US20050159392A1 - Prodrugs of phosphonate nucleotide analogues - Google Patents

Prodrugs of phosphonate nucleotide analogues Download PDF

Info

Publication number
US20050159392A1
US20050159392A1 US11/031,228 US3122805A US2005159392A1 US 20050159392 A1 US20050159392 A1 US 20050159392A1 US 3122805 A US3122805 A US 3122805A US 2005159392 A1 US2005159392 A1 US 2005159392A1
Authority
US
United States
Prior art keywords
salt
nucleotide analog
analog compound
diastereomer
solvate
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
US11/031,228
Inventor
Mark Becker
Harlan Chapman
Tomas Cihlar
Eugene Eisenberg
Gong-Xin He
Michael Kernan
William Lee
Ernest Prisbe
John Rohloff
Mark 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.)
Individual
Original Assignee
Individual
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=US20050159392(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US11/031,228 priority Critical patent/US20050159392A1/en
Publication of US20050159392A1 publication Critical patent/US20050159392A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric 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
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • 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

Definitions

  • This application relates to prodrugs of methoxyphosphonate nucleotide analogues. In particular it relates to improved methods for making and identifying such prodrugs.
  • Prodrugs of methoxyphosphonate nucleotide analogues intended for antiviral or antitumor therapy traditionally have been selected for their systemic effect.
  • 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.
  • 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 nucleotide analogue.
  • a screening method for identifying a methoxyphosphonate nucleotide analogue prodrug conferring enhanced activity in a target tissue comprising:
  • step (d) determining the relative antiviral activity conferred by the prodrug in the tissues in step (c).
  • the target tissue are sites where HIV is actively replicated and/or which serve as an HIV reservoir, and the non-target tissue is an intact animal.
  • 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),
  • Ra is H or methyl
  • compositions thereof and chirally enriched compositions thereof, salts, their free base and solvates thereof.
  • a preferred compound of this invention has the structure (2)
  • R 1 is an oxyester which is hydrolyzable in vivo, or hydroxyl
  • B is a heterocyclic base
  • R 2 is hydroxyl, or the residue of an amino acid bonded to the P atom through an amino group of the amino acid and having each carboxy substituent of the amino acid optionally esterified, but not both of R 1 and R 2 are hydroxyl;
  • E is —(CH 2 ) 2 —, —CH(CH 3 )CH 2 —, —CH(CH 2 F)CH 2 —, —CH(CH 2 OH)CH 2 , —CH(CH ⁇ CH 2 )CH 2 —, —CH(C ⁇ CH)CH 2 —, —CH(CH 2 N 3 )CH 2 —, —CH(R 6 )OCH(R 6 ′)—, —CH(R 9 )CH 2 O— or —CH(R 8 )O—, wherein the right hand bond is linked to the heterocyclic base;
  • the broken line represents an optional double bond
  • R 4 and R 5 are independently hydrogen, hydroxy, halo, amino or a substituent having 1-5 carbon atoms selected from acyloxy, alkyoxy, alkylthio, alkylamino and dialkylamino;
  • R 6 and R 6 ′ are independently H, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, or C 2 -C 7 alkanoyl;
  • R 7 is independently H, C 1 -C 6 alkyl, or are taken together to form —O— or —CH 2 —;
  • R 8 is H, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl or C 1 -C 6 haloalkyl;
  • R 9 is H, hydroxymethyl or acyloxymethyl
  • 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)
  • R 5 is methyl or hydrogen
  • R 6 independently is H, alkyl, alkenyl, alkynyl, aryl or arylalkyl, or R 6 independently is alkyl, alkenyl, alkynyl, aryl or arylalkyl which is substituted with from 1 to 3 substituents selected from alkylamino, alkylaminoalkyl, dialkylaminoalkyl, dialkylamino, hydroxyl, oxo, halo, amino, alkylthio, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, arylalkoxy, arylalkoxyalkyl, haloalkyl, nitro, nitroalkyl, azido, azidoalkyl, alkylacyl, alkylacylalkyl, carboxyl, or alkylacylamino;
  • R 7 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;
  • R 11 is amino, alkylamino, oxo, or dialkylamino
  • R 12 is amino or H
  • a preferred embodiment of this invention is the compound of structure (6), 9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine, also designated herein GS-7340
  • Another preferred embodiment of this invention is the fumarate salt of structure (5) (structure (7)), 9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine fumarate (1:1), also designated herein GS-7340-2
  • compositions containing pharmaceutically acceptable excipients are used in effective doses in the therapy or prophylaxis of viral (particularly HIV or hepadnaviral) infections.
  • a method for the facile manufacture of 9-[2-(phosphonomethoxy)propyl]adenine (hereinafter “PMPA” or 9-[2-(phosphonomethoxy)ethyl]adenine (hereinafter “PMEA”) using magnesium alkoxide comprises combining 9-(2-hydroxypropyl)adenine or 9-(2-hydroxyethyl)adenine, protected p-toluenesulfonyloxymethylphosphonate and magnesium alkoxide, and recovering PMPA or PMEA, respectively.
  • FIG. 1 shows HPLC/C-14 traces of PBMC extracts from human blood incubated for 1 h at 37° C. with TDF, GS-7340 and PMPA.
  • FIG. 2 shows PMPA and Prodrug concentration in plasma and PBMCs following oral administration of GS 7340-2 to Dogs at 10 mg-eq/kg.
  • FIG. 3 depicts Tenofovir exposure in PBMCs and plasma upon administration of 10 mg-eq/kg in dogs.
  • the methoxyphosphonate nucleotide analogue parent drugs for use in this screening method are compounds having the structure A-OCH 2 P(O)(OH) 2 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 wherein the group B is defined below and group E is defined above. Examples are described in U.S. Pat. Nos.
  • 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.
  • 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 monoamino 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. Pat. No.
  • 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.
  • pro-moieties bound to the parent drug may be the same or different.
  • 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 desirably is structurally and stereochemically 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.
  • Chiral sites are at the phosphorus atom and are also found in its substituents.
  • 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.
  • the R isomer of PMPA is preferred as it is more active than the corresponding S isomer.
  • 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.
  • 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.
  • 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%.
  • the remaining steps of the screening method of this invention are used to identify a prodrug possessing the required selectivity for the target tissue.
  • the prodrugs are labeled with a detectable group, e.g. radiolabeled, in order to facilitate detection later in tissues or cells.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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).
  • target tissue i.e., prodrugs metabolized to antivirally active forms incidental to viral infection but which remain substantially unmetabolized in uninfected cells.
  • 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 malignancies, e.g. leukemias.
  • 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.
  • 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.
  • a metabolite of the prodrug which metabolite is known to have, or is converted to, a metabolite having antiviral or antitumor activity.
  • the active metabolite is the diphosphate of the phosphonate parent compounds.
  • 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.
  • 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.
  • 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.
  • step (d) 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.
  • prodrug candidates can be undertaken before the practice of the method of this invention.
  • the prodrug will need to be capable of passing largely unmetabolized 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.
  • antiviral activity is applicable to antitumor prodrugs of methoxyphosphonate nucleotide analogues as well.
  • these include, for example, prodrugs of PMEG, the guanyl analogue of PMEA.
  • 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.
  • a prodrug is considered to be selective if the activity or concentration of parent drug in the target tissue (% dose distribution) is greater than 2 ⁇ , and preferably 5 ⁇ , that of the parent compound in non-target tissue.
  • 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 10 ⁇ concentration or activity in the target tissue as compared with the prototype, although the degree of selectivity is a matter of discretion.
  • this method comprises reacting 9-(2-hydroxypropyl)adenine (HPA) or 9-(2-hydroxyethyl)adenine (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.
  • 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.
  • 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 C 1 -C 6 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.
  • 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 B is selected from the structures
  • R 15 is H, OH, F, Cl, Br, I, OR 16 , SH, SR 16 , NH 2 , or NHR 17 ;
  • R 16 is C 1 -C 6 alkyl or C 2 -C 6 alkenyl including —CH 3 , —CH 2 CH 3 , —CH 2 C ⁇ CH, —CH 2 CH ⁇ CH 2 and —C 3 H 7 ;
  • R 17 is C 1 -C 6 alkyl or C 2 -C 6 alkenyl including —CH 3 , —CH 2 CH 3 , —CH 2 C ⁇ CH, —CH 2 CH ⁇ CH 2 , and —C 3 H 7;
  • R 18 is N, CF, CCl, CBr, CI, CR 19 , CSR 19 , or COR 19 ;
  • R 19 is H, C 1 -C 9 alkyl, C 2 -C 9 alkenyl, C 2 -C 9 alkynyl, C 1 -C 9 alkyl-C 1 -C 9 alkoxy, or C 7 -C 9 aryl-alkyl unsubstituted or substituted by OH, F, Cl, Br or I, R 19 therefore including —CH 3 , —CH 2 CH 3 , —CHCH 2 , —CHCHBr, —CH 2 CH 2 Cl,
  • —CH 2 CH 2 F —CH 2 CCH, —CH 2 CHCH 2 , —C 3 H 7 , —CH 2 OH, —CH 2 OCH 3 , —CH 2 OC 2 H 5 , —CH 2 OCCH, —CH 2 OCH 2 CHCH 2 , —CH 2 C 3 H 7 , —CH 2 CH 2 OH, —CH 2 CH 2 OCH 3 ,
  • R 20 is N or CH
  • R 21 is N, CH, CCN, CCF 3 , CC ⁇ CH or CC(O)NH 2 ;
  • R 22 is H, OH, NH 2 , SH, SCH 3 , SCH 2 CH 3 , SCH 2 C ⁇ CH, SCH 2 CH ⁇ CH 2 , SC 3 H 7 , NH(CH 3 ), N(CH 3 ) 2 , NH(CH 2 CH 3 ), N(CH 2 CH 3 ) 2 , NH(CH 2 C ⁇ CH), NH(CH 2 CHCH 2 ), NH(C 3 H 7 ), halogen (F, Cl, Br or I) or X wherein X is —(CH 2 ) m (O) n (CH 2 ) m N(R 10 ) 2 wherein each m is independently 0-2, n is 0-1, and
  • R 10 independently is H
  • C 1 -C 15 alkyl C 2 -C 15 alkenyl, C 6 -C 15 arylalkenyl, C 6 -C 15 arylalkynyl, C 2 -C 15 alkynyl, C 1 -C 6 -alkylamino-C 1 -C 6 alkyl, C 5 -C 15 aralkyl, C 6 -C 15 heteroaralkyl, C 5 -C 6 aryl, C 2 -C 6 heterocycloalkyl,
  • both R 10 are joined together with N to form a saturated or unsaturated C 2 -C 5 heterocycle containing one or two N heteroatoms and optionally an additional O or S heteroatom,
  • R 10 groups which is substituted with 1 to 3 halo, CN or N 3 ; but optionally at least one R 10 group is not H;
  • R 23 is H, OH, F, Cl, Br, I, SCH 3 , SCH 2 CH 3 , SCH 2 C ⁇ CH, SCH 2 CHCH 2 , SC 3 H 7 , OR 16 , NH 2 , NHR 17 or R 22 ;
  • R 24 is O, 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.
  • DMT 4,4′-dimethoxytrityl
  • 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 N 4 -benzoylcytosine, N 6 -benzoyladenine, N 2 -isobutyrylguanine and the like.
  • Protected bases have the formulas Xa.1, XIa.1, XIb.1, XIIa.1 or XIIIa.1
  • R 18 , R 20 , R 21 , R 24 have the meanings previously defined;
  • R 22A is R 39 or R 22 provided that R 22 is not NH 2 ;
  • R 23A is R 39 or R 23 provided that R 23 is not NH 2 ;
  • R 39 is NHR 40 , NHC(O)R 36 or CR 41 N(R 38 ) 2 wherein R 36 is C 1 -C 19 alkyl, C 1 -C 19 alkenyl, C 3 -C 10 aryl, adamantoyl, alkylanyl, or C 3 -C 10 aryl substituted with 1 or 2 atoms or groups selected from halogen, methyl, ethyl, methoxy, ethoxy, hydroxy and cyano;
  • R 38 is C 1 -C 10 alkyl, or both R 38 together are 1-morpholino, 1-piperidine or 1-pyrrolidine;
  • R 40 is C 1 -C 1a alkyl, including methyl, eth
  • R 39 is present at R 22A or R 23A , both R 39 groups on the same base will generally be the same.
  • Exemplary R 36 are phenyl, phenyl substituted with one of the foregoing R 36 aryl substituents, —C 10 H 15 (where C 10 H 15 is 2-adamantoyl), —CH 2 -C 6 H 5 , —C 6 H 5 , —CH(CH 3 ) 2 , —CH 2 CH 3 , 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-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza-8-aza derivatives of adenine, guanine, 2-aminopurine, 2,6-diaminopurine, 2-amino-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-6-chloropurine
  • 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-diaminopurinyl, 2-aminopurinyl, 6-chloro-2-aminopurinyl and 6-thio-2-aminopurinyl, or a B′ is a 1-pyrimidinyl residue selected from cytosinyl, 5-halocytosinyl, and 5-(C 1 -C 3 -alkyl)cytosinyl.
  • Preferred B groups have the formula
  • R 22 independently is halo, oxygen, NH 2 , X or H, but optionally at least one R 22 is X;
  • X is —CH 2 ) m (O) n (CH 2 ) m N(R 10 ) 2 wherein m is 0-2, n is 0-1, and
  • R 10 independently is H
  • C 1 -C 15 alkyl C 2 -C 15 alkenyl, C 6 -C 15 arylalkenyl, C 6 -C 15 arylalkynyl, C 2 -C 15 alkynyl, C 1 -C 6 -alkylamino-C 1 -C 6 alkyl, C 5 -C 15 aralkyl, C 6 -C 15 heteroaralkyl, C 5 -C 6 aryl, C 2 -C 6 heterocycloalkyl,
  • both R 10 are joined together with N to form a saturated or unsaturated C 2 -C 5 heterocycle containing one or two N heteroatoms and optionally an additional O or S heteroatom,
  • R 10 groups is substituted with 1 to 3 halo, CN or N 3 ; but optionally at least one R 10 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.
  • the E group is —CH(CH 3 )CH 2 — or —CH 2 CH 2 —.
  • 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).
  • R 1 is an in vivo hydrolyzable oxyester having the structure —OR 35 or —OR 6 wherein R 35 is defined in column 64, line 49 of U.S. Pat. No. 5,798,340, herein incorporated by reference, and R 6 is defined above.
  • R 1 is aryloxy, ordinarily unsubstituted or para-substituted (as defined in R 6 ) phenoxy.
  • R 2 is an amino acid residue, optionally provided that any carboxy group linked by less than about 5 atoms to the amidate N is esterified.
  • R 2 typically has the structure
  • n 1 or 2;
  • R 12 independently is H or C 1 -C 9 alkyl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, O, N, COOR 11 and halogen; C 3 -C 6 aryl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, O, N, COOR 11 and halogen; or C 3 -C 9 aryl-alkyl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, O, N, COOR 11 and halogen;
  • R 13 independently is C(O)—OR 11 ; amino; amide; guanidinyl; imidazolyl; indolyl; sulfoxide; phosphoryl; C 1 -C 3 alkylamino; C 1 -C 3 alkyldiamino; C 1 -C 6 alkenylamino; hydroxy; thiol; C 1 -C 3 alkoxy; C 1 -C 3 alkthiol; (CH 2 ),COOR 11 ; C 1 -C 6 alkyl which is unsubstituted or substituted with OH, halogen, SH, NH 2 , phenyl, hydroxyphenyl or C 7 -C 10 alkoxyphenyl; C 2 -C 6 alkenyl which is unsubstituted or substituted with OH, halogen, SH, NH 2 , phenyl, hydroxyphenyl or C 7 -C 10 alkoxyphenyl; and C 6 -C 12
  • R 14 is H or C 1 -C 9 alkyl or C 1 -C 9 alkyl independently substituted with OH, halogen, COOR 11 , O or N; C 3 -C 6 aryl; C 3 -C 6 aryl which is independently substituted with OH, halogen, COOR 11 , O or N; or C 3 -C 6 arylalkyl which is independently substituted with OH, halogen, COOR 11 , O or N.
  • the invention includes metabolites in which the phenoxy and isopropyl esters have been hydrolyzed to —OH.
  • the de-esterified enriched phosphonoamidate metabolites of compounds (5a), 5(b) and (6) are included within the scope of this invention.
  • Aryl and “O” or “N” substitution are defined in column 16, lines 42-58, of U.S. Pat. No. 5,798,340.
  • amino acids are in the natural or l amino acids. Suitable specific examples are set forth in U.S. Pat. No. 5,798,340, for instance Table 4 and col. 8-10 therein.
  • the prodrug compounds of this invention are provided in the form of free base or the various salts enumerated in U.S. Pat. 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. Pat. No. 5,798,340.
  • These prodrugs have the antiviral and utilities already established for the parent drugs (see U.S. Pat. No. 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.
  • HSA 9-(2-Hydroxyethyl)adenine
  • 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 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 (0.77 g, 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 sodium sulfate, and concentrated under reduced pressure to afford 2.4 g III as a tan foam.
  • Chiralpak AS is a proprietary packing material manufactured by Diacel and sold in North America by Chiral Technologies, Inc. (U.S. Pat. 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 amylosetris[(S)- ⁇ -methylbenzyl carbamate] 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 min. duration) peak to elute from the column.
  • 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.
  • 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 vacuo to yield the purified diastereomer as a friable foam.
  • the GS-7340 fraction comprised about 50% of the total recovered mass.
  • 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., 20 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.
  • GS-7171 (III) was chromatographed by reverse phase HPLC to separate the diastereomers using the following summary protocol.
  • the oil was purified by chromatography over a 15 ⁇ 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.
  • the diastereomeric 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)
  • 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)
  • Phenyl PMPA Ethyl L-Alanyl Amidate.
  • Phenyl PMPA (15.0 g, 41.3 mmol)
  • L-alanine ethyl ester hydrochloride 12.6 g, 83 mmol
  • triethylamine (11.5 mL, 83 mmol) were slurried together in 500 mL pyridine under dry N 2 .
  • This suspension was combined with a solution of triphenylphosphine (37.9 g, 145 mmol), Aldrithiol 2 (2,2′-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 ⁇ 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 1 H 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.
  • 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 GS-7340 than TDF after oral administration.
  • both prodrugs and PMPA were radiolabeled and spiked into intact human whole blood at equimolar 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.
  • GS-7340 results in 10 ⁇ and 30 ⁇ the total intracellular concentration of PMPA species in PBMCs as compared to TDF and PMPA, respectively.
  • 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 10 ⁇ 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 FIG. 1 . TABLE 2 PMPA Metabolites in Plasma, PBMCs and RBCs After 1 h Incubation of PMPA Prodrugs or PMPA in Human Blood.
  • Met. X and Met Y are shown in Table 5. Lower case “p” 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 50 values seen in vitro.
  • IC 50 values of tenofovir, TDF, GS-7340, several nucleosides and the protease inhibitor nelfinivir are listed. As shown, nelfinavir and GS-7340 are 2-3 orders of magnitude more potent than all other nucleotides or nucleosides.
  • “Phe-methylester” is the methylphenylalaninyl monoamidate, phenyl monoester of tenofovir; “gly-methylester” is the methylglycyl monoamidate, phenyl monoester of tenofovir.
  • isomer A is believed to have the same absolute stereochemistry as GS-7340 (S), and isomer B is believed to have the same absolute stereochemistry that of GS-7339.
  • the in vitro metabolism and stability of separated diastereomers were determined in PLCE, MT-2 extract and human plasma.
  • a biological sample listed below, 80 ⁇ L 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 60 ⁇ L 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.
  • reaction mixture 20 ⁇ L
  • 60 ⁇ L of methanol containing the internal standard 60 ⁇ L
  • the mixture thus obtained was centrifuiged 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.
  • the buffer systems used in the studies are as follows.
  • GS 7340 The pharmacokinetics of GS 7340 were studied in dogs after oral administration of a 10 mg-eq/kg dose.
  • PBMC Peripheral Blood Mononuclear Cell
  • the concentration of PMPA in dog plasma samples was determined by derivatizing PMPA with chloroacetaldehyde to yield a highly fluorescent N 1 , N 6 -ethenoadenine derivative (L. Naesens, J. Balzarini, and E. De Clercq, Clin. Chem. 38, 480 (1992). Briefly, plasma (100 ⁇ l) was mixed with 200 ⁇ l acetonitrile to precipitate protein. Samples were then evaporated to dryness under reduced pressure at room temperature. Dried samples were reconstituted in 200 ⁇ l 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 ⁇ l of water for HPLC analysis.
  • the HPLC system was comprised of a P4000 solvent delivery system with AS3000 autoinjector and F2000 fluorescence detector (Thermo Separation, San Jose, Calif.).
  • the column was an Inertsil ODS-2 column (4.6 ⁇ 150 mm).
  • the mobile phases used were: A, 5% acetonitrile in 25 mM potassium phosphate buffer with 5 mM tetrabutyl ammonium bromide (TBABr), pH 6.0; B, 60% acetonitrile in 25 mM potassium phosphate buffer with 5 mM TBABr, pH 6.0.
  • 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 nm and emission at 420 nm, and the injection volume was 10 ⁇ l. Data was acquired and stored by a laboratory data acquisition system (PeakPro, Beckman, Allendale, N.J.).
  • PMPA and prodrug exposures were expressed as areas under concentration curves in plasma or PBMC from zero to 24 hours (AUC).
  • AUC areas under concentration curves in plasma or PBMC from zero to 24 hours
  • FIG. 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.
  • the bar graph in FIG. 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.
  • GS-7340 isopropyl alaninyl monoamidate, phenyl monoester of tenofovir
  • GS-7340 isopropyl alaninyl monoamidate, phenyl monoester of tenofovir
  • aqueous solution 50 mM citric acid, pH 2.2
  • Plasma and peripheral blood mononuclear cells 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 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).
  • TDF the prodrug concentration in the 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.

Abstract

A novel method has led to the identification of novel mixed ester-amidates of PMPA for retroviral or hepadnaviral therapy, including compounds of structure (5a)
Figure US20050159392A1-20050721-C00001
having substituent groups as defined herein. Compositions of these novel compounds in pharmaceutically acceptable excipients and their use in therapy and prophylaxis are provided.

Description

    1. CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of Ser. No. 10/798,692, filed Mar. 11, 2004, now pending, which is a continuation of Ser. No. 10/354,207, filed Jan. 28, 2003, now pending, which is a continuation of Ser. No. 09/909,560, filed Jul. 20, 2001, now abandoned, which claims benefit under 35 U.S.C. § 119(e) to Ser. No. 60/220,021, filed Jul. 21, 2000, all of which are incorporated herein by reference.
  • 2. FIELD
  • 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(O)(OR)2 where A is the residue of a nucleoside analogue and R independently is hydrogen or various protecting or prodrug functionalities. See U.S. Pat. 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 Nimmo, W. S., ed.), pp. 121-126; Bundgaard, H. (1985) Design of Prodrugs (Bundgaard, H., ed.) pp. 70-74 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. All literature and patent citations herein are expressly incorporated by reference.
  • 3. 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 nucleotide 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) determining 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 HIV 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),
    Figure US20050159392A1-20050721-C00002
  • 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)
    Figure US20050159392A1-20050721-C00003
  • 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 invention, we provide diastereomerically enriched compounds of this invention having the structure (3)
    Figure US20050159392A1-20050721-C00004
  • which are substantially free of the diastereomer (4)
    Figure US20050159392A1-20050721-C00005
  • wherein
  • R1 is an oxyester which is hydrolyzable in vivo, or hydroxyl;
  • B is a heterocyclic base;
  • R2 is hydroxyl, or the residue of an amino acid bonded to the P atom through an amino group of the amino acid and having each carboxy substituent of the amino acid 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═CH2)CH2—, —CH(C≡CH)CH2—, —CH(CH2N3)CH2—,
    Figure US20050159392A1-20050721-C00006

    —CH(R6)OCH(R6′)—, —CH(R9)CH2O— or —CH(R8)O—, wherein the right hand bond is linked to the heterocyclic base;
  • the broken line represents an optional double bond;
  • R4 and R5 are independently hydrogen, hydroxy, halo, amino or a substituent having 1-5 carbon atoms selected from acyloxy, alkyoxy, alkylthio, alkylamino and dialkylamino;
  • R6 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 —O— 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)
    Figure US20050159392A1-20050721-C00007
  • which is substantially free of diastereomer (5b)
    Figure US20050159392A1-20050721-C00008
  • wherein
  • R5 is methyl or hydrogen;
  • R6 independently is H, 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 alkylamino, alkylaminoalkyl, dialkylaminoalkyl, dialkylamino, hydroxyl, oxo, halo, amino, alkylthio, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, arylalkoxy, arylalkoxyalkyl, haloalkyl, nitro, nitroalkyl, 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), 9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine, also designated herein GS-7340
    Figure US20050159392A1-20050721-C00009
  • Another preferred embodiment of this invention is the fumarate salt of structure (5) (structure (7)), 9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine fumarate (1:1), also designated herein GS-7340-2
    Figure US20050159392A1-20050721-C00010
  • 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, a method is provided for the facile manufacture of 9-[2-(phosphonomethoxy)propyl]adenine (hereinafter “PMPA” or 9-[2-(phosphonomethoxy)ethyl]adenine (hereinafter “PMEA”) using magnesium alkoxide, which comprises combining 9-(2-hydroxypropyl)adenine or 9-(2-hydroxyethyl)adenine, protected p-toluenesulfonyloxymethylphosphonate and magnesium alkoxide, and recovering PMPA or PMEA, respectively.
  • 4. BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows HPLC/C-14 traces of PBMC extracts from human blood incubated for 1 h at 37° C. with TDF, GS-7340 and PMPA.
  • FIG. 2 shows PMPA and Prodrug concentration in plasma and PBMCs following oral administration of GS 7340-2 to Dogs at 10 mg-eq/kg.
  • FIG. 3 depicts Tenofovir exposure in PBMCs and plasma upon administration of 10 mg-eq/kg in dogs.
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • The methoxyphosphonate nucleotide analogue parent drugs for use in this screening method are compounds having the structure A-OCH2P(O)(OH)2 wherein A is the residue of a nucleoside analogue. These compounds are known
    Figure US20050159392A1-20050721-C00011

