WO2011088549A1 - Derivatives of pyridoxine for inhibiting hiv integrase - Google Patents

Derivatives of pyridoxine for inhibiting hiv integrase Download PDF

Info

Publication number
WO2011088549A1
WO2011088549A1 PCT/CA2011/000041 CA2011000041W WO2011088549A1 WO 2011088549 A1 WO2011088549 A1 WO 2011088549A1 CA 2011000041 W CA2011000041 W CA 2011000041W WO 2011088549 A1 WO2011088549 A1 WO 2011088549A1
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
methyl
hydroxy
fluorophenyl
dihydroxy
Prior art date
Application number
PCT/CA2011/000041
Other languages
French (fr)
Inventor
Brent Stranix
Jean-Emmanuel Bouchard
Guy Milot
Original Assignee
Ambrilia Biopharma Inc.
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 Ambrilia Biopharma Inc. filed Critical Ambrilia Biopharma Inc.
Priority to EP11734279.0A priority Critical patent/EP2526091A4/en
Priority to JP2012549212A priority patent/JP2013517302A/en
Priority to CN2011800066516A priority patent/CN102834381A/en
Publication of WO2011088549A1 publication Critical patent/WO2011088549A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • 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
    • 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
    • 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
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems

Definitions

  • the present invention is directed to pyridoxine (vitamin B 6 ) derived compounds of Formula I, pharmaceutically acceptable salts or solvates thereof, pharmaceutical formulations comprising one or more compounds of Formula I, their synthesis, and use as modulators or inhibitors of human immunodeficiency virus (HIV) integrase enzyme.
  • Compounds of the present invention are useful for prophylaxis, treatment, delay in the onset or delay in the progression of human immuno-deficiency virus (HIV) infection, acquired immune deficiency syndrome AIDS, AIDS-related complex (ARC), and other diseases and conditions caused or mediated by HIV infection.
  • integration of viral DNA into the host genome.
  • HIV enzymes including reverse transcriptase, protease and integrase.
  • Various synthetic antiviral agents that block various stages of the HIV replication cycle have been developed and marketed including compounds that: interfere with viral binding to CD4 (+) T-lymphocytes (for example, soluble CD4), block viral reverse transcriptase (for example, didanosine and zidovudine (AZT)), block viral aspartyl protease (for example Ritonavir and Indinavir) and inhibit virion budding (for example interferon).
  • CD4 (+) T-lymphocytes for example, soluble CD4
  • block viral reverse transcriptase for example, didanosine and zidovudine (AZT)
  • block viral aspartyl protease for example Ritonavir and Indinavir
  • virion budding for example interferon
  • the compounds of the present invention are useful for inhibiting or modulating HIV integrase enzyme activity and, in particular, for inhibiting HIV replication and for treating HIV infection, AIDS, and HIV-mediated diseases and conditions.
  • the present invention relates to a series of integrase inhibitors derived from pyridoxine and pharmaceutically acceptable derivatives thereof (e.g., salts and solvates).
  • X is H or OH
  • Y is H or OH
  • R is H or halogen (F, CI, Br, I);
  • R 2 is H or halogen (F, CI, Br, I);
  • R 3 is H, C 1-6 alkyl, Q-6 fluoroalkyl, or benzyl;
  • R4 is H, Ci-6 alkyl, or benzyl
  • R 5 is H, or C 1-6 alkyl
  • X is OH
  • Y is H or OH
  • Ri is H or halogen (F, CI, Br, I);
  • R 2 is H or halogen (F, CI, Br, I);
  • R 3 is H, Ci-6 alkyl or benzyl
  • R4 is H, Ci-6 alkyl or benzyl
  • R5 is H or Ci-6 alkyl
  • R is H or halogen (F, CI, Br, I);
  • R 2 is H or halogen (F, CI, Br, I);
  • R 3 is H, C 1-6 alkyl or benzyl
  • R4 is H, C 1-6 alkyl or benzyl
  • R 5 is H, Ci ⁇ alkyl or benzyl
  • X is O or N-OH
  • Y is H or OH
  • R is H or halogen (F, CI, Br, I);
  • R 2 is H or halogen (F, CI, Br, I);
  • R3 is H, Ci_6 alkyl or benzyl
  • R4 is H, Ci-6 alkyl or benzyl
  • R5 is H, Ci-6 alkyl or benzyl
  • compounds of formula I selected from N5-(4- fluorobenzyl)-N2,3-dihydroxy-4-(hydroxymethyl)pyridine-2,5-dicarboxamide, N5-(3- chloro-4-fluorobenzyl)-N2,3 -dihydroxy-4-(hydroxymethyl)pyridine-2, 5 - dicarboxamide, N5-(3,4-dichlorobenzyl)-N2,3-dihydroxy-4-(hydroxymethyl)pyridine- 2,5-dicarboxamide, and 5-(benzyloxymethyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide.
  • compositions comprising a therapeutically effective amount of at least one compound of the present invention, and pharmaceutically acceptable salts or solvates thereof.
  • the compounds of the present invention inhibit HIV integrase including both HIV-1 and HIV-2 and may be used as antiviral agent against HIV, including HIV-1 and HIV-2 strains.
  • the compounds of the present invention are useful for prophylaxis, treatment or delay in the onset or progression of HIV infection, or of a disease or condition caused or mediated by HIV infection, including HIV-1 and HIV-2 infection.
  • the present invention features a method of inhibiting HIV replication, in a mammal, that includes administering to the mammal a replication- inhibiting amount of at least one compound of the present invention, or a
  • methods of inhibiting HIV replication in a cell comprising contacting the cell with an inhibiting amount of at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • kits for inhibiting HIV integrase enzyme activity comprising contacting the integrase enzyme with an integrase-inhibiting amount of at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • the method includes contacting a cell directly or administering the compound of the invention to a mammal suffering from an HIV infection.
  • Another aspect of the present invention includes methods of treating HIV infection in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • AIDS HIV-inhibiting a mammal
  • methods of treating AIDS in a mammal comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof in combination with one or more additional HIV-inhibiting agent.
  • a disease or condition caused or mediated by HIV infection in a mammal comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • kits for prophylaxis or prevention of HIV infection in a mammal comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • kits for inhibiting HIV replication in a mammal comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof and at least one other HIV-inhibiting agent.
  • methods of inhibiting HIV replication in a mammal wherein the HIV is resistant to at least one HIV protease inhibitor comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • methods of inhibiting HIV replication in a mammal, having an HIV infection, wherein the HIV is resistant to at least one HIV reverse transcriptase inhibitor comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • kits for reducing HIV viral load in a mammal infected with HIV comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
  • kits for reducing HIV viral load in a mammal infected with HIV comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof in combination with one or more additional HIV-inhibiting agents.
  • At least one compound of the present invention for the manufacture of a pharmaceutical composition for prevention or prophylaxis of AIDS or ARC.
  • At least one compound of the present invention for the manufacture of a pharmaceutical composition for prevention or prophylaxis of HIV infection.
  • the mammal e.g., human
  • the mammal e.g., human
  • HIV human immunodeficiency virus
  • HIV-1 HIV-1
  • HIV-2 HIV-2
  • HIV immunodeficiency Syndrome
  • LAV lymphadenopathy-associated virus
  • ARV AIDS-associated retrovirus
  • HIV reverse transcriptase refers to an enzyme, encoded by a retroviral genome, which catalyzes or mediates the conversion (reverse transcription) of viral RNA to DNA or generation of a provirus (Haseltine W. A. FASEB J. vol. 5, p. 2349 - 2360 (1991)).
  • reverse transcriptase inhibitor or "HIV reverse transcriptase inhibitor,” as used herein, refer to compounds or combinations of compounds that interfere with the proper functioning of the HIV reverse transcriptase enzyme that is responsible for converting single-stranded HIV viral RNA into HIV viral DNA.
  • HIV integrase or "integrase” as used herein refer to an enzyme, encoded by a retroviral genome, that catalyzes or mediates integration of provirus DNA (retroviral double stranded DNA) into the host genomic DNA.
  • the integrase enzyme can serve as a template for viral gene expression by the host transcription system, leading to viral replication (Roth et al., Cell, 1989 Jul 14; 58(l):47-54.:
  • integrase inhibitor or "HIV integrase inhibitor,” as used herein, refer to a compound or combination of compounds that interfere with the proper functioning of the HIV integrase enzyme that is responsible for inserting the genes of HIV into the DNA of a host cell.
  • integration refers to insertion of viral DNA, retroviral DNA, provirus, or provirus DNA into the host genome mediated by integrase enzyme. Integration generally occurs following association of integrase and viral DNA with the pre-integration complex (PIC) at the host nucleus and transport of the viral DNA into the host nucleus as a component of the pre-integration complex (Goldgur Y et al Proc Natl Acad Sci U S A. 1999 Nov 9;96(23): 13040-3; Sayasith , Sauve G and Yelle J. Expert Opin Ther Targets. 2001 Aug;5(4):443-464; Debyser Z et al Methods Mol Biol. 2001;160:139-55).
  • PIC pre-integration complex
  • protease inhibitor means compounds or combinations of compounds that interfere with the proper functioning of the HIV protease enzyme that is responsible for cleaving long strands of viral protein into the separate proteins making up the viral core.
  • fusion inhibitor or “HIV fusion inhibitor,” as used herein, refer to compounds or combinations of compounds that bind to the gp41 envelope protein on the surface of CD4 cells and block the structural changes necessary for the virus to fuse with the cell.
  • HIV load and "HIV viral load,” as used herein, mean the amount of HIV in the circulating blood of a mammal, such as a human.
  • the amount of HIV virus in the blood of mammal can be determined by measuring the quantity of HIV RNA in the blood using methods known to those of ordinary skill in the art.
  • Retrovirus refers to a virus belonging to the viral family Retroviridae, which includes viruses that possess an RNA genome, and that replicate via a DNA intermediate.
  • Vitamin B 6 refers to one or more of three compounds that are commonly referred to as vitamin B 6 namely pyridoxal, pyridoxamine and pyridoxine.
  • Pyridoxine differs from pyridoxamine by the substituent at the '4 position. Pyridoxine based on a pyridine ring, with hydroxyl, methyl, and hydroxymethyl substituents and is converted in vivo to pyridoxal 5-phosphate, the biologically active form of pyridoxine.
  • Ci-6 alkyl as used herein means saturated monovalent hydrocarbon radicals having straight or branched moieties and containing from 1 to 6 carbon atoms.
  • . 6 alkyl group may be substituted or unsubstituted. Examples of such groups include, but are not limited to, methyl, ethyl, propyl, iso-propyl, n-butyl, iso- butyl, and tert-butyl.
  • benzyl and “phenyl” refer to both substituted and unsubstitued benzyl and phenyl groups, respectively.
  • fluoroalkyl represents a C]- alkyl group, as defined herein, where one or more hydrogen radicals bound to the alkyl group has been replaced by a fluoride radical.
  • a fluoroalkyl group can be further substituted with any of the substituent groups described herein for C 1-6 alkyl groups. Fluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl, and the like.
  • a group e.g., a Ci- 6 alkyl group, benzyl, or phenyl group
  • the group may be substituted with, e.g., 1, 2, 3, 4, 5, or 6 substituents.
  • Optional substituents for phenyl or benzyl groups include, but are not limited to, the optional substituents described herein for alkyl groups, as well as Ci -6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 6- io aryl, and halogen (e.g., F, CI, Br, or I) substituents.
  • inhibiting HIV replication means reducing or preventing (e.g., by at least 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) human
  • HIV immunodeficiency virus
  • a cell may be present in vitro, or it may be present in vivo, such as in a mammal, such as a human.
  • Such inhibition may be accomplished by administering a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, directly to the cell, or to a mammal, in an amount sufficient to inhibit HIV replication.
  • the inhibition of HIV replication in a cell, such as in a mammal can be measured or monitored using methods known to those of ordinary skill in the art. For example, an amount of a compound of the invention may be administered to a mammal, either alone or as part of a pharmaceutically acceptable formulation.
  • Blood samples may then be withdrawn from the mammal and the amount of HIV virus in the sample may be quantified using methods known to those of ordinary skill in the art.
  • a reduction in the amount of HIV virus in the sample compared to the amount found in the blood before administration of a compound of the invention would represent inhibition of the replication of HIV virus in the mammal.
  • a reduction in the amount of HIV virus in the sample compared to the amount found in a positive reference sample e.g., the blood from a subject having HIV but not treated with a compound of the invention
  • the administration of a compound of the invention to the cell such as in a mammal, may be in the form of single dose or a series of doses. In the case of more than one dose, the doses may be administered in one day or they may be administered over more than one day.
  • HlV-inhibiting agent means a compound, including but not limited to the compounds of the present invention, or a pharmaceutically acceptable salt thereof which is capable of inhibiting the replication of HIV in a cell, such as a cell in a mammal. Such compounds may inhibit HIV replication through any mechanism known to those of ordinary skill in the art.
  • Non-limiting examples of HIV-inhibiting agents include an entry inhibitor, a protease inhibitor, a reverse transcriptase inhibitor, a fusion inhibitor, and an integrase inhibitor.
  • human immunodeficiency virus-inhibiting amount or "HIV- inhibiting amount,” as used herein, refer to the amount of an HIV-inhibiting agent, or a pharmaceutically acceptable salt of solvate thereof, required to inhibit replication of the human immunodeficiency virus (HIV) in vivo, such as in a mammal, or in vitro.
  • HIV human immunodeficiency virus
  • the amount of such compounds required to cause such inhibition can be determined without undue experimentation using methods described herein and those known to those of ordinary skill in the art.
  • inhibiting HIV integrase enzyme activity means decreasing (e.g., by at least 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) the activity or functioning of the HIV integrase enzyme either in vitro or in vivo, such as in a mammal, such as a human.
  • HIV integrase enzyme-inhibiting amount refers to the amount of an HIV-inhibiting agent or a pharmaceutically acceptable salt or solvate thereof, required to decrease the activity of the HIV integrase enzyme either in vivo, such as in a mammal, or in vitro, such as in a cultured cell line. In one example, such inhibition may take place by the compound of the present invention binding directly to the HIV integrase enzyme. In addition, the activity of the HIV integrase enzyme may be decreased in the presence of a compound of the present invention when such direct binding between the enzyme and the compound does not take place.
  • HIV integrase may be determined using in vitro or in vivo systems, or a combination of both, using methods known to those of ordinary skill in the art.
  • solvate means a pharmaceutically acceptable solvate form of a compound of the present invention that retains the biological effectiveness of such compound.
  • solvates include, but are not limited to, compounds of the invention in combination with water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine, or mixtures thereof.
  • one solvent molecule is associated with one molecule of the compounds of the present invention, such as a hydrate.
  • more than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a dihydrate.
  • solvates of the present invention include solvates of compounds of the present invention that retain the biological effectiveness of the non-hydrate form of the compounds.
  • a "pharmaceutically acceptable salt” as used herein means a salt that retains the biological effectiveness of the free acids and bases of the specified derivative, containing pharmacologically acceptable anions or cations, and is not biologically or otherwise undesirable.
  • pharmaceutically acceptable salts include, but are not limited to, acetate, acrylate, benzenesulfonate, benzoate (such as chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, and methoxybenzoate), bicarbonate, bisulfate, bisulfite, bitartrate, borate, bromide, butyne-1, 4-dioate, calcium edetate, camsylate, carbonate, chloride, caproate, caprylate, clavulanate, citrate, decanoate, dihydrochloride, dihydrogenphosphate, edetate, edislyate, estolate, esylate, ethylsuccinate, formate, fumarate
  • pharmaceutically acceptable formulation means a combination of a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, and a carrier, diluent, and/or excipients that are compatible with a compound of the present invention, and is not deleterious to the recipient thereof.
  • Pharmaceutical formulations can be prepared by procedures known to those of ordinary skill in the art.
  • the compounds of the present invention can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, and the like.
  • excipients, diluents, and carriers that are suitable for such formulations include the following: fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl pyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as povidone, sodium starch glycolate, sodium carboxymethylcellulose, agar agar, calcium carbonate, and sodium bicarbonate; agents for retarding dissolution such as paraffin; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adsorptive carriers such as keolih and bentonite; and lubricants such as talc, calcium and magnesium stearate and solid polyethylene glycols.
  • fillers and extenders such as starch, sugars, mannitol, and silicic derivatives
  • compositions of the present invention can contain more than one active ingredient.
  • such formulations may contain more than one compound according to the present invention.
  • such formulations may contain one or more compounds of the present invention and one or more additional anti-HIV agents.
  • a pharmaceutically acceptable formulation may also include but is not limited to compounds, other than the compounds of formula 1, having a structure such that, upon administration to a recipient or patient, a compound of this invention, active metabolite or residue thereof is directly or indirectly provided.
  • reduce or “inhibit” is meant the ability to cause an overall decrease of 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in the biological activity or levels being measured.
  • a therapeutically effective amount means an amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, that, when administered to a mammal in need of such treatment, is sufficient to effect treatment, as defined herein.
  • a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof is a quantity sufficient to modulate or inhibit the activity of the HIV integrase enzyme such that a disease condition that is mediated by activity of the HIV integrase enzyme is reduced or alleviated.
  • treat refers to both therapeutic treatment and prophylactic or preventative measures for an HIV infection or an HIV or AIDS mediated disease or condition.
  • therapeutic treatment refers to administering treatment to a subject already suffering from a disease to improve the subject's condition. Treatment can include modulating or inhibiting the disease or condition, (e.g., arresting its development); relieving the disease or condition, (e.g., causing regression of the disease or condition); reduction in viral load; or relieving and/or alleviating the disease or condition or the symptoms resulting from the disease or condition with or without addressing the underlying disease or condition.
  • prevent disease refers to prophylactic treatment of a subject who is not yet ill, but who is susceptible to, or otherwise at risk of, developing a particular disease.
  • Prophylactic treatment can also include the prevention of one or more symptoms associated with HIV or AIDS.
  • treating includes the administration to a mammal either for therapeutic or prophylactic purposes.
  • subject is meant a mammal, including, but not limited to, a human or non- human mammal, such as a simian, bovine, canine, equine or feline.
  • resistant refers to HIV virus demonstrating a reduction in sensitivity to a particular drug.
  • a mammal infected with HIV that is resistant to a particular anti-HIV agent or combination of agents usually manifests an increase in HIV viral load despite continued
  • Resistance may be either genotypic, meaning that a mutation in the HIV genetic make-up has occurred, or phenotypic, meaning that resistance is discovered by successfully growing laboratory cultures of HIV virus in the presence of an anti-HIV agent or a combination of such agents.
  • co-administration refers to the administration of a combination of at least a first agent and a second agent and can include two or more agents. Such co-administration can be performed such that two or multiple agents are part of the same composition or part of the same unitary dosage form, or in separate compositions or dosage forms. Co-administration also includes administering a first agent and a second agent, or more than two agents separately and as part of the same therapeutic regimen. The agents, if administered separately, need not necessarily be administered at essentially the same time, although they can be if so desired.
  • co-administration includes, for example, administering a first agent and a second agent as separate dosages or dosage forms, but at the same time.
  • Co-administration also includes separate administration at different times (e.g., sequentially or alternating one agent with the other) and in any order.
  • compound of the present invention refers to compounds of formulas I and la, lb, and Ic as well compounds provided in the Examples that follow, and includes pharmaceutically acceptable salts of these compounds.
  • HIV- 1. -2 Human immunodeficiency virus type 1 , type 2
  • HTLV-I, -II I Human T-cell lymphotropic virus type 1, type II i M Molar
  • compositions contemplated herein comprise at least one compound of the present invention, including pharmaceutically acceptable salts, solvate or formulations thereof, with a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances,
  • polyethyleneglycol sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, liposomes and lanolin.
  • Compounds of the present invention that are basic may be prepared as a salt using suitable methods known in the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid; hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid; and the like, or with an organic acid, such as acetic acid; maleic acid; succinic acid; mandelic acid; fumaric acid; malonic acid; pyruvic acid; oxalic acid; glycolic acid; salicylic acid; pyranosidyl acid, such as glucuronic acid or galacturonic acid; alpha- hydroxy acid, such as citric acid or tartaric acid; amino acid, such as aspartic acid or glutamic acid; aromatic acid, such as benzoic acid or cinnamic acid; sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid; and the like.
  • an inorganic acid such as hydrochloric acid
  • Basic compounds of the present invention can form a variety of salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, it is common practice to first isolate the compound of the present invention as a pharmaceutically unacceptable salt and then convert to a free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition salt.
  • the acid addition salts of the base compounds of this invention can be prepared by treating the base compound with a substantially equivalent amount of the selected mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or ethanol.
  • Compounds of the present invention that are acidic may be prepared as a salt using suitable methods known in the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth metal hydroxide; or the like.
  • suitable salts include organic salts derived from amino acids such as glycine and arginine;
  • ammonia primary, secondary, and tertiary amines; and cyclic amines, such as piperidine, morpholine, and piperazine; as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • Acidic compounds of the present invention can form base salts with various pharmacologically acceptable cations.
  • such salts include the alkali metal or alkaline- earth metal salts and particularly, the sodium and potassium salts, which can be prepared using conventional techniques.
  • the chemical bases suitable as reagents in preparing the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of the present invention.
  • Such non- toxic base salts include those derived from such
  • pharmacologically acceptable cations as sodium, potassium calcium and magnesium, etc.
  • These salts can be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, preferably under reduced pressure.
  • they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together, and then evaporating the resulting solution to dryness in the same manner as before.
  • stoichiometric quantities of reagents are preferably employed in order to ensure completeness of reaction and maximum yields of the desired final product.
  • a pharmaceutical composition comprising at least one of the compounds of the present invention, is administered in a pharmaceutically acceptable formulation prepared by combining a therapeutically effective amount of the compound with one or more pharmaceutically suitable carriers including diluents, excipients and auxiliaries that facilitate processing of the active compounds into a pharmaceutically acceptable formulation.
  • Carriers employed may be either solid or liquid.
  • Exemplary solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • Exemplary liquid carriers are syrup, peanut oil, olive oil, water and the like.
  • the inventive compositions may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate or the like.
  • Further additives or excipients may be added to achieve the desired formulation properties.
  • a bioavailability enhancer such: as Labrasol®, Gelucire® or the like, or formulator, such as CHIC (carboxy- methylcellulose), PG (propyleneglycol), or PEG (polyethyleneglycol)
  • CHIC carboxy- methylcellulose
  • PG propyleneglycol
  • PEG polyethyleneglycol
  • Gelucire® a semi-solid vehicle that protects active ingredients from light, moisture and oxidation, may be added, e.g., when preparing a capsule formulation.
  • the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form, or formed into a troche or lozenge.
  • the amount of solid carrier may vary, but generally will be from about 25 mg to about lg.
  • the preparation may be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or nonaqueous liquid suspension.
  • the inventive compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g., parenteral or oral
  • a pharmaceutically acceptable salt of a compound of the present invention may be dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3 M solution of succinic acid or citric acid.
  • the agent may be dissolved in a suitable co-solvent or combinations of co-solvents.
  • suitable co-solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0-60% of the total volume.
  • a compound of the present invention is dissolved in DMSO and diluted with water.
  • the composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution.
  • compositions for oral use can be obtained using a solid excipient in an admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl- cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the pharmaceutical compositions comprising the compounds of the present invention may also contain suitable solid- or gel- phase carriers or excipients. These carriers and excipients may provide marked improvement in the bioavailability of poorly soluble drugs. Examples of such carriers or excipients include calcium carbonate, calcium, phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Furthermore, additives or excipients such as Gelucire®, Capryol®, Labrafil®, Labrasol®, Lauroglycol®, Plurol®, Peceol®, Transcutol® and the like may be used. Further, the pharmaceutical composition may be incorporated into a skin patch for delivery of the drug directly onto the skin.
  • the compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties.
  • modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral bioavailability, increase solubility to allow administration by injection, alter metabolism or alter rate of excretion ⁇ Pharmacokinetic Optimization in Drug Research, Testa, B. et al, 2001, Wiley-VCH, VCHA).
  • compositions of this invention may be administered orally, intravenously, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally, or via an implanted reservoir and are preferably administered orally or parenterally.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically acceptable carriers, adjuvants or vehicles.
  • parenteral or “parenterally” as used herein includes sub-cutaneous, intracutaneous, intra-venous, intra-muscular, intra-articular, intra-synovial, intra-sternal, intra-thecal, intra-lesional and intracranial injection or infusion techniques.
  • compositions of the invention may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3- butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solutions.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • compositions of the invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspension and solutions.
  • tablets for oral and carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavouring and/or coloring agents may be added.
  • compositions of the invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax, and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • an exemplary daily dose generally employed will be from about 0.001 to about 1000 mg/kg of body weight, with courses of treatment repeated at appropriate intervals, preferably between 0.01 and about 25 mg/kg body weight per day, and more preferably between about 0.5 and about 25 mg/kg body weight per day of the active ingredient compound useful in the prevention and treatment of viral infection, including HIV infection.
  • the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 7 0 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 to about 500 mg, or from about 100 mg to about 500 mg.
  • the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 7 0 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 to about 500 mg, or from about 100 mg to about 500 mg.
  • the pharmaceutically acceptable formulations of the present invention may contain a compound of the present
  • pharmaceutically acceptable salt or solvate thereof in an amount from about 0.5 w/w % to about 95 w/w %, or from about 1 w/w % to about 95 w/w %, or from about 1 w/w % to about 75 w/w %, or from about 5 w/w % to about 75 w/w %, or from about 10 w/w % to about 75 w/w %, or from about 10 w/w % to about 50 w/w %.
  • compositions of this invention may be administered as a continuous infusion, once per day, multiple times per day (e.g., from about 1 to about 5 times per day), once per week, twice per week, three times per week, every other day, every other week or as determined by the practicing clinician.
  • administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the patient treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 75% active compound (w/w).
  • such preparations contain from about 20% to about 50% active compound.
  • a maintenance dose of a compound, composition or combination of this invention may be administered if necessary or desired. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained or maintained. When the symptoms have been alleviated to the desired level, treatment should cease, at least in principle. Patients may, however, require intermittent treatment on a long-term basis, upon any recurrence of disease symptoms, especially for AIDS.
  • the compounds of this invention are also useful as commercial reagents which effectively bind to HIV integrase.
  • the compounds of this invention, and their derivatives may be used to block integration of a target DNA molecule by integrase, or may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications.
  • the compounds of the present invention can be used alone (monotherapy) or administered in combination with one or more other HIV-inhibiting agents including but not limited to additional compounds of the invention or entry inhibitors, protease inhibitors, reverse transcriptase inhibitors, fusion inhibitors, and integrase inhibitors, examples of which are described below and known to the skilled artisan.
  • the compounds of the invention can be used in combination with an additional HIV integrase inhibitor.
  • Compounds that effectively inhibit HIV integrase may provide improved antiviral agents and compositions for treating HIV infection (Wai, J.S. et al., J. Med. Chem. 43:4923-4926 (2000); Grobler, J. et al, PNAS 99: 6661- 6666 (2002); Pals, G.C.G. et al, J. Med. Chem. 45: 3184-3194 (2002); Young, S. D., Curr. Opin. Drug Disc. & Devel. 4(4): 402-410 (2001);
  • the compounds of this invention may be administered in combination with antiviral agents which target other steps in the retroviral replication cycle.
  • the co-administered antiviral agent can be one that targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA.
  • Antiviral agents targeting such early life cycle events include, didanosine (ddl), zalcitabine (ddC), stavudine (d4T), zidovudine (AZT), polysulfated polysaccharides, sT4 (soluble CD4)— which blocks attachment or adsorption of the virus to host cells— and other compounds which block binding of virus to CD4 receptors on CD4-bearing T-lymphocytes.
  • retroviral reverse transcriptase inhibitors such as derivatives of AZT
  • retroviral reverse transcriptase inhibitors may also be co-administered with the compounds of this invention to provide therapeutic treatment for substantially reducing or eliminating viral infectivity and the symptoms associated therewith.
  • antiviral agents include ganciclovir, dideoxycytidine, trisodium phosphonoformiate, eflornithine, ribavirin, acyclovir, alpha interferon and trimenotrexate.
  • non-ribonucleoside inhibitors of reverse transcriptase such as TIBO, efavirenz, nevirapine or delavirdine
  • TIBO efavirenz
  • nevirapine nevirapine
  • delavirdine may be used to potentiate the effect of the compounds of this invention, as may viral uncoating inhibitors, inhibitors of trans-activating proteins such as tat or rev, or inhibitors of the viral protease.
  • These compounds may also be co-administered with other inhibitors of HIV integrase.
  • Combination therapies according to this invention may exert an additive or combined inhibitory effect on HIV replication because each therapeutic agent of the combination acts on a different site of HIV replication or a synergistic effect.
  • the use of such combination therapies also advantageously enables a reduction in the dosage of each anti-retroviral agent, compared to administration of either agent alone as a monotherapy, while providing an equivalent or better therapeutic or prophylactic effect.
  • Administration of lower doses of each therapeutic agent often reduces or even eliminates side effects or toxicity relative to monotherapy.
  • combination therapies reduce the potential for the development of viral resistance to the agents administered compared to monotherapy.
  • Preferred combination therapies include the administration of a compound of this invention with AZT, 3TC, ddl, ddC, d4T, tenofovir, FTC, Combivir® (AZT/3TC combination), abacavir, efavirenz, nevirapine and delavirdine.
  • the compounds of this invention may also be co-administered with other HIV protease inhibitors such as saquinavir, indinavir, nelfinavir, ritonavir, atazanavir, darunavir, and amprenavir. Combination of the compounds of this invention with such protease inhibitors may increase the therapeutic or prophylactic against various HIV viral mutants, HIV quasi species or other closely related viruses.
  • the compounds of this invention may be administered in combination with nucleoside or non-nucleoside retroviral reverse transcriptase inhibitors (e.g., nucleoside or non-nucleoside retroviral reverse transcriptase inhibitors (e.g., nucleoside or non-nucleoside retroviral reverse transcriptase inhibitors (e.g., nucleoside or non-nucleoside retroviral reverse transcriptase inhibitors (e.g.
  • HIV-entry inhibitors HIV integrase inhibitors
  • immuno-modulators e.g., bropirimine, anti-human alpha interferon antibody, IL-2, GM-CSF, methionine enkephalin, interferon alpha, diethyldithiocarbante, tumor necrosis factor, naltrexone and rEPO
  • antibiotics e.g., pentamidine isethionate
  • compositions of this invention may include a combination of an integrase inhibitor compound of the present invention and another therapeutic or prophylactic agent or HIV-inhibiting agent. Additional examples of agents useful for treating AIDS and HIV and suitable for combination therapies with the compounds of this invention are listed in Tables 1 and 2 below.
  • GW 1592 Glaxo Wellcome HIV infection, AIDS, ARC
  • BMS-232623 (CGP-73547) Bristol-Myers Squibb/ ovartis HIV infection, AIDS, ARC
  • BMS-234475 (CGP-61755) Bristol-Myers Squibb/Novartis HIV infection, AIDS, ARC
  • Cidofovir Gilead Science CMV retinitis, herpes, papillomavirus Curdlan sulfate AJI Pharma USA HIV infection
  • Delaviridine Pharmacia-Upjohn HI V infection, AIDS, ARC
  • DMP-450 protease inhibitor
  • AVID HIV infection, AIDS, ARC
  • Efavirenz (DMP 266) BMS & DuPont Merck HIV infection, AIDS, ARC
  • Reyataz® (atazanavir) Bristol-Myers Squibb HIV infection, AIDS
  • Trizivir® GSK HIV infection HIV
  • IMREG-1 Imreg New Orleans, LA
  • AIDS Kaposi's sarcoma
  • ARC ARC
  • Immunotherapeutic Anti-infectives that may be used in combination with the compounds of the present invention include, but are not limited to, atovaquone, azithromycin, clarithromycin, trimethoprim, trovafloxacin, pyrimethamine, daunorubicin, clindamycin with primaquine, fluconazole, pastill, ornidyl, eflornithine pentamidine, rifabutin, spiramycin, intraconazole-R 1211, trimetrexate, daunorubicin, recombinant human erythropoietin, recombinant human growth hormone, megestrol acetate, testerone, and total enteral nutrition.
  • Antifungals that may be used in combination with the compounds of the present invention include, but are not limited to, anidulafungin, C31G, caspofungin, DB-289, fluconzaole, itraconazole, ketoconazole, micafungin, posaconazole, and voriconazole.
  • Other compounds that may be used in combination with the compounds of the present invention include, but are not limited to, acmannan, ansamycin, LM 427, AR177, BMS-232623, BMS-234475, CI-1012, curdlan sulfate, dextran sulfate, STOCRINE EL 10, hypericin, lobucavir, novapren, peptide T octabpeptide sequence, trisodium phosphonoformate, probucol, and RBC-CD4.
  • the compounds of the present invention may be used in combination with anti-proliferative agents for the treatment of conditions such as Kaposi's sarcoma.
  • agents include, but are not limited to, inhibitors of metallo-matrix proteases, A-007, bevacizumab, BMS-275291, halofuginone, interleukin-12, rituximab, paclitaxel, porfimer sodium, rebimastat, and COL-3.
  • Agents or pharmaceutical compositions that may increase the bioavailability or slow the metabolism of the compounds herein include inhibitors of at least one isoform of the cytochrome P450 (CYP450) enzymes, preferably CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4.
  • CYP450 cytochrome P450
  • Suitable agents that may be used to inhibit CYP 3A4 include, but are not limited to, delavirdine and ritonavir.
  • Such combinations may be administered such that a compound or compounds of the present invention are present in a single formulation or in the form of separate formulations that may be administered sequentially with an appropriate period of time in between or simultaneously.
  • the choice of whether to include the compound or compounds of the present invention in the same formulation as the additional agent or agents is within the knowledge of one of ordinary skill in the art.
  • Scheme 21 consists of the deprotection and oxidation of the intermediate methyl 5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxylate XII, as described in WO09146555, to produce the corresponding aldehyde-ester XIV.
  • This intermediate is then reacted with Grignard reagents to give the (rac)alcohol ester VIII intermediate.
  • Compound VII can then be converted to the final product IX, XI following the procedures described in Scheme 1.
  • Preparative chromatography was performed by flash chromatography, using silica gel 60 (EM Science) with the indicated solvent systems and positive air pressure, to allow for a proper rate of elution, or with a Biotage SP4 rM automated chromatography system. Detection of the compounds was carried out by exposing eluted plates (analytical or preparative) to iodine, UV light and/or treating analytical plates with a 2% solution of jo-anisaldehyde in ethanol containing 3% sulfuric acid and 1% acetic acid, followed by heating.
  • analytical plates can be treated with a 0.3% ninhydrin solution in ethanol containing 3% acetic acid and/or a CAM solution made of 20 g (NH 4 )6Mo 7 024 and 8.3 g Ce(S0 4 )2 polyhydrate in water (750 mL) containing concentrated sulfuric acid (90 mL).
  • Step la preparation of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H- [1,3 Jdioxinof 4, 5-c]pyridin-5-yl)methanol
  • Step lb preparation of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H- [l,3]dioxino[4,5-c]pyridin-5-yl)methanone
  • Step lc preparation of 5-(3-Chloro-4-fluorobenzoyl)-2,2,8-trimethyl-4H- [1,3 ]dioxino[4, 5-cJpyridine 7 -oxide
  • 6.2g of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5- c]pyridin-5-yl)methanone (lb) was dissolved in 130 mL CHCI3 and 6g of 70% meta- Chloroperoxybenzoic acid (mCPBA) was added in one portion. The reaction mixture was stirred for 25 min to the completion of the reaction. 100 mL of 1M K 2 C0 3 was added and the organic phase extracted.
  • mCPBA meta- Chloroperoxybenzoic acid
  • Step Id Preparation of (3-Chloro-4-fluorophenyl)(8-(hydroxymethyl)-2,2- dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-c ]pyridin-5-yl)methanone
  • Step le Preparation of 5-(3-Chloro-4-fluorobenzoyl)-2,2-dimethyl-4H- [1,3 Jdioxinof 4, 5-c ]pyridine-8-carbaldehyde
  • Step lg Preparation of (R)-methyl 5-((3-Chloro-4- fluorophenyl) (hydroxy)methyl)-2, 2-dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-cJpyridine-8- carboxylate
  • R- methyl (CBS) 100 mg was dissolved in 10 mL THF. 3 mL 1.0 M borane THF was then added and the mixture cooled to -20°C with an argon atmosphere. 1 g of the ketone was dissolved in 10 mL THF and added drop wise over 1.5 h with the temperature slowly warming to RT. stirring was left for 2h. 10 mL of anhydrous MeOH was then added and the mixture heated to ⁇ 50°C to quench the reaction (H 2 evolution). The solvent was evaporated and replaced with anhydrous MeOH, stirred for 30 min and once again evaporated.
  • CBS R- methyl
  • Step lh preparation of (R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)- N,3-dihydroxy-4-(hydroxymethyl)picolinamide
  • the EtAc was removed and the residual oil dissolved in 3mL 70% formic acid. This solution was left to rest at RT for 30 min upon which completion of the deprotection was noted. The oil was then diluted with 20 mL of water and 70 mL of EtAc and the product was extracted into the organic phase. A black aqueous layer formed and was extracted with one 50mL portion of EtAc. Following this extraction the organic phase was concentrated (to 20 mL) and extracted with 0. IN NaOH (2 ⁇ 20 mL). The aqueous layer was then removed and washed with hexane. The aqueous layer was again removed and acidified until complete precipitation of the product (acid pH) was observed.
  • Step 2a preparation of methyl 5-((4-fluorophenyl)(hydroxy)methyl)-2,2- dimethyl-4H-[l, 3 ]dioxino[4, 5-c ]pyridine-8-carboxylate
  • a 4-fluorophenyl magnesium bromide solution (0.438 mL of 1M in tetrahydrofuran, 30.0 mmol, 1.1 eq) at -78 °C was added to a methyl 5-formyl-2,2- dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8-carboxylate solution (0.100 g, 0.398 mmol, 1 eq. in 5.0 mL of tetrahydrofurane). The reaction mixture was stirred for 20 min. at -78 °C. A saturated bicarbonate solution was added and reaction mixture was extracted with ethyl acetate.
  • Step 2b preparation of methyl 5-((4-fluorophenyl)(acetoxy)methyl)-2,2- dimethyl-4H-[ 1, 3 ]dioxino[4, 5-c ]pyridine-8-carboxylate
  • Trietylamine (0.03 lg, 0.300 mmol, 3 eq) and acetic anhydride (0.01 lg, 0.11 1 mmol, 1.1 eq) were added to a solution of methyl 5-((4- fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8- carboxylate 2a in 4.0 mL of dichloromethane. This reaction mixture was stirred for 1 hour at room temperature followed by addition of a saturated bicarbonate solution. The reaction mixture was then extracted with ethyl acetate and the organic phases were combined and concentrated under reduced pressure yielding 0.039 g of crude product 2b (100% yield) as a white solid.
  • Step 2c preparation of methyl 5-(4-fl orobenzyl)-2,2-dimethyl-4H- [1,3 Jdioxinof 4, 5-c Jpyridine-8-carboxylate
  • Step 2d preparation of 5-(4-fluorobenzyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide
  • Step 6a preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N-methoxy-2,2- dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-c Jpyridine-8-carboxamide
  • reaction mixture was stirred for 30 min at -78 °C and a saturated ammonium chloride solution was added.
  • the reaction mixture was then extracted with ethyl acetate and the resulting organic phases were combined and concentrated under reduced pressure yielding 0.136 g of crude product 6a (100% yield) as a white solid.
  • Step 6b preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-3-hydroxy-4- (hydroxymethyl)-N-methoxypicolinamide
  • Step 7a preparation of methyl 3-(benzyloxy)-5-((4- fluorophenyl)(hydroxy)methyl)-4-methylpicolinate
  • Step 7b preparation of methyl 5-(acetoxy(4fluorophenyl)methyl)-3- (benzyloxy)-4-methylpicolinate
  • Step 7c preparation of methyl 5-(4-fluorobenzyl)-3-hydroxy-4- methylpicolinate
  • step 7b The product of step 7b (0.260 g, 0.615 mmol, 1 eq) and 10% Pd/C (20 mg) in methanol (4.0 mL) were stirred under an atmospheric hydrogen overnight. The catalyst was then filtered and the crude product was purified by silica gel (40% ethyl acetate/hexane) to give 0.169 g of 7c (100%) as a white solid.
  • Step 7d preparation of 5-(4-fluorobenzyl)-N,3-dihydroxy-4- methylpicolinamide
  • Step 8a preparation of 3-(benzyloxy)-5-((4-fluorophenyl)(hydroxy)methyl)-N- hydroxy-4-methylpicolinamide
  • Step 8b preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- methylpicolinamide
  • Step 9a preparation of methyl 3-(benzyloxy)-5-(4-fluorobenzoyl)-4- methylpicolinate
  • Step 9b preparation of 3-(benzyloxy)-5-(4-fluorobenzoyl)-N-hydroxy-4- methylpicolinamide
  • the crude product was purified by Biotage reverse phase chromatography yieldingO.015 g of 5- (4-fluorobenzoyl)-N,3-dihydroxy-4-methylpicolinamide (the product step 9b, example 9) (22% yield) and 0.015 g of the corresponding oxime (E)-5-((4- fluorophenyl)(hydroxyimino)methyl)-N,3-dihydroxy-4-methylpicolinarnide (10) (22% yield), both as a white solid.
  • step la (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridin-5- yl)rnethanol
  • step lb (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridin-5- yl)methanone
  • step lc 5-(3-Chloro-4-fluorobenzoyl)-2,2,8-trimethyl-4H-[l,3]dioxino[4,5- cjpyridme 7-oxide
  • step Id (3-Chloro-4-fluorophenyl)(8-(hydroxymethyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridin-5-yl)methanone
  • step le Step e 5-(3-Chloro-4-fluorobenzoyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5- c]pyridine-8 -carbaldehyde
  • step lg (R)-methyl 5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H- [1 ,3] diox ino [4,5 -c]pyridine-8-carboxylate
  • step lh (R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide
  • Example 12 Biological Evaluation. In Vitro Integrase Inhibition Assay
  • IC50 was determined for the compounds of the inventions based on data generated in strand transfer assays.
  • the IC50 is a measure of the ability of the compounds tested to inhibit the integration of 3 '-processed oligonucleotides by recombinant HIV- 1 integrase.
  • Strand transfer assays were performed essentially as described in Hazuda, D. J.; Felock, P.; Hastings, J. C; Pramanik, B.; Wolfe, A. J. Virol. 1997, 71, 7005-7011).
  • Donor DNA 1.5 pmol/well
  • biotinylated on the 5' end of the strand processed by integrase was immobilized onto streptavidin-coated microtiter plates.
  • Recombinant integrase (250 ng/well) was assembled onto the immobilized donor oligonucleotide in reaction buffer (20 mM Hepes, pH 7.6, 5 mM B-mercaptoethanol, 50 ug/mL bovine serum albumin) containing 30 mM MnCl 2 . Excess enzyme was removed, and the complexes were washed extensively prior to the addition of the target DNA substrate. The target DNA (0.75 pmoles/well) substrate was labeled on each 3' end with FITC.
  • the FITC-labeled products were detected using an anti-FITC antibody conjugated with alkaline phosphatase (Roche) and a chemi-luminescent substrate (Tropix CSPD with Sapphire II enhancer, Applied Biosystems).
  • the assay was performed in a final concentration of 10% DMSO.
  • compounds were added after assembly, just prior to the addition of the target DNA.
  • IC50 values were determined using a sigmoidal dose-response equation.
  • the percent inhibition of HIV- 1 integrase activity was graphed against the log of the compound concentration (M).
  • IDBS ActivityBase
  • A is the lower plateau (-0%)
  • B is the higher plateau (-100%)
  • C is the IC 50
  • D is the slope
  • X is the compound concentration (M)
  • Y is the % inhibition.
  • the antiviral efficacies of the integrase inhibitor compounds of the invention were evaluated based on EC 50 measures obtained from two different in vitro HIV infection assay using cultured MT-4 cells: (1) a multi-cycle infection where cells were infected with wild type NL-4.3) and (2) a single-cycle infection where the cells were infected with a luciferase-bearing, envelope defective (env-) NL-4.3 virus pseudotyped with HIV-1 env (HXBc2.
  • the incubation period for the multi-cycle infection assay was 6 days.
  • Cell viability (cytoprotection) and EC50 were determined using the colorimetric MTT assay (A. J. Japour et al, Antimicrobial Agents and Chemotherapy, 37, 1095-1 101, 1993 and R. Pauwels et al. Journal of Virological Methods, 20, 309-321 , 1988).
  • the incubation period for the single-cycle infection assay was 48 hours.
  • EC50 was determined, as described by Chen et al., Journal of Virology, Feb. 1994, Vol. 68, No. 2, p. 654-660, based on measures of luciferase signal over a range of drug concentrations.
  • the multi-cycle antiviral activity in the absence and presence of human serum was determined by p24 ELISA measurement after 6 days of NL4.3 virus infection.

