CA2893174A1 - Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same - Google Patents

Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same Download PDF

Info

Publication number
CA2893174A1
CA2893174A1 CA2893174A CA2893174A CA2893174A1 CA 2893174 A1 CA2893174 A1 CA 2893174A1 CA 2893174 A CA2893174 A CA 2893174A CA 2893174 A CA2893174 A CA 2893174A CA 2893174 A1 CA2893174 A1 CA 2893174A1
Authority
CA
Canada
Prior art keywords
activity
target tissue
prodrug
pmpa
prodrugs
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.)
Abandoned
Application number
CA2893174A
Other languages
French (fr)
Inventor
Mark W. Becker
Harlan H. Chapman
Tomas Cihlar
Eugene J. Eisenberg
Gong-Xin He
Michael R. Kernan
William A. Lee
Ernest J. Prisbe
John C. Rohloff
Mark L. Sparacino
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.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22821718&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2893174(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Publication of CA2893174A1 publication Critical patent/CA2893174A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • 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
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity

Abstract

A method is provided for screening prodrugs of methoxyphosphonate nucleotide analogues. The method comprises providing at least one of said prodrugs;
selecting at least one therapeutic target tissue and at least one non-target tissue;
administering the prodrug to the target tissue and to said at least one non-target tissue; and determining the relative activity conferred by the prodrug in the tissues.

Description

Pro drugs of Phosphonate Nucleotide Analogues and Methods for Selecting and Making Same This application relates to prodrugs of methoxyphosphonate nucleotide analogues. In particular it relates to improved methods for making and identifying such prodrugs.
Many methoxyphosphonate nucleotide analogues are known. In general, such compounds have the structure A-OCH2P(0)(0R)2 where A is the residue of a nucleoside analogue and R independently is hydrogen or various protecting or proclrug functionalities. See U.S. Patent Nos. 5,663,159,5,977,061 and 5,798,340, Oliyai et al, "Pharmaceutical Research" 16(11):1687-1693 (1999), Stella et al., "J. Med.
Chem." 23(12):1275-1282 (1980), Aarons, L., Boddy, A. and Petrak, K. (1989) Novel Drug Delivery and Its Therapeutic Application (Prescott, L. F. and Nirnmo, W.
S., ed.), pp. 121-126; Bundgaard, H. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp.

74 and 79-92; Banerjee, P. K. and Amidon, G. L. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 118-121; Notari, R. E. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 135-156; Stella, V. J. and Himmelstein, K. J. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 177-198; Jones, G. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 199-241; Connors, T. A. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 291-316.

Summary of the Invention Prodrugs of methoxyphosphonate nucleotide analogues intended for antiviral or antitumor therapy, while known, traditionally have been selected for their systemic effect. For example, such prodrugs have been selected for enhanced bioavailability, i.e., ability to be absorbed from the gastrointestinal tract and converted rapidly to parent drug to ensure that the parent drug is available to all tissues, However, applicants now have found that it is possible to select prodrugs that become enriched at therapeutic sites, as illustrated by the studies described herein where the analogues are enriched at localized focal sites of HIV
infection.
The objective of this invention is, among other advantages, to produce less toxicity to bystander tissues and greater potency of the parental drug in tissues which are the targets of therapy with the parent methoxyphosphonate nudeotide analogue.
Accordingly, pursuant to these observations, a screening method is provided for identifying a methoxyphosphonate nucleotide analogue prodrug conferring enhanced activity in a target tissue comprising:
(a) providing at least one of said prodrugs;
(b) selecting at least one therapeutic target tissue and at least one non-target tissue;
(c) administering the prodrug to the target tissue and to said at least one non-target tissue; and (d) deterxrtining'the relative antiviral activity conferred by the prodrug in the tissues in step (c).
In preferred embodiments, the target tissue are sites where HIV is actively replicated and/or which serve as an ITN reservoir, and the non-target tissue is an intact animal. Unexpectedly, we found that selecting lymphoid tissue as the target tissue for the practice of this method for HIV led to identification of prodrugs that enhance the delivery of active drug to such tissues.
A preferred compound of this invention, which has been identified by this method has the structure (1),
2 < I
NN"
Ly04, (1) where Ra is H or methyl, and chirally enriched compositions thereof, salts, their free base and solvates thereof.
A preferred compound of this invention has the structure (2) I y NH

(2) and its enriched diasteromers, salts, free base and solvates.
In addition, we unexpectedly found that the chirality of substituents on the phosphorous atom and/or the amidate substituent are influential in the enrichment observed in the practice of this invention. Thus, in another embodiment of this
3 invention, we provide diastereomerically enriched compounds of this invention having the structure (3) B-E-OCH2-P.,111R1 1!1,2 (3) which are substantially free of the diastereomer (4) B-E-OCH2-p-41R1
(4) wherein R1 is an oxyester which is hydrolyzable in rim, or hydroxyl;
B is a heterocyclic base;
2 .
R hydroxyl or the residue of an amino acid bonded to the P atom through an amino group of the amino acid and having each c.arboxy substituent of the amino add optionally esterified, but not both of R1 and R2 are hydroxyl;
E is -(CH2)2-, -CH(CH3)CH2-, -CH(CH2F)CH2-, -CH(CH2OH)CH2-, -CH(CH=CE12)CH2-, -CH(CH)CH2-,-CH(CH2N3)CH2-, ) -CH(R9 )CH20- or -CII(R8)0-, wherein the right hand bond is linked to the heterocyclic base;
the broken line represents an optional double bond;

R4 and Rs are independently hydrogen, hydroxy, halo, amino or a sub stituent having 1-5 carboir atoms selected from acyloxy, alkyoxy, allcylthio, alkylamino and dialkylamino;
4a 6 ' R and R6 are independently H, C1-C6 alkyl, C1-C6 hydroxyalkyl, or C2-C7 alkanoyl;
R7 is independently H, C1-C6 alkyl, or are taken together to form -0- or -CH2-;
R8 is H, C1-C6 alkyl, C1-C6 hydroxyalkyl or C1-C6 haloalkyl; and R9 is H, hydroxymethyl or acyloxymethyl;
and their salts, free base, and solvates.
The diastereomers of structure (3) are designated the (S) isomers at the phosphorus chiral center.
Preferred embodiments of this invention are the diastereomerically enriched compounds having the structure (5a) KNNNN

NH
p6 (5a) which is substantially free of diastereomer (5b) R"
( 1 R

t;.
1\111 7õ,s4r.o." R6 (5b)
5 wherein R5 is methyl or hydrogen;
R6 independently is alkyl, alkenyl, alkynyl, aryl or arylalkyl, or R6 independently is alkyl, alkenyl, alkynyl, aryl or arylalkyl which is substituted with from 1 to 3 substituents selected from alkylarnino, alkylaminoalkyl, dialkylaminoalkyl, dialkylamino, hydroxyl, oxo, halo, amino, alkylthio, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, arylalkoxy, arylalkoxyalkyl, haloalkyl, nitro, nitroall<yl, azido, azidoalkyl, alkylacyl, alkylacylalkyl, carboxyl, or alkylacylamino;
R7 is the side chain of any naturally-occurring or pharmaceutically acceptable amino acid and which, if the side chain comprises carboxyl, the carboxyl group is optionally esterified with an alkyl or aryl group;
R11 is amino, alkylamino, oxo, or dialkylamino; and R12 is amino or H;
and its salts, tautomers, free base and solvates.
A preferred embodiment of this invention is the compound of structure (6), -asopropoxycarbonyflethyllamino]phenoxyphosphinyllmethoxylpropylladenine, also designated herein GS-7340 Na) < I ) Liuo NH
(6) Another preferred embodiment of this invention is the fumarate salt of structure (5) (structure (7)), 9-[(R)-2-R(S)-[[(S)-1-(isopropoxycarbonyl)ethyllaminolphenoxyphosphinylimethoxy]propylladenine fumarate (1:1), also designated herein GS-7340-2 Nfli 2 < 5 N =="14>CO Fl =
______________________________________________________ < 2 P"110 = NH
=

H3C\""r T-=
0 (7) The compounds of structures (1)-(7) optionally are formulated into compositions containing pharmaceutically acceptable excipients. Such =
compositions are used in effective doses in the therapy or prophylaxis of viral (particularly HIV or hepadnaviral) Infections.
In a further embodiment, there is provided a method for preparing 942-(phosphonomethoxy)propyliadenine (PMPA) or (phosphonomethoxy)ethyl]adenine (PMEA) comprising reacting 9-(2-hydroxypropyl)adenine (HPA) or 9-(2-hydroxyethyl)adenine (HEA), magnesium alkoxide, and protected p-toluenesulfonyloxymethylphosphonate.
In a further embodiment, there is provided the use of magnesium alkoxide to prepare 9-[2-(phosphonomethoxy)propyl] adenine (PMPA) or 942-(phosphonomethoxy)ethyl]adenine (PMEA).
In a further embodiment, a method is provided for the facile manufacture of 9[2-(phosphonomethoxy)propylladenine (hereinafter "PMPA" or 9-12- =
(phosphonomethoxy)ethyli adenine (hereinafter "PMEA") using magnesium alkoxide, Which comprises combining 9-(2-hydroxypropyl)adenine or 942-hyciroxyethyl)adenine, protected p-toluenesuifonyloxymethylphosphonate and magnesium alkoxide; and recovering PMPA or PMEA, respectively.
Detailed Description of the Invention The methoxyphosphonate nucleotide analogue parent drugs for use in this screening method are compounds having the structure A-OCH2P(0)(OH)2
7 wherein A is the residue of a nucleoside analogue. These compounds are known per se and are not part of this invention. More particularly, the parent compounds comprise a heterocyclic base B and an aglycon E, in general having the structure l I

OH
7a wherein the group B is defined below and group E is defined above. Examples are described in U.S. Patent Nos. 4,659,825, 4,808,716, 4,724,233, 5,142,051,5,130,427, 5,650,510, 5,663,159, 5,302,585,5,476,938, 5,696,263, 5,744,600, 5,688,778, 5,386,030, 5,733,896, 5,352,786, and 5,798,340, and EP 821,690 and 654,037.
The prodrugs for use in the screening method of this invention are covalently modified analogues of the parent methoxyphosphonate nucleotide analogues described in the preceding paragraph. In general, the phosphorus atom of the parent drug is the preferred site for prodrug modification, but other sites are found on the heterocyclic base B or the aglycon E. Many such prodrugs are already known. Primarily, they are esters or amidates of the phosphorus atom, but also include substitutions on the base and aglycon. None of these modifications per se is part of this invention and none are to be considered limiting on the scope of the invention herein.
The phosphorus atom of the methoxyphosphonate nucleotide analogues contains two valences for covalent modification such as amidation or esterification (unless one phosphoryl hydroxyl is esterified to an aglycon E hydroxyl substituent, whereupon only one phosphorus valence is free for substitution). The esters typically are aryloxy. The amidates ordinarily are naturally occurring monoarnino acids having free carboxyl group(s) esterified with an alkyl or aryl group, usually phenyl, cycloalkyl, or t-, n- or s- alkyl groups. Suitable prodrugs for use in the screening method of this invention are disclosed for example in U.S. Patent No.
5,798,340. However, any prodrug which is potentially believed to be capable of being converted in vivo within target tissue cells to the free methoxyphosphonate nucleotide analogue parent drug, e.g., whether by hydrolysis, oxidation, or other covalent transformation resulting from exposure to biological tissues, is suitable for use in the method of this invention. Such prodrugs may not be known at this time but are identified in the future and thus become suitable candidates available for testing in the method of this invention. Since the prodrugs are simply candidates for screening in the methods their structures are not relevant to practicing or enabling the screening method, although of course their structures ultimately are dispositive of whether or not a prodrug will be shown to be selective in the assay.
8 The pro-moieties bound to the parent drug may be the same or different.
However, each prodrug to be used in the screening assay will differ structurally from the other prodrugs to be tested. Distinct, i.e. structurally different, prodrugs generally are selected on the basis of either their stereochemistry or their covalent structure, or these features are varied in combination. Each prodrug tested, however, desirably is structurally and stereochernically substantially pure, else the output of the screening assay will be less useful. It is of course within the scope of this invention to test only a single prodrug in an individual embodiment of the method of this invention, although typically then one would compare the results with prior studies with other prodrugs.
We have found that the stereochemistry of the prodrugs is capable of influencing the enrichment in target tissues. Chiral sites are at the phosphorus atom and are also found in its substituents. For example, amino acid used in preparing amidates may be D or L forms, and the phosphonate esters or the amino acid esters can contain chiral centers as well. Chiral sites also are found on the nucleoside analogue portion of the molecules, but these typically are already dictated by the stereochemistry of the parent drug and will not be varied as part of the screen. For example the R isomer of PMPA is preferred as it is more active than the corresponding S isomer. Typically these diasteromers or enantiomers will be chirally enriched if not pure at each site so that the results of the screen will be more meaningful. As noted, distinctiveness of stereoisomers is conferred by enriching or purifying the stereoisomer (typically this will be a diastereomer rather than an enantiomer in the case of most methoxyphosphonate nucleotide analogues) free of other stereoisomers at the chiral center in question, so that each test compound is substantially homogeneous. By substantially homogeneous or chirally enriched, we mean that the desired stereoisomer constitutes greater than about 60% by weight of the compound, ordinarily greater than about 80% and preferably greater than about 95%.
9 Novel Screening Method Once at least one candidate prodrug has been selected, the remaining steps of the screening method of this invention are used to identify a prodrug possessing the required selectivity for the target tissue. Most conveniently the prodrugs are labeled with a detectable group, e.g. radiolabeled, in order to facilitate detection later in tissues or cells. However, a label is not required since other suitable assays for the prodrug or its metabolites (including the parent drug) can also be employed. These assays could include mass spectrometry, HPLC, bioassays or immunoassays for instance. The assay may detect the prodrug and any one or more of its metabolites, but preferably the assay is conducted to detect only the generation of the parent drug. This is based on the assumption (which may not be warranted in all cases) that the degree and rate of conversion of prodrug to antivirally active parent diphosphate is the same across all tissues tested.
Otherwise, one can test for the diphosphate.
The target tissue preferably will be lymphoid tissue when screening for prodrugs useful in the treatment of HIV infection. Lymphoid tissue will be known to the artisan and includes CD4 cells, lymphocytes, lymph nodes, macrophages and macrophage-like cells including monocytes such as peripheral blood monocytic cells (PBMCs) and glial cells. Lymphoid tissue also includes non-lymphoid tissues that are enriched in lymphoid tissues or cells, e.g. lung, skin and spleen.
Other targets for other antiviral drugs of course will be the primary sites of replication or latency for the particular virus concerned, e.g., liver for hepatitis and peripheral nerves for HSV. Similarly, target tissues for tumors will in fact be the tumors themselves. These tissues are all well-known to the artisan and would not require undue experimentation to select. When screening for antiviral compounds, target tissue can be infected by the virus.
Non-target tissues or cells also are screened as part of the method herein.
Any number or identity of such tissues or cells can be employed in this regard. In general, tissues for which the parent drug is expected to be toxic will be used as non-target tissues. The selection of a non-target tissue is entirely dependent upon the nature of the prodrug and the activity of the parent. For example, non-hepatic tissues would be selected for prodrugs against hepatitis, and untransformed cells of the same tissue as the tumor will suffice for the antitumor-selective prodrug screen.
It should be noted that the method of this invention is distinct from studies typically undertaken to determine oral bioavailability of prodrugs. In oral bioavailability studies, the objective is to identify a prodrug which passes into the systemic circulation substantially converted to parent drug. In the present invention, the objective is to find prodrugs that are not metabolized in the gastrointestinal tract or circulation. Thus, target tissues to be evaluated in the method of this invention generally do not include the small intestines or, if the intestines are included, then the tissues also include additional tissues other than the small intestines.
The target and non-target tissues used in the screening method of this invention typically will be in an intact living animal. Prodrugs containing esters are more desirably tested in dogs, monkeys or other animals than rodents; mice and rat plasma contains high circulating levels of esterases that may produce a misleading result if the desired therapeutic subject is a human or higher mammal.
It is not necessary to practice this method with intact animals. It also is within the scope of this invention to employ perfused organs, in vitro culture of organs (e.g. skin grafts) or cell lines maintained in various forms of cell culture, e.g.
roller bottles or zero gravity suspension systems. For example, MT-2 cells can be used as a target tissue for selecting HIV prodrugs. Thus, the term "tissue"
shall not be construed to require organized cellular structures, or the structures of tissues as they may be found in nature, although such would be preferred. Rather, the term "tissue" shall be construed to be synonymous with cells of a particular source, origin or differentiation stage.
The target and non-target tissue may in fact be the same tissue, but the tissues will be in different biological status. For example, the method herein could be used to select for prodrugs that confer activity in virally-infected tissue (target tissue) but which remain substantially inactive in virally-uninfected cells (corresponding non-target tissue). The same strategy would be employed to select prophylactic prodrugs, i.e., prodrugs metabolized to antivirally active forms incidental to viral infection but which remain substantially unmetabolized in uninfected cells. Similarly, prodrugs could be screened in transformed cells and the untransformed counterpart tissue. This would be particularly useful in comparative testing to select prodrugs for the treatment of hematological w malignancies, e.g. leukemias.
Without being limited by any particular theory of operation, tissue selective prodrugs are thought to be selectively taken up by target cells and/or selectively metabolized within the cell, as compared to other tissues or cells. The unique advantage of the methoxyphosphonate prodrugs herein is that their metabolism to the dianion at physiological pH ensures that they will be unable to diffuse back out of the cell. They therefore remain effective for lengthy periods of time and are maintained at elevated intracellular concentrations, thereby exhibiting increased potency. The mechanisms for enhanced activity in the target tissue are believed to include enhanced uptake by the target cells, enhanced intracellular retention, or both mechanisms working together. However, the manner in which selectivity or enhanced delivery occurs in the target tissue is not important. It also is not important that all of the metabolic conversion of the prodrug to the parent compound occurs within the target tissue. Only the final drug activity-conferring conversion need occur in the target tissue; metabolism in other tissues may provide intermediates finally converted to antiviral forms in the target tissue.
The degree of selectivity or enhanced delivery that is desired will vary with the parent compound and the manner in which it is measured (% dose distribution or parent drug concentration). In general, if the parent drug already possess a generous therapeutic window, a low degree of selectivity may be sufficient for the desired prodrug. On the other hand, toxic compounds may require more extensive screening to identify selective prodrugs. The relative expense of the method of this invention can be reduced by screening only in the target tissue and tissues against which the parent compound is known to be relatively toxic, e.g. for PMEA, which is nephrotoxic at higher doses, the primary focus will be on kidney and lymphoid tissues.

The step of determining the relative antiviral activity of a prodrug in the selected tissues ordinarily is accomplished by assaying target and non-target tissues for the relative presence or activity of a metabolite of the prodrug, which metabolite is known to have, or is converted to, a metabolite having antiviral or antitumor activity. Thus, typically one would determine the relative amount of the parent drug in the tissues over substantially the same time course in order to identify prodrugs that are preferentially metabolized in the target tissue to an antivirally or antitumor active metabolite or precursor thereof which in the target tissue ultimately produces the active metabolite. In the case of antiviral compounds, the active metabolite is the diphosphate of the phosphonate parent compounds. It is this metabolite that is incorporated into the viral nucleic acid, thereby truncating the elongating nucleic acid strand and halting viral replication.
Metabolites of the prodrug can be anabolic metabolites, catabolic metabolites, or the product of anabolism and catabolism together. The manner in which the metabolite is produced is not important in the practice of the method of this invention.
The method of this invention is not limited to assaying a metabolite which per se possesses antiviral or antitumor activity. Instead, one can assay inactive precursors of the active metabolites. Precursors of the antivirally active diphosphate metabolite include the monophosphate of the parent drug, monophosphates of other metabolites of the parent drug (e.g., an intermediate modification of a substituent on the heterocyclic base), the parent itself and metabolites generated by the cell in converting the prodrug to the parent prior to phosphorylation. The precursor structures may vary considerably as they are the result of cellular metabolism. However, this information is already known or could be readily determined by one skilled in the art.
If the prodrug being assayed does not exhibit antitumor or antiviral activity per se then adjustments to the raw assay results may be required. For example, if the intracellular processing of the inactive metabolite to an active metabolite occurs at different rates among the tissues being tested, the raw assay results with the inactive metabolite would need to be adjusted to take account of the differences among the cell types because the relevant parameter is the generation of activity in the target tissue, not accumulation of inactive metabolites. However, determining the proper adjustments would be within the ordinary skill. Thus, when step (d) of the method herein calls for determining the activity, activity can be either measured directly or extrapolated. It does not mean that the method herein is limited to only assaying intermediates that are active per se. For instance, the absence or decline of the prodrug in the test tissues also could be assayed.
Step (d) only requires assessment of the activity conferred by the prodrug as it interacts with the tissue concerned, and this may be based on extrapolation or other indirect measurement.
Step (d) of the method of this invention calls for determining the "relative"
activity of the prodrug. It will be understood that this does not require that each and every assay or series of assays necessarily must also contain runs with the selected non-target tissue. On the contrary, it is within the scope of this invention to employ historical controls of the non-target tissue or tissues, or algorithms representing results to be expected from such non-target tissues, in order to provide the benchmark non-target activity.
The results obtained in step (d) are then used optimally to select or identify a prodrug which produces greater antiviral activity in the target tissue than in the non-target tissue. It is this prodrug that is selected for further development.
It will be appreciated that some preassessment of prodrug candidates can be undertaken before the practice of the method of this invention. For example, the prodrug will need to be capable of passing largely Immetabolized through the gastrointestinal tract, it will need to be substantially stable in blood, and it should be able to permeate cells at least to some degree. In most cases it also will need to complete a first pass of the hepatic circulation without substantial metabolism.
Such prestudies are optional, and are well-known to those skilled in the art.
The same reasoning as is described above for antiviral activity is applicable to antitumor pro drugs of methoxyphosphonate nucleotide analogues as well.
These include, for example, prodrugs of PMEG, the guanyl analogue of PMEA. In this case, cytotoxic phosphonates such as PMEG are worthwhile candidates to pursue as their cytotoxicity in fact confers their antitumor activity.
A compound identified by this novel screening method then can be entered into a traditional preclinical or clinical program to confirm that the desired objectives have been met. Typically, a prodrug is considered to be selective if the activity or concentration of parent drug in the target tissue (% dose distribution) is greater than 2x, and preferably 5x, that of the parent compound in non-target tissue. Alternatively, a prodrug candidate can be compared against a benchmark prodrug. In this case, selectivity is relative rather than absolute. Selective prodrugs will be those resulting in greater than about 10x concentration or activity in the target tissue as compared with the prototype, although the degree of selectivity is a matter of discretion.
Novel Method for Preparation of Starting Materials or Intermediates Also included herein is an improved method for manufacture of preferred starting materials (parent drugs) of this invention, PMEA and (R)-PMPA.
Typically, this method comprises reacting 9-(2-hydroxypropyl)adenine (HPA) or (2-hydroxyethypadenine (HEA) with a magnesium alkoxide, thereafter adding the protected aglycon synthon p-toluene-sulfonyloxymethylphosphonate (tosylate) to the reaction mixture, and recovering PMPA or PMEA, respectively.
Preferably, HPA is the enriched or isolated R enantiomer. If a chiral HPA
mixture is used, R-PMPA can be isolated from the chiral PMPA mixture after the synthesis is completed.
Typically the tosylate is protected by lower alkyl groups, but other suitable groups will be apparent to the artisan. It may be convenient to employ the tosylate presubstituted with the prodrug phosphonate substituents which are capable of acting as protecting groups in the tosylation reaction, thereby allowing one to bypass the deprotection step and directly recover prodrug or an intermediate therefore.
The alkyl group of the magnesium alkoxide is not critical and can be any C1-C6 branched or normal alkyl, but is preferably t-butyl (for PMPA) or isopropyl (for PMEA). The reaction conditions also are not critical, but preferably comprise heating the reaction mixture at about 70-75 C with stirring or other moderate agitation.
If there is no interest in retaining the phosphonate substituents, the product is deprotected (usually with bromotrimethylsilane where the tosylate protecting group is alkyl), and the product then recovered by crystallization or other conventional method as will be apparent to the artisan.
Heterocyclic Base In the compounds of this invention depicted in structures (3) and (4), the heterocyclic base B is selected from the structures \/R20 0 N1{ R R
Or wherein 15 .
R H, OH, F, Cl, Br, I, OR16, SH, SR16, NH2, or NHR17;
R16 is C1-C6 alkyl or C2-C6 alkenyl including CH3, CH2CH3, CH2CCH, CH2CHCH2 and C3H7;

R is C1-C6 alkyl or C2-C6 alkenyl including CH3, CH2CH3, CH2CCH, CH2CHCH2, and C31-17;
Ris is N, CF, CC1, CBr, CI, CR19, CSR19, or C0R19;
R19 is H, C1-C9 alkyl, C2-C9 alkenyl, C2 - C9 alkynyl, C1-C9 alkyl-C1-C9 alkoxy, or C7-C9 aryl-alkyl unsubstituted or substituted by OH, F, Cl, Br or I, R19 therefore induding -CH3, -CH2CH3, -CHCH2, -CHCHBr, -CH2CH2C1, -CH2CH2F, -CH2CCH, -CH2CHCH2, -C3H7, -CH2OH, -CH2OCH3, -CH20C2H5, -CH2OCCH, -CH2OCH2CHCH2, -CH2C3H7, -CH2CH2OH, -CH2CH2OCH3, -CH2CH20C2H5, -CH2CH2OCCH, -CH2CH2OCH2CHCH2, and -CH2CH20C3F17;
R2 is N or CH;
R21 is N, CH, CCN, CCF3, CCECH or CC(0)NH2;
I222 is H, OH, NH2, SH, SCH3, SCH2CH3, SCH2CCH, SCH2CHCH2, SC3H7, NH(CH3), N(CH3)2, NH(CH2CF13), N(CH2CH3)2, NH(CH2CCH), NH(CH2CHCH2), NH(C3H7), halogen (F, Cl, Br or I) or X wherein X is -(CH2)m(0)n(CH2)mN(R10)2 wherein each m is independently 0-2, n is 0-1, and R1 independently is H, C1-C15 alkyl, C2-C15 alkenyl, C6-C15 arylalkenyl, C6-C15 arylalkynyl, C2-C15 alkynyl, C1-C6-alkylamino-C1-C6 alkyl, C5-C15 aralkyl, C6-C15 heteroaralkyl, C5-C6 aryl, C2-C6 heterocydoalkyl, C2-C15 alkyl, C3-C15 alkenyl, C6-C15 arylalkenyl, C3-C15 alkynyl, C7-C15 arylalkynyl, C1-C6-alkylamino-C1-C6 alkyl, C5-C15 aralkyl, C6-C15 heteroalkyl or C3-C6 heterocycloalkyl wherein methylene in the alkyl moiety not adjacent to N6 has been replaced by -0-, optionally both R1 are joined together with N to form a saturated or unsaturated C2-05 heterocycle containing one or two N heteroatoms and optionally an additional 0 or S heteroatom, or one of the foregoing R1 groups which is substituted with 1 to 3 halo, CN or N3; but optionally at least one R'' group is not H;
fe is H, OH, F, Cl, Br, I, SCH3, SCH2CH3, SCH2CCH, SCH2CHCH2, SC3H7, OR", NH2, NHR17 or le; and R24 is 0, S or Se.
B also includes both protected and unprotected heterocyclic bases, particularly purine and pyrimidine bases. Protecting groups for exocyclic amines and other labile groups are known (Greene et al. "Protective Groups in Organic Synthesis") and include N-benzoyl, isobutyryl, 4,4'-dimethoxytrityl (DMT) and the like. The selection of protecting group will be apparent to the ordinary artisan and will depend upon the nature of the labile group and the chemistry which the protecting group is expected to encounter, e.g. acidic, basic, oxidative, reductive or other conditions. Exemplary protected species are N4-benzoylcytosine, N6-benzoyladenine, N2-isobutyrylguanine and the like.
Protected bases have the formulas Xa.1, Xla.1, Xlb.1, XlIa.1 or XIIIa.1 R,,21 .1==_.-2i Ni:
R20, ')R20 R39%--.'N) RR24 15 0 Nn R23A R2o N R39 R20 N, NN N
(Xa.1) (XIa.1) (Xlb.1) (XIIa. 1) (XIIIa.1) 20 wherein Ria, R20õ, -21, K I224 have the meanings previously defined; RA is R39 or R22 provided that R22 is not NI-12; R23A is R" or eprovided that R23 is not NH2;
R" is NHR", NHC(0)le or CleN(Rm)2 wherein 1236 is C1-C19 alkyl, Ci-C19 alkenyl, C3-Cio aryl, adamantoyl, alkylanyl, or C3-C10 aryl substituted with 1 or 2 atoms or groups selected from halogen, methyl, ethyl, methoxy, ethoxy, hydroxy and cyano;
R" is Ci-Cio alkyl, or both R38 together are 1-morpholino, 1-piperidine or 1-pyrrolidine; Ie is C1-Ci. alkyl, including methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, hexyl, octyl and decanyl; and R41 is hydrogen or CH3.
For bases of structures Xia.1 and Xlb.1, if R39 is present at R
22A or R23A, both 39 groups on the same base will generally be the same. Exemplary R36 R are phenyl, phenyl substituted with one of the foregoing R36 aryl substituents, -CIOH15 (where CioH15 is 2-adamantoy1), -CH2-C6H5, -C6H5, -CH(CH3)2, -CH2CH3, methyl, butyl, t-butyl, heptanyl, nonanyl, undecanyl, or undecenyl.
Specific bases include hypoxanthine, guanine, adenine, cytosine, inosine, thymine, uracil, xanthine, 8-aza derivatives of 2-aminopurine, 2,6-diarninopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza-8-aza W() 02/08241 PCT/US01/23104 derivatives of adenine, guanine, 2-aminopurine, 2,6-diaminopurine, 2-arnino-6-chloropurine, hypoxanthine, inosine and xanthine; 1-deaza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-chloropurine, hypoxanthine, inosine and xanthine; 3-deaza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 6-azacytosine; 5-fluorocytosine; 5-chlorocytosine; 5-iodocytosine; 5-bromocytosine; 5-methykytosine; 5-bromovinyluracil; 5-fluorouracil; 5-chlorouracil; 5-iodouracil; 5-bromouracil; 5-trifluoromethyluracil; 5-methoxymethyluracil; 5-ethynyluracil and 5-propynyluracil.
Preferably, B is a 9-purinyl residue selected from guanyl, 3-deazaguanyl, 1-deazaguanyl, 8-azaguanyl, 7-deazaguanyl, adenyl, 3-deazaadenyl, 1-dezazadenyl, 8-azaadenyl, 7-deazaadenyl, 2,6-diarninopurinyl, 2-aminopurinyl, 6-chloro-2-arninopurinyl and 6-thio-2-aminopurinyl, or a B' is a 1-pyrimidinyl residue selected from cytosinyl, 5-halocytosinyl, and 5-(Cl-C3-alkyl)cytosinyl.
Preferred B groups have the formula \ Z
R22)%
wherein R22 independently is halo, oxygen, NH2, X or H, but optionally at least one R22 is x;
X is ¨(CH2)m(0)n(CH2)mN(R10)2 wherein m is 0-2, n is 0-1, and R10 independently is H, Cl-C15 alkyl, C2-C15 alkenyl, C6-C15 arylalkenyl, C6-C15 arylalkynyl, C2-C15 alkynyl, C1-C6-alkylamino-C1-C6 alkyl, C5-C15 aralkyl, C6-C15 heteroaralkyl, C5-C6 aryl, C2-C6 heterocycloalkyl, C2-C15 alkyl, C3-C15 alkenyl, C6-C15 arylalkenyl, C3-C15 alkynyl, C7-C15 arylalkynyl, Ci-C6-alkylamino-Cl-C6 alkyl, C5-C15 aralkyl, C6-C15 heteroalkyl or C3-C6 heterocycloalkyl wherein methylene in the alkyl moiety not adjacent to N6 has been replaced by -0-, optionally both R10 are joined together with N to form a saturated or unsaturated C2-05 heterocycle containing one or two N heteroatoms and optionally an additional 0 or S heteroatom, or one of the foregoing R10 groups is substituted with Ito 3 halo, CN
or N3; but optionally at least one R10 group is not H; and Z is N or CH, provided that the heterocyclic nucleus varies from purine by no more than one Z.
E groups represent the aglycons employed in the methoxyphosphonate nucleotide analogues. Preferably, the E group is -CH(C1-13)CH,- or -CH,CH,-.
Also, it is preferred that the side groups at chiral centers in the aglycon be substantially solely in the (R) configuration (except for hydroxymethyl, which is the enriched (S) enantiomer).
IV is an in vivo hydrolyzable oxyester having the structure -OR' or -OW
wherein R35 is defined in column 64, line 49 of U.S. Patent No. 5,798,340, herein incorporated by reference, and R6 is defined above. Preferably R1 is aryloxy, ordinarily unsubstituted or para-substituted (as defined in R6) phenoxy.
R2 is an amino acid residue, optionally provided that any carboxy group linked by less than about 5 atoms to the amidate N is esterified. R2 typically has the structure }?.1 ' 0)(\/\( 12 \P13 R
(8) wherein n is 1 or 2;
R11 is le or H; preferably le = C,-C, alkyl; C3-C9 alkyl substituted independently with OH, halogen, 0 or N; C,-C, aryl; C,-C, aryl which is independently substituted with OH, halogen, 0 or N; or C,-C,arylalkyl which is independently substituted with OH, halogen, 0 or N;
R12 independently is H or C1-C9 alkyl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, 0, N, COOR" and halogen; C,-C, aryl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, 0, N, COOR" and halogen; or C,-C, aryl-alkyl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, 0, N, COOR" and halogen;
RI' independently is C(0)-OR11; amino; amide; guanidinyl; imidazolyl;
indolyl; sulfoxide; phosphoryl; C1-C3 alkylamino; C1-C3 alkyldiarnino; C1-C6 alkenylamino; hydroxy; thiol; C1-C3 alkoxy; C1-C3 alkthiol; (CH,)COOR"; C1-C6 alkyl which is unsubstituted or substituted with OH, halogen, SH, NHõ phenyl, hydroxyphenyl or C7-C10 alkoxyphenyl; C2-C6 alkenyl which is unsubstituted or substituted with OH, halogen, SH, NHõ phenyl, hydroxyphenyl or alkoxyphenyl; and C6-C12 aryl which is unsubstituted or substituted with OH, halogen, SH, NHõ phenyl, hydroxyphenyl or C7-C10 alkoxyphenyl; and R14 is H or C1-C9 alkyl or C1-C9 alkyl independently substituted with OH, halogen, COOR", 0 or N; C,-C, aryl; C,-C, aryl which is independently substituted with OH, halogen, COOR", 0 or N; or C,-C, arylalkyl which is independently substituted with OH, halogen, COOR", 0 or N.
Preferably, R11 isC,-C, alkyl, most preferably isopropyl, V is the side chain of a naturally occurring amino acid, n = 1, IV' is H and R14 is H. In the compound of structure (2), the invention includes metabolites in which the phenoxy and isopropyl esters have been hydrolyzed to -OH. Similarly, the de-esterified enriched phosphonoamidate metabolites of compounds (5a), 5(b) and (6) are included within the scope of this invention.
Aryl and "0" or "N" substitution are defined in column 16, lines 42-58, of United States Patent No. 5,798,340.
Typically, the amino acids are in the natural or 1 amino acids. Suitable specific examples are set forth in U. S. Patent No. 5,798,340, for instance Table 4 and col. 8-10 therein.
Alkyl as used herein, unless stated to the contrary, is a normal, secondary, tertiary or cyclic hydrocarbon. Unless stated to the contrary alkyl is C1-C.
Examples are -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2, -CH2CH2CH2CH3), -CH2CH(CH3)2, -CH(CH3)CH2CH3, -C(CH3)3, -C,H2CH2CH2CH2CH3, -CH(CH3)CH2CH2CH3, -CH(CH2CF13)2, -C(CH3)2CH2CH3), -CH(CH3)CH(CH3)2, -CH2CH2CH(CH3)2), -CH2CH(CH3)CH2CH3, -CH2CH2CH2CH2CH2CH3, -CH(CH3)CH2CH2CH2CH3, -CH(CH2CH3)(CH2CH2CH3), -C(CH3)2CH2CH2CH3, -CH(CH3)CH(CH3)CH2CH3, -CH(CH3)CH2CH(CH3)2, -C(CH3)(CH2CH3)2, -CI(CH2CH3)CH(CH3)2, -C(CH3)2CH(CH3)2, and -CH(CH3)C(CH3)3. Alkenyl and alkynyl are defined in the same fashion, but contain at least one double or triple bond, respectively.
Where enol or keto groups are disclosed, the corresponding tautomers are to be construed as taught as well.
The prodrug compounds of this invention are provided in the form of free base or the various salts enumerated in U. S. Patent No. 5,798,340, and are formulated with pharmaceutically acceptable excipients or solvating diluents for use as pharmaceutical products also as set forth in U. S. Patent No.
5,798,340. These prodrugs have the antiviral and utilities already established for the parent drugs (see U. S. Patent 5,798,340 and other citations relating to the methoxyphosphonate nucleotide analogues). It will be understood that the diastereomer of structure (4) at least is useful as an intermediate in the chemical production of the parent drug by hydrolysis in vitro, regardless of its relatively unselective character as revealed in the studies herein.
The invention will be more fully understood by reference to the following examples:

Example la NCN NaOH

Adenine to PMEA using Magnesium Isopropoxide. To a suspension of adenine (16.8g, 0.124 mol) in DMF (41.9 ml) was added ethylene carbonate (12.1g, 0.137 mol) and sodium hydroxide (.100g, 0.0025 mol). The mixture was heated at 130 C
overnight. The reaction was cooled to below 50 C and toluene (62.1 ml) was added. The slurry was further cooled to 5 C for 2 hours, filtered, and rinsed with toluene (2x). The wet solid was dried in yam) at 65 C to yield 20.0g (90%) of 9-(2-hydroxyethypadenine as an off-white solid. Mp: 238-240 C.

I Ts0 (OEt)2 W NH2iPr)2* rr I IS
yov1813r,._ ft N N, NI
80gC N ¨ 0 N 0 it(oEt)2 P(OH)2 9-(2-HydTOXyethyl)adellirle (HEA) (20.0g, 0.112 mol) was suspended in DMF (125 ml) and heated to 80 C. Magnesium isopropoxide (11.2g, 0.0784 mol) , or alternatively magnesium t-butoxide, was added to the mixture followed by diethyl p-toluenesulfonyloxymethylphosphonate (66.0g, 0.162 mol) over one hour. The mixture was stirred at 80 C for 7 hours. 30 ml of volatiles were removed via vacuum distillation and the reaction was recharged with 30 ml of fresh DMF.
After cooling to room temperature, brornotrimethylsilane (69.6g, 0.450 mol) was added and the mixture heated to 80 C for 6 hours. The reaction was concentrated to yield a thick gum. The gum was dissolved into 360 ml water, extracted with 120 ml dichloromethane, adjusted to pH 3.2 with sodium hydroxide, and the resulting slurry stirred at room temperature overnight. The slurry, was cooled to 4 C
for one hour. The solids were isolated by filtration, washed with water (2x), and dried in =

vacuo at 56 C to yield 20g (65.4%) of 9112-(phosphonomethoxy)ethylladenine (PMEA) as a white solid. Mp: > 200 C dec. 1H NMR (D20) = 3.49 (t, 2H); 3.94 (t, 2H); 4.39 (t, 2H); 8.13 (s, 1H); 8.22 (s, 1H).
Example lb OH
N
teX N, 4. NaOH
DMF
N N 1309C \NN N
H /Me H Me Adenine to PMPA using Magnesium t-Butoxide. To a suspension of adenine (40g, 0.296 mol) in DMF (41.9 ml) was added (R)-propylene carbonate (34.5g, 0.338 mol) and sodium hydroxide (.480g, 0.012 mol). The mixture was heated at 130 C
overnight. The reaction was cooled to 100 C and toluene (138 ml) was added followed by methanesulfonic acid (4.7g, 0.049 mol) while maintaining the reaction temperature between 100-110 C. Additional toluene (114 ml) was added to create a homogeneous solution. The solution was cooled to 3 C over 7 hours and then held at 3 C for one hour. The resulting solid was isolated by filtration and rinsed with acetone (2x). The wet solid was dried in vacuo at 80 C to yield 42.6g (75%) of (R)-9-[2-(hydroxy)propyl]adenine (HPA) as an off-white solid. Mp: 188-190 C.
NH, I + Ts0 P(OEt)2 it(F It3t) Ni: N> -r8T)/12rrb, I N>
N N
77gC N N N
yOH i>õ0P(OEt)2 H -?ME) HMe HMe (R)-9-12-(hydroxy)propyl]adenine (HPA) (20.0g, 0.104 mol) was suspended in DMF

(44.5 ml) and heated to 65 C. Magnesium t-butoxide (14.2g, 0.083 mol), or alternatively magnesium isopropoxide, was added to the mixture over one hour followed by diethyl p-toluenesutfonyloxymethylphosphonate (66.0g, 0.205 mol) over two hours while the temperature was kept at 78 C. The mixture was stirred at 75 C for 4 hours. After cooling to below 50 C, bromotrirnethylsilane (73.9g, 0.478 mol) was added and the mixture heated to 77 C for 3 hours. When complete, the reaction was heated to 80 C and volatiles were removed via atmospheric distillation. The residue was dissolved into water (120 ml) at 50 C and then extracted with ethyl acetate (101 m1). The pH of the aqueous phase was adjusted to pH 1.1 with sodium hydroxide, seeded with authentic (R)-PMPA, and the pH of the aqueous layer was readjusted to pH 2.1 with sodium hydroxide. The resulting slurry was stirred at room temperature overnight. The slurry was cooled to 4 C
for three hours. The solid was isolated by filtration, washed with water (60 ml), and dried in vacuo at 50 C to yield 18.9g (63.5%) of crude(R)-942-(phosphonomethoxy)propylladenine (PMPA) as an off-white solid.
The crude(R)-9[2-(phosphonomethoxy)propylladenine was heated at reflux in water (255 ml) until all solids dissolved. The solution was cooled to room temperature over 4 hours. The resulting slurry was cooled at 4 C for three hours.
The solid was isolated by filtration, washed with water (56 ml) and acetone (56 ml), and dried in vacuo at 50 C to yield 15.0g (50.4%) of (R)-942-(phosphonomethoxy)propylladenine (PMPA) as a white solid. Mp: 278-280 C.

Example 2 Preparation of GS-7171 (III) ' Scheme 1 N ..-'=%---.,...-N 0 (--)_N=0=N_-0 1 N
N N 0. T-OH __________________________ I. N N 0 -.P-0 0, Li OH Et3N NMP LI OH
-(anhydrous) .NH2 v..1-y0.y.,- N-1-(---N 0 .
LNN \> 0 ,P-0 =_a22.2,... 0 1).....t_31 4...
'1N>-----i\i 0 T-0 \--/ NH
LI OH cH2a2 m 0 \

NH3)(OH

N--L'',"-I Ni\\ 0 .
N N 0 P"0 0 A N-----------1.N,\ 0 .
1 , .......11 HOy ¨ OH L,,,, 1 / ...,11 ' N N 0 P.10 Li 41 0 Li NH
).-..1. ,,...y ..: s= 0 IV 0 \ V 0 A glass-lined reactor was charged with anhydrous PMPA, (I) (14.6 kg, 50.8 mol), phenol (9.6 kg, 102 mol), and 1-methyl-2-pyrrolidinone (39 kg). The mixture was heated to 85 C and_triethylamine (6.3 kg, 62.3 mol) added. A solution of 1,3-dicyclohexylcarbodiimide (17.1 kg, 82.9 mol) in 1-methyl-2-pyrrolidinone (1.6 kg) was then added over 6 hours at 100 C. Heating was continued for 16 hours. The reaction was cooled to 45 C, water (29 kg) added, and cooled to 25 C. Solids were removed from the reaction by filtration and rinsed with water (15.3 kg). The combined filtrate and rinse was concentrated to a tan slurry under reduced pressure, water (24.6 kg) added, and adjusted to pH = 11 with NaOH (25% in water). Fines were removed by filtration through diatomaceous earth (2 kg) followed by a water (4.4 kg) rinse. The combined filtrate and rinse was extracted with ethyl acetate (28 kg). The aqueous solution was adjusted to pH = 3.1 with (37% in water) (4 kg). Crude II was isolated by filtration and washed with methanol (12.7 kg). The crude II wet cake was slurried in methanol (58 kg).
Solids were isolated by filtration, washed with methanol (8.5 kg), and dried under reduced pressure to yield 9.33 kg II as a white powder: 1H NMR (D20) 8 1.2 (d, 3H), 3.45 (q, 2H), 3.7 (q, 2H), 4 (m, 2H), 4.2 (q, 2H), 4.35 (dd, 2H), 6.6 (d, 2H), 7 (t, 1H), 7.15 (t, 2H), 8.15 (s, 1H), 8.2 (s, 1H); 31P NMR (D20) 8 15.0 (decoupled).
GS-7171 (III). (Scheme 1) A glass-lined reactor was charged with monophenyl PMPA, (II), (9.12 kg, 25.1 mol) and acetonitrile (30.7 kg). Thionyl chloride (6.57 kg, 56.7 mol) was added below 50 C. The mixture was heated at 75 C until solids dissolved. Reaction temperature was increased to 80 C and volatiles (11.4 kg) collected by atmospheric distillation under nitrogen. The pot residue was cooled to 25 C, dichloromethane (41 kg) added, and cooled to -29 C. A solution of (L)-alanine isopropyl ester (7.1 kg, 54.4 mol) in dichloromethane (36 kg) was added over 60 minutes at -18 C followed by triethylamine (7.66 kg, 75.7 mol) over 30 minutes at -18 to -11 C. The reaction mixture was warmed to room temperature and washed five times with sodium dihydrogenphosphate solution (10% in water, 15.7 kg each wash). The organic solution was dried with anhydrous sodium sulfate (18.2 kg), filtered, rinsed with dichloromethane (28 kg), and concentrated to an oil under reduced pressure. Acetone (20 kg) was charged to the oil and the mixture concentrated under reduced pressure. Acetone (18.8 kg) was charged to the resulting oil. Half the product solution was purified by chromatography over a x 38 cm bed of 22 kg silica gel 60, 230 to 400 mesh. The column was eluted with 480 kg acetone. The purification was repeated on the second half of the oil using fresh silica gel and acetone. Clean product bearing fractions were concentrated under reduced pressure to an oil. Acetonitrile (19.6 kg) was charged to the oil and the mixture concentrated under reduced pressure. Acetonitrile (66.4 kg) was charged and the solution chilled to 0 to ¨5 C for 16 hours. Solids were removed by filtration and the filtrate concentrated under reduced pressure to 5.6 kg III as a dark oil; 1H
NMR (CDC13) 5 1.1 (m 121-1), 3.7 (m, 1H), 4.0 (m, 5H), 4.2 (m, 1H), 5.0 (m, 1H), 6.2 (s, 2H), 7.05 (m, 5H), 8.0 (s, IH), 8.25 (d, 1H); 31P NMR (CDC1,3) 621.0, 22.5 (decoupled).
Alternate Method for GS-7171 (HI) Scheme 2 Si o ) ) 0 N N 0 P¨OH it N"---.N
LI OH
.,S, (anhydrous) 0"0 I II

o Hoy_ OH
Nµs N N Pi I
OH

GS-7]

Monophenyl PMPA (H). A round-bottom flask with reflux condenser and nitrogen inlet was placed in a 70 C oil bath. The flask was charged with anhydrous PMPA (I) (19.2 g, 67 mmol), N,N-dimethylformantide (0.29 g, 3.3 mmol), and tetramethylene sulfone (40 mL). Thionyl chloride (14.2 g, 119 mmol) was added over 4 hours. Heating was increased to 100 C over the same time. A homogeneous solution resulted. Phenoxytrimethylsilane (11.7 g, 70 mmol) was added to the solution over 5 minutes. Heating in the 100 C oil bath continued for two hours more. The reaction was poured into rapidly stirring acetone (400 mL) with cooling at 0 C. Solids were isolated by filtration, dried under reduced pressure, and dissolved in methanol (75 mL). The solution pH was adjusted to 3.0 with potassium hydroxide solution (45% aq.) with cooling in ice/water. The resulting solids were isolated by filtration, rinsed with methanol, and dried under reduced pressure to 20.4 g II (Scheme 2) as a white powder.
GS-7171 (III). Monophenyl PMPA (II) (3 g, 8.3 mmol), tetramethylene sulfone (5 mL), and N,N-dimethylformamide (1 drop) were combined in a round bottom flask in a 40 C oil bath. Thionyl chloride (1.96 g, 16.5 mmol) was added. After 20 minutes the clear solution was removed from heat, diluted with dichloromethane (10 ml), and added to a solution of (L)-alanine isopropyl ester (5g, 33 mmol) and diisopropylethylamine (5.33 g, 41 mmol) in dichloromethane (20 mL) at -10 C.
The reaction mixture was warmed to room temperature and washed three times with sodium dihydrogenphosphate solution (10% aq., 10 mL each wash). The organic solution was dried over anhydrous sodium sulfate and concentrated under reduced pressure to a oil. The oil was combined with fumaric acid (0.77g, 6.6 mmol) and acetonitrile (40 mL) and heated to reflux to give a homogeneous solution. The solution was cooled in an ice bath and solids isolated by filtration.
The solid GS-7171 fumarate salt was dried under reduced pressure to 3.7 g. The salt (3.16 g, 5.3 mmol) was suspended in dichloromethane (30 mL) and stirred with potassium carbonate solution (5 mL, 2.5 M in water) until the solid dissolved.
The organic layer was isolated, then washed with water (5 mL), dried over anhydrous = CA 02893174 2015-05-28 sodium sulfate, and concentrated under reduced pressure to afford 2.4 g III as a tan foam.
Example 3 A. Diastereomer Separation by Batch Elution Chromatography The diastereomers of GS-7171 (III) were resolved by batch elution chromatography using a commercially available Chiralpak AS, 20 pm, 21 x 250 mm semi-preparative HPLC column with a Chiralpak AS, 20 pm, 21 x 50 mm guard column. Chiralpak AS is a proprietary packing material manufactured by Diacel and sold in North America by Chiral Technologies, Inc. (U. S. Patent Nos.
5,202,433, RE 35,919, 5,434,298, 5,434,299 and 5,498,752). Chiralpak AS is a chiral stationary phase (CSP) comprised of amylosetrisf(S)-cc-methylbenzyl carbamateJ coated onto a silica gel support.
The GS-7171 diastereomeric mixture was dissolved in mobile phase, and approximately 1 g aliquots of GS-7171 were pumped onto the chromatographic system. The undesired diastereomer, designated GS-7339, was the first major broad (approx. 15 mM, duration) peak to elute from the column. When the GS-peak had finished eluting, the mobile phase was immediately switched to 100%
methyl alcohol, which caused the desired diastereomer, designated GS-7340 (IV), to elute as a sharp peak from the column with the methyl alcohol solvent front. The methyl alcohol was used to reduce the over-all cycle time. After the first couple of injections, both diastereomers were collected as a single large fractions containing one of the purified diastereomers (>99.0% single diastereomer). The mobile phase solvents were removed in vacua to yield the purified diastereomer as a friable foam.
About 95% of the starting GS-7171 mass was recovered in the two diastereomer fractions. The GS-7340 fraction comprised about 50% of the total recovered mass.

WO 02/08241 Periuso1/23104 The chromatographic conditions were as follows:
Mobile Phase(Initial) : GS-7171 - Acetonitrile : Isopropyl Alcohol (90:10) (Final) : 100% Methyl Alcohol Flow : 10 mL/minute Run Time : About 45 minute Detection : UV at 275 nm Temperature : Ambient Elution Profile : GS-7339 (diastereomer B) : GS-7340 (diastereomer A; (IV)) B. Diastereomer Separation of GS-7171 by SMB Chromatography For a general description of simulated moving bed (SMB) chromatography, see Strube et al., "Organic Process Research and Development" 2:305-319 (1998).
GS-7340 (IV). GS-7171 (III), 2.8 kg, was purified by simulated moving bed chromatography over 10 cm by 5 cm beds of packing (Chiral Technologies Inc., micron Chiralpak AS coated on silica gel) (1.2 kg). The columns were eluted with 30% methanol in acetonitrile. Product bearing fractions were concentrated to a solution of IV in acetonitrile (2.48 kg). The solution solidified to a crystalline mass wet with acetonitrile on standing. The crystalline mass was dried under reduced pressure to a tan crystalline powder, 1.301 kg IV, 98.7% diastereomeric purity: mp 117 - 120 C; 'I-1 NMR (CDC13) 8 1.15 (m 12H), 3.7 (t, 1H), 4.0 (m, 5H), 4.2 (dd, 1H), 5.0 (m, 1H), 6.05 (s, 2H), 7.1 (m, 5H), 8.0 (s, 1H), 8.2 (s, 1H); 31P NMR
(CDC1) 8 21.0 (decoupled).
C. Diastereomer Separation by C18 RP-HPLC
GS-7171 (III) was chromatographed by reverse phase HPLC to separate the diastereomers using the following summary protocol.

Chromatographic column: Phenomenex LunaTM C18(2), 5 pm, 100 A pore size, (Phenomenex, Torrance, CA), or equivalent Guard column: Pellicular C18 (Alltech, Deerfield, IL), or equivalent Mobile Phase: A ¨ 0.02% (85%) H3PO4 in water : acetonitrile (95:5) B ¨ 0.02% (85%) H3PO4 in water: acetonitrile (50:50) Mobile Phase Gradient:
Time % Mobile Phase % Mobile Phase A

Run Time: 50 minutes Equilibration Delay: 10 min at 100% mobile phase A
Flow Rate: 1.2 mL/min Temperature: Ambient Detection: UV at 260 run Sample Solution: 20 mM sodium phosphate buffer, pH 6 Retention Times: GS-7339, about 25 minutes GS-7340, about 27 minutes D. Diastereomer Separation by Crystalli7ation GS-7340 (IV). A solution of GS-7171 (III) in acetonitrile was concentrated to an amber foam (14.9g) under reduced pressure. The foam was dissolved in acetonitrile (20 mL) and seeded with a crystal of IV. The mixture was stirred overnight, cooled to 5 C, and solids isolated by filtration. The solids were dried to 2.3 g IV as white crystals, 98% diastereomeric purity ('P NMR): 1H NMR (CDC1) 1.15 (m 12H), 3.7 (t, 1H), 3.95 (m, 2H), 4.05 (m, 2H), 4.2 (m, 2H), 5.0 (m, 1H), 6.4 (s, 2H), 7.1 (m, 5H), 8.0 (s, 1H), 8.2 (s, 1H); 31P NMR (CDC1) 8 19.5 (decoupled).
X-ray crystal analysis of a single crystal selected from this product yielded the following data:

WO 02/08241 periusol/23104 Crystal Color, Habit colorless, column Crystal Diminsions 0.25 X 0.12 XØ08 mm Crystal System orthorhombic Lattice Type Primitive Lattice Parameters a = 8.352(1) A
b = 15.574(2) A
c = 18.253(2) A
V = 2374.2(5) A3 Space Group P212121 (#19) Z value 4 Dcalc 1.333 g/cm3 F000 1008.00 -,u(MoKa) 1.60 cm Example 4 Preparation of Fumarate Salt of GS-7340 GS-7340-02 (V). (Scheme 1) A glass-lined reactor was charged with GS-7340 (IV), (1.294 kg, 2.71 mol), furnaric acid (284 g, 2.44 mol), and acetonitrile (24.6 kg). The mixture was heated to reflux to dissolve the solids, filtered while hot and cooled to 5 C for 16 hours. The product was isolated by filtration, rinsed with acetonitrile (9.2 kg), and dried to 1329 g (V) as a white powder: mp 119.7- 121.1 C; [4,2 ¨41.7 (c 1.0, acetic acid).

Example 5 Preparation of GS-7120 (VI) Scheme 3 N.!
N N\\. 0 0 õ Et L
.,N õ N
0'L0 N N
= Li OH CH3CN CH2C12 vir 0 A 5 L round bottom flask was charged with monophenyl PMPA, (II), (200 g, 0.55 mol) and acetonitrile (0.629 kg). Thionyl chloride (0.144 kg, 1.21 mol) was added below 27 C. The mixture was heated at 70 C until solids dissolved. Volatiles (0.45 L) were removed by atmospheric distillation under nitrogen. The pot residue was cooled to 25 C, dichlorornethane (1.6 kg) was added and the mixture was cooled to -20 C. A solution of (L)-a aminobutyric acid ethyl ester (0.144 kg, 1.1 mol) in dichloromethane (1.33 kg) was added over 18 minutes at -20 to -10 C followed by triethylamine (0.17 kg, 1.65 mol) over 15 minutes at -8 to -15 C. The reaction mixture was warmed to room temperature and washed four times with sodium dihydrogenphosphate solution (10% aq., 0.3 L each wash). The organic solution was dried with anhydrous sodium sulfate (0.5 kg) and filtered. The solids were rinsed with dichloromethane (0.6 kg) and the combined filtrate and rinse was concentrated to an oil under reduced pressure. The oil was purified by chromatography over a 15 x 13 cm bed of 1.2 kg silica gel 60,230 to 400 mesh.
The column was eluted with a gradient of dichloromethane and methanol. Product bearing fractions were concentrated under reduced pressure to afford 211 g VI
(Scheme 3) as a tan foam.

Example 5a Diastereomer Separation of GS-7120 by Batch Elution Chromatography The diastereorneric mixture was purified using the conditions described for GS-7171 in Example 3A except for the following:
Mobile Phase (Initial) : GS-7120 - Acetonitrile : Isopropyl Alcohol (98:2) (Final) : 100% Methyl Alcohol Elution Profile : GS-7341 (diastereomer B) : GS-7342 (diastereomer A) Example 6 Diastereomer Separation of GS-7120 by Crystalli7ation A 1 L round bottom flask was charged with monophenyl PMPA, (II), (50 g, 0.137 mol) and acetonitrile (0.2 L). Thionyl chloride (0.036 kg, 0.303 mol) was added with a 10 C exotherm. The mixture was heated to reflux until solids dissolved.
Volatiles (0.1 L) were removed by atmospheric distillation under nitrogen. The pot residue was cooled to 25 C, dichloromethane (0.2 kg) was added, and the mixture was cooled to -20 C. A solution of (L)-a aminobutyric acid ethyl ester (0.036 kg, 0.275 mol) in dichloromethane (0.67 kg) was added over 30 minutes at -20 to -8 C
followed by triethylamine (0.042 kg, 0.41 mol) over 10 minutes at up to -6 C.
The reaction mixture was warmed to room temperature and washed four times with sodium dihydrogenphosphate solution (10% aq., 0.075 L each wash). The organic solution was dried with anhydrous sodium sulfate (0.1 kg) and filtered. The solids were rinsed with ethyl acetate (0.25 L, and the combined filtrate and rinse was concentrated to an oil under reduced pressure. The oil was diluted with ethyl acetate (0.25 L), seeded, stirred overnight, and chilled to - 15 C. The solids were isolated by filtration and dried under reduced pressure to afford 17.7 g of GS-(Table 5) as a tan powder: '1-1 NMR (CDC13) 5 0.95 (t, 3H), 1.3 (in, 6H), 1.7, (in, 2H), = CA 02893174 2015-05-28 3.7 (m, 2H), 4.1(m, 6H), 4.4 (dd, 1H), 5.8 (s, 2H), 7.1 (m, 5H), 8.0 (s, 1H), 8.4 (s, IFI);
'P NMR (CDC]) 821 (decoupled).
Example 7 Diastereorner Separation of GS-7097 The diastereomeric mixture was purified using the conditions described for GS-7171 (Example 3A) except for the following:
Mobile Phase (Initial) : GS-7120 Acetonitrile : Isopropyl Alcohol (95:5) (Final) : 100% Methyl Alcohol Elution Profile : GS-7115 (diastereomer B) : GS-7114 (diastereomer A) Example 8 Alternative Procedure for Preparation of GS-7097 GS-7097: Phenyl PMPA, Ethyl L-Alanyl Amidate. Phenyl PMPA (15.0 g, 41.3 mmol), L-alanine ethyl ester hydrochloride (12.6 g, 83 mmol) and triethylamine (11.5 mL, 83 mmol) were slurried together in 500 mL pyridine under dry N2.
This suspension was combined with a solution of triphenylphosphine (37.9 g, 145 mmol), Aldrithiol 2 (2,21-dipyridyl disulfide) (31.8 g, 145 mmol), and 120 mL
pyridine. The mixture was heated at an internal temperature of 57 C for 15 hours.
The complete reaction was concentrated under vacuum to a yellow paste, 100 g.
The paste was purified by column chromatography over a 25 x 11 cm bed of 1.1 kg silica gel 60, 230 to 400 mesh. The column was eluted with 8 liters of 2%
methanol in dichloromethane followed by a linear gradient over a course of 26 liters eluent up to a final composition of 13% methanol. Clean product bearing fractions were concentrated to yield 12.4 g crude (5), 65% theory. This material was contaminated with about 15% (weight) triethylamine hydrochloride by 1H NMR. The contamination was removed by dissolving the product in 350 mL ethyl acetate, extracting with 20 mL water, drying the organic solution over anhydrous sodium sulfate, and concentrating to yield 11.1 g pure GS-7097 as a white solid, 58%
yield.
The process also is employed to synthesize the diastereomeric mixture of GS-7003a and GS-7003b (the phenylalanyl amidate) and the mixture GS-7119 and GS-7335 (the glycyl amidate). These diastereomers are separated using a batch elution procedure such as shown in Example 3A, 6 and 7.
Example 9 In Vitro Studies of Prodrug Diastereomers The in vitro anti-HIV-1 activity and cytotoxicity in MT-2 cells and stability in human plasma and MT-2 cell extracts of GS-7340 (freebase) and tenofovir disoproxil fumarate (TDF), are shown in Table 1. GS-7340 shows a 10-fold increase in antiviral activity relative to TDF and a 200-fold increase in plasma stability. This greater plasma stability is expected to result in higher circulating levels of than TDF after oral administration.
Table 1. In Vitro Activity and Stability HIV-1 Activity Cytotoxicity_ ________________ Stability T 1/2 (min) Human MT-2 IC5ogm CCsoum Plasma Cell Extract (P/MT-2) GS 7340 0.005 >40 90.0 28.3 3.2 TDF 0.05 70 0.41 70.7 0.006 Tenofovir 5 6000 In order to estimate the relative intracellular PMPA resulting from the intracellular metabolism of I'DF as compared to that from GS-7340, both prodrugs and PMPA were radiolabeled and spiked into intact human whole blood at equirnolar concentrations. After 1 hour, plasma, red blood cells (RBCs) and peripheral blood mononuclear cells (PBMCs) were isolated and analyzed by HPLC
with radiometric detection. The results are shown in Table 2.

After 1 hour, GS-7340 results in 10x and 30x the total intracellular concentration of PMPA species in PBMCs as compared to TDF and PMPA, respectively. In plasma after 1 hour, 84% of the radioactivity is due to intact GS-7340, whereas no TDF is detected at 1 hour. Since no intact TDF is detected in plasma, the 10x difference at 1 hour between TDF and GS-7340 is the minimum difference expected in vivo. The HPLC chromatogram for all three compounds in PBMCs is shown in Figure 1.
Table 2. PMPA Metabolites in Plasma, PBMCs and RBCs After 1 h Incubation of PMPA Prodrugs or PMPA in Human Blood.
Total C-14 Metabolites (%
of Total Peak Area) Compound Matrix Recovered, PMPA PMPAp, PMPApp, Met. X, Met. Y, GS
7340, pig-eq % % % %
GS-7340 Plasma/FP 43.0 1 - 2 13 84 (60 g-eq) PBMC 1.25 45 16 21 18 -RBC/FP 12.6 8 - 24 11 57 PMPA PMPAp PMPApp Mono-POC GS-4331 GS-4331 Plasma/FP 48.1 11 - 89 (TDF) (60 ag-eq) PBMC 0.133 50 25 18 7 -RBC/FP 10.5 93 7.0 . - -, PMPA PMPAp PMPApp PMPA Plasma/FP 55.7 100 -(60 ag-eq) PBMC 0.033 86 14 -RBC/FP 3.72 74 10 16 Figure 1. HPLC/C-14 Traces of PBMC Extracts from Human Blood Incubated for 1 h at 37 C with TDF, GS-7340 or PMPA.
TDF

vir400-PMPA PMPAp PMPAPp lb 15 20 SOO

= PMPA

200: Met. X pmpAp PMPApp 0 ___ o 5 10 -200.
PMPA
o 10 5 1b 15 -- 20 Met. X and Met Y (metabolites X and Y) are shown in Table 5. Lower case designates phosphorylation. These results were obtained after 1 hour in human blood. With increasing time, the in vitro differences are expected to increase, since 84% of GS-7340 is still intact in plasma after one hour. Because intact GS-7340 is present in plasma after oral administration, the relative clinical efficacy should be related to the IC,,, values seen in vitro.
In Table 3 below, IC values of tenofovir, TDF, GS-7340, several nucleosides and the protease inhibitor nelfinivir are listed. As shown, nelfinavir and GS-are 2-3 orders of magnitude more potent than all other nucleotides or nucleosides.

WO 02/08241 PCl/US01/23104 Table 3. In Vitro Anti-HIV-1 Activities of Antiretroviral Compounds Compound ICso (11M) Adefovir (PMEA) 13.4 4.21 Tenofovir (PMPA) 6.3 3.31 AZT 0.17 0.081 3TC 1.8 0.251 d4T 3 2.5' Nelfinavir 0.006 0.0021 TDF 0.05 GS 7340 0.005 1. A. S. Mulato and J. M. Cherrington, Antiviral Research 36, 91(1997) Additional studies of the in vitro cell culture anti-HIV-4 activity and CC50 of separated diastereomers of this invention were conducted and the results tabulated below.

Table 4. Effect of Diastereomer _ Compound Diastereomer ICõ (pM) Fold change A /B activity CCõ (pM) PMPA - 5 lx - 6000 ¨
Ala-methylester Mixture 1:1 0.025 200x 20x 80 GS-6957a A 0.0075 670x GS-6957b 0.15 33x Phe-methylester Mixture 1:1 0.03 170x 10x 60 GS-7003a A 0.01 500x GS-7003b B 0.1 50x Gly-ethylester Mixture 1:1 0.5 10x 20x GS-7119 A 0.05 100x >100 GS-7335 B 1.0 5x Ala-isopropyl Mixture 1:1 0.01 500x 12x GS-7340 A 0.005 1,000x 40 GS-7339 B 0.06 83x >100 ABA-ethyl Mixture 1:1 0.008 625x 7.5x >100 GS-7342 A 0.004 1,250x GS-7341 B 0.03 170x Ala-ethyl Mixture 1:1 0.02 250x 10x 60 GS- 7114 A 0.005 1,000x GS- 7115 B 0.05 100x Assay reference: Arimilli, MN, et al., (1997) Synthesis, in vitro biological evaluation and oral bioavailability of 9{2-(phosphonomethoxy)propyl]adenine io (PMPA) proctrugs. Antiviral Chemistry and Chemotherapy 8(6):557-564.
"Phe-rnethylester" is the methylphenylalaninyl monoamidate, phenyl monoester of tenofovir; "gly-methylester" is the methylglycyl monoamidate, phenyl monoester of tenofovir.
In each instance above, isomer A is believed to have the same absolute stereochemistry as GS-7340 (S), and isomer B is believed to have the same absolute stereochernistry that of GS-7339.

The in vitro metabolism and stability of separated diastereorners were determined in PLCE, MT-2 extract and human plasma. A biological sample listed below, 80 4, was transferred into a screw-capped centrifuge tube and incubated at 37 C for 5 min. A solution containing 0.2 mg/mL of the test compound in a suitable buffer, 20 L, was added to the biological sample and mixed. The reaction mixture, 20 L, was immediately sampled and mixed with 604 of methanol containing 0.015 mg/mL of 2-hydroxymethylnaphthalene as an internal standard for HPLC analysis. The sample was taken as the time-zero sample. Then, at specific time points, the reaction mixture, 20 4, was sampled and mixed with of methanol containing the internal standard. The mixture thus obtained was centrifuged at 15,000 G for 5 min and the supernatant was analyzed with HPLC
under the conditions described below.
The biological samples evaluated are as follows.
(1) PLCE (porcine liver carboxyesterase from Sigma, 160 u/mg protein, 21 mg protein/mL) diluted 20 fold with PBS (phosphated-buffered saline).
(2) MT-2 cell extract was prepared from MT-2 cells according to the published procedure [A. Pompon, I. Lefebvre, J.-L. Imbach, S. Kahn, and D. Farquhar, "Antiviral Chemistry & Chemotherapy", 5:91-98 (1994)] except for using HEPES
buffer described below as the medium.
(3) Human plasma (pooled normal human plasma from George King Biomedical Systems, Inc.) The buffer systems used in the studies are as follows.
In the study for PLCE, the test compound was dissolved in PBS. PBS (phosphate-buffered saline, Sigma) contains 0.01 M phosphate, 0.0027 M potassium chloride, and 0.137 M sodium chloride. pH 7.4 at 37 C.
In the study for MT-2 cell extracts, the test compound was dissolved in HEPES
buffer. HEPES buffer contains 0.010 M HEPES, 0.05 M potassium chloride, 0.005 M
magnesium chloride, and 0.005 M di-clithiothreitol. pH 7.4 at 37 C.

WO 02/082-11 Pcrius01/23104 In the study for human plasma, the test compound was dissolved in TBS. TBS
(tris-buffered saline, Sigma) contains 0,05 M Tris, 0.0027 M potassium chloride, and 0.138 M sodium chloride. pH 7.5 at 37 C.
The HPLC analysis was carried out under the following conditions.
Column: Zorbax Rx-C8, 4.6 x 250 mm, 5 (MAC-MOD Analytical, Inc. Chadds Ford, PA) Detection: UV at 260 nin Flow Rate: 1.0 mL/min Run Time: 30 min Injection Volume: 20 iL
Column Temperature: Ambient temperature Mobile Phase A: 50 mM potassium phosphate (pH 6.0)/CH3CN = 95/5 (v/v) Mobile Phase B: 50 mM Potassium phosphate (pH 6.0)/CH3CN . 50/50 (v/v) Gradient Run: 0 min 100% Mobile Phase A
min 100% Mobile Phase B
25 30 min 100% Mobile Phase B
The results are shown below in Table 5 (also including selected IC50 data from Table 4).

, Table 5. In Vitro Metabolism of Isomers A and B of PMPA monoamidate at 37 C
No PMPA monoamidate HIV ICõ PLCE hydrolysis MT-2 extract Human . structure (AM) rate and hydrolysis rate Plasma product and product Stabty (HP) _ # 9 9H3 tõ., = 2.9 min tõ, = 2.9 min t112 = 148 min 1 0\,PcNH-CHOOOEt 0.005 :. Met. X & PMPA Met, X & PMPA
Met. Y
5: OPh Isomer A GS7114 2 C/ "
0 P-NH-CHCOOEt t,12 = 8.0 min t12 = 150.6 min t =
495 min : =.. \ 0.05 OPh Met, X 8( PMPA Met. X 8( PMPA
Met. Y
fi Isomer B GS7115 A, 9 9-13 t = 3.3 min t,,, = 28.3 min tõ2 = 90.0 min 3 IN(ON,PcNH-CHCO0iPr 0.005 11- OPh Met. X & PMPA Met. X & PMPA
Met. Y
Isomer A GS7340 A, 9 cH3 tõ, = 10.1 min 1'1,2> 1000 min t,õ = 231 min 4 I0\,P-NH-CHCO0iPr r. \ 0.06 Met, X & PMPA
Met. Y
5-. OPh Isomer B GS7339 1/-NH-CHCOOEt fõ2= 3,9 min t = 49.2 min tõ, = 103 mm n = \ 0.004 Met. X Met. X & PMPA
Met. Y
...I-1- OPh Isomer A GS7342 A 0 9H2CH3 t,õ = 11,3 min t,õ > 1000 min t,õ = 257 min 6 C, "
0 P-NH-CHCOOEt 0.03 Met. X Met. Y
OPh Isomer B GS7341 _ A, 9 0 II t,õ <0.14 min t,õ = 70,7 min tõ, = 0.41 min 7 LOP-OCH2OCOIPr 0,05 - \
MonoPOC PMPA monoPOC PMPA monoPOC
Gs4331 OCH20u0iPr PMPA
Met. X: Met. Y:
A 0 P-NH-CHCOOH \ ,,. N-OH- HCOOR11 0 R13 A o R13 A NH
E._ "OH a ir) NH2 A =1)7;1 N N
i =

Example 10 Plasma and PBMC Exposures Following Oral Administration Of Prodrug Diastereomers to Beagle Dogs The pharmacokinetics of GS 7340 were studied in dogs after oral administration of a 10 mg-eq/kg dose.
Formulations. The prodrugs were formulated as solutions in 50 mM citric acid within 0.5 hour prior to dose. All compounds used in the studies were synthesized by Gilead Sciences. The following lots were used:
__________________________________________________________ GSI Amidate Amino acid AA Ester Diastereoisomer Lot Number GS-7340-2 Alanine i-Propyl Isomer A 1504-187-19 GS-7339 Alanine i-Propyl Isomer B 1509-185-31 GS7114 Alanine Ethyl Isomer A 1509-181-26 GS7115 Alanine Ethyl Isomer B 1509-181-22 GS7119 Glycine Ethyl Isomer A 1428-163-28 GS7342 et-Aminobutyric Acid Ethyl Isomer A 1509-191-12 GS7341 ec-Aminobutyric Acid Ethyl Isomer B 1509-191-7 Dose Administration and Sample Collection. The in7life phase of this study was conducted in accordance with the recommendations of the "Guide for the Care and Use of Laboratory Animals" (National Institutes of Healthpublication 86-23) and was approved by art Institutional Animal Care and Use Committee. Fasted male beagle dogs (10 2 kg) were used for the studies. Each drug was administered as a single dose by oral gavage (1.5-2 ml/kg). The dose was 10 mg-equivalent of PMPA/kg. For PBMCs, blood samples were collected at 0 (pre-dose), 2,8, and 24 h post-dose. For plasma, blood samples were collected at 0 (pre-dose), 5, 15, and 30 min, and 1, 2, 3,4, 6, 8, 12 and 24h post-dose. Blood (1.0 ml) was processed immediatelyfor plasma by centrifugation at 2,000 rpm for 10 min. Plasma samples were frozen and maintained at 70 C until analyzed.
Peripheral Blood Mononuclear Cell (PBMC) preparation. Whole blood (8 ml) drawn at specified time points was mixed in equal proportion with phosphate buffered saline (PBS), layered onto 15 ml of Ficoll-Paque solution (Pharmacia Biotech,) and centrifuged at 400 x g for 40 min. PBMC layer was removed and washed once with PBS. Formed PMBC pellet was reconstituted in 0.5 ml of PBS, cells were resuspended, counted using hemocytometer and maintained at 70 C
until analyzed. The number of cells multiplied by the mean single-cell volume was used in calculation of intracellular concentrations. A reported value of 200 femtoliters/cell was used as the resting PBMC volume (B. L. Robins, R.V.
Srinivas, C. Kim, N. Bischofberger, and A. Fridland, Antixnicrob. Agents Chemother.
42,612 (1998).
Determination of PMPA and Prodrugs in plasma and PBMCs. The concentration of PMPA in dog plasma samples was determined by derivatizing PMPA with chloroacetaldehyde to yield a highly fluorescent N, N6-ethenoadenine derivative (L.
Naesens, J. Balzarini, and E. De Clercq, din. Chem. 38,480 (1992). Briefly, plasma (100 pl) was mixed with200 ul acetonitrile to precipitateprotein. Samples were then evaporated to dryness under reduced pressure at room temperature. Dried samples were reconstituted in 200 ul derivatization cocktail (0.34% chloroacetaldehyde in 100 mM sodium acetate, pH 4.5), vortexed, and centrifuged. Supernatant was then transferred to a clean screw-cap tube and incubated at 95 C for 40 min.
Derivatized samples were then evaporated to dryness and reconstituted in 100 ul of water for HPLC analysis.
Before intracellular PMPA could be determined by HPLC, the large amounts of adenine related ribonucleotides present in the PBMC extracts had to be removed by selective oxidation. We used a modified procedure of Tanaka et al (K. Tanaka, A.
Yoshioka, S. Tanaka, and Y. Wataya, Anal. Biochem., 139,35 (1984). Briefly, PBMC
samples were mixed 1:2 with methanol and evaporated to dryness under reduced pressure. The dried samples were derivatized as described in the plasma assay.
The derivatized samples were mixed with 20 - 1, of 1M rhamnose and 30 pL of 0.1M
sodium periodate and incubated at 37 C for 5 min. Following incubation, 40 pL
of 4M methylamine and 20 uL of 0.5M inosine were added. After incubation at 37 C
for 30 min, samples were evaporated to dryness under reduced pressure and reconstituted in water for HPLC analysis.

No intact prodrug was detected in any PBMC samples. For plasma samples potentially containing intact prodrugs, experiments were performed to verify that no further conversion to PMPA occurred during derivatization. Prodrug standards were added to drug-freeplasma and derivatized as described. There were no detectable levels of PMPA present in any of the plasma samples, and the projected % of conversion was less than 1%.
The HPLC system was comprised of a P4000 solvent delivery system with AS3000 autoinjector and F2000 fluorescence detector (Thermo Separation, San Jose, CA).
The columnwas an Inertsil ODS-2 column (4.6 x 150 mm). The mobile phases used were: A, 5% acetonitrile in 25 mM potassium phosphate buffer with 5mM
tetrabutyl ammonium bromide (TBABr), pH 6.0; B, 60% acetonitrile in 25 mM potassium phosphatebuffer with 5 mM TBABr, pH 6Ø The flow rate was 2 ml/min and the column temperature was maintained at 35 C by a column oven. The gradient profile was 90% A/10% B for 10 min for PMPA and 65%A/35%B for 10 min for the prodrug. Detection was by fluorescence with excitation at 236 nrn and emission at 420 nm, and the injection volume was 10 p1. Data was acquired and stored by a laboratory data acquisition system (PeakPro, Beckman, Allendale, NJ).
Pharmacokinetic Calculations. PMPA and prodrug exposures were expressed as areas under concentration curves in plasma or PBMC from zero to 24 hours (AUC).
The AUC values were calculated using the trapezoidal rule.
Plasma and PBMC Concentrations. The results of this study is shown in Figures and 3. Figure 2 shows the time course of GS 7340-2 metabolism summary of plasma and PBMC exposures following oral administration of pure diastereoisomers of the PMPA prodrugs.

WO 02/08241 PC:TRISH/23104 Figure 2. PMPA and Prodrug Concentration in Plasma and PBMCs Following Oral Administration of GS 7340-2 to Dogs at 10 mg-eq/kg.

=
to --=¨ PMPA in Plasma 7340-2 In Plasma 1 lib PMPA in PBMC

0.1 = _______________ =
0.01 Time Postdose (h) The bar graph in Figure 2 shows the AUC (0-24h) for tenofovir in dog PBMCs and plasma after administration of PMPA s.c., TDF and amidate ester prodrugs. All of the amidate prodrugs exhibited increases in PBMC exposure.
For example, GS 7340 results in a -21-fold increase in PBMC exposure as compared to PMPA s.c. and TDF; and a 6.25-fold and 1.29-fold decrease in plasma exposure, respectively.

= CA 02 8 93174 2 015-05-2 8 Figure 3. Depicts Tenofovir Exposure in PBMCs and Plasma Upon Administration of 10 mg-eq/kg in dogs AUC(0-24h) for PMPA in PBMC and Plasma Following an Oral Dose of 10 mg-eq/kg PMPA Prodrugs to Dogs.
1600 _____________________________ _ MIN Plasma 1400 - ..:\I Pgmc a D
< 1200 -E.) = _ --1 j v) woo -¨

Q-w <
a.
m i a. 600 -I

T I

'[ 114 i __ 0 ___ r ' ¨ , i ¨ r' cn - cll tl- ;
17 R. 07 r=-= E C,3 Ci) cn cn cn V) to9 0 0 Cf1 0 0 0 0 c.9 These data establish in vivo that GS 7340 can be delivered orally, minimizes systemic exposure to PMPA and greatly enhances the intracellular concentration of PMPA in the cells primarily responsible for HIV replication.

Table 6 PMPA Exposure in KWIC and Plasma from Oral Prodruqs of PMPA in Dogs PMPA AUC In Plasma PMPA AUC in PBMC Prodrug PBMC/Plasma GS# Moiety Exposure Mean StDev N Mean StDev N in Plasma Ratio GS-7114 Mono-Ala-Et-A 5.8 0.9 2 706 331 5 YES 122 GS-7115 Mono-Ala-Et-B 6.6 1.5 2 284 94 5 YES 43 GS-7340-2 Mono-Ala-iPr-A 5.0 1.1 5 805 222 5 YES 161 GS-7339 Mono-Ala-iPr-A 6.4 1.3 2 200 57 5 YES 31 GS-7119 Mono-Gly-Et-A 6,11 1.86 2 530 304 5 YES 87 GS-7342 Mono-ABA-Et-A 4.6 1.2 2 1060 511 5 YES 230 GS7341 Mono-ABA-Et-B 5.8 1.4 2 199 86 5 YES 34 Example 11 20 Biodistribution of GS-7340 As part of the preclinical characterization of GS-7340, its biodistribution in dogs was determined. The tissue distribution of GS-7340 (isopropyl alaninyl monoamidate, phenyl monoester of tenofovir) was examined following oral 25 administration to beagle dogs. Two male animals were dosed orally with 14C=GS-7340 (8.85 mg-equiv. of PMPA/kg, 33.2 I.LCi/kg; the 8-carbon of adenine is labeled) in an aqueous solution (50 mM citric acid, pH 2.2). Plasma and peripheral blood mononuclear cells (PBMCs) were obtained over the 24-hr period. Urine and feces were cage collected over 24 hr. At 24 h after the dose, the animals were sacrificed 30 and tissues removed for analysis. Total radioactivity in tissues was determined by oxidation and liquid scintillation counting.
The biodistribution of PMPA after 24 hours after a single oral dose of radiolabelled GS 7340 is shown in Table 4 along with the data from a previous study with TDF (GS-4331). In the case of TDF, the prodrug concentration in the 35 plasma is below the level of assay detection, and the main species observed in plasma is the parent drug. Levels of PMPA in the lymphatic tissues, bone marrow, and skeletal muscle are increased 10-fold after administration of GS-7340.

. CA 02893174 2015-05-28 Accumulation in lymphatic tissues is consistent with the data observed from the PBMC analyses, since these tissues are composed primarily of lymphocytes.
Likewise, accumulation in bone marrow is probably due to the high percentage of lymphocytes (70%) in this tissue.
Table 7. Excretion and Tissue Distribution of Radiolabelled GS-7340 in Dogs (Mean, N=2) Following an Oral Dose at 10 mg-eq. PMPA/kg.
Tissue/Fluid GS-4331 GS-7340 Tissue Conc.
% Dose Conc. % Dose Conc.
Ratio of GS 7340 . (ug-eq/g) (ug-eq/g) to GS-Liver 12.40 38.30 16.45 52.94 1.4 Kidney 4.58 87.90 3.78 80.21 0.9 Lungs 0.03 0.53 0.34 4.33 8.2 Iliac Lymph Nodes 0.00 0.51 0.01 5.42 10.6 Axillary Lymph Nodes 0.00 0.37 0.01 5.54 14.8 Inguinal Lymph Nodes 0.00 0.28 0.00 4.12 15.0 Mesenteric Lymph Nodes 0.00 1.20 0.04 6.88 5.7 Thyroid Gland 0.00 0.30 0.00 4.78 15.8 Pituitary Gland 0.00 0.23 0.00 1.80 7.8 Salivary Gland (L+R) 0.00 0.45 0.03 5.54 12.3 Adrenal Gland 0.00 1.90 0.00 3.47 1.8 Spleen 0.00 0.63 0.17 8.13 12.8 Pancreas 0.00 0.57 0.01 3.51 6.2 Prostate 0.00 0.23 0.00 2.14 9.1 Testes (L+R) 0.02 1.95 0.02 2.01 1.0 Skeletal Muscle 0.00 0.11 0.01 1.12 10.1 Heart 0.03 0.46 0.15 1.97 4.3 Femoral Bone 0.00 0.08 0.00 0.28 3.5 Bone Marrow 0.00 0.20 0.00 2.05 10.2 Skin 0.00 0.13 0.00 0.95 7.2 Abdominal fat 0.00 0.16 0.00 0.90 5.8 Eye (L+R) 0.00 0.06 0.00 0.23 3.7 Brain 0.00 <LOD 0.00 <LOD n.d.
Cerebrospinal Fluid 0.00 <LOD 0.00 0.00 n.d.
Spinal Cord , 0.00 <LOD 0.00 0.04 n.d.
Stomach 0.11 1.92 0.26 2.68 1.4 Jejunum 1.34 3.01 0.79 4.16 1.4 Duodenum 0.49 4.96 0.44 8.77 1.8 Ileum 0.01 0.50 0.16 4.61 9.2 Large Intestine 1.63 5.97 2.65 47.20 7.9 Gall bladder 0.00 3.58 0.04 25.02 7.0 Bile 0.00 9.63 _ 0.22 40.48 4.2 Feces 40.96 n.d. 0.19 n.d. ma.
Total GI Tract Contents 5.61 n.d. 21.64 n.d. n.a.
Urine 23.72 n.d. 14.73 n.d. n.a.
Plasma at 24 h 0.00 0.20 0.00 0.20 1.0 Plasma at 0.25 h n.a. 3.68 ma. 3.48 0.9 PBMC* 0.00 n.d. 0.00 63.20 n.d.
Whole Blood _ 0.00 0.85 0.16 0.20 0.2 Total Recovery L 81.10 68.96 I
* Calculated using typical recovery of 15 x 106 cells total, and mean PBMC
volume of 0.2 picoliters/cell n.s. = no sample, ma. = not applicable, n.d. = not determined.

Claims (18)

CLAIMS:
1. A screening method for identifying a methoxyphosphonate nucleotide analogue prodrug conferring enhanced activity in a target tissue comprising:
(a) providing at least one of said prodrugs;
(b) selecting at least one therapeutic target tissue and at least one non-target tissue;
(c) administering the prodrug to the target tissue and to said at least one non-target tissue; and (d) determining the relative activity conferred by the prodrug in the tissues in step (c).
2. The method of claim 1 wherein the activity is antiviral activity or antitumor activity.
3. The method of claim 2 wherein the activity is antiviral activity.
4. The method of claim 3 wherein the activity is anti-HIV or anti-HBV
activity.
5. The method of claim 1 wherein the prodrug is a prodrug of PMPA or PMEA.
6. The method of claim 5 wherein the prodrug is a phosphonoamidate, phosphonoester or mixed phosphonoamidate/phosphonoester.
7. The method of claim 6 wherein the amidate is an amino acid amidate.
8. The method of claim 6 wherein the ester is an aryl ester.
9. The method of claim 1 further comprising selecting a prodrug having a relative activity in the target tissue that is greater than 10 times that of the non-target tissue.
10. The method of claim 1 wherein the target and non-target tissue are in an animal, the prodrug is administered to the animal and the relative activity is determined by analysis of the animal tissues after administration of the prodrug.
11. The method of claim 1 wherein activity in the target and non-target tissues is determined by assaying the amount of at least one metabolite of the prodrug in the tissues.
12. The method of claim 12 wherein the metabolite is the parental drug.
13. The method of claim 12 wherein the metabolite is the diphosphate of the parental drug.
14. The method of claim 1 wherein the target tissue is virally infected tissue and the non-target tissue is the same tissue which is not virally infected.
15. The method of claim 1 wherein the target tissue is lymphoid tissue and the activity is anti-HIV activity.
16. The method of claim 1 wherein the target tissue is liver and the activity is anti-HBV activity.
17. The method of claim 1 wherein the target tissue is hematological and the activity is antitumor activity.
18. The method of claim 1 wherein the target tissue is malignant and the non-target tissue is the same tissue but non-malignant.
CA2893174A 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same Abandoned CA2893174A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US22002100P 2000-07-21 2000-07-21
US60/220,021 2000-07-21
US60/220,021(CIP) 2000-07-21
CA2725819A CA2725819C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA2725819A Division CA2725819C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Publications (1)

Publication Number Publication Date
CA2893174A1 true CA2893174A1 (en) 2002-01-31

Family

ID=22821718

Family Applications (3)

Application Number Title Priority Date Filing Date
CA2893174A Abandoned CA2893174A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
CA2416757A Expired - Lifetime CA2416757C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
CA2725819A Expired - Lifetime CA2725819C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Family Applications After (2)

Application Number Title Priority Date Filing Date
CA2416757A Expired - Lifetime CA2416757C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
CA2725819A Expired - Lifetime CA2725819C (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Country Status (37)

Country Link
US (10) US20040018150A1 (en)
EP (3) EP3235823A1 (en)
JP (4) JP4651264B2 (en)
KR (2) KR100767432B1 (en)
CN (2) CN1291994C (en)
AP (1) AP1466A (en)
AU (3) AU8294101A (en)
BE (1) BE2016C018I2 (en)
BG (1) BG66037B1 (en)
BR (1) BRPI0112646B8 (en)
CA (3) CA2893174A1 (en)
CY (2) CY2016008I1 (en)
CZ (2) CZ304734B6 (en)
DK (2) DK1301519T4 (en)
EA (1) EA004926B1 (en)
EE (1) EE05366B1 (en)
ES (2) ES2627903T3 (en)
FR (1) FR16C0013I2 (en)
HK (2) HK1054238A1 (en)
HR (2) HRP20160074B1 (en)
HU (2) HU230960B1 (en)
IL (1) IL153658A0 (en)
IS (1) IS2985B (en)
LT (2) LT2682397T (en)
LU (1) LU93029I2 (en)
MX (1) MXPA03000587A (en)
NL (1) NL300803I2 (en)
NO (6) NO336718B1 (en)
NZ (3) NZ523438A (en)
OA (1) OA12393A (en)
PL (1) PL213214B1 (en)
PT (2) PT2682397T (en)
SI (2) SI2682397T1 (en)
TR (1) TR200300055T2 (en)
UA (1) UA75889C2 (en)
WO (1) WO2002008241A2 (en)
ZA (1) ZA200210271B (en)

Families Citing this family (217)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
IL161901A0 (en) * 2001-11-14 2005-11-20 Biocryst Pharmaceuticals Inc A Nucleosides, preparation thereof and use as inhibiDevice for the reading and the signalling of environmental parameters tors of rna viral polymerases
US7388002B2 (en) * 2001-11-14 2008-06-17 Biocryst Pharmaceuticals, Inc. Nucleosides, preparation thereof and use as inhibitors of RNA viral polymerases
US20050239054A1 (en) * 2002-04-26 2005-10-27 Arimilli Murty N Method and compositions for identifying anti-HIV therapeutic compounds
US20060128692A1 (en) * 2002-04-26 2006-06-15 Gilead Sciences, Inc Non nucleoside reverse transcriptase inhibitors
US7214668B2 (en) 2002-05-13 2007-05-08 Metabasis Therapeutics, Inc. Phosphonic acid based prodrugs of PMEA and its analogues
AU2004206821C1 (en) * 2003-01-14 2009-10-01 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
WO2004096285A2 (en) * 2003-04-25 2004-11-11 Gilead Sciences, Inc. Anti-infective phosphonate conjugates
CN101410120A (en) * 2003-04-25 2009-04-15 吉里德科学公司 Anti-inflammatory phosphonate compounds
US7470724B2 (en) * 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
US20090247488A1 (en) * 2003-04-25 2009-10-01 Carina Cannizzaro Anti-inflammatory phosphonate compounds
US7417055B2 (en) 2003-04-25 2008-08-26 Gilead Sciences, Inc. Kinase inhibitory phosphonate analogs
WO2004096287A2 (en) * 2003-04-25 2004-11-11 Gilead Sciences, Inc. Inosine monophosphate dehydrogenase inhibitory phosphonate compounds
US7407965B2 (en) * 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
US20050261237A1 (en) * 2003-04-25 2005-11-24 Boojamra Constantine G Nucleoside phosphonate analogs
WO2005002626A2 (en) * 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
US7429565B2 (en) * 2003-04-25 2008-09-30 Gilead Sciences, Inc. Antiviral phosphonate analogs
US7432261B2 (en) * 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
JP2007508843A (en) * 2003-10-24 2007-04-12 ギリアード サイエンシーズ, インコーポレイテッド Methods and compositions for the identification of therapeutic compounds
WO2005044279A1 (en) * 2003-10-24 2005-05-19 Gilead Sciences, Inc. Purine nucleoside phosphonate conjugates
US7432273B2 (en) 2003-10-24 2008-10-07 Gilead Sciences, Inc. Phosphonate analogs of antimetabolites
US20070281907A1 (en) * 2003-12-22 2007-12-06 Watkins William J Kinase Inhibitor Phosphonate Conjugates
WO2005063751A1 (en) * 2003-12-22 2005-07-14 Gilead Sciences, Inc. 4’-substituted carbovir-and abacavir-derivatives as well as related compounds with hiv and hcv antiviral activity
US20050153990A1 (en) * 2003-12-22 2005-07-14 Watkins William J. Phosphonate substituted kinase inhibitors
PL212403B1 (en) * 2003-12-30 2012-09-28 Gilead Sciences Phosphoranians, amidomonophosphoranians, amidobisphosphoranians for treating virus diseases
AU2005209256B2 (en) * 2004-01-21 2010-11-18 Gilead Sciences, Inc. Use of adefovir or tenofovir for inhibiting MMTV-like viruses involved in breast cancer and primary biliary cirrhosis
US8416242B1 (en) 2004-05-14 2013-04-09 Nvidia Corporation Method and system for interpolating level-of-detail in graphics processors
US8432394B1 (en) 2004-05-14 2013-04-30 Nvidia Corporation Method and system for implementing clamped z value interpolation in a raster stage of a graphics pipeline
US8411105B1 (en) 2004-05-14 2013-04-02 Nvidia Corporation Method and system for computing pixel parameters
US7079156B1 (en) 2004-05-14 2006-07-18 Nvidia Corporation Method and system for implementing multiple high precision and low precision interpolators for a graphics pipeline
BRPI0511948A (en) 2004-06-08 2008-01-29 Metabasis Therapeutics Inc lewis acid mediated cyclic ester synthesis
JP2008508291A (en) * 2004-07-27 2008-03-21 ギリアード サイエンシーズ, インコーポレイテッド Nucleoside phosphonate conjugates as anti-HIV agents
JP2008538354A (en) * 2005-04-08 2008-10-23 キメリクス,インコーポレイテッド Compounds, compositions and methods for treating viral infections and other medical diseases
WO2006110655A2 (en) 2005-04-08 2006-10-19 Chimerix, Inc. Compounds, compositions and methods for the treatment of poxvirus infections
CN100359315C (en) * 2005-05-26 2008-01-02 林维宣 Animal remedy residual ability verification sample and method for preparing same
TWI375560B (en) 2005-06-13 2012-11-01 Gilead Sciences Inc Composition comprising dry granulated emtricitabine and tenofovir df and method for making the same
TWI471145B (en) 2005-06-13 2015-02-01 Bristol Myers Squibb & Gilead Sciences Llc Unitary pharmaceutical dosage form
US8076303B2 (en) 2005-12-13 2011-12-13 Spring Bank Pharmaceuticals, Inc. Nucleotide and oligonucleotide prodrugs
CN100396689C (en) * 2006-03-07 2008-06-25 中国医学科学院医药生物技术研究所 Tenoforv monoester compounds with HIV-1/HBV virus copying inhibiting activity
PL2020996T3 (en) 2006-05-16 2012-04-30 Gilead Sciences Inc Method and compositions for treating hematological malignancies
WO2008007392A2 (en) 2006-07-12 2008-01-17 Matrix Laboratories Limited Process for the preparation of tenofovir
US7951789B2 (en) * 2006-12-28 2011-05-31 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US7964580B2 (en) 2007-03-30 2011-06-21 Pharmasset, Inc. Nucleoside phosphoramidate prodrugs
WO2009038851A2 (en) * 2007-06-26 2009-03-26 University Of Wyoming Research Corporation D/B/A Western Research Institute Treatment and prevention systems for acid mine drainage and halogenated contaminants
US8441497B1 (en) * 2007-08-07 2013-05-14 Nvidia Corporation Interpolation of vertex attributes in a graphics processor
EP3085377A1 (en) * 2008-01-25 2016-10-26 Chimerix, Inc. Methods of treating viral infections
TWI444384B (en) 2008-02-20 2014-07-11 Gilead Sciences Inc Nucleotide analogues and their use in the treatment of malignancies
US8519126B2 (en) * 2008-04-25 2013-08-27 Cipla Limited Crystalline form of tenofovir disoproxil and a process for its preparation
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
ES2458358T3 (en) * 2008-07-02 2014-05-05 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2010005986A1 (en) 2008-07-08 2010-01-14 Gilead Sciences, Inc. Salts of hiv inhibitor compounds
KR20110104074A (en) * 2008-12-23 2011-09-21 파마셋 인코포레이티드 Synthesis of purine nucleosides
EP2376088B1 (en) 2008-12-23 2017-02-22 Gilead Pharmasset LLC 6-O-Substituted-2-amino-purine nucleoside phosphoramidates
AU2009329917B2 (en) 2008-12-23 2016-03-31 Gilead Pharmasset Llc Nucleoside analogs
TWI583692B (en) 2009-05-20 2017-05-21 基利法瑪席特有限責任公司 Nucleoside phosphoramidates
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
WO2011011519A1 (en) 2009-07-21 2011-01-27 Chimerix, Inc. Compounds, compositions and methods for treating ocular conditions
WO2011035250A1 (en) 2009-09-21 2011-03-24 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1'-substituted carba-nucleoside analogs
EP2534150B1 (en) 2010-02-12 2017-04-05 Chimerix, Inc. Methods of treating viral infection
HUE034239T2 (en) 2010-03-31 2018-02-28 Gilead Pharmasset Llc Process for the crystallisation of (s)-isopropyl 2-(((s)-(perfluorophenoxy)(phenoxy)phosphoryl)amino)propanoate
US8563530B2 (en) 2010-03-31 2013-10-22 Gilead Pharmassel LLC Purine nucleoside phosphoramidate
US8680071B2 (en) 2010-04-01 2014-03-25 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
AU2011248620B2 (en) 2010-04-26 2015-11-26 Chimerix, Inc. Methods of treating retroviral infections and related dosage regimes
EP2596004B1 (en) 2010-07-19 2014-09-10 Gilead Sciences, Inc. Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
MX2013000744A (en) 2010-07-22 2013-03-07 Gilead Sciences Inc Methods and compounds for treating paramyxoviridae virus infections.
TW201242974A (en) 2010-11-30 2012-11-01 Gilead Pharmasset Llc Compounds
JP5932829B2 (en) 2010-12-10 2016-06-08 シグマファーム ラボラトリーズ エルエルシー Highly stable compositions of orally active nucleotide analogs or orally active nucleotide analog prodrugs
ZA201103820B (en) 2010-12-13 2012-01-25 Laurus Labs Private Ltd Process for the preparation of tenofovir
JP2014514295A (en) 2011-03-31 2014-06-19 アイディニックス ファーマシューティカルズ インコーポレイテッド Compounds and pharmaceutical compositions for the treatment of viral infections
US9550803B2 (en) 2011-05-06 2017-01-24 University Of Southern California Method to improve antiviral activity of nucleotide analogue drugs
PT2710019T (en) 2011-05-19 2017-07-21 Gilead Sciences Inc Processes and intermediates for preparing anti-hiv agents
SG2014011548A (en) * 2011-08-16 2014-09-26 Gilead Sciences Inc Tenofovir alafenamide hemifumarate
AU2014271320B2 (en) * 2011-08-16 2017-02-23 Gilead Sciences, Inc. Tenofovir alafenamide hemifumarate
SG11201400664WA (en) 2011-09-16 2014-04-28 Gilead Pharmassett Llc Methods for treating hcv
AU2016228317B2 (en) * 2011-10-07 2018-07-19 Gilead Sciences, Inc. Methods for preparing anti-viral nucleotide analogs
AU2014215976B2 (en) * 2011-10-07 2016-06-30 Gilead Sciences, Inc. Methods for preparing anti-viral nucleotide analogs
TWI689513B (en) * 2011-10-07 2020-04-01 美商基利科學股份有限公司 Methods for preparing anti-viral nucleotide analogs
US8889159B2 (en) 2011-11-29 2014-11-18 Gilead Pharmasset Llc Compositions and methods for treating hepatitis C virus
EP2794624B1 (en) 2011-12-22 2019-05-15 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
MD20140091A2 (en) 2012-02-03 2015-01-31 Gilead Sciences, Inc. Combination therapy comprising tenofovir alafenamide hemifumarate and cobicistat for use in the treatment of viral infections
WO2013115916A1 (en) 2012-02-03 2013-08-08 Gilead Sciences, Inc. Combination therapy comprising gs-7340 and cobicistat for use in the treatment of viral infections
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
GB201215696D0 (en) * 2012-09-03 2012-10-17 Ithemba Pharmaceuticals Pty Ltd A process for the preparation of (R)-9-[2-(Phosphonometh-Oxy)propyl]adenine (PMPA)
EP2912047B1 (en) 2012-10-29 2016-08-24 Cipla Limited Antiviral phosphonate analogues and process for preparation thereof
CN102899327B (en) * 2012-11-06 2014-06-11 清华大学深圳研究生院 Antiviral small nucleic acid and temperature-sensitive type gel preparation and application thereof
BR112015011148A8 (en) * 2012-11-16 2019-10-01 Merck Sharp & Dohme compound, pharmaceutical composition, and combination
CN103848868B (en) * 2012-12-04 2017-04-12 蚌埠丰原涂山制药有限公司 method for preparing tenofovir
CN103848869B (en) * 2012-12-04 2016-12-21 上海医药工业研究院 The method preparing tenofovir
PT2950786T (en) 2013-01-31 2020-03-03 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
CN104072539B (en) * 2013-03-25 2017-03-29 安徽贝克联合制药有限公司 Double (4 acetaminophenol epoxide) esters of tenofovir and preparation method thereof and its application
WO2014187314A1 (en) * 2013-05-21 2014-11-27 成都先导药物开发有限公司 Drug target capturing method
IN2013MU01967A (en) 2013-06-07 2015-06-12 Cipla Ltd
PT3038601T (en) 2013-08-27 2020-06-30 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
WO2015040640A2 (en) * 2013-09-20 2015-03-26 Laurus Labs Private Limited An improved process for the preparation of tenofovir alafenamide or pharmaceutically acceptable salts thereof
EP2860185A1 (en) 2013-10-09 2015-04-15 Zentiva, k.s. An improved process for the preparation of Tenofovir disoproxil and pharmaceutically acceptable salts thereof
IN2013CH05455A (en) * 2013-11-27 2015-08-07 Laurus Labs Private Ltd
ES2842123T3 (en) 2014-01-14 2021-07-12 Mylan Laboratories Ltd Purification of tenofovir alafenamide and its intermediates
TWI660965B (en) 2014-01-15 2019-06-01 美商基利科學股份有限公司 Solid forms of tenofovir
CN104804042B (en) * 2014-01-24 2018-01-19 齐鲁制药有限公司 Phosphonate-nucleotide ester class compound, its pharmaceutical composition, Preparation method and use
WO2015120057A1 (en) 2014-02-05 2015-08-13 Gilead Sciences, Inc. Pharmaceutical combinations against co-infection with hiv and tuberculosis
US10449210B2 (en) 2014-02-13 2019-10-22 Ligand Pharmaceuticals Inc. Prodrug compounds and their uses
CN105814068B (en) * 2014-02-27 2017-08-04 四川海思科制药有限公司 A kind of substituted phosphoramidic acid ester derivative, its preparation method and its application
CN105001262B (en) * 2014-04-18 2017-09-01 四川海思科制药有限公司 The phosphonaminate of aryl substitution and its application medically
WO2015161785A1 (en) * 2014-04-21 2015-10-29 四川海思科制药有限公司 Method for preparing phosphoramidate derivative and intermediates thereof, and method for preparing intermediates
CN105085571A (en) * 2014-05-20 2015-11-25 四川海思科制药有限公司 Tenofovir alafenamide compound, preparation method and purpose thereof
CN105518012B (en) * 2014-06-25 2018-03-02 四川海思科制药有限公司 A kind of substituted amino acid sulfur ester, its composition and application
JP2017520545A (en) 2014-07-02 2017-07-27 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 compounds and their use
KR101703258B1 (en) 2014-12-30 2017-02-06 한미정밀화학주식회사 Preparation method for (r)-9-[2-(phosphonomethoxy)propyl]adenine with high purity
JP2017535520A (en) * 2014-09-30 2017-11-30 ハンミ・ファイン・ケミカル・カンパニー・リミテッドHanmi Fine Chemical Co., Ltd. Method for producing high purity (R) -9- [2- (phosphonomethoxy) propyl] adenine
KR101703257B1 (en) 2014-09-30 2017-02-06 한미정밀화학주식회사 Preparation method for (r)-9-[2-(phosphonomethoxy)propyl]adenine with high purity
US11311545B2 (en) 2014-10-09 2022-04-26 Board Of Regents Of The University Of Nebraska Compositions and methods for the delivery of therapeutics
TWI740546B (en) 2014-10-29 2021-09-21 美商基利科學股份有限公司 Methods for the preparation of ribosides
CN108191913A (en) * 2014-11-12 2018-06-22 四川海思科制药有限公司 A kind of tenofovir Chinese mugwort draws phenol amine crystal form a and preparation method thereof
CN104558036A (en) * 2014-12-11 2015-04-29 杭州和泽医药科技有限公司 Tenofovir alafenamide hemi-fumarate crystal form and preparation method thereof
US20170348334A1 (en) 2015-01-03 2017-12-07 Mylan Laboratories Limited Processes for the preparation of amorphous tenofovir alafenamide hemifumarate and a premix thereof
WO2016187160A1 (en) * 2015-05-16 2016-11-24 Godx, Inc. Point of need testing device and methods of use thereof
CN106188139B (en) 2015-05-29 2020-02-18 江苏天士力帝益药业有限公司 Tenofovir monobenzyl phosphoric acid amide prodrug, preparation method and application thereof
CZ2015384A3 (en) 2015-06-05 2016-12-14 Zentiva, K.S. Tenofovir alafenamide solid forms
AU2016277859B2 (en) * 2015-06-17 2019-08-01 Gilead Sciences, Inc. Co-crystals, salts and solid forms of tenofovir alafenamide
AU2016287500B2 (en) 2015-06-30 2019-05-02 Gilead Sciences, Inc. Pharmaceutical formulations comprising tenofovir and emtricitabine
AR105643A1 (en) 2015-08-10 2017-10-25 Merck Sharp & Dohme ANTI-VIRAL COMPOSITES OF B-AMINO ACID ESTER PHOSPHODIAMIDE
TWI620754B (en) * 2015-08-26 2018-04-11 Method for preparing amino phosphate derivative and preparation method thereof
TWI616452B (en) * 2015-08-26 2018-03-01 Preparation method of nucleoside analog and intermediate thereof
TWI616453B (en) * 2015-08-27 2018-03-01 Substituted amino acid thioester compounds, materials and uses thereof
WO2017037608A1 (en) * 2015-08-28 2017-03-09 Laurus Labs Private Limited Solid forms of tenofovir alafenamide and salts thereof, processes for its preparation and pharmaceutical compositions thereof
US10251904B2 (en) 2015-09-16 2019-04-09 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
NZ741957A (en) 2015-11-09 2019-04-26 Gilead Sciences Inc Therapeutic compositions for treatment of human immunodeficiency virus
CN106800573B (en) * 2015-11-25 2020-03-10 四川海思科制药有限公司 Nucleotide phosphonate monohydrate, preparation method and medical application thereof
WO2017100108A1 (en) 2015-12-10 2017-06-15 Merck Sharp & Dohme Corp. Antiviral phosphodiamide prodrugs of tenofovir
CN106866737B (en) * 2015-12-11 2020-11-20 南京圣和药物研发有限公司 Phosphonic acid derivatives and their use
WO2017106069A1 (en) 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Antiviral oxime phosphoramide compounds
WO2017118928A1 (en) 2016-01-06 2017-07-13 Lupin Limited Process for the separation of diastereomers of tenofovir alafenamide
ES2806604T3 (en) 2016-02-02 2021-02-18 Sandoz Ag Crystalline forms of tenofovir alafenamide monofumarate
CN107709288A (en) * 2016-02-03 2018-02-16 四川海思科制药有限公司 A kind of phosphinylidyne amine derivative and preparation method and purposes
CN108350007B (en) * 2016-03-01 2020-04-10 深圳市塔吉瑞生物医药有限公司 Substituted adenine compound and pharmaceutical composition thereof
CN107179355B (en) * 2016-03-11 2021-08-10 广东东阳光药业有限公司 Method for separating and detecting tenofovir alafenamide and related substances thereof
CZ2016156A3 (en) 2016-03-17 2017-09-27 Zentiva, K.S. The method of preparation of diastereomerically pure Tenofovir Alafenamide or its salts
CN107226826A (en) * 2016-03-25 2017-10-03 江苏奥赛康药业股份有限公司 Tenofovir Chinese mugwort draws phenol amine fumarate compound and its pharmaceutical composition
WO2017211325A1 (en) 2016-06-05 2017-12-14 上海诚妙医药科技有限公司 New crystal form of tenofovir alafenamide salt, preparation method and use thereof
CN106543227B (en) * 2016-06-20 2018-02-02 杭州和泽医药科技有限公司 A kind of phosphonate prodrugs of adenine derivative and its application in medicine
WO2017221189A1 (en) * 2016-06-22 2017-12-28 Laurus Labs Limited An improved process for the preparation of tenofovir alafenamide or pharmaceutically acceptable salts thereof
CN106317116A (en) * 2016-08-19 2017-01-11 张红利 Phosphamide nucleosides compound, pharmaceutically acceptable salt and application thereof, and pharmaceutical composition
EP3503895B1 (en) 2016-08-25 2021-09-15 Merck Sharp & Dohme Corp. Antiviral prodrugs of tenofovir
CN106380484A (en) * 2016-08-29 2017-02-08 杭州百诚医药科技股份有限公司 New crystal form of tenofovir alafenamide and preparation method thereof
WO2018042331A1 (en) 2016-08-31 2018-03-08 Glaxosmithkline Intellectual Property (No.2) Limited Combinations and uses and treatments thereof
WO2018051250A1 (en) 2016-09-14 2018-03-22 Viiv Healthcare Company Combination comprising tenofovir alafenamide, bictegravir and 3tc
WO2018080903A1 (en) 2016-10-26 2018-05-03 Merck Sharp & Dohme Corp. Antiviral aryl-amide phosphodiamide compounds
CN106565785B (en) * 2016-11-09 2019-11-12 周雨恬 One kind having the active nucleoside phosphoramidate class compound of Anti-HBV activity/HIV and its salt and purposes
CN108129514A (en) * 2016-12-01 2018-06-08 北京美倍他药物研究有限公司 The individual isomer and its medical usage of phosphoric acid/phosphonate derivative
EP3558322B1 (en) 2016-12-22 2021-09-29 Merck Sharp & Dohme Corp. Antiviral benzyl-amine phosphodiamide compounds
CA3047573A1 (en) 2016-12-22 2018-06-28 Merck Sharp & Dohme Corp. Antiviral aliphatic ester prodrugs of tenofovir
WO2018115046A1 (en) 2016-12-23 2018-06-28 Sandoz Ag Crystalline solid forms of tenofovir alafenamide
TWI820984B (en) 2017-01-31 2023-11-01 美商基利科學股份有限公司 Crystalline forms of tenofovir alafenamide
WO2018153977A1 (en) 2017-02-24 2018-08-30 Hexal Ag Stable composition of tenofovir alafenamide
US20190374557A1 (en) * 2017-02-28 2019-12-12 Alexandre Vasilievich Ivachtchenko Cyclobutyl (S)-2-[[[(R)-2-(6-aminopurin-9-yl)-1-methyl-ethoxy]methyl-phenoxy-phosphoryl]amino]-propanoates, and production process and application thereof
RU2659388C1 (en) 2017-02-28 2018-07-02 Васильевич Иващенко Александр Nucleotides including n-[(s)-1-cyclobutoxycarbonyl]phosphoramidate fragment, their analogs and their application
CN106866739B (en) * 2017-03-10 2018-11-02 华东师范大学 The preparation method of one kind (R) -1- (6- amino -9H- purine -9- bases) 2- phenyl esters
KR102318320B1 (en) 2017-03-14 2021-10-27 길리애드 사이언시즈, 인코포레이티드 How to treat feline coronavirus infection
CA3054822A1 (en) 2017-03-20 2018-09-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hiv post-exposure prophylaxis
CN108794530A (en) * 2017-04-26 2018-11-13 上海医药工业研究院 A kind of the third phenol of tenofovir amidic-salt crystal form and its preparation method and application
EP4219513A1 (en) 2017-05-01 2023-08-02 Gilead Sciences, Inc. Crystalline form of (s)-2-ethylbutyl 2-(((s)-(((2r,3s,4r,5r)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate
KR102379965B1 (en) * 2017-05-19 2022-03-29 주식회사 종근당 Efficient preparation method of Tenofovir
CN107266499B (en) * 2017-06-05 2019-07-02 珠海优润医药科技有限公司 A kind of antiviral compound and preparation method thereof
EP3641733A1 (en) 2017-06-30 2020-04-29 Cipla Limited Pharmaceutical compositions
CA3077489A1 (en) 2017-07-11 2019-01-17 Gilead Sciences, Inc. Compositions comprising an rna polymerase inhibitor and cyclodextrin for treating viral infections
WO2019021319A1 (en) 2017-07-27 2019-01-31 Cipla Limited Pharmaceutical compositions
WO2019027920A1 (en) 2017-08-01 2019-02-07 Gilead Sciences, Inc. Crystalline forms of ethyl ((s)-((((2r,5r)-5-(6-amino-9h-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl)-l-alaninate (gs-9131) for treating viral infections
AR112412A1 (en) 2017-08-17 2019-10-23 Gilead Sciences Inc CHOLINE SALT FORMS OF AN HIV CAPSID INHIBITOR
CN107655987B (en) * 2017-09-08 2020-11-03 厦门蔚扬药业有限公司 HPLC detection method for tenofovir alafenamide and isomer thereof
CN107522743A (en) * 2017-09-30 2017-12-29 深圳科兴生物工程有限公司 A kind of half fumaric acid tenofovir Chinese mugwort draws phenol amine industrial continuous producing method
EP3700573A1 (en) 2017-10-24 2020-09-02 Gilead Sciences, Inc. Methods of treating patients co-infected with a virus and tuberculosis
CN109942632B (en) * 2017-12-20 2021-08-31 上海博志研新药物研究有限公司 Preparation method of tenofovir alafenamide intermediate
CN109942633B (en) * 2017-12-20 2021-08-31 上海新礼泰药业有限公司 Preparation method of tenofovir alafenamide intermediate
WO2019130354A1 (en) 2017-12-30 2019-07-04 Cipla Limited Polymorphic forms of (9-[(r)-2-[[(s)-[[(s)-1- (isopropoxycarbonyl)ethyl]amino]phenoxy phosphinyl]methoxy]propyl] adenine and pharmaceutically acceptable salts thereof
JP7181938B2 (en) * 2018-01-10 2022-12-01 ヌクオリオン ファーマシューティカルズ インコーポレイテッド Phosphoro(n)amidate acetal and phospha(hona)toarcetal compounds
WO2019140365A1 (en) * 2018-01-12 2019-07-18 Board Of Regents Of The University Of Nebraska Antiviral prodrugs and formulations thereof
JP7083398B2 (en) 2018-02-15 2022-06-10 ギリアード サイエンシーズ, インコーポレイテッド Pyridine derivatives and their use for treating HIV infection
EP3752496B1 (en) 2018-02-16 2023-07-05 Gilead Sciences, Inc. Methods and intermediates for preparing a therapeutic compound useful in the treatment of retroviridae viral infection
CN108101943B (en) * 2018-02-28 2020-11-24 顾世海 Tenofovir prodrug or pharmaceutically acceptable salt and application thereof in medicine
CA3132832A1 (en) 2018-04-09 2019-10-17 Howard E. Gendelman Antiviral prodrugs and formulations thereof
WO2020018459A1 (en) 2018-07-16 2020-01-23 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
EP3823629A4 (en) 2018-07-19 2022-05-04 Merck Sharp & Dohme Corp. Phosphinic amide prodrugs of tenofovir
US20210393631A1 (en) 2018-09-19 2021-12-23 Gilead Sciences, Inc. Integrase inhibitors for the prevention of hiv
WO2021011891A1 (en) 2019-07-18 2021-01-21 Gilead Sciences, Inc. Long-acting formulations of tenofovir alafenamide
CA3146263A1 (en) 2019-07-19 2021-01-28 Jose Gerardo Garcia Lerma Hiv pre-exposure prophylaxis
EP4017476A1 (en) 2019-08-19 2022-06-29 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US20220298185A1 (en) * 2019-08-22 2022-09-22 Emory University Nucleoside Prodrugs and Uses Related Thereto
US20220372171A1 (en) * 2019-09-20 2022-11-24 Abbott Rapid Diagnostics International Unlimited Company Antibody directed against tenofovir and derivatives thereof
JP2023502530A (en) 2019-11-26 2023-01-24 ギリアード サイエンシーズ, インコーポレイテッド Capsid inhibitors for HIV prevention
TWI789695B (en) 2020-01-27 2023-01-11 美商基利科學股份有限公司 Methods for treating sars cov-2 infections
EP4085062A1 (en) 2020-02-20 2022-11-09 Cipla Limited Novel salts and/or co-crystals of tenofovir alafenamide
CN115298181A (en) 2020-03-12 2022-11-04 吉利德科学公司 Process for preparing 1' -cyanonucleosides
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021202669A2 (en) 2020-04-01 2021-10-07 Reyoung Corporation Nucleoside and nucleotide conjugate compounds and uses thereof
WO2021207049A1 (en) 2020-04-06 2021-10-14 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carbanucleoside analogs
KR20210125298A (en) 2020-04-08 2021-10-18 주식회사 파마코스텍 New process for the preparation of Tenofovir alafenamide hemi-tartrate
EP4143199A1 (en) 2020-04-21 2023-03-08 Ligand Pharmaceuticals, Inc. Nucleotide prodrug compounds
KR20230018473A (en) 2020-05-29 2023-02-07 길리애드 사이언시즈, 인코포레이티드 How to treat remdesivir
IL299202A (en) 2020-06-24 2023-02-01 Gilead Sciences Inc 1'-cyano nucleoside analogs and uses thereof
KR20230027275A (en) 2020-06-25 2023-02-27 길리애드 사이언시즈, 인코포레이티드 Capsid inhibitors for HIV treatment
CN113970612B (en) * 2020-07-22 2023-08-01 北京四环制药有限公司 Method for measuring related substances of propiophenone tenofovir by high performance liquid chromatography
EP4204421A2 (en) 2020-08-27 2023-07-05 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
CN112336695B (en) * 2020-09-28 2023-01-03 华北制药华坤河北生物技术有限公司 Propofol fumarate and tenofovir tablet, preparation method thereof and detection method of related substances
KR20230107288A (en) 2020-11-11 2023-07-14 길리애드 사이언시즈, 인코포레이티드 Method for identifying HIV patients susceptible to therapy with gp120 CD4 binding site-directed antibody
US11667656B2 (en) 2021-01-27 2023-06-06 Apotex Inc. Crystalline forms of Tenofovir alafenamide
CN113214322B (en) * 2021-04-30 2022-10-25 山东立新制药有限公司 Green and environment-friendly preparation method of tenofovir
WO2022251594A1 (en) * 2021-05-27 2022-12-01 Antios Therapeutics, Inc. Pharmacokinetics and dose-related improvments in subjects treated with phosphoramidate clevudine prodrugs
TW202342447A (en) 2021-12-03 2023-11-01 美商基利科學股份有限公司 Therapeutic compounds for hiv virus infection
US20230203071A1 (en) 2021-12-03 2023-06-29 Zhimin Du Therapeutic compounds for hiv virus infection
US11787825B2 (en) 2021-12-03 2023-10-17 Gilead Sciences, Inc. Therapeutic compounds for HIV virus infection
CN114369120A (en) * 2022-01-28 2022-04-19 石家庄龙泽制药股份有限公司 Preparation method of key intermediate of prophenoltenofovir
TW202400185A (en) 2022-03-02 2024-01-01 美商基利科學股份有限公司 Compounds and methods for treatment of viral infections
TW202400172A (en) 2022-04-06 2024-01-01 美商基利科學股份有限公司 Bridged tricyclic carbamoylpyridone compounds and uses thereof
US20240034724A1 (en) 2022-07-01 2024-02-01 Gilead Sciences, Inc. Therapeutic compounds useful for the prophylactic or therapeutic treatment of an hiv virus infection
WO2024044477A1 (en) 2022-08-26 2024-02-29 Gilead Sciences, Inc. Dosing and scheduling regimen for broadly neutralizing antibodies
WO2024076915A1 (en) 2022-10-04 2024-04-11 Gilead Sciences, Inc. 4'-thionucleoside analogues and their pharmaceutical use

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CS233665B1 (en) 1983-01-06 1985-03-14 Antonin Holy Processing of isomere o-phosphonylmethylderivative of anantiomere racemic vicinal diene
CS263951B1 (en) 1985-04-25 1989-05-12 Antonin Holy 9-(phosponylmethoxyalkyl)adenines and method of their preparation
CS263952B1 (en) 1985-04-25 1989-05-12 Holy Antonin Remedy with antiviral effect
CS264222B1 (en) 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
US5650510A (en) 1986-11-18 1997-07-22 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiviral phosphonomethoxyalkylene purine and pyrimidine derivatives
US5057301A (en) 1988-04-06 1991-10-15 Neorx Corporation Modified cellular substrates used as linkers for increased cell retention of diagnostic and therapeutic agents
US5053215A (en) * 1988-05-26 1991-10-01 University Of Florida NMR-assayable ligand-labelled trifluorothymidine containing composition and method for diagnosis of HSV infection
US5744600A (en) 1988-11-14 1998-04-28 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Phosphonomethoxy carbocyclic nucleosides and nucleotides
US5688778A (en) 1989-05-15 1997-11-18 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Nucleoside analogs
JP2648516B2 (en) 1989-07-27 1997-09-03 ダイセル化学工業株式会社 Separation of stereoisomers
US5624898A (en) * 1989-12-05 1997-04-29 Ramsey Foundation Method for administering neurologic agents to the brain
JP2925753B2 (en) 1990-02-23 1999-07-28 ダイセル化学工業株式会社 Optical isomer separation method
FI911875A (en) * 1990-04-20 1991-10-21 Bristol Myers Squibb Co KIRALA 2- (PHOSPHONOMETOXY) PROPYLGUANINER SOM ANTIVIRALA AEMNEN.
US5302585A (en) 1990-04-20 1994-04-12 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Use of chiral 2-(phosphonomethoxy)propyl guanines as antiviral agents
SK280313B6 (en) 1990-04-24 1999-11-08 �Stav Organick� Chemie A Biochemie Av �R N-(3-fluoro-2-phosphonylmethoxypropyl) derivatives of purine and pyrimidine heterocyclic bases, process for their preparation and their use
US5627165A (en) * 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
US5177064A (en) 1990-07-13 1993-01-05 University Of Florida Targeted drug delivery via phosphonate derivatives
CS276072B6 (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
AU653552B2 (en) 1990-08-10 1994-10-06 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Novel process for the preparation of nucleotides
DE69129650T2 (en) * 1990-09-14 1999-03-25 Acad Of Science Czech Republic Precursor of phosphonates
US5827819A (en) * 1990-11-01 1998-10-27 Oregon Health Sciences University Covalent polar lipid conjugates with neurologically active compounds for targeting
US5208221A (en) * 1990-11-29 1993-05-04 Bristol-Myers Squibb Company Antiviral (phosphonomethoxy) methoxy purine/pyrimidine derivatives
CZ284678B6 (en) 1991-05-20 1999-01-13 Ústav Organické Chemie A Biochemie Avčr Di(2-propyl)esters of 1-fluoro-2-phosphonomethoxy-3-p-toluenesulfonyloxypropanes, process of their preparation and use
US5498752A (en) 1991-08-22 1996-03-12 Daicel Chemical Industries, Ltd. Process for recovering optical isomers and solvent, process for using solvent by circulation and process for reusing optical isomers in optical resolution
JP3010816B2 (en) 1991-08-22 2000-02-21 ダイセル化学工業株式会社 Method for recovering optical isomer and solvent in optical resolution, method for recycling solvent, and method for reusing optical isomer
JP3497505B2 (en) 1991-10-11 2004-02-16 インスティテュート オブ オルガニック ケミストリー アンド バイオケミストリー オブ ザ アカデミー オブ サイエンシズ オブ ザ チェコ パブリック Antiviral acyclic phosphonomethoxyalkyl-substituted alkenyl and alkynylpurine and pyrimidine derivatives
US6057305A (en) 1992-08-05 2000-05-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiretroviral enantiomeric nucleotide analogs
IL106998A0 (en) * 1992-09-17 1993-12-28 Univ Florida Brain-enhanced delivery of neuroactive peptides by sequential metabolism
US6413949B1 (en) * 1995-06-07 2002-07-02 D-Pharm, Ltd. Prodrugs with enhanced penetration into cells
CA2171868A1 (en) * 1993-09-17 1995-03-23 Petr Alexander Method for dosing therapeutic compounds
US5656745A (en) * 1993-09-17 1997-08-12 Gilead Sciences, Inc. Nucleotide analogs
US5798340A (en) * 1993-09-17 1998-08-25 Gilead Sciences, Inc. Nucleotide analogs
BR9407510A (en) 1993-09-17 1997-01-07 Gilead Sciences Inc Nucleotide analogues
GB9505025D0 (en) 1995-03-13 1995-05-03 Medical Res Council Chemical compounds
US5977061A (en) 1995-04-21 1999-11-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic N6 - substituted nucleotide analagues and their use
JP2002500617A (en) * 1995-05-26 2002-01-08 ジンタ・インコーポレイテッド Synthesis method and composition of organic phosphorus derivative
NZ325704A (en) 1995-12-29 2000-02-28 Gilead Sciences Inc Nucleotide analogs
US5717095A (en) * 1995-12-29 1998-02-10 Gilead Sciences, Inc. Nucleotide analogs
US5874577A (en) * 1996-04-03 1999-02-23 Medichem Research, Inc. Method for the preparing 9-12-(Diethoxyphosphonomethoxy)ethyl!adenine and analogues thereof
US5922695A (en) * 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
CA2261619C (en) * 1996-07-26 2006-05-23 Gilead Sciences, Inc. Nucleotide analogs
US5739314A (en) 1997-04-25 1998-04-14 Hybridon, Inc. Method for synthesizing 2'-O-substituted pyrimidine nucleosides
DE69809750T2 (en) * 1997-07-25 2003-08-21 Gilead Sciences Inc NUCLEOTIDE ANALOG COMPOSITION AND SYNTHESIS PROCEDURE
US5935946A (en) 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
AU747163B2 (en) 1997-07-25 2002-05-09 Gilead Sciences, Inc. Nucleotide analog compositions
EP1037649B1 (en) * 1997-12-17 2009-09-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
EP1167972B1 (en) * 1998-01-23 2017-03-08 Kiadis Pharma Intellectual Property B.V. Enzyme catalyzed therapeutic agents
US6169078B1 (en) * 1998-05-12 2001-01-02 University Of Florida Materials and methods for the intracellular delivery of substances
WO1999067284A2 (en) * 1998-06-20 1999-12-29 Washington University Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy
US6169879B1 (en) * 1998-09-16 2001-01-02 Webtv Networks, Inc. System and method of interconnecting and using components of home entertainment system
GB9821058D0 (en) 1998-09-28 1998-11-18 Univ Cardiff Chemical compound
TWI230618B (en) 1998-12-15 2005-04-11 Gilead Sciences Inc Pharmaceutical compositions of 9-[2-[[bis[(pivaloyloxy)methyl]phosphono]methoxy]ethyl]adenine and tablets or capsules containing the same
PL204458B1 (en) 1999-02-12 2010-01-29 Univ Cardiff Phosphoramidate, and mono-, di-, and tri-phosphate esters of (1r, cis)-4-(6-amino-9h-purin-9-yl)-2-cyclopentene-1-methanol as antiviral agents
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
JP4387669B2 (en) * 2000-10-13 2009-12-16 ザイジェン エス.アー. Intracellular delivery of biological effectors with novel transporter peptide sequences
US20020119433A1 (en) * 2000-12-15 2002-08-29 Callender Thomas J. Process and system for creating and administering interview or test

Also Published As

Publication number Publication date
EP1301519B1 (en) 2015-02-25
PL360490A1 (en) 2004-09-06
AP2003002724A0 (en) 2003-06-30
CZ2003413A3 (en) 2003-12-17
NO2016006I1 (en) 2016-04-19
PT1301519E (en) 2015-06-11
HRP20160074B1 (en) 2021-09-03
IS6689A (en) 2003-01-17
PT2682397T (en) 2017-05-31
AU2001282941C1 (en) 2016-12-22
BRPI0112646B8 (en) 2021-05-25
CZ304886B6 (en) 2015-01-07
NO20120466L (en) 2003-03-20
EA200300188A1 (en) 2003-06-26
HRP20030047B1 (en) 2016-02-26
ZA200210271B (en) 2003-12-31
CN100402539C (en) 2008-07-16
NO20030270L (en) 2003-03-20
US20040018150A1 (en) 2004-01-29
KR100749160B1 (en) 2007-08-14
FR16C0013I1 (en) 2016-05-27
PL213214B1 (en) 2013-01-31
US20050009043A1 (en) 2005-01-13
UA75889C2 (en) 2006-06-15
JP5111551B2 (en) 2013-01-09
HUP0301307A3 (en) 2005-12-28
FR16C0013I2 (en) 2016-09-09
HRP20160074A2 (en) 2016-03-11
JP2010174033A (en) 2010-08-12
US20050124585A1 (en) 2005-06-09
EP3235823A1 (en) 2017-10-25
JP2009062383A (en) 2009-03-26
US7390791B2 (en) 2008-06-24
CA2725819C (en) 2015-09-22
NO20030270D0 (en) 2003-01-20
JP4651264B2 (en) 2011-03-16
NZ535408A (en) 2006-09-29
US7803788B2 (en) 2010-09-28
AU8294101A (en) 2002-02-05
US20050124583A1 (en) 2005-06-09
TR200300055T2 (en) 2004-12-21
US20060024659A1 (en) 2006-02-02
HUS1900027I1 (en) 2021-03-29
US20080227754A1 (en) 2008-09-18
BG66037B1 (en) 2010-11-30
IS2985B (en) 2017-09-15
NZ523438A (en) 2005-02-25
HK1054238A1 (en) 2003-11-21
EE05366B1 (en) 2010-12-15
JP5063554B2 (en) 2012-10-31
US20020119443A1 (en) 2002-08-29
IL153658A0 (en) 2003-07-06
BR0112646A (en) 2003-06-24
NZ536942A (en) 2006-03-31
CN1706855A (en) 2005-12-14
BG107572A (en) 2003-11-28
HRP20030047A2 (en) 2007-08-31
HU230960B1 (en) 2019-06-28
NO2023006I1 (en) 2023-02-03
SI1301519T1 (en) 2015-07-31
HUS000494I2 (en) 2021-03-29
HK1243711A1 (en) 2018-07-20
SI2682397T1 (en) 2017-08-31
ES2627903T3 (en) 2017-08-01
ES2536972T5 (en) 2022-04-06
EE200300029A (en) 2004-10-15
JP2011140506A (en) 2011-07-21
ES2536972T3 (en) 2015-06-01
NO336718B1 (en) 2015-10-26
HUP0301307A2 (en) 2003-09-29
NO20131717L (en) 2003-03-20
WO2002008241A3 (en) 2002-08-29
CN1443189A (en) 2003-09-17
EA004926B1 (en) 2004-10-28
DK2682397T3 (en) 2017-06-19
CY1119411T1 (en) 2018-03-07
OA12393A (en) 2006-04-18
US20050124584A1 (en) 2005-06-09
US20030219727A1 (en) 2003-11-27
CA2416757C (en) 2011-02-15
NO20150909L (en) 2003-03-20
MXPA03000587A (en) 2004-04-05
CZ304734B6 (en) 2014-09-10
AU2001282941B2 (en) 2006-04-27
KR20060105807A (en) 2006-10-11
DK1301519T4 (en) 2021-12-20
EP1301519A2 (en) 2003-04-16
EP2682397A1 (en) 2014-01-08
LTPA2016009I1 (en) 2016-04-25
EP2682397B1 (en) 2017-04-19
CY2016008I2 (en) 2016-12-14
NL300803I2 (en) 2016-06-30
AP1466A (en) 2005-09-22
NO2016006I2 (en) 2016-04-19
US20050159392A1 (en) 2005-07-21
KR20030022295A (en) 2003-03-15
WO2002008241A2 (en) 2002-01-31
BRPI0112646B1 (en) 2017-10-17
CA2725819A1 (en) 2002-01-31
AU2005225039B2 (en) 2008-09-25
LTC1301519I2 (en) 2023-02-27
BE2016C018I2 (en) 2020-08-20
DK1301519T3 (en) 2015-05-26
LT2682397T (en) 2017-06-12
AU2005225039A1 (en) 2005-11-10
KR100767432B1 (en) 2007-10-17
CA2416757A1 (en) 2002-01-31
CN1291994C (en) 2006-12-27
HRP20160074A8 (en) 2016-07-29
CY2016008I1 (en) 2016-12-14
EP1301519B2 (en) 2021-11-10
JP2004504402A (en) 2004-02-12
LU93029I2 (en) 2016-06-14

Similar Documents

Publication Publication Date Title
CA2725819C (en) Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
AU2001282941A1 (en) Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20150528

FZDE Discontinued

Effective date: 20171031