WO2006017369A2 - Use of a farnesyl transferase inhibitor in the treatment of viral infections - Google Patents

Use of a farnesyl transferase inhibitor in the treatment of viral infections Download PDF

Info

Publication number
WO2006017369A2
WO2006017369A2 PCT/US2005/025176 US2005025176W WO2006017369A2 WO 2006017369 A2 WO2006017369 A2 WO 2006017369A2 US 2005025176 W US2005025176 W US 2005025176W WO 2006017369 A2 WO2006017369 A2 WO 2006017369A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
substituted
unsubstituted
alkyl
aryl
Prior art date
Application number
PCT/US2005/025176
Other languages
French (fr)
Other versions
WO2006017369A3 (en
Inventor
Steven Zeichner
Vyjayanthi Krishnan
Original Assignee
Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services
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 Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services filed Critical Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services
Publication of WO2006017369A2 publication Critical patent/WO2006017369A2/en
Publication of WO2006017369A3 publication Critical patent/WO2006017369A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV

Definitions

  • the present invention relates to methods for treatment or prevention of an HTV infection and, more particularly, to use of one or more farnesyl transferase inhibitor compounds to treat a subject suffering from or susceptible to an HIV infection.
  • human immunodeficiency virus type 1 (HIV-I, also referred to as HTLV- m LAV or HTLV-IH/LAV) and, to a lesser extent, human immunodeficiency virus type 2 (HIV-2) is the etiological agent of the acquired immune deficiency syndrome (AIDS) and related disorders.
  • This disease is characterized by a long asymptomatic period followed by the progressive degeneration of the immune system and the central nervous system.
  • Studies of the virus indicate that replication is highly regulated, and both latent and lytic infection of the CD4 positive helper subset of T-lymphocytes occur in tissue culture. Zagury, et al., Science, 231:850-853 (1986).
  • the expression of the virus in infected patients also appears to be regulated as the titer of infectious virus remains low throughout the course of the disease.
  • Both HTV-I and 2 share a similar structural and function genomic organization, having regulatory genes such as tat, re ⁇ , nef, in addition to structural genes such as enx, gag and pal.
  • AIDS While AIDS, itself, does not necessarily cause death, in many individuals the immune system is so severely depressed that various other diseases (secondary infections or unusual tumors) such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus related lymphomas among others occur, which ultimately results in death. These secondary infections may be treated using other medications. However, such treatment can be adversely affected by the weakened immune system.
  • Some humans infected with the AIDS virus seem to live many years with little or no symptoms, but appear to have persistent infections. Another group of humans suffers mild immune system depression with various symptoms such as weight loss, malaise, fever and swollen lymph nodes. These syndromes have been called persistent generalized lymphadenopathy syndrome (PGL) and AIDS related complex (ARC) and may or may not develop into AIDS. In all cases, those infected with the HTV are believed to be persistently infective to others.
  • PDL persistent generalized lymphadenopathy syndrome
  • ARC AIDS related complex
  • LTR long terminal repeat
  • HTV-I The activity of HTV-I is determined by the complex interaction of positive and negative transcriptional regulators that bind to specific sequences within the LTR. Cullen, B.R., et al., Cell, 5&:423-426 (1989). Changes in the quantity or quality of these factors may underlie the activation of transcription of HTV-I and HTV-2 latent provirus by a myriad of stimuli. See, e.g. Fauci, A.S.,
  • FTase inhibitor compounds can be useful for treating cells infected by immunodeficiency viruses and methods of preventing cells from becoming infected by immunodeficiency viruses, preferably human immunodeficiency viruses such as HTV.
  • therapeutic methods that in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor compound) to mammalian cells that are infected with an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
  • a compound that inhibits farnesyl transferase a FTase inhibitor compound
  • the invention further methods that in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor) to a patient in need of treatment, such as a mammal suffering from or susceptible to an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
  • a FTase inhibitor a compound that inhibits farnesyl transferase
  • FTase inhibitor compounds can be employed in the methods of the invention.
  • suitable compounds have been reported previously including those in U.S. Patents 5,238,922; 5,571,792; and 5,571,835; WO 94/10138; WO 94/04561; WO 94/10138; WO 96/21456; and WO 97/02817.
  • FTase inhibitor compounds for use in the methods of the invention exhibit good activity in a standard in vitro FTase inhibition assay (specifically the standard assay defined below), preferably an ICso (concentration required to inhibit FTase activity by 50% relative to control) in such an assay of about 100 nM or less, more preferably an IC 50 about 50 nM or less.
  • a standard in vitro FTase inhibition assay specifically the standard assay defined below
  • ICso concentration required to inhibit FTase activity by 50% relative to control
  • FTase inhibitor compounds for use in accordance with the invention include the peptidomimetic compounds, including FTI276, FTI277 and L-744832.
  • Non-peptidomimetic FTase inhibitor compounds also are preferred, including BMS214662, Rl 15777 and SCH66336.
  • FTase inhibitor compounds used in accordance with the present invention can induce lytic replication in cells latently infected with an immunodeficiency virus such as HTV by targeting a product or products of the genes encoding proteins in the Ras and Rho signaling pathways which are differentially expressed in latently infected cells and lytic replicating cells.
  • the compounds of the present invention can treat cells infected acutely and chronically by immunodeficiency viruses, for example, HIV, preferably HIV-I, and thus can be used to treat humans infected by HTV.
  • immunodeficiency viruses for example, HIV, preferably HIV-I
  • HTV immunodeficiency virus
  • treatment of those diagnosed as having AIDS as well as those having ARC, PGL and those not yet exhibiting such conditions for example, treatment of those diagnosed as having AIDS as well as those having ARC, PGL and those not yet exhibiting such conditions.
  • Another aspect is a method of reducing latent HTV-reservoirs in a subject including administration of an effective amount of one or more FTase inhibitor compounds.
  • the latent HTV-reservoirs are collections of latent HTV-infected cells, that is cells in which the HIV-re ⁇ lication is considered to be in a latent state.
  • aspects or embodiments are a method of reducing latent HTV reservoirs in an HIV-infected subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds; a method of increasing (e.g., 10- 15 fold relative to cells treated with AZT) expression of p24 in a latently HIV-infected cell comprising administration to the cell of an effective amount of one or more FTase inhibitor compounds; a method of activating latent HlV-provirus in a cell in a subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds; and a method of activating latent HlV-provirus in a cell in a subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds
  • the methods delineated herein include administering to a subject (e.g., a human or an animal) in need thereof an effective amount of one or more FTase inhibitors, e.g., compounds as deline
  • the methods can also include the step of identifying that the subject is in need of treatment of diseases or disorders described herein, e.g., identifying that the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells.
  • the identification can be in the judgment of a subject or a health professional and can be subjective (e.g., opinion) or objective (e.g., measurable by a test or a diagnostic method).
  • Tests for HTV infection include polymerase chain reaction-based (PCR-based) amplification and detection of viral RNA; Western blot detection of anti- HIV antibodies; agglutination assays for anti-HTV antibodies; ELISA-based detection of HlV-specific antigens (e.g., p24); line immunoassay (LIA); and other methods known to one of ordinary skill in the art.
  • PCR-based polymerase chain reaction-based
  • Western blot detection of anti- HIV antibodies e.g., agglutination assays for anti-HTV antibodies
  • ELISA-based detection of HlV-specific antigens e.g., p24
  • line immunoassay line immunoassay
  • the methods of the invention can include the step of obtaining a sample of biological material (such as a bodily fluid) from a subject; testing the sample to determine the presence or absence of detectable HIV infection, HIV particles, or HIV nucleic acids; and determining whether the subject is in need of treatment according to the invention, i.e. * identifying whether the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells.
  • a sample of biological material such as a bodily fluid
  • the methods delineated herein can further include the step of assessing or identifying the effectiveness of the treatment or prevention regimen in the subject by assessing the presence, absence, increase, or decrease of a marker, including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HIV infection marker; preferably this assessment is made relative to a measurement made prior to beginning the therapy.
  • a marker including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HIV infection marker; preferably this assessment is made relative to a measurement made prior to beginning the therapy.
  • Such assessment methodologies are known in the art and can be performed by commercial diagnostic or medical organizations, laboratories, clinics, hospitals and the like.
  • the methods can further include the step of taking a sample from the subject and analyzing that sample.
  • the sample can be a sampling of cells, genetic material, tissue, or fluid (e.g., blood, plasma, sputum, etc.) sample.
  • the methods can further include the step of reporting the results of such analyzing to the subject or other
  • the invention provides a method of treating HTV infection in a subject.
  • the method comprises the steps of identifying a subject as in need of reactivation of replication processes in latent HTV-infected cells; and administrating of an effective amount of a farnesyl transferase (FTase) inhibitor to the subject to reactivate the viral replication process.
  • FTase farnesyl transferase
  • the farnesyl transferase inhibitor is one or more of FTI277, L-744832, BMS214662, Rl 15777 and SCH66336.
  • one or more peptidomimetic FTase inhibitor compounds are administered to the subject; in other preferred embodiments, one or more non-peptidomimetic FTase inhibitor compounds are administered to the subject.
  • the one or more FTase inhibitor compounds are of any one of the general formulae (a) through (gg) described herein.
  • the administered FTase inhibitor compound has an IC50 of about 100 nM or less in a standard in vitro farnesyl transferase inhibition assay.
  • the invention provides a method of inhibiting HIV replication in a subject or a cell.
  • the method comprises the steps of identifying a subject or cell as in need of reactivation of replication processes in latent HTV-infected cells; administering an effective amount of a farnesyl transferase (FTase) inhibitor to the subject or cell to reactivate the viral replication process; and administering one or more HTV antiviral agents to the subject or cell to inhibit induced lytic HIV viral replication.
  • FTase farnesyl transferase
  • the invention provides a method of treating latently HIV- infected cells in a subject comprising administration to the cells one or more FTase inhibitor compounds.
  • the invention provides a method of modulating lytic replication in an HIV-infected cell in a subject identified as in need of such treatment.
  • the method comprises the step of administration to the subject of an effective amount of one or more FTase inhibitor compounds.
  • the cell is a human cell capable of sustaining a latent provirus.
  • the cell is a lymphocytic cell.
  • the cell is a monocytic cell.
  • the cells are human cells.
  • the invention provides a method of reducing latent HTV reservoirs in an HIV-infected subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
  • Another aspect is a method of reducing latent HTV-reservoirs in a subject by controlled activation of viral replication including administration of an effective amount of a farnesyl transferase inhibitor.
  • Controlled activation is that activation initiated by administration of an effective amount of an FTase inhibitor such that the FTase inhibitor reactivates (directly or indirectly) replication processes in latent HIV- infected cells.
  • the latent HIV-reservoirs are collections of latent BDV-infected cells, that is cells in which the HlV-replication is considered to be in a latent state.
  • the method can further include administration with one or more antiviral agents, thus both depleting the latent cell reservoir and inhibiting induced viral lytic replication, whereupon the cells in that state are subjected to and susceptible to the antiretroviral therapy, which controls viral proliferation.
  • the invention provides a method of activating latent HTV-provirus in a cell in a subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
  • the method further comprises the step of administration of one or more additional anti-viral (e.g., anti- HIV) therapeutic agents to the subject or cell.
  • additional anti-viral agent(s) are a reverse transcriptase inhibitor, a protease inhibitor, or combination thereof.
  • the invention provides a method of increasing expression of p24 in a latently HIV-infected cell comprising administration to the cell of an effective amount of a farnesyl transferase inhibitor.
  • the cell is a lymphocytic cell or a monocytic cell.
  • the cell is a human cell, i.e., any human cell capable of sustaining a latent provirus.
  • the invention provides a method of screening for a compound capable of activating latent HTV-infected cells comprising contacting a FTase inhibitor test compound with an ACH-2 cell or Jl .1 cell or Ul cell and determining the level of p24 expression, hi preferred embodiments, increased expression of p24 in cells treated with a test compound relative to non-treated cells indicates a compound capable of activating latent HTV-infected cells.
  • the invention also provides pharmaceutical compositions comprising one or more FTase inhibitor compounds and a suitable carrier therefore for use in the conditions referred to above.
  • the methods delineated herein can further include the step of assessing or identifying the effectiveness of the treatment or prevention regimen in the subject by assessing the presence, absence, increase, or decrease of a marker, including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HTV infection marker.
  • a marker including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HTV infection marker.
  • the methods can further include the step of taking a sample from the subject and analyzing that sample.
  • the sample can be a sampling of cells, genetic material, tissue, or fluid (e.g., blood, plasma, sputum, etc.) sample.
  • the methods can further include the step of reporting the results of such analyzing to the subject or other health care professional.
  • Figure 1 shows the flow cytometric analysis of chronically infected ACH-2 cells before and after induction. Uninduced cells and cells from serial time points were fixed and permeabilized for intracellular p24 labeling. As an isotype control, cell samples were labeled with mouse IgGl . For each sample, 100,000 events were collected. In the figure, each sample histogram labeled for p24 (darker color) is overlaid with the control histogram labeled for the isotype control (lighter color).
  • A Uninduced ACH-2 cells, showing minimal p24 accumulation with 8.2% of cells infected
  • B ACH-2 cells at 0.5 hours post induction (p.i.) with 7.4% of cells positive for p24
  • C ACH-2 cells at 6 hours p.i, with 61.6% cells infected
  • D ACH-2 cells at 12, 18, and 24 hours p.i., respectively, showing complete infection.
  • Flow cytometric analysis was performed on all batches of cells to ensure active replication of HTV following induction with PMA. Data from one induction experiment is shown. Data indicate that viral replication occurs in an ordered manner post induction, and complete infection of cells is achieved within 12 hours post induction of chronically infected ACH-2 cells.
  • Figure 2 shows the levels of expression of multiply spliced (MS HIV-I) and unspliced (US HTV-I) mRNA prior and post induction of chronically infected ACH-2 cells.
  • Real time RT-PCR reactions were carried out using Taqman probes specific for early (multiply spliced) and late (unspliced) transcripts of HTV-I, tagged with FAM and TAMRA fluorescent dyes at the 5' and 3' ends respectively. Reactions were performed in triplicate for each time point as described in the Methods section and average values are shown. Maximal fold change in mRNA levels for early transcripts (MS HTV-I) was observed 8 hours post induction. Fold change for late transcripts (US
  • FIG. 3 shows the hierarchical clustering of differentially expressed cellular genes before and after induction of chronically infected ACH-2 cells.
  • the figure shows the hierarchical clustering of the cellular genes that showed significant differential expression (p ⁇ 0.001) across the time course (before induction up to 96 hours post induction), following reactivation of chronically infected ACH-2 cells as per the criteria described in the Methods.
  • Genes that are on the color scale ⁇ 1 showed up regulation, those on the color scale ⁇ 1 were down regulated, while those that did not show any change with respect to normalized matched control are shown in black.
  • the gray areas indicate missing data for the given gene and time point.
  • the magnified panels indicate selected kinetic profiles that are seen before and following induction into active viral replication.
  • A Up regulation of selected genes observed before induction;
  • B Up regulation of genes immediately following induction;
  • C Genes that are up regulated prior to induction and down regulated 12-24 hours post induction;
  • D Genes that are up regulated in the early stage following reactivation, but are down regulated in the intermediate stage;
  • E Genes that are down regulated before induction but are up regulated in the intermediate stage followed by down regulation in the late stage (48-96 hours p.i.).
  • Figure 4 shows the trends seen in pathways that show differential expression before and after induction of chronically infected ACH-2 cells. Pathway profiles observed prior to induction and following reactivation of ACH-2 cells with PMA over a period of 96 hours. The figure shows the number of genes in each pathway that were differentially expressed in a particular pathway,.
  • A indicates the pathways that were maximally altered prior to induction.
  • B includes the pathways that showed maximum change during the early phase of the lytic cycle, (0.5-8 hours p.i.).
  • C represents the pathways that showed maximal change during the period of 12-24 hours post induction. Most pathways did not show any change during the period of 48-96 hours post induction.
  • FIG. 5 shows the hierarchical clustering of genes that show differential expression across three chronically infected cell lines prior to induction. Hierarchical clustering of differentially expressed genes that show a significant change in expression (p ⁇ 0.001), in the chronically infected cell lines ACH-2, Ul and Jl.1. Genes shown on the color scale >1 are up regulated, those on the color scale ⁇ 1 exhibit down regulation, while black indicates normal expression. In several cases (e.g., FCGR2A, PPIB), dark gray areas indicate missing values. Many genes are altered similarly across the cell lines.
  • Each cell line also shows some unique patterns of cellular expression. Data are the average of values from eight independent samples per cell line. The magnified portions of the cluster highlight some of the patterns of gene expression across the cell lines.
  • A shows genes that are up regulated in all three cell lines;
  • B shows genes that are down regulated in all three cell lines;
  • C indicates the genes that are up regulated in ACH-2 and Jl.1 and down regulated in Ul;
  • D indicates genes which show no significant similarity in their expression in the three cell lines.
  • Figure 6 shows the effects of specific agents on HIV p24 production in latently infected JLl cells.
  • Different concentrations of the FTase inhibitor L-744,832 were tested in latently infected Jl.1 cells treated with 250 nM AZT.
  • Samples were collected 24 hours after addition of agent and p24 concentrations were determined by ELISA.
  • p24 production from cells treated by TNF-alpha was used as a positive control in J 1.1 cells compared to control (AZT treated cells).
  • p24 production from untreated cells No AZT was also determined. Experiments were performed in triplicate and are representative of three independent experiments.
  • Figure 7 shows the effect of L-744832 on several HTV latently infected Jurkat clones.
  • Figure 8 shows shows the effect of L-744832 on viral reactivation in a sample from a 1-year aviremic patient.
  • Figure 9 shows the effect of L-744832 on viral reactivation in a sample from a long-term (8-year) aviremic patient.
  • Table 1 Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells. List of selected classes of genes based on known function that are differentially expressed in latently infected ACH-2 cells, relative to uninfected parental cell line, A3.01. A number of genes involved in similar cellular functions previously not associated with presence of proviral HIV were altered coordinately even during the latent non-replicative stage.
  • Table 1 Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells.
  • farnesyl transferase (FTase) inhibitors e.g., compounds of the formulae herein, can be used to reactivate viral replication processes in cells infected by an immunodeficiency virus, preferably human cells infected with HTV and thus can be used for treatment in BXV-infected individuals.
  • Fase farnesyl transferase
  • the methods of the invention in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor) to a patient in need of treatment, such as a mammal suffering from or susceptible to an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
  • a FTase inhibitor a compound that inhibits farnesyl transferase
  • FTase inhibitor compounds can be employed in the methods of the invention.
  • suitable compounds have been reported previously including those in U.S. Patents 5,238,922; 5,571,792; and 5,571,835; WO 94/10138; WO 94/04561; WO 94/10138; WO 96/21456; and WO 97/02817.
  • FTase inhibitor compounds for use in the methods of the invention exhibit good activity in a standard in vitro FTase inhibition assay, preferably an IC50 (concentration required to inhibit FTase activity by 50% relative to control) in such an assay of about 100 nM or less, more preferably an IC 50 about 50 nM or less.
  • IC50 concentration required to inhibit FTase activity by 50% relative to control
  • a standard in vitro FTase inhibition assay includes the following steps a) through c): a) admixing in a suitable assay solution 1) a potential FTase inhibitor compound, 2) [ 3 H]farnesyl diphosphate, 3) farnesyl transferaseand 4) H-Ras; b) incubating the test mixture for 15 minutes at 37 0 C; and c) measuring utilization of [ 3 H] farnesyl diphosphate over that time relative to a control mixture that is prepared and incubated under the same conditions as the assay mixture but does not include the potential inhibitor compound.
  • a suitable assay solution includes 50 mM HEPES 5 pH 7.5, 5 mM MgCl 2 , 5 mM dithiothreitol. References herein to a standard in vitro farnesyl transferase inhibition assay are intended to refer to that protocol. That protocol also has been described in A.M. Garica et al., J. Biol Chem., 268: 18415-18418 (1993).
  • an IUPAC-type name and other designator such as L-744,832, SCH 66336, etc.
  • additional information is provided beneath the corresponding compound structure, such as the molecular formula of the structurally depicted compound.
  • FTI-277 molecular formula: C22H29N3O3S2; 2- ⁇ [5-(2-amino-3-mercapto- propylamino)-biphenyl-2-carbonyl]-amino ⁇ -4-methylsulfanyl-butyric acid methyl ester
  • any particular farnesyl transferase inhibitor in the therapeutic methods of the invention can be readily determined.
  • compounds with superior intrinsic inhibitory activity against and selectivity for farnesyl transferase can be identified through the in vitro assays discussed above and herein.
  • suitable FTase inhibitors compounds for use in the methods of the invention are disclosed below (including those compounds of groups (a) through (gg) as those groups of compounds are defined below, and other compounds defined below). It should be appreciated however that the present invention is not limited by the particular FTase inhibitor, and the invention is applicable to any such FTase inhibitor compound now known or subsequently discovered or developed.
  • FTase inhibitor compounds suitable for use in the methods of the invention will include those compounds that incorporate a cysteinyl or sulfhydryl containing moiety at the N-terminus of the molecule. More specifically, the following compounds are useful in the methods of the invention:
  • Ai an aliphatic amino acid
  • a 2 an aliphatic amino acid
  • X any amino acid
  • Xaa 2 any amino acid in the natural L-isomer form
  • Xaa any amino acid
  • Xaa the amino acid phenyl alanine or a p-fluorophenylalanine
  • Xaa 3 any amino acid
  • Xaa any amino acid in the natural L-isomer form
  • dXaa any amino acid in the natural L-isomer form
  • Xaa 3 any amino acid in the natural L-isomer form
  • X 3 Y, and Z are independently H 2 or O, provided that at least one of these is H 2 ;
  • R is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R 1 NH may be absent;
  • R 2 j R 3 and R 4 are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring; and
  • R is H or a straight or branched chain aliphatic group, which may be substituted with an aromatic or heteroaromatic group;
  • X and Y are independently H 2 or O, provided that at least one of these is H 2 ;
  • R 1 is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R NH may be absent;
  • R and R are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the.aliphatic substituents may be substituted with an aromatic or heteroaromatic ring;
  • Z is O or S; and n is 0, 1 or 2;
  • X and Y are independently H 2 or 0, provided that at least one of these is H 2 ;
  • R is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R NH may be absent;
  • R and R are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring;
  • Z is O or S; and n is 0, 1 or 2; (h) compounds of the following formula, which compounds are also disclosed in U.S.
  • X and Y are independently H 2 or O;
  • R 1 is an alkyl group, hydrogen, an acyl group, an alkylsulfonyl group or arylsulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbons atoms, which alternatively may be substituted with an aryl group;
  • R is the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heterocyclic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic s ⁇ bstituents may be substituted with an aromatic or heteroaromatic ring;
  • R 3 is an aromatic or heteroaromatic ring or in the alternative an alkyl group or an aryl or heteroaryl substituted alkane, wherein the aromatic ring is unsubstituted or in the alternative, substituted with one or more groups which maybe alkyl, halo, alkoxy, trifluoromethyl, or sulfamoyl groups, and which may be polycyclic; (i) compounds of the following formulae, which compounds are also disclosed in U.S. Patent No. 5,326,773 and PCT Publication No. WO 94/10137, incorporated herein by reference,
  • R and R a are independently selected from hydrogen, a Ci-C 6 alkyl group, a CI-CO acyl group, an aroyl group, a Ci-C 6 alkylsulfonyl group, Ci-C 6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally substituted with substituted or unsubstituted aryl or heterocycle;
  • R 2 , R 3 and R are independently selected from: a) a side chain of naturally occurring amino acids, b) an oxidized form of a side chain of naturally occurring amino acids selected from methionine sulfoxide and methionine sulfone, c) substituted or unsubstituted Ci-C 8 alkyl, C 3 -C 8 cycloalkyl, C 2 -Cs alkenyl, aryl or heterocycle groups, wherein the aliphatic substituent is optionally substituted with an aryl, heterocycle or C 3 -C 8 cycloalkyl;
  • R ,5b is a C 1 -C 6 alkyl group, a C 1 -Ce acyl group, an aroyl group, a Ci-C 6 alkylsulfonyl group, Ci-C 6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally
  • R is a substituted or unsubstituted aliphatic, aryl or heterocyclic group, wherein the aliphatic substituent is optionally substituted with an aryl or heterocyclic ring; and n is 0, 1 or 2;
  • R is selected from hydrogen, a C 1 -CO alkyl group, a Ci-Ce acyl group, an aroyl group, a Ci-Ce alkylsulfonyl group, Ci-C 6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally substituted with substituted or unsubstituted aryl or heterocycle;
  • R , R and R are independently selected from: a) a side chain of naturally occurring amino acids, b) an oxidized form of a side chain of naturally occurring amino acids selected from methionine sulfoxide and methionine sulfone, c) substituted or unsubstituted Ci-C 8 alkyl, C 3 -Cg cycloalkyl, C 2 -C 8 alkenyl, aryl or het ⁇ rocycle groups, wherein the aliphatic substituent is optionally substituted with an aryl, heterocycle or C 3 -C 8 cycloalkyl;
  • R is a substituted or unsubstituted aliphatic, aryl or heterocyclic group, wherein the aliphatic substituent is optionally substituted with an aryl or heterocyclic ring
  • n is 0, 1 or 2;
  • R 1 is hydrogen, an alkyl group, an aralkyl group, an acyl group, an aracyl group, an aroyl group, an alkylsulfonyl group, aralkylsulfonyl group or arylsulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms;
  • R 2 , R 3 and R 5 are the side chains of naturally occurring amino acids, including their oxidized forms which may be methionine sulfoxide or methionine sulfone, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring;
  • R 4 is hydrogen or an alkyl group, wherein the alkyl group comprises straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms;
  • R 6 is a substituted or unsubstituted aliphatic, aromatic or heteroaromatic group such as saturated chains of 1 to 8 carbon atoms, which may be branched or unbranched, wherein the aliphatic substituent may be substituted with an aromatic or heteroaromatic ring;
  • T is O or S(O) n ,; m is 0, 1 or 2; and n is 0, 1 or 2; (1) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 95/00497, incorporated herein by reference,
  • R and R 1 are independently selected from H, Ci -4 alkyl, or aralkyl;
  • R 2 , R 3 , R 4 , and R 5 are independently selected from H, Ci -8 alkyl, alkenyl,
  • R 2 , R 3 , R 4 , and R 5 are optionally attached to the same carbon atom;
  • Y is aryl, heterocycle, unsubstituted or substituted with one or more of:
  • Ci -4 alkyl unsubstituted or substituted with: a) C 1-4 alkoxy, b) NR 6 R 7 , c) C 3-6 cycloalkyl, d) aryl or heterocycle, e) HO,
  • W is Ha or O
  • Z is aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more of the following:
  • R , R and R are independently selected from H, Ci -4 alkyl, C 3- O cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Ci -4 alkoxy, b) aryl or heterocycle, c) halogen,
  • R ⁇ and R 7 may be joined in a ring
  • R and R may be joined in a ring
  • R 9 is C 1-4 alkyl or aralkyl.
  • R 1 is selected from: a) hydrogen, 0 b) R 8 S(O) 2 -, R 8 C(O)-, (R 8 ) 2 NC(O> or R 9 OC(O)-, and c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R 8 O-, R 8 S(O) 1n -, R 8 C(O)NR 8 -, CN 3 (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , OrR 9 OC(O)NR 8 -; R a and R are independently selected from: 5 a) hydrogen, b) Ci-C 6 alkyl unsubstituted or substituted by alkenyl, R 8 O-
  • R and R are independently selected from: a) a side chain of a naturally occurring amino acid, 5 b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted Ci-C 20 alkyl, C 2 -C 20 alkenyl, C 3 -Ci 0 O cycloalkyl, aryl or heterocyclic group, wherein the substiruent is selected from F, Cl, Br, N(R 8 ) 2 , NO 2 , R 8 O-, R 8 S(O) n O R 8 C(O)NR 8 -, CN, (R 8 ) 2 N-C(NR 8 )- 3 R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , R 9 OC(O)NR 8 - and C 1 -
  • R 5a and R 5 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C 20 alkyl, C 2 -C 20 alkenyl, C3-C10
  • R a and R are combined to form -(CH 2 ) S - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0) m , -NC(O)-, and -N(COR )-;
  • R 6 is a) substituted or unsubstituted Ci-C 8 alkyl, wherein the substituent on the alkyl is selected from:
  • R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) Ci -CO alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
  • R is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and C 1 -C
  • R is independently selected from hydrogen, Ci-C 6 alkyl and aryl
  • R 9 is independently selected from Ci-C 6 alkyl and aryl
  • R is independently selected from hydrogen and Ci-C 6 alkyl
  • R 11 is independently selected from Ci-C 6 alkyl
  • Z 1 and Z 2 are independently H 2 or O, provided that Z 1 is not O when X-Y is - C(O)N(R 7a ); m is 0, 1 or 2; q is 0, 1 or 2; s is 4 or 5; and t is 3, 4 or 5; (n) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 96109820, incorporated herein by reference,
  • R 1 is selected from: a) hydrogen, b) R 5 S(O) 2 -, R 5 C(O)-, (R 5 ) 2 NC(O)- or R 6 OC(O)-, and c) Ci-Ce alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R 5 O-, R 5 S(O) n ,-, R 5 C(O)NR 5 -, CN, (R 5 ) 2 N-C(NR 5 )- 5 R 5 C(O)-, R 5 OC(O)-, N 3 , -N(R 5 ) 2 , or R 6 OC(O)NR 5 -;
  • R a and R are independently selected from: a) hydrogen, b) Ci-C 6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R 5 O-, R
  • R is selected from: a) unsubstituted or substituted aryl, b) unsubstituted or substituted heterocycle, c) unsubstituted or substituted cycloalkyl, and d) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
  • X-Y is a)
  • R 4a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
  • R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, i) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci -Ce alkyl substituted with hydrogen or an unsubstituted or substituted
  • R 6 is independently selected from Ci-C 6 alkyl and aryl
  • X and Y are independently O or H 2 ; m is 1 or 2; n is 0 or 1 ; p is 1, 2 or 3; q is 0, 1 or 2; t is 1 to 4;
  • R, R 1 and R 2 are independently selected from H, Ci -6 alkyl, or Ci -6 aralkyl;
  • R and R are independently selected from: a) hydrogen, b) Ci-C 6 alkyl unsubstituted or substituted by C 2 -C 6 alkenyl, R 6 O-,
  • R 5 S(COq-, R 7 C(O)NR 6 -, CN, N 3 , R 6 OC(O)NR 6 -, R 6 R 7 N-C(NR 6 R 8 )-, R 6 C(O)-, R 7 R 8 NC(O)O-, R 7 R 8 NC(O)-, R 6 R 7 N-S(O) 2 -, -NR 6 S(O) 2 R 5 , R 6 OC(O)O-, -NR 6 R 7 , or R 7 R 8 NC(O)NR 6 -, c) unsubstituted or substituted cycloalkyl, alkenyl, R 6 O-, R 5 S(O)q-, R 6 C(O)NR 6 -, CN, NO 2 , R 6 R 7 N-C(NR 8 )-, R 6 C(O)-, N 3 , -NR 6 R 7 , halogen or
  • R 7 OC(O)NR 6 -, and d) Ci-C 6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C 3 -CiO cycloalkyl;
  • W is -CHR 9 - or -NR 9 -;
  • Z is unsubstituted or substituted Ci -8 alkyl, unsubstituted or substituted C 2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle; wherein the substituted group is substituted with one or more of:
  • Ci-4 alkyl unsubstituted or substituted with: a) Ci -4 alkoxy, b) NR 6 R 7 , c) C 3-6 cycloalkyl, d) aryl or heterocycle, e) HO, 2) aryl or heterocycle,
  • R 5 is C 1-4 alkyl or aralkyl
  • R , R and R are independently selected from H, C 1-4 alkyl, C 3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, e)
  • R 6 and R may be joined in. a ring
  • R 7 and R may be j oined in a ring
  • R 9 is selected from H, C 1-4 alkyl, C 3-6 cycloalkyl, heterocycle and aryl, unsubstituted, monosubstituted or disubstituted with substituents independently selected from: a) C 1-4 alkyl, b) C 1-4 alkoxy, c) aryl or heterocycle, d) halogen, ⁇ ) HO, f)
  • R 10 and R 11 are independently selected from hydrogen, Ci-C 6 alkyl, C 2 -C 4 alkenyl, benzyl and aryl; or the pharmaceutically acceptable salt thereof.
  • Compounds suitable for use in the methods of the invention also include those farnesyl transferaseinhibitors that do not incorporates a cysteinyl or sulfhydryl containing moiety at the N terminus of the molecule. Such compounds may exhibit preferred pharmacological activity, e.g. by avoiding thiol-related reactions in vivo. More specifically, the following compounds may be suitable.
  • R 1 is selected from: a) heterocycle, and b) Ci-Cio alkyl, which is substituted with heterocycle and which is optionally substituted with one or more of Ci-C 4 alkyl, hydroxy or amino groups;
  • R 2a and R 2b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C alkyl, C 2 -C 20 alkenyl, C 3 -CiO cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, NO 2 , R 8 O-, R 9 S(OV, R 8 C(O)NR 8 -, CN, (R 8 ) 2 NC(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , R 9 OC(O)NR 8 - and Ci-C 20 alkyl, and d) Ci-C 6 alkyl substituted with
  • R and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) ' an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C 1 -C 20 alkyl., C 2 -C 20 alkenyl, C 3 -C 10 cycloalkyl, axyl or heterocyclic group, wherein the substituent is selected from F, Cl 5 Br 5 N(R-V NO 2 , R 8 O-, R 9 S(O) 1n -, R 8 C(O)NR 8 -, CN, (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , R 9 OC(O)NR 8 - and C 1 -C 20
  • R and R are combined to form - (CH 2 )S -;
  • R 5a and R 5b are independently selected from: a) a side chain of a naturally occurring amino acid, 0 b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C 20 alkyl, C 2 -C 20 alkenyl, C 3 -Ci 0 5 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, N(R 8 ) 2 , NO 2 , R 8 O-, R 9 S(O) 1n -, R 8 C(O)NR 8 -, CN, (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R
  • R 7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
  • R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and Ci
  • R 8 is independently selected from hydrogen,, Ci-C 6 alkyl and aryl
  • R 9 is independently selected from Ci-C 6 alkyl and aryl
  • R is independently selected from hydrogen and Ci-Ce alkyl; R 11 is independently selected from Ci-C 6 alkyl;
  • Z is independently H 2 or O; m is 0, 1 or 2; n is O 3 1 or 2; and s is 4 or 5; (q) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 95/09000 and U.S. Patent No. 5,468,773, incorporated herein by reference,
  • V is CH 2 , O 5 S, HN 3 or R 7 N;
  • R 3 R 5 R and R are independently the side chains of naturally occurring amino acids, including their oxidized forms which maybe methionine sulfoxide or methionine sulfone., or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic substituents maybe substituted with an aromatic or heteroaromatic ring;
  • X-Y is
  • R 6 is a substituted or unsubstituted aliphatic, aromatic or heteroaromatic group such as saturated chains of 1 to 8 carbon atoms, which may be branched or unbranched, wherein the aliphatic substituent may be substituted with an aromatic or heteroaromatic ring;
  • R 7 is an alkyl group, wherein the alkyl group comprises straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, which may be substituted with an aromatic or heteroaromatic group;
  • Z is H 2 or O; m is 0, 1 or 2; n is 0, 1 or 2; and o is 0, 1, 2 or 3;
  • R is selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, (R 10 J 2 N-C(NR 1 °)-,
  • R lb is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, cycloalkyl, alkenyl, alkynyl, (R 10 ) 2 N- C(NR 10 )-, R 10 C(O)-, Or R 10 OC(O)-, and c) C 1 -C 6 alkyl
  • R and R are independently selected 1 a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C 2 O alkyl, C 2 -C 20 alkenyl, C 3 -Ci 0 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br 3
  • R a and R independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 3 -Ci 0 cycloalkyl, aryl or heterocyclic group s wherein the substiruent is selected from F, Cl, Br 3 NO 25 R 10 O-, R 11 S(OV, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(9>, N 3 , -N(R 1 V R 11 C(O)NR 10 - and C 1 -C 20 alkyl, and d) C 1 -C 6 alkyl
  • R 5a and R 5b are combined to form - (CH 2 )s - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0) m , -NC(O)-, and -N(COR 10 )-;
  • R 6 is a) substituted or unsubstituted C 1 -C 8 alkyl, wherein the substituent on the alkyl is selected from:
  • R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl; R is selected from: .
  • R 8 is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfmoroalkyl, F, Cl,
  • R is selected from: hydrogen, C 1 -C 6 alkyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , N 3 , - N(R 1 V and R 11 OC(O)NR 10 -; provided that R 9 is not R 10 C(O)NR 10 - when R la is alkenyl, V is hydrogen and X-Y iS -C(O)NR 7 -;
  • R 10 is independently selected from hydrogen, Ci-C 6 alkyl, benzyl and aryl; R is independently selected from Ci-C 6 alkyl and aryl; R 12 is independently selected from hydrogen and Ci-C 6 alkyl; R 13 is Ci-C 6 alkyl; V is selected from: a) aryl; b) heterocycle; or c) hydrogen;
  • W is -S(OV, -0-, -NHC(O)-, -C(O)NH-, -NHSO 2 -, -SO 2 NH-, N(R 7a )- or N[C(O)R 72 ]-;
  • R 1 is hydrogen, C 1 -C 6 alkyl or aryl;
  • R a and R are independently selected from: 5 a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, 0 c) substituted or unsubstituted C 1 -C 2 O alkyl, C 2 -C 2 O alkenyL C 3 -C 1 O cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, NO 2 , R 9 O-, R 10 S(O) 1n -, R 9 C(O)NR 9 -, CN, (R 9 ) 2 NC(NR 9 )-, R 9 C(O)-, R 9 OC(O)-, N 3 , -N
  • Ci-C 6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C 3 -CiO cycloalkyl; or
  • R 3 and R 4 are combined to form-(CH 2 ) s -;
  • O R 5a and R 5b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C 1 -C 2 O alkyl, C 2 -C 20 alkenyl, C 3 -C 10 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F 5 Cl, Br 5 NO 2 , R 9 O-, R 10 S(O) 1n -, R 9 C(O)NR 9 -, CN 5 (R 9 ) 2 N-C(NR 9 )- 5 R 9 C(O)-, R 9 OC(O)-, N 3 , -N(R 9 ) 2 , R 10
  • R 5a and R 5b are combined to form -(CH 2 ) S - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0) m , -NC(O)-, and -N(COR 9 )-;
  • R 6 is a) substituted or unsubstituted C 1 -C 8 alkyl, wherein the substituent on the alkyl is selected from:
  • R 7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
  • R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl., f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci-C 6 alkyl substituted with hydrogen or an unsubstir ⁇ ted or substituted group selected from aryl, heterocycle and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from
  • R 8a and R 8b are independently selected from hydrogen, F, Cl, Br, NO 2 , R 11 O-, R 10 S(O) 1n -, CN, R 9 C(O)NR 9 -, (R 9 ) 2 N-C(NR 9 )-, R 9 C(O)-, R 9 OC(O)-, N 3 , -N(R 9 ) 2 , R 10 OC(O)NR 9 -, C1-C20 alkyl, aryl, heterocycle or Ci-C 20 alkyl substituted with aryl or heterocycle; R is independently selected from hydrogen, Ci-C 6 alkyl and aryl;
  • R is independently selected from Ci-C 6 alkyl and aryl
  • R 11 is independently selected from hydrogen, Ci-C 6 alkyl and aryl, provided R is Ci-C 6 alkyl when n is O;
  • R 12 is independently hydrogen or Ci-C 6 alkyl
  • R 13 is Ci-C 6 alkyl
  • R 1 is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R 10 OR 11 S(O) 1n -, R 10 C(O)NR 10 -, CN 5 NO 2 , (R 1 ⁇ 2 NC(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 11 OC(O)NR 10 -, c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2
  • R 2a and R 2b are combined to form -(CH 2 )S-;
  • R and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 3 -C 10 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R 10 ) 2 , NO 2 , R 10 O-, R ⁇ S(O) m -, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)- , R 10 OC(O)-, N 3 , -N(R 1
  • R 5a and R 5b are combined to form -(CH 2 ) S - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0) m , -NC(O)-, and -N(COR 10 )-;
  • R 6 is a) substituted or unsubstituted Ci-Cs alkyl, wherein the substituent on the alkyl is selected from:
  • R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
  • R 7 is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloaliyl, e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, - 6S -
  • a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl
  • a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
  • R is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 35 -N(R 10 ) 2 , OrR 11 OC(O)NR 10 -, and c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NH-, CN, H 2 N-C(NH)-, R 10 C(O)
  • R 9 is selected from: a) hydrogen, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 NC(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 1 ] OC(O)NR 10 -, and c) C 1 -C 6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, (R 1 ⁇ 2 N-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3> - N(R 10 ) 2 , Or R 11 OC(O)
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl and aryl; R is independently selected from C 1 -C 6 alkyl and aryl;
  • R is independently selected from hydrogen and C 1 -C 6 alkyl
  • R 13 is independently selected from C 1 -C 6 alkyl
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl 5 d) Ci-C 2 O alkyl wherein from 0 to 4 non-terminal carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 2 O alkenyi; provided that V is not hydrogen if A 1 is S(O) m and V is not hydrogen if Ai is a bond, n is 0 and A 2 is S(O) m or a bond; W is a heterocycle; z is independently H
  • R la and R lb are independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 1 V or R 11 OC(O)NR 10 -, c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 10 O-, R 11 S(O) n ,-, R 10 C(O)NR 10 -, CN 3 (R 10
  • R 2a and R 2b are independently selected from: a) hydrogen, b) C 1 -C 6 alkyl unsubstituted or substituted by C 2 -C 6 alkenyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, N 3 , (R 10 ) 2 N-C(NR 10 )- 3 R 10 C(O)-, R 10 OC(O)-, - N(R 1 V Or R 1 C(O)NR 10 -, c) aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2; or R 11
  • R 3 and R 4 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C 2 O alkyl, C 2 -C 2 O alkenyl, C3-C10 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R 1 V NO 2 , R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, (R 10 ⁇ N-C(NR 1 °)-, R 10 C(O)- , R 10 OC(O)-, N 3 , -N(R 1 °) 2 , R 11 OC(O)NR 10 - and Ci-C 20 alkyl, and
  • R 3 and R 4 are combined to form -(CH 2 )S-;
  • R 5a and R 5 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 3 -Ci 0 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, CF 3 , N(R 10 ) 2 , NO 2 , R 10 O-, R 11 S(O) n ,-, R 10 C(O)NR 10 -, CN, (R 10 J 2 N-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 ,
  • R 6 is a) substituted or unsubstituted C 1 -C 8 aLkyl, substituted or unsubstituted C 5 -C 8 cycloalkyl, or substituted or unsubstituted cyclic amine, wherein the substituted alkyl, cycloalkyl or cyclic amine is substituted with 1 or 2 substituents independently selected from:
  • R 7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C 3 -C 10 cycloalkyl, and e) C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C 3 -C 10 cycloalkyl;
  • R 7b is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C 3 -Ci 0 cycloalkyl, f) a carbonyl
  • R 9 is selected from: a) hydrogen, b) C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-,
  • R 11 S(OV, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ⁇ N-C-(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 , - N(R 10 ) 2 , Or R 11 OC(O)NR 10 -, and c) Ci-C 6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , - N(R 1 V OrR 11 OC(O)NR 10 -;
  • R is independently selected from H, Ci-C 6 alkyl, benzyl, substituted aryl and Ci-C 6 alkyl substituted with substituted aryl;
  • R 11 is independently selected from Ci-C 6 alkyl and aryl;
  • R 12 is hydrogen or Ci-C 6 alkyl;
  • R 13 is Ci-C 6 alkyl;
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C 20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 20 alkenyl, provided that V is not hydrogen if Ai is S(0) m and V is not hydrogen if Ai is a bond, n is O and A 2 is S(0) m ; W is a heterocycle;
  • R la and R lb are independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ⁇ N-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 11 OC(O)NR 10 -, c) C 1 -Ce alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R 10 O-, R 11 S(OV, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(
  • R 2a and R 2b are independently selected from: a) hydrogen, b b)) C C 11 --CC 66 a allbkyl unsubstituted or substituted by alkenyl, R 10 O-, R ⁇ S(O) m -, R 10 C(O)NR 10 -, CN, N 3 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, -N(R 10 ) 2 , or
  • R 3 a and R 3 b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C1-C20 alkyl, C 2 -C 20 alkenyl, C 3 -C 1 O cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R 1 V NO 2 , R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )- 3 R 10 C(O)- , R 10 OC(O)-, N 3 , -N(R 10 ) 2 , R 11 OC(O)NR 10 - and C 1 -
  • R 4 and R 5 are independently selected from: a) hydrogen, and
  • R 6 is a) substituted or unsubstituted Ci-Cs alkyl or substituted or unsubstituted
  • R 7 is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , Or R 11 OC(O)NR 10 -, and c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfmoroalkyl, F, Cl, Br 3 R 10 O-, R 11 S(O) n ,-, R 10 C(O)NH-, CN, H 2 NC(NH)-, R 10 C(O)
  • R is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl 5 Br 5 R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C-(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 11 OC(O)NR 10 -, and c) Ci-C 6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br 3 R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN 3 (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , - N(R 10 ) 2 , or R 11 OC(O
  • R 10 is independently selected from hydrogen, Ci-C 6 alkyl, benzyl and aryl;
  • R 11 is independently selected from Ci-C 6 alkyl and aryl
  • R is independently selected from hydrogen and Ci-C 6 alkyl; R is independently selected from Ci-C 6 alkyl;
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C2 0 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 20 alkenyl, provided that V is not hydrogen if A is S(0) m and V is not hydrogen if A is a bond, n is O and A is S(0) m ; W is a heterocycle; Z is independently H2 or O; m is O, 1 or 2; n is 0, 1, 2, 3 or 4; - IS -
  • p is O 5 1,2, 3 or 4; q is 0, 1 or 2; r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; and u is 0 or 1;
  • R and R are combined to form -(CH 2 )S-; or R or R are combined with R to form a ring such that
  • R a , R , R 7a and R are independently selected from: a) hydrogen, b) Ci-Ce alkyl unsubstituted or substituted by alkenyl, R 10 O-, R 11 S(O) n ,-, 5 R 10 C(O)NR 10 -, CN, N 3 , (R 10 ) 2 N-CCNR 10 )-, R 10 C(O)-, R 10 OC(O)-, -N(R 10 ) 2 , or
  • R 11 OC(O)NR 10 -, c) aryl, heterocycle, cycloalkyl, alkenyl, R 10 O-,
  • R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ⁇ N-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 , - N(R 10 ) 2 , OrR 11 OC(O)NR 10 -, and 0 d) C 1 -C 6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C 3 -Ci 0 cycloalkyl;
  • R 5a and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid 5 which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C 2 O alkyl, C 2 -C 20 alkenyl
  • R 5a and R 5b are combined to form -(CH 2 ) S - wherein one of the carbon atoms is 5 optionally replaced by a moiety select from O, S(0) m , -NC(O)-, and -N(COR 10 )-;'
  • R is independently selected from hydrogen or Ci-C 6 alkyl
  • R is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, O R 10 O-, R ⁇ S(O) m -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 J 2 N-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-,
  • R is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F 5 Cl 5 Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ⁇ N-C-(NR 1 °)- 5 R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 11 OC(O)NR 10 -, and c) Ci-C 6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br,
  • R 10 is independently selected from hydrogen, Ci-C 6 alkyl, benzyl and aryl; R is independently selected from Ci-C 6 alkyl and aryl; R 1"2 is a) substituted or unsubstituted Ci-C 8 alkyl or substituted or unsubstituted C 3 -C 8 cycloalkyl, wherein the substituent on the alkyl or cycloalkyl is selected from:
  • R is independently selected from hydrogen and Ci-C 6 alkyl;
  • R 14 is independently selected from Ci-C 6 alkyl;
  • Q is a substituted or unsubstituted nitrogen-containing C 4 -C9 mono or bicyclic ring system, wherein the non-nitrogen containing ring may be an aromatic ring, a C 5 - C 7 saturated ring or a heterocycle;
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C 1 -C 20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N 9 and e) C2-C 20 alkenyl, provided that V is not hydrogen if A 1 is S (O) m and V is not hydrogen if A 1 is a bond, n is 0 and A 2 is S(O) m ;
  • W is a heterocycle
  • X, Y and Z are independently H 2 or O; m is 0, 1 or 2; n is 0, 1 5 2, 3 or 4; p is O, 1, 2, 3 or 4; q is 0, 1 or 2; . r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; t is 3, 4 or 5; and u is 0 or 1;
  • R la and R lb are independently selected from: a) hydrogen, b) aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, 5 N 3 -, -N(R 1 V or R 11 OC(O)NR 10 -, c) C 1 -C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, C 3 -Ci 0 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 10 O-, R 11 S(O) n ,-, R 10 C(O)NR 10 -, CN
  • R 2 and R 3 are independently selected from H; unsubstituted or substituted Ci -8 O alkyl, unsubstituted or substituted C 2-8 alkenyl, unsubstituted or substituted C 2 -S alkynyl, unsubstituted or substituted aryl, unsubstituted or
  • R and R are attached to the same C atom and are combined to form (CH 2 )u - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(O) m , -NC(O)-, and -N(COR 10 )-;
  • R is selected from H and CH 3 ; and any two of R , R and R are optionally attached to the same carbon atom;
  • R 6 , R 7 and R 7 are independently selected from H, Ci -4 alkyl, C 3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Ci -4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • R 6 and R 7 may be joined in a ring; R 7 and R 7a may be joined in a ring; R is independently selected from: a) hydrogen, b) aryl, heterocycle, C 3 -Ci 0 cycloalkyl, C2-C 6 alkenyl, C 2 -C 6 alkynyl, perfluoroalkyl, F, Cl 5 Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN 3 NO 2 , (R 10 ) 2 N- C(NR 10 K R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , or R 1 C(O)NR 10 -, and c) Ci-C 6 alkyl unsubstituted or substituted by aryl, heterocycle, C 3 -Ci 0 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkyn
  • R 9 is selected from: a ⁇ hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 11 S(O) 1n -,
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1-C20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 2 O alkenyl, provided that V is not hydrogen if Ai is S(O) m and V is not hydrogen if Ai is a bond, 11 is O and A 2 is S(O) m ;
  • W is a heterocycle
  • Y is aryl, heterocycle, unsubstituted or substituted with one or more of: 1) Ci -4 alkyl, unsubstituted or substituted with: a) Ci -4 allcoxy, b) NR 6 R 7 , c) C 3-6 cycloalkyl,, d) aryl or heterocycle., e) HO, f) -S(O) 1n R 6 , or g) -C(O)NR 6 R 7 ,
  • Z is aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more of the following:
  • R la is independently selected from: a) hydrogen, b) aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 2 O alkenyl, C 2 -C 20 alkynyl, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , (R 1 VN-C(NR 1 °)-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , Or R 11 OC(O)NR 10 -, c) Ci-C 6 alkyl unsubstituted or substituted by aryl, heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, R 10 O-, R ⁇ S(O) m -, R 10 C(O)NR 10 -, CN, (R
  • R is independently selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C 3 -C 10 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, R 10 O-, R 11 S(O) 1n -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 or -N(R 10 ) 2 , c) C 1 -C 6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 20 alkenyl, C 2 -
  • R 2 and R 3 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C 1 -C 20 alkyl, substituted or unsubstituted C 2 -C 20 alkenyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R 10 ) 2 , NO 2 , R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(
  • R 5 R 5 R a and R are independently selected from: a) hydrogen, b) Ci-C 6 alkyl unsubstituted or substituted by C 2 -C 2 Q alkenyl, R 10 O-, R 11 S(OV, R 10 C(O)NR 10 -, CN, N 3 , (R 10 ) 2 N- C(NR 10 )-, R 10 C(O)-, -N(R 10 ) 2s OrR 11 OC(O)NR 10 -, c) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C 3 -Ci 0 cycloalkyl, C 2 -C 20 alkenyl, R 10 O-, R 11 S(Q) 1n -, R 10 C(O)NR 10 -, CN 3 NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, N 3 , -N(R 10 ) 2 ,
  • R is selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C 3 -Ci 0 cycloalkyl, C2-C20 alkenyl, C 2 -C 20 alkynyl, Ci-C 20 perfluoroalkyl, aUyloxy, F, Cl, Br 5 R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, CN, NO 2 , R 10 IN-C(NR 1 °)-, R 10 C(O)-, N 3 , -N(R 1 V (R 12) 2 NC(O)- OrR 11 OC(O)NR 10 -, and c) C 1 -C 6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C 2 -C 20
  • R 7 is independently selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C 3 -Ci 0 cycloalkyl, and e) Ci-C 6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl;
  • R 8 is selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, Ci-C 2 O perfluoroalkyl, allyloxy, F, Cl, Br, R 10 O-, R 11 S(O) 1n -, R 10 C(O)NR 10 -, -S(O) 2 N(R 1 °) 2 , CN, NO 2 ,
  • R 10 2 N-C(NR 10 )-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , OrR 11 OC(O)NR 10 -, and c) C 1 -C 6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C 3 -CiO cycloalkyl, C 2 -C 20 alkenyl, C 2 - C 20 alkynyl, C 2 -C 20 perfluoroalkyl, F, Cl, Br, R 10 O-, R ⁇ S(O) m -, R 10 C(O)NH-, CN 5 H 2 N-C(NH)-, R 10 C(O)-, R 10 OC(O)-, N 3 , -N(R 10 ) 2 , OrR 10 OC(O)NH-; R 9 is selected from:
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl and aryl; R is independently selected from C 1 -C 6 alkyl and aryl; R 12 is independently selected from hydrogen, C 1 -C 6 alkyl and aryl, or (R ! 2) 2 forms -(CH 2 )S-;
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C 20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 20 alkenyl, provided that V is not hydrogen if Ai is S(0) m and V is not hydrogen if Ai is a bond, n is O and A 2 is S(0) m ;
  • W is a heterocycle
  • R la and R lb are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C 3 -C 6 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 8 O-, R 9 S(O) n ,-, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , OrR 9 OC(O)NR 8 -, c) C 1 -C 6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C 3 -C 6 cycloalky
  • R is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C 3 -C 6 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, Ci-C 6 perfluoioalkyl, F 5 Cl, is Br, R 8 O-, R 9 S(O) 1n -, R 8 C(O)NR 8 -, CN 3 NO 2 , (R 8 ) 2 NC(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , OrR 9 OC(O)NR 8 -, and c) Ci-C 6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C 3
  • R is independently selected from hydrogen, Ci-C 6 alkyl, substituted or unsubstituted Ci-C 6 aralkyl and substituted or unsubstituted aryl;
  • R 9 is independently selected from Ci-C 6 alkyl and aryl;
  • R is independently selected from hydrogen, C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 aralkyl and substituted or unsubstituted aryl;
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1-C2Q alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 2 O alkenyl, provided that V is not hydrogen if A 1 is S(0) m and V is not hydrogen if
  • Ai is a bond, n is O and A 2 is S(0) m ;
  • Compounds for use in the methods of the invention also may obtained by fermentation of cultures of novel organisms, such as the compounds disclosed in U.S. Patent No. 5,420,334.
  • Other suitable compounds are disclosed in U.S. Patent No. 5,420,245; European Patent Publication No. 0618 221; PCT Patent Publication Nos. WO 94/26723; WO 95/10514; WO 95/10515; WO 95/10516; WO 95/08542; WO 95/11917; and WO 95/12612.
  • monomycin is less preferred and may be excluded from preferred aspects of the invention.
  • Suitable compounds for use in the methods of the invention include the following:
  • N-(2(R)-amino-3-mercaptopropyl)-L-alaninebenzylamide N-benzyl-[2(S)-2(R)-Amino-3-mercaptopropyl)-amino]butyramide, N-(2(R)-amino--3-mercaptopropyl)-L-norleucinebenzylamide,
  • N-(2(R)-amino-3-mercaptopropyl)-L-norvalinebenzylamide N-(2(R)-ammo-3-mercaptopropyl)isoleucyl-phenylalanyl-homoserine, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-isoleucyl-homoserine, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-homoserine lactone, N-(2(R)-amino-3-merca ⁇ topropyl)isoleucyl-isoleucyl-homoserine lactone, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-homocysteine lactone,
  • R 1 and R 2 are each independently selected from: a) H; b) Ci-s alkyl; c) C 1-5 alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted, with methyl, methoxy, halogen (Cl, Br 3 F, ⁇ ) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R and R is hydrogen;
  • R 1 and R 2 are each independently selected from: a) H; b) Ci-5 is alkyl; c) C 1-5 is alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted with methyl, methoxy, halogen (Cl, Br, F, T) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R and R is hydrogen;
  • R 1 and R 2 are each independently selected from: a) H; b) Ci -5 alkyl; • c) Ci-5 alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted with methyl, methoxy, halogen (Cl, Br, F, T) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R 1 and R 2 is hydrogen;
  • n 0 to 4
  • R 1 and R 3 independently are Ci -4 alkyl, substituted with substituents selected from the group consisting of: a) aryl, which is defined as phenyl or naphthyl 5 unsubstituted or substituted with one, two, three or four substituents selected from the group consisting of: i) F 5 ii) Cl, i ⁇ ) Br, iv) nitro, v) cyano, vi) C 1-8 alkoxy, vii) Ci-8 alkylthio, vii ⁇ ) Ci -8 alkylsulfonyl, ix) sulfamoyl, or x) C 1-8 alkyl; or b) heteroaryl, which is defined as indolyl, imidazolyl or pyridyl, unsubstituted or substituted with one, two, three or four substituents selected from the group consisting of: i) F, ⁇ ) Cl, iii) Br, iv) nitro, v
  • X is CH 2 , CH(OH), C-O, CHCOR, CH(NH 2 ), CH(NHCOR), O, S(0) P , NH 5
  • p is O, l or 2;
  • Y is PO 3 RR 1 or CO 2 R;
  • R is H, lower alkyl, or CH 2 CH 2 N+Me 3 A-;
  • R 1 is H, lower alkyl, or CH 2 CH 2 N+Me 3 A-;
  • A is a pharmaceutically acceptable anion; m is O, 1, 2, or 3; and n is 0, 1, 2, or 3;
  • A is a C 2 -a saturated or unsaturated aliphatic hydrocarbon group which may have substituent(s) selected from the group consisting of a lower alkyl group, a hydroxyl group, a lower hydroxyalkyl group, a lower alkoxy group, a carboxyl group, a lower carboxyalkyl group, an aryl group and an aralkyl group; each of X and Y which are the same or different, is an oxygen atom, a sulfur atom, a carbonyl group or a group of the formula -CHR a - (wherein Ra is a hydrogen atom or a lower alkyl group) or -NR b (wherein Rb is a hydrogen atom or a lower alkyl group), or X and Y together represent a vinylene group or an ethynylene group; each of R , R 5 R 3 R and R which are
  • A is a C 2 -s saturated or unsaturated aliphatic hydrocarbon group which may have substituent(s) selected from the group consisting of a lower alkyl group, a hydroxyl group, a lower hydroxyalkyl group, a lower alkoxy group, a carboxyl group, a lower carboxyalkyl group, an aryl group and an aralkyl group;
  • Q is a group of the formula -(CH 2 )m- (wherein m is an integer of from 1 to 6) or-(CH2)n-W(CH 2 )p- (wherein W is an oxygen atom, a sulfur atom, a vinylene group or an ethynylene group; and each of n and p which are the same or different, is an integer of from 0 to 3);
  • R is a hydrogen atom, a halogen atom, a hydroxyl group, a lower al
  • each of R , R and R which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, a lower alkyl group or a lower alkoxy group
  • each of R 3 and R 4 which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a nitro group, a cyano group, a carboxyl group, a lower alkoxycarbonyl group, a carbamoyl group, a lower alkylcarbamoyl group, a lower alkyl group, a lower hydroxyalkyl group, a lower fluoroalkyl group or a lower alkoxy group;
  • R is a lower alkyl group; and R is a hydrogen atom or a lower alkyl group; or the pharmaceutically acceptable salts thereof.
  • Suitable compounds of the above type include the following: 3-Hydroxy-7,l l,15-trimethylhexadeca-6,10,14-trienoic acid,
  • N-((lRS,2RS,6E)-2-(4-chlorophenyl)-l-raethyl-7-phenyl-6-heptenyl)-N-(2- naphthylmethyl)carbamoylmethyl succinic acid N-((lRS,2RS,6E)-2-(4-chlorophenyl)-l-methyl-7-(2-naphthyl)-6-heptenyl)-N- (2-naphthylmethyl)carbamoylmethyl succinic acid
  • a further embodiment of the specific farnesyl pyrophosphate-competitive inhibitors includes: disodium (3RS.4RS)-4-[N-l(lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2- (3,4-methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- carboxyl-4-hyroxybutanoate
  • inhibitor compounds suitable for use in the methods of the invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. Unless otherwise specified, named amino acids are understood to have the natural "L" stereoconfiguration. Further, inhibitor compounds suitable for use in the methods of the invention may have enol form and keto form tautomers, depending upon the form of its substituents. The compounds of the present invention includes such enol form and keto form isomers and their mixtures.
  • such a hydroxyl group and a carboxyl group may form an intramolecular ester i.e. a 5-membered or 6-membered lactone ring.
  • Alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • Cycloalkyl is intended to include non-aromatic cyclic hydrocarbon groups having the specified number of carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • Alkenyl include those groups having the specified number of carbon atoms and having one or several double bonds.
  • alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, isoprenyl, farnesyl, geranyl, geranylgeranyl and the like.
  • aryl is intended to include any stable monocyclic, bicyclic or tricyclic carbon ring(s) of up to 7 members in each ring, wherein at least one ring is aromatic.
  • heterocycle or heterocyclic represents a stable 5 to 7 membered monocyclic or stable 8 to 11 membered bicyclic or stable 11-membered tricyclic heterocycle ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting ofN 5 O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothio- pyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, iso
  • substituted aryl As used herein, the terms “substituted aryl”, “substituted heterocycle” and “substituted cycloalkyl” are intended to include the cyclic group which is substituted with 1 or 2 substituents selected from the group which includes but is not limited to F, Cl, Br, CF 3 , NH 2 , N(C 1 -C 6 alkyl) 2 , NO 2 , CN, (Ci-C 6 alkyl)O-,-OH, (Ci-C 6 alkyl)S(O) m -, (Ci-C 6 alkyl)C(O)NH-, H 2 N-C(NH)-, (Ci-C 6 alkyl)C(O)-, (Ci-C 6 alkyl)OC(O)-, N 3 , (Ci-C 6 alkyl)OC(O)NR- and Ci-C 20 alkyl.
  • cyclic amine moiety having 5 or 6 members in the ring, such a cyclic amine which may be optionally fused to a phenyl or cyclohexyl ring.
  • a cyclic amine moiety include, but are not limited to, the following specific structures:
  • substitution on the cyclic amine moiety by R 2a , R 2b , R a and R may be on different carbon atoms or on the same carbon atom.
  • cyclic moieties are formed.
  • examples of such cyclic moieties include, but are not limited to:
  • cyclic moieties may optionally include a heteroatom(s).
  • heteroatom-containing cyclic moieties include, but are not limited to:
  • nitrogen containing C 4 -C9 mono or bicyclic ring system wherein the non-nitrogen containing ring may be a C 6 aromatic ring, a C5-C7 saturated ring or a heterocycle which defines moiety "Q" includes but is not limited to the following ring systems:
  • N(R 1 °) 2 represents-NHH, -NHCH 3 , -NHC 2 H 5 , etc. It is understood that substituents and substitution patterns on a particular inhibitor compounds suitable for use in the methods of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by known techniques.
  • the pharmaceutically acceptable salts of inhibitor compounds for use in the methods of the invention include known non-toxic salts, e.g. pharmaceutically acceptable inorganic or organic acids such as the following acids: hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenyl-acetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy- benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
  • pharmaceutically acceptable inorganic or organic acids such as the following acids: hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric
  • the pharmaceutically acceptable salts of inhibitor compounds for use in the methods of the invention can be synthesized from the corresponding inhibitor of this, invention which contain a basic moiety by conventional chemical methods.
  • the salts are prepared by reacting the free base with stoichiometric amounts or with an excess of the .desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
  • the aryl group means a phenyl group, a naphthyl group or an anthryl group. A phenyl group or a naphthyl group is preferred.
  • the heteroaromatic ring group means a 5-membered or 6-membered monocyclic aromatic heterocyclic group containing one or two heteroatoms, which are the same or different, selected from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, or a fused aromatic heterocyclic group having such a monocyclic aromatic heterocyclic group fused with the above-mentioned aryl group or having the same or different such monocyclic aromatic heterocyclic groups fused with each other, which may, for example, be a pyrrolyl group, an imidazolyl group, a pyrazolyl group, a pyridyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an oxazolyl group, an isoxazolyl group, a furyl group, a thienyl group, a thiazolyl group, an isothiazolyl group, an ind
  • a furyl group, a thienyl group, a pyridyl group, a pyrimidinyl group, an oxazolyl group, an isoxazolyl group, a thiazolyl group, a benzofuranyl group, a benzothienyl group, a benzimidazolyl group, a benzoxazolyl group, a benzothiazolyl group or a quinolyl group is preferred.
  • the lower alkyl group means a
  • C 1-6 linear or branched alkyl group which may, for example, be a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, a sec-butyl group, a tert-butyl group, a pentyl group or a hexyl group. Among them, a methyl group or an ethyl group is preferred.
  • the lower hydroxyalkyl group means the above-mentioned lower alkyl group having a hydroxyl group, i.e.
  • a Ci -6 hydroxyalkyl group such as a hydroxymethyl group, a hydroxyethyl group, a hydroxypropyl group or a hydroxybutyl group. Among them, a hydroxymethyl group or a hydroxyethyl group is preferred.
  • the lower alkoxy group means a C 1-6 alkoxy or alkylenedioxy group, which may, for example, be a methoxy group, an ethoxy group, a propoxy group, an isopropoxy group, a butoxy group, a tert-butoxy group, a methylenedioxy group, an ethylenedioxy group or a trimethylenedioxy group.
  • the lower carboxyalkyl group means the above-mentioned lower alkyl group having a carboxyl group, i.e. a C 1-7 carboxyalkyl group, such as a carboxymethyl group, a carboxyethyl group, a carboxypropyl group or a carboxybutyl group. Among them, a carboxymethyl group or a carboxyethyl group is preferred.
  • the aralkyl group means the above-mentioned lower alkyl group having the above-mentioned aryl group, such as a benzyl group, a phenethyl group, a 3-phenylpropyl group, a 1-naphthylmethyl group, a 2-naphthylmethyl group or a l-(2- naphthyl)ethyl group.
  • a benzyl group, a phenethyl group or a 2- naphthylmethyl group is preferred.
  • the saturated aliphatic hydrocarbon group may, for example, be an ethylene group, a trimethylene group, a tetramethylene group, a pentamethylene group, a hexamethylene group, a heptamethylene group or an octamethylene group.
  • a trimethylene group, a tetramethylene group or a pentamethylene group is preferred.
  • the unsaturated aliphatic hydrocarbon group means an unsaturated aliphatic hydrocarbon group having one or more, preferably one or two double bonds, at optional position(s) on the carbon chain, which may, for example, be a vinylene group, apropenylene group, a 1-butenylene group, a 2-butenylene group, a 1,3- butadienylene group, a 1-pentenylene group, a 2-pentenylene group, a 1,3- pentadienylene group, a 1,4-pentadienylene group, a 1-hexenylene group, a 2- hexenylene group, a 3-hexenylene group, a 1,3-hexadienylene group, a 1,4- hexadienylene group, a 1,5-hexadienylene group, a 1,3,5-hexatrienylene group, a 1- heptenylene group, a 2-heptenylene group,
  • halogen atom may be a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.
  • a fluorine atom or a chlorine atom is preferred.
  • the lower alkoxycarbonyl group means a C 1-7 alkoxycarbonyl group, such as a methoxycarbonyl group, an ethoxycarbonyl group, a propoxycarbonyl group, a butoxycarbonyl group or a tert-butoxycarbonyl group. Among them, a methoxycarbonyl group or an ethoxycarbonyl group is preferred.
  • the lower alkylcarbamoyl group means a carbamoyl group mono-substituted or di-substituted by the above-mentioned lower alkyl group, such as a methylcarbamoyl group, an ethylcarbamoyl group, a dimethylcarbamoyl group or a diethylcarbamoyl group.
  • the lower fluoroalkyl group means the above-mentioned lower alkyl group having fluorine atom(s), i.e.
  • a C 1-6 fluoroalkyl group such as a fluoromethyl group, a difluoromethyl group, a trifluoromethyl group, a 1-fluoroethyl group, a 2-fluoroethyl group, a 2,2,2-trifluoroethyl group or a pentafluoroethyl group.
  • the salt of the compound of a formula (ff) or (gg) may be a pharmaceutically acceptable common salt, which may, for example, be a base- addition salt of the terminal carboxyl group or of a carboxyl group when R 4 and/or R 5 or R 3 and/or R 4 is a carboxyl group, or when a carboxyl group or a lower carboxyalkyl group is present on a saturated or unsaturated aliphatic hydrocarbon group represented by A in the formulas (ff) and (gg), or an acid-addition salt of an amino group when R and/or R or R 3 and/or R 4 is an amino group, or of a basic heteroaromatic ring when such a basic heteroaromatic ring is present.
  • a pharmaceutically acceptable common salt which may, for example, be a base- addition salt of the terminal carboxyl group or of a carboxyl group when R 4 and/or R 5 or R 3 and/or R 4 is a carboxyl group, or when a carboxyl group
  • the base-addition salt may, for example, be an alkali metal salt such as a sodium salt or a potassium salt; an alkaline earth metal salt such as a calcium salt or a magnesium salt; an ammonium salt; or an organic amine salt such as a trimethylamine salt, a triethylamine salt, a dicyclohexylamine salt, an ethanolamine salt, a diethanolamine salt, a triethanolamine salt, a procaine salt or an N,N'- dibenzylethylenediamine salt.
  • an alkali metal salt such as a sodium salt or a potassium salt
  • an alkaline earth metal salt such as a calcium salt or a magnesium salt
  • an ammonium salt or an organic amine salt such as a trimethylamine salt, a triethylamine salt, a dicyclohexylamine salt, an ethanolamine salt, a diethanolamine salt, a triethanolamine salt, a procaine salt or an N,N'- dibenz
  • the acid-addition salt may, for example, be an inorganic acid salt such as a hydrochloride, a sulfate, a nitrate, a phosphate or a perchlorate; an organic acid salt such as a maleate, a fumarate, a tartrate, a citrate, an ascorbate or a trifluoroacetate; or a sulfonic acid salt such as a methanesulfonate, an isethionate, a benzenesulfonate or a p-toluenesulfonate.
  • an inorganic acid salt such as a hydrochloride, a sulfate, a nitrate, a phosphate or a perchlorate
  • an organic acid salt such as a maleate, a fumarate, a tartrate, a citrate, an ascorbate or a trifluoroacetate
  • a sulfonic acid salt
  • the ester of a compound of the formula (ff) or (gg) means a pharmaceutically acceptable common ester of the terminal carboxyl group or of a carboxyl group when R and/or R or R and/or R is a carboxyl group, or when a carboxyl group or a lower carboxyalkyl group is present on the saturated or unsaturated aliphatic hydrocarbon group represented by A in the formulas (ff) and (gg).
  • prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing the parent compounds described herein (see Goodman and Gilman's, The Pharmacological basis of Therapeutics, 8th ed., McGraw-Hill, Int. Ed. 1992, “Biotransformation of Drugs”).
  • inhibitor compounds useful in the methods of the invention can be synthesized from their constituent amino acids by conventional peptide synthesis techniques, and the additional methods described below. Standard methods of peptide synthesis are disclosed, for example, in the following works: Schroeder et al., The Peptides, Vol. 1, Academic Press 1965, or Bodanszky et al., Peptide Synthesis, Interscience Publishers, 1966, or McOmie (ed.) "Protective Groups in Organic Chemistry", Plenum Press, 1973, or Barany et al., "The Peptides: Analysis, Synthesis, Biology” 2, Chapter 1, Academic Press, 1980, or Stewart et al., “Solid Phase Peptide Synthesis", Second Edition, Pierce Chemical Company, 1984.
  • FTase inhibitor compounds are also commercially available. Some FTase inhibitor compounds may have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. AU such isomeric forms of these compounds are expressly included in the present invention.
  • the compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). AU such isomeric forms of such compounds are expressly included in the present invention. AU crystal forms of the compounds described herein are expressly included in the present invention.
  • the compounds of the present invention provide effective therapy of latently infected cells (i.e. cells infected by a virus which is an immunodeficiency virus such as FIV, SIV, HTV, etc.) as evidenced by the induction of lytic replication in latently infected cells.
  • latently infected cells i.e. cells infected by a virus which is an immunodeficiency virus such as FIV, SIV, HTV, etc.
  • the present invention can be used in treating those diagnosed as having AIDS as well as those having ARC, PGL and those seropositive but asymptomatic patients.
  • an effective amount of an FTase inhibitor compound can also be used prophylactically as a preventative for high risk individuals.
  • Compounds of the present invention can be used to treat cells, especially mammalian cells and in particular human cells, infected by an immunodeficiency virus such as HIV.
  • an immunodeficiency virus such as HIV.
  • P24 a major structural protein (product of gag), has been widely used for monitoring HIV-I replication in cells and vireamia in individuals.
  • the increase can be determined relative to untreated uninfected cells.
  • the compounds of the present invention can be administered to HTV infected individuals or to individuals at high risk for HIV infection, for example, those having sexual relations with an HIV infected partner, intravenous drug users, etc. Because of their effect of inducing lytic viral replication, the compounds of the present invention and pharmaceutical compositions comprising one or more compounds of formula I can be used prophylactically as a method of prevention for such individuals to minimize their risk of cells becoming latently infected.
  • the compound is adminstered in an effective amount as set forth below by methodology such as described herein.
  • Preferred compounds used in accordance with the present invention may induce lytic replication of HIV-I LTR and HIV-I in latently infected cells.
  • preferred compounds of the present invention in a dose dependent fashion may cause latently infected cells to lyrically replicate.
  • such induction is provided with essentially no adverse effects on cell survival or cellular mRNA or total cellular RNA synthesis.
  • compounds of the present invention will have utility in clearing latent infections of an HIV infection and other retroviral infections in cells and in a human, and (in preferred embodiments) to ultimately entirely clear virus from an infected subject.
  • the methods of the invention can further include administration of one or more antiviral agents to the subject, thus both depleting the latent cell reservoir and inducing viral lytic replication whereupon the cells in that state are subjected to and susceptible to the antiretroviral therapy, which controls viral proliferation.
  • one or more compounds of the invention is administered in an amount sufficient to activate viral lytic replication in at least about 25 percent of infected cells, more preferably an amount sufficient to induce lytic replication in at least about 50 percent of the infected cells and still more preferably induce lytic replication in at least about 75 percent of latently infected cells.
  • a preferred effective dose of one or more therapeutic compounds can be readily determined based on known factors such as efficacy of the particular therepautic agent used, age, weight and gender of the patient, and the like. See dosage guidelines as set forth e.g. in Remington, The Science and Practice of Pharmacy, 20 th Edition.
  • a FTase inhibitor compound may be administered to a mammal (e.g. human) in the range 0.1 mg to 5g per kilogram body weight of recipient per day, more preferably in the range of 0.1 mg to 1,000 mg per kilogram body weight per day, and still more preferably in the range of 1 to 600 mg per kilogram of body weight per day.
  • the desired dose is suitably administered once or several more sub-doses administered at appropriate intervals throughout the day, or other appropriate schedule.
  • a therapeutic compound used in accordance with the invention will be in an isolated form distinct as it may be naturally found and in a comparatively pure form, e.g., at least 85% by weight pure, more preferably at least 95% pure.
  • administered compound of formula I be at least 98% or even greater than 99% pure.
  • Such a material would be considered sterile for pharmaceutical purposes.
  • Potential contaminants include side products that may result upon synthesis of a compound of the invention or materials that may be otherwise associated with the compound prior to its isolation and purification.
  • the present compounds should preferably be sterile and pyrogen free. Purification techniques known in the art may be employed, for example chromatography.
  • the terms “subject” and “patient” are used interchangeably.
  • the terms “subject” and “subjects” refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, ape, monkey, or human), and more preferably a human, hi one embodiment, the subject is an immunocompromised or immunosuppressed mammal, preferably a human (e.g., an HIV infected patient).
  • a non-primate e.g., a cow, pig, horse, cat, dog, rat, and mouse
  • a primate e.g., a monkey, ape, monkey, or human
  • the subject is an immunocompromised or immunosuppressed mammal, preferably a human (e.g., an HIV infected patient).
  • the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat), hi a preferred embodiment, the subject is a human.
  • farm animal e.g., a horse, a cow, a pig, etc.
  • pet e.g., a dog or a cat
  • the subject is a human.
  • Administration of the compounds of the invention may be by any suitable route including oral, rectal, nasal, topical (including buccal and sublingual), vaginal andparenteral (including subcutaneous, intramuscular, intravenous and intradermal) with oral or parenteral being preferred. It will be appreciated that the preferred route may vary with, for example, the condition and age of the recipient.
  • the administered ingredients may be used in therapy in conjunction with other medicaments such as reverse transcriptase inhibitors such as dideoxynucleosides, e.g. zidovudine (AZT), 2',3'-dideoxymosine (ddT) and 2',3'-dideoxycytidine (ddC), lamivudine (3TC), stavudine (d4T), and TRIZIVIR (abacavir + zidovudine + lamivudine), nonnucleosides, e.g., efavirenz (DMP-266, DuPont Pharmaceuticals/Bristol Myers Squibb), nevirapine (Boehringer higleheim), and delaviridine (Pharmacia-Upjohn), TAT antagonists such as Ro 3-3335 and Ro 24- 7429, protease inhibitors, e.g., indinavir (Merck), ritonavir (Abbott), saquina
  • a FTase inhibitor compound may be administered in coordination or conunction with an entry inhibitor e.g. T20 (enfuvirtide, Roche/Trimeris) or UK-427,857 (Pfizer). Because many of these drugs are directed to different targets, e.g., viral integration, it is anticipated that an additive or synergistic result will be obtained by this combination. Further, FTase inhibitors can be combined with the use of other viral lytic replication activators, for example, proteasome inhibitors or Egrl activators, as disclosed in U.S. provisional patent application Nos. 60/588,013 and 60/588,301, respectively, both filed on July 13, 2004 and incorporated herein by reference.
  • one or more compounds of the formulae herein are used in conjunction with one or more therapeutic agents useful for treatment or prevention of HIV, a symptom associated with HTV infection, or other disease or disease symptom such as a secondary infection or unusual tumor such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus-related lymphomas among others, that can result in FUV immunocompromised subjects.
  • a symptom associated with HTV infection or other disease or disease symptom
  • a secondary infection or unusual tumor such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus-related lymphomas among others, that can result in FUV immunocompromised subjects.
  • one or more FTase inhibitor compounds are used in conjunction with a standard HIV antiviral treatment regimen.
  • This combination is advantageous in that the one or more FTase inhibitor compounds can activate latent HIV infected cells to replicate by stimulating lytic viral replication, thus making them susceptible to the co-administered standard HTV antiviral treatment regimens.
  • the latent or secondary reservoirs of HIV-infected cells are depleted through "controlled" activation (rather then serendipitous or uncontrolled activation), resulting in more complete elimination of virus, while controlling the spread of viral infection.
  • the treatment methods herein include administration of a so-called FJTV-drug "cocktail” or combination therapy, wherein a combination of reverse transcriptase inhibitor(s) and HTV protease inhibitor(s) is co-administered.
  • a highly active anti-retroviral therapy (HAART) treatment regime is combined with treatment with an FTase inhibitor according to the invention.
  • a combination therapy according to the invention includes administration of an FTase inhibitor together with an abl kinase inhibitor such as imatinib (use of imatinib for HIV treatment is described more fully in co- pending U.S. Provisional Patent Application No. 60/588,015, filed June 13, 2004).
  • the methods involve modulation of any gene that exhibits altered expression in chronically HlV-infected cells compared to uninfected parental cells, prior to induction into lytic replication.
  • the methods herein can involve, or target, any of the genes listed in the tables herein. This modulation can be direct or indirect, that is, it can be by direct control of expression or binding activity of the target, or by indirect control of the expression or binding activity of the target.
  • another aspect is modulation of viral replication activity of latent HIV- infected cells.
  • the methods involve modulation of lyn, cdc42, MNDA, CEBP alpha orMeisl by administration of the compounds of the formulae herein.
  • the present invention includes use of both racemic mixtures and optically active stereoisomers of FTase inhibitor compounds.
  • One or more FTase inhibitor compounds may be administered alone, or as part of a pharmaceutical composition, comprising at least one FTase inhibitor compound together with one or more acceptable carriers thereof and optionally other therapeutic ingredients, including those therapeutic agents discussed above.
  • the carrier(s) should be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions of the compounds of the invention used in combination with other compounds (e.g., reverse transcriptase inhibitors, protease inhibitors, and the like) maybe employed alone or in combination with acceptable carriers such as those described below.
  • a suitable effective dose of a compound in such a composition will be in the range of 1 to 5,000 mg per kilogram body weight of recipient per day, preferably in the range of 10 to ' 4,000 mg per kilogram body weight of recipient per day. When multiple compounds having complementary activity are administered together it is expected one can use the lower portion of these ranges (or even less).
  • stable refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds).
  • the compounds delineated herein are commercially available or readily synthesized by one of ordinary skill in the art using methodology known in the art.
  • compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the formulations may conveniently be presented in unit dosage form, e.g., tablets and sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy.
  • compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion, or packed in liposomes and as a bolus, etc.
  • a tablet maybe made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein.
  • compositions suitable for topical administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the ingredient to be administered in a suitable liquid carrier.
  • compositions suitable for topical administration to the skin may be presented as ointments, creams, gels and pastes comprising one or more compounds of the present invention and a pharmaceutically acceptable carrier.
  • a suitable topical delivery system is a transdermal patch containing the ingredient to be administered.
  • compositions suitable for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate.
  • suitable base comprising, for example, cocoa butter or a salicylate.
  • Compositions suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size, for example, in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • Suitable formulations wherein the carrier is a liquid, for administration, as for example, a nasal spray or as nasal drops include aqueous or oily solutions of the active ingredient.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • compositions suitable for parenteral administration include aqueous and non ⁇ aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the ' sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions maybe prepared from sterile powders, granules and tablets of the kind previously described.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.
  • ACH-2, A3.01, Jl.1, and Ul cells were obtained through the NTH AJDS Research and Reference Reagent Program, Division of AIDS, NIAJD, NJU.
  • U-937-cells were obtained from American Type Culture Collection (Manassas, VA).
  • ACH-2, Jl.1 and Ul are chronically infected cell lines harboring HIV-I LAV strain, while A3.01, Jurkat, and U-937 are the corresponding parental uninfected cell lines.
  • Cells were grown in RPMI- 1640 (Jnvitrogen, San Diego, CA) with 10% fetal bovine serum (FBS, hivitrogen), 5% penicillin- streptomycin (Lrvitrogen), and 2mM glutamine (Invitrogen). Cells were maintained at a concentration of IxIO 6 cells/ml in T- 175 flasks. Cell concentrations and cell viability were monitored throughout the experiment at all time points studied. Cells were induced by addition of 20 ng/niL of phorbol myristyl acetate (PMA or TPA, Sigma, St Louis, MO) for one hour, after which the cells were washed with phosphate buffered saline (PBS).
  • PMA or TPA phorbol myristyl acetate
  • HIV-infected and uninfected cells maintained and harvested in parallel with the PMA-treated cells but not induced with PMA were also harvested.
  • 3'-azido- 3'-deoxythymidine (AZT, Sigma) was not added to the ACH-2 or A3.01 cells, in order to keep conditions as close to an acute infection as allowed by the experimental model.
  • Flow Cytometry To. confirm viral replication following PMA induction, we . measured the accumulation of intracellular p24 over a period of 48 hours by measuring cell populations labeled with anti-p24 FITC-labeled antibody by flow cytometry.
  • Cells (ACH-2 and A3.01) were washed twice with ice-cold PBS and suspended in 50 ⁇ L ice-cold permeabilization buffer (BD Biosciences, San Jose, CA), and incubated at 4 0 C in the dark for 30 minutes. The cells were fixed using the CytoFix/CytoPerm kit (BD Biosciences) and 5 ⁇ L KC57-FITC-labeled p24 antibody (Beckman Coulter), was added to detect intracellular p24.
  • RNA from the samples was subjected to DNase treatment to remove contaminating DNA, and the DNAse was inactivated using the DNase Free kit (Amersham Biosciences, Piscataway, NJ) according to manufacturer's protocols.
  • RNA reverse transcribed using the Taqman RT kit from ABI per manufacturer's specifications. Briefly, the reaction mixture (50 ⁇ l) was incubated at 65 0 C for 5 minutes followed by 37 0 C for 45 minutes, 94 0 C for 5 minutes, and then cooled on ice. 1/40 th aliquots of the corresponding samples were then used in a real-time PCR reaction using Taqman probes labeled with FAM and TAMRA at the 5' and 3 ' ends respectively. Primer probe pairs were designed using PrimerExpress (ABI). The reactions were carried out in triplicate for each time point and the fold changes observed were normalized to GAPDH, for each time point.
  • ABI PrimerExpress
  • the sequence for the labeled probe was 5'CTTCTCTATCAAAGCAGACCCACCTCCS' which overlapped with the splice site of HTV-I Rev sequence.
  • the sequence detection primers for unspliced. or late RNA were SK38 and SK39 from the HIV-I Gene Amplimer kit (ABI).
  • a TAMRA labeled probe with sequence identical to SKl 9 (ABT) was used for Real time PCR quantitation of the late viral RNA species. Standards from the kit were diluted to calculate copy number of virus based on gag mRNA concentrations. Real time RT-PCR analysis was also carried out for selected cellular genes using gene specific primer probe pairs and Taqman detection primers.
  • RNA obtained from induced chronically infected and corresponding uninfected parental cells were used for microarray experiments. For each time point, RNA from the induced chronically infected ACH-2 cells and RNA from the corresponding induced, uninfected A3.01 cells were compared to minimize effects due to PMA induction. Microarrays were obtained from the National Cancer Institute Microarray Facility, Advanced Technology Center (Gaithersburg, MD). The microarrays (Hs. Um ' Gem2) contained 10,395 cDNA spots on each glass slide.
  • cDNAs were selected for spotting on the slides based on their known or probable involvement in oncogenesis, signal transduction, apoptosis, immune function, inflammatory pathways, cellular transport, transcription, protein translation and other important cellular functions.
  • a number of expressed sequence tags (ESTs) from unknown genes homologous to known genes and cDNAs encoding housekeeping genes were also included in these gene sets.
  • ESTs expressed sequence tags
  • 50 ⁇ g of total RNA from PMA induced ACH-2 cells and 70 ⁇ g of total RNA from PMA induced A3.01 cells was labeled with Cy-3-dUTP and Cy-5-dUTP respectively as previously described (34, 60). Higher amounts of RNA were used for Cy-5 labeling to minimize the disparities in dye incorporation.
  • RNA from PMA- induced, infected cells from a particular time point was compared with RNA from the corresponding PMA-induced, uninfected cells from the same time point for subsequent hybridization to the same array to ensure accurate comparisons and to eliminate inter-array variability.
  • the labeled cDNAs were then combined and purified using MicroCon YM-30 (Millipore, Bedford, MA) spin column filters, to remove any unincorporated nucleotides. 8-10 ⁇ g each of Cot-1 DNA, (Boehringer Mannheim, Indianapolis, IN), yeast tRNA (Sigma) and polyA (Amersham Biosciences) were added to the reaction mixture and heated at 100 0 C for 1 minute.
  • Hybridization of the labeled cDNA to the microarray was carried out at 65 0 C overnight, followed by washes with IX SSC, 0.2X SSC and 0.05X SSC respectively. The slides were dried by centrifugation at 1000 rpm for 3 minutes and then scanned as described below. RNA samples from three identical but independently conducted time course experiments were tested. Microarray experiments were performed at least twice for each time point (technical replicates) of each experiment. We also compared AZT- treated ACH-2 cells to untreated ACH-2 cells to determine whether any differences in gene expression might be solely due to AZT.
  • RNA from the same samples labeled with Cy5 (70 ⁇ g RNA) and Cy3 (50 ⁇ g RNA) were co-hybridized to the same array, scanned, and data were analyzed for all the cell lines studied, using identical filtering and statistical tests, and genes showing dye incorporation bias were eliminated from further analysis as described below.
  • Microarray Scanning and Data Analysis The slides were scanned using an Axon GenePix 4000 scanner (Axon Instruments, Union City, CA). The photomultiplier tube values (PMT) were adjusted to obtain equivalent intensities at both wavelengths used, 635 nm and 532 nm for the Cy5 and Cy3 channels respectively. Image analysis was performed using GenePix analysis software (Axon Instruments) and data analysis was performed using the microArray Database (mAdb) system hosted by the Center for Information Technology and Center for Cancer Research at NTH (http://nciarray.nci.nih.gov). Each array was normalized using Lowess normalization (71).
  • Filtering criteria were as follows: a) For each spot, signal intensity must be at least twice that of the background intensity; b) Each gene must have values in at least 70% of the arrays; c). Each array must have values for at least 70% of the gene spots. Genes that showed dye labeling bias in a particular cell line after normalization were excluded from that gene set prior to further analysis. This was determined using a one sample t-test on mean log ratios for replicate arrays with the same sample labeled with both Cy3 and Cy5.
  • the random variance model enabled variance information to be shared across genes without assuming that all genes have the same variance (69).
  • significance was based on p ⁇ 0.001 for a parametric one-sample random variance t-test.
  • a multivariate permutation test based on the one-sample random variance t-statistic was used in which the proportion of false discoveries was limited to 0.10 with 90% confidence (36, 62).
  • Hierarchical clustering analyses on the resulting data sets were done using the mAdb system as well as Cluster and TreeView software programs (Stanford University, CA).
  • pathway analysis of the various genes that showed significant differential expression was performed by utilizing analysis tools provided by the NIH mAdb database (http://nciarray.nci.nih.gov) and querying the database of the Cancer Genome Anatomy Project (CGAP), (http://cgap.nci.nih.gov/) with pathway information provided by KEGG (www.genome.ad.jp/kegg/) and Biocarta (www.biocarta.com) pathway databases.
  • CGAP Cancer Genome Anatomy Project
  • Latency Reactivation Studies Cells (ACH-2, Jl.1 and Ul) were seeded at a concentration of 2xlO 5 cells/mL in 24 well plates in a volume of 1 mL. L-744,832 (structure shown above; FTase inhibitor) was dissolved in sterile dimethylsulfoxide (DMSO) and further diluted with media to obtain the desired final concentrations. The final concentration of DMSO in contact with the cells was never greater than 0.001% at any dose tested.
  • DMSO sterile dimethylsulfoxide
  • AZT (250 nM) was added, to the chronically infected cells in order to inhibit p24 production that maybe caused due to low levels of actively replicating virus present along with the chronically infected cells and to ensure that any increases in p24 expression would be attributable to activation of latent provirus and not due to . subsequent amplification via additional rounds of viral replication.
  • Cells were incubated with different concentrations of L-744,832 at 37 0 C. 200 ⁇ L samples of cell supernatant were collected at 24 hours after treatment.
  • TNF-alpha tumor necrosis factor alpha
  • the plates were washed, and rabbit anti-HIV p24 antibody was added at 1 :400 dilution. Following incubation for one hour, the plates were washed and goat anti-rabbit IgG peroxidase labeled antibody at 1 :300 dilution was added. The plates were incubated for one hour at 37 0 C, followed by washing and addition of a two-component substrate.
  • Substrate solution consisted of equal volumes of TMB peroxidase substrate and peroxidase solution B (Kirkegaard and Perry Laboratories, Gaithersburg, MD). Samples were incubated for 30 minutes at room temperature and reactions were stopped by addition of IN hydrochloric acid solution. The absorbance was measured at 450 nm using a SpectraMax250 spectrophotometer (Molecular Devices Corporation, Sunnyvale, CA). The samples were assayed in duplicate and experiments were performed at least thrice using independent cell samples.
  • the late time period (48-96 hours pi) showed the least change with 566 genes exhibiting significant altered expression (p ⁇ 0.001).
  • Many of the genes that were differentially expressed in the early time period also showed either similar or the opposite trend in their expression patterns during the other time periods, hence some genes were included in the analysis of both the time periods.
  • a number of discrete patterns of gene regulation were observed.
  • Several cellular genes showed distinct temporal expression patterns during the lytic replication cycle, an expected finding, but more interestingly, a smaller number of genes appeared to be differentially expressed in the latently infected ACH-2 cells compared to their parental, uninfected cells, even before induction of the lytic cycle.
  • genes encoding transcription factors, components ofproteasom.es, factors that control immune function, apoptosis and other functional classes were included.
  • gene classes that were annotated in the gene ontology database GO database, www.geneontology.org
  • observed/expected ratio for the number of genes within a functional class that were differentially expressed was set at greater than one (O/E > 1), so as to eliminate functional classes where the number of genes differentially expressed was not greater than that randomly expected.
  • O/E > 1 the gene ontology database
  • Table 1 An abbreviated listing of the genes grouped according to known functions that were differentially expressed before induction is given in Table 1.
  • PSMB4 has peptidase activity, which is inhibited by Tat during viral replication.
  • Tat competes with the 1 IS regulatory subunit, for binding to the 2OS core complex due to presence of a common binding site in Tat and the HS regulator alpha subunit (32, 59).
  • Proteasomes are also involved in processing certain regions of HIV-I Nef preferentially, which leads to production of Nef-speciflc CTLs (cytotoxic T-lymphocytes) (44).
  • Many other classes of genes encoding immune response modulators, integrins, cell cycle modulators (such as Egrl), nuclear import factors, and G-protein signaling molecules were also differentially expressed.
  • a listing of genes that were differentially expressed prior to induction, based on their functional classification is given (Table 1).
  • a list of pathways that were affected in the uninduced, chronically infected cells is given (supplemental data, Table S2) .
  • HTV cell lines including J 1.1, a chronically infected T-lymphocytic cell line derived from Jurkat cells and Ul, a promonocyte chronically infected cell line derived from U937 cells, were studied using microarrays to determine the similarities and differences in their expression profiles.
  • the p24 expression in all the latently infected cell lines was below lng/mL (0.2-0.8 ng/mL) indicating that the cells were not lytically active at the time of harvesting the cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Molecular Biology (AREA)
  • Emergency Medicine (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Treatment of cells or humans carrying or infected with a virus capable of causing an immunodeficiency disease by administration of one or more compounds that inhibit farnesyl transferase.

Description

TREATMENT OF VIRAL INFECTIONS
CROSS-1RFPFKKNCF, TO RF.T.ATKD APPLICATIONS
This application claims the benefit of U.S Provisional Patent Application No. 60/587,771, filed July 13S 2004, the entire teachings of which are hereby incorporated by reference.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
Research supporting this application was carried out by the United States of
America as represented by the Secretary, Department of Health and Human Services.
BACKGROUND OF THE INVENTION 1. Field of the invention.
The present invention relates to methods for treatment or prevention of an HTV infection and, more particularly, to use of one or more farnesyl transferase inhibitor compounds to treat a subject suffering from or susceptible to an HIV infection.
2. Background.
The human immunodeficiency virus type 1 (HIV-I, also referred to as HTLV- m LAV or HTLV-IH/LAV) and, to a lesser extent, human immunodeficiency virus type 2 (HIV-2) is the etiological agent of the acquired immune deficiency syndrome (AIDS) and related disorders. Barre-Sinoussi, et al., Science, 2211:868-871 (1983); GaUo, et al., Science, 224:500-503 (1984); Levy, et al., Science, 225:840-842 (1984); Popovic, et al., Spienr.e, 224:497-500 (1984); Sarngadharan, et al., Science, 224:506- 508 (1984); Siegal, et al., N. F,ng1. .T. Med., 3ϋ5_: 1439-1444 (1981); Clavel, F., ATDS, 1:135-140. This disease is characterized by a long asymptomatic period followed by the progressive degeneration of the immune system and the central nervous system. Studies of the virus indicate that replication is highly regulated, and both latent and lytic infection of the CD4 positive helper subset of T-lymphocytes occur in tissue culture. Zagury, et al., Science, 231:850-853 (1986). The expression of the virus in infected patients also appears to be regulated as the titer of infectious virus remains low throughout the course of the disease. Both HTV-I and 2 share a similar structural and function genomic organization, having regulatory genes such as tat, reϋ, nef, in addition to structural genes such as enx, gag and pal.
While AIDS, itself, does not necessarily cause death, in many individuals the immune system is so severely depressed that various other diseases (secondary infections or unusual tumors) such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus related lymphomas among others occur, which ultimately results in death. These secondary infections may be treated using other medications. However, such treatment can be adversely affected by the weakened immune system. Some humans infected with the AIDS virus seem to live many years with little or no symptoms, but appear to have persistent infections. Another group of humans suffers mild immune system depression with various symptoms such as weight loss, malaise, fever and swollen lymph nodes. These syndromes have been called persistent generalized lymphadenopathy syndrome (PGL) and AIDS related complex (ARC) and may or may not develop into AIDS. In all cases, those infected with the HTV are believed to be persistently infective to others.
The activation of the latent HTV provirus from the asymptomatic period has been reported to be governed by a long terminal repeat (LTR) in the viral DNA. See, e.g. Ranki, A., et al., T.anr,ftt ii- 589-593 (1987); Fauci, A.S., et al., .Sdenne, 7.39:617- 622 (1988); Zagury, D., et al., Sr,iκnπft, 211:850-853 (1985); Mosca, J.D., Nature (London), 325:67-70 (1987). The activity of HTV-I is determined by the complex interaction of positive and negative transcriptional regulators that bind to specific sequences within the LTR. Cullen, B.R., et al., Cell, 5&:423-426 (1989). Changes in the quantity or quality of these factors may underlie the activation of transcription of HTV-I and HTV-2 latent provirus by a myriad of stimuli. See, e.g. Fauci, A.S.,
SriftTiπft, 23-9_:617-622 (1988); Griffin, G.E., et al., Nature (London), 339-70-73 (1989); Nabel, G., et al., Sdenr^ 233:1299-1302 (1988). Specifically, phorbol 12- myristate-13-acetate (PMA) and Tumor Necrosis Factor-α (TNPa) are believed to be potent activators, in particular, TNFcc is present in markedly enhanced levels in HTV infected individuals, suggesting that the cytokine plays an important role in the pathogenesis of AIDS. Lahdevirta, T Am T MRH J £5:289-291 (1988).
Most known methods for treating individuals infected by HTV have focused on preventing integration of the provirus into the host cell's chromosome. One overlooked area of interest has been drugs that affect latently infected cells so that virus may be cleared from the infected individual completely. Many of the proposed therapeutic methods, however, have not proven clinically effective. While current antiretroviral agents can control HIV replication, they have not eliminated latently infected cells. Since viral reactivation is necessary for targeting by antiviral drugs (Brooks, et al. 2003. Molecular characterization, reactivation, and depletion of latent HTV. Immunity 19:413-23), identification of new approaches that eject HTV from latency when used together with effective antiviral therapy can lead to the reduction of latent HIV reservoirs in an infected patient.
It thus would be desirable to have a new compound that can treat cells latently infected with HIV. It would be particularly desirable to have a new therapy that can be used to treat cells already infected, by means other than by preventing integration of the virus.
SUMMARY OF THE INVENTION
We have now discovered that certain compounds that inhibit farnesylation, e.g., of protein (farnesyl transferase (FTase) inhibitor compounds), can be useful for treating cells infected by immunodeficiency viruses and methods of preventing cells from becoming infected by immunodeficiency viruses, preferably human immunodeficiency viruses such as HTV.
More particularly, we now provide therapeutic methods that in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor compound) to mammalian cells that are infected with an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
The invention further methods that in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor) to a patient in need of treatment, such as a mammal suffering from or susceptible to an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
A wide variety of FTase inhibitor compounds can be employed in the methods of the invention. For example, suitable compounds have been reported previously including those in U.S. Patents 5,238,922; 5,571,792; and 5,571,835; WO 94/10138; WO 94/04561; WO 94/10138; WO 96/21456; and WO 97/02817. Particularly preferred FTase inhibitor compounds for use in the methods of the invention exhibit good activity in a standard in vitro FTase inhibition assay (specifically the standard assay defined below), preferably an ICso (concentration required to inhibit FTase activity by 50% relative to control) in such an assay of about 100 nM or less, more preferably an IC50 about 50 nM or less.
Specifically preferred FTase inhibitor compounds for use in accordance with the invention include the peptidomimetic compounds, including FTI276, FTI277 and L-744832. Non-peptidomimetic FTase inhibitor compounds also are preferred, including BMS214662, Rl 15777 and SCH66336.
FTase inhibitor compounds used in accordance with the present invention can induce lytic replication in cells latently infected with an immunodeficiency virus such as HTV by targeting a product or products of the genes encoding proteins in the Ras and Rho signaling pathways which are differentially expressed in latently infected cells and lytic replicating cells.
In certain embodiments, the compounds of the present invention can treat cells infected acutely and chronically by immunodeficiency viruses, for example, HIV, preferably HIV-I, and thus can be used to treat humans infected by HTV. For example, treatment of those diagnosed as having AIDS as well as those having ARC, PGL and those not yet exhibiting such conditions.
Another aspect is a method of reducing latent HTV-reservoirs in a subject including administration of an effective amount of one or more FTase inhibitor compounds. The latent HTV-reservoirs are collections of latent HTV-infected cells, that is cells in which the HIV-reρlication is considered to be in a latent state.
Other aspects or embodiments are a method of reducing latent HTV reservoirs in an HIV-infected subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds; a method of increasing (e.g., 10- 15 fold relative to cells treated with AZT) expression of p24 in a latently HIV-infected cell comprising administration to the cell of an effective amount of one or more FTase inhibitor compounds; a method of activating latent HlV-provirus in a cell in a subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds; and a method of activating latent HlV-provirus in a cell in a subject comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds The methods delineated herein include administering to a subject (e.g., a human or an animal) in need thereof an effective amount of one or more FTase inhibitors, e.g., compounds as delineated herein. The methods can also include the step of identifying that the subject is in need of treatment of diseases or disorders described herein, e.g., identifying that the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells. The identification can be in the judgment of a subject or a health professional and can be subjective (e.g., opinion) or objective (e.g., measurable by a test or a diagnostic method). Tests for HTV infection are known in the art and include polymerase chain reaction-based (PCR-based) amplification and detection of viral RNA; Western blot detection of anti- HIV antibodies; agglutination assays for anti-HTV antibodies; ELISA-based detection of HlV-specific antigens (e.g., p24); line immunoassay (LIA); and other methods known to one of ordinary skill in the art. In each of these methods, a sample of biological material, such as blood, plasma, semen, or saliva, is obtained fom the subject to be tested. Thus, the methods of the invention can include the step of obtaining a sample of biological material (such as a bodily fluid) from a subject; testing the sample to determine the presence or absence of detectable HIV infection, HIV particles, or HIV nucleic acids; and determining whether the subject is in need of treatment according to the invention, i.e.* identifying whether the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells.
The methods delineated herein can further include the step of assessing or identifying the effectiveness of the treatment or prevention regimen in the subject by assessing the presence, absence, increase, or decrease of a marker, including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HIV infection marker; preferably this assessment is made relative to a measurement made prior to beginning the therapy. Such assessment methodologies are known in the art and can be performed by commercial diagnostic or medical organizations, laboratories, clinics, hospitals and the like. As described above, the methods can further include the step of taking a sample from the subject and analyzing that sample. The sample can be a sampling of cells, genetic material, tissue, or fluid (e.g., blood, plasma, sputum, etc.) sample. The methods can further include the step of reporting the results of such analyzing to the subject or other health care professional. The method can further include additional steps wherein (such that) the subject is treated for the indicated disease or disease symptom.
In one aspect, the invention provides a method of treating HTV infection in a subject. The method comprises the steps of identifying a subject as in need of reactivation of replication processes in latent HTV-infected cells; and administrating of an effective amount of a farnesyl transferase (FTase) inhibitor to the subject to reactivate the viral replication process. In preferred embodiments, the farnesyl transferase inhibitor is one or more of FTI277, L-744832, BMS214662, Rl 15777 and SCH66336. In certain preferred embodiments, one or more peptidomimetic FTase inhibitor compounds are administered to the subject; in other preferred embodiments, one or more non-peptidomimetic FTase inhibitor compounds are administered to the subject. In certain prefered embodiments, the one or more FTase inhibitor compounds are of any one of the general formulae (a) through (gg) described herein. In certain embodiments, the administered FTase inhibitor compound has an IC50 of about 100 nM or less in a standard in vitro farnesyl transferase inhibition assay.
In another aspect, the invention provides a method of inhibiting HIV replication in a subject or a cell. The method comprises the steps of identifying a subject or cell as in need of reactivation of replication processes in latent HTV-infected cells; administering an effective amount of a farnesyl transferase (FTase) inhibitor to the subject or cell to reactivate the viral replication process; and administering one or more HTV antiviral agents to the subject or cell to inhibit induced lytic HIV viral replication.
In another aspect, the invention provides a method of treating latently HIV- infected cells in a subject comprising administration to the cells one or more FTase inhibitor compounds.
In still another aspect, the invention provides a method of modulating lytic replication in an HIV-infected cell in a subject identified as in need of such treatment. The method comprises the step of administration to the subject of an effective amount of one or more FTase inhibitor compounds.
In certain preferred embodiments, the cell is a human cell capable of sustaining a latent provirus. In certain preferred embodiments, the cell is a lymphocytic cell. In certain preferred embodiments, the cell is a monocytic cell. In certain preferred embodiments, the cells are human cells.
In another aspect, the invention provides a method of reducing latent HTV reservoirs in an HIV-infected subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
Another aspect is a method of reducing latent HTV-reservoirs in a subject by controlled activation of viral replication including administration of an effective amount of a farnesyl transferase inhibitor. Controlled activation is that activation initiated by administration of an effective amount of an FTase inhibitor such that the FTase inhibitor reactivates (directly or indirectly) replication processes in latent HIV- infected cells. The latent HIV-reservoirs are collections of latent BDV-infected cells, that is cells in which the HlV-replication is considered to be in a latent state. The method can further include administration with one or more antiviral agents, thus both depleting the latent cell reservoir and inhibiting induced viral lytic replication, whereupon the cells in that state are subjected to and susceptible to the antiretroviral therapy, which controls viral proliferation.
In yet another aspect, the invention provides a method of activating latent HTV-provirus in a cell in a subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
Ih any of the above methods, in preferred embodiments, the method further comprises the step of administration of one or more additional anti-viral (e.g., anti- HIV) therapeutic agents to the subject or cell. In preferred embodiments, the additional anti-viral agent(s) are a reverse transcriptase inhibitor, a protease inhibitor, or combination thereof.
In another aspect, the invention provides a method of increasing expression of p24 in a latently HIV-infected cell comprising administration to the cell of an effective amount of a farnesyl transferase inhibitor.
hi preferred embodiments of any of the methods described above, the cell is a lymphocytic cell or a monocytic cell. In preferred embodiments, the cell is a human cell, i.e., any human cell capable of sustaining a latent provirus.
In still another aspect, the invention provides a method of screening for a compound capable of activating latent HTV-infected cells comprising contacting a FTase inhibitor test compound with an ACH-2 cell or Jl .1 cell or Ul cell and determining the level of p24 expression, hi preferred embodiments, increased expression of p24 in cells treated with a test compound relative to non-treated cells indicates a compound capable of activating latent HTV-infected cells.
The invention also provides pharmaceutical compositions comprising one or more FTase inhibitor compounds and a suitable carrier therefore for use in the conditions referred to above.
The methods delineated herein can further include the step of assessing or identifying the effectiveness of the treatment or prevention regimen in the subject by assessing the presence, absence, increase, or decrease of a marker, including a marker or diagnostic measure of HTV infection, HIV replication, viral load, or expression of an HTV infection marker. Such assessment methodologies are known in the art and can be performed by commercial diagnostic or medical organizations, laboratories, clinics, hospitals and the like. The methods can further include the step of taking a sample from the subject and analyzing that sample. The sample can be a sampling of cells, genetic material, tissue, or fluid (e.g., blood, plasma, sputum, etc.) sample. The methods can further include the step of reporting the results of such analyzing to the subject or other health care professional.
Other aspects of the invention are disclosed infra.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the flow cytometric analysis of chronically infected ACH-2 cells before and after induction. Uninduced cells and cells from serial time points were fixed and permeabilized for intracellular p24 labeling. As an isotype control, cell samples were labeled with mouse IgGl . For each sample, 100,000 events were collected. In the figure, each sample histogram labeled for p24 (darker color) is overlaid with the control histogram labeled for the isotype control (lighter color). (A) Uninduced ACH-2 cells, showing minimal p24 accumulation with 8.2% of cells infected, (B) ACH-2 cells at 0.5 hours post induction (p.i.) with 7.4% of cells positive for p24, (C) ACH-2 cells at 6 hours p.i, with 61.6% cells infected, (D), (E), and (F) ACH-2 cells at 12, 18, and 24 hours p.i., respectively, showing complete infection. Flow cytometric analysis was performed on all batches of cells to ensure active replication of HTV following induction with PMA. Data from one induction experiment is shown. Data indicate that viral replication occurs in an ordered manner post induction, and complete infection of cells is achieved within 12 hours post induction of chronically infected ACH-2 cells.
Figure 2 shows the levels of expression of multiply spliced (MS HIV-I) and unspliced (US HTV-I) mRNA prior and post induction of chronically infected ACH-2 cells. Real time RT-PCR reactions were carried out using Taqman probes specific for early (multiply spliced) and late (unspliced) transcripts of HTV-I, tagged with FAM and TAMRA fluorescent dyes at the 5' and 3' ends respectively. Reactions were performed in triplicate for each time point as described in the Methods section and average values are shown. Maximal fold change in mRNA levels for early transcripts (MS HTV-I) was observed 8 hours post induction. Fold change for late transcripts (US
HTV-I) showed maximal increase 18 hours post induction. Figure 3 shows the hierarchical clustering of differentially expressed cellular genes before and after induction of chronically infected ACH-2 cells. The figure shows the hierarchical clustering of the cellular genes that showed significant differential expression (p < 0.001) across the time course (before induction up to 96 hours post induction), following reactivation of chronically infected ACH-2 cells as per the criteria described in the Methods. Genes that are on the color scale <1 showed up regulation, those on the color scale <1 were down regulated, while those that did not show any change with respect to normalized matched control are shown in black. The gray areas indicate missing data for the given gene and time point. The magnified panels indicate selected kinetic profiles that are seen before and following induction into active viral replication. (A) Up regulation of selected genes observed before induction; (B) Up regulation of genes immediately following induction; (C) Genes that are up regulated prior to induction and down regulated 12-24 hours post induction; (D) Genes that are up regulated in the early stage following reactivation, but are down regulated in the intermediate stage; (E) Genes that are down regulated before induction but are up regulated in the intermediate stage followed by down regulation in the late stage (48-96 hours p.i.).
Figure 4 shows the trends seen in pathways that show differential expression before and after induction of chronically infected ACH-2 cells. Pathway profiles observed prior to induction and following reactivation of ACH-2 cells with PMA over a period of 96 hours. The figure shows the number of genes in each pathway that were differentially expressed in a particular pathway,. (A) indicates the pathways that were maximally altered prior to induction. (B) includes the pathways that showed maximum change during the early phase of the lytic cycle, (0.5-8 hours p.i.). (C) represents the pathways that showed maximal change during the period of 12-24 hours post induction. Most pathways did not show any change during the period of 48-96 hours post induction. The groups above are a selected representation of the various pathways that changed differentially prior to induction and/or over the time course studied. Classification of the altered genes into various pathways was performed using the CGAP pathway databases. Figure 5 shows the hierarchical clustering of genes that show differential expression across three chronically infected cell lines prior to induction. Hierarchical clustering of differentially expressed genes that show a significant change in expression (p < 0.001), in the chronically infected cell lines ACH-2, Ul and Jl.1. Genes shown on the color scale >1 are up regulated, those on the color scale <1 exhibit down regulation, while black indicates normal expression. In several cases (e.g., FCGR2A, PPIB), dark gray areas indicate missing values. Many genes are altered similarly across the cell lines. Each cell line also shows some unique patterns of cellular expression. Data are the average of values from eight independent samples per cell line. The magnified portions of the cluster highlight some of the patterns of gene expression across the cell lines. (A) shows genes that are up regulated in all three cell lines; (B) shows genes that are down regulated in all three cell lines; (C) indicates the genes that are up regulated in ACH-2 and Jl.1 and down regulated in Ul; (D) indicates genes which show no significant similarity in their expression in the three cell lines.
Figure 6 shows the effects of specific agents on HIV p24 production in latently infected JLl cells. Different concentrations of the FTase inhibitor L-744,832 were tested in latently infected Jl.1 cells treated with 250 nM AZT. Samples were collected 24 hours after addition of agent and p24 concentrations were determined by ELISA. p24 production from cells treated by TNF-alpha was used as a positive control in J 1.1 cells compared to control (AZT treated cells). p24 production from untreated cells (No AZT) was also determined. Experiments were performed in triplicate and are representative of three independent experiments.
Figure 7 shows the effect of L-744832 on several HTV latently infected Jurkat clones.
Figure 8 shows shows the effect of L-744832 on viral reactivation in a sample from a 1-year aviremic patient.
Figure 9 shows shows the effect of L-744832 on viral reactivation in a sample from a long-term (8-year) aviremic patient. Table 1 : Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells. List of selected classes of genes based on known function that are differentially expressed in latently infected ACH-2 cells, relative to uninfected parental cell line, A3.01. A number of genes involved in similar cellular functions previously not associated with presence of proviral HIV were altered coordinately even during the latent non-replicative stage.
Table 1 : Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells.
Figure imgf000014_0001
Figure imgf000015_0001
DETAILED DESCRIPTION OF THE INVENTION
It has been discovered that farnesyl transferase (FTase) inhibitors, e.g., compounds of the formulae herein, can be used to reactivate viral replication processes in cells infected by an immunodeficiency virus, preferably human cells infected with HTV and thus can be used for treatment in BXV-infected individuals.
The methods of the invention in general comprise administration of a therapeutically effective amount of a compound that inhibits farnesyl transferase (a FTase inhibitor) to a patient in need of treatment, such as a mammal suffering from or susceptible to an immunodeficiency virus, particularly a human immunodeficiency virus such as HTV.
A variety of FTase inhibitor compounds can be employed in the methods of the invention. For example, suitable compounds have been reported previously including those in U.S. Patents 5,238,922; 5,571,792; and 5,571,835; WO 94/10138; WO 94/04561; WO 94/10138; WO 96/21456; and WO 97/02817.
Particularly preferred FTase inhibitor compounds for use in the methods of the invention exhibit good activity in a standard in vitro FTase inhibition assay, preferably an IC50 (concentration required to inhibit FTase activity by 50% relative to control) in such an assay of about 100 nM or less, more preferably an IC50 about 50 nM or less. As referred to herein, a standard in vitro FTase inhibition assay includes the following steps a) through c): a) admixing in a suitable assay solution 1) a potential FTase inhibitor compound, 2) [3H]farnesyl diphosphate, 3) farnesyl transferaseand 4) H-Ras; b) incubating the test mixture for 15 minutes at 370C; and c) measuring utilization of [3H] farnesyl diphosphate over that time relative to a control mixture that is prepared and incubated under the same conditions as the assay mixture but does not include the potential inhibitor compound. A suitable assay solution includes 50 mM HEPES5 pH 7.5, 5 mM MgCl2, 5 mM dithiothreitol. References herein to a standard in vitro farnesyl transferase inhibition assay are intended to refer to that protocol. That protocol also has been described in A.M. Garica et al., J. Biol Chem., 268: 18415-18418 (1993).
Specifically preferred FTase inhibitor compounds for use in the methods of the invention include the following where the compound is structurally depicted above one or more chemical names thereof (i.e. one or both of an IUPAC-type name and other designator (such as L-744,832, SCH 66336, etc.) are listed); and pharmaceutically acceptable salts of the compounds. In some instances, additional information is provided beneath the corresponding compound structure, such as the molecular formula of the structurally depicted compound.
Figure imgf000018_0001
L-744,832; (2S)-2[[(2S)-2-[(2S,3S)-2-[(2R)-2-Amino-3-mercaptopropyl]amino]-3- niethylpentyl]oxophenylpropyl]amino-4-(methylsulfonyl)butanoic acid 1-methylethyl ester; molecular formula: C26H4SN3O6S2.
Figure imgf000018_0002
SCH 66336; 4-{2-[4-(3,10-dibromo-8-chloro-6,l l-dihydro-5H- benzo[5,6]cyclohepta[l,2-b]pyridin-ll-yl)piperidin-l-yl]-2-oxo-ethyl}-piperidine-l- carboxylic acid amide.
Figure imgf000019_0001
R- 11577; (R)-6-amino[(4-chlorophenyl)( 1 -methyl-lH-imidazol-5-yl)methyl]-4-(3 - chlorophenyl)-l-methyl-2(lH)-quinolinone
Figure imgf000019_0002
BMS 214662; 3-benzyl-l-(3H-imidazol-4-ylmethyl)-4-(thiophene-2-sulfonyl)-2,3,4,5- tetrahydro- IH-benzo [e] [ 1 ,4]diazepine-7-caτbonitrile
Figure imgf000019_0003
FTI-276; molecular formula: C21H27N3O3S2
Figure imgf000020_0001
FTI-277; molecular formula: C22H29N3O3S2; 2-{[5-(2-amino-3-mercapto- propylamino)-biphenyl-2-carbonyl]-amino } -4-methylsulfanyl-butyric acid methyl ester
Figure imgf000020_0002
(2(.S)-[2(5)-[2(i?)-amino-3-mercapto]-propylamino-3(6)-methyl]penryloxy-3- henylpropionyl-methionine-sulphone isopropyl ester).
Figure imgf000020_0003
(2(S)-[2(S)-[2(R )-amino-3-mercapto]-propylamino-3(S)-methyl]pentyIoxy-3- phenylpropionyl-methionine-sulphone).
Figure imgf000021_0001
(N--[2(S)-[2(R )-amino-3-mercaptopropylamino]-3-methylbutyl]-L-phenylalanmyl-L- methionine).
Figure imgf000021_0002
(N-[2(S)-N'-(l-(4-cyanophenylmethyl)-lH-imidazol-5-yl-acetyl)amino-3(S)- methylpentyl]-N-l-naphthylmetliyl-glycyl-methionme-sulphone methylester).
The efficacy of any particular farnesyl transferase inhibitor in the therapeutic methods of the invention can be readily determined. For example, compounds with superior intrinsic inhibitory activity against and selectivity for farnesyl transferase can be identified through the in vitro assays discussed above and herein. In addition to the above discussed preferred FTase inhibitors, suitable FTase inhibitors compounds for use in the methods of the invention are disclosed below (including those compounds of groups (a) through (gg) as those groups of compounds are defined below, and other compounds defined below). It should be appreciated however that the present invention is not limited by the particular FTase inhibitor, and the invention is applicable to any such FTase inhibitor compound now known or subsequently discovered or developed.
FTase inhibitor compounds suitable for use in the methods of the invention will include those compounds that incorporate a cysteinyl or sulfhydryl containing moiety at the N-terminus of the molecule. More specifically, the following compounds are useful in the methods of the invention:
(a) a peptide that comprises the amino acids CA1A2X, wherein: C = cysteine;
Ai = an aliphatic amino acid; A2 = an aliphatic amino acid; and X = any amino acid;
(b) Cys - Xaa - Xaa - Xaa - NRR , wherein Cys = cysteine; Xaa1 = any amino acid in the natural L-isomer form;
Xaa2 = any amino acid in the natural L-isomer form; and
Xaa3 = NRR1 = an amide of any amino acid in the natural L isomer form, wherein R and R1 are independently selected from hydrogen, Ci-Ci2 alkyl, aralkyl, or unsubstituted or substituted aryl; (c) Cys - Xaa - Xaa - Xaa , wherein Cys = cysteine;
Xaa = any amino acid;
Xaa = the amino acid phenyl alanine or a p-fluorophenylalanine; and
Xaa3 = any amino acid;
(d) Cys - Xaa1 - dXaa2 - Xaa3, wherein
Cys = cysteine;
Xaa = any amino acid in the natural L-isomer form; dXaa = any amino acid in the natural L-isomer form; and
Xaa3 = any amino acid in the natural L-isomer form;
(e) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,238,922, incorporated herein by reference,
Figure imgf000023_0001
wherein:
X3 Y, and Z are independently H2 or O, provided that at least one of these is H2;
R is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R1NH may be absent;
R2j R3 and R4 are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring; and
R is H or a straight or branched chain aliphatic group, which may be substituted with an aromatic or heteroaromatic group;
(f) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,340,828, incorporated herein by reference,
Figure imgf000023_0002
wherein:
X and Y are independently H2 or O, provided that at least one of these is H2; R1 is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R NH may be absent;
R and R are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the.aliphatic substituents may be substituted with an aromatic or heteroaromatic ring;
Z is O or S; and n is 0, 1 or 2;
(g) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,340,828, incorporated herein by reference,
Figure imgf000024_0001
wherein:
X and Y are independently H2 or 0, provided that at least one of these is H2; R is H, an alkyl group, an acyl group, an alkylsulfonyl group or aryl sulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, or in the alternative, R NH may be absent; R and R are the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring; Z is O or S; and n is 0, 1 or 2; (h) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,352,705, incorporated herein by reference, (h) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,352,705, incorporated herein by reference,
Figure imgf000025_0001
wherein:
X and Y are independently H2 or O; R1 is an alkyl group, hydrogen, an acyl group, an alkylsulfonyl group or arylsulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbons atoms, which alternatively may be substituted with an aryl group;
R is the side chains of naturally occurring amino acids, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heterocyclic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic sύbstituents may be substituted with an aromatic or heteroaromatic ring;
R3 is an aromatic or heteroaromatic ring or in the alternative an alkyl group or an aryl or heteroaryl substituted alkane, wherein the aromatic ring is unsubstituted or in the alternative, substituted with one or more groups which maybe alkyl, halo, alkoxy, trifluoromethyl, or sulfamoyl groups, and which may be polycyclic; (i) compounds of the following formulae, which compounds are also disclosed in U.S. Patent No. 5,326,773 and PCT Publication No. WO 94/10137, incorporated herein by reference,
Figure imgf000025_0002
II
Figure imgf000026_0001
in or
Figure imgf000026_0002
rv
wherein in said formulae I, II, EI and IV:
R and R a are independently selected from hydrogen, a Ci-C6 alkyl group, a CI-CO acyl group, an aroyl group, a Ci-C6 alkylsulfonyl group, Ci-C6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally substituted with substituted or unsubstituted aryl or heterocycle;
R2, R3 and R are independently selected from: a) a side chain of naturally occurring amino acids, b) an oxidized form of a side chain of naturally occurring amino acids selected from methionine sulfoxide and methionine sulfone, c) substituted or unsubstituted Ci-C8 alkyl, C3-C8 cycloalkyl, C2-Cs alkenyl, aryl or heterocycle groups, wherein the aliphatic substituent is optionally substituted with an aryl, heterocycle or C3-C8 cycloalkyl; R ,5b is a C1-C6 alkyl group, a C1-Ce acyl group, an aroyl group, a Ci-C6 alkylsulfonyl group, Ci-C6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally substituted with substituted or unsubstituted aryl or heterocycle;
R is a substituted or unsubstituted aliphatic, aryl or heterocyclic group, wherein the aliphatic substituent is optionally substituted with an aryl or heterocyclic ring; and n is 0, 1 or 2;
(j) compounds of the following formulae, which compounds are also disclosed in U.S. Patent No. 5,504,212, incorporated herein by reference,
Figure imgf000027_0001
Figure imgf000027_0002
II
Figure imgf000028_0001
πi or
Figure imgf000028_0002
IV
wherein in said formulae I, π, IH and IV:
R is selected from hydrogen, a C1-CO alkyl group, a Ci-Ce acyl group, an aroyl group, a Ci-Ce alkylsulfonyl group, Ci-C6 aralkylsulfonyl group or arylsulfonyl group wherein the alkyl group and acyl group is optionally substituted with substituted or unsubstituted aryl or heterocycle;
R , R and R are independently selected from: a) a side chain of naturally occurring amino acids, b) an oxidized form of a side chain of naturally occurring amino acids selected from methionine sulfoxide and methionine sulfone, c) substituted or unsubstituted Ci-C8 alkyl, C3-Cg cycloalkyl, C2-C8 alkenyl, aryl or hetβrocycle groups, wherein the aliphatic substituent is optionally substituted with an aryl, heterocycle or C3-C8 cycloalkyl;
X is CH2CH2 or trans CH=CH; R is a substituted or unsubstituted aliphatic, aryl or heterocyclic group, wherein the aliphatic substituent is optionally substituted with an aryl or heterocyclic ring; and n is 0, 1 or 2;
(k) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 94/10138, incorporated herein by reference,
Figure imgf000029_0001
II
Figure imgf000030_0001
IV wherein in said formulae I, π, IE and IV,
R1 is hydrogen, an alkyl group, an aralkyl group, an acyl group, an aracyl group, an aroyl group, an alkylsulfonyl group, aralkylsulfonyl group or arylsulfonyl group, wherein alkyl and acyl groups comprise straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms;
R2, R3 and R5 are the side chains of naturally occurring amino acids, including their oxidized forms which may be methionine sulfoxide or methionine sulfone, or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic substituents may be substituted with an aromatic or heteroaromatic ring; R4 is hydrogen or an alkyl group, wherein the alkyl group comprises straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms;
R6 is a substituted or unsubstituted aliphatic, aromatic or heteroaromatic group such as saturated chains of 1 to 8 carbon atoms, which may be branched or unbranched, wherein the aliphatic substituent may be substituted with an aromatic or heteroaromatic ring;
T is O or S(O)n,; m is 0, 1 or 2; and n is 0, 1 or 2; (1) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 95/00497, incorporated herein by reference,
Figure imgf000031_0001
Figure imgf000031_0002
wherein in said formulae A, B and C: X is O or H2; m is 1 or 2; nis O or 1; t is 1 to 4;
R and R1 are independently selected from H, Ci-4 alkyl, or aralkyl;
R2, R3, R4, and R5 are independently selected from H, Ci-8 alkyl, alkenyl,
Figure imgf000032_0001
alkynyl, aryl, heterocycle, unsubstituted or substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl,
Figure imgf000032_0002
d) halogen,
2) C3-6 cycloalkyl,
3) OR6,
4) SR6, S(O)R6, SO2R6,
5) -NR6R7,
6)
7)
Figure imgf000032_0003
8)
NR6R7
O 9) 9)
Figure imgf000033_0001
10)
Figure imgf000033_0002
11) -SO2-NR6R7,
12)
R0
-N-SO2-R7
13)
R6
, or O
14)
OR6
O
and any two of R2, R3, R4, and R5 are optionally attached to the same carbon atom; Y is aryl, heterocycle, unsubstituted or substituted with one or more of:
1) Ci-4 alkyl, unsubstituted or substituted with: a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO,
2) aryl or heterocycle, 3) halogen, 4) OR6,
5) NR6R7,
6) CN,
7) NO2, or 8) CF3;
W is Ha or O;
Z is aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more of the following:
1) C1-4 alkyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, or e) HO,
2) aryl or heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN,
7) NO2, or
8) CF3;
6 7 S
R , R and R are independently selected from H, Ci-4 alkyl, C3-O cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) aryl or heterocycle, c) halogen,
Φ HO, e)
Figure imgf000034_0001
f) -SO2R9, or g) NRR1, wherein
Rδ and R7 may be joined in a ring, and
R and R may be joined in a ring; and
R9 is C1-4 alkyl or aralkyl.
(m) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 96/09821, incorporated herein by reference,
Figure imgf000035_0001
II
Figure imgf000035_0002
II! IV or
wherein in said formulae I, π, IH and IV: R1 is selected from: a) hydrogen, 0 b) R8S(O)2-, R8C(O)-, (R8)2NC(O> or R9OC(O)-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R8O-, R8S(O)1n-, R8C(O)NR8-, CN3 (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, OrR9OC(O)NR8-; R a and R are independently selected from: 5 a) hydrogen, b) Ci-C6 alkyl unsubstituted or substituted by alkenyl, R8O-, R8S(O)n,-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, c) aryl, heterocycle, cycloalkyl, alkenyl, R8O-, R8S(O)1n-, R8C(O)NR8-, CN, O NO2, (R8)2NC(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-CiO cycloalkyl;
R and R are independently selected from: a) a side chain of a naturally occurring amino acid, 5 b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted Ci-C20 alkyl, C2-C20 alkenyl, C3-Ci0 O cycloalkyl, aryl or heterocyclic group, wherein the substiruent is selected from F, Cl, Br, N(R8)2, NO2, R8O-, R8S(O)nO R8C(O)NR8-, CN, (R8)2N-C(NR8)-3 R8C(O)-, R8OC(O)-, N3, -N(R8)2, R9OC(O)NR8- and C1-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl; or R3 and R4 are combined to form - (CH2)s -;
R5a and R5 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C20 alkyl, C2-C20 alkenyl, C3-C10
• cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br5 N(R8)2, NO2, R8O-, R8S(O)1n-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, " R8OC(O)-, N3, -N(R8)2, R9OC(O)NR8- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl; or
R a and R are combined to form -(CH2)S- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0)m, -NC(O)-, and -N(COR )-; R6 is a) substituted or unsubstituted Ci-C8 alkyl, wherein the substituent on the alkyl is selected from:
1) aryl,
2) heterocycle, 3) -N(R9)2,
4) -OR8, or b)
R10
O I I l
\
X-Y is a)
R7'
O * b)
R 7b
^NΛ >
c)
Figure imgf000038_0001
d)
(O)m
e)
Figure imgf000038_0002
f) -CH2-CH2- ;
R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) Ci -CO alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl; R is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
R is independently selected from hydrogen, Ci-C6 alkyl and aryl;
R9 is independently selected from Ci-C6 alkyl and aryl;
R is independently selected from hydrogen and Ci-C6 alkyl;
R11 is independently selected from Ci-C6 alkyl;
Z1 and Z2 are independently H2 or O, provided that Z1 is not O when X-Y is - C(O)N(R7a); m is 0, 1 or 2; q is 0, 1 or 2; s is 4 or 5; and t is 3, 4 or 5; (n) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 96109820, incorporated herein by reference,
Figure imgf000039_0001
wherein: R1 is selected from: a) hydrogen, b) R5S(O)2-, R5C(O)-, (R5)2NC(O)- or R6OC(O)-, and c) Ci-Ce alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R5O-, R5S(O)n,-, R5C(O)NR5-, CN, (R5)2N-C(NR5)-5 R5C(O)-, R5OC(O)-, N3, -N(R5)2, or R6OC(O)NR5-; R a and R are independently selected from: a) hydrogen, b) Ci-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R5O-, R5S(O)n,-, R5C(O)NR5-, CN, (R5)2N-C(NR5)-, R5C(O)-, R5OC(O)-, N3, -N(R5)2, OrR6OC(O)NR5-, and c) aryl, heterocycle, cycloalkyl, alkenyl, R5O-, R5S(O)1nR5C(O)NR5-, CN, NO2, (R5)2N-C(NR5)-, R5C(O)-, R5OC(O)-, N3, -N(R5)2, OrR6OC(O)NR5-,
R is selected from: a) unsubstituted or substituted aryl, b) unsubstituted or substituted heterocycle, c) unsubstituted or substituted cycloalkyl, and d) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl; X-Y is a)
Figure imgf000040_0001
b)
Figure imgf000040_0002
Figure imgf000040_0003
d)
(O)m
e)
Figure imgf000041_0001
f) -CH2-CH2- ; R4a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, i) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci -Ce alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl; R5 is independently selected from hydrogen, Ci-C6 alkyl and aryl;
R6 is independently selected from Ci-C6 alkyl and aryl;
Z is independently H2 or O; m is 0, 1 or 2, provided that m is O when R5 = hydrogen; n is θ3 1, 2, 3 or 4; and t is 3, 4 or 5.
(o) compounds of the following formulae:
Figure imgf000042_0001
Figure imgf000042_0002
wherein in said formula A, B3 C and D:
X and Y are independently O or H2; m is 1 or 2; n is 0 or 1 ; p is 1, 2 or 3; q is 0, 1 or 2; t is 1 to 4;
R, R1 and R2 are independently selected from H, Ci-6 alkyl, or Ci-6 aralkyl;
R and R are independently selected from: a) hydrogen, b) Ci-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R6O-,
R5S(COq-, R7C(O)NR6-, CN, N3, R6OC(O)NR6-, R6R7N-C(NR6R8)-, R6C(O)-, R7R8NC(O)O-, R7R8NC(O)-, R6R7N-S(O)2-, -NR6S(O)2R5, R6OC(O)O-, -NR6R7, or R7R8NC(O)NR6-, c) unsubstituted or substituted cycloalkyl, alkenyl, R6O-, R5S(O)q-, R6C(O)NR6-, CN, NO2, R6R7N-C(NR8)-, R6C(O)-, N3, -NR6R7, halogen or
R7OC(O)NR6-, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-CiO cycloalkyl;
W is -CHR9- or -NR9-; Z is unsubstituted or substituted Ci-8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle; wherein the substituted group is substituted with one or more of:
1) Ci-4 alkyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, 2) aryl or heterocycle,
3) halogen, 4) OR6,
5) NR6R7,
6) CN,
7) NO2, or
9) CF3;
R5 is C1-4 alkyl or aralkyl;
R , R and R are independently selected from H, C1-4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) C1-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, e)
Figure imgf000044_0001
f) -SO2R5, or g) -NR6R7, or
R6 and R may be joined in. a ring, and
R7 and R may be j oined in a ring;
R9 is selected from H, C1-4 alkyl, C3-6 cycloalkyl, heterocycle and aryl, unsubstituted, monosubstituted or disubstituted with substituents independently selected from: a) C1-4 alkyl, b) C1-4 alkoxy, c) aryl or heterocycle, d) halogen, β) HO, f)
Figure imgf000044_0002
g) -SO2R5, and h) -NR6R7;
V is selected from -C(R11J=C(R11J-, CsC-, -C(O)-, -C(R1 V, -C(OR1 ^R11-, - CN(Rn)2Rπ-, -OC(R1V3 -NR11C(R1V. -C(RU)2θ-, -C(Ru)2NRn-3 -C(O)NR11-, - NR11C(O)-, O, -NC(O)R11-, -NC(O)OR11-, -S(O)2N(R11)-, -N(R1 ^S(O)2-, or S(O)n-;
R10 and R11 are independently selected from hydrogen, Ci-C6 alkyl, C2-C4 alkenyl, benzyl and aryl; or the pharmaceutically acceptable salt thereof.
Compounds suitable for use in the methods of the invention also include those farnesyl transferaseinhibitors that do not incorporates a cysteinyl or sulfhydryl containing moiety at the N terminus of the molecule. Such compounds may exhibit preferred pharmacological activity, e.g. by avoiding thiol-related reactions in vivo. More specifically, the following compounds may be suitable.
(p) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO-95/09001, incorporated herein by reference,
Figure imgf000045_0001
Figure imgf000045_0002
ill or
Figure imgf000046_0001
IV wherein in said formulae I, 33, HI and IV:
R1 is selected from: a) heterocycle, and b) Ci-Cio alkyl, which is substituted with heterocycle and which is optionally substituted with one or more of Ci-C4 alkyl, hydroxy or amino groups;
R2a and R2b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C alkyl, C2-C20 alkenyl, C3-CiO cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, NO2, R8O-, R9S(OV, R8C(O)NR8-, CN, (R8)2NC(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, R9OC(O)NR8- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-CiO cycloalkyl; or R2a and R2b are combined to form - (CH2)s -;
R and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) ' an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C1-C20 alkyl., C2-C20 alkenyl, C3-C10 cycloalkyl, axyl or heterocyclic group, wherein the substituent is selected from F, Cl5 Br5 N(R-V NO2, R8O-, R9S(O)1n-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, R9OC(O)NR8- and C1-C20 alkyl, and
5 d) C1-Ce alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-1O cycloalkyl; or
R and R are combined to form - (CH2)S -; R5a and R5b are independently selected from: a) a side chain of a naturally occurring amino acid, 0 b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C20 alkyl, C2-C20 alkenyl, C3-Ci0 5 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, N(R8)2, NO2, R8O-, R9S(O)1n-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2> R9OC(O)NR8- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl; or O R5a and R5b are combined to form - (CEb)S - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0)m, -NC(O)-, and -N(COR8)-; R6 is a) substituted or unsubstituted Ci-C8 alkyl, wherein the substituent on the alkyl is selected from: 5 1) aryl,
2) heterocycle,
3) -N(R9)2,
4) -OR8, or b) O X-Y is a)
R7a
Figure imgf000047_0001
O b)
Figure imgf000048_0001
C)
Figure imgf000048_0002
d)
(O)m
e)
Figure imgf000048_0003
f) -CH2-CH2- ;
R7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl; R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
R8 is independently selected from hydrogen,, Ci-C6 alkyl and aryl;
R9 is independently selected from Ci-C6 alkyl and aryl;
R is independently selected from hydrogen and Ci-Ce alkyl; R11 is independently selected from Ci-C6 alkyl;
Z is independently H2 or O; m is 0, 1 or 2; n is O3 1 or 2; and s is 4 or 5; (q) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 95/09000 and U.S. Patent No. 5,468,773, incorporated herein by reference,
Figure imgf000049_0001
m* and
Figure imgf000050_0001
rv wherein in said formula I, II, III and IV:
V is CH2, O5 S, HN3 or R7N;
R 3 R 5 R and R are independently the side chains of naturally occurring amino acids, including their oxidized forms which maybe methionine sulfoxide or methionine sulfone., or in the alternative may be substituted or unsubstituted aliphatic, aromatic or heteroaromatic groups, such as allyl, cyclohexyl, phenyl, pyridyl, imidazolyl or saturated chains of 2 to 8 carbon atoms which may be branched or unbranched, wherein the aliphatic substituents maybe substituted with an aromatic or heteroaromatic ring; X-Y is
Figure imgf000050_0002
Figure imgf000050_0003
Figure imgf000050_0004
Figure imgf000050_0005
f)
-CH2-CH2"
R6 is a substituted or unsubstituted aliphatic, aromatic or heteroaromatic group such as saturated chains of 1 to 8 carbon atoms, which may be branched or unbranched, wherein the aliphatic substituent may be substituted with an aromatic or heteroaromatic ring;
R7 is an alkyl group, wherein the alkyl group comprises straight chain or branched chain hydrocarbons of 1 to 6 carbon atoms, which may be substituted with an aromatic or heteroaromatic group;
Z is H2 or O; m is 0, 1 or 2; n is 0, 1 or 2; and o is 0, 1, 2 or 3;
(r) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 96/09836, incorporated herein by reference,
Figure imgf000051_0001
Il
Figure imgf000052_0001
IfI and
Figure imgf000052_0002
wherein in said formulae I, II, HE and IV: R is selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, (R10J2N-C(NR1 °)-,
R10C(O)-, Or R10OC(O)-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)NR10-; Rlb is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, cycloalkyl, alkenyl, alkynyl, (R10)2N- C(NR10)-, R10C(O)-, Or R10OC(O)-, and c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-
C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)NR10-; provided that Rlb is not R10C(O)NR10- ^ V is hydrogen and X-Y is -C(O)NR7a-; R and R are independently selected 1 a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C2O alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br3
NO2, R10O-, R1 (O)1n-, R10C(O)NR10O-, CN, (R10^N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or R a and R are combined to form - (CH2)S -; R3 and R4 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl,
Br, N(R10)2, NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)- , R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or R3 and R4 are combined to form - (CH2)S -;
R a and R independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocyclic groups wherein the substiruent is selected from F, Cl, Br3 NO25 R10O-, R11S(OV, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(9>, N3, -N(R1V R11C(O)NR10- and C1-C20 alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl; or
R5a and R5b are combined to form - (CH2)s - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0)m, -NC(O)-, and -N(COR10)-;
R6 is a) substituted or unsubstituted C1-C8 alkyl, wherein the substituent on the alkyl is selected from:
1) aryl,
2) heterocycle,
3) -N( R11)2,
4) -OR10, or b)
Figure imgf000054_0001
Figure imgf000054_0002
b)
Figure imgf000054_0004
Figure imgf000054_0003
d)
/m
\ -A y
Figure imgf000055_0001
f) -CH2-CH2- ; '~" ~
R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl; R is selected from: . a) hydrogen, r b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
R8 is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfmoroalkyl, F, Cl,
Br, R10O-, R11S(OV, R10C(O)NR10-, CN, NO2, R102N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)n,-, R10C(O)NH-, CN, H2N-C(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R1 °)2, or R11OC(O)NH-;
R is selected from: hydrogen, C1-C6 alkyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, N3, - N(R1V and R11OC(O)NR10-; provided that R9 is not R10C(O)NR10- when Rla is alkenyl, V is hydrogen and X-Y iS -C(O)NR7-;
R10 is independently selected from hydrogen, Ci-C6 alkyl, benzyl and aryl; R is independently selected from Ci-C6 alkyl and aryl; R12 is independently selected from hydrogen and Ci-C6 alkyl; R13 is Ci-C6 alkyl; V is selected from: a) aryl; b) heterocycle; or c) hydrogen;
W is -S(OV, -0-, -NHC(O)-, -C(O)NH-, -NHSO2-, -SO2NH-, N(R7a)- or N[C(O)R72]-;
Z is independently H2 or O; m is O, 1 or 2; n is O, 1, 2, 3 or 4, provided that n is not O when V is hydrogen and W is -S(O)1n-; p is O, 1 , 2, 3 or 4, provided that p = O when R9 is not hydrogen or Ci-C6 lower alkyl; q is O, 1 or 2; r is O or 1; s is 4 or 5; and t is 0, 1 or 2, provided that t = 0 when V is hydrogen;
(s) compounds of the following formulae which compounds are also disclosed in PCT Publication WO 96/10011, incorporated herein by reference,
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000057_0003
and
Figure imgf000057_0004
wherein in said formulae I5 II, HI and IV: R1 is hydrogen, C1-C6 alkyl or aryl; R a and R are independently selected from: 5 a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, 0 c) substituted or unsubstituted C1-C2O alkyl, C2-C2O alkenyL C3-C1O cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, NO2, R9O-, R10S(O)1n-, R9C(O)NR9-, CN, (R9)2NC(NR9)-, R9C(O)-, R9OC(O)-, N3, -N(R9)2, R10OC(O)NR9- and Ci-C20 alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group 5 selected from aryl, heterocycle and C3-CiO cycloalkyl; or R2a and R2b are combined to form -(CH2)S-; R3 and R4 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid O which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C2O alkyl, C2-C2O alkenyl, C3-CiO cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, 5 Br, NO2, R9O-, R10S(O)1n-, R9C(O)NR9-, CN, (R9)2NC(NR9)-, R9C(O)-, R9OC(O)-,
N3, -N(R9)2, R10OC(O)NR9- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-CiO cycloalkyl; or
R3 and R4 are combined to form-(CH2)s-; O R5a and R5b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C2O alkyl, C2-C20 alkenyl, C3-C10 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F5 Cl, Br5 NO2, R9O-, R10S(O)1n-, R9C(O)NR9-, CN5 (R9)2N-C(NR9)-5 R9C(O)-, R9OC(O)-, N3, -N(R9)2, R10OC(O)NR9- and C1-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl; or
R5a and R5b are combined to form -(CH2)S- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0)m, -NC(O)-, and -N(COR9)-; R6 is a) substituted or unsubstituted C1-C8 alkyl, wherein the substituent on the alkyl is selected from:
1) aryl,
2) heterocycle,
3) -N(R10)2,
4) -OR9, or b)
Figure imgf000059_0001
X-Y is a)
Figure imgf000059_0002
b)
Figure imgf000060_0001
c)
Figure imgf000060_0002
f) -CH2-CH2- ; R7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
R is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl., f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstirαted or substituted group selected from aryl, heterocycle and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl;
R8a and R8b are independently selected from hydrogen, F, Cl, Br, NO2, R11O-, R10S(O)1n-, CN, R9C(O)NR9-, (R9)2N-C(NR9)-, R9C(O)-, R9OC(O)-, N3, -N(R9)2, R10OC(O)NR9-, C1-C20 alkyl, aryl, heterocycle or Ci-C20 alkyl substituted with aryl or heterocycle; R is independently selected from hydrogen, Ci-C6 alkyl and aryl;
R is independently selected from Ci-C6 alkyl and aryl;
R11 is independently selected from hydrogen, Ci-C6 alkyl and aryl, provided R is Ci-C6 alkyl when n is O;
R12 is independently hydrogen or Ci-C6 alkyl; R13 is Ci-C6 alkyl;
is aryl or 1,2,3,4-tetrahydronaphthyl;
Z is independently H2 or O; m is 0, 1 or 2; n is independently 0 to 4; p is O or l; q is 0, 1 or 2; and s is 4 or 5; (t) compounds of the following formulae, which compounds are also disclosed in
PCT Publication 96/10034, incorporated herein by reference,
Figure imgf000062_0001
Figure imgf000062_0002
Figure imgf000062_0003
or
Figure imgf000062_0004
wherein in said formulae I, H, EI and IV: R1 is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R10OR11S(O)1n-, R10C(O)NR10-, CN5 NO2, (R1 ^2NC(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN,
Figure imgf000063_0001
R10C(O)-, R10OC(O)-, N3, -N(R10)2, OrR11OC(O)NR10-; R2a and R2b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10J2N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and C1-C20 alkyl, and d) Ci-Ce alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or
R2a and R2b are combined to form -(CH2)S-; R and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-C10 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R10)2, NO2, R10O-, RπS(O)m-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)- , R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and Ci-C20 alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-CiO cycloalkyl; or R3 and R4 are combined to form -(CH2)S-; R5a and RΛ are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C2O alkyl, C2-C2O alkenyl, C3-C10 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, N(Rl0)23 NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-5 R10C(O)- , R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and C1-C20 alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-C1 o cycloalkyl; or
R5a and R5b are combined to form -(CH2)S- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(0)m, -NC(O)-, and -N(COR10)-; R6 is a) substituted or unsubstituted Ci-Cs alkyl, wherein the substituent on the alkyl is selected from:
1) aryl, 2) heterocycle,
3) -N(Rπ)2,
4) -OR10, or b)
Figure imgf000064_0001
X-Y is a)
Figure imgf000064_0002
Figure imgf000065_0001
c)
Figure imgf000065_0002
Figure imgf000065_0003
Figure imgf000065_0004
f) -CH2-CH2- ; R a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloalkyl, and e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl; R7 is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted cycloaliyl, e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, - 6S -
f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocyclic, cycloalkyl and C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocyclic and cycloalkyl;
R is independently selected from: a) hydrogen, b) aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N35 -N(R10)2, OrR11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NH-, CN, H2N-C(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NH-;
R9 is selected from: a) hydrogen, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2NC(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R1 ] OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R1^2N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3> - N(R10)2, Or R11OC(O)NR10-;
R10 is independently selected from hydrogen, C1-C6 alkyl and aryl; R is independently selected from C1-C6 alkyl and aryl;
R is independently selected from hydrogen and C1-C6 alkyl;
R13 is independently selected from C1-C6 alkyl;
Ai and A2 are independently selected from a bond, -CH=CH-, -C=C-, -C(O)-, -C(O)NR10-, O, -N(R10)-, -NR10C(O)-, -S(O)2N(R10)-, -N(R10)S(O)2- or S(O)n,; V is selected from: a) hydrogen, b) heterocycle, c) aryl5 d) Ci-C2O alkyl wherein from 0 to 4 non-terminal carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C2O alkenyi; provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if Ai is a bond, n is 0 and A2 is S(O)m or a bond; W is a heterocycle; z is independently H2 or O; m is O, l or 2; n is 0, 1, 2, 3 or 4; p is O5 1, 2, 3 or 4; q is 0, 1 or 2; r is 0 to 5, provided that r is 0 when V is hydrogen; and s is 4 or 5;
(u) compounds of the following formulae, which compounds are also disclosed in PCT Publication No. WO 96/10035, incorporated herein by reference,
Figure imgf000067_0001
- S l -
Figure imgf000068_0001
Figure imgf000068_0002
wherein in said formulae I, II, DI and TV:
Rla and Rlb are independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R1V or R11OC(O)NR10-, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)n,-, R10C(O)NR10-, CN3 (R10^N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R1 \ Or R11OC(O)-NR10-;
R2a and R2b are independently selected from: a) hydrogen, b) C1 -C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-3 R10C(O)-, R10OC(O)-, - N(R1V Or R1 C(O)NR10-, c) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, R11 S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2; or R11OC(O)NR10-, and d) C1-CO alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-CiO cycloalkyl;
R3 and R4 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C2O alkyl, C2-C2O alkenyl, C3-C10 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R1V NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10^N-C(NR1 °)-, R10C(O)- , R10OC(O)-, N3, -N(R1 °)2, R11OC(O)NR10- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl; or
R3 and R4 are combined to form -(CH2)S-; R5a and R5 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, Br, CF3, N(R10)2, NO2, R10O-, R11S(O)n,-, R10C(O)NR10-, CN, (R10J2N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R1 \ R11OC(O)NR10- and C1-C20 alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or R a and R are combined to form -(CH2)S- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(O)m, -NC(O)-, and -N(COR10)-;
R6 is a) substituted or unsubstituted C1-C8 aLkyl, substituted or unsubstituted C5-C8 cycloalkyl, or substituted or unsubstituted cyclic amine, wherein the substituted alkyl, cycloalkyl or cyclic amine is substituted with 1 or 2 substituents independently selected from:
1) C1-C6 alkyL
2) aryl,
3) heterocycle,
4) -N(R11)2,
5) -OR10,or b)
X-Y is a)
b)
c)
d)
Figure imgf000070_0001
e)
Figure imgf000071_0001
f) -CH2-CH2- R7a is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl; R7b is selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl, f) a carbonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, C3-CiO cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl, and g) a sulfonyl group which is bonded to an unsubstituted or substituted group selected from aryl, heterocycle, C3-Ci0 cycloalkyl and Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-CiO cycloalkyl; R is independently selected from: a) hydrogen, _ H Λ —
b) aryl, heterocycle, C3-Ci0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N- C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R1V or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-CiO cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, RπS(O)m-5 R10C(O)NH-, CN, H2N-C(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R10OC(O)NH-;
R9 is selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-,
R11S(OV, R10C(O)NR10-, CN, NO2, (R10^N-C-(NR1 °)-, R10C(O)-, R10OC(O)-, N3, - N(R10)2, Or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, - N(R1V OrR11OC(O)NR10-;
R is independently selected from H, Ci-C6 alkyl, benzyl, substituted aryl and Ci-C6 alkyl substituted with substituted aryl;
R11 is independently selected from Ci-C6 alkyl and aryl; R12 is hydrogen or Ci-C6 alkyl; R13 is Ci-C6 alkyl;
Ai and A2 are independently selected from a bond, -CH=CH-, -C≡C-, -C(O)-, - C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -(O)2N(R10)-, -N(R10)S(O)2-, or S(O)1n; V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if Ai is S(0)m and V is not hydrogen if Ai is a bond, n is O and A2 is S(0)m; W is a heterocycle;
Z is independently H2 or 0; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is O, I5 2, 3 or 4; q is O3 1 or 2; r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; t is 3, 4 or 5; and u is 0 or 1;
(v) compounds of the following formulae,
Figure imgf000073_0001
Figure imgf000073_0002
wherein in formulae I, II, IE and IV:
Rla and Rlb are independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10^N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) C1-Ce alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(OV, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)-NR10-;
R2a and R2b are independently selected from: a) hydrogen, b b)) C C11--CC66 a allbkyl unsubstituted or substituted by alkenyl, R10O-, RπS(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or
R11OC(O)NR10-, c) aryl, heterocycle, cycloalkyl, alkenyl, R , 10 O, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R1O)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -
N(R1V Or R11OC(O)NR10-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl;
R3a and R3b are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C1-C20 alkyl, C2-C20 alkenyl, C3-C1O cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R1V NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-3 R10C(O)- , R10OC(O)-, N3, -N(R10)2, R11OC(O)NR10- and C1-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from, heterocycle and C3-CiO cycloalkyl; or R3a and R3b are combined to form -(CH2)S- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(O)1n, -NC(O)-, and -N(COR10)-;
R4 and R5 are independently selected from: a) hydrogen, and
Figure imgf000075_0001
R6 is a) substituted or unsubstituted Ci-Cs alkyl or substituted or unsubstituted
C3-C8 cycloalkyl, wherein the substituent on the alkyl is selected from:
1) aryl,
2) heterocycle,
3) -N(R11K 4) -OR10, or b)
Figure imgf000075_0002
R7 is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfmoroalkyl, F, Cl, Br3 R10O-, R11S(O)n,-, R10C(O)NH-, CN, H2NC(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R10K or R10OC(O)NH-;
R is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl5 Br5 R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C-(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br3 R10O-, R11S(O)1n-, R10C(O)NR10-, CN3 (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, - N(R10)2, or R11OC(O)NR10-;
R10 is independently selected from hydrogen, Ci-C6 alkyl, benzyl and aryl;
R11 is independently selected from Ci-C6 alkyl and aryl;
R is independently selected from hydrogen and Ci-C6 alkyl; R is independently selected from Ci-C6 alkyl;
1 O
A and A are independently selected from a bond, -CH=CH-, -C≡C-, -C(O)-, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2-, or S(O)1n;
V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if A is S(0)m and V is not hydrogen if A is a bond, n is O and A is S(0)m; W is a heterocycle; Z is independently H2 or O; m is O, 1 or 2; n is 0, 1, 2, 3 or 4; - IS -
p is O51,2, 3 or 4; q is 0, 1 or 2; r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; and u is 0 or 1;
(w) compounds of the following formulae.
Figure imgf000077_0001
Figure imgf000077_0002
wherein in said formulae I, II, IH and W: R Rllaa a annddR Rllbb a arree i inndependently selected from: a) hydrogen, b) aryl5 heterocycle, cycloalkyl, alkenyl, alkynyl, R10O-, R11S(O)1n-,
R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) Ci-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, cycloalkyl, alkenyl, alkynyl, R10O-, RπS(O)m-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R1V or R11OC(O)-NR10-; R and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted Ci-C20 alkyl, C2-C20 alkenyl, C3-Ci0 cycloalkyl, aryl or heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R10)2, NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10J2N-C(NR1 °)-, R10C(O)- , R10OC(O)-, N3, -N(R1V R11OC(O)NR10- and Ci-C20 alkyl, and d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or
R and R are combined to form -(CH2)S-; or R or R are combined with R to form a ring such that
Figure imgf000078_0001
R a, R , R7a and R are independently selected from: a) hydrogen, b) Ci-Ce alkyl unsubstituted or substituted by alkenyl, R10O-, R11S(O)n,-, 5 R10C(O)NR10-, CN, N3, (R10)2N-CCNR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or
R11OC(O)NR10-, c) aryl, heterocycle, cycloalkyl, alkenyl, R10O-,
R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10^N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, - N(R10)2, OrR11OC(O)NR10-, and 0 d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-Ci0 cycloalkyl; R5a and R are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid 5 which is: i) methionine sulfoxide, or ii) methionine sulfone, c) substituted or unsubstituted Ci-C2O alkyl, C2-C20 alkenyl, C3-CiO cycloalkyl, aryl or heterocycle group, wherein the substituent is selected from F, Cl, O Br, N(R10)2, NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)- , R10OC(O)-, N3, -N(R10)2, R11OC(O)NR10- and Ci-C20 alkyl, d) Ci-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or
R5a and R5b are combined to form -(CH2)S- wherein one of the carbon atoms is 5 optionally replaced by a moiety select from O, S(0)m, -NC(O)-, and -N(COR10)-;'
R is independently selected from hydrogen or Ci-C6 alkyl; o
R is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, O R10O-, RπS(O)m-, R10C(O)NR10-, CN, NO2, (R10J2N-C(NR1 °)-, R10C(O)-, R10OC(O)-,
N3, -N(R10)2, OrR11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, alkynyl, perfluoroalkyl, F, Cl5 Br3 R10O-, R11S(OV, R10C(O)NH-, CN5 H2NC(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, OrR10OC(O)NH-
R is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F5 Cl5 Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10^N-C-(NR1 °)-5 R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br,
R10O-, R11S(OV-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -
N(R10)2, OrR11OC(O)NR10-;
R10 is independently selected from hydrogen, Ci-C6 alkyl, benzyl and aryl; R is independently selected from Ci-C6 alkyl and aryl; R 1"2 is a) substituted or unsubstituted Ci-C8 alkyl or substituted or unsubstituted C3-C8 cycloalkyl, wherein the substituent on the alkyl or cycloalkyl is selected from:
1) aryl,
2) heterocycle, 3) -N(R1 \ -
4) -OR10, or b)
Figure imgf000080_0001
13
R is independently selected from hydrogen and Ci-C6 alkyl; R14 is independently selected from Ci-C6 alkyl; Ai and A2 are independently selected from a bond, -CH=CH-, -C-=C-, -C(O)-, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2-, or S(0)m; Q is a substituted or unsubstituted nitrogen-containing C4-C9 mono or bicyclic ring system, wherein the non-nitrogen containing ring may be an aromatic ring, a C5- C7 saturated ring or a heterocycle;
V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N9 and e) C2-C20 alkenyl, provided that V is not hydrogen if A1 is S (O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
W is a heterocycle;
X, Y and Z are independently H2 or O; m is 0, 1 or 2; n is 0, 15 2, 3 or 4; p is O, 1, 2, 3 or 4; q is 0, 1 or 2; . r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; t is 3, 4 or 5; and u is 0 or 1;
(x) compounds of the following formulae,
Figure imgf000081_0001
Figure imgf000081_0002
Figure imgf000082_0001
0 wherein in said formulae A, B and C:
Rla and Rlb are independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, 5 N3-, -N(R1V or R11OC(O)NR10-, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, C3-Ci0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)n,-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)-NR10-;
R2 and R3 are independently selected from H; unsubstituted or substituted Ci-8 O alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted C2-S alkynyl, unsubstituted or substituted aryl, unsubstituted or
V m5R7.
O "*^ γ0*
5 substituted heterocycle, wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl, b) (CH2)p0R6, 0 c) (CH2)PNR6R7, d) halogen, 2) C3-6 cycloalkyl, 3) OR6 4) SR6, S(O)R6, SO2R6, 5) -NR6R7
Figure imgf000083_0001
13)
^r R6 or
14)
^ir ORS 1 or
R and R are attached to the same C atom and are combined to form (CH2)u - wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S(O)m, -NC(O)-, and -N(COR10)-;
R is selected from H and CH3; and any two of R , R and R are optionally attached to the same carbon atom;
R6, R7 and R7 are independently selected from H, Ci-4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
Figure imgf000084_0001
f) -SO2R11, or g) N(R10)2;or
R6 and R7 may be joined in a ring; R7 and R7a may be joined in a ring; R is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-Ci0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl5 Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN3 NO2, (R10)2N- C(NR10K R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R1 C(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-Ci0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NH-, CN, H2N-C(NH)-, R1OC(O)-, R10OC(O)-, N3, -N(R10)2, or R10OC(O)NH-;
R9 is selected from: a^ hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-,
R10C(O)NR10-, CN, NO2, (R10)2N-C-(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F5 Cl, Br, R10O-, RuS(O)m-, R10C(O)NR10-, CN, (R10^N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, - N(R10)2, or R1 C(O)NR10-;
R10 is independently selected from hydrogen, Ci-C6 alkyl, benzyl and aryl; R11 is independently selected from Ci-C6 alkyl and aryl; Ai and A2 are independently selected from a bond, -CH=CH-, -C=C-, -C(O)-, - C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2-, or S(O)1n; G is H2 or O;
V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1-C20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C2O alkenyl, provided that V is not hydrogen if Ai is S(O)m and V is not hydrogen if Ai is a bond, 11 is O and A2 is S(O)m;
W is a heterocycle; X is -CH2-, -C(=O)-, or -S(=O)m-;
Y is aryl, heterocycle, unsubstituted or substituted with one or more of: 1) Ci-4 alkyl, unsubstituted or substituted with: a) Ci-4 allcoxy, b) NR6R7, c) C3-6 cycloalkyl,, d) aryl or heterocycle., e) HO, f) -S(O)1nR6, or g) -C(O)NR6R7,
2) aryl or heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN,
7) NO2, CF3,
9) -S(O)mR6,
10) -C(O)NR6R7, or
H) C3-C6 cycloalkyl;
Z is aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more of the following:
1 ) C1-4 alkyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR6, or g) -C(O)NR6R7,
2) aryl or heterocycle, 3) halogen,
4) OR6, 5) NRV5
6) CN5
7) NO2,
8) CF3; 9) -S(O)mR6,
10) -C(O)NR6R7, or
11) C3-C6 cycloalkyl; m is O3 1 or 2; n is O, 1, 2, 3 or 4; p is 0, 1, 2, 3 or 4; r is 0 to 5, provided that r is 0 when V is hydrogen; is 0 to 1; t is 0 to 1 ; and u is 4 or 5;
(y) compounds of the following formula,
Figure imgf000087_0001
V - A1(CR1a 2)nA2(CR1a 2)n - -'
Figure imgf000087_0002
wherein: ' Rla is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C2O alkenyl, C2-C20 alkynyl, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R1VN-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)NR10-, c) Ci-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, C3-C10 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, R10O-, RπS(O)m-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, or R11OC(O)-NR10-;
R is independently selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, R10O-, R11S(O)1n-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3 or -N(R10)2, c) C1-C6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic, C3-C10 cycloalkyl, C2-C20 alkenyl, C2-
C20 alkynyl, R10O-, R11S(O)1n-, CN, (R10)2N-C(NR10)-5 R10C(O)-, R10OC(O)-, N3 or - N(R10)2;
R2 and R3 are independently selected from: a) a side chain of a naturally occurring amino acid, b) an oxidized form of a side chain of a naturally occurring amino acid which is: i) methionine sulfoxide, or ii) methionine sulfone, and c) substituted or unsubstituted C1-C20 alkyl, substituted or unsubstituted C2-C20 alkenyl, substituted or unsubstituted C3-C10 cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heterocyclic group, wherein the substituent is selected from F, Cl, Br, N(R10)2, NO2, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, R11OC(O)NR10- and C1- C2o alkyl, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocycle and C3-Ci0 cycloalkyl; or R and R are combined to form ~(CH2)S-; or R or R are combined with R to form a ring such that
Figure imgf000088_0001
R 5 R 5 R a and R are independently selected from: a) hydrogen, b) Ci-C6 alkyl unsubstituted or substituted by C2-C2Q alkenyl, R10O-, R11S(OV, R10C(O)NR10-, CN, N3, (R10)2N- C(NR10)-, R10C(O)-, -N(R10)2s OrR11OC(O)NR10-, c) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Ci0 cycloalkyl, C2-C20 alkenyl, R10O-, R11S(Q)1n-, R10C(O)NR10-, CN3 NO2, (R10)2N-C(NR10)-, R10C(O)-, N3, -N(R10)2, OrR11OC(O)NR10-, and d) Ci-Ce alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl;
R is selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-Ci0 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, Ci-C20 perfluoroalkyl, aUyloxy, F, Cl, Br5 R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, R10IN-C(NR1 °)-, R10C(O)-, N3, -N(R1V (R 12)2NC(O)- OrR11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, C2-C20 perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)n,-, R10C(O)NH-, CN, H2N-C(NH)-, R10C(O)-, N3, -N(R10)2, OrR10OC(O)NH-;
R7 is independently selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-Ci0 cycloalkyl, and e) Ci-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and C3-C10 cycloalkyl;
R8 is selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C2-C20 alkenyl, C2-C20 alkynyl, Ci-C2O perfluoroalkyl, allyloxy, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, -S(O)2N(R1 °)2, CN, NO2,
(R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, OrR11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-CiO cycloalkyl, C2-C20 alkenyl, C2- C20 alkynyl, C2-C20 perfluoroalkyl, F, Cl, Br, R10O-, RπS(O)m-, R10C(O)NH-, CN5 H2N-C(NH)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, OrR10OC(O)NH-; R9 is selected from: a) hydrogen, b) C2-C20 alkenyl, C2-C20 alkynyl, C2-C20 perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)1n-, R10C(O)NR10-, CN, NO2, (R1 ^2N-C-(NR1 °>, R10C(O)-, R10OC(O)-, N3, - N(R10)2, Or R11OC(O)NR10-, and c) Ci-C6 alkyl unsubstituted or substituted by C2-C20 perfluoroalkyl, F,
Cl, Br7 R10O-, RnS(O)m-, R10C(O)NR10-, CN, (R10^N-C(NR1 °)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, Or R11OC(O)NR10-;
R10 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl; R is independently selected from C1-C6 alkyl and aryl; R12 is independently selected from hydrogen, C1-C6 alkyl and aryl, or (R!2)2 forms -(CH2)S-;
A1, A2 and A3 are independently selected from a bond, -CH=CH-, -C=C-, - C(O)-, -C(O)NR7-, -NR7C(O)-, O, -N(R7)-, -S(O)2N(R7)-, -N(R7)S(O)2-, or S(O)1n;
V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) Ci-C20 alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if Ai is S(0)m and V is not hydrogen if Ai is a bond, n is O and A2 is S(0)m;
W is a heterocycle;
Z is independently H2 or O; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is 0, 1, 2, 3 or 4; q is 0, 1, 2, 3 or 4; r is 0 to 5, provided that r is 0 when V is hydrogen; s is 4 or 5; and t is 3, 4 or 5; and (z) compounds of the following formula,
Figure imgf000091_0001
wherein:
Rla and Rlb are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)n,-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, OrR9OC(O)NR8-, c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N- C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)-NR8-; R2a, R2b and R3 are independently selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R O-, R9S(O)1n-, R8C(O)NR8-, CN, N3, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or
R9OC(O)NR8-, c) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted cycloalkyl, alkenyl, R8O-, R9S (O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, halogen or R9OC(O)NR8-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-CiO cycloalkyl; R4 and R5 are independently selected from: a) hydrogen, and
b)
Figure imgf000092_0001
V - A1(CR1a 2)nA2(CR1a 2)n
Figure imgf000092_0002
R is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, Ci-C6 perfluoioalkyl, F5 Cl, is Br, R8O-, R9S(O)1n-, R8C(O)NR8-, CN3 NO2, (R8)2NC(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, OrR9OC(O)NR8-, and c) Ci-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)n,-, R8C(O)NH-, CN, H2N-C(NH)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R8OC(O)NH-; R is selected from: a) hydrogen, b) C2-C6 alkenyl, C∑-Cβ alkynyl, C1-Ce perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)1n-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, Or R9OC(O)NR8-, and c) Ci-C6 alkyl unsubstituted or substituted by C1-C6 perfluoroalkyl, F, Cl,
Br, R8O-, R9S(OV, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, - N(R8)2, or R9OC(O)NR8-; o
R is independently selected from hydrogen, Ci-C6 alkyl, substituted or unsubstituted Ci-C6 aralkyl and substituted or unsubstituted aryl; R9 is independently selected from Ci-C6 alkyl and aryl; R is independently selected from hydrogen, C1-C6 alkyl, substituted or unsubstituted C1-C6 aralkyl and substituted or unsubstituted aryl;
Ai and A2 are independently selected from a bond, -CH=CH-, -C=C-, is - C(O)-, -C(O)NR8-, -MR8C(O)-, O, -N(R8)-, -S(O)2N(R8)-, -N(R8)S(O)2-, or S(0)m; V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1-C2Q alkyl wherein from O to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C2-C2O alkenyl, provided that V is not hydrogen if A1 is S(0)m and V is not hydrogen if
Ai is a bond, n is O and A2 is S(0)m;
W is a heterocycle; Y is selected from a bond, -C(R1 ^=C(R1 °)-, -C≡C-, -C(O)-, -C(R10)2-, -
C(0R10)R10-, -CN(R10)2R10-, -OC(R1V, -NR10C(R1V, -C(R10)2O-, -C(R10)2NR10, - C(O)NR10-, -NR10C(O)-, O, -NC(O)R10-, -NC(O)OR10-, -S(O)2N(R10)-, -N(R10)s(O)2-, or S(0)m;
Z is H2 or O; m is O, 1 or 2; n is O, 1, 2, or 4; p is 0, 1, 2, 3 or 4; r is O to 5, provided that r is O when V is hydrogen; and u is O or 1; or the pharmaceutically acceptable salts thereof.
Compounds for use in the methods of the invention also may obtained by fermentation of cultures of novel organisms, such as the compounds disclosed in U.S. Patent No. 5,420,334. Other suitable compounds are disclosed in U.S. Patent No. 5,420,245; European Patent Publication No. 0618 221; PCT Patent Publication Nos. WO 94/26723; WO 95/10514; WO 95/10515; WO 95/10516; WO 95/08542; WO 95/11917; and WO 95/12612. In certain embodiments of the invention, monomycin is less preferred and may be excluded from preferred aspects of the invention.
Specifically suitable compounds for use in the methods of the invention include the following:
Figure imgf000094_0001
Figure imgf000094_0002
Figure imgf000094_0003
or the pharmaceutically acceptable salts thereof.
Other specifically suitable compounds include the following:
5(S)-[2(R)-ammo-3-mercaptopropylamirio]--6(S)-methyl-2(R)-n-propyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone, 5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-methyl-3:>4-E- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-E-octenoyl- homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-i-propyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-n-butyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaρtopropylamino]-6(S)-methyl-2(R)-s-butyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone, 5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-t-butyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-cyclohexyl-3,4- E-octenoyl-homo serine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-cyclopentyl-3,4- E-octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-benzyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-i-propyl-3,4-E- octenoyl-homoserine, and the corresponding homoserine lactone, 5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-2(R)-i-propyl-3,4-E- octenoyl-methionine, and the corresponding methyl ester,
5(S)-[2(R)-amino-3-mercaptoproρylamino]-6(S)-methyl-2(R)-n-butyl-3,4-E- octenoyl-methionine, and the corresponding methyl ester,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-benzyl-3,4-E- octenoyl-methionine, and the corresponding methyl ester, 5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-n-propyl- octenoyl-homoserine, and the corresponding homoserine lactone,
5(S)-[2(R)-amino-3-mercaptopropylamino]-6(S)-methyl-2(R)-benzyl- octenoyl-homoserine, and the corresponding homoserine lactone, N-(3-phenyl-2(S)-(mercaptopropionylamino)prop-l-yl)isoleucyl-methionme,
N-(2(R)-arnino-3-rnercaptopropyl)isoleucyl-phenylalanyl-methionine,
N-(3-mercaptopropyl)isoleucyl-phenylalanyl-metliionine,
N-(3-mercaptopropyl)valyl-isoleucyl-methionine,
N-(2(R)-amino-3-mercaptopropyl)valyl-isoleucyl-methionine, N-(3 -methyl-2(S)-(cysteinylamino)but- 1 -yl)phenylalanyl-methionine5
N-(3 -methyl-2(S)-(mercaptopropionylamino)but- 1 -yl)-phenylalanyl methionine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3(S)methylpentyl]- phenylalanyl-methionine, N-[2(S)-(3-mercaptopropylamino)-3-(S)methylpentyl]-phenylalanyl- methiom'ne,
N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-(methionme sulfone),
N-(2(R)-ammo-3-mercaptopropyl)isoleucyl)-(p-iodophenylalanyl)-methionine, N-[2(R)-(cysteinyl-isoleucylamiiio)-3(S)-methylpentyl]-methionine,
N-[2(R)-(Nl-(2(R)-amino-3-mercaptopropyl)-isoleucylamino)-3-phenyl- propyljmethionine,
N-[2(R)-(N'-(2(R)-amino-3-mercaptopropyl)-isoleucylamino)-3(S)- methylpentyljmethionine, N-(3-mercaptopropyl)valyl-isoleucyl-methionine methyl ester,
N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-methionine ethyl ester,
N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-methionine benzyl ester, N-[2(R)-amrno-3-mercaptopropyl]-L-isoleucine-phenethylamide,
N- [2(R)-amino-3 -mercaptopropyl] -L-isoleucine-benzylamide, N-[2(R)-amiiio-3-mercaptopropyl]-L-isoleucine-3-methylbutylamide5 N-[2(R)-amino-3-mercaptopropyl]-L-isoleucme-3-phenylpropylamide, N-p^-amino-S-mercaptopropyy-L-isoleucyl-L-phenylalaninol, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-N'-methylbenzylarnide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(4-methoxybenzyl)aniide3
N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2,4-dichlorobenzyl)amide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(4- trifluoromethylbenzyljamide,
N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2,4-dichloro- ρhenethyl)amino,
N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2- benzimidazolylmethyl)amide,
N-[2(R)-ammo-3-mercaptopropyl] -L-isoleucine-(l -indanyl)amide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2,4-diniethylbenzyl)amide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2,3-dichlorobenzyl)amide,
N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(4-sulfamoylbenzyl)amide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucineanilide, N-[2(R)-ammo-3-mercaptopropyl]-L-isoleucine-(2,4-dimethylphenyl)amide, N-[2(R)-amino-3-mercaptopropyl]-L-isoleucine-(2,3-dimethylphenyl)amide, L-cysteinyl-L-isoleucine-phenethylamide,
N-[2(S)-[2(R)-amino-3-mercaptopropylamino]-3- methylpentyljphenethylamide,
N-(2(R)-amino-3-mercaptopropyl)-L-alaninebenzylamide, N-benzyl-[2(S)-2(R)-Amino-3-mercaptopropyl)-amino]butyramide, N-(2(R)-amino--3-mercaptopropyl)-L-norleucinebenzylamide,
N-(2(R)-amino-3-mercaptopropyl)-L-norvalinebenzylamide, N-(2(R)-ammo-3-mercaptopropyl)isoleucyl-phenylalanyl-homoserine, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-isoleucyl-homoserine, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-homoserine lactone, N-(2(R)-amino-3-mercaρtopropyl)isoleucyl-isoleucyl-homoserine lactone, N-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl-homocysteine lactone,
N-[2(S)-(2(R)-amino-3-mercaptopropyl)-3(S)-methylpentyl]-isoleucyl homoserine lactone, N-[Nr-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl]-3-(S)amino- tetrahydropyran-2-one,
N- [N'-(2(R)-amino-3 -mercaptopropytyisoleucy.1-isoleucyl] -3 -(S)- aminotetrahydropyran-2-one,
N-(2(R)-amino-3-mercaptopropyl)isoleucyl-isoleucyl-homocysteine lactone, N-[2(S)-(2(R)-amino-3 -mercaptopropylamino)-3 (S)-methylpentyl]isoleucyl- homoserine,
N-[N'-(2(R)-amino-3-mercaptopropyl)isoleucyl-phenylalanyl]-3(S)-amino-4- hydroxypentanoic acid, .
N-[N'-(2(R)-amino-3-mercaptopropyl)isoleucyl-isoleucyl]-3-(S)-amino-4- hydroxypentanoic acid,
N-[2(S)-(2(R)-ammo-3-mercaptopropylammo)-3-(S)-methylpentyl]-N-methyl- isoleucyl-homoserine,
N-[2(S)-(2(R)-ammo-3-mercaptopropylamino)-3(S)-methylpentyl]-N-methyl- isoleucyl-homoserine lactone, N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3(S)-meth.ylpentyl]-N-meth.yl- phenylalanyl-homoserine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3(S)-methylpentyl]-N-methyl- phenylalanyl-homoserine lactone,
N-[2(S)-(2(R)-amino-3-mercaptopropylammo)-3-methyl-butyl]-N-methyl- phenylalanyl-homoserine lactone,
3(S)-(N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylρentyl)-N- methyl-isoleucylamino)-3-methyltetra-hydropyran-2-one,
2(S)-(N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3(S)-methylpentyl]-N- methyl-isoleucylamino)-2-methyl-5-hydroxypentanoic acid, 2(S)-(N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N- methyl-isoleucylamino)-5-methyl-5-hydroxyhexanoic acid, N-[2(S)-(2(R)-amino-3-mercaptopropylatnmo)-3(S)-methylpentyl]-N-methyl- norvalyl-homoserine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-metliylpentyl]-N-metliyl- norvalyl-homoserine lactone, N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- isoleucyl-methionine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-mettiyl- isoleucyl-methionine methyl ester,
N-[2(S)-(2(R)-ardno-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- phenylalanyl-methionine,
N-[2(S)-(2(R)-amino-3-mercaptopropylarnino)-3(S)-methylpentyl]-N-methyl- phenylalanyl-methionine methyl ester,
3(S)-(N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N- methyl-isoleucylamino)-6,6-dimethyl-tetrahydropyran-2-one, N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- norvalyl-methionine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- norvalyl-methionine methyl ester,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- D-norvalyl-homoserine,
N-[2(S)-(2(R)-amino-3-mercaptopropylamino)-3-(S)-methylpentyl]-N-methyl- D-norvalyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylpropionyl-homoserine lactone, 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylpropionyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-2- methyl-3-phenylpropionyl-homoserine lactone,
2(S)-[2(S)-[2(^)-ammo-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-2- methyl-3-phenylpropionyl-homoserine, 2(S)-[2(S)-[2(R)-εmiino-3-mercapto]propylainiiio-3(S)-methyl]-pentyloxy-4- pentenoyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-4- pentenoyl-homoserines, 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]- pentyloxypentanoyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto)propylamino-3(S)-methyl]- pentyloxypentanoyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-4- methylpentanoyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-4- methylpentanoyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- methylbutanoyl-homoserine lactone, 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- methylbutanoyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylbutanoyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylbutanoyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]pentylthio-2- methyl-3 phenylpropionyl-homόserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentylthio-2- methyl-3-phenylpropionyl-homoserine, 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentylsulfonyl-
2-methyl-3-phenylpropionyl-homoserine lactone,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentylsulfonyl- 2-methyl-3-phenylpropionyl-homoserine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylpropionyl-methionine methyl ester, 2(S)-[2(S)-[2(R)-ainino-3-mercapto]propylarnino-3(S)-inetliyl]-pentyloxy-3- phenylpropionyl-methionine,
2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylpropionyl-methionine sulfone methyl ester, 2(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- phenylpropionyl-methionine sulfone,
2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3-(S)-methyl]-pentyloxy-3- naphth-2-yl-propionyl-methionine sulfone methyl ester,
2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- naphth-2-yl-propionyl-methionine sulfone,
2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3-(S)-methyl]pentyloxy-3- naphth-1-yl-propionyl-methionine sulfone methyl ester,
2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3(S)-methyl]-pentyloxy-3- naphth- 1 -yl-propionyl-methionine sulfone, 2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3-(S)-methyl]pentyloxy-3- methybutanoyl-methionine methyl ester,
2-(S)-[2(S)-[2(R)-amino-3-mercapto]propylamino-3-(S)-methyl]pentyloxy-3- methybutanoyl-methionine,
Disulfide of 2(S)-[2(S)-[2(R)-amino-3-mercaρto]ρropylamino-3(S)- methyl]pentyloxy-3-phenylpropionyl-homoserine lactone,
Disulfide of 2(S)-[2(S)-[2(R)-amino-3-mercaρto]propylamino-3(S)- methyl]pentyloxy-3-phenylpropionyl-homoserine,
Disulfide of 2(S)-[2(S)-[2(R)-amino-3-mercapto]ρropylammo-3(S)- methyl]pentyloxy-3-methylbutanoyl-methionine methyl ester, l-[2-(R)-amino-3-mercaptopropyl]-2(S)-(l-butyl)-4-(2,3- dimethylbenzoyl)piperazine dihydrochloride, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(n-butyl)-4-(l-naphthoyl)piperazine, l-[2(R)-amino-3-mercaρtoρropyl]-2(S)-benzyl-4-[l-(2,3- dimethyl)benzoyl]pip erazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(2-methoxy)ethyl-4-[l-(2,3- dimethyl)benzoyl]piperazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(2-methylthio)ethyl-4-[l-(2,3- dimethyl)benzoyl]piperazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(π-butyl)-4-[7-(23- dihydrobenzofuroyl)Jpiperazine, l-(2(R)-amino-3-mercaptopropyl)-4-(l-naphthoyl)-2(S)-pyridinylcarboxyl-4- piperazine dihydrochloride,
Methyl 4-(2(R)-amino-3-mercaptopropyl)-l -(I -naphthylmethyl)piperazine-2- carboxylate hydrochloride, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(2-methoxyethyl)-4-(l- naphthoyl)piperazine, l-[2(R)-amino-3-mercaptoρropyl]-2(S)-n-butyl-4-(8- quinolinylcarbonyl)piperazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(2-(l-propoxy)ethyl)-4-(l- naphthoyl)piperazine, l-[2(R)-amino-3-mercaptopropyl)-2(S)-(3-methoxy-l-propyl)-4-(l- naphthoyl)piperazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-(2-(l-proρoxy)ethyl)-4-(8- quinolinoyl)piperazine, l-[2(R)-amino-3-mercaptopropyl]-2(S)-[(3-pyridyl)rriethoxyethyl)]-4-(l- naphthoyl)piperazine, l-[2(R)-amino-3-mercaptopropyl]-4-naphthoyl-2(S)-(2- phenylsulfonylethyl)piperazine dihydrochloride, bis- 1, V- [2(R)-amino-3 -(2(S)-(2-methoxyethyl)-4-naphthoyl- 1 - piperazinyl)]ρropyl disulfide tetrahydrochloride, &w-l,l'-[2(R)-amino-3-(4-naphthoyl-2(S)-(2-ρhenylsulfonylethyl)-l- piperazinyl)]propyl disulfide tetrahydrochloride, l-[2(R)-amino-3-mercaptopropyl]-4-naphthoyl-2(S)-(2- cyclopropyloxyethyl)piperazine dihydrochloride, l-[2(R)-amino-3-mercaptopropyl]-4-(l-naphthoyl)-2(S)-(4- acetamidobutyl)piperazine dihydrochloride, l-[2(R)-amino-3-mercaptopropyl]-4-naphthoyl-2(S)-(2- cyclopropylmethylsulfonylethyl)piperazine dihydrochloride,
Pyroglutamyl-valyl-phenylalanyl-methionine,
Pyroglutamyl-valyl-plienylalanyl-methionine methyl ester, Pyroglutamyl-valyl-isoleucyl-methionine,
Pyroglutamyl-valyl-isoleucyl-methionine methyl ester,
Nicotinoyl-isoleucyl-phenylalanyl-methionine,
Nicotinoyl-isoleucyl-phenylalanyl-methioπine methyl ester,
N-[2(S)-(L-pyroglutamylamino)-3-methylbutyl]phenylalanyl-methionine, N-[2(S)-(L-pyroglutamylamino)-3-methylbutyl]phenylalanyl- methioninemethyl ester,
N-[5(S)-(L-pyroglutamylamino)-6(S)-methyl-2(R)-butyl-3,4(E)octenoyl]- methionine,
N-[5(S)-(L-ρyroglutamylamino)-6(S)-methyl-2(R)-butyl-3,4(E)octenoyl]- methionine methyl ester,
N-[5(S)-((Imidazol-4-yl)acetylammo)-6(S)-methyl-2(R)-butyl- 3,4(E)octenoyl]-methionine,
N-[5(S)-((Imidazol-4-yl)acetylamino)-6(S)-methyl-2(R)-butyl- 3,4(E)octenoyl]-methionine methyl ester, N-[5(S)-((Imidazol-4-yl-carbonylamino)-6(S)-methyl-2(R)-butyl-
3,4(E)octenoyl]-methionine,
N-[5(S)-((Imidazol-4-yl-carbonylamino)-6(S)-methyl-2(R)-butyl- 3,4(E)octenoyl]-methionine methyl ester,
N-[2(S)-(2(S)-(Imidazol-4-yl)acetylammo)-3(S)-methylpentyloxy)-3- phenylpropionyl]-methionine,
N-[2(S)-(2(S)-(Imidazol-4-yl)acetylammo)-3(S)-methylpentyloxy)-3- phenylpropionylj-methionine methyl ester,
N-[2(S)-(2(S)-Pyroglutamylamino-3(S)-methylpentyloxy)-3-phenylpropionyl]- methionine, N-[2(S)-(2(S)-Pyroglutamylamino-3(S)-methylpentyloxy)-3-phenylpropionyl]- methionine methyl ester, N-[2(S)-(2(S)-Imidazol-4-yl-carbonyl)amino)-3(S)-methylpentyloxy)-3- phenylpropionyl]-methionine,
N-[2(S)-(2(S)-Imidazol-4-yl-caxbonyl)amino)-3(S)-methylpentyloxy)-3- phenylpropionyl]-methionine.methyl ester, N-[2(S)-(2(S)-((3-Picolinyl)amino)-3(S)-methylpentyloxy)-3- phenylpropionyl] -methionine,
N-[2(S)-(2(S)-((3-Picolinyl)amino)-3(S)-meth.ylρentyloxy)-3- phenylpropionyl]-methionine methyl ester,
N-[2(S)-(2(S)-((Histidyl)amino)-3(S)-methylpentyloxy)-3-phenylproρionyl]- methionine,
N-[2(S)-(2(S)-((Histidyl)amino)-3(S)-methylρentyloxy)-3-ρhenylpropionyl]- methionine methyl ester,
N-Benzyl-N-[2(S)-((Imidazol-4-yl-carbonyl)arnino)-3(S)-methylpentyl]glycyl- methionine, N-Berizyl-N-[2(S)-((Lxιidazol-4-yl-carbonyl)amino)-3(S)-methylpentyl]glycyl- methionine methyl ester,
N-Benzyl-N-[2(S)-((Imidazol-4-yl-acetyl)amino)-3(S)-methylρentyl]glycyl- methionine,
N-Benzyl-N-[2(S)-((Imidazol-4-yl-acetyl)amino)-3(S)-methylpentyl]glycyl- methionine methyl ester,
N-Benzyl-N-[2(S)-((Pyroglutamyl)amino)-3(S)-methylpentyl]- glycyhnethionine,
N-Benzyl-N-[2(S)-((Pyroglutamyl)amino)-3(S)-methylpentyl]- glycylmethionine methyl ester, N-(I -Naphthylmethyl)-N-[2(S)-((imidazol-4-yl-carbonyl)ammo)-3(S)- methylpentylj-glycyl-methionine,
N-(l-Naphthylmethyl)-N-[2(S)-((imidazol-4-yl-carbonyl)amino)-3(S)- methylpentylj-glycyl-methionine methyl ester,
N-(l-Naphthylmethyl)-N-[2(S)-((imidazol-4-yl-acetyl)amino)-3(S)- methylpentylj-glycyl-methionine, N-(l-Naphthylmethyl)-N-[2(S)-((imidazol-4-yl-acetyl)amino)-3(S)- methylpentyl]-glycyl-methionine methyl ester,
N-(l-Naphthylmethyl)-N-[2(S)-((pyroglutamyl)amino-3(S)-methylpentyl]- glycyl-methioriine., N-(l-Naρhthylmetliyl)-N-[2(S)-(φyroglutamyl)ammo-3(S)-inethylpentyl]- glycyl-methionine methyl ester,
N-[I -(Pyroglutamylamino)cyclopent- 1 -yl~methyl]-N-( 1 -naphthylmethyl)- glycyl-methionine methyl ester,
N-[l-(Pyroglutaniylamino)-cyclopent-l-yl-methyl]-N-(l-naphthylrnethyl)- glycyl-methionine,
N-(2(S)-L-Histidylarnino-3(S)-methylpentyl)-N-(benzylmethyl)- glycylmethionine methyl ester,
N-(2(S)-L-Histidylamino-3(S)-methylpentyl)-N- (benzylmethyl)glycylmethionine, N-(2(S)-L-Histidylamino-3(S)-methylpentyl)-N-(l- naρhthyrmethyl)glycyrmethionine methyl ester,
N-(2(S)-L-Histidylamino-3(S)-methylpentyl)-N-(l-naphthylmethyl)glycyl- methionine,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-methylbutanoyl- methionine methyl ester,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-methylbutanoyl- methionine,
2(S)-[2(S)-(Imidazol-4-yl-acetyl)amino-3(S)-methylpentyloxy]-3- methylbutanoyl-methionine methyl ester, 2(S)-[2(S)-(Imidazol-4-yl-acetyl)ammo-3(S)-me%lpentyloxy]-3- methylbutanoyl-methiomne,
N-(Benzyl)-N-[2(S)-(2-oxoρyrrolidm-5(R,S)-yl-methyl)amino- 3(S)methylpentyl]-glycyl-metbionine methyl ester,
N-(Benzyl)-N-[2(S)-(2-oxoρyrrolidin-5(R,S)-yl-methyl)amino- 3(S)methylpentyl]-glycyl-methionine, N-(Ben2yl)-N-(2(S)-[((D,L)-2-thiazolyl)alaiiyl)amino]-3(S)methylρentyl)- glycyl-methionine methyl ester,
N-(Benzyl)-N-(2(S)-[((D,L)-2-thiazolyl)alanyl)amino]-3(S)methylpentyl)- glycyl-methionine, N-(BeiKyl)-N-[2(S)-(3-pyridylmethyl)amino-3(S)-methylpentyl]- glycylmethionine methyl ester,
N-(Beri2yl)-N-[2(S)-(3-pyridylmethyl)amino-3(S)-methylpentyl]- glycylmethionine,
2(S)-[2(S)-(2-Oxopyrrolidin-5(S)-yl-methyl)amino-3(S)methylpentyloxy]-3- phenylpropionyl-methionine methyl ester,
2(S)-[2(S)-(2-Oxopyrrolidin-S(S)-yl-methyl)amino-3(S)-methyl-pentyloxy]-3- phenylpropionyl-methionine,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylρentyloxy]-3-(l- naphthyl)propionyl-methionine sulfone methyl ester, 2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-(l- naphthyl)propionyl-methionine sulfone,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-(2- naphthyl)propionyl-methionine sulfone methyl ester,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-(2- naphthyl)propionyl-methionine sulfone,
2(S)-[2(S)-(Imidazol-4-yl-acetyl)ammo-3(S)-methylρentyloxy]-3-(l- naphthyl)propionyl-methionine sulfone methyl ester,
2(S)-[2(S)-(itnidazol-4-yl-acetyl)amino-3 (S)-methylρentyloxy]-3-(l - naphthyl)propionyl-methionine sulfone, 2(S)-[2(S)-(Imidazol-4-yl-acetyl)amino-3(S)-methylρentyloxy]-3-(2- naρhthyl)propionyl-methionine sulfone methyl ester,
2(S)-[2(S)-(lmidazol-4-yl-acetyl)ammo-3(S)-methylpentyloxy]-3-(2- naphthyl)propionyl-methionine sulfone,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(3- quinolylmethyl)glycyl-methionine methyl ester, N-[2(S)-(L-pyiOglutamyl)amino-3(S)-methylpentyl]-N-(3- quinolylmethyl)glycyl-methionine,
N-(Benzyl)-N-[2(S)-(tetrazol-l-yl-acetyl)amino-3(S)-methylpentyl]glycyl- methionine methyl ester, N-(Benzyl)-N-[2(S)-(tetrazol- 1 -yl-acetyl)amino-3 (S)-methylpentyl]glycyl- methionine,
N-(B enzyl)-N- [2(S)-nicotinoylamino-3 (S)-methylpentyl] -glycylmethionine- methyl ester,
N-(Beπzyl)-N-[2(S)-mcotmoylamino-3(S)-methylpentyl]-glycylmethionine, N-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyl]-N-(l-naρhthylmethyl)- glycyl-methionine sulfoxide methyl ester,
N-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyl]-N-(l-naphthylmethyl)- glycyl-methionine sulfoxide,
2(S)-(2(S)-[2(S,R)-(Imidazol-4-yl)-2-aminoacetyl)ammo]-3(S)- methylpentyloxy)-3-phenylpropionyl-methionine sulfone methyl ester,
2(S)-(2(S)-[2(S,R)-(Imidazol-4-yl)-2-aminoacetyl)amino]-3(S)- methylpentyloxy)-3 -phenylpropionyl-methionine sulfone,
2(S)-(2(S)-[2(R,S)-(Imidazol-4-yl)-2-ammoacetyl)amino]-3(S)- methylpentyloxy)-3 -phenylpropionyl-methionine sulfone methylester, 2(S)-(2(S)-[2(R,S)-(hnidazol-4-yl)-2-aminoacetyl)amino]-3(S)- methylpentyloxy)-3-phenylpropionyl-methionine sulfone,
N-(2(S)-[2(S,R)-(]midazol-4-yl)-2-aminoacetyl]amino-3(S)-methylρentyl)-N- (1 -naphthyhnethyl)-glycyl-methioninemethyl ester,
N-(2(S)-[2(S,R)-(hnidazol-4-yl)-2-aminoacetyl]amino-3(S)-methylpentyl)-N- (1 -naphthylmethyl)-glycyl-methiomne,
N-(2(S)-[2(R,S)-(Imidazol-4-yl)-2-aminoacetyl]ammo-3(S)-methylpentyl)-N- ( 1 -naphthyhnethy^-glycyl-methioninemethyl ester,
N-(2(S)-[2(R,S)-(Imidazol-4-yl)-2-aminoacetyl]ammo-3(S)-methylpentyl)-N- ( 1 -naphthyhnethyl)-glycyl-methionine, N-(2(S)-[(Imidazol-4-yl)methyl]amino-3(S)-methylpentyl)-N-(l- naphthylmethyl)-glycyl-methionine methyl ester, N-(2(S)-[(Imidazol-4-yl)methyl]amino-3(S)-methylpentyl)-N-(l- naphthylmethyl)-glycyl-methionine,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylρentyl]-N-(l- naphthylmethyl)glycyl-methionine isopropyl ester, N-[2(S)-(L-pyiOglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-methionine t-butyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(4-quinolyl-5- methyl)glycyl-methionine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(4- quinolylmethyl)glycyl-methionine,
N-(2(S)-[3 -(Imidazol-4-yl)propyl] amino-3 (S)-methylpentyl)-N-(l - naphthylmethyl)glycyl-methionine methyl ester,
N-(2(S)-[3-(Tmidazol-4-yl)propyl]amino-3(S)-methylpentyl)-N-(l- naphthyhnethyl)glycyl-methionine, N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-norleucine,
N-[2(S)-(L-pyroglutamyl)amino-3 (S)-methylpentyl]-N-(l - naphthylmethyl)glycyl-norleucine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l - naphthylmethyl)glycyl-glutamine,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyknethyl)glycyl-glutamine t-butyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-[5- (dimethylamino)naphthylsulfonyl]glycyl-methionine methyl ester, N-[2(S)-(3-pyridylmethyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-methionine,
2(S)-(2(S)-[2-(Itnidazol-4-yl)ethyl]amino-3(S)-methylpentyloxy)-3- phenylpropionyl-methionine sulfone methyl ester,
2(S)-(2(S)-[2-(Imidazol-4-yl)ethyl]amino-3(S)-methylpentyloxy)-3- phenylpropionyl-methionine sulfone, N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-serine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-(D5L)-serine, N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-(L,D)-serine3
N-[2(S)-(L-pyroglutamyl)amino-3(S)-metliylpentyl]-N-(l- naphthylmethyl)glycyl-homoserine lactone,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-homoserine,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(ciniiamyl)glycyl- methionine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylρentyl]-N-(cuinamyl)glycyl- methionine, N-(2(S)-[2-(Imidazol-4-yl)ethyl]amino-3(S)-methylpentyl)-N-(l- naphthyhnethyl)glycyl-methionine methyl ester,
N-(2(S)-[2-(Imidazol-4-yl)ethyl]amino-3(S)-methylpentyl)-N-(l- naphthylmethyl) glycyl-methionine,
N-[2(S)-(L-ρyroglutamyl)amino-3(S)-methylpentyl]-N-(l-naρhthyl- methyl)glycyl-alanine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l-naphthyl-5- methy^glycyl-alam'ne,
N-[2(S)-(D-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-methionine methyl ester, N-[2(S)-(D-pyroglutamyl)aiBino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-methionine,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-phenylpropionyl- methionine sulfone methyl ester,
2(S)-[2(S)-(L-Pyroglutamyl)amino-3(S)-methylpentyloxy]-3-phenylpropionyl- methionine sulfone, N-[2(S)-CL-ρyroglutamyl)amino-3(S)-methylpentyl]-N-(233- methylenedioxybenzyl)glycyl-methionine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(2,3- methylenedioxybenzyl)glycyl-methioπine, N-[2(S)-(Imidazol-4-yl-acetyl)ainino-3(S)-methylpentyl]-N-(2,3- dihydrobenzofuran-7-yl-methyl)glycyl-methionine methyl ester,
N-[2(S)-(Imidazol-4-yl-acetyl)amino-3(S)-methylpentyl]-N-(2,3- dihydrobenzoflιran-7-yl-methyl)glycyl-rαethionine,
N-(2(S)-[3-(3-indolyl)propionyl]amino-3(S)-methylpentyl)-N-(l- naphthylmethyl)glycyl-methionine methyl ester,
N-(2(S)-[3-(3-indolyl)propionyl]amino-3(S)-methylpentyl)-N-(l- naphthylmethyl)glycyl-methionine,
N-(2(S)- [3-(l -indolyl)propionyl] amino-3(S)-methylpentyl)-N-( 1 - naρhthyhnethyl)glycyl-methionine methyl ester, N-(2(S)-[3-(l-indolyl)proρionyl]amino-3(S)-methylpentyl)-N-(l- naphthyhnethyl)glycyl-methionine,
N-[2(S)-(L-pyroglutamyl)amino-3 (S)-methylpentyl]-N-( 1 - naphthyhnethyl)glycyl-histidine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-histidine,
N-[2(S)-(L-ρyroglutamyl)amino-3(S)-methylρentyl]-N- (cyclopropyhnethyl)glycyl-methionine methylester,
N-[2(S)-(L-ρyroglutamyl)amino-3(S)-methylρentyl]-N- (cyclopropylmethyl)glycyl-methionine, N-[2(S)-(Imidazol-4-yl-acetyl)ammo-3 (S)-methylpentylj-N-
(cyclopropylmethy^glycyl-methionine methylester,
N-[2(S)-(Imidazol-4-yl-acetyl)amino-3(S)-methylpentyl]-N-
(cyclopropylmethyl)glycyl-methionine,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(2,3- dihydrobenzofuran-7-yl-methyl)glycyl-methionine methyl ester, N-[2(S)-(L-ρyroglutatnyl)amino-3(S)-methylpentyl]-N-(2,3- dihydrobenzofuran-7-yl-methyl)glycyl-methionine,
2(S)-[2(S)-N-(L-PyiOglutamyl)-N-methylamino-3(S)-methylpentyloxy]-3- phenylpropionyl-methionine methyl ester, 2(S)-[2(S)-N-(L-Pyroglutamyl)-N-methylamino-3(S)-methylpentyloxy]-3- phenylpropionyl-methionine,
N-[2(S)-(L-ρyroglutamyl)amino-3(S)-N-ethylρentyl]-N-(l- naphthylmethyl)glycyl-O-methylserine methyl ester,
N-[2(S)-(L-pyroglutamyl)arnino-3(S)-methylperityl]-N-(l- naphthylmethyl)glycyl-O-methylserine,
N-(l-Naρhthylmethyl)-N-[2(S)-(N'-(L-pyroglutamyl)-N'-methylamino)-3(S)- methylpentylj-glycyl-methionine methyl ester,
N-(l-Naρhthylmethyl)-N-[2(S)-(N'-(L-pyroglutamyl)-Nr-methylamino)-3(S)- methylpentyl]-glycyl-methionine, N-[I -(Pyroglutamylamino)cyclopent- 1 -yl-methyl] -N-(I- naphthylmethyl)glycyl-methionine methyl ester,
N- [ 1 -(Pyroglutamylamino)-cyclopent- 1 -yl-methyl] -N-(I- naphthylmethyl)glycyl-methionine,
N-[2(S)-(Pyridm-2-on-6-yl-carbonyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-methionine methyl ester,
N-[2(S)-(Pyridin-2-on-6-yl-carbonyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-methionine,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(3-chlorobenzyl)glycyl- methionine methyl ester, N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(3-chlorobenzyl)glycyl- methionine,
N-[2(S)-(L-ρyroglutamyI)amino-3(S)-methylρentyl]-N-(l- naphthyhnethyl)glycyl-O-methylhomoserine methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyhnethyl)glycyl-O-methylhomoserine, - Ill -
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(2,3- dimethylbenzyl)glycyl-methiordne methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(2,3- dimeth.ylbenzyl)glycyl-methionine9
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthylmethyl)glycyl-(2-thienyl)alaiιiαe methyl ester,
N-[2(S)-(L-pyroglutamyl)amino-3(S)-methylpentyl]-N-(l- naphthyhτiethyl)glycyl-(2-thienyl)alanine,
N-[2(S)-(pyrrolidin-2-on-l-yl)-3-methylbutanoyl]-isoleucyl-methionine,
N-[2(S)-(piperidin-2-on-l-yl)-3-methylbutanoyl]-isoleucyl-methionine, or the pharmaceutically acceptable salts or optical isomers thereof.
(aa) compounds of the following formulae, which compounds are also disclosed in U.S. Patent No. 5,260,465, incorporated herein by reference,
Figure imgf000112_0001
wherein:
X-X is: CH=CH (cis);
CH = CH (trans); or CH2CH2; R1 and R2 are each independently selected from: a) H; b) Ci-s alkyl; c) C1-5 alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted, with methyl, methoxy, halogen (Cl, Br3 F, ϊ) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R and R is hydrogen;
(bb) compounds of the following formula, which compounds are also disclosed in U.S. Patent No. 5,420,157, incorporated herein by reference,
Figure imgf000113_0001
wherein: . . .
R1 and R2 are each independently selected from: a) H; b) Ci-5 is alkyl; c) C1-5 is alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted with methyl, methoxy, halogen (Cl, Br, F, T) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R and R is hydrogen;
(cc) compounds of the following formulae, which compounds are also disclosed in U.S. Patent Nos. 5,245,061 and -5,350,867, incorporated herein by reference, wherein:
Figure imgf000114_0001
X-X is: CH=CH(cis); CH = CH (trans); or
CH2CH2; R1 and R2 are each independently selected from: a) H; b) Ci-5 alkyl; c) Ci-5 alkyl substituted with a member of the group consisting of: i) phenyl; ii) phenyl substituted with methyl, methoxy, halogen (Cl, Br, F, T) or hydroxy; or a pharmaceutically acceptable salt of a compound of formula (T) in which at least one of R1 and R2 is hydrogen;
(dd) compounds of the following formula, which compounds are also disclosed in PCT Publication No. WO 96/10037, incorporated herein by reference,
O2F
Figure imgf000114_0002
or the pharmaceutically acceptable salts, hydrates, esters or amides thereof, wherein: n is 0 to 4,
R1 and R3 independently are Ci-4 alkyl, substituted with substituents selected from the group consisting of: a) aryl, which is defined as phenyl or naphthyl5 unsubstituted or substituted with one, two, three or four substituents selected from the group consisting of: i) F5 ii) Cl, iϋ) Br, iv) nitro, v) cyano, vi) C1-8 alkoxy, vii) Ci-8 alkylthio, viiϊ) Ci-8 alkylsulfonyl, ix) sulfamoyl, or x) C1-8 alkyl; or b) heteroaryl, which is defined as indolyl, imidazolyl or pyridyl, unsubstituted or substituted with one, two, three or four substituents selected from the group consisting of: i) F, ϋ) Cl, iii) Br, iv) nitro, v) cyano, vi) Ci-8 alkoxy, vii) Ci-8 alkylthio, viii) Ci-8 alkylsulfonyl, ix) sulfamoyl, or x) Ci-8 alkyl; R2 is: Ci-6 alkyl, which is unsubstituted or substituted with a substituent selected from the group consisting of: a) unsubstituted or substituted aryl, as defined in R (a), b) unsubstituted or substituted heteroaryl, as defined in R!(b), c) C3-S cycloalkyl, d) C1-8 alkylthio, e) C1-S alkylsulfonyl., f) C1-8 alkoxy, or g) aryl Ci-8 alkyl sulfonyl; and R4 is H;
(ee) compounds of the following formula, which are also disclosed in U.S. Patent Nos. 5,298,655 and -5,362,906, incorporated herein by reference, .
Figure imgf000116_0001
wherein: X is CH2, CH(OH), C-O, CHCOR, CH(NH2), CH(NHCOR), O, S(0)P, NH5
NHCO' • o O
Il Il
- CNH -, - NH - P - ; OH
p is O, l or 2;
Y is PO3RR1 or CO2R; R is H, lower alkyl, or CH2CH2N+Me3A-; R1 is H, lower alkyl, or CH2CH2N+Me3A-;
A is a pharmaceutically acceptable anion; m is O, 1, 2, or 3; and n is 0, 1, 2, or 3;
(ff) compounds of the following formula, which compounds are also disclosed in PCT Publication No. WO 96/05169, incorporated herein by reference,
Figure imgf000117_0001
wherein each of
Figure imgf000117_0002
which are the same or different, is an aryl group or a heteroaromatic ring group; A is a C2-a saturated or unsaturated aliphatic hydrocarbon group which may have substituent(s) selected from the group consisting of a lower alkyl group, a hydroxyl group, a lower hydroxyalkyl group, a lower alkoxy group, a carboxyl group, a lower carboxyalkyl group, an aryl group and an aralkyl group; each of X and Y which are the same or different, is an oxygen atom, a sulfur atom, a carbonyl group or a group of the formula -CHRa- (wherein Ra is a hydrogen atom or a lower alkyl group) or -NRb (wherein Rb is a hydrogen atom or a lower alkyl group), or X and Y together represent a vinylene group or an ethynylene group; each of R , R 5 R 3 R and R which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, a lower alkyl group or a lower alkoxy group; each of R and R5 which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a nitro group, a cyano group, a carboxyl group, a lower alkoxycarbonyl group, a carbamoyl group, a lower alkylcarbamoyl group, a lower alkyl group, a lower hydroxyalkyl group, a lower fluόroalkyl group or a lower alkoxy group; R6 is a lower alkyl group; and R7 is a hydrogen atom or a lower alkyl group, provided that when one of X and Y is an oxygen atom, a sulfur atom or a group of the formula -NR (wherein Rb is as defined above), the other is a carbonyl group or a group of the formula -CHRa- (wherein Ra is as defined above);
(gg) compounds of the following formula, which compounds are also disclosed in PCT Publication No. WO 96/05168, incorporated herein by reference,
Figure imgf000118_0001
wherein each of
Figure imgf000118_0002
which are the same or different, is an aryl group or a heteroaromatic ring group; A is a C2-s saturated or unsaturated aliphatic hydrocarbon group which may have substituent(s) selected from the group consisting of a lower alkyl group, a hydroxyl group, a lower hydroxyalkyl group, a lower alkoxy group, a carboxyl group, a lower carboxyalkyl group, an aryl group and an aralkyl group; Q is a group of the formula -(CH2)m- (wherein m is an integer of from 1 to 6) or-(CH2)n-W(CH2)p- (wherein W is an oxygen atom, a sulfur atom, a vinylene group or an ethynylene group; and each of n and p which are the same or different, is an integer of from 0 to 3); R is a hydrogen atom, a halogen atom, a hydroxyl group, a lower alkyl group, a lower alkoxy group, or an aryl or heteroaromatic ring group which may have substituent(s) selected from the group consisting of a halogen atom, a lower alkyl
0 1 R group and a lower alkoxy group; each of R , R and R which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, a lower alkyl group or a lower alkoxy group; each of R3 and R4 which are the same or different, is a hydrogen atom, a halogen atom, a hydroxyl group, an amino group, a nitro group, a cyano group, a carboxyl group, a lower alkoxycarbonyl group, a carbamoyl group, a lower alkylcarbamoyl group, a lower alkyl group, a lower hydroxyalkyl group, a lower fluoroalkyl group or a lower alkoxy group; R is a lower alkyl group; and R is a hydrogen atom or a lower alkyl group; or the pharmaceutically acceptable salts thereof.
Specifically suitable compounds of the above type include the following: 3-Hydroxy-7,l l,15-trimethylhexadeca-6,10,14-trienoic acid,
[2-Oxo-6, 10, 14-trimethylpentadec-5,9, 13-trienyl]phosphonic acid, [2-Hydroxy-6,10,14-trimetliylpentadec 5,9,13-trienyl]phosphonic acid, [ 1 -Acetyl-4,8, 12-trimethylpentadeca-3,7, 11 -trienyl]phosphonic acid, [2-[(E,E)-3,7,l l-Trimethyl-2,6,10-dodecatrienylamino]-2-oxo- ethyljphosphonic acid,
[(E,E)-4,8, 12-Trimethyl-3,7, 11 -tridecatrienyl]thiomethyl-phosphonic acid, 3-[(E,E)-3,7,l l-Trimethyl-2,6,10-dodecatrienylamino]-3-oxo-propionic acid, [2-[(E,E)-3,7, 11 -Trimethyl-2,6, 10-dodecatrienylamino]-2- oxoethyljphosphonic acid monomethyl ester, [2-[(E,E)-3,7,l 1 -Trimethyl-2,6, 10-dodecatrienylamino]-l- oxomethyljphosphonic acid,
[l-Hydroxy-(E,E)-3,7,ll-trimethyl-2,6,10-dodecatrienyl]-phosphonic acid, [ 1 -Hydroxy-(E,E)-5 ,9,13 -trimethyl-4, 8 , 12-tetradecatrienyl]-phosphonic acid, [l-Hydroxy-(E,E)-4,8,12-trimethyl-3,7,ll-tridecatrienyl]-phosphonic acid, [2-Acetamido-(E,E)-4, 8, 12-trimethyl-3 ,7, 11 -tridecatrienyl]-phosphonic acid,
[2-Hydroxy-(E,E)-4,8, 12-trimethyl-3 ,7,1 l-tridecatrienyl]-phosphonic acid, N-((lRS,2RS,4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4-pentenyl)-N- (2-naphthylmethyl)-carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-2-(4-chlorophenyl)-l-methyl-5-(l-naplithyl)-4-pentenyl)-N- (2-naphthylmethyl)-carbamoylmetliyl succinic acid, N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)ρentyl)-N-(2S naphthylmethyl)carbamoylniethyl succinic acid5
N-((lRS,2RS)-2-(4-ch.lorophenyl)-l-methyl-4-(2-naphthoxy)butyl)-N(2- naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-4-(2-naphthyl)butyl)-N-(2- naphtliylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-6-(2-naphthyl)hexyl)-N-(2- naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-5-phenyl-4-pentynyl)-N-(2- naphthylmethyl)carbamoylmethyl succinic acid, N-((lRS,2RS,4E)-2-(4-methoxyphenyl> 1 -methyl-5-(2-naphthyl)-4-pentenyl)-
N-(2-naphthylmethyl)carbamoylmethyl succinic acid,
' N-((lRS,2RS,4E)-l-methyl-2-(4-methylphenyl)-5-(2-naphthyl)-4-ρentenyl)-N- (2-naphthyl-methyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-l-methyl-5-(2-naphthyl)-2-(4-nitrophenyl)-4-pentenyl)-N- (2-naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-2-(4-fluorophenyl)-l-methyl-5-(2-naphthyl)-4-pentenyl)-N- (2-naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-l-methyl-5-(2-naphtbyl)-2-(4-trifluoromethylphenyl)4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl succinic acid, N-((1RS,2RS,4E)-1 -methyl-5-(2-naphthyl)-2-phenyl-4-pentenyl)-N-(2- naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-l-methyl-2-(6-methyl-3-pyridyl)-5-(2-naphthyl)-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,6E)-2-(4-chlorophenyl)-l-raethyl-7-phenyl-6-heptenyl)-N-(2- naphthylmethyl)carbamoylmethyl succinic acid, N-((lRS,2RS,6E)-2-(4-chlorophenyl)-l-methyl-7-(2-naphthyl)-6-heptenyl)-N- (2-naphthylmethyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-2-(4-clilorophenyl)-l-metliyl-5-(2-naphthyl)-4-pentenyl)-N- (3-quinolylmethyl)carbamoylmethyl succinic acid, N-((lRS,2RS54E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4-pentenyl)-N-
(3,4-dijQuorobenzyl)carbamoylmethyl succinic acid,
N-(2-benzoxazolylmethyl)-N-((lRS,2RS,4E)-2-(4-chlorophenyl)-l-methyl-5- (2-naphthyl)-4-pentenyl)carbamoylmethyl succinic acid,
N-(2-benzo[b]tbienylmethyl)-N-((lRS,2RS,4E)-2-(4-chloiOphenyl)-l-methyl- 5-(2-naphthyl)-4-pentenyl)carbamoylmethyl succinic acid,
N-((lRS,2RS,4E)-l-methyl-2-(3,4-methylenedioxyphenyl)-5-(2-naphthyl)-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl succinic acid,
(2R*)-2-[N-((lS*,2S*,4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4- pentenyl)-N-(2-naphtliylmethyl)carbamoylmeth.yl] succinic acid, (2R*)-2-[N-((lR*,2R*,4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmetb.yl] succinic acid,
(2S *)-2-[N-((lR*,2R*,4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl] succinic acid,
(2S *)-2-[N-((lS*,2S*?4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4- pentenyl)-N-(2-naphtiiylnieth.yl)carbamoylnieth.yl] succinic acid,
5-[N-((lRS,2RS,4E)-2-(4-chlorophenyl)-l-methyl-5-(2-naphthyl)-4-pentenyl)- N-(2-naphthylmethyl)carbamoyl]pentanoic acid,
(2R*)-2-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naph1iιylmethyl)carbamoylinetb.yl]succinic acid,
(2R*)-2-[N-((lRS,2RS,4Z)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl]succinic acid,
(2R*)-2-[N-(2-benzo[b]furanylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)- 1 -methyl-2-(3,4-methylenedioxyphenyl)-4-pentenyl)carbamoylmethyl]succinate, (2R*)-2-[N^(2-benzo[b]tMenylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)- l-methyl-2-(334-methylenedioxyphenyl)-4-pentenyl)carbamoylmetliyl]succinic acid,
(2R*)-2-[N-[(lRS,2RS34E)-5-(2-benzoxazolyl)-2-(334- bis(methoxycarbonyl)phenyl)- 1 -methyl-4-pentenyl]-N-(2- naphthylmethyl)carbamoylmethyl] succinic acid,
(2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)- 2-(4-methoxycarbonylphenyl)-l-methyl-4-pentenyl)carbamoylmethyl]succinic acid5
(2R*)-2-[N-(2-benzo[b]furanylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)- 2-(4-methoxycarbonylphenyl)-l-methyl-4-pentenyl)carbainoylmethyl]succmic acid, (2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-
2-(4-cyanophenyl)- 1 -methyl-4-pentenyl)carbamoylmethyl] succinic acid,
(2R*)-2-[N-(5-benzo[b]tbienylmethyl)-N-((lRS,2RS,4E)-5-(2-benzoxazolyl)- 2-(4-methoxycarbonylphenyl)-l-methyl-4-pentenyl)carbamoylmethyl]succinic acid,
N-((lRS,2RS,4E)-5-(3-chloro-4-methylphenyl)-2-(4-chloroplienyl)-l-metliyl- 4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccmic acid,
N-((lRS,2RS,4Z)-5-(3-chloro-4-methylρhenyl)-2-(4-chloroρhenyl)-l-metliyl- 4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-((lRS,2RS,4E)-5-(2-benzo[b]furanyl)-2-(4-chlorophenyl)-l-methyl-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, N-((lRS,2RS,4Z)-5-(2-benzo[b]furanyl)-2-(4-chlorophenyl)-l-methyl-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-2-(4-chlorophenyl)-l-methyl-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-((lRS,2RS,4Z)-5-(2-benzoxazolyl)-2-(4-chlorophenyl)-l-methyl-4- pentenyl)-N-(2-naphthylmethyl)carbanioylmethyl succinic acid,
N-((lRS,2RS,4E)-5-(2-benzimidazolyl)-2-(4-chlorophenyl)-l-methyl-4- pentenyl)-N-(2-naphthylniethyl)carbamoylmethylsuccmic acid,
N-((lRS,2RS,4E)-2-(4-chlorophenyl)-l-methyl-5-(3,4- metb.ylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, N-((lRS,2RS,4E)-5-(2-benzothiazolyl)-2-(4-clilorophenyl)-l-methyl-4-' pentenyl)-N-(2-naphthylmethyl)carbampylmethylsuccinic acid,
N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-2-(4-cyanoplienyl)-l-metliyl-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmeth.ylsuccinic acid, 4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-l,2,3- butanetricarboxylic acid,
3-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl~2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmetliyl)carbamoyl]l,2,2- propanetricarboxylic acid,
(2S33R)-4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl]-3- carboxy-2-hydroxybutanoic acid,
4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3-carboxy-4- methoxybutanoic acid,
5-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-carboxy-3- carboxymethyl pentanoic acid, l-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-2-(4-methoxycarbonylphenyl)-l- methyl-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-l,2,3-propanetricarboxylic acid,
(3R*)-4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- methoxybutanoic acid, (3S*)-4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- methoxybutanoic acid,
N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-2-(4-carboxyphenyl)-l-methyl-4- pentenyl)-N-(2-naphthyknethyl)carbamoyhnethylsuccinic acid, N-((lRS,2RS,4E)-5~(2-benzσxazolyl)-l-methyl-2-(4-(N- methylcarbamoyl)phenyl)-4-pentenyl]-N-(2-naphthylmethyl)carbam acid,
(2R*)-2-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-2-(4-hydroxy-3- methoxyphenyl)- 1 -methyl-4-pentenyl)-N-(2-naphthylmethyl)carbamoylmeth.ylsuccinic acid,
N-((lRS,2RS,4E)-2-(4-hydroxymethylpb.enyl)-l-methyl-5-(2-naplitliyl)-4- pentenyl)-N-(2-naphthylmethyl)carbamoylmetb.ylsuccinic acid,
N-(IRS ,2RS54E)-2-(4-aminoρhenyl)-l-methyl-5-(2-napb.thyl)-4-ρentenyl)-N- (2-iiaphthylmethyl)carbamoylrnethylsuccinic acid, disodium (3RS,4RS)-4-[N-((lRS,2RS54E)-5-(2-benzoxazolyl)-l-methyl-2- (3,4-methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3-carboxy- 4-hyroxybutahoate, . . . .
N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)-5-oxotetrahydrofuran-2- carboxyamide, sodium 4-[N-((lRS,2Rs,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)]carbamoyl-4- hyroxybutanoate, . 4-[N-((lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-2- oxotetrahydrofuran-3-yl-acetic acid,
(2R*)-2-[N-((lR*,2R*,4E)-5-(2-benzoxazolyl)-2-(4-methoxycarbonylphenyl)- l-methyl-4-pentenyl)-N-(2-naphtiiylmethyl)carbamoylmethyl]succinic acid, (2R*)-2-[N-((l S*;2S*54E)-5-(2-benzoxazolyl)-2-(4-methoxycarbonylρhenyl)-
1 -methyl-4-pentenyl)-N-(2-naphthylmethyl)carbamoylmethyl]succinic acid,
(2R*)-2-[N-(2-benzo[b]tliienylmethyl)-N-(lS*,2S*,4B)-5-(2-benzoxazolyl)-l- methyl-2-(3,4-methylenedioxyphenyl)-4-pentenyl)carbamoylmethyl]succinic acid,
(2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-((lR*,2R*,4E)-5-(2-benzoxazolyl)- l-meώyl-2-(3,4-methylenedioxyphenyl)-4-pentenyl)carbamoylmetb.yl]succiiiic acid, (2R*)-2-[N-((lRS,2RS)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- metliylenedioxyphenyl)pentyl)-N-(2-na
(2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-((lRS,2RS)-5-(2-benzoxazolyl)-l- methyl-2-(3 ,4-meth.ylenedioxyphenyl)pentyl)carbamoylmethyl] succinic acid,
(2R*)-2-[N-((lR*,2R*)-5-(2-benzoxazolyl)-2-(4-methoxycarbonylρhenyl)-l- methylpen1yl)-N-(2-naphthylmethyl)carbamoylmethyl]succinic acid3 disodium (3S,4S)-4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2- (3,4-methylenedioxyphenyl)-4-pentenyl)-N-(2-naph1iiylmefeyl)carbamoyl]-3-carboxy- 4-hyroxybutanoate,
Figure imgf000125_0001
sodium (3S,45)-4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(334- methylenedioxyphenyl)-4-pentenyl)-N-(2-naρhthylrneth.yl)carbamoyl]-3- ethoxycarbonyl-4-hyroxybutanoate,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)- 1 -methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3-tert- butoxycarbonyl-4-hydroxy-3 -butenoic acid,
Figure imgf000125_0002
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydiOxy-3- niethoxycarbonyl-3-butenoic acid,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(354- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmetliyl)carbamoyl]-4-hydroxy-3- isopropoxycarbonyl-3-butenoic acid, 4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- cyclohexyloxycarbonyl-4-hydroxy-3 -butenoic acid,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydroxy-3- (2-methoxyethoxy)carbonyl-3-butenoic acid,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- benzyloxycarbonyl-4-hydroxy-3-butenoic acid,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphth.ylmethyl)carbamoyl]-3- cyclopentyloxycarbonyl-4-hydroxy-3-butenoic acid,
4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydroxy-3- (3-tetrahydrofuranyloxycarbonyl)-3-butenoic acid, 4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydroxy-3- (2-hydroxy- 1 -hydroxymethylethoxycarbonyl)-3 -butenoic acid,
3-aπyloxycarbonyl-4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-metb.yl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydroxy-3- butenoic acid, 4-[N-(( 1 R,2R?4E)-5-(2-benzoxazolyl)-2-(3 ,4-methylenedioxyphenyl)- 1 - methyl-4-pentenyl)-N-(2-naphthylmethyl)carbai-noyl]-3-carboxymetliylcarbonyl-4- hydroxy-3-butenoic acid,
5-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-metliyl-2-(334- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4- ethoxycarbonyl-5-hydroxy-4-pentenoic acid,
5-N-((lR,2R34E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4-methylenedioxyphenyl)- 4-pentenyl)-N-(2-naphthyknethyl)carbamoyl]-4-tert-butoxycarbonyl-5-hydroxy-4- pentenoic acid, 4-N-((lR,2R34E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4-methylenedioxyphenyl)-
4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-4-hydroxy-3-hydroxyinethyl-3-butenoic acid,
4-[N-((lRS,2RS,4E)-6-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-5-hexenyl)-N-(2-naplithylmethyl)carbainoyl]-3-tert- butoxycarbonyl-4~hydroxy-3-butenoic acid,
(2S*,3R*)-4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-l,2,3- butanetricarboxylic acid,
(2R*,3S*)-4-[N-((lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(3,4- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-l,2,3- butanetricarboxylic acid,
N-[(lRS,2RS)-l-methyl-2-(4-nitroρhenyl)-3-(5-(phenylcarbamoyl)-2- fiuyl)-propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-[(lRS,2RS)-3-(5-(3,4-dimethoxyphenylcarbamoyl)-2-furyl)-l-methyl-2-(4- nitrophenyl)propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-[(lRS,2RS)-3-(5-(2-hydroxyphenylcarbamoyl)-2-furyl)-l-methyl-2-(4- rdtrophenyl)propyl]-N-(2-naphthylinetb.yl)carbarnoylmethylsuccinic acid,
N-[(lRS,2RS)-l-methyl-3-(5-(N-methylphenylcarbamoyl)-2-furyl)-2-(4- nitrophenyl)propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, N-[(lRS,2RS)-l-metiiyl-2-(4-nitrophenyl)-3-(5-(3-pyridylcarbamoyl)-2-furyl)- propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, N-[(lRS,2RS)-l-methyl-2-(4-πitrophenyl)-3-(5-(4-pyridylcarbamoyl)-2-furyl)- propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-[(lRS,2RS)-l-methyl-2-(4-nitroρhenyl)-3-(5-(spyrimidinylcarbainoyl)-2- furyl)-propyl]-N-(2-naphthylmefliyl)carbamoylmethylsuccinic acid, N-[(lRS,2RS)-l-methyl-2-(4-nitrophenyl)-3-(5-(2-tbiazolylcarbainoyl)-furyl)- propyl] -N-(2-naphthylmethyl)carbamoylmethylsuccinic acid
N-[(lRS,2RS)-2<4-chlorophenyl)-l-methyl-3-(5-(phenylcarbamoyl)-2-foryl)- propyl] -N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-3-(3- phenylcarbamoylphenyl)propyl)-N-(2-naphthylmethyl)carbamoylmetliylsuccinic acid?
N-[(lRS,2RS)-2-(4-chlorophenyl)-l-methyl-3-(3-(phenylcarbamoyl)-5- isoxazolyl)-propyl]-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid,
N-[(lRS,2RS)-2-(4-chlorophenyl)-l-methyl-3-(4-(phenylcarbamoyl)-2- pyridy l)-propyl] -N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, (2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-[(lRS,2RS)-l-methyl-2-(3,4- methylenedioxyphenyl)-3-(5-(phenylcarbamoyl)-2-furyl)-propyl]carbamoylmethyl] succinic acid,
(2R*)-2-[N-(2-benzo[b]thienylmethyl)-N-[(lRS,2RS)-l-methyl-2-(3,4- methylenedioxyphenyl)-3-(5-(3-pyridylcarbamoyl)-2-furyl)propyl] carbamoylmethyl] succinic acid, monoρivaloyloxymethyl (2R:!:)-2-[N-(2-benzo[b)thienylmethyl)-N[(lRS,2RS)- 1 -methyl-2-(3 ,4-methylenedioxyphenyl)-3 -(5 -(pheny lcarbamoyl)-2- furyl)propyl] carbamoylmethyl] succinate
(2R*)-2-[N-((lRSJ2RS)-2-(4-methoxycarbonylphenyl)-l-methyl-3-(3- ρhenoxymetb.ylphenyl)propyl)-N-2-naρhthylmethyl)carbamoylmethyl]succinic acid,
(2R*)-2-[N-[(lRS,2RS)-2-(4-methoxycarbonylphenyl)-l-methyl-3-(3- (phenoxymethyl)-5-(l,2,4-oxadiazolyl))propyl]-N-(2- naphthylmeth.yl)carbamoylmethyl] succinic acid,
(2R*)-2-[N-[(lRS,2RS)-2-(4-methoxycarbonylphenyl)-l-methyl-3-((E)-3- styiylphenyl)ρropyl]-N-(2-naph1iLylmethyl)carbamoylrnethyl]succinic acid, (2R*)-2-[N-[(lRSi2RS)-2-(4-methoxycarbonylphenyl)-l-methyl-3-(3-(2- phenylethyl)phenyl)propyl]-N-(2-naphthylmethyl)carbamoylmethyl]succimc acid,
N-((lRS,2RS)-2-(4-chlorophenyl)-l-methyl-3-(4- phenyle1iiynylphenyl)propyl)-N-(2-naphthylmethyl)carbamoylmethylsuccinic acid, N-[(lRS,2RS)-2-(4-chlorophenyl)-l-methyl-3-((E)-3-styrylphenyl)piOPyl]-N-
(2-naphthylmethyl)carbamoylmethylsuccinic acid
N-((lRS,2RS)-2-(4-methoxycarbonylphenyl)-l-methyl-3-(5-phenoxymethyl-2- furyl)propyl)-N-(2-naphthylmetliyl)carbamoylmethylsuccinic acid,
4-[N-[(lRS,2RS)-l-methyl-2-(4-nitrophenyl)-3-(5-(plienylcarbamoyl)-2- furyl)propyl]-N-(2-naphthylmethyl)carbamoyl]-l,2,3-butanetricarboxylic acid, disodium (3RS,4RS)-3-carboxylate-4-hydroxy-4-[N-[(lRS,2RS)-l-methyl-2- (4-nitrophenyl)-3-(5-(phenylcarbamoyl)-2-furyl)propyl]-N-(2- naphthylmethyl)carbamoyl]butanoate, disodium (3RS,4RS)-3-carboxylate-4-hydroxy-4-[N-[(lRS,2RS)-l-methyl-2- (4-nitrophenyl)-3-(5-(phenylcarbamoyl)-2--furyl)propyl]-]Sf-(2- naphthylmetliyl)carbamoyl]butanoate,
3-tert-butoxycarbonyl-4-hydroxy-4-[N-[(lRS,2RS)-l-metliyl-2-(4- nitrophenyl)-3-(5-(phenylcarbamoyl)-2-furyl)propyl}-N-(2- naphthylmethyl)carbamoyl]-3-butenoic acid, 3-tert-butoxycarbonyl-4-hydroxy-4-[N-[(lRS,2RS)-l-metliyl-2-(3,4- methylenedioxyphenyl)-3-(5-(phenylcarbamoyl)-2-furyl)propyl]-N-(2- naphthylmethyl)carbamoyl]-3-butenoic acid,
3-tert-butoxycarbonyl-4-hydroxy-4-[N-((lRS,2RS)-l-methyl-2-(4- vnitrophenyl)-3-(3-phenoxymethylphenyl)propyl)-N-(2-Naphthylmethyl)carbamoyl]-3- butenoic acid,
4-hydroxy-3-methoxycarbonyl-4-[N-[(lRS32RS)-l-metliyl-2-(3,4- methylenedioxyphenyl)-3-(5-(ρhenylcarbamoyl)-2-furyl)propyl]-N-(2- naphthylmethyl)carbamoyl]-3-butenoic acid,
3-allyloxycarbonyl-4-hydroxy-4-[N-[(lRS,2RS)-l-methyl-2-(3,4- metibLylenedioxyphenyl)-3-(5-(phenylcarbamoyl)-2-fυryl)propyl]-N-(2- naphthylmethyl)carbamoyl]-3-butenoic acid, 5-hydroxy-4-isopropylcarbonyl-5-[N-[(lRS,2RS)-l-methyl-2-(4-niUoplienyl)- 3-(5-(phenylcarbamoyl)-2-furyl)propyl]-N-(2-naphthylmethyl)carbamoyl]-4-pentenoic acid,
3-tert-butoxycarbonyl-4-(N-(233-dichlorobenzyl)-N-[(lRS,2RS)-l-methyl-2- (4-miJophenyl)-3-(5-(phenylcarbamoyl)-2-furyl)propyl]carbamoyl]-4-liydroxy-3- butenoic acid, or a pharmaceutically acceptable salt or optical isomer thereof. A further embodiment of the specific farnesyl pyrophosphate-competitive inhibitors includes: disodium (3RS.4RS)-4-[N-l(lRS,2RS,4E)-5-(2-benzoxazolyl)-l-methyl-2- (3,4-methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3- carboxyl-4-hyroxybutanoate
Figure imgf000130_0001
and sodium 4-[N-l(lR,2R,4E)-5-(2-benzoxazolyl)-l-methyl-2-(334- methylenedioxyphenyl)-4-pentenyl)-N-(2-naphthylmethyl)carbamoyl]-3-tert- butoxycarbonyl-4-hydroxy-3-butenoate
Figure imgf000130_0002
Other faπiesyl transferaseinhibitor compounds that are suitable for use in the methods of the invention are disclosed in the following publications: European Patent Publication Nos. 0 537 008; and 0 540 782; PCT Patent Publication Nos. WO 94/1935; WO 95/12572; and WO 95/08546.
The inhibitor compounds suitable for use in the methods of the invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. Unless otherwise specified, named amino acids are understood to have the natural "L" stereoconfiguration. Further, inhibitor compounds suitable for use in the methods of the invention may have enol form and keto form tautomers, depending upon the form of its substituents. The compounds of the present invention includes such enol form and keto form isomers and their mixtures. Additionally, when a hydroxyl group is present at the γ or δ-position of the terminal carboxyl group or of a carboxyl group when such a carboxyl group or a lower carboxyalkyl group is present on the saturated or unsaturated aliphatic hydrocarbon group represented by A in the formulas (ff) and (gg), such a hydroxyl group and a carboxyl group may form an intramolecular ester i.e. a 5-membered or 6-membered lactone ring.
The following definitions apply to the above-discussed compounds, including those of the above general formulae (a) through (ee):
"Alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. "Cycloalkyl" is intended to include non-aromatic cyclic hydrocarbon groups having the specified number of carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. "Alkenyl" groups include those groups having the specified number of carbon atoms and having one or several double bonds. Examples of alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, isoprenyl, farnesyl, geranyl, geranylgeranyl and the like. As used herein, "aryl" is intended to include any stable monocyclic, bicyclic or tricyclic carbon ring(s) of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of aryl groups include phenyl, naphthyl, anthracenyl, biphenyl, tetrahydronaphthyl, indanyl, phenanthrenyl and the like. The term heterocycle or heterocyclic, as used herein, represents a stable 5 to 7 membered monocyclic or stable 8 to 11 membered bicyclic or stable 11-membered tricyclic heterocycle ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting ofN5 O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothio- pyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, 2- oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyridyl N-oxide, pyridonyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinolinyl N-oxide, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydro-quinolinyl, thiamorpholinyl, thiamoφholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl.
As used herein, the terms "substituted aryl", "substituted heterocycle" and "substituted cycloalkyl" are intended to include the cyclic group which is substituted with 1 or 2 substituents selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N(C1-C6 alkyl)2, NO2, CN, (Ci-C6 alkyl)O-,-OH, (Ci-C6 alkyl)S(O)m-, (Ci-C6 alkyl)C(O)NH-, H2N-C(NH)-, (Ci-C6 alkyl)C(O)-, (Ci-C6 alkyl)OC(O)-, N3, (Ci-C6 alkyl)OC(O)NR- and Ci-C20 alkyl. The following structure:
Figure imgf000133_0001
represents a cyclic amine moiety having 5 or 6 members in the ring, such a cyclic amine which may be optionally fused to a phenyl or cyclohexyl ring. Examples of such a cyclic amine moiety include, but are not limited to, the following specific structures:
Figure imgf000133_0002
It is also understood that substitution on the cyclic amine moiety by R2a, R2b, R a and R may be on different carbon atoms or on the same carbon atom.
When R2a and R2b,and R3 and R4 are combined to form (CH2O)5-, cyclic moieties are formed. Examples of such cyclic moieties include, but are not limited to:
Figure imgf000133_0003
When R5a and R5b are combined to form-(CH2)s-, cyclic hereinabove for R3 and R4 are formed, ha addition, such cyclic moieties may optionally include a heteroatom(s). Examples of such heteroatom-containing cyclic moieties include, but are not limited to:
Figure imgf000134_0001
As used herein, the phrase "nitrogen containing C4-C9 mono or bicyclic ring system wherein the non-nitrogen containing ring may be a C6 aromatic ring, a C5-C7 saturated ring or a heterocycle" which defines moiety "Q" includes but is not limited to the following ring systems:
V.' '
Figure imgf000134_0002
Figure imgf000135_0001
It is intended that the definition of any substituent or variable (e.g., R10, Z, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus, N(R1 °)2 represents-NHH, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on a particular inhibitor compounds suitable for use in the methods of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by known techniques.
The pharmaceutically acceptable salts of inhibitor compounds for use in the methods of the invention include known non-toxic salts, e.g. pharmaceutically acceptable inorganic or organic acids such as the following acids: hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric, acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenyl-acetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy- benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like. The pharmaceutically acceptable salts of inhibitor compounds for use in the methods of the invention can be synthesized from the corresponding inhibitor of this, invention which contain a basic moiety by conventional chemical methods. Generally, the salts are prepared by reacting the free base with stoichiometric amounts or with an excess of the .desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
The following definitions apply to compounds of the above general formulae (ff) through (gg): The aryl group means a phenyl group, a naphthyl group or an anthryl group. A phenyl group or a naphthyl group is preferred.
The heteroaromatic ring group means a 5-membered or 6-membered monocyclic aromatic heterocyclic group containing one or two heteroatoms, which are the same or different, selected from the group consisting of an oxygen atom, a nitrogen atom and a sulfur atom, or a fused aromatic heterocyclic group having such a monocyclic aromatic heterocyclic group fused with the above-mentioned aryl group or having the same or different such monocyclic aromatic heterocyclic groups fused with each other, which may, for example, be a pyrrolyl group, an imidazolyl group, a pyrazolyl group, a pyridyl group, a pyrazinyl group, a pyrimidinyl group, a pyridazinyl group, an oxazolyl group, an isoxazolyl group, a furyl group, a thienyl group, a thiazolyl group, an isothiazolyl group, an indolyl group, a benzofuranyl group, a benzothienyl group, a benzimidazolyl group, a benzoxazolyl group, a benzisoxazolyl group, a benzothiazolyl group, a benzisothiazolyl group, an indazolyl group, a purinyl group, a quinolyl group, an isoquinolyl group, a phthalazinyl group, a naphthylidinyl group, a quinoxalinyl group, a quinazolinyl group, a cinnolinyl group or a pteridinyl group. Among them, a furyl group, a thienyl group, a pyridyl group, a pyrimidinyl group, an oxazolyl group, an isoxazolyl group, a thiazolyl group, a benzofuranyl group, a benzothienyl group, a benzimidazolyl group, a benzoxazolyl group, a benzothiazolyl group or a quinolyl group is preferred. The lower alkyl group means a
C1-6 linear or branched alkyl group, which may, for example, be a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, a sec-butyl group, a tert-butyl group, a pentyl group or a hexyl group. Among them, a methyl group or an ethyl group is preferred. The lower hydroxyalkyl group means the above-mentioned lower alkyl group having a hydroxyl group, i.e. a Ci-6 hydroxyalkyl group, such as a hydroxymethyl group, a hydroxyethyl group, a hydroxypropyl group or a hydroxybutyl group. Among them, a hydroxymethyl group or a hydroxyethyl group is preferred. The lower alkoxy group means a C1-6 alkoxy or alkylenedioxy group, which may, for example, be a methoxy group, an ethoxy group, a propoxy group, an isopropoxy group, a butoxy group, a tert-butoxy group, a methylenedioxy group, an ethylenedioxy group or a trimethylenedioxy group. Among them, a methoxy group, an ethoxy group or a methylenedioxy group is preferred. The lower carboxyalkyl group means the above-mentioned lower alkyl group having a carboxyl group, i.e. a C1-7 carboxyalkyl group, such as a carboxymethyl group, a carboxyethyl group, a carboxypropyl group or a carboxybutyl group. Among them, a carboxymethyl group or a carboxyethyl group is preferred. The aralkyl group means the above-mentioned lower alkyl group having the above-mentioned aryl group, such as a benzyl group, a phenethyl group, a 3-phenylpropyl group, a 1-naphthylmethyl group, a 2-naphthylmethyl group or a l-(2- naphthyl)ethyl group. Among them, a benzyl group, a phenethyl group or a 2- naphthylmethyl group is preferred. The saturated aliphatic hydrocarbon group may, for example, be an ethylene group, a trimethylene group, a tetramethylene group, a pentamethylene group, a hexamethylene group, a heptamethylene group or an octamethylene group. For example, a trimethylene group, a tetramethylene group or a pentamethylene group is preferred.
The unsaturated aliphatic hydrocarbon group means an unsaturated aliphatic hydrocarbon group having one or more, preferably one or two double bonds, at optional position(s) on the carbon chain, which may, for example, be a vinylene group, apropenylene group, a 1-butenylene group, a 2-butenylene group, a 1,3- butadienylene group, a 1-pentenylene group, a 2-pentenylene group, a 1,3- pentadienylene group, a 1,4-pentadienylene group, a 1-hexenylene group, a 2- hexenylene group, a 3-hexenylene group, a 1,3-hexadienylene group, a 1,4- hexadienylene group, a 1,5-hexadienylene group, a 1,3,5-hexatrienylene group, a 1- heptenylene group, a 2-heptenylene group, a 3-heptenylene group, a 1,3- heptadienylene group, a 1,4-heptadienylene group, a 1,5-heptadienylene group, a 1,6- heptadienylene group, a 1,3,5-heptatrienylene group, a 1-octenylene group, a 2- octenylene group, a 3-octenylene group, a 4-octenylene group, a 1,3-octadienylene group, a 1,4-octadienylene group, a 1,5-octadienylene group, a 1,6-octadienylene group, a 1,7-octadienylene group, a 2,4-octadienylene group, a 2,5-octadienylene group, a 2,6-octadienylene group, a 3,5-octadienylene group, a 1,3,5-octatrienylene group, a 2,4,6-octatrienylene group or a 1,3,5,7-octatetraenylene group. Among them, a propenylene group, a 1-butenylene group, a 1,3-butadienylene group or a 1- pentenylene group is preferred. The halogen atom may be a fluorine atom, a chlorine atom, a bromine atom or an iodine atom. For example, a fluorine atom or a chlorine atom is preferred.
The lower alkoxycarbonyl group means a C1-7 alkoxycarbonyl group, such as a methoxycarbonyl group, an ethoxycarbonyl group, a propoxycarbonyl group, a butoxycarbonyl group or a tert-butoxycarbonyl group. Among them, a methoxycarbonyl group or an ethoxycarbonyl group is preferred. The lower alkylcarbamoyl group means a carbamoyl group mono-substituted or di-substituted by the above-mentioned lower alkyl group, such as a methylcarbamoyl group, an ethylcarbamoyl group, a dimethylcarbamoyl group or a diethylcarbamoyl group. The lower fluoroalkyl group means the above-mentioned lower alkyl group having fluorine atom(s), i.e. a C1-6 fluoroalkyl group, such as a fluoromethyl group, a difluoromethyl group, a trifluoromethyl group, a 1-fluoroethyl group, a 2-fluoroethyl group, a 2,2,2-trifluoroethyl group or a pentafluoroethyl group.
The salt of the compound of a formula (ff) or (gg) may be a pharmaceutically acceptable common salt, which may, for example, be a base- addition salt of the terminal carboxyl group or of a carboxyl group when R4 and/or R5 or R3 and/or R4 is a carboxyl group, or when a carboxyl group or a lower carboxyalkyl group is present on a saturated or unsaturated aliphatic hydrocarbon group represented by A in the formulas (ff) and (gg), or an acid-addition salt of an amino group when R and/or R or R3 and/or R4 is an amino group, or of a basic heteroaromatic ring when such a basic heteroaromatic ring is present. The base-addition salt may, for example, be an alkali metal salt such as a sodium salt or a potassium salt; an alkaline earth metal salt such as a calcium salt or a magnesium salt; an ammonium salt; or an organic amine salt such as a trimethylamine salt, a triethylamine salt, a dicyclohexylamine salt, an ethanolamine salt, a diethanolamine salt, a triethanolamine salt, a procaine salt or an N,N'- dibenzylethylenediamine salt. The acid-addition salt may, for example, be an inorganic acid salt such as a hydrochloride, a sulfate, a nitrate, a phosphate or a perchlorate; an organic acid salt such as a maleate, a fumarate, a tartrate, a citrate, an ascorbate or a trifluoroacetate; or a sulfonic acid salt such as a methanesulfonate, an isethionate, a benzenesulfonate or a p-toluenesulfonate.
The ester of a compound of the formula (ff) or (gg) means a pharmaceutically acceptable common ester of the terminal carboxyl group or of a carboxyl group when R and/or R or R and/or R is a carboxyl group, or when a carboxyl group or a lower carboxyalkyl group is present on the saturated or unsaturated aliphatic hydrocarbon group represented by A in the formulas (ff) and (gg). It may, for example, be an ester with a lower alkyl group such as a methyl group, an ethyl group, a propyl group, an isopropyl group, a butyl group, a sec-butyl group, a tert-butyl group, a cyclopropyl group or a cyclopentyl group, an ester with an aralkyl group such as a benzyl group or a phenethyl group, an ester with a lower alkenyl group such as an allyl group or a 2- butenyl group, an ester with a lower alkoxyalkyl group such as a methoxymethyl group, a 2-methoxyethyl group or a 2-ethoxyethyl group, an ester with a lower alkanoyloxyalkyl group such as an acetoxymethyl group, a pivaloyloxymethyl group or a pivaloyloxyethyl group, an ester with a lower alkoxycarbonylalkyl group such as a memoxycarbonylmethyl group or an isopropoxycarbonylmethyl group, an ester with a lower carboxyalkyl group such as a carboxymethyl group, an ester with a lower alkoxycarbonyloxyalkyl group such as a l-(ethoxycarbonyloxy) ethyl group or a 1- (cyclohexyloxycarbonyloxy)ethyl group, an ester with an lower carbamoyloxyalkyl group such as a carbamoyloxymethyl group, an ester with a phthalidyl group, or an ester with a (5-substituted-2-oxo-l,3-dioxol-4-yl)methyl group such as a (5-methyl-2- oxo-l,3-dioxol-4yl)methyl group. While compounds having the precise structure of the formulae herein are preferred, as the terms are defined herein compounds coming within the formulae herein include structurally related compounds, including those compounds that are pharmaceutically acceptable salts, solvates, hydrates, and prodrugs of compounds delineated herein. Examples of prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing the parent compounds described herein (see Goodman and Gilman's, The Pharmacological basis of Therapeutics, 8th ed., McGraw-Hill, Int. Ed. 1992, "Biotransformation of Drugs").
Some of the inhibitor compounds useful in the methods of the invention can be synthesized from their constituent amino acids by conventional peptide synthesis techniques, and the additional methods described below. Standard methods of peptide synthesis are disclosed, for example, in the following works: Schroeder et al., The Peptides, Vol. 1, Academic Press 1965, or Bodanszky et al., Peptide Synthesis, Interscience Publishers, 1966, or McOmie (ed.) "Protective Groups in Organic Chemistry", Plenum Press, 1973, or Barany et al., "The Peptides: Analysis, Synthesis, Biology" 2, Chapter 1, Academic Press, 1980, or Stewart et al., "Solid Phase Peptide Synthesis", Second Edition, Pierce Chemical Company, 1984. Also useful in exemplifying syntheses of specific unnatural amino acid residues are European Patent Application No. 0 350 163 A2 (particularly page 51-52) and J.E. Baldwin et al., Tetrahedron, 50:5049-5066 (1994). With regards to the synthesis of the above discussed compounds containing a (β-acetylamino)alanine residue at the C-terminus, use of the commercially available N-Z-L-2,3-diaminopropionic acid (Fluka) as a starting material is preferred. In general, methods for preparation of the above discussed compounds are known in the art and disclosed e.g. in the above-mentioned publications. Detailed synthetic procedures are also disclosed in PCT/US96/11022. Useful FTase inhibitor compounds are also commercially available. Some FTase inhibitor compounds may have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. AU such isomeric forms of these compounds are expressly included in the present invention. The compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). AU such isomeric forms of such compounds are expressly included in the present invention. AU crystal forms of the compounds described herein are expressly included in the present invention.
It is believed that the compounds of the present invention provide effective therapy of latently infected cells (i.e. cells infected by a virus which is an immunodeficiency virus such as FIV, SIV, HTV, etc.) as evidenced by the induction of lytic replication in latently infected cells.
Hence, in one preferred embodiment the present invention can be used in treating those diagnosed as having AIDS as well as those having ARC, PGL and those seropositive but asymptomatic patients. For example, as a preventative, an effective amount of an FTase inhibitor compound can also be used prophylactically as a preventative for high risk individuals.
Compounds of the present invention can be used to treat cells, especially mammalian cells and in particular human cells, infected by an immunodeficiency virus such as HIV. As a result of treatment with compounds of the present invention the number of latently infected cells can be significantly reduced.
P24, a major structural protein (product of gag), has been widely used for monitoring HIV-I replication in cells and vireamia in individuals. Use of FTase inhibitor compounds in accordance with the present invention at concentrations that <lo not adversely affect cells, can dramatically reduce the number of cells latently infected with HIV3 e.g. preferably a reduction of HIV-I latently infected cells as shown by an increase (e.g., X-fold increase, where x is 2, 3, 4, 5, 10, 15, 20, 30, 40, or any integer between about 2 and about 50) in P24 levels in cells treated with test compound (e.g., farnesyl transferase inhibitors) relative to levels in untreated latent HIV-infected cells. In another aspect the increase can be determined relative to untreated uninfected cells.
The compounds of the present invention can be administered to HTV infected individuals or to individuals at high risk for HIV infection, for example, those having sexual relations with an HIV infected partner, intravenous drug users, etc. Because of their effect of inducing lytic viral replication, the compounds of the present invention and pharmaceutical compositions comprising one or more compounds of formula I can be used prophylactically as a method of prevention for such individuals to minimize their risk of cells becoming latently infected. The compound is adminstered in an effective amount as set forth below by methodology such as described herein.
Preferred compounds used in accordance with the present invention may induce lytic replication of HIV-I LTR and HIV-I in latently infected cells. In particular, preferred compounds of the present inventionin a dose dependent fashion may cause latently infected cells to lyrically replicate. Moreover, such induction is provided with essentially no adverse effects on cell survival or cellular mRNA or total cellular RNA synthesis. Thus, it is believed compounds of the present invention will have utility in clearing latent infections of an HIV infection and other retroviral infections in cells and in a human, and (in preferred embodiments) to ultimately entirely clear virus from an infected subject.
The methods of the invention can further include administration of one or more antiviral agents to the subject, thus both depleting the latent cell reservoir and inducing viral lytic replication whereupon the cells in that state are subjected to and susceptible to the antiretroviral therapy, which controls viral proliferation. Preferably, for inducing lytic replication, one or more compounds of the invention is administered in an amount sufficient to activate viral lytic replication in at least about 25 percent of infected cells, more preferably an amount sufficient to induce lytic replication in at least about 50 percent of the infected cells and still more preferably induce lytic replication in at least about 75 percent of latently infected cells.
In general for the treatment of immunodeficiency viral infections, for example an HIV infection, a preferred effective dose of one or more therapeutic compounds can be readily determined based on known factors such as efficacy of the particular therepautic agent used, age, weight and gender of the patient, and the like. See dosage guidelines as set forth e.g. in Remington, The Science and Practice of Pharmacy, 20th Edition. For certain preferred dosages, a FTase inhibitor compound may be administered to a mammal (e.g. human) in the range 0.1 mg to 5g per kilogram body weight of recipient per day, more preferably in the range of 0.1 mg to 1,000 mg per kilogram body weight per day, and still more preferably in the range of 1 to 600 mg per kilogram of body weight per day. The desired dose is suitably administered once or several more sub-doses administered at appropriate intervals throughout the day, or other appropriate schedule.
Preferably a therapeutic compound (e.g., a compound of the formulae herein) used in accordance with the invention will be in an isolated form distinct as it may be naturally found and in a comparatively pure form, e.g., at least 85% by weight pure, more preferably at least 95% pure. For some treatments in accordance with the present invention, it may be desirable that administered compound of formula I be at least 98% or even greater than 99% pure. Such a material would be considered sterile for pharmaceutical purposes. Potential contaminants include side products that may result upon synthesis of a compound of the invention or materials that may be otherwise associated with the compound prior to its isolation and purification. The present compounds should preferably be sterile and pyrogen free. Purification techniques known in the art may be employed, for example chromatography. As used herein, the terms "subject" and "patient" are used interchangeably. As used herein, the terms "subject" and "subjects" refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, ape, monkey, or human), and more preferably a human, hi one embodiment, the subject is an immunocompromised or immunosuppressed mammal, preferably a human (e.g., an HIV infected patient). In another embodiment, the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat), hi a preferred embodiment, the subject is a human.
Administration of the compounds of the invention may be by any suitable route including oral, rectal, nasal, topical (including buccal and sublingual), vaginal andparenteral (including subcutaneous, intramuscular, intravenous and intradermal) with oral or parenteral being preferred. It will be appreciated that the preferred route may vary with, for example, the condition and age of the recipient.
The administered ingredients may be used in therapy in conjunction with other medicaments such as reverse transcriptase inhibitors such as dideoxynucleosides, e.g. zidovudine (AZT), 2',3'-dideoxymosine (ddT) and 2',3'-dideoxycytidine (ddC), lamivudine (3TC), stavudine (d4T), and TRIZIVIR (abacavir + zidovudine + lamivudine), nonnucleosides, e.g., efavirenz (DMP-266, DuPont Pharmaceuticals/Bristol Myers Squibb), nevirapine (Boehringer higleheim), and delaviridine (Pharmacia-Upjohn), TAT antagonists such as Ro 3-3335 and Ro 24- 7429, protease inhibitors, e.g., indinavir (Merck), ritonavir (Abbott), saquinavir (Hoffmann LaRoche), nelfmavir (Agouron Pharmaceuticals), 141 W94 (Glaxo- Wellcome), atazanavir (Bristol Myers Squibb), amprenavir (GlaxoSmithKline), fosamprenavir (GlaxoSmithKline), tipranavir (Boehringer higleheim), KALETRA (lopinavir + ritonavir, Abbott)and other agents such as 9-(2- hydroxyethoxymethyl)guanine (acyclovir), interferon, e.g., alpha-interferon, interleukin π, and phosphonoformate (Foscarnet) or in conjunction with other immune modulation agents or treatments including bone marrow or lymphocyte transplants or other medications such as levamisol or thymosin which would increase lymphocyte numbers and/or function as is appropriate. Additionally, a FTase inhibitor compound may be administered in coordination or conunction with an entry inhibitor e.g. T20 (enfuvirtide, Roche/Trimeris) or UK-427,857 (Pfizer). Because many of these drugs are directed to different targets, e.g., viral integration, it is anticipated that an additive or synergistic result will be obtained by this combination. Further, FTase inhibitors can be combined with the use of other viral lytic replication activators, for example, proteasome inhibitors or Egrl activators, as disclosed in U.S. provisional patent application Nos. 60/588,013 and 60/588,301, respectively, both filed on July 13, 2004 and incorporated herein by reference.
In one embodiment, one or more compounds of the formulae herein are used in conjunction with one or more therapeutic agents useful for treatment or prevention of HIV, a symptom associated with HTV infection, or other disease or disease symptom such as a secondary infection or unusual tumor such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus-related lymphomas among others, that can result in FUV immunocompromised subjects.
In certain embodiments of the invention, one or more FTase inhibitor compounds are used in conjunction with a standard HIV antiviral treatment regimen. This combination is advantageous in that the one or more FTase inhibitor compounds can activate latent HIV infected cells to replicate by stimulating lytic viral replication, thus making them susceptible to the co-administered standard HTV antiviral treatment regimens. In this manner, the latent or secondary reservoirs of HIV-infected cells are depleted through "controlled" activation (rather then serendipitous or uncontrolled activation), resulting in more complete elimination of virus, while controlling the spread of viral infection.
In certain embodiments, the treatment methods herein include administration of a so-called FJTV-drug "cocktail" or combination therapy, wherein a combination of reverse transcriptase inhibitor(s) and HTV protease inhibitor(s) is co-administered. In a preferred embodiment, a highly active anti-retroviral therapy (HAART) treatment regime is combined with treatment with an FTase inhibitor according to the invention.
In yet other embodiments, a combination therapy according to the invention includes administration of an FTase inhibitor together with an abl kinase inhibitor such as imatinib (use of imatinib for HIV treatment is described more fully in co- pending U.S. Provisional Patent Application No. 60/588,015, filed June 13, 2004).
In certain embodiments, the methods involve modulation of any gene that exhibits altered expression in chronically HlV-infected cells compared to uninfected parental cells, prior to induction into lytic replication. The methods herein can involve, or target, any of the genes listed in the tables herein. This modulation can be direct or indirect, that is, it can be by direct control of expression or binding activity of the target, or by indirect control of the expression or binding activity of the target. In any case, another aspect is modulation of viral replication activity of latent HIV- infected cells. In another aspect, the methods involve modulation of lyn, cdc42, MNDA, CEBP alpha orMeisl by administration of the compounds of the formulae herein.
The present invention includes use of both racemic mixtures and optically active stereoisomers of FTase inhibitor compounds.
One or more FTase inhibitor compounds may be administered alone, or as part of a pharmaceutical composition, comprising at least one FTase inhibitor compound together with one or more acceptable carriers thereof and optionally other therapeutic ingredients, including those therapeutic agents discussed above. The carrier(s) should be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
Compositions of the compounds of the invention (e.g., compounds of the formulae herein) used in combination with other compounds (e.g., reverse transcriptase inhibitors, protease inhibitors, and the like) maybe employed alone or in combination with acceptable carriers such as those described below. For the treatment of immunodeficiency viral infections, for example an HTV infection, a suitable effective dose of a compound in such a composition will be in the range of 1 to 5,000 mg per kilogram body weight of recipient per day, preferably in the range of 10 to ' 4,000 mg per kilogram body weight of recipient per day. When multiple compounds having complementary activity are administered together it is expected one can use the lower portion of these ranges (or even less).
Combinations of substiruents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds). The compounds delineated herein are commercially available or readily synthesized by one of ordinary skill in the art using methodology known in the art.
The compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. The formulations may conveniently be presented in unit dosage form, e.g., tablets and sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy.
Such methods include the step of bringing into association the to be administered ingredients with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers or both, and then if necessary shaping the product. Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion, or packed in liposomes and as a bolus, etc.
A tablet maybe made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein.
Compositions suitable for topical administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the ingredient to be administered in a suitable liquid carrier.
Compositions suitable for topical administration to the skin may be presented as ointments, creams, gels and pastes comprising one or more compounds of the present invention and a pharmaceutically acceptable carrier. A suitable topical delivery system is a transdermal patch containing the ingredient to be administered.
Compositions suitable for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate. Compositions suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size, for example, in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations wherein the carrier is a liquid, for administration, as for example, a nasal spray or as nasal drops, include aqueous or oily solutions of the active ingredient.
Compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Compositions suitable for parenteral administration include aqueous and non¬ aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the ' sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions maybe prepared from sterile powders, granules and tablets of the kind previously described.
It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.
The use of the term "or" is unless otherwise indicated, to be construed as being inclusive. That is, the recitation of A, B or C is meant include A, B or C each alone, or in any combination (e.g., A and B, B and C, A and C, and A and B and C) thereof. All documents mentioned herein (including patents, patent applications, and other references) are incorporated herein by reference.
The present invention is further illustrated by the following examples. These examples are provided to aid in the understanding of the invention and are not to be construed as limitations thereof.
KYAMPTP. 1
MATERIALS AND METHODS
Cells: ACH-2, A3.01, Jl.1, and Ul cells (refs. H5 15, 24, 25, 48) were obtained through the NTH AJDS Research and Reference Reagent Program, Division of AIDS, NIAJD, NJU. U-937-cells were obtained from American Type Culture Collection (Manassas, VA). ACH-2, Jl.1 and Ul are chronically infected cell lines harboring HIV-I LAV strain, while A3.01, Jurkat, and U-937 are the corresponding parental uninfected cell lines. Cells were grown in RPMI- 1640 (Jnvitrogen, San Diego, CA) with 10% fetal bovine serum (FBS, hivitrogen), 5% penicillin- streptomycin (Lrvitrogen), and 2mM glutamine (Invitrogen). Cells were maintained at a concentration of IxIO6 cells/ml in T- 175 flasks. Cell concentrations and cell viability were monitored throughout the experiment at all time points studied. Cells were induced by addition of 20 ng/niL of phorbol myristyl acetate (PMA or TPA, Sigma, St Louis, MO) for one hour, after which the cells were washed with phosphate buffered saline (PBS). Cells were harvested by centrifugation at 1000 rpm for 10 minutes, at the following times after induction, 0.5, 3, 6, 8, 12, 18, 24, 48, 72, and 96 hour(s). HIV-infected and uninfected cells maintained and harvested in parallel with the PMA-treated cells but not induced with PMA were also harvested. For the time course experiment, 3'-azido- 3'-deoxythymidine (AZT, Sigma) was not added to the ACH-2 or A3.01 cells, in order to keep conditions as close to an acute infection as allowed by the experimental model. Harvested cells were washed thrice with ice-cold PBS to remove media; cell pellets were snap frozen using ethanol-dry ice mixture and stored at -8O0C for subsequent RNA extraction. Three independent time course experiments (biological replicates) were performed using the protocols described above to ensure reproducibility.
To compare expression profiles of chronically infected cell lines, ACH-2, Ul, and Jl.1 and their uninfected parental cell lines, A3.01, U-937 and Jurkat cells . respectively, were grown under identical conditions but in presence of AZT (250 nM) in growth media and cells were harvested as described above. In these studies, no inducing agent was used in either chronically infected or uninfected parental cell lines so as to study changes in cellular gene expression cells latently infected with HTV and uninfected cells.
Flow Cytometry: To. confirm viral replication following PMA induction, we . measured the accumulation of intracellular p24 over a period of 48 hours by measuring cell populations labeled with anti-p24 FITC-labeled antibody by flow cytometry. Cells (ACH-2 and A3.01) were washed twice with ice-cold PBS and suspended in 50 μL ice-cold permeabilization buffer (BD Biosciences, San Jose, CA), and incubated at 40C in the dark for 30 minutes. The cells were fixed using the CytoFix/CytoPerm kit (BD Biosciences) and 5 μL KC57-FITC-labeled p24 antibody (Beckman Coulter), was added to detect intracellular p24. Labeling A3.01 samples with FITC-labeled p24 antibody served as controls to ensure that the parental cell line • did not show any p24 accumulation over samples labeled with FITC-labeled mouse IgGl (Immunotech, Hialeah, FL), which was used as isotype control. Following incubation on ice for 30 minutes in the dark, the cells were washed thrice with permeabilization buffer, and resuspended in 300 μL of permeabilization buffer and analyzed using a Becton Dickinson FACSCAN instrument (BD Biosciences) in conjunction with CellQuest software (BD Biosciences) for flow cytometric analysis.
Total RNA Extraction: Total RNA was extracted using RNEasy Midiprep Kits per manufacturer's protocol (Qiagen, Valencia, CA). RNA concentrations and purity were measured by spectrophotometry, RNA quality (absence of RNA degradation) was assessed by gel electrophoresis. RNA concentration was adjusted to the levels required for subsequent microarray experiment protocols by concentration in a SpeedVac (Savant Instruments, Holbrook, CA). RNA samples (6-7 μg/μL) were stored in 100 μL TE buffer at -8O0C.
Real Time RT-PCR Quantitation of Viral RNA and Cellular RNA: Quantitation of HTV viral mRNA was carried out by real-time PCR using an ABI 7000 instrument (Applied Biosystems (ABI), Foster City, CA). A housekeeping gene, glyceraldehyde phosphate dehydrogenase (GAPDH) whose expression was unchanged in ACH-2 cells before and after PMA, was used as a normalization control. RNA from the samples was subjected to DNase treatment to remove contaminating DNA, and the DNAse was inactivated using the DNase Free kit (Amersham Biosciences, Piscataway, NJ) according to manufacturer's protocols. 2 μg of RNA was reverse transcribed using the Taqman RT kit from ABI per manufacturer's specifications. Briefly, the reaction mixture (50 μl) was incubated at 650C for 5 minutes followed by 370C for 45 minutes, 940C for 5 minutes, and then cooled on ice. 1/40 th aliquots of the corresponding samples were then used in a real-time PCR reaction using Taqman probes labeled with FAM and TAMRA at the 5' and 3 ' ends respectively. Primer probe pairs were designed using PrimerExpress (ABI). The reactions were carried out in triplicate for each time point and the fold changes observed were normalized to GAPDH, for each time point. Melting curve analysis and an assessment of the efficiency of the PCR reactions over a given concentration range were performed to determine if any errors occurred during the reactions, for example, primer-dimer formation or poor priming effects. Real time PCR reactions were set up as described above using primers and probes specific for early (multiply spliced mRNA, MS HTV- 1) and late (unspliced mRNA, US HTV-I) respectively. The sequences for 5' and 3' primers to quantitate early mRNA were 5'CGAAGAGCTCATCAGAACAGTCAS' and 5'TTGGGAGGTGGGTCTGCTTS'. The sequence for the labeled probe was 5'CTTCTCTATCAAAGCAGACCCACCTCCS' which overlapped with the splice site of HTV-I Rev sequence. The sequence detection primers for unspliced. or late RNA were SK38 and SK39 from the HIV-I Gene Amplimer kit (ABI). A TAMRA labeled probe with sequence identical to SKl 9 (ABT) was used for Real time PCR quantitation of the late viral RNA species. Standards from the kit were diluted to calculate copy number of virus based on gag mRNA concentrations. Real time RT-PCR analysis was also carried out for selected cellular genes using gene specific primer probe pairs and Taqman detection primers. Fold differences in mRNA expression in uninduced ACH-2 samples and the corresponding A3.01 samples, was determined using the protocol described for quantitation of viral mRNA. Real time RT-PCR quantitation was performed for genes PSMC5,p44slO (proteasome subunits) Egi-1 (early growth response 1), HDACl (histone deacetylase I)5 NK4, EIF4, SFRS3, to confirm that these genes were differentially expressed in the latently infected ACH- 2 cells compared to the uninfected A3.01 parental cells. Primer-probe pairs specific for each gene were designed using PrimerExpress (ABI). The sequences for the detection primers and probes for each gene are available as supplemental data, Table Sl.
Microarray Studies: Total RNA obtained from induced chronically infected and corresponding uninfected parental cells were used for microarray experiments. For each time point, RNA from the induced chronically infected ACH-2 cells and RNA from the corresponding induced, uninfected A3.01 cells were compared to minimize effects due to PMA induction. Microarrays were obtained from the National Cancer Institute Microarray Facility, Advanced Technology Center (Gaithersburg, MD). The microarrays (Hs. Um'Gem2) contained 10,395 cDNA spots on each glass slide. The cDNAs were selected for spotting on the slides based on their known or probable involvement in oncogenesis, signal transduction, apoptosis, immune function, inflammatory pathways, cellular transport, transcription, protein translation and other important cellular functions. A number of expressed sequence tags (ESTs) from unknown genes homologous to known genes and cDNAs encoding housekeeping genes were also included in these gene sets. For each time point, 50 μg of total RNA from PMA induced ACH-2 cells and 70 μg of total RNA from PMA induced A3.01 cells was labeled with Cy-3-dUTP and Cy-5-dUTP respectively as previously described (34, 60). Higher amounts of RNA were used for Cy-5 labeling to minimize the disparities in dye incorporation. Each sample of RNA from PMA- induced, infected cells from a particular time point was compared with RNA from the corresponding PMA-induced, uninfected cells from the same time point for subsequent hybridization to the same array to ensure accurate comparisons and to eliminate inter-array variability. The labeled cDNAs were then combined and purified using MicroCon YM-30 (Millipore, Bedford, MA) spin column filters, to remove any unincorporated nucleotides. 8-10 μg each of Cot-1 DNA, (Boehringer Mannheim, Indianapolis, IN), yeast tRNA (Sigma) and polyA (Amersham Biosciences) were added to the reaction mixture and heated at 1000C for 1 minute. Hybridization of the labeled cDNA to the microarray was carried out at 650C overnight, followed by washes with IX SSC, 0.2X SSC and 0.05X SSC respectively. The slides were dried by centrifugation at 1000 rpm for 3 minutes and then scanned as described below. RNA samples from three identical but independently conducted time course experiments were tested. Microarray experiments were performed at least twice for each time point (technical replicates) of each experiment. We also compared AZT- treated ACH-2 cells to untreated ACH-2 cells to determine whether any differences in gene expression might be solely due to AZT.
Microarray experiments for each chronically infected cell line (Jl.1, Ul and ACH-2) were also conducted as per the protocol described above. Samples from eight independent experiments per cell line were used for studying the gene expression patterns in chronically infected cell lines. To compensate for dye labeling bias that might be due to differences in Cy5 and Cy3 labeling efficiency and preferential dye incorporation by some mRNA species, RNA from the same samples labeled with Cy5 (70 μg RNA) and Cy3 (50 μg RNA) were co-hybridized to the same array, scanned, and data were analyzed for all the cell lines studied, using identical filtering and statistical tests, and genes showing dye incorporation bias were eliminated from further analysis as described below.
Microarray Scanning and Data Analysis: The slides were scanned using an Axon GenePix 4000 scanner (Axon Instruments, Union City, CA). The photomultiplier tube values (PMT) were adjusted to obtain equivalent intensities at both wavelengths used, 635 nm and 532 nm for the Cy5 and Cy3 channels respectively. Image analysis was performed using GenePix analysis software (Axon Instruments) and data analysis was performed using the microArray Database (mAdb) system hosted by the Center for Information Technology and Center for Cancer Research at NTH (http://nciarray.nci.nih.gov). Each array was normalized using Lowess normalization (71). Normalization across arrays over the time course was not feasible since no particular time point could be established as a median for cellular gene expression data, since viral gene expression and associated host cell gene expression was different for different time points. For each time point, there were at least 6 data sets (2 technical replicates X 3 biological replicates). For each time period (0.5-8 hours, 12-24 hours, 48-96 hours post induction.), there were at least 18 datasets (at least 3 time points per time period). Only arrays that passed initial spot size and intensity criteria, and whose normalization ratio (between the two signal intensities) was close to one (0.85-1.15), were analyzed. Filtering criteria were as follows: a) For each spot, signal intensity must be at least twice that of the background intensity; b) Each gene must have values in at least 70% of the arrays; c). Each array must have values for at least 70% of the gene spots. Genes that showed dye labeling bias in a particular cell line after normalization were excluded from that gene set prior to further analysis. This was determined using a one sample t-test on mean log ratios for replicate arrays with the same sample labeled with both Cy3 and Cy5.
Statistical Analysis: Comparison of expression profiles of infected versus uninfected (both induced by PMA) cell lines at a given time point was performed using univariate parametric and multivariate permutation tests based on the one sample random variance t-statistic in BRB-ArrayTools (http://linus.nci.nih.gov/BRB- ArrayTools) (62). Since RNA for infected and uninfected cell lines corresponding to the same time point were paired and co-hybridized on the same array, inter-array sources of variation were minimized and differential expression could be detected by a statistically significant non-zero mean log-ratio in biologically independent replicates. All biological replicates that passed the filtering criteria described above were used in the analyses. Technical replicates were averaged. The random variance model enabled variance information to be shared across genes without assuming that all genes have the same variance (69). For comparison of expression for latently infected versus uninfected cell lines, significance was based on p < 0.001 for a parametric one-sample random variance t-test. For evaluation of differential expression between infected and uninfected cell lines at fixed times after induction, a multivariate permutation test based on the one-sample random variance t-statistic was used in which the proportion of false discoveries was limited to 0.10 with 90% confidence (36, 62). Hierarchical clustering analyses on the resulting data sets were done using the mAdb system as well as Cluster and TreeView software programs (Stanford University, CA). Since statistically significant gene classes need not necessarily indicate the biological relevance of genes in a particular class, pathway analysis of the various genes that showed significant differential expression was performed by utilizing analysis tools provided by the NIH mAdb database (http://nciarray.nci.nih.gov) and querying the database of the Cancer Genome Anatomy Project (CGAP), (http://cgap.nci.nih.gov/) with pathway information provided by KEGG (www.genome.ad.jp/kegg/) and Biocarta (www.biocarta.com) pathway databases. Gene ontology summary analyses, which allow for grouping of genes based on their molecular function were also performed to ensure that changes observed in our studies were not due to general dysregulation of all genes studied. Where possible, observed/expected ratio (O/E) for each functional class of genes was determined to ensure that observed changes in number of genes differentially expressed within a gene class were greater than that expected by chance (O/E > 1). This provided stringent thresholds for pathway and functional classification of the differentially expressed genes.
Latency Reactivation Studies: Cells (ACH-2, Jl.1 and Ul) were seeded at a concentration of 2xlO5 cells/mL in 24 well plates in a volume of 1 mL. L-744,832 (structure shown above; FTase inhibitor) was dissolved in sterile dimethylsulfoxide (DMSO) and further diluted with media to obtain the desired final concentrations. The final concentration of DMSO in contact with the cells was never greater than 0.001% at any dose tested. AZT, (250 nM) was added, to the chronically infected cells in order to inhibit p24 production that maybe caused due to low levels of actively replicating virus present along with the chronically infected cells and to ensure that any increases in p24 expression would be attributable to activation of latent provirus and not due to . subsequent amplification via additional rounds of viral replication. Cells were incubated with different concentrations of L-744,832 at 370C. 200 μL samples of cell supernatant were collected at 24 hours after treatment. Cells incubated with tumor necrosis factor alpha (TNF-alpha, 0.5 μg/mL), also in the presence of AZT, served as a positive control. Cells treated with AZT alone served as a negative control. Cells not treated with AZT were also examined. Samples were mixed with lysing buffer (10% Triton-X-100, Sigma) to inactivate virus and diluted 5-fold with sample diluent (1% bovine serum albumin, 0.2% Tween-20 in RPMI-1640). p24 expression was assayed by ELISA using HIV-I p24 antigen capture kits (ADDS Vaccine Program, ' Frederick, MD) per manufacturer's specifications. Briefly, plates were washed with plate wash buffer and samples were added in duplicate wells. The samples (100 μL) were incubated for 2 hours at 370C. The plates were washed, and rabbit anti-HIV p24 antibody was added at 1 :400 dilution. Following incubation for one hour, the plates were washed and goat anti-rabbit IgG peroxidase labeled antibody at 1 :300 dilution was added. The plates were incubated for one hour at 370C, followed by washing and addition of a two-component substrate. Substrate solution consisted of equal volumes of TMB peroxidase substrate and peroxidase solution B (Kirkegaard and Perry Laboratories, Gaithersburg, MD). Samples were incubated for 30 minutes at room temperature and reactions were stopped by addition of IN hydrochloric acid solution. The absorbance was measured at 450 nm using a SpectraMax250 spectrophotometer (Molecular Devices Corporation, Sunnyvale, CA). The samples were assayed in duplicate and experiments were performed at least thrice using independent cell samples.
KXAMPT P, ?,
We treated the ACH-2 chronically infected cell line with phorbol myristyl aceate (PMA of TPA) to trigger the initiation and completion of the lytic replication cycle. Virus production was confirmed by flow cytometry for the HIV late protein p24 (Figure 1). p24 production was low in the absence of PMA (8.2%) in ACH-2 cells not treated with AZT. A3.01, the parental uninfected cell line, did not show any p24 specific staining over isotype control (data not shown). At 6 hours post induction (pi), 62% of the ACH-2 cells showed p24 production, and from 12 hours pi. up to 96 hours p. L, nearly all cells were positive for p24 production, indicating that a lytic HTV infection was underway in essentially all the cells in the culture (Figure. 1). By 48 hours post induction, flow cytometry analysis showed high levels of p24 production, but with increased cell death due to cytopathic effects (data not shown).
Cell viability before and after induction of viral lytic replication was carefully monitored so as to ensure that changes in gene expression could be associated with the process of lytic replication and not due to excessive cell death due to HIV replication. From the start of PMA induction to up to 24 hours p.i., cell viability of induced ACH- 2 cells remained at similar levels to that of uninfected, induced parental A3.01cells and also to uninduced ACH-2 cells, indicating that cell death due to HTV lytic replication was not a significant factor in changes in gene expression for that period. Beyond 24 hours p.i., (48- 96 hr p.i.) cytopathic effects and increased cell death were observed in the induced ACH-2 cells (also confirmed by flow cytometry), indicating that changes in cellular gene expression during the 48- 96 hr p.i. may be attributed to a combination of viral replication as well as cellular mechanisms involved in cell death. RNA yields decreased for this time period, however RNA quality and purity were similar to the previous time points, suggesting that changes in cellular gene expression were not due to RNA degradation in these cells. Since nearly all cells undergoing lytic replication were positive for p24 antigen by 24 hours p.i. when cellular viability was unaffected, the changes in cellular gene expression from 0.5 hour p.i. up to 24 hours p.i. are most likely associated with the process of lytic replication in ACH-2 cells.
-RY A MPT /R 3 Viral mRNA Expression by Real Time RT-PCR
To confirm that production of early and late viral mRNA was underway in lyrically induced cells, and to determine relative proportions of early (multiply spliced; tat, rev and nef) and late (unspliced; gag, pol, env) HIV-I mRNA over the time course, we used real time RT-PCR to quantitate the message classes, ha our system, the multiply spliced mRNA expression increased after lytic induction and showed a maximum of 40 fold increase over uninduced ACH-2 cells at 8 hours post induction. Unspliced mRNA concentrations showed a gradual increase with a 148-fold increase at 18 hours post induction (Figure 2). The results indicate that viral RNA expression in our lyrically induced cells followed known kinetic expression patterns (6, 35). There was a clear distinction in the peak expression of the early and late viral mRNA indicating that viral RNA expression followed a discrete temporal pattern in these cells.
P.yAMPT.F.4
Effects on ACH-2 Cellular Gene Expression Before and After Induction
For the cellular gene expression studies, we compared the cellular gene expression pattern of ACH-2 with that of its uninfected parental line, A3.01. Both the infected and uninfected parental lines were subjected to identical PMA treatments and sampling procedures over a 96 hour time course, and RNA samples from three separate time course experiments were each tested in duplicate, hi all, 66 arrays were examined. The filtering criteria yielded 9122 analyzable gene spots out of the 10395 gene spots printed on each microarray. The expression of most cellular genes was similar in the uninfected and chronically infected cells and did not change during activation into a lytic infection cycle. We also performed experiments to detect any gene spots that may be false positive due to bias in dye labeling under our experimental conditions. A small set of genes that showed dye bias (43 genes), in our dye-labeling bias experiments were excluded from the data set. Data from technical and biological replicates for each time point, were normalized for each array as described in the Methods section. Statistical analyses to identify genes that were significantly differentially expressed were performed using univariate and multivariate random variance one sample t-test, using all the replicates that passed the filtering criteria. Based on the statistical analyses of the genes showing altered expression, 131 genes showed altered expression even prior to induction. 1740 of all the analyzable spots showed statistically significant (p < 0.001) altered gene expression at some point, either before induction or over the, entire period of the lytic replication cycle (Figure 3).
TtYAMPT .P. S
Cellular Gene Expression Profiles Before Induction and During Lytic Replication
The changes observed during lytic replication following induction occurred in an orderly, time dependent manner. To better appreciate the time-dependent changes in cellular gene expression that accompany the portion of the lytic replication cycle occurring after activation of the chronically infected cells by PMA, we grouped our observations into three time periods after induction, a) Early (0.5-8 hours pi), b) Intermediate (12-24 hours pi) and c) Late (48-96 hours pi), roughly corresponding to the times during lytic replication when the early and the late viral rnRNA peak, and the end of the lytic cycle. Statistical analyses and hierarchical clustering of the data also grouped the various cellular genes into these time periods (data not shown). The changes in gene expression observed during the late period, (48-96 hours pi.) cannot be solely associated with the process of lytic replication, since during this time period cells showed cytopathic effects. However, the data is presented in order to provide the most complete possible view of the cellular environment following lytic replication. Some genes show differential expression only during the early stages of lytic replication and return to levels observed in the uninfected cells, while certain other - genes are initially unaltered and show differential expression at later time points. Certain groups of genes show similar patterns of regulation following induction (Figure 3). IQ the early time period 1334 genes were differentially expressed. In the intermediate time period, 756 genes were differentially expressed. The late time period (48-96 hours pi) showed the least change with 566 genes exhibiting significant altered expression (p < 0.001). Many of the genes that were differentially expressed in the early time period also showed either similar or the opposite trend in their expression patterns during the other time periods, hence some genes were included in the analysis of both the time periods. A number of discrete patterns of gene regulation were observed. Several cellular genes showed distinct temporal expression patterns during the lytic replication cycle, an expected finding, but more interestingly, a smaller number of genes appeared to be differentially expressed in the latently infected ACH-2 cells compared to their parental, uninfected cells, even before induction of the lytic cycle. A total of 131 genes already showed significant change (p < 0.001) in their expression prior to induction. They included genes encoding transcription factors, components ofproteasom.es, factors that control immune function, apoptosis and other functional classes. For gene classes that were annotated in the gene ontology database (GO database, www.geneontology.org) (5), observed/expected ratio for the number of genes within a functional class that were differentially expressed was set at greater than one (O/E > 1), so as to eliminate functional classes where the number of genes differentially expressed was not greater than that randomly expected. However, not all genes that were significantly differentially expressed are annotated in the database. Extensive literature studies for functional significance and classification were conducted in such cases to ensure that important classes of genes were still included in the analyses. An abbreviated listing of the genes grouped according to known functions that were differentially expressed before induction is given in Table 1.
FYAMPTF. fi
Genes and Pathways Affected Prior to Induction
While a large number of pathways are altered during lytic replication, a fewer number of pathways are also altered prior to induction of a lytic replication cycle. This observation was interesting and is important for understanding the mechanisms involved in latency maintenance, hence we conducted further analyses on this data set. Pathways involved in cell-cell signaling, signal transduction, inhibition of T-cell receptor signaling, protein translation, and cell cycle transition/regulation show altered expression prior to induction. Most notably, a number of genes encoding different subunits of proteasomes, including those that constitute the 20S/26S core complex (PSMB4, PSMA6, PSMA5) as well as regulatory subunits like PSMD 13 (1 IS regulatory subunit) were up regulated prior to induction. PSMB4 has peptidase activity, which is inhibited by Tat during viral replication. Tat competes with the 1 IS regulatory subunit, for binding to the 2OS core complex due to presence of a common binding site in Tat and the HS regulator alpha subunit (32, 59). Proteasomes are also involved in processing certain regions of HIV-I Nef preferentially, which leads to production of Nef-speciflc CTLs (cytotoxic T-lymphocytes) (44). Many other classes of genes encoding immune response modulators, integrins, cell cycle modulators (such as Egrl), nuclear import factors, and G-protein signaling molecules were also differentially expressed. A listing of genes that were differentially expressed prior to induction, based on their functional classification is given (Table 1). A list of pathways that were affected in the uninduced, chronically infected cells is given (supplemental data, Table S2) .
Real time RT PCR analysis was performed on a selected set of cellular genes using gene specific sequence detection primers and probes to determine if microarray results correlated with the actual normalized fold differences in the corresponding ACH-2 and A3.01 samples, prior to induction. For genes that were found to be statistically and/or biologically significant, the fold difference in expression levels was confirmed by their RT-PCR quantitation (supplemental data, Table S3).
BY A MPT .F. 7
Trends Seen in Pathway Profiles During Lytic Replication
While a number of genes that had altered expression during lytic replication could be classified based on their functionality, an alternate method was also used to determine if the genes that showed differential expression grouped into known cellular pathways. Genes encoding components of several distinct pathways were regulated in a coordinated fashion during lytic viral replication. Of the 1740 genes that were . differentially expressed over the lytic infection cycle in ACH-2 cells, 697 genes were assigned to various known pathways using the CGAP pathway databases, used in conjunction with the microArray database (mAdb) data analysis tools (http://nciarray.nci.nih.gov). Based on the information in the database, these genes were found to lie in a total of 385 known pathways. The remainder of the genes could not be classified into any known pathways listed in the CGAP database. The principal pathways affected over the time course and classified based on the number of genes involved in a particular pathway that are differentially expressed are shown in Figure 4 (and in supplemental data, Table S2). We observed that a number of pathways involved in signaling, cell cycle, and transcription showed maximum changes even prior to induction. During the early stage post induction (0.5- 8 hours p.i.), a number of metabolic pathways and signaling pathways show similar patterns. The intermediate stage (12-24 hours p.i) showed maximal changes in pathways involved in immune response modulation and cell survival. These patterns correlate well with the levels of early (multiply spliced) and late (unspliced) HTV protein gene expression and known viral replication effects on the host 0611(32, 44, 59).
RyAMPT, F, 8
Microarray Analysis of Chronically Infected Cell Lines
Our initial experiments with the ACH-2 cell line were aimed at studying the changes in cellular host gene expression profiles during a lytic infection following activation. However, our results showed that even prior to induction, many cellular genes were differentially expressed. This led us to study cellular gene expression in other chronically infected cell lines, in order to assess whether other chronically infected cell lines also showed alterations in cellular gene expression in the absence of active viral replication and if so, whether different cell lines showed similar patterns of altered gene expression. Different chronically infected HTV cell lines including J 1.1, a chronically infected T-lymphocytic cell line derived from Jurkat cells and Ul, a promonocyte chronically infected cell line derived from U937 cells, were studied using microarrays to determine the similarities and differences in their expression profiles. The p24 expression in all the latently infected cell lines was below lng/mL (0.2-0.8 ng/mL) indicating that the cells were not lytically active at the time of harvesting the cells. Experiments were performed on eight independent cell samples for each cell line and similar parameters were applied for filter criteria, gene selection and statistical analysis as with the ACH-2 cell line, as described in the Methods section, hi these sets of experiments, 24 arrays were analyzed and 8902 of the 10395 gene spots passed the selection criteria. Statistical analysis of expression ratios of AZT treated ACH-2 cells and untreated ACH-2 cells over their respective A3.01 controls did not show any significant differences in gene expression profiles, indicating that changes in gene expression were not due to low levels of actively replicating viral population (data not shown). Genes that had shown differential dye incorporation in our dye labeling bias experiments were excluded from each cell line data (43 genes in ACH-2, 18 genes in Jurkat, and 22 genes in U937 cell lines). Upon analyzing the resulting datasets, we found that 131 genes were differentially expressed in ACH-2, 65 genes were differentially expressed in Jl.1 and 155 genes were differentially expressed in Ul cells compared to their respective uninfected AZT treated parental cell lines. While stringent statistical thresholds were used for genes that were differentially expressed in each cell line, we reasoned that if a gene showed up as significantly differentially expressed (p < 0.001) in at least one cell line, then the expression data for those genes in the other cell lines may be important, even if not found statistically significant for that cell line. We found that ACH-2 and Jl.1 were more similar in their profiles, which may be due to their common T-cell lineage as compared to Ul, which is a promonocytic cell line (Figure 5). '
KY A MPT .R 9
Gene and Pathway Profile Analysis of Chronically Infected Cell Lines
An analysis of the gene expression profiles showed a limited number of genes that changed similarly across the three cell lines tested (Figure 5). These genes include cdc42, Lyn, MNDA, CEBPalpha, Meisl and others, which were down regulated in all cell lines. Genes that showed up regulation, include those encoding BTGl, BTG3, CDTl, pinin, and many others. Cdc42 is critical for activation of Nef associated kinase (PAO)5 while Lyn is required for binding to the PXXP motif of HrV-Nef (43), (57). MNDA, CEBP alpha mAMeisl are all tightly clustered. The proteins encoded by these genes are known to be critical in the progress of certain leukemias (18, 54, 65), but have not been hitherto related to HTV" latency. Certain genes show similar differential expression in ACH-2 and Jl .1 but not in Ul cells. Also, some genes show opposite trends in the cell lines tested. For example, proteasome subunits are up regulated in ACH-2, while they are down regulated in Ul . A list of common pathways affected and some pathways that change selectively is given (supplemental data, Table S4). The list of differentially expressed genes common to all three cell lines is given along with their expression ratios (supplemental data, Table S5).
RXA MPT ,F. 10
Latency reactivation studies were conducted with J 1.1 cells treated with L- 744,832 as described above. Results are set forth in FIG. 6.
Table Sl: List of sequence detection primers and probe pairs for real time RT- PCR quantitation of selected genes.
Figure imgf000165_0001
Figure imgf000166_0001
Table Sl : List of sequence detection primers and probe pairs for real time RT- PCR quantitation of selected genes.
Sequence detection primers and labeled probes were designed by PrimerExpress software from Applied Biosystems (ABI). The primer and probe names indicate the position in the cDNA sequence. F= forward, R= reverse, T= Taqman probe.
Table S2: Pathways that changed significantly prior and post induction of ACH-2 cells.
Figure imgf000167_0001
Figure imgf000168_0001
Table S2: Pathways that changed significantly prior and post induction of ACH-2 cells.
List of pathways that show altered expression based upon the number of genes involved in the pathways that showed significant differential expression during the four stages of the time course studied: Uninduced, 0.5-8 hours post induction (pi.), 12-24 hour p.i., and 48-96 hours p.i. The values highlighted in each column indicate the pathways that were maximally altered during that time period. The pathway profiles that are shown in Figure 3 are included here, along with the respective pathway descriptions. During 48-96 hours p.i, not many pathways changed appreciably in relation to the other time periods, hence data for those pathways is not shown.
Table S3: Verification of differentially expressed gene expression levels by real time RT-PCR quantitation.
Figure imgf000169_0001
Table S3: Verification of differentially expressed gene expression levels by real time RT-PCR quantitation.
Comparison of fold change in gene expression in latently infected ACH-2 cells prior to PMA induction, as compared to gene expression in uninfected parental A3.01 cells prior to PMA induction, by microarray and real time RT-PCR. RT-PCR data for the selected genes was normalized to data for GAPDH for each cell line, before assessing fold change in ACH-2 cells with respect to A3.01 cells. Differential expression of the selected genes was confirmed by RT-PCR quantitation.
Table S4: Pathways that change significantly in three chronically infected cell lines.
Figure imgf000170_0001
Figure imgf000171_0001
Table S4: Pathways that changed significantly in three chronically infected cell lines.
List of common cellular pathways differentially expressed in three chronically infected cell lines, ACH-2, Ul, and Jl.1. Classification was performed using the CGAP pathway database. The pathways are classified on the basis of the number of genes that showed significant differential expression in the chronically infected cell line over the corresponding uninfected parental cell line. Bold italicized values indicate similar number of genes affected in the pathways across all the three cell lines. Values in bold indicate pathways which are affected in ACH-2 and Jl.1 but not appreciably in Ul. Values in italics indicate pathways affected in ACH-2 and Ul, but not in Jl.1. Values in normal text indicate pathways that show some change in Jl.1 and Ul but not in ACH-2 cells. Table S5: Genes that showed similar differential expression in all three chronically infected cell lines.
Figure imgf000172_0001
Figure imgf000173_0001
Table S5: Genes that showed similar differential expression in all three chronically infected cell lines.
Genes that were up regulated or down regulated in all three latently infected cell lines. Normalized gene expression ratios that passed the various filter criteria and were shown to be significant by univariate (p < 0.001) and multivariate T-test for each cell line are listed.
FXAMPT P. 1 1
Effect of the FTase inhibitor L-744832 on HIV latently infected Jurkat clones.
Different HTV latently infected cloned Jurkat-derived cell lines produced in our laboratory showed different maximal response to reactivation of latent HTV pro virus by 1 μM L-744832, with effects ranging from 18-250% of the effect caused by TNF- alpha across the latently infected cell lines (Figure 7).
The results show that L-744832 (lμM) causes reactivation of latent proviras in these latently infected cell lines, indicating that L-744832 may have broad activity in activating latently infected cells, although differently latently infected cells have different sensitivity to L-744832. Cellular viability was unaffected at the concentration tested.
PY AMPT P. 19.
Effect of the FTase inhibitor L-744832 on viral reactivation from aviremic patient samples. Aliquots of resting CD4+ lymphocytes from HIV patients treated with HAART who had sustained viral loads below the limits of detection were tested with different concentrations of L-744832 and assayed for p24 expression 5 days post drug addition. The results are shown in Figures 8 and 9. p24 levels are expressed as a percentage of p24 expression caused by TNF-alpha (a known reactivating agent). A dose-dependent increase in p24 expression was observed with L-744832, ranging from 150-400% of TNF alpha induced p24 expression levels for the sample from the 1-year aviremic patient and ranging from 1000-1580% of TNF alpha induced p24 expression levels for the sample from the 8-year aviremic patient.
A patient treated with HAART can have undetectable viral loads but presumably continues to have latent viral reservoirs. These data show that L-744832 has a strong and potent effect on viral reactivation in samples from patients aviremic for 1 year (Figure 8) and 8 years (Figure 9). This indicates that even after very long periods of undetetectable viral loads as measured by standard methods, latent viral reservoirs may be present in patient which respond to reactivation by reactivating agents, ending viral latency in these cells. Also, the dose at which reactivation was observed in the patient samples was about 10-fold lower than that observed in cell lines, indicating that a much lower concentration range of L-744832 (or other FTase inhibitors) may be sufficient to reactivate latent viral reservoirs from primary cells from patients. Moreover, the extent of reactivation was different among patient samples, suggesting that viral reactivation can vary under different cellular conditions.
The following specific references, also incorporated by reference, are indicated in the examples and discussion above by a number in parentheses.
REFERENCES
1. Adams, J. 2003. Potential for proteasome inhibition in the treatment of cancer. Drug Discov Today 8:307-15. 2. Adams, J. 2001. Proteasome inhibition in cancer: development of PS-341. Semin Oncol 28:613-9.
3. Adams, J. 2003. The proteasome: structure, function, and role in the cell. Cancer Treat Rev 29 Suppl 1:3-9.
4. Arendt, C. W., and D. R. Littman. 2001. HIV: master of the host cell. Genome Biol 2:Reviews 1030.
5. Ashburner, M., C. A. Ball, J. A. Blake, D. Botstein5 H. Butler, J. M. Cherry, A. P. Davis, K. Bolinski, S. S. Dwight, J. T. Eppig, M. A. Harris, D. P. Hill, L. Issel-Tarver, A. Kasarskis, S. Lewis, J. C. Matese, J. E. Richardson, M. Ringwald, G. M. Rubin, and G. Sherlock. 2000. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25:25-9.
6. Bagnarelli, P., A. Valenza, S. Menzo, R. Sampaolesi, P. E. Varaldo, L. Butini, M. Montroni, C. F. Perno, S. Aquaro, D. Mathez, J. Leibowitch, C. Balotta, and M. Clementi. 1996. Dynamics and modulation of human immunodeficiency virus type 1 transcripts in vitro and in vivo. J Virol 70:7603-13.
7. Biswas, N., V. Sanchez, and D. H. Spector. 2003. Human cytomegalovirus infection leads to accumulation of geminin and inhibition of the licensing of cellular DNA replication. J Virol 77:2369-76.
8. Blankson, J. N., D. Persaud, and R. F. Siliciano. 2002. The challenge of viral reservoirs in HIV-I infection. Annu Rev Med 53:557-93.
9. Brooks, D. G., D. H. Hamer, P. A. Arlen, L. Gao, G. Bristol, C. M. Kitchen, E. A. Berger, and J. A. Zack. 2003. Molecular characterization, reactivation, and depletion of latent HIV. Immunity 19:413-23. 10. Brooks, D. C5 and J. A. Zack. 2002. Effect of latent human immunodeficiency virus infection on cell surface phenotype. J Virol 76:1673- 81.
11. Buttke, T. M., and T. M. Folks. 1992. Complete replacement of membrane cholesterol with 4,4',14-trimethyl sterols in a human T cell line defective in lanosterol demethylation. J Biol Chem 267:8819-26.
12. Chang, Y. E., and L. A. Laimins. 2001. Interferon-inducible genes are major targets of human papillomavirus type 31: insights from microarray analysis. Dis Markers 17:139-42.
13. Chun, T. W., and A. S. Fauci. 1999. Latent reservoirs of HIV: obstacles to the eradication of virus. Proc Natl Acad Sci U S A 96:10958-61.
14. Chun, T. W., L. Stuyver, S. B. Mizell, L. A. Ehler, J. A. Mican, M.
Baseler, A. L. Lloyd, M. A. Nowak, and A. S. Fauci. 1997. Presence of an inducible HTV-I latent reservoir during highly active antiretro viral therapy. Proc Natl Acad Sci U S A 94: 13193-7.
15. Clouse, K. A., D. Powell, L Washington, G. PoIi, K. Strebel, W. Farrar, P. Barstad, J. Kovacs, A. S. Fauci, and T. M. Folks. 1989. Monokine regulation of human immunodeficiency virus- 1 expression in a chronically infected human T cell clone. J Immunol 142:431-8.
16. Corbeil, J., D. Sheeter, D. Genini, S. Rought, L. Leoni, P. Du, M. Ferguson, D. R. Masys, J. B. Welsh, J. L. Fink, R. Sasik, D. Huang, J. Drenkow, D. D. Richman, and T. Gingeras. 2001. Temporal gene regulation during HIV-I infection of human CD4+ T cells. Genome Res 11:1198-204.
17. Coull, J. J., F. Romerio, J. M. Sun, J. L. Volker, K. M. Galvin, J. R. Davie, Y. Shi, U. Hansen, and D. M. Margolis. 2000. The human factors YYl and LSF repress the human immunodeficiency virus type 1 long terminal repeat via recruitment of histone deacetylase 1. J Virol 74:6790-9. 18. Cousar, J. B.? and R. C. Briggs. 1990. Expression of human myeloid cell nuclear differentiation antigen (MND A) in acute leukemias. Leuk Res 14:915- 20.
19. Craiu5 A., M. Gacgynska, T. Akopian, C. F. Gramma, G. Fenteany, A. L. Goldberg, and K. L. Rock. 1997. Lactacystin and clasto-Lactacystin beta - Lactone Modify Multiple Proteasome beta -Subunits and Inhibit Intracellular Protein Degradation and Major Histocompatibility Complex Class I Antigen Presentation. J. Biol. Chem. 272:13437-13445.
20. de Ia Fuente, C, F. Santiago, L. Deng, C. Eadie, i. Zilberman, K. Kehn, A. Maddukurij S. Baylor, K. Wu, C. G. Lee, A. Pumfery, and F. Kashanchi. 2002. Gene expression profile of HTV-I Tat expressing cells: a close interplay between proliferative and differentiation signals. BMC Biochem 3:14.
21. El Kharroubi, A., G. Piras, R. Zensen, and M. A. Martin. 1998. Transcriptional activation of the integrated chromatin-associated human rmmunodeficiency virus type 1 promoter. MoI Cell Biol 18:2535-44.
22. Emerman, M., and M. H. Malim. 1998. HIV-I regulatory/accessory genes: keys to unraveling viral and host cell biology. Science 280:1880-4.
23. Finzi, D., J. Blankson, J. D. Siliciano, J. B. Margolick, K. Chadwick, T. Pierson, K. Smith, J. Lisziewicz, F. Lori, C. Flexner, T. C. Quinn, R. E. Chaisson, E. Rosenberg, B. Walker, S. Gange, J. Gallant, and R. F. Siliciano. 1999. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HTV-I, even in patients on effective combination therapy. Nat Med 5:512-7.
24. Folks, T. M., K. A. Clouse, J. Justement, A. Rabson, E. Dun, J. H. Kehrl, and A. S. Fauci. 1989. Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci U S A. 86:2365-8. 25. Folks, T. M., J. Justement, A. IOnter, C. A. Bmarello5 and A. S. Fauci. 1987. Cytokine-induced expression of HTV-I in a chronically infected promonocyte cell line. Science 238:800-2.
26. FoIkS5 T. M., J. Justement, A. Kinter, 8. Schnittman, J. Orenstein? G. PoIi, and A. S. Fauci. 1988. Characterization of a promonocyte clone chronically infected with HTV and inducible by 13-phorbol-12-myristate acetate. J Immunol 140:1117-22.
, 27. Fukushi, M., M. Higuchi, M. Oie, T. Tetsuka, F. Kasolo, K. Ichiyama, N. Yamamoto, H. Katano, T. Sata, and M. Fujii. 2003. Latency-associated nuclear antigen of Kaposi's sarcoma-associated herpesvirus interacts with human myeloid cell nuclear differentiation antigen induced by interferon alpha. Virus Genes 27:237-47.
28. Gautier-Bert, K., B. Murol, A. S. Jarrousse, L. Ballut, S. Badaoui, F.
Petit, and H. P. Schmid. 2003. Substrate affinity and substrate specificity of proteasomes with RNase activity. MoI Biol Rep 30:1-7.
29. Geiss, G. K., R. E. Bumgarner, M. C. An, M. B. Agy, A. B. van 't Wout, E. Hammersmark, V. S. Carter, D. Upchurch, J. I. Mullins, and M. G. Katze. 2000. Large-scale monitoring of host cell gene expression during HTV- 1 infection using cDNA microarrays. Virology 266:8-16.
30. Graziosi, C, G. Pantaleo, L. Butini, J. F. Demarest, M. S. Saag, G. M. Shaw, and A. S. Fauci. 1993. Kinetics of human immunodeficiency virus type 1 (EHV-I) DNA and RNA synthesis during primary HTV-I infection. Proc Natl Acad Sci U S A 90:6405-9.
31. Ho, D. D., A. U. Neumann, A. S. Perelson, W. Chen, J. M. Leonard, and M. Markowitz. 1995. Rapid turnover of plasma virions and CD4 lymphocytes in HTV-I infection. Nature 373:123-6. 32. Huang, X.5 U. Seifβrt, U. Salzmann, P. Henklein5 R. Prβissner, W. Henke, A. J. SiJtS9 P. M. Kloefeel, and V/. DubieL 2002. The RTP site shared by the HIV-I Tat protein and the HS regulator subunit alpha is crucial for their effects on proteasome function including antigen processing. J MoI Biol 323:771-82.
33. Jones, J. ©., and A. M. Arvin. 2003. Microarray analysis of host cell gene transcription in response to varicella-zoster virus infection of human T cells and fibroblasts in vitro and SCIDhu skin xenografts in vivo. J Virol 77:1268- 80.
34. Khan, J.s R. Simon, M. Bittner, Y. Chen, S. B. Leighton, T. Pohida, P. D. Smith, Y. Jiang, G. C. Gooden, J. M. Trent, and P. S. Meltzer. 1998. Gene expression profiling of alveolar rhabdomyosarcoma with cDNA microarrays. Cancer Res 58:5009-13.
35. Kim, S., R. Byrn, J. Groopman, and D. Baltimore. 1989. Temporal aspects of DNA and RNA synthesis during human immunodeficiency virus infection: evidence for differential gene expression. J Virol 63:3708-3713.
36. Korn , E. L., J. F. Troendle, L. M. McShane, and R. Simon. 2004. Controlling the number of false discoveries: Application to high-dimensional genomic data. Journal of Statistical Planning and Inference In press.
37. Kramer, M. F., W. J. Cook, F. P. Roth, J. Zhu, H. Holman, D. M. Knipe, and D. M. Coen. 2003. Latent herpes simplex virus infection of sensory neurons alters neuronal gene expression. J Virol 77:9533-41.
38. Krones-Herzig, A., E. Adamson, and Dl Mercola. 2003. Early growth response 1 protein, an upstream gatekeeper of the p53 tumor suppressor, controls replicative senescence. Proc Natl Acad Sci U S A 100:3233-8.
39. Kundu, M., S. Sharma, A. De Luca, A. Giordano, J. Rappaport, K.
Khalili, and S. Amini. 1998. HIV-I Tat elongates the Gl phase and indirectly promotes HtV-I gene expression in cells of glial origin. J Biol Chem 273:8130-6.
40. Laughlin, M. A., S. Zeichner, D. Kolson, J. C. Alwine, T. Sβshamma,, R. J. Pomerante, and F, Gonzalez-Scarano. 1993. Sodium butyrate treatment of cells latently infected with HTV-I results in the expression of unspliced viral RNA. Virology 196:496-505.
41. Lifsøn, J. D., G. R. Reyes, M. S. McGrath, B. S. Stein, and E. G. Engleman. 1986. AIDS retrovirus induced cytopathology: giant cell formation and involvement of CD4 antigen. Science 232:1123-7.
42. Liu, C5 V. M. Rangnekar5 E. Adamson, and D. Mercola. 1998. Suppression of growth and transformation and induction of apoptosis by EGR- 1. Cancer Gene Ther 5:3-28.
43. Lu, X., X. Wu, A. Plemenitas, H. Yu, E. T. Sawai, A. Abo, and B. M. Peterlin. 1996. CDC42 and Racl are implicated in the activation of the Nef- associated kinase and replication of HTV-I. Curr Biol 6:1677-84.
44. Lucchiari-Hartz, M., V. Lindo, N. Hitziger, S. Gaedicke, L. Saveanu, P. M. van Endert, F. Greer, K. Eichmann, and G. Niedermann. 2003. Differential proteasomal processing of hydrophobic and hydrophilic protein regions: contribution to cytotoxic T lymphocyte epitope clustering in HTV-I- Nef. Proc Natl Acad Sci U S A 100:7755-60.
45. Lucchiari-Hartz, M., P. M. van Endert, G. Lauvau, R. Maier, A. Meyerhans, D. Mann, K. Eichmann, and G. Niedermann. 2000. Cytotoxic T lymphocyte epitopes of HTV-I Nef: Generation of multiple definitive major histocompatibility complex class I ligands by proteasomes. J Exp Med 191:239-52. 46. Mikovits, J. A., N. C. Lohrey, R. Schulof, J. Courtless, and F. W. Ruseetti. 1992. Activation of infectious virus from latent human immunodeficiency virus infection of monocytes in vivo. J Clin Invest 90:1486-91.
47. Nabel, C5 and D. Baltimore. 1987. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature
326:711-3.
48. Perez, V. L., T. Rowe, J. S. Justement, S. T. Butera, C. H. June, and T. M. Folks. 1991. An HTV-I -infected T cell clone defective in JL-2 production and Ca2+ mobilization after CD3 stimulation. J Immunol 147:3145-8.
49. Persaud, D., T. Pierson, C. Ruff, D. Finzi, K. R. Chadwick, J. B. Margolick, A. Ruff, N. Button, S. Ray, and R. F. Siliciano. 2000. A stable latent reservoir for HTV-I in resting CD4(+) T lymphocytes in infected children. J Clin Invest 105:995-1003.
50. Poon, B., K. Grovit-Ferbas, S. A. Stewart, and L S. Chen. 1998. Cell cycle arrest by Vpr in HTV-I virions and insensitivity to antiretroviral agents. Science 281:266-9.
51. Radhakrishnan, S., J. Otte, S. Enam, L. Del Valle, K. Khalili, and J. Gordon. 2003. JC virus-induced changes in cellular gene expression in primary human astrocytes. J Virol 77: 10638-44.
52. Ragione, F. D., V. Cucciolla, V. Criniti, S. Indaco, A. Borriello, and V. Zappia. 2003. p21Cipl gene expression is modulated by Egrl: a novel regulatory mechanism involved in the resveratrol antiproliferative effect. J Biol Chem 278:23360-8.
53. Ramratnam, B., J. E. Mittler, L. Zhang, D. Boden, A. Hurley, F. Fang, C. A. Macken, A. S. Perelson, M. Markowitz, and D. D. Ho. 2000. The decay of the latent reservoir of replication-competent HTV-I is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy. Nat Med 6:82-5.
54. Rozov§kaia5 T., E. Feinstem, O. Mor, R. Foa, J. Blechman, T. Nakamura, C. M. Croce, G. Cimino, and E. Canaaui. 2001. Upregulation of Meisl and HoxA9 in acute lymphocytic leukemias with the t(4 : 11) abnormality. Oncogene 20:874-8.
55. Ruff, C. T., S. C. Ray, P. Kwon? R. ZUm5 A. Pendleton, N. Button, R. Ashworth, S. Gange, T. C. Quinn, R. F. Silicianø, and D. Persaud. 2002. Persistence of wild-type virus and lack of temporal structure in the latent reservoir for human immunodeficiency virus type 1 in pediatric patients with extensive antiretroviral exposure. J Virol 76:9481-92.
56. Sakamoto, K. M., C. Bardeleben, K. E. Yates, M. A. Raines, D. W. Golde, and J. C. Gasson. 1991. 5! upstream sequence and genomic structure of the human primary response gene, EGR-1/TIS8. Oncogene 6:867-71.
57. Saksela, K., G. Cheng, and D. Baltimore. 1995. Proline-rich (PxxP) motifs in HΓV-1 Nef bind to SH3 domains of a subset of Src kinases and are required for the enhanced growth of Nef+ viruses but not for down-regulation of CD4. EMBO J 14:484-91.
58. Schwartz, O., V. Marechal, B. Friguet, F. Arenzana-Seisdedos, and J. M. Heard. 1998. Antiviral activity of the proteasome on incoming human immunodeficiency virus type 1. J Virol 72:3845-50.
59. Seeger, M., K. Ferrell, R. Frank, and W. Dubiel. 1997. HIV-I tat inhibits the 20 S proteasome and its 11 S regulator-mediated activation. J Biol Chem 272:8145-8.
60. Shaheduzzaman, S., V. Krishnan, A. Petrovic, M. Bittner, P. Meltzer, J. Trent, S. Venkatesan, and S. Zeichner. 2002. Effects of HTV-I Nef on cellular gene expression profiles. J Biomed Sci 9:82-96. 61. Simmons, A., V. Aluvihare, and A. McMichael. 2001. Nef triggers a transcriptional program in T cells imitating single-signal T cell activation and inducing HTV virulence mediators. Immunity 14:763-77.
62. Simon, R. M., E. L. Korn, L. M. McShane, M. D. Radmacher, G. M. Wright, and Y. Zhao. 2003. Design and Analysis of DNA Microarray Investigations- An Artificial Intelligence Milestone. Spinger Verlag, New York.
63. Song, Y. J,, and M. F. Stinski. 2002. Effect of the human cytomegalovirus IE86 protein on expression of E2F-responsive genes: a DNA microarray analysis. Proc Natl Acad Sci U S A 99:2836-41.
64. Sukhatme, V. P., X. M. Cao, L. C. Chang, C. H. Tsai-Morris, D. Stamenkovich, P. C. Ferreira, D. R. Cohen, S. A. Edwards, T. B. Shows,
T. Curran, and et al. 1988. A zinc finger-encoding gene coregulated with c- fos during growth and differentiation, and after cellular depolarization. Cell 53:37-43.
65. Tenen, D. G. 2001. Abnormalities of the CEBP alpha transcription factor: a major target in acute myeloid leukemia. Leukemia 15:688-9.
66. van 't Wout, A. B., G. K. Lehrman, S. A. Mikheeva, G. C. O'Keeffe, M. G. Katze, R. E. Bumgarner, G. K. Geiss, and J. I. Mullins. 2003. Cellular gene expression upon human immunodeficiency virus type 1 infection of CD4(+)- T-cell lines. J Virol 77:1392-402.
67. Van Lint, C, S. Emiliani, M. Ott, and E. Verdin. 1996. Transcriptional activation and chromatin remodeling of the HIV-I promoter in response to histone acetylation. EMBO J 15:1112-20.
68. Wiebusch, L., R. Uecker, and C. Hagemeier. 2003. Human cytomegalovirus prevents replication licensing by inhibiting MCM loading onto chromatin. EMBO Rep 4:42-6. 69. Wright, G. W., and R. M. Simon. 2003. A random variance model for detection of differential gene expression in small microarray experiments. Bioinformatics 19:2448-55.
70. Yang5 X., Y. Chen, and D. Gabuzda. 1999. EKEC MAP kinase links cytokine signals to activation of latent HIV-I infection by stimulating a cooperative interaction of AP-I and NF-kapρaB. J Biol Chem 274:27981-8.
71. Yang5 Y. H., S. Dudoit, P. Luu, D. M. Lin, V. Peng, J. Ngai, and T. P. Speed. 2002. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res 30:el5.
72. Zhu, H., J. P. Cong, G. Mamtora, T. Gingeras, and T. Shenk. 1998. Cellular gene expression altered by human cytomegalovirus: global monitoring with oligonucleotide arrays. Proc Natl Acad Sci U S A 95:14470-5.
See also V. Krishnan and SX. Zeichner, J. Virology, 78(17): 9458-9473 (2004).
All documents (including patents, patent applications and literature references) mentioned herein are incorporated herein by reference.
AU of the features disclosed in this specification maybe combined in any combination. Each feature disclosed in this specification may be replaced by an alternative feature serving the same, equivalent, or similar purpose. Thus, unless expressly stated otherwise, each feature disclosed is only an example of a generic series of equivalent or similar features.
From the above description, one skilled in the art can easily ascertain the essential characteristics of the present invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions. Thus, other embodiments are within the appended claims.

Claims

What is claimed is:
1. A method of treating HTV infection in a subject comprising identifying a subject as in need of reactivation of replication processes in latent HTV-infected cells; and administration of an effective amount of a farnesyl transferase (FTase) inhibitor to reactivate the replication process.
2. The method of claim 1 wherein the farnesyl transferase inhibitor is one or more of FTI277, L-744832, BMS214662, Rl 15777 and SCH66336.
3. The method of claim 1 wherein one or more peptidomimetic FTase inhibitor compounds are administered to the subject.
4. The method of claim 1 wherein one or more non-peptidomimetic FTase inhibitor compounds are administered to the subject.
5. The method of claim 1 wherein the one or more FTase inhibitor compounds are of any one of the general formulae (a) through (gg) above.
6. A method of any one of claims 1 or 2 wherein the administered FTase inhibitor compound has an IC50 of about 100 nM or less in a standard in vitro farnesyl transferase inhibition assay.
7. A method of inhibiting HIV replication in a cell or a subject comprising identifying a subject as in need of reactivation of replication processes in latent HTV-infected cells; administration of an effective amount of a farnesyl transferase (FTase) inhibitor to reactivate the replication process; and administration of one or more HIV antiviral agents to inhibit induced lytic viral replication.
8. A method of treating latently HTV-infected cells in a subject comprising administration to the cells one or more FTase inhibitor compounds.
9. A method of modulating lytic replication in an HTV-infected cell in a subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
10. The method of any of claims 7-9, wherein the cell is a lymphocytic cell.
11. The method of any of claims 7-9, wherein the cell is a monocytic cell.
12. The method of any one of claims 7-11 wherein the cells are human cells.
13. A method of reducing latent HIV reservoirs in an HTV-infected subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
14. A method of activating latent HTV-provirus in a cell in a subject identified as in need of such treatment comprising administration to the subject of an effective amount of one or more FTase inhibitor compounds.
15. The method of any of claims 1-14 further comprising administration of one or more additional anti-HIV therapeutic agents.
16. The method of claim 15 wherein the additional agent(s) are a reverse transcriptase inhibitor, a protease inhibitor, or combination thereof.
17. A method of increasing expression of p24 in a latently HTV-infected cell comprising administration to the cell of an effective amount of a farnesyl transferase inhibitor.
18. A method of screening for a compound capable of activating latent HIV- infected cells comprising contacting a FTase inhibitor test compound with an ACH-2 cell or Jl.1 cell or Ul cell and determining the level of p24 expression.
19. The method of claim 18, wherein increased expression of p24 in cells . treated with a test compound relative to non-treated cells indicates a compound capable of activating latent HIV-infected cells.
PCT/US2005/025176 2004-07-13 2005-07-13 Use of a farnesyl transferase inhibitor in the treatment of viral infections WO2006017369A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US58777104P 2004-07-13 2004-07-13
US60/587,771 2004-07-13

Publications (2)

Publication Number Publication Date
WO2006017369A2 true WO2006017369A2 (en) 2006-02-16
WO2006017369A3 WO2006017369A3 (en) 2006-11-23

Family

ID=35839815

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/025176 WO2006017369A2 (en) 2004-07-13 2005-07-13 Use of a farnesyl transferase inhibitor in the treatment of viral infections

Country Status (1)

Country Link
WO (1) WO2006017369A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999027929A1 (en) * 1997-12-04 1999-06-10 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5919780A (en) * 1995-06-16 1999-07-06 Warner Lambert Company Tricyclic inhibitors of protein farnesyltransferase
WO2002024686A2 (en) * 2000-09-25 2002-03-28 Janssen Pharmaceutica N.V. Farnesyl transferase inhibiting 6-heterocyclylmethyl quinoline and quinazoline derivatives

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919780A (en) * 1995-06-16 1999-07-06 Warner Lambert Company Tricyclic inhibitors of protein farnesyltransferase
WO1999027929A1 (en) * 1997-12-04 1999-06-10 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
WO2002024686A2 (en) * 2000-09-25 2002-03-28 Janssen Pharmaceutica N.V. Farnesyl transferase inhibiting 6-heterocyclylmethyl quinoline and quinazoline derivatives

Also Published As

Publication number Publication date
WO2006017369A3 (en) 2006-11-23

Similar Documents

Publication Publication Date Title
Liu et al. Cathepsin L-selective inhibitors: A potentially promising treatment for COVID-19 patients
DE69534549T2 (en) Combination of retroviral protease inhibitors
KR20080081358A (en) Methods for improving the pharmacokinetics of hiv integrase inhibitors
WO2008097654A1 (en) Methods of using saha for treating hiv infection
Desimio et al. The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity
US20160143884A1 (en) Use of tetrahydroindazolylbenzamide and tetrahydroindolylbenzamide derivatives for the treatment of human immunodeficiency virus (hiv) and acquired immune deficiency syndrome (aids)
KR20170105113A (en) Compositions and methods for inhibiting viral infection
US20050256181A1 (en) Treating a viral disorder
WO2021071809A1 (en) Dosages for hdac treatment with reduced side effects
US6448245B1 (en) Methods of and compounds for inhibiting calpains
WO2018049400A1 (en) Use of atr and chk1 inhibitor compounds
US20160151365A1 (en) Trizol-1-ol analogs anti-retroviral latency drugs
US11666575B2 (en) Pyrazolo[1,5]pyrimidine-based compounds and methods of their use to treat viral infections
US11730723B2 (en) Compounds and methods for treating viral infections
WO2010099169A2 (en) A novel in vitro hiv-1 latency model for screening reactivation agents of hiv-1
US9352010B2 (en) Treatment of HIV-1 infection and AIDS
WO2006017346A1 (en) Treatment of viral infections by means o proteasome inhibitors
WO2006019841A2 (en) Treatment of viral infections with egr 1 activators
WO2011139636A1 (en) Small molecule inhibitors of functions of the hiv-1 matrix protein
WO2006017369A2 (en) Use of a farnesyl transferase inhibitor in the treatment of viral infections
WO2006017353A2 (en) Treatment of viral infections
CA2881967C (en) Inhibitors of protein phosphatase-1 and uses thereof
JPH10504036A (en) Combinations of HIV protease inhibitors
CZ269699A3 (en) Quinoxalines in treble combination with protease inhibitors and inhibitors of reversible transcriptase as medicaments for treating AIDS

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase