WO2015035003A1 - Thioether prodrug compositions as anti-hiv and anti-retroviral agents - Google Patents

Thioether prodrug compositions as anti-hiv and anti-retroviral agents Download PDF

Info

Publication number
WO2015035003A1
WO2015035003A1 PCT/US2014/054019 US2014054019W WO2015035003A1 WO 2015035003 A1 WO2015035003 A1 WO 2015035003A1 US 2014054019 W US2014054019 W US 2014054019W WO 2015035003 A1 WO2015035003 A1 WO 2015035003A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
salt
virus
use according
group
Prior art date
Application number
PCT/US2014/054019
Other languages
French (fr)
Inventor
Daniel Appella
Pankaj Kumar
Nathaniel SHANK
Matthew David HASSINK
Original Assignee
The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The United States Of America, As Represented By The Secretary, Department Of Health And Human Services filed Critical The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
Publication of WO2015035003A1 publication Critical patent/WO2015035003A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/62Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • 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

Definitions

  • NCp7 The nucleocapsid protein, NCp7, is a target for the development of new antiretroviral drugs.
  • Current combination therapies against enzymatic or viral fusion processes reduce viral replication to very low levels, but are hindered by the development of viral resistance.
  • new therapies against other viral targets are desired to improve the therapeutic armament against HIV/AIDS.
  • NC nucleocapsid proteins
  • NCp7 The zinc fingers of NCp7 are critical for their multiple roles during the viral replication cycle (recently reviewed in Thomas and Gorelick, Virus Res. 134, 39-63 (2008)). This protein functions as a nucleic acid chaperone, and as such, facilitates RNA
  • NCp7 assists in tRNA annealing to genomic RNA, initiation and processivity of reverse transcription, plus- and minus-strand transfer reactions, 3' DNA processing by integrase, and integrase-mediated strand transfer.
  • NCp7 is encoded in the viral genome as part of the Gag polyprotein, which is specifically cleaved by the viral protease in immature virions to form the four substituent proteins: matrix (MA), capsid (CA), NCp7, and p6, as well as two spacer proteins, SP1 and SP2.
  • MA matrix
  • CA capsid
  • SP1 and SP2 two spacer proteins
  • R is hydrogen or a linear or branched alkyl.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound or salt of the invention and a pharmaceutically acceptable carrier.
  • the invention further provides a method for dissociating a metal ion from a zinc finger-containing protein, the method comprising contacting said zinc finger-containing protein with a compound or salt of the invention.
  • the invention additionally provides method for inactivating a virus, the method comprising contacting a virus with a compound or salt of the invention, whereby contacting the virus with said compound or salt inactivates the virus.
  • the invention also provides a method for inhibiting the transmission of a virus, the method comprising contacting a virus with a compound or salt of the invention, whereby contacting the virus with said compound inhibits the transmission thereof.
  • the activity of the inventive compound against NCp7 leads to inactivation of the virus by blocking infectivity without affecting cellular proteins. Due to the high sequence conservation of the NCp7, it is likely that this target will not evolve resistance to the compound. Mutations that disrupt the zinc finger motifs abolish genomic RNA encapsidation and viral infectivity. In contrast, use of all known anti-HIV drugs approved to date is complicated by the development of resistance and substantial side effects.
  • Figure 1 illustrates a synthetic scheme to prepare a compound 1 of formula (I) in accordance with an embodiment of the invention.
  • Figure 2 depicts the proposed conversion of a compound of formula (I) to a virucidal 2-acetylmercaptobenzamide.
  • Figure 3 depicts the structures of metabolites of a compound of formula (I) when incubated in human plasma, dog plasma, or rat whole blood.
  • D is a thiol metabolite obtained by hydrolysis of the ester group followed by 1 ,2-elimination of formaldehyde.
  • E is an isothiazole obtained by oxidative cyclization of D.
  • F is obtained from D via alpha hydroxylation.
  • G is obtained via hydrolysis of the terminal amide of F.
  • R is hydrogen or a linear or branched alkyl.
  • alkyl means a straight-chain or branched alkyl substituent containing from, for example, 1 to about 6 carbon atoms, preferably from 1 to about 4 carbon atoms, more preferably from 1 to 2 carbon atoms.
  • substituents include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, and the like.
  • R is Ci-Q linear or branched alkyl.
  • the compound is the compound is:
  • the compound is the compound is:
  • the compound or salt of formula (I) exists in the racemic form, in the form of its pure optical isomers, or in the form of a mixture wherein one isomer is enriched relative to the other.
  • the inventive compounds when the inventive compounds have a single asymmetric carbon atom, the inventive compounds may exist as racemates, i.e., as mixtures of equal amounts of optical isomers, i.e., equal amounts of two enantiomers.
  • the compound or salt of formula (I) exists in the form of a single enantiomer, and more preferably in the form of a single levorotatory enantiomer.
  • single enantiomer is intended to mean a compound that comprises more than 50% of a single enantiomer.
  • Single levorotatory enantiomer therefore, means that more than 50% of the levorotatory enantiomer is present along with less than 50% of the dextrorotatory enantiomer (this can also be referred to as a single levorotatory enantiomer), and vice versa (this can also be referred to as a single dextrorotatory enantiomer).
  • a levorotatory enantiomer is defined as an enantiomer having a specific rotation at a light wavelength of 589 nm that is negative.
  • a dextrorotatory enantiomer is defined as having a specific rotation at a light wavelength of 589 nm that is positive.
  • the single enantiomer comprises at least 75% of a single enantiomer (50% enantiomeric excess) ("e.e.”), more preferably at least 90% of a single enantiomer (80% e.e.), still more preferably at least 95% of a single enantiomer (90% e.e.), even more preferably at least 97.5% of a single enantiomer (95% e.e.), and most preferably at least 99% of a single enantiomer (98% e.e.).
  • a single enantiomer 50% enantiomeric excess
  • the compound or salt has more than one chiral center, and can therefore exist as a mixture of diastereomers, preferably the compound or salt exists in the form of a single diastereomer.
  • single diastereomer is intended to mean a compound that comprises more than 50% of a single diastereomer.
  • solvates refers to a molecular complex wherein the solvent molecule, such as the
  • solvates include hydrates, alcoholates such as methanolates and ethanolates, acetonitrilates and the like. These compounds can also exist in polymorphic forms.
  • Contacting refers to the act of bringing components of an interaction (e.g., a compound or salt of formula (I) with a zinc finger protein or a viral protein) or a reaction into adequate proximity such that the interaction or reaction can occur. More generally, as used herein, the term “contacting” can be used interchangeably with the following: bound to, combined with, added to, mixed with, passed over, flowed over, etc.
  • nucleocapsid protein or "NC protein” refers to the retroviral nucleocapsid protein, which is an integral part of the virion nucleocapsid where it coats the dimeric RNA genome, as described by Huang (1997) J Virol. 71 : 4378-4384;
  • NCp7 nucleocapsid protein
  • Gag protein or “Gag-Pol protein” refers to the polyprotein translation product of HIV- 1 or other retroviruses, as described, e.g., by
  • the "Gag protein” is processed by a viral protease to yield mature viral proteins, see, e.g., Humphrey (1997) Antimicrob. Agents & Chemotherapy 41 : 1017-1023; Karacostas (1993) Virology 193: 661-671.
  • retrovirus refers to viruses of the Retroviridae family. These viruses can have dsRNA or ssRNA genomes transcribed by reverse transcriptase, as described by, e.g., P. K. Vogt, "Historical introduction to the general properties of retroviruses.” in Retroviruses, eds. J. M. Coffin, S. H. Hughes and H. E. Varmus, Cold Spring Harbor Laboratory Press, 1997, pp. 1 -26; Murphy et al. (eds.) Archives of
  • Retroviridae Virology/Supplement 10, 586 pp. (1995) Springer Verlag, Wien, N.Y.
  • Retroviridae For a general description of the Retroviridae family, see the Committee on International Taxonomy of Viruses, Virology Division of the International Union of Microbiology Societies viral classifications and taxonomy.
  • Retroviridae family members containing zinc finger motif- containing polypeptides and whose activity, e.g., replication or infectivity, can be inhibited by the compounds of the present invention include, e.g., avian sarcoma and leukosis retroviruses (alpharetroviruses), mammalian B-type retroviruses (betaretroviruses) (e.g., mouse mammary tumor virus), human T-cell leukemia and bovine leukemia retroviruses (deltaretroviruses) (e.g., human T-lymphotropic virus 1 ), murine leukemia-related group (gammmaretroviruses), D-type retroviruses (epsilonretroviruses (e.g., Mason-Pfizer monkey vims), and lentiviruses.
  • alpharetroviruses alpharetroviruses
  • mammalian B-type retroviruses betaretroviruses
  • Lentiviruses include, e.g., bovine, equine feline, ovine/caprine, and primate lentivirus groups, such as human imnmunodeficiency virus type 1 (HIV-1).
  • HIV-1 HIV-2
  • SIV BIV
  • EIAV Visna
  • CaEV CaEV
  • HTLV-1 HTLV-1
  • BLV BLV
  • MPMV MPMV
  • MMTV MMTV
  • RSV MuLV
  • FeLV FeLV
  • BaEV BaEV
  • SSV retroviruses retroviruses.
  • zinc finger refers to a polypeptide motif consisting of cysteines and/or histidines that coordinate metal ions giving rise to structures involved in protein/nucleic acid and/or protein/protein interactions.
  • the compounds or salts of the present invention are capable of modifying the structure of zinc finger peptides in such a way that allows eventual dissociation of the metal ions.
  • the metal ion is a divalent cation, such as those of zinc or cadmium.
  • a zinc finger motif-containing protein is commonly a highly conserved and essential structure in viruses.
  • Zinc finger motifs are found in human papilloma virus (HPV), particularly, HPV E6 and E7 proteins (see, e.g., Ullman (1996) Biochem. J. 319: 229-239) and influenza virus (see, e.g., Nasser (1996) J. Virol. 70: 8639-8644).
  • HPV human papilloma virus
  • retro viridae including avian sarcoma and leukosis retroviruses, mammalian B-type retroviruses, human T-cell leukemia and bovine leukemia retroviruses, D-type retroviruses, and lentiviruses
  • the invariable zinc finger motif is the most highly conserved structure.
  • Retroviral nucleocapsid, Gag and Gag-Pol proteins have zinc finger motifs.
  • the zinc finger motif typically consists of 14 amino acid residues, with four residues being invariant; one exemplary zinc finger motif is described as Cys(X) 2 Cys(X) 4 His(X) 4 Cys and is refen-ed to as a "CCHC zinc finger” (Henderson ( 1981 ) J. Biol. Chem. 256: 8400).
  • Zinc fingers chelate zinc through their histidine imidazole and cysteine thiolates (Berg ( 1986) Science 232: 485; Bess (1992) J. Virol. 66: 840; Chance ( 1992) Proc. Natl. Acad. Sci.
  • CCHC zinc fingers perform essential functions in retroviral infectivity, such as packaging genomic RNA. They are also essential for early events in virus infection.
  • the active form of the compounds of the present invention acts as a zinc fmger inhibitor that does not disrupt all zinc fingers. It has been determined that the compound of interest is specific to the particular configuration of the zinc fingers in NCp7.
  • the cysteine at position 49 is the most reactive and was the expected, most likely target for the nucleophilic attack of the sulfur in this thioether drug since all other molecules that disrupt HIV NCp7 attack at cysteine 49. This is not what happens here.
  • This thiol attacks cysteine at position 36 and the resulting acyl subsequently adds the lysine at position 38. With the presence of tryptophan (W37) and glutamine (Q 45) in key locations, the result is the disruption of the metal coordination and ultimate ejection of the zinc component of NCp7.
  • antiviral activity means a compound has demonstrated some degree of antiviral activity in any assay, e.g., the XTT cytoprotection assay or p24
  • virucidal includes any degree of viral attenuation, including, but not limited to, complete inactivation or killing of a virus.
  • viral infectivity or “index of infectivity” refer to the capacity of virus to pass from an infected cell to an uninfected cell, bringing about productive infection of the uninfected cell.
  • measurements of infectivity may be carried out by the MAGI assay, wherein the uninfected recipient cells are HeLa CD4 HIV
  • the terms “inhibit the transmission of the virus” and “antiviral activity” mean the ability of a compound to negatively affect viral replicative capacity in any way. Such inhibition of transmission, e.g., loss in replicative capacity, can be measured using any means known in the art.
  • a compound inhibits the transmission of the virus (has antiviral activity) if it diminishes a virus' ability to produce progeny, (when in the form of a virion) fuse with a cell, enter a cell, bud from a cell, survive intracellularly or
  • RNA genome extracellularly, reverse transcribe its RNA genome, translate viral proteins, process polyproteins with proteases, effect intracellular assembly of viral components into a capsid, and the like.
  • the ability of a compound of the present invention to inhibit the transmission of a virus is not limited by any chemical or biological mechanism or pathway.
  • a compound can inhibit infectivity or transmission (decrease replicative capacity) of a virus by, e.g.: binding to a nucleocapsid protein, such as NCp7; preventing binding of NCp7 to viral RNA or another nucleic acid; being involved in a specific chemical attack resulting in a stable or transient adduct; promoting the formation of inter- and intramolecular disulfide bonds through consequent destabilization of the NCp7 zinc finger loops; interacting with other conserved or non-conserved residues within the NCp7 protein which results in loss of function; and the like.
  • a nucleocapsid protein such as NCp7
  • NCp7 nucleocapsid protein
  • the present invention is further directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one compound or salt described herein.
  • the pharmaceutically acceptable carrier be one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
  • compositions of the present invention are merely exemplary and are in no way limiting.
  • compositions for parenteral administration that comprise a solution or suspension of the inventive compound or salt dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous isotonic sterile injection solutions.
  • Such solutions can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the compound or salt of the present invention may be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl- l ,3-dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or
  • Oils useful in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanol amine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations can contain preservatives and buffers.
  • such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17.
  • HLB hydrophile-lipophile balance
  • the quantity of surfactant in such formulations will typically range from about 5 to about 15% by weight.
  • Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • Topical formulations including those that are useful for transdermal drug release, are well-known to those of skill in the art and are suitable in the context of the invention for application to skin.
  • Topically applied compositions are generally in the form of liquids, creams, pastes, lotions and gels. Topical administration includes application to the oral mucosa, which includes the oral cavity, oral epithelium, palate, gingival, and the nasal mucosa.
  • the composition contains at least one active component and a suitable vehicle or carrier. It may also contain other components, such as an anti-irritant.
  • the carrier can be a liquid, solid or semi-solid.
  • the composition is an aqueous solution.
  • the composition can be a dispersion, emulsion, gel, lotion or cream vehicle for the various components.
  • the primary vehicle is water or a biocompatible solvent that is substantially neutral or that has been rendered substantially neutral.
  • the liquid vehicle can include other materials, such as buffers, alcohols, glycerin, and mineral oils with various emulsifiers or dispersing agents as known in the art to obtain the desired pH, consistency and viscosity.
  • the compositions can be produced as solids, such as powders or granules. The solids can be applied directly or dissolved in water or a biocompatible solvent prior to use to form a solution that is substantially neutral or that has been rendered substantially neutral and that can then be applied to the target site.
  • the vehicle for topical application to the skin can include water, buffered solutions, various alcohols, glycols such as glycerin, lipid materials such as fatty acids, mineral oils, phosphoglycerides, collagen, gelatin and silicone based materials.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as a therapeutically effective amount of the inventive compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules, (c) powders, (d) suspensions in an appropriate liquid, and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, com starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • the compound or salt of the present invention can be made into aerosol formulations to be administered via inhalation.
  • the compounds are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of active compound are 0.01%-20% by weight, preferably 1%-10%.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • Such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
  • Mixed esters such as mixed or natural glycerides may be employed.
  • the surfactant may constitute 0.1 %-20% by weight of the composition, preferably 0.25%-5%. The balance of the composition is ordinarily propellant.
  • a carrier can also be included as desired, e.g., lecithin for intranasal delivery.
  • aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations may be used to spray mucosa.
  • pressurized propellants such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • non-pressured preparations such as in a nebulizer or an atomizer.
  • Such spray formulations may be used to spray mucosa.
  • the compound or salt of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration may be presented as vaginal rings (i.e., intravaginal rings), pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • formulations presented as vaginal rings provide a slow release formulation of the compound or salt within the vagina over a period of time, for example, over about one week, or over about two weeks, or over about three weeks, or over about one month.
  • vaginal rings can be made of any suitable material, non-limiting examples of which include silicone elastomers and ethylene-co-vinyl acetate.
  • suitable material non-limiting examples of which include silicone elastomers and ethylene-co-vinyl acetate.
  • Non-limiting examples of vaginal rings can be found in, e.g., U.S. Patents 4,155,991 , 5,989,581 , and 6,126,958, the disclosures of which are incorporated totally herein by reference.
  • Other suitable vaginal ring products and formulations suitable for use therewith that are useful in connection with the compound or salt of the present invention will be readily apparent to those of ordinary skill in the medical and pharmaceutical arts.
  • the compound or salt of the present invention may be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.
  • inclusion complexes such as cyclodextrin inclusion complexes, or liposomes.
  • Liposomes serve to target the compounds to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of the inventive compound.
  • Liposomes useful in the present invention include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like.
  • the active agent to be delivered is incorporated as part of a liposome, alone or in conjunction with a suitable chemotherapeutic agent.
  • liposomes filled with a desired inventive compound or salt thereof can be directed to the site of a specific tissue type, for example hepatic cells, where the liposomes then deliver the selected compositions.
  • Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, for example, liposome size and stability of the liposomes in the blood stream.
  • a liposome suspension containing a compound or salt of the present invention may be administered intravenously, locally, topically, etc. in a dose that varies according to the mode of administration, the agent being delivered, and the stage of disease being treated.
  • the invention further provides a method for dissociating a metal ion from a zinc finger-containing protein, the method comprising contacting said zinc finger-containing protein with a compound or salt of the invention.
  • the motif can be an isolated peptide or polypeptide, or, it can be a substructure of a viral protein or a virion.
  • the method includes contacting the zinc finger with a compound of the present invention and subsequently detecting the dissociation of the metal ion from the zinc finger protein.
  • the cation is commonly zinc. Any methodology known in the art can be used to detect the dissociation of the metal ion.
  • Exemplary means include, e.g., capillary electrophoresis, immune-blotting, nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), detecting release of radioactive zinc-65, detecting fluorescence, or detecting gel mobility shift, and other techniques which would be apparent to one of skill upon review of this disclosure. These procedures can be practiced with any protocol known in the art, which are well described in the scientific and patent literature. A few exemplary techniques are set forth below.
  • the invention provides a genus of novel compounds capable of dissociating a metal ion from a zinc finger in vitro
  • detection of the dissociation of the metal ion identifies some of the compounds within the scope of the present invention.
  • a zinc ejection assay can be used as a screen to identify compounds within the scope of the present invention.
  • One strategy for such screening uses the XTT cytoprotection assay to monitor anti-viral activity.
  • Alternative strategies use a Trp37 zinc ejection assay (see, e.g., U.S. Pat. No.
  • NPG N-propyl gallate
  • the zinc finger-containing protein is a viral protein.
  • the viral protein is selected from the group consisting of a nucleocapsid protein, a Gag protein, and a Gag-Pol protein. The contacting of the protein with the compound or salt is performed in vitro or in vivo.
  • the protein is that of a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • the protein is that of an HIV-1 retrovirus.
  • the zinc-finger protein is present in a mammal. In certain preferred embodiments, the zinc-finger protein is that of a retrovirus present in a mammal. In certain embodiments, the zinc-finger protein is present on a surface of a mammal. In certain preferred embodiments, the zinc-finger protein is that of a retrovirus present on a surface of a mammal.
  • the invention further provides a method for inactivating a virus, the method comprising contacting a virus with a compound or salt of the present invention, whereby contacting the virus with said compound or salt inactivates the virus.
  • the contacting can be as described herein.
  • the virus can be any suitable virus.
  • suitable viruses include an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • the virus is an HIV-1 retrovirus.
  • the virus is selected from the group consisting of a retrovirus, an avian sarcoma and leukosis retroviral group, a mammalian B-type retroviral group, a human T cell leukemia and bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group.
  • a retrovirus an avian sarcoma and leukosis retroviral group
  • a mammalian B-type retroviral group a human T cell leukemia and bovine leukemia retroviral group
  • a D-type retroviral group a murine leukemia-related group
  • lentivirus group preferably the contacting of the virus with the compound or salt is performed in vivo.
  • the compound or salt of the invention is administered to inhibit the transmission of the virus.
  • a purpose for inhibiting the transmission of the virus refers to transmission of the virus from an infected individual to another individual.
  • inhibition of the transmission of the virus can be achieved by administration of the compound or salt intra- vaginally or intra-rectally.
  • inhibition of the transmission of the virus can be achieved by administration of the compound or salt parenterally, intrathecally, subcutaneously, or orally.
  • the compound or salt of the invention can be administered to a human as a pharmaceutical formulation.
  • the compound or salt of the invention can be administered to an animal as a veterinary pharmaceutical formulation.
  • the pharmaceutical formulation is administered systemically to the human or animal.
  • the pharmaceutical formulation is administered topically to the human or animal.
  • the term "animal” includes a mammal such as, without limitation, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order
  • Artiodactyla including Bovines (cows) and Swine (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
  • the compound or salt thereof is administered in a dose sufficient to dissociate a zinc ion from a zinc finger-containing protein or for inactivating a virus.
  • doses are known in the art (see, for example, the Physicians ' Desk Reference (2004)).
  • the compounds can be administered using techniques such as those described in, for example, Physicians ' Desk Reference, 58th ed., Thomson PDR (2004).
  • Suitable doses and dosage regimens can be determined by conventional range- finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound of the present invention. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached.
  • the present method can involve the administration of about 0.1 ⁇ g to about 50 mg of at least one compound of the invention or salt thereof per kg body weight of the individual. For a 70 kg patient, dosages of from about 10 ⁇ g to about 200 mg of the compound or salt of the invention would be more commonly used, depending on a patient's physiological response, e.g., as determined by measuring viral load or other measurable parameters related to the viral load of a patient.
  • the method further comprises contacting the virus with another anti -retroviral agent.
  • the anti-retroviral agent can be any suitable anti-retroviral agent.
  • suitable anti-retroviral agents include anti-retroviral agents selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, an entry inhibitor, a maturation inhibitor, and a protease inhibitor.
  • the anti-retroviral agent is a nucleoside analogue which is an AZT, a ddCTP or a ddl.
  • the said another anti-retroviral agent is selected from the group consisting of enfuviritide, maraviroc, delavirdine, etravirine, efavirenz, nevirapine, zidovudine, lamivudine, emtricitabine, didanosine, tenofovir, stavudine, abacavir, raitegravir, dolutegravir, amprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, and nelfinavir, and combinations thereof.
  • the contacting of the virus with the compound or salt is performed on a blood product, blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food, drink, implant or prosthesis.
  • the contacting of the virus with the compound or salt is performed in vitro.
  • the antiviral activity of the inventive compounds can be further assessed via methods disclosed in U.S. Patent 7,528,274, the disclosure of which is totally incorporated herein by reference.
  • the compounds of the invention undergo intracellular hydrolysis and elimination to liberate 2- mercaptobenzamides, which are then acetylated by acetyl-Co-A to produce 2- acetylmercaptobenzamides.
  • Compound B is acetylated to produce the 2-acetylmercaptobenzamide derivative C.
  • the 2-acetylmercaptobenzamides are believed to specifically react with the C-terminal zinc finger of NCp7 and eject its coordinated zinc ion, irreversibly destroying protein function.
  • the compounds of the invention desirably exhibit low toxicity.
  • compound 1 has been shown to have a maximum tolerated dose in rats of greater than 2 g per kg of body weight.
  • the compounds of the invention desirably exhibit oral bioavailability.
  • pharmacokinetic studies in rats indicate that compound 1 is orally bioavailable.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs Banded PBMCs were gently aspirated from the resulting interface and subsequently washed three times with PBS by low speed centrifugation. After the final wash, cells were enumerated by Trypan Blue dye exclusion and re-suspended at 1 x 10 6 cells/mL in RPMI 1640 with 15 % Fetal Bovine Serum (FBS), 2 mmol/L L-glutamine, 2 ⁇ ig/mL PHA-P, 100 U/mL penicillin and 100 ⁇ g/mL streptomycin and allowed to incubate for 48 - 72 hour at 37 °C.
  • FBS Fetal Bovine Serum
  • PBMCs were centrifuged and resuspended in tissue culture medium (RPMI 1640 with 15% FBS, 2 mmol/L L- glutamine, 100 U/mL penicillin, 100 ⁇ ig/mL streptomycin and 3.6 ng/mL recombinant human IL-2).
  • tissue culture medium RPMI 1640 with 15% FBS, 2 mmol/L L- glutamine, 100 U/mL penicillin, 100 ⁇ ig/mL streptomycin and 3.6 ng/mL recombinant human IL-2).
  • the cultures were maintained until use by 1 ⁇ 2 culture volume change with fresh IL-2 containing tissue culture medium every 3 days.
  • Assays were initiated with PBMCs that have been induced to proliferate for 72 hr.
  • PHA-P-stimulated PBMCs from three donors were pooled together to minimize the variability between individual donors, and resuspended in fresh tissue culture medium at 1 x 10 6 cells/mL and plated in the interior wells of a 96-well round bottom microtiter plate at 50 ⁇ . Then, 100 xL of 2x concentrations of compound-containing medium (200, 62.5, 20, 6.25, 2, and 0.625 nM of CTP-51 8 or ATZ) was transferred to the round-bottom 96-well plate containing the cells in 50 ⁇ , of the medium. AZT (1 , 0.32, 0.1 , 0.032, 0.01 , and 0.0032 ⁇ ) was evaluated as an internal assay standard.
  • PBMCs were exposed in triplicate to vims and cultured in the presence or absence of the test material at varying concentrations as described above in the 96-well microtiter plates. After 7 days in culture, HIV-1 replication was quantified in the tissue culture supernatant by measurement of reverse transcriptase activity. Wells with cells and virus alone were used for virus control. Separate plates were identically prepared without virus for drug cytotoxicity studies using the tetrazolium dye XTT.
  • This example illustrates the in vitro metabolism of compounds 1 and 2 in human and dog plasma and in rat whole blood.
  • a “-” indicates that a particular metabolite was not observed, a “++” indicates that the metabolite was present in 1-10% relative HPLC peak area, and a “+++” indicates that the metabolite was present in >10% relative HPLC peak area.
  • This example demonstrates the anti-HIV evaluation of compounds 1 and B in human PBMCs in accordance with an embodiment of the invention.
  • PHA-P stimulated PBMCs from two donors were pooled together and re- suspended in fresh tissue culture medium at 1 x 10 6 cells/mL and plated in the interior wells of a 96 well round bottom microplate at 50 ⁇ ⁇ .
  • a 100 volume of 2X concentrations of compound-containing media were transferred to the round-bottom 96-well plate containing the cells in triplicate.
  • Fifty microliters (50 ⁇ ) of HIV- 1 at a pre-determined dilution was added.
  • Each plate contained cell control wells and virus control wells in parallel with the experimental wells.
  • efficacy was evaluated by measuring the reverse transcriptase in the culture supernatants and the cells were stained with the tetrazolium dye XTT to evaluate cytotoxicity. The results are set forth in Table 4.
  • Compounds 1 and B were solubilized at 40 mM in DMSO and stored at -20° C. Compounds 1 and B were evaluated at concentrations of 4, 2, 1 , 0.5, and 0.25 ⁇ in combination with nine concentrations of an existing HIV inhibitor in at least two replicate antiviral combination assays. The second known anti-HIV agent was evaluated using a high test concentration of two-times the EC 5 o value as determined in the cytoprotection assay. Table 5 lists the class of inhibitor and compound source. Compounds were diluted in cell culture medium at 4 times the high test concentration, serially diluted, and added to the microliter plate in the appropriate format in a volume of 50 ⁇ . per well.
  • CEM-SS cells were passaged in RPMI1640 supplemented with 10% heat inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin and 100 ⁇ g/mL streptomycin in T- 75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1 :2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification were performed using a hemocytometer and Trypan Blue dye exclusion. Cell viability must be greater than 95% for the cells to be utilized in the assay. The cells were resuspended at 5 x 10 4 cells per ml tissue culture medium and added to the drug-containing microtiter plates.
  • the virus used for the cytoprotection assays was the lymphocyte-tropic virus HIV-lrrrs.
  • the virus was obtained from the NIH AIDS Research and Reference Reagent Program and stock virus pools were produced in CEM-SS cells. A pretitered aliquot of virus was removed from the freezer (-80° C) and allowed to thaw slowly to room temperature in a biological safety cabinet. Virus was resuspended and diluted into tissue culture medium such that the amount of virus added to each well in a volume of 50 was the amount determined to yield 85 to 95% cell killing at 6 days post-infection.
  • Each plate contained cell control wells (cells only), virus control wells (cells plus virus), drug toxicity wells (cells plus drug only), drug colorimetric control wells (drug only) as well as experimental wells (drug plus cells plus virus). Samples were tested in triplicate.
  • test plates were stained with the tetrazolium dye XTT (2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-5- [(phenylamino)carbonyi]-2H-tetrazolium hydroxide).
  • XTT-tetrazolium is metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product.
  • XTT solution was prepared daily as a stock of 1 mg/mL RPMI1640.
  • Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/mL PBS and stored in the dark at -20°C.
  • XTT/PMS stock was prepared immediately before use by adding 40 ⁇ PMS per mL of XTT solution. Fifty microliters of XTT/PMS was added to each well of the plate prior to reincubation for 4 hr at 37° C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble formazan product. The plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader.
  • synergy is defined as drug combinations yielding synergy volumes greater than 50 ⁇ 2 % at the 95% confidence interval. Slightly synergistic activity and highly synergistic activity have been defined as yielding synergy volumes of 50 to 100 ⁇ 2 % and > 100 ⁇ 2 %, respectively. Synergy volumes between -50 and 50 ⁇ 2 % are considered additive and synergy volumes less than -50 ⁇ % are considered antagonistic.
  • Combination Therapy Evaluations Compounds 1 and B were evaluated in combination with twenty-three known anti-HIV agents for the inhibition of cytopathic effect in CEM-SS cells using the IIIB strain of HIV- 1. The percent of virus replication inhibition above expected at each concentration for each two-drug combination was calculated at the 95%, 99% and 99.9% confidence interval. The data obtained at the 95% confidence value were plotted three dimensionally.
  • the synergy volumes for the combinations at the 95% confidence interval of two replicate assays are summarized in Table 6 for B.
  • the synergy volumes for the combinations at the 95% confidence interval of two replicate assays are summarized in Tabic 7 for 1.
  • Enfuviritide 57.5A26.8 Slightly 104/-38.7 Synergistic synergistic
  • Lamivudine 27 1 /-36.9 Synergistic 90.1 /0 Slightly synergistic
  • B or 1 in combination with reverse transcriptase inhibitors yielded additive to synergistic interactions.
  • B combinations with delavirdine, efavirenz, nevirapine, zidovudine, tenofovir or stavudine yielded the mean synergy volumes of 80.1 ⁇ 2 %, 84.7 ⁇ 2 %, 74.4 ⁇ 2 %, 1 1 1 ⁇ 2 %, 1 16 ⁇ 2 %, and 87.7 ⁇ 2 %, respectively.
  • ⁇ combinations with etravirine, lamivudine, emtricitabine and didanosine resulted in an additive interaction from two assay replicates.
  • B and 1 resulted in additive interactions.
  • B in combination with dolutegravir demonstrated a synergistic interaction with a mean synergy volume of 95.7 ⁇ 2 %.
  • 1 in combination with dolutegravir resulted in an additive interaction.
  • B or 1 in combination with protease inhibitors resulted in additive to highly synergistic interactions.
  • 1 in combination with tipranavir resulted in an additive interaction from two assay replicates.
  • B yielded mean synergy volumes ranging from 65.1 (saquinavir) to 498 ⁇ % (nelfinavir).
  • 1 in combination with tipranavir or indinavir resulted in additive interactions.
  • 1 yielded mean synergy volumes ranging from 95.1 (saquinavir) to 282 ⁇ 2 % (nelfinavir).
  • This example demonstrates the maximum tolerated dose (MTD) of compound 1 in Sprague-Dawley rats following a dose escalation design via oral administration (Phase A).
  • this example demonstrates the potential for repeat dose toxicity following once daily oral administration of compound 1 for 14 consecutive days (Phase B). In this phase, the reversibility, progression and potential delayed effects were determined following a 14-day recovery period.
  • Parameters evaluated in Phase A included cageside and detailed clinical observations, body weights and body weight changes, and food consumption.
  • Control Article CA; 2% CMC-Na in Water for injection
  • compound 1 were administered once daily for 14 consecutive days via oral administration at a dose volume of 20 mL/kg.
  • Blood samples for clinical pathology analyses were collected from the mam study animals prior to terminal sacrifice on Day 15 and from the recovery animals prior to recovery sacrifice on Day 29. All main study animals were euthanized and necropsied following the final blood collection on Day 15. All recovery study animals were euthanized and necropsied following the final blood collection on Day 29. Parameters evaluated included cageside and detailed clinical observations, body weights and body weight changes, food consumption, clinical pathology (clinical chemistry, hematology, coagulation and urinalysis), absolute and relative organ weights, gross pathology and microscopic pathology.
  • Coagulation There are no compound 1-related effects for any coagulation parameter from blood samples taken on Day 15 and no evidence of a delayed compound 1-related effect for any coagulation parameter from samples taken on Day 29.
  • the gross pathology findings including red multifocal discoloration of mandibular lymph node, were sporadically observed in one female given 500 mg/kg/day and one male given 1000 mg/kg/day. Those changes were considered to be not related to compound 1 treatment, due to low incidence and lack of correlated histopathological findings.
  • Duplicate bone marrow smears were prepared from the sternum of all euthanized rats during the terminal necropsy on Day 15 (2 rats/sex/group) and the recovery necropsy on Day 29 (2 rats/sex/group). All smears from all animals were stained with Wright-Giemsa and evaluated by Pharmaron Pathology Laboratory. Megakaryocyte precursors were estimated as decreased (D), normal (N) or increased (I), based on a semi-quantitative subjective evaluation of megakaryocyte precursor density from the smears.
  • a bone marrow differential count (200 cells) was used for the quantification of myeloid, erythroid, and other cell lineages (including lymphocytes, macrophages/monocytes, and plasma cells).
  • a Myeloid : Erythroid (M:E) ratio was calculated for each animal by dividing the total number of myeloid cells by the total number of erythroid cells. Eosinophilic and basophilic granulocytes were not separated from neutrophilic granulocytes unless there were apparent differences in the proportions of eosinophils and basophils between groups.
  • R is hydrogen or a linear or branched alkyl.
  • a pharmaceutical composition comprising a compound of any one of aspects 1 -4 and a pharmaceutically acceptable earner. 6. The pharmaceutical composition of aspect 5, further comprising another anti- retro viral agent other than the compound of formula (I).
  • composition of aspect 6, wherein the another anti- retroviral agent is a member selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
  • a composition comprising a compound of any one of aspects 1 -4 or a pharmaceutically acceptable salt thereof and further comprising blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food or drink.
  • a method for dissociating a zinc ion from a zinc finger-containing protein comprising contacting said zinc finger-containing protein with a compound of any one of aspects 1-4 and pharmaceutically acceptable salts thereof.
  • NCp7 protein is selected from the group consisting of a nucleocapsid protein, a Gag protein and a Gag-Pol protein.
  • the protein is that of a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • the zinc finger-containing protein is that of a virus selected from the group consisting of an avian sarcoma retroviral group, a mammalian B-type retroviral group, a human T cell leukemia retroviral group, a bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group.
  • a virus selected from the group consisting of an avian sarcoma retroviral group, a mammalian B-type retroviral group, a human T cell leukemia retroviral group, a bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group.
  • HPLC chromatography
  • a method for inactivating a virus comprising contacting a virus with a compound of any one of aspects 1 -4 or a pharmaceutically acceptable salt thereof, whereby contacting the virus with said compound or salt inactivates the virus.
  • the virus is selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1, a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
  • the virus is selected from the group consisting of a retrovirus of an avian sarcoma and leukosis retroviral group, a mammalian B- type retroviral group, a human T cell leukemia and bovine leukemia retroviral group, a D- type retroviral group, a murine leukemia-related group and a lentivirus group.
  • said another anti-retroviral agent is selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
  • nucleoside analogue is an AZT, a ddCTP or a ddl.
  • said another anti-retroviral agent is selected from the group consisting of enfuviritide, maraviroc, delavirdine, etravirine, efavirenz, nevirapine, zidovudine, lamivudine, emtricitabine, didanosine, tenofovir, stavudine, abacavir, raitegravir, dolutegravir, amprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, and nelfinavir.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed are inhibitors of retroviral growth of formula (I), that are useful in treatment of retroviral infections such as HIV. Also disclosed are a composition comprising a pharmaceutically acceptable carrier and at least one compound or salt of the invention, a method for inactivating a virus, a method for dissociating a metal ion from a zinc finger-containing protein method, and a method for inhibiting the transmission of a virus, wherein R is hydrogen or a linear or branched alkyl.

Description

THIOETHER PRODRUG COMPOSITIONS AS ANTI-HIV AND ANTI-RETROVIRAL
AGENTS
CROSS REFERENCE TO A RELATED APPLICATION
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 61/874, 182, filed September 5, 2013, which is incorporated by reference.
BACKGROUND OF THE INVENTION
[0002] The nucleocapsid protein, NCp7, is a target for the development of new antiretroviral drugs. Current combination therapies against enzymatic or viral fusion processes reduce viral replication to very low levels, but are hindered by the development of viral resistance. Thus, new therapies against other viral targets are desired to improve the therapeutic armament against HIV/AIDS.
[0003] The nucleocapsid proteins (NC) of all orthoretroviruses contain zinc fingers with a strictly conserved motif containing seven amino acids including cysteine and histidine. The nucleocapsid of most orthoretroviruses, including HIV-1 NCp7, has two zinc fingers that comprise the primary structural elements of the protein. In addition to the zinc fingers, the surrounding residues in NCp7 are also highly conserved among the clades of HIV-1.
Mutation of any of the zinc-coordinating residues renders the virus non-infectious, including conservative changes at positions that maintain the ability to coordinate zinc.
[0004] The zinc fingers of NCp7 are critical for their multiple roles during the viral replication cycle (recently reviewed in Thomas and Gorelick, Virus Res. 134, 39-63 (2008)). This protein functions as a nucleic acid chaperone, and as such, facilitates RNA
conformation-dependent reactions in the cell. Early after viral infection, NCp7 assists in tRNA annealing to genomic RNA, initiation and processivity of reverse transcription, plus- and minus-strand transfer reactions, 3' DNA processing by integrase, and integrase-mediated strand transfer. NCp7 is encoded in the viral genome as part of the Gag polyprotein, which is specifically cleaved by the viral protease in immature virions to form the four substituent proteins: matrix (MA), capsid (CA), NCp7, and p6, as well as two spacer proteins, SP1 and SP2. As part of Gag, NCp7 is required for packaging of genomic RNA into the new virion and assists the formation of a mature dimerized double-stranded RNA genome.
[0005] While several compounds have been proposed targeting NCp7, none have advanced through clinical trials due to toxicity or lack of specificity. [0006] In view of the foregoing, there is a desire to provide new antiretroviral compounds, in particular, compounds that target NCp7.
BRIEF SUMMARY OF THE INVENTION [0007] The invention provides a compound of formula (I):
(I)
[0008] wherein R is hydrogen or a linear or branched alkyl.
[0009] The invention also provides a pharmaceutical composition comprising a compound or salt of the invention and a pharmaceutically acceptable carrier.
[0010] The invention further provides a method for dissociating a metal ion from a zinc finger-containing protein, the method comprising contacting said zinc finger-containing protein with a compound or salt of the invention.
[0011] The invention additionally provides method for inactivating a virus, the method comprising contacting a virus with a compound or salt of the invention, whereby contacting the virus with said compound or salt inactivates the virus.
[0012] The invention also provides a method for inhibiting the transmission of a virus, the method comprising contacting a virus with a compound or salt of the invention, whereby contacting the virus with said compound inhibits the transmission thereof.
[0013] Advantageously, the activity of the inventive compound against NCp7 leads to inactivation of the virus by blocking infectivity without affecting cellular proteins. Due to the high sequence conservation of the NCp7, it is likely that this target will not evolve resistance to the compound. Mutations that disrupt the zinc finger motifs abolish genomic RNA encapsidation and viral infectivity. In contrast, use of all known anti-HIV drugs approved to date is complicated by the development of resistance and substantial side effects.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S)
[0014] Figure 1 illustrates a synthetic scheme to prepare a compound 1 of formula (I) in accordance with an embodiment of the invention. [0015] Figure 2 depicts the proposed conversion of a compound of formula (I) to a virucidal 2-acetylmercaptobenzamide.
[0016] Figure 3 depicts the structures of metabolites of a compound of formula (I) when incubated in human plasma, dog plasma, or rat whole blood. D is a thiol metabolite obtained by hydrolysis of the ester group followed by 1 ,2-elimination of formaldehyde. E is an isothiazole obtained by oxidative cyclization of D. F is obtained from D via alpha hydroxylation. G is obtained via hydrolysis of the terminal amide of F.
DETAILED DESCRIPTION OF THE INVENTION
[0017] The invention provides a compound of formula (I):
Figure imgf000004_0001
(I)
[0018] wherein R is hydrogen or a linear or branched alkyl.
[0019] Referring now to terminology used generically herein, the term "alkyl" means a straight-chain or branched alkyl substituent containing from, for example, 1 to about 6 carbon atoms, preferably from 1 to about 4 carbon atoms, more preferably from 1 to 2 carbon atoms.
Examples of such substituents include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, and the like.
[0020] In certain embodiments, R is Ci-Q linear or branched alkyl.
[0021] In a preferred embodiment, the compound is the compound is:
Figure imgf000004_0002
[0022] In another preferred embodiment, the compound is the compound is:
Figure imgf000004_0003
[0023] In any of the above embodiments, the compound or salt of formula (I) exists in the racemic form, in the form of its pure optical isomers, or in the form of a mixture wherein one isomer is enriched relative to the other. In particular, in accordance with the present invention, when the inventive compounds have a single asymmetric carbon atom, the inventive compounds may exist as racemates, i.e., as mixtures of equal amounts of optical isomers, i.e., equal amounts of two enantiomers. Preferably the compound or salt of formula (I) exists in the form of a single enantiomer, and more preferably in the form of a single levorotatory enantiomer. As used herein, "single enantiomer" is intended to mean a compound that comprises more than 50% of a single enantiomer. "Single levorotatory enantiomer," therefore, means that more than 50% of the levorotatory enantiomer is present along with less than 50% of the dextrorotatory enantiomer (this can also be referred to as a single levorotatory enantiomer), and vice versa (this can also be referred to as a single dextrorotatory enantiomer). As used herein, a levorotatory enantiomer is defined as an enantiomer having a specific rotation at a light wavelength of 589 nm that is negative. By contrast, a dextrorotatory enantiomer is defined as having a specific rotation at a light wavelength of 589 nm that is positive.
[0024] Preferably, the single enantiomer comprises at least 75% of a single enantiomer (50% enantiomeric excess) ("e.e."), more preferably at least 90% of a single enantiomer (80% e.e.), still more preferably at least 95% of a single enantiomer (90% e.e.), even more preferably at least 97.5% of a single enantiomer (95% e.e.), and most preferably at least 99% of a single enantiomer (98% e.e.).
[0025] When the compound or salt has more than one chiral center, and can therefore exist as a mixture of diastereomers, preferably the compound or salt exists in the form of a single diastereomer. As used herein, "single diastereomer" is intended to mean a compound that comprises more than 50% of a single diastereomer.
[0026] It is further understood that the above compounds may form solvates, or exist in a substantially uncomplexed form, such as the anhydrous form. As used herein, the term "solvate" refers to a molecular complex wherein the solvent molecule, such as the
crystallizing solvent, is incorporated into the crystal lattice. When the solvent incorporated in the solvate is water, the molecular complex is called a hydrate. Pharmaceutically acceptable solvates include hydrates, alcoholates such as methanolates and ethanolates, acetonitrilates and the like. These compounds can also exist in polymorphic forms. [0027] "Contacting" refers to the act of bringing components of an interaction (e.g., a compound or salt of formula (I) with a zinc finger protein or a viral protein) or a reaction into adequate proximity such that the interaction or reaction can occur. More generally, as used herein, the term "contacting" can be used interchangeably with the following: bound to, combined with, added to, mixed with, passed over, flowed over, etc.
[0028] As used herein, the term "nucleocapsid protein" or "NC protein" refers to the retroviral nucleocapsid protein, which is an integral part of the virion nucleocapsid where it coats the dimeric RNA genome, as described by Huang (1997) J Virol. 71 : 4378-4384;
Lapadat-Tapolsky (1997) J. Mol. Biol. 268: 250-260. HIV-l 's nucleocapsid protein is termed "NCp7," see also Demene (1994) Biochemistry 33: 1 1707-1 1716.
[0029] As used herein, the term "Gag protein" or "Gag-Pol protein" refers to the polyprotein translation product of HIV- 1 or other retroviruses, as described, e.g., by
Fehrmann (1997) Virology 235: 352-359; Jacks (1988) Nature 331 : 280-283. The "Gag protein" is processed by a viral protease to yield mature viral proteins, see, e.g., Humphrey (1997) Antimicrob. Agents & Chemotherapy 41 : 1017-1023; Karacostas (1993) Virology 193: 661-671.
[0030] The term "retrovirus" as used herein refers to viruses of the Retroviridae family. These viruses can have dsRNA or ssRNA genomes transcribed by reverse transcriptase, as described by, e.g., P. K. Vogt, "Historical introduction to the general properties of retroviruses." in Retroviruses, eds. J. M. Coffin, S. H. Hughes and H. E. Varmus, Cold Spring Harbor Laboratory Press, 1997, pp. 1 -26; Murphy et al. (eds.) Archives of
Virology/Supplement 10, 586 pp. (1995) Springer Verlag, Wien, N.Y. For a general description of the Retroviridae family, see the Committee on International Taxonomy of Viruses, Virology Division of the International Union of Microbiology Societies viral classifications and taxonomy. Retroviridae family members containing zinc finger motif- containing polypeptides and whose activity, e.g., replication or infectivity, can be inhibited by the compounds of the present invention include, e.g., avian sarcoma and leukosis retroviruses (alpharetroviruses), mammalian B-type retroviruses (betaretroviruses) (e.g., mouse mammary tumor virus), human T-cell leukemia and bovine leukemia retroviruses (deltaretroviruses) (e.g., human T-lymphotropic virus 1 ), murine leukemia-related group (gammmaretroviruses), D-type retroviruses (epsilonretroviruses (e.g., Mason-Pfizer monkey vims), and lentiviruses. Lentiviruses include, e.g., bovine, equine feline, ovine/caprine, and primate lentivirus groups, such as human imnmunodeficiency virus type 1 (HIV-1). Examples of particular species of viruses whose replicative capacity could be inactivated by the compounds of the present invention include HIV-1 , HIV-2, SIV, BIV, EIAV, Visna, CaEV, HTLV-1 , BLV, MPMV, MMTV, RSV, MuLV, FeLV, BaEV, and SSV retroviruses.
[0031] As used herein, the term "zinc finger" refers to a polypeptide motif consisting of cysteines and/or histidines that coordinate metal ions giving rise to structures involved in protein/nucleic acid and/or protein/protein interactions. The compounds or salts of the present invention are capable of modifying the structure of zinc finger peptides in such a way that allows eventual dissociation of the metal ions. Typically, the metal ion is a divalent cation, such as those of zinc or cadmium. A zinc finger motif-containing protein is commonly a highly conserved and essential structure in viruses. Zinc finger motifs are found in human papilloma virus (HPV), particularly, HPV E6 and E7 proteins (see, e.g., Ullman (1996) Biochem. J. 319: 229-239) and influenza virus (see, e.g., Nasser (1996) J. Virol. 70: 8639-8644). In most subfamilies of Retro viridae, including avian sarcoma and leukosis retroviruses, mammalian B-type retroviruses, human T-cell leukemia and bovine leukemia retroviruses, D-type retroviruses, and lentiviruses, the invariable zinc finger motif is the most highly conserved structure. Retroviral nucleocapsid, Gag and Gag-Pol proteins have zinc finger motifs. In retroviruses, the zinc finger motif typically consists of 14 amino acid residues, with four residues being invariant; one exemplary zinc finger motif is described as Cys(X)2Cys(X)4His(X)4Cys and is refen-ed to as a "CCHC zinc finger" (Henderson ( 1981 ) J. Biol. Chem. 256: 8400). Zinc fingers chelate zinc through their histidine imidazole and cysteine thiolates (Berg ( 1986) Science 232: 485; Bess (1992) J. Virol. 66: 840; Chance ( 1992) Proc. Natl. Acad. Sci. U.S.A. 89: 10041 ; South (1990) Adv. Inorg. Biochem. 8: 199; South ( 1990) Biochem. Pharmacol. 40: 123-129). CCHC zinc fingers perform essential functions in retroviral infectivity, such as packaging genomic RNA. They are also essential for early events in virus infection.
[0032] The active form of the compounds of the present invention acts as a zinc fmger inhibitor that does not disrupt all zinc fingers. It has been determined that the compound of interest is specific to the particular configuration of the zinc fingers in NCp7. The cysteine at position 49 is the most reactive and was the expected, most likely target for the nucleophilic attack of the sulfur in this thioether drug since all other molecules that disrupt HIV NCp7 attack at cysteine 49. This is not what happens here. This thiol attacks cysteine at position 36 and the resulting acyl subsequently adds the lysine at position 38. With the presence of tryptophan (W37) and glutamine (Q 45) in key locations, the result is the disruption of the metal coordination and ultimate ejection of the zinc component of NCp7.
[0033] As used herein, the term "antiviral activity" means a compound has demonstrated some degree of antiviral activity in any assay, e.g., the XTT cytoprotection assay or p24
ELISA assays. As used herein, the term "virucidal" includes any degree of viral attenuation, including, but not limited to, complete inactivation or killing of a virus.
[0034] As used herein, terms such as "viral infectivity" or "index of infectivity" refer to the capacity of virus to pass from an infected cell to an uninfected cell, bringing about productive infection of the uninfected cell. For example, measurements of infectivity may be carried out by the MAGI assay, wherein the uninfected recipient cells are HeLa CD4 HIV
LTR Gal cells.
[0035] As used herein, the terms "inhibit the transmission of the virus" and "antiviral activity" mean the ability of a compound to negatively affect viral replicative capacity in any way. Such inhibition of transmission, e.g., loss in replicative capacity, can be measured using any means known in the art. For example, a compound inhibits the transmission of the virus (has antiviral activity) if it diminishes a virus' ability to produce progeny, (when in the form of a virion) fuse with a cell, enter a cell, bud from a cell, survive intracellularly or
extracellularly, reverse transcribe its RNA genome, translate viral proteins, process polyproteins with proteases, effect intracellular assembly of viral components into a capsid, and the like. The ability of a compound of the present invention to inhibit the transmission of a virus is not limited by any chemical or biological mechanism or pathway. A compound can inhibit infectivity or transmission (decrease replicative capacity) of a virus by, e.g.: binding to a nucleocapsid protein, such as NCp7; preventing binding of NCp7 to viral RNA or another nucleic acid; being involved in a specific chemical attack resulting in a stable or transient adduct; promoting the formation of inter- and intramolecular disulfide bonds through consequent destabilization of the NCp7 zinc finger loops; interacting with other conserved or non-conserved residues within the NCp7 protein which results in loss of function; and the like.
[0036] The present invention is further directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one compound or salt described herein. [0037] It is preferred that the pharmaceutically acceptable carrier be one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
[0038] The choice of carrier will be determined in part by the particular compound of the present invention chosen, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the
pharmaceutical composition of the present invention. The following formulations for oral, aerosol, nasal, pulmonary, parenteral, subcutaneous, intravenous, intra-arterial, intramuscular, intraperitoneal, intrathecal, intratumoral, topical, rectal, and vaginal administration are merely exemplary and are in no way limiting.
[0039] The pharmaceutical composition can be administered parenterally, e.g., intravenously, intraarterially, subcutaneously, intradermally, or intramuscularly. Thus, the invention provides compositions for parenteral administration that comprise a solution or suspension of the inventive compound or salt dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous isotonic sterile injection solutions.
[0040] Overall, the requirements for effective pharmaceutical carriers for parenteral compositions are well known to those of ordinary skill in the art. See, e.g., Banker and Chalmers, eds., Pharmaceutics and Pharmacy Practice, J. B. Lippincott Company,
Philadelphia, pp. 238-250 (1982), and Toissel, ASHP Handbook on Injectable Drugs, 4th ed., pp. 622-630 (1986). Such solutions can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound or salt of the present invention may be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl- l ,3-dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants.
[0041] Oils useful in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
[0042] Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanol amine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
[0043] The parenteral formulations can contain preservatives and buffers. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
[0044] Topical formulations, including those that are useful for transdermal drug release, are well-known to those of skill in the art and are suitable in the context of the invention for application to skin. Topically applied compositions are generally in the form of liquids, creams, pastes, lotions and gels. Topical administration includes application to the oral mucosa, which includes the oral cavity, oral epithelium, palate, gingival, and the nasal mucosa. In some embodiments, the composition contains at least one active component and a suitable vehicle or carrier. It may also contain other components, such as an anti-irritant. The carrier can be a liquid, solid or semi-solid. In embodiments, the composition is an aqueous solution. Alternatively, the composition can be a dispersion, emulsion, gel, lotion or cream vehicle for the various components. In one embodiment, the primary vehicle is water or a biocompatible solvent that is substantially neutral or that has been rendered substantially neutral. The liquid vehicle can include other materials, such as buffers, alcohols, glycerin, and mineral oils with various emulsifiers or dispersing agents as known in the art to obtain the desired pH, consistency and viscosity. It is possible that the compositions can be produced as solids, such as powders or granules. The solids can be applied directly or dissolved in water or a biocompatible solvent prior to use to form a solution that is substantially neutral or that has been rendered substantially neutral and that can then be applied to the target site. In embodiments of the invention, the vehicle for topical application to the skin can include water, buffered solutions, various alcohols, glycols such as glycerin, lipid materials such as fatty acids, mineral oils, phosphoglycerides, collagen, gelatin and silicone based materials.
[0045] Formulations suitable for oral administration can consist of (a) liquid solutions, such as a therapeutically effective amount of the inventive compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules, (c) powders, (d) suspensions in an appropriate liquid, and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a
pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of lactose, sucrose, mannitol, com starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
[0046] The compound or salt of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. The compounds are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of active compound are 0.01%-20% by weight, preferably 1%-10%. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. The surfactant may constitute 0.1 %-20% by weight of the composition, preferably 0.25%-5%. The balance of the composition is ordinarily propellant. A carrier can also be included as desired, e.g., lecithin for intranasal delivery. These aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations may be used to spray mucosa.
[0047] Additionally, the compound or salt of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as vaginal rings (i.e., intravaginal rings), pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate. Desirably, formulations presented as vaginal rings provide a slow release formulation of the compound or salt within the vagina over a period of time, for example, over about one week, or over about two weeks, or over about three weeks, or over about one month. The vaginal rings can be made of any suitable material, non-limiting examples of which include silicone elastomers and ethylene-co-vinyl acetate. Non-limiting examples of vaginal rings can be found in, e.g., U.S. Patents 4,155,991 , 5,989,581 , and 6,126,958, the disclosures of which are incorporated totally herein by reference. Other suitable vaginal ring products and formulations suitable for use therewith that are useful in connection with the compound or salt of the present invention will be readily apparent to those of ordinary skill in the medical and pharmaceutical arts. [0048] It will be appreciated by one of ordinary skill in the art that, in addition to the afore described pharmaceutical compositions, the compound or salt of the present invention may be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes. Liposomes serve to target the compounds to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of the inventive compound. Liposomes useful in the present invention include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations, the active agent to be delivered is incorporated as part of a liposome, alone or in conjunction with a suitable chemotherapeutic agent. Thus, liposomes filled with a desired inventive compound or salt thereof, can be directed to the site of a specific tissue type, for example hepatic cells, where the liposomes then deliver the selected compositions. Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, for example, liposome size and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, for example, Szoka et al., Ann. Rev. Biophys. Bioeng., 9, 467 (1980), and U.S. Patents 4,235,871 , 4,501,728, 4,837,028, and 5,019,369. For targeting to the cells of a particular tissue type, a ligand to be
incorporated into the liposome can include, for example, antibodies or fragments thereof specific for cell surface determinants of the targeted tissue type. A liposome suspension containing a compound or salt of the present invention may be administered intravenously, locally, topically, etc. in a dose that varies according to the mode of administration, the agent being delivered, and the stage of disease being treated.
[0049] The invention further provides a method for dissociating a metal ion from a zinc finger-containing protein, the method comprising contacting said zinc finger-containing protein with a compound or salt of the invention. The motif can be an isolated peptide or polypeptide, or, it can be a substructure of a viral protein or a virion. The method includes contacting the zinc finger with a compound of the present invention and subsequently detecting the dissociation of the metal ion from the zinc finger protein. The cation is commonly zinc. Any methodology known in the art can be used to detect the dissociation of the metal ion. Exemplary means include, e.g., capillary electrophoresis, immune-blotting, nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), detecting release of radioactive zinc-65, detecting fluorescence, or detecting gel mobility shift, and other techniques which would be apparent to one of skill upon review of this disclosure. These procedures can be practiced with any protocol known in the art, which are well described in the scientific and patent literature. A few exemplary techniques are set forth below.
[0050] As the invention provides a genus of novel compounds capable of dissociating a metal ion from a zinc finger in vitro, detection of the dissociation of the metal ion identifies some of the compounds within the scope of the present invention. For example, a zinc ejection assay can be used as a screen to identify compounds within the scope of the present invention. One strategy for such screening uses the XTT cytoprotection assay to monitor anti-viral activity. Alternative strategies use a Trp37 zinc ejection assay (see, e.g., U.S. Pat. No. 6,046,228) or a N-propyl gallate (NPG) fluorescence zinc ejection assay to identify compounds of the present invention that are able to act at the cellular level, e.g., on the NCp7 protein or its Gag or Gag-Pol precursors.
[0051] In certain embodiments, the zinc finger-containing protein is a viral protein. In certain embodiments, the viral protein is selected from the group consisting of a nucleocapsid protein, a Gag protein, and a Gag-Pol protein. The contacting of the protein with the compound or salt is performed in vitro or in vivo.
[0052] In certain preferred embodiments, the protein is that of a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus. In a preferred embodiment, the protein is that of an HIV-1 retrovirus.
[0053] In certain embodiments, the zinc-finger protein is present in a mammal. In certain preferred embodiments, the zinc-finger protein is that of a retrovirus present in a mammal. In certain embodiments, the zinc-finger protein is present on a surface of a mammal. In certain preferred embodiments, the zinc-finger protein is that of a retrovirus present on a surface of a mammal.
[0054] The invention further provides a method for inactivating a virus, the method comprising contacting a virus with a compound or salt of the present invention, whereby contacting the virus with said compound or salt inactivates the virus. The contacting can be as described herein. The virus can be any suitable virus. Non-limiting examples of suitable viruses include an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus. In certain embodiments, the virus is an HIV-1 retrovirus. In certain other embodiments, the virus is selected from the group consisting of a retrovirus, an avian sarcoma and leukosis retroviral group, a mammalian B-type retroviral group, a human T cell leukemia and bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group. In the above-described embodiments, preferably the contacting of the virus with the compound or salt is performed in vivo.
[0055] In some embodiments, the compound or salt of the invention is administered to inhibit the transmission of the virus. In this regard, a purpose for inhibiting the transmission of the virus refers to transmission of the virus from an infected individual to another individual. In some embodiments, inhibition of the transmission of the virus can be achieved by administration of the compound or salt intra- vaginally or intra-rectally. In certain other embodiments, inhibition of the transmission of the virus can be achieved by administration of the compound or salt parenterally, intrathecally, subcutaneously, or orally. In these embodiments, the compound or salt of the invention can be administered to a human as a pharmaceutical formulation. In other embodiments, the compound or salt of the invention can be administered to an animal as a veterinary pharmaceutical formulation. In certain embodiments, the pharmaceutical formulation is administered systemically to the human or animal. In certain other embodiments, the pharmaceutical formulation is administered topically to the human or animal.
[0056] In accordance with the invention, the term "animal" includes a mammal such as, without limitation, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order
Artiodactyla, including Bovines (cows) and Swine (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
[0057] The compound or salt thereof is administered in a dose sufficient to dissociate a zinc ion from a zinc finger-containing protein or for inactivating a virus. Such doses are known in the art (see, for example, the Physicians ' Desk Reference (2004)). The compounds can be administered using techniques such as those described in, for example, Physicians ' Desk Reference, 58th ed., Thomson PDR (2004).
[0058| Suitable doses and dosage regimens can be determined by conventional range- finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound of the present invention. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. The present method can involve the administration of about 0.1 μg to about 50 mg of at least one compound of the invention or salt thereof per kg body weight of the individual. For a 70 kg patient, dosages of from about 10 μg to about 200 mg of the compound or salt of the invention would be more commonly used, depending on a patient's physiological response, e.g., as determined by measuring viral load or other measurable parameters related to the viral load of a patient.
[0059] In other embodiments of the above-described method, the method further comprises contacting the virus with another anti -retroviral agent. The anti-retroviral agent can be any suitable anti-retroviral agent. Non-limiting examples of suitable anti-retroviral agents include anti-retroviral agents selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, an entry inhibitor, a maturation inhibitor, and a protease inhibitor. In certain preferred embodiments, the anti-retroviral agent is a nucleoside analogue which is an AZT, a ddCTP or a ddl.
[0060] In certain embodiments, the said another anti-retroviral agent is selected from the group consisting of enfuviritide, maraviroc, delavirdine, etravirine, efavirenz, nevirapine, zidovudine, lamivudine, emtricitabine, didanosine, tenofovir, stavudine, abacavir, raitegravir, dolutegravir, amprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, and nelfinavir, and combinations thereof.
[0061] In other embodiments, the contacting of the virus with the compound or salt is performed on a blood product, blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food, drink, implant or prosthesis. In these embodiments, the contacting of the virus with the compound or salt is performed in vitro.
[0062] The antiviral activity of the inventive compounds, such as viral inactivation and removal of metal ions from the zinc fingers of viral associated proteins, can be further assessed via methods disclosed in U.S. Patent 7,528,274, the disclosure of which is totally incorporated herein by reference.
[0063] Without wishing to be bound by any particular theory, it is believed that the compounds of the invention undergo intracellular hydrolysis and elimination to liberate 2- mercaptobenzamides, which are then acetylated by acetyl-Co-A to produce 2- acetylmercaptobenzamides. For example, as depicted in FIG. 2, it is believed that compounds of formula (I) undergo hydrolysis of the fragment RC(=0) by esterase or by esterase to liberate compound A, which then undergoes 1 ,2-elimination to liberate the free thiol B. Compound B is acetylated to produce the 2-acetylmercaptobenzamide derivative C. The 2-acetylmercaptobenzamides are believed to specifically react with the C-terminal zinc finger of NCp7 and eject its coordinated zinc ion, irreversibly destroying protein function.
[0064] The compounds of the invention desirably exhibit low toxicity. For example, compound 1 has been shown to have a maximum tolerated dose in rats of greater than 2 g per kg of body weight.
[0065] The compounds of the invention desirably exhibit oral bioavailability. For example, pharmacokinetic studies in rats indicate that compound 1 is orally bioavailable.
[0066] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
[0067] All chemicals and solvents were obtained from Sigma-Aldrich (Milwaukee, WI).
EXAMPLE 1
[0068] This example demonstrates the synthesis of ((2-((3-amino-3- oxopropyl)carbamoyl)phenyl)thio)methyl butyrate 1. The reaction scheme is depicted in FIG. 1.
[0069] Thiosalicylic acid (10 g, 65 mmole) and HBTU (25.8 g, 68 mmole) were dissolved in -200 mL DMF. DIEA (39.52 mL, 227 mmole) was then added and the solution was allowed to stir for 15 minutes, β-alaninamide HC1 (8.48 g, 96.2 mmole) was then added and the solution was allowed to stir at room temperature for 24 hours. Chloromethyl butyrate (8.12 mL, 64 mmole) was then added at room temperature and the solution was again allowed to stir 24 hours. The solvent was removed under reduced pressure (at ~65°C) and then the residue was allowed to solidify at 4°C. The white semi-solid was recrystallized from a mixture of 9: 1 ethyl acetate/hexanes adding slightly more EtOAc as needed. Overall yield with two recrystallizations was 43%. Ή NMR (CDC13) 5 7.589 (d, I H), 7.489 (d, I H), 7.406 (t, I H), 7.303 (t, I H), 6.905 (s, IH), 5.979 (s, I H), 5.585 (s, I H), 5.408 (s, 2H), 3.722 (q, 2H), 2.599 (t, 2H), 2.346 (t, 2H), 1.660 (m, 2H), 0.950 (t, 2H). EXAMPLE 2
[0070] This example demonstrates the virucidal activity of two compounds in accordance with embodiments of the invention, ((2-((3-amino-
3-oxopropyl)carbamoyl)phenyl)thio)methyl butyrate (compound 1) and ((2-((3-amino-3- oxopropyl)carbamoyl)phenyl)thio)methyl 2-ethylbutanoate (compound 2)
[0071] Fresh human peripheral blood mononuclear cells (PBMCs) were obtained from a commercial source (Biological Specialty Corporation; Colmar, PA) and determined to be seronegative for HIV and HBV. Depending on the volume of the donor blood received, the leukophoresed blood cells were washed several times with PBS. After washing, the leukophoresed blood was diluted 1 : 1 with Dulbecco's phosphate buffered saline (PBS) and layered over 15 mL of Ficoll-Hypaque density gradient in a 50 mL conical centrifuge tube. These tubes were centrifuged for 30 min at 600g. Banded PBMCs were gently aspirated from the resulting interface and subsequently washed three times with PBS by low speed centrifugation. After the final wash, cells were enumerated by Trypan Blue dye exclusion and re-suspended at 1 x 106 cells/mL in RPMI 1640 with 15 % Fetal Bovine Serum (FBS), 2 mmol/L L-glutamine, 2 ^ig/mL PHA-P, 100 U/mL penicillin and 100 μg/mL streptomycin and allowed to incubate for 48 - 72 hour at 37 °C. After incubation, PBMCs were centrifuged and resuspended in tissue culture medium (RPMI 1640 with 15% FBS, 2 mmol/L L- glutamine, 100 U/mL penicillin, 100 ^ig/mL streptomycin and 3.6 ng/mL recombinant human IL-2). The cultures were maintained until use by ½ culture volume change with fresh IL-2 containing tissue culture medium every 3 days. Assays were initiated with PBMCs that have been induced to proliferate for 72 hr.
[0072] PHA-P-stimulated PBMCs from three donors were pooled together to minimize the variability between individual donors, and resuspended in fresh tissue culture medium at 1 x 106 cells/mL and plated in the interior wells of a 96-well round bottom microtiter plate at 50 μίΛνεΙΙ. Then, 100 xL of 2x concentrations of compound-containing medium (200, 62.5, 20, 6.25, 2, and 0.625 nM of CTP-51 8 or ATZ) was transferred to the round-bottom 96-well plate containing the cells in 50 μΐ, of the medium. AZT (1 , 0.32, 0.1 , 0.032, 0.01 , and 0.0032 μΜ) was evaluated as an internal assay standard.
[0073] Immediately following test material addition to the wells, 50 μΐ, of a
predetermined dilution of HIV virus (prepared at 4X of final in well concentration) was added, and mixed well. The virus was CCR5 tropic clade B HIV- l 92/us/727- For infection, 50- 150 TCIDso of virus was added per well (final MOI=0.05-0.10). PBMCs were exposed in triplicate to vims and cultured in the presence or absence of the test material at varying concentrations as described above in the 96-well microtiter plates. After 7 days in culture, HIV-1 replication was quantified in the tissue culture supernatant by measurement of reverse transcriptase activity. Wells with cells and virus alone were used for virus control. Separate plates were identically prepared without virus for drug cytotoxicity studies using the tetrazolium dye XTT.
[0074] The EC and TC and calculated therapeutic index at 25%, 50%, and 95% of the virus control are set forth in Table 1.
Table 1.
Figure imgf000019_0001
[0075] As is apparent from the results set forth in Table 1 , both compounds 1 and 2 exhibited virucidal activity in the antiviral assay against CCR5 tropic clade B HIV- I 92/US/727 while exhibiting essentially no cellular toxicity.
EXAMPLE 3
[0076] This example illustrates the in vitro metabolism of compounds 1 and 2 in human and dog plasma and in rat whole blood.
[0077] Compounds 1 and 2 were incubated in human plasma, dog plasma, and SD rat whole blood at a concentration of 10 μΜ with 0.5% DMSO. After 1 h and 48 h, the samples were quenched with 5 volumes of acetonitrile followed by centrifugation for 15 min at 16, 100 g. Equal volumes of purified water were added into the supernatant, and the samples were analyzed by LC-MS/MS. [0078] The major metabolites D-G were identified by mass spectrometry, and their structures are illustrated in FIG. 3. The metabolite profiles at 1 h and at 48 h for compounds land 2 are set forth in Tables 2 and 3, respectively. A "-" indicates that a particular metabolite was not observed, a "++" indicates that the metabolite was present in 1-10% relative HPLC peak area, and a "+++" indicates that the metabolite was present in >10% relative HPLC peak area.
Table 2. Metabolic profile for compound 1.
Figure imgf000020_0001
EXAMPLE 4
[0079] This example demonstrates the anti-HIV evaluation of compounds 1 and B in human PBMCs in accordance with an embodiment of the invention.
[0080] PHA-P stimulated PBMCs from two donors were pooled together and re- suspended in fresh tissue culture medium at 1 x 106 cells/mL and plated in the interior wells of a 96 well round bottom microplate at 50 μΐ ννεΐΐ. A 100 volume of 2X concentrations of compound-containing media were transferred to the round-bottom 96-well plate containing the cells in triplicate. Fifty microliters (50 μί) of HIV- 1 at a pre-determined dilution was added. Each plate contained cell control wells and virus control wells in parallel with the experimental wells. After 7 days in culture, efficacy was evaluated by measuring the reverse transcriptase in the culture supernatants and the cells were stained with the tetrazolium dye XTT to evaluate cytotoxicity. The results are set forth in Table 4.
Table 4
Figure imgf000021_0001
EXAMPLE 5
[0081] This example demonstrates the evaluation of compounds 1 and B in combination with 23 known anti-HIV inhibitors for antiviral activity against HIV-1 HIB using microtiter in vitro assay systems.
[0082] Compounds 1 and B were solubilized at 40 mM in DMSO and stored at -20° C. Compounds 1 and B were evaluated at concentrations of 4, 2, 1 , 0.5, and 0.25 μΜ in combination with nine concentrations of an existing HIV inhibitor in at least two replicate antiviral combination assays. The second known anti-HIV agent was evaluated using a high test concentration of two-times the EC5o value as determined in the cytoprotection assay. Table 5 lists the class of inhibitor and compound source. Compounds were diluted in cell culture medium at 4 times the high test concentration, serially diluted, and added to the microliter plate in the appropriate format in a volume of 50 μΐ. per well.
Table 5
Figure imgf000022_0001
[0083] Cell Preparation [0084] CEM-SS cells were passaged in RPMI1640 supplemented with 10% heat inactivated FBS, 2 mM L-glutamine, 100 U/mL penicillin and 100 μg/mL streptomycin in T- 75 flasks prior to use in the antiviral assay. On the day preceding the assay, the cells were split 1 :2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification were performed using a hemocytometer and Trypan Blue dye exclusion. Cell viability must be greater than 95% for the cells to be utilized in the assay. The cells were resuspended at 5 x 104 cells per ml tissue culture medium and added to the drug-containing microtiter plates.
[0085] Virus Preparation
[0086] The virus used for the cytoprotection assays was the lymphocyte-tropic virus HIV-lrrrs. The virus was obtained from the NIH AIDS Research and Reference Reagent Program and stock virus pools were produced in CEM-SS cells. A pretitered aliquot of virus was removed from the freezer (-80° C) and allowed to thaw slowly to room temperature in a biological safety cabinet. Virus was resuspended and diluted into tissue culture medium such that the amount of virus added to each well in a volume of 50 was the amount determined to yield 85 to 95% cell killing at 6 days post-infection.
[0087] Plate Format
[0088] Each plate contained cell control wells (cells only), virus control wells (cells plus virus), drug toxicity wells (cells plus drug only), drug colorimetric control wells (drug only) as well as experimental wells (drug plus cells plus virus). Samples were tested in triplicate.
[0089] Efficacy and Toxicity XTT
[0090] Following incubation at 37° C in a 5% CO? incubator, the test plates were stained with the tetrazolium dye XTT (2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-5- [(phenylamino)carbonyi]-2H-tetrazolium hydroxide). XTT-tetrazolium is metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product. XTT solution was prepared daily as a stock of 1 mg/mL RPMI1640. Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/mL PBS and stored in the dark at -20°C. XTT/PMS stock was prepared immediately before use by adding 40 μΕ PMS per mL of XTT solution. Fifty microliters of XTT/PMS was added to each well of the plate prior to reincubation for 4 hr at 37° C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble formazan product. The plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader.
[0091] Data Analysis [0092] Raw data was collected from the Softmax Pro 4.6 software and imported into the Prichard and Shipman MacSynergy II software template (Prichard et at. 1993. Antiviral Research 14: 181 -206). Effects of the drug combination are calculated based on the activity of the two compounds when tested alone. The expected additive antiviral protection is subtracted from the experimentally determined antiviral activity at each combination concentration resulting in a positive value (synergy, or potentiation), a negative value (antagonism), or zero (additivity).
[0093] The results of the combination assays are presented three dimensionally at each combination concentration, yielding a surface of activity extending above (synergy) or below (antagonism) the plane of additivity. The volume of the surface is calculated and expressed as a synergy volume (μΜ2%) calculated at the 95% confidence interval.
[0094] For these studies, synergy is defined as drug combinations yielding synergy volumes greater than 50 μΜ2% at the 95% confidence interval. Slightly synergistic activity and highly synergistic activity have been defined as yielding synergy volumes of 50 to 100 μΜ2 % and > 100 μΜ2%, respectively. Synergy volumes between -50 and 50 μΜ2% are considered additive and synergy volumes less than -50 μΜ % are considered antagonistic.
[0095] Combination Therapy Evaluations: Compounds 1 and B were evaluated in combination with twenty-three known anti-HIV agents for the inhibition of cytopathic effect in CEM-SS cells using the IIIB strain of HIV- 1. The percent of virus replication inhibition above expected at each concentration for each two-drug combination was calculated at the 95%, 99% and 99.9% confidence interval. The data obtained at the 95% confidence value were plotted three dimensionally. The synergy volumes for the combinations at the 95% confidence interval of two replicate assays are summarized in Table 6 for B. The synergy volumes for the combinations at the 95% confidence interval of two replicate assays are summarized in Tabic 7 for 1.
Table 6
CEM-SS/HIV- I IHB Assay 1 CEM-SS/HIV- l niB Assay 2
Compound B Synergy/Antagonism Definition Synergy/Antagonism Definition tested in Volume (μΜ2%) of Volume (μΜ2%) of combination Interaction Interaction with:
Enfuviritide 57.5A26.8 Slightly 104/-38.7 Synergistic synergistic
Maraviroc 0/- 12.1 Additive 20.5/- 1 1 .2 Additive
Delavirdine 86.2/- 14.0 Slightly 74.0/-2.40 Slightly synergistic synergistic
Etravirine 38.3A6.58 Additive 35.7A29.8 Additive
Efavirenz 67.9Λ-73.7 Slightly 102/-5.8 Synergistic synergistic
Nevirapine 60.4/-0.67 Slightly 88.3A32.6 Slightly synergistic synergistic
Zidovudine 147/-5.04 Synergistic 74.9A33 Slightly synergistic
Lamivudine 29.8A34.5 Additive 27.9/0 Additive
Emtricitabine 38.5/- 1 .97 Additive 15.9/-33.8 Additive
Didanosine 8.50/- 14.6 Additive 46.1/-13.2 Additive
Tenofovir 63.2/-30.1 Slightly 168/-86.7 Synergistic synergistic
Stavudine 1 17/0 Synergistic 58 J/-26.9 Slightly synergistic
Abaca vir 22.4A6.27 Additive 25.8/-24.1 Additive
Raltegravir 6.95A23.7 Additive 3 1 .4/-0.6 Additive
Dolutegravir 127/-8.92 Synergistic 64.4/- 1 3.9 Slightly synergistic
Amprenavir 58.2/- 1 .8 1 Slightly 144/- 17.9 Synergistic synergistic Tipranavir 3.3/-28.5 Additive 28.8/-13.9 Additive
Indinavir 73.0/-3.00 Slightly 105/-36.3 Synergistic synergistic
Saquinavir 76.2/- 12.7 Slightly 53.0/0 Slightly synergistic synergistic
Lopinavir 154/- 195 Synergistic 62.6/-20.2 Slightly synergistic
Ritonavir 203.0/-5.1 Synergistic 361 /-6.3 Synergistic
Darunavir 101 /-30.4 Synergistic 62.9/-2.05 Slightly synergistic
Nelfmavir 701/-7.1 Synergistic 293A9.83 Synergistic
Table 7
CEM-SS/HIV-Ι π,Β Assay 1 CEM-SS/HIV- I HIB Assay 2
Compound 1 Synergy/Antagonism Definition Synergy/ Antagoni sm Definition tested in Volume (μΜ2%) of Volume (μΜ2%) of combination Interaction Interaction with:
Enfuviritide 86.7/-0.4 Slightly 143/-0 Synergistic synergistic
Maraviroc 28.9/-27.5 Additive 20.7/- 16.3 Additive
Delavirdine 44.4/-3 1 .5 Additive 35.9/-21 .5 Additive
Etravirine 0/-25.6 Additive 22.8Λ-98.8 Additive
Efavirenz 26.5A29.8 Additive 42.1 /-66.8 Additive
Nevirapine 25.7/0 Additive 5.3Λ5.3 Additive
Zidovudine 1 2.3/-32.5 Additive 16.8/-35.0 Additive
Lamivudine 27 1 /-36.9 Synergistic 90.1 /0 Slightly synergistic
Emtricitabine 42.5/- 17.1 Additive 24.1 /-85.9 Additive
Didanosine 30.4/- 1 5.0 Additive 45.7/- 10.3 Additive
Tenofovir 28.6/- 1 2.1 Additive 35.6/-33. 1 Additive Stavudine 158/-1 .34 Synergistic 62.2/-0.3 1 Slightly
synergistic
Abacavir 26.4/-21.4 Additive 29.7/-26.1 Additive
Raltegravir 3.91/-8.40 Additive 17.8/-22.2 Additive
Dolutegravir 46.2/0 Additive 34.5/-18.3 Additive
Amprenavir 131/-0.78 Synergistic 91.7/-17.1 Synergistic
Tipranavir 2.2A44.4 Additive 0/-5.63 Additive
Indinavir 39.2/-86.0 Additive 38.5Λ77.4 Additive
Saquinavir 122/-2.50 Synergistic 68.3/-1 1.8 Slightly
synergistic
Lopinavir 256/-1 1.9 Synergistic 168/-24.8 Synergistic
Ritonavir 1 19/-47.2 Synergistic 167/-20.2 Synergistic
Darunavir 79.0/-4.09 Slightly 178/-56.7 Synergistic synergistic
Nelfinavir 325A45.6 Synergistic 238/0 Synergistic
[0096] Importantly, the combination assays demonstrated that within the concentration ranges employed in the combination antiviral assays, no evidence of either antagonism or synergistic toxicity was observed with any of the compound combinations, suggesting that the test compounds could be safely used with all approved HIV-1 inhibitors.
[0097] Compounds 1 and B were evaluated in combination with two entry/fusion inhibitors. Enfuviritide (fusion inhibitor) in combination with the test compounds yielded slightly synergistic interactions of cytoprotection (mean synergy volume of 80.8 μΜ2% for B and 1 14.6 μ 2% for 1). Maraviroc (CCR5-receptor inhibitor; was inactive alone versus the predominantly CXCR4-receptor tropic IIIB strain of HIV-1 ) in combination with the test compounds yielded an additive interaction.
[0098] B or 1 in combination with reverse transcriptase inhibitors yielded additive to synergistic interactions. B combinations with delavirdine, efavirenz, nevirapine, zidovudine, tenofovir or stavudine yielded the mean synergy volumes of 80.1 μΜ2%, 84.7 μΜ2%, 74.4 μΜ2%, 1 1 1 μΜ2%, 1 16 μΜ2%, and 87.7 μΜ2%, respectively. Β combinations with etravirine, lamivudine, emtricitabine and didanosine resulted in an additive interaction from two assay replicates. 1 in combination with lamivudine or stavudine resulted in a mean synergy volume from two assay replicates of 180 or 1 10 μΜ %, respectively. 1 in combination with all other RT inhibitors resulted in additive interactions.
[0099] In combination with the integrase inhibitor raltegravir, B and 1 resulted in additive interactions. B in combination with dolutegravir demonstrated a synergistic interaction with a mean synergy volume of 95.7 μΜ2%. 1 in combination with dolutegravir resulted in an additive interaction.
[00100] B or 1 in combination with protease inhibitors resulted in additive to highly synergistic interactions. 1 in combination with tipranavir resulted in an additive interaction from two assay replicates. In combination with the other protease inhibitors, B yielded mean synergy volumes ranging from 65.1 (saquinavir) to 498 μΜ % (nelfinavir). 1 in combination with tipranavir or indinavir resulted in additive interactions. In combination with the other protease inhibitors, 1 yielded mean synergy volumes ranging from 95.1 (saquinavir) to 282 μΜ2% (nelfinavir).
[00101] These results show that there was no significant combination antagonism or synergistic toxicity for any of the combination therapy assays at the concentrations tested. Of the twenty-three combination assays performed with representatives of the various classes of HIV inhibitors, B and the prodrug 1 resulted in additive to synergistic interactions with all of the approved inhibitors, with the greatest levels of synergy observed with compounds which inhibited protease (ritonavir, lopinavir and nelfinavir).
EXAMPLE 6
[0100] This example demonstrates the maximum tolerated dose (MTD) of compound 1 in Sprague-Dawley rats following a dose escalation design via oral administration (Phase A). In addition, this example demonstrates the potential for repeat dose toxicity following once daily oral administration of compound 1 for 14 consecutive days (Phase B). In this phase, the reversibility, progression and potential delayed effects were determined following a 14-day recovery period.
[0101] Phase A:
[0102] Forty rats were assigned to Phase A as shown in Table 8. Compound 1 was dosed once via oral administration at a dose volume of 10 mL/kg (Groups 1 , 2 and 3) or 20 mL/kg (Groups 4 and 5). After each dose, there was a 4-day observation period. There was at least a one-day interval between doses, in order to allow observation of abnormal clinical signs and decision of the dose level for the next group. The vehicle for Groups 1 -4 was 5% DMSO and 70% PEG 400 in water, and for Group 52% CMC-Na in water.
Table 8
Figure imgf000029_0001
[0103] Parameters evaluated in Phase A included cageside and detailed clinical observations, body weights and body weight changes, and food consumption.
[0104] Phase B:
[0105] Thirty-two rats were assigned to Phase B as shown in Table 9. Control Article (CA; 2% CMC-Na in Water for injection) or compound 1 were administered once daily for 14 consecutive days via oral administration at a dose volume of 20 mL/kg.
Table 9
Figure imgf000029_0002
[0106] Main= Main study animals, sacrificed on Day 15
[0107] Recovery^ Recovery study animals, sacrificed on Day 29
[0108] Blood samples for clinical pathology analyses were collected from the mam study animals prior to terminal sacrifice on Day 15 and from the recovery animals prior to recovery sacrifice on Day 29. All main study animals were euthanized and necropsied following the final blood collection on Day 15. All recovery study animals were euthanized and necropsied following the final blood collection on Day 29. Parameters evaluated included cageside and detailed clinical observations, body weights and body weight changes, food consumption, clinical pathology (clinical chemistry, hematology, coagulation and urinalysis), absolute and relative organ weights, gross pathology and microscopic pathology.
[0109] Animal Disposition and Observations
[0110] There was no mortality during Phase A or Phase B. All animals survived to their scheduled sacrifice.
[0111] There were no test article-related clinical signs observed in study animals throughout Phase A or Phase B.
[0112] Body Weights and Body Weight Changes
[0113] Treatment with compound 1 had no effect on body weights or body weight changes in Phase A or Phase B.
[0114] Food Consumption
[0115] Treatment with compound 1 had no effect on food consumption in Phase A or Phase B.
[0116] Clinical Chemistry
[0117] There are no test article-related effects for any clinical chemistry parameter from blood samples taken on Day 15 and no evidence of a delayed test article-related effect for any clinical chemistry parameter from samples taken on Day 29.
[0118] Hematology
[0119] There are no test article-related effects for any hematology parameter from blood samples taken on Day 15 and no evidence of a delayed test article-related effect for any hematology parameter from samples taken on Day 29.
[0120] There are no appreciable differences in the values from control and compound 1 -treated rats with the exception of terminal sacrifice Group 4 male 80970. The WBC, ABLYMP, and ABMONO values for this rat were noticeably higher than control values on Day 15. On Day 29, the WBC and ABNEUT values were appreciably higher in one male rat (80972). There was no gross or histopathological correlation, and the absence of similar findings for any other Group 4 rat suggests the findings are individual animal variation and unlikely test article-related.
[0121] Coagulation [0122] There are no compound 1-related effects for any coagulation parameter from blood samples taken on Day 15 and no evidence of a delayed compound 1-related effect for any coagulation parameter from samples taken on Day 29.
[0123] Urinalysis
[0124] Cloudy or flocculent urine samples from Day 15 (terminal sacrifice) were noted for two Group 3 (1000 mg/kg/day) rats (one male and one female) and all four Group 4 (2000 mg/kg/day) rats. Unidentified amorphous crystals were identified in the Day 15 urine samples from all Group 3 and Group 4 rats. The number of crystals per low power field increased with dose. The toxicological significance of these findings is probably low as they only occur at very high dose levels and were not associated with changes in other urine analysis parameters or sediment evaluations. Furthermore, they do not correlate with any kidney-related clinical chemistry findings or any histopathological changes in the urinary system.
[0125] Cloudy urine samples from Day 29 were noted in one Group 1 (control) male, two Group 2 (500 mg/kg/day) males, and two Group 4 rats (one male and one female). The Day 29 urine sediment analysis did not reveal any significant differences among control and NS 1040 treated rats. The cause for the cloudy appearance of urine on Day 29 is unknown and not likely to be relating to the test article.
[0126] Organ Weights
[0127] There are no compound 1-related effects in organ weights following 14 days of dosing (Day 15 terminal sacrifice) or evidence of delayed effects following a 14-day recovery (Day 29 recovery sacrifice).
[0128] Slight increases in liver organ weight, organ-to-body weight ratios and organ-to- brain weight ratios were observed in some animals given 2000 mg/kg/day. These findings were not associated to any gross or histopathology findings and not considered to be toxicologically significant.
[0129] Gross Pathology
[0130] There are no compound 1-related effects following 14 days of dosing (Day 15 terminal sacrifice) or evidence of delayed effects following a 14-day recovery (Day 29 recovery sacrifice).
[0131] The gross pathology findings, including red multifocal discoloration of mandibular lymph node, were sporadically observed in one female given 500 mg/kg/day and one male given 1000 mg/kg/day. Those changes were considered to be not related to compound 1 treatment, due to low incidence and lack of correlated histopathological findings.
[0132] Bone Marrow Smear Evaluation
[0133] Duplicate bone marrow smears were prepared from the sternum of all euthanized rats during the terminal necropsy on Day 15 (2 rats/sex/group) and the recovery necropsy on Day 29 (2 rats/sex/group). All smears from all animals were stained with Wright-Giemsa and evaluated by Pharmaron Pathology Laboratory. Megakaryocyte precursors were estimated as decreased (D), normal (N) or increased (I), based on a semi-quantitative subjective evaluation of megakaryocyte precursor density from the smears. A bone marrow differential count (200 cells) was used for the quantification of myeloid, erythroid, and other cell lineages (including lymphocytes, macrophages/monocytes, and plasma cells). A Myeloid : Erythroid (M:E) ratio was calculated for each animal by dividing the total number of myeloid cells by the total number of erythroid cells. Eosinophilic and basophilic granulocytes were not separated from neutrophilic granulocytes unless there were apparent differences in the proportions of eosinophils and basophils between groups.
[0134] Terminal Sacrifice (Day 15)
[0135] One male (80954) and one female (80958) given 500 mg/kg/day, as well as one female (80965) given 1000 mg/kg/day and one female (80974) given 2000 mg/kg/day had slightly increased total myeloid cell counts, which correlated to slight increases in M:E ratio. The relation to the test article treatment is not clear, but the findings are most likely of low toxicological significance.
[0136] Recovery Sacrifice (Day 29)
[0137] Both males (80955 and 80956) and one female (80960) given 500 mg/kg/day had slightly increased total myeloid cell count relative to concurrent controls, which correlated to slightly increased M:E ratio. In addition, slightly increased total myeloid cell count was observed in one male (80972) given 2000 mg/kg/day, but without con-elation to any other findings.
[0138] There were no obvious quantitative alterations in Megakaryocyte precursors and abnormal morphology changes in all examined bone marrow smears from all treated animals. The relation of the above changes on cell counts to the compound 1 treatment is not clear, but the findings are most likely of low toxicological significance.
[0139] Histopathology [0140] There are no compound 1-related effects following 14 days of dosing (Day 15 terminal sacrifice) or evidence of delayed effects following a 14-day recovery (Day 29 recovery sacrifice).
[0141] Early changes of spontaneous nephropathy (tubular epithelial
degeneration/regeneration and tubular dilatation) were observed with a similar incidence and severity in control and compound 1 treated rats. One Group 3 male (80962; terminal sacrifice) had harderian gland inflammation that may be due to chronic infection and one Group 3 male (80963; recovery sacrifice) had an area in the myocardium of proliferative histiocytic cells. The above changes can be regarded as spontaneous or incidental, and not related to the compound 1 treatment.
[0142] The following describes aspects of the present invention.
formula (I):
Figure imgf000033_0001
(I)
wherein R is hydrogen or a linear or branched alkyl.
2. The compound of aspect 1 , wherein R is Ci-C8 linear or branched alkyl.
of aspect 2, wherein the compound is:
Figure imgf000033_0002
of aspect 2, wherein the compound is:
Figure imgf000033_0003
5. A pharmaceutical composition comprising a compound of any one of aspects 1 -4 and a pharmaceutically acceptable earner. 6. The pharmaceutical composition of aspect 5, further comprising another anti- retro viral agent other than the compound of formula (I).
7. The pharmaceutical composition of aspect 6, wherein the another anti- retroviral agent is a member selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
8. A composition comprising a compound of any one of aspects 1 -4 or a pharmaceutically acceptable salt thereof and further comprising blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food or drink.
9. A method for dissociating a zinc ion from a zinc finger-containing protein, the method comprising contacting said zinc finger-containing protein with a compound of any one of aspects 1-4 and pharmaceutically acceptable salts thereof.
10. The method of aspect 9, wherein the zinc finger-containing protein is NCp7.
1 1. The method of aspect 10, wherein the NCp7 protein is selected from the group consisting of a nucleocapsid protein, a Gag protein and a Gag-Pol protein.
12. The method of aspect 1 1 , wherein the contacting of the protein with the compound is performed in vitro.
13. The method of aspect 1 1 , wherein the contacting of the protein with the compound is performed in vivo.
14. The method of aspect 9 or aspect 10, wherein the protein is that of a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
15. The method of aspect 14, wherein the protein is that of an HIV-1 retrovirus.
16. The method of aspect 9 or aspect 10, wherein the zinc finger-containing protein is that of a virus selected from the group consisting of an avian sarcoma retroviral group, a mammalian B-type retroviral group, a human T cell leukemia retroviral group, a bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group.
17. The method of aspect 9, wherein the zinc finger-containing protein is part of an intact virus. 18. The method of aspect 9, further comprising detecting the dissociation of the metal ion from the zinc finger-containing protein.
19. The method of aspect 18, wherein detecting the dissociation of the metal ion is carried out by a method selected from the group consisting of capillary electrophoresis, immunoblotting, nuclear magnetic resonance (NMR), high performance liquid
chromatography (HPLC), a method of detecting release of radioactive zinc-65, a method of detecting fluorescence, and a method of detecting gel mobility shift.
20. The method of any one of aspects 16-19, wherein the zinc finger-containing protein is present in a mammal.
21. The method of any one of aspects 16-19, wherein the zinc finger-containing protein is present on a surface of a mammal.
22. A method for inactivating a virus, the method comprising contacting a virus with a compound of any one of aspects 1 -4 or a pharmaceutically acceptable salt thereof, whereby contacting the virus with said compound or salt inactivates the virus.
23. The method of aspect 22, wherein the virus is selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1, a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
24. The method of aspect 23, wherein the virus is an HIV-1 retrovirus.
25. The method of aspect 23, wherein the virus is selected from the group consisting of a retrovirus of an avian sarcoma and leukosis retroviral group, a mammalian B- type retroviral group, a human T cell leukemia and bovine leukemia retroviral group, a D- type retroviral group, a murine leukemia-related group and a lentivirus group.
26. The method of aspect 22, wherein the contacting of the virus with the compound or salt is performed in vivo.
27. The method of aspect 26, wherein the compound or salt is administered to inhibit the transmission of the virus.
28. The method of aspect 26, wherein the compound or salt is administered intra- vaginally or intra-rectally to inhibit the transmission of the virus.
29. The method of aspect 26, wherein the compound or salt is administered parenterally, intrathecally, subcutaneously, or orally.
30. The method of aspect 26, wherein the compound or salt is administered to a human as a pharmaceutical formulation. 31. The method of aspect 26, wherein the compound or salt is administered to an animal as a veterinary pharmaceutical formulation.
32. The method of any one of aspects 26-31 , wherein the method further comprises contacting the virus with another anti -retroviral agent.
33. The method of aspect 22, wherein said another anti-retroviral agent is selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
34. The method of aspect 33, wherein the nucleoside analogue is an AZT, a ddCTP or a ddl.
35. The method of aspect 32, wherein said another anti-retroviral agent is selected from the group consisting of enfuviritide, maraviroc, delavirdine, etravirine, efavirenz, nevirapine, zidovudine, lamivudine, emtricitabine, didanosine, tenofovir, stavudine, abacavir, raitegravir, dolutegravir, amprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, and nelfinavir.
36. The method of aspect 22, wherein the contacting of the virus with the compound or salt is performed on a blood product, blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food, drink, implant or prosthesis.
[0143] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0144] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and
"containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
(0145] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

CLAIM(S):
of formula (I):
Figure imgf000038_0001
(I)
wherein R is hydrogen or a linear or branched alkyl.
2. The compound of claim 1 , wherein R is Ci-Cg linear or branched alkyl.
3. The compound of claim 2, wherein the compound is:
Figure imgf000038_0002
The compound of claim 2, wherein the compound
Figure imgf000038_0003
5. A pharmaceutical composition comprising a compound of any one of claims 1 -4 and a pharmaceutically acceptable carrier.
6. The pharmaceutical composition of claim 5, further comprising another anti- retroviral agent other than the compound of formula (I).
7. The pharmaceutical composition of claim 6, wherein the another anti- retroviral agent is a member selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
8. A composition comprising a compound of any one of claims 1 -4 or a pharmaceutically acceptable salt thereof and further comprising blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food or drink.
9. The compound of any one of claims 1-4 and pharmaceutically acceptable salts thereof for use in dissociating a zinc ion from a zinc fmger-containing protein, the use comprising contacting said zinc finger-containing protein with the compound or salt.
10. The compound or salt of any one of claims 1-4 for use according to claim 9, wherein the zinc finger-containing protein is NCp7.
1 1. The compound or salt of any one of claims 1 -4 for use according to claim 10, wherein the NCp7 protein is selected from the group consisting of a nucleocapsid protein, a Gag protein and a Gag-Pol protein.
12. The compound or salt of any one of claims 1-4 for use according to claim 1 1 , wherein the contacting of the protein with the compound is performed in vitro.
13. The compound or salt of any one of claims 1 -4 for use according to claim 1 1 , wherein the contacting of the protein with the compound is performed in vivo.
14. The compound or salt of any one of claims 1-4 for use according to claim 9 or claim 10, wherein the protein is that of a retrovirus selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1 , a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
15. The compound or salt of any one of claims 1 -4 for use according to claim 14, wherein the protein is that of an HIV-1 retrovirus.
16. The compound or salt of any one of claims 1-4 for use according to claim 9 or claim 10, wherein the zinc finger-containing protein is that of a virus selected from the group consisting of an avian sarcoma retroviral group, a mammalian B-type retroviral group, a human T cell leukemia retroviral group, a bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lentivirus group.
17. The compound or salt of any one of claims 1 -4 for use according to claim 9, wherein the zinc finger-containing protein is part of an intact virus.
18. The compound or salt of any one of claims 1 -4 for use according to claim 9, further comprising detecting the dissociation of the metal ion from the zinc fmger-containing protein.
19. The compound or salt of any one of claims 1 -4 for use according to claim 1 8, wherein detecting the dissociation of the metal ion is carried out by a method selected from the group consisting of capillary electrophoresis, immunoblotting, nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), a method of detecting release of radioactive zinc-65, a method of detecting fluorescence, and a method of detecting gel mobility shift.
20. The compound or salt of any one of claims 1-4 for use according to any one of claims 16-19, wherein the zinc finger-containing protein is present in a mammal.
21. The compound or salt of any one of claims 1 -4 for use according to any one of claims 16-19, wherein the zinc finger-containing protein is present on a surface of a mammal.
22. The compound of any one of claims 1-4 and pharmaceutically acceptable salts thereof for use in inactivating a virus, the use comprising contacting a virus with a compound of any one of claims 1-4 or a pharmaceutically acceptable salt thereof, whereby contacting the virus with said compound or salt inactivates the virus.
23. The compound or salt of any one of claims 1 -4 for use according to claim 22, wherein the virus is selected from the group consisting of an HIV-1 , an HIV-2, an SIV, a BIV, an EIAV, a Visna, a CaEV, an HTLV-1, a BLV, an MPMV, an MMTV, an RSV, an MuLV, a FeLV, a BaEV and an SSV retrovirus.
24. The compound or salt of any one of claims 1-4 for use according to claim 23, wherein the virus is an HIV-1 retrovirus.
25. The compound or salt of any one of claims 1 -4 for use according to claim 23, wherein the virus is selected from the group consisting of a retrovirus of an avian sarcoma and leukosis retroviral group, a mammalian B-type retroviral group, a human T cell leukemia and bovine leukemia retroviral group, a D-type retroviral group, a murine leukemia-related group and a lenti virus group.
26. The compound or salt of any one of claims 1 -4 for use according to claim 22, wherein the contacting of the virus with the compound or salt is performed in vivo.
27. The compound or salt of any one of claims 1 -4 for use according to claim 26, wherein the compound or salt is for administration to inhibit the transmission of the virus.
28. The compound or salt of any one of claims 1 -4 for use according to claim 26, wherein the compound or salt is for administration intra-vaginally or intra-rectally to inhibit the transmission of the virus.
29. The compound or salt of any one of claims 1 -4 for use according to claim 26, wherein the compound or salt is for administration parenterally, intrathecally,
subcutaneously, or orally.
30. The compound or salt of any one of claims 1 -4 for use according to claim 26, wherein the compound or salt is for administration to a human as a pharmaceutical formulation.
31. The compound or salt of any one of claims 1 -4 for use according to claim 26, wherein the compound or salt is for administration to an animal as a veterinary
pharmaceutical formulation.
32. The compound or salt of any one of claims 1-4 for use according to any one of claims 26-31 , wherein the use further comprises contacting the virus with another anti- retro viral agent.
33. The compound or salt of any one of claims 1-4 for use according to claim 32, wherein said another anti-retroviral agent is selected from the group consisting of a nucleoside analogue, a nucleotide analogue, a reverse transcriptase inhibitor, an integrase inhibitor, a fusion inhibitor, and a protease inhibitor.
34. The compound or salt of any one of claims 1-4 for use according to claim 33, wherein the nucleoside analogue is an AZT, a ddCTP or a ddl.
35. The compound or salt of any one of claims 1-4 for use according to claim 32, wherein said another anti-retroviral agent is selected from the group consisting of
enfuviritide, maraviroc, delavirdine, etravirine, efavirenz, nevirapine, zidovudine, lamivudine, emtricitabine, didanosine, tenofovir, stavudine, abacavir, raitegravir,
dolutegravir, amprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, and nelfmavir.
36. The compound or salt of any one of claims 1 -4 for use according to claim 22, wherein the contacting of the virus with the compound or salt is performed on a blood product, blood plasma, nutrient media, protein, a pharmaceutical, a cosmetic, a sperm or oocyte preparation, cells, cell cultures, bacteria, viruses, food, drink, implant or prosthesis.
PCT/US2014/054019 2013-09-05 2014-09-04 Thioether prodrug compositions as anti-hiv and anti-retroviral agents WO2015035003A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361874182P 2013-09-05 2013-09-05
US61/874,182 2013-09-05

Publications (1)

Publication Number Publication Date
WO2015035003A1 true WO2015035003A1 (en) 2015-03-12

Family

ID=51570884

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/054019 WO2015035003A1 (en) 2013-09-05 2014-09-04 Thioether prodrug compositions as anti-hiv and anti-retroviral agents

Country Status (1)

Country Link
WO (1) WO2015035003A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019197468A1 (en) * 2018-04-12 2019-10-17 Bayer Aktiengesellschaft N-(cyclopropylmethyl)-5-(methylsulfonyl)-n-{1-[1-(pyrimidin-2-yl)-1h-1,2,4-triazol-5-yl]ethyl}benzamide derivatives and the corresponding pyridine-carboxamide derivatives as pesticides
CN111647034A (en) * 2020-05-13 2020-09-11 山东大学 2-mercaptobenzamide thioester compound and preparation method and application thereof
CN112203655A (en) * 2018-05-16 2021-01-08 爱维斯健有限公司 Pharmaceutical composition for preventing or treating AIDS comprising rhodanine derivative
WO2021127133A1 (en) * 2019-12-17 2021-06-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services N-(3-amino-3-oxopropyl)-2-[(1-methyl-4-nitro-1h-imidazol-5-yl)thio]benzamide and its use for treating hiv infection

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4155991A (en) 1974-10-18 1979-05-22 Schering Aktiengesellschaft Vaginal ring
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US5989581A (en) 1997-04-11 1999-11-23 Akzo Nobel N.V. Drug delivery system for two or more active substances
US6046228A (en) 1996-07-05 2000-04-04 The United States Of America As Represented By The Department Of Health And Human Services Anti-viral pharmaceutical compositions containing saturated 1,2-dithiaheterocyclic compounds and uses thereof
US6126958A (en) 1996-07-31 2000-10-03 The Population Council, Inc. Intravaginal rings with insertable drug-containing core
WO2003014062A1 (en) * 2001-08-03 2003-02-20 The Government Of The United States As Represented By The Secretary Of The Department Of Health And Human Services Acylthiols and component thiol compositions as anti-hiv and anti-retroviral agents
WO2011156674A2 (en) * 2010-06-10 2011-12-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Thioether prodrug compositions as anti-hiv and anti-retroviral agents

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4155991A (en) 1974-10-18 1979-05-22 Schering Aktiengesellschaft Vaginal ring
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US6046228A (en) 1996-07-05 2000-04-04 The United States Of America As Represented By The Department Of Health And Human Services Anti-viral pharmaceutical compositions containing saturated 1,2-dithiaheterocyclic compounds and uses thereof
US6126958A (en) 1996-07-31 2000-10-03 The Population Council, Inc. Intravaginal rings with insertable drug-containing core
US5989581A (en) 1997-04-11 1999-11-23 Akzo Nobel N.V. Drug delivery system for two or more active substances
WO2003014062A1 (en) * 2001-08-03 2003-02-20 The Government Of The United States As Represented By The Secretary Of The Department Of Health And Human Services Acylthiols and component thiol compositions as anti-hiv and anti-retroviral agents
US7528274B2 (en) 2001-08-03 2009-05-05 The United States Of America As Represented By The Department Of Health And Human Services Acylthiols and component thiol compositions as anti-HIV and anti-retroviral agents
WO2011156674A2 (en) * 2010-06-10 2011-12-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Thioether prodrug compositions as anti-hiv and anti-retroviral agents

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Archives of Virology", vol. 10, 1995, SPRINGER VERLAG, pages: 586
"Pharmaceutics and Pharmacy Practice", 1982, J. B. LIPPINCOTT COMPANY, pages: 238 - 250
"Physicians ' Desk Reference", 2004, THOMSON PDR
BERG, SCIENCE, vol. 232, 1986, pages 485
BESS, J. VIROL., vol. 66, 1992, pages 840
CHANCE, PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 10041
DEMENE, BIOCHEMISTRY, vol. 33, 1994, pages 11707 - 11716
FEHRMANN, VIROLOGY, vol. 235, 1997, pages 352 - 359
HENDERSON, J. BIOL. CHEM., vol. 256, 1981, pages 8400
HUANG, J VIROL., vol. 71, 1997, pages 4378 - 4384
HUMPHREY, ANTIMICROB. AGENTS & CHEMOTHERAPY, vol. 41, 1997, pages 1017 - 1023
JACKS, NATURE, vol. 331, 1988, pages 280 - 283
KARACOSTAS, VIROLOGY, vol. 193, 1993, pages 661 - 671
LAPADAT-TAPOLSKY, J. MOL. BIOL., vol. 268, 1997, pages 250 - 260
NASSER, J. VIROL., vol. 70, 1996, pages 8639 - 8644
P. K. VOGT: "Retroviruses", 1997, COLD SPRING HARBOR LABORATORY PRESS, article "Historical introduction to the general properties of retroviruses", pages: 1 - 26
PHYSICIANS' DESK REFERENCE, 2004
PRICHARD, ANTIVIRAL RESEARCH, vol. 14, 1993, pages 181 - 206
SOUTH, ADV. INORG. BIOCHEM., vol. 8, 1990, pages 199
SOUTH, BIOCHEM. PHANNACOL., vol. 40, 1990, pages 123 - 129
SZOKA ET AL., ANN. REV. BIOPHYS. BIOENG., vol. 9, 1980, pages 467
THOMAS; GORELICK, VIRUS RES., vol. 134, 2008, pages 39 - 63
TOISSEL: "ASHP Handbook on Injectable Drugs", 1986, pages: 622 - 630
ULLMAN, BIOCHEM. J., vol. 319, 1996, pages 229 - 239

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019197468A1 (en) * 2018-04-12 2019-10-17 Bayer Aktiengesellschaft N-(cyclopropylmethyl)-5-(methylsulfonyl)-n-{1-[1-(pyrimidin-2-yl)-1h-1,2,4-triazol-5-yl]ethyl}benzamide derivatives and the corresponding pyridine-carboxamide derivatives as pesticides
IL277777B1 (en) * 2018-04-12 2023-06-01 Bayer Ag N-(cyclopropylmethyl)-5-(methylsulfonyl)-n-{1-[1-(pyrimidin-2-yl)-1h-1,2,4-triazol-5-yl]ethyl}benzamide derivatives and the corresponding pyridine-carboxamide derivatives as pesticides
CN112203655A (en) * 2018-05-16 2021-01-08 爱维斯健有限公司 Pharmaceutical composition for preventing or treating AIDS comprising rhodanine derivative
JP2021531239A (en) * 2018-05-16 2021-11-18 アヴィックスジェン・インコーポレイテッド A pharmaceutical composition for the prevention or treatment of AIDS containing a rhodanine derivative.
WO2021127133A1 (en) * 2019-12-17 2021-06-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services N-(3-amino-3-oxopropyl)-2-[(1-methyl-4-nitro-1h-imidazol-5-yl)thio]benzamide and its use for treating hiv infection
CN111647034A (en) * 2020-05-13 2020-09-11 山东大学 2-mercaptobenzamide thioester compound and preparation method and application thereof
CN111647034B (en) * 2020-05-13 2021-05-28 山东大学 2-mercaptobenzamide thioester compound and preparation method and application thereof

Similar Documents

Publication Publication Date Title
KR101822348B1 (en) Methods for treating filoviridae virus infections
KR101883750B1 (en) Antibiral therapy
WO2015035003A1 (en) Thioether prodrug compositions as anti-hiv and anti-retroviral agents
CA2718175C (en) Use of 4'-thio-2'-deoxynucleosides as anti orthopoxvirus agents
AU2012300274B2 (en) HIV replication inhibitors
Hartman et al. Preclinical evaluation of a mercaptobenzamide and its prodrug for NCp7-targeted inhibition of human immunodeficiency virus
EP2580191A2 (en) Thioether prodrug compositions as anti-hiv and anti-retroviral agents
WO2013016441A1 (en) Hiv integrase inhibitory oxoisoindoline sulfonamides
US9181294B2 (en) Nucleoside analogues for the treatment of a viral infection, and method for evaluating the sensitivity to said treatment
EP3016714B1 (en) Inhibitors of viral replication, their process of preparation and their therapeutical uses
WO2020023849A1 (en) Compositions and methods for targeting a viral infection
WO2019021319A1 (en) Pharmaceutical compositions
NZ622558B2 (en) Hiv replication inhibitors
EP1722782A2 (en) Compounds, compositions and methods for inhibiting or treating hiv-1

Legal Events

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

Ref document number: 14767228

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14767228

Country of ref document: EP

Kind code of ref document: A1