    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 wherein the group B is defined below and group E is defined above. Examples are described in U.S. Pat. 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 monoamino 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. Pat. 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.
  • 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 stereochemically 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%.
  • 5.1 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 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 unmetabolized 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 prodrugs 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 2×, and preferably 5×, 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 10× concentration or activity in the target tissue as compared with the prototype, although the degree of selectivity is a matter of discretion.
  • 5.2 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 9-(2-hydroxyethyl)adenine (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.
  • 5.3 Heterocyclic Base
  • In the compounds of this invention depicted in structures (3) and (4), the heterocyclic base B is selected from the structures
    Figure US20050159392A1-20050721-C00012
  • wherein
  • R15 is H, OH, F, Cl, Br, I, OR16, SH, SR16, NH2, or NHR17;
  • R16 is C1-C6 alkyl or C2-C6 alkenyl including —CH3, —CH2CH3, —CH2C≡CH, —CH2CH═CH2 and —C3H7;
  • R17 is C1-C6 alkyl or C2-C6 alkenyl including —CH3, —CH2CH3, —CH2C≡CH, —CH2CH═CH2, and —C3H7;
  • R18 is N, CF, CCl, CBr, CI, CR19, CSR19, or COR19;
  • 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 including —CH3, —CH2CH3, —CHCH2, —CHCHBr, —CH2CH2Cl,
  • —CH2CH2F, —CH2CCH, —CH2CHCH2, —C3H7, —CH2OH, —CH2OCH3, —CH2OC2H5, —CH2OCCH, —CH2OCH2CHCH2, —CH2C3H7, —CH2CH2OH, —CH2CH2OCH3,
  • —CH2CH2OC2H5, —CH2CH2OCCH, —CH2CH2OCH2CHCH2, and
  • —CH2CH2OC3H7;
  • R20 is N or CH;
  • R21 is N, CH, CCN, CCF3, CC≡CH or CC(O)NH2;
  • R22 is H, OH, NH2, SH, SCH3, SCH2CH3, SCH2C≡CH, SCH2CH═CH2, SC3H7, NH(CH3), N(CH3)2, NH(CH2CH3), N(CH2CH3)2, NH(CH2C≡CH), NH(CH2CHCH2), NH(C3H7), halogen (F, Cl, Br or I) or X wherein X is —(CH2)m(O)n(CH2)mN(R10)2 wherein each m is independently 0-2, n is 0-1, and
  • R10 independently is H,
  • C1-C15 alkyl, C2-C15 alkenyl, C6-C15 arylalkenyl, C6-C 15 arylalkynyl, C2-C 15 alkynyl, C1-C6-alkylamino-C1-C6 alkyl, C5-C15 aralkyl, C6-C 15 heteroaralkyl, C5-C6 aryl, C2-C6 heterocycloalkyl,
  • C2-C 15 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 —O—,
  • optionally both R10 are joined together with N to form a saturated or unsaturated C2-C5 heterocycle containing one or two N heteroatoms and optionally an additional O or S heteroatom,
  • or one of the foregoing R10 groups which is substituted with 1 to 3 halo, CN or N3; but optionally at least one R10 group is not H;
  • R23 is H, OH, F, Cl, Br, I, SCH3, SCH2CH3, SCH2C≡CH, SCH2CHCH2, SC3H7, OR16, NH2, NHR17 or R22; and
  • R24is O, 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, XIa.1, XIb.1, XIIa.1 or XIIIa.1
    Figure US20050159392A1-20050721-C00013
  • wherein R18, R20, R21, R24 have the meanings previously defined; R22A is R39 or R22 provided that R22 is not NH2; R23A is R39 or R23 provided that R23 is not NH2; R39 is NHR40, NHC(O)R36 or CR41N(R38)2 wherein R36 is C1-C19 alkyl, C1-C19 alkenyl, C3-C10 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; R38 is C1-C10 alkyl, or both R38 together are 1-morpholino, 1-piperidine or 1-pyrrolidine; R40 is C1-C1a 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 XIb.1, if R39 is present at R22A or R23A, both R39 groups on the same base will generally be the same. Exemplary R36 are phenyl, phenyl substituted with one of the foregoing R36 aryl substituents, —C10H15 (where C10H15 is 2-adamantoyl), —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-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza-8-aza derivatives of adenine, guanine, 2-aminopurine, 2,6-diaminopurine, 2-amino-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-6-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-methylcytosine; 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-diaminopurinyl, 2-aminopurinyl, 6-chloro-2-aminopurinyl and 6-thio-2-aminopurinyl, or a B′ is a 1-pyrimidinyl residue selected from cytosinyl, 5-halocytosinyl, and 5-(C1-C3-alkyl)cytosinyl.
  • Preferred B groups have the formula
    Figure US20050159392A1-20050721-C00014
  • wherein
  • R22 independently is halo, oxygen, NH2, X or H, but optionally at least one R22 is X;
  • X is —CH2)m(O)n(CH2)mN(R10)2 wherein m is 0-2, n is 0-1, and
  • R10 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 heterocycloalkyl,
  • 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—O—,
  • optionally both R10 are joined together with N to form a saturated or unsaturated C2-C5 heterocycle containing one or two N heteroatoms and optionally an additional O or S heteroatom,
  • or one of the foregoing R10 groups is substituted with 1 to 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(CH3)CH2— or —CH2CH2—. 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).
  • R1 is an in vivo hydrolyzable oxyester having the structure —OR35 or —OR6 wherein R35 is defined in column 64, line 49 of U.S. Pat. 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
    Figure US20050159392A1-20050721-C00015
  • wherein
  • n is 1 or 2;
  • R11 is R6 or H; preferably R6=C3-C9 alkyl; C3-C9 alkyl substituted independently with OH, halogen, O or N; C3-C6 aryl; C3-C6 aryl which is independently substituted with OH, halogen, O or N; or C3-C6 arylalkyl which is independently substituted with OH, halogen, O 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, O, N, COOR11 and halogen; C3-C6 aryl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, O, N, COOR11 and halogen; or C3-C9 aryl-alkyl which is unsubstituted or substituted by substituents independently selected from the group consisting of OH, O, N, COOR11 and halogen;
  • R13 independently is C(O)—OR11; amino; amide; guanidinyl; imidazolyl; indolyl; sulfoxide; phosphoryl; C1-C3 alkylamino; C1-C3 alkyldiamino; C1-C6 alkenylamino; hydroxy; thiol; C1-C3 alkoxy; C1-C3 alkthiol; (CH2),COOR11; C1-C6 alkyl which is unsubstituted or substituted with OH, halogen, SH, NH2, phenyl, hydroxyphenyl or C7-C10 alkoxyphenyl; C2-C6 alkenyl which is unsubstituted or substituted with OH, halogen, SH, NH2, phenyl, hydroxyphenyl or C7-C10 alkoxyphenyl; and C6-C12 aryl which is unsubstituted or substituted with OH, halogen, SH, NH2, phenyl, hydroxyphenyl or C7-C10 alkoxyphenyl; and
  • R14 is H or C1-C9 alkyl or C1-C9 alkyl independently substituted with OH, halogen, COOR11, O or N; C3-C6 aryl; C3-C6 aryl which is independently substituted with OH, halogen, COOR11, O or N; or C3-C6 arylalkyl which is independently substituted with OH, halogen, COOR11, O or N.
  • Preferably, R11 is C1-C6 alkyl, most preferably isopropyl, R13 is the side chain of a naturally occurring amino acid, n=1, R12 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 “O” or “N” substitution are defined in column 16, lines 42-58, of U.S. Pat. No. 5,798,340.
  • Typically, the amino acids are in the natural or l amino acids. Suitable specific examples are set forth in U.S. Pat. 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-C12. Examples are —CH3, —CH2CH3, —CH2CH2CH3, —CH(CH3)2, —CH2CH2CH2CH3), —CH2CH(CH3)2, —CH(CH3)CH2CH3, —C(CH3)3, —CH2CH2CH2CH2CH3, —CH(CH3)CH2CH2CH3, —CH(CH2CH3)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, —CH(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. Pat. 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. Pat. No. 5,798,340. These prodrugs have the antiviral and utilities already established for the parent drugs (see U.S. Pat. No. 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.
  • 6. EXAMPLES
  • The invention will be more fully understood by reference to the following examples:
  • Example 1a
  • Figure US20050159392A1-20050721-C00016
  • Adenine to PMEA Using Magnesium Isopropoxide
  • To a suspension of adenine (16.8 g, 0.124 mol) in DMF (41.9 ml) was added ethylene carbonate (12.1 g, 0.137 mol) and sodium hydroxide (0.100 g, 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 (2×). The wet solid was dried in vacuo at 65° C. to yield 20.0 g (90%) of 9-(2-hydroxyethyl)adenine as an off-white solid. Mp: 238-240° C.
    Figure US20050159392A1-20050721-C00017
  • 9-(2-Hydroxyethyl)adenine (HEA) (20.0 g, 0.112 mol) was suspended in DMF (125 ml) and heated to 80° C. Magnesium isopropoxide (11.2 g, 0.0784 mol) , or alternatively magnesium t-butoxide, was added to the mixture followed by diethyl p-toluenesulfonyloxymethylphosphonate (66.0 g, 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, bromotrimethylsilane (69.6 g, 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 (2×), and dried in vacuo at 56° C. to yield 20 g (65.4%) of 9-[2-(phosphonomethoxy)ethyl]adenine (PMEA) as a white solid. Mp: >200° C. dec. 1H NMR (D2O) δ 3.49 (t, 2H); 3.94 (t, 2H); 4.39 (t, 2H); 8.13 (s, 1H); 8.22 (s, 1H).
  • Example 1b
  • Figure US20050159392A1-20050721-C00018
  • Adenine to PMPA Using Magnesium t-Butoxide
  • To a suspension of adenine (40 g, 0.296 mol) in DMF (41.9 ml) was added (R)-propylene carbonate (34.5 g, 0.338 mol) and sodium hydroxide (0.480 g, 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 (4.7 g, 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 (2×). The wet solid was dried in vacuo at 80° C. to yield 42.6 g (75%) of (R)-9-[2-(hydroxy)propyl]adenine (HPA) as an off-white solid. Mp: 188-190° C.
    Figure US20050159392A1-20050721-C00019
  • (R)-9-[2-(hydroxy)propyl]adenine (HPA) (20.0 g, 0.104 mol) was suspended in DMF (44.5 ml) and heated to 65° C. Magnesium t-butoxide (14.2 g, 0.083 mol), or alternatively magnesium isopropoxide, was added to the mixture over one hour followed by diethyl p-toluenesulfonyloxymethylphosphonate (66.0 g, 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., bromotrimethylsilane (73.9 g, 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 ml). 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 18.9 g (63.5%) of crude(R)-9-[2-(phosphonomethoxy)propyl]adenine (PMPA) as an off-white solid.
  • The crude(R)-9-[2-(phosphonomethoxy)propyl]adenine 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 15.0 g (50.4%) of (R)-9-[2-(phosphonomethoxy)propyl]adenine (PMPA) as a white solid. Mp: 278-280° C.
  • Example 2 Preparation of GS-7171 (III)
  • Figure US20050159392A1-20050721-C00020
    Figure US20050159392A1-20050721-C00021
  • 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 HCl (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 (D2O) δ 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 (D2O) δ 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 38×38 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 (CDCl3) δ 1.1 (m, 12H), 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, 1H), 8.25 (d, 1H); 31P NMR (CDCl3) δ 21.0, 22.5 (decoupled).
  • Alternate Method for GS-7171(III)
  • Figure US20050159392A1-20050721-C00022
  • Monophenyl PMPA (II).
  • 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-dimethylformamide (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 (5 g, 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 (0.77 g, 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 sodium sulfate, and concentrated under reduced pressure to afford 2.4 g III as a tan foam.
  • Example 3 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 μm, 21×250 mm semi-preparative HPLC column with a Chiralpak AS, 20 μm, 21×50 mm guard column. Chiralpak® AS is a proprietary packing material manufactured by Diacel and sold in North America by Chiral Technologies, Inc. (U.S. Pat. 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 amylosetris[(S)-α-methylbenzyl carbamate] 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 min. duration) peak to elute from the column. When the GS-7339 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 vacuo 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.
  • 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))

    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., 20 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.; 1H NMR (CDCl3) δ 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 (CDCl3) δ 21.0 (decoupled).
  • 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 Phenomenex Luna ™ C18(2), 5 μm, 100 Å pore size,
    column: (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 A % Mobile Phase B
     0 100 0
     5 100 0
     7 70 30
    32 70 30
    40 0 100
    50 0 100
    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 nm
    Sample Solution: 20 mM sodium phosphate buffer, pH 6
    Retention Times: GS-7339, about 25 minutes
    GS-7340, about 27 minutes

    Diastereomer Separation by Crystallization
  • GS-7340 (IV).
  • A solution of GS-7171 (III) in acetonitrile was concentrated to an amber foam (14.9 g) 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 (31P NMR): 1H NMR (CDCl3) δ 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 (CDCl3) δ 19.5 (decoupled). X-ray crystal analysis of a single crystal selected from this product yielded the following data:
    Crystal Color, Habit colorless, column
    Crystal Dimensions 0.25 × 0.12 × 0.08 mm
    Crystal System orthorhombic
    Lattice Type Primitive
    Lattice Parameters a = 8.352(1) Å
    b = 15.574(2) Å
    c = 18.253(2) Å
    V = 2374.2(5) Å3
    Space Group P212121 (#19)
    Z value 4
    Dcalc 1.333 g/cm3
    F000 1008.00
    μ(MoKα) 1.60 cm−1
  • 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), fumaric 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.; [α]D 20 −41.7° (c 1.0, acetic acid).
  • Example 5
  • Figure US20050159392A1-20050721-C00023
  • 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., dichloromethane (1.6 kg) was added and the mixture was cooled to −20° C. A solution of (L)-α 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 15×13 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 diastereomeric 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 Crystallization
  • 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)-α 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-7342 (Table 5) as a tan powder: 1H NMR (CDCl3) δ 0.95 (t, 3H), 1.3 (m, 6H), 1.7, (m, 2H), 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, 1H); 31P NMR (CDCl3) δ 21 (decoupled).
  • Example 7 Diastereomer 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 (2,2′-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 25×11 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 GS-7340 than TDF after oral administration.
    TABLE 1
    In Vitro Activity and Stability
    Stability T ½ (min)
    HIV-1 MT-2
    Activity Cytotoxicity Human Cell
    IC50 μM CC50 μM Plasma 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 TDF as compared to that from GS-7340, both prodrugs and PMPA were radiolabeled and spiked into intact human whole blood at equimolar 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 10× and 30× 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 10× 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 FIG. 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)
    Recovered, GS
    Compound Matrix μg-eq PMPA % PMPAp, % PMPApp, % Met. X, % Met. Y, % 7340, %
    GS-7340 Plasma/ 43.0 1 2 13 84
    (60 μg-eq) FP
    PBMC 1.25 45 16 21 18
    RBC/FP 12.6 8 24 11 57
    Mono-
    PMPA PMPAp PMPApp POC GS-4331
    GS-4331 Plasma/ 48.1 11 89
    (TDF) FP
    (60 μg-eq) PBMC 0.133 50 25 18 7
    RBC/FP 10.5 93 7.0
    PMPA PMPAp PMPApp
    PMPA Plasma/ 55.7 100
    (60 μg-eq) FP
    PBMC 0.033 86 14
    RBC/FP 3.72 74 10 16
  • Met. X and Met Y (metabolites X and Y) are shown in Table 5. Lower case “p” 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 IC50 values seen in vitro.
  • In Table 3 below, IC50 values of tenofovir, TDF, GS-7340, several nucleosides and the protease inhibitor nelfinivir are listed. As shown, nelfinavir and GS-7340 are 2-3 orders of magnitude more potent than all other nucleotides or nucleosides.
    TABLE 3
    In Vitro Anti-HIV-1 Activities of Antiretroviral Compounds
    Compound IC50 (μM)
    Adefovir (PMEA)  13.4 ± 4.21
    Tenofovir (PMPA)  6.3 ± 3.31
    AZT  0.17 ± 0.081
    3TC  1.8 ± 0.251
    d4T    8 ± 2.51
    Nelfinavir 0.006 ± 0.0021
    TDF 0.05
    GS 7340 0.005

