WO2011046901A2 - Antiviral à large spectre et procédés d'utilisation - Google Patents

Antiviral à large spectre et procédés d'utilisation Download PDF

Info

Publication number
WO2011046901A2
WO2011046901A2 PCT/US2010/052255 US2010052255W WO2011046901A2 WO 2011046901 A2 WO2011046901 A2 WO 2011046901A2 US 2010052255 W US2010052255 W US 2010052255W WO 2011046901 A2 WO2011046901 A2 WO 2011046901A2
Authority
WO
WIPO (PCT)
Prior art keywords
virus
dna
hydrogen
rna
administering
Prior art date
Application number
PCT/US2010/052255
Other languages
English (en)
Other versions
WO2011046901A3 (fr
Inventor
Dale M. Walker
Vernon E. Walker
Original Assignee
Walker Dale M
Walker Vernon E
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 Walker Dale M, Walker Vernon E filed Critical Walker Dale M
Publication of WO2011046901A2 publication Critical patent/WO2011046901A2/fr
Publication of WO2011046901A3 publication Critical patent/WO2011046901A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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

Definitions

  • a method for the prevention or treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses infection by administering an effective amount of a compound of Formula (I), Formula (II) or a derivative thereof to an individual in need is provided.
  • Viruses can be divided into several (arbitrary) classifications based upon the type of nucleic acid that they carry and the mode of expression.
  • Table 1 illustrates one classification scheme based upon "The International Code of Virus Classification and Nomenclature", and the classification system developed by Dr. David Baltimore (incorporated herein by reference) TABLE 1
  • a limited number of chronic viral diseases which affect millions of people world-wide (e.g., 40 million individuals are HIV-infected), can be controlled to some degree, but no cures are currently available.
  • infected individuals provide a reservoir for the respective virus through which naive individuals become infected, thereby, perpetuating the problem of viral infection of significant human populations with these pathogens.
  • antiviral drugs such as the nucleoside reverse transcriptase inhibitors (NRTIs) used to inhibit viral replication of specific pathogenic viruses, have resulted in recognizable improvements in the ability to control infections with these pathogens and to improve the quality and length of life of infected individuals.
  • NRTIs nucleoside reverse transcriptase inhibitors
  • a partial list of currently available antiviral drugs used to inhibit viral replication of specific pathogenic viruses is provided in Appendix 2 (for eg. the NRTIs).
  • the therapies listed have resulted in recognizable improvements in the ability to control infections from these pathogens and to improve the quality and length of life of infected individuals.
  • currently available classes of antiviral agents have limited utilities due to their narrow scope of activities against different viruses and/or problems with significant drug- induced toxicities.
  • the modes of action of the NRTIs and other drugs in current clinical use predispose to the development of drug resistance through viral mutations.
  • many currently available drugs have considerable side effects that prevent their wide spread use to achieve treatment or prevention goals.
  • compositions and methods of the preferred embodiments provide such agents and associated methods of treatment.
  • Amifostine is a pro-drug that is metabolized by alkaline phosphatase to the reduced free thiol (WR- 1065); oxidation of WR-1065 leads to formation of the disulfide (WR-33278).
  • WR-1065 and WR-33278 H 2 N(CH 2 ) 3 NH(CH2) 2 (CH 2 ) 2 NH(CH 2 ) 3 NH 2 ) and WR-1065 and WR-33278.
  • Spermine is associated with nucleic acids and is thought to stabilize helical structure, particularly in viruses.
  • WR-1065 appears to function as an analog of the polyamine spermine, and to compete with spermine for sites on DNA, and probably also on other nucleic acids and proteins.
  • analogs of WR-1065 and the other compounds of preferred embodiments, as discussed below, may function as spermine or other polyamine analogs, and may mimic the antiviral and polyamine-like activity of WR-1065.
  • These functions include, but are not limited to, (i) protection against radiation induced cytotoxicity and cell killing, (ii) protection against radiation-induced mutagenesis/carcinogenesis, (iii) modulation of topoisomerase I and topoisomerase II alpha activities, (iv) modulation of conformational changes in chromatid structure, (v) inhibition of cell cycle progression, (vi) inhibition of endonuclease activity, (vii) competition with spermine in polyamine transport systems, (viii) induction and repression of gene expression (effect dependent upon the particular gene).
  • Other activities include detoxifying cisplatin and other alkylating agents, scavenging free radicals, modulating apoptosis, and modifying the activity of specific enzymes/proteins.
  • tissues are especially effective at uptake of the drugs (and/or their metabolites) and retention of those metabolites; these tissues include: kidney, liver, salivary gland, and lung (Rasey et al., (1984) , Radial Res. 97(3): 598-607). It is possible that other tissues that have not been tested also perform like the above mentioned tissues. These studies establish the ability to obtain therapeutic levels of these drugs and/or their metabolites in the cells of these tissues/organ systems. Therapeutic drug levels are also expected to be obtained in tissues or organs systems whe-ie drug pharmacokinetics resemble that of the above mentioned tissues.
  • PLGA/amifostine nanoparticles showed high levels of retention in ail evaluated tissues 30 minutes after
  • liver Retention of drug four hours after administration was demonstrated in the following tissues (in order from highest to lowest): liver, jejunum, stomach, ileum, duodenum, bone marrow, and spleen.
  • cytoprotective effects of amifostine appear to be dependent upon a number of factors including, but not limited to, oxygen tension, pH, gene status (including the presence of a functional p53 gene), and
  • tumor cells and normal, nontumorigenic tissue.
  • amifostine, phosphonol and their derivatives and analogs are particularly useful
  • DNA or RNA reverse transcribing virus infection in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of a compound or a pharmaceutically
  • X is selected from the group consisting of -P0 3 H 2 , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R-i, R 2> and R 3 is independently selected from hydrogen and C, ⁇ alkyl, and wherein n is an integer of from 1 to 10.
  • the compound is of Formula (I), R, is methyl, R 2 is hydrogen, R 3 is hydrogen, and n is 3. [0015] In an embodiment of the first aspect, the compound is of Formula (I), R-i is methyl, R 2 is hydrogen, R 3 is hydrogen, and X is hydrogen.
  • the compound is of Formula (I), R, is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
  • the compound is of Formula (I), Rt is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and X is hydrogen.
  • the compound is of Formula (li), is methyl, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
  • the compound is of Formula (II), R-t is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
  • the compound is administered to the individual at a daily dosage of from about 200 mg/m 2 to about 3000 mg/m z .
  • the step of administering is selected from the group consisting of orally administering, subcutaneously administering, intravenously administering, parenterally administering, and administering by inhalation.
  • the method further comprises the step of administering to the individual an effective antiviral amount of an antiviral drug, for example (see the list of antiviral drugs in Appendix 2) to treat or prevent an viral infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus.
  • an antiviral drug for example (see the list of antiviral drugs in Appendix 2) to treat or prevent an viral infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus.
  • a method of treating or preventing a human or animal viral infection caused by a DNA virus, a RNA virus, a DNA or a RNA reverse transcribing virus in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of amifostine, the free thiol form of amifostine, the disulfide of amifostine (WR-33278), a combination of both of the free thiol and the disulfide of amifostine, or other structurally and functionally related compounds as described in this document.
  • a method of treating or preventing an infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus, or combination thereof in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of phosphonol, the free thiol form of phosphonol, the disulfide of phosphonol, a combination of both the free thiol and the disulfide of phosphonol, or other structurally and functionally related compounds as described in this document.
  • the viral infection is caused by a dsDNA virus.
  • the viral infection is caused by a dsDNA virus.
  • the viral infection is caused by a dsRNA virus.
  • the viral infection is caused by a dsRNA virus.
  • the viral infection is caused by a (+)ssRNA virus.
  • the viral infection is caused by a (+)ssRNA virus.
  • the viral infection is caused by a (-)ssRNA virus.
  • the viral infection is caused by a (-)ssRNA virus.
  • the viral infection is caused by a non Retroviridae ssRNA reverse transcribing virus.
  • the viral infection is caused by a non Retroviradae ssRNA reverse transcribing virus.
  • the viral infection is caused by a dsDNA reverse transcribing virus.
  • the viral infection is caused by a dsDNA reverse transcribing virus.
  • the viral infection is caused by a ssDNA reverse transcribing virus.
  • the viral infection is caused by a ssDNA reverse transcribing virus.
  • a pharmaceutical kit comprising a pharmaceutical composition comprising a compound or pharmaceutically acceptable salt or solvate thereof in a pharmaceutically accepta
  • X is selected from the group consisting of -P0 3 H r , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R 1 ; R 2 , and R 3 is independently selected from hydrogen and d-e alkyl, and wherein n is an integer of from 1 to 10; and
  • RNA virus or a DNA or RNA reverse transcribing virus.
  • the kit further comprises an antiviral drug, selected on the basis of its effectiveness for treating the given viral agent, in a pharmaceutically acceptable carrier.
  • the kit further comprises an antiviral drug in a pharmaceutically acceptable carrier and directions for administering the antiviral drug in a pharmaceutically acceptable carrier to the patient.
  • Amifostine is an organic thiophosphate which selectively protects normal tissues but not tumors against cytotoxicity of ionizing radiations, DNA-binding chemotherapeutic agents (e.g., classical alkylating agents such as cyclophosphamide and non-classical alkylating agents such as mitomycin-C and platinum analogs). Amifostine is a prodrug that is dephosphorylated to the active metabolite, the free thiol form, by alkaline phosphatase and exits the bloodstream rapidly.
  • DNA-binding chemotherapeutic agents e.g., classical alkylating agents such as cyclophosphamide and non-classical alkylating agents such as mitomycin-C and platinum analogs.
  • Amifostine is a prodrug that is dephosphorylated to the active metabolite, the free thiol form, by alkaline phosphatase and exits the bloodstream rapidly.
  • the compounds of preferred embodiments including amifostine and its derivatives and analogs, are particularly effective antiviral compounds for use in inhibiting replication of diverse species of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses.
  • Amifostine (referred to as "WR-2721") is marketed under the name Ethyoi® by Schering-Plough Pty Ltd. It has the chemical name S-2-(3-aminopropyl)aminoethyl phosphorothioic acid and the structure:
  • a particularly preferred antiviral compound for use in inhibiting replication of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses includes the free thiol form of amifostine.
  • the free thiol form (referred to as "WR-1065”) has the chemical name 2-(3-aminopropylamino)ethanethiol and the following structure:
  • the disulfide form of amifostine (referred to as WR-33278) has the chemical name
  • Phosphonol (referred to as "WR-3689”) is structurally similar to amifostine, the only difference being the presence of a terminal methyl group.
  • Phosphonol has the chemical name S-2-(3- (methylamino)propylamino)ethyl phosphorothioic acid and the following structure: OH
  • WR-255591 The free thiol form of phosphonol (referred to as WR-255591) has the chemical name 2-(3- (methylamino)propylamino) ethanethiol and the following structure:
  • the disulfide form of phosphonol (referred to as WR-33278) has the following structure:
  • the compounds of preferred embodiments include prodrug forms of the above-described free thiol forms of amifostine, phosphonol, and analogs thereof.
  • prodrugs include compounds of the structure:
  • R 1 R 2 , and R 3 is independently selected from hydrogen and lower alkyl
  • -(C n H 2n )- is lower alkyl and n is 1 , 2, 3, 4, 5, or (up to 10), and
  • X is a suitable leaving group.
  • Suitable leaving groups include -P0 3 H 2 , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl); however, any other suitable leaving group can be employed that yields the active free thiol form of the compound when metabolized in vivo.
  • lower alkyl as used herein is a broad term, and is to be given its ordinary and customary meaning to a person of ordinary skill in the art (and is not to be limited to a special or customized meaning), and refers without limitation to a straight chain or branched chain, acyclic or cyclic, saturated aliphatic hydrocarbon containing 1 , 2, 3, 4, 5, or 6 carbon atoms.
  • Saturated straight chain lower alkyis include methyl, ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl; while representative saturated branched chain alkyis include isopropyl, sec-butyl, isobutyl, ferf-butyl, and isopentyl.
  • aryl as used herein is a broad term and is used in its ordinary sense, including, without limitation, to refer to an aromatic carbocyclic moiety such as phenyl or naphthyl, including mono-, di-, and poly-homocyclic aromatic ring systems ⁇ e.g. , C 6 -ie aryl)- [0051]
  • the prodrug forms described above are metabolized into active thiols of the formula:
  • each of R ⁇ R 2 , R 3 , -(C n H 2n )-, and n is as defined above.
  • -(C n H 2n )- is a straight alkyl chain having three carbon atoms. It is also particularly preferred that R 2 and R 3 are both hydrogen, and that R-, is hydrogen or methyl.
  • disulfide forms also exhibit antiviral activity, for example, a disulfide of the following structure:
  • cysteamine-like group i.e., a group containing the moiety >N-(CH 2 ) 2 -S-
  • a moiety with the structure of WR 2721 or WR-1065 tethered to a DNA or nucleic acid binding agent, or other agents with some affinity for nucleic acids or proteins.
  • the compounds of preferred embodiments are particularly effective antiviral agents for
  • the compounds of preferred embodiments are generally administered at dosages equal to or less than the oral radioprotective dosage of arnifostine ⁇ e.g. , 1456 mg total dose, or 910 mg/m 2 for a 60 kg body weight (BW) adult human) or phosphonol (e.g. , 725 mg/m 2 ).
  • the recommended starting dose of arnifostine is 910 mg/m 2 for a 60 kg BW adult human administered once daily as a 15-minute i.v. infusion, starting 30 minutes prior to chemotherapy.
  • Dosages can be converted from mg/m 2 to total mg or mg/kg BW (see, e.g. , Freireich et al. (1966), Cancer Chemother. Reports, 50 (4) 219-244) as in Table 2.
  • arnifostine or other compounds of preferred embodiments
  • dosages 910 mg/m 2 or less.
  • This dosage is equivalent to 24.3 ,g/kg BW for a 60 kg BW adult human being (or a total dose of 1456 mg for a 60 kg BW adult, as described above); however, in certain embodiments it can be desirable to administer the compounds of preferred embodiments at higher dosage levels.
  • children have been given up to a 2700 mg/m 2 total dose of amifostine prior to administration of a chemotherapeutic agent. In some individuals such high doses are associated with side effects.
  • a dose of 740 mg/m 2 amifostine (1148 mg total, for a 60 kg BW human adult) is associated with fewer side effects (List et al. (1997), Blood 90(9): 3364-9), and is thus generally preferred.
  • 200-340 mg/m 2 of amifostine (544 mg total dose for a 60 kg BW adult) is generally preferred (Schuchter (1996), Semin Oncol 23(4 Suppl 8): 40-3; Santini et al. (1999), Haematologlca 84(1 1): 1035-42).
  • the maximally tolerated dose (MTD) for WR-2721 in mice was 432 mg/kg (BW) administered i.p. and 720 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD.
  • the MTD was 893 mg/kg BW administered i.p. and 1488 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD.
  • All of the aminothiols have MTDs in rodents of greater than 400 mg/kg BW.
  • Amifostine and WR-1065 can be efficacious at very low concentrations, for example, down to 0.4 micromolar concentrations in some in vitro studies.
  • a drug delivery system that obtains relatively constant intracellular concentrations over a period of time that could extend from several days to up to one year or more is desired.
  • drug delivery systems include subcutaneous administration, formulation in nanoparticles, were formulation in other slow release systems.
  • Table 3 provides plasma concentrations of amifostine metabolites (see Geary et al., Res. Comm. Chem. Path. Pharmacol. , 65(2), 147-159 (1989)) and phosphonol metabolites (see Buckpitt er a . , Contract #DAMD 17-86-C-6177, reference obtained from Dr. D. Grdina, personal communication) after duodenal administration of 150 mg/kg BW of each drug in rhesus macaques. This data was collected as part of work performed to evaluate the radioprotective activity of the compounds. These data may be useful in estimating plasma concentrations in humans.
  • amifostine While it is generally preferred to formulate amifostine or the other compounds of preferred embodiments for oral administration, the compounds of preferred embodiments can be formulated so as to allow them to be administered by other routes, as discussed herein. It can be desirable in certain embodiments to formulate amifostine for intravenous administration in order to maximize efficacy. Because of the structural similarities between amifostine and phosphonol, especially the similarities in the sulfhydryl ends of the molecules, phosphonol is expected to behave in a manner similar to amifostine rather than WR-151327 (chemical formula CH3 H(CH2)3NH(CH2) 3 SP0 3 H 2 ). WR-151327 has been previously shown to have anti-HIV activity; this activity was attributed to the ability of the compound to modulate cytokine levels in a manner that inhibited HIV replication and/or proliferation (Kalebic et al., 1994).
  • Phosphonol may not be quite as efficacious as amifostine, based upon the work of Gutschow et al. , who found lower activity when a methyl group was substituted for a hydrogen atom in a position similar to that of the methyl group of phosphonol that distinguishes its structure frcm that of amifostine (Gutschow et al. (1995), Pharmazie 50(10): 672-5).
  • phosphonol is expected to be significantly more efficacious than WR-151327. This consideration should be viewed in light of the fact that the overall structures of the compounds tested by Gutschow et al. were significantly different from the structures of the preferred embodiments. The overall structures of the compounds tested by Gutschow ef al. are significantly different from the structures of the preferred embodiments, and it is expected that phosphonol will be significantly more efficacious than WR-151327.
  • Amifostine, its analogs, and its derivatives can be administered in combination with other antiviral agents employed to treat HIV infection.
  • One of the benefits of such combination therapies is that lower doses of the other antiviral agents can be administered while still achieving a similar level of antiviral efficacy.
  • Such lower dosages can be particularly advantageous for drugs known to have genotoxicity and mitochondrial toxicity (for example, some nucleoside analogs).
  • greater efficacy might be achieved using therapeutic doses of two drugs than could be achieved using only a single drug.
  • nucleoside analogs members of the class of drugs known as nucleoside analogs.
  • Other agents that could be used potentially are included in the list of antiviral drugs included in Appendix 2.
  • nucleoside analogs has been associated with a variety of side effects due to the fact that these drugs are analogs of naturally occurring nucleosides, are incorporated into host cell DNA, and function as obligate DNA chain terminators. As such, these drugs are associated with the induction of mutations in host cell DNA, increased risks for certain types of cancer, as well as increased risks for a variety of diseases associated with mutation induction in nuclear and mitochondrial DNA.
  • Nucleoside analogs are also associated with mitochondrial toxicity to a degree that varies depending upon the specific nucleoside analog in question. Thus, it is desirable to be able to minimize the doses of nucleoside analogs used, and/or to use these drugs in combination with other drugs with demonstrated antiviral efficacy that does not compromise the effectiveness of the other treatment, and that also has demonstrated antimutagenic efficacy.
  • compositions Comprising Amifostine and Analogs Thereof:
  • the compounds of preferred embodiments can be incorporated into a pharmaceutically acceptable carrier , including incorporation into nanoparticles for administration to an individual having a viral infection (for example, an adenoviral infection as an example of a DNA virus, an influenza infection as an example of a RNA virus, or an HIV infection as an example of a DNA or RNA reverse transcribing virus) or can be administered
  • a viral infection for example, an adenoviral infection as an example of a DNA virus, an influenza infection as an example of a RNA virus, or an HIV infection as an example of a DNA or RNA reverse transcribing virus
  • the compounds of preferred embodiments can be employed as the sole agent in the prevention or treatment of DNA viruses, RNA viruses, or DNA or RNA reverse transcribing viruses, and/or combinations thereof two or more such compounds can be employed, optionally in combination with other therapeutic agents, e.g., conventional or newly developed drugs employed in the treatment of AIDS or HIV, or other viral infections.
  • reducing agents including, but not limited to, vitamin C and vitamin E.
  • Other reducing agents include organic aldehydes, hydroxyl-containing aldehydes, and reducing sugars such as glucose, mannose, galactose, xylose, ribose, and arabinose.
  • Other reducing sugars containing hemiacetal or keto groupings can be employed, for example, maltose, sucrose, lactose, fructose, and sorbose.
  • reducing agents include alcohols, preferably polyhydric alcohols, such as glycerol, sorbitol, glycols, especially ethylene glycol and propylene glycol, and polyglycols such as polyethylene and polypropylene glycols.
  • polyhydric alcohols such as glycerol, sorbitol, glycols, especially ethylene glycol and propylene glycol, and polyglycols such as polyethylene and polypropylene glycols.
  • pharmaceutically acceptable salts and "a pharmaceutically acceptable salt thereof” as used herein are broad terms, and are to be given their ordinary and customary meaning to a person of ordinary skill in the art (and are not to be limited to a special or customized meaning), and refer without limitation to salts prepared from pharmaceutically acceptable, non-toxic acids or bases. Suitable
  • salts include metallic salts, e.g., salts of aluminum, zinc, alkali metal salts such as lithium, sodium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts; organic salts, e.g. , salts of lysine, ⁇ , ⁇ '-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and tris; salts of free acids and bases; inorganic salts, e.g.
  • Any suitable constituent can be selected to make a salt of the therapeutic agents discussed herein, provided that it is non-toxic and does not substantially interfere with the desired activity.
  • pharmaceutically acceptable precursors and derivatives of the compounds can be employed.
  • compositions and methods of preferred embodiments can also be suitable for use in compositions and methods of preferred embodiments. While it may be possible to administer the compounds of the preferred embodiments in the form of pharmaceutically acceptable salts, it is generally preferred to administer the compounds in neutral form. . [0067] It is generally preferred to administer the compounds of preferred embodiments orally; however, other routes of administration are contemplated. Contemplated routes of administration include but are not limited to oral, sublingual, parenteral, transcutaneous, subcutaneous, intravenous, and by inhalation. Compounds of preferred embodiments can be formulated into liquid preparations for, e.g. , oral administration. Suitable forms for such administration include suspensions, syrups, and elixirs.
  • compositions of preferred embodiments are preferably isotonic with the blood or other body fluid of the recipient.
  • the isotonicity of the compositions can be attained using sodium tartrate, propylene glycol or other inorganic or organic solutes.
  • Sodium chloride is particularly preferred.
  • Buffering agents can be employed, such as acetic acid and salts, citric acid and salts, boric acid and salts, and phosphoric acid and salts.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like.
  • a reducing agent such as vitamin C, vitamin E, or other reducing agents as are known in the pharmaceutical arts, in the formulation.
  • Viscosity of the pharmaceutical compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent.
  • Methylcellulose is preferred because it is readily and economically available and is easy to work with.
  • Other suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like. The preferred concentration of the thickener will depend upon the thickening agent selected. An amount is preferably used that will achieve the selected viscosity. Viscous compositions are normally prepared from solutions by the addition of such thickening agents.
  • a pharmaceutically acceptable preservative can be employed to increase the shelf life of the pharmaceutical compositions.
  • Benzyl alcohol can be suitable, although a variety of preservatives including, for example, parabens, thimerosal, chlorobutanol, or benzalkonium chloride can also be employed.
  • a suitable concentration of the preservative is typically from about 0.02% to about 2% based on the total weight of the composition, although larger or smaller amounts can be desirable depending upon the agent selected.
  • Reducing agents as described above, can be advantageously used to maintain good shelf life of the formulation.
  • the compounds of preferred embodiments can be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, or the like, and can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • a suitable carrier diluent, or excipient
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • Such preparations can include complexing agents, metal ions, polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, dextran, and the like, liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts.
  • Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. The presence of such additional components can influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance, and are thus chosen according to the intended application, such that the characteristics of the carrier are tailored to the selected route of administration.
  • compositions intended for oral use can be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can include one or more of the following agents: sweeteners, flavoring agents, coloring agents and preservatives.
  • Aqueous suspensions can contain the active ingredient in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • Formulations for oral use can also be provided as hard gelatin capsules, wherein the active ingredient(s) are mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate, or kaolin, or as soft gelatin capsules.
  • an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin
  • the active compounds can be dissolved or suspended in suitable liquids, such as water or an oil medium, such as peanut oil, olive oil, fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • Stabilizers and microspheres formulated for oral administration can also be used.
  • Capsules can include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredient in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • binders such as starches
  • lubricants such as talc or magnesium stearate
  • stabilizers optionally, stabilizers.
  • a reducing agent in the capsule.
  • Tablets can be uncoated or coated by known methods to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period of time.
  • a time delay material such as glyceryl monostearate can be used.
  • the solid form typically comprises from about 0.001 wt. % or less to about 50 wt. % or more of active ingredient(s), preferably from about 0.005, 0.01 , 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1 wt. % to about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or 45 wt. %.
  • Tablets can contain the active ingredients in admixture with non-toxic pharmaceutically acceptable excipients including inert materials.
  • a tablet can be prepared by compression or molding, optionally, with one or more additional ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding, in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • each tablet or capsule contains from about 10 mg or less to about 1 ,000 mg or more of a compound of the preferred embodiments, more preferably from about 20, 30, 40, 50, 60, 70, 80, 90, or 100 mg to about 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, or 900 mg.
  • tablets or capsules are provided in a range of dosages to permit divided dosages to be administered. A dosage appropriate to the patient and the number of doses to be administered daily can thus be conveniently selected.
  • Suitable inert materials include diluents, such as carbohydrates, mannitol, lactose, anhydrous lactose, cellulose, sucrose, modified dextrans, starch, and the like, or inorganic salts such as calcium triphosphate, calcium phosphate, sodium phosphate, calcium carbonate, sodium carbonate, magnesium carbonate, and sodium chloride.
  • diluents such as carbohydrates, mannitol, lactose, anhydrous lactose, cellulose, sucrose, modified dextrans, starch, and the like
  • inorganic salts such as calcium triphosphate, calcium phosphate, sodium phosphate, calcium carbonate, sodium carbonate, magnesium carbonate, and sodium chloride.
  • Disintegrants or granulating agents can be included in the formulation, for example, starches such as corn starch, alginic acid, sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite, insoluble cationic exchange resins, powdered gums such as agar, karaya or tragacanth, or alginic acid or salts thereof.
  • starches such as corn starch, alginic acid, sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite, insoluble cationic exchange resins, powdered gums such as agar, karaya or tragacanth, or alginic acid or salts thereof.
  • Binders can be used to form a hard tablet. Binders include materials from natural products such as acacia, tragacanth, starch and gelatin, methyl cellulose, ethyl cellulose, carboxymethyl cellulose, polyvinyl pyrrolidone, hydroxypropylmethyl cellulose, and the like.
  • Lubricants such as stearic acid or magnesium or calcium salts thereof, polytetrafluoroethylene, liquid paraffin, vegetable oils and waxes, sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol, starch, talc, pyrogenic silica, hydrated silicoaluminate, and the like, can be included in tablet formulations.
  • Surfactants can also be employed, for example, anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate, cationic such as benzalkonium chloride or benzethonium chloride, or nonionic detergents such as polyoxyethylene hydrogenated castor oil, glycerol monostearate, polysorbates, sucrose fatty acid ester, methyl cellulose, or carboxymethyl cellulose.
  • anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate
  • cationic such as benzalkonium chloride or benzethonium chloride
  • nonionic detergents such as polyoxyethylene hydrogenated castor oil, glycerol monostearate, polysorbates, sucrose fatty acid ester, methyl cellulose, or carboxymethyl cellulose.
  • Controlled release formulations can be employed wherein the amifostine or analog(s) thereof is incorporated into an inert matrix that permits release by either diffusion or leaching mechanisms. Slowly degenerating matrices can also be incorporated into the formulation. Other delivery systems can include timed release, delayed release, or sustained release delivery systems.
  • Coatings can be used, for example, nonenteric materials such as methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl-methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols, or enteric materials such as phthalic acid esters.
  • Dyestuffs or pigments can be added for identification or to characterize different combinations of active compound doses.
  • a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils can be added to the active ingredient(s).
  • Physiological saline solution, dextrose, or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol are also suitable liquid carriers.
  • the pharmaceutical compositions can also be in the form of oil-in-water emulsions.
  • the oily phase can be a vegetable oil, such as olive or arachis oil, a mineral oil such as liquid paraffin, or a mixture thereof.
  • Suitable emulsifying agents include naturally-occurring gums such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan mono-oleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan mono-oleate.
  • the emulsions can also contain sweetening and flavoring agents.
  • a compound of the preferred embodiments When a compound of the preferred embodiments is administered by intravenous, parenteral, or other injection, it is preferably in the form of a pyrogen-free, parenterally acceptable aqueous solution or oleaginous suspension.
  • Suspensions can be formulated according to methods well known in the art using suitable dispersing or wetting agents and suspending agents. The preparation of acceptable aqueous solutions with suitable pH, isotonicity, stability, and the like, is within the skill in the art.
  • a preferred pharmaceutical composition for injection preferably contains an isotonic vehicle such as 1 ,3-butanediol, water, isotonic sodium chloride solution, Ringer's solution, dextrose solution, dextrose and sodium chloride solution, lactated Ringer's solution, or other vehicles as are known in the art.
  • an isotonic vehicle such as 1 ,3-butanediol, water, isotonic sodium chloride solution, Ringer's solution, dextrose solution, dextrose and sodium chloride solution, lactated Ringer's solution, or other vehicles as are known in the art.
  • sterile fixed oils can be employed
  • any bland fixed oil can be employed including synthetic mono or diglycerides.
  • fatty acids such as oleic acid can likewise be used in the formation of injectable preparations.
  • the pharmaceutical compositions can also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
  • the duration of the injection can be adjusted depending upon various factors, and can comprise a single injection administered over the course of a few seconds or less, to 0.5, 0.1 , 0.25, 0.5, 0.75, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, or 24 hours or more of continuous intravenous administration.
  • compositions of the preferred embodiments can additionally employ adjunct components conventionally found in pharmaceutical compositions in their art-established fashion and at their art- established levels.
  • the compositions can contain additional compatible pharmaceutically active materials for combination therapy (such as supplementary antimicrobials, antipruritics, astringents, local anesthetics, anti-inflammatory agents, reducing agents, and the like), or can contain materials useful in physically formulating various dosage forms of the preferred embodiments, such as excipients, dyes, thickening agents, stabilizers, preservatives or antioxidants.
  • the compounds of the preferred embodiments can be provided to an administering physician or other health care professional in the form of a kit.
  • the kit is a package which houses a container which contains the compound(s) in a suitable pharmaceutical composition, and instructions for administering the pharmaceutical composition to a subject.
  • the kit can optionally also contain one or more additional therapeutic agents, e.g., antiviral nucleoside analog, peptide analog, protease inhibitor, monoclonal antibody, or any other antiviral used for the treatment or prevention of virus disease (see for example Appendix 2).
  • kits containing one or more compositions comprising compound(s) of the preferred embodiments in combination with one or more additional antiviral, antibacterial, and/or anti-infective agents can be provided, or separate pharmaceutical compositions containing a compound of the preferred embodiments and additional therapeutic agents can be provided.
  • the kit can also contain separate doses of a compound of the preferred embodiments and additional therapeutic agents.
  • the kit can optionally contain one or more diagnostic tools and instructions for use.
  • the kit can contain suitable delivery devices, e.g. , syringes, and the like, along with instructions for administering the compound(s) and any other therapeutic agent.
  • the kit can optionally contain instructions for storage, reconstitution (if applicable), and administration of any or all therapeutic agents included.
  • the kits can include a plurality of containers reflecting the number of administrations to be given to a subject.
  • a kit for the treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses, and combinations thereof is provided that includes amifostine or another compound of a preferred embodiment and one or more antiviral agents currently employed to treat the virus.
  • Antiviral agents include for example, nucleoside analogs such as acyclovir, reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC; non-nucleoside reverse transcriptase inhibitors; protease inhibitors; cytokines; immunomodulators, and antibodies but are not limited thereto.
  • nucleoside analogs such as acyclovir
  • reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC
  • non-nucleoside reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC
  • non-nucleoside reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC
  • non-nucleoside reverse transcriptase inhibitors such as zidov
  • the compounds of the preferred embodiments can be administered prophylactically for the prevention of DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection.
  • therapy is preferably initiated as early as possible following the onset of signs and symptoms of a viral infection, or following exposure to a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection.
  • the administration route, amount administered, and frequency of administration will vary depending on the age of the patient, the severity of the infection, and any associated conditions.
  • Contemplated amounts, dosages, and routes of administration for the compounds of preferred embodiments for treatment of a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection are similar to those established for conventional antiviral agents.
  • anti-viral therapies combining an aminothiol with another antiviral therapy will have an additive or synergistic activity, making it possible to reduce the doses of the drugs currently used to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection, or making it possible to achieve higher efficacy by using therapeutic doses of two or more agents together.
  • the combination of an aminothiol with a nucleoside analog is expected to simultaneously provide improved antiviral therapy and protection against nucleoside analog-induced side effects, especially those associated with nuclear DNA or mitochondrial DNA damage. It is further anticipated that an aminothiol could protect against deleterious side effects of another antiviral drugs, especially those that induce nuclear DNA or mitochondrial DNA damage.
  • contemplated amounts of the compounds of the preferred embodiments for oral administration to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection are from about 10 mg or less to about 2000 mg or more administered from about every 4 hours or less to about every 6 hours or more (or from about 4 times daily to about 6 times daily) for about 5 days or less to about 10 days or more (40 mg/day or less to about 15,000 mg/day or more) or until there is a significant improvement in the condition.
  • doses of from about 10 mg or less to about 1000 mg or more are orally administered once, twice, or multiple times a day, typically for up to about 12 months, or, in certain circumstances, indefinitely (from about 10 mg/day to about 1 ,000 mg/day).
  • doses of from about 10 mg or less to about 1000 mg or more are orally administered once, twice, or multiple times a day, typically for up to about 12 months, or, in certain circumstances, indefinitely (from about 10 mg/day to about 1 ,000 mg/day).
  • it can be desirable to vary the dosage employing a higher dosage early in the treatment, and a lower dosage later in the treatment.
  • the single highest dose of amifostine administered to an adult human as documented in the literature was 1330 mg/m 2 . Children have been administered single doses of amifostine of up to 2700 mg/m 2 with no untoward effects.
  • the literature indicates that multiple doses (up to three times the recommended single dose of 740 to 910 mg/m 2 ) have been safely administered within a 24-hour period. Repeated administration of amifostine at two and four hours after the initial dose does not appear to result in an increase in side effects, especially nausea, vomiting, or hypotension. It appears that the most significant deleterious side effect from the administration of amifostine is hypotension.
  • contemplated amounts of the compounds of the preferred embodiment are of the compounds of the preferred embodiment.
  • methods of administration, and treatment schedules for individuals with infections caused by DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses are generally similar to those described above for the prevention of and/or treatment of radiation- or chemotherapy-induced cytotoxicity; they may also be similar to those used for the treatment of myelodysplasia syndrome.
  • Suitable side effects of amifostine include decrease in systolic blood pressure, nausea, and vomiting. If such side effects are observed for the particular thiophosphate administered, it is generally preferred to administer an antiemetic medication prior to, or in conjunction with the thiophosphate.
  • Suitable antiemetic medications include antihistamines (e.g., buclizine, cyclizine, dimenhydrinate, diphenhydramine, meclizine), anticholinergic agents (e.g. , scopolamine), dopamine antagonists (e.g. , chlorpromazine, droperidol, metoclopramide, prochlorperazine, promethazine), serotonin antagonists (e.g. , dolasetron, granisetron, ondansetron), or other agents (e.g. , dexamethasone, methylprednisolone, trimethobenzamide).
  • antihistamines e.g., buclizine,
  • WR-1065 The purity of the WR-1065 used in the experiments described below is unknown. WR-1065 is sensitive to oxidation (and possibly other reactions) and can undergo reaction to forms that appear to lack antiviral activity. Accordingly, in the tables below and in the preferred dosages provided above, the indicated concentrations represent the maximum amount of active compound that could be present; the true concentration of active compound is less than that indicated but cannot be determined definitively. In addition, for concentrations of WR-1065 of less than 100 ⁇ , estimates of the true efficacy of the compound are limited by the fact that the compound has been found to be inactivated by a variety of medium components (Grdina et al. (2000), Drug Metabol Drug Interact 16(4): 237-79.). Below approximately 50 ⁇ , this problem becomes especially severe.
  • A549 or MDCK cells were used for the plaque reduction assays.
  • Cells were grown to near confluence in Minimal Essential Medium (MEM) in six-well plates.
  • MEM Minimal Essential Medium
  • PFUs plaque forming units
  • Cell monolayers were subsequently overlaid with 0.6% agarose in MEM.
  • the overlay medium was prepared to contain a final concentration of 10 ⁇ , 33 ⁇ and 100 ⁇ amount of WR 1065. Each concentration was assayed in triplicate.
  • MDCK cells growing in 6 well plates were infected with 100 plaque forming units (PFUs) of virus in 100 ⁇ and adsorption was allowed for one hour at 37 °C.
  • Cell monolayers were subsequently overlaid with 0.6% agarose in MEM.
  • the overlay medium was prepared to contain a final concentration of 10 ⁇ , 33 ⁇ and 100 ⁇ amount of WR 1065. Each concentration was assayed in triplicate.
  • Wells overlaid with media without WR-1065 were set up as 100% infectious virus yield controls.
  • Cultures were maintained at 37°C in 5% C0 2 for 3 days. At the end of this time period, cell cultures were fixed with 1 % formaldehyde and stained with Crystal Violet for visualization of plaques and evaluation of plaque size and number. Reduction of plaque formation was expressed as a percentage of the untreated controls.
  • Table 5 includes data from an experiment to measure WR-1065 protection against cytopathic effects in MDCK cell monolayers infected with influenza A/Puerto Rico/8/34 (H1 1 ), using densitometry readings of the stained cell monolayers to estimate relative cell survival. Similar results were obtained in a second independent experiment using influenza A Puerto Rico/8/34 (H1 N1 ), and in an experiment using the same approach with influenza B/Lee/40. In a third experiment with influenza A, WR 1065 was added to an agarose overlay following a one-hour adsorption with a different virus strain, A/HKx31 (H3N2), and a plaque reduction assay was performed (Table 5). The results illustrate protection of DCK cells from influenza A by WR 1065, based upon densitometry readings (experiment 1 ) or a plaque reduction assay (experiment 2)
  • the reduced form of amifostine (WR 2721 or ethyol), was the most active moiety in the observed antiviral effects.
  • WR- 1065 was the most active moiety in the observed antiviral effects.
  • a common medium component WR 2721 or ethyol
  • Densitometry readings mean counts/mm 2 ⁇ standard deviation using Quantity One (Bio-Rad) plate reader.
  • PB Cs Peripheral blood mononuclear cells obtained from the NIH Blood Bank were cultured with phytohemagglutinin (PHA) for 48 hours to create PHA blasts.
  • PHA phytohemagglutinin
  • the cells were cultured in RPMI-1640 medium with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin and glutamine, and 10% lnterleukin-2 (IL-2).
  • FBS fetal bovine serum
  • IL-2 lnterleukin-2
  • WR-1065 from the NCI Chemical Carcinogen Repository; see Hoffman ef a/. (2001), Env. Mol. Mut. 37:1 17 was weighed (Mol. Wt. 134) and stored frozen in RPMI-1640 medium as a 10 mg/ml solution (13.4 ⁇ L ⁇ of the solution added to 1 ml of RPMI-1640 yields 1000 ⁇ solution).
  • AZT SIGMA, Mol. Wt.
  • HIV infection status was monitored in five experimental groups by measuring p24 using an ELISA kit (RETRO-TEK HIV-1 , p24 Extended Range ELISA, ZMC catalog # 0801 137).
  • the HIV infection status for the five experimental groups is provided in Table 3 (see Experiment #1 data).
  • Table 7 illustrates the antiviral efficacy of AZT and WR-1065 on PHA blasts. Subsequent experiments confirmed this antiretroviral effect, and demonstrated that the ability of WR 1065 to inhibit viral replication was dose-dependent and comparable in concentration and effect to that seen with AZT alone using this in vitro assay system.
  • WR-1065 was prepared from a 10 mg/ml solution in RPMI-1640 medium and diluted to the desired concentrations. Cell viability was measured 72 hours after exposure to WR-1065. Percentage of viable cells (compared to unexposed controls) is provided in Table 8.)
  • test results demonstrated acceptable cell viability for all concentrations tested, and particularly good cell viability for concentrations of 50 ⁇ and lower.
  • Cysteamine (chemical formula H 2 NCH 2 CH 2 SH) has been demonstrated to have anti-HIV activity (Ho et al. (1995), AIDS Res Hum Retroviruses 11 (4): 451-9; Bergamini et al. (1996), J Infect Dis 174(1 ); 214-8).
  • 200 ⁇ cysteamine effectively suppressed (-100%) HIV replication.
  • WR-1065 effectively suppressed (>99.9%) HIV replication at a concentration of less than 100 ⁇ .
  • the in vitro anti-HIV activity of WR-1065 is over 2- fold higher than that of cysteamine.
  • cysteamine's duration of action was determined to be very short; to achieve an anti-HIV effect, fresh cysteamine had to be added to the cell culture system every 12 hours.
  • WR-1065 had a long duration of action - it only had to be added to the culture system once in a 72 hour period.
  • Cystamine (chemical formula H 2 N(CH 2 ) 2 SS(CH 2 ) 2 NH 2 ), the oxidized form of cysteamine, has been demonstrated to have anti-HIV activity, DNA binding capacity, radioprotective capacity, and the ability to shift the equilibrium of DNA from the A-form towards the B-form (Allegrs et al. (2002), Amino Acids 22(2): 155-66).
  • WR-1065 the active form of WR-2721 , has also been shown to bind to DNA in the minor groove, and also to shift the B/A-DNA equilibrium towards the B-form.
  • cystamine and its reduced form cysteamine are capable of binding to other nucleic acids in addition to DNA, and have some ability to bind to and/or to interact with proteins. It should be noted that the anti-HIV activity of cystamine is considered to be due, at least in large part, to its rapid in vivo conversion to cysteamine.
  • WR-1065's antiviral activity is due in part to its ability to bind to and to block critical sites on nucleic acids and/or proteins.
  • Blockage of nucleic acid sites and/or proteins is considered to be a possible mechanism for the aminothiols' modulation of enzyme function (Brekken et al. (1986), J Biol Chem 273(41 ): 26317-22).
  • the mechanism by which an aminothiol could bind to and/or block nucleic acids and/or proteins without inducing significant cytotoxicity to eukaryotic cells is unknown at this time.
  • one or more of the compounds of the present invention function as an antiviral agent because its structure renders it bi-functional, combining two distinct properties.
  • the -(CH 2 ) 3 NH 2 portion of WR-1065 is believed to be responsible for binding to nucleic acids (DNA, RNA), and possibly also to some proteins. .
  • a second portion of WR-1065 -NH(CH 2 ) 2 SH is believed to be largely responsible for the antiretroviral/antiviral effects that have been observedTo be effective against some viruses, it is believed that the sulfhydryl group needs to be in the reduced state in order for antiretroviral/antiviral effects to be observed for the compound; however, it is possible that the sulfhydryl group may remain functional if oxidized to the disulfide (as in WR-33278). Both of these components contribute to maximizing antiretroviral/antiviral effects of the compounds of preferred embodiments (e.g.
  • both the -(CH 2 ) 3 NH 2 and the -NH(CH 2 ) 2 SH moieties of amifostine).
  • the first portion of the molecule functions to align the molecule in close proximity to critical binding sites on nucleic acids and/or proteins, and the second portion of the molecule functions in a reaction that contributes to antiviral efficacy.
  • Variations to the chemical structure of the moiety -(CH 2 ) 3 NH 2 may be tolerated without losing or altering the antiretroviral/antiviral efficacy of the compound, so long as the resulting structure retains a binding capacity/affinity that is similar to that observed with -(CH 2 ) 3 NH 2 itself.
  • Such variations may include more or less than three carbon atoms in the alkyl group, and a branched alkyl chain, lower alkyl substituents on the amino group.
  • Gutschow et al. describe two compounds that are demonstrated to display significant
  • the significantly higher ratio of proteins to nucleic acids of viruses would allow for enhanced interaction between proteins associated with nucleic acids, the nucleic acids themselves, and a molecule with nucleic acid and protein binding affinity.
  • the enhanced opportunity for binding between WR- 1065 and viral nucleic acids and proteins could result in deleterious effects upon viral nucleic acid production, maintenance, or functionality. A similar effect would not be expected to be exerted at the same magnitude upon the nucleic acids of higher organisms.
  • WR-1065 and similar compounds could block replication initiation through interactions with a defined replicator sequence on viral nucleic acids, while not exerting a similar effect upon the nucleic acids of higher organisms because of the degenerate nature of their initiation-site selection. Binding of WR-1065 to replication initiation sites of eukaryotic cells would be hypothesized to slow down nucleic acid processing and, hence, cell division, but not block it completely. This hypothesis is supported by the fact that WR-1065 has been shown to delay the progression of cells through the cell cycle, but the exact mechanism remains unclear.
  • replication origin recognition complexes of higher animals share many similarities with analogous complexes in simple life forms, including viruses and archaeal cells (Gai et al., 2004). However, critical differences have been noted in the components and in the mode of action of these molecular machines (Gai et al., 2004),
  • the molecular machine known as the origin recognition complex consists of the oncogenic large tumor antigen that is formed as a monomer; six monomers then form a hexamer, and two hexamers assemble together, along with replication protein A, topoisomerase I, and polymerase-alpha/primase. These molecules form the replication initiation complex for SV-40.
  • This complex is highly dependent upon the conformation of individual monomers, as well as the conformation of the hexamer (Gai et al. 2004).
  • the functionality of the entire replication initiation complex is also dependent upon the conformation of the target nucleic acids and the target initiation site (Gai et al., 2004). Because WR-1065 is known to have the capacity to alter the conformation of both nucleic acids and proteins, a complex such as that described for SV-40 would be especially vulnerable to induced
  • origin recognition complexes of eukaryotic cells do not appear to require an origin of replication site, but rather are capable of using a variety of sites, it is reasonable to hypothesize that these latter origin recognition complexes could potentially tolerate a degree of conformational change that SV-40 and other virally-associated origin recognition complexes cannot tolerate.
  • Thymosin alpha I (TA1) Synthetic peptide Increases T cell function Viramidine (ICN3142) Amidine prodrug of
  • HCV / MF59 Enhances immune response
  • Rhinovirus Rupintrivir AG7088 Pfizer Inc: Protease inhibitor
  • Picornavirus WIN51711 Bind to viral capsid to prevent / Poliovirus uncoating
  • the subject matter described herein includes a potential broad-spectrum class of antiviral drugs that can be used for treatment of DNA viral, RNA viral, and retroviral infections.
  • this class include Amifostine, phosphonol and functional derivatives thereof.
  • these organic thiophosphate reducing agents inhibit viral growth in influenza and adenoviral-infected human alveolar epithelial cell lines and/or kidney cells, and in lymphocytes infected with HIV-1 , without destroying the cells.
  • a range of effective doses and methods for administration, and pharmaceutical compositions, is provided.
  • the class of antiviral drugs described has application for prevention and/or treatment of acute and/or chronic viral infections such as HIV, adenovirus, Influenza, and pathogenic Avian Influenza strains.
  • Ethyol® by Medimmune for application as a selective cytoprotective agent, including, but not limited to, treatment for moderate to severe xerostomia (dry mouth) in patients undergoing post-operative radiation treatment for head and neck cancer
  • Amifostine selectively protects normal tissues but not tumors against the cytotoxicity of ionizing radiations and DNA-binding chemotherapeutic agents. It is a prodrug that is dephosphotylated by alkaline phosphatase to the active metabolite, WR1065.

Landscapes

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

Abstract

La présente invention concerne un procédé de prévention ou de traitement d'une infection par le virus de la grippe ou d'une infection par un adénovirus par administration d'une quantité efficace d'un composé de formule (I) ou de formule (II), ou d'un composé similaire à un individu le nécessitant.
PCT/US2010/052255 2009-10-12 2010-10-12 Antiviral à large spectre et procédés d'utilisation WO2011046901A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25071509P 2009-10-12 2009-10-12
US61/250,715 2009-10-12

Publications (2)

Publication Number Publication Date
WO2011046901A2 true WO2011046901A2 (fr) 2011-04-21
WO2011046901A3 WO2011046901A3 (fr) 2011-08-11

Family

ID=43876820

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/052255 WO2011046901A2 (fr) 2009-10-12 2010-10-12 Antiviral à large spectre et procédés d'utilisation

Country Status (1)

Country Link
WO (1) WO2011046901A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019500407A (ja) * 2015-11-17 2019-01-10 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX9205549A (es) * 1991-09-30 1993-05-01 Jess G Thoene Metodo para el tratamiento de vih.
WO2006136516A2 (fr) * 2005-06-23 2006-12-28 Ciba Specialty Chemicals Holding Inc. Colorants de sulfure
AU2007335962B2 (en) * 2006-12-20 2012-09-06 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Antiviral indoles

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019500407A (ja) * 2015-11-17 2019-01-10 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法
JP2021130674A (ja) * 2015-11-17 2021-09-09 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法
JP7121425B2 (ja) 2015-11-17 2022-08-18 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法
JP2022160543A (ja) * 2015-11-17 2022-10-19 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法
US11554174B2 (en) 2015-11-17 2023-01-17 The Burlington Hc Research Group, Inc. Methods for improved protection and delivery of aminothiols and analogs thereof
JP7323960B2 (ja) 2015-11-17 2023-08-09 ザ バーリントン エイチシー リサーチ グループ インコーポレイテッド アミノチオール及びその類似体の改善された保護及び送達のための方法

Also Published As

Publication number Publication date
WO2011046901A3 (fr) 2011-08-11

Similar Documents

Publication Publication Date Title
US9849143B2 (en) Broad spectrum antiviral and methods of use
KR0180019B1 (ko) 개량된 화학적 화합물
IL193148A (en) Medicine combined with hepatitis B virus
AU2009223064B2 (en) Use of 4'-Thio-2'-Deoxynucleosides as Anti Orthopoxvirus Agents
EP3426671A1 (fr) Composés et compositions pour le traitement d'infections
US20160374967A1 (en) Organic thiophosphate antiretroviral agents
JP2004538319A (ja) 抗−hiv治療のためのr−nsaid化合物の用途
AU775459B2 (en) Gallium complexes of 3-hydroxy-4-pyrones to treat infection by intracellular prokaryotes, DNA viruses and retroviruses
KR100413312B1 (ko) 항바이러스 복합약제
WO2011046901A2 (fr) Antiviral à large spectre et procédés d'utilisation
Morris-Natschke et al. Phospholipid analogs against HIV-1 infection and disease
RU2726210C2 (ru) Комбинация противовирусных средств, набор и способ лечения на ее основе
JP2014532636A (ja) ウイルス感染の処置のためのヌクレオシドアナログおよび該処置に対する感受性を評価するための方法
US20020173491A1 (en) DAPD combination therapy with inosine monophosphate dehydrogenase inhibitor
US20230078120A1 (en) Methods for Treating Coronavirus Infections
WO2015044491A1 (fr) Utilisation d'un composé indolique avec des substitutions nucléophiles en c3 et ses variants dimères dans la préparation d'une composition pharmaceutique utile pour le traitement de néoplasies lymphoïdes b
EP4149454A1 (fr) Utilisation de composés d'aminoacétonitrile pour le traitement d'une infection et d'une maladie
EA045000B1 (ru) Комбинация противовирусных средств, набор и способ лечения на ее основе

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: 10823923

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase in:

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 16.07.2012)

122 Ep: pct application non-entry in european phase

Ref document number: 10823923

Country of ref document: EP

Kind code of ref document: A2