CA2681718A1 - Hiv-1 protease inhibitors - Google Patents

Hiv-1 protease inhibitors Download PDF

Info

Publication number
CA2681718A1
CA2681718A1 CA002681718A CA2681718A CA2681718A1 CA 2681718 A1 CA2681718 A1 CA 2681718A1 CA 002681718 A CA002681718 A CA 002681718A CA 2681718 A CA2681718 A CA 2681718A CA 2681718 A1 CA2681718 A1 CA 2681718A1
Authority
CA
Canada
Prior art keywords
alkyl
compound
heterocyclyl
amino
amido
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002681718A
Other languages
French (fr)
Inventor
Akbar Ali
Michael D. Altman
Saima Ghafoor Anjum
Hong Cao
Sripriya Chellappan
Miguel X. Fernandes
Michael Gilson
Visvaldas Kairys
Nancy King
Ellen Nalivaika
Moses Prabu
Tariq M. Rana
Kiran Kumar Reddy Garudammagari Sai
Celia A. Schiffer
Bruce Tidor
Madhavi Naga Laxmi Nalam
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Massachusetts UMass
Massachusetts Institute of Technology
University of Maryland at College Park
Original Assignee
University Of Massachusetts
University Of Maryland Biotechnology Institute
Massachusetts Institute Of Technology
Akbar Ali
Michael D. Altman
Saima Ghafoor Anjum
Hong Cao
Sripriya Chellappan
Miguel X. Fernandes
Michael Gilson
Visvaldas Kairys
Nancy King
Ellen Nalivaika
Moses Prabu
Tariq M. Rana
Kiran Kumar Reddy Garudammagari Sai
Celia A. Schiffer
Bruce Tidor
Madhavi Naga Laxmi Nalam
University Of Maryland, College Park
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Massachusetts, University Of Maryland Biotechnology Institute, Massachusetts Institute Of Technology, Akbar Ali, Michael D. Altman, Saima Ghafoor Anjum, Hong Cao, Sripriya Chellappan, Miguel X. Fernandes, Michael Gilson, Visvaldas Kairys, Nancy King, Ellen Nalivaika, Moses Prabu, Tariq M. Rana, Kiran Kumar Reddy Garudammagari Sai, Celia A. Schiffer, Bruce Tidor, Madhavi Naga Laxmi Nalam, University Of Maryland, College Park filed Critical University Of Massachusetts
Publication of CA2681718A1 publication Critical patent/CA2681718A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/08Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D263/16Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/18Oxygen atoms
    • C07D263/20Oxygen atoms attached in position 2
    • C07D263/24Oxygen atoms attached in position 2 with hydrocarbon radicals, substituted by oxygen atoms, attached to other ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors

Abstract

Described are novel protease inhibitors and methods for using said protease inhibitors in the treatment of human immunodeficiency virus (HIV) infection.

Description

HIV-1 Protease Inhibitors RELATED APPLICATIONS

The present application claims priority to U.S. Provisional Application Serial No.
60/919,896, filed March 23, 2007, U.S. Provisional Application Serial No.
60/919,819, filed March 23, 2007, U.S. Provisional Application Serial No. 60/941,786, filed June 4, 2007, and U.S. Provisional Application Serial No. 60/941,829, filed June 4, 2007, all of which are hereby incorporated by reference in their entirety.

GOVERNMENT SUPPORT

This invention was made with government support under Grant No. P01 GM
066524 awarded by the National Institutes of Health/National Institute of Allergy and Infectious Diseases. The government has certain rights in the invention.

BACKGROUND OF THE INVENTION

Human immunodeficiency virus type 1 (HIV-1) protease plays a critical role in the virus life cycle by processing the viral Gag and Gag-Pol polyproteins into structural and functional proteins essential for viral maturation. Inhibition of HIV-1 protease leads to the production of noninfectious virus particles and hence is a promising therapeutic target for antiviral therapy in AIDS patients.

Protease inhibitors (PIs) are potent antiretroviral drugs for the treatment of patients infected with Human Immunodeficiency Virus (HIV). Several known PIs are recommended as part of the "preferred regimen" for patients in the guidelines of International AIDS Society-USA (IAS-USA) and the U.S. Department of Health and Human Services (DHHS). However, use of these drugs has sometimes been associated with the development of irreversible HIV resistance, due to mutation of the virus.

In fact, HIV-1 protease inhibitors represent the most potent anti-AIDS drugs reported to date and are essential components of highly active antiretroviral therapy (HAART). In the last decade, structure based drug design has led to the discovery of eight FDA approved drugs and several others in advanced clinical trials. Currently marketed HIV-1 protease inhibitors, saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir, atazanavir, and tipranavir, are all competitive inhibitors that bind in the active site of the enzyme. Except the newly approved drug tipranavir, all approved inhibitors have been developed based on transition state mimetic concept and contain various noncleavable dipeptide isosteres as core scaffolds to mimic the transition state of HIV-1 protease substrates. The development and clinical introduction of anti-AIDS HIV-1 protease inhibitors is regarded as major success of structure based drug design.

Anti-AIDS chemotherapy based on HIV-1 protease and reverse-transcriptase inhibitors has been remarkably successful in decreasing the mortality rate in HIV-1 infected patients. However, the emergence of HIV-1 mutants that are resistant to current drug regimens is a critical factor in the clinical failure of antiviral therapy. In general, drug resistance occurs when mutations in a target protein allow that protein to retain function while in the presence of a drug. In the case of HIV-1 protease, drug resistence typically occurs when, even in the presence of protease inhibitors, the enzyme is able to cleave the Gal and Pol polypeptides in at least nine different locations, allowing viral mutation. Viral resistance is regarded as a critical factor in clinical failure of antiviral therapy. The relatively rapid appearance of resistant viral mutants among treated HIV
patients is attributable to the virus' high rate of replication, coupled with a high intrinsic rate of mutation due to the infidelity of the HIV reverse transcriptase. Further, current HIV-1 protease inhibitors were designed to inhibit a single variant of HIV-1 protease.

For most of the currently approved protease inhibitors, the emergence of multi drug resistant (MDR) mutants poses a great challenge to the efficacy of these drugs. (Condra, J.
H. et al. Nature 1995, 374, 569-571; and Clavel, F. et al. N. Engl. J. Med.
2004, 350, 1023-1035.) Development of next generation HIV-1 protease inhibitors active against MDR virus has been the focus of intense research efforts in recent years.
(Koh, Y. et al.
Antimicrob. Agents Chemother. 2003, 47, 3123-3129; Surleraux, D. L. N. G. et al. J. Med.
Chem. 2005, 48, 1813-1822; and Surleraux, D. L. N. G. et al.. J Med. Chem.
2005, 48, 1965-1973.) Developing different classes of therapeutic agents is not likely to be an adequate solution to the problem of resistance to protease inhibitors, primarily because the same basic mechanisms readily generate viral strains resistant to other agents.
Thus, resistance is a major clinical problem for the other major class of HIV drugs, the reverse transcriptase inhibitors, and resistance to newer, preclinical agents, such as the fusion inhibitors is readily elicited in culture.
-2-The challenge for the research community is therefore to develop drugs, e.g., protease inhibitors, that are less vulnerable to drug resistance and/or more active against current protease resistant HIV-1 isolates. The present inventions address this challenge by integrating clinical data, in vitro virology, protein crystallography, computational modeling and chemical design, and high-throughput chemistry and compound screening. HIV
protease is a particularly appealing target, as inhibition of its activity is clinical effective;
however, it can evolve to tolerate extensive mutation that confers drug resistance while retaining enzymatic function. As the design of the initial protease inhibitors was structure based, a huge knowledge reservoir exists for this protein.

SUMMARY OF THE INVENTION

The present invention is based, at least in part, on the discovery of new small molecule protease inhibitors (PIs). These inhibitors, and methods of making and using them, are described herein. Because these inhibitors do not protrude beyond the substrate binding envelope on the protease, it is expected that these inhibitors will be less likely to induce the development of resistant strains.

In one aspect, the invention features PIs described herein, or an enantiomer, diastereomer or a pharmaceutically acceptable salt thereof, and pharmaceutical compositions for inhibiting HIV protease that include a pharmaceutical carrier and a therapeutically effective amount of a PI described herein.

In another aspect, the invention features methods for treating HIV in a subject, by administering a therapeutically effective amount of a compound or pharmaceutical composition described herein. In some embodiments, the methods further include administering a second therapeutic agent, e.g., a non-nucleoside reverse transcriptase inhibitor (NNRTI) such as efavirenz (SustivaTM), nevirapine (ViramuneTM) and delavirdine (RescriptorTM); an nucleoside reverse transcriptase inhibitor (NRTI) such as AZT
(zidovudine, RetrovirTM)/3TC (lamivudine, EpivirTM) and d4T (stavudine, ZeritTM)/3TC, and d-drugs (ddl [didanosine, VidexTM/VidexECTM], ddC [zalcitabine, HividTM], d4T); a nucleotide reverse transcriptase inhibitor, such as tenofovir (VireadTM); and a fusion inhibitor, such as enfuvirtide (FuzeonTM). In some embodiments, the compound or pharmaceutical composition is administered as part of a highly active antiretroviral therapy (HAART) regimen.
-3-Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.

BRIEF DESCRIPTION OF DRAWINGS

Figures la-b depict possible synthetic routes to selected inventive compounds.
Figures 2a-k depict anti-HIV drugs by class.

Figure 3 depicts the synthesis of protease inhibitors containing [A] a hydroyethylamine (HEA) core or [B] a hydroxyethylene (HE) core. Key: (a) EtOH, 70 C;
(b) aq. Na2CO3, CH2C12, r. t; (c) TFA, CH2C12; (d) Et3N, THF; (e) R4X2CO2H, EDCI, HOBt, DIPEA, 0 C to r. t.; (f) H2, Pd/C, MeOH, r. t; and (g) aq. NaHCO3, EtOAc, 0 C to r. t..

Figure 4 depicts the synthesis of protease inhibitors containing an aza-hydroxyethylamine (Aza-HEA) core. Key: (a) (CH3)2CHOH, 80 C (b) H2, Pd/C, MeOH, r. t; (c) R4X2CO2H, EDCI, HOBt, DIPEA, 0 C to r. t.; (d) TFA, CH2C12; and (e) R3XI COZH, EDCI, HOBt, DIPEA, 0 C to r. t.; aq. NaHCO3, EtOAc.

Figure 5 depicts a comparison of isobutyl and (S)-2-methylbutyl moieties as PI' ligands: Inhibitory activities against wild-type and MDR mutant variants of HIV-1 protease.
Figure 6 depicts selected compounds of formula I and associated K; values.

Figure 7 depicts selected compounds of formula I.

Figure 8 depicts the chemical structures of amprenavir (APV) 1, TMC114 2 and selected compounds of the invention 3.
-4-Figure 9 depicts a scheme showing the synthesis of intermediates N-Phenyloxazolidine-5-carboxylic acids 9 and 10. Key: (a) n-BuLi, THF, -78 C to r. t.
overnight; (b) RuC13.H20, CH3CN-CC14-H20 (2:2:3), 0 C to r. t. 4-10 h.

Figure 10 depicts a scheme showing the synthesis of inventive compounds 20-29.
Key: (a) EtOH, 80 C, 3-4 h; (b) aq. NazCO3, CH2C12, 0 C to r. t., 4-8 h; (c) TFA, CH2C12, 1 h; (d) (OCOCI)2, r. t., overnight; (e) Et3N, THF, 0 C to r. t., 4-8 h; (f) SnC12.2H20, EtOAc, 70 C, 2 h.

Figure 11 depicts a scheme showing the synthesis of compounds 36-39. Key: (a) iPrOH or EtOH, 80 C, 3-4 h; (b) aq. Na2CO3, CH2C12, 0 C to r. t., 4-8 h; (c) TFA, CH2C12, 1 h; (d) (OCOCI)2, r. t., overnight; (e) Et3N, THF, 0 C to r. t., 4-8 h.

Figure 12 depicts selected compounds of formula XVA/XVB and associated K;
values.

Figure 13 depicts selected compounds of formula XIIIA/XIIIB and associated K;
values.

Figure 14 depicts selected compounds of formula XVIIA/XVIIB and associated K;
values.

Figure 15a-b depict possible synthetic routes to selected inventive compounds.
Figure 16 depicts inhibitory activities of selected compounds against wild-type and MDR mutant variants of HIV-1 protease.

Figure 17a-j depicts selected compounds and, in some cases, associated K;
values.
DETAILED DESCRIPTION

One aspect of the present invention addresses the challenge of developing HIV-protease inhibitors that are less vulnerable to drug resistance and/or more active against current protease-resistant HIV-1 isolates than other HIV drugs. The present invention addresses this challenge by integrating clinical data, in vitro virology, and high-throughput chemistry and compound screening. HIV protease is a particularly appealing target because inhibiton of its activity is clinically effective; however, it can evolve to tolerate extensive mutations conferring drug resistance while retaining enzymatic function.

Remarkably. certain compounds of the invention are competitive inhibitors that appear to bind in the center of the "substrate envelope" (i.e., at the active site of the protease). Importantly, the compounds of the invention are designed such that when bound
-5-they do not significantly protrude beyond the substrate envelope; therefore, they are less likely to induce escape mutations. The protease inhibitors of the invention are useful in the treatment of HIV in susceptible mammals, e.g., humans and certain other primates. In addition the compounds have shown activity against a panel of multi-drug resistant (MDR) mutant variants of HIV-1 protease. Moreover, as mentioned above, the inhibitors of the invention can be administered as a monotherapy, or in combination with other therapeutic agents, e.g., as part of a highly active antiretroviral therapy (HAART) regime.

Selected Protease Inhibitors of the Invention. One aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula I:

0 J~ R2 0 0 R3. Ra I
wherein, independently for each occurrence, X, is absent, -0-, -S-, -NR- or X2 is absent, -0-, -S-, -NR- or Rl is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and
-6-the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

-__clr0 O-~p N
provided that when Xl is absent; R3 is not R3A or N-R3A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is or X2 is or R3 is amino, 0 CH3 Ci H CH3 CH3 H3C~a, H3C,O~~ H3C0 0 ~ H3 O CH3 H3COJJ \/~~~~X H3C0 0 0 0 0 > >

H

F3C1~N0 ~f0 , H2N` H2N1 ~ HZN II H3C O N/~
0 `` , ~`'J , O , 0 0 O NH2, H3CuN,/~ O O H H H H
IOI ~ O O`/
O NHZ H H H or ~`H ; or R6 ls CH3 or CH3.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein XI is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is ~

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X2 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein Xl is absent; and X2 is absent.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein Rl is OH.
-7-In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is aryl or heteroaryl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is alkyl, aryl or heteroaryl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R5 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R5 is alkyl, (cycloalkyl)alkyl or (heterocyclyl)alkyl.

Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula II:

O Ph II OõO
R3.XJ~H N,S.R4 OH R

II

wherein, independently for each occurrence, X, is absent, -0- or R3 is alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, heteroaryl, aralkyl or heteroaralkyl; and
-8-
9 PCT/US2008/058004 R6 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
~lr0 O O

provided that when X, is absent; R3 is not N R3A or N-R3A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and c~ z H3CY---'-~ H3C
provided that X, is or R3 is amino, 0 , 0 CI H CH3 CH3 H

C~,,YN,,-,/ H3CO,~K/ H3CO 1 ~3 J\~~3 F3C II N~~
O O O H3C0/\ ` H3CO
> > > > > o O O O CH3 GH3 H H3C N H3C N~~~
~~ HZNHZN~ HZN
~UO O O~ NH~

I 2, 2 9 ~ ~~~ CCH3 H
H H or ; or R6 is 3 or CHg cH3.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X1 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is ~.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is aryl or heteroaryl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is alkyl, aryl or heteroaryl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R6 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R6 is alkyl, (cycloalkyl)alkyl or (heterocyclyl)alkyl.

In certain embodiments, the present invention relates to the aforementioned H3C N,N
Y'l-compound and any of the attendant definitions, wherein R3 is CH3 O
H
H3C\ /~.. O CH3 H3C\ ~ H3C

0 , T0( ~O(~ ~O( 0 N~ ~-H3C0 ~ ~ H3C N\ ~ N HO N~
N/ N I/ I/
OH
O
JH O H
Oi~+ O O O H O/~
> > > > > >
N
S
C H 3 CF3 S /i CH3 F3C H3C C/~/~' Br ~
> > > , > >
F

HO F CH3 ~ HO ~,. HO OH `~,L CH3 F I~ I~ OH
F HO / HO / H3C'-"~\
CI Br S H3CUNCH3 CI I~~.. HO I
II

O I N ^ H3C N Y~ H2N
3 ~ I
H C OH
CH3 CH 3 0 ~ O
-10-F H
~ (~j` CH3 CH3 C' H CH3 I ~\v H3C\ H3C~1 Cl1,,rN,/,~`` H3CO
F 0( 0 0 , 0 H3C0~/ 0 ~H3 0 CHs F3CyN~~ 0 0 O HzN ~/
0 H3~J-~~``'s H3CO~~ O O H H

H N ~H3 H N N H3C N H3CUN~~~1 O O H
z ~ 2y ~~'~ O I0I ~"',".
O O , 0 NH2 0 NH2 /a~ , H
H H H
O Ol``~ O
H H ,or V`H .

In certain embodiments, the present invention relates to the aforementioned H3CY---)--~
compound and any of the attendant definitions, wherein R3 is 0 CH3 CI H CH3 H3C~~ CI 1~N\/~~ H3C0~, H3CO 0 CH3 0 CH3 / H3COJ-~,,,s H3CO
O O O O

H ~~ O~ (o~ H2NH2N H2NH3C O NJ ~

O II \~~ u II II NHZ
O O O

H3CUN,/~ O O H H H H

IOI ~ '', O O OLO 0 NHz ~` H H H , or H

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is F

N H3 NI~ NO H3C N ~
I ~ ( ~ N
O ~ ~ II ~}-CH3 O ~ /
5 ~ OCH3 / > N CH CH3, , > 3 F

F

\
~/~
\ CH3 S
F ,~- O NH2 H3C H3C

CH
F or ~ 3
-11-In certain einbodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R6 is H3C ~ ~ HO HO
CH3 ~L S ~L / \~ ~L~CH3 '~,L/-CH3 ~CH3 > > > > > > >

CH3 CH3 ~OH

O
,CHg CHa OH CH3 or CH3.

In certain embodiments, the present invention relates to the aforementioned ~CH3 ~CH3 compound and any of the attendant definitions, wherein R6 is CH3 or CH3.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent; and R3 is ~ \

H3C' NH3C\ /~.. CH3 H3C
~( ' f( CH3 , 0 , 0 O O

H3CH3CO ~H3C I N~ N~ HO I N~ ~,.
II N / N~ /

> > > > >
N ~..

O H O H NQ
/ O O O ''O/~ ~
OH O , H H
S

F3C~ H3C~
F

CH3 F HO I ~ ~
/
F HO HO
> > > > >
-12-OH S H3CUNCH3 CI I~~

H3C~~~' IOI CH3 HO X HO `~L H3CyN H3Cu N ~
~ H3C I O II
OH CHg CH3 0 CH3 > > > > > >
F H
\ nj \~ CH3 CH3 Ci H
H2N~~ O H3CY_"~ H3Cy \/ Ci-'yN_--\/
O , F , 0 , 0 , 0 CH3 CH3 H ~~/
O O
~CO H3CO` ^~ 0 CH3 0 CH3 F3CUN~~
O lOlf H3C0H3C0/~J~/=\ II

> > > ~ ~H N H3 H N QH3 H N H H3C 5:- H3C ~z~zz OO~

0 , 0 IOI 0 NH2, or 0 NH2 Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of Ph O 0 Ph OCH3 O O O O
HZNN N'~O HZNH ~rN
// H OH lT /CH3 L~ OH ~CH3 O 0 Ph O\ C Ph O

H2NH N'~O H3C N N~N'O
LL OH ~ H OH OH

N
N
O O Ph /J S 0 0 Ph O\ ~\ S
~
~
HZN N N~O HZNH N' O
~H (:~~

~~
0 O Ph O Cj5 S Ph N
~ /~~ H O N
H2N' 'H NXIO H3C.N'""~' N~N'S=O
-13-CH3 O Ph O S O Ph F " ~ ^
~ N~N O HO ~ N N~O
I ~
/ OH ~CH3 HO H 1~
~ OH CH3 ~
CH3 `CH3 NH2 OCH3 O O Ph ~ ~ Ph ~ ~
ON~ O, O H3C N~ OS~.
H y _ H~N " O
OH ~CH3 0 CH3 OH ~CH3 CH3 and CH3 Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of Ph OCH3 Ph OCH3 O
I O / I
CH3j, 0 \ ~ I CH
H3C~ON~N.S H3C~/~/~ OJ~NN"

H OH ~CH3 0 H OH O
~

CI 0 Ph O I OCH, Ph 0- OCH3 N \ \ H CO CH3 ~
CI'~ N~ N"3 ~O N~ N
O H OH ~CH3 0 H OH ~CH3 CO CH3~ O C0 H3 Ph O \ I
Ph ~~O N~ N"~ H3CO O N~ N"0 O H OH ~CH3 H OH ~CH3 Ph ~ ~~ Ph a OC

CH3) O\ ~ ~ H Q H3C0 O N 1 N"~O F3C II N~\OxN~N" O
H OH ~CH3 0 H OH CH3 O Ph ~~
CH3 CH3 O 0 Ph ~~ U'OIJ~N' ~ Q~ ~ ~S

H OH ~CH3 H OH ~CH3 , OCH3 Ph , OCH3 CH3 Q Ph Q ~ I CH3 r I ~ ~ I
Fi2N'OxN~N,S~ FizN ONJ~' N (~~
H OH OCH3 ~ H
O OH ~~ H3 ~

Ph , OCH3 Ph OCH3 H ~ Q H3C N
H2NUN~, O N~N y O N O I N

C~ CH3 Ph OCH3 , OCH3 H Q Q CH3 O Ph p\ ~ ~
H3CUN", N~N ~O H3C~N SO

IOI ONH H OH ~CH3 0 H OH ~CH3 H C CH3 0 Ph O` \ C' N~~ 0 Ph p \ I
~ N~N ~
ci O
3O N~N ~
O H OH ~CH3 0 H OH ~CH3 CH3 CH3 Ph OCH3 OCH3 3 CH3 OI Ph H3COC pJZN~N ~\ I H3CO~OJ~N~N.
~ H O H O
O OH CH3 O OH ~C%

CH3 CH3 OCH3 , OCH3 O CH3~ Ph p\ Cr Q CH3 Q Ph p\ ~ ~
H3C0~0 N~N % H3CO~xN~N' ~
H OH ~CH3 H OH ~CH3 CH3 CH3 Ph OCH3 O Ph OCH3 H ~ 0\ O
~
F3CUN~~OJN~N~S~O OxN~N SO
IOI H OH ~CH3 H OH ~CH3 CH3 CH3 O Ph OCH3 Ph OCH3 p ~ CH3 Q p\ ~ ~
oN N~S6 HZN~o~N~N O
~ O
H OH ~CH3 O H OH ~CH3 Ph OCH3 Ph OCH3 N~. CH3 O N
\ \ I ~~
Hz N Q
H ~ \ I
z O N N~ yO N~N p ~ O H
O H OH ,,'CH3 0 OH ~CH3 CH3 CH3 OCH3 Ph OCH3 Ph H3Cu O H3C N~~
N
O N N
~ = p NN O

Q O NHz H OH CH3 O Q~-NHz OH CH3 ~

CH3 CH3 OCH3 Ph / OCH3 CH3 O Ph Q~ CH3 O p\ ~ I
\\
H3C~Q~N~N- H3CpH -(~- N' O
H
p OH L~'~ O OH

OCH3 Ph / OCH3 CI O Ph CH3 O O\ ~ ~ H CI N~~O~N N'~ H3CO~p~N~N ~~
~ ~ O H
O H OH O OH

H3CO / OCH3 Ph / OCH3 C
N N ' H3~ Ph p\ \ I 0 CH03~ N~ NO~ ~ I
p ' ~ O H3C0 ~ H 0 O H OH OH

OCH3 Ph / OCH3 O CH3 O Ph O I F3C N~~ OS ~ ~
H~Ol~N N~ O N~N p 3CO ~ O y H
H OH O OH

/ OCH3 O Ph / OCH3 O O Ph Cl" ~\ I 'J~ ~ \ I
O N~N L"~ O N~NO H OH H OH O

a OCH3 Ph OCH3 CH3 O Ph CH3 O Q ~ ~

O N N` HzN~O N~N
~ 0 O H
O H OH O OH

Ph / OCH3 Ph / OCH3 \
rI O ~ p O ~
O~N~Ng HzN Np~NJ~-N.~O/ \~ H3CUN O
O H OH IOI JN Hz H OH

Ph OCH3 CH3 O Ph p O O \
H3CUNpN~N-~O H3CO~H N ~
IOI p~NH H OH O OH ~CH3 z Ph OCH3 Ph ~OCH3 H C CH3 0 p\ CI N~/~ ~
3 O N~N !` CI~ O N~N
Y~\ O

~ 3 ~ 3 Ph CH Ph O p ~
H3CO CH31O\ H3(.O 3~
O N~N O N O
~
O OH ~CH3 O H OH ~CH

CH3 CH3 Ph OCH3 Ph OCH3 CH3 R p\ \ ~ Q CH3 Q O~
H3COOxN~N ~ H3CO~~~OxN~N ~
H OH ~CH3 H OH ~CH3 CH3 CH3 N Ph OCH3 p p Ph , OCH3 FC ~ qs \ , \ I
a O N~N ,~ O &-N-%
O OH

~ ~CH3 CH3 CH3 , OCH3 , OCH3 op Ph \ ~ H2N CH3 Q Ph p\ \ ~
O N~N ~ ~OxN~N ~
H OH ~CH3 O H OH ~CH3 Ph OCH3 Ph 0OCH3 H
H N CH3 ~L O H2 N N~, z O N~N ~O y O NN ~
~ H OH CH3 0 H OH
~ ~CHz ~OCH3 Ph OCH3 Ph p {
p p { , H3Cu N CO N N;S H3C N~ O N N~ \
II ~ O y = O
H ~-H
0 O NH2 OH ~CH3 O OTNH2 OH CH3 Ph CH3 p Ph p {
N~N:4 \ OCH3 H3C C p~N~N g\ I pCH3 H3Cp~ H O

H OH ~CH3 0 /
C4 H 0 Ph 0\ ~{ CH3 0 Ph p\ ~ ~
CI N- '~ON~N.~` OCH3 H3CO~O~N~N.~ 0 ` OCH3 -ly H ~ H3 0 H
0 OH OH ~CH3 ~

3 Ph // Ph a H3Cp C p~N~N ~ O ~{ OCH3 F3CUN f.pN~ 44OCH3 H
II
0 H OH ~CH3 O H OH ~CH3 O Ph OOCH3 0 N~pJ+~N~ 4S~ OCH3 H OH ~CH3 H OH ~CH3 CH3 CH3 CH3 Ph Q ~ I CH3 Ph 0 O ~{
H2N pJ~N~N.SOCH3 HZN pJl N~N4 OCH3 ~ H OH ~CH3 ~ H OH ~CH3 CH3 CH3 , ~ ph a Ph / {
HZN N~~N~ 4O OCH3 H3CUN O NN,~O OCH3 H OH ~CH3 O I I pH2 H
~
Cli3 CH3 Ph Ph /
H Q ~ CH3 0 II
H3C N~~ pN~N ~~ { OCH3 H3C~pJ~N~N OCH3 ~ - H O
p O~NH OH ~CH3 0 OH ~CH3 z ~
CH3 0 Ph CI H O Ph O ~ ~ 11 HaC~p~N~N- Q OCH3 CIyNO~N N,~` OCH3 H ~ O
O OH ~CH3 0 H OH 5.CH3 CHs p Ph CH3 O Ph / ~
H3CO~p~N~N. OCH3 H3CO~p~N~N;~' \ OCH3 H p H O
O OH ~CH3 O OH ,.CH3 O CH3 O Ph O CH3 O Ph aOCH3 H3CO~O~N~N" ~` \ OCH3 H3CO " O NN'O
H OH CHS H OH ~ H3 p Ph p p p Ph ~
H F3CUN~~O N~ 4- \ OCH3 O~N N S` \ I OCH3 II -]C~ O
H O
OH ~CH3 H
O OH ,CH3 op Ph a CH3~ Ph O N~N~S~ OCH3 H2N~O N-- N' OCH3 H OH CH3 0 H OH ~ H3 ~
Ph ~ Ph HzN ~N~N ~\ I OCH3 HZNu N~~ O
~ \ I OCH3 H p ~I p NN ~ O
O OH ~CH3 O H OH ,CH3 Ph Ph p\ \ p p H3CUN O N~N ~ OCH3 H3CUN~~ ~ S OCH3 H II O N~N ~
O p 3 H2 OH 55.CH3 O O~NH2 H OH 5,.CH

~
CH3 0 Ph O ~ CHs O Ph O ~ ~
HsC~p~N~N.~ \ OCH3 H3CN N'~ OCH3 H ~ O

C Ph p CH3 1N Ph p CI ~N~-p~N~N.~ OCH3 H3CO~pJ~~N.~ OCH3 CH3 O Ph / ~ 0 CH3 O Ph / ~
\ \
H3CO~O~N -(;- N ~ OCH3 H3CO~O~N~N,~O OCH3 Ph Ph ~~3 ~ ~ \ I OCH3 F3C N~~ OS \ I OCH
H3CO O N~N p y O N~N `~ 3 CH3 Y",CH3 p O Ph O~ OCH3 O J~ Ph O \ I OCH3 0 0 N~N p 0 N~N 0 H OH H OH

o 3 Ph p /( CH3 0 rPh p / ~
\ OCH3 H2N N~N;Sy` OCH3 H2N pNJ: N.S~\

0 Ph p Ph r 0 ~

HzNUN"" O N N~0 \ OCH3 H3CUN pN~T^N.S OCH3 IOI H~OH IOI O NH H O
OH
z Ph p / CH3 0 rPh / ~
Sy\ OCH3 H3CAp~NJ~~N'~ \ OCH3 N _ ~~ O ~ N N: H

0 O-~'-NH H OH O OH ~CH

Ph Ph ~
H3C~p~NJ~~N \\ OCH3 CI C~N~~p~NJ~~N \ I OCH3 ~ 3 ~ 3 Ph Ph O ~ I
H3CO~0~N~N OCH3 H3CO. \ OCH3 Q 3 Ph p \ I
` ; H3~ Ph ! Cri H3CO O N OCH3 H3CO~ 0 N r N,O OCH3 H OH ~CH3 H OH ~CH3 CH3 CH3 rPh p / ~ o~p Q Ph H
F3CUNO NJ~' N.SO \ OCH3 OxN~N. O OCHS
IOI H OH ~CH H OH ~CH

CH3 CH3 U'01 0 Ph ~ ~ CH3 O Ph Q ~ ~
N~N,~` OCH3 H2N~0 N~N OCH3 H O H O
OH CH3 0 OH ~CH3 CH3 CH3 CH ~ Ph ~ Ph aOCH3 H2N O N~N OCH3 HZNUN~, O N~N SyO'O II

0 H OH CH 0 H OH ~CH2 c3 CH3 CH3 p Ph ~ I Ph H

3CU0 N4S\ OCH3 H3CUNOJ~NLN ' II H I II H
0 0 NH2 OH ~CH3 0 O~NHz OH ~CH3 CH3 CH3 Ph N 3 Ph 0 N\~
CH3 0 p\ // ~~~ S CH 0 S/
H3C~ON~ NSO H3C~O)~ N~ N

O H OH ~CH3 0 H OH COH3 CI N 0 Ph O \ I S H CO CH3~ Ph O\ S
CI~ ~~O N~N 3 -,-J-O N-- N q\

0 H OH ~CH3 0 H OH ~CH3 CH30 Ph 0 N~ ~0 ~ ~, C H3~ rPh 0 I N~
S
~cp~O H S H3C0 O H 7^N 4 O OH ~CH3 OH ~CH3 Ph N Ph Q qH3 q ( I q H3C0OJ~NJ~~ 4~O S F3CUN~~OJ~NJ~~ O S
H OH ~CH3 0 H OH ~CH3 CH3 CH3 O Ph \ 0 Ph N

S
N~N~
~~OIN--N% S OJ~/
H OH ~CH3 H 0 OH ~CH3 CH3 CH3 cH3 q P" \ I N~ OH3 Q Ph q, <)-S
N
J~ s - J~ HzN~O N~N~S HzN-Tfl~O N N 0 -(~- O H OH ~CH3 0 H OH CH3 CH3 CH3 Ph N Ph H H
S H3C q S
HZNy NN~N'~~ y 0 N -(~~ N~~~
H OH ~CH3 0 O~H
O OH ~CH3 z H

N / N
H Ph O ~ S CH3 0 P" 0 ~ ~ ~
H3C II NO N~N O H3C~~O~N~N O
z H OH ~CH3 0 H OH ~CH3 0 O11~1, NH

CH3 CH3 CH3 0 Ph p\ CI H 0 Ph 0 ~ ~ N~
S S
H3CO~N~N'~`O CI~N~\O't~N N' O
0 H OH ~CH3 0 H OH ~,CH3 CH3 CH3 Ph N 3 Ph ~
T3 O \ ~ C_H O

~N
O~N~N. ` H3C0 O~N
H O ~ ~ H 0 0 OH ~,CH3 0 OH ~C%

Ph N Ph N
3~ p~ S ~3x p~ I S
H3C0 O N--N ~ H3CO
O N~N 0 H OH ~CH3 H OH ~CH3 Ph ~ N p Ph i N
~( S~
F3C N Jk 4\ \ S aOlll--N
O N
y ~ N0 O H OH ~CH3 H OH ~CH3 CH3 CH3 p Ph Nl Ph / N

~ J~ 4\ \ I S/ CH3 ~ O ~ I S
O N~N ~ HZN~O N~N ~
H OH ~CH3 0 H OH ~CH3 CH3 CH3 ^ Ph N Ph N

CHa }x{ O O ~ H O ~ ~ ~
S S
HZN~O N''N'% HZNyNO N 06 O H OH ~,CH3 0 H OH ~CH3 CH3 CH3 Ph ~N Ph N

p O ~ O ~
H3CUN 0 N~N,~p S H3CUN~~ IN~N;~O
O \ S
OH H OH ,CH3 IOI O~NH H OH CH3 2 2 ~
CH3 CH3 H3C CH3 0 Ph p\ ~ S~ H3C CHa O Ph O S

Y~OIkN~N, O ~~Ot`N-(~-N, CI N 0 Ph O \ I S/ H CO CH3~ Ph O ~ ~ S~
CI~ N~N ~ 3 ~O N~N ~
O H OH O H OH

Ph N Ph \
CH3~ 0 CH3~ Q ~ I \/

O N~N~ ~ H3C0~0 N~N

Ph N Ph ~-Il N
S
~~- 3 JL q~ F3C N JL q H3CO O H N'~ ~ O H J~N"~ OH O OH

0 O~ Ph N
O\~\ I ~\ 0 O Ph O \ ( ~\ N
~ S/ ~ S
O H N N~ O O r Hr^N~ O
OH OH

H N CH3~ Ph q H N CH31 Ph O ~ ~ ~
~ S S
z~O N N O N~N

Ph O N
0 0 Ph \ S H3CN N~N:S S
HZNUNOJ,N(~'N.~O O
IOI H OH

,~' IOI H OH L"~

Ph ~ CN Ph N
H C CH3~L 0 I S~ H C CHs~ O, ~ S~
3O N~N O 3 II O N~N~~ O
O H OH ~CH 0 H OH CH

Ph N\ Ph N

N t~ O S H CO ~ S
CI~ ~~O N~N ~ 3 ~O N-(~-10~
O H OH ~CH3 0 H OH ~CH

> >
Ph ~ N Ph ~ N
~~ I ~ Q~ q~ I
cH3~
H3co ~O N~N ~ s H3c0 ;H 3 s 0JN~N~ %
O H OH ~CH H OH ~CH

CH3 CH3 Ph N\ Ph N

~ 3~ q \ S/ F C N J~ q` S~
H3C0 O N~N % 3~~~0 N~N SO
H OH ~CH3 0 H OH ~CH3 N
l O N\/
~ ^ S ^
~ \i O Ph O
O Ph O O S

H H N O

CH3 CH3 H N CH3 Ph O S H N CH3~ Ph O`

2 ~ ~N~N 0 2 ~O J~N-%
O H OH ~CH3 0 H OH ~CH3 2 ~\ J~ Ph / N Ph z N~I S H NuN O H NS/ O H3CU~0 H N.S S
O
O OH ~CHZ 0 O NH2 OH ~CH3 CH3 CH3 , and S
Ph z NH3C N~\ J~ 4\%
y = O N H
O O~NH2 OH CH3 Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of ~ NHZ 0 / OCH3 \ , 0 I \
o \ \~ ~0 l~ O " / ~ R S \ O o \ /
0 N N'S O N N' 0 O N'S
H OH H OH H OH 1~

> > >
OH N N
0 \ I 0 O O \ I > O O ~ \ 0 H N. \0 aOH N.SO S aO~H N.S 0 OH OH OH
NH2 4ci OCH3 O
H 0 o o o ~ o ~ SJI~
O O ~ H OH N H O O H OH N `O O H OH NH > > >

O ' P-- OH N \ N

O .O N YHx ~'0 O O\~ H~ O `
N.S 0 ~N N'~S ~ S 'O N N"
~ 0 H H OH H H OH H H OH

\ NHz OCH3 p H O p ~/ H 0 \ p H 0 \ 0 p 'OxN N S 0 'ON N'SO O O~N N'Sp H H OH H H OH ~ H H OH > > IV\ \ OH N \ N

H O \ H O p ~\ H 0 p ~\ ~

p 'ON SO O OxN N ~ p 'ON N-O
H H OH H OH ~ H H OH

> > >

O
H H =-, J~ ~` I/
[=-,O N N O O N Nq~

\ O I/ O H O IO S
~ O H NqO ~ O H H 0 H O OH ~C~ OH ~CH3 H l ~( O N N O Q"O N N

O~
O NH2 H ao ~
I-/ "H O H N O H O H N O
OH CH3 OH ~CH3 CH3 CH3 H O ~( O N COH
H
~S
O H N O ~ O H N O
OH CH3 OH ~CH3 CH3 , and CHs Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula III:

R3,XxHN ~ ^ /NUX2 1 ~ ~ II

III

wherein, independently for each occurrence, Xl is absent, -0-, -S-, -NR- or X2 is absent, -0-, -S-, -NR- or Rl is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R7 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

~jO
-_clr0 O1 provided that when X1 is absent; R3 is not N R3a or N-R3a; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

0~o 0 N
provided that when X2 is absent; R4 is not R4A or N-R4A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and ^
-'0 provided that X1 is or X2 is R3 is amino, ~CO' v~` ' H3CN.H H3C)~ N.H H3C'k N.H H3CLNH F~C)~ N.H
, OH -0`~ H3C~~` H3C~` F1CH3C~ H3C~' F > > CH3 CH3 CH3 CH3 CH3 H C''NH H3C N.H H3C N.H

\
H3C~ ~ ~ 0 / I OH

CH3 H3C CH3 , or H3C CH3 ; or IZ4 is %CO~, F , H3C)~N.H H3C'kN.H H3C''N.H H3CkNH ~C~NH H3C~NH H3C)LN.H
H3C'--",-` H3C I / H3C~ H3C~ HbC H3Cr)~, > > > > > o ~
H3CJ~N.H

Or H3CPCH3 , In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein XI is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X2 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein Xl is absent; and X2 is absent.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein RI is OH.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is aryl or heteroaryl.

In certain embodiments, the present invention relates to the aforementioned O , OH
~
~` F ~ ) compound and any of the attendant definitions, wherein R3 is ~CO' v^

H3CNH H3C)~ N.H H3C'J~ NH H3CNH HC1~1 NH H3C'J~ N.H
H3C H3C~ ~C/~ H3 H3C~ H3C~

H3CN H3CJ~NH

H3C~CH3 , pr H3CPCH3 In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is alkyl, aryl or heteroaryl.
In certain embodiments, the present invention relates to the aforementioned O , OH

compound and any of the attendant definitions, wherein R4 is NCO' v` F~ I
, H3C~NH H3CNH H3C'k NH H3CNH H3Ct~N.H H3CN.H
O~ H3CH3C~ ~C~ H3C~ H3C~ H3C~
CHg CH3 CH3 CH3 CH3 CH3 H3CJ~N.H H3CJ~NH

H3CCH3~ , or Hs~C~~

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R5 is hydrogen.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R7 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R7 is alkyl, (cycloalkyl)alkyl or aralkyl.

Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of fonnula IV:

iR3,XN NuR4 H II

IV

wherein, independently for each occurrence, Xl is absent or -0-;

R3 is alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, amino, (amino)a1ky1, amido, (amido)alkyl, heterocyclyl, (heterocyclyl)alkyl, heteroaryl, aralkyl or heteroaralkyl; and R7 is alkyl, cycloalkyl, (cycloalkyl)alkyl or aralkyl;

0~o o O

provided that when X1 is absent; R3 is not R3A or ---CN-R3A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

~0 -_clro O--~o N
provided that when X2 is absent; R4 is not R4A or N-R4A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and s ~
provided that X1 is or X2 is R3 is amino, ~CO , H3C~LN.H H3CN.H H3C11~ N.H H3CNH F~C'k N.H
, OH
\ I O H3C~ H3C~ H3C~'~` H3C~` ~C
F ~ CH3 CH3 CH3 CH3 CH3 OII

H CN.H H3C N.H H3C N.H

CH3c H3C CH3 , or H3C CH3 , or R4 is H3C0~
H F~

II

H3C~N.H H3CJ~N.H H3CJ~N.H H3CJ~NH 1~CJI~N.H H3CJ~NH H3C N.
H3C~ H3C~ H3C~ H3C~ H3C~ H3C~

> > > > > > >
H3CI N.H

Or H3CPCH3 =

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X1 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned O , OH
~
~ F ~
compound and any of the attendant definitions, wherein R3 is %CO' v^ ~ \

H3CNH H3C)~ N.H H3C~N.H H3C~N.H F13C'k N.H H3C)~ N.H
O H3C~ H3C~ H3C H3C~,` Fi3C~ H3C~

OII
H3CN.H H3C N.H

H3C CH3~, or H3CP

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is aryl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl (heterocyclyl)alkyl or heterocyclyl.

In certain embodiments, the present invention relates to the aforementioned O , OH
^ ~
' compound and any of the attendant definitions, wherein R4 is ~CO' v~` F 5 H3CN.H H3C1~1 N.H H3C'k NH H3CNH H3CNH H3CNH
O~ H3C H3C--~ II3C~ H3H3C H3C~

H3CN.H H3CI N.H

~
H3C~CH3 , or H3C CH3 .

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R7 is alkyl, cycloalkyl or (cycloalkyl)alkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent; and R3 is amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent; R3 is amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; R4 is aryl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl (heterocyclyl)alkyl or heterocyclyl; and R7 is alkyl, cycloalkyl or (cycloalkyl)alkyl.

In certain embodiments, the present invention relates to the aforementioned ~ ( compound and any of the attendant definitions, wherein R3 is F H3C Oy CH3 \ ~ NH
F \ / OCH3 O H3C
CY, HO .

3 NH /\O O 0''~ 0 H3C ~ I c C H3 O p H3CO~
> > > > >
H3C'U, N.H H3CN.H H3C'1~ N.H H3C~NH H3CN.H
/ OH
~CY~
\ I OI /~, H3CH3C~ CH ' 3C CH H3C CH CH
F ~ CH3 3 3 3 3 0 H C)~N H H3CNH H3C N-H
H3Cyly ~ P
CH3 H3C CH3 , or H3C CH3 In certain embodiments, the present invention relates to the aforementioned ~ OCH3 ~
compound and any of the attendant definitions, wherein R4 is F
~ Q H3C Oy CH3 H3C Oy CH3 O H3C NH H3CNH ry--0) OCH3 .~,,,,, -> >
H3C O\\/CH3 H3C O\'CH3 H3C O\\/CH3 ~ O H3C NH H3C NH H3C\ NH

, H3CCH3CH3 H3C~CH3 H3C CH3 H3C~CHg HCOy CH3 H3C 3 NH ~ NH
O ~
~OCH ~ N O N
~ O OCH3~ O 3, H H, O
OH O H3CN.H H3C)~ N.H H3C)~ N.H HCNH
~
\ I l'ol H3C---/ 3 y H3C~~ ` Fi3C~ H3C
H3CO F CH CH3 CH3 CH3 OI~ OII

CN.H H CN.H H3CNH H3C N.
~ 3 ~ ~
H3C\~,.~ H3C
ICH3 , CH3 H3C CH3 , or H3C CH3 In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R7 is ~CH3 , or ~

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R7 is Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of H C Ph H3CICH3 F Ph H3CvCH3 s N~N N7N0CH3 F /\ N~N N~N~CH3 ~O H OH Ph0 H ~O H OH Ph0 H
O , O

F3C O Ph H3C,_,CH3 fph N

= N~HCH3 O N N~O O
N
N I H
O OH Ph0 H OH O
O , Ph Ph H N Ph H N

ON N~ ~O

CH3 Ph N Ph O H OH ~.......0 OHH OH !f 0 it. .~ NJ1. ~.,.
~J=, ^.1fN` H N
F Il H
C) Q 0 0 , .._ ,...._( = ~:, ri, ~/f ....._..._ ; ._..._...
n L/
0'H OH ~....0 NH H OH ~0 ~ ;u.v Q.... .
N 1 N. '~. O
y ~ IT = 11 r ~ li ~
O_..l O
F~ ,; ,, =.
.... t .
, ..... . , C) Fj {J ~i NH H OH / O "wN H H OH >...._ 0 N, N
H
0 O H C) '.
......._.~
.__ .~

N., N
H 1i O ~. ._ ... .. ~
O O O

\:........:F , ,..........: , h_..., r~...-.. ~~
-,r O iH OH ~.. U 0 H OH j.....-...0 ll'NH H OH ~.....-c, ` ,%
'N 11, ~ u t = II
) N N^ N = N/
F' ~f II
t~ = H U G H ..O = 0 H
e e ~

) I =j ~
NH OH OH ~ 0 NH OH O
{,J Nill, N..`- N J~' I ~ N.ti ~=~ r N. ~~\ \
C) H H H O
,...._~' , `,:........:F ,.........t \.........:7 _ ;.........\ F' ._...:\ %.

0 UH ~......... ~~.,~ OH OH O =
0~, N N.. F'' I=~N. H

IOI._._~ H HN. i C) ,l HN
~ ;..... 1(^
0 iI 0 ,........., ;,_, ;........,~
= \, ?i H OH 0 , NH H OH ~,.C =
N , J. N
~~. - ~ N = I ; ~l ",.~~ N
H H
HN, = C) HN =.
- -_=' ~i(' :-_l ,fT
.......... %' =
, ........... , ,~ .
O
K
NH OH 0 _ NH OH ~-(7 N, H
~= _ U -:-.H.. H~
o HN...II 0 H- ~
= _l O C
õ ,: .. ,.
~
~ , ,`- ,`=

NH H OH C OH
. , ~. , i~ . N.- ,,~= ~~ ,:., ,r' Jl~ N.
. ... . ...:
C " HN,. ,-' O l " HN ,~-. 11 ~...... li o 0 ~.., . ........._, , OH OH H OH ~-O
F N N, j.. N-.JJ. ~. . N.
, ~.. , . , O H "
HN 0.-1 HN, C

,__. .........., 11~ j/ ~=~
O i C
j..-,~ ~ ~....
NH H '?H NH OH 0 H
, ~ . .,.
N, = = - . . ' . ra' = C) H HN., H H. 5 ~;... i , ,._. , .

0 NH OH NH OH ~.....-...(~
N..1.~
H ^ N=. ~`., N: ~J. ~ ~ ~. H
H H -HN, i O HN.. i ;. l i 0 {, } 0 , ~...........: , ,.........~~ , C H OH 0 t~H OH ~.._.-...0 N ~~= N-'~J 1 I. ~ N\ix N:
..,....
F.
H =
HN,. H
~ ' O HN-,Cr ^f r , v., ? l? ! ; , -.......... ~ ,_~

0 0 (.}' OH ~n 'NH H OH C' NH H OH - U
N.ti .~ i. N.A tiTI, N,ti r~- N~~=, N N ll ~^~
C) H HN. = 0~ H HN1i H HN.
~~-' 0 p :. :
: ti,.... .
c) c) - ,1 - i JH OH ~...._( ~
H II ~ NH OH O
J. "
j N
H H
0 l~. v s. .
HN ~ O H HN
i, ~
~

and `:

Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula V:

R3, XxHN ~N~X2R4 Rl R5 V
wherein, independently for each occurrence, X1 is absent, -0-, -S-, -NR- or X2 is absent, -0-, -S-, -NR- or R, is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

o 0 provided that when Xl is absent; R3 is not 'R3A or -N-R3A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

~lro o o provided that when X2 is absent; R4 is not R4A or -1N- R4A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X 1 is or X2 is or R3 is amino.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X1 is -0-.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X2 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent or -0-;
and X2 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein Rl is OH.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl or heteroaralkyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is heterocyclyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is (heterocyclyl)alkyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R5 is alkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X, is absent or -0-; X2 is absent;
RI is OH; R2 is aralkyl; R3 is heterocyclyl; R4 is alkyl, aryl or heteroaryl;
and R5 is alkyl.

Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula VII:

R3.XJ~HN N.NYX2 R, R5 0 VII
wherein, independently for each occurrence, XI is absent, -0-, -S-, -NR- or X2 is absent, -0-, -S-, -NR- or R1 is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

~lr0 p-~p N
provided that when Xl is absent; R3 is not R3A or N-R3A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

O--~p N
provided that when X2 is absent; R4 is not R4A or N-R4A; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and ~ ~
provided that X1 is ~; or XZ is or R3 is amino.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X1 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein X2 is absent.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein Xl is absent; and X2 is absent.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R, is -OH or -NH2.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R2 is aralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R3 is (amido)alkyl or heterocyclyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R4 is (amido)alkyl or heterocyclyl.
In certain embodiments, the present invention relates to the aforementioned compound and any of the attendant definitions, wherein R5 is alkyl or aryl.

One aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula XII:

R3_N/~H N.SO2R4 ~0/ OH

XII
wherein, independently for each occurrence, ~ \ / ~ \ /
R3is > >

~ \ / OCH3 ~ \ / \ / ~ \ /
> > >
O OH OH
CH3 H3C H3C O HO / \
CF3 CH3, H3C~ ~
O CH3 HO,, N N ~+CH3 OH
H3C Ci Ci \ / CH3 O ~CH3 H3C~ O H3C ~ O H3C O O
CH3 NH N HN H f! H2N
CH3, H3C CH3 > > > > >
H3C-N CH3 / ~ - N}-" H3C~S~
N HsC

> > > > > > >
q---H
CH3 - f - 02N H2N I`N

N~

HC F F F

H3C>-N hi H O\O O H
H3C O2N ~ S' ~---N

H3C // H3CO L or H
> > > > >
O H

F

1S \ / OCH3 F\ / ~ \ / NH2 1 \ / OCH3 R4 >

O

F CF3 / CF3 ~ \ / N" CH3' -S~ -~ -OI -O,N N
"~ \ / " \ /
N OH
> > > > > >
H H
SYw YW "~Ny CH3 - " " II CH3 N ~N O O ~S
or \ / O
; wherein, independently for each occurrence, W is selected, from the group consisting of -NHR7 or -NHR(CHz)pN(R')Z; R7 is selected from the group consisting of hydrogen, alkyl, aralkyl, heteroaralkyl and acyl; and p is 1-10 inclusive; and R6 is F-~-CH3 or F--~-CH3.

In certain embodiments, the present invention relates to any of the aforementioned H3~CH3 N
H / \
compounds and any of the attendant definitions, wherein R3 is - ~, H H / \ F / N F3C / N
- ~ - ~ - > > > >
/N~H3C S I)H H3C N N

o > > > > > >

02N H2N ~ N ~N
/
~ ~N
0-~ 0-1 H3C H3C \
- -F , F , F , F , / \ , OH

F r \ ~
r \ ' OH O H
H3C O HO HO,; OZN/ \ H3 C N/ \

O H O H
~ 4S'O ~- H3C S-N
H3CO or O

In certain embodiments, the present invention relates to any of the aforementioned \/ \/OCH3 compounds and any of the attendant definitions, wherein R4 is ~
F
- F F O

\ / 1NH2~ JO ~CF3 F \ / ~ \ /OCH3' \ / ~ \ /

N O\\- S~
I
CF3 ~ \ /NH CH3 pH O \ / N OH N
> >
O O O, O
N ~ C I O N or > > >

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is CH3.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is CH3.

Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of:

H3C--~ O \ a O H3C/ O
HN O > N O II >
N~ O H ~ \~O
- ~O H OH ~~ F.~3 N O H OH ~CH3 H3CN O \ / OCH3 H3C~N O \ / OCH3 H O. I H O.
N~H N ~O ~H
J N,S`O
O OH CH3 ~O OH CH3 ~(CH3 / ~(CH3 H3C \N O 0 / N H3C \N O / N
N q~~ ~ I S N q~ ~ I S
O H N ~ 0 H OH N O
OH ~

O O

H3C~ O \ ~ / NH2 H3C~ O \ / NH2 N
N R o I ~ o H N'H N'o H /\ N H N~~o O OH ~CH3 ~O OH CH3 H3CN 0 O H3CN O O c N NOCH3 OCH3 H O _ H O
tO OH ~CH3 ~O OH ~CH3 H3CF6 O O COCH, H3C ~
O O N~~ N 4H N//\\~~ N N:5 \ I OCH3 O H OH CH3 - O H OH ,OCH3 O ~ O

H3c o ~ I ~ o H3c o I
I J
/\ N O~ O HN O`S.` O
HN

O OH CH3 O H OH ~CH3 N
H I N N O OCH3 H N~H ~ ~O\ OCH3 - ~O OH ~CH3 ~-O OH ~CH3 H3C O ~( ~ OCH3 H3C OCH3 N O ~ N 0, H NH N=~O\ H NH N~O~
- ~O OH ~CH3 ~O OH ,CH3 HAN <)-- N H3C~N , N
O O. ~
H N~H N%.OS H /\ N~H N.O s ~-O OH ~CH3 - ~O OH ~CH3 H O~~ H O~~ ~ I

- ~O OH ~CH3 ~O OH ~CH3 H3C Or OCH3 H3C O , OCH3 H N ~\HN N o ~ I
N'' N

O
H2N O ~ cc O H2N O \ a - :~ O:\ , O,\ / O
N~N N S O NN N\O
H H
- ~O OH ~CH3 ~-O OH ~CH3 H2N O \ p / H2N O \ O
N;~p\ OCHs N~H pOCH3 N H
/~-O OH CH3 - O OH ~CH

HZN O \ p / I NHZ HZN O \ p / ~ NHz .. \ \
H N~p H N~O
0 0 OH CH3 O O OH ~CH3 CH3 CH3 ~N O \ / OCH3 O \ / OCH3 N O;~ HZN N p`~
N~H N p N~H N p O OH CH3 - ~O OH ~CH3 H2N O \ p ~/ I N O
H2N p / I N
:-; v S `S
N~N N p Z"rlN N
/\/- 0 H OH ~CH3 O H OH ~CH3 HzN 0 J \ p / H3C-N O \ /
N~N N-S. OCH3 /~ N N. OCH3 H H
O OH /CH3 O OH y CH

~C"H3 CH3 NCH3 O~ H3C-N CH3 IO 4yo O
H3C_ ~
~0 -~N~H N p N N p O OH ~ H OH
CH3 ~O ~CH3 CH3 H p OCH3 CH3 I OCH3 H3C N p I H3C N p . \ ~ \
N N-~ N~N N-~p O OH ~ O H OH
CH3 ~CH3 H3C-N CH3 OCH3 H3C-N ~
.~ \ . \
N H N 0 N^~ H N s~o 0 OH ~
~CH3 ~-O OH
,CH3 H3C- O H3C- o / I
N ON:~O\ OCH3 O H OH CH3 - ~O H OH 0 0 ~CH, N~H N o NN N O
O OH - O H OH
CH3 ~ CH3 N ,CHa H3C- C ~ / I H3C- o / I N~
\ .. \
/-~ H NO S N~H N~O S
/~ N
- 0 O OH CH3 ~O OH ~CH

O a o>0o>
N O / N
F N~H N O F N N O
~~O OH CH3 ~ H OH CH3 N o \ I N o F L NH N'SO F NN N'~o ~O OH CH3 ~O OH CH3 CH3 CH3 \ I / I N \ I / I N

N O~~S/ N O~ S~

O H OH CH3 ~-O H OH O O ~,,,CH3 P~o NH2 NHz C~' O-N O
FN~ N H N F _ N N N o O p CH3 CH3 \ I \ a / N ~/ IOCH3 / N
\
F N~H N p F _ N~N N~O 0 CH3 0 OH CH3 ~-o H OH CH3 \) /~
O. o> O o N
~p N ~
F3C N~H NO F3C N NO
/~
J
~ ~O OH CH3 ~ H OH ,,,CH3 O ~ p ~

CH3 CH3 \ / OCH3 \ I / OCH3 p~\ / N p,. \\ ~

N o F3C NN N~o J J
H OH CH3 O H OH ~CH3 O ~ p /I /
~ \
N p I
F3C / N 0O \
H OCHa F3C / N/ H N0-~ OCH3 \ p J
O OH CH3 ~O OH
O ~ p ~CH3 $ `CH3 CH3 N I N

/
J -S\ g / N S
F3C NH N 0 F3C _ N"'~N N :S`O
O OH CH3 ~-O H OH ,,,CH3 O ~ p CH3 LCH3 \ / NH2 P
N

o-/ N o;
F3C NN N p F3C NN Np ~O H OH CH3 ~O H OH ~CH3 O ~ O

CH3 `CH3 -48-i I p\ / o N O:~ p/ N ~ \
N~H N O N~N N~O
~O OH CH3 ~O H OH ,CH3 H3C Q H3C Q ~

, OCH3 JD\ , I OCH3 N ~
. \ N ," v N~N N p N~N N p ~/O H OH CH3 ~~-p H OH CH3 H3C Q ~ H3C 0 ~

N N NO OCHs N N N0 ~O H OH CH3 ~O H OH CH3 H3C p ~ H3C p N\ N
N N O~
s ~S
N~N NQ N/ N Np ~O H OH CH3 ~~-O H OH CH3 H3C O ~ H3C Q ~

NH2 , NHZ

/ N N Q
N N` O N/ NO
O H OH CH3 ~~-O H OH CH3 H3C p H3C Q ~
CH3 `CH3 O
Q \ / I O>
N .~ p / N p H N-~p ~ N~N N-~p ~Q OH CHs ~-O H OH ,,,CH3 H3C CH3 O ~ H3 CH3 C 0 CH3 , CH3 I aOCH3 p%,/ N pCl {a N N p N NO
- ~O H OH CH3 - ~O H OH ,,,CH3 \( / I N\ \ / I N
O O%5 N~H N p N p O OH CH3 p H OH ~CH3 / / NHz \ IN
O /
NN N=S,~\ Ni\hN ;p\
H - / H
O OH CH3 O OH , CH3 3 H3 H3('+YS/ / I p\ H3C!S/ / I O

N O O/ N Q\
N p N~N N
H
~O OH ~CH3 ~p H OH p ~

,O' ~ p,~ N p~ \ ( N
N N O N N N'~, ~ H H
OH p OH
CH3 ~ O

~

CH3 CH3 H3CYS~ / I H3CS/ \ Q

O:" " OCH3 N OCH3 N~N N p ~N N p p H OH CH3 O H OH ,CH3 ~

H3C N s / I N H3C-S/ \ ~ / I N
O~.~S N O~
-N~N N p N~N N p p H OH CH3 p H OH ~CH3 H3CS \ 1 NH2 H3CV1S / I NH2 O ~ ~ O, ~
N \
N . \
N H NO H N~O
0 OH ~CH3 ~O OH ~CH3 N%\ S 0 N%~ / 0 o ~I o NN N O NN
0 H OH CH3 ~ H N O

CH3 CH3 N~ OCH3 N~ /
( OCH3 `
N~H N N'~O
O OH ~CH3 0 H OH ,,.CH3 ~
CH3 CH3 N~

0 OCH3 - ~ $~/ I
~OCH3 N/ H -((~N 0 NzN N 0 O OH ~CH3 0 H OH `,,,CH3 CH3 LCH3 N~ a N\ N~ ( N~ 0 ~ S ~/ ~ ;~/ S

NH N O N~N N 0 ~

CH3 CH3 N a NH2 ^ NH2 N s ~\ \ I
NH N~O N~N N ~O
O OH ~CH3 ~ H OH ,,.CH3 CH3 CHa \
CH3 / I 0\ CH3 0/
N \ ~~/v'0/ \ 0' IN 0 N~N N O
S ~O H OH CH3 H3CS ~O H
H3C OH ~CH3 O ~ O

CH3 O aOCH3 N_NH N;$, N\\~N~N OCH3 /-/
H3C S ~0 OH ~ H3 H3C S H N
OH ~ H3 O ~ O ~

N
CH3 \ ( / I \ CH3 \ I / I
\
C~N N \ ~S
IH N p I-N N 0 H3CS ~O OH CH3 H3C)S ~_ O H OH ~CH3 O O

~~ \
~N H N O NH N ~O
H3C S /- O OH CH3 H3CS /~- O OH ,.,CH3 CH3 CH3 \ I / NH2 \ I / NH2 H3 p ~ H3 Q .~ \ Q~
N NH N N,p NN\~N N, / H O
H3C S ~-O OH CH3 H3C S /-O OH ~CH3 CH3 CH3 q O

N \ ~ O~
H N~p O N H N~p O
O OH CH3 O OH ~CH3 CH3 CH3 - \ ~ / OCH3 OCH3 N\ /
J p~ \ ~ p\S\
NZ H N p N N' H p ~-o OH ~CH3 ~-O OH ~CH3 \ o 0OCH3 \ ~ J
N \ I OCH3 NOO H OH N,~p CH3 ~-O H OH .,,CH3 O ~ O ~
CH3 CH3 q\/ N N I N J \~ Q\ \ ~ S

N~H N.S~p S N~N N.,p ~O OH ~CH3 ~p H OH .CH3 \/ N 0 H NSp~ OCH3 NNH
Z ~ p ~
N:~O OCH3 N
/~-p OH ~ /CH3 ~p OH ~ /CH3 ~C"H3 C" H3 ft$' p O
O N ~ \ / ~
~~ ~ v `O
, ~/
NH N NH N p O OH CH3 ~-O OH ~ CH3 ftNNNSOCH3 ~ N p N~H H p OCH3 O OH ~CH3 O OH ~CH3 CH3 CH3 qN/' , OCH3 - / OCH3 J J Q\ \ N/ Q. \ I

NH N' ~p NH N~~p O OH CH3 ~O OH ~CH3 CH3 CH3 / ~ I:N / N

N 0 \S N ~ ~
\
N~H N N~H N p O OH ~CH3 ~O OH ~CH3 NQ N/ O..
N"~'IH N,p NH N p O OH CH3 ~-O OH ",CH3 CH3 CH3 ftNN I

\ p,~ o o O~

O OH ~CH3 O OH 1CH3 p~ p~
N/-~NH N ~

O OH ~CH3 ~p H OH CH3 ~

CH3 CH3 qNH N;S O

p I pCH3 H OH CH3 ~O OH CCH3 O ~ J
CH3 CH3 N \ / N

O
~' `S q-/'~N S
N~H N p NN 0 ~
CH3 CH3 p O ~ \ ~ / I p N ~N ,~/`O
N N O - N~N N O
O OH CH3 ~~O H OH O ~ O CH3 N p- N p, 3 N~N N p NN N p ~O H OH CH3 ~o H OH CH3 ~
N ~. \ OCH3 CNN ;~" " ~OCH3 N N \ N O N N N O
H H
O OH ~CH3 ~O OH ~CH3 / N O~ S / N O~S
H o N N O
~O OH CH3 ~ H OH ,,,CH3 O ~ O ~

i O I
H3C 0 0OCH3 H3C 0 ONNN N ~ S
H O H O
O OH CH3 )- 0 OH ~CH3 CH3 , and CH3 Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of I , OCH3 , OCH3 ;~ Os ~ O_N/- o Os ~ I
H N' I N N 0 ~ OH 0 H OH
0// ~
\ / OCH3 / OCH3 O 0\
~ F F 0 0 \ ~

I N N'S\ N S

~

, , OCH3 0 \ , OCH3 F3C 0 OS \ O OS

O-N ~0 H OH 1-0 H OH
//

~
0 0 ~ ~ H3CO 0 OS ~ ~
OY I N N SO N f-~ N
N ~0 H OH b-N/
~0 H OH

0 ~ 0 Fb-N/ O 0S I O F_ ~ 0 ~S ~ I O
N NO F~N /I N N 0 O H OH ~ O H OH ~
Oro> 0 S I
F3C 0 oO /~ 0~N N/

N I / O
O OH ~O H OH
~ 0// ~

/ NHz 0 \ I0 ~ I 0\ - IL o \ I
f"~
0 S\ \ O/ ~N I` N N' N~ H OH N' O ~p H OH
~ ~ 0 \ > >

0 \ OS \ ~ F 0 \ OS \( ~0 OH b-N ~0 OH ~
i ^ /NHZ NH2 F 0 0~ ~ F3C 0 1 /--N S ~N N'S`

F ~/ ~ H-~~
OH N~p H

NHZ
, NH2 O
OS \ ~ 0 ~ ( I H N' 0 N I H N SO
0 OH ~0 OH

/~ 0 \ OS \ I F F /~ 0 oS \ I F
N I N N/ F~N I N N
b-to H OH ~ / 0 H OH
H\F , OCH3 I OCH3 F3~H p N/ s \( F
~N I N N \0 /

~-0 H OH 0 OH
and ~I
0 \
0 ~~
0 N~N N.SO F
~ / 0 H OH
~

Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of O \ OS ~ ~ F 0 \ OS ~ ~

H N 0 F~N I H N' F ~0 OH O OH

CF3 0 OS ~ I OH O OS
~ ~ I f-r:~ j( ~N I N N N N~O H OH 0_,,/-I ~-O H OH

F3CO O 0 ~ ~ _ O
N/
N I H N So \~ N I H
o OH 0 OH ~ ~ //

0 \ I o, / I O> O 0, ~/ ~
.O F /~~ `g ~~/ `O
F~N H NI F~N I H N/
~ ~-O OH 0 OH

CF3 O OS ~ I O OH O OS :I O
6-N/-~H
N j( N
N ~ H N ~O
O OH O OH
~ ~

N

F3C0 0 > 0 OS I
_ 0 N~N N' O F~N N N \0 ~ ~ 0 H OH ~-o H OH

NHZ
F 0 / I NH2 CF3 0 O~
_ ~I ` S
/~/~N N~S \ N N~
F N~ OI H OH ~O H OH
// //

OH O 0~ F3CO 0 0`

N Nb-N /~N NS`
~OH H
O
~ p ~0 OH

0 \ IO /
0 F ~
\ IO ~/ N N
SS b-N/-~H
;N H N NN N

~ ~

N N
0 0S S F O 0S S~
F~N I N N' F~N I N N' ~O
~ ~ ~O H OH ~ ~0 H OH

F p p ~ pF3 p p~

. S 6N/-~H
S F N
~N ~/ 0 H OH O OH
~
N~ \ I / N
F3C - 0 OS \ S 0 O ~ I S
~N N N N
~~ N p H OH F3C N~H OH
p//
/
/ N
OH 0 O\ \ \ HO 0 \ 0 ~H S S H S/ 'S
N I N N ~ N N
OH p //-p OH 0 0 0 / ~ O O \ IO ~/ ~ >

N N
O-N /~ ;5,~/\S /~ ;S
I N N N I N N ~0 H OH ~0 H OH

N >1-1 -N
F3CO 0 OS ~ S~ 0 - 0 OS
\~
~p H OH 0 OH
b-N ~NN/ p N H N/ \

~

0 ~ IO / I OH F 0 IO OH
~
-(J~~ .S~ b-N/ a-N/ I N N N N rO H OH O H
OH

OOH p OH
F \
F~N I N N SO F~N I N N SO
\ / rO H OH \ / ~O H OH

0 p OH CF3 0 / I OH
S N N N S \
~O
F \/ N0 H OH N' cb/ //-O H OH

F3C 0 0` \ OH p 0 / I OH
S
Z]~-Np H OH N.Sp F3C \/ N //~H OH
N\0 OH ~ / OH
0 OS ~ F3CO 0 IOS I
I N N 0 //~N N
// -0 H OH t/-N -O H OH
0 0 ~
/
O
OH ~ I ao 0 p / I 0 H O p>
`S ~ O L N N
'S \ N~N N O
NH OH 0 \/ -p H OH
' //
~/ 0 OH 0 ~N\\ O~
I O
lI
S HO
N/-/~N N SO \ HO
pl H OH N\HN OH
~ O//
0 p / ~ H2N 0 \ 0~\
H O S\ S \S \ S
N 0 " j( ~H

r r p 0 and Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula XII:

O
R3_N-^~N N.SOzR4 ~-O H OH

XII
wherein, independently for each occurrence, H3CS~ O H3C~ H3C, O O CH3 N HN JV HzN H3C-N
H
R3 is - , - , - ~ - > >
/ N N^S H3C S CH3 ~ (''H \`~z5> > H3C S

02N H2N ~-N H H3C >-N H
N ~ H3C 0-1 H3C N

F F F F

, OH OH

0 HO HO, - , ~ H3C ~ H3C ~ H3C

N S' ~N H3C-S-N
F13 C / \ H3C H3CO b-A, O , or F
OCH3 ~- F F
is ~ \ /OCH3 F\ / NHZ ~ \ / OCH3 ~ > > > > >

- - N- ~CH3 ~ \ / F> ~ \ / CF3> ~ \ / CFs> ~ \ /NH

- S~ -O~ OI 4IN
N_OH, , H H
SYW - OYW N I Ny CH3 N I Ny CH3 N ~N O O S
, \ / , or (::~; wherein, independently for each occurrence, W is selected, from the group consisting of -NHR7 or -NHR(CHz)pN(R')zi R' is selected from the group consisting of hydrogen, alkyl, aralkyl, heteroaralkyl and acyl; and p is 1-10 inclusive; and R6 1S ~~ H3, ~~3 CH3' O O N
~
N , or In certain embodiments, the present invention relates to any of the aforementioned H3C-N 0-~

/ \ compounds and any of the attendant definitions, wherein R3 is , F, HzN N N
H
L\-l H3C 0-~ H3C H2N O2N ~ ~

F F , F

H3CO p or In certain embodiments, the present invention relates to any of the aforementioned N~S H3C~S
N
compounds and any of the attendant definitions, wherein R3 is NZ N

, or In certain embodiments, the present invention relates to any of the aforementioned Hs~CH3 N
H
compounds and any of the attendant definitions, wherein R3 is - ~, H3C~ H3C O O OH OH

"
- ~ - ~ - ~ - ~ - ~ H3C O H

HO HO / \ ~C
"aC , HsC , or In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R4 is ~ \/OCH3, F

- -\ /F - - FOCH3 \ / O, 1-N"2 \ / ~ ~ ~-\ /~ 1 1 ~ F \ / O F \ / CF3 > > > >
O - S~
CF3 ~ \ /N~C"3 \ / O ~ \ / \
, OH, , N OH, O O O, ~ \ / \O
N
\ / N or >

In certain embodiments, the present invention relates to any of the aforementioned "3 compounds and any of the attendant definitions, wherein R6 is ZS--1\CH3.

Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of:

H3C , I

H / \ ~N N:~O\ OCH3 - ~O H OH I/CH3 O
H /\ N 4S~ \ OCH3 F3C /~ N~N N-C. OCH3 O H OH 1/OCH3 OO H OH y0 TCH3 CH3 O I ~~ N~ O I

-~N / N O:\~OCH3 O%C, OCH3 - NH OH N CO NH OH N O
~ ~ C'.H3 ~ CH3 ~
H3C O y 0 CH3 CH3 CH3 0 ~ I 0 O ~

~\\~N~H
N.~. \ OCH3 ~ H N N-~ \ 0 CH3 O
J
H3C S ~O OH O NO ~/CH3 H3C ~ OH `
C"/CH3 1H3 CH3 ~C"H3 H3C g I

Z Y~ 0 o I \ 0 o OCH3 O
CH3 CH3 i i O NH2 H3C O ~ NHZ
/ ~
HZN O O,\ \ ~ N O.\\
Fj H N H N-~
~-O OH ` /CH3 O OH ` /CH3 1C" H3 ~C" H3 CH3 H3 CH3 H3C~ a NHz 3C~ NHz N O ON O ~ / ~

H /\ N~H N/vO H N / H NSO
O OH y 0 H3 1C" H3 O / N H3C O \ I / N
H2N O O ~ N O O~
\ H \ \
N~H OH N S N O H OH NO S
/ ~ /CH3 ~ ~

CH3 ~H3 CH3 CH3 H3 CH3 H3CN O O I O I N H3C~N p 0 ~ N 1 10 ~S p. S

N~H N ~ H N~N N
O OH `/ 0 CI CH3 ~O H OH I/CH3 H3 C" H3 H2 a N p O\ H3CN O 1 O>
O
N~H OH O'~ // O O H /\ N~H OH NO O
~CH3 ~CH3 H3C CH3 ~~C~
~N O 0 O\ H3C O 1 /
H /\ O\~~O/ HN p p\\ ~ >
N I H N 0 N~N N'~p p O
OH ~CH3 - ~p H OH `/CH3 0 CH3 C"H3 H2N O O \ O. OH H3CN O O 0 ~ OH
\ ~
~~

~ ~CH3 ~/ OH ~CH3 H3C-~C O \ ~ / H3C CH3 N 0 OH H3C~N 0 O /
0 \ OH
H N,~O H /~ N N~

0 CH3 CH3 - O O H OH 1 ~/OCH3 ~ H3 C" H3 02N O O~ H2N 0 /~~
O.= \
.\\
H N~O O-N /'~-( H N' ~O
OH 1 /CH3 F O OH ~CH3 ~C"H3 CH3 -64-O NH O NH2 HsC NH 0 NHz /

H3C N O,`/\i H3~/ t~N ~-N~" OH N O NH OH N O
F ~ ~CH3 F CH3 CH3 CH3 02N O \ I ~ N~ H2N O ~~ ~ N

N ~' S
t ~ - S
L N/ ~\ H
N N
H
p OH CH3 OH CH3 F 1~ F O 1~

CH3 CH3 O H I , N H3C H / N

H3C 9 \ S> H3C /\ ~ ~ \ I S
Nl--r"" OH N o N/ I H OH N O
F p ~CH3 F 0 y CH3 J O> H2N 0 4)0>
/\ NN N~\ /\ N~N
H Np /% OH CH H
3 - p OH CH3 F O ~ F ~

CH3 CH3 O H I =~ p H3C H ( O

~ N/ \ ~\ p ;~
H3C p o H3C o ~" OH NI - 0 H OH N O
F ~ YCH3 F 0 1~CH3 02N O ~ pH H2N p OOH J
NJ( ~iN~' N NO ~H OH N O
" OH CH3 CH3 y F O

" 0 OH H3C ~NH 0 O\ OH
\

N H N'S.O H3C NN N-~ \
/ H ~CH3 ~/O " OH Y~ H3 H3C H3C J / ~ N~H N ~O NH N'SO

- ~-O OH 1 /CH3 ~O OH ~ /CH3 O T p ~"

y H3C OH 0 / N O / NuCH3 O~( g/ O` ~ ~ IOI
N/-~H
J
N'~O o-N~H OH N O
~p OH y CH 3 CH3 O 0O\ Ng O OH
J \ /
N~H Np N~H N O

OH y CH3 H3C ~ ~CH3 C H3 CH3 a_s p \\ /~ O, ~\ O
HO /
~ N O,~
~N N 0 F~N~~ N
pCH3 H OH CH3 O H OH y O y pCH

F O N 02N O p O O
O
/ ~ S ~~p F~N~
~O H OH N~ y p CH3 ~O H OH N `/ 0 Hg C" CH3 H2N O / ~ O O~NH p / p>
O p O H3C 0:~.0 / ~ N~N N ~N~N N O
p// H OH ~CH3 p// O H OH yCH3 H3CO ~N O O\ ~ 02N p p.~ ~ I
\
N N N, O
~O H OH ~CH3 ~O H OH 1/CH3 H2N O OCH3 O H O x \( / OCH3 ~N ~

H N O H3C L~-N H N'\O
J
O OH 1 /CH3 ~O OH ~ /CH3 ~C" H3 ~C" CH3 OH / I OCH3 02N 0 / I N

~N 0 ~

~~N O` ~ / ~ OS
I N N~O N O H OH N O
/O H OH 1 /CH3 ~ ~CH3 0 ~C"H3 CH3 0 H I

H2N O O. > H~N 0 O.. ~
/~ N~H N'~O S 3 /~ N~H N.~O S
~O OH `/CH3 ~O OH ~CH3 ~C" H3 CH3 NO O~~ H3C-S-N O
N C)c H3C0 NS O/~ H NO S

r OH 1 /CH3 O OH ~CH3 0 ~C"H3 CH3 qS.O O \ ~ / I OCH3 ~S.O 0 \ I / I O

O
N O
b NH N'~O H
O O OH ~ /CH3 /O OH ~CH3 ~C"H3 , CH3 sA O I N

NSI\
~O H OH O

1~
and oFb Another aspect of the present invention relates to a compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of:
~
~ OCH3 I , OCH3 0 ~ 0 ~ I F 0jI 0 ~ I
N NS F~~ N I H NSO
F N~ I0 H O \ OH 0 OH

/ OCH3 \ / OCH3 CF3 0 p~ OH 0 p \
N H NSp N H N
O
\ / ~0 OH ~-O OH IY

O
F3CO p \ Ip OCH3 0 1 0 O \ I O\ O \ IOS \ O
/ ~N N
NN N SO ~N N S p F \/ N~ H OH
H OH \/ Nyp H OH O
//

p 0 / p F 0 \ OS / > -CF3~ \ 0 \ I > -OH 0 0~ \ I /

_ O N H N.O 0 N H N0 F \/ N~H OH N 0 \/ OH \/ O 0 OH 1/
0 , +

F3CO Q \ OS \ ~ - O R S \ I
x 0 -Tx t 0 H OH \ / t0 H OH

F 0 OS \ - CF3 0 \ OS \ I 6N pH o OS
N/_y~_N N NI lj`N NI 0 F N N N
\ / yp H OH 0 H OH yp H OH + //

/
NHZ \ / N \ / N
S
F3CO b--~H-~- Q 0II O \ I S~ b-N Q 0 \( N/,N N SO N I H N SO /,}~N"Sb OH 1~0 OH ~pH
p OH

OS \ S F 0 F O O
N N
/~/ 0II~
N N. S~
F~N H N ~N N p "S S ~H
\/ 0 OH F \/ N0 H OH F N0 OH t o' O i Y
p \ / N N
\
CF3 OII OS S F3C O QS \ ~ S 0 O
N N H NI p _ g S
d-NY p H OH 0 pH F30 \/ ~O H NI
OH
i i 0 i , \

O \ I /( N\\ HO b-N~f H N I 2 S H OH p N y~H N O
O H OH O // O OH

0 OS \ S~ F3CO 0 p OH NI SO
H 0 ~/ N~H

\ IO ~ I OH

JI"~ b-N Sp N N p N~O Ory N ~H N 0 N~
0 H OH p O OH
0// i YI O
F p \ IpS ~ OH F p pS / I OH
1I \ ll . \
~O H OH N~0 F ~/ Npl H OH N 0 i ~
pH 0 \ ~p / OH
0II 0 \ I CF3 oS \
F N H N SO ~N H N
/ O OH ~ / 0 OH
p O

0 OH p O I OH
F3C II \ II ~S
N H N SO F3C-N H N\0 1~0 OH \r 0 OH

p p \ p , OH F3CO p p \ I oH
II \ N~N NS
N H N b'-j yp H OH / 0 OH //

+

\ I .S~OH OH \ I S OCH3 O 0~ \ I 0 0 ~N N ~ H N
p H OH O p OH IY // 1 >
H
N 0 N H N p ~~ OS \ 0 ~ pS \ S
r `
// IT T I

/ N

/
~
O'I p \ ~ 0 0 0 \ ) S
P
HON H SO HO ( -H SO
~ / 1~0 OH ~~/ 0 OH
// , and 0 Another aspect of the present invention relates to a compound, or a pharmaceutically acceptable salt thereof, of formula iso-XII:

I
O
R3-N N NSp2R4 "-)"., 1~
~O H OH
p Rs iso-XII
wherein, independently for each occurrence, R3 is alkyl, (amino)alkyl, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, heteroaryl, aralkyl or heteroaralkyl; and R6 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R3 is aryl or heteroaryl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R3 is aryl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R3 is1--0,~ O ,1--0, &F J-0 F ~ &OCH3 , , , O OH

~ O CF3 ~ c O
OH

O HO / \ I HO / \1 I ~ N
CH3, H3C H3C Cl Ci O H3~C~
~-- O

~
, CH ~ ' CH3 H3C ~ - ~
~~3 H3 ~ ~ N` CHg N

H3C~ H3q O O CH3 N fl H2N H3C-N

H N N
- ~ - > > > > >

/ NS ~ N/\\ 5 \ / N /
H3C ~-`-~ S
H3C ~ CH3 > > > H3C > > >

O2N HzN ~-N H H3C N H 0 0-1 H3C 0-~ H3C 0-~ 2N ~-"

F F , F , F
O
4S'O H O H
~-N H3C-S-N
H3CO / \
/ \ or In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R4 is aryl or heteroaryl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R4 is aryl.

In certain embodiments, the present invention relates to any of the aforementioned \/ \/OCH3, compounds and any of the attendant definitions, wherein R4 is ~
F
- F F O

\ / ~ ~ 1O ~~F ~CF
F \ / "Hz~ \ / OCH3, \ / ~ \ / ~ \ / 3' O - S~
~ 0 - 0-~~ N
\ / CF3 i \ / "H CH3~ H~ \ / N-OH, \ /
O O O," N S~w OYw " '"I Z \ / I"I

H H
- NO YNy CH3 - N IS N II CH3 O O
\/ or ~\/ ; wherein, independently for each occurrence, W
is selected, from the group consisting of -NHR7 or -NHR(CH2)pN(R7)2, R7 is selected from the group consisting of hydrogen, alkyl, aralkyl, heteroaralkyl and acyl; and p is 1-10 inclusive.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is alkyl, heterocyclyl, aryl or heteroaryl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is alkyl.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is CH3,CH3 , CH3, ~ ~ I ~ ~CH3 , CH3 N
H

~ ~ /N' ~HJ~0 or ~0 In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is CH3.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R6 is CH3 or F-~--CH3.

In certain embodiments, the present invention relates to any of the aforementioned compounds and any of the attendant definitions, wherein R3 is aryl or heteroaryl; and R4 is aryl or heteroaryl.

Synthesis of Selected Compounds of the Invention. The protease inhibitors I, II, III, IV, V and VII can be synthesized using the synthetic schemes outlined in Figures 1 a-b.
The definition of each of the variables may be the same as shown in formulae shown above.
Protease inhibitors I, II, and V can be prepared using the synthetic scheme shown in Figure 1 a (top). As shown therein, an epoxide, for example, can be reacted with an amine in a stereoselective manner to yield amine 2. Amine 2 is reacted with sulfonyl chloride or an acyl chloride to yield 3. Deprotection followed by reaction with an acid chloride, for example, yields inhibitor I, II, or V.

Protease inhibitor III and IV can be prepared using the synthetic scheme shown in Figure 1 a (bottom). Amino acid 5 can be converted to amine 6 using standard synthetic procedures. Reaction with an acid yields amide 7. Deprotection followed by reaction with an acid chloride yields inhibitor III or IV.

Protease inhibitor IV can be prepared using the synthetic scheme in Figure lb.
As shown in the scheme, an epoxide, for example, can be reacted with a protected hydrazine in a stereoselective manner to yield hydrazine 9, after deprotection. Hydrazine 9 is reacted with an acid to yield amide 10. Under certain conditions, further deprotection yields amine 11 followed by reaction with acid chloride yields inhibitor VII.

As can be seen from Figures la and lb, the R groups of the inhibitors are determined by choosing suitable reagents and starting material. Similarly, the stereochemistry of the inhibitors is determined by choosing appropriate starting material and reagents.

Protease inhibitors XII can be prepared using the synthetic scheme shown in Figure 15a (top). As shown therein, an epoxide, for example, can be reacted with an amine in a stereoselective manner to yield amine 2. Amine 2 is reacted with sulfonyl chloride or an acyl chloride to yield 3. Deprotection followed by reaction with an acid chloride, for example, yields inhibitor XI, XII, XV or XVI.

As can be seen from Figures 15a and 15b, the R groups of the inhibitors are determined by choosing suitable reagents and starting material. Similarly, the stereochemistry of the inhibitors is determined by choosing appropriate starting material and reagents.

For example, chiral N-phenyloxazolidine-5-carboxylic acids 9 and 10 used in the synthesis of designed inhibitors, were prepared following the literature procedure as outlined in Figure 9. (Brickner, S. J. et al. J. Med. Chem. 1996, 39, 673-679;
Hester, J. B.
WO 2003/006440, hereby incorporated by reference; and Thomas, R. C. et al. WO
2003/072553, hereby incorporated by reference). The intermediate chiral alcohols, 5-(hydroxymethyl)-3-aryl-oxazolidine-2-ones 7-8, were obtained from substituted anilines in two steps. The reaction of CBZ protected anilines 4a-g with either (R)- or (S)-enantiomer of glycidyl butyrate promoted by n-BuLi provided chiral alcohols 7a and 8a-g.
This one pot, three step cascade reaction involves the initial ring opening of chiral epoxide with N-lithium species followed by an intramolecular cyclization and finally an in situ ester hydrolysis. (Brickner, S. J. et al. J. Med. Chem. 1996, 39, 673-679.) Oxidation of the resulting chiral alcohols using catalytic ruthenium chloride provided the desired N-phenyloxazolidine-5-carboxylic acids 9a and lOa-g (Scheme 1). In case of unsubstituted phenyloxazolidines, both (R)- and (S)- enantiomers, 9a and 10a respectively, were prepared from the corresponding chiral epoxide. All other compounds with substituted phenyl ring, 10b-g, were prepared only as (S)-enantiomers.

The synthetic route applied for the preparation of designed protease inhibitors is illustrated in Figure 10. The Boc protected intermediate (R)-(hydroxyethylamino)sulfonamides 14-19 were prepared following literature procedure.
(Koh, Y. et al. Antimicrob. Agents Chemother. 2003, 47, 3123-3129; and Surleraux, D. L.
N. G. et al. J. Med. Chem. 2005, 48, 1813-1822.) Briefly, ring opening of commercially available chiral epoxide, (1S,2S)-(1-oxiranyl-2-phenylethyl)carbamic acid tert-butyl ester, 11 with isobutylamine provided the amino alcohol 12. Reactions of substituted phenylsulfonyl chlorides with 12 afforded the sulfonamides 14-19 that were coupled with phenyloxazolidine fragments. Initially, four compounds were synthesized using either unsubstituted (R)- or (S)-3-phenyloxazolidine-5-carboxylic acid 9a or l0a attached to the (R)-(hydroxyethylamino)-sulfonamide isostere at P2 position. Previously optimized phenylsulfonamides, 4-methoxyphenylsulfonamide and 4-aminophenylsulfonamide, were utilized as P2' ligands. Thus, removal of the Boc protection of sulfonamides
14-15 followed by the reactions of the resulting amino alcohols with either (R)- or (S)-enantiomer of the activated carboxylic acids 9a or l0a provided the target compounds 20a-23a (Figure 10). In the case of compounds 22a and 23a, the nitro group was reduced using tin chloride to afford the corresponding amino derivatives 24a and 25a. It has to be noted that attempts to use the standard amide coupling conditions, EDCI/HOBt/DIEA, were not very successful and resulted in poor yields mainly because of very slow reactions even with DMF as solvent. In all subsequent reactions the carboxylic acids 9 and 10 were converted to the corresponding acid chlorides using oxalyl chloride.

Series of inhibitors were synthesized using substituted (S)-phenyloxazolidines at P2 and different phenylsufonamide groups at P2' position for structure-activity relationship (SAR) studies. Following the deprotection of sulfonamide intermediates 14-19, the resulting amines were reacted with (S)-N-phenyloxazolidine-5-carbonyl chlorides obtained by the activation of the corresponding carboxylic acids 10b-g to afford the target compounds 21 and 25-29 (Figure 10). The compounds 23b-f containing 4-nitrophenylsulfonamide group at P2' position were transformed to the corresponding 4-aminophenylsulfonamide derivatives 25b-f by the reduction of the nitro group.

In addition to the compounds described above, series of compounds were prepared with variations at three different positions. The isobutyl group at P1O
position was replaced with three cyclic primary amines. Again, starting from commercially available chiral epoxide 11, the target compounds were synthesized using an analogous synthetic route (Figure 11). In brief, ring opening of epoxide 11 with primary amines 30a-c provided amino alcohols 31a-c. Reactions of various substituted phenylsulfonyl chlorides with 31a-c provided sulfonylamides 32-35. After deprotection of intermediate compounds 32-35, the resulting amines were reacted with (S)-N-phenyloxazolidine-5-carbonyl chlorides prepared from the corresponding carboxylic acids 10 to afford the target compounds 36-39 (Figure 11).

Pharmaceutical Compositions. The methods described herein include the manufacture and use of pharmaceutical compositions, which include the protease inhibitors described herein as active ingredients. Also included are the pharmaceutical compositions themselves. These compositions can be administered using routes of administration and dosages similar to those used for known HIV protease inhibitors.

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. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable salts, esters, salts of such esters, or a pro-drug or other adduct or derivative of a compound of this invention which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.

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. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences 1977, 66: 1-19, 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, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, 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, quatemary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
Additionally, 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.

Furthermore, the term "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the 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-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S.
Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.

Methods of formulating pharmaceutical compositions are known in the art; see, e.g., Remington: The Science and Practice of Pharmacy, 20th Ed. (Baltimore, MD:
Lippincott Williams & Wilkins, 2000). Pharmaceutical compositions typically include a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" includes saline, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions.

Pharmaceutical compositions are typically formulated to be compatible with their intended route(s) of administration. Examples of routes of administration include parenteral, e.g., by intravenous, intradermal, or subcutaneous injection; or mucosal (e.g., by oral ingestion, inhalation, or rectal or vaginal administration) administration. Compositions intended for parenteral administration can include the following components: a sterile diluent, such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents, such as benzyl alcohol or methyl parabens; antioxidants, such as ascorbic acid or sodium bisulfite;
chelating agents, such as ethylenediaminetetraacetic acid; buffers, such as acetates, citrates or phosphates and agents for the adjustment of tonicity, such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide, as appropriate. A parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.

Pharmaceutical compositions suitable for injectable use can include sterile aqueous solutions (where the active ingredient is water soluble) or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent necessary to allow administration via syringe. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols, such as mannitol, sorbitol, and/or sodium chloride in the composition.
Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

Oral compositions generally include an inert diluent or an edible carrier. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules, e.g., gelatin capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder, such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient, such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant, such as colloidal silicon dioxide;
a sweetening agent, such as sucrose or saccharin; or a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring.

For administration by inhalation, the compounds can be delivered in the form of an aerosol spray from a pressured container or dispenser that contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Such methods include those described in U.S. Patent No. 6,468,798; hereby incorporated by reference.

Systemic administration of a therapeutic compound as described herein can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art. The pharmaceutical compositions can also be prepared in the form of suppositories (e.g., with conventional suppository bases, such as cocoa butter and other glycerides) or retention enemas for rectal delivery.

In one embodiment, the therapeutic compounds are prepared with carriers that will protect the therapeutic compounds against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such forrnulations can be prepared using standard techniques. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811; hereby incorporated by reference.

The pharmaceutical compositions can be included in a container, kit, pack, or dispenser, optionally with instructions for administration. A kit may comprise one or more compounds described herein and/or one or more other therapeutic compounds and/or a device for their administration, e.g., a syringe.

Biological Evaluation of Selected Compounds of the Invention. HIV protease inhibitor activities were determined by fluorescence resonance energy transfer (FRET) method. (Matayoshi, E. D. et al. Science 1990, 247, 954-958.) Protease substrate (Arg-Glu(EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg) was labeled with the energy transfer donor (EDANS) and acceptor (DABCYL) dyes at its two ends to perform FRET. Inhibitor binding dissociation constant (Ki value) was obtained by nonlinear regression fitting to the plot of initial velocity as a function of inhibitor concentration based on Morrison equation. (Greco, W. R. et al. J. Biol. Chem. 1979, 254, 12104-12109.) The activities of all the synthesized inhibitors against wild type HIV-1 protease (Q7K) were determined in triplicate. Chemical structures of inhibitors and their inhibitory activities (Ki values) are presented in the figures.

In addition, a small set of protease inhibitors with potent activities against wild type protease was studied against a panel of multidrug-resistant (MDR) mutants of proteases each representing different paradigms of resistance. The mutant variants were selected by examining the Stanford HIV-1 Drug Resistance Database (http://hivdb.stanford.edu) of HIV-1 infected-patient sequences of viral isolates. The three selected protease variants represent the pattern of resistance mutations that occur under the selective pressure of three or more currently prescribed protease inhibitors.
(Wu, T. D. et al. J. Virol. 2003, 77, 4836-4847.) These MDR mutant variants are L10I, G48V, I54V, L63P, V82A (M1), D30N, L63P, N88D (M2), and L10I, L63P, A71V, G73S, 184V, L90M
(M3). The inhibitory activities of selected protease inhibitors against M1-M3 mutant HIV-1 proteases were examined. For comparison two currently marketed drugs amprenavir (APV) and lopinavir (LPV) were also studied against the selected panel of mutant proteases.

Methods of Treatment. The methods described herein include methods for the treatment or prevention of a viral infection, e.g., an HIV, infection and Acquired Immunodeficiency Syndrome (AIDS) or AIDS Related Complex (ARC). Generally, the methods include administering a therapeutically effective amount of a protease inhibitor described herein, to a subject (e.g., a human or other primate) in need thereof, or who has been determined to be in need of, such treatment, e.g., a subject who is (or is determined to be) infected with HIV. A subject who is likely to be infected with HIV, e.g., a person in a high risk group, may also be treated as indicated herein. Subjects also include women who are expecting a child (pregnant women) and in whom a treatment reduces the liklihood of transmission of HIV to the child.

In addition to HIV-1 infections, the methods described herein are also expected to be beneficial for treating or preventing HIV-2 infections. Among HIV-1 viruses, it is expected that the methods will be effective against any HIV-1 strain, such as those of group M, 0 and N, and subtypes A, B, C, D, E, F, Q H, I, J and K and "circulating recombinant forms" or CRFs thereof. The compounds described herein may also be used for treating any other viral infections in which the viral agent has a protease inhibitor that can be inhibited by the compounds described herein.

As used in this context, to "treat" means to ameliorate at least one clinical symptom or parameter of HIV infection or preventing it from worsening or preventing the transmission of HIV, e.g., from mother to child. For example, a treatment can result in a reduction in viral load, and/or an increase in number of CD4+ T cells ("CD4 count").
When a subject has achieved a reduction in viral load, and/or an increase in CD4 count, then treatment may also include maintaining the reduction in viral load, and/or the increased CD4 count, e.g., preventing a resurgence of viral load and/or a decrease in CD4 count. These, and other clinically relevant parameters, can be measured using methods known in the art. For example, viral load can be measured, e.g., using PCR or branched DNA (bDNA) assays known in the art. CD4 counts can be measured, e.g., using hematology, DYNAbeadsTM (Dynal Biotech/Invitrogen Corp., Brown Deer, WI), flow cytometry (e.g., FACSCountTM, BD Biosciences, Franklin Lakes, NJ) or enzyme-linked immunosorbent assay (ELISA) methods (see, e.g., Lyamuya et al., J. Immunol.
Methods 195(1-2):103-12 (1996); Paxton et al., Clin. Diagn. Lab. Immunol. 2(1):104-114 (1995);
Saah et al. Arch. Pathol. Lab. Med. 121(9):960-2 (1997); Mwaba et al., Lancet (2003)). Healthy adults and teenagers generally have a CD4 count of at least 800 cells per cubic millimeter of blood; a CD4 count below 200 is associated with severe risk of illness (e.g., AIDS-related diseases, such as Kaposi's sarcoma or pneumocystic pneumonia).
Current guidelines suggest treatment for HIV should be started when the CD4 count is less than about 350 and/or the viral load is greater than about 50,000.

A "therapeutically effective amount" is an amount sufficient to effect a desired therapeutic effect, e.g., a reduction in viral load, and/or an increase in number of CD4+ T
cells. An effective amount can be administered in one or more administrations, applications or dosages. A therapeutically effective amount of a composition may depend on the composition selected. The compositions can be administered once, one or more times per day, and/or one or more times per week; including once every other day. In certain embodiments, the compositions will be administered two or three times per day.
The skilled artisan will appreciate that certain factors may influence the dosage and timing required to treat effectively a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and any other indications present. Treatment of a subject with a therapeutically effective amount of a protease inhibitor described herein can include a single treatment or a series of treatments.

Dosage, toxicity and therapeutic efficacy of the compounds can be determined, e.g., by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
Compounds that exhibit high therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to select a dose and administration schedule that minimizes severe side effects while maximizing therapeutic efficacy.

The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in a method described herein, a therapeutically effective dosage range can be estimated initially from cell culture assays. A dose can be further formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to determine more accurately useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. In some embodiments, a therapeutically effective amount of a new protease inhibitor described herein ranges from about 0.1 to 10 mg per day, or about 0.3 to 5 mg/day.

In some embodiments, one or more of the protease inhibitors described herein will be administered in combination with one or more other therapeutic agents, e.g., as part of a highly active antiretroviral therapy (HAART) regimen that includes one or more other anti-retroviral agents. For example, the methods may include administration of one or more of a non-nucleoside reverse transcriptase inhibitor (NNRTI), such as efavirenz (SustivaTM), nevirapine (ViramuneTM) and delavirdine (RescriptorTM), 8 and 9-Cl TIBO
(tivirapine), loviride, TMC-125, dapivirine, MKC-442, UC 781, UC 782, Capravirine, DPC 961, DPC963, DPC082, DPC083, calanolide A, SJ-1366, TSAO, 4"-deaminated TSAO, MV150 and MV026048; a nucleoside reverse transcriptase inhibitor (NRTI), such as AZT
(zidovudine, RetrovirTM)/3TC (lamivudine, EpivirTM), emtricitabine (EmtrivaTM) and d4T
(stavudine, ZeritTM)/3TC, and d-drugs (ddl [didanosine, VidexTM/VidexECTM], ddC
[zalcitabine, HividTM], d4T), Abacavir, FTC, DAPD, dOTC, and DPC 817; a nucleotide reverse transcriptase inhibitor, such as tenofovir (VireadTM) and PMEA; a fusion inhibitor, such as enfuvirtide (FuzeonTM), T20, T1249, 5-helix and D-peptide ADS-Jl; an entry inhibitor; a co-receptor binding inhibitor, such as AMD 3100, AMD-3465, AMD7049, AMD3451 (Bicyclams), TAK 779; SHC-C (SCH351125), SHC-D, PRO-14ORT inhibitors, such as foscamet and prodrugs; an RNAse H inhibitor, such as SP1093V and PD126338; a TAT inhibitor, such as RO-5-3335, K12 and K37; an integrase inhibitor, such as L 708906, L 731988 and S-1360; another protease inhibitor, such as amprenavir and prodrug GW908, nelfinavir, saquinavir, indinavir, lopinavir, palinavir, BMS 186316, atazanavir, DPC 681, DPC 684, tipranavir, AG1776, mozenavir, GS3333, KNI-413, KNI-272, L754394, L756425, LG-71350, PD161374, PD173606, PD177298, PD178390, PD178392, PNU
140135, TMC114 maslinic acid and U-140690; a glycosylation inhibitor, such as castanospermine, deoxynojirimycine; or a binding inhibitor, such as dextran sulfate, suramine, polyanions, soluble CD4, PRO-542 and BMS-806. Other drugs include those set forth at http://aidsinfo.nih.gov/, hereby incorporated by reference.

Other therapeutic agenets that may be coadministered with with one or more agents described herein are agents that inhibit metabolic enzymes, e.g., inhibitors of cytochrome P450 (CYP450) enzymes. For example, a compound described herein may be administered, simultaneously or not, with an inhibitor of CYP3A4, e.g., Ritonavir, or an inhibitor of CYP2C19, CYP1A2, CYP2D6, or CYP2C9. Exemplary inhibitors of 2C9 are described, e.g., in U.S. publication No. 2006.0069042, hereby incorporated by reference.

The compounds of the present invention may also be administered in combination with immunomodulators (e.g., bropirimine, anti-human alpha interferon antibody, IL-2, methionine enkephalin, interferon alpha, HE-2000 and naltrexone), antibiotics (e.g., pentamidine isothiorate), cytokines (e.g. Th2), modulators of cytokines, chemokines or the receptors thereof (e.g. CCR5) or hormones (e.g. growth hormone), to ameliorate, combat, or eliminate HIV infection and its symptoms.

In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is selected from the group consisting of amprenavir (Agenerase ; APV), tipranavir (Aptivus ; TPV), indinavir (Crixivan(M; IDV), saquinavir (Invirase ; SQV), lopinavir and ritonavir (Kaletra ; LPV), fosamprenavir (Lexiva ; FPV), ritonavir (Norvir ; RTV), atazanavir (Reyataz ;
ATZ), nelfinavir (Viracept ; NFV), brecanavir, and darunavir.

In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is ritonavir (Kaletra ; LPV).
In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is selected from the group consisting of zidovudine (AZT; Azidothymidine; Retrovir ), didanosine (Dideoxyinosine;
ddl; Videx ), zalcitabine (Dideoxycytidine; ddC; Hivid ), lamivudine (3TC;
Epivir ), stavudine (2',3'-didehydro-3'-deoxythymidine; D4T; Zerit ), abacavir succinate (1592U89 succinate; Ziagen ABC), Combivir (lamivudine & zidovudine; (-)-3TC & AZT), and Trizivir (abacavir & lamivudine & zidovudine; ABC & (-)-3TC & AZT).

In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is selected from the group consisting of nevirapine (BI-RG-587; Viramune ), delavirdine (BHAP; U-90152;
Rescriptor ), and (efavirenz; DMP-266; Sustiva ).

In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is T-20 (Fuzeon ;
Enfuvirtide; DP-178; Pentafuside; GP41 127-162 AA).

In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is TMCC114, or TMCC114 in combination with a reverse transcriptase inhibitor. In some embodiments, the methods further comprise administering a second therapeutic agent, wherein the second therapeutic agent is Lipinavir, or Lupanivir in combination with a reverse transcriptase inhibitor.

Combination therapy in different formulations may be administered simultaneously, separately or sequentially. Alternatively, such combination may be administered as a single formulation, whereby the active ingredients are released from the formulation simultaneously or separately. Compositions comprising at least two inhibitors described herein and/or one or more other protease inhibitors and/or other therapeutic agents are also provided herein. In certain embodiments the compounds of the invention can be combined with one or more of any anti-HIV compounds (e.g. those listed in Figures 6a-k). Additional compounds which may be combined with one or more of the inventive compounds, and further discussion of combination therapy can be found in Yeni, P. G. et al.
JAMA 2004, 292(2), 251-265; Pozniak, A. et al. Business Briefing: Clinical Virology &
Infectious Disease 2004, 1-7; and Chittick, G. E. et al. Antimicrobial Agents and Chemotherapy 2006, 1304-1310; all of which are hereby incorporated by reference.

Definitions. All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.

The indefinite articles "a" and "an," as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean "at least one."

The term "HIV" is known to one skilled in the art to refer to Human Immunodeficiency Virus. There are two types of HIV: HIV-1 and HIV-2. There are many different strains of HIV-1. The strains of HIV-1 can be classified into three groups: the "major" group M, the "outlier" group 0 and the "new" group N. These three groups may represent three separate introductions of simian immunodeficiency virus into humans.
Within the M-group there are at least ten subtypes or clades: e.g., clade A, B, C, D, E, F, G, H, I, J, and K. A"clade" is a group of organisms, such as a species, whose members share homologous features derived from a common ancestor. Any reference to HIV in this application includes all of these tupes and strains.

As known to one skilled in the art, "retroviruses" are diploid positive-strand RNA
viruses that replicate through an integrated DNA intermediate (proviral DNA).
In particular, upon infection by the RNA virus, the lentiviral genome is reverse-transcribed into DNA by a virally encoded reverse transcriptase that is carried as a protein in each retrovirus. The viral DNA is then integrated pseudo-randomly into the host cell genome of the infecting cell, forming a "provirus" which is inherited by daughter cells.
The retrovirus genome contains at least three genes: gag codes for core and structural proteins of the virus;
pol codes for reverse transcriptase, protease and integrase; and env codes for the virus surface proteins. Within the retrovirus family, HIV is classified as a lentivirus, having genetic and morphologic similarities to animal lentiviruses such as those infecting cats (feline immunodeficiency virus), sheep (visna virus), goats (caprine arthritis-encephalitis virus), and non-human primates (simian immunodeficiency virus).

The term "heteroatom" is art-recognized and refers to an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium.

The term "alkyl" is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has about 30 or fewer carbon atoms in its backbone (e.g., CI -C30 for straight chain, C3-C30 for branched chain), and alternatively, about 20 or fewer. Likewise, cycloalkyls have from about 3 to about 10 carbon atoms in their ring structure, and alternatively about 5, 6 or 7 carbons in the ring structure.

Unless the number of carbons is otherwise specified, "lower alkyl" refers to an alkyl group, as defmed above, but having from one to about ten carbons, alternatively from one to about six carbon atoms in its backbone structure. Likewise, "lower alkenyl"
and "lower alkynyl" have similar chain lengths.

The term "aralkyl" is art-recognized and refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).

The terms "alkenyl" and "alkynyl" are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.

The term "aryl" is art-recognized and refers to 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, naphthalene, anthracene, pyrene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or "heteroaromatics." The aromatic ring may be substituted at one or more ring positions with such substituents as described herein, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF3, -CN, or the like. The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.

The terms ortho, meta and para are art-recognized and refer to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.

The terms "heterocyclyl", "heteroaryl", or "heterocyclic group" are art-recognized and refer to 3- to about 10-membered ring structures, alternatively 3- to about 7-membered rings, whose ring structures include one to four heteroa toms. Heterocycles may also be polycycles. Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxanthene, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like.
The heterocyclic ring may be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, or the like.

The terms "polycyclyl" or "polycyclic group" are art-recognized and refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged"
rings. Each of the rings of the polycycle may be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, or the like.

The term "carbocycle" is art-recognized and refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.

The term "nitro" is art-recognized and refers to -NO2i the term "halogen" is art-recognized and refers to -F, -Cl, -Br or -I; the term "sulfhydryl" is art-recognized and refers to -SH; the term "hydroxyl" means -OH; and the term "sulfonyl" is art-recognized and refers to -SO2 ."Halide" designates the corresponding anion of the halogens, and "pseudohalide" has the definition set forth on page 560 of "Advanced Inorganic Chemistry"
by Cotton and Wilkinson.

The terms "amine" and "amino" are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas:
-N(R5 1)(R50) or [-N(R50)(R52)(R53)]+, wherein R50, R5 1, R52 and R53 each independently represent a hydrogen, an alkyl, an alkenyl, -(CHz),n R61, or R50 and R51 or R52, taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R61 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8.
In other embodiments, R50 and R51 (and optionally R52) each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH2)rõR61. Thus, the term "alkylamine"
includes an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R50 and R51 is an alkyl group.

The term "acylamino" is art-recognized and refers to a moiety that may be represented by the general formula: -N(R50)-C(=O)R54, wherein R50 is as defined above, and R54 represents a hydrogen, an alkyl, an alkenyl or -(CH2)m R61, where m and R61 are as defined above.

The term "amido" is art recognized as an amino-substituted carbonyl and includes a moiety that may be represented by the general formula: -C(=O)N(R5 0)(R5 1), wherein R50 and R51 are as defined above. Certain embodiments of the amide in the present invention will not include imides which may be unstable.

The term "alkylthio" refers to an alkyl group, as defined above, having a sulfur radical attached thereto. In certain embodiments, the "alkylthio" moiety is represented by one of -S-alkyl, -S-alkenyl, -S-alkynyl, and -S-(CH2)õ,-R61, wherein m and R61 are defined above. Representative alkylthio groups include methylthio, ethyl thio, and the like.

The term "carboxyl" is art recognized and includes such moieties as may be represented by the general formulas: -C(=O)-X50-R55 or -X50-C(=O)-R56, wherein X50 is a bond or represents an oxygen or a sulfur, and R5 5 and R5 6 represents a hydrogen, an alkyl, an alkenyl, -(CH2)r,-R61 or a pharmaceutically acceptable salt, R56 represents a hydrogen, an alkyl, an alkenyl or -(CH2)m R61, where m and R61 are defined above.
Where X50 is an oxygen and R55 or R56 is not hydrogen, the formula represents an "ester". Where X50 is an oxygen, and R55 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R55 is a hydrogen, the formula represents a "carboxylic acid". Where X50 is an oxygen, and R56 is hydrogen, the formula represents a "formate". In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a"thiolcarbonyl" group. Where X50 is a sulfur and R55 or R56 is not hydrogen, the formula represents a "thiolester." Where X50 is a sulfur and R55 is hydrogen, the formula represents a "thiolcarboxylic acid." Where X50 is a sulfur and R56 is hydrogen, the formula represents a"thiolformate." On the other hand, where X50 is a bond, and R55 is not hydrogen, the above formula represents a "ketone" group.
Where X50 is a bond, and R55 is hydrogen, the above formula represents an "aldehyde"
group.

The term "carbamoyl" refers to -O(C=O)NRR', where R and R' are independently H, aliphatic groups, aryl groups or heteroaryl groups.

The term "oxo" refers to a carbonyl oxygen (=0).

The terms "oxime" and "oxime ether" are art-recognized and refer to moieties that may be represented by the general formula: -C(R75)(=NOR), wherein R75 is hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, aralkyl, or -(CH2),,,-R61. The moiety is an "oxime"
when R is H; and it is an "oxime ether" when R is alkyl, cycloalkyl, alkenyl, alkynyl, aryl, aralkyl, or -(CH2)m R61.

The terms "alkoxyl" or "alkoxy" are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as may be represented by one of -0-alkyl, -0-alkenyl, -0-alkynyl, -0-(CH2)m-R61, where m and R61 are described above.
The term "sulfonate" is art recognized and refers to a moiety that may be represented by the general formula: -S(=O)20R57, in which R57 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.

The term "sulfate" is art recognized and includes a moiety that may be represented by the general formula: -OS(=0)20R57, in which R57 is as defined above.

The term "sulfonamido" is art recognized and includes a moiety that may be represented by the general formula: -N(R50)-S(=O)20R56, in which R50 and R56 are as defined above.

The term "sulfamoyl" is art-recognized and refers to a moiety that may be represented by the general formula: -S(=O)2N(R50)(R51), in which R50 and R51 are as defined above.

The term "sulfonyl" is art-recognized and refers to a moiety that may be represented by the general formula: -S(=O)2R58, in which R58 is one of the following:
hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl.

The term "sulfoxido" is art-recognized and refers to a moiety that may be represented by the general formula: -S(=O)R58, in which R58 is defined above.

Analogous substitutions may be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.

The definition of each expression, e.g., alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.

The abbreviations Me, Et, Ph, Tf, Nf, Ts, and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and methanesulfonyl, respectively. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations.

It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.

The term "substituted" is also contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein above. The permissible substituents may be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.

For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, "Handbook of Chemistry and Physics", 67th Ed., 1986-87, inside cover.
EXEMPLIFICATION
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention General Experimental Procedures. 'H and 13C NMR spectra were recorded on a Varian 400 MHz NMR spectrometer, operating at 400 MHz for 'H and 100 MHz for 13C.
Chemical shifts are reported in ppm relative to the solvent signal, and coupling constant (J) values are reported in Hertz (Hz). Thin-layer chromatography (TLC) was performed on E.
Merck silica gel 60-F-254 plates and spots were visualized with UV light.
Flash column chromatography was performed using 230-400 mesh silica gels (E-Merck). High resolution mass spectra (HRMS) were recorded on Waters Q-TOF Premier mass spectrometer by direct infusion of solutions of each compound using electrospray ionization (ESI) in positive mode. Low resolution mass spectra were obtained using Waters Alliance HT/Micromass ZQ system (ESI). Tetrahydrofuran was distilled from sodium/benzophenone. Anhydrous Dichloromethane, N,N-dimethylformamide, benzene, and toluene were purchased from Aldrich and used as such. All reagents and chemicals were purchased from commercial vendors and used as received.

Analytical reversed-phase high performance liquid chromatography (HPLC) was performed on a Waters Separation Module 2695 system equipped with an auto sampler and a Waters 996 photodiode array detector. Purity of the final compounds was determined using two different chromatographic systems. First system: column, Waters Nova-Pak RP-C18 (4,um, 3.9 mm x 150 mm); mobile phase A, 10 mM ammonium acetate in water;
mobile phase B, acetonitrile. Using a flow rate of 0.8 mL/min, gradient elution was performed from 50% B to 100% B over 10 min. Second system: column, Agilent Zorbax 300SB-C8 (5 ^m, 4.6 mm x 250 mm); mobile phase A, 0.1% trifluoroacetic acid in water;
mobile phase B, 0.1% trifluoroacetic acid in acetonitrile. Gradient elution was performed from 50% B to 100% B over 10 min at a flow rate of 1 mL/min. A table containing the retention time and purity of selecyed compound is shown in Figure 20.

GENERAL PROCEDURE FOR THE CBZ PROTECTION OF SUBSTITUTED
ANILINES

Solid NaHCO3 (32.65 g, 388.5 mmols) was added to an ice-cooled solution of aniline derivative (185 mmols) in acetone-water mixture (2:1) (300 mL) followed by the slow addition of benzyl chloroformate (27 mL, 190 mmols). The resulting slurry was warmed to ambient temperature and stirred overnight. Reaction mixture was poured onto ice and the resulting precipitate was filtered, washed with water and dried.
Product was purified by recrystallization from a mixture of hexanes and ethylacetate to provide the pure product as crystalline solid. Compounds 4a-g were prepared following this general procedure.

GENERAL PROCEDURE FOR THE SYNTHESIS OF 5-(HYDROXYMETHYL)-A solution of CBZ protected aniline derivative 4 (34.7 mmols) in dry THF (150 mL) was cooled to -78 C under dry N2 atmosphere. A solution of n-BuLi (1.6 M in hexanes; 25 mL, 40 mmols) was slowly added keeping the temperature below -70 C. After stirring the reaction mixture at -78 C for 45 minutes, a solution of chiral glycidyl butyrate (5 g, 34.7 mmols) in dry THF was slowly added. The resulting mixture was stirred at -78 C for 2 hours and then slowly warmed to room temperature and stirred overnight.
Reaction was quenched by the addition of saturated aqueous NH4C1 solution. Ethyl acetate and water were added and layers separated. The aqueous layer was further extracted with ethyl acetate (3 times). Combined organic extract was washed with saturated aqueous NaC1 solution, dried (Na2SO4), filtered and evaporated to yield a pale yellow solid. This solid was triturated with a mixture of chloroform and hexanes and filtered to provide pure alcohol as off white solid. Compounds 8a-g were prepared following this general procedure.

CARBOXYLIC ACIDS 9 AND 10.

To an ice-cooled solution of Na104 (35 mmols) in water (75 mL) was added a solution of the alcohol 7 or 8(10 mmols) in a mixture of CH3CN and CC14 (1:1) (100 mL).
Solid RuC13.HZ0 (0.5 mmol) was added and the reaction mixture was stirred at 0 C for 30 minutes, warmed to room temperature and stirred for 4-6 hours. Reaction was quenched by adding CH2C12and layers were separated. The aqueous layer was further extracted with CH2C12, combined organic extract was dried (NaZSO4) and evaporated to provide a gummy solid. Crude product was purified by column chromatography on silica gel using a mixture of 25% CH3CN in CH2C12 + 1% HCOzH as eluent. This method provided the desired phenyloxazolidine-5-carboxylic acids as solids in excellent yields. The following compounds were prepared by this general procedure:

GENERAL PROCEDURE FOR THE RING OPENING OF EPOXIDE WITH AMINES.
A solution of the chiral epoxide 11 (1-Oxiranyl-2-phenylethyl)carbamic acid tert-butyl ester) (10 mmol) in EtOH (75 mL) was added to isobutyl amine (10 mL;
large excess) and the mixture was heated at 80 C for 3 hours. After cooling to room temperature, solvents were removed under reduced pressure. Product was purified by recrystallization from ethyl acetate-hexanes mixture to provide the product as white solid in excellent yield.

GENERAL PROCEDURE FOR THE SYNTHESIS OF (HYDROXYETHYLAMINO)-SULFONAMIDES

To an ice-cooled solution of the secondary amine 12 (5 mmol) in CH2C12 (20 mL) was added an aqueous solution of Na2CO3 (8 mmol in 5 mL H20) followed by the slow addition of sulfonyl chloride (5 mmol) solution in CH2C12 (5 mL). After 15 minutes the reaction mixture was warmed to ambient temperature and stirred till no starting material was detected by tlc. Reaction mixture was diluted with CH2C12 and layers were separated.
Organic extract was washed with saturated aqueous NaCl solution, dried (Na2SO4), filtered and evaporated. Product was purified by flash chromatography on silica gel using mixture of ethyl acetate and hexanes as eluent to afford pure product.

The following compounds were prepared by this general procedure: N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[isobutyl[(4-methoxyphenyl)sulfonyl]amino]propyl]carbamic acid tert-butyl ester (14); N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[isobutyl[(4-nitrophenyl)sulfonyl]-amino]propyl]carbamic acid tert-butyl ester (15); N-[(1S,2R)-3-[[(Benzo-[1,3]-dioxole-5-sulfonyl)](isobutyl)amino]-1-benzyl-2-hydroxypropyl]carbamic acid tert-butyl ester (16);
N-[(1 S,2R)-1-Benzyl-3-[[(3-fluoro-4-methoxyphenyl)sulfonyl] (isobutyl)amino]-hydroxypropyl]-carbamic acid tert-butyl ester (17); N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[isobutyl[[(4-trifluoro-methoxy)phenyl]sulfonyl]amino]propyl]carbamic acid tert-butyl ester (18); N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[isobutyl[(3-methoxyphenyl)sulfonyl]amino]propyl]carbamic acid tert-butyl ester (19);1V-[(1S,2R)-1-Benzyl-3 -[(cyclopropylmethyl) [(3 -methoxyphenyl)sulfonyl] -amino] -2-hydroxypropyl]carbamic acid tert-butyl ester (32); N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[[(3-methoxyphenyl)sulfonyl](2-thiophenylmethyl)]amino]propyl]carbamic acid tert-butyl ester (33); N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[(2-thiophenylmethyl)[(2,4,5-trifluorophenyl)sulfonyl]-amino]propyl]carbamic acid tert-butyl ester (34);
and N-[(1S,2R)-1-Benzyl-2-hydroxy-3-[[(3-methoxyphenyl)sulfonyl] [(R)-(tetrahydro-2-furanyl)methyl]amino]propyl]carbamic acid tert-butyl ester (35).

GENERAL PROCEDURE FOR THE COUPLING REACTION.

Excess oxalyl chloride was added to solid phenyloxazolidinone-5-carboxylic acid (0.5 mmols) and the resulting mixture was stirred at room temperature overnight. Oxalyl chloride was removed by distillation under reduced pressure and residue dried under high vacuum for 30 minutes. A solution of the resulting acid chloride in dry THF (5 mL) was used in the coupling reaction.

To an ice-cooled mixture of the Boc deprotected amine (0.5 mmols) in dry THF
(5 mL) was added Et3N (1.1 mmols) followed by slow addition of the acid chloride solution.
After 15 minutes the reaction mixture was warmed to room temperature and stirred until reaction was complete (monitored by tlc). Small amount of water and ethyl acetate were added and layers were separated. The organic extract was washed with saturated aqueous NaC1 solution, dried (NaZSO4), filtered and evaporated. Flash chromatography on silica gel using mixture of ethyl acetate and hexanes (in some cases, methanol/chloroform mixture) as eluent, provided the target compound as solid.

GENERAL PROCEDURE FOR THE REDUCTION OF THE NITRO GROUP

A mixture of the nitro compound (0.4 mmols) and SnC12.2H20 (0.45 g, 2 mmols) in ethyl acetate (10 mL) was heated at 80 C for 2-3 hours. Reaction mixture was allowed to cool to ambient temperature and treated with saturated aqueous NaHCO3 solution (10 m). It was diluted with ethyl acetate and layers were separated, aqueous layer was further extracted with ethyl acetate (2 x). Combined organic extract was washed with saturated aqueous NaCl solution, dried (Na2SO4) and evaporated to yield a foamy solid.
Flash chromatography on silica gel using mixture of methanol in chloroform as eluent, provided the target compound as solid.

HIV-1 protease inhibitor activities were determined by fluorescence resonance energy transfer (FRET) method (Matayoshi, E. D.; Wang, G. T.; Krafft, G. A.;
Erickson, J.
Novel fluorogenic substrates for assaying retroviral proteases by resonance energy transfer.
Science 1990, 247, 954-958.) Protease substrate (Arg-Glu(EDANS)-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Lys(DABCYL)-Arg) was purchased from Molecular Probe. The energy transfer donor (EDANS) and acceptor (DABCYL) were labeled at its two ends respectively to perform FRET. Fluorescence measurements were carried out on PTI
fluorescence spectrophotometer (Photon Technology International) at 30 C. Excitation and emission wavelengths were set at 340 nm and 490 nm, respectively. Each reaction was recorded for about 10 min. Wide type HIV-1 protease (Q7K) and its MDR mutants M1 (L10I, G48V, 154V, L63P, V82A), M2 (D30N, L63P, N88D), and M3 (L10I, L63P, A71V, G73S, 184V, L90M) were desalted through PD-10 columns (Amersham Biosciences). Sodium Acetate (20 mM, pH 5) was used as elution buffer. Apparent protease concentrations were around 50 nM estimated by UV spectrophotometer (Shimadzu) at 280 nm. All inhibitors were dissolved in DMSO and diluted to appropriate concentrations. Protease (2 L) and inhibitor (2 L) or DMSO were mixed and incubated for 20-30 min at room temperature before initialing substrate cleavage reaction. Throughout this work, 150 L of 1 M
substrate was used. Substrate buffer is composite of 0.1 M sodium acetate, 1 M sodium chloride, 1 mM

ethylenediaminetetraacetic acid (EDTA), 1 mM dithiothreitol (DTT), 2%
dimethylsulfoxide (DMSO) and 1 mg/mL bovine serum albumin (BSA) with an adjusted pH 4.7.
Inhibitor binding dissociation constant (Ki) was obtained by nonlinear regression fitting (GraFit 5, Erithacus software) to the plot of initial velocity as a function of inhibitor concentrations based on Morrison equation. (Greco, W. R.; Hakala, M. T. Evaluation of methods for estimating the dissociation constant of tight binding enzyme inhibitors. J.
Biol. Chem. 1979, 254, 12104-12109.) The initial velocities were derived from the linear range of reaction curves.

SYNTHESIS OF PROTEASE INHIBITORS CONTAINING A
HYDROXYETHYLAMINE (HEA) CORE

The designed inhibitors with a hydroxyethylamine (HEA) core isostere can be synthesized in four steps starting with commercially available chiral epoxide (1S,2S
enantiomer) 12. Ring opening of epoxide 12 with various primary and secondary amines provided compounds 13. Reaction of 13 with various sulfonyl chlorides gave compounds 14. After deprotecting the Boc group, the resulting amines 15 were coupled with either (R) or (S) isomer of activated carboxylic acids to provide the designed inhibitors 16 (Figure 7A).

SYNTHESIS OF PROTEASE INHIBITORS CONTAINING CYCLIC CARBAMATES:
HE SERIES

The synthesis of protease inhibitors containing hydroxyethylene (HE) isostere starts with the synthesis of the core 17, which was obtained from L-phenylalanine in 5 steps.
After coupling of R4X2CO2H to 17, the dibenzyl protection was removed and the free amine 19 was coupled to the an activated acid to provide inhibitors 20 (Figure 7B).

SYNTHESIS OF PROTEASE INHIBITORS CONTAINING CYCLIC CARBAMATES:
AZA-HEA SERIES

The synthesis of protease inhibitors containing aza-hydroxyethylamine (Aza-HEA) isostere is outlined in Figure 8. The ring opening of chiral epoxide (1S,2R
enantiomer) 21 with CBz protected hydrazine derivative 22 provided compound 23. Deprotection of CBz followed by the coupling with R4X2CO2H gave compounds 24. Removal of the Boc protection and coupling with R3X1COZH provided the desired inhibitors 27.

INCORPORATION BY REFERENCE

The contents of all cited references (including literature references, issued patents, published patent applications and GenBank Accession numbers as cited throughout this application) are hereby expressly incorporated by reference. When definitions of terms in documents that are incorporated by reference herein conflict with those used herein, the definitions used herein govern.

EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (107)

We claim:
1. A compound, or a pharmaceutically acceptable salt thereof, of formula I:
I
wherein, independently for each occurrence, X1 is absent, -O-, -S-, -NR- or ;
X2 is absent, -O-, -S-, -NR- or ;

R1 is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

provided that when X1 is absent; R3 is not ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is ; or X2 is ; or R3 is amino, ,
2. The compound of claim 1, wherein X1 is absent.
3. The compound of claim 1, wherein X1 is
4. The compound of claim 1, wherein X2 is absent.
5. The compound of claim 1, wherein X1 is absent; and X2 is absent.
6. The compound of claim 1, wherein R1 is OH.
7. The compound of claim 1, wherein R2 is aralkyl or heteroaralkyl.
8. The compound of claim 1, wherein R2 is aralkyl.
9. The compound of claim 1, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
10. The compound of claim 1, wherein R3 is aryl or heteroaryl.
11. The compound of claim 1, wherein R4 is alkyl, aryl or heteroaryl.
12. The compound of claim 1, wherein R5 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
13. The compound of claim 1, wherein R5 is alkyl, (cycloalkyl)alkyl or (heterocyclyl)alkyl.
14. A compound, or a pharmaceutically acceptable salt thereof, of formula II:
II

wherein, independently for each occurrence, X! is absent, -O- or ;

R3 is alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, heteroaryl, aralkyl or heteroaralkyl; and R6 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
provided that when X1 is absent; R3 is not or ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is ; or R3 is amino, , ,
15. The compound of claim 14, wherein X1 is absent.
16. The compound of claim 14, wherein X1 is .
17. The compound of claim 14, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
18. The compound of claim 14, wherein R3 is aryl or heteroaryl.
19. The compound of claim 14, wherein R4 is alkyl, aryl or heteroaryl.
20. The compound of claim 14, wherein R6 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
21. The compound of claim 14, wherein R6 is alkyl, (cycloalkyl)alkyl or (heterocyclyl)alkyl.
2. The compound of claim 14, wherein R3 is , ,
23. The compound of claim 14, wherein R3 is
24. The compound of claim 14, wherein R4 is
25. The compound of claim 14, wherein R6 is
26. The compound of claim 14, wherein R6 is
27. The compound of claim 14, wherein X1 is absent; and R3 is
28. A compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of
29. A compound, or a pharmaceutically acceptable salt thereof, of formula III:

wherein, independently for each occurrence, X1 is absent, -O-, -S-, -NR- or ;

X2 is absent, -O-, -S-, -NR- or R1 is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R7 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

provided that when X1 is absent; R3 is not ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

provided that when X2 is absent; R4 is not ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is or X2 is ; R3 is amino, ,
30. The compound of claim 29, wherein X1 is absent.
31. The compound of claim 29, wherein X2 is absent.
32. The compound of claim 29, wherein X1 is absent; and X2 is absent.
33. The compound of claim 29, wherein R1 is OH.
34. The compound of claim 29, wherein R2 is aralkyl or heteroaralkyl.
35. The compound of claim 29, wherein R2 is aralkyl.
36. The compound of claim 29, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
37. The compound of claim 29, wherein R3 is aryl or heteroaryl.
38. The compound of claim 29, wherein R3 is
39. The compound of claim 29, wherein R4 is alkyl, aryl or heteroaryl.
40. The compound of claim 29, wherein R4 is
41. The compound of claim 29, wherein R5 is hydrogen.
42. The compound of claim 29, wherein R7 is alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
43. A compound, or a pharmaceutically acceptable salt thereof, of formula IV:

wherein, independently for each occurrence, X1 is absent or -O-;

R3 is alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, amino, (amino)alkyl, amido, (amido)alkyl, heterocyclyl, (heterocyclyl)alkyl, heteroaryl, aralkyl or heteroaralkyl; and R7 is alkyl, cycloalkyl, (cycloalkyl)alkyl or aralkyl;

provided that when X1 is absent; R3 is not ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

provided that when X2 is absent; R4 is not ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is or X2 is R3 is amino,
44. The compound of claim 43, wherein X1 is absent.
45. The compound of claim 43, wherein R3 is amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
46. The compound of claim 43, wherein R3 is
47. The compound of claim 43, wherein R4 is aryl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl (heterocyclyl)alkyl or heterocyclyl.
48. The compound of claim 43, wherein R4 is
49. The compound of claim 43, wherein R7 is alkyl, cycloalkyl or (cycloalkyl)alkyl.
50. The compound of claim 43, wherein X1 is absent; and R3 is amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
51. The compound of claim 43, wherein R3 is ,
52. The compound of claim 43, wherein R4 is
53. The compound of claim 43, wherein R7 is .
54. The compound of claim 43, wherein R7 is .
55. A compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of
56. A compound, or a pharmaceutically acceptable salt thereof, of formula V:

wherein, independently for each occurrence, X1 is absent, -O-, -S-, -NR- or X2 is absent, -O-, -S-, -NR- or R1 is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

provided that when X1 is absent; R3 is not wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

provided that when X2 is absent; R4 is not wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is or X2 is or R3 is amino.
57. The compound of claim 56, wherein X1 is absent.
58. The compound of claim 56, wherein X1 is -O-.
59. The compound of claim 56, wherein X2 is absent.
60. The compound of claim 56, wherein X1 is absent or -O-; and X2 is absent.
61. The compound of claim 56, wherein R1 is OH.
62. The compound of claim 56, wherein R2 is aralkyl or heteroaralkyl.
63. The compound of claim 56, wherein R2 is aralkyl.
64. The compound of claim 56, wherein R3 is heterocyclyl.
65. The compound of claim 56, wherein R4 is (heterocyclyl)alkyl.
66. The compound of claim 56, wherein R5 is alkyl.
67. The compound of claim 56, wherein X1 is absent or -O-; X2 is absent; R1 is OH; R2 is aralkyl; R3 is heterocyclyl; R4 is alkyl, aryl or heteroaryl; and R5 is alkyl.
68. A compound, or a pharmaceutically acceptable salt thereof, of formula VII:

wherein, independently for each occurrence, X1 is absent, -O-, -S-, -NR- or X2 is absent, -O-, -S-, -NR- or R1 is -OH, -SH or -NHR;

R is hydrogen, alkyl, aralkyl, heteroaralkyl or acyl;

R2 is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R3 is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R5 is hydrogen, alkyl, (cycloalkyl)alkyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and the stereochemical configuration at any stereocenter is R, S, or a mixture of these configurations;

provided that when X1 is absent; R3 is not wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

provided that when X2 is absent; R4 is not or ; wherein R3A is hydrogen, alkyl, alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl; and provided that X1 is ; or X2 is ; or R3 is amino.
69. The compound of claim 68, wherein X, is absent.
70. The compound of claim 68, wherein X2 is absent.
71. The compound of claim 68, wherein X1 is absent; and X2 is absent.
72. The compound of claim 68, wherein R1 is -OH or -NH2.
73. The compound of claim 68, wherein R2 is aralkyl or heteroaralkyl.
74. The compound of claim 68, wherein R2 is aralkyl.
75. The compound of claim 68, wherein R3 is alkenyl, amino, (amino)alkyl, amido, (amido)alkyl, (keto)alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl.
76. The compound of claim 68,wherein R3 is (amido)alkyl or heterocyclyl.
77. The compound of claim 68, wherein R4 is (amido)alkyl or heterocyclyl.
78. The compound of claim 68, wherein R5 is alkyl or aryl.
79. A compound, or a pharmaceutically acceptable salt thereof, of formula XII:

wherein, independently for each occurrence, ; wherein, independently for each occurrence, W is selected, from the group consisting of -NHR7 or -NHR(CH2)p N(R7 )2; R7 is selected from the group consisting of hydrogen, alkyl, aralkyl, heteroaralkyl and acyl; and p is inclusive; and R6 is
80. The compound of claim 79, wherein R3 is
81. The compound of claim 79, wherein R4 is
82. The compound of claim 79, wherein R6 is
83. The compound of claim 79, wherein R6 is
84. A compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of:

85. A compound, or a pharmaceutically acceptable salt thereof, of formula XII:

wherein, independently for each occurrence, R3 is R4 is wherein, independently for each occurrence, W is selected, from the group consisting of -NHR7 or -NHR(CH2)p N(R7)2; R7 is selected from the group consisting of hydrogen, alkyl, aralkyl, heteroaralkyl and acyl; and p is inclusive; and
86. The compound of claim 85, wherein R3 is
87. The compound of claim 85, wherein R3 is
88. The compound of claim 85, wherein R3 is
89. The compound of claim 85, wherein R4 is
90. The compound of claim 85, wherein R6 is
91. A compound, or pharmaceutically acceptable salt thereof, selected from the group consisting of:
92. A compound, or a pharmaceutically acceptable salt thereof, of formula iso-XII:

wherein, independently for each occurrence, R3 is alkyl, (amino)alkyl, (amido)alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, (heterocyclyl)alkyl, aralkyl or heteroaralkyl;

R4 is aryl, heteroaryl, aralkyl or heteroaralkyl; and R6 is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl.
93. A pharmaceutical composition, comprising a pharmaceutical carrier and a therapeutically effective amount of a compound of any one of claims 1-92.
94. A method for treating an HIV infection, comprising administering to a mammal in need thereof a therapeutically effective amount of a compound of any one of claims 1-92, or a therapeutically effective amount of a pharmaceutical composition of claim 93.
95. The method of claim 94, wherein the compound is administered as part of a highly active antiretroviral therapy (HAART) regimen.
96. The method of claim 94, wherein the pharmaceutical composition is administered as part of a highly active antiretroviral therapy (HAART) regimen.
97. The method of any one of claims 94-96, further comprising administering a second therapeutic agent.
98 The method of claim 97, wherein the second therapeutic agent is a non-nucleoside reverse transcriptase inhibitor (NNRTI), a nucleoside reverse transcriptase inhibitor (NRTI), a nucleotide reverse transcriptase inhibitor, an entry inhibitor, an integrase inhibitor, a fusion inhibitor, a protease inhibitor, or an inhibitor of a metabolic enzyme.
99. The method of claim 97, wherein the second therapeutic agent is selected from the group consisting of efavirenz (Sustiva.TM.), nevirapine (Viramune.TM.), delavirdine (Rescriptor.TM.), AZT (zidovudine, Retrovir.TM.)/3TC (lamivudine, Epivir.TM.), d4T
(stavudine, Zerit.TM.)/3TC, ddI (didanosine, Videx.TM./VidexEC.TM.), ddC
(zalcitabine, Hivid.TM.), d4T, tenofovir (Viread.TM.), and enfuvirtide (Fuzeon.TM.).
100. The method of claim 97, wherein the second therapeutic agent is selected from the group consisting of amprenavir (Agenerase®; APV), tipranavir (Aptivus®; TPV), indinavir (Crixivan®; IDV), saquinavir (Invirase®; SQV), lopinavir and ritonavir (Kaletra®; LPV), fosamprenavir (Lexiva®; FPV), ritonavir (Norvir®;
RTV), atazanavir (Reyataz®; ATZ), nelfinavir (Viracept®; NFV), brecanavir, and darunavir.
101. The method of claim 97, wherein the second therapeutic agent is ritonavir (Kaletra®; LPV).
102. The method of claim 97, wherein the second therapeutic agent is selected from the group consisting of zidovudine (AZT; Azidothymidine; Retrovir®), didanosine (Dideoxyinosine; ddI; Videx®), zalcitabine (Dideoxycytidine; ddC;
Hivid®), lamivudine (3TC; Epivir®), stavudine (2',3'-didehydro-3'-deoxythymidine;
D4T;
Zerit®), abacavir succinate (1592U89 succinate; Ziagen® ABC), Combivir®
(lamivudine & zidovudine; (-)-3TC & AZT), and Trizivir® (abacavir &
lamivudine & zidovudine; ABC & (-)-3TC & AZT).
103. The method of claim 97, wherein the second therapeutic agent is selected from the group consisting of nevirapine (BI-RG-587; Viramune®), delavirdine (BHAP;
U-90152; Rescriptor®), and (efavirenz; DMP-266; Sustiva®).
104. The method of claim 97, wherein the second therapeutic agent is T-20 (Fuzeon®;
Enfuvirtide; DP-178; Pentafuside; GP41 127-162 AA).
105. The method of claim 97, wherein the second therapeutic agent is TMCC114.
106. The method of claim 97, further comprising administering a reverse transcriptase inhibitor.
107. The method of claim 97, wherein the second therapeutic agent is lupinavir.
CA002681718A 2007-03-23 2008-03-24 Hiv-1 protease inhibitors Abandoned CA2681718A1 (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US91981907P 2007-03-23 2007-03-23
US91989607P 2007-03-23 2007-03-23
US60/919,896 2007-03-23
US60/919,819 2007-03-23
US94178607P 2007-06-04 2007-06-04
US94182907P 2007-06-04 2007-06-04
US60/941,786 2007-06-04
US60/941,829 2007-06-04
PCT/US2008/058004 WO2008118849A2 (en) 2007-03-23 2008-03-24 Hiv-1 protease inhibitors

Publications (1)

Publication Number Publication Date
CA2681718A1 true CA2681718A1 (en) 2008-10-02

Family

ID=39789249

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002681718A Abandoned CA2681718A1 (en) 2007-03-23 2008-03-24 Hiv-1 protease inhibitors

Country Status (4)

Country Link
EP (1) EP2139883A4 (en)
CN (1) CN101702908A (en)
CA (1) CA2681718A1 (en)
WO (1) WO2008118849A2 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010135424A1 (en) * 2009-05-19 2010-11-25 Glaxosmithkline Llc Chemical compounds
WO2011061590A1 (en) 2009-11-17 2011-05-26 Hetero Research Foundation Novel carboxamide derivatives as hiv inhibitors
JOP20180009A1 (en) 2017-02-06 2019-01-30 Gilead Sciences Inc Hiv inhibitor compounds
AR112413A1 (en) 2017-08-17 2019-10-23 Gilead Sciences Inc SOLID FORMS OF AN HIV CAPSID INHIBITOR
AR112412A1 (en) 2017-08-17 2019-10-23 Gilead Sciences Inc CHOLINE SALT FORMS OF AN HIV CAPSID INHIBITOR
CN108220482A (en) * 2018-02-11 2018-06-29 广西医科大学 The primer for the treatment of AIDS drug NRTIs resistant mutational sites G73S and its application
CA3089590C (en) 2018-02-15 2022-12-06 Gilead Sciences, Inc. Pyridine derivatives and their use for treating hiv infection
KR20210033492A (en) 2018-07-16 2021-03-26 길리애드 사이언시즈, 인코포레이티드 Capsid inhibitors for the treatment of HIV
TWI829205B (en) 2018-07-30 2024-01-11 美商基利科學股份有限公司 Anti-hiv compounds
WO2020150369A1 (en) * 2019-01-15 2020-07-23 Ghosh Arun K Hiv-1 protease inhibitors and uses thereof
US11787825B2 (en) 2021-12-03 2023-10-17 Gilead Sciences, Inc. Therapeutic compounds for HIV virus infection

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040122000A1 (en) * 1981-01-07 2004-06-24 Vertex Pharmaceuticals Incorporated. Inhibitors of aspartyl protease
US5877334A (en) * 1996-07-23 1999-03-02 Abbott Laboratories Intermediates useful in solid phase synthesis method
AU4828199A (en) * 1998-06-23 2000-01-10 Board Of Trustees Of The University Of Illinois, The Multi-drug resistant retroviral protease inhibitors and associated methods
TWI260322B (en) * 1999-02-12 2006-08-21 Vertex Pharma Inhibitors of aspartyl protease
TWI286476B (en) * 2001-12-12 2007-09-11 Tibotec Pharm Ltd Combination of cytochrome P450 dependent protease inhibitors
EA200401437A1 (en) * 2002-04-26 2005-04-28 Джилид Сайэнс, Инк. Nucleoside reverse transcriptase inhibitors for treatment of HIV infection, PROCESS FOR THEIR PREPARATION (VARIANTS) AND A METHOD OF INCREASING half-life of non-nucleoside compounds active against retroviruses, and methods of preparing a pharmaceutical composition having specificity for leukocytes
ATE371652T1 (en) * 2002-05-17 2007-09-15 Tibotec Pharm Ltd SUBSTITUTED BENZISOXAZOLE SULFONAMIDE WITH BROAD-BAND HIV PROTEASE INHIBITING EFFECT
JP2007528423A (en) * 2004-03-11 2007-10-11 セコイア、ファーマシューティカルズ、インコーポレイテッド Antiviral resistance retroviral protease inhibitors
WO2006104646A1 (en) * 2005-03-11 2006-10-05 Smithkline Beecham Corporation Hiv protease inhibitors

Also Published As

Publication number Publication date
WO2008118849A2 (en) 2008-10-02
EP2139883A2 (en) 2010-01-06
EP2139883A4 (en) 2011-06-22
WO2008118849A3 (en) 2008-12-18
CN101702908A (en) 2010-05-05

Similar Documents

Publication Publication Date Title
CA2681718A1 (en) Hiv-1 protease inhibitors
US8933075B2 (en) Compounds useful as antiviral agents, compositions, and methods of use
CA2658545A1 (en) Bisfuranyl protease inhibitors
US20110178092A1 (en) HIV-1 Protease Inhibitors
AU2002302363B2 (en) Broadspectrum 2-(substituted-amino)-benzothiazole sulfonamide HIV protease inhibitors
TWI385173B (en) Substituted aminophenylsulfonamide compounds as hiv protease inhibitor
CA2667445A1 (en) Hiv-1 protease inhibitors
AU2003262561B2 (en) Broadspectrum 2-amino-benzothiazole sulfonamide HIV protease inhibitors
AU2002361235B2 (en) Broadspectrum heterocyclic substituted phenyl containing sulfonamide HIV protease inhibitors
CA2485903C (en) Broadspectrum substituted benzisoxazole sulfonamide hiv protease inhibitors
US7148243B2 (en) Antivirals
EP2196453A1 (en) Novel substituted aryl derivatives, their process of preparation and their therapeutical uses as anti-HIV agents
CA2667449A1 (en) Hiv-1 protease inhibitors, and methods of making and using them
AU2009301131B2 (en) New amide compounds as boosters of antivirals
EP1960381B1 (en) Aminophenylsulfonamide derivatives as hiv protease inhibitor
IL163960A (en) Broadspectrum substituted benzimidazole sulfonamide hiv protease inhibitors and compositions comprising such

Legal Events

Date Code Title Description
FZDE Discontinued