMR (400 MHz, CD3OD) 5 8.51 (m, 1H), 8.08 (m, 1H), 8.01 (m, 1H), 7.94 (m, 1H), 7.84 (m, 2H), 7.70 (m, 3H), 7.58 (m, 1H), 7.36 and 7.04 (m, 1H), 5.07 (m, 1H), 4.78 and 4.95 (m, 2H), 3.79 and 4.08 (m, 2H), 3.45 (m, 1H), 3.17 (m, 1H), 3.09 (s, 3H), 2.97 (m, 2H) ppm; ESI-MS (m/z): (M+H4) 626.10.
[0304] EXAMPLE 14
[0305] This example describes the synthesis of

[0306] which was prepared according to Scheme 9 and the procedure below. [0307] SCHEME 9

[0308] a) To a solution of 3-carboxylbenzenesulfonyl chloride (3.54 g, 16 mmol) in ethyl acetate (50 mL) at 0 °C was added concentrated ammonia (2.5 mL). The reaction was neutralized with HC1 in dioance (20 mL), diluted with ethyl acetate (100 mL), dried with anhydrous sodium sulfate and filtered. Concentration of the filtrate yielded the title compound, which was used without purification.
96

WO 2005/044817 PCT/US2004/036942
[0309] b) Crude compound 14.1 was dissolved in THF (50 mL), to it was added borane (1.0 M in THF, 50 mL) over 20 minute period. After the reaction was stirred at room temperature for 15 hours, the reaction was diluted with brine (20 mL) and water (10 mL), extracted with ethyl acetate (100 mL). The organic extract was dried over anhydrous sodium sulfate and filtered. Concentration of the filtrate yielded the title compound, which was used without further purification. [0310] c) To crude compound 14.2 solution in DCM (100 mL) was added activated 4A molecular sieve powder (8 g), pyridinium dichromate (7.55 g, 20 mmol). After the reaction was stirred at room temperature for 2 hours, the reaction mixture was filtered through silica gel (50 g), rinsed with ethyl acetate. The residue after concentration of the filtrate was purified by silca gel column with 30-50% ethyl acetate in hexane to give compound 14.3 (477mg, 16%, 3 steps). ESI-MS (m/z): (M+H+)186.

which was prepared according to Scheme 10 and the procedure below.
97
[0311] d) Compound 14.4 was made according to Example 8e except that compound 14.3 was used instead of compound 8.7. MS (ESI4) m/z: 260 (M+H+). [0312] e) Compound 14 was made according to Example 3g except that compound 14.4 was used instead of compound 3.4. JH NMR (400 MHz, CD3OD) 5 7.89 (s, 1 H), 7.80 (s, 1 H), 7.75 (m, 2 H), 7.64 (s, 1 H), 7.57(d, 1 H), 7.34 (d, 2 H), 6.93 9s, 1 H), 5.00 (m, 1 H), 3.99 (m, 1 H), 3.73 (m, 1 H), 3.40 (dd, 1 H), 3.12 (dd, 1 H), 2.89 (m, 2 H) ppm; ESI-MS (m/z): 616 (M+H+). [0313] EXAMPLE 15 [0314] This example describes the synthesis of

WO 2005/044817 PCT/US2004/036942

[0316] a) To a solution of 0.2 mol of fuxan in 200 mL of dry THF was added 0.2 mol of n-BuLi (1.6 M in hexanes) at -78 °C, the resulting solution was stirred at room temperature for 4 hours. Subsequently, the mixture was cooled to -78 °C and treated with 0.21 mol of dimethyl disulfide, and the mixture was stirred at room temperature overnight, followed by adding 10 mL of saturated aqueous NH4CI. The mixture was concentrated at room temperature, and the residue was diluted with 200 mL of saturated aqueous NH4CI and extracted with ether. The extract was then washed with brine and dried with anhydrous Na2SO4. The solvent was removed, and the residue was distilled to collect, the fraction at 135-140 °C/760 mmHg to give compound 15.1 in 55% yield. 1H NMR (400 MHz, CD3C1): 5 7.50 (s, 1H), 6.45 (m, 1H), 6.39 (s, 1H), 2.42 (s, 3H) ppm.
[0317] b) To a solution of 0.1 mol of compound 15.1 in 100 mL of dry THF was added 0.1 mol of rc-BuLi (1.6 M in hexanes) at -78 °C, the resulting solution was stirred at room temperature for 4 hours. Subsequently, the mixture was cooled to -78 °C and treated with 0.12 mol of dry DMF, and the mixture was stirred at room temperature overnight. The reaction was quenched by adding 10 mL of saturated aqueous NH4CI, and the mixture was concentrated. The residue was diluted with 100 mL of brine and extracted with EtOAC. The extract was washed with brine and dried with anhydrous Na2SO4. The solvent was removed and the residue was purified to give the title compound in 65% yield. !H NMR (400 MHz, CD3CI): 5 9.52 (s, 1H), 7.24 (d, J= 3.4 Hz, 1H), 6.42 (d, J= 3.4Hz, 1H), 2.60 (s, 3H) ppm; ESI-MS (m/z): (M+H4) 143.1.
98

WO 2005/044817 PCT/US2004/036942
[0318] c) A mixture of 50 mmol of compound 15.2 and 120 mmol of m-CPBA
in 100 mL of CH2CI2 was stirred at room temperature overnight. The mixture was
diluted with 150 mL of CH2CI2, and the mixture was washed with saturated aqueous
NaHCC>3 for several times. The solution was then dried with anhydrous Na2SO4 and
concentrated. The residue was purified to give compound 15.3 in 70% yield. !H
NMR (400 MHz, CD3C1): 8 9.83 (s, 1H), 7.33 (m, 2H), 3.27 (s, 3H) ppm; ESI-MS
(m/z): (M+H+) 175.0.
[0319] d) Compound 15.4 was made according to Example 8e except that
compound 15.3 was used 'instead of 8.7. ESI-MS (m/z): (M+H4) 248.1.
[0320] e) Compound 15 was made according to Example except that compound
15.4 was used instead of 3.4. JH NMR (400 MHz, CD3OD): 5 7.92 (s, 1H), 7.76 (m,
1H), 7.67 (s, 1H), 7.34 (m, 1H), 7.13 (s, 1H), 6.69 (s, 1H), 6.49 (s, 1H), 5.11 (m,
1H), 4.73 and 4.88 (m, 2H), 3.76 and 4.02 (m, 2H), 3.46 (m, 1H), 3.30 (m, 1H), 3.17
(s, 3H), 2.94 (m, 2H) ppm; ESI-MS (m/z): (M+H") 605.05.
[0321] EXAMPLE 16
[0322] This example describes the synthesis of

.OH o o B' I *a °
which was prepared according to the procedure below.
[0323] To a solution of 0.2 mmol of compound 8.4 (Example 8c or 8e) in 1 mmol of Et3N and 5 mL of dry CH2CI2 was added 0.22 mmol of 2,6-dichlorobenzoyl chloride at 0°C, the resulting mixture was stirred at room temperature for 12 hours. The solvent was removed and the residue was dried in vacuo. Subsequently, the residue was treated with 0.8 mmol of LiOH-H2O in 2 mL of THF and 0.5 mL of H2O. After stirring at room temperature for 30 minutes, the reaction mixture was added 1.0 mL of 1.0N aq. HC1. The organic solvent was removed, and the residue was diluted with 10 mL of brine. The mixture was extracted with EtOAc and the extract was dried with anhydrous Na2SO4. The solvent was removed and the residue was dried in vacuo to give the desired compound in
99

WO 2005/044817 PCT/US2004/036942
65% yield. 1H NMR (400 MHz, CD3OD) 5 7.92 (s, 1 H), 7.82 (d, J=6.85 Hz, 1 H),
7.71 (d, J=6.85 Hz, 1 H), 7.56 (t, J=7.83 Hz, 1 H), 7.34 (m, 3 H), 5.08 (dd, J=9.78,
4.89 Hz, 1 H), 3.45 (dd, J=14.67, 4.89 Hz, 1 H), 3.14 (dd, J=14.67, 9.78 Hz, 1 H),
3.08 (s, 3 H) ppm; ESI-MS (m/z): (M+H+) 416.00.
[0324] EXAMPLE 17
[0325] This example describes the synthesis of

which was prepared according to the procedure below.
[0326] Compound 6.1 (Example 6a, 0.2 mmol) in DCM (1 mL) was treated with HC1 in dioxane (4.0 M, 1 mL). After lhour, the solvent was evaporated. The residue and 1 mmol of Et3N and 5 mL of dry CH2CI2 was added 0.22 mmol of 2,6-dichlorobenzoyl chloride at 0 °C, the resulting mixture was stirred at room temperature for 15 hours. The solvent was removed and the residue was dried in vacuo. Subsequently, the residue was treated with 0.8 mmol of LiOH.H2O in 2 mL of THF and 0.5 mL of H2O. After stirring at room temperature for 30 minutes, the reaction mixture was added 1.0 mL of 1.0 N aqueous HC1. The organic solvent was removed, and the residue was diluted with 10 mL of brine. The mixture was extracted with EtOAc and the extract was dried with anhydrous Na2SO4. The solvent was removed and the residue was dried in vacuo to give the desired compound.
[0327] EXAMPLE 18

[0329] which is prepared according to Scheme 11 and the procedure below.
100
[0328] This example describes the synthesis of

WO 2005/044817 PCT/US2004/036942

[0331] a) 1 equivalent of LiAIH4 (1.0M in THF) was added to a 0 °C solution of 1 equivalent of compound ethyl 6-indazolecarboxylate (Batt,D.G., J.Med. Chem. 43; 41-58 (2000)) at -78 °C. The reaction was stirred at -78 °C for another 30 minutes, and then warmed to 0°C. An aqueous solution of 1 equivalent of 1M NaOH was added slowly. The resulting slurry was filtered thru a plug of Celite and washed with a copious amount of ethyl acetate. The combined organics were dried with MgSO4 and concentrated in vacuo to provide the alcohol 18.1 in high enough purity to be used without further purification.
[0332] b) 1.1 equivalent of Dess-Martin periodinane was added to 1 equivalent of 18.1 in dichloromethane. After stirring the reaction for 3 hours at room temperature, the resulting precipitate was removed by filtering thru a plug of celite. The celite plug was washed with dichloromethane. The combined organics were
101

WO 2005/044817 PCTYUS2004/036942
concentrated to provide the aldehyde 18.2 in high enough purity to be used without further purification.
[0333] c) 2.1 equivalents of ethyl magnesium bromide (0.5M in THF) were added to a pre-cooled solution containing 18.2 in THF at 0°C. After 30 minutes, the reaction was warmed to room temperature and stirred for an additional 2 hours. The resulting reaction mixture was diluted with ethyl acetate and washed with water. The organic layer was then dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 18.3. [0334] d) To a mixture of 4-amino-2,6-dichlorophenol (1 equivalent) in 3:2 THF/H2O was added NaHCO3 (1.1 equivalent) and Boc2O (1.1 equivalent), after stirring overnight, the reaction was extracted with ether, and dried with MgSO4, filtered, and concentrated in vacuo. The residue 18.4 was used without purification. [0335] e) To a solution of phenol 18.4 (1 equivalent) and 2,6-lutidine (2.2 equivalent) in DCM at -78°C was added triflic anhydride (1.2 equivalent). After the reaction mixture was gradually warmed to room temperature overnight, the reaction mixture was diluted with ether, washed with water, dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column (gradient elution using ethyl acetate and hexanes) to provide pure compound 18.5. [0336] f) A mixture of lOmmol of the triflate 18.5 , l.Ommol of dppp and 40mmol of DIEA (in lOOmL of, dry DMF and 50mL of anhydrous CH3OH was flushed with CO for 15 minutes, and then l.Ommol of Pd(OAc)2 was added under the atmosphere of CO. Subsequently, the resulting mixture was stirred at 70 °C overnight under an atmosphere of CO. The solvent was removed and the residue was purified by column chromatography with 10-30% EtOAc in hexane to give compound 18.6.
[0337] g) 1 equivalent of Boc-aniline 18.6 was dissolved carefully in 6 M aqueous H2SO4, and the mixture was then cooled to 0 °C. To it was added slowly with vigorous stirring sodium nitrite (1.1 equivalent in water), followed by sodium iodide (5 equivalent) in 1.5 hours. After the reaction was stirred at room temperature for overnight, the reaction was diluted with ether, washed with water, dried with MgSO4, filtered, and concentrated in vacuo. The residue was then
102

WO 2005/044817 PCT/US2004/036942
purified on silica gel column (gradient elution using ethyl acetate and hexanes) to provide pure compound 18.7.
[0338] h) 1 equivalent of iodide 18.7, 1 equivalent of alkyne 18.3, 0.05 equivalent of Cul, and 5 equivalents of triethylamine were dissolved in benzene and the solution degassed by bubbling N2 thru a syringe needle and into the solution for 15 minutes. 0.05 equivalent of PdCl2(dppf). DCM was added. After 4 hours, the reaction was diluted with ethyl acetate and washed with water, brine. The organic layer was then dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 18.8.
[0339] i) 1 equivalent of 18.8 was dissolved in MeOH and 5 % Rh/Al2O3 (20 weight%) was added. Under reduced pressure, oxygen was removed from the flask. The internal pressure was restored by the addition of hydrogen gas delivered using a hydrogen filled balloon. The reaction was stirred under an atmosphere of hydrogen gas for 14 hours. The reaction was filtered thru a pad of celite and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 18.9. [0340] j) 4 equivalents of Lil was added to 1 equivalent of compound 18.9 in pyridine. The reaction was refruxed for 14 hours, then allowed to cool to room temperature. The reaction was concentrated and the resulting residue was partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were dried over MgSO4, filtered, and concentrated. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and methanol) to provide pure compound 18.10. [0341] k) 1 equivalent of compound 18.10, 1 equivalent ofcompound 8.4 (Example 8c or 8e), and 3 equivalents of DIEA were dissolved in DMF. 1.1 equivalent of HATU was added. The reaction was stirred at room temperature for 14 hours. The reaction mixture was diluted with ethyl acetate and washed with water, brine. The combined organics were dried with MgSO4, filtered, and concentrated. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure intermediate ester. The ester was dissolved in methanol followed by addition of 2 equivalents of 1M
103

WO 2005/044817 PCT/US2004/036942
LiOH(aq). Upon completion, the excess solvents were removed under reduced
pressure and the resulting acid was then purified by reverse phase HPLC to give
pure compound 18.
[0342] EXAMPLE 19
[0343] This example describes the synthesis of

which was prepared by treating a solution of compound 18 (Example 18) in DCM7TFA (1:1 ratio) with triethylsilane (10 equivalent). After LC-MS showed that the starting material was completely consumed, the reaction was concentrated, and the residue was purified by reverse phase HPLC to give the title compound.
[0344] EXAMPLE 19
[0345] This example describes the synthesis of
which was prepared according to Scheme 12 and the procedure below.
104

WO 2005/044817 PCT/US2004/036942
[0346] SCHEME 12
i
i
[0347] a) A solution of 1 equivalent of compound 18.7 (Example 18g) in THF at -40 °C was treated with 1.0 equivalent of isopropylmagnesium bromide. After 0.5 hour, DMF (5 equivalents) was added, and the reaction was warmed to room temperature over night. The reaction mixture was diluted with ethyl acetate and washed with water, brine. The organic layer was then dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 20.1.
[0348] b) To a solution of aldehyde 20.1 (1 equivalent) in THF at -78 °C was added 2.1 equivalents of ethnyl magnesium bromide (0.5 M in THF). After the reaction was warmed to room temperature and stirred for an additional 2 hours. The resulting reaction mixture was diluted with ethyl acetate and washed with water. The organic layer was then dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 20.2. [0349] c) 1 equivalent of compound 20.2, 1 equivalent of l-chloro-4-iodobenzene, 0.05 equivalent of Cul, and 5 equivalents of triethylamine were dissolved in benzene and the solution degassed by bubbling N2 thru a syringe needle and into the solution for 15 minutes. 0.05 equivalent of PdCl2(dppf).DCM was added. After 4 hours, the reaction was diluted with ethyl acetate and washed with water, brine. The organic layer was then dried with MgSO4, filtered, and concentrated in vacuo. The residue was then purified on silica gel column
105

WO 2005/044817 PCTYUS2004/036942
chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 20.3.
106
[0350] d) 1 equivalent of compound 20.3 was dissolved in MeOH. 5% Rh/Al2O3 was added. Under reduced pressure, oxygen was removed from the flask. The internal pressure was restored by the addition of hydrogen gas delivered using a hydrogen filled balloon. The reaction was stirred under an atmosphere of hydrogen gas for 14 hours. The reaction was filtered thru a pad of celite and concentrated in vacuo. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 20.4. [0351] e) 4 equivalents of Lil was added to 1 equivalent of compound 20.4 in pyridine. The reaction was refiuxed for 14 hours, then allowed to cool to room temperature. The reaction was concentrated and the resulting residue was , partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were dried over MgSO4, filtered, and concentrated. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and methanol) to provide pure compound 20.5. [0352] f) 1 equivalent of compound 20.5, 1 equivalent of compound 8.4 (Example 8c or 8e), and 3 equivalents of DIEA were dissolved in DMF. 1.1 equivalent of HATU was added. The reaction was stirred at room temperature for 14 hours. The reaction mixture was diluted with ethyl acetate and washed with water, brine. The combined organics were dried with MgSO4, filtered, and concentrated. The residue was then purified on silica gel column chromatography (gradient elution using ethyl acetate and hexanes) to provide pure compound 20.6. [0353] g) 1 equivalent of compound 20.6 was dissolved in methanol followed by 2 equivalents of 1M LiOH(aq). Upon completion, the excess solvents were removed under reduced pressure and the resulting acid was then purified by reverse phase HPLC and lyophilized to a pure powder compound 20. [0354] EXAMPLE 21 [0355] This example describes the synthesis of


WO 2005/044817 PCT/US2004/036942
which was prepared according to Scheme 13 and the procedure helow. [0356] SCHEME 13

[0357] a) A solution of bromine (461 uL, 9.00 mmol) in methanol (6.0 mL) was prepared at -78°C. The cold solution was added dropwise to a mixture of KCN (1.85 g, 19.0 mmol) in methanol (6.0 mL) under nitrogen at -78°C. After 20 minutes, a solution of pyrrole (0.624 mL, 9.00 mmol) in methanol (20 mL) was added. The mixture was allowed to reach -40°C and stirred for 0.5 hour. It was then poured into ice-water and extracted with ether (3x). The organic layers were combined and washed with saturated sodium thiosulfate and brine. The organic phase was then dried (Na2SO4) and concentrated. The crude residue was purified by flash chromatography (0-50% ethyl acetate in hexane) to yield 734 mg (66%) of compound 21.1 as a clear oil, Rf 0.24 (10% ethyl acetate in hexane). lH NMR (CDC13) 5 8.68 (br s51 H), 7.03 (s, 1 H), 6.69 (s, 1 H), 6.32 (s, 1 H). ES (+) MS m/e = 125(M + H)+.
[0358] b) To a solution of 21.1 (614 mg, 4.95 mmol) and iodomethane (0.340 mL, 5.45 mmol) in methanol (40 mL) under nitrogen at -10°C was added dropwise
107

WO 2005/044817 PCT/US2004/036942
aqueous sodium hydroxide (9.90 mL, 1 M, 9.90 mmol). The mixture is warmed to ambient temperature and stirred for 0.5 h. Excess sodium hydroxide is then quenched by the addition of dry ice. The mixture is tiluted with brine and extracted with dichloromethane (3x). The combined organic layers are dried (Na2SO4) and concentrated to yield 504 mg (90%) of compound 21.2 as a dark oil, Rf 0.35 (10% ethyl acetate in hexane). 1H NMR (CDC13) 5 8.31 (br s, 1H), 6.84 (s, 1 H), 6.38 (s, 1 H), 6.24 (s, 1 H), 2.36 (s, 3 H).
[0359] c) To a 0°C solution of compound 21.2 (100 mg, 0.884 mmol) in methanol (4.0 mL) was added dropwise sodium periodate (208 mg, 0.972 mmol) in water (4.0 mL). The mixture is allowed to reach room temperature and after 15 minutes LC/MS and TLC indicated complete disappearance of compound 21.2. The mixture was then filtered, concentrated, and the residue was partitioned between ethyl acetate and water. The aqueous layer was extracted twice with ethyl acetate and the combined organic phases were washed with brine, dried (Na2SO4) and concentrated to yield 63 mg (55%) of compound 213 as a dark oil, Rf 0.25 (ethyl acetate). 1HNMR (CDCI3) 5 11.74 (br s, 1 H), 7.00 (s, 1 H), 6.65 (s, 1 H), 6.20 (s, 1 H), 3.03 (s, 3 H). ES (+) MS m/e = 130 (M + H)+.
[0360] d) To a solution of compound 21.3 (60 mg, 0.464 mmol) in dichloromethane (2.0 mL) was added trifluoroacetic acid (1.0 mL). After 15 minutes, LC/MS and TLC indicated complete consumption of compound 21.3. The solvent was removed in vacuo and the residue was dried under high vacuum to yield 60 mg (100%) of compound 21.4 as an oil, Rf0.11 (ethyl acetate). 1H NMR (CDC13) 5 9.16 (br s, 1 H), 7.28 (s, 1 H), 6.94 (s, 1 H), 6.66 (s, 1 H), 3.02 (s, 3 H). ES (+) MS m/e=130(M + H)+.
[0361] e) To a mixture of compound 21.4 (60 mg, 0.464 mmol) and CS2CO3 (378 mg, 1.16 mmol) was added a solution of compound 21.5 (Ferreira et al, Tetrahedron Letters, 39: 9575 (1998); 140 mg, 0.464 mmol) in acetonitrile. The resulting mixture was stirred at 60°C until LC/MS and TLC indicated complete consumption of the starting material (~1 hour). The mixture was cooled to room temperature, diluted with ethyl acetate and washed with water and brine. The organic layer was dried (Na2SO4) and concentrated. The crude residue was purified by flash chromatography (0-100% ethyl acetate in hexane) to yield 114 mg (57%) of
108

WO 2005/044817 PCT/US2004/036942
compound 21.6 as a viscous oil, 12/0.31 (ethyl acetate). 1H NMR (CDC13) 5 7.05 (s, 1 H), 6.75 (s, 1 H), 6.50 (s, 1 H), 5.25 (m, 1 H), 4.65 (m, 1 H), 4.40 (m, 1 H), 3.79 (s, 3 H), 2.79 (s, 3 H), 1.47 (s, 18 H). ES (+) MS m/e = 275 (M - Boc - ?-Bu+ 2 H)+. [0362] f) To a solution of compound 21.6 (114 mg, 0.265 mmol) in dichloromethane (1.00 mL) is added mCPBA (89.0 mg, 0.397 mmol) portionwise. After 5 minutes at room temperature, LC/MS and TLC indicated complete disappearance of compound 21.6. The mixture was filtered and concentrated. The crude residue was purified by flash chromatography (0-50% ethyl acetate in hexane) to yield 89.0 mg (75%) of compound 21.7 as a clear oil, Rf0A5 (50% ethyl acetate in hexane). 1H NMR (CDCI3) 5 7.22 (s, 1 H), 6.69 (s, 1 H), 6.50 (s, 1 H), 5.27 (m, 1 H), 4.65 (m, 1 H), 4.45 (m, 1 H), 3.79 (s, 3 H), 305 (s, 3 H), 1.48 (s, 18 H). ES (+) MS m/e = 247 (M - 2 Boc + 3 H)+.
[0363] g) To a solution of compound 21.7 (89.0 mg, 0.0.199 mmol) in dichloromethane (0.50 mL) was added HC1 (4.00 mL, 4.0 M in dioxane). The resulting mixture was stirred at ambient temperature until LC/MS indicated complete deprotection (~1 hour). The mixture was concentrated and the residue was dried under high-vacuum to yield 53 mg (100%) of compound 21.8 as white powder. ES (+) MS m/e = 247 (M + H)+.
[0364] h) A mixture of compound 21.8 (53.0 mg, 0.199 mmol), compound 4.1 (Example 4a, 77.0 mg, 0.199 mmol), HATU (79.0 mg, 0.209 mmol), and triethyl amine (0.111 mL, 0.796 mmol) in DMF (1.00 mL) was stirred at room temperature over night. The mixture was then diluted with ethyl acetate and washed with 1.0 M aqueous HC1, saturated NaHCO3, and brine. The organic layer was dried (Na2SO4) and concentrated. The crude residue was purified by flash chromatography (0-100% ethyl acetate in hexane) to yield 70.3 mg (58%) of compound 21.9 as a white solid, 5/0.16 (75 %ethyl acetate in hexane). 1H NMR (CDCI3) 5 7.46-7.39 (m, 5 H), 7.28 (s, 1 H), 6.73 (s, 1 H), 6.69 (br s, 1 H), 6.48 (s, 1 H), 5.13 (m, 1 H), 4 83 (br s, 1 H), 4.56 (m, 1 H), 3.85 (s, 3 H), 3.70 (m, 1 H), 3.04 (s, 3 H), 2.91 (br s, 2 H), 2.81 (s, 2H). ES (+) MS m/e = 614 (M + H)+.
[0365] i) To a solution of 21.9 (70.3 mg, 0.115 mmol) in THF (1.00 mL) was added LiOH (aqueous 1.0 M, 0.360 mL, 0.360 mmol). The resulting mixture was stirred at room temperature until TLC and LC/MS indicated complete hydrolysis
109

WO 2005/044817 PCT/US2004/036942
(-0.5 hour). The reaction was then quenched by the addition of 1.0 M aqueous HC1 (0.400 mL) and concentrated to dryness. The residue was taken up in dimethylsulfoxide ("DMSO"; 4.0 mL) and purified by preparative RP-HPLC. The fractions containing pure compound were consolidated and concentrated. The residue was lyophilized under high-vacuum for 48 hours to yield 33.8 mg (49%) of compound 21 as a white powder. 1H NMR (CDC13) 8 7.46-7.39 (m, 4 H), 7.28 (m, 2 H), 7.15 (br s, 1 H), 6.77 (s, 1 H), 6.38 (s, 1 H), 5.74 (br s, 2 H), 5.04 (m, 1 H), 4.83 (br s, 1 H), 4.53 (m, 3 H), 3.69 (m, 1 H), 3.00 (s, 3 H), 2.85 (br s, 2 H). ES (+) MS m/e = 600(M + H)+. [0366] EXAMPLE 22 [0367] This example describes the synthesis of

which was prepared according to Scheme 14 and the procedure below. [0368] SCHEME 14

[0369] a) A solution of 5.0g (28.2 mmol) of compound 22.1 (Plobeck et al, J.MedChem. 43:3878-3894 (2000)) and sulfuryl chloride (100 mmol each, added at the beginning of the reaction and after 15 hours) in acetic acid (50 mL) was refluxed for 36 hours. The off white solid after concentration of the reaction mixture was rinsed with ether, and to the resulting crude product was added DCM (50 mL), followed by BBr3 (1.0 M in DCM, 100 mL). After 6 hours, the reaction mixture is
110

WO 2005/044817 PCT/US2004/036942
concentrated, and water (50 mL) was carefully added. The resulting precipitate is collected by suction filtration and dried to give crude compound 22.2 in quantitative yield.
[0370] b) The crude compound 22.2 was dissolved in DCM/pyridine (50 mL/50 mL) and cooled to 0 °C. To it was slowly added triflic anhydride (42.3 mmol), and the reaction was then stirred at room temperature for 6 hours. The reaction mixture was partitioned between ethyl acetate (200 mL) and water (50 mL), and the organic layer was washed with water (30 mL, twice) and brine, dried over anhydrous sodium sulfate and filtered. The residue after concentration of the filtrate was purified by silica gel column chromatography to give compound compound 22.3 (3.16 g, 32%). ESI-MS (m/z): (M+H+) 364/366.
[0371] c) Carbon monoxide gas was bubbled through a mixture of compound 22.3 (581 mg, 1.6 mmol), B2MAP (0. 2 mmol), palladium acetate (0.2 mmol), triethylamine (1 mL), anhydrous methanol (3 mL) and anhydrous DMF (3 mL) for 10 min, then the reaction was heated at 65 °C under a carbon monoxide balloon for 15 hours. The reaction mixture was partitioned between ethyl acetate (100 mL) and water (25 mL), and the organic layer was washed with water (25 mL, twice) and brine, dried over anhydrous magnesium sulfate and filtered. The residue after concentration of the filtrate was purified by silica gel column to give compound 22.4 (213 mg, 49 %). ESI-MS (m/z): (M+H+) 274/276.
[0372] d) To a suspension of sodium hydride (24 mg, 1.0 mmol) in THF (2 mL) was added compound 22.4 (63 mg, 0.23 mmol), 4-chlorobenzyl chloride (55 mg, 0.34 mmol) and tetrabutylammonium iodide (10 mg). After 6 hours, the reaction was diluted with ether and filtered through silica gel, rinsed with ether. The residue after concentration of the filtrate was purified by silica gel column to give compound 22.5 (50 mg, 55 %). ESI-MS (m/z): (M+H1) 398/400. [0373] e) A mixture of compound 22.5 (50 mg) and 1 mmol of Lil in 2 mL of pyridine was reflux overnight. The reaction was concentrated in vacuo and the residue was further dried by high vacuum for 2 hour. The resulting crude compound 22.6 was used without further purificaiton. ESI-MS (m/z): (M+l), 384. [0374] f) Compound 22 was prepared according to Example 3g except that compound 22.6 was used instead of compound 3.7 (yield: 82%). 1H NMR (400
111

WO 2005/044817 PCT/US2004/036942
MHz, dmso-d6): 9.12 (d, 1 H), 8.54 (t, 1 H), 7.90 (s, 1 H), 7.77 (dd, 1 H), 7.72 (dd,
1 H), 7.42 (d, 2 H), 735 (d, 2 H), 7.16 (dd, 1 H), 4.77 (m, 1 H), 4.70 (s, 2 H), 3.64
(m, 2 H), 3.53 (t, 2 H), 3.01 (t, 2 H) ppm. ESI-MS (m/z): (M+H4) 580.
[0375] EXAMPLE 23
[0376] This example describes the synthesis of

' which was prepared according to Scheme 15 and the procedure below. [0377] SCHEME 15

[0378] a) To a solution of Trit-Ser-Ome (compound 23.1, 10 mmol) and triethylamine in DCM (40 mL) was added slowly methanesulfhonyl chloride (11 mml), after 12 hours, the reaction was extracted with ether (100 mL), washed with water, dried over anhydrous magnesium sulfate and filtered. Concentration of the filtrate gave compound 23.2, which was used without further purification. [0379] b) Solution of the crude compound 23,2, sodium azide (20 mmol) in DMF was stirred for 15 hours. The reaction was extracted with ether (100 mL), washed with water, dried over anhydrous magnesium sulfate and filtered. The residue after concentration of the filtrate was purified by column, eluting with 0-20% ethyl acetate in hexane to give compound 23.3.
112

WO 2005/044817 PCT/US2004/036942
[0380] c) A mixture of 1 mmol of compound 23.3, 1.5 mmol of
cyclopropylacetylene, 0.02 mmol of Cul, and 0.02 mmol of Et3N in 6 mL of CH3CN
was stirred at room temperature overnight. The solvent was removed and the residue
was purified to give compound 23.4 in 65% yield. 1H NMR (400 MHz, CD3OD): 5
7.80 (s, 1H), 7.29-7.31 (m, 6H), 7.15-7.23 (m, 9H), 4.49 (m, 2H), 3.73 (m, 1H), 3.16
(s, 3H), 1.99 (m, 1H), 0.99 (m, 2H), 0.80 (m, 2H) ppm; ESI-MS (m/z): (M+H+)
453.15.
[0381] d) A mixture of 0.5 mmol of compound 23.4 in 2 mL of 4.0 N HC1 in
dioxane was stirred at room temperature for 1 hours. The solvent was removed and
the residue was diluted with 10 mL of water. The mixture was extracted with ether
for 3 rimes, and the aqueous phase was dried with lyophilizer to give compound 23.5
in quantitative yield. ESI-MS (m/z): (M+H+) 212.15.
[0382] e) Compound 23 was prepared according to Example 3g except that
compound 4.4 was used instead of compound 3.7 and compound 23.5 was used
instead of compound 3.4. !H NMR (400 MHz, CD3OD): 5 7.74 (s, 1H), 7.33 and
7.49 (m, 5H), 5.25 (m, 1H), 4.63-4.92 (m, 4H), 3.99 and 3.68 (m, 2H), 2.89 (m, 2H),
1.91 (m, 1H), 0.95 (m, 2H), 0.75 (m, 2H) ppm; ESI-MS (m/z): (M+H+) 562.10.
[0383] EXAMPLE 24
[0384] This example describes the synthesis of

which was prepared according to Scheme 16 and the procedure below.
113

WO 2005/044817 PCT/US2004/036942
[0385] SCHEME 16

[0386] a) To a solution of 50 mmol of l-bromo-3,5-difluorobenzene in 100 mL of dry DMF was added 50 mmol of NaSCH3 at 0 °C, the resulting mixture was stirred at room temperature overnight and treated with 10 mL of saturated aqueous NH4CI. The mixture was diluted with 1 L of water, extracted with hexane for several times. The extract was washed with waster and dried with anhydrous Na2SO4. The solvent was removed and the residue was purified to give compound 24.2 in 90% yield. 1H NMR (400 MHz, CD3C1): 5 7.15 (s, 1H), 7.02 (d, J= 8.3 Hz, 1H), 6.89 (d, J= 9.2 Hz, 1H), 2.50 (s, 3H) ppm.
[0387] b) A mixture of 40 mmol of compound 24.2 and 42 mmol of CuCN in 100 mL of dry DMF was stirred at 150 °C overnight. The mixture was diluted with 500 mL of water, extracted with ether for several times. The mixture was then washed with diluted aqueous NH4OH and water and dried with anhydrous Na2SO4. The solvent was removed; the residue was purified to give compound 24.3 in 50% yield. JH NMR (400 MHz, CD3C1): 5 7.28 (s, 1H), 7.17 (d, /= 9.2 Hz, 1H), 7.11 (d, J= 6.8 Hz, 1H), 2.53 (s, 3H) ppm; ESI-MS (m/z): (M+H+) 168.0. [0388] c) A mixture of 20 mmol of compound 24.3 and 22 mmol of KOH in 25 mL of EtOH and 35 mL of H2O was stirred at 60 °C for 30 minutes. The mixture was concentrated, the residue was diluted with 100 mL of water, extracted with EtOAc for several time. The extract was dried with anhydrous Na2SO4. The solvent was removed and the residue was dried in vacuo to give a crude product of compound 24.4. The crude was carried on the next step without further purification. ESI-MS (m/z): (M+H+) 187.0.
[0389] d) To a solution of 20 mmol of compound 24.4 in 60 mL of dry THF was added 24 mmol of LiAlHt (1.0 M in THF) at 0 °C. After stirring at room
114

WO 2005/044817 PCT/US2004/036942
temperature overnight, the reaction mixture was carefully added saturated aqueous
NH4CI. The resulting suspension was then concentrated. The residue was dissolved
in 200 mL of 1.0 N HC1 and extracted with EtOAc for several times. The extract
was dried with anhydrous Na2SO4 and concentrated. Subsequently, the residue was
purified to give 5-fluoro-3-methylmercapto-l-benzeyl alcohol in 81% yield. 1H
NMR (400 MHz, CD3C1): 5 7.03 (s, 1H), 6.86 (m, 2H), 4.69 (s, 2H), 2.51 (s, 3H)
ppm; ESI-MS (m/z): (M+H+) 173.1.
[0390] A mixture of 15 mmol of 5-imoro-3-methylmercapto-l-benzeyl alcohol
and 20 mmol of SOCI2 in 30 mL of dry CH2CI2 was refiuxed for several hours. The
mixture was then diluted with 100 mL of CH2CI2, washed with saturated aqueous
NaHCO3, saturated aqueous NH4CI, and brine and dried with anhydrous Na2SO4.
The solvent was removed and the residue was purified to give compound 24.5 in
85% yield.
[0391] e) Compound 24.6 was prepared according to Example 3a-c except that
compound 24.5 was used instead of 3-methylthiobenzyl chloride.
[0392] f) Compound 24 was prepared'according to Example 3g except that
compound 24.6 was used instead of compound 3.7. 1H NMR (400 MHz, CD3OD): 8
7.92 (s, 1H), 7.80 (s, 1H), 7.75 (d, J= 7.83 Hz, 1H), 7.74 (s, 1H), 7.59 (d, J= 134
Hz, 1H), 7.49 (d, J= 9.29 Hz, 1H), 7.07 and 7.35 (m, 2H), 6.96 (s, 1H), 5.10 (dd, J=
9.78, 4.40 Hz, 1H), 4.72 and 4.91 (m, 2H), 3.77 and 4.00 (m, 2H), 3.50 (dd, J=
14.18, 4.40 Hz, 1H), 3.20 (m, 1H), 2.91 (m, 2H) ppm; ESI-MS (m/z): (M+H4)
633.10.
[0393] EXAMPLE 25

which was prepared according to Scheme 17 and the procedure below.
115
[0394] This example describes the synthesis of

WO 2005/044817 PCT/US2004/036942
[0395] SCHEME 17

[0396] a) To a solution of 5.0 mmol of compound 3.2 (Example 3 a), 0.25 mmol
of Pd(PPh2)2Cl2, and 0.25 mmol of Cul in 15 mmol of degassed Et3N and 40 mL of
degassed toluene was added 5.5 mmol of isobutyryl chloride at 0 °C. The resulting
mixture was stirred at room temperature overnight and treated with 20 mL of
saturated aqueous NaHCO3. The organic layer was separated and dried with
anhydrous Na2SO4. The solvent was removed and the residue was purified by
column chromatography to give compound 25.1 in 80% yield. !H NMR (400 MHz,
CD3OD): 5 4.38 (m, 1H), 3.74 (s, 3H), 2.91 (m, 2H), 2.61 (m, 1H), 1.44 (s, 9H),
1.14 (d, J=6.85 Hz, 6H) ppm; ESI-MS (m/z): (M+H+) 320.01.
[0397] b) To a solution of 1.0 mmol of compound 25.1 in 3 mL of CH3OH was
added 1.0 mmol of NH2NH2 at 0 °C, the resulting mixture was stirred for another 30
minutes. The solvent was removed, and the residue was purified give compound
25.2 in 65% yield.
[0398] 1HNMR (400 MHz, CD3OD): 6.28 and 5.95 (s, s, 1H), 4.39 and 4.19 (m,
1H), 3.72 (s, 3H), 2.91 (m, 2H), 2.70 and 2.98 (m, 1H), 1.43 and 1.45 (s,s, 9H), 1.12
and 1.27 (m, 6H) ppm; ESI-MS (m/z): (M+H+) 312.20.
[0399] c) A mixture of 0.5 mmol of compound 25.2 in 4 mL of 4.0 N HC1 in
dioxane was stirred at room temperature for 12 hours. The solvent was removed and
the residue was dried in vacuo to give compound 25.3. ESI-MS (m/z): (M+H+)
212.10.
[0400] d) Compound 25 was prepared according to Example 3g except that
compound 4.4 was used instead of compound 3.7 and compound 25.3 was used
instead of compound 3.4.
116

WO 2005/044817 PCT/US2004/036942
[0401] 1H NMR (400 MHz, CD3OD): 7.15-7.54 (m, 5H), 6.38 (s, 1H), 5.06 (dd,
J= 9.78, 4.89 Hz, 1H), 4.66 and 4.88 (m, 2H), 4.01 and 3.71 (m, 2H), 3.41 (dd, J=
15.41, 4.65 Hz, 1H), 3.18 (dd, J= 15.16, 4.65 Hz, 1H), 3.07 (m, 1H), 2.92 (m, 2H),
1.32 (d, J= 7.34 Hz, 6H) ppm; ESI-MS (m/z): (M+H+) 563.10.
[0402] EXAMPLE 26
[0403] This example describes the synthesis of

which was synthesized according Scheme 18 and the procedure below. [0404] Scheme 18
117

WO 2005/044817 PCT/US2004/036942

a) A solution of compound 26.1 (12.4 g, 75 mmol) and NH4BF4 (10.5 g, 100 mmol) in water (85 mL) was treated with concentrated HC1 (15 mL), cooled to 3 °C, and treated dropwise over 25 minutes with a solution of NaNO2 (5.18 g, 75 mmol) in water (12 mL). The resulting thick slurry was stirred for 35 minutes, and the solid was collected by filtration, rinsed with water, methanol, and ether, and dried under N2. The solid was added in one portion to a stirred mixture of KOAc (8.1 g, 82.5 mmol) and 18-crown-6 (0.5 g, 1.9 mmol) in chloroform (170 mL). After 70 minutes, water (170 mL) was added, and the layers were separated. The aqueous phase was extracted with chloroform, and the combined organic layers were rinsed with water, dried, and concentrated. The residue was triturated with hexane and the
118

WO 2005/044817 PCT/US2004/036942
resulting solid isolated by filtration to provide 8.85 g (67% yield) of compound 26.2 as a dull yellow powder.1H NMR (CDC13) 5 3.96 (s, 3H), 7.80 - 7.85 (m, 2H), 8.14 (s, 1H), 8.27 (s, 1H); ES (+) MS m/e = 177(M+1).
b) A solution of compound 26.2 (5.0 g, 28.4 mmol) in THF (56 mL) was treated with LiOH (21 mL of a 2M aqueous solution, 42 mmol), and the reaction mixture is stirred at 50 °C. After 4 hours, the reaction mixture was cooled to room temperature and diluted with water. The basic aqueous layer was rinsed with diethyl ether, acidified to pH 3-4 by the addition of 1 M HC1, and extracted with ethyl acetate. The aqueous layer was extracted further with ethyl acetate, and the combined organic layers were rinsed with brine, dried over MgSO4, and concentrated to afford 4.0 g (87% yield) of compound 26.3. !H NMR (CD3OD) 5 7.79 - 7.87 (m, 2H), 8.14 (s, 1H), 8.29 (s, 1H); ES (+) MS m/e = 163(M+1).
c) A solution of compound 1.1 (7.5 g, 20.8 mmol) in DCM (30 mL) was treated
with TFA (10 mL). After 1 hour, the reaction mixture was concentrated to afford
7.8 g (100% yield) of compound 26.4. ES (+) MS m/e = 261(M+1).
d) A solution of compound 26.3 (7.8 g, 20.8 mmol), compound 26.4 (3.4 g,
20.8 mmol), 1-hydroxybenzotriazole hydrate ("HOBt", 3.5 g, 22.3 mmol), and
diisopropylethylamine ("DIEA",14 mL, 83.3 mmol) in DMF (100 mL) was treated
with EDCI (4.4 g, 22.3 mmol). After 2 h, the reaction mixture was treated with 1 M
HC1 and extracted with ethyl acetate. The combined organic extracts were rinsed
with NaHCO3 (sat'd), rinsed with brine, rinsed with water, dried over MgSO4, and
concentrated to afford 8.4 g (99% yield) of the title compound. ES (+) MS m/e =
404(M+l).
e) A solution of compound 26.5 (8.4 g, 20.8 mmol) in pyridine (70 mL) was
treated with lithium iodide (11.1 g, 83.1 mmol), and the reaction mixture was heated
to 100 °C. After 16 hours, the reaction mixture was cooled to room temperature and
diluted with 1 M NaOH (aq). The basic aqueous layer was rinsed with diethyl ether
to remove most of the pyridine. The aqueous portion was then carefully acidified
119