Abstract

The present invention relates to compounds of Formula I or pharmaceutically acceptable salts, solvates or compositions thereof wherein X, Y, R1-R5 are defined in the specification. Compounds of Formula I inhibit HIV-integrase enzyme and are useful for preventing and treating of HIV infection, AIDS or AIDS-related complex (ARC), and other diseases and conditions caused or mediated by HIV infection.

Description

DERIVATIVES OF PYRIDOXINE FOR INHIBITING HIV INTEGRASE
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims benefit of U.S. Provisional Application No. 61/297,450, filed January 22, 2010, which is hereby incorporated by reference.
FIELD OF THE INVENTION
The present invention is directed to pyridoxine (vitamin B6) derived compounds of Formula I, pharmaceutically acceptable salts or solvates thereof, pharmaceutical formulations comprising one or more compounds of Formula I, their synthesis, and use as modulators or inhibitors of human immunodeficiency virus (HIV) integrase enzyme. Compounds of the present invention are useful for prophylaxis, treatment, delay in the onset or delay in the progression of human immuno-deficiency virus (HIV) infection, acquired immune deficiency syndrome AIDS, AIDS-related complex (ARC), and other diseases and conditions caused or mediated by HIV infection.
BACKGROUND OF THE INVENTION
Retroviruses designated as human immunodeficiency virus (HIV), particularly strains known as HIV-1 and HIV-2, are the etiological agent of AIDS, ARC, and other diseases or conditions caused or mediated by HIV. HIV infection and AIDS are difficult to treat due to the ability of retroviruses to rapidly replicate, mutate and acquire drug resistance. To date, the treatment of AIDS and HIV infection and the development of new drugs for AIDS and HIV infection have focused primarily on the inhibition of HIV replication by targeting key steps in retroviral replication, such as conversion of viral RNA to viral DNA (reverse transcription) and insertion
(integration) of viral DNA into the host genome. These steps rely on the activity of HIV enzymes including reverse transcriptase, protease and integrase. Various synthetic antiviral agents that block various stages of the HIV replication cycle have been developed and marketed including compounds that: interfere with viral binding to CD4 (+) T-lymphocytes (for example, soluble CD4), block viral reverse transcriptase (for example, didanosine and zidovudine (AZT)), block viral aspartyl protease (for example Ritonavir and Indinavir) and inhibit virion budding (for example interferon). Some of these agents have proved ineffective in clinical tests and others, primarily those that target the early stages of viral replication, have no effect on the production of infectious virions in chronically infected cells.
Furthermore, administration of therapeutic doses of these agents has commonly led to cell-toxicity, unwanted side effects, such as anemia, neurotoxicity and bone marrow suppression, and rapid emergence of drug resistance which limits safe and effective treatment of AIDS, HIV infection and other HIV-caused diseases.
The use of combination therapy has suppressed the emergence of resistance relative to monotherapy, however even with combination therapy there is a loss of efficacy in 30-50% of patients due to the development of viral resistance.
Considering the shortcomings of reverse transcriptase and protease inhibitors, even when used as part of a drug cocktail (combination therapy), there is a need for new antiviral drugs and in particular drugs that do not lead to cross-resistance with the current standard of care.
SUMMARY OF THE INVENTION
The compounds of the present invention are useful for inhibiting or modulating HIV integrase enzyme activity and, in particular, for inhibiting HIV replication and for treating HIV infection, AIDS, and HIV-mediated diseases and conditions. The present invention relates to a series of integrase inhibitors derived from pyridoxine and pharmaceutically acceptable derivatives thereof (e.g., salts and solvates).
In one aspect the present invention are compounds of formula I,
Figure imgf000003_0001
I
wherein:
X is H or OH;
Y is H or OH;
R, is H or halogen (F, CI, Br, I); R2 is H or halogen (F, CI, Br, I);
R3 is H, C1-6 alkyl, Q-6 fluoroalkyl, or benzyl;
R4 is H, Ci-6 alkyl, or benzyl; and
R5 is H, or C1-6 alkyl; or
pharmaceutically acceptable salts or solvates thereof.
Further provided herein are compounds of formula la,
Figure imgf000004_0001
la
wherein:
X is OH;
Y is H or OH;
Ri is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, Ci-6 alkyl or benzyl;
R4 is H, Ci-6 alkyl or benzyl; and
R5 is H or Ci-6 alkyl; or
pharmaceutically acceptable salts or solvates thereof.
Further provided herein are compounds of formula lb,
Figure imgf000004_0002
lb
wherein:
X is OH; Y is H or OH;
R, is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, C1-6 alkyl or benzyl;
R4 is H, C1-6 alkyl or benzyl; and
R5 is H, Ci^ alkyl or benzyl; or
pharmaceutically acceptable salts or solvates thereof.
Further provided herein are compounds of formula Ic:
Figure imgf000005_0001
Ic
wherein:
X is O or N-OH;
Y is H or OH;
R, is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, Ci_6 alkyl or benzyl;
R4 is H, Ci-6 alkyl or benzyl; and
R5 is H, Ci-6 alkyl or benzyl; or
pharmaceutically acceptable salts or solvates thereof.
Further provided herein are compounds of formula I selected from N5-(4- fluorobenzyl)-N2,3-dihydroxy-4-(hydroxymethyl)pyridine-2,5-dicarboxamide, N5-(3- chloro-4-fluorobenzyl)-N2,3 -dihydroxy-4-(hydroxymethyl)pyridine-2, 5 - dicarboxamide, N5-(3,4-dichlorobenzyl)-N2,3-dihydroxy-4-(hydroxymethyl)pyridine- 2,5-dicarboxamide, and 5-(benzyloxymethyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide.
Further provided herein are compounds selected from (R)-5-((3-Chloro-4- fluorophenyl)(hydroxy)methy])-N,3-dihydroxy-4-(hydroxymethyl)picolinamide; 5-(4-fluorobenzyl)-N,3-dihydroxy-4-(hydroxymethyl)picolinamide; 5-((4- fluorophenyl)(hydroxy)memyl)-N,3-dihy ^
5-((4-fluorophenyl)(hydroxy)methyl)-N-hydroxy-N,2,2-trimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxamide; 5-((4-fluorophenyl)(hydroxy)methyl)- N,3-dihydroxy-4-(hydroxymethyl)-N-methylpicolinamide; 5-((4- fluorophenyl)(hydroxy)methyl)-3-hydroxy-4-(hydroxyrnethyl)-N- methoxypicolinamide; 5-(4-fluorobenzyl)-N,3-dihydroxy-4-methylpicolinamide; 5- ((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4-methylpicolinamide; 5-(4- fluorobenzoyl)-N,3-dihydroxy-4-methylpicolinamide;
(E)-5-((4-fluorophenyl)(hydroxyimino)memyl)-N,3-dihydroxy-4-memylpicolinamide; and (S)-5-((3-Chloro-4-£luorophenyl)(liydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide.
Further provided herein are pharmaceutical compositions, and pharmaceutically acceptable formulations, comprising a therapeutically effective amount of at least one compound of the present invention, and pharmaceutically acceptable salts or solvates thereof.
The compounds of the present invention inhibit HIV integrase including both HIV-1 and HIV-2 and may be used as antiviral agent against HIV, including HIV-1 and HIV-2 strains.
The compounds of the present invention are useful for prophylaxis, treatment or delay in the onset or progression of HIV infection, or of a disease or condition caused or mediated by HIV infection, including HIV-1 and HIV-2 infection.
In one aspect, the present invention features a method of inhibiting HIV replication, in a mammal, that includes administering to the mammal a replication- inhibiting amount of at least one compound of the present invention, or a
pharmaceutically acceptable salt or solvate thereof.
Further provided are methods of inhibiting HIV replication in a cell, comprising contacting the cell with an inhibiting amount of at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of inhibiting HIV integrase enzyme activity, comprising contacting the integrase enzyme with an integrase-inhibiting amount of at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof. The method includes contacting a cell directly or administering the compound of the invention to a mammal suffering from an HIV infection.
Another aspect of the present invention includes methods of treating HIV infection in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of treating AIDS in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of treating AIDS in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof in combination with one or more additional HIV-inhibiting agent.
Further provided are methods of treating a disease or condition caused or mediated by HIV infection in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of prophylaxis or prevention of HIV infection in a mammal, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of inhibiting HIV replication in a mammal comprising administering to the mammal at least one compound of the present invention or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of inhibiting HIV replication in a mammal comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof and at least one other HIV-inhibiting agent.
Further provided are methods of inhibiting HIV replication in a mammal wherein the HIV is resistant to at least one HIV protease inhibitor, the method comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof. Further provided are methods of inhibiting HIV replication in a mammal, having an HIV infection, wherein the HIV is resistant to at least one HIV reverse transcriptase inhibitor, the method comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of reducing HIV viral load in a mammal infected with HIV, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof.
Further provided are methods of reducing HIV viral load in a mammal infected with HIV, comprising administering to the mammal at least one compound of the present invention, or a pharmaceutically acceptable salt, solvate or formulation thereof in combination with one or more additional HIV-inhibiting agents.
Further provided is the use of at least one compound of the present invention for the manufacture of a pharmaceutical composition for treatment of HIV infection.
Further provided is the use of at least one compound of the present invention for the manufacture of a pharmaceutical composition for treatment of AIDS or ARC.
Further provided is the use of at least one compound of the present invention for the manufacture of a pharmaceutical composition for prevention or prophylaxis of AIDS or ARC.
Further provided is the use of at least one compound of the present invention for the manufacture of a pharmaceutical composition for prevention or prophylaxis of HIV infection.
For any of the above aspects of the invention, the mammal (e.g., human) may have or be suspected of having an HIV infection or an AIDS or HIV mediated disease or condition. The mammal (e.g., human) may or may not have been previously treated with anti-viral or other therapeutic compounds for the HIV infection or AIDS or HIV mediated disease or condition.
Definitions
The terms "human immunodeficiency virus," "HIV," "HIV-1," or "HIV-2" as used herein refer to a retrovirus that is the causative agent for acquired
immunodeficiency Syndrome (AIDS) and diseases, conditions or opportunistic infections associated with AIDS. Previous names for HIV include human T- lymphotropic virus-Ill (HTLV-III), lymphadenopathy-associated virus (LAV), and AIDS-associated retrovirus (ARV).
The terms "HIV reverse transcriptase," "reverse transcriptase, "or "RT" as used herein refer to an enzyme, encoded by a retroviral genome, which catalyzes or mediates the conversion (reverse transcription) of viral RNA to DNA or generation of a provirus (Haseltine W. A. FASEB J. vol. 5, p. 2349 - 2360 (1991)).
The terms "reverse transcriptase inhibitor" or "HIV reverse transcriptase inhibitor," as used herein, refer to compounds or combinations of compounds that interfere with the proper functioning of the HIV reverse transcriptase enzyme that is responsible for converting single-stranded HIV viral RNA into HIV viral DNA.
The terms "HIV integrase" or "integrase" as used herein refer to an enzyme, encoded by a retroviral genome, that catalyzes or mediates integration of provirus DNA (retroviral double stranded DNA) into the host genomic DNA. The integrase enzyme can serve as a template for viral gene expression by the host transcription system, leading to viral replication (Roth et al., Cell, 1989 Jul 14; 58(l):47-54.:
Bukrinsky M. I., Proc. Natn. Acad. Sci. USA 1992, vol. 89 p.6580 - 6584; Gallay et al., Cell. 1995 Nov 17;83(4):569-76).
The terms "integrase inhibitor" or "HIV integrase inhibitor," as used herein, refer to a compound or combination of compounds that interfere with the proper functioning of the HIV integrase enzyme that is responsible for inserting the genes of HIV into the DNA of a host cell.
The term "integration" as used herein refers to insertion of viral DNA, retroviral DNA, provirus, or provirus DNA into the host genome mediated by integrase enzyme. Integration generally occurs following association of integrase and viral DNA with the pre-integration complex (PIC) at the host nucleus and transport of the viral DNA into the host nucleus as a component of the pre-integration complex (Goldgur Y et al Proc Natl Acad Sci U S A. 1999 Nov 9;96(23): 13040-3; Sayasith , Sauve G and Yelle J. Expert Opin Ther Targets. 2001 Aug;5(4):443-464; Debyser Z et al Methods Mol Biol. 2001;160:139-55).
The terms "protease inhibitor" or "HIV protease inhibitor" as used herein mean compounds or combinations of compounds that interfere with the proper functioning of the HIV protease enzyme that is responsible for cleaving long strands of viral protein into the separate proteins making up the viral core. The terms "fusion inhibitor" or "HIV fusion inhibitor," as used herein, refer to compounds or combinations of compounds that bind to the gp41 envelope protein on the surface of CD4 cells and block the structural changes necessary for the virus to fuse with the cell.
The terms "viral load" and "HIV viral load," as used herein, mean the amount of HIV in the circulating blood of a mammal, such as a human. The amount of HIV virus in the blood of mammal can be determined by measuring the quantity of HIV RNA in the blood using methods known to those of ordinary skill in the art.
The term "retrovirus" as used herein refers to a virus belonging to the viral family Retroviridae, which includes viruses that possess an RNA genome, and that replicate via a DNA intermediate.
The term "Vitamin B6" as used herein refers to one or more of three compounds that are commonly referred to as vitamin B6 namely pyridoxal, pyridoxamine and pyridoxine. Pyridoxine differs from pyridoxamine by the substituent at the '4 position. Pyridoxine based on a pyridine ring, with hydroxyl, methyl, and hydroxymethyl substituents and is converted in vivo to pyridoxal 5-phosphate, the biologically active form of pyridoxine.
The terms "comprising" and "including" are used in their open, non-limiting sense.
The term "Ci-6 alkyl" as used herein means saturated monovalent hydrocarbon radicals having straight or branched moieties and containing from 1 to 6 carbon atoms. The C|.6 alkyl group may be substituted or unsubstituted. Examples of such groups include, but are not limited to, methyl, ethyl, propyl, iso-propyl, n-butyl, iso- butyl, and tert-butyl.
As used herein, the terms "benzyl" and "phenyl" refer to both substituted and unsubstitued benzyl and phenyl groups, respectively.
The term "fluoroalkyl," as used herein, represents a C]- alkyl group, as defined herein, where one or more hydrogen radicals bound to the alkyl group has been replaced by a fluoride radical. A fluoroalkyl group can be further substituted with any of the substituent groups described herein for C1-6 alkyl groups. Fluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl, and the like.
Where a group (e.g., a Ci-6 alkyl group, benzyl, or phenyl group) is substituted, the group may be substituted with, e.g., 1, 2, 3, 4, 5, or 6 substituents. Optional substituents for Q.6 alkyl groups include, but are not limited to: C3-9 cycloalkyl, C3-g cycloalkenyl, a heterocyclyl having 3-9 ring atoms, a heteroaryl having 5-12 ring atoms, azido(-N3), nitro (-N02), cyano (-CN), acyloxy(-OC(=0)R'), acyl (-C(=0)R')5 alkoxy (-OR'), amido (-NR'C(=0)R" or -C(=0)NRR'), amino (-NRR'), carboxylic acid (-C02H), carboxylic ester (-C02R'), carbamoyl
(-OC(=0)NR'R" or -NRC(=0)OR'), hydroxy (-OH), isocyano (-NC), sulfonate (-S(=0)2OR), sulfonamide (-S(=0)2NRR' or -NRS(=0)2R'), or sulfonyl (-S(=0)2R), where each R or R' is selected, independently, from H, Ci-6 alkyl, C2-6 alkenyl,
C2-6 alkynyl, C3-9 cycloalkyl, a heterocyclyl having 3-9 ring atoms, C6-10 aryl, or a heteroaryl having 5-12 ring atoms. Optional substituents for phenyl or benzyl groups include, but are not limited to, the optional substituents described herein for alkyl groups, as well as Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-io aryl, and halogen (e.g., F, CI, Br, or I) substituents.
The term "inhibiting HIV replication" means reducing or preventing (e.g., by at least 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) human
immunodeficiency virus (HIV) replication in a cell. Such a cell may be present in vitro, or it may be present in vivo, such as in a mammal, such as a human. Such inhibition may be accomplished by administering a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, directly to the cell, or to a mammal, in an amount sufficient to inhibit HIV replication. The inhibition of HIV replication in a cell, such as in a mammal, can be measured or monitored using methods known to those of ordinary skill in the art. For example, an amount of a compound of the invention may be administered to a mammal, either alone or as part of a pharmaceutically acceptable formulation. Blood samples may then be withdrawn from the mammal and the amount of HIV virus in the sample may be quantified using methods known to those of ordinary skill in the art. A reduction in the amount of HIV virus in the sample compared to the amount found in the blood before administration of a compound of the invention would represent inhibition of the replication of HIV virus in the mammal. In another example, a reduction in the amount of HIV virus in the sample compared to the amount found in a positive reference sample (e.g., the blood from a subject having HIV but not treated with a compound of the invention) would represent inhibition of the replication of HIV virus in the mammal. The administration of a compound of the invention to the cell, such as in a mammal, may be in the form of single dose or a series of doses. In the case of more than one dose, the doses may be administered in one day or they may be administered over more than one day.
The terms "HlV-inhibiting agent," "HIV antiviral agent," or "anti-HIV agent" as used herein means a compound, including but not limited to the compounds of the present invention, or a pharmaceutically acceptable salt thereof which is capable of inhibiting the replication of HIV in a cell, such as a cell in a mammal. Such compounds may inhibit HIV replication through any mechanism known to those of ordinary skill in the art. Non-limiting examples of HIV-inhibiting agents include an entry inhibitor, a protease inhibitor, a reverse transcriptase inhibitor, a fusion inhibitor, and an integrase inhibitor.
The terms "human immunodeficiency virus-inhibiting amount" or "HIV- inhibiting amount," as used herein, refer to the amount of an HIV-inhibiting agent, or a pharmaceutically acceptable salt of solvate thereof, required to inhibit replication of the human immunodeficiency virus (HIV) in vivo, such as in a mammal, or in vitro. The amount of such compounds required to cause such inhibition can be determined without undue experimentation using methods described herein and those known to those of ordinary skill in the art.
The term "inhibiting HIV integrase enzyme activity," as used herein, means decreasing (e.g., by at least 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) the activity or functioning of the HIV integrase enzyme either in vitro or in vivo, such as in a mammal, such as a human.
The term "HIV integrase enzyme-inhibiting amount," as used herein, refers to the amount of an HIV-inhibiting agent or a pharmaceutically acceptable salt or solvate thereof, required to decrease the activity of the HIV integrase enzyme either in vivo, such as in a mammal, or in vitro, such as in a cultured cell line. In one example, such inhibition may take place by the compound of the present invention binding directly to the HIV integrase enzyme. In addition, the activity of the HIV integrase enzyme may be decreased in the presence of a compound of the present invention when such direct binding between the enzyme and the compound does not take place.
Furthermore, such inhibition may be competitive, non-competitive, or uncompetitive. Inhibition of HIV integrase may be determined using in vitro or in vivo systems, or a combination of both, using methods known to those of ordinary skill in the art.
Φ u The term "solvate," as used herein, means a pharmaceutically acceptable solvate form of a compound of the present invention that retains the biological effectiveness of such compound. Examples of solvates include, but are not limited to, compounds of the invention in combination with water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine, or mixtures thereof. In one embodiment of the present invention, one solvent molecule is associated with one molecule of the compounds of the present invention, such as a hydrate. In another embodiment of the present invention, more than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a dihydrate. Additionally, it is specifically contemplated that in the present invention less than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a hemihydrate. Furthermore, solvates of the present invention include solvates of compounds of the present invention that retain the biological effectiveness of the non-hydrate form of the compounds.
A "pharmaceutically acceptable salt" as used herein means a salt that retains the biological effectiveness of the free acids and bases of the specified derivative, containing pharmacologically acceptable anions or cations, and is not biologically or otherwise undesirable. Examples of pharmaceutically acceptable salts include, but are not limited to, acetate, acrylate, benzenesulfonate, benzoate (such as chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, and methoxybenzoate), bicarbonate, bisulfate, bisulfite, bitartrate, borate, bromide, butyne-1, 4-dioate, calcium edetate, camsylate, carbonate, chloride, caproate, caprylate, clavulanate, citrate, decanoate, dihydrochloride, dihydrogenphosphate, edetate, edislyate, estolate, esylate, ethylsuccinate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, heptanoate, hexyne-l,6-dioate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, y- hydroxybutyrate, iodide, isobutyrate, isothionate, lactate, lactobionate, laurate, malate, maleate, malonate, mandelate, mesylate, metaphosphate, methane-sulfonate, methylsulfate,
monohydrogenphosphate, mucate, napsylate, naphthalene- 1 -sulfonate, naphthalene-2- sulfonate, nitrate, oleate, oxalate, pamoate (embonate), palmitate, pantothenate, phenylacetates, phenylbutyrate, phenylpropionate, phthalate, phospate/diphosphate, polygalacturonate, propanesulfonate, propionate, propiolate, pyrophosphate, pyrosulfate, salicylate, stearate, subacetate, suberate, succinate, sulfate^, sulfonate, sulfite, tannate, tartrate, teoclate, tosylate, triethiodode, valerate salts, and cations, such as sodium, potassium, calcium, magnesium, ammonium, and
tetraalkylammonium, among others.
The term "pharmaceutically acceptable formulation," as used herein, means a combination of a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, and a carrier, diluent, and/or excipients that are compatible with a compound of the present invention, and is not deleterious to the recipient thereof. Pharmaceutical formulations can be prepared by procedures known to those of ordinary skill in the art. For example, the compounds of the present invention can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, and the like. Examples of excipients, diluents, and carriers that are suitable for such formulations include the following: fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl pyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as povidone, sodium starch glycolate, sodium carboxymethylcellulose, agar agar, calcium carbonate, and sodium bicarbonate; agents for retarding dissolution such as paraffin; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adsorptive carriers such as keolih and bentonite; and lubricants such as talc, calcium and magnesium stearate and solid polyethylene glycols. Final pharmaceutical forms may be pills, tablets, powders, lozenges, saches, cachets, or sterile packaged powders, and the like, depending on the type of excipient used. Additionally, it is specifically contemplated that pharmaceutically acceptable formulations of the present invention can contain more than one active ingredient. For example, such formulations may contain more than one compound according to the present invention. Alternatively, such formulations may contain one or more compounds of the present invention and one or more additional anti-HIV agents. A pharmaceutically acceptable formulation may also include but is not limited to compounds, other than the compounds of formula 1, having a structure such that, upon administration to a recipient or patient, a compound of this invention, active metabolite or residue thereof is directly or indirectly provided. By "reduce" or "inhibit" is meant the ability to cause an overall decrease of 10%, 20% 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in the biological activity or levels being measured.
The term "therapeutically effective amount," as used herein, means an amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, that, when administered to a mammal in need of such treatment, is sufficient to effect treatment, as defined herein. Thus, a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, is a quantity sufficient to modulate or inhibit the activity of the HIV integrase enzyme such that a disease condition that is mediated by activity of the HIV integrase enzyme is reduced or alleviated.
The terms "treat," "treating," or "treatment" refers to both therapeutic treatment and prophylactic or preventative measures for an HIV infection or an HIV or AIDS mediated disease or condition. To "treat disease" or use for "therapeutic treatment" refers to administering treatment to a subject already suffering from a disease to improve the subject's condition. Treatment can include modulating or inhibiting the disease or condition, (e.g., arresting its development); relieving the disease or condition, (e.g., causing regression of the disease or condition); reduction in viral load; or relieving and/or alleviating the disease or condition or the symptoms resulting from the disease or condition with or without addressing the underlying disease or condition. To "prevent disease" refers to prophylactic treatment of a subject who is not yet ill, but who is susceptible to, or otherwise at risk of, developing a particular disease. Prophylactic treatment can also include the prevention of one or more symptoms associated with HIV or AIDS. Thus, in the claims and embodiments, treating includes the administration to a mammal either for therapeutic or prophylactic purposes.
By "subject" is meant a mammal, including, but not limited to, a human or non- human mammal, such as a simian, bovine, canine, equine or feline.
The terms "resistant," "resistance," and "resistant HIV," as used herein, refer to HIV virus demonstrating a reduction in sensitivity to a particular drug. A mammal infected with HIV that is resistant to a particular anti-HIV agent or combination of agents usually manifests an increase in HIV viral load despite continued
administration of the agent or agents. Resistance may be either genotypic, meaning that a mutation in the HIV genetic make-up has occurred, or phenotypic, meaning that resistance is discovered by successfully growing laboratory cultures of HIV virus in the presence of an anti-HIV agent or a combination of such agents.
The terms "co-administration," "co-administering," "co-administer," "coadministered," or "combination therapy" as used herein, refer to the administration of a combination of at least a first agent and a second agent and can include two or more agents. Such co-administration can be performed such that two or multiple agents are part of the same composition or part of the same unitary dosage form, or in separate compositions or dosage forms. Co-administration also includes administering a first agent and a second agent, or more than two agents separately and as part of the same therapeutic regimen. The agents, if administered separately, need not necessarily be administered at essentially the same time, although they can be if so desired. Thus, co-administration includes, for example, administering a first agent and a second agent as separate dosages or dosage forms, but at the same time. Co-administration also includes separate administration at different times (e.g., sequentially or alternating one agent with the other) and in any order.
The term "compound of the present invention" refers to compounds of formulas I and la, lb, and Ic as well compounds provided in the Examples that follow, and includes pharmaceutically acceptable salts of these compounds.
The abbreviations used herein refer to the following:
! Abbreviation I Meaning
j AcOH ! Acetic acid
Ar \ Argon
AIDS ; Acquired Immunodeficiency Syndrome
AZT i 3-Azido-3-deoxythymine (Zidovudine)
! BSA ! Bovine serum albumin
! DMF ; Dimethylforrnamide
DMA Deoxyribonucleic acid
i EtOH Ethyl alcohol
& gram
! HPLC High pressure liquid chromatography
HIV- 1. -2 ] Human immunodeficiency virus type 1 , type 2
HTLV-I, -II I Human T-cell lymphotropic virus type 1, type II i M Molar
: MeOH Methyl alcohol
mg ; Milligram
: HP i Melting point
min : Minute
: mL : Milliliter
' mmol Millimole
n.M Nanomolar
RNA Ribonucleic acid j RBF Round bottomed flask
i THF " \ etrahydrofuran _ i
Detailed Description
Pharmaceutical compositions contemplated herein comprise at least one compound of the present invention, including pharmaceutically acceptable salts, solvate or formulations thereof, with a pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances,
polyethyleneglycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, liposomes and lanolin.
It is understood by those skilled in the art that the compounds of the present invention, salts, or solvates thereof may exist in different crystal or polymorphic forms that are within the scope of the present invention and specified formulas.
Compounds of the present invention that are basic may be prepared as a salt using suitable methods known in the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid; hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid; and the like, or with an organic acid, such as acetic acid; maleic acid; succinic acid; mandelic acid; fumaric acid; malonic acid; pyruvic acid; oxalic acid; glycolic acid; salicylic acid; pyranosidyl acid, such as glucuronic acid or galacturonic acid; alpha- hydroxy acid, such as citric acid or tartaric acid; amino acid, such as aspartic acid or glutamic acid; aromatic acid, such as benzoic acid or cinnamic acid; sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid; and the like.
Basic compounds of the present invention can form a variety of salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, it is common practice to first isolate the compound of the present invention as a pharmaceutically unacceptable salt and then convert to a free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition salt. The acid addition salts of the base compounds of this invention can be prepared by treating the base compound with a substantially equivalent amount of the selected mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or ethanol.
Compounds of the present invention that are acidic may be prepared as a salt using suitable methods known in the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth metal hydroxide; or the like. Examples of suitable salts include organic salts derived from amino acids such as glycine and arginine;
ammonia; primary, secondary, and tertiary amines; and cyclic amines, such as piperidine, morpholine, and piperazine; as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
Acidic compounds of the present invention can form base salts with various pharmacologically acceptable cations. Examples of such salts include the alkali metal or alkaline- earth metal salts and particularly, the sodium and potassium salts, which can be prepared using conventional techniques. The chemical bases suitable as reagents in preparing the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of the present invention. Such non- toxic base salts include those derived from such
pharmacologically acceptable cations as sodium, potassium calcium and magnesium, etc. These salts can be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, preferably under reduced pressure. Alternatively, they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together, and then evaporating the resulting solution to dryness in the same manner as before. In either case, stoichiometric quantities of reagents are preferably employed in order to ensure completeness of reaction and maximum yields of the desired final product.
To treat or prevent diseases or conditions caused or mediated by HIV, a pharmaceutical composition, comprising at least one of the compounds of the present invention, is administered in a pharmaceutically acceptable formulation prepared by combining a therapeutically effective amount of the compound with one or more pharmaceutically suitable carriers including diluents, excipients and auxiliaries that facilitate processing of the active compounds into a pharmaceutically acceptable formulation. Carriers employed may be either solid or liquid. Exemplary solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary liquid carriers are syrup, peanut oil, olive oil, water and the like. Similarly, the inventive compositions may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate or the like. Further additives or excipients may be added to achieve the desired formulation properties. For example, a bioavailability enhancer, such: as Labrasol®, Gelucire® or the like, or formulator, such as CHIC (carboxy- methylcellulose), PG (propyleneglycol), or PEG (polyethyleneglycol), may be added. Gelucire®, a semi-solid vehicle that protects active ingredients from light, moisture and oxidation, may be added, e.g., when preparing a capsule formulation.
If a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form, or formed into a troche or lozenge. The amount of solid carrier may vary, but generally will be from about 25 mg to about lg. If a liquid carrier is used, the preparation may be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or nonaqueous liquid suspension. The inventive compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g., parenteral or oral
administration.
To obtain a stable water-soluble dose form, a pharmaceutically acceptable salt of a compound of the present invention may be dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3 M solution of succinic acid or citric acid. If a soluble salt form is not available, the agent may be dissolved in a suitable co-solvent or combinations of co-solvents. Examples of suitable co-solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0-60% of the total volume. In an exemplary embodiment, a compound of the present invention is dissolved in DMSO and diluted with water. The composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution.
Pharmaceutical preparations for oral use can be obtained using a solid excipient in an admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl- cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
The pharmaceutical compositions, comprising the compounds of the present invention may also contain suitable solid- or gel- phase carriers or excipients. These carriers and excipients may provide marked improvement in the bioavailability of poorly soluble drugs. Examples of such carriers or excipients include calcium carbonate, calcium, phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Furthermore, additives or excipients such as Gelucire®, Capryol®, Labrafil®, Labrasol®, Lauroglycol®, Plurol®, Peceol®, Transcutol® and the like may be used. Further, the pharmaceutical composition may be incorporated into a skin patch for delivery of the drug directly onto the skin.
Methods of prophylaxis and treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques.
The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral bioavailability, increase solubility to allow administration by injection, alter metabolism or alter rate of excretion {Pharmacokinetic Optimization in Drug Research, Testa, B. et al, 2001, Wiley-VCH, VCHA).
The pharmaceutical compositions of this invention may be administered orally, intravenously, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally, or via an implanted reservoir and are preferably administered orally or parenterally. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically acceptable carriers, adjuvants or vehicles. The term "parenteral" or "parenterally" as used herein includes sub-cutaneous, intracutaneous, intra-venous, intra-muscular, intra-articular, intra-synovial, intra-sternal, intra-thecal, intra-lesional and intracranial injection or infusion techniques.
For intravenous administration, pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solutions. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
Pharmaceutical compositions of the invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspension and solutions. In the case of tablets for oral and carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavouring and/or coloring agents may be added.
Pharmaceutical compositions of the invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax, and polyethylene glycols.
The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
It will be appreciated that the actual dosages of the agents of this invention will vary according to the particular agent being used, the particular composition formulated, the mode of administration, and the particular site, host, and disease being treated. Those skilled in the art using: conventional dosage-determination tests in view of the experimental data for a given compound may ascertain optimal dosages for a given set of conditions. For oral administration, an exemplary daily dose generally employed will be from about 0.001 to about 1000 mg/kg of body weight, with courses of treatment repeated at appropriate intervals, preferably between 0.01 and about 25 mg/kg body weight per day, and more preferably between about 0.5 and about 25 mg/kg body weight per day of the active ingredient compound useful in the prevention and treatment of viral infection, including HIV infection.
Furthermore, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 7 0 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 to about 500 mg, or from about 100 mg to about 500 mg. Additionally, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a
pharmaceutically acceptable salt or solvate thereof, in an amount from about 0.5 w/w % to about 95 w/w %, or from about 1 w/w % to about 95 w/w %, or from about 1 w/w % to about 75 w/w %, or from about 5 w/w % to about 75 w/w %, or from about 10 w/w % to about 75 w/w %, or from about 10 w/w % to about 50 w/w %.
The pharmaceutical compositions of this invention may be administered as a continuous infusion, once per day, multiple times per day (e.g., from about 1 to about 5 times per day), once per week, twice per week, three times per week, every other day, every other week or as determined by the practicing clinician. Such
administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the patient treated and the particular mode of administration. A typical preparation will contain from about 5% to about 75% active compound (w/w). Preferably, such preparations contain from about 20% to about 50% active compound.
Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered if necessary or desired. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained or maintained. When the symptoms have been alleviated to the desired level, treatment should cease, at least in principle. Patients may, however, require intermittent treatment on a long-term basis, upon any recurrence of disease symptoms, especially for AIDS.
As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician.
With respect to the compounds of the present invention, the particular pharmaceutical formulation, the dosage, and the number of doses given per day to a mammal requiring such treatment, are all choices within the knowledge of one of ordinary skill in the art and can be determined without undue experimentation. For example, see "Guidelines for the Use of Antiretroviral Agents in HIV-1 Infected Adults and Adolescents," United States Department of Health and Human Services, available at http://aidsinfo.nih.gov/Guidelines/Default.aspx?MenuItem=Guidelines.
The compounds of this invention are also useful as commercial reagents which effectively bind to HIV integrase. As commercial reagent, the compounds of this invention, and their derivatives, may be used to block integration of a target DNA molecule by integrase, or may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications. These and other uses which characterize commercial integrase inhibitors will be evident to those of ordinary skill in the art.
The compounds of the present invention can be used alone (monotherapy) or administered in combination with one or more other HIV-inhibiting agents including but not limited to additional compounds of the invention or entry inhibitors, protease inhibitors, reverse transcriptase inhibitors, fusion inhibitors, and integrase inhibitors, examples of which are described below and known to the skilled artisan.
