WO2006130217A2 - Substituted phosphate esters of nucleoside phosphonates - Google Patents

Substituted phosphate esters of nucleoside phosphonates Download PDF

Info

Publication number
WO2006130217A2
WO2006130217A2 PCT/US2006/012121 US2006012121W WO2006130217A2 WO 2006130217 A2 WO2006130217 A2 WO 2006130217A2 US 2006012121 W US2006012121 W US 2006012121W WO 2006130217 A2 WO2006130217 A2 WO 2006130217A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
phospho
alkyl
alkenyl
hpmpa
Prior art date
Application number
PCT/US2006/012121
Other languages
French (fr)
Other versions
WO2006130217A3 (en
Inventor
Karl Y. Hostetler
James R. Beadle
Jacqueline C. Ruiz
Original Assignee
The Regents Of The University Of California
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 Regents Of The University Of California filed Critical The Regents Of The University Of California
Priority to US11/887,501 priority Critical patent/US20090156545A1/en
Publication of WO2006130217A2 publication Critical patent/WO2006130217A2/en
Publication of WO2006130217A3 publication Critical patent/WO2006130217A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/6512Six-membered rings having the nitrogen atoms in positions 1 and 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids

Definitions

  • FIELD Provided herein are antiviral and anticancer phosphonate drugs, their preparation, and their use for treatment of viral infections and cancers. Also provided are methods for synthesizing anhydrides containing alkyl phosphate or alkoxyalkyl phosphate coupled to nucleoside phosphonate drugs. The new conjugates have greater antiviral and/or antiproliferative activity when compared with the parent nucleoside phosphonate.
  • BACKGROUND Phosphonate nucleosides are well known in the art and are in clinical use as antiviral and anticancer agents ⁇ see, Holy, A., Phosphonomethoxyalkyl analogs of nucleotides, Current Pharmaceutical Design 9(31), 2567-92, 2003). Their limitations relate to poor oral bioavailability, poor target cell uptake and toxicity in kidneys. In general, nucleoside phosphonate uptake into target cells is poor because of the dual negative charges on the phosphonate moiety.
  • nucleoside phosphonate diphosphate Once in the cell, they require two subsequent anabolic phosphorylations to achieve activity as the nucleoside phosphonate diphosphate. Some nucleoside phosphonates are hampered by slow phosphorylation. There is a continuing need for less toxic, more effective pharmaceutical agents to treat a variety of disorders associated with viral infection, and cell proliferation.
  • nucleoside phosphonates linked via their phosphonate residue to the phosphate of alkoxyalkyl-phosphate, alkylglycerol-phosphate or alkyl- phosphate and pharmaceutically acceptable derivatives thereof are provided herein.
  • the nucleoside phosphonates or acyclic nucleoside phosphonates linked to the phosphate of alkoxyalkyl-phosphate, alkylglycerol-phosphate or alkyl-phosphate result in orally available compounds which exhibit greater antiviral or anticancer activity by promoting cell uptake and favorable cellular metabolism which yields the nucleoside phosphonate monophosphate, bypassing the need for the first of two anabolic phosphorylations.
  • compounds provided herein exhibit greater antiviral or anticancer activity than the unmodified nucleoside phosphonates.
  • compositions and methods of using the compounds and compositions for the treatment of various diseases are provided.
  • compounds and compositions provided herein have antiviral activity.
  • compounds and compositions that are useful in the treatment, prevention, or amelioration of one or more symptoms associated with cell proliferation are provided.
  • the compounds for use in the compositions and methods provided herein have formula I:
  • R L is a lipophilic group
  • Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound, coupled to the phosphate group by an anhydride linkage and y is 1 or 2.
  • pharmaceutically-acceptable derivatives including salts, esters, enol ethers, enol esters, solvates, hydrates and prodrugs of the compounds described herein.
  • pharmaceutical compositions containing the compounds provided herein and a pharmaceutically acceptable carrier are formulated for single dosage administration. Methods of treatment, prevention or amelioration using the compounds and compositions provided herein are provided. Such methods encompass treating, preventing or ameliorating one or more symptoms of diseases associated with viral infections and cell proliferation. In practicing the methods, effective amounts of the compounds or compositions containing therapeutically effective concentrations of the compounds are administered.
  • Articles of manufacture are provided containing packaging material, a compound or composition provided herein which is useful for treating, preventing, or ameliorating one or more symptoms of diseases or disorders associated with viral infections or cell proliferation using the compounds and compositions provided herein, and a label that indicates that the compound or composition is useful for treating, preventing, or ameliorating one or more symptoms of diseases or disorders associated with viral infections or cell proliferation.
  • DETAILED DESCRIPTION A Definitions
  • phosphonate and phosphonate group mean a functional group or moiety within a molecule that comprises at least one phosphorus- carbon bond, and at least one phosphorus-oxygen double bond.
  • the phosphorus atom is further substituted with oxygen, sulfur, and nitrogen substituents. These substituents may be part of a prodrug moiety.
  • phosphonate and “phosphonate group” include molecules with phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, phosphondiamidate, and phosphonthioate functional groups.
  • nucleoside refers to a molecule composed of a heterocyclic base and a carbohydrate.
  • a nucleoside is composed of a heterocyclic nitrogenous base in N-glycosidic linkage with a sugar.
  • Nucleosides are recognized in the art to include natural bases (standard), and non-natural bases well known in the art.
  • the carbohydrates include the true sugars found in natural nucleosides or a species replacing the ribofuranosyl moiety or acyclic sugars.
  • the heterocyclic nitrogenous bases are generally located at the 1' position of a nucleoside sugar moiety. Nucleosides generally contain a base and sugar group.
  • the nucleosides can be unmodified or modified at the sugar, and/or base moiety, (also referred to interchangeably as nucleoside analogs, modified nucleosides, non-natural nucleosides, non-standard nucleosides; see for example, Eckstein et al, International PCT Publication No. WO 92/07065 and Usman et al, International PCT Publication No. WO 93/15187).
  • the heterocyclic base is thymine, uracil, cytosine, adenine or guanine.
  • acyclic sugars contain 3-6 carbon atoms and include, for example, the acyclic sugar moieties present in acyclovir (-CH 2 -O-CH 2 -CH 2 -OH), ganciclovir (-CH 2 -O-CH(CH 2 OH)-CH 2 -OH), and the like.
  • Natural nucleosides have the ⁇ -D-configuration. The term “nucleoside” shall be understood to encompass unnatural configurations and species replacing the true sugar that lack an anomeric carbon. In natural nucleosides the heterocyclic base is attached to the carbohydrate through a carbon-nitrogen bond. The term “nucleoside” shall be understood to encompass species wherein the heterocyclic base and carbohydrate are attached through a carbon-carbon bond (C -nucleosides).
  • nucleoside contains 1 or more functional groups that may be reactive to form undesired products under the reaction conditions of the present process
  • functional groups may be blocked using the protecting groups commonly employed in nucleoside chemistry.
  • the amino group of adenine and cytosine may be protected by benzoyl; the 6-oxo and 2-amino groups of guanine may be protected by the triphenylmethyl (trityl) group.
  • Trityl triphenylmethyl
  • nucleoside base refers to natural and non-natural purine and pyrimidine bases, including adenine, thymine, cytosine, guanine and uracil and analogs thereof.
  • nucleoside phosphonate and acyclic nucleoside phosphonate refer to the group of phosphonomethoxyalkyl or phosphono substituted nucleoside derivatives that are biologically active, for example, as anti-viral, anti-cancer or antiparasitic drugs.
  • lipophilic refers to the cyclic, branched or straight chain chemical groups that when covalently linked to a phosphonic acid to form a phosphonate monoester increase oral bioavailability and enhance activity of the nucleoside phosphonates as compared with the parent nucleoside phosphonates.
  • lipophilic groups include, but are not limited to alkyl, alkoxyalkyl, and alkylglyceryl.
  • lipophilic monoesters of nucleoside phosphonates refers to compound where a lipophilic group is covalently attached to a nucleoside phosphonate via an ester linkage.
  • pharmaceutically acceptable derivatives of a compound include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives may be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced may be administered to animals or humans without substantial toxic effects and either are pharmaceutically active or are prodrugs.
  • salts include, but are not limited to, amine salts, such as but not limited to N,N'-dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzylphenethylamine, 1 -para-chlorobenzyl-2-pyrrolidin- 1 '-ylmethyl- benzimidazole, diethylamine and other alkylamines, piperazine and tris(hydroxymethyl)aminomethane; alkali metal salts, such as but not limited to lithium, potassium and sodium; alkali earth metal salts, such as but not limited to barium, calcium and magnesium; transition metal salts, such as but not limited to zinc; and other metal salts, such as but not limited to sodium hydrogen phosphate and disodium phosphate; and also including, but not limited to, nitrates, borates, me
  • esters include, but are not limited to, alkyl, alkenyl, alkynyl, and cycloalkyl esters of acidic groups, including, but not limited to, carboxylic acids, phosphoric acids, phosphinic acids, sulfonic acids, sulfinic acids and boronic acids.
  • Pharmaceutically acceptable solvates and hydrates are complexes of a compound with one or more solvent or water molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4, solvent or water molecules.
  • treatment means any manner in which one or more of the symptoms of a disease or disorder are ameliorated or otherwise beneficially altered.
  • amelioration of the symptoms of a particular disorder by administration of a particular compound or pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.
  • ECs 0 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
  • a prodrug is a compound that, upon in vivo administration, is metabolized by one or more steps or processes or otherwise converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • the pharmaceutically active compound is modified such that the active compound will be regenerated by metabolic processes.
  • the prodrug may be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • prodrugs of the compound can design prodrugs of the compound (see, e.g., Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392). Other prodrugs for use herein are described elsewhere herein.
  • the compounds provided herein may contain chiral centers. Such chiral centers may be of either the (R) or (S) configuration, or may be a mixture thereof.
  • the compounds provided herein may be enantiomerically pure, or be stereoisomeric or diastereomeric mixtures.
  • the compounds provided herein encompass any racemic, optically active, polymorphic, or steroisomeric form, or mixtures thereof, of a compound provided herein, which possesses the useful properties described herein, it being well known in the art how to prepare optically active forms and how to determine antiproliferative activity using the standard tests described herein, or using other similar tests which are well known in the art.
  • Examples of methods that can be used to obtain optical isomers of the compounds provided herein include the following: i) physical separation of crystals- a technique whereby macroscopic crystals of the individual enantiomers are manually separated.
  • This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; ii) simultaneous crystallization- a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state; iii) enzymatic resolutions — a technique whereby partial or complete separation of a racemate by viitue of differing rates of reaction for the enantiomers with an enzyme iv) enzymatic asymmetric synthesis — a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enatiomerically pure or enriched synthetic precursor of the desired enantiomer; v) chemical asymmetric synthesis — a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce assymetry (i.e., chirality)
  • first- and second-order asymmetric transformations a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiorner perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer.
  • kinetic resolutions this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors — a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; x) chiral liquid chromatography — a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase.
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
  • chiral gas chromatography a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;
  • extraction with chiral solvents a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent;
  • xiii) transport across chiral membranes a technique whereby a racemate is placed in contact with a thin membrane barrier.
  • the barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the. non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
  • substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), gel electrophoresis, high performance liquid chromatography (HPLC) and mass spectrometry (MS), used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance.
  • TLC thin layer chromatography
  • HPLC high performance liquid chromatography
  • MS mass spectrometry
  • alkyl refers to a monovalent straight or branched chain or cyclic radical.
  • the alkyl group contains from one to twenty-four carbon atoms, including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl, octadecyl, nonadecyl, eicosyl, 18-methyl-nonadecyl, 19- methyl-eicosyl, and the like.
  • lower alkyl refers to alkyl groups of 1 to 6 carbon atoms.
  • substituted alkyl refers to alkyl groups further bearing one or more substituents, including, but not limited to substituents selected from lower alkyl, hydroxy, alkoxy (of a lower alkyl group), mercapto (of a lower alkyl group), cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryloxy, substituted aryloxy, halogen, trifluoromethyl, cyano,azido, nitro, nitrone, amino, amido, -C(O)H, acyl, oxyacyl, carboxyl, carbamate, sulfonyl, sulfonamide, and sulfuryl, which may be protected or unprotected as necessary, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Ed
  • alkenyl refers to straight or branched chain hydrocarbon group having one or more carbon-carbon double bonds. In certain embodiments, the alkenyl group contains from 2 up to 24 carbon atoms, and "substituted alkenyl” refers to alkenyl groups further bearing one or more substituents as set forth above.
  • alkynyl refers to straight or branched chain hydrocarbon group having one or more carbon-carbon triple bonds. In certain embodiments, the alkynyl group contains from 2 up to 24 carbon atoms, and "substituted alkynyl” refers to alkynyl groups further bearing one or more substituents as set forth above.
  • aryl refers to aromatic groups having in the range of 6 up to 14 carbon atoms and “substituted aryl” refers to aryl groups further bearing one or more substituents as set forth above.
  • heteroaryl refers to aromatic groups containing one or more heteroatoms (e.g., N, O, S, or the like) as part of the ring structure, and having in the range of 3 up to 14 carbon atoms and "substituted heteroaryl” refers to heteroaryl groups further bearing one or more substituents as set forth above.
  • heteroatoms e.g., N, O, S, or the like
  • subject is an animal, such as a mammal, including human, such as a patient.
  • effective amount means an amount required for prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders associated including those associated with viral infection, cell proliferation and/or bone metabolism .
  • haloalkyl may include one or more of the same or different halogens.
  • parenteral includes subcutaneous, intravenous, intraarterial, intramuscular or intravitreal injection, or infusion techniques.
  • topically encompasses administration rectally and by inhalation spray, as well as the more common routes of the skin and mucous membranes of the mouth and nose and in toothpaste.
  • Oleyloxyethyl OLE
  • Oleyloxypropyl OLP
  • Phosphonomethoxyethyl-adenine PMEA
  • Phosphonomethoxy-propyladenine PMPA (tenofovir)
  • Hexadecyloxypropyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine HDP-phospho-(S)-HPMPA
  • Oleyloxyethyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine OLE-phospho-(S)-HPMPA
  • Oleyloxypropyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine OLP- phospho-(S)-HPMPA
  • 5-Phosphono-pent-2-en-l-yl cytosine PPen-C
  • 5-Phosphono-pent-2-en-l-yl guanine PPen-G
  • the compound for use in the compositions and methods provided herein has formula II:
  • R 1 and R lx are each independently -H, optionally substituted -0(C 1 - C 24 )alkyl, -O(C 1 -C 24 )alkenyl, -O(C 1 -C 24 )acyl, -S(C r C 24 )alkyl, -S(C 1 -C 24 )alkenyl, or -S(C 1 -C 24 )acyl, wherein at least one of R 1 and R lx is not -H, and wherein the alkenyl or acyl optionally have 1 to about 6 double bonds;
  • R 2 and R 2x are each independently -H, optionally substituted -0(C 1 - C 7 )alkyl, -O(C 1 -C 7 )alkenyl, S(C 1 -C 7 )alkyl, -S(C 1 -C 7 )alkenyl, -O(C 1 -C 7 )acyl, -S(C 1 - C 7 )HCyI, -N((C 1 -C 7 )alkyl) 2 , halogen, -NH 2 , -OH, or -SH;
  • Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound, coupled to the phosphate group by an anhydride linkage;
  • L is a valence bond or a bifunctional linking molecule of the formula -J-(CR x R y ) t -G-, wherein t is an integer from 1 to 24; J and G are independently -O-, -S-, -C(O)O- or - NH-; R x and R y are each independently -H, substituted or unsubstituted alkyl, or alkenyl; m is an integer from 0 to 6; y is 1 or 2; and n is 0 or 1.
  • the compounds for use in the compositions and methods provided herein have formula III:
  • Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound and R q has formula:
  • R p is a pharmacologically active nucleoside or an analog thereof.
  • R 1 and R lx are each independently -H, or optionally substituted -0(C 1 - C 24 )alkyl; wherein at least one of R 1 and R lx is not -H; R 2 and R 2x are each independently -H, or optionally substituted -0(C 1 - C 7 )alkyl;
  • Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound of formula:
  • Rp is a pharmacologically active nucleoside or analog thereof
  • L is a valence bond or a bifunctional linking molecule of the formula — J- (CR x R y )t-G-, wherein t is an integer from 1 to 24, J and G are each independently -O-, and R x and R y are each independently -H, substituted or unsubstituted alkyl, or alkenyl; n is 0 or 1 ; and n 1 is O to 3.
  • R p is
  • R 3 , R 4 and R 5 are each independently H, hydroxy, halo, azido, C 1-6 alkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • B is a purine or pyrimidine base or an analog thereof
  • R 3x is H, azido, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl or substituted or unsubstituted C 2-6 alkynyl;
  • R 4x is H, C 1-6 substituted or unsubstituted alkyl, C 2-6 substituted or unsubstituted alkenyl or C 2-6 substituted or unsubstituted alkynyl; and R 3z is H, C 1-6 alkyl, hydroxylC 1-6 alkyl, haloC 1-6 alkyl, azidoC 1 . 6 alkyl or OH.
  • R p has formula:
  • R 3 is H, azido, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl or substituted or unsubstituted C 2-6 alkynyl. In certain embodiments, R 3 is H, azido, substituted or unsubstituted C 1-6 alkyl. In certain embodiments, R is H or azido. In certain embodiments, R is azido. In certain embodiments, R 3 is H. In certain embodiments, R 4 and R 5 are each independently selected from hydrogen, halo and hydroxyalkyl. In certain embodiments, R 4 and R 5 are each independently selected from halo and hydroxyalkyl. In certain embodiments, R 4 and R 5 are each independently selected from fluoro and hydroxymethyl. In certain embodiments, R is selected from fluoro and hydroxymethyl. In certain embodiments, R 5 is selected from fluoro and hydroxymethyl.
  • R p has formula:
  • R 3x is H, azido, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl or substituted or unsubstituted C 2-6 alkynyl;
  • R 4x is H, C 1-6 substituted or unsubstituted alkyl, C 2-6 substituted or unsubstituted alkenyl or C 2-6 substituted or unsubstituted alkynyl and other variables are as defined elsewhere herein.
  • R 3x is H, azido or substituted or unsubstituted C 1-6 alkyl.
  • R 4x is H, C 1-6 substituted or unsubstituted alkyl, C 2-6 substituted or unsubstituted alkenyl or C 2-6 substituted or unsubstituted alkynyl. In certain embodiments, R 4x is H, or C 1-6 alkyl. In certain embodiments, R x is H, or methyl.
  • R p has formula:
  • R 3z is H, C 1-6 alkyl, hydroxyl C 1-6 alkyl, halo C 1-6 alkyl, azido C 1-6 alkyl or OH and the other variables are as defined elsewhere herein.
  • R 3z is hydrogen, C 1-6 alkyl or hydroxyC] -6 alkyl.
  • R 3z is hydrogen or hydroxymethyl.
  • R 3z is hydrogen.
  • R 3z is hydroxymethyl.
  • the OH group is protected, for example as an ester or an ether.
  • R 3z may be in S or R configuration.
  • R p has formula:
  • R 3y is H, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl or substituted or unsubstituted C 2-6 alkynyl, or OH and the other variables are as defined elsewhere herein.
  • R 3y is hydrogen, C 1-6 alkyl or hydroxyl C 1-6 alkyl.
  • R 3y is hydrogen or hydroxymethyl.
  • R 3y may be in S or R configuration.
  • R p has formula:
  • R p has formula:
  • R L has formula:
  • R 1 and R lx are as defined elsewhere herein.
  • R L has formula:
  • R 1 and R lx are as defined elsewhere herein.
  • R L has formula:
  • R 1 and R lx are as defined elsewhere herein.
  • RL is hexadecyloxypropyl, octadecyloxypropyl, oleyloxyethyl, oleyloxypropyl, octadecyloxyethyl, 15-methylhexadecyloxypropyl or 17-methyloctadecyloxyethy 1.
  • R 1 is an alkoxy group having the formula -O-(CH 2 ) t - CH 3 wherein t is 0-24.
  • t is 8, 10, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
  • t is 13, 14, 15, 16, 17, 18, 19 or 20.
  • t is 15, 16, 17, 18, 19 or 20.
  • t is 17, 18, 19 or 20.
  • t is 15 or 17.
  • RL is a substituted or unsubstituted C 8 -C 24 alkyl, substituted or unsubstituted C 8 -C 24 alkenyl having from 1 to 6 double bonds or substituted or unsubstituted C 8 -C 24 alkynyl having from 1 to 6 triple bonds, wherein substituents when present are selected from one or more, in some embodiments, 1 to 4 or 1 or 2 substituents selected from halogen, alkyl, -OR W , -SR W , cycloalkyl or epoxide, where R w is hydrogen or alkyl and where the alkyl, alkenyl, alkynyl groups may be further substituted or unsubstituted.
  • R L is an alkyl, alkenyl or alkynyl group and contains 8, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms and can be a straight or branched chain moiety.
  • R L is a C 16 -C 23 straight or branched chain alkyl or C 16 -C 23 straight or branched chain alkenyl.
  • R L is a C 17 -C 19 straight or branched chain alkyl or C 17 -C 19 straight or branched chain alkenyl.
  • R L is C 17 -alkyl, C 18 -alkyl or C 19 alkyl.
  • R L is C 17 -alkenyl, C 18 -alkenyl or C 19 alkenyl. In other embodiments, R L is C 17 -C 22 alkyl. In other embodiments, R L is C 17 alkyl, C 18 alkyl, C 19 alkyl, C 20 alkyl, C 21 alkyl, or C 22 alkyl.
  • R L is substituted with one or more groups selected from lower alkyl and halo. In certain embodiments, R L is substituted with one or more methyl groups. In certain embodiments, R L is substituted with one or more fluoro groups. In certain embodiments, R L is C 16 -C 23 alkyl and is substituted with one or more methyl or fluoro groups. In certain embodiments, the methyl group or the fluoro group substituent is present on the penultimate carbon of the alkyl, alkenyl, or alkynyl chain.
  • R L is 7-methyl-octyl, 8-methyl-nonyl, 9-methyl-decyl, 10- methyl-undecyl, 11-methyl-dodecyl, 12-methyl-tridecyl, 13-methyl-tetradecyl, 14- methyl-pentadecyl, 15-methyl-hexadecyl, 16-methyl-heptadecyl, 17-methyl-octadecyl, 18-methyl-nonadecyl, 19-methyl-eicosyl, 20-methyl-heneicosyl, 21-methyl-docosyl, 22-methyl-tricosyl, 7-fluoro-octyl, 8- fluoro-nonyl, 9- fluoro-decyl, 10- fluoro-undecyl, 11- fluoro-dodecyl, 12- fluoro-tridecyl, 13-fluoro-tetradecy
  • B is selected from a natural or non natural purine or pyrimidine base.
  • B is wherein R 3a is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, hydroxy, halo, aryl or heteroaryl;
  • R 6 is H or C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or cycloalkyl;
  • R 7 is H, hydroxy, halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl or NR 4 R 5 ;
  • R 8 is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or cycloalkyl;
  • R 9 is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, halo Or NR 4 R 5 , where R 4 and R 5 are each independently H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, or C 3-6 cycloalkyl.
  • R 3a is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, hydroxy, halo, aryl or heteroaryl. In other embodiments, R 3a is H, halo or C 1-6 alkyl. In other embodiments, R 3a is H. In other embodiments, R 3a is methyl. In other embodiments, R 3a is fluoro.
  • R 4 and R 5 are each independently H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, or C 3-6 cycloalkyl. In other embodiments, R 4 is H, C 1-6 alkyl or C 3-6 cycloalkyl. In other embodiments, R 4 is H, methyl or cyclopropyl. In other embodiments, R 5 is H, C 1-6 alkyl or C 3-6 cycloalkyl. In other embodiments, R 5 is H, methyl or cyclopropyl.
  • R 6 is H or C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or cycloalkyl. In other embodiments, R 6 is H or C 1-6 alkyl. In other embodiments, R 6 is H or methyl. In other embodiments, R 6 is H. In other embodiments, R 6 is methyl. In other embodiments, R 7 is H, hydroxy, halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl or NR 4 R 5 . In other embodiments, R 7 is H, C 1-6 alkyl, or NR 4 R 5 . In other embodiments, R 7 is methyl. In other embodiments, R 7 is NR R 5 . In other embodiments, R 7 is NH 2 .
  • R 8 is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or cycloalkyl. In other embodiments, R 8 is H.
  • R 9 is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, halo or NR 4 R 5 . In other embodiments, R 9 is H.
  • B is selected from pyrimidin-1-yl, pyrimidin-3-yl, purin- 3-yl, purin-7-yl and purin-9-yl residue.
  • B is thymin-1-yl, cytosine-1-yl, adenine-9-yl or guanine-9-yl.
  • B is selected from:
  • the alkyl, alkenyl and alkynyl groups in the compounds provided herein are substituted with one or more, in one embodiment, one, two, three or four substituents selected from alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • substituents selected from alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • the compounds herein are phosphate esters of antiviral and anticancer nucleoside phosphonates.
  • the compounds provided herein are analogs of (S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]cytosine (HPMPC, cidofovir), (S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine ((S)-HPMPA), phosphonomethoxyethyl-guanine (PMEG), phosphonomethoxyethyl-adenine (PMEA) and phosphonomethoxy-propyladenine (PMPA, tenofovir).
  • Nucleoside analogs with antiviral activity against hepatitis C may be converted to their 5 '-phosphonates or 5 '-methylene phosphonates for use in the compounds provided herein.
  • Some exemplary nucleosides include: 2'-C-methyl adenosine; 2'-C- methyl guanosine; 7-deaza-2'-C-methyl adenosine, 2'-C-methyl cytosine.
  • Other nucleosides which can be used in the compounds provided herein, after conversion fo their 5 '-phosphonates or 5'-methylene-phosphonates are described in the following patents, which are hereby incorporated by reference in their entirety.
  • Nucleoside analogs with antiviral activity against hepatitis B may be converted to their 5'-phosphonates or 5 '-methylene phosphonates for use herein.
  • Exemplary nucleosides include 3TC, FTC, DAPD, L-FMAU, entecavir, telbivudine and various ⁇ - L-2'-deoxycytidine, ⁇ -L-2'-deoxyadenine and ⁇ -L-2'-deoxythymidine analogs described by Bryant et ah, Anti-viral L-nucleosides specific for Hepatitis B infection, Antimicrob. Agents Chemother., 45:229-235, 2001.
  • the nucleosides for use herein include, but are not limited to tenofovir, adefovir, and the 5- phosphono-pent-2-en-l-yl nucleosides, such as 5-phosphono-pent-2-en-l-yl adenine (PPen-A), 5-phosphono-pent-2-en-l-yl cytosine (PPen-C), 5-phosphono-pent-2-en-l-yl guanine (PPen-G), 5-phosphono-pent-2-en-l-yl thymine (PPen-T) and 5-phosphono- pent-2-en-l-yl uracil (PPen-U) and others disclosed in U.S.
  • PPen-A 5-phosphono-pent-2-en-l-yl adenine
  • Pen-C 5-phosphono-pent-2-en-l-yl cytosine
  • PPen-G 5-phosphono-p
  • the compounds have anti-hepatitis B activity.
  • Certain other Nucleoside 5'- monophosphates for use herein are described by Prakash et a in J. Med. Chem. 2005, 48, 1199-1210.
  • the compounds provided herein are selected from hexadecyloxypropyl-phospho-(S)-HPMPA (HDP-phospho-(S)-HPMPA), octadecyloxyethyl-phos ⁇ ho-(S)-HPMPA (ODE-phospho-(S)-HPMPA), oleyloxyethyl- phospho-(S)-HPMPA (OLE-phospho-(S)-HPMPA), oleyloxypropyl-phospho-(S)- HPMPA (OLP- phospho-(S)-HPMPA), 15-methyl-hexadecyloxy-propyl-phospho-(S)- HPMPA, and 17-methyl-octadecyloxy-ethyl- phospho-(S)-HPMPA.
  • HDP-phospho-(S)-HPMPA octadecyloxyethyl-phos ⁇ ho-(S)-HPMPA
  • the compound is selected from HDP-phospho-(S)-HPMPA
  • HDP-phospho-Phosphonomethoxy-1 -methyl cytidine HDP-phospho-PM-2 -O-methyl cytidine
  • Schemes 1 and 2 Exemplary methods for the preparation of nucleoside phosphonate- phosphate ester conjugates provided herein are depicted in Schemes 1 and 2.
  • Scheme 1 outlines the synthesis of alkoxyalkyl phospho-morpholidates 3 and 4. Detailed synthesis is described in detail in example 1.
  • (S)-l-(3-hydroxy-2-phosphonomethoxypropyl)cytosine (HPMPC) is treated with dimethoxytritylchloride in DMSO by the method of Otmar et.
  • Compoj ⁇ nd 9 is prepared from the condensation of compound 8 9-(2- phosphonylmethoxyethyl)adenine (PMEA) and compound 3 in pyridine and acetic acid as catalyst.
  • the synthesis of compound 11 is achieved by the reaction between 3'-azido-3'- deoxythymidine and diethyl[p-toluenesulfonyl-oxy]methylphosphonate in the presence of NaH, followed by hydrolysis with TMSBr to obtain the phosphonate intermediate 10 which is finally reacted with 3 to give the hexadecyloxypropyl-phospho conjugate 11.
  • compositions provided herein contain therapeutically effective amounts of one or more of the compounds provided herein that are useful in the prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders associated with viral infections and inappropriate cell proliferation and a pharmaceutically acceptable carrier.
  • Pharmaceutical carriers suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • the compounds may be formulated as the sole pharmaceutically active ingredient in the composition or may be combined with other active ingredients.
  • compositions contain one or more compounds provided herein.
  • the compounds are, in one embodiment, formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers.
  • suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers.
  • the compounds described above are formulated into pharmaceutical compositions using techniques and procedures well known in the art ((see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Seventh Edition 1999).
  • compositions effective concentrations of one or more compounds or pharmaceutically acceptable derivatives thereof is (are) mixed with a suitable pharmaceutical carrier.
  • the compounds may be derivatized as the corresponding salts, esters, enol ethers or esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs prior to formulation, as described above.
  • concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration, that treats, prevents, or ameliorates one or more of the symptoms of diseases or disorders associated with associated with viral infections or inappropriate cell proliferation.
  • the compositions are formulated for single dosage administration. To formulate a composition, the weight fraction of a compound is dissolved, suspended, dispersed or otherwise mixed in a selected carrier at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
  • the active compound is included in the pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful effect in the absence of undesirable side effects on the patient treated.
  • the therapeutically effective concentration may be determined empirically by testing the compounds in in vitro and in vivo systems well known to those of skill in the art and then extrapolated therefrom for dosages for humans.
  • the concentration of active compound in the pharmaceutical composition will depend on absorption, inactivation and excretion rates of the active compound, the physicochemical characteristics of the compound, the dosage schedule, and amount administered as well as other factors known to those of skill in the art.
  • the amount that is delivered is sufficient to ameliorate one or more of the symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation, as described herein.
  • a therapeutically effective dosage should produce a serum concentration of active ingredient of from about 0.1 ng/ml to about 50- 100 ⁇ g/ml.
  • the pharmaceutical compositions in another embodiment, should provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day.
  • Pharmaceutical dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500mg, 1000 mg or 2000 mg, and in one embodiment from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • solubilizing compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEN®, or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions. Upon mixing or addition of the compound(s), the resulting mixture may be a solution, suspension, emulsion or the like. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.
  • cosolvents such as dimethylsulfoxide (DMSO)
  • surfactants such as TWEEN®
  • dissolution in aqueous sodium bicarbonate such as sodium bicarbonate
  • the pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds or pharmaceutically acceptable derivatives thereof.
  • the pharmaceutically therapeutically active compounds and derivatives thereof are, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit-dose forms as used herein refer to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.
  • unit-dose forms include ampoules and syringes and individually packaged tablets or capsules.
  • Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form.
  • multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons.
  • multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art.
  • the contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%.
  • compositions are lactose-free compositions containing excipients that are well known in the art and are listed, for example, in the U.S.
  • lactose-free compositions contains active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Particular lactose-free dosage forms contain active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
  • anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf- life or the stability of formulations over time. See, e.g., Jens T.
  • Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are generally packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs. 1. Compositions for oral administration
  • Oral pharmaceutical dosage forms are either solid, gel or liquid.
  • the solid dosage forms are tablets, capsules, granules, and bulk powders.
  • Types of oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated.
  • Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art.
  • Solid compositions for oral administration In certain embodiments, the formulations are solid dosage forms, in one embodiment, capsules or tablets.
  • the tablets, pills, capsules, troches and the like can contain one or more of the following ingredients, or compounds of a similar nature: a binder; a lubricant; a diluent; a glidant; a disintegrating agent; a coloring agent; a sweetening agent; a flavoring agent; a wetting agent; an emetic coating; and a film coating.
  • binders include microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, molasses, polvinylpyrrolidine, povidone, crospovidones, sucrose and starch paste.
  • Lubricants include talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • Diluents include, for example, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • Glidants include, but are not limited to, colloidal silicon dioxide.
  • Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate.
  • Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors.
  • Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene laural ether.
  • Emetic-coatings include fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
  • Film coatings include hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
  • the compound, or pharmaceutically acceptable derivative thereof could be provided in a composition that protects it from the acidic environment of the stomach.
  • the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
  • the composition may also be formulated in combination with an antacid or other such ingredient.
  • the dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil.
  • dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • the compounds can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like.
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics.
  • the active ingredient is a compound or pharmaceutically acceptable derivative thereof as described herein. Higher concentrations, up to about 98% by weight of the active ingredient may be included.
  • tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.
  • enterically digestible coating such as phenylsalicylate, waxes and cellulose acetate phthalate.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Aqueous solutions include, for example, elixirs and syrups. Emulsions are either oil-in- water or water-in-oil. Elixirs are clear, sweetened, hydroalcoholic preparations.
  • Pharmaceutically acceptable carriers used in elixirs include solvents. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative.
  • An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid.
  • Pharmaceutically acceptable carriers used in emulsions are non-aqueous liquids, emulsifying agents and preservatives. Suspensions use pharmaceutically acceptable suspending agents and preservatives.
  • Pharmaceutically acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form include diluents, sweeteners and wetting agents.
  • Pharmaceutically acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form include organic acids and a source of carbon dioxide. Coloring and flavoring agents are used in all of the above dosage forms.
  • Solvents include glycerin, sorbitol, ethyl alcohol and syrup.
  • preservatives include glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol.
  • non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil.
  • emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • Suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia.
  • Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as saccharin.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • Organic acids include citric and tartaric acid.
  • Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof.
  • Flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds which produce a pleasant taste sensation.
  • the solution or suspension in for example, propylene carbonate, vegetable oils or triglycerides, is in one embodiment encapsulated in a gelatin capsule.
  • a gelatin capsule Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Patent Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the solution e.g., for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be easily measured for administration.
  • a pharmaceutically acceptable liquid carrier e.g., water
  • liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells.
  • Other useful formulations include those set forth in U.S. Patent Nos. RE28,819 and 4,358,603.
  • such formulations include, but are not limited to, those containing a compound provided herein, a dialkylated mono- or poly-alkylene glycol, including, but not limited to, 1,2- dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
  • BHT butylated
  • formulations include, but are not limited to, aqueous alcoholic solutions including a pharmaceutically acceptable acetal.
  • Alcohols used in these formulations are any pharmaceutically acceptable water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol.
  • Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes such as acetaldehyde diethyl acetal. 2. Injectables, solutions and emulsions
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • the injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol.
  • compositions to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • a compound provided herein is dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, poly
  • Parenteral administration of the compositions includes intravenous, subcutaneous and intramuscular administrations.
  • Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions.
  • the solutions may be either aqueous or nonaqueous.
  • suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • PBS physiological saline or phosphate buffered saline
  • thickening and solubilizing agents such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
  • aqueous vehicles examples include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection.
  • Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
  • Antimicrobial agents in bacteriostatic or fungistatic concentrations must be added to parenteral preparations packaged in multiple-dose containers which include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
  • Isotonic agents include sodium chloride and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (TWEEN® 80). A sequestering or chelating agent of metal ions include EDTA. Pharmaceutical carriers also include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • the concentration of the pharmaceutically active compound is adjusted so that an injection provides an effective amount to produce the desired pharmacological effect.
  • the exact dose depends on the age, weight and condition of the patient or animal as is known in the art.
  • the unit-dose parenteral preparations are packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration must be sterile, as is known and practiced in the art.
  • intravenous or intraarterial infusion of a sterile aqueous solution containing an active compound is an effective mode of administration.
  • Another embodiment is a sterile aqueous or oily solution or suspension containing an active material injected as necessary to produce the desired pharmacological effect.
  • Injectables are designed for local and systemic administration.
  • a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1% w/w up to about 90% w/w or more, in certain embodiments more than 1% w/w of the active compound to the treated tissue(s).
  • the compound may be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug.
  • the form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle.
  • the effective concentration is sufficient for ameliorating the symptoms of the condition and may be empirically determined.
  • Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; 6,699,500 and 6,740,634, each of
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled- release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time.
  • the drug In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al, Surgery 88:507 (1980); Saudek et al, N. Engl. J. Med. 321:57 '4 (1989).
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical
  • a controlled release device is introduced into a subject in proximity of the site of inappropriate immune activation or a tumor.
  • Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990).
  • the active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene- vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neo
  • Lyophilized powders Of interest herein are also lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. They may also be reconstituted and formulated as solids or gels.
  • the sterile, lyophilized powder is prepared by dissolving a compound provided herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent.
  • the solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder.
  • Excipients that may be used include, but are not limited to, an antioxidant, a buffer and a bulking agent.
  • the excipient is selected from dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose and other suitable agent.
  • the solvent may contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, at about neutral pH.
  • the resulting solution will be apportioned into vials for lyophilization.
  • Each vial will contain a single dosage or multiple dosages of the compound.
  • the lyophilized powder can be stored under appropriate conditions, such as at about 4 0 C to room temperature.
  • Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration.
  • the lyophilized powder is added to sterile water or other suitable carrier. The precise amount depends upon the selected compound. Such amount can be empirically determined.
  • Topical mixtures are prepared as described for the local and systemic administration.
  • the resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
  • the compounds or pharmaceutically acceptable derivatives thereof may be formulated as aerosols for topical application, such as by inhalation (see, e.g., U.S. Patent Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment of inflammatory diseases, particularly asthma).
  • These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation will, in one embodiment, have diameters of less than 50 microns, in one embodiment less than 10 microns.
  • the compounds may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application.
  • Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies.
  • Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered.
  • the preparation may contain an esterified phosphonate compound dissolved or suspended in a liquid carrier, in particular, an aqueous carrier, for aerosol application.
  • the carrier may contain solubilizing agents such as propylene glycol, surfactants, absorption enhancers such as lecithin or cyclodextrin, or preservatives.
  • solubilizing agents such as propylene glycol, surfactants, absorption enhancers such as lecithin or cyclodextrin, or preservatives.
  • These solutions particularly those intended for ophthalmic use, may be formulated as 0.01% - 10% isotonic solutions, pH about 5-7, with appropriate salts.
  • compositions for other routes of administration are provided.
  • transdermal patches including iontophoretic and electrophoretic devices, and rectal administration, are also contemplated herein.
  • Transdermal patches including iotophoretic and electrophoretic devices, are well known to those of skill in the art.
  • such patches are disclosed in U.S. Patent Nos. 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010715, 5,985,317, 5,983,134, 5,948,433, and 5,860,957.
  • pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum which melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients.
  • compositions utilized in rectal suppositories are bases or vehicles and agents to raise the melting point.
  • bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used.
  • Agents to raise the melting point of suppositories include spermaceti and wax.
  • Rectal suppositories may be prepared either by the compressed method or by molding.
  • the weight of a rectal suppository in one embodiment, is about 2 to 3 gm. Tablets and capsules for rectal administration are manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
  • the compounds provided herein, or pharmaceutically acceptable derivatives thereof, may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated. Many such targeting methods are well known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Patent Nos.
  • liposomal suspensions including tissue-targeted liposomes, such as tumor-targeted liposomes, may also be suitable as pharmaceutically acceptable carriers.
  • tissue-targeted liposomes such as tumor-targeted liposomes
  • liposome formulations may be prepared according to methods known to those skilled in the art.
  • liposome formulations may be prepared as described in U.S. Patent No. 4,522,811. Briefly, liposomes such as multilamellar vesicles (MLVs) may be formed by drying down egg phosphatidyl choline and brain phosphatidyl serine (7:3 molar ratio) on the inside of a flask.
  • MLVs multilamellar vesicles
  • a solution of a compound provided herein in phosphate buffered saline lacking divalent cations (PBS) is added and the flask shaken until the lipid film is dispersed.
  • PBS phosphate buffered saline lacking divalent cations
  • the compounds or pharmaceutically acceptable derivatives may be packaged as articles of manufacture containing packaging material, a compound or pharmaceutically acceptable derivative thereof provided herein, which is effective for for treatment, prevention or amelioration of one or more symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation, within the packaging material, and a label that indicates that the compound or composition, or pharmaceutically acceptable derivative thereof, is used for the treatment, prevention or amelioration of one or more symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation.
  • the articles of manufacture provided herein contain packaging materials.
  • Packaging materials for use in packaging pharmaceutical products are well known to those of skill in the art. See, e.g., U.S. Patent Nos. 5,323,907, 5,052,558 and 5,033,252.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • a wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease or disorder associated with viral infections or inappropriate cell proliferation. E. Dosages
  • the physician will determine the dosage regimen that is most appropriate according to a preventive or curative treatment and according to the age, weight, stage of the disease and other factors specific to the subject to be treated.
  • the pharmaceutical compositions in another embodiment, should provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day.
  • Pharmaceutical dosage unit forms are prepared, e.g., to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500mg, 1000 mg or 2000 mg, and in one embodiment from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
  • the amount of active ingredient in the formulations provided herein, which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof, will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
  • Exemplary doses of a formulation include milligram or microgram amounts of the active compound per kilogram of subject or sample weight (e.g., from about 1 micrograms per kilogram to about 50 milligrams per kilogram, from about 10 micrograms per kilogram to about 30 milligrams per kilogram, from about 100 micrograms per kilogram to about 10 milligrams per kilogram, or from about 100 microgram per kilogram to about 5 milligrams per kilogram).
  • compositions provided herein are also encompassed by the above described dosage amounts and dose frequency schedules.
  • the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the composition or it may be decreased to reduce one or more side effects that a particular subject is experiencing.
  • administration of the same formulation provided herein may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • the activity of the compounds as antivirals can be measured in standard assays known in the art.
  • Exemplary assays include, but are not limited to, plaque reduction assay in HFF cells, DNA reduction assay in MRC-5 cells, p24 reduction assay in MT-2 cells, CPE assay in HFF cells and EBV Elisa assay in Daudi cells.
  • the methods provided herein are for the preventing, or ameliorating one or more symptoms of diseases associated with viral infections, including, but not limited to influenza; hepatitis B and C virus; cytomegalovirus (CMV); herpes infections, such as those caused by Varicella zoster virus, Herpes simplex virus types 1 & 2, Epstein-Barr virus, Herpes type 6 (HHV-6) and type 8 (HHV-8); Varicella zoster virus infections such as shingles or chicken pox; Epstein Barr virus infections, including, but not limited to infectious mononucleosis/glandular; retroviral infections including, but not limited to SIV, HIV-I and HIV-2; ebola virus; adenovirus and papilloma virus
  • the methods provided herein are for treating, preventing, treating, or ameliorating one or more symptoms of diseases associated with viral infections caused by orthopox viruses, such as variola major and minor, vaccinia, smallpox, cowpox, camelpox, and monkeypox.
  • the disease is drug resistant hepatitis B.
  • the methods provided herein are for treating, preventing, or ameliorating one or more symptoms of diseases associated with cell proliferation, including, but not limited to cancers.
  • cancers include, but are not limited to, lung cancer, head and neck squamous cancers, colorectal cancer, prostate cancer, breast cancer, acute lymphocytic leukemia, adult acute myeloid leukemia, adult non Hodgkin's lymphoma, brain tumors, cervical cancers, childhood cancers, childhood sarcoma, chronic lymphocytic leukemia, chronic myeloid leukemia, esophageal cancer, hairy cell leukemia, kidney cancer, liver cancer, multiple myeloma, neuroblastoma, oral cancer, pancreatic cancer, primary central nervous system lymphoma, and skin cancer.
  • lung cancer head and neck squamous cancers, colorectal cancer
  • prostate cancer breast cancer
  • acute lymphocytic leukemia adult acute myeloid leukemia, adult non Hodgkin's lymphoma
  • brain tumors cervical cancers
  • childhood cancers childhood sarcoma
  • chronic lymphocytic leukemia chronic myeloid
  • the compounds and compositions provided herein may also be used in combination with one or more other active ingredients.
  • the compounds may be administered in combination, or sequentially, with another therapeutic agent.
  • Such other therapeutic agents include those known for treatment, prevention, or amelioration of one or more symptoms associated with viral infections or inappropriate cell proliferation.
  • therapeutic agents include, but are not limited to, antiviral agents and anti-neoplastic agents.
  • a second agent effective for the treatment, prevention or amelioration of viral infection such as HIV and/or HCV infection.
  • the second agent can be any agent known to those of skill in the art to be effective for the treatment, prevention or amelioration of viral infections, such as the HIV and/or HCV infection.
  • the second agent can be a second agent presently known to those of skill in the art, or the second agent can be second agent later developed for the treatment, prevention or amelioration of viral infections.
  • the second agent is presently approved for the treatment or prevention of HIV and/or HCV.
  • a compound provided herein is administered in combination with one second agent.
  • a second agent is administered in combination with two second agents.
  • a second agent is administered in combination with two or more second agents.
  • the second antiviral agent for the treatment of HIV can be a reverse transcriptase inhibitor (a "RTI"), which can be either a synthetic nucleoside (a "NRTI”) or a non-nucleoside compound (a "NNRTI”).
  • RTI reverse transcriptase inhibitor
  • NRTI non-nucleoside compound
  • the second (or third) antiviral agent can be a protease inhibitor.
  • the second (or third) compound can be a pyrophosphate analog, or a fusion binding inhibitor.
  • compounds for combination or alternation therapy for the treatment of HBV include, but are not limited to 3TC, FTC, L-FMAU, interferon, ⁇ -D- dioxolanyl-guanine (DXG), ⁇ -D-dioxolanyl-2,6-diaminopurine (DAPD), and ⁇ -D-dioxolanyl- 6-chloropurine (ACP), famciclovir, penciclovir, BMS-200475, bis pom PMEA (adefovir, dipivoxil); lobucavir, ganciclovir, and ribavarin.
  • DXG ⁇ -D- dioxolanyl-guanine
  • DAPD ⁇ -D-dioxolanyl-2,6-diaminopurine
  • ACP ⁇ -D-dioxolanyl- 6-chloropurine
  • famciclovir penciclovir
  • examples of antiviral agents that can be used in combination or alternation with the compounds disclosed herein for HIV therapy include cis-2- hydroxymethyl-5-(5-fluorocytosin-l-yl)-l,3-oxathiolane (FTC); the (-;)-enantiomer of 2- hydroxymethyl-5-(cytosin-l-yl)-l,3-oxathiolane (3TC); carbovir, acyclovir, foscarnet, interferon, AZT, DDI, DDC, D4T, CS-87 (3'-azido-2',3'-dideoxy-uridine), and ⁇ -D-dioxolane nucleosides such as ⁇ -D-dioxolanyl-guanine (DXG), ⁇ -D-dioxolanyl-2,6-diaminopurine (DAPD), and ⁇ -D-dioxolanyl-6-chlor
  • protease inhibitors include crixivan, nelfinavir, ritonavir, saquinavir, DMP-266 and DMP-450.
  • Nevirapine 11-cyclopropy 1-5,1 l-dihydro-4-methyl-6H-dipyridol[3,2-b:2',3'-e]diazepin-6- one; NSC648400:l-benzyloxymethyl-5-ethyl-6-(alpha-pyridylthio)uracil (E-BPTU); P9941: [2-pyridylacetyl-IlePheAla-y(CHOH)]2; PFA: phosphonoformate; PMEA: 9-(2- phosphonylmethoxyethyl)adenine; PMPA: (R)-9-(2-phosphonyl-methoxypro ⁇ yl)adenine; Ro 31-8959: hydroxyethylamine derivative HIV-I protease inhibitor; RPI-312: peptidyl protease inhibitor, l-[(3s)-3-(n-alpha-benzyloxycarbonyl)-l-a
  • suitable second agents include small-molecule, orally bioavailable inhibitors of the HCV enzymes, nucleic-acid-based agents that attack viral RNA, agents that can modulate the host immune response.
  • exemplary second agents include: (i) current approved therapies (peg-interferon plus ribavirin), (ii) HCV-enzyme targeted compounds, (iii) viral-genome-targeted therapies (e.g., RNA interference or RNAi), and (iv) immunomodulatory agents such as ribavirin, interferon (INF) and Toll-receptor agonists.
  • the second agent is a modulator of the NS3-4A protease.
  • the NS3-4A protease is a heterodimeric protease, comprising the amino-terminal domain of the NS3 protein and the small NS4A cofactor. Its activity is essential for the generation of components of the viral RNA replication complex.
  • BILN 2061 (Ciluprevir; Boehringer Ingelheim), a macrocyclic mimic of peptide product inhibitors. Although clinical trials with BILN 2061 were halted (preclinical cardiotoxicity), it was the first NS3 inhibitor to be tested in humans. See Lamarre et al, 2003, Nature 426:186-189, the contents of which are hereby incorporated by reference in their entirety.
  • NS3-4A protease inhibitor is VX-950 (Vertex/Mitsubishi), a protease- cleavage-product-derived peptidomimetic inhibitor of the NS3-4A protease. It is believed to be stabilized into the enzyme's active site through a ketoamide. See, e.g., Lin et al, 2005, J Biol Chem. Manuscript M506462200 (epublication); Summa, 2005, Curr Opin Investig Drugs. 6:831-7, the contents of which are hereby incorporated by reference in their entireties.
  • the second agent is a modulator of the HCV NS5B
  • NM283 Valopicitabine; Idenix/Novartis
  • NM283 is an oral prodrug (valine ester) of NMl 07 (2 -C-methyl-cytidine) in phase II trials for the treatment or prevention of HCV infection. See, e.g., U.S. Patent Application Publication No. 20040077587, the contents of which are hereby incorporated by reference in their entirety.
  • RdRp Other useful modulators of RdRp include 7-deaza nucleoside analogs.
  • 7-Deaza-2'-C-methyl-adenosine is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties. Olsen et al., 2004, Antimicrob. Agents Chemother. 48:3944-3953, the contents of which are hereby incorporated by reference in their entirety.
  • the second agent is a non-nucleoside modulator of NS5B.
  • NNI non-nucleoside inhibitors
  • Useful non-nucleoside modulators of NS5B include JTK-003 and JTK-009. JTK-003 has been advanced to phase II.
  • Useful non-nucleoside modulators of NS5B include the 6,5- fused heterocyclic compounds based on a benzimidazole or indole core. See, e.g., Hashimoto et al, WO 00147883, the contents of which are hereby incorporated by reference in their entirety.
  • NNIs include R803 (Rigel) and HCV-371, HCV-086 and HCV-796 (ViroPharma/Wyeth).
  • Additional useful NNIs include thiophene derivatives that are reversible allosteric inhibitors of the NS5B polymerase and bind to a site that is close to, but distinct from, the site occupied by benzimidazole-based inhibitors. See, e.g., Biswal, et al, 2005, J. Biol. Chem. 280, 18202-18210 (2005).
  • NNIs for the methods provided herein include benzothiadiazines, such as benzo-l,2,4-thiadiazines.
  • benzothiadiazines such as benzo-l,2,4-thiadiazines.
  • Derivatives of benzo-l,2,4-thiadiazine have been shown to be highly selective inhibitors of the HCV RNA polymerase. Dhanak, et al, 2002, J. Biol. Chem. 277:38322-38327, the contents of which are hereby incorporated by reference in their entirety.
  • NNIs for the methods provided herein, and their mechanisms, are described in LaPlante et al, 2004 Angew Chem. Int. Ed. Engl. 43:4306-4311; Tomei et al, 2003, J. Virol. 77:13225-13231; Di Marco et al, 2005, J. Biol Chem. 280:29765-70; Lu, H., WO 2005/000308; Chan et al, 2004, Bioorg. Med. Chem. Lett. 14:797-800; Chan et al, 2004, Bioorg. Med. Chem. Lett. 14:793-796; Wang et al, 2003, J. Biol Chem.
  • the second agent is an agent that is capable of interfering with HCV RNA such as small inhibitory RNA (siRNA) or a short hairpin RNA (shRNA) directed to an HCV polynucleotide.
  • siRNA small inhibitory RNA
  • shRNA short hairpin RNA
  • siRNA and vector-encoded short hairpin RNA shRNA directed against the viral genome, effectively block the replication of HCV replicons. See, e.g., Randall et al., 2003, Proc. Natl Acad. Sci. USA 100:235-240, the contents of which are hereby incorporated by reference in their entirety.
  • the second agent is an agent that modulates the subject's immune response.
  • the second agent can be a presently approved therapy for HCV infection such as an interferon (IFN), a pegylated IFN, an IFN plus ribavirin or a pegylated IFN plus ribavirin.
  • interferons include IFN ⁇ , IFN ⁇ 2a and IFN ⁇ 2b, and particularly pegylated IFN ⁇ 2a (PEGASYS®) or pegylated IFN ⁇ 2b (PEG-INTRON®).
  • the second agent is a modulator of a Toll-like receptor (TLR). It is believed that TLRs are targets for stimulating innate anti-viral response.
  • TLR Toll-like receptor
  • Suitable TLRs include, bur are not limited to, TLR3, TLR7, TLR8 and TLR9. It is believed that toll-like receptors sense the presence of invading microorganisms such as bacteria, viruses and parasites. They are expressed by immune cells, including macrophages, monocytes, dendritic cells and B cells. Stimulation or activation of TLRs can initiate acute inflammatory responses by induction of antimicrobial genes and pro-inflammatory cytokines and chemokines.
  • the second agent is a polynucleotide comprising a CpG motif.
  • Synthetic oligonucleotides containing unmethylated CpG motifs are potent agonists of TLR-9. Stimulation of dendritic cells with these oligonucleotides results in the production of tumour necrosis factor-alpha, interleukin-12 and IFN-alpha.
  • TLR-9 ligands are also potent stimulators of B-cell proliferation and antibody secretion.
  • One useful CpG-containing oligonucleotide is CPG-10101 (Actilon; Coley Pharmaceutical Group) which has been evaluated in the clinic.
  • ANA975 Another useful modulator of a TLR is ANA975 (Anadys). ANA975 is believed to act through TLR-7, and is known to elicit a powerful anti-viral response via induction and the release of inflammatory cytokines such as IFN-alpha.
  • the second agent is Celgosivir.
  • Celgosivir is an alpha- glucosidase I inhibitor and acts through host-directed glycosylation. In preclinical studies, celgosivir has demonstrated strong synergy with IFN ⁇ plus ribavirin. See, e.g., Whitby et al., 2004, Antivir Chem Chemother. 15(3):141-51. Celgosivir is currently being evaluated in a Phase II monotherapy study in chronic HCV patients in Canada.
  • the compounds provided herein may be administered in combination with one or more anti-cancer agents.
  • Anti-cancer agents for use in combination with the instant compounds include, but are not limited to, an antifolate, a 5-fluoropyrimidine (including 5-fluorouracil), a cytidine analogue such as ⁇ -L-l,3-dioxolanyl cytidine or ⁇ -L- 1,3-dioxolanyl 5-fluorocytidine, antimetabolites (including purine antimetabolites, cytarabine, fudarabine, floxuridine, 6-mercaptopurine, methotrexate, and 6-thioguanine), hydroxyurea, mitotic inhibitors (including CPT-11, Etoposide (VP-21), taxol, and vinca alkaloids such as vincristine and vinblastine, an alkylating agent (including but not limited to busulfan, chlorambucil, cyclophosphamide, if
  • the compounds provided herein can also be used in combination with enzyme therapy agents and immune system modulators such as an interferon, interleukin, tumor necrosis factor, macrophage colony-stimulating factor and colony stimulating factor. It should be understood that any suitable combination of the compounds provided herein with one or more of the above-mentioned compounds and optionally one or more further pharmacologically active substances are considered to be within the scope of the present disclosure. In another embodiment, the compound provided herein is administered prior to or subsequent to the one or more additional active ingredients.
  • the following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention.
  • HDP-phospho-morpholidate was prepared using the procedure described in example 1 using 1.92g (15 mmol) of 3-(hexadecyloxypropyl) dihydrogen phosphate, 1.3g (15 mmol) of morpholine and 35 mmol of DCC. HDP-phospho-morpholidate was obtained as an oil. Mass spectrum: (ESI) m/z 449 (M-H) " .
  • Example 3
  • HDP-P-HPMPC (l-(4- aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2yloxy)-methylphosphonic(3-hexadecyloxy)propyl-phosphoric) anhydride (6)
  • ODE-P-HPMPC (l-(4-aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2-yloxy)methylphosphonic-(3- octadecyloxy)ethyl phosphoric) anhydride
  • ADV adefovir, phosphonomethoxyethyadenine
  • HDP-P-ADV (2-(6amino-9H- purin-9-yl)ethoxy)methylphosphonic(3-hexadecyloxy)-propylphosphonic)anhydride
  • Antiviral Assays 'Cow

Abstract

Compounds and compositions are provided for treatment, prevention, or amelioration of a variety of medical disorders associated with viral infections and/or cell proliferation. The compounds provided herein are obtained by attaching the phosphonate nucleoside of interest to alkyloxyalkyl-phosphate or alkyl-phosphate in a phosphate-phosphono anhydride linkage to provide a modified nucleoside phosphonate drug.

Description

SUBSTITUTED PHOSPHATE ESTERS OF NUCLEOSIDE PHOSPHONATES
GRANT INFORMATION This invention was made with government support under Grant No. AI29164 awarded by the National Institute of Allergy and Infectious Diseases/ National Health Institute and Grant No. DAMD 17-01-2-0071 awarded by United States Army. The United States government has certain rights in this invention. RELATED APPLICATION DATA This application claims priority under 35 U.S.C. §119(e) to U.S. provisional application Serial No. 60/667,739, entitled "Substituted Phosphate Esters Of Nucleoside Phosphonates" to Hostetler et al., filed April 1, 2005. The contents of the provisional application are incorporated by reference herein in their entirety. FIELD Provided herein are antiviral and anticancer phosphonate drugs, their preparation, and their use for treatment of viral infections and cancers. Also provided are methods for synthesizing anhydrides containing alkyl phosphate or alkoxyalkyl phosphate coupled to nucleoside phosphonate drugs. The new conjugates have greater antiviral and/or antiproliferative activity when compared with the parent nucleoside phosphonate.
In another embodiment, provided herein are methods of treatment, prevention, or amelioration of a variety of medical disorders associated with viral infections and cell proliferation using the compounds and compositions provided herein. BACKGROUND Phosphonate nucleosides are well known in the art and are in clinical use as antiviral and anticancer agents {see, Holy, A., Phosphonomethoxyalkyl analogs of nucleotides, Current Pharmaceutical Design 9(31), 2567-92, 2003). Their limitations relate to poor oral bioavailability, poor target cell uptake and toxicity in kidneys. In general, nucleoside phosphonate uptake into target cells is poor because of the dual negative charges on the phosphonate moiety. Once in the cell, they require two subsequent anabolic phosphorylations to achieve activity as the nucleoside phosphonate diphosphate. Some nucleoside phosphonates are hampered by slow phosphorylation. There is a continuing need for less toxic, more effective pharmaceutical agents to treat a variety of disorders associated with viral infection, and cell proliferation. SUMMARY
Provided herein are nucleoside phosphonates linked via their phosphonate residue to the phosphate of alkoxyalkyl-phosphate, alkylglycerol-phosphate or alkyl- phosphate and pharmaceutically acceptable derivatives thereof. In certain embodiments, the nucleoside phosphonates or acyclic nucleoside phosphonates linked to the phosphate of alkoxyalkyl-phosphate, alkylglycerol-phosphate or alkyl-phosphate result in orally available compounds which exhibit greater antiviral or anticancer activity by promoting cell uptake and favorable cellular metabolism which yields the nucleoside phosphonate monophosphate, bypassing the need for the first of two anabolic phosphorylations. In certain embodiments, compounds provided herein exhibit greater antiviral or anticancer activity than the unmodified nucleoside phosphonates.
Also provided are compositions and methods of using the compounds and compositions for the treatment of various diseases. In one embodiment, compounds and compositions provided herein have antiviral activity. In another embodiment, provided herein are compounds and compositions that are useful in the treatment, prevention, or amelioration of one or more symptoms associated with cell proliferation.
In one embodiment, the compounds for use in the compositions and methods provided herein have formula I:
Figure imgf000003_0001
or pharmaceutically acceptable derivatives thereof, wherein RL is a lipophilic group, Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound, coupled to the phosphate group by an anhydride linkage and y is 1 or 2. Also provided are pharmaceutically-acceptable derivatives, including salts, esters, enol ethers, enol esters, solvates, hydrates and prodrugs of the compounds described herein. Further provided are pharmaceutical compositions containing the compounds provided herein and a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical compositions are formulated for single dosage administration. Methods of treatment, prevention or amelioration using the compounds and compositions provided herein are provided. Such methods encompass treating, preventing or ameliorating one or more symptoms of diseases associated with viral infections and cell proliferation. In practicing the methods, effective amounts of the compounds or compositions containing therapeutically effective concentrations of the compounds are administered.
Articles of manufacture are provided containing packaging material, a compound or composition provided herein which is useful for treating, preventing, or ameliorating one or more symptoms of diseases or disorders associated with viral infections or cell proliferation using the compounds and compositions provided herein, and a label that indicates that the compound or composition is useful for treating, preventing, or ameliorating one or more symptoms of diseases or disorders associated with viral infections or cell proliferation. DETAILED DESCRIPTION A. Definitions
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, applications, published applications and other publications are incorporated by reference in their entirety. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.
As used herein the terms "phosphonate" and "phosphonate group" mean a functional group or moiety within a molecule that comprises at least one phosphorus- carbon bond, and at least one phosphorus-oxygen double bond. The phosphorus atom is further substituted with oxygen, sulfur, and nitrogen substituents. These substituents may be part of a prodrug moiety. As used herein, "phosphonate" and "phosphonate group" include molecules with phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, phosphondiamidate, and phosphonthioate functional groups. As used herein, the term "nucleoside" refers to a molecule composed of a heterocyclic base and a carbohydrate. A nucleoside is composed of a heterocyclic nitrogenous base in N-glycosidic linkage with a sugar. Nucleosides are recognized in the art to include natural bases (standard), and non-natural bases well known in the art. The carbohydrates include the true sugars found in natural nucleosides or a species replacing the ribofuranosyl moiety or acyclic sugars. The heterocyclic nitrogenous bases are generally located at the 1' position of a nucleoside sugar moiety. Nucleosides generally contain a base and sugar group. The nucleosides can be unmodified or modified at the sugar, and/or base moiety, (also referred to interchangeably as nucleoside analogs, modified nucleosides, non-natural nucleosides, non-standard nucleosides; see for example, Eckstein et al, International PCT Publication No. WO 92/07065 and Usman et al, International PCT Publication No. WO 93/15187). In natural nucleosides the heterocyclic base is thymine, uracil, cytosine, adenine or guanine. In certain embodiments, acyclic sugars contain 3-6 carbon atoms and include, for example, the acyclic sugar moieties present in acyclovir (-CH2-O-CH2-CH2-OH), ganciclovir (-CH2-O-CH(CH2OH)-CH2-OH), and the like. Natural nucleosides have the β-D-configuration. The term "nucleoside" shall be understood to encompass unnatural configurations and species replacing the true sugar that lack an anomeric carbon. In natural nucleosides the heterocyclic base is attached to the carbohydrate through a carbon-nitrogen bond. The term "nucleoside" shall be understood to encompass species wherein the heterocyclic base and carbohydrate are attached through a carbon-carbon bond (C -nucleosides).
Where the nucleoside contains 1 or more functional groups that may be reactive to form undesired products under the reaction conditions of the present process, for example, the amino groups of cytosine and adenine and the 2-amino and 6-oxo groups of guanine, such functional groups may be blocked using the protecting groups commonly employed in nucleoside chemistry. For example, the amino group of adenine and cytosine may be protected by benzoyl; the 6-oxo and 2-amino groups of guanine may be protected by the triphenylmethyl (trityl) group. The selection of methods for introducing and subsequent removal of such protecting groups are well known to one of ordinary skill in the pertinent art.
As used herein, the term "nucleoside base" refers to natural and non-natural purine and pyrimidine bases, including adenine, thymine, cytosine, guanine and uracil and analogs thereof. The terms "nucleoside phosphonate" and "acyclic nucleoside phosphonate" refer to the group of phosphonomethoxyalkyl or phosphono substituted nucleoside derivatives that are biologically active, for example, as anti-viral, anti-cancer or antiparasitic drugs. As used herein, the terms "lipophilic" or "long-chain" refer to the cyclic, branched or straight chain chemical groups that when covalently linked to a phosphonic acid to form a phosphonate monoester increase oral bioavailability and enhance activity of the nucleoside phosphonates as compared with the parent nucleoside phosphonates. These lipophilic groups include, but are not limited to alkyl, alkoxyalkyl, and alkylglyceryl.
As used herein, the term "lipophilic monoesters of nucleoside phosphonates" refers to compound where a lipophilic group is covalently attached to a nucleoside phosphonate via an ester linkage. As used herein, pharmaceutically acceptable derivatives of a compound include salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives may be readily prepared by those of skill in this art using known methods for such derivatization. The compounds produced may be administered to animals or humans without substantial toxic effects and either are pharmaceutically active or are prodrugs. Pharmaceutically acceptable salts include, but are not limited to, amine salts, such as but not limited to N,N'-dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzylphenethylamine, 1 -para-chlorobenzyl-2-pyrrolidin- 1 '-ylmethyl- benzimidazole, diethylamine and other alkylamines, piperazine and tris(hydroxymethyl)aminomethane; alkali metal salts, such as but not limited to lithium, potassium and sodium; alkali earth metal salts, such as but not limited to barium, calcium and magnesium; transition metal salts, such as but not limited to zinc; and other metal salts, such as but not limited to sodium hydrogen phosphate and disodium phosphate; and also including, but not limited to, nitrates, borates, methanesulfonates, benzenesulfonates, toluenesulfonates, salts of mineral acids, such as but not limited to hydrochlorides, hydrobromides, hydroiodides and sulfates; and salts of organic acids, such as but not limited to acetates, trifluoroacetates, maleates, oxalates, lactates, malates, tartrates, citrates, benzoates, salicylates, ascorbates, succinates, butyrates, valerates and fumarates. Pharmaceutically acceptable esters include, but are not limited to, alkyl, alkenyl, alkynyl, and cycloalkyl esters of acidic groups, including, but not limited to, carboxylic acids, phosphoric acids, phosphinic acids, sulfonic acids, sulfinic acids and boronic acids. Pharmaceutically acceptable enol ethers include, but are not limited to, derivatives of formula C=C(OR) where R is hydrogen, alkyl, alkenyl, alkynyl, and cycloalkyl. Pharmaceutically acceptable enol esters include, but are not limited to, derivatives of formula C=C(OC(O)R) where R is hydrogen, alkyl, alkenyl, alkynyl, or cycloalkyl. Pharmaceutically acceptable solvates and hydrates are complexes of a compound with one or more solvent or water molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4, solvent or water molecules.
As used herein, treatment means any manner in which one or more of the symptoms of a disease or disorder are ameliorated or otherwise beneficially altered. As used herein, amelioration of the symptoms of a particular disorder by administration of a particular compound or pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.
As used herein, ECs0 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
As used herein, a prodrug is a compound that, upon in vivo administration, is metabolized by one or more steps or processes or otherwise converted to the biologically, pharmaceutically or therapeutically active form of the compound. To produce a prodrug, the pharmaceutically active compound is modified such that the active compound will be regenerated by metabolic processes. The prodrug may be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those of skill in this art, once a pharmaceutically active compound is known, can design prodrugs of the compound (see, e.g., Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392). Other prodrugs for use herein are described elsewhere herein. It is to be understood that the compounds provided herein may contain chiral centers. Such chiral centers may be of either the (R) or (S) configuration, or may be a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, or be stereoisomeric or diastereomeric mixtures. It is understood that the compounds provided herein encompass any racemic, optically active, polymorphic, or steroisomeric form, or mixtures thereof, of a compound provided herein, which possesses the useful properties described herein, it being well known in the art how to prepare optically active forms and how to determine antiproliferative activity using the standard tests described herein, or using other similar tests which are well known in the art. Examples of methods that can be used to obtain optical isomers of the compounds provided herein include the following: i) physical separation of crystals- a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; ii) simultaneous crystallization- a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state; iii) enzymatic resolutions — a technique whereby partial or complete separation of a racemate by viitue of differing rates of reaction for the enantiomers with an enzyme iv) enzymatic asymmetric synthesis — a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enatiomerically pure or enriched synthetic precursor of the desired enantiomer; v) chemical asymmetric synthesis — a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce assymetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries; vi) diastereomer separations — a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer; vii) first- and second-order asymmetric transformations — a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiorner perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer; viii) kinetic resolutions — this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors — a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; x) chiral liquid chromatography — a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; xi) chiral gas chromatography — a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase; xii) extraction with chiral solvents — a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; xiii) transport across chiral membranes — a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the. non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
As used herein, substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), gel electrophoresis, high performance liquid chromatography (HPLC) and mass spectrometry (MS), used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance. Methods for purification of the compounds to produce substantially chemically pure compounds are known to those of skill in the art. A substantially chemically pure compound may, however, be a mixture of stereoisomers. In such instances, further purification might increase the specific activity of the compound.
As used herein, the term "alkyl" refers to a monovalent straight or branched chain or cyclic radical. In certain embodiments, the alkyl group contains from one to twenty-four carbon atoms, including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl, octadecyl, nonadecyl, eicosyl, 18-methyl-nonadecyl, 19- methyl-eicosyl, and the like. As used herein lower alkyl refers to alkyl groups of 1 to 6 carbon atoms. As used herein, "substituted alkyl" refers to alkyl groups further bearing one or more substituents, including, but not limited to substituents selected from lower alkyl, hydroxy, alkoxy (of a lower alkyl group), mercapto (of a lower alkyl group), cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryloxy, substituted aryloxy, halogen, trifluoromethyl, cyano,azido, nitro, nitrone, amino, amido, -C(O)H, acyl, oxyacyl, carboxyl, carbamate, sulfonyl, sulfonamide, and sulfuryl, which may be protected or unprotected as necessary, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Ed. 1991, hereby incorporated by reference. As used herein, "alkenyl" refers to straight or branched chain hydrocarbon group having one or more carbon-carbon double bonds. In certain embodiments, the alkenyl group contains from 2 up to 24 carbon atoms, and "substituted alkenyl" refers to alkenyl groups further bearing one or more substituents as set forth above.
As used herein, "alkynyl" refers to straight or branched chain hydrocarbon group having one or more carbon-carbon triple bonds. In certain embodiments, the alkynyl group contains from 2 up to 24 carbon atoms, and "substituted alkynyl" refers to alkynyl groups further bearing one or more substituents as set forth above. As used herein, "aryl" refers to aromatic groups having in the range of 6 up to 14 carbon atoms and "substituted aryl" refers to aryl groups further bearing one or more substituents as set forth above.
As used herein, "heteroaryl" refers to aromatic groups containing one or more heteroatoms (e.g., N, O, S, or the like) as part of the ring structure, and having in the range of 3 up to 14 carbon atoms and "substituted heteroaryl" refers to heteroaryl groups further bearing one or more substituents as set forth above.
As used herein "subject" is an animal, such as a mammal, including human, such as a patient. The phrase "effective amount" as used herein means an amount required for prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders associated including those associated with viral infection, cell proliferation and/or bone metabolism .
Where the number of any given substituent is not specified (e.g., haloalkyl), there may be one or more substituents present. For example, "haloalkyl" may include one or more of the same or different halogens.
As used herein, the term "parenteral" includes subcutaneous, intravenous, intraarterial, intramuscular or intravitreal injection, or infusion techniques.
The term "topically" encompasses administration rectally and by inhalation spray, as well as the more common routes of the skin and mucous membranes of the mouth and nose and in toothpaste.
As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (see, (1972) Biochem. 11 :942-944).
Some abbreviations used herein are as follows:
Hexadecyloxypropyl = HDP
Octadecyloxyethyl = ODE
Oleyloxyethyl = OLE, Oleyloxypropyl = OLP,
(S)-9-[3-hydroxy-2-(phosphonomethoxy)-propyl]cytosine = HPMPC (cidofovir), (S)-9-[3-hydroxy-2-(phosphonomethoxy)-propyl]adenine = (S)-HPMPA,
Phosphonomethoxyethyl-guanine = PMEG,
Phosphonomethoxyethyl-adenine = PMEA,
Phosphonomethoxy-propyladenine = PMPA (tenofovir), Hexadecyloxypropyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine = HDP-phospho-(S)-HPMPA,
Octadecyloxyethyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine = ODE-phospho-(S)-HPMPA,
Oleyloxyethyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine = OLE-phospho-(S)-HPMPA,
Oleyloxypropyl-phospho-(S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine= OLP- phospho-(S)-HPMPA,
5-Phosphono-pent-2-en-l-yl adenine = PPen-A,
5-Phosphono-pent-2-en-l-yl cytosine = PPen-C, 5-Phosphono-pent-2-en-l-yl guanine = PPen-G,
5-Phosphonopent-2-en-l-yl thymine = PPen-T and
5-Phosphono-pent-2-en-l-yl uracil = PPen-U.
B. Compounds
In certain embodiments, the compound for use in the compositions and methods provided herein has formula II:
Figure imgf000012_0001
or a pharmaceutically active derivatives thereof, wherein;
R1 and Rlx are each independently -H, optionally substituted -0(C1- C24)alkyl, -O(C1-C24)alkenyl, -O(C1-C24)acyl, -S(CrC24)alkyl, -S(C1-C24)alkenyl, or -S(C1-C24)acyl, wherein at least one of R1 and Rlx is not -H, and wherein the alkenyl or acyl optionally have 1 to about 6 double bonds;
R2 and R2x are each independently -H, optionally substituted -0(C1- C7)alkyl, -O(C1-C7)alkenyl, S(C1-C7)alkyl, -S(C1-C7)alkenyl, -O(C1-C7)acyl, -S(C1- C7)HCyI,
Figure imgf000013_0001
-N((C1-C7)alkyl)2, halogen, -NH2, -OH, or -SH;
Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound, coupled to the phosphate group by an anhydride linkage;
X, when present, is:
Figure imgf000013_0002
L is a valence bond or a bifunctional linking molecule of the formula -J-(CRxRy)t-G-, wherein t is an integer from 1 to 24; J and G are independently -O-, -S-, -C(O)O- or - NH-; Rx and Ry are each independently -H, substituted or unsubstituted alkyl, or alkenyl; m is an integer from 0 to 6; y is 1 or 2; and n is 0 or 1.
In certain embodiments, the compounds for use in the compositions and methods provided herein have formula III:
Figure imgf000013_0004
or pharmaceutically active derivatives thereof, wherein Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound and Rq has formula:
Figure imgf000013_0003
wherein Rp is a pharmacologically active nucleoside or an analog thereof. In certain embodiments, in the compounds of formula II, R1 and Rlx are each independently -H, or optionally substituted -0(C1- C24)alkyl; wherein at least one of R1 and Rlx is not -H; R2 and R2x are each independently -H, or optionally substituted -0(C1- C7)alkyl;
Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound of formula:
Figure imgf000014_0001
Rp is a pharmacologically active nucleoside or analog thereof;
L is a valence bond or a bifunctional linking molecule of the formula — J- (CRxRy)t-G-, wherein t is an integer from 1 to 24, J and G are each independently -O-, and Rx and Ry are each independently -H, substituted or unsubstituted alkyl, or alkenyl; n is 0 or 1 ; and n1 is O to 3.
In certain embodiments, Rp is
Figure imgf000014_0002
wherein
R3, R4 and R5 are each independently H, hydroxy, halo, azido, C1-6 alkyl, C2-6 alkenyl or C2-6 alkynyl;
B is a purine or pyrimidine base or an analog thereof;
R3x is H, azido, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl;
R4x is H, C1-6 substituted or unsubstituted alkyl, C2-6 substituted or unsubstituted alkenyl or C2-6 substituted or unsubstituted alkynyl; and R3z is H, C1-6 alkyl, hydroxylC1-6alkyl, haloC1-6 alkyl, azidoC1.6 alkyl or OH.
In certain embodiments, Rp has formula:
Figure imgf000015_0001
wherein the variables are as described elsewhere herein. In certain embodiments, R3 is H, azido, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl. In certain embodiments, R3 is H, azido, substituted or unsubstituted C1-6 alkyl. In certain embodiments, R is H or azido. In certain embodiments, R is azido. In certain embodiments, R3 is H. In certain embodiments, R4 and R5 are each independently selected from hydrogen, halo and hydroxyalkyl. In certain embodiments, R4 and R5 are each independently selected from halo and hydroxyalkyl. In certain embodiments, R4 and R5 are each independently selected from fluoro and hydroxymethyl. In certain embodiments, R is selected from fluoro and hydroxymethyl. In certain embodiments, R5 is selected from fluoro and hydroxymethyl.
In certain embodiments, Rp has formula:
Figure imgf000015_0002
wherein the variables are as described elsewhere herein. In certain embodiments, R3x is H, azido, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl; R4x is H, C1-6 substituted or unsubstituted alkyl, C2-6 substituted or unsubstituted alkenyl or C2-6 substituted or unsubstituted alkynyl and other variables are as defined elsewhere herein. In certain embodiments, R3x is H, azido or substituted or unsubstituted C1-6 alkyl. In certain embodiments, R4x is H, C1-6 substituted or unsubstituted alkyl, C2-6 substituted or unsubstituted alkenyl or C2-6 substituted or unsubstituted alkynyl. In certain embodiments, R4x is H, or C1-6 alkyl. In certain embodiments, R x is H, or methyl.
In certain embodiments, Rp has formula:
Figure imgf000016_0001
wherein the variables are as described elsewhere herein.
In certain embodiments, R3z is H, C1-6 alkyl, hydroxyl C1-6 alkyl, halo C1-6 alkyl, azido C1-6 alkyl or OH and the other variables are as defined elsewhere herein. In certain embodiments, R3z is hydrogen, C1-6 alkyl or hydroxyC]-6 alkyl. In certain embodiments, R3z is hydrogen or hydroxymethyl. In certain embodiments, R3z is hydrogen. In certain embodiments, R3z is hydroxymethyl. In certain embodiments, the OH group is protected, for example as an ester or an ether. In certain embodiments, R3z may be in S or R configuration.
In certain embodiments, Rp has formula:
Figure imgf000016_0002
wherein the variables are as described elsewhere herein.
In certain embodiments, R3y is H, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl, or OH and the other variables are as defined elsewhere herein. In certain embodiments, R3y is hydrogen, C1-6 alkyl or hydroxyl C1-6 alkyl. In certain embodiments, R3y is hydrogen or hydroxymethyl. In certain embodiments, R3y may be in S or R configuration.
In certain embodiments, Rp has formula:
Figure imgf000016_0003
wherein the variables are as described elsewhere herein. In certain embodiments, Rp has formula:
Figure imgf000017_0001
In certain embodiments, m = O, 1 or 2. In certain embodiments, m = O or 1. In certain embodiments, m = O. In certain embodiments, m = 1. In certain embodiments, R2 and R2xare H.
In certain embodiments, RL has formula:
Figure imgf000017_0002
wherein R1 and Rlx are as defined elsewhere herein. In certain embodiments, RL has formula:
Figure imgf000017_0003
wherein R1 and Rlx are as defined elsewhere herein. In certain embodiments, RL has formula:
Figure imgf000017_0004
wherein R1 and Rlx are as defined elsewhere herein. In certain embodiments, RL is hexadecyloxypropyl, octadecyloxypropyl, oleyloxyethyl, oleyloxypropyl, octadecyloxyethyl, 15-methylhexadecyloxypropyl or 17-methyloctadecyloxyethy 1.
In certain embodiments, R1 is an alkoxy group having the formula -O-(CH2)t- CH3 wherein t is 0-24. In other embodiments, t is 8, 10, 12, 13, 14, 15, 16, 17, 18, 19 or 20. In other embodiments, t is 13, 14, 15, 16, 17, 18, 19 or 20. In other embodiments, t is 15, 16, 17, 18, 19 or 20. In other embodiments, t is 17, 18, 19 or 20. In other embodiments, t is 15 or 17. In certain embodiments, RL is a substituted or unsubstituted C8-C24 alkyl, substituted or unsubstituted C8-C24 alkenyl having from 1 to 6 double bonds or substituted or unsubstituted C8-C24 alkynyl having from 1 to 6 triple bonds, wherein substituents when present are selected from one or more, in some embodiments, 1 to 4 or 1 or 2 substituents selected from halogen, alkyl, -ORW, -SRW, cycloalkyl or epoxide, where Rw is hydrogen or alkyl and where the alkyl, alkenyl, alkynyl groups may be further substituted or unsubstituted.
In certain embodiments, RL is an alkyl, alkenyl or alkynyl group and contains 8, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 carbon atoms and can be a straight or branched chain moiety. In certain embodiments, RL is a C16-C23 straight or branched chain alkyl or C16-C23 straight or branched chain alkenyl. In other embodiments, RL is a C17-C19 straight or branched chain alkyl or C17-C19 straight or branched chain alkenyl. In other embodiments, RL is C17-alkyl, C18-alkyl or C19 alkyl. In other embodiments, RL is C17-alkenyl, C18-alkenyl or C19 alkenyl. In other embodiments, RL is C17-C22 alkyl. In other embodiments, RL is C17 alkyl, C18 alkyl, C19 alkyl, C20 alkyl, C21 alkyl, or C22 alkyl.
In certain embodiments, RL is substituted with one or more groups selected from lower alkyl and halo. In certain embodiments, RL is substituted with one or more methyl groups. In certain embodiments, RL is substituted with one or more fluoro groups. In certain embodiments, RL is C16-C23 alkyl and is substituted with one or more methyl or fluoro groups. In certain embodiments, the methyl group or the fluoro group substituent is present on the penultimate carbon of the alkyl, alkenyl, or alkynyl chain. In certain embodiments, RL is 7-methyl-octyl, 8-methyl-nonyl, 9-methyl-decyl, 10- methyl-undecyl, 11-methyl-dodecyl, 12-methyl-tridecyl, 13-methyl-tetradecyl, 14- methyl-pentadecyl, 15-methyl-hexadecyl, 16-methyl-heptadecyl, 17-methyl-octadecyl, 18-methyl-nonadecyl, 19-methyl-eicosyl, 20-methyl-heneicosyl, 21-methyl-docosyl, 22-methyl-tricosyl, 7-fluoro-octyl, 8- fluoro-nonyl, 9- fluoro-decyl, 10- fluoro-undecyl, 11- fluoro-dodecyl, 12- fluoro-tridecyl, 13-fluoro-tetradecyl, 14- fluoro-pentadecyl, 15- fluoro -hexadecyl, 16- fluoro-heptadecyl, 17- fluoro-octadecyl, 18- fluoro-nonadecyl, 19- fluoro-eicosyl, 20- fluoro-heneicosyl, 21- fluoro-docosyl or 22- fluoro-tricosyl.
In certain embodiments, B is selected from a natural or non natural purine or pyrimidine base. In certain embodiments, B is
Figure imgf000019_0001
wherein R3a is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, hydroxy, halo, aryl or heteroaryl;
R6 is H or C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl;
R7 is H, hydroxy, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl or NR4R5;
R8 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl; and
R9 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, halo Or NR4R5, where R4 and R5 are each independently H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C3-6 cycloalkyl.
In other embodiments, R3ais H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, hydroxy, halo, aryl or heteroaryl. In other embodiments, R3a is H, halo or C1-6 alkyl. In other embodiments, R3a is H. In other embodiments, R3a is methyl. In other embodiments, R3a is fluoro.
In other embodiments, R4 and R5 are each independently H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C3-6 cycloalkyl. In other embodiments, R4 is H, C1-6 alkyl or C3-6 cycloalkyl. In other embodiments, R4 is H, methyl or cyclopropyl. In other embodiments, R5 is H, C1-6 alkyl or C3-6 cycloalkyl. In other embodiments, R5 is H, methyl or cyclopropyl.
In other embodiments, R6 is H or C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl. In other embodiments, R6 is H or C1-6 alkyl. In other embodiments, R6 is H or methyl. In other embodiments, R6 is H. In other embodiments, R6 is methyl. In other embodiments, R7 is H, hydroxy, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl or NR4R5. In other embodiments, R7 is H, C1-6 alkyl, or NR4R5. In other embodiments, R7 is methyl. In other embodiments, R7 is NR R5. In other embodiments, R7 is NH2.
In other embodiments, R8 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl. In other embodiments, R8 is H.
In other embodiments, R9 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, halo or NR4R5. In other embodiments, R9 is H.
In other embodiments, B is selected from pyrimidin-1-yl, pyrimidin-3-yl, purin- 3-yl, purin-7-yl and purin-9-yl residue. In certain embodiments, B is thymin-1-yl, cytosine-1-yl, adenine-9-yl or guanine-9-yl.
In other embodiments, B is selected from:
Figure imgf000020_0001
In certain embodiments, the alkyl, alkenyl and alkynyl groups in the compounds provided herein are substituted with one or more, in one embodiment, one, two, three or four substituents selected from alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl. Exemplary compounds
In certain embodiments, the compounds herein are phosphate esters of antiviral and anticancer nucleoside phosphonates. In certain embodiments, the compounds provided herein are analogs of (S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]cytosine (HPMPC, cidofovir), (S)-9-[3-hydroxy-2-(phosphonomethoxy)- propyl]adenine ((S)-HPMPA), phosphonomethoxyethyl-guanine (PMEG), phosphonomethoxyethyl-adenine (PMEA) and phosphonomethoxy-propyladenine (PMPA, tenofovir). Many other acyclic nucleoside phosphonates can be modified by conjugation to alkoxyalkyl-phosphate and alkyl-phosphates as described herein. Certain exemplary compounds that can be modified as provided herein are described in the documents listed in Table 1. All the documents listed herein are hereby incorporated by reference in their entirety. Table 1
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Nucleoside analogs with antiviral activity against hepatitis C may be converted to their 5 '-phosphonates or 5 '-methylene phosphonates for use in the compounds provided herein. Some exemplary nucleosides include: 2'-C-methyl adenosine; 2'-C- methyl guanosine; 7-deaza-2'-C-methyl adenosine, 2'-C-methyl cytosine. Other nucleosides which can be used in the compounds provided herein, after conversion fo their 5 '-phosphonates or 5'-methylene-phosphonates are described in the following patents, which are hereby incorporated by reference in their entirety.
Table Ia
Figure imgf000024_0002
Figure imgf000025_0001
Nucleoside analogs with antiviral activity against hepatitis B may be converted to their 5'-phosphonates or 5 '-methylene phosphonates for use herein. Exemplary nucleosides include 3TC, FTC, DAPD, L-FMAU, entecavir, telbivudine and various β- L-2'-deoxycytidine, β-L-2'-deoxyadenine and β-L-2'-deoxythymidine analogs described by Bryant et ah, Anti-viral L-nucleosides specific for Hepatitis B infection, Antimicrob. Agents Chemother., 45:229-235, 2001. In certain embodiments, the nucleosides for use herein include, but are not limited to tenofovir, adefovir, and the 5- phosphono-pent-2-en-l-yl nucleosides, such as 5-phosphono-pent-2-en-l-yl adenine (PPen-A), 5-phosphono-pent-2-en-l-yl cytosine (PPen-C), 5-phosphono-pent-2-en-l-yl guanine (PPen-G), 5-phosphono-pent-2-en-l-yl thymine (PPen-T) and 5-phosphono- pent-2-en-l-yl uracil (PPen-U) and others disclosed in U.S. Application S. No. 60/667,740, which incorporated by reference in its entirety. In certain embodiments, the compounds have anti-hepatitis B activity. Certain other Nucleoside 5'- monophosphates for use herein are described by Prakash et a in J. Med. Chem. 2005, 48, 1199-1210.
In certain embodiments, the compounds provided herein are selected from hexadecyloxypropyl-phospho-(S)-HPMPA (HDP-phospho-(S)-HPMPA), octadecyloxyethyl-phosρho-(S)-HPMPA (ODE-phospho-(S)-HPMPA), oleyloxyethyl- phospho-(S)-HPMPA (OLE-phospho-(S)-HPMPA), oleyloxypropyl-phospho-(S)- HPMPA (OLP- phospho-(S)-HPMPA), 15-methyl-hexadecyloxy-propyl-phospho-(S)- HPMPA, and 17-methyl-octadecyloxy-ethyl- phospho-(S)-HPMPA.
Additional description of the compounds provided herein, including general structural formulas are given Table 2.
Table 2
Figure imgf000027_0001
Figure imgf000027_0002
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
In certain embodiments, the compound is selected from HDP-phospho-(S)-HPMPA
Figure imgf000037_0001
ODE-phospho-(S)-HPMPA
Figure imgf000037_0002
OLE-phospho-(S)-HPMPA
Figure imgf000037_0003
OLP-phospho-(S)-HPMPA
Figure imgf000037_0004
15-methyl-hexadecyloxy-propyl-phospho-(S)-HPMPA
Figure imgf000038_0001
-methyl-octadecyloxy-ethyl-phospho-(S)-HPMPA
Figure imgf000038_0002
-fluoro-hexadecyloxy-propyl-phospho-(S)-HPMPA
Figure imgf000038_0003
-fluoro-octadecyloxy-ethyl-phospho-(S)-HPMPA
Figure imgf000038_0004
-methyl-hexadecyloxy-ethyl-phospho-(S)-HPMPA
Figure imgf000039_0001
HDP-phospho-cidofovir
Figure imgf000039_0002
ODE-phospho-cidofovir
Figure imgf000039_0003
OLE-phospho-cidofovir
Figure imgf000039_0004
OLP-phospho-cidofovir
Figure imgf000039_0005
HDP-phospho-PMEG
Figure imgf000040_0001
ODE-phospho-PMEG
Figure imgf000040_0002
HDP-phospho-PME-DAP
Figure imgf000040_0003
HDP-phospho-PME-N6cPr-DAP
Figure imgf000040_0004
OLE-phospho-PME-N6cPr-DAP
Figure imgf000040_0005
HDP- phospho-PPMG
Figure imgf000041_0001
HDP-phospho-PPM-DAP
Figure imgf000041_0002
HDP-phospho-PPM-N6cPr-DAP
Figure imgf000041_0003
HDP-phospho- PME-5FU
Figure imgf000041_0004
HDP-phospho-PME-5FC
Figure imgf000042_0001
HDP-phospho-HPMP-5FC
Figure imgf000042_0002
HDP-phospho-HPMP-5FU
Figure imgf000042_0003
HDP-phospho-Phosphonomethoxy-STC
Figure imgf000042_0004
HDP-phospho-Phosphonomethoxy-2 ' -C-methyl ribo-guanine
Figure imgf000042_0005
HDP-phospho-Phosphonomethoxy-1 -methyl cytidine
Figure imgf000043_0001
HDP-phospho-PM-2 -O-methyl cytidine
Figure imgf000043_0002
ODE-phospho-PM-2 -C-methyl adenosine
Figure imgf000043_0003
C. Preparation of the Compounds
Exemplary methods for the preparation of nucleoside phosphonate- phosphate ester conjugates provided herein are depicted in Schemes 1 and 2. Scheme 1 outlines the synthesis of alkoxyalkyl phospho-morpholidates 3 and 4. Detailed synthesis is described in detail in example 1. In scheme 2, (S)-l-(3-hydroxy-2-phosphonomethoxypropyl)cytosine (HPMPC) is treated with dimethoxytritylchloride in DMSO by the method of Otmar et. al, An Alternative Synthesis of HPMPC and HPMPA diphosphoryl derivatives, Collection Symposium Series 2 (Chemistry of Nucleic Acid Components), 252-54, 1999, to give the intermediate 5 that is condensed with hexadecyloxypropyl-phosphate morpholidate ( 3) or octadecyloxyethyl-phosphate morpholidate ( 4) in pyridine, tributylamine and catalytic acetic acid at room temperature. Finally, hydrolysis with TFA in CHCl3 gives compounds 6, hexadecyloxypropyl-phospho-cidofovir (HDP-phospho-ΗPMPC), and 7, octadecyloxyethyl- phospho-cidofovir (ODE-phospho-HPMPC). Scheme Il
Figure imgf000044_0001
i) Pyridine, acetic acid, ii) HCCI3:TFA
Compojμnd 9 is prepared from the condensation of compound 8 9-(2- phosphonylmethoxyethyl)adenine (PMEA) and compound 3 in pyridine and acetic acid as catalyst. The synthesis of compound 11 is achieved by the reaction between 3'-azido-3'- deoxythymidine and diethyl[p-toluenesulfonyl-oxy]methylphosphonate in the presence of NaH, followed by hydrolysis with TMSBr to obtain the phosphonate intermediate 10 which is finally reacted with 3 to give the hexadecyloxypropyl-phospho conjugate 11.
D. Formulation of pharmaceutical compositions
The pharmaceutical compositions provided herein contain therapeutically effective amounts of one or more of the compounds provided herein that are useful in the prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders associated with viral infections and inappropriate cell proliferation and a pharmaceutically acceptable carrier. Pharmaceutical carriers suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
In addition, the compounds may be formulated as the sole pharmaceutically active ingredient in the composition or may be combined with other active ingredients.
The compositions contain one or more compounds provided herein. The compounds are, in one embodiment, formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers. In one embodiment, the compounds described above are formulated into pharmaceutical compositions using techniques and procedures well known in the art ((see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Seventh Edition 1999).
In the compositions, effective concentrations of one or more compounds or pharmaceutically acceptable derivatives thereof is (are) mixed with a suitable pharmaceutical carrier. The compounds may be derivatized as the corresponding salts, esters, enol ethers or esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs prior to formulation, as described above. The concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration, that treats, prevents, or ameliorates one or more of the symptoms of diseases or disorders associated with associated with viral infections or inappropriate cell proliferation. In one embodiment, the compositions are formulated for single dosage administration. To formulate a composition, the weight fraction of a compound is dissolved, suspended, dispersed or otherwise mixed in a selected carrier at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
The active compound is included in the pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful effect in the absence of undesirable side effects on the patient treated. The therapeutically effective concentration may be determined empirically by testing the compounds in in vitro and in vivo systems well known to those of skill in the art and then extrapolated therefrom for dosages for humans.
The concentration of active compound in the pharmaceutical composition will depend on absorption, inactivation and excretion rates of the active compound, the physicochemical characteristics of the compound, the dosage schedule, and amount administered as well as other factors known to those of skill in the art. For example, the amount that is delivered is sufficient to ameliorate one or more of the symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation, as described herein.
In one embodiment, a therapeutically effective dosage should produce a serum concentration of active ingredient of from about 0.1 ng/ml to about 50- 100 μg/ml. The pharmaceutical compositions, in another embodiment, should provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day. Pharmaceutical dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500mg, 1000 mg or 2000 mg, and in one embodiment from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
In instances in which the compounds exhibit insufficient solubility, methods for solubilizing compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEN®, or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions. Upon mixing or addition of the compound(s), the resulting mixture may be a solution, suspension, emulsion or the like. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.
The pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds or pharmaceutically acceptable derivatives thereof. The pharmaceutically therapeutically active compounds and derivatives thereof are, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms. Unit-dose forms as used herein refer to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form.
Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975.
Dosage forms or compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%.
In certain embodiments, the compositions are lactose-free compositions containing excipients that are well known in the art and are listed, for example, in the U.S.
Pharmacopeia (USP) 25-NF20 (2002). In general, lactose-free compositions contains active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts. Particular lactose-free dosage forms contain active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate. Further provided are anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water (e.g., 5%) is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf- life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations. Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are generally packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs. 1. Compositions for oral administration
Oral pharmaceutical dosage forms are either solid, gel or liquid. The solid dosage forms are tablets, capsules, granules, and bulk powders. Types of oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated. Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art. a. Solid compositions for oral administration In certain embodiments, the formulations are solid dosage forms, in one embodiment, capsules or tablets. The tablets, pills, capsules, troches and the like can contain one or more of the following ingredients, or compounds of a similar nature: a binder; a lubricant; a diluent; a glidant; a disintegrating agent; a coloring agent; a sweetening agent; a flavoring agent; a wetting agent; an emetic coating; and a film coating. Examples of binders include microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, molasses, polvinylpyrrolidine, povidone, crospovidones, sucrose and starch paste. Lubricants include talc, starch, magnesium or calcium stearate, lycopodium and stearic acid. Diluents include, for example, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate. Glidants include, but are not limited to, colloidal silicon dioxide. Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose. Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate. Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors. Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene laural ether. Emetic-coatings include fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates. Film coatings include hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
The compound, or pharmaceutically acceptable derivative thereof, could be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.
When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents. The compounds can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
The active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics. The active ingredient is a compound or pharmaceutically acceptable derivative thereof as described herein. Higher concentrations, up to about 98% by weight of the active ingredient may be included.
In all embodiments, tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient. Thus, for example, they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate. b. Liquid compositions for oral administration
Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Aqueous solutions include, for example, elixirs and syrups. Emulsions are either oil-in- water or water-in-oil. Elixirs are clear, sweetened, hydroalcoholic preparations. Pharmaceutically acceptable carriers used in elixirs include solvents. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative. An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid. Pharmaceutically acceptable carriers used in emulsions are non-aqueous liquids, emulsifying agents and preservatives. Suspensions use pharmaceutically acceptable suspending agents and preservatives. Pharmaceutically acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents. Pharmaceutically acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic acids and a source of carbon dioxide. Coloring and flavoring agents are used in all of the above dosage forms. Solvents include glycerin, sorbitol, ethyl alcohol and syrup. Examples of preservatives include glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil. Examples of emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate. Suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia. Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as saccharin. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate. Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof. Flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds which produce a pleasant taste sensation.
For a solid dosage form, the solution or suspension, in for example, propylene carbonate, vegetable oils or triglycerides, is in one embodiment encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Patent Nos. 4,328,245; 4,409,239; and 4,410,545. For a liquid dosage form, the solution, e.g., for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be easily measured for administration.
Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Patent Nos. RE28,819 and 4,358,603. Briefly, such formulations include, but are not limited to, those containing a compound provided herein, a dialkylated mono- or poly-alkylene glycol, including, but not limited to, 1,2- dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
Other formulations include, but are not limited to, aqueous alcoholic solutions including a pharmaceutically acceptable acetal. Alcohols used in these formulations are any pharmaceutically acceptable water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol. Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes such as acetaldehyde diethyl acetal. 2. Injectables, solutions and emulsions
Parenteral administration, in one embodiment characterized by injection, either subcutaneously, intramuscularly or intravenously is also contemplated herein. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. The injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
Implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Patent No. 3,710,795) is also contemplated herein. Briefly, a compound provided herein is dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The compound diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject.
Parenteral administration of the compositions includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions. The solutions may be either aqueous or nonaqueous. If administered intravenously, suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
Examples of aqueous vehicles include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection. Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil. Antimicrobial agents in bacteriostatic or fungistatic concentrations must be added to parenteral preparations packaged in multiple-dose containers which include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride. Isotonic agents include sodium chloride and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (TWEEN® 80). A sequestering or chelating agent of metal ions include EDTA. Pharmaceutical carriers also include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
The concentration of the pharmaceutically active compound is adjusted so that an injection provides an effective amount to produce the desired pharmacological effect. The exact dose depends on the age, weight and condition of the patient or animal as is known in the art.
The unit-dose parenteral preparations are packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration must be sterile, as is known and practiced in the art.
Illustratively, intravenous or intraarterial infusion of a sterile aqueous solution containing an active compound is an effective mode of administration. Another embodiment is a sterile aqueous or oily solution or suspension containing an active material injected as necessary to produce the desired pharmacological effect. Injectables are designed for local and systemic administration. In one embodiment, a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1% w/w up to about 90% w/w or more, in certain embodiments more than 1% w/w of the active compound to the treated tissue(s).
The compound may be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the condition and may be empirically determined. 3. Sustained Release Dosage Form
Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; 6,699,500 and 6,740,634, each of which is incorporated herein by reference. Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled- release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
All controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts. Ideally, the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance. In addition, controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
Most controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
In certain embodiments, the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al, Surgery 88:507 (1980); Saudek et al, N. Engl. J. Med. 321:57 '4 (1989). In another embodiment, polymeric materials can be used. In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical
Applications of Controlled Release, vol. 2, pp. 115-138 (1984). In some embodiments, a controlled release device is introduced into a subject in proximity of the site of inappropriate immune activation or a tumor. Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990). The active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene- vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The active ingredient then diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active ingredient contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the needs of the subject.
4. Lyophilized powders Of interest herein are also lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. They may also be reconstituted and formulated as solids or gels.
The sterile, lyophilized powder is prepared by dissolving a compound provided herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent. The solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder. Excipients that may be used include, but are not limited to, an antioxidant, a buffer and a bulking agent. In some embodiments, the excipient is selected from dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose and other suitable agent. The solvent may contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, at about neutral pH. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. In one embodiment, the resulting solution will be apportioned into vials for lyophilization. Each vial will contain a single dosage or multiple dosages of the compound. The lyophilized powder can be stored under appropriate conditions, such as at about 4 0C to room temperature.
Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration. For reconstitution, the lyophilized powder is added to sterile water or other suitable carrier. The precise amount depends upon the selected compound. Such amount can be empirically determined.
5. Topical administration
Topical mixtures are prepared as described for the local and systemic administration. The resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
The compounds or pharmaceutically acceptable derivatives thereof may be formulated as aerosols for topical application, such as by inhalation (see, e.g., U.S. Patent Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment of inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation will, in one embodiment, have diameters of less than 50 microns, in one embodiment less than 10 microns.
The compounds may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered. For nasal administration, the preparation may contain an esterified phosphonate compound dissolved or suspended in a liquid carrier, in particular, an aqueous carrier, for aerosol application. The carrier may contain solubilizing agents such as propylene glycol, surfactants, absorption enhancers such as lecithin or cyclodextrin, or preservatives. These solutions, particularly those intended for ophthalmic use, may be formulated as 0.01% - 10% isotonic solutions, pH about 5-7, with appropriate salts.
6. Compositions for other routes of administration
Other routes of administration, such as transdermal patches, including iontophoretic and electrophoretic devices, and rectal administration, are also contemplated herein.
Transdermal patches, including iotophoretic and electrophoretic devices, are well known to those of skill in the art. For example, such patches are disclosed in U.S. Patent Nos. 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010715, 5,985,317, 5,983,134, 5,948,433, and 5,860,957. For example, pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum which melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point. Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The weight of a rectal suppository, in one embodiment, is about 2 to 3 gm. Tablets and capsules for rectal administration are manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
7. Targeted Formulations
The compounds provided herein, or pharmaceutically acceptable derivatives thereof, may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated. Many such targeting methods are well known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Patent Nos. 6,316,652, 6,274,552, 6,271,359, 6,253,872, 6,139,865, 6,131,570, 6,120,751, 6,071,495, 6,060,082, 6,048,736, 6,039,975, 6,004,534, 5,985,307, 5,972,366, 5,900,252, 5,840,674, 5,759,542 and 5,709,874.
In one embodiment, liposomal suspensions, including tissue-targeted liposomes, such as tumor-targeted liposomes, may also be suitable as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art. For example, liposome formulations may be prepared as described in U.S. Patent No. 4,522,811. Briefly, liposomes such as multilamellar vesicles (MLVs) may be formed by drying down egg phosphatidyl choline and brain phosphatidyl serine (7:3 molar ratio) on the inside of a flask. A solution of a compound provided herein in phosphate buffered saline lacking divalent cations (PBS) is added and the flask shaken until the lipid film is dispersed. The resulting vesicles are washed to remove unencapsulated compound, pelleted by centrifugation, and then resuspended in PBS.
8. Articles of manufacture The compounds or pharmaceutically acceptable derivatives may be packaged as articles of manufacture containing packaging material, a compound or pharmaceutically acceptable derivative thereof provided herein, which is effective for for treatment, prevention or amelioration of one or more symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation, within the packaging material, and a label that indicates that the compound or composition, or pharmaceutically acceptable derivative thereof, is used for the treatment, prevention or amelioration of one or more symptoms of diseases or disorders associated with viral infections or inappropriate cell proliferation.
The articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging pharmaceutical products are well known to those of skill in the art. See, e.g., U.S. Patent Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. A wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease or disorder associated with viral infections or inappropriate cell proliferation. E. Dosages
In human therapeutics, the physician will determine the dosage regimen that is most appropriate according to a preventive or curative treatment and according to the age, weight, stage of the disease and other factors specific to the subject to be treated. The pharmaceutical compositions, in another embodiment, should provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day. Pharmaceutical dosage unit forms are prepared, e.g., to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500mg, 1000 mg or 2000 mg, and in one embodiment from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
The amount of active ingredient in the formulations provided herein, which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof, will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered. The frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
Exemplary doses of a formulation include milligram or microgram amounts of the active compound per kilogram of subject or sample weight (e.g., from about 1 micrograms per kilogram to about 50 milligrams per kilogram, from about 10 micrograms per kilogram to about 30 milligrams per kilogram, from about 100 micrograms per kilogram to about 10 milligrams per kilogram, or from about 100 microgram per kilogram to about 5 milligrams per kilogram).
It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with subject response.
Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to prevent, manage, treat or ameliorate such disorders, but insufficient to cause, or sufficient to reduce, adverse effects associated with the composition provided herein are also encompassed by the above described dosage amounts and dose frequency schedules. Further, when a subject is administered multiple dosages of a composition provided herein, not all of the dosages need be the same. For example, the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the composition or it may be decreased to reduce one or more side effects that a particular subject is experiencing. In certain embodiments, administration of the same formulation provided herein may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months. F. Evaluation of the activity of the compounds
The activity of the compounds as antivirals can be measured in standard assays known in the art. Exemplary assays include, but are not limited to, plaque reduction assay in HFF cells, DNA reduction assay in MRC-5 cells, p24 reduction assay in MT-2 cells, CPE assay in HFF cells and EBV Elisa assay in Daudi cells.
In Table 3, EC50 for the compounds provided herein and tested against various viruses in vitro are provided.
G. Methods of use of the compounds and compositions
Methods of treating, preventing, or ameliorating one or more symptoms of diseases associated with viral infections or inappropriate cell proliferation using the compounds and compositions are provided. In practicing the methods, effective amounts of the compounds or compositions containing therapeutically effective concentrations of the compounds are administered. In certain embodiments, the methods provided herein are for the preventing, or ameliorating one or more symptoms of diseases associated with viral infections, including, but not limited to influenza; hepatitis B and C virus; cytomegalovirus (CMV); herpes infections, such as those caused by Varicella zoster virus, Herpes simplex virus types 1 & 2, Epstein-Barr virus, Herpes type 6 (HHV-6) and type 8 (HHV-8); Varicella zoster virus infections such as shingles or chicken pox; Epstein Barr virus infections, including, but not limited to infectious mononucleosis/glandular; retroviral infections including, but not limited to SIV, HIV-I and HIV-2; ebola virus; adenovirus and papilloma virus.
In further embodiments, the methods provided herein are for treating, preventing, treating, or ameliorating one or more symptoms of diseases associated with viral infections caused by orthopox viruses, such as variola major and minor, vaccinia, smallpox, cowpox, camelpox, and monkeypox. In certain embodiments, the disease is drug resistant hepatitis B. In certain embodiments, the methods provided herein are for treating, preventing, or ameliorating one or more symptoms of diseases associated with cell proliferation, including, but not limited to cancers. Examples of cancers include, but are not limited to, lung cancer, head and neck squamous cancers, colorectal cancer, prostate cancer, breast cancer, acute lymphocytic leukemia, adult acute myeloid leukemia, adult non Hodgkin's lymphoma, brain tumors, cervical cancers, childhood cancers, childhood sarcoma, chronic lymphocytic leukemia, chronic myeloid leukemia, esophageal cancer, hairy cell leukemia, kidney cancer, liver cancer, multiple myeloma, neuroblastoma, oral cancer, pancreatic cancer, primary central nervous system lymphoma, and skin cancer. H. Combination Therapy
The compounds and compositions provided herein may also be used in combination with one or more other active ingredients. In certain embodiments, the compounds may be administered in combination, or sequentially, with another therapeutic agent. Such other therapeutic agents include those known for treatment, prevention, or amelioration of one or more symptoms associated with viral infections or inappropriate cell proliferation. Such therapeutic agents include, but are not limited to, antiviral agents and anti-neoplastic agents. Recently, it has been demonstrated that the efficacy of a drug against HIV infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principle drug. Alternatively, the pharmacokinetics, biodistribution, or other parameter of the drug can be altered by such combination or alternation therapy.
In certain embodiments, provided herein are methods of treatment, prevention or amelioration that encompass administration of a second agent effective for the treatment, prevention or amelioration of viral infection, such as HIV and/or HCV infection. The second agent can be any agent known to those of skill in the art to be effective for the treatment, prevention or amelioration of viral infections, such as the HIV and/or HCV infection. The second agent can be a second agent presently known to those of skill in the art, or the second agent can be second agent later developed for the treatment, prevention or amelioration of viral infections. In certain embodiments, the second agent is presently approved for the treatment or prevention of HIV and/or HCV.
In certain embodiments, a compound provided herein is administered in combination with one second agent. In further embodiments, a second agent is administered in combination with two second agents. In still further embodiments, a second agent is administered in combination with two or more second agents.
In one embodiment, the second antiviral agent for the treatment of HIV can be a reverse transcriptase inhibitor (a "RTI"), which can be either a synthetic nucleoside (a "NRTI") or a non-nucleoside compound (a "NNRTI"). In an alternative embodiment, in the case of HIV, the second (or third) antiviral agent can be a protease inhibitor. In other embodiments, the second (or third) compound can be a pyrophosphate analog, or a fusion binding inhibitor. In some embodiments, compounds for combination or alternation therapy for the treatment of HBV include, but are not limited to 3TC, FTC, L-FMAU, interferon, β-D- dioxolanyl-guanine (DXG), β-D-dioxolanyl-2,6-diaminopurine (DAPD), and β-D-dioxolanyl- 6-chloropurine (ACP), famciclovir, penciclovir, BMS-200475, bis pom PMEA (adefovir, dipivoxil); lobucavir, ganciclovir, and ribavarin.
In another embodiment, examples of antiviral agents that can be used in combination or alternation with the compounds disclosed herein for HIV therapy include cis-2- hydroxymethyl-5-(5-fluorocytosin-l-yl)-l,3-oxathiolane (FTC); the (-;)-enantiomer of 2- hydroxymethyl-5-(cytosin-l-yl)-l,3-oxathiolane (3TC); carbovir, acyclovir, foscarnet, interferon, AZT, DDI, DDC, D4T, CS-87 (3'-azido-2',3'-dideoxy-uridine), and β-D-dioxolane nucleosides such as β-D-dioxolanyl-guanine (DXG), β-D-dioxolanyl-2,6-diaminopurine (DAPD), and β-D-dioxolanyl-6-chloropurine (ACP), MKC442 (6-benzyl-l-(ethoxymethyl)- 5-isopropyl uracil.
The protease inhibitors include crixivan, nelfinavir, ritonavir, saquinavir, DMP-266 and DMP-450.
Further compounds that can be administered in combination or alternation with any of the compounds provided herein include (lS,4R)-4-[2-amino-6-cyclopropyl-amino)-9H-purin- 9-yl]-2-cyclopentene-l -methanol succinate ("1592", a carbovir analog); 3TC; -β-L-2',3'- dideoxy-3'-thiacytidine; a-APA R18893: a-nitro-anilino-phenylacetamide; A-77003; C2 symmetry -based protease inhibitor; A-75925: C2 symmetry-based protease inhibitor; AAP- BHAP: bisheteroarylpiperazine analog; ABT-538: C2 symmetry-based protease inhibitor; AzddU:3'-azido-2',3'-dideoxyuridine; AZT: 3'-azido-3'-deoxythymidine; AZT-p-ddI:3'- azido-3'-deoxythymidilyl-(5',5')-2',3'-dideoxyinosinic acid; BHAP: bisheteroarylpiperazine; BILA 1906: N-{lS-[[[3-[2S-{(l,l-dimethylethyl)amino]carbonyl}-4R-]3- pyridinylmethyl)thio]-l-piperidinyl]-2R-hydroxy-lS-(phenylmethyl)propyl]amino]carbonyl]- 2-methylpropyl}-2-quinolinecarboxamide; BILA 2185: N-(l,l-dimethylethyl)-l-[2S-[[2-2,6- dimethyphenoxy)-l-oxoethyl]amino]-2R-hydroxy-4-phenylbutyl]4R-pyridinylthio)-2- piperidine-carboxamide; BM+51.0836: thiazolo-isoindolinone derivative; BMS 186,318: aminodiol derivative HIV-I protease inhibitor; d4API: 9-[2,5-dihydro-5- (phosphonomethoxy)-2-furanyl]adenine; d4C: 2',3'-didehydro-2',3'-dideoxycytidine; d4T: 2',3'-didehydro-3'-deoxythymidine; ddC; 2',3'-dideoxycytidine; ddl: 2',3'-dideoxyinosine; DMP-266: a l,4-dihydro-2H-3,l-benzoxazin-2-one; DMP-450: {[4R-(4-a,5-a,6-b,7-b)]- hexabydro-5,6-bis(hydroxy)- 1 ,3-bis(3-amino)phenyl]methyl)-4,7-bis(phenylmethyl)-2H- 1,3- diazepin-2-one}-bismesylate; DXG:(-;)-β-D-dioxolane-guanosine; EBU-dM:5-ethyl-l- ethoxymethyl-6-(3,5-dimethylbenzyl)uracil; E-EBU: 5-ethyl-l-ethoxymethyl-6-benzyluracil; DS: dextran sulfate; E-EPSeU:l-(ethoxymethyl)-(6-phenylselenyl)-5-ethyluracil; E-EPU: 1- (ethoxymethyl)-(6-phenyl-thio)-5-ethyluracil; FTC:β-2',3'-dideoxy-5-fluoro-3'-thiacytidine (Triangle); HBY097:S~4-isopropoxycarbonyl-6-methoxy-3-(methylthio-methyl)-3,4- dihydroquinoxalin-2(lH)-thione; HEPT: 1 -[(2-hydroxyethoxy)methyl]-6- (phenylthio)thymine; HIV-l:human immunodeficiency virus type 1; JM2763: 1,1'-(1,3- propanediy l)-bis- 1,4,8,11 -tetraazacy clotetradecane ; JM3100:1, 1 '- [ 1 ,4-pheny lenebis- (methylene)]-bis- 1,4,8,11-tetraazacyclotetradecane; KNI-272: (2S,3S)-3-amino-2-hydroxy-4- phenylbutyric acid-containing tripeptide; L-697,593;5-ethyl-6-methyl-3-(2-phthalimido- ethyl)pyridin-2(lH)-one; L-735,524:hydroxy-aminopentane amide HIV-I protease inhibitor; L-697,661: 3-{[(-4,7-dichloro-l,3-benzoxazol-2-yl)methyl]amino}-5-ethyl-6-methylpyridin - 2(1 H)-one; L-FDDC: (-;)-β-L-5-fluoro-2',3'-dideoxycytidine; L-FDOC:(-;)-β-L-5-fluoro- dioxolane cytosine; MKC442:6-benzyl-l-ethoxymethyl-5-isopropyluracil (I-EBU);
Nevirapine: 11-cyclopropy 1-5,1 l-dihydro-4-methyl-6H-dipyridol[3,2-b:2',3'-e]diazepin-6- one; NSC648400:l-benzyloxymethyl-5-ethyl-6-(alpha-pyridylthio)uracil (E-BPTU); P9941: [2-pyridylacetyl-IlePheAla-y(CHOH)]2; PFA: phosphonoformate; PMEA: 9-(2- phosphonylmethoxyethyl)adenine; PMPA: (R)-9-(2-phosphonyl-methoxyproρyl)adenine; Ro 31-8959: hydroxyethylamine derivative HIV-I protease inhibitor; RPI-312: peptidyl protease inhibitor, l-[(3s)-3-(n-alpha-benzyloxycarbonyl)-l-asparginyl)-amino-2-hydroxy-4- phenylbutyryl]-n-tert-butyl-l -proline amide; 2720: 6-chloro-3,3-dimethyl-4- (isopropenyloxycarbonyl)-3,4-dihydro-quinoxalin-2(lH)thione; SC-52151: hydroxyethylurea isostere protease inhibitor; SC-55389A: hydroxyethyl-urea isostere protease inhibitor; TIBO R82150: (+)-(5S)-4,5,6,7-tetrahydro-5-methyl-6-(3-methyl-2-butenyl)imidazo[4,5,l-jk][l,4]- benzodiazepin-2(lH)-thione; TIBO 82913: (+)-(5S)-4,5,6,7,-tetrahydro-9-chloro-5-methyl-6- (3-methyl-2-butenyl)imidazo[4,5,ljk]-[l,4]benzo-diazepin-2(lH)-thione; TSAO-m3T:[2',5'- bis-O-(tert-butyldimethylsilyl)-3'-spiro-5'-(4'-amino-l',2'-oxathiole-2',2'-dioxide)]-b-D- pentofiaranosyl-N3-methylthymine; U90152: 1 -[3-[(I -methylethyl)-amino]-2-pyridinyl]-4- [[5-[(methylsulphonyl)-amino]-lH -indol-2yl]carbonyl]-piperazine; UC: thiocarboxanilide derivatives (Uniroyal); UC-78 l=N-[4-chloro-3-(3-methyl-2-butenyloxy)phenyl]-2-methyl-3- furancarbothioamide; UC-82=N-[4-chloro-3-(3-methyl-2-butenyloxy)phenyl]-2-methyl-3- thiophenecarbothioamide; VB 11,328: hydroxyethyl-sulphonamide protease inhibitor; VX- 478:hydroxyethylsulphonamide protease inhibitor; XM 323: cyclic urea protease inhibitor.
In certain embodiments, suitable second agents include small-molecule, orally bioavailable inhibitors of the HCV enzymes, nucleic-acid-based agents that attack viral RNA, agents that can modulate the host immune response. Exemplary second agents include: (i) current approved therapies (peg-interferon plus ribavirin), (ii) HCV-enzyme targeted compounds, (iii) viral-genome-targeted therapies (e.g., RNA interference or RNAi), and (iv) immunomodulatory agents such as ribavirin, interferon (INF) and Toll-receptor agonists.
In certain embodiments, the second agent is a modulator of the NS3-4A protease. The NS3-4A protease is a heterodimeric protease, comprising the amino-terminal domain of the NS3 protein and the small NS4A cofactor. Its activity is essential for the generation of components of the viral RNA replication complex.
One useful NS3-4A protease inhibitor is BILN 2061 (Ciluprevir; Boehringer Ingelheim), a macrocyclic mimic of peptide product inhibitors. Although clinical trials with BILN 2061 were halted (preclinical cardiotoxicity), it was the first NS3 inhibitor to be tested in humans. See Lamarre et al, 2003, Nature 426:186-189, the contents of which are hereby incorporated by reference in their entirety.
Another useful NS3-4A protease inhibitor is VX-950 (Vertex/Mitsubishi), a protease- cleavage-product-derived peptidomimetic inhibitor of the NS3-4A protease. It is believed to be stabilized into the enzyme's active site through a ketoamide. See, e.g., Lin et al, 2005, J Biol Chem. Manuscript M506462200 (epublication); Summa, 2005, Curr Opin Investig Drugs. 6:831-7, the contents of which are hereby incorporated by reference in their entireties.
In certain embodiments, the second agent is a modulator of the HCV NS5B The RNA-dependent RNA polymerase (RdRp). Contained within the NS5B protein, RdRp synthesizes RNA using an RNA template. This biochemical activity is not present in mammalian cells.
One useful modulator of RdRp is NM283 (Valopicitabine; Idenix/Novartis). NM283, is an oral prodrug (valine ester) of NMl 07 (2 -C-methyl-cytidine) in phase II trials for the treatment or prevention of HCV infection. See, e.g., U.S. Patent Application Publication No. 20040077587, the contents of which are hereby incorporated by reference in their entirety.
Other useful modulators of RdRp include 7-deaza nucleoside analogs. For instance, 7-Deaza-2'-C-methyl-adenosine is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties. Olsen et al., 2004, Antimicrob. Agents Chemother. 48:3944-3953, the contents of which are hereby incorporated by reference in their entirety.
In further embodiments, the second agent is a non-nucleoside modulator of NS5B. At least three different classes of non-nucleoside inhibitors (NNI) ofNS5B inhibitors are being evaluated in the clinic.
Useful non-nucleoside modulators of NS5B include JTK-003 and JTK-009. JTK-003 has been advanced to phase II. Useful non-nucleoside modulators of NS5B include the 6,5- fused heterocyclic compounds based on a benzimidazole or indole core. See, e.g., Hashimoto et al, WO 00147883, the contents of which are hereby incorporated by reference in their entirety.
Further useful polymerase NNIs include R803 (Rigel) and HCV-371, HCV-086 and HCV-796 (ViroPharma/Wyeth). Additional useful NNIs include thiophene derivatives that are reversible allosteric inhibitors of the NS5B polymerase and bind to a site that is close to, but distinct from, the site occupied by benzimidazole-based inhibitors. See, e.g., Biswal, et al, 2005, J. Biol. Chem. 280, 18202-18210 (2005).
Further useful NNIs for the methods provided herein include benzothiadiazines, such as benzo-l,2,4-thiadiazines. Derivatives of benzo-l,2,4-thiadiazine have been shown to be highly selective inhibitors of the HCV RNA polymerase. Dhanak, et al, 2002, J. Biol. Chem. 277:38322-38327, the contents of which are hereby incorporated by reference in their entirety.
Further useful NNIs for the methods provided herein, and their mechanisms, are described in LaPlante et al, 2004 Angew Chem. Int. Ed. Engl. 43:4306-4311; Tomei et al, 2003, J. Virol. 77:13225-13231; Di Marco et al, 2005, J. Biol Chem. 280:29765-70; Lu, H., WO 2005/000308; Chan et al, 2004, Bioorg. Med. Chem. Lett. 14:797-800; Chan et al, 2004, Bioorg. Med. Chem. Lett. 14:793-796; Wang et al, 2003, J. Biol Chem.
278:9489-9495; Love, et al, 2003, J. Virol. 77:7575-7581; Gu et al, 2003, J. Biol. Chem. 278:16602-16607; Tomei et al, 2004, J. Virol 78:938-946; and Nguyen et al, 2003, Antimicrob. Agents Chemother. 47:3525-3530; the contents of each are hereby incorporated by reference in their entireties. In a further embodiment, the second agent is an agent that is capable of interfering with HCV RNA such as small inhibitory RNA (siRNA) or a short hairpin RNA (shRNA) directed to an HCV polynucleotide. In tissue culture, siRNA and vector-encoded short hairpin RNA shRNA directed against the viral genome, effectively block the replication of HCV replicons. See, e.g., Randall et al., 2003, Proc. Natl Acad. Sci. USA 100:235-240, the contents of which are hereby incorporated by reference in their entirety.
In a further embodiment, the second agent is an agent that modulates the subject's immune response. For instance, in certain embodiments, the second agent can be a presently approved therapy for HCV infection such as an interferon (IFN), a pegylated IFN, an IFN plus ribavirin or a pegylated IFN plus ribavirin. In certain embodiments, interferons include IFNα, IFNα2a and IFNα2b, and particularly pegylated IFNα2a (PEGASYS®) or pegylated IFNα2b (PEG-INTRON®).
In a further embodiment, the second agent is a modulator of a Toll-like receptor (TLR). It is believed that TLRs are targets for stimulating innate anti-viral response.
Suitable TLRs include, bur are not limited to, TLR3, TLR7, TLR8 and TLR9. It is believed that toll-like receptors sense the presence of invading microorganisms such as bacteria, viruses and parasites. They are expressed by immune cells, including macrophages, monocytes, dendritic cells and B cells. Stimulation or activation of TLRs can initiate acute inflammatory responses by induction of antimicrobial genes and pro-inflammatory cytokines and chemokines.
In certain embodiments, the second agent is a polynucleotide comprising a CpG motif. Synthetic oligonucleotides containing unmethylated CpG motifs are potent agonists of TLR-9. Stimulation of dendritic cells with these oligonucleotides results in the production of tumour necrosis factor-alpha, interleukin-12 and IFN-alpha. TLR-9 ligands are also potent stimulators of B-cell proliferation and antibody secretion. One useful CpG-containing oligonucleotide is CPG-10101 (Actilon; Coley Pharmaceutical Group) which has been evaluated in the clinic.
Another useful modulator of a TLR is ANA975 (Anadys). ANA975 is believed to act through TLR-7, and is known to elicit a powerful anti-viral response via induction and the release of inflammatory cytokines such as IFN-alpha.
In another embodiment, the second agent is Celgosivir. Celgosivir is an alpha- glucosidase I inhibitor and acts through host-directed glycosylation. In preclinical studies, celgosivir has demonstrated strong synergy with IFNα plus ribavirin. See, e.g., Whitby et al., 2004, Antivir Chem Chemother. 15(3):141-51. Celgosivir is currently being evaluated in a Phase II monotherapy study in chronic HCV patients in Canada.
Further immunomodulatory agents, and their mechanisms or targets, are described in Schetter& Vollmer, 2004, Curr. Opin. DrugDiscov. Dev. 7:204-210; Takeda et al, 2003, Annu. Rev. Immunol. 21:335-376; Lee et al, 2003, Proc. Natl Acad. Sci. USA
100:6646-6651 ; Hosmans et al. , 2004, Hepatology 40 (Suppl. 1), 282A; and U.S. Patent No.
6,924,271; the contents of each are hereby incorporated by reference in their entireties.
In certain embodiments, the compounds provided herein may be administered in combination with one or more anti-cancer agents. Anti-cancer agents for use in combination with the instant compounds include, but are not limited to, an antifolate, a 5-fluoropyrimidine (including 5-fluorouracil), a cytidine analogue such as β-L-l,3-dioxolanyl cytidine or β-L- 1,3-dioxolanyl 5-fluorocytidine, antimetabolites (including purine antimetabolites, cytarabine, fudarabine, floxuridine, 6-mercaptopurine, methotrexate, and 6-thioguanine), hydroxyurea, mitotic inhibitors (including CPT-11, Etoposide (VP-21), taxol, and vinca alkaloids such as vincristine and vinblastine, an alkylating agent (including but not limited to busulfan, chlorambucil, cyclophosphamide, ifofamide, mechlorethamine, melphalan, and thiotepa), nonclassical alkylating agents, platinum containing compounds, bleomycin, an anti-tumor antibiotic, an anthracycline such as doxorubicin and dannomycin, an anthracenedione, topoisomerase II inhibitors, hormonal agents (including but not limited to corticosteroids (dexamethasone, prednisone, and methylprednisone), androgens such as fluoxymesterone and methyltestosterone, estrogens such as diethylstilbesterol, antiestrogens such as-tamoxifen, LHRH analogues such as leuprolide, antiandrogens such as flutamide, aminoglutethimide, megestrol acetate, and medroxyprogesterone), asparaginase, carmustine, lomustine, hexamethyl-melamine, dacarbazine, mitotane, streptozocin, cisplatin, carboplatin, levamasole, and leucovorin. The compounds provided herein can also be used in combination with enzyme therapy agents and immune system modulators such as an interferon, interleukin, tumor necrosis factor, macrophage colony-stimulating factor and colony stimulating factor. It should be understood that any suitable combination of the compounds provided herein with one or more of the above-mentioned compounds and optionally one or more further pharmacologically active substances are considered to be within the scope of the present disclosure. In another embodiment, the compound provided herein is administered prior to or subsequent to the one or more additional active ingredients. The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention. EXAMPLES Example 1
2-(octadecyloxy)ethyl dihydrogen phosphate (2).
To a cold solution of phosphorus oxychloride, 3ml (32 mmol) in THF was added dropwise a solution of 2-octadecyloxy-l-ethanol (5g,16 mmol) and triethylamine (4.4 ml, 32 mmol) in THF, while the temperature was maintained below 20 0C. After addition was complete, the mixture was kept an additional hour, then water was added and the stirring was continued overnight. The mixture was then extracted with ethyl ether and the ether layer was concentrated. The crude solid was recrystallized from hexane to give octadecyloxyethyl phosphate as a white solid in 72% yield.
2-(octadecyIoxy)ethyl hydrogen morpholinophosphate (4). To a solution of 3g (7.6 mmol) of 2-(octadecyloxy)ethyl dihydrogen phosphate in tert- butyl alcohol was added 2g (24 mmol) of morpholine and 5.8g (30 mmol) of DCC, added in four portions and refluxed over 48 h. Ethyl ether was added to the mixture, then it was filtered, and the filtrate concentrated to give 2-(octadecyloxy)ethyl phosphonomorpholidate as an oil. Mass spectrum: (ESI) m/z 462 (M-H)" Example 2
3-(hexadecyloxy)propyI dihydrogen phosphate (1).
1 was prepared following the same procedure described in example 1, except that 3- (hexadecyloxy)- 1 -propanol was phosphorylated.
3-(hexadecyloxy)propyI hydrogen mopholinophosphate (3). HDP-phospho-morpholidate was prepared using the procedure described in example 1 using 1.92g (15 mmol) of 3-(hexadecyloxypropyl) dihydrogen phosphate, 1.3g (15 mmol) of morpholine and 35 mmol of DCC. HDP-phospho-morpholidate was obtained as an oil. Mass spectrum: (ESI) m/z 449 (M-H)". Example 3
(S)-l-(3-(4,4'-dimethoxytrityloxy)-2-phosphonomethoxypropyl)cytosine (5). (S)-l-(3-hydroxy-2-phosphonomethoxypropyl)cytosine dihydrate (free acid, 0.25g, 0.75 mmol) was mixed with methanol and 3 ml of tributylamine. After solution was achieved, solvents were removed to dryness and the solid residue was dissolved in DMSO. To this solution was added 0.9g of tributylamine and 0.7 g of dimethoxytrityl chloride, and the mixture was stirred overnight at room temperature. Solvents were removed and the solid residue was recrystallized from ethyl acetate and dried under vacuum to obtain 0.35g (91%) of the product 5 as a white solid. MS (ESI) m/z 943 (M-H)".
Example 4
(l-(4-aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2- yloxy)methylphosphonic(3-hexadecyloxy)propylphosphoric) anhydride (ΗDP-phospho- cidofovir, 6).
To a suspension of (5) 0.1 g (0.17 mmol ) in pyridine was added 0.1 ml of tributylamine. After solution was achieved, a solution of Ig of 3 in pyridine was added along with 0.1 ml of acetic acid. The reaction was stirred at room temperature for 48h. Solvents were removed to dryness and the residue was mixed with a solution of CHCl3 and TFA
(5:0.5) and stirred at room temperature. Solvents were removed and the crude mixture was purified by flash column chromatography using silica gel and eluting with 70:30:3:3 (CHCl3 -.Methanol -.Ammonium hydroxide: Water) to obtain 0.04g (40 %) of 6 as a white solid. MS (ESI) m/z 642 (M-H)-. 1HNMR 300MHz (DMSO) δ ppm: 7.47 (d,1H, 6.9Hz), 5.66 (d, lH,7.2Hz), 3.8-3.1 (mm, 16H), 1.7 (m, 2H), 1.4 (m,2H), 1.2 (s, 26H), 0.8 (t,3H). 31P NMR (DMSO-d6) 12.5 (s, Pl), 0.4(s, P2).
Example 5
(l-(4-aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2- yloxy)methylphosphonic-(3-octadecyloxy)ethyl phosphoric) anhydride (ODE-phospho- cidofovir, 7).
To a suspension of 5 (0.14 g, 0.17 mmol ) in pyridine was added 0.1 ml of tributylamine. After solution was achieved, a solution of Ig of 4 in pyridine was added along with 0.1 ml of acetic acid. The reaction was stirred at room temperature for 48h. Solvents were removed to dryness and the residue was mixed with a solution of CHC13TFA (5:0.5) and stirred at room temperature. After complete deprotection the crude mixture was purified by column chromatography using silica gel and eluting with 70:30:3:3 (CHCl3:Methanol: Ammonium hydroxide: Water) to obtain 0.03g (20 %) of 7 as a white solid. MS (ESI) m/z 654 (M-H)-. 1H NMR 300MHz (CDCl3/CD3OD) δ ppm: 6 (d,lH, J=8Hz), 5.8 (d, lH,J=7.2Hz), 4.2-3.4 (mm, 16H), 1.7 (m, 2H), 1.58 (m,2H), 1.27 (s, 28H), 0.89 (t,3H). 31P NMR (CDCl3/CD3OD), 12.38 (m.Pl), 5.6 (d.P2,J=25.5Hz). Example 6
(2-(6amino-9H-purin-9-yI)ethoxy)methylphosphonic(3- hexadecyloxy)propylphosphonic)anhydride (HDP-phospho-PMEA, 9).
9-(2-phosphonylmethoxyethyl)adenine (PMEA) 0.5 g was dissolved in pyridine and a solution of Ig of 3 in pyridine was added along with 1 ml of acetic acid. The reaction mixture was heated at 40 °C overnight . Solvents were removed and the oily residue was washed with 10% methanol in ethyl ether and filtered. The filtrate was concentrated and purified by column chromatography using silica gel and eluting with 25% methanol in dichloromethane to obtain 0.19g of 9 (17 %) as a white solid. Mass spectrum: (ESI) m/z 635(M-H)", 636(M+H)+. 1H NMR 300MHz (DMSO) δ 8.35 (s,lH), 8.09 (s,lH), 7.86 (bs,2H), 7.12 (bs,2h), 0.83 (s,3H). 31P NMR (DMSO) δl.4 (d,Pl,JPP= 24.32Hz), -12 (d,P2,JPP= 24.32Hz).
Example 7
5', 3'-dideoxy-5'-(oxymethyIphosphonic acid)-3'-azidothymidine (10). To a suspension of 1.2 g of NaH (60% oil dispersion) in DMF, was added a solution of 2.67g (10 mmol) of 3'-deoxy-3'-azidothymidine (AZT). After 30 min of reaction at room temperature, a solution of 3.22g (10 mmol) of diethyl p- toluenesulfonyloxymethylphosphonate in DMF was added. The mixture was stirred for 3 days at room temperature, then neutralized with acetic acid and the solvent was removed to dryness. The solid residue was purified by column chromatography using 5% methanol in dichloromethane to obtain 1.4g (34%) of diethyl 5', 3'-dideoxy-5'-(oxymethylphosphonate)- 3'-azidothymidine. This product was treated with TMSBr in dicholoromethane to obtain a mixture that was purified by ion exchange using DEAE in HCO3 form, eluting with ammonium bicarbonate solution to obtain 10 as a white solid. Mass spectrum: (ESI) m/z 360(M-H)". 31P NMR (D2O) 14.3(s,Pl). Example 8
5', 3'-dideoxy-5'-(oxymethylphosphonic acid)-3'-azidothymidine, (3- hexadecyloxy)propyl phosphoric anhydride (HDP-phospho-phosphonomethoxy AZT),
H)- To a solution of 0.18g (0.49 mmol) of compound 10 in pyridine was added a solution of Ig (2.2 mmol) of 3 in pyridine and 1 ml of acetic acid. The mixture was heated at 40 °C overnight. Solvents were removed and the residue was purified by flash column chromatography on silica gel using 70:30:3:3 (CHCl3 Methanol: Ammonium hydroxide: Water) to obtain product 11 as a white solid. Mass spectrum: (ESI) m/z 722(M-H)". 31P NMR (DMSOd6) 5.6 (d,Pl, ,JPP =26.6 Hz), 9.5 (d,P2, JPP =25.6 Hz). 1HNMR 300MHz (DMSO-d6) δ 7.5 (s,IH), 6.01 (m,lH), 4.4-4.3 (m, IH), 3.7-3.48 (m,6H), 2.0-2.3 (m,4H), 1.8- 1.6 (m,5H), 1.4 (m,2H), 1.2 (bs,24H), 0.83 (t,3H).
Example 9 Antiviral Activity of Substituted Phosphate Esters Of Nucleoside Phosphonates.
Compounds provided herein were prepared and tested against various viruses in vitro. In Table 3, EC50 for exemplary compounds are provided as follows:
Table 3
Figure imgf000072_0001
Results expressed as 50% effective concentration, EC50 in μM; Abbreviations: HDP-P-HPMPC: (l-(4- aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2yloxy)-methylphosphonic(3-hexadecyloxy)propyl-phosphoric) anhydride (6), ODE-P-HPMPC: (l-(4-aminooxopyrimidin-l(2H)-yl)-3-hydroxypropan-2-yloxy)methylphosphonic-(3- octadecyloxy)ethyl phosphoric) anhydride, ADV: adefovir, phosphonomethoxyethyadenine; HDP-P-ADV: (2-(6amino-9H- purin-9-yl)ethoxy)methylphosphonic(3-hexadecyloxy)-propylphosphonic)anhydride; Antiviral Assays: 'Cowpox Brighton, plaque reduction assay in HFF cells; 2Vaccinia WR plaque reduction assay in HFF cells, 3AD 169 plaque reduction assay in HFF cells; "Plaque reduction assay in HFF cells, 5HSV-I DNA reduction assay in MRC-5 Cells, 6HSV-I plaque reduction assay; 7HSV-2 plaque reduction assay, 8HTV-ILai p24 reduction assay in MT-2 cells.
ND = not determined.
Since modifications will be apparent to those of skill in the art, it is intended that the invention be limited only by the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula I:
Figure imgf000073_0001
or a pharmaceutically active derivative thereof, wherein RL is a lipophilic group, Rq is a pharmacologically active phosphonate, and y is 1 or 2.
2. The compound of claim 1, wherein the compound has formula II:
Figure imgf000073_0002
or a pharmaceutically active derivative thereof, wherein; R1 and Rlx are each independently -H, -O(C1-C24)alkyl, -O(C2-C24)alkenyl,
-0(C1-C2-OaCyI, -S(C1-C24)alkyl, -S(C2-C24)alkenyl, or -S(C1-C24)acyl, wherein at least one of R1 and Rlx is not -H, and wherein said alkenyl or acyl optionally have 1 to about 6 double bonds,
R2 and R2x are each independently -H, -O(C1-C7)alkyl, -O(C2-C7)alkenyl, S(C1- C7)alkyl, -S(C2-C7)alkenyl, -O(C1-C7)acyl, -S(d-C7)acyl, -N(CrC7)acyl, NH(C1-C7)alkyl, - N((CrC7)alkyl)2, halogen, -NH2, -OH, or -SH; X, when present, is:
Figure imgf000073_0003
L is a valence bond or a bifunctional linking molecule of the formula -J-(CRxRy)t-G-, wherein t is an integer from 1 to 24; J and G are independently -O-, -S-, -C(O)O- or -NH-; Rx and Ry are each independently -H, alkyl, or alkenyl; m is an integer from 0 to 6; n is 0 or 1 ; and wherein R1, Rlx, R2, R2x, Rx and Ry are optionally substituted with one to four substituents selected each independently selected from alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
3. The compound of claim 2, wherein R and R x are each independently -H, optionally substituted -O(C1-C24)alkyl; wherein at least one of R1 and Rlx is not -H;
R2 and R2x are each independently -H, optionally substituted -O(C1-C7)alkyl; Rq is a pharmacologically active phosphonate or a phosphonate derivative of a pharmacologically active compound of formula:
Figure imgf000074_0001
Rp is a pharmacologically active nucleoside or analog thereof;
L is a valence bond or a bifunctional linking molecule of the formula -J-(CRxRy)t-G-, wherein t is an integer from 1 to 24, J and G are independently -O-, and Rx and Ry are each independently -H, substituted or unsubstituted alkyl, or alkenyl; n is 0 or 1 ; and n1 is O to 3.
4. The compound of any of claims 1-3, wherein Rp is
Figure imgf000074_0002
wherein
R3, R4 and R5 are each independently H, hydroxy, halo, azido, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl; and wherein the substituents on the alkyl and alkenyl groups, when present, are selected from one to four alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl or heteroaryl.
B is a purine or pyrimidine base or analog thereof; R3x is H, azido, substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted
C2-6 alkenyl or substituted or unsubstituted C2-6 alkynyl;
R4x is H, C1-6 substituted or unsubstituted alkyl, C2-6 substituted or unsubstituted alkenyl or C2-6 substituted or unsubstituted alkynyl;
R3z is H, substituted or unsubstituted C1-6 alkyl, hydroxylC1-6 alkyl, ImIoC1-6 alkyl, azidoC1-6 alkyl or OH; and wherein the alkyl, alkenyl and alkynyl groups when substituted, are substituted with one to four substituents each independently selected from alkyl, alkenyl, alkynyl, halo, hydroxyl, pseudohalo, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
5. The compound of claim 1 , wherein RL has formula:
Figure imgf000075_0001
6. The compound of claim 1, wherein RL has formula:
Figure imgf000075_0002
7. The compound of claim 1, wherein RL has formula:
Figure imgf000075_0003
8. The compound of claim 1, wherein RL is hexadecyloxypropyl, octadecyloxypropyl, or octadecyloxyethyl.
9. The compound of claim 4, wherein B is
Figure imgf000076_0001
wherein R3a is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, hydroxy, halo, aryl or heteroaryl;
R6 is H or C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl;
R7 is H, hydroxy, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl OrNR4R5;
R8 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl or cycloalkyl and
R9 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, halo or NR4R5, where R4 and R5 are each independently H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C3-6 cycloalkyl.
10. The compound of claim 9, wherein B is selected from:
Figure imgf000076_0002
11. The compound according to claim 1, wherein Rq is an acyclic nucleoside phosphonate.
12. The compound of claim 11, wherein the acyclic nucleoside phosphonate is cidofovir.
13. The compound of claim 11 , wherein the acyclic nucleoside phosphonate is
(S)-HPMPA.
14. The compound of claim 11 , wherein the acyclic nucleoside phosphonate is adefovir.
15. The compound of claim 11 , wherein the acyclic nucleoside phosphonate is tenofovir.
16. The compound of claim 11, wherein the acyclic nucleoside phosphonate is PMEG.
17. The compound of claim 1, wherein Rq is a nucleoside-5 '-phosphonate or a nucleoside-5 '-methylene phosphonate.
18. The compound of claim 17, wherein the 5'-methylene phosphonate is azidothymidine.
19. The compound of claim 18, wherein the 5'-methylene phosphonate is 2'-O- methyl cytosine.
20. The compound of claim 18, wherein the 5'-methylene phosphonate is a β-D-1'- methyl ribofurano analog of cytidine, guanosine, uridine, adenosine, inosine or thymidine.
21. The compound of claim 18, wherein the 5 '-methylene phosphonate is a β-D-2'- C-methyl ribofurano analog of cytidine, guanosine, uridine, adenosine, inosine or thymidine.
22. The compound of claim 18, wherein the 5 '-methylene phosphonate is a β-D-2'- O-methyl ribofurano analog of cytidine, guanosine, uridine, adenosine, inosine or thymidine.
23. The compound of claim 1 , wherein the compound has formula:
Figure imgf000077_0001
24. The compound of claim 1 , wherein the compound has formula:
Figure imgf000077_0002
25. The compound according to claim 23, wherein Rq is an acyclic nucleoside phosphonate.
26. The compound of claim 25, wherein the acyclic nucleoside phosphonate is cidofovir.
27. The compound of claim 25, wherein the acyclic nucleoside phosphonate is
(S)-HPMPA.
28. The compound of claim 25, wherein the acyclic nucleoside phosphonate is adefovir.
29. The compound of claim 25, wherein the acyclic nucleoside phosphonate is tenofovir.
30. The compound of claim 26, wherein the acyclic nucleoside phosphonate is PMEG.
31. The compound of claim 1 selected from HDP-phospho-(S)-HPMPA, ODE- phospho-(S)-HPMPA, OLE-phospho-(S)-HPMPA, OLP-phospho-(S)-HPMPA, 15-methyl- hexadecyloxy-propyl-phospho-(S)-HPMPA, 17-methyl-octadecyloxy-ethyl-phospho-(S)- HPMPA, 16-fluoro-hexadecyloxy-propyl-phospho-(S)-HPMPA, 18-fluoro-octadecyloxy- ethyl-phospho-(S)-HPMPA, 15-methyl-hexadecyloxy-ethyl-phospho-(S)-HPMPA, HDP- phospho-cidofovir, ODE-phospho-cidofovir, OLE-phospho-cidofovir, OLP-phospho- cidofovir, HDP-phospho-PMEG, ODE-phospho-PMEG, HDP-phospho-PME-DAP, HDP- phospho-PME-N6cPr-DAP, OLE-phospho-PME-N6cPr-DAP, HDP- phospho-PPMG, HDP- phospho-PPM-DAP, HDP-phospho-PPM-N6cPr-DAP, HDP-phospho- PME-5FU, HDP- phospho-PME-5FC, HDP-phospho-HPMP-5FC, HDP-phospho-HPMP-5FU, HDP-phospho- Phosphonomethoxy-3TC, HDP-phospho-Phosphonomethoxy-2 -C-methyl ribo-guanine, HDP-phospho-Phosphonomethoxy-1 -methyl cytidine, HDP-phospho-PM-2 -O-methyl cytidine and ODE-phospho-PM-2 -C-methyl adenosine.
32. The compound of claim 1 selected from HDP-phospho-(S)-HPMPA), ODE- phospho-(S)-HPMPA, OLE-phospho-(S)-HPMPA, OLP- phospho-(S)-HPMPA, 15-methyl- hexadecyloxy-propyl-phospho-(S)-HPMPA, and 17-methyl-octadecyloxy-ethyl- phospho- (S)-HPMPA.
33. A pharmaceutical composition comprising a compound of any of claims 1-32 and a pharmaceutically acceptable carrier.
34. A method for treating a viral infection, wherein the method comprises administering an effective amount of a compound of any of claims 1-32.
35. The method of claim 34, wherein the viral infection is a caused by influenza, hepatitis B virus, hepatitis C virus, cytomegalovirus, Varicella zoster virus, Herpes simplex virus types 1 and 2, Epstein-Barr virus, Herpes type 6 and type 8, Varicella zoster virus, Epstein Barr virus infections, retroviral infections, orthopox viruses, vaccinia, ebola virus, adenovirus and papilloma virus.
36. The method of claim 35, wherein the viral infection is Hepatitis B.
37. A method for treating a growing neoplasm, wherein the method comprises administering an effective amount of a compound of any of claims 1-32.
38. A method for modulating cell proliferation, wherein the method comprises administering an effective amount of a compound of any of claims 1-32.
39. A method for treating a cancer, wherein the method comprises administering an effective amount of a compound of any of claims 1-32.
40. The method of claim 39, wherein the cancer is selected from lung cancer, head and neck squamous cancers, colorectal cancer, prostate cancer, breast cancer, acute lymphocytic leukemia, adult acute myeloid leukemia, adult non Hodgkin's lymphoma, brain tumors, cervical cancers, childhood cancers, childhood sarcoma, chronic lymphocytic leukemia, chronic myeloid leukemia, esophageal cancer, hairy cell leukemia, kidney cancer, liver cancer, multiple myeloma, neuroblastoma, oral cancer, pancreatic cancer, primary central nervous system lymphoma, and skin cancer.
41. An article of manufacture, comprising packaging material and a compound of any of claims 1-32, contained within the packaging material, wherein the compound is effective for treatment of a disease associated with a viral infection or cell proliferation and the packaging material includes a label that indicates that the compound is used for treatment, prevention or amelioration of a disease associated with a viral infection or cell proliferation.
42. The compound of any of claims 1-32 for use in the treatment of a viral infection.
43. The compound of any of claims 1-32 for use in the treatment of a cell proliferative disease.
44. Use of a compound of any of claims 1-32 for the formulation of a medicament for the treatment of a viral infection.
45. Use of a compound of any of claims 1-32 for the formulation of a medicament for the treatment of a cell proliferative disease.
PCT/US2006/012121 2005-04-01 2006-03-30 Substituted phosphate esters of nucleoside phosphonates WO2006130217A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/887,501 US20090156545A1 (en) 2005-04-01 2006-03-30 Substituted Phosphate Esters of Nucleoside Phosphonates

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US66773905P 2005-04-01 2005-04-01
US60/667,739 2005-04-01

Publications (2)

Publication Number Publication Date
WO2006130217A2 true WO2006130217A2 (en) 2006-12-07
WO2006130217A3 WO2006130217A3 (en) 2007-03-01

Family

ID=37460185

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/012121 WO2006130217A2 (en) 2005-04-01 2006-03-30 Substituted phosphate esters of nucleoside phosphonates

Country Status (2)

Country Link
US (1) US20090156545A1 (en)
WO (1) WO2006130217A2 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009094191A2 (en) * 2008-01-25 2009-07-30 Chimerix, Inc. Methods of treating viral infections
WO2011011710A1 (en) * 2009-07-24 2011-01-27 Chimerix, Inc. Methods of treating viral infections
US8445457B2 (en) 2007-07-12 2013-05-21 Novartis Ag Oral pharmaceutical solutions containing telbivudine
WO2013095684A1 (en) * 2011-12-22 2013-06-27 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US8609627B2 (en) 2009-02-06 2013-12-17 Rfs Pharma, Llc Purine nucleoside monophosphate prodrugs for treatment of cancer and viral infections
US8614200B2 (en) 2009-07-21 2013-12-24 Chimerix, Inc. Compounds, compositions and methods for treating ocular conditions
US8642577B2 (en) 2005-04-08 2014-02-04 Chimerix, Inc. Compounds, compositions and methods for the treatment of poxvirus infections
US9006218B2 (en) 2010-02-12 2015-04-14 Chimerix Inc. Nucleoside phosphonate salts
US9694024B2 (en) 2010-04-26 2017-07-04 Chimerix, Inc. Methods of treating retroviral infections and related dosage regimes
US9828410B2 (en) 2015-03-06 2017-11-28 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US10946033B2 (en) 2016-09-07 2021-03-16 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
US11690860B2 (en) 2018-04-10 2023-07-04 Atea Pharmaceuticals, Inc. Treatment of HCV infected patients with cirrhosis
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds
US11773122B2 (en) 2020-08-24 2023-10-03 Gilead Sciences. Inc. Phospholipid compounds and uses thereof

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6849605B1 (en) 1999-03-05 2005-02-01 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of viral infections
WO2013059638A1 (en) 2011-10-21 2013-04-25 Abbvie Inc. Combination treatment (eg. with abt-072 or abt -333) of daas for use in treating hcv
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
EP2583677A3 (en) 2011-10-21 2013-07-03 Abbvie Inc. Methods for treating HCV comprising at least two direct acting antiviral agent, ribavirin but not interferon.
BR112016010862B1 (en) 2013-11-15 2022-11-29 Chimerix, Inc MORPHIC FORMS OF HEXADECYLOXYPROPYLPHOSPHONATE ESTERS, THEIR METHOD OF PREPARATION AND COMPOSITION INCLUDING SAID morphic FORM
MA39986A (en) * 2014-04-25 2017-03-01 Vitae Pharmaceuticals Inc Purine derivatives as cd73 inhibitors for the treatment of cancer
US11192914B2 (en) 2016-04-28 2021-12-07 Emory University Alkyne containing nucleotide and nucleoside therapeutic compositions and uses related thereto
WO2018067424A1 (en) * 2016-10-03 2018-04-12 Arcus Biosciences, Inc. Inhibitors of adenosine 5'-nucleotidase
WO2018144640A1 (en) 2017-02-01 2018-08-09 Atea Pharmaceuticals, Inc. Nucleotide hemi-sulfate salt for the treatment of hepatitis c virus

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001039724A2 (en) * 1999-12-03 2001-06-07 The Regents Of The University Of California, San Diego Phosphonate compounds
WO2003087298A2 (en) * 2001-11-14 2003-10-23 Biocryst Pharmaceuticals, Inc. Nucleosides preparation thereof and use as inhibitors of rna viral polymerases

Family Cites Families (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) * 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) * 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3710795A (en) * 1970-09-29 1973-01-16 Alza Corp Drug-delivery device with stretched, rate-controlling membrane
US4044126A (en) * 1972-04-20 1977-08-23 Allen & Hanburys Limited Steroidal aerosol compositions and process for the preparation thereof
US3845770A (en) * 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
USRE28819E (en) * 1972-12-08 1976-05-18 Syntex (U.S.A.) Inc. Dialkylated glycol compositions and medicament preparations containing same
US4008719A (en) * 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4328245A (en) * 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4410545A (en) * 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4409239A (en) * 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
CS233665B1 (en) * 1983-01-06 1985-03-14 Antonin Holy Processing of isomere o-phosphonylmethylderivative of anantiomere racemic vicinal diene
KR890002631B1 (en) * 1984-10-04 1989-07-21 몬산토 캄파니 Composition of prolonged release of biologically active somatotropin
IE58110B1 (en) * 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
CS263951B1 (en) * 1985-04-25 1989-05-12 Antonin Holy 9-(phosponylmethoxyalkyl)adenines and method of their preparation
CS263952B1 (en) * 1985-04-25 1989-05-12 Holy Antonin Remedy with antiviral effect
CS264222B1 (en) * 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
US5650510A (en) * 1986-11-18 1997-07-22 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiviral phosphonomethoxyalkylene purine and pyrimidine derivatives
US5033252A (en) * 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) * 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
IT1229203B (en) * 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
PH30995A (en) * 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5585112A (en) * 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
IT1246382B (en) * 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
CZ285420B6 (en) * 1990-04-24 1999-08-11 Ústav Organické Chemie A Biochemie Avčr N-(3-fluoro-2-phosphonylmethoxypropyl) derivatives of purine and pyrimidine heterocyclic bases, processes of their preparation and use
US5733566A (en) * 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5210548A (en) * 1991-08-01 1993-05-11 Xerox Corporation Method and system for reducing surface reflections from a photosensitive imaging member
US5580578A (en) * 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US6010715A (en) * 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) * 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5323907A (en) * 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5532225A (en) * 1992-07-31 1996-07-02 Sri International Acyclic purine phosphonate nucleotide analogs as antiviral agents, and related synthetic methods
US6057305A (en) * 1992-08-05 2000-05-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiretroviral enantiomeric nucleotide analogs
TW333456B (en) * 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
US6444656B1 (en) * 1992-12-23 2002-09-03 Biochem Pharma, Inc. Antiviral phosphonate nucleotides
US6005107A (en) * 1992-12-23 1999-12-21 Biochem Pharma, Inc. Antiviral compounds
GB9226879D0 (en) * 1992-12-23 1993-02-17 Iaf Biochem Int Anti-viral compounds
US5591767A (en) * 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6274552B1 (en) * 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US5817647A (en) * 1993-04-01 1998-10-06 Merrell Pharmaceuticals Inc. Unsaturated acetylene phosphonate derivatives of purines
US5523092A (en) * 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US6087324A (en) * 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
EP0719274A1 (en) * 1993-09-17 1996-07-03 Gilead Sciences, Inc. Method for dosing therapeutic compounds
US5798340A (en) * 1993-09-17 1998-08-25 Gilead Sciences, Inc. Nucleotide analogs
US5656745A (en) * 1993-09-17 1997-08-12 Gilead Sciences, Inc. Nucleotide analogs
IT1270594B (en) * 1994-07-07 1997-05-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION OF LIQUID SUSPENSION MOGUISTEIN
US5759542A (en) * 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5660854A (en) * 1994-11-28 1997-08-26 Haynes; Duncan H Drug releasing surgical implant or dressing material
WO1997000262A1 (en) * 1995-06-15 1997-01-03 Mitsubishi Chemical Corporation Phosphonate nucleotide derivatives
ATE268591T1 (en) * 1995-06-27 2004-06-15 Takeda Chemical Industries Ltd METHOD FOR PRODUCING DELAYED RELEASE PREPARATIONS
TW448055B (en) * 1995-09-04 2001-08-01 Takeda Chemical Industries Ltd Method of production of sustained-release preparation
US6039975A (en) * 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US5717095A (en) * 1995-12-29 1998-02-10 Gilead Sciences, Inc. Nucleotide analogs
US5877166A (en) * 1996-04-29 1999-03-02 Sri International Enantiomerically pure 2-aminopurine phosphonate nucleotide analogs as antiviral agents
TW345603B (en) * 1996-05-29 1998-11-21 Gmundner Fertigteile Gmbh A noise control device for tracks
US6264970B1 (en) * 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) * 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
NZ334914A (en) * 1996-10-01 2000-09-29 Stanford Res Inst Int Taste-masked microcapsule compositions and methods of manufacture using a phase seperation-coacervation technique
CA2217134A1 (en) * 1996-10-09 1998-04-09 Sumitomo Pharmaceuticals Co., Ltd. Sustained release formulation
PT839525E (en) * 1996-10-31 2004-10-29 Takeda Chemical Industries Ltd PROLONGED LIBERATION PREPARATION
US6131570A (en) * 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
DE69719367T2 (en) * 1996-12-20 2003-10-16 Takeda Chemical Industries Ltd METHOD FOR PRODUCING A COMPOSITION WITH DELAYED DELIVERY
US5891474A (en) * 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5860957A (en) * 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US6120751A (en) * 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) * 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US5948433A (en) * 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
DE69839179T2 (en) * 1997-10-28 2009-02-19 Bando Chemical Industries, Ltd., Kobe DERMATOLOGICAL PLASTER AND METHOD FOR PRODUCING ITS BASE LAYER
IL136951A0 (en) * 1998-01-16 2001-06-14 Takeda Chemical Industries Ltd Sustained-release composition, method of its production and use thereof
US6613358B2 (en) * 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6048736A (en) * 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
KR19990085365A (en) * 1998-05-16 1999-12-06 허영섭 Biodegradable polymer microspheres capable of continuously controlled controlled release and preparation method thereof
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6271359B1 (en) * 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6256533B1 (en) * 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
US6261595B1 (en) * 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
JP2003532643A (en) * 2000-04-13 2003-11-05 フアーマセツト・リミテツド 3'- or 2'-hydroxymethyl-substituted nucleoside derivatives for treating hepatitis virus infection
MY164523A (en) * 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
CN101099745A (en) * 2000-05-26 2008-01-09 艾登尼科斯(开曼)有限公司 Methods and compositions for treating flaviviruses and pestiviruses
US20030008841A1 (en) * 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
WO2002032920A2 (en) * 2000-10-18 2002-04-25 Pharmasset Limited Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
DK1355916T3 (en) * 2001-01-22 2007-05-07 Merck & Co Inc Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
KR20020097384A (en) * 2001-06-20 2002-12-31 미쯔비시 도꾜 세이야꾸 가부시끼가이샤 A Medicament which is Effective for a Virus Having a Resistance Mutation against a Known Medicament
EP1404694A1 (en) * 2001-06-21 2004-04-07 Glaxo Group Limited Nucleoside compounds in hcv
US20040023928A1 (en) * 2001-10-31 2004-02-05 Colacino Joseph Matthew Phosphonate nucleotide and thiadiazole compounds for the treatment of smallpox
YU45204A (en) * 2001-11-27 2006-08-17 Anadys Pharmaceuticals Inc. 3-betha-d-ribofuranosylthiazolo/4,5-d/ pyrimidine nucleosides and uses thereof
US7247621B2 (en) * 2002-04-30 2007-07-24 Valeant Research & Development Antiviral phosphonate compounds and methods therefor
AU2003248748A1 (en) * 2002-06-28 2004-01-19 Idenix (Cayman) Limited 2'-c-methyl-3'-o-l-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
US20040067877A1 (en) * 2002-08-01 2004-04-08 Schinazi Raymond F. 2', 3'-Dideoxynucleoside analogues for the treatment or prevention of Flaviviridae infections
SI2604620T1 (en) * 2003-05-30 2016-10-28 Gilead Pharmasset LLC c/o Gilead Sciences, Inc. Modified fluorinated nucleoside analogues
WO2005090370A1 (en) * 2004-02-05 2005-09-29 The Regents Of The University Of California Pharmacologically active agents containing esterified phosphonates and methods for use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001039724A2 (en) * 1999-12-03 2001-06-07 The Regents Of The University Of California, San Diego Phosphonate compounds
WO2003087298A2 (en) * 2001-11-14 2003-10-23 Biocryst Pharmaceuticals, Inc. Nucleosides preparation thereof and use as inhibitors of rna viral polymerases

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8642577B2 (en) 2005-04-08 2014-02-04 Chimerix, Inc. Compounds, compositions and methods for the treatment of poxvirus infections
US8445457B2 (en) 2007-07-12 2013-05-21 Novartis Ag Oral pharmaceutical solutions containing telbivudine
WO2009094190A3 (en) * 2008-01-25 2009-10-01 Chimerix, Inc. Methods of treating viral infections
WO2009094191A3 (en) * 2008-01-25 2009-10-01 Chimerix, Inc. Methods of treating viral infections
WO2009094191A2 (en) * 2008-01-25 2009-07-30 Chimerix, Inc. Methods of treating viral infections
US9173893B2 (en) 2009-02-06 2015-11-03 Cocrystal Pharma, Inc. Purine nucleoside monophosphate prodrugs for treatment of cancer and viral infections
US8609627B2 (en) 2009-02-06 2013-12-17 Rfs Pharma, Llc Purine nucleoside monophosphate prodrugs for treatment of cancer and viral infections
US8614200B2 (en) 2009-07-21 2013-12-24 Chimerix, Inc. Compounds, compositions and methods for treating ocular conditions
WO2011011710A1 (en) * 2009-07-24 2011-01-27 Chimerix, Inc. Methods of treating viral infections
US9765100B2 (en) 2010-02-12 2017-09-19 Chimerix, Inc. Nucleoside phosphonate salts
US9006218B2 (en) 2010-02-12 2015-04-14 Chimerix Inc. Nucleoside phosphonate salts
US9694024B2 (en) 2010-04-26 2017-07-04 Chimerix, Inc. Methods of treating retroviral infections and related dosage regimes
US9956239B2 (en) 2010-04-26 2018-05-01 Chimerix, Inc. Methods of treating retroviral infections and related dosage regimes
WO2013095684A1 (en) * 2011-12-22 2013-06-27 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US9593137B2 (en) 2011-12-22 2017-03-14 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US10035814B2 (en) 2011-12-22 2018-07-31 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US11279720B2 (en) 2011-12-22 2022-03-22 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US10562926B2 (en) 2011-12-22 2020-02-18 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
US10005811B2 (en) 2015-03-06 2018-06-26 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US9828410B2 (en) 2015-03-06 2017-11-28 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10000523B2 (en) 2015-03-06 2018-06-19 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10815266B2 (en) 2015-03-06 2020-10-27 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10870672B2 (en) 2015-03-06 2020-12-22 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10870673B2 (en) 2015-03-06 2020-12-22 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10875885B2 (en) 2015-03-06 2020-12-29 Atea Pharmaceuticals, Inc. β-d-2′-deoxy-2′-α-fluoro-2′-β-c-substituted-2-modified-n6-substituted purine nucleotides for HCV treatment
US10239911B2 (en) 2015-03-06 2019-03-26 Atea Pharmaceuticals, Inc. Beta-D-2′-deoxy-2′-alpha-fluoro-2′-beta-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10946033B2 (en) 2016-09-07 2021-03-16 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
US11690860B2 (en) 2018-04-10 2023-07-04 Atea Pharmaceuticals, Inc. Treatment of HCV infected patients with cirrhosis
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11707480B2 (en) 2020-02-27 2023-07-25 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11738038B2 (en) 2020-02-27 2023-08-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11813278B2 (en) 2020-02-27 2023-11-14 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11773122B2 (en) 2020-08-24 2023-10-03 Gilead Sciences. Inc. Phospholipid compounds and uses thereof
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides

Also Published As

Publication number Publication date
US20090156545A1 (en) 2009-06-18
WO2006130217A3 (en) 2007-03-01

Similar Documents

Publication Publication Date Title
WO2006130217A2 (en) Substituted phosphate esters of nucleoside phosphonates
EP1866319B1 (en) Phosphono-pent-2-en-1-yl nucleosides and analogs
EP2012799B1 (en) Metabolically stable alkoxyalkyl esters of antiviral or antiproliferative phosphonates, nucleoside phosphonates and nucleoside phosphates
US7652001B2 (en) Pharmacologically active agents containing esterified phosphonates and methods for use thereof
ES2498046T3 (en) Phosphonate ester antiviral compounds
EP1831235B1 (en) Lung-targeted drugs
AU5009496A (en) Chemical compounds
KR20060008297A (en) METHODS OF MANUFACTURE OF 2'-DEOXY-beta-L-NUCLEOSIDES
JP2012153731A (en) Pyrimidine compound having phosphonate group as antiviral nucleotide analog
NO20131091L (en) 6- [2- (phosphonomethoxy) alkoxy] pyrimidine derivatives with antiviral activity
JP2006527702A5 (en)
AU2002315625A1 (en) 6-2'-(phosphonomethoxy)alkoxy pyrimidine derivatives having antiviral activity
Raju et al. Synthesis and biological properties of purine and pyrimidine 5'-deoxy-5'-(dihydroxyphosphinyl)-. beta.-D-ribofuranosyl analogs of AMP, GMP, IMP, and CMP
US20130018018A1 (en) Novel nucleoside phosphonates and analogs
WO2011069688A1 (en) Novel phosph(on)ate- and sulf(on)ate-based phosphate modified nucleosides useful as substrates for polymerases and as antiviral agents
ZA200204194B (en) Phosphonate compounds.

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06784342

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 11887501

Country of ref document: US