WO 2005/044817 PCT/US2004/036942
with concentrated HC1 to pH 3-4. The resulting slurry was filtered. The precipitate was collected and dissolved in THF, while the filtrate was extracted with ethyl acetate. The THF and ethyl acetate solutions were combined, rinsed with brine, dried over MgSO4, and concentrated to afford 7.1 g (88% yield) of compound 26.6. ES(+)MSm/e = 390(M+l).
f) A solution of compound 26.6 (3.06 g, 7.83 mmol) and DIEA (4.6 mL, 25.4 mmol) in dimethylformamide ("DMF") was treated with HATU (3.06 g, 8.06 mmol), and the resulting mixture was stirred at room temperature. After 20 minutes, the reaction mixture was treated sequentially with HCl\H-DAP(Boc)-OMe (2.18 g, 8.59 mmol) and tyiV-dimethylaminopyridine ("DMAP", 0.568 g, 4.65 mmol). After 2.5 hours, the reaction was diluted with ethyl acetate, washed with three portions of water, washed with one portion of brine, dried over MgSO4, and concentrated. Flash column chromatography afforded 3.91 g (84% yield) of compound 26.7. 1H NMR (400 MHz, chloroform-d) 5: 1.42 (s, 9H), 2.81 (s, 2H), 3.70 (m, 2H), 3.75 (2H), 3.81 (s, 3H), 4.82 (m, 21H), 4.99 (m, 1H), 7.21 (d, 2H), 7.59 (s, 1H), 7.81 (d, 1H), 8.10 (s, 1H). MS (API-ES+) m/z: 590.2 (M+H+), 534.1(M-tButyl+H4), 490.1(M-Boc+H+).
g) A solution of compound 26.7 (3.91 g, 6.62 mmol) in DCM was treated with HC1 (8.3 mL of a 4 M dioxane soln, 33.2 mmol), and the resulting mixture was stirred at room temperature. After 2 hours, the reaction mixture was concentrated to afford the HC1 salt, which was used without further purification. The HC1 salt (3.94 g, 6.99 mmol) and triethylamine ("TEA", 3.0 mL, 21.5 mmol) in methanol was treated with A'-cyanoirnido-5)Sr-dunethyl-dithiocarbonate (1.37 g, 8.43 mmol), and the reaction mixture was stirred at 50 °C. After 3.5 hours, the reaction mixture was concentrated to remove most of the methanol, diluted with ethyl acetate, washed with two portions of water, washed with one portion of brine, dried over MgSC>4, and concentrated. Flash column chromatography afforded 3.27 g (80% yield) of compound 26.8 (MS (API-ES*) m/z: 5%%2QA+Yt). A solution of compound 26.8 (0.10 mmol) was made in 4:1 methanol/dichloroethane ("DCE", 2.5 mL) and treated sequentially with 2 M methanolic ammonia (0.25 mmol) and silver nitrate (0.10
120

WO 2005/044817 PCT/US2004/036942
mmol). The reaction mixture was stiired at 50 °C until complete conversion is observed through monitoring by LCMS. The reaction mixture was then filtered through celite, and KOH (0.1 mL of a 2 M methanolic soln, 0.2 mmol) was added. The reaction mixture was stirred once again at 50 °C. After 2-4 hours the reaction mixture was directly subjected to preparatory HPLC purification to afford compound 26.
[0405] EXAMPLE 27
[0406] This example describes the synthesis of

where RA and RB are each independently hydrogen, aliphatic, aromatic, heteroaromatic, or together form a cyclic moiety. These compounds are made according to the procedure of Example 26 except that a substituted amine of the formula HNRARB is used instead of ammonia in step g. Illustrative examples of substituted amines and the resulting compounds are shown in Table 1.
121
TABLE 1


122
WO 2005/044817 PCT/US2004/036942


WO 2005/044817

PCT/US2004/036942


[0407] EXAMPLE 28
[0408] This example describes the synthesis of

which is made according to Scheme 19 and the procedure below.
123

WO 2005/044817 PCT/US2004/036942
[0409] SCHEME 19

a) Compound 28.4 is prepared from 28.1 in three chemical steps, following the
procedure published in Okada, T. et al Chem. Phar. Bull. 1993,41(1), 126-131;
Frigola, J. et all Med. Chem. 1993,36(7), 801-810 as shown above. A solution of
commercially available 28.1 in DCM is treated with trifluoromethanesulfonyl
chloride in the presence of base to provide 36.2. Next, this product is dissolved in
dimethoxyethane ("DME"), and the mesylate moiety is displaced by dimethylamine.
Finally, Pd/C-catalyzed hydrogenolysis in MeOH at 45 PSIH2 (g) affords
compound 28.4.
b) Compound 28 is synthesized according to the procedure of Example 26
except 3-(dimethylamino)cyclobutanol (35.1) is used instead of ammonia in step g.
[0410] EXAMPLE 29
[0411] This example describes the synthesis of

which is made according to Scheme 20 and the procedure below. [0412] SCHEME 21

a) To a solution of benzyl-3-pyrroline-l-carboxylate (compound 29.1,10 mmol) in THF (15 mL) was added N-methyl morphline (22 mmol) and OsO4 (2 mL of a 2.5 wt% in t-BuOH), and the resulting mixture was stirred at room temperature overnight. The solvent was removed; the residue was dissolved in EtOAc (100 mL),
124

WO 2005/044817 PCT/US2004/036942
washed with dilute aq. Na2SO3, sat. aq. NH4CI, and brine, and dried with anhydrousNa2SO4. The solvent was removed and the residue was purified by column chromatography to give compound 29.2 in 55% yield. EIMS (m/z): calcd. for Ci2H15NO4 (M+)+Na 260.1, found 260.1; 1HNMR (CD3OD, 400MHz): 6 7.31-7.38 (m, 5H), 5.13 (s, 2H), 4.17 (m, 2H), 3.58 (m, 2H), 3.34 (m, 2H) ppm.
b) A mixture of compound 29.2 (1.0 mmol) and 10% Pd/C (0.1 mmol) in
methanol (5 mL) is stirred at room temperature for several hours under an
atmosphere of H2. The reaction mixture is filtered, the filtrate is concentrated, and
the residue is dried in vacuo to give compound 29.3.
c) Compound 29 is synthesized according to the procedure of Example 26
except that (3R, 4S)-(dihydroxy)pyrrolidine (29.3) is used instead of ammonia in
step g.
[0413] EXAMPLE 30
[0414] This example describes the synthesis of

which was prepared according to Scheme 21 and the procedure below. [0415] SCHEME 21
a) A mixture of (3R, 4R)-benzyl-3,4-pyrrolidindiol (compound 30.1,1 mmol) and 20% Pd(OH)2/C (0.1 mmol) in methanol (10 mL) is shaken at room temperature for several hours under 45 psi of H2. (g). The reaction mixture is filtered, the filtrate is concentrated, and the residue is dried in vacuo to give compound 30.2.
125

WO 2005/044817 PCT/US2004/036942
b) Compound 30 is synthesized according to the procedure of Example 26 except that compound 30.2 is used instead of ammonia in step g.
[0416] EXAMPLE 31
[0417] This example describes the synthesis of

which is prepared according to Scheme 22 and the procedure below. [0418] SCHEME 22
a) A mixture of commercially available benzyl 3-pyrroline-l-carboxylate (compound 28.1,10 mmol) and m-CPBA (12 mmol) in DCM (50 mL) was stirred at room temperature overnight. The reaction mixture was diluted with DCM (100 mL) and washed sequentially with sat aq. Na2SO3, and brine. The organic layer was dried with anhydrous Na2SO4 and then concentrated. The residue was purified by chromatography to give compound 31.1 in 80% yield. EIMS (m/z): calcd. for C12H13NO3 (M+)+Na242.1, found242.1; 1HNMR (CDCI3,400MHz): 6 7.36-7.37 (m, 5H), 5.13 (s, 2H), 3.89 (m, 2H), 3.70 (m, 2H), 3.41 (m, 2H) ppm.
126

WO 2005/044817 PCT/US2004/036942
b) A mixture of compound 31.1 (5 mmol) in cone. aq. NH3 (20 mL) is stirred at
65°C overnight. The reaction mixture was concentrated and dried in vacuo to give
compound 31.2. This material is used without further purification.
c) A solution of compound 31.2 (10 mmol) and Et3N (20 mmol) in dry THF
(100 mL) at -20° C is treated dropwise with TFAA (10 mmol) over 1 hour. After 1
h, the reaction mixture is quenched with sat. aq. NH4CI (1 mL). The solvent is
removed and the residue is dissolved in DCM (100 mL). The mixture is
subsequently washed sequentially with sat. aq. NH4CI, sat. aq. NaHCOs, and brine.
The organic layer is dried with anhydrous Na2SO4, the solvent was removed, and the
residue is purified by chromatography to afford compound 31.3.
d) A solution of compound 31.3 (5 mmol) and Et3N (10 mmol) dry DCM (20
mL) at 0°C is treated dropwise with MsCl (5.5 mmol), and the mixture is allowed to
come gradually to room temperature. After 1 hour at room temperature, the reaction
mixture containing in situ generated 31.4 is treated with DBU (30 mmol), and the
resulting mixture is stirred at for several hours. The solvent is removed, and the
residue is purified by chromatography to afford compound 31.5.

e) A mixture of compound 31.5 (3 mmol) and K2CO3 (6 mmol) in 2/1 (v/v)
MeOH/H2O (15 mL) is stirred at room temperature. After 24 h, the solvent is
removed; the residue is treated with sat aq. NaHCO3 (20 mL), and the mixture is
extracted with DCM several times. The extract is dried withNa2CO3., the solvent is
removed, and the residue is purified by chromatography to afford compound 31.6.
f) A mixture of compound 31.6 (2 mmol), NaHCO3 (3 mmol), and Boc2O (2.2
mmol) in 1:1 1,4-dioxane/water (20 mL) is stirred at room temperature for several
hours. The mixture is diluted with brine (50 mL) and extracted with EtOAc several
times. The combined extracts are washed with brine, dried with anhydrous Na2SO4,
and concentrated. The residue is purified by chromatography to give the N-Boc
protected intermediate. A mixture of this intermediate (1 mmol) and 10% Pd/C (0.1
mmol) in MeOH (5 mL) is stirred at room temperature for several hours under an
127

WO 2005/044817 PCT/US2004/036942
atmosphere of H2 (g). The reaction mixture is filtered, the filtrate is concentrated, and the residue is dried in vacuo to afford compound 31.7.
g) Compound 31 is prepared according to Example 26g except that compound 31.7 is used instead of ammonia.
[0419] EXAMPLE 32
[04201 This example describes the synthesis of


which is prepared according to Scheme 23 and the procedure below. [0421] SCHEME 23

a) A mixture of compound 31.2 (2 mmol), NaHCO3 (3 mmol), and Boc2O (2.2
mmol) in 1:11,4-dioxane/water (20 mL) is stirred at room temperature for several
hours. The mixture is diluted with brine (50 mL) and extracted with EtOAc several
times. The combined extracts are washed with brine, dried with anhydrous Na2SO4,
and concentrated. The residue is purified by chromatography to afford compound
32.3.
b) A mixture of compound 323 (1 mmol) and 10% Pd/C (0.1 mmol) in MeOH
(5 mL) is stirred at room temperature for several hours under an atmosphere of H2
(g). The reaction mixture is filtered, the filtrate is concentrated, and the residue is
dried in vacuo to afford compound 32.4.
128

WO 2005/044817 PCT/US2004/036942
c) Compound 32 is prepared according to Example 26g except that compound 32.4 is used instead of ammonia.
[0422] EXAMPLE 33
[0423] This example describes the synthesis of

which is prepared according to Scheme 24 and the procedure below. [0424] SCHEME 24
a) A solution of compound 33.1 (10 mmol) and DIEA (25 mmol) in DCM (20
mL) at 0 °C is treated dropwise with benzyl chloroformate (10 mmol), and the
reaction mixture is allowed to come to room temperature. After 2 h at RT, the
reaction mixture is diluted with ethyl acetate (100 mL), rinsed with 1 M HC1 (50
mL), rinsed with brine, dried over MgSO4, and concentrated to afford compound
33.2.
b) A solution of compound 33.2 (10 mmol) and N-methyl morpholine (22
mmol) in THF (15 mL) is treated with OSO4 (2 mL of a 2.5 wt% in /-BuOH), and
the resulting mixture is stirred at room temperature overnight. The solvent is
removed. The residue is dissolved in EtOAc (100 mL), washed with dilute aq.
Na2SO3, sat. aq. NH4CI, and brine, and then dried over anhydrous Na2SO4. The
solvent is removed, and the residue is purified by column chromatography to give
the title compound.
c) A mixture of compound 33.3 (1.0 mmol) and 10% Pd/C (0.1 mmol) in
methanol (5 mL) is stirred at room temperature for several hours under an
129

WO 2005/044817 PCT/US2004/036942
atmosphere of H2. The reaction mixture is filtered, the filtrate is concentrated, and the residue is dried in vacuo to give compound 33.4.
d) Compound 33 is prepared according to Example 26g except that compound 33.4 is used instead of ammonia.
[0425] EXAMPLE 34
[0426] This example describes the synthesis of

which is prepared according to Scheme 25 and the procedure below. [0427] SCHEME 25

a) A mixture of Tl(OAc) (17.6 g, 54.5 mmol) in dried acetic acid (40 mL) is
retluxed with stirring for 1 hour and then cooled to room temperature. Compound
32.2 (34.6 mmol) and iodine (8.46 g, 33.3 mmol) are added, and the resulting
suspension is heated to reflux. After 9 hours, the reaction mixture is cooled to room
temperature, and the yellow TII precipitate is removed by filtration with ether rinses.
The filtrate is concentrated, and the residue is dissolved in ethyl acetate, dried over
MgSC«4, and reconcentrated to afford compound 34.1.
b) A mixture of 34.1 (1.0 mmol) and 10% Pd/C (0.1 mmol) in methanol (5 mL)
is stirred at room temperature for several hours under an atmosphere of H2. The
reaction mixture is filtered, the filtrate is concentrated, and the residue is dried in
vacuo to give compound 34.2.
130

WO 2005/044817 PCT/US2004/036942
c) Compound 34 is prepared according to Example 26g except that compound 34.2 is used instead of ammonia.
[0428] EXAMPLE 35
[0429] This example describes the synthesis of

which is prepared according to Scheme 26 and the procedure below. [0430] SCHEME 26

a) A mixture of compound 35.1 (10 mmol) in ethanol (20 mL) is saturated with HC1 (g) and stirred at room temperature overnight. The solvent is removed, the residue is dissolved in DCM (100 mL), the resulting solution is treated sequentially with TEA (30 mmol) and BnBr (11 mmol), and the reaction mixture is heated to reflux. After 12 h, the reaction mixture is cooled to room temperature and concentrated. The residue is dissolved in EtOAc (150 mL), washed with brine, and dried over anhydrous Na2SO4. The solvent is removed and the residue is purified by chromatography to afford compound 35.2.
131

WO 2005/044817 PCT/US2004/036942
b) A solution of L1BH4 (20 mmol) in dry THF (20 mL) at room temperature is
treated in a dropwise fashion with a solution of compound 35.2 (5 mmol) in THF (5
mL). After stirring overnight, the reaction mixture is quenched by adding several
drops of water. The mixture is concentrated, diluted with brine (50 mL), and
extracted several times with 9/1 (v/v) EtOAc/z-PrOH. The combined extracts are
dried over anhydrous Na2SO4, the solvent is removed, and the residue is purified by
chromatography to afford compound 35.3.
c) A solution of compound 35.3 (2 mmol) and TEA (2.2 mmol) in dry THF (10
mL) at -78°C is treated dropwise with trifluoroacetic anhydride ("TFAA", 2.2
mmol). After several hours, the mixture is treated with TEA (6 mmol), and the
reaction mixture is heated to reflux. The mixture then is concentrated, and the
residue is dissolved in THF (10 mL) and treated with water (2.5 mL). This mixture
is treated with NaOH (10 mmol) with vigorous stirring at room temperature for
several hours. The solvent is removed, and the residue is treated with sat aq.
NaHCO3 (20 mL) and extracted several times with 9/1 (v/v) EtOAc/z-PrOH. The
combined extracts are dried over anhydrous Na2SO4, the solvent is removed, and the
residue is purified by chromatography to afford compound 35.4.

d) A mixture of compound 35.4 (1 mmol) and 20% Pd(OH)2/C (0.1 mmol) in
methanol (10 mL) is shaken at room temperature for several hours under 45 psi of
H2. (g). The reaction mixture is filtered, the filtrate is concentrated, and the residue
is dried in vacuo to give compound 35.5.,
e) Compound 35 is prepared according to Example 26g except that compound
35.5 is used instead of ammonia.
[0431] EXAMPLE 36
[0432] This example describes the synthesis of
132

WO 2005/044817 PCT/US2004/036942

which was prepared according to Example 35 except that (2R, 4S)-4-hydroxyproline (trans-D-Hyp-OH) was used instead of compound 35.1.
[0433] EXAMPLE 37
[0434] This example describes the synthesis of

which was prepared according to Example 35 except that (2S, 4R)-4-hydroxyproline (trans-L-Hyp-OH) was used instead of compound 35.1.
[0435] EXAMPLE 38
[0436] This example describes the synthesis of

which is prepared according to Scheme 27 and the procedure below. [0437] SCHEME 27
133

WO 2005/044817 PCT/US2004/036942

a) A solution of HCl-H-DAP(Boc)-OMe (10 mmol) and DIEA (11 mmol) in
DCM (50 mL) at 0 °C is treated dropwise with benzoyl chloride (11 mmol). Over 3
h the reaction mixture is allowed to warm to room temperature. The reaction
mixture is concentrated, and the residue is purified by chromatography. This
intermediate is treated with a solution of 4 M HC1 in dioxane, and the resulting
mixture is stirred at room temperature. After 1 h, the solvent is removed to afford
compound 38.1, which is used without further purification.
b) A solution of compound 38.1 (10 mmol) and DIEA (12 mmol) in DCM (50
mL) at 0 °C is treated dropwise with p-nitrophenyl chloroformate (11 mmol), and
the reaction mixture is allowed to warm to room temperature. After 2 hours, the
reaction mixture is concentrated, and the residue is purified by chromatography to
afford compound 38.2.
c) A solution of compound 38.2 (10 mmol) and TEA (25 mmol) in 1:1
DCE/DMF (10 mL) is treated with 2 M methanolic ammonia (18 mmol), and the
mixture is heated to 40 °C. After 15 hours, the reaction mixture is concentrated, and
the residue is purified by chromatography to afford compound 38.3.
134

WO 2005/044817 PCT/US2004/036942
d) A mixture of compound 38.3 (1.0 mmol) and 10% Pd/C (0.1 mmol) in
methanol (10 mL) is shaken at room temperature for several hours under 45 psi of
H2. (g). The reaction mixture is filtered, the filtrate is concentrated, and the residue
is dried in vacuo to give compound 38.4.
e) A solution of compound 26.6 (8.0 mmol) and DIEA (25. mmol) in DMF is
treated with HATU (8.0 mmol), and the resulting mixture was stirred at room
temperature. After 20 minutes, the reaction mixture is treated sequentially with
compound 38.4 (8.6 mmol) and DMAP (0.5 mmol), and the mixture is then heated
to 60 °C. After 2.5 hours, the reaction is diluted with ethyl acetate, washed with
three portions of water, washed with one portion of brine, dried over MgSO4, and
concentrated. Flash column chromatography affords compound 38.5.
f) A solution of compound 38.5 (0.15 mmol) in methanol (1 mL) was treated
with 2 M methanolic KOH (0.45 mmol), and the reaction mixture is heated to 50 °C.
After 3 hours, the reaction mixture is concentrated to dryness and the residue is
subjected to preparatory HPLC purification to afford compound 38.
[0438] EXAMPLE 39
[0439] This example describes the synthesis of

where RA and RB are each independently hydrogen, aliphatic, aromatic, heteroaromatic, or together form a cyclic moiety. These compounds are made according to the procedure in Example 38 except that a substituted arnine of the formula HNRARB is used instead of ammonia in step c. Illustrative examples of substituted amines and the resulting compounds are shown in Table 2.
135

WO 2005/044817

PCT/US2004/036942

TABLE 2

136

WO 2005/044817

PCT/US2004/036942


137

WO 2005/044817

PCT/US2004/036942


[0440] EXAMPLE 40
[0441] This example describes the synthesis of

which is prepared according to Scheme 28 and the procedure below.
[0442] SCHEME 28
138

WO 2005/044817 PCT/US2004/036942

a) Commercially available compound 40.1 (10 mmol) in THF (50 mL) is treated with sodium hydrosulfite (50 mmol) in water (20 mL). After 8 hours at room temperature, the reaction is extracted with ethyl acetate (100 mL), and the organic extract is washed with water and brine, dried over anhydrous magnesium sulfate and filtered to give the crude compound 40.2.
b) To a slurry of compound 40.2 (10 mmol), ammonium tetrafluoroborate (12.5 mmol) in water (12 mL) is added concentrated HC1 (2 mL). The reaction is cooled to 0 °C, and to it is added sodium nitrite (10 mmol). After the reaction is stirred for 1 hour at 0 °C, the solid is collected by filtration, rinsed with methol, ether and dried under vacuum. The resulting solid is added to a stirring solution of HO Ac (3 mL), 18-crown-6 (0.3 mmol) in chloroform (20 mL). After 1 hour, water (10 mL) and DCM (20 mL) are added. The organic layer is separated, dried by magnesium sulfate and filtered. The residue after concentration of the filtrate is triturated with
hexane to give product 40.3.
139

WO 2005/044817 PCT/US2004/036942
c) Compound 40.4 was made according to the procedure for the preparation of
compound 6.1 except that i?-3-(+)-pyrrolidinol was used instead of pyrrolidine.
d) Compound 40.4 is desolved in DCM, and is treated with triethylamine (1.5
eq) and acetic anhydride (1.2 eq). The resulting solution is filtered through silica
gel, concentrated. The residue is then purified by silica gel column chromagraphy to
yield compound 40.5.
e) Compound 40.5 in DCM is treated with anhydrous 4 N HC1 in dioxane (2.0
eq). After starting material disappears, the reaction is concentrated to give
compound 40.6.
f) Compound 40.7 is made according to the procedure for the preparation of
compound 1.5 except that compound 40.6 is used instead of 1.3.
g) Compound 40.8 is made according to the procedure for the preparation of
compound 1.6 except that compounds 40.7 and 40.3 are used instead of compounds
1.5 and 1.11.
h) Compound 40 is made according to the procedure for the preparation of compound 1 except that compound 40.8 is used instead of 1.6.
[0443] EXAMPLE 41

which is prepared according to Scheme 29 and the procedure below.
140
[0444] This example describes the synthesis of

WO 2005/044817 PCT/US2004/036942
[0445] SCHEME 29
i
Compound 41 is made according to the procedure for the preparation of compound 3 except that compounds 18.10 and 40.6 are used instead of 3.4 and 3.7. The enantiomerically pure compounds are isolated using chiral column chromatography.
[0446] EXAMPLE 42
[0447] This example describes the synthesis of

which is prepared according to Scheme 30 and the procedure below. [0448] SCHEME 30

a) A solution of compound 18.7 (10 mmol), 1-ethoxy-l-ethenyltributyltin (10.5 mmol), Pd(PPh)4 (0.5 mmol) in dimethoxyethane (DME, 50 mL) is heated at 80 °C until compound 18.7 disappears. The reaction is cooled to room temperature, and to
141

WO 2005/044817 PCT/US2004/036942
it is added 4N aqueous HC1 (5 mL). The reaction is stirred for 3 hours and extracted with ether (80 mL). The organic extract is washed with brine, dried with anhydrous magnesium sulfate, filtered and concentrated. The residue is purified by column chromatography to give compound 42.1.
b) A solution of compound 42.1 in THF at -78 °C is treated with LDA (2.0 eq).
After 1 hour, a solution of compound 18.2 (1.0 eq) in THF is added to the dry ice
cooled reaction. After another 3 hours, saturated aqueous NH4CI is added to the
reaction and the mixture is allowed to warm to room temperature. The reaction
mixture is partitioned between ethyl acetate and water, and the organic layer is
washed with water and brine, dried with anhydrous magnesium sulfate and filtered.
The residue after concentration of the filtrate is purified by silica gel column to give
compound 42.2.
c) A solution of compound 42.2 in ethanol is treated with sodium borohydride
(2.0 eq). After 1 hour, the reaction mixture is partitioned between ethyl acetate and
water, and the organic layer is washed brine,, dried with anhydrous magnesium
sulfate and filtered. The residue after concentration of the filtrate is purified by
silica gel column to give compound 42.3.

d) A mixture of 42.3 Lil (3 eq) in pyridine is reflux overnight. The solvent is
removed and the residue is dissolved in EtOAc. The resulting solution is then
washed with saturated aqueous NH4CI and dried with anhydrous Na2SC>4. The
solvent is removed and the residue is dried in vacuo to give a quantitative yield of
compound 42.4. The crude product was carried on the next step without further
purification.
e) Compound 42 is made according to the procedure for the preparation of
compound 3 except that compounds 42.4 and 40.6 are used instead of 3.4 and 3.7.
The enantiomerically pure compounds are isolated using chiral column
chromatography.
142

WO 2005/044817

PCT/US2004/036942

[0449] EXAMPLE 43
[0450] This example describes the synthesis of

which is prepared according to Scheme 31 and the procedure below. [0451] SCHEME 31

a) A solution of compound 18.2 in ethanol is treated with hydroxylamine (1.05
eq). After 10 hours, the reaction is concentrated and the residue is dried under
vacuum to give compound 43.1.
b) A solution of 43.1 is hydrogenated with 20% Pd(OH)2/C as a catalyst at 45
psi of hydrogen to give compound 43.2.
c) A solution of commercially available 43.3 in carbon tetrachloride is treated
with JV-chlorosuccinimide (NCS, 3 eq). The reaction mixture is then diluted with
ethyl acetate, washed with IN NaOH, water and brine, dried over anhydrous MgSC>4
and filtered. The crude product is recrystalized from hot ethanol to give compound
43.4.
143

WO 2005/044817 PCT/US2004/036942
d) A solution of 43.4 in THF is treated with LiOH (2 eq, 2.0 N aqueous). After
most starting material is consumed, the mixture is then diluted with ethyl acetate,
washed with saturated ammonium chloride, water and brine, dried over anhydrous
MgSO4 and filtered. The crude product is recrystalized from hot ethanol to give
compound 43.5.
e) A solution of 43.5,43.2 (1.1 eq) and EDC (1.0 eq) in DMF is stirred for 10
hours at room temperature. The mixture is then diluted with ethyl acetate, washed
with saturated ammonium chloride, water and brine, dried over anhydrous MgSO4
and filtered. The crude product is then refluxed with formic acid until LC-MS
indicates the reaction is completed. Solvent is evaporated, and the crude product is
purified by silica gel column to give compound 43.6.
f) A mixture of 43.6 Lil (3 eq) in pyridine is reflux overnight. The solvent is
removed and the residue is dissolved in EtOAc. The resulting solution is then
washed with saturated aqueous NH4CI and dried with anhydrous Na2SC>4. The
solvent is removed and the residue is dried in vacuo to give a quantitative yield of
compound 43.7. The crude product was carried on the next step without further
purification.
g) Compound 43 is made according to the procedure for the preparation of
compound 3 except that compounds 43.7 and 40.6 are used instead of 3.4 and 3.7.
[0452] Diversification:
[0453] It will also be appreciated that each of the components used in the synthesis of inventive compounds can be diversified either before synthesis or alternatively after the construction of the core structure of formula (I). As used herein, the term "diversifying" or "diversify" means reacting an inventive compound (I) or any of the precursor fragments (or any classes or subclasses thereof) at one or more reactive sites to modify a functional moiety or to add a functional moiety {e.g., nucleophilic addition of a substrate). Described generally herein are a variety of
144

WO 2005/044817 PCT/US2004/036942
schemes to assist the reader in the synthesis of a variety of compounds, either by diversification of the intermediate components or by diversification of the core structures as described herein, and classes and subclasses thereof. It will be appreciated that a variety of diversification reactions can be employed to generate compounds other than those described in the Exemplification herein. As but a few examples, where a double bond is present in the compound structure, epoxidation and aziridation can be conducted to generate epoxide and aziridine derivatives of compounds described herein. For additional guidance available in the art, the practitioner is directed to "Advanced Organic Chemistry", March, J. John Wiley & Sons, 2001, 5th ed., the entire contents of which are hereby incorporated by reference.
[0454] 2) Biological Data:
[0455] As discussed above, LFA-ICAM interactions have been directly implicated in numerous inflammatory disease states including, but not limited to graft rejection, dermatitis, psoriasis, asthma and rheumatoid arthritis. Thus, compounds capable of modulating adhesion between intracellular adhesion molecules (e.g., ICAM-1, -2 and -3) and the leukocyte integrin family of receptors would be useful in the development of novel therapeutics. Described below are certain assays used for the determination of ICAM-1 :LFA Receptor binding, Human T-Cell Adhesion, and T-Cell proliferation which are described in published PCT applications WO 99/49856 and WO 02/05114, the entire contents of which are hereby incorporated by reference. WO 99/49856 also describes the preparation and purification of full-length LFA-1 from 293 cells, the preparation of a plasmid for expression of a human ICAM-1 immunoadhesion, and the generation of ICAM-1 immunoadhesion expressing 293 cell line.
[0456] ICAM-1:LFA Receptor Binding Assay (protein/protein assay); [0457] Competitive inhibition of the GDI 1 a/CD 18-ICAM-l interaction is quantitated by adding known amounts of inhibitors according to the two protein/protein assay systems described below: [0458] Forward Format LFA-1 :ICAM-1 Assay (PPFF):
145

WO 2005/044817 PCT/US2004/036942
[0459] Purified full length recombinant human LFA-1 protein is diluted to 2.5 Hg/ml in 0.02 M Hepes, 0.15M NaCl, and 1 mM MnCl2 and 96-well plates (50 ul/well) are coated overnight at 4°C. The plates are washed with wash buffer (0.05% Tween in PBS) and blocked for lh at room temperature with 1% BSA in 0.02M Hepes, 0.15 M NaCl, and 1 mM MnCl2. Plates are washed. 50 ul/well inhibitors, appropriately diluted in assay buffer (0.5% BSA in 0.02M Hepes, 0.15M NaCl, and 1 mM MnCb), are added to a 2X final concentration and incubated for lh at room temperature. 50 ul/well of purified recombinant human 5 domain ICAM-Ig, diluted to 50ng/ml in assay buffer, is added and incubated 2h at room temperature. Plates are washed and bound ICAM-Ig is detected with Goat anti-HuIgG(Fc)-HRP for lh at room temperature. Plates are washed and developed with 100 ul/well TMB substrate for 10-30' at room temperature. Colorimetric development is stopped with 100 ul/well 1M H2PO4 and read at 450 nM on aplatereader.
[0460] An alternative protein/protein assay system below also quantitates competitive inhibition of the CD 11 a/CD 18-ICAM-1 interaction. [0461] PLM2 Antibody Capture LFA-1 :ICAM-1 Assay (PLM2) [0462] A non-function blocking monoclonal antibody against human CD18, PLM-2 (as described by Hildreth, et at, Molecular Immunology, Vol. 26, No. 9, pp. 883-895, 1989), is diluted to 5 u.g/ml in PBS and 96-well flat-bottomed plates are coated with 100 ^I/well overnight at 4°C. The plates are blocked with 0.5% BSA in assay buffer (0.02 M Hepes, 0.15 M NaCl, and 1 mM MnCl2) 1 h at room temperature. Plates are washed with 50 mM Tris pH 7.5,0.;1M NaCl, 0.05% Tween 20 and 1 mM MnCb- Purified full-length recombinant human LFA-1 protein is diluted to 2 u-g/ml in assay buffer and 100 ml/well is added to plates and incubated at 1 h at 37°C. Plates are washed 3X. 50 ul/well inhibitors, appropriately diluted in assay buffer, are added to a 2X final concentration and incubated for 30' at 37°C. 50 fil/well of purified recombinant human 5 domain ICAM-Ig, diluted to 161 ng/ml (for a final concentration of 80 ng/ml) in assay buffer, is added and incubated 2 h at 37°C. Plates are washed and bound ICAM-Ig is detected with Goat anti-HuIgG(Fc)-HRP for lh at room temperature. Plates are washed and developed with 100 ul/well
146

WO 2005/044817 PCT/US2004/036942
TMB substrate for 5-10' at room temperature. Colorimetric development is stopped with 100 pi/well 1M H3PO4 and read at 450 nM on a platereader.
[0463] Human T-Cell Adhesion Assay (cell attachment assay) [0464] The T-cell adhesion assay is performed using a human T-lymphoid cell line HuT 78. Goat anti-Hu!gG(Fc) is diluted to 2 ug/ml in PBS and 96-well plates are coated with 50 ul/well at 37°C for 1 h. Plates are washed with PBS and blocked for 1 h at room temperature with 1% BSA in PBS. 5 domain ICAM-Ig is diluted to 100 ng/ml in PBS and 50 pi/well was added to the plates O/N at 4°C. HuT 78 cells are centrifuged at 100 g and the cell pellet is treated with 5 mM EDTA for ~ 5' at 37°C in a 5% CO2 incubator. Cells are washed in 0.14 M NaCl, 0.02 M Hepes, 0.2% glucose and 0.1 mM MhCh (assay buffer) and centrifuged. The cells are resuspended in assay buffer to 3.0 x 106c/ml. Inhibitors are diluted in assay buffer to a 2X final concentration and pre-incubated with HuT78 cells for 30' at room temperature. 100 ul/well of cells and inhibitors are added to the plates and incubated at room temperature for 1 h. 100 ul/well PBS is added and the plates are sealed and centrifuged inverted at 100 g for 5'. Unattached cells are flicked out of the plate and excess PBS is blotted on a paper towel. 60 ul/well p-nitrophenyl n-acetyl-p-D-glucosaminide (0.257 g to 100 ml citrate buffer) is added to the plate and incubated for 1.5 h at 37°C. The enzyme reaction is stopped with 90ul/well 50 mM glycine/5mM EDTA and read on a platereader at 405 nM. HUT 78 cell adhesion to 5dICAM-Ig is measured using the p-nitrophenyl n-acetyl-p-D-glucosaminide method of Landegren, U. (1984). J. Immunol. Methods 57,379-388. [0465] T-Cell Proliferation Assay:
[0466] This assay is an in vitro model of lymphocyte proliferation resulting from activation, induced by engagement of the T-cell receptor and LFA-1, upon interaction with antigen presenting cells (Springer, Nature 346:425 (1990)). [0467] Microtiter plates (Nunc 96 well ELISA certified) are pre-coated overnight at 4°C with 50 ul of 2 ug/ml of goat anti-human Fc(Caltag H10700) and 50 ul of 0.07 ug/ml monoclonal antibody to CD3 (Immunotech 0178) in sterile PBS. The next day coat solutions are aspirated. Plates are then washed twice with PBS
147

WO 2005/044817 PCT/US2004/036942
and 100 ul of 17 ng/ml 5d-ICAM-l-IgG is added for 4 hours at 37°C. Plates are washed twice with PBS prior to addition of CD4+ T cells. Lymphocytes from peripheral blood are separated from heparinized whole blood drawn from healthy donors. An alternative method is to obtain whole blood from healthy donors through leukophoresis. Blood is diluted 1:1 with saline, layered and centrifuged at 2500 x g for 30 minutes on LSM (6.2 g Ficoll and 9.4 g sodium diztrizoate per 100 ml) (Organon Technica, NJ). Monocytes are depleted using a myeloid cell depletion reagent method (Myeloclear, Cedarlane Labs, Hornby, Ontario, Canada). PBLs are resuspended in 90% heat-inactivated Fetal Bovine serum and 10% DMSO, aliquoted, and stored in liquid nitrogen. After thawing, cells are resuspended in RPMI 1640 medium (Gibco, Grand Island, NY) supplemented with 10% heat-inactivated Fetal Bovine serum (Intergen,. Purchase, NY), lmM sodium pyruvate, 3 mM L-glutamine, lmM nonessential amino acids, 500 |ig/ml penicillin, 50 ng/ml streptomycin, 50 u,g/ml gentamycin (Gibco).
[0468] Purification of CD4+ T cells are obtained by negative selection method (Human CD4 Cell Recovery Column Kit # CL110-5 Accurate). 100,000 purified CD4+ T cells (90% purity) per microtiter plate well are cultured for 72 hours at 37°C in 5% CO2 in 100 ml of culture medium (RPMI 1640 (Gibco) supplemented with 10% heat inactivated FBS (Intergen), O.lmM non-essential amino acids, 1 nM Sodium Pyruvate, 100 units/ml Penicillin, 100 ng/ml Streptomycin, 50 u-g /ml Gentamicin, 10 mM Hepes and 2 mM Glutamine). Inhibitors are added to the plate at the initiation of culture. Proliferative responses in these cultures are measured by addition of 1 uCi/well titrated thymidine during the last 6 hours before harvesting of cells. Incorporation of radioactive label is measured by liquid scintillation counting (Packard 96 well harvester and counter). Results are expressed in counts per minute (cpm).
[0469] In vitro Mixed Lymphocyte Culture Model: [0470] The mixed lymphocyte culture model, which is an in vitro model of transplantation (A. J. Cunningham, "Understanding Immunology, Transplantation Immunology" pages 157-159 (1978) examines the effects of various LFA-1 antagonists in both the proliferative and effector arms of the human mixed lymphocyte response.
148

WO 2005/044817 PCT/US2004/036942
[0471] Isolation of Cells: Mononuclear cells from peripheral blood (PBMC) are separated from heparanized whole blood drawn from healthy donors. Blood is diluted 1:1 with saline, layered, and centrifuged at 2500 x g for 30 minutes on LSM (6.2 g Ficoll and 9.4 g sodium diztrizoate per 100 ml) (Organon Technica, NJ). An alternative method is to obtain whole blood from healthy donors through leukophoresis. PBMCs are separated as above, resuspended in 90% heat inactivated Fetal Bovine serum and 10% DMSO, aliquoted and stored in liquid nitrogen. After thawing, cells are resuspended in RPMI1640 medium (Gibco, Grand Island, NY) supplemented with 10% heat-inactivated Fetal Bovine serum (Intergen, Purchase, NY), 1 mM sodium pyruvate, 3 mM L-glutamine, 1 mM nonessential amino acids, 500 ug/ml penicillin, 50 jig /ml streptomycin, 50 ug /ml gentamycin (Gibco). [0472] Mixed Lymphocyte Response (MLR): One way human mixed lymphocyte cultures are established are in 96-well flat-bottomed microtiter plates. 1.5 x 10s responder PBMCs are co-cultured with an equal number of allogeneic irradiated (3000 rads for 3 minutes, 52 seconds stimulator PBMSc in 200 ul of complete medium. LFA-1 antagonists are added at the initiation of cultures. Cultures are incubated at 37°C hi 5% CO% for 6 days, then pulsed with 1 uCi/well of 3H-thymidine (6.7 Ci/mmol, NEN, Boston, MA) for 6 hours. Cultures are harvested on a Packard cell harvester (Packard, Canberra, Canada). [3H] TdR incorporation is measured by liquid scintillation counting. Results are expressed as counts per minute (cpm).
149

WO 2005/044817 PCT/US2004/036942
CLAIMS
1. An isolated compound having the structure (I):

and pharmaceutically acceptable derivatives thereof;
wherein R1 and R2 are each independently hydrogen, an amino acid side
chain, -(CH2)mOH, -(CH2)maryl, -(CH2)mheteroaryl, wherein m is 0-6, -
CH(R1A)(OR1B), -CH(R1A)(NHR1B), U-T-Q, or an aliphatic, alicyclic,
heteroaliphatic or heteroalicyclic moiety optionally substituted with U-T-Q, wherein
U is absent, -O-, -S(O)0.2-, -SO2NT(R1A), -N(R1A)-, -N(R1A)C(=O)-, -N(R1A)C(=O)-
O-, -N(R1A)C(=O)-N(RIB)-, -N(R1A)-SOr, -C(=O)-, -C(=O)-O-, -O-C(=O)-, aryl,
heteroaryl, alkylaryl, alkylheteroaryl, -C(=O)-N(R1A)-, -O-C(=O)-N(RIA)-, -C(=N-
R1E)-, -C(=N-R1E)-O-, -C(=N-R1E)-N(R1A)-, -O-C(=N-R1E)-N(R1A)-, -N(R1A)C(=N-
R1E)-, -N(R1A)C(=N-R1E>0-, N(R1A)C(=N-R1E)-N(R1B)-, -P(=O)(OR1A)-0-, or -
P(=O)(R1A)-O-; T is absent, an aliphatic, heteroaliphatic, aryl, heteroaryl, alkylaryl
or alkylheteroaryl moiety; and Q is hydrogen, halogen, cyano, isocyanate, -OR1B, -
SR1B; -N(R1B)2, -NHC(=O)ORIB, -NHC(=O)N(RIB)2, -NHC(=O)R1B, -NHSO2R1B, -
NHSO2N(R1B)2, -NHSO2NHC(=O)OR1B, -NHC(=O)NHSO2R1B,
C(=O)NHC(=O)OR1B, -C(=O)NHC(=O)R1B, -C(=O)NHC(=O)N(R1B)2,
C(=O)NHSO2R1B, -C(=O)NHSO2N(R1B)2, -C(=S)N(R1B)2, -SO2R1B, -SO2-O-R1B, -
SO2-N(R1B)2, -SO2-NHC(=O)OR1B, -SO2-NHC(=O)-N(R1B)2, -SO2-NHC(=O)R1B, -
O-C(=O)N(R1B)2, -O-C(=O) R1B, -O-C(=O)NHC(=O)R1B, -O-C(=O)NH-SO2R1B, -
O-SO2R1B, or an aliphatic heteroaliphatic, aryl or heteroaryl moiety, or wherein R1
and R2 taken together are an alicyclic or heterocyclic moiety, or together are
wherein each occurrence of R1A and R1B is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, -COR1C, or -C0NRICR1D; wherein each occurrence of Rlc and R1D is independently hydrogen, hydroxyl, or an aliphatic, heteroaliphatic, aryl,
150

WO 2005/044817 PCT/US2004/036942
heteroaryl, alkylaryl or alkylheteroaryl moiety; and R1E is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, -CN, -OR1C, -NR1CR1D or -SO2R1C;
R3 is -C(=O)OR3A, -C(=O)H, -CH2OR3A, -CH2O-C(=O)-alkyl, -C(=O)NH(R3A), - CH2X°; wherein each occurrence of R3A is independently hydrogen, a protecting group, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl or heteroalkylheteroaryl moiety, or R3A, taken together with R1 or R2, forms a heterocyclic moiety; wherein X° is a halogen selected from F, Cl, Br or I;
R4, for each occurrence, is independently hydrogen, halogen, -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR°2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O>, -NRG2C(=O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety;
n is an integer from 0-4;
AR1 is a monocyclic or polycyclic aryl, heteroaryl, alkylaryl, alkylheteroaryl, alicyclic or heterocyclic moiety;
A, B, D and E are connected by either a single or double bond, as valency permits; wherein each occurrence of A, B, D and E is independently C=O, CR'R", NR1, CR1, N, O, S, S(=O) or SO2; wherein each occurrence of R1 is independently hydrogen, halogen^ -CN, -NO2, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NR^CCO)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or any two adjacent occurrences of R1, taken together, represent an alicyclic, heteroalicyclic, aryl, or heteroaryl moiety;
p is an integer from 0-4; and
L is absent or is V-W-X-Y-Z, wherein each occurrence of V, W, X, Y and Z is independently absent, C=O, NRL1, -O-, -C(RL1)=, =C(RL1)-, -CCRL1)(RU), C(=N-O-RL1), C(=N-RL1), -N=, S(O)0-2; a substituted or unsubstiruted Ci.6alkylidene or C2.
151

WO 2005/044817 PCT/US2004/036942
6alkenylidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O)-, -CO2-5 -C(=O)C(=O)-, -C(=O)NRU-, -OC(=O)-, -OC(=O)NRU-, -NR^NR14-, -NRL3NRUC(=O)-, -NRUC(=O>, -NRUCO2-, -NRUC(=O)NRM-, -S(=O>, -SO2-, -NRUSO2-, -SC^NR13-, -NRI3SO2NRL4-, -O-, -S-, or -NR^-; wherein each occurrence of Ru and RM is independently hydrogen, alkyl, heteroalkyl, aryl, heteroaryl or acyl; or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and each occurrence of RL1 and Ru is independently hydrogen, hydroxyl, protected hydroxyl, amino, protected amino, thio, protected thio, halogen, cyano, isocyanate, carboxy, carboxyalkyl, formyl, formyloxy, azido, nitro, ureido, thioureido, thiocyanato, alkoxy, aryloxy, mercapto, sulfonamido, benzamido, tosyl, or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or wherein one or more occurrences of RL1 and R12, taken together, or taken together with one of V, W, X, Y or Z form an alicyclic or heterocyclic moiety or form an aryl or heteroaryl moiety.
2. The compound of claim 1, wherein R3 is carboxyl, protected carboxyl or a
prodrug thereof, wherein R3 is C(=O)R3A, wherein R3A is hydroxy, alkoxy,
cycloalkoxy, aralkoxy, arcycloalkoxy, aryloxy, alkylcarbonyloxyalkyloxy,
alkoxycarbonyloxyalkyloxy, alkoxycarbonylalkyloxy,
cycloalkylcarbonyloxyalkyloxy, cycloalkoxycarbonyloxyalkyloxy,
cycloalkoxycarbonylalkyloxy, arylcarbonyloxyalkyloxy,
aryloxycarbonyloxyalkyloxy, arylcarbonyloxyalkyloxy,
alkoxyalkylcarbonyloxyalkyloxy, or one of the structures:
152

WO 2005/044817 PCT/US2004/036942

3. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RB1, RB2 and RE are independently hydrogen or substituted or unsubstituted lower alkyl.
4. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl.
5. The compound of claim 1 wherein the compound has the structure:
153

WO 2005/044817 PCT/US2004/036942

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl.
6. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RE is hydrogen or substituted or unsubstituted lower alkyl.
7. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA is hydrogen, lower alkyl or acyl; and RE is hydrogen or substituted or unsubstituted lower alkyl.
8. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA1, R*2, RB1 and RB2 are independently hydrogen or substituted or unsubstituted lower alkyl.
154

WO 2005/044817 PCT/US2004/036942
9. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA and RB are independently hydrogen or substituted or unsubstituted lower alkyl.
10. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA is hydrogen or substituted or unsubstituted lower alkyl.
11. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RB is hydrogen or substituted or unsubstituted lower alkyl.
12. The compound of claim 1 wherein the compound has the structure:
155

WO 2005/044817 PCTAJS2004/036942

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and RA is hydrogen or substituted or unsubstituted lower alkyl.
13. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA, RB and RE are independently hydrogen or substituted or unsubstituted lower alkyl.
14. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; RA, RB and RE are independently hydrogen or substituted or unsubstituted lower alkyl.
15. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and A and B are independently N or CH.
156

WO 2005/044817 PCT/US2004/036942
16. The compound of claim 1 wherein the compound has the structure:

wherein R4A and R4B are independently a halogen selected from F, Cl, Br or I; and A and B are independently N or CH.
17. The compound of claim 1, wherein - CC^NHCCR1)^2)!?3 is a moiety
having the following structure:

wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and Rs is hydrogen, alkyl, heteroalkyl, aryl, heteroaryl, or is -G°RG1 wherein G° is -O-, -S- or -NR02-, and RG1 and RG2 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aromatic or heteroaromatic moiety.
18. The compound of claim 1, wherein - C(=O)NHC(R1)(R2)R3 is a moiety
having the following structure:
wherein R1A is Ar2> -OR1B, -SR1B or -NR1BR1C; or an alkyl or heteroalkyl moiety; and Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; wherein R1B and RIC are independently hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocyclic, aryl, heteroaryl, or R1B and R1C, taken together with the nitrogen atom to which they are attached, form a heterocylic or heteroaryl moiety.
19. The compound of claim 1, wherein - C(=O)NHC(R1)(R2)R3 is a moiety
having the following structure:
157

WO 2005/044817 PCT/US2004/036942

wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and R2A is hydrogen, C1.6alkyl, C2.6alkenyl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; or R2A, taken together with a substituent on Ar2, forms a substituted or unsubstituted heterocyclic or heteroaryl moiety.
20. The compound of claim 1, wherein -C(=O)NHC(Ri)(R2)R3 is a moiety
having the following structure:

wherein t is 1-3; and RP3 is alkyl, cycloalkyl, heterocyclic, aryl or heteroaryl moiety.
21. The compound of claim 20, wherein t is 2 and RP3 is, alkyl, cycloalkyl,
!!' heterocyclic, aryl or heteroaryl moiety.
22. The compound of claim 1, wherein - C(=O)NHC(R1)(R2)R3 is a moiety
having the following structure:

wherein Ar2 is a cycloalkyl, heterocyclic, aryl or heteroaryl moiety.
23. The compound of claim 1, wherein - C(=O)NHC(RI)(R2)R3 is a moiety
having the following structure:
158

WO 2005/044817 PCT/US2004/036942

wherein R2A is hydrogen, C^aUcyl, C2.6alkenyl, aryl, heteroaryl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; or R2A, taken together with R2C or R2D, forms a substituted or unsubstituted heterocyclic or heteroaryl moiety; R2C is hydrogen, CN, -C=NMe, =NO2, =NC(=O)NH2, =NS(O)2R, =NS(O)2NRR\ -SO2R2G, or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic,'aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; wherein R end R' are each independently hydrogen or methyl, and R2G is lower alkyl; and R2D is Ar2, hydrogen, halogen, CN, NO2, an aliphatic, heteroaliphatic, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NR^CeO)- or -SOjNR02-, and RG1 and R02 are independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety.
24. The compound of claim 20, , wherein
C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D is a moiety having the following structure:

wherein R2E and R2F are each independently hydrogen, or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or R2E and R2F , taken together, form a substituted or unsusbtituted heterocyclic or heteroaryl moiety.
159

WO 2005/044817 PCT/US2004/036942
25. The compound of claim 20, wherein
C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D is a moiety having one of the following structures:

wherein R2C is hydrogen, CN, -C=NMe, =NO2, =NC(==O)NH2, =NS(O)2R, or =NS(O)2NRR'; wherein R and R' are each independently hydrogen or methyl.
26. The compound of claim 20, wherein
C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D is a moiety having one of the following structures:

or bioisostere thereof;
wherein R2E and R2F are each independently hydrogen, or an aliphatic, alicyclic, heteroaliphatic, heteroalicyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or R2E and R2F , taken together, form a substituted or unsusbtituted heterocyclic or heteroaryl moiety.
27. The compound of claim 23, wherein the bioiostere has one of the following structures:
160

WO 2005/044817 PCT/US2004/036942

wherein R2C is lower alkyl.
28. The compound of claim 23, wherein R2D is, or R2E and R2F together with the nitrogen atom to which they are attached form, a moiety having one of the structures:

wherein s is an integer between 0 and 6; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O>, -NRG2C(=O> or -SOaNR02-, and RG1 and R02 are
161

WO 2005/044817 PCT/US2004/036942
independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of R is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is O RP5 may also be hydrogen; and each occurrence of RP2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two adjacent occurrences of RP1 and R1"2, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety.
29. The compound of claim 25, wherein R2D is, or R2E and R2F together with the
nitrogen atom to which they are attached form, a moiety having one of the
structures:
wherein each occurrence of RP1 is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3, or -N(CH3)2.
30. The compound of claim 26, wherein R2D is, or R2E and R2F together with the
nitrogen atom to which they are attached form, a moiety having one of the
structures:
31. The compound of claim 26, wherein
C(=O)NHCH(CO2R3A)CH2N(R2A)C(=NR2C)R2D has the structure:
162

WO 2005/044817 PCT/US2004/036942

or bioisostere thereof;
wherein each occurrence of RP1 is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3 or -N(CH3)2; R2A is hydrogen, C^alkyl, C2.6alkenyl, aryl, heteroaryl, -C(=O)R2B or -SO2R2B, wherein R2B is alkyl, cycloalkyl, heteroalkyl, heterocyclyl, aryl or heteroaryl; and q is 1 or 2.
32. The compound of claim 31, wherein the bioisostere has one of the following structures:

wherein q is 1 or 2; and R is lower alkyl.
33. The compound of claim 17, wherein R3A is independently hydrogen, lower
alkyl, phenyl or benzyl.
34. The compound of claim 17 wherein Ar2 is one of the following structures:

163

WO 2005/044817 PCTYUS2004/036942

wherein s is an integer from 0-2; each occurrence of Rpl is independently hydrogen, halogen, CN, isocyanate, NO2, -ORG1, -S RG1, -NRG1RG2-, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently lower alkyl, or when Y is O RP5 may also be hydrogen; each occurrence of RK is independently hydrogen, alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; RP3 is lower alkyl or — N(R )2; and R and R are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety.
3 5. The compound of claim 3 0 wherein Ar2 is one of the following structures:

36. The compound of claim 17 wherein Ar2 is one of the following structures:

wherein Rp3 is lower alkyl and RG1 is hydrogen or lower alkyl.
37. The compound of claim 18 wherein R1A is -NH2 or a moiety having the
structure:

164

WO 2005/044817 PC17US2004/036942
wherein RP1 is independently hydrogen, hydroxyl, lower alkyl or lower heteroalkyl; and each occurrence of RK is independently hydrogen or lower alkyl.
38. The compound of claim 37 wherein R1A is -NH2 or a moiety having the
structure:
wherein R is hydrogen or lower alkyl.
39. The compound of claim 18 wherein R1A is a moiety having one of the
structures:

wherein s is an integer between 0 and 2; each occurrence of RP1 is independently lower alkyl or is -GRG1 wherein G is -O- or -NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and each occurrence of RP2 is independently hydrogen, lower alkyl, aryl or heteroaryl.
40. The compound of claim 39 wherein R1A is a moiety having one of the structures:

165

WO 2005/044817 PCT/US2004/036942
wherein G is -0- or -NR -, and R and R are independently hydrogen or lower alkyl.
41. The compound of claim 19, wherein -NH(R2A)Ar2 is one of the following structures:

wherein Xi is N or CRP1; s is an integer from 0-5; and each occurrence of RP1 is independently hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SOr, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and RP3 is alkyl, heteroalkyl, aryl or heteroaryl.
42. The compound of claims 41, wherein -NH(R2A)Ar2 has the following
structure:

wherein RP1 is hydrogen, halogen or lower alkyl.
43. The compound of claim 22, wherein -C(=O)NHC(R1)(R2)R3 is one of the
following structures:
166

WO 2005/044817 PCT/US2004/036942
wherein RP3 is lower alkyl or aryl; Xi and X2 are independently N or CRP1; X3 is O, S or NRP2; wherein RP1 is hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O>, -NR^C^O)- or -SO2NRG2-, and RG1 and R02 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and R 2 is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety.
44. The compound of claim 43, wherein R1"3 is lower alkyl; and RM is hydrogen
or lower alkyl.
45. The compound of any one of claims 17,18, 19 and 22, or claim 23 where
R2D is Ar2, wherein Ar2 is one of the following structures:

wherein s, X1, X2 and X3 are as defined in xx) above; X5 is O, S or NRP2; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(=O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl,
167

WO 2005/044817 PCT/US2004/036942
alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(O)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; each occurrence of RP5 is independently alkyl, heteroalkyl, aryl or heteroaryl, or when Y is 0 Rps may also be hydrogen; each occurrence of RP2 is independently hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group; wherein any two adjacent occurrences of RP1 and JtP, taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety; and each occurrence of Rp3 is independently alkyl, aryl, heteroaryl or -N(RP2)2.
46. The compound of claim 45 wherein Ar2 is one of the following structures:

wherein Xi is N or CRP1; s is an integer from 0-6; each occurrence of RPI is independently hydrogen, halogen, CN, NO2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NR02-, -CO-, -SO-, -SO2-, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NRG2C(=O)- or -SO-jNR02-, and RG1 and R02 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety; and Rp3 is independently lower alkyl or aryl.
47. The compound of claim 1, wherein -CC^NHCCR^CR^R3 has the structure
-C(=O)NHC(=C(Rs)Ar2)CO2R3A wherein R3Aand Rs, taken together, form a
substituted or unsubstituted heterocyclic moiety.
48. The compound claim 47, wherein -C(=O)NHC(=C(Rs)Ar2)CO2R3A is one of
the following structures:
168

WO 2005/044817 PCT/US2004/036942

wherein Ar2 is heterocycle, aryl or heteroaryl; and Xi is 0, S or NH.
49. The compound of claim 47, wherein -C(=O)NHC(=C(Rs)Ar2)CO2R3A is one of the following structures:

wherein Xi is O, S or NH; and X2 is N or CH.
50. The compound of claim 1, wherein L is absent, -C(=O), -CH2C(=O)NH-, -CH2NH-C(=O)-, -O-CH2-C(=O)-, -CH2-CH2-C(=O)-, -CH=CH-C(=O)NH-CH2-, -CH(OH)-CH2-O-3 -CH(OH)-CH2-N(CH3>, -CH(OH)-CH2-CH2-, -CH2-CH2-CH(OH)-3 -O-CH2-CH(OH)-, -O-CH2-CH(OH)-CH2-, -O-CH2-CH2-CH(OH)-, O-CH2-CH2-O-, -CH2-CH2-CH2-O-, -CH2-CH(OH)-CH2-O, -CH2-CH2-O-, -CH-(CH3> NH-C(O)-, -CH2-NH-SO2-, -NH-SO2-CH2-, -CH2-SO2-NH-? -SO2NH-CH2-, -C(=O)-NH-C(=O)-, -NH-C(=O)-NH-, -NH-C(=O)-NH-CH2-, -CH2-NH-C(=O)-NH-, -C(=O)-NH-CH2-C(=O)-NH, -NH-C(=0)-O, or -O-C(-O)-NH-; or a substituted or unsubstituted Ci.6alkylidene or C2^alkenylidene chain wherein up to two non-adjacent methylene units are independently optionally replaced by -C(=O)-, -CO2-, -C(=O)C(=O>, -C(=0)NR13-, -OC(=O)-, -OC(=O)NRu-5 -NR^NR14-, -NRL3NRL4C(=O)-, -NR^CC^O)-, -NRUCO2-, -NRUC(=O)NRM-, -S(=O)-, -SO2-, -NRUSO2-, -SO2NR°-, -NRUSO2NRW-, -O-, -S-, or -NR13-; wherein each occurrence of R13 and Rw is independently hydrogen, alkyl, heteroalkyl, aryl, heteroaryl or acyl.
169

WO 2005/044817 PCT/US2004/036942
51. The compound of claim 1, wherein L is absent, -C(=O), -CH2C(=O)NH-, -
CH2NH-C(O)-, -O-CH2-C(=O)-, -CH2-CH2-C(=O)-, -CH=CH-C(=O)NH-CH2-, -
CH(OH)-CH2-O-3 -CH(OH)-CH2-N(CH3)-, -CH(OH)-CH2-CH2-, -CH2-CH2-
CH(OH)-, -O~CH2-CH(OH)-, -O-CH2-CH(OH)-CH2-5 -O-CH2-CH2-CH(OH)-, O-
CH2-CH2-O-, -CH2-CH2-CH2-O-, -CH2-CH(OH)-CH2-O, -CH2-CH2-O-, -CH-(CH3)-
NH-C(=O)-, -CH2-NH-SO2-, -NH-SO2-CH2-, -CH2-SO2-NH-, -SO2NH-CH2-, -
C(=O)-NH-C(=O)-, -NH-C(=O)-NH-, -NH-C(=O)-NH-CH2~, -CH2-NH-C(=O)-NH-
, -C(=O)-NH-CH2-C(=O)-NH, -NH-C(=O)-O- or -O-C(=O)-NH.
52. The compound of claim 1, wherein L is absent, -C(=O), or a substituted or
unsubstituted Ci^alkylidene or C2-6alkenylidene chain wherein up to two non-
adjacent methylene units are independently optionally replaced by -C(=O)-, -CO2-, -
C(=O)C(=O)-, -CH^NR13-, -OC(=O)-, -OCC^NR13-, -NRUNRM-, -
NR^NR^COO)-, -NRUC(=O)-, -NRuCO2-5 -NRUC(=O)NRM-, -S(=O)-, -SO2-, -
NRL3SO2-3 -SO2NRU-, -NRUSO2NRL4-, -O-, -S-, or -NRU-; wherein each
occurrence of Ru and RM is independently hydrogen or methyl.

53. The compound of claim 1, wherein L is absent
54. The compound of claim 1, wherein L is -C(=O).
55. The compound of claim 1, wherein L is a substituted or unsubstituted d.
6alkylidene or C2.6alkenylidene chain wherein up to two non-adjacent methylene
units are independently optionally replaced by -C(=O>, -C(=O)NR°-5 -OC(=O>, -
NRL3C(=O)-, -S(-O)-, -SO2-, -NRL3SO2-, -SO2NRU-, -O- or -NR13-; wherein each
occurrence of Ru and Ru is independently hydrogen or methyl.
170
56. The compound of claim 1, wherein L is -(CH2)q- wherein q is 1 -5.
57. The compound of claim 1, wherein L is a moiety having the structure:


WO 2005/044817 PCT/US2004/036942
58. The compound of claim 1, wherein AR1 is one of the following structures:

wherein each occurrence of r is an integer from 0-6; Xi, X2, X3 and X4 is independently N or CRQ1; X5 is O, S or NRQ2; AR3 is a heterocyclic, aryl or heteroaryl moiety; each occurrence of RQ1 is independently hydrogen, ORQ3, OCF3, SRQ3, halogen, CN, isocyanate, NO2, CF3, NRQ3QRQ4, -SC>2RQ3, alkyl-NR^R^, alkyl-C(=O)-NRQ3RQ4, alkyl-C(=O)RQ3, or an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety, wherein each occurrence of RQ3 and RQ4 is independently hydrogen, a protecting group, or an aliphatic, heteroaliphatic, aryl or heteroaryl moiety; and RQ2 is hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl, alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or a nitrogen protecting group.
59. The compound of claim 58, wherein AR1 is one of the following structures:

171

WO 2005/044817 PCT/US2004/036942

wherein X° is F or Cl; X2 is N or CRQ1; X5 is CH, 0, S or NH; RQ1 is hydrogen, methyl, -CF3, -OCH3,- OCF3 or halogen.
60. The compound of claim 59, wherein ARl is one of the following structures:

61. The compound of claim 60, wherein AR1 is one of the following structures:

62. The compound of claim 1, wherein AR^-L- is one of the following structures:

63. The compound of claim 62, wherein AR*-L- is one of the following
structures:
172

WO 2005/044817 PCT/US2004/036942
64. The compound any one of claims 3-16 wherein AR^L- is a moiety having one of the following structures:

and -C(=O)NHC(R1)(R2)R3 is a moiety having one of the following structures:
or bioisosteres thereof;
wherein R2A and R3A are as defined in classes and subclasses herein; and R2D is a moiety having one of the following structures:

wherein s is an integer between 0 and 6; each occurrence of RP1 is independently hydrogen, halogen, CN, isocyanate, NO2, -P(-O)(YRP5)2, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylaryl or alkylheteroaryl moiety, or is -GRG1 wherein G is -O-, -S-, -NRG2-, -CO-, -SO-, -SO2~, -C(=O)O-, -C(=O)NRG2-, -OC(=O)-, -NR^CCO)- or -SO2NRG2-, and RG1 and RG2 are independently hydrogen, an alkyl, cycloalkyl, heteroalkyl, heterocyclic, aryl, heteroaryl, alkylary] or alkylheteroaryl moiety; each occurrence of Y is independently a bond or O; eacl occurrence of Rps is independently alkyl, heteroalkyl, aryl or heteroaryl, or when "V is O RP5 may also be hydrogen; and each occurrence of Rp2 is independent!} hydrogen, an aliphatic, alicyclic, heteroaliphatic, heterocyclic, aryl, heteroaryl alkylaryl, alkylheteroaryl, heteroalkylaryl, or heteroalkylheteroaryl moiety or 1 nitrogen protecting group; wherein any two adjacent occurrences of Rp and R taken together, may form a cycloalkyl, heterocyclic, aryl or heteroaryl moiety.
173

WO 2005/044817 PCT/US2004/036942
65. The compound of claim 64 wherein R2A and R3A are each hydrogen.
66. The compound of claim 64 wherein R2D is a moiety having one of the
structures:
wherein each occurrence of RPI is independently hydrogen, halogen, methyl, -OCH3, -OH, -NH2, -NHCH3, or-N(CH3)2.
67. The compound of claim 66 wherein R D is a moiety having one of the
structures:
68. A pharmaceutical composition comprising a compound of any one of claims
1-67
and pharmaceuticaily acceptable derivatives thereof; and a pharmaceuticaily acceptable carrier or diluent, and optionally further comprising an additional therapeutic agent.
69. The pharmaceutical composition of claim 68, wherein the compound is
present in an amount effective to modulate adhesion between intracellular adhesion
molecules (e.g., ICAM-1, -2 and -3) and the leukocyte integrin family of receptors.
> 70. The pharmaceutical composition of claim 68, wherein the compound is present in an amount effective to antagonize CD11/CD18 receptors associated with leukocytes.
174

WO 2005/044817 PCT/US2004/036942
71. The pharmaceutical composition of claim 68, wherein the compound is
present in an amount effective to antagonize Mac-1 and/or LFA-1.
72. A method of treating, preventing or ameliorating an immune or inflammatory
response or disorder in a subject mediated through the CD11/CD18 family of
cellular adhesion molecules comprising administering to a subject a therapeutically
effective amount of a compound of any one of claims 1-67.
73. The method of claim 72, wherein the CD11/CD18 family of cellular
adhesion molecules is LFA-1.

74. A method of treating, preventing or ameliorating an immune or inflammatory
response or disorder in a subject comprising administering to a subject in need
thereof a compound of any one of claims 1-67.
75. The method of claim 74, wherein the disorder is selected from T-cell
inflammatory responses such as inflammatory skin diseases including psoriasis;
responses associated with inflammatory bowel disease (such as Crohn's disease and
ulcerative colitis); adult respiratory distress syndrome, dermatitis, meningitis,
encephalitis, uveitic, allergic conditions such as eczema and asthma and other
conditions involving infiltration of T-cells and chronic inflammatory responses, skin
hypersensitivity reactions (including poison ivy and poison oak), atherosclerosis,
leukocyte adhesion deficiency, autoimmune diseases such as rheumatoid arthritis,
systemic lupus erythematosus (SLE), diabetes mellitus, multiple sclerosis,
Reynaud's syndrome, autoimmune thyroiditis, experimental autoimmune
encephalomyelitis, Sjorgen's syndrome, type 1 diabetes, juvenile onset diabetes, and
immune responses associated with delayed hypersensitivity mediated by cytokines
and T-Iymphocytes typically found in tuberculosis, sarcoidosis, polymyositis,
granulomatosis, and vasculitis, pernicious anemia, diseases involving leukocyte
diapedesis, CNS inflammatory disorder, multiple organ injury syndrome secondary
to septicaemia or trauma, autoimmune haemolytic anemia, myethamia gravis,
175

WO 2005/044817

PCT/US2004/036942

antigen-antibody complex mediated diseases, and all types of transplantations, including graft vs. host or host vs. graft disease.
176

Documents:

01233-kolnp-2006-abstract.pdf

01233-kolnp-2006-claims.pdf

01233-kolnp-2006-correspondence other.pdf

01233-kolnp-2006-correspondence-1.1.pdf

01233-kolnp-2006-description complete.pdf

01233-kolnp-2006-form 1.pdf

01233-kolnp-2006-form 3.pdf

01233-kolnp-2006-form 5.pdf

01233-kolnp-2006-form-18.pdf

01233-kolnp-2006-international publication.pdf

01233-kolnp-2006-international search report.pdf

1233-KOLNP-2006-ABSTRACT.pdf

1233-KOLNP-2006-AMANDED CLAIMS 1.1.pdf

1233-KOLNP-2006-AMANDED CLAIMS.pdf

1233-KOLNP-2006-ASSIGNMENT-1.1.pdf

1233-KOLNP-2006-ASSIGNMENT-1.2.pdf

1233-KOLNP-2006-ASSIGNMENT.pdf

1233-kolnp-2006-assignment1.3.pdf

1233-KOLNP-2006-CORRESPONDENCE 1.1.pdf

1233-KOLNP-2006-CORRESPONDENCE 1.2.pdf

1233-KOLNP-2006-CORRESPONDENCE 1.3.pdf

1233-KOLNP-2006-CORRESPONDENCE-1.1.pdf

1233-KOLNP-2006-CORRESPONDENCE-1.2.pdf

1233-KOLNP-2006-CORRESPONDENCE-1.3.pdf

1233-KOLNP-2006-CORRESPONDENCE-1.4.pdf

1233-kolnp-2006-correspondence.pdf

1233-KOLNP-2006-DESCRIPTION (COMPLETE).pdf

1233-kolnp-2006-examination report.pdf

1233-KOLNP-2006-FORM 1 1.2.pdf

1233-KOLNP-2006-FORM 1-1.1.pdf

1233-KOLNP-2006-FORM 1.pdf

1233-KOLNP-2006-FORM 13-1.1.pdf

1233-KOLNP-2006-FORM 13-1.2.pdf

1233-kolnp-2006-form 13.3.pdf

1233-KOLNP-2006-FORM 13.pdf

1233-kolnp-2006-form 18.pdf

1233-KOLNP-2006-FORM 2 1.2.pdf

1233-KOLNP-2006-FORM 2-1.1.pdf

1233-KOLNP-2006-FORM 2.pdf

1233-kolnp-2006-form 3.1.pdf

1233-KOLNP-2006-FORM 3.pdf

1233-KOLNP-2006-FORM 5 1.1.pdf

1233-kolnp-2006-form 5.2.pdf

1233-KOLNP-2006-FORM 5.pdf

1233-kolnp-2006-form 6.1.pdf

1233-KOLNP-2006-FORM 6.pdf

1233-KOLNP-2006-FORM-27.pdf

1233-kolnp-2006-gpa.pdf

1233-kolnp-2006-granted-abstract.pdf

1233-kolnp-2006-granted-claims.pdf

1233-kolnp-2006-granted-description (complete).pdf

1233-kolnp-2006-granted-form 1.pdf

1233-kolnp-2006-granted-form 2.pdf

1233-kolnp-2006-granted-specification.pdf

1233-KOLNP-2006-OTHER PCT FORM.pdf

1233-KOLNP-2006-OTHERS 1.2.pdf

1233-KOLNP-2006-OTHERS-1.1.pdf

1233-KOLNP-2006-OTHERS.pdf

1233-KOLNP-2006-PA 1.2.pdf

1233-KOLNP-2006-PA-1.1.pdf

1233-KOLNP-2006-PA.pdf

1233-KOLNP-2006-PETITION UNDER RULE 137-1.1.pdf

1233-KOLNP-2006-PETITION UNDER RULE 137.pdf

1233-KOLNP-2006-REPLY TO EXAMINATION REPORT.pdf

1233-kolnp-2006-reply to examination report1.1.pdf

1233-kolnp-2006-translated copy of priority document.pdf

abstract-01233-kolnp-2006.jpg


Patent Number 250181
Indian Patent Application Number 1233/KOLNP/2006
PG Journal Number 50/2011
Publication Date 16-Dec-2011
Grant Date 14-Dec-2011
Date of Filing 11-May-2006
Name of Patentee SARCODE CORPORATION
Applicant Address 1000 MARINA BOULEVARD, SUITE 250, BRISBANE, CA
Inventors:
# Inventor's Name Inventor's Address
1 SHEN, WANG 1719 MONTICELLO ROAD, SAN MATEO, CA 94402
2 OSLOB, JOHAN, D. 1191 LOCHINVAR AVENUE SUNNYVALE, CA 94087
3 ZHONG, MIN 912 BEACH PARK BOULEVARD UNIT 93 FOSTER CITY, CA 94404
4 ZHONG, MIN 839 CATAMARAN STREET,# 4 FOSTER CITY, CA 94404
5 BARR, KENNETH 4016 18TH STREET, SAN FRANCISCO, CA 94114
PCT International Classification Number C07D 401/06
PCT International Application Number PCT/US2004/036942
PCT International Filing date 2004-11-05
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/517,535 2003-11-05 U.S.A.
2 60/560,517 2004-04-08 U.S.A.