In one example, the compounds of the invention can be used in combination with an additional HIV integrase inhibitor. Compounds that effectively inhibit HIV integrase may provide improved antiviral agents and compositions for treating HIV infection (Wai, J.S. et al., J. Med. Chem. 43:4923-4926 (2000); Grobler, J. et al, PNAS 99: 6661- 6666 (2002); Pals, G.C.G. et al, J. Med. Chem. 45: 3184-3194 (2002); Young, S. D., Curr. Opin. Drug Disc. & Devel. 4(4): 402-410 (2001);
Godwin, C.G. et al, J. Med. Chem. 45: 3184-3194 (2002); Opar, A. Nature Reviews, Drug Discovery, vol. 6, p. 258-259, (2007)). Other integrase inhibitors known in the art include those disclosed in patent applications WO200510305, WO2004039803, WO2004067531, WO2008/048538, WO2003082881 WO2007000043, and
WO09146555.
The compounds of this invention may be administered in combination with antiviral agents which target other steps in the retroviral replication cycle. For example, the co-administered antiviral agent can be one that targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA. Antiviral agents targeting such early life cycle events include, didanosine (ddl), zalcitabine (ddC), stavudine (d4T), zidovudine (AZT), polysulfated polysaccharides, sT4 (soluble CD4)— which blocks attachment or adsorption of the virus to host cells— and other compounds which block binding of virus to CD4 receptors on CD4-bearing T-lymphocytes. Other retroviral reverse transcriptase inhibitors, such as derivatives of AZT, may also be co-administered with the compounds of this invention to provide therapeutic treatment for substantially reducing or eliminating viral infectivity and the symptoms associated therewith. Examples of other antiviral agents include ganciclovir, dideoxycytidine, trisodium phosphonoformiate, eflornithine, ribavirin, acyclovir, alpha interferon and trimenotrexate. Additionally, non-ribonucleoside inhibitors of reverse transcriptase, such as TIBO, efavirenz, nevirapine or delavirdine, may be used to potentiate the effect of the compounds of this invention, as may viral uncoating inhibitors, inhibitors of trans-activating proteins such as tat or rev, or inhibitors of the viral protease. These compounds may also be co-administered with other inhibitors of HIV integrase.
Combination therapies according to this invention may exert an additive or combined inhibitory effect on HIV replication because each therapeutic agent of the combination acts on a different site of HIV replication or a synergistic effect. For example, the use of such combination therapies also advantageously enables a reduction in the dosage of each anti-retroviral agent, compared to administration of either agent alone as a monotherapy, while providing an equivalent or better therapeutic or prophylactic effect. Administration of lower doses of each therapeutic agent often reduces or even eliminates side effects or toxicity relative to monotherapy. Furthermore, combination therapies reduce the potential for the development of viral resistance to the agents administered compared to monotherapy.
Preferred combination therapies include the administration of a compound of this invention with AZT, 3TC, ddl, ddC, d4T, tenofovir, FTC, Combivir® (AZT/3TC combination), abacavir, efavirenz, nevirapine and delavirdine. The compounds of this invention may also be co-administered with other HIV protease inhibitors such as saquinavir, indinavir, nelfinavir, ritonavir, atazanavir, darunavir, and amprenavir. Combination of the compounds of this invention with such protease inhibitors may increase the therapeutic or prophylactic against various HIV viral mutants, HIV quasi species or other closely related viruses.
The compounds of this invention may be administered in combination with nucleoside or non-nucleoside retroviral reverse transcriptase inhibitors (e.g.
derivatives of AZT or HIV aspartyl protease inhibitors) HIV-entry inhibitors, HIV integrase inhibitors, immuno-modulators (e.g., bropirimine, anti-human alpha interferon antibody, IL-2, GM-CSF, methionine enkephalin, interferon alpha, diethyldithiocarbante, tumor necrosis factor, naltrexone and rEPO); antibiotics (e.g., pentamidine isethionate), vaccines or a combination thereof.
Administration of the compounds of this invention in combination therapies with other agents to patients may be sequential or concurrent. Furthermore, pharmaceutical or prophylactic compositions of this invention may include a combination of an integrase inhibitor compound of the present invention and another therapeutic or prophylactic agent or HIV-inhibiting agent. Additional examples of agents useful for treating AIDS and HIV and suitable for combination therapies with the compounds of this invention are listed in Tables 1 and 2 below.
Table 1
Antiviral Drug Manufacturer Indication
097 (non-nucleoside reverse Hoechst/Bayer HIV infection, AIDS.ARC transcriptase inhibitor)
Amprenavir 141 W94 GW Glaxo Wellcome HIV infection, AIDS, ARC
141 (protease inhibitor)
GW 1592 (RT inhibitor) Glaxo Wellcome HIV infection, AIDS, ARC
Acemannan Carrington Labs (Irving, TX)
Acyclovir Burroughs Wellcome HIV infection, AIDS, ARC, in
combination with AZT
AD-439 Tanox Biosystems HIV infection, AIDS, ARC
AD-519 Tanox Biosystems HIV infection, AIDS, ARC
Adefovir dipivoxil AL-721 Gilead Sciences Ethigen (Los HIV infection, ARC, PGL HIV positive,
Angeles, CA) AIDS
Alpha Interferon HIV in Glaxo Wellcome Kaposi's sarcoma
combination w/Retrovir
Ansamycin LM 427 Adria Laboratories (Dublin, OH) ARC
Erbamont (Stamford, CT)
Antibody which Neutralizes Advanced Biotherapy Concepts AIDS, ARC
pH Labile alpha aberrant (Rockville, MD)
Interferon
AR 177 Aronex Pharm HIV infection, AIDS, ARC
Beta-fluoro-ddA Nat'l Cancer Institute AIDS-associated diseases
BMS-232623 (CGP-73547) Bristol-Myers Squibb/ ovartis HIV infection, AIDS, ARC
(protease inhibitor)
BMS-234475 (CGP-61755) Bristol-Myers Squibb/Novartis HIV infection, AIDS, ARC
(protease inhibitor)
CI- 1012 Warner-Lambert HIV-1 infection
Cidofovir Gilead Science CMV retinitis, herpes, papillomavirus Curdlan sulfate AJI Pharma USA HIV infection
Cytomegalovirus Immune Medlmmune CMV retinitis
globin
Cytovene Ganciclovir Syntex Sight threatening CMV peripheral, CMV retinitis
Darunavir (PI) Tibotec/J&J HIV infection, AIDS, ARC
Delaviridine (RT inhibitor) Pharmacia-Upjohn HI V infection, AIDS, ARC
Dextran Sulfate Ueno Fine Chem. Ind. Ltd. AIDS, ARC, HIV positive asymptomatic
(Osaka, Japan)
ddCDideoxycytidine Hoffman-La Roche HIV infection, AIDS, ARC ddl Dideoxyinosine Bristol-Myers Squibb HIV infection, AIDS, ARC; combination with AZT/d4T
DMP-450 (protease inhibitor) AVID (Camden, NJ) HIV infection, AIDS, ARC
Efavirenz (DMP 266) BMS & DuPont Merck HIV infection, AIDS, ARC
EL10 Elan Corp, PLC (Gainesville, HIV infection
Figure imgf000027_0001
0041
Reyataz® (atazanavir) Bristol-Myers Squibb HIV infection, AIDS
Fuzeon (Enfuvirtide, T-20) Rocheh Trirneris HIV infection, AIDS, viral fusion
inhibitor
Trizivir® GSK HIV infection, AIDS
aietra® Abbott HIV infection, AIDS, ARC
Table 2
Immuno-Modulator Manufacturer Indication
Carrington Labs Inc. (Irving,
Acemannan TX) AIDS, ARC
Alpha-2 Interferon Schering Plough Kaposi's sarcoma w/AZT, AIDS
Wyeth-Ayerst/Pharmacia
AS-101 Bropirimine Upjohn AIDS, advanced AIDS
American Cyanamid Lederle
CL246,738 Labs AIDS, Kaposi's sarcoma
Elan Corp PLC (Gainesville,
EL10 GA) HIV infection
FP-2139 Fuki ImmunoPharm Blocks HIV fusion with CD4+ cells
ARC, in combination w/TNF (tumor
Gamma Interferon Genentech necrosis factor)
Granulocyte Macrophage
Colony Stimulating Factor Genetics Institute/Sandoz AIDS
Granulocyte Macrophage
Colony Stimulating Factor I loethst- Rousse IMm in unex AIDS
Granulocyte Macrophage
Colony Stimulating Factor Schering-Plough AIDS, combination w/AZT
HIV Core Particle
Immunostimulant Rorer Seropositive HIV
IL-2 Interleukin-2 Cetus AIDS, in combination w/AZT
AIDS, ARC, HIV, in combination
IL-2 Interleukin-2 Hoffman-LaRoche/Immunex w/AZT
IL-2 Interleukin-2 (aldeslukin) Chiron AIDS, increase in CD4 cell counts
IMERG-2 Imreg (New Orleans, LA) AIDS, Kaposi's sarcoma, ARC, PGL
Immune Globulin Intravenous Cutter Biological (Berkeley,
(human) CA) Pediatric AIDS, in combination w/AZT
IMREG-1 Imreg (New Orleans, LA) AIDS, Kaposi's sarcoma, ARC, PGL
Imuthiol Diethyl Dithio
Carbamate Merieux Institute AIDS, ARC
Kaposi's sarcoma, AIDS, in combination
Interferon Alfa 2a Hoffinan-La Roche w/AZT ARC
TNI Pharmaceutical (Chicago,
Methionine-Enkephalin II-) AIDS, ARC
MTP-PE Muramyl-f ripeptide
Granulocyte Colony Kaposi's sarcoma AIDS in combination
Stimulating Factor Ciba-Geigy Corp./Amgen w/AZT
rCD4 Soluble Human CD4 AIDS, ARC Recombinant AIDS, ARC rCD4-IgG Genentech hybrids
Recombinant Soluble Human
CD4 Biogen AIDS, ARC
Remune Immune Response Corp. Immunotherapeutic Anti-infectives that may be used in combination with the compounds of the present invention include, but are not limited to, atovaquone, azithromycin, clarithromycin, trimethoprim, trovafloxacin, pyrimethamine, daunorubicin, clindamycin with primaquine, fluconazole, pastill, ornidyl, eflornithine pentamidine, rifabutin, spiramycin, intraconazole-R 1211, trimetrexate, daunorubicin, recombinant human erythropoietin, recombinant human growth hormone, megestrol acetate, testerone, and total enteral nutrition.
Antifungals that may be used in combination with the compounds of the present invention include, but are not limited to, anidulafungin, C31G, caspofungin, DB-289, fluconzaole, itraconazole, ketoconazole, micafungin, posaconazole, and voriconazole.
[0454] Other compounds that may be used in combination with the compounds of the present invention include, but are not limited to, acmannan, ansamycin, LM 427, AR177, BMS-232623, BMS-234475, CI-1012, curdlan sulfate, dextran sulfate, STOCRINE EL 10, hypericin, lobucavir, novapren, peptide T octabpeptide sequence, trisodium phosphonoformate, probucol, and RBC-CD4.
In addition, the compounds of the present invention may be used in combination with anti-proliferative agents for the treatment of conditions such as Kaposi's sarcoma. Such agents include, but are not limited to, inhibitors of metallo-matrix proteases, A-007, bevacizumab, BMS-275291, halofuginone, interleukin-12, rituximab, paclitaxel, porfimer sodium, rebimastat, and COL-3.
Compounds of the present invention may be administered in combination with an additional agent or pharmaceutical composition that increases the bioavailability or slows the metabolism of the compounds. Agents or pharmaceutical compositions that may increase the bioavailability or slow the metabolism of the compounds herein include inhibitors of at least one isoform of the cytochrome P450 (CYP450) enzymes, preferably CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4. Suitable agents that may be used to inhibit CYP 3A4 include, but are not limited to, delavirdine and ritonavir. Such combinations may be administered such that a compound or compounds of the present invention are present in a single formulation or in the form of separate formulations that may be administered sequentially with an appropriate period of time in between or simultaneously. The choice of whether to include the compound or compounds of the present invention in the same formulation as the additional agent or agents is within the knowledge of one of ordinary skill in the art. Preparation of Intermediates and Compounds
Four general approaches (synthetic schemes) were use to prepare the compounds of the present invention.
The first approach (Scheme I) starts from pyridoxine which is modified to produce Intermediate aldehyde I using methodologies described in Paul et al. J. Med. Chem., 1977, 20 p745. Reaction of this intermediate with aryl Grignard reagents leads to an intermediate alcohol II. Through a series of controlled oxidations, an intermediate keto-ester VII is obtained which can be selectively reduced to chiral alcohol-ester VIII using established chiral borane chemistry. The final compound IX is then formed by deprotection of the acetonide with formic acid followed by conversion of the ester to a hydroxamic acid with hydroxylamine.
The second approach (Scheme 21 consists of the deprotection and oxidation of the intermediate methyl 5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxylate XII, as described in WO09146555, to produce the corresponding aldehyde-ester XIV. This intermediate is then reacted with Grignard reagents to give the (rac)alcohol ester VIII intermediate. Compound VII can then be converted to the final product IX, XI following the procedures described in Scheme 1.
The third approach (Scheme 3) starts from methyl 3-(benzyloxy)-5-formyl-4- methylpicolinate XV an intermediate obtained by the selective benzylation of methyl 3-(hydroxy)-5-formyl-4-methylpicolinate as described in WO09146555. Further reaction with an aryl Grignard reagent leads to an alcohol ester XVI. This alcohol XVI is acylated with acetic anhydride and reduced to the methylene XVIII through catalytic hydrogenation. Further transformation to the final compound XIX can be carried out using the methods described in Scheme 1.
The fourth (Scheme 4) approach begins with the alcohol-ester intermediate XVI obtained in Scheme 3. This alcohol is oxidized to the ketone XX and reacted with hydroxylamine. Deprotection followed by chromatography provided two compounds, the bis and mono addition products of hydroxylamine XXIII and XXII respectively. Scheme 1
Figure imgf000031_0001
Scheme 2
Grignard
Figure imgf000032_0001
Scheme 3
Figure imgf000032_0002
Scheme 4
Figure imgf000033_0001
General Procedures
Preparative chromatography was performed by flash chromatography, using silica gel 60 (EM Science) with the indicated solvent systems and positive air pressure, to allow for a proper rate of elution, or with a Biotage SP4rM automated chromatography system. Detection of the compounds was carried out by exposing eluted plates (analytical or preparative) to iodine, UV light and/or treating analytical plates with a 2% solution of jo-anisaldehyde in ethanol containing 3% sulfuric acid and 1% acetic acid, followed by heating. Alternatively, analytical plates can be treated with a 0.3% ninhydrin solution in ethanol containing 3% acetic acid and/or a CAM solution made of 20 g (NH4)6Mo7024 and 8.3 g Ce(S04)2 polyhydrate in water (750 mL) containing concentrated sulfuric acid (90 mL).
Unless otherwise indicated: all starting materials were purchased from a commercial source such as Aldrich Co. or Sigma Co; melting points (mp) were determined on a Buchi 530 melting point apparatus in capillary tubes (uncorrected); mass spectra were recorded on a Hewlett Packard LC/MSD 1100 system APCI either in negative mode or positive mode; nuclear magnetic resonance ( MR) spectra were recorded on a Bruker AMX 400 equipped with a reversed or QNP probe.
Samples were dissolved in deutero-chloroform (CDC13), deuterium oxide (D20) or deutero-dimethylsulfoxide (DMSO-d^) for data acquisition and tetramethylsilane was used as internal standard. Chemical shifts (δ) are expressed in parts per million (ppm), coupling constants (J) are expressed in hertz (Hz) and multiplicities are denoted as s for singlet, d for doublet, dd for doublet of doublets, t for triplet, q for quartet, quint for quintet, m for multiplet, and br s for broad singlet.
EXAMPLES
Example 1: Preparation of (R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)- N,3-dihydroxy-4-(hydroxymethyl)picolinamide
Step la: preparation of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H- [1,3 Jdioxinof 4, 5-c]pyridin-5-yl)methanol
1.2g of 50 mmol magnesium turnings were washed, placed in a flame dried 3N RBF and 30 mL ether was added. 8.3 g of 40 mmol (l-Bromo-4-Fluoro-3- Chloro)-benzene was diluted in 20 mL ether and added drop wise to the Mg turnings under argon. The Grignard reaction was then initiated by heating briefly and was complete after ~ 2h. 6.2g of 30mmol 2, 8-dimethyl-4H-[l,3]dioxino[4,5-c]pyridine- 5-carbaldehyde was diluted in 25 mL of ether and added drop wise at RT to the Grignard reagent.
A thick paste formed near the end of the addition of the Grignard reagent. After completion the mixture was sonicated briefly to break up clumps of the Mg salt. After 30 min the mixture was quenched by addition of aq. NH4C1, filtered to remove MgO and partitioned. The ether phase was collected and dried with MgSCH then evaporated yielding thick oil. The thick oil was then diluted with EtOAc whereupon the product crystallized and precipitated. 0.1 volume of hexane was added and the mixture cooled. Filtration gave a clean white product (6.4 g), and further standing of the concentrated liquor gave another 1.2g (20.8 mmol) (70% yield).
Step lb: preparation of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H- [l,3]dioxino[4,5-c]pyridin-5-yl)methanone
6.6g of (4-fluorophenyl)(2,2,8-trimethyl-4H-[l ,3]dioxino[4,5-c]pyridin-5- yl)methanol was dissolved with heat in 150mL of CHCI3. 12g of Mn02 was added to this solution and the suspension was refluxed for 2h. The Mn(¾ was then filtered over a 1cm pad of silica gel and the clear organic phase evaporated to yield pure (3- Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridin-5- yl)methanone.
Step lc: preparation of 5-(3-Chloro-4-fluorobenzoyl)-2,2,8-trimethyl-4H- [1,3 ]dioxino[4, 5-cJpyridine 7 -oxide 6.2g of (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5- c]pyridin-5-yl)methanone (lb) was dissolved in 130 mL CHCI3 and 6g of 70% meta- Chloroperoxybenzoic acid (mCPBA) was added in one portion. The reaction mixture was stirred for 25 min to the completion of the reaction. 100 mL of 1M K2C03 was added and the organic phase extracted. The remaining aqueous phase was extracted with two 50mL portions of CHC13 and the organic phases were combined. 25g of CaC03 was added to the organic phase and stirred for 15 min. Filtration of the CaC03 gave a light yellow solution of clean product. Evaporation afforded 6.2 g of the desired product (lc).
Step Id: Preparation of (3-Chloro-4-fluorophenyl)(8-(hydroxymethyl)-2,2- dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-c ]pyridin-5-yl)methanone
5.5g of5-(4-fluorobenzoyl)-2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridine 7- oxide was dissolved in 20 mL dichloromethane (DCM) in 5 mL of Trifluoroacetic anhydride (TFAA) followed by vigorous boiling, /hen the boiling subsided an additional 30 mL of TFAA was added and the sol! >n was stirred overnight (14h).
TLC revealed completion of the reaction (Rf EtOAc; (s.m. 0.2), product 0.9). The DCM was removed by evaporation and the solvent switched to MeOH (100 mL). This solution was stirred for 30 min after which evaporation affording thick oil. Toluene was added and removed under high vacuum yielding a foam containing the desired product (Id).
Step le: Preparation of 5-(3-Chloro-4-fluorobenzoyl)-2,2-dimethyl-4H- [1,3 Jdioxinof 4, 5-c ]pyridine-8-carbaldehyde
(4-fluorophenyl)(8-(hydroxymethyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5- c]pyridin-5-yl)methanone (total yield from step Id) was added to 200 mL of CHC13 and 15g Mn02. The resulting suspension was refluxed for 4h to completion. Mn02 was then removed by filtration through Celite and washed 2X with CHCL3.
Evaporation of the solvent gave 5g of crude product which was used without further purification.
Step If: preparation of methyl 5-(3-Chloro-4-fluorobenzoyl)-2,2-dimethyl-4H- [1,3 ]dioxino[ 4, 5-c ]pyridine-8-carboxylate
4.0g (11.5 mrnol) of crude 5-(4-fluorobenzoyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carbaldehyde was added to a solution of 4g potassium trimethylsilanoate in 150 mL MeOH, followed by 4g of I2 powder. This mixture was stirred for lh at RT. The methanol was then removed and replaced by EtOAc which was then extracted vs. saturated NaHC03 containing metabisulfite (5% lvol). The organic phase was dried and evaporated, residual oil was dissolved in hot hexane and activated charcoal was added. This mixture was filtered hot yielding a concentrated light yellow solution that was allowed to cool to 4°C whereupon a solid separated. 4.0g of this solid was then filtered, dried and dissolved in hot TBME (also with a few drops of DCM to clarify). This solution was then cooled at -10°C for lh. An off- white solid was filtered off and dried overnight to yield 2.6g (60% yield) of desired product (If). The supernatant was allowed to evaporate giving a sticky solid containing the desired crude product.
Step lg: Preparation of (R)-methyl 5-((3-Chloro-4- fluorophenyl) (hydroxy)methyl)-2, 2-dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-cJpyridine-8- carboxylate
100 mg of R- methyl (CBS) was dissolved in 10 mL THF. 3 mL 1.0 M borane THF was then added and the mixture cooled to -20°C with an argon atmosphere. 1 g of the ketone was dissolved in 10 mL THF and added drop wise over 1.5 h with the temperature slowly warming to RT. stirring was left for 2h. 10 mL of anhydrous MeOH was then added and the mixture heated to ~50°C to quench the reaction (H2 evolution). The solvent was evaporated and replaced with anhydrous MeOH, stirred for 30 min and once again evaporated.
Step lh: preparation of (R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)- N,3-dihydroxy-4-(hydroxymethyl)picolinamide
The crude product from step lg, (R)-methyl 5-((3-Chloro-4- fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H-[l ,3]dioxino[4,5-c]pyridine-8- carboxylate, was dissolved in 4 mL of pyridine and 1.5 mL of 50% hydroxylamine in water producing a clear yellow solution that was then heated to 80°C for lh (complete conversion). The aqueous pyridine was then evacuated by vacuum distillation and the residual oil dissolved in EtAc. Extraction with 10% citric acid removed residual pyridine and hydroxylamine. The organic phase became cloudy as deprotection of the acetonide was initiated. The EtAc was removed and the residual oil dissolved in 3mL 70% formic acid. This solution was left to rest at RT for 30 min upon which completion of the deprotection was noted. The oil was then diluted with 20 mL of water and 70 mL of EtAc and the product was extracted into the organic phase. A black aqueous layer formed and was extracted with one 50mL portion of EtAc. Following this extraction the organic phase was concentrated (to 20 mL) and extracted with 0. IN NaOH (2 χ 20 mL). The aqueous layer was then removed and washed with hexane. The aqueous layer was again removed and acidified until complete precipitation of the product (acid pH) was observed. The precipitate was then extracted into EtAc and the aqueous layer back extracted. Evaporation of the solvent gave a foam product. MeCN (lmL) was added to dissolve followed by hexane producing a biphasic mixture. Stirring for 30 min gave a crystalline powder precipitate which was filtered off yielding 0.56g off-white powder.
Ή-NMR (400 MHz, dmso) : δ = 13.01 (s,lH), 12.0 (s, 1H), 9.51 (s 1H), 9.02 (t 1H), 8.11 (s, 1H), 7.60 (d 1H), 7.40 (m, 2H), 5.1 (s, 1H), 4.48 (s, 2H); MS-ESI m/z 343 [MH]+.
Example 2: Preparation of 5-(4-fluorobenzyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide
Step 2a: preparation of methyl 5-((4-fluorophenyl)(hydroxy)methyl)-2,2- dimethyl-4H-[l, 3 ]dioxino[4, 5-c ]pyridine-8-carboxylate
A 4-fluorophenyl magnesium bromide solution (0.438 mL of 1M in tetrahydrofuran, 30.0 mmol, 1.1 eq) at -78 °C was added to a methyl 5-formyl-2,2- dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8-carboxylate solution (0.100 g, 0.398 mmol, 1 eq. in 5.0 mL of tetrahydrofurane). The reaction mixture was stirred for 20 min. at -78 °C. A saturated bicarbonate solution was added and reaction mixture was extracted with ethyl acetate. The organic phases were combined, dried over magnesium sulphate and concentrated under reduced pressure yielding 0.065 g of crude product methyl 5-((4-fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H- [l ,3]dioxino[4,5-c]pyridine-8-carboxylate 2a (47% yield) as a white solid.
Ή-NMR. MS-ESI m/z 348 [MH]+.
Step 2b: preparation of methyl 5-((4-fluorophenyl)(acetoxy)methyl)-2,2- dimethyl-4H-[ 1, 3 ]dioxino[4, 5-c ]pyridine-8-carboxylate
Trietylamine (0.03 lg, 0.300 mmol, 3 eq) and acetic anhydride (0.01 lg, 0.11 1 mmol, 1.1 eq) were added to a solution of methyl 5-((4- fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8- carboxylate 2a in 4.0 mL of dichloromethane. This reaction mixture was stirred for 1 hour at room temperature followed by addition of a saturated bicarbonate solution. The reaction mixture was then extracted with ethyl acetate and the organic phases were combined and concentrated under reduced pressure yielding 0.039 g of crude product 2b (100% yield) as a white solid.
^- MR. MS-ESI m/z 390 [MH]+.
Step 2c: preparation of methyl 5-(4-fl orobenzyl)-2,2-dimethyl-4H- [1,3 Jdioxinof 4, 5-c Jpyridine-8-carboxylate
0.039 g of Methyl 5-((4-fluorophenyl)(acetoxy)methyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxylate (0.100 mmol, 1 eq) and 5 mg of 10% Pd/C were added to 4.0 mL of methanol and stirred under atmospheric hydrogen overnight. The catalyst was filtered and the crude product was purified by silica gel (40% ethyl acetate/hexane) yielding 0.016 g of the desired product 2c (50% yield) as a white solid.
MS-ESI m/z 332 [MH]+.
Step 2d: preparation of 5-(4-fluorobenzyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide
5.0 mL of a hydroxylamine solution (50 wt. % in water) was added to a solution of 0.015 g of methyl 5-(4-fluorobenzyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5- c]pyridine-8-carboxylate (0.045 mmol, 1 eq) in 5.0 mL of tetrahydrofuran. This reaction mixture was stirred at reflux 6hrs. The pH was adjusted to 6 and reaction mixture was extracted with ethyl acetate. Organic phases were combined, dried over magnesium sulphate and concentrated under vacuum to give 0.006 g of the desired product (2d) (40% yield) as a white solid.
Ή-NMR (400MHz, MeOD) : δ = 8.17 (s, 1H), 7.44 (m, 2H), 7.09 (t, 2H), 4.78 (s, 2H), 4.58 (s, 2H); MS-ESI m/z 293 [MH]+.
Example 3: Preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy- 4-(hyd roxy methy l)picolinamide
Hydroxylamine 50% in 3.0 mL of water was added to methyl 5-((4- fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8- carboxylate 2a (0.030 g, 0.086 mmol, 1 eq) in 5.0 mL of tetrahydrofuran. The reaction mixture was stirred at reflux overnight. The pH was adjusted to 6 and reaction mixture was extracted with ethyl acetate. The resulting organic phases were combined, dried over magnesium sulphate and concentrated under vacuum. The crude product was purified by Biotage reverse phase chromatography yielding 0.015 g of 3 (58% yield) as a white solid, δ = 8.19 (s, 1H), 7.46 (m, 2H), 7.09 (t, 2H), 5.08 (s, 1H), 4.58 (s, 2H); MS-ESI m/z 309 [MH]+.
Example 4: Preparation of 5-((4-fluorophenyI)(hydroxy)methyl)-N-hydroxy- N,2,2-trimethyl-4H-[1 ]dioxino[4,5-c]pyridine-8-carboxamide
1.8 mL of lithium bis(trimethylsilyl)amide solution (1.809 mmol, 9 eq, 1 in tetrahydrofuran) was added to 0.041 g of N-methylhydroxylamine hydrochloride (0.249 mmol, 1.1 eq.) in 5.0 mL of tetrahydrofuran (-78 °C) and stirred for 10 min at 78 °C. 0.070 g of methyl 5-((4-fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxylate solution (0.201 mmol, 1 eq.) in 3.0 mL of tetrahydrofuran was then added followed by stirring for 30 min at -78 °C. Water was then added to the reaction mixture followed by extraction with ethyl acetate. The resulting organic phases were then combined and concentrated under reduced pressure yielding 0.01 g of crude product (14% yield) as a white solid.
MS-ESI m/z 363 [MH]+.
Example 5: Preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-rN,3-dihydroxy- 4-(hydroxymethyl)-N-methyIpicolinamide
2.0 mL of formic acid was added to 5-((4-fluorophenyl)(hydroxy)methyl)-N- hydroxy-N,2,2-trimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8-carboxamide (0.010 g, 0.028 mmol, 1 eq). This reaction mixture was stirred at room temperature for 10 min. The formic acid was then concentrated under vacuum yielding 0.005 g of the desired product (5) (56% yield) as a yellow oil.
MS-ESI m/z 323 [MH]+..
Example 6; Preparation of 5-((4-fluorophenyI)(hydroxy)methyl)-3-hydroxy-4- (hydroxymethyl)-N-methoxypicoIinamide
Step 6a: preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N-methoxy-2,2- dimethyl-4H-[ 1, 3 ]dioxino[ 4, 5-c Jpyridine-8-carboxamide
Lithium bis(trimethylsilyl)amide (2.25 mL of a 1 M solution in
tetrahydrofuran, 2.25 mmol, 6 eq. at -78 °C) was added to 0.034 g of methoxylamine hydrochloride (0.413 mmol, 1.1 eq) in 5.0 mL of tetrahydrofuran. This reaction mixture was stirred for 10 min. and a solution of 0.130 g methyl 5-((4- fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5-c]pyridine-8- carboxylate (0.375 mmol, 1 eq) in 5.0 mL of tetrahydrofuran was added. The reaction mixture was stirred for 30 min at -78 °C and a saturated ammonium chloride solution was added. The reaction mixture was then extracted with ethyl acetate and the resulting organic phases were combined and concentrated under reduced pressure yielding 0.136 g of crude product 6a (100% yield) as a white solid.
MS-ESI m/z 390 [MH]+.
Step 6b: preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-3-hydroxy-4- (hydroxymethyl)-N-methoxypicolinamide
2.0 mL of formic acid was added to 0.136 g of the product of step 6a (0.376 mmol, 1 eq) and stirred at room temperature 1 hr. The formic acid was then concentrated under vacuum yielding 0.105 g of a yellow oil containing the desired product 6
(88%).
MS-ESI m/z 323 [MH]+.
Example 7; Preparation of 5-(4-fluorobenzyI)-N,3-dihydroxy-4- methylpicolinamide
Step 7a: preparation of methyl 3-(benzyloxy)-5-((4- fluorophenyl)(hydroxy)methyl)-4-methylpicolinate
1.0 g of methyl 3-(benzyloxy)-5-formyl-4-methylpicolinate (3.50 mmol, 1 eq) was dissolved in 20.0 mL of tetrahydrofuran and 4.2 mL of a 4-fluorophenyl magnesium bromide solution (1M in tetrahydrofuran, 4.20 mmol, 1.2 eq) was added at -78 °C over a period of lhr. The reaction mixture was then stirred for 20 min. -78 °C. A saturated bicarbonate solution was added and reaction mixture was extracted with ethyl acetate. The resulting organic phases were combined, dried over magnesium sulphate and concentrated under reduced pressure. The crude product was purified by silica gel (50% ethyl acetate/hexane) yielding 0.70 g of colourless oil containing the desired product 7a (53% yield).
MS-ESI m/z 382 [MH .
Step 7b: preparation of methyl 5-(acetoxy(4fluorophenyl)methyl)-3- (benzyloxy)-4-methylpicolinate
0.70 g of 5-(4-fluorobenzyl)-N,3-dihydroxy-4-methylpicolinamide was dissolved in 10.0 ml of dichloromethane and 0.28 lg of triethylamine (2.78 mmol, 5 eq.), 2.0 mg of DMAP and 0.170g of acetic anhydride (1.669 mmol, 3 eq.) were added to the solution. This reaction mixture was stirred 1 hour at room temperature and a saturated solution of bicarbonate was added. The reaction mixture was extracted with ethyl acetate and organic phases were combined and concentrated under reduced pressure yielding 0.260 g of crude product, 7b, (93% yield) as a white solid.
MS-ESI m/z 424 [MH]+.
Step 7c: preparation of methyl 5-(4-fluorobenzyl)-3-hydroxy-4- methylpicolinate
The product of step 7b (0.260 g, 0.615 mmol, 1 eq) and 10% Pd/C (20 mg) in methanol (4.0 mL) were stirred under an atmospheric hydrogen overnight. The catalyst was then filtered and the crude product was purified by silica gel (40% ethyl acetate/hexane) to give 0.169 g of 7c (100%) as a white solid.
MS-ESI m/z 276 [MH]+.
Step 7d: preparation of 5-(4-fluorobenzyl)-N,3-dihydroxy-4- methylpicolinamide
5.0 mL of hydroxylamine solution (50 wt. % in water) was added to a solution of 0.169 g of methyl 5-(4-fluorobenzyl)-3-hydroxy-4-methylpicolinate 7c (0.615 mmol, 1 eq) in 5.0 mL of tetrahydrofuran. The reaction mixture was stirred at reflux overnight. The pH was adjusted to 6 and reaction mixture was extracted with ethyl acetate. The organic phases were combined, dried over magnesium sulphate and concentrated under vacuum. The crude product was purified by Biotage reverse phase chromatography yielding 0.120 g of the desired product 7 (71% yield) as a white solid.
MS-ESI m/z 277 [MH]+.
Example 8: Preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy- 4-methyIpicolinamide
Step 8a: preparation of 3-(benzyloxy)-5-((4-fluorophenyl)(hydroxy)methyl)-N- hydroxy-4-methylpicolinamide
5.0 mL of a hydroxylamine solution (50 wt. % in water) was added to a solution of 0.239 g of methyl 3-(benzyloxy)-5-((4-fluorophenyl)(hydroxy)methyl)-4- methylpicolinate (0.627 mmol, 1 eq.; product of step 7a, example 7) in 5.0 mL of tetrahydrofuran. The reaction mixture was stirred at reflux overnight followed by extraction with ethyl acetate. The resulting organic phases were combined, dried over magnesium sulphate and concentrated under vacuum yielding 0.200 g of the desired product 8a (84% yield) as a white solid. MS-ESI m/z 383 [MH]+.
Step 8b: preparation of 5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- methylpicolinamide
3-(benzyloxy)-5-((4-fluorophenyl)(hydroxy)methyl)-N-hydroxy-4- methylpicolinamide (0.1 10 g, 0.288 mmol, 1 eq) and 10% Pd/C (5 mg) were dissolved in 4.0 mL of methanol and stirred under atmospheric hydrogen for 1 hour. The catalyst was then filtered and reaction mixture was concentrated under vacuum. The crude product was purified by Biotage reverse phase chromatography yielding 0.055 g of the desired product 8 (65% yield) as a white solid.
MS-ESI m/z 293 [MH]+.
Example 9: Preparation of 5-(4-fluorobenzoyl)-N,3-dihydroxy-4- methylpicolinamide
Step 9a: preparation of methyl 3-(benzyloxy)-5-(4-fluorobenzoyl)-4- methylpicolinate
2-iodoxybenzoic acid (0.577g, 2.06 mmol, 3 eq.) was added to a solution of 0.239 g of methyl 3-(benzyloxy)-5-((4-fluorophenyl)(hydroxy)methyl)-4- methylpicolinate (product of step 7a, example 7) in 250 mL of ethyl acetate (0.627 mmol, 1 eq.) and stirred at reflux for 1 nr. The solid was then filtered and the solution concentrated. The crude product was purified by silica gel (10% ethyl
acetate/hexane) yielding 0.222 g of the desired product 9a (85% yield) as a white solid.
MS-ESI m/z 380 [MH]+.
Step 9b: preparation of 3-(benzyloxy)-5-(4-fluorobenzoyl)-N-hydroxy-4- methylpicolinamide
5.0 mL of hydroxylamine solution (50 wt. % in water) was added to a solution of 0.222 g of methyl 3-(benzyloxy)-5-(4-fluorobenzoyl)-4-methylpicolinate (9a) in 5.0 mL of tetrahydrofuran (0.586 mmol, 1 eq). The reaction mixture was stirred at reflux overnight followed by extraction with ethyl acetate. The resulting organic phases were combined, dried over magnesium sulphate and concentrated under vacuum yielding 0.200g of the desired product 9b (89 % yield) as a white solid. MS-ESI m/z 430 [MH]+. Example 10: Preparation of (E)-5-((4-fluorophenyI)(hydroxyimino)methyl)-N,3- dihydroxy-4-methyIpicolinamide
0.090 g of 3-(benzyloxy)-5-(4-fluorobenzoyl)-N-hydroxy-4- methylpicolinamide (9b; 0.237 mmol, 1 eq.) and 5 mg of 10% Pd/C were dissolved in 4.0 mL of methanol and stirred under an atmospheric hydrogen for 1 hr. The catalyst was then filtered and reaction mixture was concentrated under vacuum. The crude product was purified by Biotage reverse phase chromatography yieldingO.015 g of 5- (4-fluorobenzoyl)-N,3-dihydroxy-4-methylpicolinamide (the product step 9b, example 9) (22% yield) and 0.015 g of the corresponding oxime (E)-5-((4- fluorophenyl)(hydroxyimino)methyl)-N,3-dihydroxy-4-methylpicolinarnide (10) (22% yield), both as a white solid.
MS-ESI z 291 [MH]+
MS-ESI m/z 306 [MH]+
Example 11: Preparation of (S)-5-((3-Chloro-4- fluorophenyl)(hydroxy)methyI)-N -dihydroxy-4-(hydroxymethyl)picolinamide
The procedure described in Example 1 was followed using (S)-CBS as a chiral reducing agent in step lg.
Table 3: Listing of Compounds in Examples
Compound Name
(R)-5-((3-Chloro-4-fiuorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxyrnethyl)picolinamide
step la (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridin-5- yl)rnethanol
step lb (3-Chloro-4-fluorophenyl)(2,2,8-trimethyl-4H-[l,3]dioxino[4,5-c]pyridin-5- yl)methanone
step lc 5-(3-Chloro-4-fluorobenzoyl)-2,2,8-trimethyl-4H-[l,3]dioxino[4,5- cjpyridme 7-oxide
step Id (3-Chloro-4-fluorophenyl)(8-(hydroxymethyl)-2,2-dimethyl-4H- [l,3]dioxino[4,5-c]pyridin-5-yl)methanone
step le Step e 5-(3-Chloro-4-fluorobenzoyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5- c]pyridine-8 -carbaldehyde
step If methyl 5-(3-ChIoro-4-fluorobenzoyl)-2,2-dimethyl-4H-[l,3]dioxino[4,5- c]pyridine-8-carboxylate
step lg (R)-methyl 5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-2,2-dimethyl-4H- [1 ,3] diox ino [4,5 -c]pyridine-8-carboxylate
step lh (R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide
Figure imgf000044_0001
Example 12: Biological Evaluation. In Vitro Integrase Inhibition Assay
IC50 was determined for the compounds of the inventions based on data generated in strand transfer assays. The IC50 is a measure of the ability of the compounds tested to inhibit the integration of 3 '-processed oligonucleotides by recombinant HIV- 1 integrase. Strand transfer assays were performed essentially as described in Hazuda, D. J.; Felock, P.; Hastings, J. C; Pramanik, B.; Wolfe, A. J. Virol. 1997, 71, 7005-7011). Donor DNA (1.5 pmol/well), biotinylated on the 5' end of the strand processed by integrase, was immobilized onto streptavidin-coated microtiter plates. Recombinant integrase (250 ng/well) was assembled onto the immobilized donor oligonucleotide in reaction buffer (20 mM Hepes, pH 7.6, 5 mM B-mercaptoethanol, 50 ug/mL bovine serum albumin) containing 30 mM MnCl2. Excess enzyme was removed, and the complexes were washed extensively prior to the addition of the target DNA substrate. The target DNA (0.75 pmoles/well) substrate was labeled on each 3' end with FITC. After strand transfer, the FITC-labeled products were detected using an anti-FITC antibody conjugated with alkaline phosphatase (Roche) and a chemi-luminescent substrate (Tropix CSPD with Sapphire II enhancer, Applied Biosystems). The assay was performed in a final concentration of 10% DMSO. To specifically evaluate inhibition of strand transfer, compounds were added after assembly, just prior to the addition of the target DNA.
The results of the integrase strand transfer assay are reported as IC50 values. IC50 values were determined using a sigmoidal dose-response equation. The formula used for calculating % inhibition was: % Inhibition = [1 -(sample counts/average of positive control)]* 100. The percent inhibition of HIV- 1 integrase activity was graphed against the log of the compound concentration (M). Using GraphPad Prism or ActivityBase (IDBS) software IC50 was determined using following sigmoidal dose-response equation:
Y = (A+((B-A)/(l+((C/XrD))))
Where A is the lower plateau (-0%), B is the higher plateau (-100%), C is the IC50, D is the slope, X is the compound concentration (M), and Y is the % inhibition.
Inhibition of strand transfer, as determined by their IC50, demonstrates that the compounds of the present invention inhibit HIV integrase and have IC50S similar to that of Raltegravir, a marketed HIV integrase inhibitor, and L-708906, an integrase inhibitor currently in clinical development (Table 4). Table 4: Strand Transfer Assay IC5o Data
Compound ST IC50
Raltegravir (MK-0518) 0.065
L-708906 (Merck) 0.045
Compound of Example 1 0.056
Compound of Example 2 0.035
Compound of Example 11 0.023
Example 13: Antiviral Efficacy
The antiviral efficacies of the integrase inhibitor compounds of the invention were evaluated based on EC 50 measures obtained from two different in vitro HIV infection assay using cultured MT-4 cells: (1) a multi-cycle infection where cells were infected with wild type NL-4.3) and (2) a single-cycle infection where the cells were infected with a luciferase-bearing, envelope defective (env-) NL-4.3 virus pseudotyped with HIV-1 env (HXBc2.
The incubation period for the multi-cycle infection assay was 6 days. Cell viability (cytoprotection) and EC50 were determined using the colorimetric MTT assay (A. J. Japour et al, Antimicrobial Agents and Chemotherapy, 37, 1095-1 101, 1993 and R. Pauwels et al. Journal of Virological Methods, 20, 309-321 , 1988).
The incubation period for the single-cycle infection assay was 48 hours. EC50 was determined, as described by Chen et al., Journal of Virology, Feb. 1994, Vol. 68, No. 2, p. 654-660, based on measures of luciferase signal over a range of drug concentrations.
The results of these assays are shown in Table 5 and integrase inhibitors of the invention were prepared using the synthetic methods described in Schemes 1-15; and the examples described herein. The reference numbers of the compounds listed in Table 5 (Ex. No.) correspond to the example numbers of examples 1 to 11 described above. These data demonstrate the antiviral efficacy of the compounds of the invention as integrase inhibitors and for treatment of HIV infection and AIDS. The compounds tested display potent antiviral activity (EC50 < 100 nM) and are selective cellular integrase. Furthermore, similar antiviral activity was observed for these compounds when the HIV-1 envelope was replaced with VSV-G, validating that the compounds of the invention are post-entry inhibitors. Table 5: Results of Cytoprotection-Cytotoxicity Assay
Figure imgf000047_0002
Example 14: Effect of Protein Binding on Antiviral Activity
The multi-cycle antiviral activity in the absence and presence of human serum was determined by p24 ELISA measurement after 6 days of NL4.3 virus infection.
The results, showing a moderate effect of protein binding on antiviral activity, are provided in Table 6.
Table 6: Effect of Protein Binding on Antiviral Activity
Figure imgf000047_0001
Other Embodiments
The examples, synthetic schemes and procedures provided herein are for the purpose of illustration only. They are not intended to be exhaustive or to limit the scope of the invention to the specific examples, synthetic schemes, and procedures described herein. Although the invention has been described with reference to several embodiments, it will be understood by one of ordinary skill in the art that various modifications can be made without departing from the spirit and the scope of the invention, as set forth in the claims. Other embodiments are in the claims.
What is claimed is:

Claims

Claims
1. A compound of formula I,
Figure imgf000049_0001
I
wherein:
X is H or OH;
Y is H or OH;
Ri is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, C] -6 alkyl, Ci-6 fluoroalkyl, or benzyl;
R4 is H, Ci-6 alkyl, or benzyl; and
R5 is H or C1-6 alkyl; or
pharmaceutically acceptable salts or solvates thereof. 2. A compound according to claim 1 and of formula la,
Figure imgf000049_0002
la
wherein:
X is OH;
Y is H or OH;
Ri is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, C].6 alkyl or benzyl; R4 is H, Q.6 alkyl or benzyl; and
R5 is H or Ci-6 alkyl.
3. A compound according to claim 1 and of formula lb,
Figure imgf000050_0001
lb
wherein:
X is OH;
Y is H or OH;
Ri is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, Ci-6 alkyl or benzyl;
R4 is H, d- alkyl or benzyl; and
R5 is H or Ci-6 alkyl.
A compound
Figure imgf000050_0002
Ic
wherein:
X is O or N-OH;
Y is H or OH;
Ri is H or halogen (F, CI, Br, I);
R2 is H or halogen (F, CI, Br, I);
R3 is H, Ci-6 alkyl or benzyl;
R4 is H, Ci-6 alkyl or benzyl; and R is H or C 1-6 alkyl; or
pharmaceutically acceptable salts or solvates thereof.
5. A compound selected from:
(R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide;
5-(4-fluorobenzyl)-N,3-dihydroxy-4-(hydroxymethyl)picolinamide;
5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4-
(hydroxymethyl)picolinamide;
5-((4-fluorophenyl)(hydroxy)methyl)-N-hydroxy-N,2,2-trimethyl-4H- [l,3]dioxino[4,5-c]pyridine-8-carboxamide;
5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4-(hydroxymethyl)-N- methylpicolinamide;
5-((4-fluorophenyl)(hydroxy)methyl)-3-hydroxy-4-(hydroxymethyl)-N- methoxypicolinamide;
5-(4-fluorobenzyl)-N,3-dihydroxy-4-methylpicolinamide;
5-((4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4-methylpicolinamide; 5-(4-fluorobenzoyl)-N,3-dihydroxy-4-methylpicolinamide;
(E)-5-((4-fluorophenyl)(hydroxyimino)methyl)-N,3-dihydroxy-4- methylpicolinamide; and
(S)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide; or
pharmaceutically acceptable salts or solvates thereof.
6. The compound of claim 1 selected from the group consisting of:
(R)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4- (hydroxymethyl)picolinamide;
5-(4-fluorobenzyl)-N,3-dihydroxy-4-(hydroxymethyl)picolinamide; and
(S)-5-((3-Chloro-4-fluorophenyl)(hydroxy)methyl)-N,3-dihydroxy-4-
(hydroxymethyl)picolinamide.
7. A pharmaceutical composition comprising a compound according to any one of claims 1 to 6 and a pharmaceutically acceptable carrier of diluent.
8. A pharmaceutical composition comprising a compound according to any one of claims 1 to 6, at least one additional HIV-inhibiting agent, and a pharmaceutically acceptable carrier of diluent.
9. Use of a compound according to any one of claims 1 to 6 or a
pharmaceutically acceptable salt thereof, in the manufacture of a drug for treatment of HIV infection.
10. Use of a compound according to any one of claims 1 to 6 or a
pharmaceutically acceptable salt thereof, in the manufacture of a drug for treatment of acquired immune deficiency syndrome (AIDS) or AIDS-related complex.
11. Use of a compound according to any one of claims 1 to 6 or a
pharmaceutically acceptable salt thereof, in the manufacture of a drug for treatment of hepatitis C infection.
12. A method of treating or preventing an HIV infection in a mammal, said
method comprising administering to said mammal a compound according to any one of claims 1 to 6 in an amount effective for the treatment or prevention of said HIV infection.
13. A method of treating or preventing AIDS or AIDS-related complex in a
mammal, said method comprising administering to said mammal a compound according to any one of claims 1 to 6 in an amount effective for the treatment or prevention of said AIDS or AIDS-related complex.
14. A method of inhibiting HIV replication in a mammal, said method comprising administering to said mammal a compound according to any one of claims 1 to 6 in an amount effective to inhibit HIV replication in a mammal.
15. A method of inhibiting HIV replication in a cell, said method comprising contacting said cell with a compound according to any one of claims 1 to 6 in an amount sufficient to inhibit HIV replication.
16. The method of any one of claims 12-15, said method further comprising
administering to said mammal or contacting said cell with at least one additional HIV inhibiting agent.
17. The method of claim 16, wherein said HIV inhibiting agent is selected from the group consisting of an entry inhibitor, a protease inhibitor, a reverse transcriptase inhibitor, a fusion inhibitor, and an integrase inhibitor.
18. The method of claim 12, 14, or 15, wherein said HIV is resistant to at least one HIV inhibiting agent.
PCT/CA2011/000041 2010-01-22 2011-01-13 Derivatives of pyridoxine for inhibiting hiv integrase WO2011088549A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP11734279.0A EP2526091A4 (en) 2010-01-22 2011-01-13 Derivatives of pyridoxine for inhibiting hiv integrase
JP2012549212A JP2013517302A (en) 2010-01-22 2011-01-13 Pyridoxine derivatives for inhibiting HIV integrase
CN2011800066516A CN102834381A (en) 2010-01-22 2011-01-13 Derivatives of pyridoxine for inhibiting HIV integrase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29745010P 2010-01-22 2010-01-22
US61/297,450 2010-01-22

Publications (1)

Publication Number Publication Date
WO2011088549A1 true WO2011088549A1 (en) 2011-07-28

Family

ID=44306336

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2011/000041 WO2011088549A1 (en) 2010-01-22 2011-01-13 Derivatives of pyridoxine for inhibiting hiv integrase

Country Status (6)

Country Link
US (2) US8283366B2 (en)
EP (1) EP2526091A4 (en)
JP (1) JP2013517302A (en)
CN (1) CN102834381A (en)
TW (1) TW201129360A (en)
WO (1) WO2011088549A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020237374A1 (en) * 2019-05-28 2020-12-03 Montreal Heart Institute Picolinic acid derivatives and use thereof for treating diseases associated with elevated cholesterol

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5401652B2 (en) * 2006-09-21 2014-01-29 タイメッド バイオロジクス インコーポレイテッド Protease inhibitor
US8283366B2 (en) 2010-01-22 2012-10-09 Ambrilia Biopharma, Inc. Derivatives of pyridoxine for inhibiting HIV integrase
JP2021501210A (en) * 2017-10-30 2021-01-14 モントリオール ハート インスティテュート How to Treat Elevated Plasma Cholesterol

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009146555A1 (en) * 2008-06-04 2009-12-10 Ambrilia Biopharma Inc. Hiv integrase inhibitors from pyridoxine
WO2010122980A1 (en) * 2009-04-20 2010-10-28 田辺三菱製薬株式会社 Novel thyroid hormone β receptor agonist

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998011073A1 (en) * 1996-09-10 1998-03-19 Pharmacia & Upjohn Company 8-hydroxy-7-substituted quinolines as anti-viral agents
CA2280518A1 (en) 1997-02-06 1998-08-13 Deborah L. Zink Hiv integrase inhibitors
US6124327A (en) * 1997-07-29 2000-09-26 Merck & Co., Inc. HIV integrase inhibitors
US6403347B1 (en) * 1998-02-03 2002-06-11 Merck & Co., Inc. HIV integrase inhibitors
US6110716A (en) 1998-02-04 2000-08-29 Merck & Co., Inc. HIV integrase inhibitors
US6262055B1 (en) * 1998-06-03 2001-07-17 Merck & Co., Inc. HIV integrase inhibitors
WO1999062520A1 (en) * 1998-06-03 1999-12-09 Merck & Co., Inc. Hiv integrase inhibitors
US6380249B1 (en) * 1998-06-03 2002-04-30 Merck & Co., Inc. HIV integrase inhibitors
US6306891B1 (en) * 1998-06-03 2001-10-23 Merck & Co., Inc. HIV integrase inhibitors
WO1999062513A1 (en) 1998-06-03 1999-12-09 Merck & Co., Inc. Hiv integrase inhibitors
AU3118200A (en) * 1998-12-14 2000-07-03 Merck & Co., Inc. Hiv integrase inhibitors
US6245806B1 (en) * 1999-08-03 2001-06-12 Merck & Co., Inc. HIV integrase inhibitors
WO2001027309A1 (en) * 1999-10-13 2001-04-19 Merck & Co., Inc. Hiv integrase inhibitors
WO2001098248A2 (en) * 2000-06-16 2001-12-27 Bristol-Myers Squibb Company Hiv integrase inhibitors
US6541515B2 (en) * 2000-08-09 2003-04-01 Merck & Co., Inc. HIV integrase inhibitors
EP1326611B1 (en) 2000-10-12 2007-06-13 Merck & Co., Inc. Aza- and polyaza-naphthalenyl-carboxamides useful as hiv integrase inhibitors
US6506786B2 (en) * 2001-02-13 2003-01-14 Pharmacor Inc. HIV protease inhibitors based on amino acid derivatives
EP1441734B1 (en) 2001-10-26 2007-02-28 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Dihydroxypyrimidine carboxamide inhibitors of hiv integrase
CN1700918B (en) 2001-10-26 2011-06-08 P·安杰莱蒂分子生物学研究所 N-substituted hydroxypyrimidinone carboxamide inhibitors of HIV integrase
MXPA04005623A (en) 2001-12-12 2004-12-06 Squibb Bristol Myers Co Hiv integrase inhibitors.
DE60315796T2 (en) 2002-01-17 2008-06-05 Merck & Co., Inc. HYDROXYNAPHTHYRIDINONCARBOXYL ACID AMIDES SUFFICIENT AS INHIBITORS OF THE HIV INTEGRASE
CA2374362C (en) * 2002-03-04 2008-01-22 Brent Richard Stranix Urea derivatives as hiv aspartyl protease inhibitors
AU2003248872A1 (en) 2002-07-09 2004-01-23 Bristol-Myers Squibb Company Hiv integrase inhibitors
US20050043370A1 (en) 2002-09-25 2005-02-24 Walker Michael A. HIV integrase inhibitors
WO2004035577A2 (en) 2002-10-16 2004-04-29 Gilead Sciences, Inc. Pre-organized tricyclic integrase inhibitor compounds
US6632816B1 (en) 2002-12-23 2003-10-14 Pharmacor Inc. Aromatic derivatives as HIV aspartyl protease inhibitors
WO2004062613A2 (en) 2003-01-13 2004-07-29 Bristol-Myers Squibb Company Hiv integrase inhibitors
US7037908B2 (en) 2003-04-24 2006-05-02 Bristol-Myers Squibb Company HIV integrase inhibitors
BRPI0409873A (en) 2003-04-28 2006-05-16 Tibotec Pharm Ltd hiv integrase inhibitors
BRPI0507628A (en) 2004-02-11 2007-07-03 Smithkline Beecham Corp pharmaceutically acceptable compound or salt thereof, method of treating a viral infection in a human, use of a compound pharmaceutical composition, and process for preparing a compound
EA200601654A1 (en) 2004-03-09 2007-02-27 Мерк Энд Ко., Инк. HIV INTEGRASE INHIBITORS
CN1930161A (en) 2004-03-09 2007-03-14 P·安杰莱蒂分子生物学研究所 HIV integrase inhibitors
US7820680B2 (en) 2004-03-09 2010-10-26 Merck & Co., Inc. HIV integrase inhibitors
US7619086B2 (en) 2004-03-09 2009-11-17 Merck & Co., Inc. HIV integrase inhibitors
WO2005110415A1 (en) 2004-05-07 2005-11-24 Merck & Co., Inc. Hiv integrase inhibitors
US7115601B2 (en) 2004-05-18 2006-10-03 Bristol-Myers Squibb Company HIV integrase inhibitors
CN101146540A (en) 2004-06-09 2008-03-19 默克公司 HIV integrase inhibitors
US7388008B2 (en) 2004-08-02 2008-06-17 Ambrilia Biopharma Inc. Lysine based compounds
JP2008512442A (en) * 2004-09-07 2008-04-24 ファイザー・インク Inhibitor of HIV integrase enzyme
WO2006103399A1 (en) 2005-03-31 2006-10-05 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Hiv integrase inhibitors
EP1881825B1 (en) 2005-05-10 2013-07-24 Merck Sharp & Dohme Corp. Hiv integrase inhibitors
JP2009503081A (en) 2005-08-04 2009-01-29 スミスクライン ビーチャム コーポレーション HIV integrase inhibitor
WO2007019100A2 (en) 2005-08-04 2007-02-15 Smithkline Beecham Corporation Hiv integrase inhibitors
EP1909578A4 (en) 2005-08-04 2010-07-14 Glaxosmithkline Llc Hiv integrase inhibitors
US20080214503A1 (en) 2005-08-04 2008-09-04 Smithkline Beecham Corporation Hiv Integrase Inhibitors
JP2009513640A (en) 2005-10-27 2009-04-02 メルク エンド カムパニー インコーポレーテッド HIV integrase inhibitor
WO2007059125A2 (en) 2005-11-15 2007-05-24 Bristol-Myers Squibb Company Hiv integrase inhibitors
US7902182B2 (en) 2005-11-16 2011-03-08 Bristol-Myers Squibb Company HIV integrase inhibitors
US8039458B2 (en) 2005-11-17 2011-10-18 Bristol-Myers Squibb Company HIV integrase inhibitors
US20070129379A1 (en) 2005-12-01 2007-06-07 Bristol-Myers Squibb Company Hiv integrase inhibitors
US7547300B2 (en) 2006-04-12 2009-06-16 Icu Medical, Inc. Vial adaptor for regulating pressure
US7893055B2 (en) 2006-06-28 2011-02-22 Bristol-Myers Squibb Company HIV integrase inhibitors
US8465926B2 (en) 2006-08-11 2013-06-18 The Hong Kong University Of Science And Technology Method and system for real time quantification and monitoring of nucleic acid amplification using electroconductive or electrochemically active labels
JP5401652B2 (en) 2006-09-21 2014-01-29 タイメッド バイオロジクス インコーポレイテッド Protease inhibitor
JP2010506913A (en) 2006-10-18 2010-03-04 メルク エンド カムパニー インコーポレーテッド HIV integrase inhibitor
US7763630B2 (en) 2007-06-06 2010-07-27 Bristol-Myers Squibb Company HIV integrase inhibitors
US8129398B2 (en) 2008-03-19 2012-03-06 Bristol-Myers Squibb Company HIV integrase inhibitors
WO2010042392A2 (en) 2008-10-06 2010-04-15 Merck & Co., Inc. Hiv integrase inhibitors
US8383639B2 (en) 2009-10-15 2013-02-26 Bristol-Myers Squibb Company HIV integrase inhibitors
US8283366B2 (en) * 2010-01-22 2012-10-09 Ambrilia Biopharma, Inc. Derivatives of pyridoxine for inhibiting HIV integrase

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009146555A1 (en) * 2008-06-04 2009-12-10 Ambrilia Biopharma Inc. Hiv integrase inhibitors from pyridoxine
WO2010122980A1 (en) * 2009-04-20 2010-10-28 田辺三菱製薬株式会社 Novel thyroid hormone β receptor agonist

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2526091A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020237374A1 (en) * 2019-05-28 2020-12-03 Montreal Heart Institute Picolinic acid derivatives and use thereof for treating diseases associated with elevated cholesterol

Also Published As

Publication number Publication date
EP2526091A1 (en) 2012-11-28
US8283366B2 (en) 2012-10-09
US8664248B2 (en) 2014-03-04
US20110184028A1 (en) 2011-07-28
US20120329827A1 (en) 2012-12-27
JP2013517302A (en) 2013-05-16
CN102834381A (en) 2012-12-19
EP2526091A4 (en) 2013-06-12
TW201129360A (en) 2011-09-01

Similar Documents

Publication Publication Date Title
US8742123B2 (en) HIV integrase inhibitors from pyridoxine
CA2472372C (en) Hydroxynaphthyridinone carboxamides useful as hiv integrase inhibitors
EP1937639B1 (en) Pyridinaminosulfonyl substituted benzamides as inhibitors of cytochrome p450 3a4 (cyp3a4)
JP5765965B2 (en) 1,3,4,8-Tetrahydro-2H-pyrido [1,2-a] pyrazine derivatives and their use as HIV integrase inhibitors
EP1943221B1 (en) Piperazine amidines as antiviral agents
IL295649A (en) Heterocyclic amides useful as protein modulators
US20090105203A1 (en) Compounds for treating viral infections
AU2003216049A1 (en) Hydroxynaphthyridinone carboxamides useful as HIV integrase inhibitors
EA019558B1 (en) Macrocyclic integrase inhibitors
KR20190080951A (en) Pyrimido [5,4-b] indolizine or pyrimido [5,4-b] pyrrolizine compounds, their preparation methods and uses
EP2601174B1 (en) Substituted indole and azaindole oxoacetyl piperazinamide derivatives
US8664248B2 (en) Derivatives of pyridoxine for inhibiting HIV integrase
KR20170032324A (en) Phenyl and tertbutylacetic acid substituted pyridinones having anti-hiv effects
JPH07502972A (en) Antiproliferative substituted naphthalene compounds
CN114761410A (en) Pyrrolopyrimidinone compounds and application thereof
US20230106880A1 (en) Inhibitors of human immunodeficiency virus replication
JP2009511463A (en) Inhibitor of HIV integrase enzyme
US20180094019A1 (en) Crystalline forms of modified triterpenoid hydrochloride salts with hiv maturation inhibitor activity
AU2016290152A1 (en) Pyridin-3-yl acetic acid derivatives as inhibitors of human immunodeficiency virus replication
MXPA06008598A (en) Compositions comprising hiv protease inhibitor and cytochrome p450 enzyme activity inhibitor.

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180006651.6

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11734279

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011734279

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012549212

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE