CA2485702C - Novel phosphonic acid based prodrugs of pmea and its analogues - Google Patents

Novel phosphonic acid based prodrugs of pmea and its analogues Download PDF

Info

Publication number
CA2485702C
CA2485702C CA2485702A CA2485702A CA2485702C CA 2485702 C CA2485702 C CA 2485702C CA 2485702 A CA2485702 A CA 2485702A CA 2485702 A CA2485702 A CA 2485702A CA 2485702 C CA2485702 C CA 2485702C
Authority
CA
Canada
Prior art keywords
compound
pmea
group
acid
adenine
Prior art date
Legal status (The legal status 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 status listed.)
Expired - Lifetime
Application number
CA2485702A
Other languages
French (fr)
Other versions
CA2485702A1 (en
Inventor
K. Raja Reddy
Mark D. Erion
Michael C. Matelich
Joseph J. Kopcho
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Metabasis Therapeutics Inc
Original Assignee
Metabasis Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Metabasis Therapeutics Inc filed Critical Metabasis Therapeutics Inc
Publication of CA2485702A1 publication Critical patent/CA2485702A1/en
Application granted granted Critical
Publication of CA2485702C publication Critical patent/CA2485702C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus
    • C07F9/65742Esters of oxyacids of phosphorus non-condensed with carbocyclic rings or heterocyclic rings or ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • 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
    • C07F9/6561Heterocyclic 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

Abstract

Prodrugs of Formula I, their uses, their intermediates, and their method of manufacture are described: Formula 1wherein:M and V are cis to one another and MPO3H2 is a phosphonic acid selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxy propyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxy ethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonyl methoxypropyl)adenine;V is selected from a group consisting of phenyl, 2-pyridyl , 3-pyridyl, 4-pyridyl, 2-furanyl, 3-furanyl, 2-thienyl, and 3-thienyl, all optionally substituted with 1-3 substituents selected from a group consisting of F, Cl, Br, C1-C3 alkyl, CF3 and OR6;R6 is selected from the group consisting of C1-C3 alkyl, and CF3;and pharmaceutically acceptable salts thereof.

Description

NOVEL PHOSPHONIC ACID BASED PRODRUGS
OF PMEA AND ITS ANALOGUES

FIELD OF THE INVENTION
The present invention is directed towards novel phosphonate prodrugs with antiviral and anticancer activity, to their preparation, to their synthetic intermediates, and to their uses. More specifically, the invention relates to the area of optionally substituted cyclic 1,3-propanyl-l-aryl esters of phosphonic acid antivirals based on PMEA
or related analogues.

BACKGROUND OF THE INVENTION
The following description of the background of the invention is provided to aid in understanding the invention, but is not admitted to be, or to describe, prior art to the invention.
9-(2-phosphonylmethoxyethyl)adenine (PMEA) and related analogues (U.S.
4,808,716; U.S. 5,142,051; De Clercq, et al., Antiviral Res. 8(5-6):261-72 (1987)) are phosphonic- acids that exhibit antiviral activity, including activity against hepatitis B and HIV. The dipivaloyloxy methylene ester of PMEA ("BisPOM PMEA") is,in clinical trials for the treatment of hepatitis B. (Benhamou, et al., Lancet 358(9283):718-23 (2001)) PMEA is thought to act by blocking DNA polymerase of HBV. In addition, some studies have shown that these compounds also show anticancer activity.
(Murono, et al.,. Cancer Res., 61(21):7875-7 (2001)) The biologically active compound is thought to be the diphosphate, such as PMEApp, which is produced from the phosphonic acid most likely as result of specific mammalian intracellular kinases.
Compounds containing phosphonic acids and their salts are highly charged at physiological pH and therefore frequently exhibit poor oral bioavailability, poor cell penetration and limited tissue distribution (e.g., CNS). In addition, these acids are also commonly associated with several other properties that hinder their use as drugs, including short plasma half-life due to rapid renal clearance, as well as toxicities (e.g., renal, gastrointestinal, etc.) (e.g., Antimicrob Agents Chemother. 42(5): 1146-50 (1998)).

= i For example, PMEA exhibits a very low volume of distribution, presumably due to its high negative charge. In humans, 98% of the dose is excreted renally as a result of the presence of organic anion transporters on the basolateral surface of the kidney tubule cells which help facilitate the renal clearance of PMEA. PMEA therapy is associated with severe renal toxicity possibly due to the exposure and accumulation of PMEA and related phosphorylated species in the kidney. Thus, there is a need for means to reduce the toxicity of PMEA and related analogues.
Cyclic phosphonate esters have also been described for PMEA and related analogues. The numbering for these cyclic esters is shown below:

O
-p\ 2' 3' Unsubstituted cyclic 1',3'-propanyl esters of PMEA were prepared but showed no in vivo activity. EP 0 481 214 B 1 discloses examples of cyclic prodrugs of PMEA
wherein the 1' and 3' positions are unsubstituted. The application and a subsequent publication by the inventors (Starrett et al., J Med. Chem. 37:1857-1864 (1994)) further disclose their findings with the compounds, namely that these compounds showed no oral bioavailability and no biological activity. The compounds were shown to be unstable at low pH, e.g., the cyclic 2',2'-difluoro-1',3'-propane ester is reported to be hydrolytically unstable with rapid generation of the ring-opened monoester.
Cyclic prodrugs with aryl groups at 1' are described for phosphonates that are known to be particularly useful in glucose lowering activity and therefore are useful in treating diabetes. (US 5,658,889, WO 98/39344, WO 98/39343, and WO 98/39342) In addition, US 6,312,662 discloses the use of this strategy for the liver-specific delivery of various drugs and compound classes to the liver for the treatment of patients with liver diseases such as hepatitis B and hepatocellular carcinoma.
Furthermore, diseases of the liver, such as hepatitis and liver cancer, remain poorly treated with current therapies due to dose-limiting extrahepatic side effects or inadequate delivery of chemotherapeutic agents to the target tissue.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure,l. Depicts the ketoconazole dependent inhibition of Compound 4 activation in Human Liver Microsomes.
Figure 2a. Depicts the Liver PMEApp levels following i.v. administration.
Figure 2b. Depicts the Liver PMEApp levels following oral administration.
Figure 3. Depicts the Oral Bioavailability of Compound 56 through the use of a liver concentration-time profile of PMEApp.
Figure 4. Depicts the PMEApp accumulation in rat hepatocytes with Compound 56 and PMEA.
Figure Sa. Depicts Liver tissue distribution of Compound 4 and bisPOM PMEA.
Figure 5b. Depicts Kidney Tissue distribution of Compound 4 and bisPOM PMEA.
Figure 5c. Depicts Small Intestine tissue distribution of Compound 4 and bisPOM PMEA.

SUMMARY OF THE INVENTION
The present invention is directed towards novel cyclic 1, 3-propanyl-l-aryl phosphonate.cyclic esters of PMEA and related analogues having cis relative stereochemistry between groups M & V, their preparation, their synthetic intermediates, and their uses. - In one embodiment, the cis cyclic esters have S
stereochemistry where V , is attached.
In one aspect, the invention is directed towards the use of these cyclic esters to treat viral infections. Another aspect of the invention is the use of these cyclic esters of PMEA and like analogues to treat diseases that benefit from enhanced dzug distribution to the liver and like tissues and cells, including hepatitis B, hepatitis C
and liver cancer. Another aspect of the invention is the use of these compounds to enhance oral delivery and/or prolong the pharmacodynamic half-life of PMEA and like analogues.
In addition, the compounds of the current invention are used to achieve sustained delivery of PMEA and like analogues and/or to increase the therapeutic index of the drug.
In another aspect of the invention, a method of making the cis prodrugs is described. In another aspect, a method of making substantially enantiomerically pure cis cyclic esters having S stereochemistry where V is attached is described.
One aspect of the present invention concerns compounds that are converted in vitro or in vivo to the corresponding MP032 , MP2063 and MP3094 and are of Formula I:
V

/P
M O
Formula I
wherein:
M and V are cis to one another and MP03H2 is a phosphonic acid selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine;
V is selected from a group consisting of phenyl, 2-pyridyl , 3-pyridyl, 4-pyridyl, 2-furanyl,.3-furanyl, 2-thienyl, and 3-thienyl, all optionally substituted with 1-3 substituents selected from a group consisting of F, Cl, Br, Cl-C3 alkyl, CF3 and OR6;;
R6 is selected from the group consisting of C1-3 alkyl, and CF3;
and pharmaceutically acceptable salts thereof.
A method for the preparation of the compounds described in this invention is described and relies on the reaction of PMEA or a PMEA analogue either as the phosphonic acid or in the activated form (e.g., dichloridate) with a 1,3-propane diol in'a manner that preferentially favors the production of the cis stereoisomer. In addition, methods and salt forms are described that enable isolation and purification of the desired isomer.
Since these compounds have asymmetric centers, the present invention is directed not only to racemic and diastereomeric mixtures of these compounds, but also to individual -stereoisomers. The present invention also includes pharmaceutically acceptable and/or useful salts of the compounds of Formula I, including acid addition salts.

Definitions In accordance with the present invention and as used herein, the following terms are defined with the following meanings, unless explicitly stated otherwise.
The term "alkyl" refers to saturated aliphatic groups including straight-chain, branched chain and cyclic groups. Suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl, and cyclopropyl.
The term "aryl" refers to aromatic groups which have 5-6 ring atoms. Suitable aryl groups include phenyl, furanyl, pyridyl, and thienyl. Aryl groups may be substituted.
The term "pharmaceutically acceptable salt" includes salts of compounds of Formula I derived from the combination of a compound of this invention and an organic or inorganic acid or base, such that they are acceptable to be safely administered to animals. Suitable acids include acetic acid, adipic acid, benzenesulfonic acid, (+)-7,7-dimethyl-2-oxobicyclo[2.2.1]heptane-l-methanesulfonic acid, citric acid, 1,2-ethanedisulfonic acid, dodecyl sulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glucoronic acid, hippuric acid, hydrochloride hemiethanolic acid, HBr, HCl, HI, 2-hydroxyethanesulfonic acid, lactic acid, lactobionic acid, maleic acid, methanesulfonic acid, methylbromide acid, methyl sulfuric acid, 2-naphthalenesulfonic acid, nitric acid, oleic acid, 4,4'-methylenebis[3-hydroxy-2-naphthalenecarboxylic acid], phosphoric acid, polygalacturonic acid, stearic acid, succinic acid, sulfuric acid, sulfosalicylic acid, tannic acid, tartaric acid, terphthalic acid, and p-toluenesulfonic acid.
The term "prodrug" as used herein refers to any M compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each. Standard prodrugs are formed using groups attached to functionality, e.g., HO-, HS-, HOOC-, R2N-, associated with the drug, that cleave in vivo. Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate. The groups illustrated are exemplary, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Such prodrugs of the compounds of Formula I, fall within the scope of the present invention. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, pharmacodynamic half-life, etc. The biologically active compounds include, for example, anticancer agents, and antiviral agents.
The term "cyclic 1',3'-propane ester", "cyclic 1,3-propane ester", "cyclic 1',3'-propanyl ester", and "cyclic 1,3-propanyl ester" refers to the following:

\ D
O 3, The term "cis" stereochemistry refers to the relationship of the V group and M
group positions on the six-membered ring. Formula II below shows a cis stereochemistry.
V
O\\ P O H
M O

II
The term "N6-substituted" refers to the substitution at the amine attached at the 6-position of a purine ring system. N6- is generally substituted with a dialkylaminomethylene group wherein R1 groups include but are not limited to acyclic alkyl, C5-C6 cyclic alkyl, benzyl, phenethyl, or R1 groups together form piperdine, morpholine, and pyrrolidine.

NO
6 1"5 N R1 l 2\N

The term "dialkylaminomethyleneimine" refers to the following structure:

NR
\R1 wherein R1 groups include but are not limited to Cl-C4 acyclic alkyl, C5-C6 cyclic alkyl, benzyl, phenethyl, or R1 groups together form piperdine, morpholine, and pyrrolidine.
The term "liver" refers to liver and to like tissues and cells that contain the CYP3A4 isozyme or any other P450 isozyme found to oxidize the cyclic prodrugs of the invention. Based on Example D, we have found that compounds of Formula I are selectively oxidized by the cytochrome P450 isoenzyme CYP3A4. According to DeWaziers et al. (J. Pharm. Exp. Ther. 253:387-394 (1990)), CYP3A4 is located in humans in the following tissues (determined by immunoblotting and enzyme measurements):
Tissues % of liver. activity Liver 100 Duodenum 50 jejunum 30 ileum 10 colon <5 (only P450 isoenzyme found) stomach <5 esophagus <5 kidney not detectable Thus, "liver" more preferably refers to the liver, duodenum, jejunum, ileum, colon, stomach, and esophagus. Most preferably, liver refers to the liver organ.
The term "enhancing" refers to increasing or improving a specific property.
The term "liver specificity" refers to the ratio:

[parent drug or a drug metabolite in liver tissue]
[parent drug or a drug metabolite in blood, urine or another non-hepatic tissue]

as measured in animals treated with the drug or a prodrug. The ratio can be determined by measuring tissue levels of the parent drug, or drug metabolite(s) including the biologically active drug metabolite, or both at a specific time or may represent an AUC
(area under curve) based on values measured at three or more time points.
The term "increased or enhanced liver specificity" refers to an increase in the liver specificity ratio in animals treated with the prodrug relative to animals treated with the parent drug.
The term "enhanced oral bioavailability" refers to an increase of at least 50%
of the absorption of the dose of the parent drug or prodrug (not of this invention) from the gastrointestinal tract. More preferably it is at least 100%. Measurement of oral bioavailability usually refers to measurements of the prodrug, drug, or drug metabolite in blood, tissues, or urine following oral administration compared to measurements following systemic administration.
The term "drug metabolite" refers to any compound produced in vivo or in vitro from the parent drug, or its prodrugs.
The term "pharmacodynamic half-life" refers to the time after administration of the drug or prodrug to observe a diminution of one half of the measured pharmacological response. Pharmacodynamic half-life is enhanced when the half-life is increased by preferably at least 50%.
The term "pharmacokinetic half-life" refers to the time after administration of the drug or prodrug to observe a diminution of one-half of the drug concentration in plasma or tissues.
The term "therapeutic index" refers to the ratio of the dose of a drug or prodrug that produces a therapeutically beneficial response relative to the dose that produces an undesired response such as death, an elevation of markers that are indicative of toxicity, and/or pharmacological side effects.
The term "sustained delivery" refers to an increase in the period in which there is a prolongation of therapeutically-effective drug levels due to the presence of the prodrug.
The term "bypassing drug resistance" refers to the loss or partial loss of therapeutic effectiveness of a drug (drug resistance) due to changes in the biochemical pathways and cellular activities important for producing and maintaining the biological activity of the drug and the ability of an agent to bypass this resistance through the use of alternative pathways or the failure of the agent to induce changes that tend to resistance.
The term "therapeutically effective amount" refers to an amount that has any beneficial effect in treating a disease or condition.
The term "phosphonate" refers to compounds attached via carbon to P032-.
The term "parent drug" refers to PMEA, or its analogues, when MPO3H2 is PMEA, or its analogues, respectively.
The term "biologically active drug or agent" refers to the chemical entity that produces the biological effect. In this invention, biologically active agents refers to MP032-, MP2063 or MP3094" where M can be the same M as in the parent drug or a metabolite.
The term "PMEA analogue" refers to compounds that have a nucleotide base connected to a phosphoric acid via a chain of atoms. Suitable PMEA analogues include (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, (2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine.
The term "percent enantiomeric excess (% ee)" refers to optical purity. It is obtained by using the following formula:
R - S X100=%R-%S=%eeRisomer [R] + [S]

where [R] is the amount of the R isomer and [S] is the amount of the S isomer.
This formula provides the % ee when R is the dominant isomer.
The terms "chiral alcohol" or "chiral diol" refers to a 1,3-diol where % ee is 50%.
The term "amelioration of gastrointestinal toxicity" refers to blocking, suppressing, or lessening toxicity to gastrointestinal tissues and organs relative to the toxicity that would otherwise occur with bisPOM PMEA.
The term "amelioration of renal toxicity" refers to blocking, suppressing, or lessening toxicity to renal tissues and organs relative to the toxicity that would otherwise occur with bisPOM PMEA.
The term "amelioration of extrahepatic toxicity" refers to blocking, suppressing, or lessening toxicity to tissues and organs outside of the liver relative to the toxicity that would otherwise occur with bisPOM PMEA.
The term "resistant to antiviral therapy" refers to not fully responding to or not fully affected by antiviral therapy treatment.
The term "administered simultaneously" refers to the administration of one drug at or near the same time in which another drug is administered. Preferably administration is within 30 minutes of one another.
The term "interferon" refers to a family of species-specific vertebrate proteins that confer non-specific resistance to a broad range of viral infections, affect cell proliferation and modulate immune responses. Suitable interferons include interferon alpha, interferon gamma, Omega interferon, Ominferon, Roferon-A, Albuferon, Alferon, Infergen, Intron A, PEG-intron, Pegasys, interferon fusions, interferon derivatives, and small molecule interferon inducers.
The term "pegylated interferon" refers to an interferon that has been modified by the attachment of polyethylene glycol molecules, in order to increase its half-life by decreasing in vivo clearance, and thereby increasing its duration of action.
The following well known drugs are referred to in the specification and the claims. Abbreviations and common names are also provided.
ara-A; 9-beta-D-Arabinofuranosyladenine (Vidarabine) AZT; 3'-Azido-2',3'-dideoxythymdine (Zidovudine) d4T; 2',3'-Deoxy-2',3'-Didehydrothymidine (Stavudine) ddl; 2',3'-Dideoxyinosine (Didanosine) ddA; 2',3'-Dideoxyadenosine ddC; 2',3'-Dideoxycytidine (Zalcitabine) L-ddC; L-2',3'-dideoxycytidine L-FddC; L-2',3'-dideoxy-5-fluorocytidine L-d4C; L-2',3'-Dideoxy-2',3'-didehydrocytidine L-Fd4C; L-2',3'-Dideoxy-2',3'-didehydro-5-fluorocytidine (ACH
126,443) 3TC; (-)-2',3'-Dideoxy-3'-thiacytidine; (-)-1-((2R,5S)-2-(Hydroxymethyl)-1,3-oxathiolan-5-yl)cystosine (Lamivudine) 1-beta-D-Ribofuranosyl-1,2,4-triazole-3-carboxamide (Ribavirin) (Virazole) FIAU; 1-(2-Deoxy-2-fluoro-b-D-arabinofuranosyl)-5-iodouridine FIAC; 1-(2-Deoxy-2-fluoro-b-D-arabinofuranosyl)-5-iodocytosine L-FMAU; 2'-Fluoro-5-methyl-(3-L-arabino-furanosyluracil (Clevudine) BvaraU; 1-beta-D-Arabinofuranosyl-E-5-(2-bromovinyl)uracil (Sorivudine) E-5-(2-bromovinyl)-2'-deoxyuridine TFT; Trifluorothymidine (Trifluorothymidine) 5-Propynyl- 1 -arabinosyluracil (Zonavir) CDG; carbocyclic 2'-deoxyguanosine DAPD; beta-D-2-Hydroxymethyl-5-(2,6-diaminopurin-9-yl)-1,3-doxalane FDOC; (-)-beta-D- 5-Fluoro-l-[2-(hydroxymethyl)-1,3-dioxolane]cytosine d4C; 2',3'-Dideoxy-2',3'-didehydrocytidine DXG; dioxolane guanosine FEAU; 1-(2'-deoxy-2'-fluoro-l-j3-D-arabinofuranosyl)-5-ethyluracil FLG; 2',3'-Dideoxy-3'-fluoroguanosine FLT; 2',3'-Dideoxy-3'-fluorothymidine FTC; (-)-cis-5-Fluoro-l-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine (Coviracil) 5-yl-carbocyclic 2'-deoxyguanosine (BMS-200475) (Entecavir) [1-(4'-Hydroxy-1',2'-butadienyl)cytosine] (Cytallene) Oxetanocin A; 9-((2R,3R,4S)-3,4-Bis(hydroxymethyl)-2-oxetanyl)adenine NK 84-0218 Oxetanocin G; 9-((2R,3R,4S)-3,4-Bis(hydroxymethyl)-2-oxetanyl)guanine ddAPR; 2,6-diaminopurine-2',3'-dideoxyriboside Cyclobut A; ( /-)-9-[(1-beta,2-alpha,3-beta)-2,3-Bis(hydroxymethyl)-1-cyclobutyl]adenine Cyclobut G; (+I-)-9-[(1-beta,2-alpha,3-beta)-2,3-Bis(hydroxymethyl)-1-cyclobutyl]guanine (Lobucavir) 5-fluoro-2'-deoxyuridine (Floxuridine) dFdC; 2',2'-difluorodeoxycytidine (Gemcitabine) araC; arabinosylcytosine (Cytarabine) BUdR; 5-Bromodeoxyuridine (Broxine) IDU; 5-Iodo-2'-deoxyuridine (Idoxuridine) CdA; 2-Chloro-2'-deoxyadenosine (Cladribine) F-ara-A; 2-Fluoroarabinosyladenosine (Fludarabine) ACV; 9-(2-Hydroxyethoxylmethyl)guanine (Acyclovir) GCV; 9-((1,3-Dihydroxy-2-propoxy)methyl)guanine (gancyclovir) 9-(4-Hydroxy-3-(hydroxymethyl)but-1-yl)guanine (Penciclovir) (R)-9-(3,4-Dihydroxybutyl)guanine (Buciclovir) Phosphonoformic acid (Foscarnet) PPA; Phosphonoacetic acid PMEA; 9-(2-Phosphonylmethoxyethyl) adenine (Adefovir) PMEDAP; 9-(2-Phosphonylmethoxyethyl)-2,6-diaminopurine HPMPC; (S)-1-(3-Hydroxy-2-phosphonylmethoxypropyl) cytosine (Cidofovir) HPMPA; (S)-9-(3-Hydroxy-2-phosphonylmethoxypropyl) adenine FPMPA; 9-(3-Fluoro-2-phosphonylmethoxypropyl) adenine PMPA; (R)9-(2-phosphonylmethoxypropyl) adenine Ara-T; 9-beta-D-Arabinofuranosylthymidine FMdC; (E)-2'-deoxy-2'(fluoromethylene)cytidine AICAR; 5-aminoimidazole-4-carboxamido-l-ribofuranosyl AM365; acylic guanosine nucleoside analogue L-dT; beta-L-2'-deoxythymidine (NV-02B) L-dC; beta-L-2'-deoxycytosine, valine prodrug derivatives of beta-L-2'-deoxycytosine MCC478; 2-aminophosphonomethoxy ethyl purine analogue Interferon alpha Pegylated interferons Famciclovir, 2-[2-(2-amino-9H-purin-9-yl)ethyl]-1,3-propanediol diacetate XTL 001; monoclonal antibody combination (Hepe X-B) Theradigm Zadaxin, thymosin alpha HBV DNA vaccine EHT 899, viral protein ICN 2001, nucleoside analogue Fluor L and D nucleosides, nucleoside analogue Racivir Robustaflavone, naturally occurring biflavanoid DETAILED DESCRIPTION OF THE INVENTION
The invention is directed to the discovery that the cis stereochemistry of cyclic 1,3-propanyl-l-aryl esters of PMEA or related analogues are effective prodrugs. It is believed that these cyclic ester prodrugs of PMEA and PMEA analogues require activation through the action of P450 enzymes to generate the biologically active drug in vivo. The high levels of P450 enzymes such as CYP3A4 in the liver enables the activation of the compounds to be largely in the liver and therefore to increase liver levels of the biologically active drug and/or decrease levels of the biologically active drug outside of the liver. Liver specificity is dependent not only on P450 tissue distribution, but also prodrug activation rate and subsequent phosphorylation of PMEA
or its analogues relative to pathways that export PMEA out of the hepatocyte and into the blood stream or bile. Poor activation would result in low PMEA levels in the liver (as well as extrahepatic tissues). Good activation but slow conversion to the PMEA
diphosphate ("PMEApp") by downstream kinases would limit PMEApp production.
Rapid export of PMEA into the blood stream would decrease the liver specificity and limit the improvement of the therapeutic index. (Mulato et al., J. Phannacol.
Exp. Ther.
295:10-15 (2001); Gilead Sciences, Inc. Press Release (6/22/01) Foster City, CA).
Compounds of the present invention are metabolized in the liver to produce the phosphonate which can be further metabolized to the biologically active drug.
Decreased extrahepatic toxicity can also enable higher doses and, therefore, increased liver levels of the biologically active agent in the liver.
In another aspect of the invention, the cyclic esters of the present invention were found to exhibit good stability in aqueous solutions and good oral bioavailability. Good stability can enable the prodrug to exist in the gastrointestinal environment (pHs 1-9) for a time sufficient to ensure good oral absorption. In addition, oral bioavailability is dependent on GI stability to enzymes. Given that the small intestine, which is the predominant site for drug absorption, expresses relatively high levels of CYP3A4 (50%
compared to the liver), compounds of the type described could be associated with poor oral bioavailability due to intestinal prodrug activation and conversion to the corresponding phosphonic acid. As described in this invention, however, the compounds of the present invention show good oral bioavailabilities.
In another aspect of the invention, the cyclic esters of the present invention can also be used to enhance the pharmacodynamic half-life relative to the parent drug or prodrug esters [esterase sensitive] that are sensitive to esterases. Compounds of the invention are slowly cleared by the kidney relative to PMEA and other phosphonic acid analogues and, therefore, provide a depot of drug that is released over time.
Thus, the compounds of the present invention can produce a sustained therapeutic effect.
In contrast, prodrug esters such as the acyloxyalkyl esters like bisPOM are rapidly cleaved in vivo and therefore are dependent on the pharmacokinetics of the phosphonic acid.
(Noble, S. and Goa, K.L., Drugs 58(3):479-487 (1999)).
In another aspect, methods of preparing the cyclic phosphonate compounds are described.

These aspects are described in greater detail below.

Increased Therapeutic Index of Phosphonic Acid Drugs The compounds of this invention can significantly increase the therapeutic index ("TI") of PMEA and PMEA analogues. In many cases, the increased TI is a result of the high liver specificity which results in higher liver levels of the biologically active drug and therefore allows for a lower dose to be administered to achieve the same therapeutic benefit. Alternatively, an increased TI can result from decreased exposure of the drug to extrahepatic tissues and therefore a decrease in toxicity at doses that achieve similar or higher liver levels of the biologically active drug.
Renal toxicity is a common toxicity associated with phosphonic acids. The toxicity results from transport, e.g., via the organic anion transporters located on the basolateral membrane of the renal proximal tubule, of the negatively charged drug into e.g., tubular cells which then accumulate the drug to high concentrations unless there is an equally efficient transport of the drug out of the cell via luminal transport mechanisms (e.g., anion-exchange or facilitated diffusion). Many examples have been reported in the literature of nephrotoxic phosphonic acids. Renal toxicity is associated with bisPOM PMEA in both animals and humans. The toxicity is associated with rapid conversion of bisPOM PMEA to PMEA which is then cleared via the kidneys. In fact, 98% of an i.v. dose of PMEA is renally cleared.
Renal toxicity is associated with serum increases in creatinine and decreases in phosphate and ultimately decreased kidney function. The poor therapeutic index associated with bisPOM PMEA therapy is a function of an undesirable tissue distribution profile, more specifically, poor distribution of PMEA to liver relative to kidney. The undesirable distribution profile of PMEA can be attributed to its esterase-sensitive (bisPOM) prodrug as these ubiquitous enzymes are highly expressed in plasma and the gastrointestinal tissues resulting in high systemic exposure of PMEA. BisPOM
PMEA
must be administered at low doses in humans (10 mg/day) to avoid such toxicity, thereby minimizing its potential efficacy (Gilead Sciences, Inc. Press Release (6/22/0 1) Foster City, CA).

In contrast to bisPOM PMEA, compounds of this invention result in enhanced PMEA delivery to liver while minimizing systemic and kidney PMEA exposure.
Compound 54 is well-absorbed following oral administration and readily distributes to most tissues as the intact prodrug via passive diffusion. Compound 54 will remain stable in blood and in most tissues until it is activated in the liver by the cytochrome P450 enzyme, CYP3A4. This isoform is abundantly expressed in liver where it accounts for approximately 30% of all P450 activity. Liver specificity is gained because no other tissues express CYP3A4 to high levels. Compound 54 results in increased efficacy in the target organ with significantly reduced peripheral drug exposure and consequential toxicities. Alternatively, Compound 54 is less toxic than bisPOM PMEA, due to reduced systemic PMEA exposure, and higher doses are administered resulting in improved efficacy compared to bisPOM PMEA. In one study, the levels of PMEA and PMEA
associated metabolites were measured in rats administered either compound 4 or bisPOM PMEA (Example N and 0). These studies showed that liver organ levels of PMEApp were higher in animals treated with compound 4 whereas kidney levels and urine levels of PMEA were lower.
Another common toxicity associated with phosphonic acid drugs is gastrointestinal toxicity due to, for example, accumulation of the phosphonic acid or its metabolites (e.g., phosphorylated phosphonic acids) in the gut epithelial cells.
Accumulation of drugs in the gut epithelial cells is dependent on many factors.
Compounds that are hydrophobic and low molecular weight rapidly diffuse across the gut epithelial cells and enter the blood stream. Compounds that are rapidly metabolized by enzymes in the gut epithelial cells to a charged and highly polar compound can be trapped inside the cells and accumulate to levels that are cytotoxic. Drug metabolism is dependent on the substrate and enzyme specific activity and on other factors, including residence time, i.e. the time in which the drug is exposed to the gut epithelial cells.
Factors that affect residence time include drug molecular weight and polarity since these factors increase the diffusion time. In addition, other proteins such as p-glycoproteins can increase the residence time by preventing the drug from transversing the gut by pumping the drug back into the gastrointestinal tract.

BisPOM PMEA therapy in humans is associated with a significant incidence of GI adverse events (Decks et al., J. Infect. Dis. 176:1517-1523 (1997)).
The gastrointestinal tract contains esterases that rapidly cleave bisPOM PMEA
to PMEA. Accordingly, radiolabeled PMEA and PMEA metabolites were found trapped in the small intestine following administration of bisPOM PMEA to rats (Example N).
CYP3A4 is also expressed in the small intestine. In contrast to bisPOM PMEA, only low levels of PMEA and PMEA metabolites were associated with the small intestine following oral administration of Compound 4 (Example N).

Increased Oral Bioavailability Phosphonic acids are highly negatively charged molecules at physiological pH.
Accordingly, most phosphonic acids exhibit poor oral bioavailability. For example, PMEA has an oral bioavailability of 11 % in rats, < 1 % in monkeys, and <12%
in humans. (Cundy, K.C., Clin. Pharniacokinet. 36(2):127-143 (1999)). Certain prodrugs of PMEA have been shown to increase oral bioavailability. For example, the esterase-sensitive prodrug, bisPOM PMEA has an oral bioavailability of 38%, 25% and 41%
in rats, monkeys and humans, respectively. (Shaw et al., Drug Metab. and Disp.
25(3):362-366 (1997)). Other prodrugs of PMEA have shown poor oral bioavailability, including 2' and 2',2' disubstituted 1,3-propanyl cyclic prodrugs, due either to poor conversion to PMEA in vivo or to instability to aqueous solutions and therefore production of charged metabolites or PMEA in the GI tract.
Oral bioavailability of P450 substrates is generally low due to the presence of P450s in the gut. For example, drugs such as Cyclosporine, Midazolam and Felodipine all show low oral bioavailability due to P450 metabolism. (Wacher, V.J. et al., Adv.
Drug Deliv. Rev. 46:89-102 (2001)) Prodrugs of this invention were shown to enhance the oral bioavailability of PMEA and to show good oral bioavailabilities despite being good substrates for CYP3A4 (Example Q. Oral bioavailability was dependent on the aryl group and its substituents, on the diol stereoisomer used to prepare the prodrug and the salt form. For example, V = 3-chlorophenyl appeared to exhibit better oral bioavailability relative to V
= phenyl. (Example Q. Oral bioavailability also improved when the S-diol (V =

chlorophenyl) was used relative to the R-diol. (Example I).

Prodrug Activation Prodrugs of this invention are activated by P450s, e.g., CYP3A4 in humans.
Activation is highly dependent on the structural features of the prodrug moiety as well as the parent drug. Catalytic efficiencies (Vmax/Km) for each substrate are determined using liver microsomes, supersomes or recombinant enzymes and monitoring either production of the phosphonic acid or its byproduct or the disappearance of the prodrug.
As shown in Example P, catalysis is dependent on the ring stereochemistry with the trans isomer of compound 9-{2-[2,4-trans-(S)-(+)-4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl}adenine acting as a weak inhibitor whereas the cis isomer is readily oxidized and converted to PMEA.
Prodrug activation was also monitored in hepatoctyes by monitoring the production of PMEApp. As shown in Example B, V = 3-chlorophenyl , 4-pyridyl, and phenyl were identified as the prodrug moieties with the best activity in rat hepatocytes.
Prodrug activation was also dependent on the prodrug stereoisomer. The S-diol was the most readily activated enantiomer in vitro in human liver microsomes at both low and high prodrug concentrations. (Example H).
Prodrug activation is an important parameter in drug pharmacokinetics, including duration and oral bioavailability as well as efficacy, e.g., liver levels of the biologically active drug. Prodrugs that are activated efficiently in vivo can result in higher liver levels of PMEApp and in greater conversion of the prodrug to the active drug. The latter can lead to improved oral bioavailabilties. Prodrug activation in vivo is readily monitored with radiolabeled parent drug. Prodrug, PMEA and PMEA metabolites were monitored following administration of Compound 4 to rats (Example N and 0). Unreacted prodrug was measured in the feces but undetected in liver organ or the small intestine.

Prodrug Stability Prodrug stability is a critical factor influencing bulk drug and product shelf life, oral bioavailability, liver distribution, drug toxicity and pharmacokinetic and pharmacodynamic half-life. Phosphonate prodrugs are often unstable in aqueous solutions especially in non-neutral pH solutions. Acyloxyalkyl esters are associated with instability as are other common phosphonate prodrugs. (Oliyai, et al., Nucleosides, Nucleotides, Nucleic Acids 20(4-7):1295-8 (2001)) Cyclic prodrugs are associated with instability stemming from the ring strain. Substituted cyclic prodrugs are reported to exhibit poor stability in aqueous solutions. (Starrett, et al., J. Med. Chem.
37(12):1857-64(1994)).
Instability of prodrugs often leads to production of a monoanion byproduct which is poorly absorbed due to its negative charge. Low bioavailability has been attributed to prodrug instability in the GI tract, including both bisPOM PMEA (Benzaria, et al., J.
Med. Chen. 39(25):4958-65 (1996); Serafinowska, et al., J. Med. Chem.
38(8):1372-9 (1995)) and cyclic prodrugs of PMEA (Starrett, et al., J. Med. Chem.
37(12):1857-64 (1994))). Instability to esterases can lead to rapid prodrug cleavage and at sites not associated with the disease. Esterases are thought to be relatively widely distributed with high levels often observed in blood, kidney, gastrointestinal tissue and liver. Rapid cleavage by an esterase can therefore limit oral bioavailability by producing the charged intermediate in the gut prior to absorption. Esterase activity outside of the liver can limit liver distribution of the drug by producing the charged intermediate in extrahepatic tissues (e.g., kidney) or in the blood where upon the drug is then charged and may be limited in its ability to enter hepatocytes and/or may exhibit increased drug clearance through anionic transporters in the kidney.
Prodrugs of this invention exhibited excellent stability in aqueous solutions as well as in rat and human plasma (Examples F and G). The stability of compounds and 54 compared favorably to that of bisPOM PMEA (J. Med. Chem. 39:4958-4965 (1996)).

Sustained Delivery Drugs that undergo rapid elimination in vivo often require multiple administrations of the drug to achieve therapeutically effective blood levels over a significant period of time. Other methods are also available including sustained release formulations and devices. Prodrugs that breakdown over time can also provide a method for achieving sustained drug levels. In general, this property has not been possible with the known phosphonate prodrugs since either they undergo rapid hydrolysis in vivo (e.g., acyloxyalkyl esters) or very slow conversion (e.g., di-aryl prodrugs).

The cyclic phosphonate prodrugs of the invention are capable of providing sustained drug release by providing a steady release of the drug over time.
Studies in rats and dogs (Examples R and S) indicate that Compound 4 has a preferable pharmacokinetic profile by circulating longer as the intact prodrug, while systemic PMEA exposure was minimized, compared to bisPOM PMEA.
Sustained delivery of the drugs is achievable by selecting the prodrugs of Formula I that are hydrolyzed in vivo at a rate capable of maintaining therapeutically effective drug levels over a period of time. Accordingly, prodrugs of this invention can also improve the pharmacodynamic half-life of the drug. The cleavage rate of the drug may depend on a variety of factors, including the rate of the P450 oxidation, which is dependent on both the substituents on the prodrug moiety, the stereochemistry of these substituents and the drug itself. Moreover, sustained drug production will depend on the rate of elimination of the intermediate generated after oxidation and the rate and availability of the prodrug to the liver, which is the major site of oxidation.
Identification of the prodrug with the desired properties is readily achieved by screening the prodrugs in an assay that monitors the rate of drug production in the presence of the major P450 enzyme involved in the metabolism, in the presence of liver microsomes or in the presence of hepatocytes. Standard PK assays (e.g. microsome metabolism, prodrug levels in plasma) indicate that compounds of this invention can be selected for parameters which will improve their ability to achieve "sustained delivery."

Specific Prodrugs of the Invention The biologically active agent is detected in the liver following administration of drugs of Formula I. Prodrugs of this type undergo oxidation to produce the phosphonic acid, e.g., PMEA, and the aryl vinyl ketone byproduct as shown below:

Ar Ar OH O O
~ oxidation ~ ~ ~ OH ~J O
-P 0 -P~ 0 -P\ Lf--Ar o -IP-OH + (EAr O O O OH
Although the esters in the invention are not limited by the above mechanisms, in general, each ester contains a group or atom susceptible to microsomal oxidation, which in turn generates an intermediate that breaks down to the parent compound in aqueous solution via B-elimination of the phosphonate diacid. It is believed that the hydrogen geminal to the Ar (aryl) group is susceptible to microsomal oxidation.

Use in Treating Liver Cancer PMEA has been shown to exhibit anticancer activity (Hatse, S., Verh K. Acad.
Geneeskd Belg. 62(5):373-384 (2000)). Prodrugs of the invention are envisioned to be useful for treating cancers wherein the tumor cells express P450s, especially CYP3A4.
For example, prodrugs of this invention are useful in treating hepatocelluar carcinoma or other liver-associated cancers. In some cases, the prodrugs are combined with agents that induce the expression of P450s, especially CYP3A4 in order to enhance activity and/or specificity.

Treatment of Metastatic Cancer:
Prodrugs of this invention may be used in combination with another oncolytic agent. They may be administered separately from the other oncolytic agent or simultaneously with the other oncolytic agent. This combination may help prevent further growth of the hepatocellular carcinoma tumor and/or the combination may help both the hepatocelluar carcinoma tumor (primarily with compounds of this invention) and extrahepatic metastases, especially metastases that have decreased CYP3A4 expression (with the other oncolytic agent).
Administration of the prodrug may occur at or near the time in which the other oncolytic agent is administered or at a different time.

Treatment of Viral Infections:
PMEA is a known drug with potent activity against hepatitis B virus. Oral administration of PMEA is characterized by low oral bioavailability (<10%) whereas the bis acyloxyalkyl prodrug, bisPOM PMEA, exhibits good oral bioavailability and good antiviral activity in a variety of animal models as well as in HBV-infected humans. In humans, doses of bisPOM PMEA that achieve the largest viral titer reductions and greatest improvements in liver histology and other parameters of liver function (e.g., 30 -60 mg/day) are associated with kidney toxicity whereas lower doses (e.g., 10 mg/day) are reported to show less antiviral activity but no kidney toxicity after one year of therapy.
Prodrugs of this invention achieve higher levels of the biologically active form of PMEA, i.e. PMEApp, in the liver relative to animals treated with bisPOM PMEA
at equal PMEA molar doses (Example N). At these doses, the prodrugs of this invention also result in significantly lower levels of PMEA in the plasma, kidney and urine.
Accordingly, prodrugs of this invention are expected to achieve greater viral titer reductions with less risk of renal toxicity.
Increased viral titer reduction is expected to benefit HBV-infected patients through increased seroconversion and decreased risk of liver damage and chronic liver disease (e.g., cirrhosis and hepatocellular carcinoma). In addition, increased viral titer reduction is associated with decreased generation of viral mutants which are associated with drug resistance.
Another preferred aspect of the invention is the combination of the PMEA
prodrugs of this invention with other antiviral agents in order to achieve even greater decreases of viral titer and associated therapeutic benefits. HBV viral DNA
has been detected in extrahepatic tissues, including kidney. Extrahepatic HBV may provide virus particles that can infect hepatocytes. Since prodrugs of this invention target the liver, therapies that combine these prodrugs with other anti-HBV drugs is a preferred aspect of this invention, since the drug combination inhibits viral replication in the liver as well as at other sites throughout the body. Moreover, a preferred aspect of this invention is the combination of prodrugs of this invention with other well known agents that result in viral titer reduction (e.g., HBV antibody therapies) and improved viral-directed immune responses, e.g., interferons or pegylated interferons. The combination can also benefit HBV-infected patients through a reduction in the effective dose and therefore decreased side effects associated with the therapy. The drug combination can be administered to the HBV patient at the same time or at different times.
Antiviral drugs useful in the combination include Vidarabine; Zidovudine;
Stavudine; Didanosine; ddA; Zalcitabine; L-ddC; L-FddC; L-d4C; Lamivudine;
Ribavirin; FIAU; FIAC; BHCG; BvaraU; E-5-(2-bromovinyl)-2'-deoxyuridine; TFT;
Zonavir; CDG; DAPD; FDOC; d4C; d4T; DXG; FEAU; FLG; FLT; Clevudine;
Coviracil; Entecavir; Cytallene; Oxetanocin A; Oxetanocin G;NK 84-0218; ddAPR;
Cyclobut A; Cyclobut G; Floxuridine; dFdC; araC; 5-bromodeoxyuridine; IDU;
CdA; F-ara-A; ACV; GCV; Penciclovir; Buciclovir; Foscarnet; PPA; PMEA; PMEDAP;
HPMPC; HPMPA; FPMPA; PMPA; araT; FMdC; AICAR; AM365; L-dT; L-dC,,beta-L-2'-deoxycytosine, valine prodrug derivatives of beta-L-2'-deoxycytosine; ACH
126,443; ddl; ddA; ddC; MCC478; Interferon alpha; Pegylated interferons;
famciclovir;
XTL001; HBV DNA vaccine; ICN 2001; Fluor L and D nucleosides; Racivir;
Robustaflavone; 9-(arabinofuranosyl)-2,6-diaminopurine; 9-(2'-deoxyribofuranosyl)-2,6-diaminopurine; 9-(2'-deoxy-2'-fluororibofuranosyl)-2,6-diaminopurine; 9-(arabinofuranosyl)guanine; 9-(2'-deoxyribofuranosyl)guanine; 9-(2'-deoxy 2'-fluororibofuranosyl)guanine; interferons-all analogues; human monoclonal antibodies;
and non-interferon enhancers, such as Theradigm, thymosin alpha-1 and EHT899.
Prodrugs of this invention may be used in combination with another antiviral agent. Administration of the prodrug may occur at or near the time in which the other antiviral agent is administered or at a different time.
Prodrugs of this invention may be used to treat viral infections of the liver in an animal that is resistant to antiviral therapy when the resistance is a result of mutations in HBV polymerase.

Use of Prodrugs with CYP Inhibitors to Reduce Toxicity and/or Increase Oral Bioavailability In some cases, enhanced CYP activity may lead to unwanted drug metabolism.
For example, enhanced activity of CYPs not involved in prodrug activation can result in increased drug metabolism and therefore decreased efficacy. In addition, increased CYP
activity in other tissues, e.g., CYP3A4 in the gastrointestinal tract, could result in decreased prodrug absorption and liver drug levels. Inhibitors of CYP activity are known that might be useful in minimizing unwanted drug metabolism. For example, grapefruit juice is known to inactivate gastrointestinal CYP3A4 and to result in enhanced absorption of numerous drugs metabolized by CYP3A4. CYP3A4 inhibitors known to enhance oral bioavailability of drugs include ketoconazole and erythromycin.
(Wacher, V.J. et al., Adv. Drug Deliv. Rev. 46:89-102 (2001); US 5,665,386; US
5,716,928; US
5,962,522; US 6,004,927; US 6,028,054; US 6,121,234; US 6,180,666). CYP
inhibitors are also known for many of the CYP subfamilies that can be useful for attenuating unwanted drug metabolism while maintaining CYP activity important for prodrug cleavage. For example, the CYP3A4 inhibitor TAO was used to modulate cyclophosphamide metabolism in vivo in a manner that decreased the formation of toxic metabolites that do not contribute to its antitumor activity.

Compounds of the Invention The compounds of the invention are substituted 6-membered cyclic 1,3-propane diester prodrugs of PMEA and analogues as represented by Formula I:
V
O O
P
M O
Formula I
wherein:
M and V are cis to one another and MPO3H2 is phosphonic acid selected from a group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl) adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonyl-methoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine;
V is selected from a group consisting of phenyl, 2-pyridyl , 3-pyridyl, 4-pyridyl, 2-furanyl, 3-furanyl, 2-thienyl, and 3-thienyl, all optionally substituted with 1-3 substituents selected from a group consisting of F, Cl, Br, C1-C3 alkyl, CF3 and OR6;

R6 is selected from the group consisting of C1-C3 alkyl, and CF3;
and pharmaceutically acceptable salts thereof.
Another aspect of the invention are the compounds of Formula II:
V
O\ O
P
M O

Formula II
wherein:
MPO3H2 is a phosphonic acid selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3 -hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine;
V is selected from a group consisting of phenyl, 2-pyridyl , 3-pyridyl, 4-pyridyl, 2-furanyl, 3-furanyl, 2-thienyl, and 3-thienyl, all optionally substituted with 1-3 substituents selected from a group consisting of F, Cl, Br, C 1-C3 alkyl, CF3 and OR6;
R6 is selected from group consisting of Cl-C3 alkyl, and CF3;
and pharmaceutically acceptable salts thereof.
Another aspect of the invention are the compounds of Formula III:
V
O\ O 0 H
P
MO
Formula III
wherein:
MPO3H2 is a phosphonic acid selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine;
V is selected from a group consisting of phenyl, 2-pyridyl , 3-pyridyl, 4-pyridyl, 2-furanyl, 3-furanyl, 2-thienyl, and 3-thienyl, all optionally substituted with 1-3 substituents selected from a group consisting of F, Cl, Br, C1-C3 alkyl, CF3 and OR6;
R6 is selected from the group consisting of C1-C3 alkyl, and CF3;
and pharmaceutically acceptable salts thereof.
Another aspect of the invention are the compounds of Formula IV:
CI
O\PO H

Formula IV
wherein:
MPO3H2 is 9-(2-phosphonylmethoxyethyl)adenine;
and pharmaceutically acceptable salts thereof.
In another aspect, the invention is directed to compounds of Formulae I, II, III or N, where MPO3H2 is a phosphonic acid selected from the group consisting of 9-(2-phosphonylmethyloxyethyl) adenine, and (R)-9-(2-phosphonyrnethoxypropyl) adenine.
In another aspect, the invention is directed to compounds where V is phenyl, 3-pyridyl, and 4-pyridyl, all optionally substituted with 1-2 substituents selected from F, Br, Cl, CH3, OCH3, and CF3.
In another aspect, the invention is directed to compounds where V is 4-pyridyl, 2-bromophenyl, and 3-chlorophenyl.
In another aspect, the invention is directed to compounds of Formula I, II, III, or N that are salts formed with acetic acid, HBr, HCI, citric acid, maleic acid, sulfuric acid, and tartaric acid.
Another aspect is directed to salts formed with methanesulfonic acid or succinic acid.

Another aspect is directed to salts formed with methanesulfonic acid.
In one aspect, oral bioavailability is at least 5%. In another aspect, oral bioavailability is at least 10%.
P450 oxidation can be sensitive to stereochemistry which might either be at phosphorus or at the carbon bearing the aromatic group (V). The compounds of the present invention have two isomeric forms around the phosphorus. In one aspect, the stereochemistry enables both oxidation and the elimination reaction.
P450 oxidation can also be sensitive to stereochemistry at C l' where V is attached. In one aspect, the compounds of the present invention have S
stereochemistry where V is attached.
In one aspect the M groups include 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)gdenine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine, and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine. More preferred are 9-(2-phosphonylmethoxyethyl)adenine, and (R)-9-(2-phosphonylmethoxypropyl)adenine.
In one aspect, antiviral agents for use in the combination therapy with the compounds of Formula I include: Vidarabine; Zidovudine; Stavudine; Didanosine;
ddA;
Zalcitabine; L-ddC; L-FddC; L-d4C; Lamivudine; Ribavirin; FIAU; FIAC; BHCG;
BvaraU; E-5-(2-bromovinyl)-2'-deoxyuridine; TFT; Zonavir; CDG; DAPD; FDOC;
d4C; d4T; DXG; FEAU; FLG; FLT; Clevudine; Coviracil; Entecavir; Cytallene;
Oxetanocin A; Oxetanocin G;NK 84-0218; ddAPR; Cyclobut A; Cyclobut G;
Floxuridine; dFdC; araC; 5-bromodeoxyuridine; IDU; CdA; F-ara-A; ACV; GCV;
Penciclovir; Buciclovir; Foscarnet; PPA; PMEA; PMEDAP; HPMPC; HPMPA;
FPMPA; PMPA; araT; FMdC; AICAR; AM365; L-dT; L-dC, beta-L-2'-deoxycytosine, valine prodrug derivatives of beta-L-2'-deoxycytosine; ACH 126,443; ddl; ddA;
ddC;
MCC478; Interferon alpha; Pegylated interferons; famciclovir; valine prodrug derivatives of beta-L-2'-deoxycytosine; XTL001; HBV DNA vaccine; ICN 2001;
Fluor L and D nucleosides; Racivir; Robustaflavone; 9-(arabinofuranosyl)-2,6-diaminopurine;
9-(2'-deoxyribofuranosyl)-2,6-diaminopurine; 9-(2'-deoxy-2'-fluororibofuranosyl)-2,6-diaminopurine; 9-(arabinofuranosyl)guanine; 9-(2'-deoxyribofuranosyl)guanine;
9-(2'-deoxy 2'-fluororibofuranosyl)guanine; interferons-all analogues; human monoclonal antibodies; and non-interferon enhancers, such as Theradigm, thymosin alpha-1 and EHT899.
In another aspect, antiviral agents include: lamivudine, entecavir, coviracil, DAPD, clevudine, AM365, L-dT, L-dC, ACH 126,443, MCC478, lobucavir, foscarnet, PPA, interferon alpha, pegylated interferon alfa, famciclovir, ara-A, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, Lamivudine, Ribavirin, FIAU, FIAC, BHCG, BvaraU, E-5-(2-bromovinyl)-2'-deoxyuridine, TFT, zonavir, CDG, FDOC, d4C, DXG, FEAU, FLG, FLT, FTC, 5-yl-carbocyclic 2'deoxyguanosine, Oxetanocin A, Oxetanocin G, ddAPR, Cyclobut A, Cyclobut G, dFdC, IDU, araT, ddAPR, Foscarnet; PMEDAP, HPMPC, HPMPA, FPMPA, PMPA, ACV, GCV, Penciclovir, 9-(arabinofuranosyl)-2,6-diaminopurine, 9-(2'-deoxyribofuranosyl)-2,6-diaminopurine, 9-(2'-deoxy-2' fluororibofuranosyl)-2,6-diaminopurine, 9-(arabinofuranosyl)guanine, 9-(2'-deoxyribofuranosyl)guanine, and 9-(2'-deoxy 2'-fluororibofuranosyl)guanine.
In another aspect, antiviral agents include: lamivudine, entecavir, coviracil, DAPD, clevudine, AM365, L-dT, L-dC, ACH 126,443, MCC478, lobucavir, foscamet, PPA, interferon alpha, and pegylated interferon alfa.
In another aspect, antiviral agents include interferons-all analogues, human monoclonal antibodies, and non-interferon enhancers, such as Theradigm, thymosin alpha-1 and EHT899.
In one aspect, oncolytic agents for use in the combination therapy with the compounds of Formula I include: agents that alkylate DNA, including alkylating agents such as busulfan, carboplatin, temozolomide, thiotepa, cisplatin, miriplatin, and nitrogen mustards such as melphalan, ifosfamide, cyclophosphamide, chlorambucil, and nechlorethamine.
Agents that are from the antibiotic class of oncolytic drugs, including doxorubicin, duanorubicin, actinomycin D, epirubicin, idarubicin, plicamycin, pentostatin, mitoxantrone, valrubicin, and dactinomycin.
Agents that are from the antimetabolite class of oncolytic drugs, including cytarabine, fludarabine, gemcitabine, floxuridine, fluorouracil, cladribine, mercaptopurine, thioguanine, capecitabine, methotrexate, and mitomycin.

Other well known oncolytic agents include dacarbazine, mitoxantrone, piroxantrone, bleomycin, epipodophyllotoxins, such as etoposide and teniposide, vinca alkaloids, including vincrisitne, and vinblastine, taxanes, including paclitaxel, docetaxel, the tecan class, including camptothecin, irinotecan, 9-aminocamptothecin, topotecan, and lurotecan.
In another aspect, oncolytic agents include: agents that alkylate DNA, including alkylating agents such as busulfan, carboplatin, temozolomide, thiotepa, cisplatin, miriplatin, and nitrogen mustards such as melphalan, ifosfamide, cyclophosphamide, and chlorambucil.
Agents that are from the antibiotic class of oncolytic drugs, including doxorubicin, duanorubicin, epirubicin, idarubicin, plicamycin, valrubicin, and dactinomycin.
Agents that are from the antimetabolite class of oncolytic drugs, including gemcitabine, floxuridine, fluorouracil, mercaptopurine, thioguanine, capecitabine, methotrexate, and mitomycin.
Other oncolytic agents including etoposide, paclitaxel, docetaxel, irinotecan, topotecan, and lurotecan.
In another aspect, oncolytic agents include: doxorubicin, gemcitabine, irinotecan, and cisplatin.

Synthesis of Compounds of Formula I
Acyclic nucleoside phosphonate antivirals such as (S)-1 -(3 -hydroxy-2-phosphonyl-methoxy propyl) cytosine (HPMPC, cidofovir); 9-(2-phosphonyl methoxyethyl) adenine (PMEA, adefovir); (R)-9-(2-phosphonylmethoxy propyl) adenine (PMPA, Tenofovir) and their analogues are well described in literature.
(Naesens and De Clercq, Nucleosides Nucleotides 16:983 (1997); Naesens et al., Antiviral Chem Chemother, 8:23, (1997); Bronson, et al.,Nucleotide Analogues as Antiviral Agents, ACS
symposium series 401, Martin, J. C., Ed., American Chemical Society (1989)).
Synthesis of the compounds encompassed by the present invention includes the following steps: (I) synthesis of prodrugs of acyclic nucleoside phosphonates;
(II) synthesis of 1-(aryl)propane-1,3-diol.

1. Synthesis of Prodrugs of Acyclic Nucleoside Phosphonates:
Prodrugs can be introduced at different stages of synthesis of acyclic nucleoside phosphonates. Most often they are made at a later stage, because of their lability.
Advantageously, when chemical stability is not an issue during subsequent reaction conditions, the prodrug can be introduced at an earlier stage of synthesis.
The synthesis of prodrugs of acyclic nucleoside phosphonates is further organized into: 1) synthesis of prodrugs via parent phosphonic acids, 2) synthesis of prodrugs via parent drug esters by trans-esterification, and 3) synthesis of prodrugs starting from cyclic phosphonate moiety.
1.1. Synthesis of Prodrugs via Parent Phosphonic Acids:
Phosphonate prodrugs are synthesized by reaction of the dichlorophosphonate generated in situ and an alcohol. For example, the reaction of dichlorophosphonate of MPO3H21 with substituted 1,3-diols in the presence of a base (such as pyridine, triethylamine, etc) yields compounds of Formula I - III (Khamnei, et al., J.
Med. Chem.
39:4109 (1996)).
HO
MP03H2 + Formula I - III
HO

V

Such reactive dichlorophosphonate intermediates can be prepared from the corresponding phosphonic acids and chlorinating agents, e.g. thionyl chloride (Starrett, et al., J. Med. Chem. 1857 (1994)), oxalyl chloride (Stowell, et al., Tetrahedron Lett.
31:3261 (1990)), and phosphorus pentachloride (Quast, et al., Synthesis 490 (1974)).
Alternatively, these dichlorophosphonates can also be generated from disilyl phosphonate esters (Bhongle, et al., Synth. Commun. 17:1071 (1987)) and dialkyl phosphonate esters (Still, et al., Tetrahedron Lett. 24:4405 (1983); Patois, et al., Bull.
Soc. Chim. Fr. 130:485 (1993)).
Alternatively, these prodrugs are prepared from phosphonic acids by coupling with diols under Mitsunobu reaction conditions (Mitsunobu, Synthesis 1 (1981);
Campbell, J. Org. Chem. 52:6331 (1992)), and other acid coupling reagents including, but not limited to, carbodiimides (Alexander, et al., Collect. Czech. Chem.
Commun.

59:1853 (1994); Casara, et al., Bioorg. Med. Chem. Lett. 2:145 (1992); Ohashi, et al., Tetrahedron Lett. 29:1189 (1988)), and benzotriazolyloxytris-(dimethylamino) phosphonium salts (Campagne, et al., Tetrahedron Lett. 34, 6743 (1993)).
Phosphonic acids also undergo cyclic prodrug formation with cyclic acetals or cyclic ortho esters of 1 -substituted propane-1,3-diols to result in prodrugs as in the case of carboxylic acid esters (Brechbuhler, et al.., Hely. Chim. Acta. 48:1746 (1965)).
Alternatively, more reactive cyclic sulfites or sulfates are also suitable as coupling precursors when reacted with phosphonic acid salts. These precursors can be made from the corresponding diols as described in the literature.
O
>-Y + MP03H2 -- - Formula I - II I E MP03H2 -I- ~g 0 ~0 X

Y = -NMe2, -OMe; X = O or null Optically pure prodrugs and corresponding salts are also made starting from optically pure diols which are synthesized as described in the following section 2. These prodrug syntheses are accomplished in three steps from parent phosphonic acids.
In the first step, chlorination of PME, PMP, HPMP analogues, is achieved using oxalyl chloride and N,N-diethylformamide to give N-protected-dichloridate. A
variety of other chlorinating agents such as thionyl chloride, phosphorus pentachloride in presence of N,N-dialkyl formamides are also used for the purpose. N,N-dialkylformamide used in the chlorination step not only forms a Vilsmeyer chlorinating agent, but also protects the NH2 group. The protected chloridate intermediate results in favorable solubility properties necessary to improve the overall yield and diastereomeric ratio of the product.
Use of other protecting groups, such as acyl, alkoxycarbonyl, aryloxycarbonyl, aralkyloxycarbonyl, Fmoc, etc., may also enhance the recovery and diastereomeric ratio of the desired product.
Coupling of chloridate intermediate and chiral alcohol in the presence of a base (e.g., triethylamine) in dichloromethane at lower temperature results in a protected intermediate. Deprotection of the protected intermediate under mild acidic conditions, in presence of ethanol-acetic acid followed by acidification (e.g., methanesulfonic acid) gives rise to prodrug as a crystalline salt with high chemical purity. A
second crystallization of this material to further purify from trans isomer in solvents such as ethanol gives prodrug in >96% diastereomeric purity. The use of other acids, including and not limited to, mineral acids, such as, sulfuric, nitric, hydrochloric, phosphoric, sulfonic acids, such as, alkyl and aryl sulfonic acids, and carboxylic acids, such as, tartaric, citric, maleic, malic, malonic, lactic, oxalic acids and the like, may lead to better recovery and isomeric ratio of the product.

1.2. Synthesis of Prodrugs via Parent Drug Esters by Trans-Esterification:
Phosphonate esters, such as 5 are also utilized in preparation of prodrugs by trans esterification reaction with 1-substituted propane diol under suitable conditions. Mixed anhydride of parent phosphonic acids generated in situ under appropriate conditions react with diols to give prodrugs as in the case of carboxylic acid esters (Inanaga, et al., Bull.
Chem. Soc. Jpn. 52:1989 (1979)). The resulting derivatives are unmasked to give the required prodrugs. Aryl esters of phosphonates are also known to undergo transesterification with alkoxy intermediates (Moriarty, et al., Tetrahedron Lett. 38:2597 (1997); Tawfik, et al., Synthesis 968 (1993)). Generation of the prodrugs under such mild conditions may be advantageous in improving diastereomeric ratio.
HO
MP03R2 + Formula I - III
HO
V

R= -C(O)OR', aryl etc., 1.3. Synthesis of Prodrugs Starting from Cyclic Phosphonate Moiety:
Prodrug moiety can also be introduced at an earlier stage in the synthesis.
Advantageously, such syntheses allow definition of the stereochemistry of prodrugs earlier in the synthesis. Phosphonate intermediate 6 containing a leaving group X may be utilized in the case of phosphonomethylenoxyethyl substituted drugs 8 (Chu, et al., J.
Het. Chem. 23:289 (1986)). The cyclic phosphonate prodrug moiety 6 may be synthesized via diol and phosphonic acid fragment as described in the earlier sections or by formylation of a cyclic phosphite followed by conversion of a hydroxyl group to a leaving group (Phillion, et al., Tetrahedron Lett. 27:1477 (1986)). Such stereodefined intermediates can be coupled under mild conditions to appropriately masked hydroxyethylsubstituted bases 7 under mild conditions to prepare prodrugs of PME, PMP or HPMP derivatives.
Such convergent synthesis can also be achieved via intermediate 10, which upon coupling with an appropriately masked base 9 results in the required prodrug 8.
Intermediate 10 containing cyclic prodrug moiety may be synthesized via Arbuzov reaction of cyclic Arbuzov chlorophosphonate (Holy, et al., Antiviral Res.
13:295 (1990)) with corresponding chloromethyl ether derivatives.

O N\V o' O N\V X.O ,~~\U
X~\\o0D" / + L0H 0~ BH + O~\\,.0 l 6 7 8 9 10 R R R
J
X = Leaving group, B = nucleoside base, R = H, Me, -CH2OP, P = H, Protective group Prodrug stereochemistry (cis- and trans-) is defined by chemical shift of benzylic methine proton in proton NMR spectra. The cis- isomer benzylic methine proton is consistently more deshielded and downfield (with a difference of 0.2 ppm) than corresponding trans- isomer. The difference is enhanced in polar NMR solvents such as DMSO-d6. The isomers can also be differentiated by phosphorus NMR chemical shifts.
2. Synthesis of 1-(Aryl)-Propane-1,3-Diols:
A variety of synthetic methods are known to prepare 1,3-diols. These suitable methods are divided into two types as following: 1) synthesis of racemic 1-(aryl)-propane-1, 3-diol; 2) synthesis of chiral 1-(aryl)-propane-1, 3-diol.

2.1. Synthesis of Racemic 1-(Aryl)-Propane-1,3-Diols:
1,3-Dihydroxy compounds can be synthesized by several well known methods in literature. Substituted aromatic aldehydes are utilized to synthesize racemic 1-(aryl)-propane-1,3-diol via addition of lithium enolate of alkyl acetate followed by ester reduction (path A) (Turner, J. Org. Cheat. 55:4744 (1990)). Alternatively, aryl Grignard additions to 1 -hydroxy propan-3-al also give 1-aryl-substituted propan-1,3-diols (path B). This method will enable conversion of various substituted aryl halides to arylsubstituted- 1,3 -propane diols (Coppi, et al., J. Org. Chern. 53:911 (1988)). Aryl halides can also be used to synthesize 1 -substituted propane diols by Heck coupling of 1,3-diox-4-ene followed by reduction and hydrolysis (Sakamoto, et al., Tetrahedron Lett.
33:6845 (1992)). Pyridyl, quinoline, isoquinoline propan-3-ol derivatives can be oxygenated to 1-substituted-l,3-diol by N-oxide formation followed by rearrangement in acetic anhydride conditions (path C) (Yamamoto, et al., Tetrahedron 37:1871 (1981)).
A variety of aromatic aldehydes can also be converted to 1-substituted-1, 3-diols by vinyl Grignard addition followed by hydroboration reaction (path D).

VCHO + CH3CO2R A B VMX + OHCCH2CH2OR' HO

HO~
V

VCHO + CH2=CHMX VCH2C i 2CH2OH
V = Aryl, R = Alkyl, R' = benzyl, M=Mg or Li, X=Halide or null 2.2. Synthesis of Chiral 1-(aryl)-Propane-1,3-Diols:
A variety of known methods for chiral resolution of secondary alcohols via chemical or enzymatic agents may be utilized for preparation of diol enantiomers (Harada, et al., Tetrahedron Lett. 28:4843 (1987)). Transition metal catalyzed hydrogenation of substituted 3-aryl-3-oxo propionic acids or esters is an efficient method to prepare R or S isomers of optically pure beta hydroxy acids or esters (Comprehensive Asymmetric Catalysis, Jacobsen, E. N., Pfaltz, A., Yamamoto, H. (Eds), Springer, (1999); Asymmetric Catalysis in Organic Synthesis, Noyori, R., John Wiley, (1994)).
These beta hydroxy acid or ester products can be further reduced to give required chiral 1-(aryl)-propane-1,3-diols. (path A). The (3-keto acid or ester substrates for high pressure hydrogenation or hydrogen transfer reactions may be prepared by a variety of methods such as condensation of acetophenone with dimethylcarbonate in the presence of a base (Chu, et al., .T. Het. Chem. 22:1033 (1985)), by ester condensation (Turner, et al., J. Org. Chem. 54:4229 (1989)) or from aryl halides (Kobayashi, et al., Tetrahedron Lett. 27:4745 (1986)). Alternatively, enantiomerically pure 1,3-diols can be obtained by chiral borane reduction of (3-hydroxyethyl aryl ketone derivatives or (3-keto acid derivatives (path B) (Ramachandran, et al., Tetrahedron Lett. 38:761 (1997)).
In another method, commercially available cinnamyl alcohols may be converted to epoxy alcohols under catalytic asymmetric epoxidation conditions. These epoxy alcohols are reduced by Red-Al to result in enantiomerically pure 1,3-diols (path C) (Gao, et al., J.
Org. Chem.
53:4081 (1980)). Aldol condensation is another well described method for synthesis of the chiral 1,3-oxygenated functionality starting from aromatic aldehydes.
(path D) (Mukaiyama, Org. React. 28:203 (1982)).

VCOCH2CO2R VCOCH2R' A B/
HO or HOD

HO HO
V V
Z'Ie D
VCHO VCH=CHCH2OH
V = Aryl, R = Alkyl or H, R'= -CH2OH, CO2R
Formulations Compounds of the invention are administered orally in a total daily dose of about 0.01 mg/kg/dose to about 30 mg/kg/dose, preferably from about 0.05 mg/kg/dose to about 10 mg/kg/dose. The most preferred dose range is from 0.1 to 3 mg/kg. The use of time-release preparations to control the rate of release of the active ingredient may be preferred. The dose may be administered in as many divided doses as is convenient.
Compounds of this invention when used in combination with other antiviral agents or oncolytic agents may be administered as a daily dose or an appropriate fraction of the daily dose (e.g. bid). Administration of the prodrug may occur at or near the time in which the other antiviral or oncolytic agent is administered or at a different time.
For the purposes of this invention, the compounds may be administered by a variety of means including orally, parenterally, by inhalation spray, topically, or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles.
The term parenteral as used here includes subcutaneous, intravenous, intramuscular, and intraarterial injections with a variety of infusion techniques. Intraarterial and intravenous injection as used herein includes administration through catheters. Oral administration is generally preferred.
Pharmaceutically acceptable salts include acetate, adipate, besylate, bromide, camsylate, chloride, citrate, edisylate, estolate, fumarate, gluceptate, gluconate, glucoranate, hippurate, hyclate, hydrobromide, hydrochloride, iodide, isethionate, lactate, lactobionate, maleate, mesylate, methylbromide, methylsulfate, napsylate, nitrate, oleate, pamoate, phosphate, polygalacturonate, stearate, succinate, sulfate, sulfosalyicylate, tannate, tartrate, terphthalate, tosylate, and triethiodide.
Oxalate salts are not pharmaceutically acceptable, but oxalate salts can be used as intermediates. As intermediates, oxalate salts often improve yields.
Pharmaceutical compositions containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and, preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.

Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives.
Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase maybe a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents.
Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain 20 to 2000 mol (approximately 10 to mg) of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions.
It is preferred that the pharmaceutical composition be prepared which provides easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion should contain from about 0.05 to about 50 mol (approximately 0.025 to 25 mg) of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
As noted above, formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid;
or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be administered as a bolus, electuary or paste.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethyl cellulose),, lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methylcellulose in varying proportions to provide the desired release profile.
Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach. This is particularly advantageous with the compounds of Formula 1 when such compounds are susceptible to acid hydrolysis.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Preferred unit dosage formulations are those containing a daily dose or unit, daily sub-dose, or an appropriate fraction thereof, of a drug.
It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion;
other drugs which have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those skilled in the art.

EXAMPLES
The compounds used in this invention and their preparation can be understood further by the examples, which illustrate some of the processes by which these compounds are prepared. These examples should not however be construed as specifically limiting the invention and variations of the compounds, now known or later' developed, are considered to fall within the scope of the present invention as hereinafter claimed.
Compounds of Formula I-III are prepared according to the literature procedures with modifications and additions well understood by those skilled in the art.
The TLC
conditions given are utilizing plates of Analtech UNIPLATE, silica gel GHLF, scored 10 X 20 cm, 250 micron.

SYNTHESIS OF RACEMIC 1-(ARYL)PROPANE-1,3-DIOLS:

Example 1: Preparation of 1-(2' Furanyl) Propane 1,3 Diol via Grignard Addition and Hydroboration:
To a solution of 2-furaldehyde (3 g, 31.2 mmol) in THE (60 mL) was added 1M
vinyl magnesium bromide in THE (34 mL) at 0 C. After stirring for an hour, a solution of 1M BH3 THE complex in THE was added. The reaction was quenched with 3N
NaOH (20 mL) and 30% hydrogen peroxide (10 mL) at 0 C. The organic fraction was separated and concentrated. The crude product was chromatographed by eluting with 5% methanol-dichloromethane to give 1-(2'-furyl)propane-1,3-diol (1 g, 22%).
Example 2: Preparation of 1-(2' Pyridyl) Propane-1,3Diol via Benzylic Oxidation:
Step A: (J. Org. Client. 22:589 (1957)) To a solution of 3-(2'-pyridyl)propane-l-ol (10g, 72.9mmol) in acetic acid (75mL) was added 30% hydrogen peroxide slowly. The reaction mixture was heated to 80 C for 16h. The reaction was concentrated under vacuum and the residue was dissolved in acetic anhydride (100mL) and heated at 110 C overnight. Acetic anhydride was evaporated upon completion of the reaction. Chromatography of the mixture by eluting with methanol-methylene chloride (1:9) resulted in 10.5g (60%) of pure diacetate.
Step B:
To a solution of diacetate (5g, 21.1mmol) in methanol-water (3:1, 40mL) was added potassium carbonate (14.6g, 105.5mmol). After stirring for 3h at room temperature, the reaction mixture was concentrated. The residue was chromatographed by eluting with methanol-methylene chloride (1:9) to give 2.2g (68%) of crystalline diol.
Example 3: Preparation Oft-(Aryl) Propane-1,3 Diol from Propane 1,3-Diol via Grignard Addition:
Step A: (J. Org. Chem. 53:911 (1988)) To a solution of oxalyl chloride (5.7 mL, 97 mmol) in dichloromethane (200 mL) at -78 C was added dimethyl sulfoxide (9.2 mL, 130 mmol). The reaction mixture was stirred at -78 C for 20 min before addition of 3-(benzyloxy)propan-l-ol (11 g, 65 mmol) in dichloromethane (25 mL). After an hour at -78 C, reaction was quenched with triethylamine (19 mL, 260 mmol) and warmed to room temperature. Work-up and column chromatography by elution with dichloromethane resulted in 8 g (75%) of (b enzyloxy)propan- l -al.
Step B:

To a solution of 3-(benzyloxy)propan-l-al (1 g, 6.1 mmol) in THE at 0 C was added a 1M solution of 4-fluorophenylmagnesium bromide in THE (6.7 mL, 6.7 mmol).
The reaction was warmed to room temperature and stirred for lh. Work-up and column chromatography by elution with dichloromethane resulted in 0.7 g (44%) of alcohol.

Step C:
To a solution of benzyl ether (500 mg) in ethyl acetate (10 mL) was added 10%
Pd(OH)2-C (100 mg). The reaction was stirred under hydrogen gas for 16h. The reaction mixture was filtered through celite and concentrated. Chromatography of the residue by elution with ethyl acetate-dichioromethane (1:1) resulted in 340mg (79%) of product.
Example 4: General Procedure for Preparation oft Aryl Substituted Propane-1,3 Diol From Aryl Aldehyde:
Step A: Q. Org. Chem. 55:4744 (1990)) To a -78 C solution of diisopropylamine (2 mmol) in THE (0.7 ml/mmol diisopropylamine) was slowly added n-butyllithium (2 mmol, 2.5M solution in hexanes).
The reaction was then stirred for 15 min at -78 C before a solution of ethyl acetate (2 mmol) in THE (0.14 ml/mmol ethyl acetate) was slowly introduced. After stirring an additional 30 min at - 78 C, aTHF solution containing the aryl aldehyde (1.0 mmol in 0.28 ml THF) was added. The reaction was then stirred at -78 C for 30 min, warmed to room temperature and stirred an additional 2 h. After aqueous work up (0.5 M
HCl), the organic layer was concentrated to a crude oil (beta-hydroxyester).
Step B:
The crude hydroxyester was dissolved in ether (2.8 ml/mmol), cooled to ice bath temperature, and lithium aluminum hydride (3 mmol) was added batch wise. The reaction was stirred allowing the cooling bath to melt and the reaction to reach room temperature. After stirring overnight at room temperature, the reaction was cooled back to ice bath temperature and'quenched with ethyl acetate. Aqueous work up (0.5M
HCl) afforded the crude diol, which was purified either by chromatography or distillation.

SYNTHESIS OF CHIRAL 1-(ARIL)-PROPANE-1,3-DIOLS:
Example 5 General Procedure for resolution ofracemic 1,3-dials:
Racemic diols synthesized as in examples 1-4 may be resolved to result in chiral form as described in the following procedure.
Step A:
To a solution of diol (1.0 mmole) in THE (1.0 ml) was added hexamethyldisilazide (2.1 mmole) followed by a catalytic amount of trimethylsilyltriflate (2 - 3 drops). After stirring at room temperature for 1 h, the reaction was diluted with hexane (4 ml) and subjected to work up with ice-cold water. The resulting disilylether was either purified by chromatography or, if sufficiently pure, used crude in the next reaction.
Step B:
To a solution of disilylether (1.0 mmole) and (-)-menthone (1.1 mmole) in dichloromethane (2.0 nil) at -40 C, was slowly added trimethylsilyltriflate (0.11 mmole). The reaction was then kept at -50 to -60 C for 48 h, at which time pyridine was added to quench the reaction. After warming to room temperature, the crude mixture was diluted with hexane (4.0 ml) and subjected to aqueous work up. The two ketals were separated by chromatography.
Step C:
The separated ketals were hydrolyzed by adding a catalytic amount of concentrated hydrochloric acid to a methanol (4.0 ml/mmole) solution of each.
After stirring overnight at room temperature, the methanol was removed under vacuum and the residue was subjected to aqueous work up. The resolved diols were further purified by either chromatography or distillation.
Example 6: Synthesis of Chiral 3-(3 '.-Chloropbenyl)-1,3-Dibydoxypropane via Sharpless Asymmetric Epoxidation:
Step A:
To a dispersion of m-chloro-cinnamic acid (25 g, 137 mmol) in ethanol (275 mL) was added conc. sulphuric acid (8 mL) at room temperature. The reaction was refluxed overnight and concentrated. Ice-cold water was added to the crude and precipitated white solid was filtered and washed with cold water. Precipitate was dried under vacuum overnight to give 25 g (87%) of ester. (R 0.50 in dichloromethane on silica) Step B:
To a solution of m-chlorocinnamic acid ethyl ester (23g, 109.5mmol) in dichloromethane at-78 C was added 1M DIBAL-H in dichloromethane (229mL, 229 mmol) dropwise over 1h. The reaction was stirred at -78 C for an additional 3h.
Ethylacetate was added to quench excess DIBAL-H and saturated aq. potassium sodium tartrate was added and stirred reaction at room temperature for 3h. Organic layer was separated and salts were washed with ethyl acetate. Combined organic portions were concentrated and distilled at 120 C/0.1mm to give 14g (76%) of pure allylic alcohol.
(Rf=0.38 in 1:1 ethylacetate:hexane on silica) Step C:
To a solution of in-chlorocinnamyl alcohol (5g, 29.76 mmol) in dichloromethane (220 mL) was added activated 4A molecular sieves powder (2.5 g) and the mixture was cooled to -20 T. (+)-Diethyl tartrate (0.61 mL, 3.57 mmol) was added at -20 C
and stirred for 15min before adding titanium tetraisopropoxide (0.87, 2.97 mmol).
The reaction was stirred for additional 30 min and 5-6M t-butylhydroperoxide in heptane (10 mL, 60 mmol) was added dropwise while maintaining the internal temperature at -20 to -25 T. The mixture was stirred for additional 3 h at -20 C and 10% sodium hydroxide in saturated aq sodium chloride (7.5 mL) followed by ether (25 mL) was added.
The reaction was warmed to 10 C and stirred for 15 min before adding anhydrous magnesium sulphate (10g) and celite (1.5g). The mixture was further stirred for additional 15 min, filtered and concentrated at 25 C to give crude epoxy alcohol.
(Rf-~-0.40 in 1:1 ethylacetate:hexane on silica).
Step D:
To a solution of crude m-chloroepoxycinnamyl alcohol obtained from earlier reaction in dimethoxyethane (300 mL) was added a 65% Red-Al solution in toluene (18.63 mL, 60 mmol) dropwise under nitrogen at 0 C. After stirring at room temperature for three hours, the solution was diluted with ethyl acetate (400 mL) and quenched with aq saturated sodium sulphate solution (50 mL). After stirring at room temperature for 30 min, the resulting white precipitate formed was filtered and washed with ethyl acetate. The filtrate was dried and concentrated. The crude product was distilled at 125-130 C/0.lmm to give 3.75g (67%) of pure (R)-3-(3'-chlorophenyl)-l,3-dihydoxypropane. (R 0.40 in 1:1 ethylacetate:dichloromethane) Optical purities were defined as diacetates (prepared by treatment of diols with acetic anhydride, triethylamine, cat.DMAP in dichloromethane) by HPLC ((S,S) Whelko-0, 2.5 cmX4.Omm ID purchased from Regis).
(R)-3-(3'-chlorophenyl)-1,3-dihydoxypropane: 91% ee (+)Diisopropyltartrate provided >96% ee of (R)-3-(3'-chlorophenyl)-1,3-dihydoxypropane.
(S)-3-(3'-Chlorophenyl)-1,3-dihydoxypropane was also prepared under identical conditions via asymmetric epoxidation and reduction protocol utilizing (-)-tartrate in similar yields.

(S)-3-(3'-Chlorophenyl)-1,3-dihydoxypropane: 79% ee Example 7: Synthesis of Chiral 3-(3'-Cbloropbenyl)-1,3 Dhydoxypropane via Hydrogen Transfer Reaction:
Step A: Preparation of Methyl 3-(3'-Chlorophenyl)-3-Oxo-Propionate:
A 22 L, 3-neck round bottom flask was equipped with a mechanical stirrer, thermowell/ thermometer and nitrogen inlet (bubbler in-line). The flask was flushed with nitrogen and charged sequentially with THE (6 L), potassium t-butoxide (1451 g), and THE (0.5 L). The resulting mixture was stirred at ambient temperature for minutes and a 20 C water bath was applied. A 3 L round bottom flask was charged with 3'-chloroacetophenone (1000 g) and diethylcarbonate (1165 g), and the resulting yellow solution was added slowly to the stirred potassium t-butoxide solution, maintaining the temperature between 16 and 31 C. After the addition was complete (1 h, 10 min.), the cooling bath was removed and the solution was stirred for 1 h, 30 min. TLC
indicated that the reaction was complete. A 5 gallon stationary separatory funnel was charged with ice water (4 L) and concentrated hydrochloric acid (1.3 L of 12 M solution).
The dark red reaction solution was quenched into the aqueous acid and the mixture was stirred for 15 minutes. The layers were separated and the aqueous phase (lower) was extracted again with toluene (4 L). The combined organic extracts were washed with saturated brine (2 X 3 L, 10 minute stirring time each), dried (MgSO4), filtered and concentrated under reduced pressure to provide 1480 g of a brown oil. The oil was placed under high vacuum (10 torr) overnight to give 1427 g. The material was vacuum distilled (short path column, fraction cutter receiver) and the fraction at 108-128 C/1-0.5 torr was collected to provide 1273.9 g of a yellow oil (92.6%). (R0.36 in 20% ethyl acetate/hexanes).
Step B: Preparation of Methyl (S)-3-(3'-Chlorophenyl)-3-Hydroxypropionate:
A 12 L, 3-neck round bottom flask was equipped with a mechanical stirrer, thermometer, addition funnel ( 500 mL) and nitrogen inlet (bubbler in-line).
The flask was flushed with nitrogen and charged with formic acid (292 mL, 350 g).
Triethylamine (422 mL, 306 g) was charged to the addition funnel, then added slowly with stirring, maintaining the temperature <45 C. After the addition was complete (1 h, 30 min.), the solution was stirred with the ice bath applied for 20 min., then at ambient temperature for an additional 1 h. The flask was charged sequentially with methyl 3-(3'-chlorophenyl)-3-oxo-propionate (1260 g), DMF (2.77 L including rinsing volume) and (S,S)-Ts-DPEN-Ru-Cl-(p-cymene) (3.77 g). The flask was equipped with a heating mantle and the addition funnel was replaced with a condenser (5 C circulating coolant for condenser).
The stirred reaction solution was slowly heated to 60 C (90 min. to attain 60 C) and the contents were maintained at 60 C for 4.25 h. HPLC indicated 3% starting material remained. The solution was stirred at 60 C for an additional 8 h, then gradually cooled to ambient temperature overnight. HPLC indicated 0.5% starting material. A 5 gallon stationary separatory funnel was charged with water (10 L) and MTBE (1 L). The reaction solution was poured into the aqueous mixture and the reaction flask was rinsed into the separatory funnel with an additional 1 L of MTBE. The contents were stirred for several minutes and the layers were separated. The aqueous phase was extracted with additional MTBE (2 X 1 L), and the combined organic extracts were washed with brine (1 L), and concentrated under reduced pressure to provide 1334 g of a red oil (105%).
The oil was used without further purification for the next step.
The crude hydroxyester (10 mg, 0.046 mmol) was dissolved in dichloromethane (1 mL). Acetic anhydride (22 .IL, 0.23 mmol) and 4-(dimethylamino)pyridine (22 mg, 0.18 mmol) were added and the solution was stirred at ambient temperature for 15 min.
The solution was diluted with dichloromethane (10 mL) and washed with 1 M
hydrochloric acid (3 X 3 mL). The organic phase was dried (MgSO4), filtered and concentrated under reduced pressure. The residual oil was dissolved in methanol and analyzed by chiral HPLC (Zorbax Rx-C18, 250 X 4.6 mm; mobile phase: 65/35 (v/v) water/acetonitrile, isocratic; flow rate= 1.5 mL/min; inj. volume= 15 L;UV
detection at 220 nm. Retention times: Product= 9.3 min, starting material= 17.2 min.). The hydroxyester was derivatized to the acetate for analysis by chiral HPLC and shown to give 91% ee. (HPLC conditions: Column: Pirkle covalent (S,S) Whelk-O 10/100 krom FEC, 250 X 4.6 mm; mobile phase: 70/30 (v/v) methanol/water, isocratic; flow rate: 1.5 mL/min;inj. volume= 10 .tL; UV detection at 220 nm. Retention times: S-hydroxyester (acetate)= 9.6 min, R-hydroxyester (acetate)= 7.3 min.) Step C: Preparation of (S)-3-(3'-Chlorophenyl)-3-hydroxypropanoic acid:
To the crude hydroxyester in a 10 L rotary evaporator flask was added sodium hydroxide solution (2.5 L of 2 M solution). The resulting solution was stirred on the rotary evaporator at ambient pressure and temperature for 2 h. HPLC indicated 5%
starting material still remained (HPLC conditions: Column: Zorbax Rx-C 18, 250 X 4.6 mm; mobile phase: 65/35 (v/v) water/acetonitrile, isocratic; flow rate= 1.5 mL/min; inj.
volume= 15 L; UV detection at 220 nm. Retention times: Product=3.8 min, starting material= 18.9 min.). The pH of the solution was 11 (wide range pH paper).
Additional 2 M NaOH solution was added to adjust the pH to 14 (approx. 100 mL), and the solution was stirred for an additional 30 min. HPLC indicated the reaction was complete. The solution was transferred to a 5 gallon stationary separatory funnel and extracted with MTBE (2 L). The layers were separated and the organic extract was discarded.
The aqueous phase was transferred back to the separatory funnel and acidified with HCl solution (600 mL). The mixture was extracted with MTBE (1 X 2 L, 2 X 1 L).
The combined acidic organic extracts were dried (MgSO4), filtered and concentrated under reduced pressure to give 1262 g of a brown, oily semi-solid. The residue was slurried with ethyl acetate (1 L) and transferred to a 12 L, 3-neck round bottom flask equipped with a mechanical stirrer, heating mantle, condenser and thermometer. The stirred mixture was heated to dissolve all solids (28 C) and the dark solution was cooled to 10 C (a precipitate formed at 11 C). The mixture was slowly diluted with hexanes (4 L
over 1 h) and the resulting mixture was stirred at <10 C for 2 h. The mixture was filtered and the collected solid was washed with cold 4/1 hexanes/ethyl acetate (1 L), and dried to constant weight (-30 in. Hg, 50 C, 4 h). Recovery= 837 g of a beige solid (70.4%). mp=94.5-95.5 C
A 50 mg sample of hydroxyacid was reduced to the diol with borane-THF (see Step D). The resulting crude diol was diacetylated (as described in Step B)) and analyzed by chiral HPLC Retention times: S-diol (diacetate)= 12.4 min, R-diol (diacetate)= 8.8 min.) ee = 98%
A second crop of hydroxyacid was isolated. The filtrate from above was concentrated under reduced pressure to give 260 g of a brown sludge. The material was dissolved in ethyl acetate (250 mL) and the stirred dark solution was slowly diluted with hexanes (1000 mL) and the resulting mixture was stirred at ambient temperature overnight. The mixture was filtered and the collected solid was washed with hexanes/ethyl acetate (200 mL), and dried to constant weight (-30 in. Hg, 50 C, 16 h).
Recovery= 134 g of a beige solid (11.2%). ee = 97%

Step D: Preparation of (S)-(-)-1-(3'-Chlorophenyl)-1,3-Propanediol:
A 22 L, 3- neck round bottom flask was equipped with a mechanical stirrer, thermowell/thermometer and nitrogen inlet (outlet to bubbler). The flask was charged with 2M borane-TIF (3697 g, 4.2 L) and the stirred solution was cooled to 5 C. A
solution of (S)-3-(3-chlorophenyl)-3-hydroxypropanoic acid (830 g) in THE
(1245 mL) was prepared with stirring (slightly endothermic). The reaction flask was equipped with an addition funnel (1 L) and the hydroxyacid solution was slowly added to the stirred borane solution, maintaining the temperature < 16 C. After the addition was complete (3 h), the mixture was stirred at ice bath temperature for 1.5 h. The reaction was quenched by careful addition of water (2.5 L). After the addition was complete (30 min), 3M
NaOH solution (3.3 L) was added (temperature increased to 35 C) and the resulting mixture was stirred for an additional 20 minutes (temperature= 30 C). The reaction mixture was transferred to a 5 gallon stationary separatory funnel and the layers were separated. The aqueous phase was extracted with MTBE (2.5 L) and the combined organic extracts (THF and MTBE) were washed with 20 wt% NaCl solution (2 L) and stirred with MgSO4 (830 g) for 30 minutes. The mixture was filtered through Celite and concentrated under reduced pressure to provide 735 g of a thick, brown oil.
The oil was purified by vacuum distillation and the fraction at 135-140 C/
0.2 mmHg was collected to provide 712.2 g of a colorless oil (92.2%).
The diol was diacetylated and analyzed by chiral HPLC (e.e.= 98%) (see Step B). Retention times: S-diol (diacetate)= 12.4 min, R-diol (diacetate)= 8.9 min. [a]D= -51.374 (5 mg/mL in CHC13) Example S. Synthesis of Chiral 3-(3'--chloropheayl)-1,3-inYbydroxypropaae via (-) ;B chlorodiisopinocampheylborane (DIPCI) Reduction Step A: Preparation of 3-(3'-Chlorophenyl)-3-oxo-propanoic acid:
A 12 L, 3-neck round bottom flask was equipped with a mechanical stirrer and addition funnel (2 L). The flask was flushed with nitrogen and charged with diisopropylamine (636 mL) and THE (1.80 L). A thermocouple probe was immersed in the reaction solution and the stirred contents were cooled to -20 C. n-Butyllithium (1.81 L of a 2.5 M solution in hexanes) was charged to the addition funnel and added slowly with stirring, maintaining the temperature between -20 and -28 C.
After the addition was complete (30 min), the addition funnel was rinsed with hexanes (30 mL) and the stirred solution was cooled to -62 C. Trimethylsilyl acetate (300 g) was added slowly with stirring, maintaining the temperature <-60 C. After the addition was complete (30 min), the solution was stirred at -60 C for 15 min. 3-Chlorobenzoyl chloride (295 mL) was added slowly with stirring, maintaining the temperature <-60 C.
After the addition was complete (65 min), the cooling bath was removed and the reaction solution was stirred for 1.25 h, with gradual warming to 0 C. The reaction flask was cooled with an ice bath, then water (1.8 L) was added to the stirred solution.
The reaction mixture was stirred for 10 minutes, then diluted with t-butyl methyl ether (1.0 L). The lower aqueous phase was separated and transferred to a 12 L, 3-neck round bottom flask equipped with a mechanical stirrer. t-Butyl methyl ether was added (1.8 L) and the stirred mixture was cooled to <10 C (ice bath). Concentrated HCl solution (300 mL of 12 M solution) was added and the mixture was vigorously stirred. The layers were separated and aqueous phase was further acidified with conc. HCl (30 mL) and extracted again with t-butyl methyl ether (1.0 L). The combined MTBE extracts were washed with brine (1 L), dried (MgSO4, 70 g), filtered and concentrated under reduced pressure to give 827 g of a yellow solid. The crude solid was slurried in hexanes (2.2 L) and transferred to a 5 L, 3-neck round bottom flask equipped with a mechanical stirrer. The mixture was stirred at <10 C (ice bath) for 1 h, then filtered, washed with hexanes (4 X
100 mL) and dried to constant weight (-30 in. Hg, ambient temperature, 14 h).
Recovery = 309 g of a pale yellow powder (68.6%).
Step B: Preparation of (S)-3-(3'-Chlorophenyl)-3-Hydroxypropanoic Acid:
A 12 L, 3-neck round bottom flask was equipped with a mechanical stirrer and addition funnel (1 L). The flask was flushed with nitrogen and charged with 3-(3'-chlorophenyl)-3-oxo-propanoic acid (275.5 g) and dichloromethane (2.2 L). A
thermocouple probe was immersed in the reaction slurry and the stirred contents were cooled to -20 C. Triethylamine (211 mL) was added over 5 minutes to the stirred slurry and all solids dissolved. A dichloromethane solution of (-)-B-chlorodiisopinocampheylborane (1.60 M, 1.04 L) was charged to the addition funnel, then added slowly with stirring, maintaining the temperature between -20 and -25 C.
After the addition was complete (35 min), the solution was warmed to ice bath temperature (2-3 C) and stirred for 4 h. An in-process NMR analysis indicated the starting material was <4%. Water (1.2 L) was added to the cloudy orange reaction mixture, followed by 3 M NaOH solution (1.44 L). The mixture was vigorously stirred for 5 min, then transferred to a separatory funnel. The layers were separated and the basic aqueous phase was washed with ethyl acetate (1.0 L). The aqueous phase was acidified with conc. HC1(300 mL) and extracted with ethyl acetate (2 X 1.3 L).
The two acidic ethyl acetate extracts were combined, washed with brine (600 mL), dried (MgSO4, 130 g), filtered and concentrated under reduced pressure to provide 328 g of a yellow oil (the oil crystallized on standing). The solid was slurried in ethyl acetate (180 mL) and transferred to a 2 L, 3-neck round bottom flask, equipped with a mechanical stirrer. The stirred mixture was cooled to <10 C (ice bath), then diluted with hexanes (800 mL).
The resulting mixture was stirred at ice bath temperature for 4 h, then filtered. The collected solid was washed with 4:1 hexanes: ethyl acetate (3 X 50 mL) and dried to constant weight (-30 in. Hg, ambient temperature, 12 h). Recovery= 207.5 g of a white powder (74.5%).
Step C: Preparation of (S)-(-)-1-(3'-Chlorophenyl)-1,3-Propanediol:
A 12 L, 3-neck round bottom flask was equipped with a mechanical stirrer, addition funnel (2 L) and thermometer. The flask was flushed with nitrogen and charged with (S)-3-(3'-chlorophenyl)-3-hydroxypropanoic acid (206.7 g) and THE (850 mL), and the stirred solution was cooled to 5 C (ice bath). A 1M borane in THE
solution (2.14 L) was charged to the addition funnel, then added slowly with stirring maintaining the temperature <10 C. After the addition was complete (1 h), the cooling bath was removed and the solution was stirred at ambient temperature for 1 h. The reaction solution was slowly and cautiously quenched with water (600 mL), followed by 3 M
NaOH solution (850 mL). The mixture was stirred for 10 min, then transferred to a separatory funnel. The layers were separated and the aqueous phase was back extracted with ethyl acetate (600 mL). The combined organic phase was washed with brine (500 mL), dried (MgS04, 322 g), filtered and concentrated under reduced pressure to provide 189.0 g of a pale yellow oil (101%). The oil was purified by vacuum distillation and the fraction at 125-155 C/ 0.15 mmHg was collected to provide 180.9 g of a colorless oil (94.0%).
The diol (5.0 mg, 0.026 mmol) was dissolved in dichloromethane (2.0 mL).
Acetic anhydride (15 L, 0.15 mmol) and 4-(dimethylamino)pyridine (13 mg, 0.10 mmol) were added and the solution was stirred at ambient temperature for 15 min. The reaction solution was quenched with 1 M HC1 solution (3 mL) and the lower organic phase was separated, passed through a plug of MgSO4, and concentrated with a stream of nitrogen. The residue was dissolved in methanol (1 mL) and analyzed by chiral HPLC
(see, Example 7; Step B). ee > 98%.

Example 9: The Preparation of1,3 Diols via Catalytic Asymmetric Hydrogenation:
Step A:
Beta-ketoester starting material was synthesized as described in Example 7, step A.
Step B:

A solution containing beta-ketoester (1 mmole) in either methanol or ethanol (5-10 ml/mmole ketoester) was degassed through several pump/vent (N2) cycles at room temperature. The degassed solution was moved into a glove bag and under an atmosphere of N2 was poured into a stainless steel bomb containing a stir bar and 1.0 mole % Ru-BINAP catalyst. The bomb was sealed, removed from the glove bag and purged with H2 prior to stirring 18-24 h at room temperature and 150 psi 112.
After venting the hydrogen pressure, the bomb was opened and the reaction mixture was removed and concentrated. The crude beta-hydroxyester was used for hydrolysis.
Step C:
Crude beta-hydroxy ester was hydrolyzed as described in Example 7, step C.
Step D:
Optically active beta-hydroxy acid was reduced as described in Example 7, step D.

SYNTHESIS OF PRODRUG VIA COUPLING OF PHOSPHONIC ACIDS AND
1-(ARYL)PROPANE 1,3-DIOLS:
Example 10: General Procedure for Synthesis of Prodrugs by Thionyl Chloride Reaction:
A suspension of 1 mmol of phosphonic acid in 5 mL of thionyl chloride was heated at reflux temperature for 4 h. The reaction mixture was cooled and evaporated to dryness. To the resulting residue was added a solution of 1 mmol of diol and 2.5 mmol pyridine in 3 mL of methylene chloride. After stirring at 25 C for 4 h the reaction was subjected to work up and chromatography.

Example 11: General Procedure for Synthesis of Prodrugs via DCC
Coupling:
To a solution of PMEA (410mg, 1.5mmol) in DMF (15mL) and pyridine (3mL) was added 1,3-dicyclohexylcarbodiimide (DCC) (925mg, 4.5mmol) followed by 1(3-chlorophenyl)propane-l,3-diol from Step B (295mg, 1.57mmol). The reaction mixture was heated overnight at 100 C. The mixture was concentrated under reduced pressure and azeotroped with toluene (2X1OmL). Crude compound was chromatographed on a silica gel column (3:97 to 10:90 methanol-dichloromethane) to result in pure cyclic prodrug (310mg).

SYNTHESIS OF PRODRUGS VIA OXALYL CHLORIDE MEDIATED
COUPLING:
Example 12: Preparation 9-{2-[2,4-cis-(S)-(+)-4-(3'-Cbloropbenyl) 2-oxo-1,3,2-dioxapbospborinan 2 ylJmetboxyetbyl}Adenine MetbaneSulfonate (18).
Example 12.1: Formation ofDichloridate (11) N^NEt2 \N I \IN
N NJ
A 2L, 3-neck round bottom flask was equipped with a mechanical stirrer, condenser, addition funnel (125mL) and heating mantle. The flask was flushed with nitrogen and charged with PMEA (50.0g), dichloromethane (650mL) and N,N-diethylformamide (22.5mL). Oxalyl chloride (58.OmL) was charged to the addition funnel, and added slowly to the stirred reaction mixture. After the addition was complete (15 minutes), the addition funnel was removed and the vigorously stirred mixture was heated at reflux for 2 hours. The solution remained a slurry during this process. The reaction mixture was cooled slightly, and additional oxalyl chloride (1.OmL) and N,N-diethylformamide (0.4ml) were added. The addition of N,N-diethylformamide produced vigorous gas evolution. The resulting mixture was heated at reflux until all solids were dissolved (additional 2.5 hours, total reaction time was approximately 4.5 hours). HPLC

analysis of the reaction solution indicated the product 11 at 83 Area %. The reaction was monitored for formation of the dichloridate. A sample of the reaction mixture (approximately 50 L) was removed and quenched in anhydrous methanol (1L) containing 1 drop of triethylamine. The resulting methyl phosphonate(s) were analyzed by HPLC.
HPLC Conditions:
YMC-Pack R & D, R-33-5 S-5 120A, 250 X 4.6 mm; mobile phase: Solvent A=
20 mM potassium phosphate, pH 6.2; Solvent B= acetonitrile; gradient: 10-60%B/

min., 60-10%B/ 2 min., 10%B/ 3 min.; 1.4 mL/ min.; inj. vol.= 10 AL; UV
detection at 270 nm.
Retention Times:

Dimethylphosphonate 12 = 8.5 min., monomethyl phosphonate 13 = 5.8 min.
N^NEt~ N^NEt /
N N J SIN N N
I
N N
O

O P-OMe O
I O P-OMe OMe SOH
The reaction solution was cooled slightly and the condenser was replaced with a distillation head with thermometer, condenser and collection flask (250 mL).
The reaction solution was heated to reflux and 250 mL of distillate was collected.
The pot solution was diluted with dichloromethane (250 mL) and an additional 250mL of distillate was collected. The distillation head was removed and the reaction flask was placed under nitrogen. The solution was diluted with dichloromethane (100 mL) and cooled to ice bath temperature. HPLC analysis of the reaction solution indicated the product at 89 area %.
HPLC Conditions:
YMC-Pack R & D, R-33-5 S-5 120A, 250 X 4.6 mm; mobile phase: Solvent A=
20 mM potassium phosphate, pH 6.2; Solvent B= acetonitrile; gradient: 10-60%B/

min., 60-10%B/ 2 min., 10%B/ 3 min.; 1.4 mL/ min.; inj. vol.= 10 L; UV
detection at 270 nm.
Retention Times: Product 11 = 8.5 min., starting material = 5.9 min Pyridine (18 mL) was added slowly to the stirred solution. After the addition was complete (5 minutes), the resulting pale orange solution was stored at ice bath temperature until used (30 minutes).
Example 12.2: CouplingReaction A 2L, 3-neck round bottom flask was equipped with a mechanical stirrer, and addition funnel (1L). The flask was flushed with nitrogen and charged with (S)-(-)-(3'-chlorophenyl)- 1,3-propanediol (34.1 g), as a solution in dichloromethane (500mL) and triethylamine (125 ml). A thermocouple probe was immersed in the reaction solution and the stirred contents were cooled to -71 C (dry ice/ isopropanol). The dichloridate solution 11 was charged to the addition funnel, then added slowly with stirring, maintaining the temperature <-67 C. After the addition was complete (1.25 h), the cooling bath was removed and the stirred mixture was warmed to 0 C over 30 min. The reaction mixture was washed with water (550 mL) and the layers were separated.
The dichloromethane phase was diluted with ethyl acetate (500 mL) and washed with 5%
NaCl solution (600 mL). The organic phase was dried (MgSO4, 50 g), filtered through diatomaceous earth (Celite 521), and concentrated under reduced pressure to provide 108g of a dark red sludge. The samples were dissolved in methanol.
HPLC Conditions:
YMC-Pack R & D, R-33-5 S-5 120A, 250 X 4.6 mm; mobile phase: Solvent A=
20 mM potassium phosphate, pH 6.2; Solvent B= acetonitrile; gradient: 10-60%B/

min., 60-10%B/ 2 min., 10%B/ 3 min.; 1.4 mL/ min.; inj. vol.= 10 L; UV
detection at 270 nm.

Retention Times: cis 14 = 12.5 min., trans 15 = 13.0 min.
N^NEt2 N^NEt2 N N < I
N
O N "
O~,,OD CI vP OD
O " LO CI

The material was dissolved in ethanol (500 mL) and transferred to a 2 L round bottom flask equipped with magnetic stirring, condenser and heating mantle.
Acetic acid (55 mL) was added and the red solution was heated at reflux for 8 hours. HPLC
indicated the reaction was complete. The samples were dissolved in methanol.
HPLC Conditions:
YMC-Pack R & D, R-33-5 S-5 120A, 250 X 4.6 mm; mobile phase: Solvent A=
20 mM potassium phosphate, pH 6.2; Solvent B= acetonitrile; gradient: 10-60%B/

min., 60-10%B/ 2 min., 10%B/ 3 min.; 1.4 mL/ min.; inj. vol.= 10 gL; UV
detection at 270 nm.
Retention Tmes: cis 16 = 9.5 min., trans 17 = 9.8 min.

NHZ NHZ
N N
<'N INJ <'N N

O

O~ = Ip'o,= CI OAP' CI
O

Example 12.3: Crystallization of9-{2-[2,4-cis-(S)-(+)-4-(3'-Cblorophenyl) 2-oxo 1,3,2-dioxaphospborinan2 ylJmetboxyetbyl}adenine metbanesulfonate (18) Methanesulfonic acid (21.5 mL) was added and a precipitate formed after 15 min.
The mixture was diluted with ethanol (400 mL) and heated until all solids dissolved (pot temperature= 70 C). The solution was cooled with stirring and a precipitate formed at 46 C. The resulting mixture was stirred for 2 h, with cooling to ambient temperature, then at ice bath temperature for 2.5 h. The mixture was filtered and the collected solid was washed with ethanol (2 X 15 mL) and dried to constant weight (-30 in. Hg, 55 C, 14 h). Recovery= 49.4 g of a white powder 18 (51.9%). The solid contained 6.5 area%
of the trans diastereomer.
Chiral HPLC: Pirkle covalent (S,S) Whelk-O 1 10/100 krom FEC 250 X 4.6 mm;
mobile phase= 55:45, methanol: 0.1 % HOAc in water; isocratic; 1.0 mL/ min.;
inj.
vol.= 10 gL; UV detection at 260 nm; sample preparation= 2.0 mg/ mL in water.
Retention times: cis-(R)-9-(2-Hydroxyethyl)adenine = 24.6 min., trans-(R)-9-(2-diethylphosphonylmethoxyethyl)adenine = 27.5 min., cis-(S)-9-(2-Phosphonylmethoxyethyl)adenine = 18.0 min.

'H NMR (D20) was used to confirm structure of components.
NH2 . MsOH
<N I LN
N N"
011'0 Example 12.4: Recrystalln'ation of 9 {2 [2,4-cis-(S)-(+)-4-(3'-Chlorophenyl) 2-oxo 1,3,2-dioxaphosphorinan 2 yl]methoxyethyl}adenine methanesulfonate (18) A 3L, 3-neck round bottom flask was equipped with a mechanical stirrer, condenser, heating mantle and thermometer. The flask was charged with 2 batches of crude mesylate salt 18 and ethanol (1.4 L). The stirred mixture was heated at reflux (pot temperature was 78 C) until all solids dissolved (approximately 10 minutes).
The stirred mixture was gradually cooled to ambient temperature over 1.5 hours (a precipitate formed at 56 C). The mixture was stirred at ambient temperature for an additional 2 hrs., then filtered. The collected solid was washed with ethanol (2 x 15 mL) and dried to constant weight (-30 in Hg, 65 C, 60 hrs.).
Color: off white granular solid Purity = 97% (HPLC) Optical purity (Chiral HPLC) >99.5%.
M.P.( C): 186.5-188 Specific Rotation (MeOH, 25 C, 589 nn): +16.429 Composition: C, 41.58; H, 4.56; N, 13.37 [Theoretical: C, 41.50; H, 4.53; N, 13.35]
1H NMR (D2O): 5=1.30-1.60 (m, 111), 1.80-1.95 (m, 1H), 2.60 (s, 311), 3.70-3.90 (m, 4H),4.10-4.50 (m, 2H), 4.60 (s, 311), 5.15-5.40 (m, 1H), 6.70-6.80 (in, 2H), 7.00-7.10 (m, 211), 8.00 (s, 1H), 8.10 (s, 1H).
Preferred compounds of the invention are listed in Table I. Table I contains the structural formulas of the V group, nomenclature, and physical data for the preferred compounds.

Examples of use of the method of the invention include the following. It will be understood that these examples are exemplary and that the method of the invention is not limited solely to these examples.
For the purposes of clarity and brevity, chemical compounds are referred to by compound numbers (from the Table above) in the biological examples below.
BIOLOGICAL EXAMPLES
Example A: In vitro Activation ofPMEA Pro drug Analogues by Rat Liver Microsomes.
PMEA prodrug analogues were tested for activation to PMEA in reactions catalyzed by the microsomal fraction of rat liver.
Methods:
Prodrugs (25 and 250 M) were tested for activation by liver microsomes isolated from rats induced with dexamethasone to enhance CYP3A4 activity (Human Biologics Inc., Phoenix AZ). Reactions were conducted in 0.1 M KHH2P04, pH
7.4, in the presence of 2 mM NADPH and liver microsomes (1 mg/mL). Reaction mixtures were incubated for 5 minutes in an Eppendorf Thermomixer 5436 (37 C, speed 6).
Reactions were terminated by the addition of 1.5 volumes of methanol. The resulting extracts were clarified by centrifugation at 14,000 rpm in an Eppendorf microfuge (20 minutes). The supernatants (200 L) were evaporated under vacuum and heat to dryness before resuspending in 80 L of buffer A (see below) for PMEA analysis. Spiked standards of 9-[2-(phosophonomethoxy)ethyl]adenine, (PMEA, lot 980397, Metabasis Therapeutics) were prepared in the same reaction mixture and processed in an identical fashion. After resuspension, samples were analyzed by reverse phase HPLC
(Altima C-18 column) with use of an ion-pairing buffer (Buffer A) consisting of 10mM
ammonium phosphate, 2.5 mM octyltriethylammonium phosphate, pH 5.5. Samples were loaded in Buffer A and eluted with a methanol gradient from 40% to 80% over 20 minutes.
Detection was at 265 nm. The retention time for PMEA was approximately 14.7 minutes.

Results:
Compound Activation Rate, Dex. Rat Liver Activation Rate, Dex. Rat Liver Microsomes, 25 pM (nmol/min/mg) Microsomes, 250 pM (nmol/min/mg) 3 2.2 1.9 4 6.2 10.9 1.3 1.5 8 4 X0.3 18 < 0.5 0.6 Conclusion:

The majority of the prodrugs tested activated to form the compound of interest, 5 PMEA. Activation rates ranged from <0.5 to 6.2 nmoles/min/mg microsomal protein at the lower drug concentration tested (25 M). Compound 4 had the highest rate of conversion to PMEA in this system.

Example B. PMEApp Accumulation in Hepatocytes Following Incubation with PMEA ProdrugAnalogues PMEA prodrugs that showed a sufficiently high level of activation in rat liver microsomes (Example A) were evaluated for their ability to generate PMEApp, a known HBV polymerase inhibitor, in freshly isolated rat hepatocytes. PMEA prodrugs were tested.
Methods:
Hepatocytes were prepared from fed Sprague-Dawley rats (250-300g) according to the procedure of Berry and Friend (Berry, M.N. Friend, D.S., J. Cell Biol.
43:506-520 (1969)) as modified by Groen (Groen, A.K. et al., Eur. J. Biochem 122:87-93 (1982)).
Hepatocytes (60 mg/ml wet weight, >85% trypan blue viability) were incubated in 2 ml of krebs-bicarbonate buffer containing 20 mM glucose, and 1 mg/ml BSA for 4 hours in the presence of 250 gM PMEA prodrugs (from 25 mM stock solutions of prodrugs in methanol). At appropriate time points throughout the incubation (0, 1, 2, 4 hours), 400-l aliquots of the cell suspension were taken and centrifuged through a silicon/mineral oil layer into 10% perchloric acid to extract intracellular nucleotides. The acidic cell extracts were neutralized with 0.3 volumes of 3M KOH/ 3M KH2CO3 before evaluating PMEApp levels by ion exchange HPLC (Hewlett Packard 1050) using a Whatman Partisphere SAX (5 m, 4.6 x 125 mm) column. Samples were loaded onto the column in 0.3M-ammonium phosphate buffer pH 3.5 and eluted from the column with a gradient to 0.8 M ammonium phosphate pH 3.5. The retention time of PMEApp was 18.6 minutes.
Results:
Following the incubation of PMEA prodrugs with primary rat hepatocytes, PMEApp formation was observed over the course of 0-4 hours. The AUC (area under the curve) values for PMEApp levels (0-4h) are shown below. Compound 4, Compound 2, and Compound 1 produced the highest levels of PMEApp.
Compound Rat Hepatocytes, PMEApp formation, 250 NM, AUC 0-4h (nmol/g*h) 1 149.1 2 106.8 3 64.4 4 109.3 7 70.3 Conclusion:
Compounds of this invention show an ability to generate PMEApp in freshly isolated rat hepatocytes.
Example C. PMEApp Accumulation in Liver Organ Following Oral Administration of PMEA Prodrug Analogues to Normal Fasted Rats:
PMEA prodrugs were tested in vivo to identify prodrugs with suitable oral bioavailability and favorable intrahepatic activation rates.
Methods:
Following oral administration of 30-mg/kg prodrug (PMEA equivalents) to normal, fasted rats, liver organ samples were freeze-clamped at 8 hr. Liver organ concentrations of PMEApp were determined following perchloric acid extraction, neutralization by reverse phase HPLC (Hewlett Packard 1050) using a Whatman Partisphere SAX (5 gm, 4.6 x 125 mm) column. Samples were loaded onto the column in 0.3 M ammonium phosphate buffer pH 3.5 and eluted from the column with 0.8 M
ammonium phosphate pH 3.5. The retention time of PMEApp was 18.6 minutes.

Results:

Compound Rats, PMEApp levels in liver organ @ 8hr., 30 mg/kg, p.o.
(nmol/g) 4 6.6 2.6 6 7.1 7 2.2 2.2 1 0.6 Conclusion:
PMEApp was readily detected in liver organ following oral administration of the 5 PMEA prodrugs tested.

Example D: In vitro Activation of Compound 4 by Human Liver Microsomes; CYP3A4 Selectivity of the Reaction.
The kinetics of activation of Compound 4 by the microsomal fraction of human liver were determined. P450 isoform specificity of the reaction was determined by 10 evaluating the effects of the known CYP3A4-selective inhibitor, ketoconazole.
Methods:

Human liver microsomes were purchased from In Vitro Technologies (IVT). Lot 1011 was prepared from a pool of 10 male donors with documented CYP3A4 activity (testosterone 60-hydroxylation rate of 5.7 nmol/mg/min; IVT). Compound 4 (lot 990301) was synthesized at Metabasis Therapeutics and solubilized in methanol.
Ketoconazole was purchased from Research Biochemicals International (lot SJG-597A) and solubilized in methanol. Following a 2-minute preincubation at 37 C, reaction mixtures containing 100 mM KH2PO4, 2 mg/mL human liver microsomes, and 0, 30, 60, 100, 200 and 400 M Compound 4 were started by addition of NADPH to 2 mM.
The reaction was carried out for 10 minutes in an Eppendorf Thermomixer 5436 at 37 C, speed 6. Inhibition studies were performed in the same fashion with 100 M
Compound 4 and ketoconazole concentrations of 0, 0.01, 0.1, 1, 10 and 100 M. Reactions were terminated by the addition of 1.5 volumes of methanol and extracts were pelleted at 14,000 rpm in an Eppendorf microfuge for 20 minutes. The supernatants (200 Al) were evaporated under vacuum and heat to dryness before resuspending in 80 L buffer A (see below). Spiked standards of 9-[2-(phosophonomethoxy)ethyl]adenine, (PMEA, lot 980397, Metabasis Therapeutics) were prepared in the reaction mixture, and quenched and processed in an identical fashion. The ion-pairing buffer (Buffer A) consisted of 10mM ammonium phosphate and 2.5 mM octyltriethylammonium phosphate, pH 5.5.
After resuspension, samples were analyzed for PMEA by reverse phase HPLC.
Samples were loaded onto an Altima C-18 nucleotide column in Buffer A and eluted with methanol at a gradient from 40% to 80% over 20 minutes with detection at 265 nm. The retention time for PMEA using this method was approximately 14.7 minutes.
Results:
Compound 4 was activated to PMEA in human liver microsomes in a time- and protein concentration-dependent manner. A Km of 111 gM and a Vmax of 1.6 nmol/min/mg were determined for the activation of Compound 4 in this system.
At a concentration of 1 M, ketoconazole was found to inhibit 97% of Compound 4 conversion to PMEA (Figure 1).
Conclusion:
The results thus indicate that Compound 4 turnover in human liver microsomes is catalyzed primarily by CYP3A4 and that the prodrug is a good substrate for this isozyme.
Comparative studies were performed with the single enantiomers of Compound 4 (Compound 57 and Compound 54) to select the enantiomer with the most optimal profile in terms of physico-chemical properties, activation in human liver microsomes, and in vivo accumulation of liver PMEApp following oral or intravenous administration to rats.
Example E: Solubility of Compound 4 Enantiomers The solubility of enantiomers Compound 57 and Compound 54 was assessed in water.
Methods:
Five mg of each enantiomer was weighed out in quadruplicate and the appropriate volume of water added to each sample to achieve a final concentration of 50 mg/mL. The samples were vortexed vigorously and sonicated at room temperature for 15 min. After standing at room temperature for 1 hr, the samples were centrifuged on a table-top microfuge (Eppendorf) at top speed for 2 min at room temperature.
The supernatants were then passed through a 0.45 M filter, and the filtrates diluted and analyzed by HPLC against known standards to assess drug concentration. HPLC
analysis was conducted on an HP1090 system with use of a Beckman Ultrasphere column (4.6 x 150 mm, 5 M) at 40 C. The column was eluted at 1.5 mL/min with a linear gradient from 20 mM potassium phosphate buffer pH 6.2 to 80%
acetonitrile over 15 minutes. The column effluent was monitored at 260 mn.
Results:

The R and S enantiomers of Compound 4, i.e., Compound 57 and Compound 54, were roughly of equivalent and high solubility in water.
Solubility In Water Of Compound 57 And Compound 54 Compound Solubility (water) 57 14 mg/ml 54 16 mg/ml Conclusion:
The results indicate that the solubility is not affected by the enantiomeric form.
Example F.= Stability of Compound 4 Enantiomers in Phosphate Buffer.
The stability of enantiomers Compound 57 and Compound 54 was assessed in phosphate buffer at pH 3, 7, and 9 (RT).
Methods:
The stability of the enantiomers was determined at room temperature in 100 mM
potassium phosphate buffer at pH 3, 7, and 9. Solutions (200 jig/mL) of the enantiomers were prepared at each pH and stability analyzed by monitoring prodrug concentration by HPLC at regular intervals for 75 hours. HPLC conditions were identical to those described in Example E.
Results:
The stability of the enantiomers was identical; they exhibited high stability at acidic (3) and neutral (7) pH. Prodrug decomposition (hydrolysis) occurred only under basic conditions (pH 9).
Stability of Compound 57 and Compound 54 in Buffer (pH 3, 7, 9) Buffer (pH 3) Buffer (pH 7) Buffer (pH 9) Compound Stability (RT) Stability (RT) Stability (RT) 57 T90% > 75h T90% > 75h T1/2 = 11.6 h 54 T90% > 75h T90% > 75h T1/2 = 11.6 h Conclusion:
The results indicate that the stability is not affected by the enantiomeric form.
Example G: Stability in Rat and Human Plasma.
The stability of enantiomers Compound 57 and Compound 54 was assessed in rat and human plasma at 37 C.
Methods:
The stability of the enantiomers was determined in duplicate samples in heparinized rat and human plasma at 37 C. At t=0, the blank plasma samples (-1 mL) were spiked separately with each enantiomer to a final drug concentration of 50 gg/mL
and incubated at 37 C. At preselected times, an aliquot of 100 L of the plasma samples was mixed with the quench cocktail (150 AL) consisting of 98%v/v acetonitrile and 2% v/v acetic acid. Extracts were vortexed and centrifuged to remove the precipitate. The supernatant was analyzed for drug by the HPLC method described above in Example E.
Results:
The stability of the Compound 4 enantiomers in heparinized rat or human plasma at 37 C was found to be similar.
Stability of Compound 57 and Compound 54 in Rat and Human Plasma Human Plasma Rat Plasma Compound Stability (37 C) Stability (37 C) 57 T112 = 8.2h T1/2 = 4.9h 54 T112 = 8.6h T1/2 = 3.9h Conclusion:
The stability of these compounds compares favorably to that of bisPOM PMEA, known to decompose rapidly in human serum with a half-life of < 5 minutes (J.
Med.
Chenz. 39:4958-4965 (1996)).
Example H.= In vitro Activation in Human Liver Microsomes The two enantiomers were compared for activation in human liver microsomes.
Methods:
Human liver microsomes were purchased from In Vitro Technologies (IVT1032).
The comparative study was performed at 2mg/mL human liver microsomes, 100mM
KHH2PO4 , 10 mM glutathione, 25 M or 250 M compound, and 2mM NADPH for 0-7.5 minutes in an Eppendorf Thermomixer 5436 at 37 C, speed 6. The reactions were initiated by addition of NADPH following a 2-minute preincubation. Reactions were quenched with 60% methanol at 0, 2.5, 5, and 7.5 minutes. L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl)cysteinylglycine, a glutathione adduct of the by-product of prodrug activation, 3-Cl-phenyl vinyl ketone, was quantified following extraction of the reaction with 1.5 volumes of methanol. The extracted samples were centrifuged at 14,000 rpm in an Eppendorf microfuge and the supernatant analyzed by HPLC for L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl)cysteinylglycine content. Spiked L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl)cysteinylglycine (lot 20000127) standards (1-30 M) were prepared in 2 mg/ml microsomes under reaction conditions and then quenched and processed in an identical fashion to unknown samples.
For HPLC analysis, the loading mobile phase buffer (Buffer A) consisted of a 9:1 ratio (v/v) of 20 mM potassium phosphate, pH 6.2 and acetonitrile. Extract (100 uL) was injected onto a Beckman Ultrasphere ODS column (4.6 x 250 mM, part# 235329). The column was eluted with a gradient to 60% acetonitrile. The elution of L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl)cysteinylglycine (retention time 10.4 minutes) was monitored at 245 nm.
Results:
Activation of Compound 4 Enantiomers in Human Liver Microsomes:
Activation (25 M) Activation (250 1M) Compound (pmol/m min) (pmol/m min) 4 (racemate) 38 283 Conclusion:
Formation of product, L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl) cysteinylglycine was linear with respect to protein concentration and time for these measurements. The S-enantiomer, Compound 54 was the most readily activated enantiomer in vitro in human liver microsomes at both low (25 M) and high (250 M) prodrug concentrations.

Example I: Assessment ofPMEApp Accumulation in Rat Liver Organ with Compound 57 and Compound 54.
A comparison of hepatic PMEApp accumulation following i.v. and oral administration of Compound 54 (S enantiomer of Compound 4) and Compound 57 (R
enantiomer of Compound 4) was performed to identify the enantiomer with the highest oral bioavailability.
Methods:
Following i.v. or oral administration of 30 mg/kg (in terms of PMEA
equivalents) of either enantiomer to normal, fasted rats, liver organ samples were freeze-clamped at 20 min, 1, 3, 5, 8, 12, and 24 hr post dose. Following perchloric acid extraction and neutralization, liver organ samples were analyzed by anion exchange HPLC
(Hewlett Packard 1050) using a Whatman Partisphere SAX column (5 gm, 4.6 x 125 mm). The column was developed with a gradient from 0.3M ammonium phosphate buffer pH
3.5 to 0.8M ammonium phosphate pH 3.5. Detection was at 254 nm. The retention time of PMEApp was 18.6 minutes.
Results:
Conversion to PMEApp in Rat Liver Organ (30 mg/kg PMEA Equivalents) Compound Route AUC PMEApp 0-24h OBAV

57 i.v. 435.1 nmol*h/g 57 P.O. 93.0 nmol*h/g 21%
54 i.v. 684.6 nmol*h/g 54 P.O. 213.2 nmol*h/g 31%
Conclusion:
Compound 54 demonstrated 1.57-fold higher and 2.29-fold higher PMEApp accumulation (AUC) in liver organ following intravenous administration and oral administration, respectively, compared to Compound 57 (Figures 2A and 2B).
Compound 54 had higher oral bioavailability than Compound 57 based on the AUC
of liver organ PMEApp.

Example J. Selection of Compound 54 Salt Form.
Selection of an appropriate Compound 54 salt form was required to aid in selective crystallization of the cis-diastereomer following prodrug coupling.
Methods:
Ten Compound 54 salt forms (L-tartartric, succinic, maleic, L-malic, D-malic, citric, hydrochloric, phosphoric, nitric, methane-sulfonic acids) were evaluated to identify the best salt form to selectively crystallize out the cis-diastereomer from cis:trans mixtures following the prodrug coupling procedure. Solubility and stability of the mesylate salt of Compound 54 were analyzed in water.
Results:
The methane-sulfonic acid form, designated as Compound 56 was identified as the best salt form to selectively crystallize the cis-diastereomer from the initial 75 (cis):
25 (trans) mixture. A single crystallization with mesylate salt improved the ratio to 93 (cis): 7 (trans) mixture, compared to 82 (cis): 18 (trans) for the succinic acid salt or no improvement for the remainder of salt forms. In water, Compound 56 was highly soluble (>300 mg/ml) and dosing solutions (30 and 300 mg/ml) were found to be stable over 5 days at room temperature (< 10% decomposition).
Conclusion:
The methanesulfonic acid salt was the best salt tested to selectively crystallize the cis-diastereomer.

Example K= Determination of Compound 56Kinetic Parameters in Liver Microsomes from Various Species The kinetic parameters of activation of Compound 56, a prodrug of PMEA, were compared in liver microsomes from male and female mouse (CD-1), rat (Sprague-Dawley), dog (beagle), monkey (cynomolgus), woodchuck, and human pools.
Methods:

The P450-catalyzed activation of Compound 56 to PMEA was monitored by a glutathione by-product capture HPLC assay as described in Example H. Kinetic parameters of Km and Vmax were calculated using SigmaPlot Enzyme Kinetics Module v.1.1. The intrinsic clearance (Vm/Km), a measure of catalytic efficiency, was also evaluated. Protein concentrations were determined by a commercially-available Bradford method.

Results:
Kinetic studies show the activation of Compound 56 in various species, including mouse, rat, dog, monkey, human, and woodchuck. For example, the (V,,,ax/Kn,)[[tL/min/mg] values were 74.8 +/- 7.7 (male) and 77.5 +/- 13.0 (female) in monkey; 10.8 +/- 1.2 (male) and 23.2 +/- 1.0 (female) in human; and 31.5 +/-0.8 (male) in woodchuck.
Conclusion:
Kinetic parameters of activation of the prodrug Compound 56 to its parent compound PMEA in microsomal preparations vary by species; it is highest in monkey.
High activation of the prodrug by monkey microsomes justifies the use of this species in toxicological studies. Kinetic parameters in woodchuck were similar to human supporting the potential use of woodchuck for preclinical efficacy studies.
Example L: Assessment Of the Oral Bioavailability of Compound 561n Normal Male Rats.
The oral bioavailability (OBAV) of the highly water-soluble mesylate salt of the S enantiomer of the cis form of Compound 4, i.e., Compound 56, was evaluated in the normal male rat.
Methods:
Compound 56 was solubilized in water for intravenous and oral administration.
The OBAV was assessed by calculating the ratio of the AUC values of the liver organ concentration-time profile of PMEApp following oral and i.v. administration of mg/kg (in terms of PMEA equivalents) of Compound 56 to groups of four rats.
Liver organ samples were taken at 20 min and 1, 3, 5, 8, 12, and 24 hrs following dosing.
Liver organ concentrations of PMEApp were determined as indicated in Example I.
Results:
The liver organ concentration-time profile of PMEApp is shown in Figure 3. The OBAV was estimated to be 42% per the above definition. This value is at minimum equivalent than the OBAV determined for the free base (31%) in Example I.
Conclusion:
Compound 56 showed good bioavailability.

Example M.. PMEApp Accumulation in Hepatocytes folio wing Incubation with Compound 56 andPMEA.
Compound 56 and PMEA, the parent antiviral drug, were evaluated for their ability to generate PMEApp, a known HBV polymerise inhibitor, in freshly isolated rat hepatocytes.
Methods:
Hepatocytes were prepared from fed Sprague-Dawley rats (250-300g) according to the procedure of Berry and Friend (Berry, M.N. Friend, D.S. J. Cell Biol.
43:506-520 (1969)) as modified by Groen (Groen, A.K. et al., Eur. J. Biochem 122:87-93 (1982)).
Hepatocytes (20 mg/ml wet weight, >85% trypan blue viability) were incubated in 2 ml of krebs-bicarbonate buffer containing 20 mM glucose, and 1 mg/ml BSA for 4 hours in the presence of 25 gM PMEA prodrugs (from 25 mM stock solutions of prodrugs in methanol). At appropriate time points throughout the incubation (0, 1, 2, 4 hours), 1600-l aliquots of the cell suspension were taken and centrifuged to pellet hepatocytes.
Hepatocyte pellets were immediately sonicated in 300 l of ice-cold acetonitrile followed by the addition of 200 gl of water. Hepatocyte extracts were centrifuged for 20 minutes in an Eppendorf centrifuge at 4 C and supernatants from the extracts were transferred to a fresh tube. Hepatocyte extract supernatants were evaporated to dryness under vacuum and then resuspended in 200 l of water. PMEApp concentrations were quantified by LC-MS/MS (negative-ion mode) based on authentic PMEApp standards spiked into hepatocyte extracts. Ten microliter samples were loaded onto a Phenomenex Luna 5g C8 column (50x2 mm) in 20mM N-N-dimethylhexylamine, 10mM propionic acid and eluted with a gradient of methanol from 20-64% over 5.5 minutes. An API
2000 triple quadrupole mass spectrometer fitted with a pneumatically-assisted electrospray interface was used to quantify the negative ion (P03") using a Multiple Reaction Monitoring (MRM) technique.
Results:
The AUC O-4h (area under the curve) values for PMEApp levels were 756 nmoles*h/g following incubation with Compound 56 compared to 84 nmoles*h/g following incubation with PMEA (Figure 4).
Conclusion:
These results suggest that Compound 56 generates PMEApp more efficiently than PMEA in isolated rat hepatocytes.

Example N.= Tissue Distribution ofPMEA Follo wing Oral Administration of Compound 4 and BisPOMPMEA.
The liver specificity of Compound 4 [adenine-2, 8_3 H], was compared relative to bisPOM PMEA [adenine-83H] in kidney and small intestine, the organs in which PMEA
toxicities have been reported.
Methods:
Compound 4 [adenine-2, 83H] or bisPOM PMEA [adenine-83H] was administered at 30 mg/kg PMEA equivalents to fasted rats by oral gavage. Total tritium counts were analyzed in solubilized samples of liver organ, kidney, plasma, urine, red blood cells, small intestine, as well as small intestine contents and feces obtained at various time points over 24 hours. Liver organ specificity was assessed by comparison of the temporal profiles of total tritium in liver organ versus kidney, small intestine, plasma, and red blood cells. Metabolite profiles in perchloric acid extracts of liver organ, kidney, small intestine, feces and urine were analyzed by HPLC.
Results:
Compound 4 generated a 3.1-fold higher AUC of PMEA equivalents in liver organ relative to bisPOM PMEA while decreasing the exposure of PMEA
equivalents in kidney (3.8-fold) and small intestine (25-fold) (Figures 5A, 5B and 5C).
Compound 4 and bisPOM PMEA generated similar ratios of PMEA:PMEA-monphosphate:PMEA-diphosphate in the tissues examined; the intact prodrugs, however, were not detected in these tissues. The majority of the Compound 4 dose ('- 60%) was found as intact prodrug in feces after 24 hours. BisPOM PMEA, in contrast, was readily metabolized in the intestinal tract; 43% of the dose was excreted in feces in the form of PMEA.
Liver Organ Targeting Index of Compound 4 and bisPOM PMEA

Liver organ Liver organ selectivity Liver Small selectivity index index (Liver Compound organ Kidney Intestine (Liver organ/Kidney) organ/Small Intestine) bisPOM PMEA 284 742 3206 0.38 0.09 Compound 4 884 196 118 4.51 7.47 Compound 4/
bisPOM PMEA
(fold difference) 3.11 0.26 0.04 11.8 84.38 Conclusion:
These results indicate significant improvements in the liver organ targeting indices (see Table above) for Compound 4 compared to bisPOM PMEA. Compound 4 is thus likely to have a better safety and efficacy profile than bisPOM PMEA.
Example O: Plasma, and Urine Distribution of PMEA Following Oral Administration of Compound 4 and bisPOMPMEA.
In a parallel study, PMEA levels were quantified in plasma and urine following administration of Compound 4 vs. bisPOM PMEA.
Method:
Compound 4 or bisPOM PMEA was administered at 30 mg/kg PMEA
equivalents to fasted rats by oral gavage. PMEA levels (0-12h) were quantified in plasma by fluorescence HPLC following chloroacetaldehyde derivatization. PMEA
levels in urine were quantified 48 hours after treatment.
Plasma drug levels: PMEA was determined in plasma by a modification of the procedure described in the literature (Shaw JP, Louie MS, Krishnamurthy VV, Arimilli MN, Jones RJ, Bidgood AM, Lee WA, & Cundy KC, Drug Metabolism and Disposition, 25:362-366 (1997)). Plasma (100 gL) samples were extracted with 2 volumes of 0.1%
TFA in acetonitrile. Following centrifugation to remove the precipitate, the supernatant was evaporated to dryness. The dried plasma was reconstituted with 200 gL of the derivatization cocktail consisting of 0.34% chloroacetaldehyde in 100 mM
sodium acetate pH 4.5 and incubated at 95 C for 40 min. The samples were evaporated to dryness and reconstituted with mobile phase for analysis by HPLC. The derivatized PMEA was analyzed by an HPLC system (Hewlett Packard 1090) consisting of a Beckman Ultrasphere ODS 4.6 x 150 mm (5 gm) column developed with a 20-min gradient of 0-30% acetonitrile in 20 mM potassium phosphate pH 6.2.
Fluorescence was monitored at an excitation and emission wavelength of 240 and 420 nm, respectively.
The column temperature was 40 C and the flow rate was 1.5 mL/min. The derivatized PMEA was detected and quantified by comparison to an authentic standard prepared in plasma and eluted at approximately 6 minutes. The limit of quantitation of PMEA, was 0.25 gg/mL.
Urinary excretion studies: The percent excretion of PMEA in urine was assessed by a modification of a previously described procedure (Russell JW, Marrero D, Whiterock VJ, & Klunk LJ, J Chromatog, 572:321-326 (1991)). Aliquots (0.25-0.5 mL) of urine samples and spiked standards were incubated with an equal volume of 17% (v/v) of 50% chloroacetaldehyde in 100 mM sodium acetate pH 4.5 at 50 C for 4 hrs.
Following centrifugation, the supernatant was analyzed by the HPLC method described in the previous section.
Results:
The AUCo_12h of PMEA in plasma following Compound 4 administration was 6-fold lower than following bisPOM PMEA administration (3.5 vs 21.3 nmol*h/mL).
The Cmax of PMEA in plasma following Compound 4 administration was 13-fold lower than following bisPOM PMEA administration (0.3 vs 4.1 nmoles/mL). Consistent with the reduced kidney levels of PMEA, urinary levels of PMEA following Compound 4 administration were 5-fold lower than following bisPOM PMEA administration (4.1%
dose vs. 20.6% dose).
Conclusion:
Thus, the reduced systemic PMEA exposure with Compound 4 led to reduced PMEA exposure in kidney, as assessed by urine levels of PMEA, and is therefore expected to lead to reduced PMEA-related toxicity.
Example P: Comparison of Cis (Compound 56) and Trans (9-{2 [2,4-trans-(S)-(+)-4-(3-chloropbenyl) 2-oxo 1,3,2-dioxapbospborinan 2-yl]metboxyethyl}adenine) Prodrugs for Activation in Human Liver Microsomes The activation rates of cis (Compound 56, lot #20010093) and trans (9-{2-[2,4-trans-(S)-(+)-4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl}adenine, (lot 20010195) PMEA prodrugs was compared in order to establish which prodrug diastereomer is activated the most efficiently in human liver microsomes.
Methods:
Human liver microsomes (mixed pool) were purchased from In Vitro Technologies (Lot RQX). The comparative study was performed at 2mg/mL human liver microsomes, 100mM KH2PO4 , 10 mM glutathione, 100 M prodrug, and 2mM
NADPH (used to initiate reaction), for 5 minutes in an Eppendorf Thermomixer 5436 at 37 C, speed 6. Reactions were quenched with 60% methanol. L-Glutamyl-L-(S-(3-oxo-3-(3-chlorophenyl)propyl)cysteinylglycine, a glutathione adduct of the by-product, 3-Cl-phenyl vinyl ketone, was quantified according to the procedures outlined in Example 8 (In Vitro Activation in Human Liver Microsomes).

Results:
The stereochemical purity and activation rates for cis (Compound 56) and trans (9- {2-[2,4-trans-(S)-(+)-4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl} adenine) prodrug configurations are shown in the table below.
The results indicate that the cis (Compound 56) prodrug form is activated in human liver microsomes much more efficiently than the trans form. Activation of the trans form (9-{2-[2,4-trans-(S)-(+)-4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl}adenine) was below the measureable detection limit (.05 nmol/mg/min.).
Compound Stereochemical Purity Activation Rate (nmol/mg/min.) cis- 98.6% cisl 1.4% trans 0.45 0.01 trans- 1.6% cisl 98.4% trans < 0.05 Conclusion:
Thus, Compound 56 was selected as the superior prodrug diastereomer based on its higher rate of bioactivation in human liver microsomes.
Example Q; Non-Toxic Pro drug by-Products in Isolated Rat Hepatocytes Studies were performed in isolated rat hepatocytes to evaluate the effect of Compound 4 and its metabolites on hepato-toxicity parameters in normal and induced primary rat hepatocytes relative to acetaminophen, a drug known to cause acute injury as a result of its microsomal metabolism in the liver.
Method:
Freshly isolated hepatocytes were prepared (according to methods described in example 13) from normal rats or rats pre-treated with dexamethasone. Male Sprague Dawley rats were induced with dexamethasone to increase CYP3A4 enzyme content and accordingly maximize Compound 4 metabolism. Normal fed Sprague-Dawley rats weighing 280-31Og were administered dexamethasone intraperitoneally at 50 mg/kg once a day for four days. Dexamethasone was purchased from Sigma (lot#119H1328) and formulated as a suspension in corn oil (Sigma lot#107H1649) at 50 mg/mL.
On the fifth day rats were used for hepatocyte isolations. Freshly isolated hepatocytes were treated with Compound 4 (250 gM and 1 mM) or acetaminophen (1-10 mm) in a suspension assay for up to 6 hours. Effects on glutathione levels were assessed by the DTNB (Ellman's reagent) method and viability was determined by the trypan blue exclusion assay and hepatic enzyme leakage assays (LDH, AST). The leakage of lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) into the supernatant was measured using colorimetric assay kits purchased from Sigma (#500 and #505).
Both assays were adapted from the protocol insert for a test tube assay to a microliter plate assay.
Results:
Following treatment with 3 mM or 10 mM acetaminophen, glutathione stores were depleted, viability was reduced to about 5% by trypan blue exclusion criteria, and enzyme leakage was increased by greater than two-fold in hepatocytes isolated from both normal and dexamethasone induced rats. In normal hepatocytes, Compound 4 treatment (1mM) caused no significant decrease in glutathione levels and no significant changes with respect to viability or hepatic enzyme leakage. In hepatocytes isolated from dexamethasone pre-treated rats, Compound 4 treatment (250 M, and 1mM) resulted in significant glutathione decreases (250 M= 50% reduction, 1mM Compound 4 >95%
reduction) after a 2-hour incubation, however, no significant changes were observed with respect to viability or hepatic enzyme leakage following glutathione depletion.
Conclusion:
These studies demonstrate that, following extremely high doses of Compound 4, the Compound 4 by-product can reduce intracellular glutathione content in vitro in CYP3A4 induced rat hepatocytes. However, unlike acetaminophen, the Compound 4 by-product/metabolite is not directly cytotoxic to the glutathione-depleted hepatocytes.
Example R: Pharmacokinetics of Compound 4 in Rat.
The pharmacokinetic profile of Compound 4, a cyclic prodrug of PMEA, was compared to bisPOM PMEA following oral administration to rats.
Method:
Rats (n=4 per time point) were administered 30 mg/kg (in terms of PMEA
equivalents) of the prodrugs of Compound 4 and bisPOM PMEA by oral gavage.
Intact prodrug and the metabolite 9-[2-(phosophonomethoxy)ethyl] adenine (PMEA) and the respective mono-acids were measured in the plasma at 0 (predose), 0.33, 1, 3, 5, 8, and 12 hrs post dose by HPLC using fluorescence detection. The pharmacokinetic parameters were calculated based on noncompartmental analysis of the concentration-time profile.
Results:
Following oral administration of 30 mg/kg of Compound 4 (in terms of PMEA
equivalents) to rats, the intact prodrug, Compound 4 and its breakdown products 9-{2-[1-{(3-chlorophenyl)-l-propyl-3-hydroxy}phosphonomethoxy]ethyl} adenine and 9-[2-(phosophonomethoxy)ethyl]adenine (PMEA) were detectable in the plasma. The plasma half-life of Compound 4, 9-{2-[1-{(3-chlorophenyl)-1-propyl-3-hydroxy}phosphonomethoxy]
ethyl}adenine, and 9-[2-(phosophonomethoxy)ethyl]adenine was approximately 3.2, 2.1, and 5.5 hrs, respectively. The Cma,, of PMEA was 0.11 g/mL at a Tmax of 5 hr.
The AUC to 12 hrs was 0.94 mg*hr/mL. Following oral administration of 30 mg/kg of bisPOM PMEA (in terms of PMEA equivalents) to rats, only PMEA was detectable in the plasma with a half-life of 6.1 hrs. The Cmax of PMEA was 1.11 g/mL at a Tmax of 20 min. The AUC to 12 hrs was 5.83 mg*hr/mL.
Conclusions:

Compared to bisPOM PMEA, the systemic exposure of PMEA as measured by plasma AUC and Cmax was substantially reduced in the rat when delivered as the Compound 4 prodrug. Intact Compound 4 was observed in plasma whereas bisPOM PMEA was not detected, consistent with literature reports (Noble et al., Drugs 58:3 (1999)) that claim bisPOM PMEA is rapidly hydrolyzed and not detected in vivo.
These results indicate that Compound 4 had a preferable pharmacokinetic profile by circulating longer as intact prodrug, while systemic PMEA exposure was minimized, compared to bisPOM PMEA.
Example S. Pharmacokinetics of Compound 4 in Dog.
The pharmacokinetics (PK) and oral bioavailability (OBAV) of Compound 4 were determined in the beagle dog and compared to a literature report for bisPOM PMEA.
Method:
Following an i.v. bolus and oral administration of 8 and 40 mg/kg (prodrug equivalent dose), respectively, of Compound 4 in PEG-400 to male beagle dogs (n=4), blood samples were collected at the following times: 5, 15, 30, 45 min and 1, 2, 4, 6, 8, 12, and 24 hrs post each dosing occasion. In addition, urine was collected over the following intervals: 0-12,12-24,24-48, and 48-72 hrs. Intact Compound 4 and PMEA
were measured in the plasma and urine samples using an LC-MS/MS method. PK
parameters were determined by non-compartmental analysis. Animals were observed for signs of ill health and mortality.
Results:
Compound 4 was well tolerated by the beagle dogs with no sign of ill health or mortality. Following i.v. administration of Compound 4, the plasma concentrations of intact Compound 4 declined mono-exponentially with a mean terminal elimination half-life t1/2 of 0.8 hr. Compound 4 was cleared from the plasma at 1.77 L/hr/kg and was widely distributed with a volume of distribution (Vss) of 1.70 L/kg. The mean residence time (MRT;,) was estimated to be 1.0 hr. Of the administered i.v. dose, approximately 30% was excreted in the urine as intact Compound 4 whereas only 2% was excreted as PMEA. Following oral administration, Compound 4 was rapidly absorbed with a mean peak plasma concentration (C,,,) of 10.2 g/mL ('24 M) achieved after 0.8 hr (TmaX).
The terminal ti/2 after p.o. dosing was 1.0 hr. Of the administered p.o. dose, 40% was excreted in the urine as intact prodrug and 1.7% was excreted as PMEA. The mean plasma Cmaõ of PMEA following oral administration of Compound 4 was 0.24 gg/mL
(0.88 M) at a Tmax of 1.4 hr. The half-life of PMEA after oral dosing of the prodrug was 5.4 hr. OBAV of Compound 4 was estimated to be 112% and 82% based on dose normalized oral-to-i.v. AUC ratios and urinary excretion of Compound 4, respectively.
Conclusions:
Compound 4 was rapidly absorbed (Tmax 0.81 hr) and highly orally bioavailable (%F=100%) in the beagle dog. Compared to literature reports for bisPOM PMEA in dog (Cundy et al., J. Phann Sci. 86:12 (1997)), the systemic exposure of PMEA
as measured by dose-normalized plasma AUC was reduced by 18.8 fold when delivered as the Compound 4 prodrug (PMEA AUC = 10.9 for bisPOM PMEA vs. 0.58 for Compound 4). Intact Compound 4 was observed in plasma whereas bisPOM PMEA was not reportedly detected. These results indicate that Compound 4 had a preferable pharmacokinetic profile by circulating longer as intact prodrug while minimizing systemic PMEA exposure, compared to bisPOM PMEA.

Claims (31)

CLAIMS:
1. A compound of Formula III:

wherein:

M is attached to the phosphorous of the compound of Formula III
through a carbon, an oxygen or a nitrogen atom of the M group, and M is an agent, that when attached to PO3 2- is a biologically active agent in vivo;

and M-PO3H2 is a phosphonic acid residue selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine, (R)-9-(2-phosphonylmethoxypropyl)adenine, 9-(2-phosphonylmethoxyethyl)guanine, 9-(2-phosphonylmethoxyethyloxy)adenine, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine, (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, 9-(3-hydroxy-2-phosphonylmethoxypropyl)guanine and (S)-9-(3-fluoro-2-phosphonylmethoxypropyl)adenine;

V is selected from the group consisting of phenyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furanyl, 3-furanyl, 2-thienyl and 3-thienyl, all optionally substituted with 1 to 3 substituents selected from the group consisting of F, Cl, Br, C1-C3 alkyl, CF3 and OR6;

R6 is selected from the group consisting of C1-C3 alkyl and CF3;
or a pharmaceutically acceptable salt thereof.
2. A compound of claim 1, wherein M-PO3H2 is selected from the group consisting of 9-(2-phosphonylmethoxyethyl)adenine and (R)-9-(2-phosphonylmethoxypropyl)adenine.
3. A compound of claim 1 or 2, wherein V is selected from the group consisting of phenyl, 3-pyridyl and 4-pyridyl, all optionally substituted with 1 or 2 substituents selected from the group consisting of F, Br, Cl, CH3, OCH3 and CF3.
4. A compound of claim 1 or 2, wherein V is selected from the group consisting of 4-pyridyl, 2-bromophenyl and 3-chlorophenyl.
5. A compound of claim 4, wherein said compound is a compound of Formula IV:

wherein:

M-PO3H2 is 9-(2-phosphonylmethoxyethyl)adenine; or a pharmaceutically acceptable salt thereof.
6. A compound of claim 5, wherein the pharmaceutically acceptable salt is formed with an acid selected from the group consisting of acetic acid, hydrobromic acid, hydrochloric acid, citric acid, maleic acid, methanesulfonic acid, nitric acid, phosphoric acid, succinic acid, sulfuric acid and tartaric acid.
7. A compound of claim 6, wherein the pharmaceutically acceptable salt is formed with an acid selected from the group consisting of methanesulfonic acid and succinic acid.
8. The compound of claim 7, wherein the pharmaceutically acceptable salt is formed with methanesulfonic acid.
9. Use of a compound or salt according to any one of claims 1 to 8, in the treatment of a disease associated with the liver selected from the group consisting of hepatitis B, hepatitis C, hepatocellular carcinoma and colorectal carcinoma.
10. Use of a compound or salt according to any one of claims 1 to 8, in the preparation of a medicament for the treatment of a disease associated with the liver selected from the group consisting of hepatitis B, hepatitis C, hepatocellular carcinoma and colorectal carcinoma.
11. Use of a compound or salt according to any one of claims I to 8, together with another antiviral agent, in the treatment of a viral infection of the liver selected from the group consisting of hepatitis B and hepatitis C.
12. Use of a compound or salt according to any one of claims 1 to 8, together with another antiviral agent, in the preparation of a medicament for the treatment of a viral infection of the liver selected from the group consisting of hepatitis B and hepatitis C.
13. The use of claim 11 or 12, wherein said antiviral agent is selected from the group consisting of lamivudine, entecavir, coviracil, DAPD, clevudine, AM365, L-dT, L-dC, ACH 126,443, MCC478, lobucavir, foscarnet, PPA, interferon alpha and pegylated interferon alpha.
14. The use of claim 11 or 12, wherein the compound is a compound of Formula IV:

or a pharmaceutically acceptable salt thereof;

and the antiviral agent is selected from the group consisting of lamivudine, entecavir, coviracil, DAPD, clevudine, AM365, LAT, L-dC, ACH 126, 433, MCC478, lobucavir, foscamet, PPA, interferon alpha and pegylated interferon alpha.
15. The use of claim 14, wherein said viral infection is hepatitis B and the antiviral agent is selected from the group consisting of lamivudine, entecavir, coviracil, DAPD, clevudine, AM365, L-dT, L-dC and ACH 126, 443, or a pharmaceutically acceptable salt thereof.
16. The use of claim 15, wherein the compound of Formula IV is:
or a pharmaceutically acceptable salt thereof;

and said antiviral agent is lamivudine.
17. The use of claim 14, wherein the antiviral agent is selected from the group consisting of interferon alpha and pegylated inteferon alpha.
18. The use of any one of claims 14 to 17, wherein said antiviral agent and said compound of Formula IV are used separately.
19. The use of any one of claims 14 to 17, wherein said antiviral agent and said compound are used simultaneously.
20. A pharmaceutical composition comprising a compound or salt according to any one of claims 1 to 8; an antiviral agent, or a salt thereof; and a pharmaceutically acceptable excipient.
21. Use of a composition of claim 20, as defined in any one of claims 11 to 19.
22. Use of a compound or salt according to any one of claims 1 to 8, together with another oncolytic agent, for the treatment of a cancer associated with the liver selected from the group consisting of hepatocellular carcinoma and colorectal carcinoma.
23. Use of a compound or salt according to any one of claims 1 to 8, together with another oncolytic agent, in the preparation of a medicament for the treatment of a cancer associated with the liver selected from the group consisting of hepatocellular carcinoma and colorectal carcinoma.
24. The use of claim 22 or 23, wherein said oncolytic agent is selected from the group consisting of busulfan, carboplatin, cisplatin, miriplatin, temozolomide, thiotepa, melphalan, ifosfamide, cyclophosphamide, chlorambucil, doxorubicin, duanorubicin, epirubicin, idarubicin, plicamycin, valrubicin, dactinomycin, gemcitabine, floxuridine, fluorouracil, mercaptopurine, thioguanine, methotrexate, mitomycin, etoposide, paclitaxel, docetaxel, irinotecan, topotecan and lurotecan.
25. The use of any one of claims 22 to 24, wherein said cancer is hepatocellular carcinoma.
26. The use of claim 25, wherein said oncolytic agent is selected from the group consisting of doxorubicin, gemcitabine, irinotecan and cisplatin.
27. The use of any one of claims 22 to 26, wherein the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof.
28. A pharmaceutical composition comprising a compound or salt according to any one of claims 1 to 8; an oncolytic agent, or a salt thereof; and a pharmaceutically acceptable excipient.
29. Use of a composition of claim 28, as defined in any one of claims 22 to 27.
30. A compound or salt as defined in any one of claims 1 to 8, or a composition defined in claim 20, for the use as defined in any one of claims 9 to 19.
31. A compound or salt as defined in any one of claims 1 to 8, or a composition defined in claim 28, for the use as defined in any one of claims 22 to 27.
CA2485702A 2002-05-13 2003-05-12 Novel phosphonic acid based prodrugs of pmea and its analogues Expired - Lifetime CA2485702C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US38054502P 2002-05-13 2002-05-13
US60/380,545 2002-05-13
PCT/US2003/014821 WO2004037161A2 (en) 2002-05-13 2003-05-12 Cyclic prodrugs of pmea one of its analogues

Publications (2)

Publication Number Publication Date
CA2485702A1 CA2485702A1 (en) 2004-05-06
CA2485702C true CA2485702C (en) 2011-07-19

Family

ID=32176361

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2485702A Expired - Lifetime CA2485702C (en) 2002-05-13 2003-05-12 Novel phosphonic acid based prodrugs of pmea and its analogues

Country Status (9)

Country Link
US (1) US7214668B2 (en)
EP (2) EP2223927B1 (en)
JP (1) JP4476811B2 (en)
AU (1) AU2003299492B2 (en)
CA (1) CA2485702C (en)
IL (2) IL164809A0 (en)
NZ (1) NZ536328A (en)
TW (1) TWI347325B (en)
WO (1) WO2004037161A2 (en)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6312662B1 (en) * 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
AU2003235501A1 (en) * 2002-05-13 2003-11-11 Metabasis Therapeutics, Inc. PMEA and PMPA cyclic producing synthesis
JP4476811B2 (en) 2002-05-13 2010-06-09 メタバシス・セラピューティクス・インコーポレイテッド New phosphonic acid prodrugs of PMEA and its analogs
BR0315795A (en) * 2002-10-31 2005-09-13 Metabasis Therapeutics Inc Cytarabine Monophosphate prodrugs
US20050239753A1 (en) * 2004-01-21 2005-10-27 Gilead Sciences, Inc. Methods of inhibition of MMTV-like viruses
US20050182252A1 (en) * 2004-02-13 2005-08-18 Reddy K. R. Novel 2'-C-methyl nucleoside derivatives
RU2006138907A (en) 2004-06-08 2008-07-20 Метабазис Терапеутикс SYNTHESIS OF COMPLEX CYCLIC ETHERS USING LEWIS ACID
BRPI0518013A (en) * 2004-11-15 2008-10-21 Ceptyr Inc protein tyrosine phosphatase inhibitors, pharmaceutical composition, complex and methods of use thereof
EP1907391A1 (en) * 2005-07-26 2008-04-09 Hetero Drugs Limited Novel process for acyclic phosphonate nucleotide analogs
US20070042501A1 (en) * 2005-08-20 2007-02-22 Institute Of Nuclear Energy Research A novel technology for the purity assay of TRODAT-1 raw material
EP2020996B1 (en) * 2006-05-16 2011-11-23 Gilead Sciences, Inc. Method and compositions for treating hematological malignancies
CN101371841A (en) * 2007-08-23 2009-02-25 浙江医药股份有限公司新昌制药厂 Crystallization type Entecavir formulation as well as preparation method and use thereof
JP2009196959A (en) * 2008-02-25 2009-09-03 Yamaguchi Univ Pharmaceutical composition for treatment of cancer
CN101475594B (en) * 2009-02-06 2012-05-30 廖国超 Liver targeted antivirus precursor medicament annular phosphoester and use thereof
CN101659676B (en) * 2009-09-07 2012-06-20 徐奎 Sulfo-Adefovir and Tenofovir liver targeting ester prodrug
WO2011133963A2 (en) * 2010-04-23 2011-10-27 University Of Southern California Tyrosine-based prodrugs of antiviral agents
US8884027B2 (en) 2010-10-22 2014-11-11 University Of Rochester Melampomagnolide B derivatives as antileukemic and cytotoxic agents
CN103221037A (en) * 2010-11-12 2013-07-24 葛兰素惠尔康制造业私人有限公司 Novel pharmaceutical compositions
KR101880724B1 (en) 2010-12-10 2018-08-17 시그마팜 래보러토리즈, 엘엘씨 Highly stable compositions of orally active nucleotide analogues or orally active nucleotide analogue prodrugs
WO2012154698A2 (en) 2011-05-06 2012-11-15 Mckenna Charles E Method to improve antiviral activity of nucleotide analogue drugs
WO2013095684A1 (en) 2011-12-22 2013-06-27 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
CN103665043B (en) 2012-08-30 2017-11-10 江苏豪森药业集团有限公司 A kind of tenofovir prodrug and its application in medicine
CN103333209B (en) * 2013-07-05 2015-09-30 西安新通药物研究有限公司 The new crystal of tynofovir prodrug (HTS)
CN104710477A (en) * 2013-12-16 2015-06-17 安徽贝克联合制药有限公司 Tenofovir cyclic phosphonate compound and pharmaceutically acceptable salt thereof, and preparation methods and applications thereof
CN114404427A (en) 2014-02-13 2022-04-29 配体药物公司 Prodrug compound and use thereof
KR102466389B1 (en) 2014-03-14 2022-11-11 올텍 법인회사 Compositions of Selenoorganic Compounds and Methods of Use Thereof
CN106687118A (en) 2014-07-02 2017-05-17 配体药物公司 Prodrug compounds and uses thereof
WO2017048252A1 (en) 2015-09-15 2017-03-23 Alltech, Inc. Compositions of selenoorganic compounds and methods of use thereof
WO2017220028A1 (en) 2016-06-24 2017-12-28 浙江柏拉阿图医药科技有限公司 Liver delivery-based antiviral precursor drug nucleoside cyclophosphate compound and use thereof
CN107540710B (en) * 2016-06-24 2020-10-16 浙江柏拉阿图医药科技有限公司 Liver delivery antiviral prodrug nucleoside cyclic phosphate ester compound and application thereof
CN108863797B (en) * 2018-07-02 2021-01-15 吉首大学 Preparation method of 3- (substituted/unsubstituted phenyl) -3-hydroxy-propionyl hydroxamic acid

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL99688C (en) 1956-12-20
CS233665B1 (en) 1983-01-06 1985-03-14 Antonin Holy Processing of isomere o-phosphonylmethylderivative of anantiomere racemic vicinal diene
US4579849A (en) 1984-04-06 1986-04-01 Merck & Co., Inc. N-alkylguanine acyclonucleosides as antiviral agents
FR2562543B1 (en) 1984-04-10 1987-09-25 Elf Aquitaine NOVEL CYCLIC PHOSPHONITES, THEIR PREPARATION AND APPLICATIONS
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
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
EP0338372A3 (en) 1988-04-22 1991-10-09 American Cyanamid Company Solubilized pro-drugs
ATE128623T1 (en) 1988-08-02 1995-10-15 Nissan Chemical Ind Ltd AGENT FOR IMPROVING DRUG EFFECTS FOR ANTITUMOR DRUGS.
US5658889A (en) 1989-01-24 1997-08-19 Gensia Pharmaceuticals, Inc. Method and compounds for aica riboside delivery and for lowering blood glucose
CS387190A3 (en) 1990-08-06 1992-03-18 Ustav Organicke Chemie A Bioch (2r)-2-/di(2-propyl)phosphonylmethoxy/-3-p-toluenesulfonyloxy -1- trimethylacetoxypropane and process for preparing thereof
DE10399025I2 (en) 1990-09-14 2007-11-08 Acad Of Science Czech Republic Active substance precursors of phosphonates
US5157027A (en) 1991-05-13 1992-10-20 E. R. Squibb & Sons, Inc. Bisphosphonate squalene synthetase inhibitors and method
GB9205917D0 (en) 1992-03-18 1992-04-29 Smithkline Beecham Plc Pharmaceuticals
US5514798A (en) 1993-06-02 1996-05-07 Gilead Sciences, Inc. Method and cyclic carbonates for nucleotide analogues
DE69426904T2 (en) 1993-06-29 2001-10-11 Mitsubishi Chem Corp Phosphonate nucleotide ester derivatives
JP4086314B2 (en) 1993-09-17 2008-05-14 ギリアード サイエンシーズ, インコーポレイテッド Nucleotide analogs
US5567592A (en) 1994-02-02 1996-10-22 Regents Of The University Of California Screening method for the identification of bioenhancers through the inhibition of P-glycoprotein transport in the gut of a mammal
ATE199721T1 (en) 1994-07-04 2001-03-15 Takeda Chemical Industries Ltd PHOSPHONIC ACID COMPOUNDS IN PRODUCTION AND USE
DE4445829A1 (en) 1994-12-22 1996-06-27 Leybold Ag Countercurrent sniffing leak detector
US5716928A (en) 1995-06-07 1998-02-10 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5665386A (en) 1995-06-07 1997-09-09 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
EP0871454B1 (en) 1995-07-17 2003-11-12 Cephalon, Inc. Phosphorous-containing cysteine and serine protease inhibitors
US5962440A (en) 1996-05-09 1999-10-05 Bristol-Myers Squibb Company Cyclic phosphonate ester inhibitors of microsomal triglyceride transfer protein and method
WO1998039342A1 (en) 1997-03-07 1998-09-11 Metabasis Therapeutics, Inc. Novel indole and azaindole inhibitors of fructose-1,6-bisphosphatase
DK0970095T3 (en) 1997-03-07 2004-03-08 Metabasis Therapeutics Inc Novel benzimidazoline inhibitors for fructose-1,6-bisphosphase
AU6452098A (en) 1997-03-07 1998-09-22 Metabasis Therapeutics, Inc. Novel purine inhibitors of fructose-1,6-bisphosphatase
AU747163B2 (en) 1997-07-25 2002-05-09 Gilead Sciences, Inc. Nucleotide analog compositions
US6180666B1 (en) 1997-09-05 2001-01-30 Anmax, Inc. Use of gallic acid esters to increase bioavailability of orally administered pharmaceutical compounds
US5962522A (en) 1997-09-05 1999-10-05 Avmax, Inc. Propyl gallate to increase bioavailability of orally administered pharmaceutical compounds
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
NZ510308A (en) 1998-09-09 2003-06-30 Metabasis Therapeutics Inc Heteroaromatic alkylene substituted phosphonate derivatives useful as inhibitors of Fructose 1,6-bisphosphatase
AU771039B2 (en) 1998-12-24 2004-03-11 Metabasis Therapeutics, Inc. A combination of FBPase inhibitors and insulin sensitizers for the treatment of diabetes
NZ513728A (en) 1999-03-05 2001-09-28 Metabasis Therapeutics Inc Novel phosphorus-containing prodrugs
JP4979866B2 (en) * 1999-09-08 2012-07-18 リガンド・ファーマシューティカルズ・インコーポレイテッド Prodrug for drug delivery specific to the liver
CN1291994C (en) 2000-07-21 2006-12-27 吉里德科学公司 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
AU2003235501A1 (en) * 2002-05-13 2003-11-11 Metabasis Therapeutics, Inc. PMEA and PMPA cyclic producing synthesis
JP4476811B2 (en) 2002-05-13 2010-06-09 メタバシス・セラピューティクス・インコーポレイテッド New phosphonic acid prodrugs of PMEA and its analogs
BR0315795A (en) 2002-10-31 2005-09-13 Metabasis Therapeutics Inc Cytarabine Monophosphate prodrugs
AU2004312546B2 (en) * 2003-12-30 2010-10-14 Gilead Sciences, Inc. Phosphonates, monophosphonamidates, bisphosphonamidates for the treatment of viral diseases
RU2006138907A (en) * 2004-06-08 2008-07-20 Метабазис Терапеутикс SYNTHESIS OF COMPLEX CYCLIC ETHERS USING LEWIS ACID

Also Published As

Publication number Publication date
TW200426152A (en) 2004-12-01
US7214668B2 (en) 2007-05-08
AU2003299492A1 (en) 2004-05-13
CA2485702A1 (en) 2004-05-06
NZ536328A (en) 2007-11-30
EP2223927B1 (en) 2014-10-15
WO2004037161A3 (en) 2005-03-31
IL164809A0 (en) 2005-12-18
IL164809A (en) 2012-08-30
EP2223927A3 (en) 2010-09-15
EP2223927A2 (en) 2010-09-01
EP1532157A4 (en) 2009-02-25
WO2004037161A2 (en) 2004-05-06
JP2006511490A (en) 2006-04-06
AU2003299492B2 (en) 2010-06-10
EP1532157A2 (en) 2005-05-25
US20030229225A1 (en) 2003-12-11
JP4476811B2 (en) 2010-06-09
TWI347325B (en) 2011-08-21

Similar Documents

Publication Publication Date Title
CA2485702C (en) Novel phosphonic acid based prodrugs of pmea and its analogues
CA2171743C (en) Nucleotide analogs
JP4741725B2 (en) New prodrugs for phosphorus-containing compounds
EP1778251B1 (en) Nucleoside phosphonate conjugates as anti hiv agents
JP5063837B2 (en) New phosphorus-containing prodrug
KR101026294B1 (en) Novel Cyclic Phosphate Diesters of 1,3-Propane-1-Aryl Diols and their Use in Preparing Prodrugs
JP2009504677A (en) Novel 2&#39;-C-methyl and 4&#39;-C-methyl nucleoside derivatives
US20070203339A1 (en) Process for Preparation of Cyclic Prodrugs of PMEA and PMPA
US20070179114A1 (en) Novel 2&#39;-C methyl nucleoside derivatives
MX2012011324A (en) Stereoselective synthesis of phosphorus containing actives.
CN103435672A (en) Structure and synthesis of novel nucleoside phosphate prodrug containing substituted benzyl
JP2008508319A (en) Novel nucleoside derivatives
Kasthuri et al. Synthesis and study of (R)-and (S)-β-hydroxyphosphonate acyclonucleosides as structural analogues of (S)-HPMPC (cidofovir)

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20230512