    1A. S. Mulato and J. M. Cherrington, Antiviral Research 36, 91 (1997)
  • Additional studies of the in vitro cell culture anti-HIV-1 activity and CC50 of separated diastereomers of this invention were conducted and the results tabulated below.
    TABLE 4
    Effect of Diastereomer
    Diastereomer Fold A/B CC50
    Compound residue IC50 (μM) change activity (μM)
    PMPA 5  1× 6000
    Ala-methylester Mixture 1:1 0.025 200× 20× 80
    GS-6957a A 0.0075 670×
    GS-6957b 0.15  33×
    Phe-methylester Mixture 1:1 0.03 170× 10× 60
    GS-7003a A 0.01 500×
    GS-7003b B 0.1    50××
    Gly-ethylester Mixture 1:1 0.5  10× 20×
    GS-7119 A 0.05 100× >100
    GS-7335 B 1.0  5×
    Ala-isopropyl Mixture 1:1 0.01 500× 12×
    GS-7340 A 0.005 1,000×   40
    GS-7339 B 0.06  83× >100
    ABA-ethyl Mixture 1:1 0.008 625× 7.5×  >100
    GS-7342 A 0.004 1,250×  
    GS-7341 B 0.03 170×
    Ala-ethyl Mixture 1:1 0.02 250× 10× 60
    GS-7114 A 0.005 1,000×  
    GS-7115 B 0.05   100××
  • Assay reference: Arimilli, M N, et al., (1997) Synthesis, in vitro biological evaluation and oral bioavailability of 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) prodrugs. Antiviral Chemistry and Chemotherapy 8(6):557-564.
  • “Phe-methylester” 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 stereochemistry that of GS-7339.
  • The in vitro metabolism and stability of separated diastereomers were determined in PLCE, MT-2 extract and human plasma. A biological sample listed below, 80 μL, 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 μL, was added to the biological sample and mixed. The reaction mixture, 20 μL, was immediately sampled and mixed with 60 μL 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 μL, was sampled and mixed with 60 μL of methanol containing the internal standard. The mixture thus obtained was centrifuiged 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 dl-dithiothreitol. pH 7.4 at 37° C.
    • 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 × 250 mm, 5μ
    (MAC-MOD Analytical, Inc. Chadds Ford, PA)
    Detection: UV at 260 nm
    Flow Rate: 1.0 mL/min
    Run Time: 30 min
    Injection Volume: 20 μL
    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
    25 min 100% Mobile Phase B
    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.
    HIV MT-2 extract Human
    IC50 PLCE hydrolysis hydrolysis rate Plasma
    No. PMPA monoamidate structure (□M) rate and product and product Stability (HP)
    1
    Figure US20050159392A1-20050721-C00024
    0.005 t1/2 = 2.9 min Met. X & PMPA t1/2 = 2.9 min Met. X & PMPA t1/2 = 148 min Met. Y
    2
    Figure US20050159392A1-20050721-C00025
    0.05 t1/2 = 8.0 min Met. X & PMPA t1/2 = 150.6 min Met. X & PMPA t1/2 = 495 min Met. Y
    3
    Figure US20050159392A1-20050721-C00026
    0.005 t1/2 = 3.3 min Met. X & PMPA t1/2 = 28.3 min Met. X & PMPA t1/2 = 90.0 min Met. Y
    4
    Figure US20050159392A1-20050721-C00027
    0.06 t1/2 = 10.1 min Met. X & PMPA t1/2 > 1000 min t1/2 = 231 min Met. Y
    5
    Figure US20050159392A1-20050721-C00028
    0.004 t1/2 = 3.9 min Met. X & PMPA t1/2 = 49.2 min Met. X & PMPA t1/2 = 103 min Met. Y
    6
    Figure US20050159392A1-20050721-C00029
    0.03 t1/2 = 11.3 min Met. X & PMPA t1/2 > 1000 min t1/2 = 257 min Met. Y
    7
    Figure US20050159392A1-20050721-C00030
    0.05 t1/2 < 0.14 min MonoPOC PMPA t1/2 = 70.7 min MonoPOC PMPA t1/2 = 0.41 min MonoPOC PMPA
    Figure US20050159392A1-20050721-C00031
    Figure US20050159392A1-20050721-C00032
    Figure US20050159392A1-20050721-C00033
  • 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:
    Amidate
    Amino AA
    GSI acid 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 □- Ethyl Isomer A 1509-191-12
    Aminobutyric
    Acid
    GS7341 □- Ethyl Isomer B 1509-191-7
    Aminobutyric
    Acid
  • Dose Administration and Sample Collection
  • The in-life 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 Health publication 86-23) and was approved by an 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 immediately for 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×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, Antimicrob. 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 N1, N6-ethenoadenine derivative (L. Naesens, J. Balzarini, and E. De Clercq, Clin. Chem. 38, 480 (1992). Briefly, plasma (100 μl) was mixed with 200 μl acetonitrile to precipitate protein. Samples were then evaporated to dryness under reduced pressure at room temperature. Dried samples were reconstituted in 200 μl 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 μl 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 μL of 1M rhamnose and 30 μL of 0.1M sodium periodate and incubated at 37° C. for 5 min. Following incubation, 40 μL of 4M methylamine and 20 μL 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-free plasma 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, Calif.). The column was an Inertsil ODS-2 column (4.6×150 mm). The mobile phases used were: A, 5% acetonitrile in 25 mM potassium phosphate buffer with 5 mM tetrabutyl ammonium bromide (TBABr), pH 6.0; B, 60% acetonitrile in 25 mM potassium phosphate buffer with 5 mM TBABr, pH 6.0. 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 nm and emission at 420 nm, and the injection volume was 10 μl. Data was acquired and stored by a laboratory data acquisition system (PeakPro, Beckman, Allendale, N.J.).
  • 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 FIGS. 2 and 3. FIG. 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.
  • The bar graph in FIG. 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.
  • 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 PBMC and Plasma from Oral Prodrugs of PMPA in Dogs
    PMPA AUC PMPA AUC PBMC/Plasma
    in Plasma in PBMC Prodrug Exposure
    GS# Moiety 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 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 administration to beagle dogs. Two male animals were dosed orally with 14C=GS-7340 (8.85 mg-equiv. of PMPA/kg, 33.2 μCi/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 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 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.
  • 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
    Conc.
    GS-4331 Ratio of
    Conc. GS-7340 GS 7340
    % (ug-eq/ % Conc. to
    Tissue/Fluid Dose g) Dose (ug-eq/g) GS-4331
    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 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. n.a.
    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 n.a. 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 81.10 68.96

    Calculated using typical recovery of 15 × 106 cells total, and mean PBMC volume of 0.2 picoliters/cell

    n.s. = no sample,

    n.a. = not applicable,

    n.d. = not determined.

Claims (26)

1. A method of inhibiting replication of an HIV virion, comprising contacting a cell comprising an HIV virion with a nucleotide analog compound according structure (Ia):
Figure US20050159392A1-20050721-C00034
and/or a salt and/or solvate thereof, wherein:
R6a is selected from hydrogen and phenyl;
R6b is selected from hydrogen, methyl, ethyl and isopropyl; and
R7 is selected from methyl, ethyl and benzyl, and which comprises at least about 60% by weight a diastereomer according to structure (Ib):
Figure US20050159392A1-20050721-C00035
2. The method of claim 1 in which the nucleotide analog compound, salt and/or solvate comprises at least about 80% by weight the diastereomer of structure (Ib).
3. The method of claim 1 in which the nucleotide analog compound, salt and/or solvate comprises at least about 95% by weight the diastereomer of structure (Ib).
4. The method of claim 1 in which the nucleotide analog compound, salt and/or solvate is substantially free of the diastereomer of structure (Ic):
Figure US20050159392A1-20050721-C00036
5. The method of claim 1 in which in which the nucleotide analog compound, salt and/or solvate is selected from Metabolite Y, Metabolite X, GS-6957a, GS-7003a, GS-7119, GS-7114, GS-7340, and GS-7342.
6. The method of claim 1 in which the nucleotide analog compound is in the form of a salt.
7. The method of claim 6 in which the salt is a 1:1 fumarate salt.
8. A method of treating an HIV infection, comprising administering to a subject having an HIV infection a therapeutically effective amount of a nucleotide analog compound according structure (Ia):
Figure US20050159392A1-20050721-C00037
and/or a salt and/or solvate thereof, wherein:
R6a is selected from hydrogen and phenyl;
R6b is selected from hydrogen, methyl, ethyl and isopropyl; and
R7 is selected from methyl, ethyl and benzyl, and which comprises at least about 60% by weight a diastereomer according to structure (Ib):
Figure US20050159392A1-20050721-C00038
9. The method of claim 8 in which the nucleotide analog compound, salt and/or solvate comprises at least about 80% by weight the diastereomer of structure (Ib).
10. The method of claim 8 in which the nucleotide analog compound, salt and/or solvate comprises at least about 95% by weight the diastereomer of structure (Ib).
11. The method of claim 8 in which the nucleotide analog compound, salt and/or solvate is substantially free of the diastereomer of structure (Ic):
Figure US20050159392A1-20050721-C00039
12. The method of claim 8 in which in which the nucleotide analog compound, salt and/or solvate is selected from Metabolite Y, Metabolite X, GS-6957a, GS-7003a, GS-7119, GS-7114, GS-7340, and GS-7342.
13. The method of claim 8 in which the nucleotide analog compound is in the form of a salt.
14. The method of claim 13 in which the salt is a 1:1 fumarate salt.
15. The method of claim 8 in which the nucleotide analog compound is administered in the form of a pharmaceutical composition comprising the nucleotide analog compound and a pharmaceutically acceptable excipient.
16. The method of any one of claim 8 in which the nucleotide analog compound is administered orally.
17. A method of preventing an HIV infection in a subject, comprising administering to the subject a prophylactically effective amount of a nucleotide analog compound according structure (Ia):
Figure US20050159392A1-20050721-C00040
and/or a salt and/or solvate thereof, wherein:
R6a is selected from hydrogen and phenyl;
R6b is selected from hydrogen, methyl, ethyl and isopropyl; and
R7 is selected from methyl, ethyl and benzyl, and which comprises at least about 60% by weight a diastereomer according to structure (Ib):
Figure US20050159392A1-20050721-C00041
18. The method of claim 17 in which the nucleotide analog compound, salt and/or solvate comprises at least about 80% by weight the diastereomer of structure (Ib).
19. The method of claim 17 in which the nucleotide analog compound, salt and/or solvate comprises at least about 95% by weight the diastereomer of structure (Ib).
20. The method of claim 17 in which the nucleotide analog compound, salt and/or solvate is substantially free of the diastereomer of structure (Ic):
Figure US20050159392A1-20050721-C00042
21. The method of claim 17 in which in which the nucleotide analog compound, salt and/or solvate is selected from Metabolite Y, Metabolite X, GS-6957a, GS-7003a, GS-7119, GS-7114, GS-7340, and GS-7342.
22. The method of claim 17 in which the nucleotide analog compound is in the form of a salt.
23. The method of claim 22 in which the salt is a 1:1 fumarate salt.
24. The method of claim 17 in which the nucleotide analog compound is administered in the form of a pharmaceutical composition comprising the nucleotide analog compound and a pharmaceutically acceptable excipient.
25. The method of claim 17 in which the nucleotide analog compound is administered orally.
26. A method of selectively delivering (R)-9-[2-(phosphonomethoxy)propyl]adenine to lymphoid tissue and/or non-lymphoid tissue enriched in lymphoid cells, comprising administering to a subject comprising such lymphoid and/or non-lymphoid tissue a prodrug compound according to structural formula (Ia):
Figure US20050159392A1-20050721-C00043
and/or a salt and/or solvate thereof, wherein:
R6a is selected from hydrogen and phenyl;
R6b is selected from hydrogen, methyl, ethyl and isopropyl; and
R7 is selected from methyl, ethyl and benzyl, and which comprises at least about 60% by weight a diastereomer according to structure (Ib):
Figure US20050159392A1-20050721-C00044
US11/031,228 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues Abandoned US20050159392A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/031,228 US20050159392A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US22002100P 2000-07-21 2000-07-21
US09/909,560 US20020119443A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/354,207 US20030219727A1 (en) 2000-07-21 2003-01-28 Prodrugs of phosphonate nucleotide analogues
US10/798,692 US7390791B2 (en) 2000-07-21 2004-03-11 Prodrugs of phosphonate nucleotide analogues
US11/031,228 US20050159392A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/798,692 Continuation US7390791B2 (en) 2000-07-21 2004-03-11 Prodrugs of phosphonate nucleotide analogues

Publications (1)

Publication Number Publication Date
US20050159392A1 true US20050159392A1 (en) 2005-07-21

Family

ID=22821718

Family Applications (10)

Application Number Title Priority Date Filing Date
US09/909,560 Abandoned US20020119443A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/333,107 Abandoned US20040018150A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/354,207 Abandoned US20030219727A1 (en) 2000-07-21 2003-01-28 Prodrugs of phosphonate nucleotide analogues
US10/785,497 Abandoned US20060024659A1 (en) 2000-07-21 2004-02-24 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/798,692 Active 2025-04-17 US7390791B2 (en) 2000-07-21 2004-03-11 Prodrugs of phosphonate nucleotide analogues
US11/031,252 Abandoned US20050124585A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US11/031,250 Abandoned US20050124583A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US11/031,251 Abandoned US20050124584A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US11/031,228 Abandoned US20050159392A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US12/110,829 Expired - Fee Related US7803788B2 (en) 2000-07-21 2008-04-28 Prodrugs of phosphonate nucoleotide analogues

Family Applications Before (8)

Application Number Title Priority Date Filing Date
US09/909,560 Abandoned US20020119443A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/333,107 Abandoned US20040018150A1 (en) 2000-07-21 2001-07-20 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/354,207 Abandoned US20030219727A1 (en) 2000-07-21 2003-01-28 Prodrugs of phosphonate nucleotide analogues
US10/785,497 Abandoned US20060024659A1 (en) 2000-07-21 2004-02-24 Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US10/798,692 Active 2025-04-17 US7390791B2 (en) 2000-07-21 2004-03-11 Prodrugs of phosphonate nucleotide analogues
US11/031,252 Abandoned US20050124585A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US11/031,250 Abandoned US20050124583A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues
US11/031,251 Abandoned US20050124584A1 (en) 2000-07-21 2005-01-06 Prodrugs of phosphonate nucleotide analogues

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/110,829 Expired - Fee Related US7803788B2 (en) 2000-07-21 2008-04-28 Prodrugs of phosphonate nucoleotide analogues

Country Status (37)

Country Link
US (10) US20020119443A1 (en)
EP (3) EP1301519B2 (en)
JP (4) JP4651264B2 (en)
KR (2) KR100749160B1 (en)
CN (2) CN1291994C (en)
AP (1) AP1466A (en)
AU (3) AU2001282941C1 (en)
BE (1) BE2016C018I2 (en)
BG (1) BG66037B1 (en)
BR (1) BRPI0112646B8 (en)
CA (3) CA2725819C (en)
CY (2) CY2016008I2 (en)
CZ (2) CZ304886B6 (en)
DK (2) DK2682397T3 (en)
EA (1) EA004926B1 (en)
EE (1) EE05366B1 (en)
ES (2) ES2536972T5 (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) NZ535408A (en)
OA (1) OA12393A (en)
PL (1) PL213214B1 (en)
PT (2) PT2682397T (en)
SI (2) SI1301519T1 (en)
TR (1) TR200300055T2 (en)
UA (1) UA75889C2 (en)
WO (1) WO2002008241A2 (en)
ZA (1) ZA200210271B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10479810B2 (en) 2016-06-05 2019-11-19 Shanghai Begreat Pharmatech Crystal form of tenofovir alafenamide salt, preparation method and use thereof

Families Citing this family (215)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1301519B2 (en) * 2000-07-21 2021-11-10 Gilead Sciences, Inc. Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
EA200400690A1 (en) * 2001-11-14 2005-06-30 Байокрист Фармасьютикалз, Инк. NUCLEOSIDES, THEIR PREPARATIONS AND THEIR APPLICATION AS VIRAL RNA POLYMERASE INHIBITORS
US7388002B2 (en) * 2001-11-14 2008-06-17 Biocryst Pharmaceuticals, Inc. Nucleosides, preparation thereof and use as inhibitors of RNA viral polymerases
EP1501841A2 (en) * 2002-04-26 2005-02-02 Gilead Sciences, Inc. Non nucleoside reverse transcriptase inhibitors
US20050239054A1 (en) * 2002-04-26 2005-10-27 Arimilli Murty N Method and compositions for identifying anti-HIV therapeutic compounds
CA2485702C (en) 2002-05-13 2011-07-19 Metabasis Therapeutics, Inc. Novel phosphonic acid based prodrugs of pmea and its analogues
EP1923063A3 (en) * 2003-01-14 2009-04-08 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US7427636B2 (en) 2003-04-25 2008-09-23 Gilead Sciences, Inc. Inosine monophosphate dehydrogenase inhibitory phosphonate compounds
US7300924B2 (en) 2003-04-25 2007-11-27 Gilead Sciences, Inc. Anti-infective phosphonate analogs
US20050261237A1 (en) * 2003-04-25 2005-11-24 Boojamra Constantine G Nucleoside phosphonate analogs
US7432261B2 (en) 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
US7407965B2 (en) * 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
WO2005002626A2 (en) * 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
US20090247488A1 (en) * 2003-04-25 2009-10-01 Carina Cannizzaro Anti-inflammatory phosphonate compounds
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
AU2004233897A1 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Kinase inhibitor phosphonate conjugates
US7470724B2 (en) * 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
WO2004096286A2 (en) * 2003-04-25 2004-11-11 Gilead Sciences, Inc. Antiviral phosphonate analogs
CN101410120A (en) * 2003-04-25 2009-04-15 吉里德科学公司 Anti-inflammatory phosphonate compounds
WO2005044279A1 (en) 2003-10-24 2005-05-19 Gilead Sciences, Inc. Purine nucleoside phosphonate conjugates
WO2005044308A1 (en) * 2003-10-24 2005-05-19 Gilead Sciences, Inc. Phosphonate analogs of antimetabolites
JP2007508845A (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
US20050153990A1 (en) * 2003-12-22 2005-07-14 Watkins William J. Phosphonate substituted kinase inhibitors
BRPI0418031A (en) * 2003-12-22 2007-04-17 Gilead Sciences Inc phosphonate-substituted kinase inhibitors
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
DE602004028763D1 (en) * 2003-12-30 2010-09-30 Gilead Sciences Inc TE FOR THE TREATMENT OF 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
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
US8416242B1 (en) 2004-05-14 2013-04-09 Nvidia Corporation Method and system for interpolating level-of-detail in graphics processors
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
US8411105B1 (en) 2004-05-14 2013-04-02 Nvidia Corporation Method and system for computing pixel parameters
JP5142716B2 (en) 2004-06-08 2013-02-13 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 Lewis acid mediated synthesis of cyclic esters
DE602005027466D1 (en) * 2004-07-27 2011-05-26 Gilead Sciences Inc NUCLEOSIDE PHOSPHONATE CONJUGATES AS ANTI HIV MEDIUM
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
JP2008535862A (en) 2005-04-08 2008-09-04 キメリクス,インコーポレイテッド 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
TWI471145B (en) 2005-06-13 2015-02-01 Bristol Myers Squibb & Gilead Sciences Llc Unitary pharmaceutical dosage form
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
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
WO2007136650A2 (en) 2006-05-16 2007-11-29 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
AU2008302676B2 (en) * 2007-06-26 2013-02-28 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
AU2009206673B2 (en) * 2008-01-25 2015-04-23 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
EP2937356A1 (en) 2008-04-25 2015-10-28 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
JP2011526893A (en) * 2008-07-02 2011-10-20 イデニク プハルマセウティカルス,インコーポレイテッド Compounds and pharmaceutical compositions for the treatment of viral infections
AP3347A (en) 2008-07-08 2015-07-31 Gilead Sciences Inc Salts of HIV inhibitor compounds
SG172361A1 (en) 2008-12-23 2011-07-28 Pharmasset Inc Nucleoside analogs
EP2376088B1 (en) 2008-12-23 2017-02-22 Gilead Pharmasset LLC 6-O-Substituted-2-amino-purine nucleoside phosphoramidates
US8716263B2 (en) * 2008-12-23 2014-05-06 Gilead Pharmasset Llc Synthesis of purine nucleosides
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
TWI598358B (en) 2009-05-20 2017-09-11 基利法瑪席特有限責任公司 Nucleoside phosphoramidates
WO2011011519A1 (en) 2009-07-21 2011-01-27 Chimerix, Inc. Compounds, compositions and methods for treating ocular conditions
KR101848099B1 (en) 2009-09-21 2018-04-11 길리애드 사이언시즈, 인코포레이티드 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
PL2552930T3 (en) 2010-03-31 2016-02-29 Gilead Pharmasset Llc Crystalline (s)-isopropyl 2-(((s)-(((2r,3r,4r,5r)-5-(2,4-dioxo-3,4-dihydropyrimidin-1-(2h)-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate
PL3290428T3 (en) 2010-03-31 2022-02-07 Gilead Pharmasset Llc Tablet comprising crystalline (s)-isopropyl 2-(((s)-(((2r,3r,4r,5r)-5-(2,4-dioxo-3,4-dihydropyrimidin-1 (2h)-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate
AR094621A1 (en) 2010-04-01 2015-08-19 Idenix Pharmaceuticals Inc PHARMACEUTICAL COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF VIRAL INFECTIONS
WO2011139709A2 (en) 2010-04-26 2011-11-10 Chimerix, Inc. Methods of treating retroviral infections and related dosage regimes
WO2012012465A1 (en) 2010-07-19 2012-01-26 Clarke, Michael, O'neil Hanrahan Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
BR122020020745B8 (en) 2010-07-22 2023-10-31 Gilead Sciences Inc Antiviral compound for the treatment of paramyxoviridae infections and pharmaceutical composition comprising it
JP6069215B2 (en) 2010-11-30 2017-02-01 ギリアド ファーマセット エルエルシー Compound
WO2012079035A1 (en) 2010-12-10 2012-06-14 Sigmapharm Laboratories, Llc Highly stable compositions of orally active nucleotide analogues or orally active nucleotide analogue prodrugs
ZA201103820B (en) 2010-12-13 2012-01-25 Laurus Labs Private Ltd Process for the preparation of tenofovir
CN103842369A (en) 2011-03-31 2014-06-04 埃迪尼克斯医药公司 Compounds and pharmaceutical compositions for the treatment of viral infections
WO2012154698A2 (en) * 2011-05-06 2012-11-15 Mckenna Charles E Method to improve antiviral activity of nucleotide analogue drugs
JP5951757B2 (en) * 2011-05-19 2016-07-13 ギリアード サイエンシーズ, インコーポレイテッド Processes and intermediates for preparing anti-HIV agents
AU2014271320B2 (en) * 2011-08-16 2017-02-23 Gilead Sciences, Inc. Tenofovir alafenamide hemifumarate
MX336627B (en) * 2011-08-16 2016-01-26 Gilead Sciences Inc Tenofovir alafenamide hemifumarate.
PE20141056A1 (en) 2011-09-16 2014-09-05 Gilead Pharmasset Llc METHODS FOR THE TREATMENT OF HCV
AR088109A1 (en) 2011-10-07 2014-05-07 Gilead Sciences Inc METHOD FOR PREPARING ANTIVIRAL NUCLEOTIDAL ANALOGS
AU2014215976B2 (en) * 2011-10-07 2016-06-30 Gilead Sciences, Inc. Methods for preparing anti-viral nucleotide analogs
AU2016228317B2 (en) * 2011-10-07 2018-07-19 Gilead Sciences, Inc. 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
ES2734495T3 (en) 2011-12-22 2019-12-10 Geron Corp Guanine analogs such as telomerase substrates and telomere length affecters
AU2012327170A1 (en) 2012-02-03 2013-08-22 Gilead Sciences, Inc. Therapeutic compounds
US20150105350A1 (en) * 2012-02-03 2015-04-16 Gilead Sciences, Inc. Combination therapy comprising tenofovir alafenamide hemifumarate and cobicistat for use in the treatment of viral infections
CN107312039B (en) 2012-08-30 2019-06-25 江苏豪森药业集团有限公司 A kind of preparation method of tenofovir prodrug
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)
WO2014068265A1 (en) * 2012-10-29 2014-05-08 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
JP6297582B2 (en) * 2012-11-16 2018-03-20 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Purine inhibitors of human phosphatidylinositol 3-kinase delta
CN103848869B (en) * 2012-12-04 2016-12-21 上海医药工业研究院 The method preparing tenofovir
CN103848868B (en) * 2012-12-04 2017-04-12 蚌埠丰原涂山制药有限公司 method for preparing tenofovir
MD4595B1 (en) 2013-01-31 2018-10-31 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
WO2014195724A1 (en) 2013-06-07 2014-12-11 Cipla Limited An efficient process for separation of diastereomers of 9-[(r)-2-[[(r,s)-[[(s)-1-(isopropoxycarbonyl)ethyl]amino]-phenoxyphosphinyl] methoxy]propyl]adenine
NZ716840A (en) 2013-08-27 2017-06-30 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
WO2015107451A2 (en) 2014-01-14 2015-07-23 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
US9463194B2 (en) 2014-02-05 2016-10-11 Gilead Sciences, Inc. Methods of treating patients co-infected 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
CN105518011B (en) * 2014-04-21 2018-07-27 四川海思科制药有限公司 The preparation method of phosphoramidic acid ester derivative Preparation Method And Their Intermediate and intermediate
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
EP3164136A4 (en) 2014-07-02 2018-04-04 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
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
KR101703258B1 (en) 2014-12-30 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
TWI698444B (en) 2014-10-29 2020-07-11 美商基利科學股份有限公司 Methods for the preparation of ribosides
CN108148094A (en) * 2014-11-12 2018-06-12 四川海思科制药有限公司 A kind of tenofovir Chinese mugwort draws phenol amine fumarate crystal form C and its preparation method and application
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
BR112017014085A2 (en) 2015-01-03 2018-01-09 Mylan Laboratories Ltd Processes for the preparation of amorphous tenofovir alafenamide hemifumarate and a premixture thereof
US20180148774A1 (en) * 2015-05-16 2018-05-31 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
MA46677A (en) * 2015-06-17 2019-09-11 Gilead Sciences Inc CO-CRYSTALS, SALTS AND SOLID FORMS OF TENOFOVIR ALAFENAMIDE
SI4070788T1 (en) 2015-06-30 2023-06-30 Gilead Sciences, Inc. Pharmaceutical formulations
CA2994720C (en) 2015-08-10 2020-11-03 Merck Sharp & Dohme Corp. Antiviral beta-amino acid ester phosphodiamide compounds
TWI616452B (en) * 2015-08-26 2018-03-01 Preparation method of nucleoside analog and intermediate thereof
TWI620754B (en) * 2015-08-26 2018-04-11 Method for preparing amino phosphate derivative and preparation method 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
ES2909419T3 (en) 2015-09-16 2022-05-06 Gilead Sciences Inc Methods for treating coronaviridae infections
PL3346995T3 (en) 2015-11-09 2020-03-31 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
WO2017134089A1 (en) 2016-02-02 2017-08-10 Sandoz Ag Crystalline forms of tenofovir alafenamide monofumarate
WO2017133517A1 (en) * 2016-02-03 2017-08-10 四川海思科制药有限公司 Phosphamide derivative, method for manufacturing the same, and uses thereof
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
CN107698621A (en) * 2016-06-20 2018-02-16 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
US10449208B2 (en) 2016-08-25 2019-10-22 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
US10736908B2 (en) 2016-10-26 2020-08-11 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
CA3046029A1 (en) * 2016-12-22 2018-06-28 Merck Sharp & Dohme Corp. Antiviral benzyl-amine phosphodiamide compounds
JP6938637B2 (en) 2016-12-22 2021-09-22 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Tenofovir antiviral aliphatic ester prodrug
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
CA3056072C (en) 2017-03-14 2022-08-23 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
EP3600332B1 (en) 2017-03-20 2023-12-13 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
CA3059777C (en) 2017-05-01 2023-02-21 Gilead Sciences, Inc. Crystalline forms 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-dihydroxytetrahydrofuran-2-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
CN111587107B (en) 2017-06-30 2023-03-31 希普拉有限公司 Pharmaceutical composition
US10675296B2 (en) 2017-07-11 2020-06-09 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
EP3661937B1 (en) 2017-08-01 2021-07-28 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
WO2019084020A1 (en) 2017-10-24 2019-05-02 Gilead Sciences, Inc. Methods of treating patients co-infected with a virus and tuberculosis
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
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
US20200407382A1 (en) 2017-12-30 2020-12-31 Cipla Limited Polymorphic forms of (9-[(r)-2-[[(s)-[[(s)-1-(isopropoxycarbonyl)ethyl]amino]phenoxy phosphinyl]methoxy]propyl] adenine and pharmaceutically acceptable salts thereof
MX2020007393A (en) * 2018-01-10 2020-11-24 Nucorion Pharmaceuticals Inc Phosphor(n)amidatacetal and phosph(on)atalcetal compounds.
EP3737359A4 (en) * 2018-01-12 2021-11-03 Board of Regents of the University of Nebraska Antiviral prodrugs and formulations thereof
PL3752495T3 (en) 2018-02-15 2024-01-15 Gilead Sciences, Inc. Pyridine derivatives and their use for treating hiv infection
CA3175557A1 (en) 2018-02-16 2019-08-22 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
WO2019199756A1 (en) 2018-04-09 2019-10-17 Board Of Regents Of The University Of Nebraska Antiviral prodrugs and formulations thereof
EP3823621A1 (en) 2018-07-16 2021-05-26 Gilead Sciences, Inc. Capsid inhibitors for the treatment of hiv
WO2020018399A1 (en) 2018-07-19 2020-01-23 Merck Sharp & Dohme Corp. Phosphinic amide prodrugs of tenofovir
KR20210060573A (en) 2018-09-19 2021-05-26 길리애드 사이언시즈, 인코포레이티드 Integrase inhibitor for HIV prevention
WO2021011891A1 (en) 2019-07-18 2021-01-21 Gilead Sciences, Inc. Long-acting formulations of tenofovir alafenamide
US20220265689A1 (en) 2019-07-19 2022-08-25 THE UNITED STATES OF AMERICA , as represented by the Secretary, Department of Health and Human Hiv pre-exposure prophylaxis
US20220296619A1 (en) 2019-08-19 2022-09-22 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
EP4017499A4 (en) * 2019-08-22 2024-01-10 Univ Emory Nucleoside prodrugs and uses related thereto
KR20230013013A (en) * 2019-09-20 2023-01-26 애보트 라피드 다이어그노스틱스 인터내셔널 언리미티드 컴퍼니 Antibodies directed against tenofovir and its derivatives
CN114727999A (en) 2019-11-26 2022-07-08 吉利德科学公司 Capsid inhibitors for the prevention of HIV
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
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
CA3169348A1 (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
JP2023523415A (en) 2020-04-21 2023-06-05 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 Nucleotide prodrug compounds
KR20230018473A (en) 2020-05-29 2023-02-07 길리애드 사이언시즈, 인코포레이티드 How to treat remdesivir
EP4172160A2 (en) 2020-06-24 2023-05-03 Gilead Sciences, Inc. 1'-cyano nucleoside analogs and uses thereof
CN115996925A (en) 2020-06-25 2023-04-21 吉利德科学公司 Capsid inhibitors for the treatment of HIV
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
PE20231983A1 (en) 2020-08-27 2023-12-12 Gilead Sciences Inc COMPOUNDS AND METHODS FOR THE 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
WO2022103758A1 (en) 2020-11-11 2022-05-19 Gilead Sciences, Inc. METHODS OF IDENTIFYING HIV PATIENTS SENSITIVE TO THERAPY WITH gp120 CD4 BINDING SITE-DIRECTED ANTIBODIES
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
TW202342448A (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
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
WO2023102239A1 (en) 2021-12-03 2023-06-08 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
TW202400165A (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
TW202402280A (en) 2022-07-01 2024-01-16 美商基利科學股份有限公司 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

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
CS263952B1 (en) 1985-04-25 1989-05-12 Holy Antonin Remedy with antiviral effect
CS263951B1 (en) 1985-04-25 1989-05-12 Antonin Holy 9-(phosponylmethoxyalkyl)adenines and method of their preparation
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
ATE124050T1 (en) * 1990-04-20 1995-07-15 Acad Of Science Czech Republic CHIRAL 2-(PHOSPHONOMETHOXY)PROPYL-GUANINE AS ANTIVIRAL AGENTS.
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
CZ285420B6 (en) 1990-04-24 1999-08-11 Ústav Organické Chemie A Biochemie Avčr N-(3-fluoro-2-phosphonylmethoxypropyl) derivatives of purine and pyrimidine heterocyclic bases, processes of their preparation and use
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
JP3116079B2 (en) 1990-08-10 2000-12-11 インスティチュート オブ オーガニック ケミストリ アンド バイオケミストリ アカデミー オブ サイエンス オブ ザ チェコ リパブリック Novel nucleotide production method
DE10399025I2 (en) 1990-09-14 2007-11-08 Acad Of Science Czech Republic Active substance precursors 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
US5656745A (en) * 1993-09-17 1997-08-12 Gilead Sciences, Inc. Nucleotide analogs
EP0719274A1 (en) * 1993-09-17 1996-07-03 Gilead Sciences, Inc. Method for dosing therapeutic compounds
US5798340A (en) * 1993-09-17 1998-08-25 Gilead Sciences, Inc. Nucleotide analogs
DE69435319D1 (en) 1993-09-17 2010-12-16 Gilead Sciences Inc nucleotide analogs
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
PT828749E (en) * 1995-05-26 2003-11-28 Genta Inc COMPOSITIONS AND METHODS FOR SYNTHESIS OF ORGANOPHOSPHORUS DERIVATIVES
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
JP4033494B2 (en) * 1996-07-26 2008-01-16 ギリヤド サイエンシーズ, インコーポレイテッド Nucleotide analogs
US5739314A (en) 1997-04-25 1998-04-14 Hybridon, Inc. Method for synthesizing 2'-O-substituted pyrimidine nucleosides
EP0996430B1 (en) 1997-07-25 2002-11-27 Gilead Sciences, Inc. Nucleotide analog compositions
US5935946A (en) 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
NZ501287A (en) 1997-07-25 2001-09-28 Gilead Sciences Inc Nucleotide analog composition and synthesis method comprising 9-(2-hydroxypropyl)adenine and lithium alkoxide in a cystalline form.
WO1999030727A1 (en) * 1997-12-17 1999-06-24 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
EP1045897B1 (en) * 1998-01-23 2002-01-30 Newbiotics Inc. Enzyme catalyzed therapeutic agents
US6169078B1 (en) * 1998-05-12 2001-01-02 University Of Florida Materials and methods for the intracellular delivery of substances
AU755564B2 (en) * 1998-06-20 2002-12-12 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
US7115590B1 (en) 1999-02-12 2006-10-03 University College Cardiff Consultants Limited 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
EP1301519B2 (en) * 2000-07-21 2021-11-10 Gilead Sciences, Inc. Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
AU2002220979A1 (en) * 2000-10-13 2002-04-22 Xigen Sa Intracellular delivery of biological effectors by 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

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10479810B2 (en) 2016-06-05 2019-11-19 Shanghai Begreat Pharmatech Crystal form of tenofovir alafenamide salt, preparation method and use thereof

Also Published As

Publication number Publication date
US20050124585A1 (en) 2005-06-09
NO336718B1 (en) 2015-10-26
LTC1301519I2 (en) 2023-02-27
FR16C0013I2 (en) 2016-09-09
ZA200210271B (en) 2003-12-31
DK1301519T3 (en) 2015-05-26
NO20120466L (en) 2003-03-20
US7803788B2 (en) 2010-09-28
JP2011140506A (en) 2011-07-21
NO2016006I1 (en) 2016-04-19
EE05366B1 (en) 2010-12-15
US20020119443A1 (en) 2002-08-29
HK1054238A1 (en) 2003-11-21
NZ523438A (en) 2005-02-25
WO2002008241A2 (en) 2002-01-31
ES2536972T3 (en) 2015-06-01
LTPA2016009I1 (en) 2016-04-25
NO20030270D0 (en) 2003-01-20
OA12393A (en) 2006-04-18
CZ304734B6 (en) 2014-09-10
CN100402539C (en) 2008-07-16
NL300803I2 (en) 2016-06-30
US7390791B2 (en) 2008-06-24
CA2416757C (en) 2011-02-15
CN1706855A (en) 2005-12-14
PT1301519E (en) 2015-06-11
JP2010174033A (en) 2010-08-12
HU230960B1 (en) 2019-06-28
BR0112646A (en) 2003-06-24
EP1301519A2 (en) 2003-04-16
BRPI0112646B1 (en) 2017-10-17
AP2003002724A0 (en) 2003-06-30
CZ2003413A3 (en) 2003-12-17
EP2682397A1 (en) 2014-01-08
CY2016008I1 (en) 2016-12-14
EP3235823A1 (en) 2017-10-25
AP1466A (en) 2005-09-22
NO20131717L (en) 2003-03-20
JP5063554B2 (en) 2012-10-31
CZ304886B6 (en) 2015-01-07
CA2416757A1 (en) 2002-01-31
IS6689A (en) 2003-01-17
LU93029I2 (en) 2016-06-14
EA200300188A1 (en) 2003-06-26
NZ535408A (en) 2006-09-29
AU2005225039B2 (en) 2008-09-25
TR200300055T2 (en) 2004-12-21
NZ536942A (en) 2006-03-31
IS2985B (en) 2017-09-15
HUS000494I2 (en) 2021-03-29
AU2001282941C1 (en) 2016-12-22
CY2016008I2 (en) 2016-12-14
HRP20030047A2 (en) 2007-08-31
AU2005225039A1 (en) 2005-11-10
LT2682397T (en) 2017-06-12
KR20060105807A (en) 2006-10-11
ES2627903T3 (en) 2017-08-01
NO20150909L (en) 2003-03-20
PL213214B1 (en) 2013-01-31
JP2004504402A (en) 2004-02-12
SI1301519T1 (en) 2015-07-31
DK2682397T3 (en) 2017-06-19
MXPA03000587A (en) 2004-04-05
AU8294101A (en) 2002-02-05
JP4651264B2 (en) 2011-03-16
FR16C0013I1 (en) 2016-05-27
BG107572A (en) 2003-11-28
BE2016C018I2 (en) 2020-08-20
WO2002008241A3 (en) 2002-08-29
PL360490A1 (en) 2004-09-06
KR100767432B1 (en) 2007-10-17
KR100749160B1 (en) 2007-08-14
EP1301519B2 (en) 2021-11-10
BG66037B1 (en) 2010-11-30
EP1301519B1 (en) 2015-02-25
CN1291994C (en) 2006-12-27
HRP20160074A8 (en) 2016-07-29
HK1243711A1 (en) 2018-07-20
US20080227754A1 (en) 2008-09-18
NO2023006I1 (en) 2023-02-03
AU2001282941B2 (en) 2006-04-27
CA2893174A1 (en) 2002-01-31
US20040018150A1 (en) 2004-01-29
US20050009043A1 (en) 2005-01-13
HUS1900027I1 (en) 2021-03-29
US20030219727A1 (en) 2003-11-27
EA004926B1 (en) 2004-10-28
US20050124584A1 (en) 2005-06-09
ES2536972T5 (en) 2022-04-06
DK1301519T4 (en) 2021-12-20
NO2016006I2 (en) 2016-04-19
HUP0301307A2 (en) 2003-09-29
KR20030022295A (en) 2003-03-15
HRP20030047B1 (en) 2016-02-26
IL153658A0 (en) 2003-07-06
HUP0301307A3 (en) 2005-12-28
US20060024659A1 (en) 2006-02-02
SI2682397T1 (en) 2017-08-31
EP2682397B1 (en) 2017-04-19
JP2009062383A (en) 2009-03-26
BRPI0112646B8 (en) 2021-05-25
CY1119411T1 (en) 2018-03-07
HRP20160074B1 (en) 2021-09-03
UA75889C2 (en) 2006-06-15
EE200300029A (en) 2004-10-15
HRP20160074A2 (en) 2016-03-11
JP5111551B2 (en) 2013-01-09
US20050124583A1 (en) 2005-06-09
CA2725819A1 (en) 2002-01-31
NO20030270L (en) 2003-03-20
CA2725819C (en) 2015-09-22
CN1443189A (en) 2003-09-17
PT2682397T (en) 2017-05-31

Similar Documents

Publication Publication Date Title
US7803788B2 (en) Prodrugs of phosphonate nucoleotide analogues
AU2001282941A1 (en) Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION