US20060246130A1 - Compositions and methods for combination antiviral therapy - Google Patents

Compositions and methods for combination antiviral therapy Download PDF

Info

Publication number
US20060246130A1
US20060246130A1 US10/540,794 US54079404A US2006246130A1 US 20060246130 A1 US20060246130 A1 US 20060246130A1 US 54079404 A US54079404 A US 54079404A US 2006246130 A1 US2006246130 A1 US 2006246130A1
Authority
US
United States
Prior art keywords
emtricitabine
tenofovir disoproxil
disoproxil fumarate
hiv
pharmaceutical formulation
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
US10/540,794
Other languages
English (en)
Inventor
Terrence Dahl
Mark Menning
Reza Oliyai
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=32776014&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20060246130(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US10/540,794 priority Critical patent/US20060246130A1/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Assigned to GILEAD SCIENCES, INC. reassignment GILEAD SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DAHL, TERRENCE C., MENNING, MARK M., OLIYAI, REZA
Publication of US20060246130A1 publication Critical patent/US20060246130A1/en
Priority to US12/195,161 priority patent/US8592397B2/en
Priority to US12/204,174 priority patent/US8716264B2/en
Priority to US14/227,653 priority patent/US20140213556A1/en
Priority to US14/523,758 priority patent/US9457036B2/en
Priority to US14/523,783 priority patent/US9744181B2/en
Priority to US15/586,500 priority patent/US20170232019A1/en
Priority to US15/622,484 priority patent/US20170273994A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention relates generally to combinations of compounds with antiviral activity and more specifically with anti-HIV properties. In particular, it relates to chemically stable combinations of structurally diverse anti-viral agents.
  • HIV-1 Human immunodeficiency virus (HIV) infection and related diseases are a major public health problem worldwide.
  • Human immunodeficiency virus type 1 (HIV-1) encodes at least three enzymes which are required for viral replication: reverse transcriptase (RT), protease (Prt), and integrase (Int).
  • RT reverse transcriptase
  • Prt protease
  • Int integrase
  • drugs targeting reverse transcriptase and protease are in wide use and have shown effectiveness, particularly when employed in combination, toxicity and development of resistant strains have limited their usefulness (Palella, et al N. Engl. J. Med . (1998) 338:853-860; Richman, D. D. Nature (2001) 410:995-1001).
  • HIV-1 protease Prt
  • the HIV Prt cleaves the viral Gag and Gag-Pol polyproteins to produce viral structural proteins (p17, p24, p7 and p6) and the three viral enzymes.
  • Combination therapy with RT inhibitors has proven to be highly effective in suppressing viral replication to unquantifiable levels for a sustained period of time. Also, combination therapy with RT and Prt inhibitors (PI) have shown synergistic effects in suppressing HIV replication.
  • AZT zidovudineTM, 3′-azido, 3′-deoxythymidine
  • AZT demonstrates synergistic antiviral activity in vitro in combination with agents that act at HIV-1 replicative steps other than reverse transcription, including recombinant soluble CD4 castanospermine and recombinant interferon- ⁇ .
  • agents that act at HIV-1 replicative steps other than reverse transcription including recombinant soluble CD4 castanospermine and recombinant interferon- ⁇ .
  • combinations of compounds can give rise to increased cytotoxicity.
  • AZT and recombinant interferon- ⁇ have an increased cytotoxic effect on normal human bone marrow progenitor cells.
  • Chemical stability of combinations of antiviral agents is an important aspect of co-formulation success and the present invention provides examples of such combinations.
  • the present invention provides combinations of antiviral compounds, in particular compositions and methods for inhibition of HIV.
  • the invention includes a composition including tenofovir disoproxil fumarate and emtricitabine which has anti-HIV activity.
  • the composition of tenofovir DF and emtricitabine is both chemically stable and either synergistic and/or reduces the side effects of one or both of tenofovir DF and emtricitabine. Increased patient compliance is likely in view of the lower pill burden and simplified dosing schedule.
  • the present invention relates to therapeutic combinations of [2-(6-amino-purin-9-yl)-1-methyl-ethoxymethyl]-phosphonic acid diisopropoxycarbonyloxymethyl ester fumarate (tenofovir disoproxil fumarate, tenofovir DF, TDF, Viread®) and (2R,5S,cis)-4-amino-5-fluoro-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one (emtricitabine, EmtrivaTM, ( ⁇ )-cis FTC) and their use in the treatment of HIV infections including infections with HIV mutants bearing resistance to nucleoside and/or non-nucleoside inhibitors.
  • the present invention is also concerned with pharmaceutical compositions and formulations of said combinations of tenofovir disoproxil fumarate and emtricitabine.
  • Another aspect of the invention is a pharmaceutical formulation comprising a physiologically functional derivative of tenofovir disoproxil fumarate or a physiologically functional derivative of emtricitabine.
  • Therapeutic combinations and pharmaceutical compositions and formulations of the invention include the combination of PMAE or PMPA (tenofovir) compounds with emtricitabine or (2R,5S,cis)-4-amino-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one (3TC, lamivudine, EpivirTM), and their use in the treatment of HIV infections.
  • One aspect of the invention is a method for the treatment or prevention of the symptoms or effects of an HIV infection in an infected animal which comprises administering to, i.e. treating, said animal with a therapeutically effective amount of a combination comprising [2-(6-amino-purin-9-yl)-1-methyl-ethoxymethyl]-phosphonic acid diisopropoxycarbonyloxymethyl ester fumarate (tenofovir DF, DIP) or a physiologically functional derivative thereof, and (2R,5S,cis)-4-amino-5-fluoro-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one (emtricitabine) or a physiologically functional derivative thereof.
  • Another aspect of the invention is a unit dosage form of a therapeutic combination comprising tenofovir disoproxil fumarate and emtricitabine, or physiological functional derivatives thereof.
  • the unit dosage form may be formulated for administration by oral or other routes and is unexpectedly chemically stable in view of the properties of the structurally diverse components.
  • Another aspect of the invention is directed to chemically stable combination antiviral compositions comprising tenofovir disoproxil fumarate and emtricitabine.
  • the chemically stable combinations of tenofovir disoproxil fumarate and emtricitabine further comprise a third antiviral agent.
  • the unique chemical stability of tenofovir disoproxil fumarate and emtricitabine is taken advantage of in order to enable the combination with the third antiviral agent.
  • Particularly useful third agents include, by way of example and not limitation, those of Table A.
  • the third component is an agent approved for antiviral use in humans, more preferably, it is an NNRTI or a protease inhibitor (PI), more preferably yet, it is an NNRTI.
  • the invention is directed to a combination of the chemically stable mixture of tenofovir disoproxil fumarate and emtricitabine together with efavirenz.
  • Another aspect of the invention is a patient pack comprising at least one, typically two, and optionally, three active ingredients and other antiviral agents selected from tenofovir disoproxil fumarate and emtricitabine, and an information insert containing directions on the use of tenofovir disoproxil fumarate and emtricitabine together in combination.
  • Another aspect of the invention is a process for preparing the combinations hereinbefore described, which comprises bringing into association tenofovir DF and emtricitabine of the combination in a medicament to provide an antiviral effect.
  • a combination of the present invention in the manufacture of a medicament for the treatment of any of the aforementioned viral infections or conditions.
  • chemical stability means that the two primary antiviral agents in combination are substantially stable to chemical degradation. Preferably, they are sufficiently stable in physical combination to permit commercially useful shelf life of the combination product.
  • “chemically stable” means that a first component of the mixture does not act to degrade a second component when the two are brought into physical combination to form a pharmaceutical dosage form. More typically, “chemically stable” means that the acidity of a first component does not catalyzes or otherwise accelerate the acid decomposition of a second component.
  • “chemically stable” means that tenofovir disoproxil fumarate is not substantially degraded by the acidity of emtricitabine.
  • “Substantially” in this context means at least about less than 10%, preferably less than 1%, more preferably less than 0.1%, more preferably yet, less than 0.01% acid degradation of tenofovir disoproxil fumarate over a 24-hour period when the products are in a pharmaceutical dosage form.
  • synergy and “synergistic” mean that the effect achieved with the compounds used together is greater than the sum of the effects that results from using the compounds separately, i.e. greater than what would be predicted based on the two active ingredients administered separately.
  • a synergistic effect may be attained when the compounds are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. in separate tablets, pills or capsules, or by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • a synergistic antiviral effect denotes an antiviral effect which is greater than the predicted purely additive effects of the individual compounds of the combination.
  • physiologically functional derivative means a pharmaceutically active compound with equivalent or near equivalent physiological functionality to tenofovir DF or emtricitabine when administered in combination with another pharmaceutically active compound in a combination of the invention.
  • physiologically functional derivative includes any: physiologically acceptable salt, ether, ester, prodrug, solvate, stereoisomer including enantiomer, diastereomer or stereoisomerically enriched or racemic mixture, and any other compound which upon administration to the recipient, is capable of providing (directly or indirectly) such a compound or an antivirally active metabolite or residue thereof.
  • Bioavailability is the degree to which the pharmaceutically active agent becomes available to the target tissue after the agent's introduction into the body. Enhancement of the bioavailability of a pharmaceutically active agent can provide a more efficient and effective treatment for patients because, for a given dose, more of the pharmaceutically active agent will be available at the targeted tissue sites.
  • the compounds of the combinations of the invention may be referred to as “active ingredients” or “pharmaceutically active agents.”
  • prodrug refers to any compound that when administered to a biological system generates the drug substance, i.e. active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s).
  • Prodrug moiety means a labile functional group which separates from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, “Design and Application of Prodrugs” in Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113-191). Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimize drug delivery, bioavailability and efficacy. A “prodrug” is thus a covalently modified analog of a therapeutically-active compound.
  • Alkyl means a saturated or unsaturated, branched, straight-chain, branched, or cyclic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene, or alkyne.
  • Typical alkyl groups consist of 1-18 saturated and/or unsaturated carbons, such as normal, secondary, tertiary or cyclic carbon atoms.
  • Examples include, but are not limited to: methyl, Me(-CH 3 ), ethyl, Et(-CH 2 CH 3 ), acetylenic (—C ⁇ CH), ethylene, vinyl (—CH ⁇ CH 2 ), 1-propyl, n-Pr, n-propyl (—CH 2 CH 2 CH 3 ), 2-propyl, i-Pr, i-propyl (—CH(CH 3 ) 2 ), allyl (—CH 2 CH ⁇ CH 2 ), propargyl (—CH 2 C ⁇ CH), cyclopropyl (—C 3 H 5 ), 1-butyl, n-Bu, n-butyl (—CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1-propyl, i-Bu, i-butyl (—CH 2 CH(CH 3 ) 2 ), 2-butyl, s-Bu, s-butyl (—CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl, t-Bu
  • Aryl means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like.
  • Arylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with an aryl radical.
  • Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like.
  • the arylalkyl group 6 to 20 carbon atoms e.g., the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, of the arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 5 to 14 carbon atoms.
  • Substituted alkyl mean alkyl, aryl, and arylalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a substituent.
  • Typical substituents include, but are not limited to, —X, —R, —O ⁇ , —OR, —SR, —S ⁇ , —NR 2 , —NR 3 , ⁇ NR, —CX 3 , —CN, —OCN, —SCN, —N ⁇ C ⁇ O, —NCS, —NO, —NO 2 , ⁇ N 2 , —N 3 , NC( ⁇ O)R, —C( ⁇ O)R, —C( ⁇ O)NRR—S( ⁇ O) 2 O ⁇ , —S( ⁇ O) 2 OH, —S( ⁇ O) 2 R, —OS( ⁇ O) 2 OR, —S( ⁇ O) 2 NR, —S( ⁇ O)R, —OP( ⁇ O)O 2 RR, —P( ⁇ O)O 2 RR—P( ⁇ O)(O ⁇ ) 2 , —P( ⁇ O)(OH) 2 ,
  • Heteroaryl and Heterocycle refer to a ring system in which one or more ring atoms is a heteroatom, e.g. nitrogen, oxygen, and sulfur. Heterocycles are described in: Katritzky, Alan R., Rees, C. W., and Scriven, E. Comprehensive Heterocyclic Chemistry (1996) Pergamon Press; Paquette, Leo A.; Principles of Modern Heterocyclic Chemistry W. A. Benjamin, New York, (1968), particularly Chapters 1, 3, 4, 6, 7, and 9; “The Chemistry of Heterocyclic Compounds, A series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28.
  • heterocycles include but are not limited to substituents, i.e. radicals, derived from pyrrole, indole, furan, benzofuran, thiophene, benzothiophene, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-quinolyl, 3-quinolyl, 4-quinolyl, 2-imidazole, 4-imidazole, 3-pyrazole, 4-pyrazole, pyridazine, pyrimidine, pyrazine, purine, cinnoline, pthalazine, quinazoline, quinoxaline, 3-(1,2,4-N)-triazolyl, 5-(1,2,4-N)-triazolyl, 5-tetrazolyl, 4-(1-O,3-N)-oxazole, 5-(1-O,3-N)-oxazole, 4-(1-S,3-N)-thiazole, 5-(1-S,3-N)-thi
  • d and L or (+) and ( ⁇ ) are employed to designate the sign of rotation of plane-polarized light by the compound, with ( ⁇ ) or 1 meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these compounds, called stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer is also referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate.
  • racemic mixture and racemate refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • the present invention provides novel combinations of two or more active ingredients being employed together.
  • a synergistic antiviral effect is achieved.
  • a chemically stable combination is obtained.
  • the combinations include at least one active ingredient selected from (1) tenofovir disoproxil fumarate and physiologically functional derivatives, and at least one active ingredient selected from (2) emtricitabine and physiologically functional derivatives.
  • the term “synergistic antiviral effect” is used herein to denote an antiviral effect which is greater than the predicted purely additive effects of the individual components (a) and (b) of the combination.
  • Tenofovir disoproxil fumarate also known as Viread®, Tenofovir DF, Tenofovir disoproxil, TDF, Bis-POC-PMPA (U.S. Pat. Nos. 5,935,946, 5,922,695, 5,977,089, 6,043,230, 6,069,249) is a prodrug of tenofovir, and has the structure:
  • fumarate salt HO 2 CCH 2 CH 2 CO 2 ⁇ .
  • Tenofovir disoproxil include: [2-(6-amino-purin-9-yl)-1-methyl-ethoxymethyl]-phosphonic acid diisopropoxycarbonyloxymethyl ester; 9-[(R)-2-[[bis[[(isopropoxycarbonyl)oxy]methoxy]phosphinyl]methoxy]propyl]adenine; and 2,4,6,8-tetraoxa-5-phosphanonanedioic acid, 5-[[(1R)-2-(6-amino-9H-purin-9-yl)-1-methylethoxy]methyl]-, bis(1-methylethyl)ester, 5-oxide.
  • the CAS Registry numbers include: 201341-05-1; 202138-50-9; 206184-49-8. It should be noted that the ethoxymethyl unit of tenofovir has a chiral center.
  • the R (rectus, right handed configuration) enantiomer is shown. However, the invention also includes the S isomer.
  • the invention includes all enantiomers, diastereomers, racemates, and enriched stereoisomer mixtures of tenofovir (PMPA) and physiologically functional derivatives thereof.
  • PMPA or tenofovir (U.S. Pat. Nos. 4,808,716, 5,733,788, 6,057,305) has the structure:
  • the chemical names of PMPA, tenofovir include: (R)-9-(2-phosphonylmethoxypropyl)adenine; and phosphonic acid, [[(1R)-2-(6-amino-9H-purin-9-yl)-1-methylethoxy]methyl].
  • the CAS Registry number is 147127-20-6.
  • Tenofovir disoproxil fumarate is a nucleotide reverse transcriptase inhibitor approved in the United States in 2001 for the treatment of HIV-1 infection in combination with other antiretroviral agents.
  • Tenofovir disoproxil fumarate or Viread® is the fumarate salt of tenofovir disoproxil.
  • Viread® may be named as: 9-[(R)-2-[[bis[[(isopropoxycarbonyl)oxy]methoxy]phosphinyl]methoxy]propyl]adenine fumarate (1:1); or 2,4,6,8-tetraoxa-5-phosphanonanedioic acid, 5-[[(1R)-2-(6-amino-9H-purin-9-yl)-1-methylethoxy]methyl]-, bis(1-methylethyl)ester, 5-oxide, (2E)-2-butenedioate (1:1).
  • the CAS Registry number is 202138-50-9.
  • Physiologically functional derivatives of tenofovir disoproxil fumarate include PMEA (adefovir, 9-((R)-2-(phosphonomethoxy)ethyl)adenine) and PMPA compounds.
  • Exemplary combinations include a PMEA or PMPA compound in combination with emtricitabine or 3TC.
  • PMEA and PMPA compounds have the structures: where PMEA (R 3 is H) and PMPA (R3 is C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, or CH 2 OR 8 where R 8 is C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl or C 1 -C 6 haloalkyl.
  • R 6 and R 7 are independently H or C 1 -C 6 alkyl.
  • R 4 and R 5 are independently H, NH 2 , NHR or NR 2 where R is C 1 -C 6 alkyl.
  • R 1 and R 2 are independently H, C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 6 -C 20 arylalkyl, C 6 -C 20 substituted arylalkyl, acyloxymethyl esters —CH 2 C( ⁇ O)R 9 (e.g. POM) or acyloxymethyl carbonates —CH 2 C( ⁇ O)OR 9 (e.g.
  • R 9 is C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl or C 6 -C 20 substituted aryl.
  • R 1 and R 2 may be pivaloyloxymethoxy, POM, —CH 2 C( ⁇ O)C(CH 3 ) 3 ; —CH 2 C( ⁇ O)OC(CH 3 ) 3 ; or POC, —CH 2 C( ⁇ O)OCH(CH 3 ) 2 .
  • tenofovir has the structure where R 3 is CH 3 , and R 1 , R 2 , R 4 , R 5 , R 6 and R 7 are H.
  • Dialkyl phosphonates may be prepared according to the methods of: Quast et al (1974) Synthesis 490; Stowell et al (1990) Tetrahedron Lett. 3261; U.S. Pat. No. 5,663,159.
  • the PMPA compound may be enantiomerically-enriched or purified (single stereoisomer) where the carbon atom bearing R 3 may be the R or S enantiomer.
  • the PMPA compound may be a racemate, i.e. a mixture of R and S stereoisomers.
  • Adefovir (9-(2-phosphonomethoxyethyl)adenine where R 1 -R 7 ⁇ H) is an exemplary PMEA compound (U.S. Pat. Nos. 4,808,716, 4,724,233).
  • Adefovir dipivoxil has demonstrated minor to moderate synergistic inhibition of HIV replication in combination with other compounds with anti-HIV activity including PMPA, d4T, ddC, nelfinavir, ritonavir, and saquinavir (Mulato et al (1997) Antiviral Research 36:91-97).
  • the invention includes all enantiomers, diastereomers, racemates, and enriched stereoisomer mixtures of PMEA and PMPA, and physiologically functional derivatives thereof.
  • Emtricitabine ( ⁇ )-cis-FIC, EmtrivaTM), a single enantiomer of FTC, is a potent nucleoside reverse transcriptase inhibitor approved for the treatment of HIV (U.S. Pat. Nos. 5,047,407, 5,179,104, 5,204,466, 5,210,085, 5,486,520, 5,538,975, 5,587,480, 5,618,820, 5,763,606, 5,814,639, 5,914,331, 6,114,343, 6,180,639, 6,215,004; WO 02/070518).
  • the single enantiomer emtricitabine has the structure:
  • the chemical names for emtricitabine include: ( ⁇ )-cis-FIC; ⁇ -L-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane; (2R,5S)-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine; and 4-amino-5-fluoro-1-(2-hydroxymethyl-[1,3]-(2R,5S)-oxathiolan-5-yl)-1H-pyrimidin-2-one.
  • the CAS Registry numbers include: 143491-57-0; 143491-54-7.
  • FTC contains two chiral centers, at the 2 and 5 positions of the oxathiolane ring, and therefore can exist in the form of two pairs of optical isomers (i.e. enantiomers) and mixtures thereof including racemic mixtures.
  • FTC may be either a cis or a trans isomer or mixtures thereof.
  • Mixtures of cis and trans isomers are diastereomers with different physical properties.
  • Each cis and trans isomer can exist as one of two enantiomers or mixtures thereof including racemic mixtures.
  • the invention includes all enantiomers, diastereomers, racemates, and enriched stereoisomer mixtures of emtricitabine and physiologically functional derivatives thereof.
  • the invention includes physiological functional derivatives such as the 1:1 racemic mixture of the enantiomers (2R,5S,cis)-4-amino-5-fluoro-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one (emtricitabine) and its mirror image (2S,5R, cis)-4-amino-5-fluoro-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one, or mixtures of the two enantiomers in any relative amount.
  • the invention also includes mixtures of cis and trans forms of FTC.
  • Physiologically functional derivatives of emtricitabine include 1,3 oxathiolane nucleosides having the structure:
  • B is a nucleobase including any nitrogen-containing heterocyclic moiety capable of forming Watson-Crick hydrogen bonds in pairing with a complementary nucleobase or nucleobase analog, e.g. a purine, a 7-deazapurine, or a pyrimidine.
  • a complementary nucleobase or nucleobase analog e.g. a purine, a 7-deazapurine, or a pyrimidine.
  • B include the naturally occurring nucleobases: adenine, guanine, cytosine, uracil, thymine, and minor constituents and analogs of the naturally occurring nucleobases, e.g.
  • Nucleobases B may be attached in the configurations of naturally-occurring nucleic acids to the 1,3 oxathiolane moiety through a covalent bond between the N-9 of purines, e.g. adenin-9-yl and guanin-9-yl, or N-1 of pyrimidines, e.g. thymin-1-yl and cytosin-1-yl (Blackburn, G. and Gait, M. Eds. “DNA and RNA structure” in Nucleic Acids in Chemistry and Biology, 2 nd Edition, (1996) Oxford University Press, pp. 15-81).
  • purines e.g. adenin-9-yl and guanin-9-yl
  • N-1 of pyrimidines e.g. thymin-1-yl and cytosin-1-yl
  • R is H, C 1 -C 18 alkyl, C 1 -C 18 substituted alkyl, C 2 -C 18 alkenyl, C 2 -C 18 substituted alkenyl, C 2 -C 18 alkynyl, C 2 -C 18 substituted alkynyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 2 -C 20 heterocycle, C 2 -C 20 substituted heterocycle, phosphonate, phosphophosphonate, diphosphophosphonate, phosphate, diphosphate, triphosphate, polyethyleneoxy, or a prodrug moiety.
  • Physiologically functional derivatives of emtricitabine also include 3TC (lamivudine, Epivir®), a reverse transcriptase inhibitor approved in the United States for the treatment of HIV-1 infection in combination with AZT as Combivir® (GlaxoSmithKline).
  • 3TC lamivudine, Epivir®
  • Re transcriptase inhibitor approved in the United States for the treatment of HIV-1 infection in combination with AZT as Combivir® (GlaxoSmithKline).
  • Lamivudine U.S. Pat. Nos. 5,587,480, 5,696,254, 5,618,820, 5,756,706, 5,744,596, 568,164, 5,466,806, 5,151,426) has the structure:
  • 3TC may be a physiologically functional derivative of emtricitabine in combination with tenofovir DF or a physiologically functional derivative of tenofovir DF.
  • tenofovir DF and emtricitabine may exist in keto or enol tautomeric forms and the use of any tautomeric form thereof is within the scope of this invention.
  • Tenofovir DF and emtricitabine will normally be utilized in the combinations of the invention substantially free of the corresponding enantiomer, that is to say no more than about 5% w/w of the corresponding enantiomer will be present.
  • the invention includes all prodrugs of tenofovir and emtricitabine.
  • An exemplary prodrug of tenofovir is tenofovir disoproxil fumarate (TDF, Viread®).
  • TDF tenofovir disoproxil fumarate
  • a large number of structurally-diverse prodrugs have been described for phosphonic acids (Freeman and Ross in Progress in Medicinal Chemistry 34: 112-147 (1997).
  • a commonly used prodrug class is the acyloxyalkyl ester, which was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al (1983) J. Pharm. Sci. 72: 324; also U.S. Pat. Nos.
  • acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bioavailability.
  • a close variant of the acyloxyalkyl ester strategy, the alkoxycarbonyloxyalkyl ester, may also enhance oral bioavailability as a prodrug moiety in the compounds of the combinations of the invention.
  • Aryl esters of phosphorus groups, especially phenyl esters, are reported to enhance oral bioavailability (DeLambert et al (1994) J. Med. Chem. 37: 498).
  • Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described (Khamnei and Torrence, (1996) J. Med. Chem. 39:4109-4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ortho- or para-position may accelerate the hydrolysis. Benzyl analogs with an acylated phenol or an alkylated phenol may generate the phenolic compound through the action of enzymes, e.g. esterases, oxidases, etc., which in turn undergoes cleavage at the benzylic C—O bond to generate the phosphoric acid and the quinone methide intermediate.
  • enzymes e.g. esterases, oxidases, etc.
  • prodrugs examples include Mitchell et al (1992) J. Chem. Soc. Perkin Trans. 12345; Brook et al WO 91/19721. Still other benzylic prodrugs have been described containing a carboxylic ester-containing group attached to the benzylic methylene (Glazier et al WO 91/19721). Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphonate drugs. These proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide.
  • Prodrug esters in accordance with the invention are independently selected from the following groups: (1) mono-, di-, and tri-phosphate esters of tenofovir or emtricitabine or any other compound which upon administration to a human subject is capable of providing (directly or indirectly) said mono-, di, or triphosphate ester; (2) carboxylic acid esters (3) sulphonate esters, such as alkyl- or aralkylsulphonyl (for example, methanesulphonyl); (4) amino acid esters (for example, alanine, L-valyl or L-isoleucyl); (5) phosphonate; and (6) phosphonamidate esters.
  • Ester groups (1)-(6) may be substituted with; straight or branched chain C 1 -C 18 alkyl (for example, methyl, n-propyl, t-butyl, or n-butyl); C 3 -C 12 cycloalkyl; alkoxyalkyl (for example, methoxymethyl); arylalkyl (for example, benzyl); aryloxyalkyl (for example, phenoxymethyl); C 5 -C 20 aryl (for example, phenyl optionally substituted by, for example, halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, or amino; acyloxymethyl esters —CH 2 C( ⁇ O)R 9 (e.g.
  • ester groups may be: —CH 2 C( ⁇ O)C(CH 3 ) 3 , —CH 2 C( ⁇ O)OC(CH 3 ) 3 or —CH 2 C( ⁇ O)OCH(CH 3 ) 2 .
  • An exemplary aryl moiety present in such esters comprises a phenyl or substituted phenyl group.
  • Many phosphate prodrug moieties are described in U.S. Pat. No. 6,312,662; Jones et al (1995) Antiviral Research 27:1-17; Kucera et al (1990) AIDS Res. Hum. Retro Viruses 6:491-501; Piantadosi et al (1991) J. Med. Chem. 34:1408-14; Hosteller et al (1992) Antimicrob. Agents Chemother. 36:2025-29; Hostetler et al (1990) J. Biol. Chem. 265:611127; and Siddiqui et al (1999) J. Med. Chem. 42:4122-28.
  • prodrugs refer to a compound that is metabolized in the host, for example hydrolyzed or oxidized, by either enzymatic action or by general acid or base solvolysis, to form an active ingredient.
  • Typical examples of prodrugs of the active ingredients of the combinations of the invention have biologically labile protecting groups on a functional moiety of the active compound.
  • Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, esterified, deesterified, alkylated, dealkylated, acylated, deacylated, phosphorylated, dephosphorylated, or other functional group change or conversion involving forming or breaking chemical bonds on the prodrug.
  • the chemical stability of the active ingredients in a pharmaceutical formulation is of concern to minimize the generation of impurities and ensure adequate shelf-life.
  • the active ingredients, tenofovir disoproxil fumarate and emtricitabine, in the pharmaceutical formulations of the invention have relatively low pKa values, indicative of the potential to cause acidic hydrolysis of the active ingredients.
  • Emtricitabine, with a pKa of 2.65 is subject to hydrolytic deamination of the 5-fluoro cytosine nucleobase to form the 5-fluoro uridine nucleobase.
  • Tenofovir disoproxil fumarate with a pKa of 3.75 (Yuan L. et al “Degradation Kinetics of Oxycarbonyloxymethyl Prodrugs of Phosphonates in Solution”, Pharmaceutical Research (2001) Vol. 18, No. 2, 234-237), is subject also to hydrolytic deamination of the exocyclic amine of the adenine nucleobase, and to hydrolysis of one or both of the POC ester groups (U.S. Pat. No. 5,922,695). It is desirable to formulate a therapeutic combination of tenofovir disoproxil fumarate and emtricitabine, and the physiological functional derivatives thereof, with a minimum of impurities and adequate stability.
  • the combinations of the present invention provide combination pharmaceutical dosage forms which are chemically stable to acid degradation of: (1) a first component (such as tenofovir disoproxil fumarate, and physiological functional derivatives; (2) a second component (such as emtricitabine, and physiological functional derivatives; and (3) optionally a third component having antiviral activity.
  • the third component includes anti-HIV agents and include: protease inhibitors (PI), nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI), and integrase inhibitors.
  • Exemplary third active ingredients to be administered in combination with first and second components are shown in Table A. First and second components are as defined in the above section entitled: ACTIVE INGREDIENTS OF THE COMBINATIONS.
  • any reference to any of the compounds in the compositions of the invention also includes any physiologically acceptable salt thereof.
  • physiologically acceptable salts of tenofovir DF, emtricitabine and their physiologically functional derivatives include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth (for example, magnesium), ammonium and NX 4 + (wherein X is C 1 -C 4 alkyl), or an organic acid such as fumaric acid, acetic acid, succinic acid.
  • Physiologically acceptable salts of an hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, sulfuric, phosphoric and sulfamic acids.
  • organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids
  • organic sulfonic acids such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids
  • Physiologically acceptable salts of a compound of an hydroxy group include the anion of said compound in combination with a suitable cation such as Na + and NX 4 + (wherein X is independently selected from H or a C 1 -C 4 alkyl group).
  • salts of active ingredients of the combinations of the invention will be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base.
  • salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived from a physiologically acceptable acid or base, are within the scope of the present invention.
  • a two-part or three-part combination may be administered simultaneously or sequentially.
  • the combination may be administered in one, two, or three administrations.
  • two-part or three-part combinations are administered in a single pharmaceutical dosage form. More preferably, a two-part combination is administered as a single oral dosage form and a three-part combination is administered as two identical oral dosage forms. Examples include a single tablet of tenofovir disoproxil fumarate and emtricitabine, or two tablets of tenofovir disoproxil fumarate, emtricitabine, and efavirenz.
  • the compounds of the combination may be administered: (1) simultaneously by combination of the compounds in a co-formulation or (2) by alternation, i.e. delivering the compounds serially, sequentially, in parallel or simultaneously in separate pharmaceutical formulations.
  • alternation therapy the delay in administering the second, and optionally a third active ingredient, should not be such as to lose the benefit of a synergistic therapeutic effect of the combination of the active ingredients.
  • the combination should be administered to achieve peak plasma concentrations of each of the active ingredients.
  • a one pill once-per-day regimen by administration of a combination co-formulation may be feasible for some HIV-positive patients.
  • Effective peak plasma concentrations of the active ingredients of the combination will be in the range of approximately 0.001 to 100 ⁇ M. Optimal peak plasma concentrations may be achieved by a formulation and dosing regimen prescribed for a particular patient. It will also be understood that tenofovir DF and emtricitabine, or the physiologically functional derivatives of either thereof, whether presented simultaneously or sequentially, may be administered individually, in multiples, or in any combination thereof. In general, during alternation therapy (2), an effective dosage of each compound is administered serially, where in co-formulation therapy (1), effective dosages of two or more compounds are administered together.
  • the references hereinafter to formulations refer unless otherwise stated to formulations containing either the combination or a component compound thereof. It will be understood that the administration of the combination of the invention by means of a single patient pack, or patient packs of each formulation, within a package insert diverting the patient to the correct use of the invention is a desirable additional feature of this invention.
  • the invention also includes a double pack comprising in association for separate administration, formulations of tenofovir disoproxil fumarate and emtricitabine, or a physiologically functional derivative of either or both thereof.
  • the combination therapies of the invention include: (1) a combination of tenofovir DF and emtricitabine or (2) a combination containing a physiologically functional derivative of either or both thereof.
  • the combination may be formulated in a unit dosage formulation comprising a fixed amount of each active pharmaceutical ingredient for a periodic, e.g. daily, dose or subdose of the active ingredients.
  • compositions according to the present invention comprise a combination according to the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.
  • Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared ( Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.).
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including antioxidants, sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • inert diluents such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example pregelatinized starch, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example pregelatinized starch, calcium phosphate or kaolin
  • water or an oil medium such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate).
  • a suspending agent
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose, sucralose or saccharin.
  • preservatives such as ethyl or n-propyl p-hydroxybenzoate
  • coloring agents such as a coloring agent
  • flavoring agents such as sucrose, sucralose or saccharin.
  • sweetening agents such as sucrose, sucralose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an antioxidant such as ascorbic acid, BHT, etc.
  • Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium tartrate
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions or liposome formulations.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these.
  • Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic
  • compositions of the invention may be injected parenterally, for example, intravenously, intraperitoneally, intrathecally, intraventricularly, intrastemally, intracranially, intramuscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • compositions of the invention may also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurized container or a nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFC 134a), carbon dioxide or other suitable gas.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFC 134a), carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurized container or nebuliser may contain a solution or suspension of the composition, e.g. using a mixture of ethanol and the propellant as the solvent, which may additional contain a lubricant, e.g. sorbitan trioleate.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the formula (I) and a suitable powder base such as lactose or starch. Aerosol or dry powder formulations are preferably arranged so that each metered dose or “puff” contains from 20 ⁇ g to 20 mg of a composition for delivery to the patient.
  • the overall daily dose with an aerosol will be in the range of from 20 ⁇ g to 20 mg which may be administered in a single dose or, more usually, in divided doses throughout the day.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be administered as a bolus, electuary or paste.
  • the combinations of the invention may conveniently be presented as a pharmaceutical formulation in a unitary dosage form.
  • a convenient unitary dosage formulation contains the active ingredients in any amount from 1 mg to 1 g each, for example but not limited to, 10 mg to 300 mg.
  • the synergistic effects of tenofovir DF in combination with emtricitabine may be realized over a wide ratio, for example 1:50 to 50:1 (tenofovir DF:emtricitabine). In one embodiment, the ratio may range from about 1:10 to 10:1. In another embodiment, the weight/weight ratio of tenofovir to emtricitabine in a co-formulated combination dosage form, such as a pill, tablet, caplet or capsule will be about 1, i.e.
  • tenofovir DP and emtricitabine there may be more or less tenofovir than FTC.
  • 300 mg tenofovir DF and 200 mg emtricitabine can be co-formulated in a ratio of 1.5:1 (tenofovir DF: emtricitabine).
  • each compound will be employed in the combination in an amount at which it exhibits antiviral activity when used alone.
  • Exemplary Formulations A, B, C, D, E, and F (Examples) have ratios of 12:1 to 1:1 (tenofovir DF: emtricitabine).
  • Exemplary Formulations A, B, C, D, E, and F use amounts of tenofovir DF and emtricitabine ranging from 25 mg to 300 mg. Other ratios and amounts of the compounds of said combinations are contemplated within the scope of the invention.
  • a unitary dosage form may further comprise tenofovir DF and emtricitabine, or physiologically functional derivatives of either thereof, and a pharmaceutically acceptable carrier.
  • the amount of active ingredients in the combinations of the invention required for use in treatment will vary according to a variety of factors, including the nature of the condition being treated and the age and condition of the patient, and will ultimately be at the discretion of the attending physician or health care practitioner.
  • the factors to be considered include the route of administration and nature of the formulation, the animal's body weight, age and general condition and the nature and severity of the disease to be treated.
  • doses ranging from 25 mg to 200 mg twice-a-day for two weeks.
  • a 98-percent (1.75 Iog10) or greater viral suppression was observed.
  • a once-a-day dose of 200 mg of emtricitabine reduced the viral load by an average of 99 percent (1.92 Iog10).
  • Viread® tenofovir DF
  • EmtrivaTM emtricitabine
  • any two of the active ingredients in a unitary dosage form for simultaneous or sequential administration with a third active ingredient may be administered simultaneously or sequentially. When administered sequentially, the combination may be administered in two or three administrations.
  • Third active ingredients have anti-HIV activity and include protease inhibitors (PI), nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI), and integrase inhibitors.
  • PI protease inhibitors
  • NRTI nucleoside reverse transcriptase inhibitors
  • NRTI non-nucleoside reverse transcriptase inhibitors
  • integrase inhibitors integrase inhibitors.
  • Another aspect of the present invention is a three-part combination comprising tenofovir DF, FIC, and 9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine, also designated herein as GS-7340, which has the structure:
  • GS-7340 is a prodrug of tenofovir and the subject of commonly owned, pending application, U.S. Ser. No. 09/909,560, filed Jul. 20, 2001 and Becker et al WO 02/08241.
  • a ternary unitary dosage may contain 1 mg to 1000 mg of tenofovir disoproxil fumarate, 1 mg to 1000 mg of emtricitabine, and 1 mg to 1000 mg of the third active ingredient.
  • a unitary dosage form may further comprise tenofovir DF, emtricitabine, the third active ingredient, or physiologically functional derivatives of the three active ingredients thereof, and a pharmaceutically acceptable carrier.
  • Combinations of the present invention enable patients greater freedom from multiple dosage medication regimens and ease the needed diligence required in remembering and complying with complex daily dosing times and schedules.
  • the desired daily regimen may be presented in a single dose or as two or more sub-doses per day.
  • the combination of co-formulated tenofovir DF and emtricitabine may be administered as a single pill, once per day.
  • a further aspect of the invention is a patient pack comprising at least one active ingredient: tenofovir disoproxil fumarate, emtricitabine, or a physiologically functional derivative of either of the combination and an information package or product insert containing directions on the use of the combination of the invention.
  • Segregation of active ingredients in pharmaceutical powders and granulations is a widely recognized problem that can result in inconsistent dispersions of the active ingredients in final dosage forms.
  • Some of the main factors contributing to segregation are particle size, shape and density. Segregation is particularly troublesome when attempting to formulate a single homogenous tablet containing multiple active ingredients having different densities and different particle sizes.
  • Glidants are substances that have traditionally been used to improve the flow characteristics of granulations and powders by reducing interparticulate friction. See Lieberman, Lachman , & Schwartz, Pharmaceutical Dosage Forms: Tablets , Volume 1, p. 177-178 (1989), incorporated herein by reference.
  • Glidants are typically added to pharmaceutical compositions immediately prior to tablet compression to facilitate the flow of granular material into the die cavities of tablet presses.
  • Glidants include: colloidal silicon dioxide, asbestos free talc, sodium aluminosilicate, calcium silicate, powdered cellulose, microcrystalline cellulose, corn starch, sodium benzoate, calcium carbonate, magnesium carbonate, metallic stearates, calcium stearate, magnesium stearate, zinc stearate, stearowet C, starch, starch 1500, magnesium lauryl sulfate, and magnesium oxide.
  • Exemplary Tablet Formulation A has colloidal silicon dioxide (Examples).
  • Glidants can be used to increase and aid blend composition homogeneity in formulations of anti-HIV drugs (U.S. Pat. No. 6,113,920).
  • the novel compositions of the present invention may contain glidants to effect and maintain homogeneity of active ingredients during handling prior to tablet compression.
  • the present invention provides pharmaceutical formulations combining the active ingredients tenofovir DF and emtricitabine, or physiologically functional derivatives thereof, in a sufficiently homogenized form, and a method for using this pharmaceutical formulation.
  • An object of the present invention is to utilize glidants to reduce the segregation of active ingredients in pharmaceutical compositions during pre-compression material handling.
  • Another object of the present invention is to provide a pharmaceutical formulation combining the active ingredients tenofovir DF and emtricitabine, or physiologically functional derivatives thereof, with a pharmaceutically acceptable glidant, resulting in a mixture characterized by a pharmaceutically acceptable measure of homogeneity.
  • Formulations include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods represent a further feature of the present invention and include the step of bringing into association the active ingredients with the carrier, which constitutes one or more accessory ingredients, and maintaining chemical stability.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, caplets, cachets or tablets each containing a predetermined amount of the active ingredients; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g. povidone, gelatin, hydroxypropyl methylcellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycollate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent.
  • Molded tablets may be made by molding a mixture of the powdered compound moistened with an inert liquid diluent in a suitable machine.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein using, for example, cellulose ether derivatives (e.g., hydroxypropyl methylcellulose) or methacrylate derivatives in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • cellulose ether derivatives e.g., hydroxypropyl methylcellulose
  • methacrylate derivatives in varying proportions to provide the desired release profile.
  • Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredients in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylates.
  • Topical administration may also be by means of a transdermal iontophoretic device.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Formulations suitable for penile administration for prophylactic or therapeutic use may be presented in condoms, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • compositions suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories.
  • Suitable carriers include cocoa butter and other materials commonly used in the art.
  • the suppositories may be conveniently formed by admixture of the active combination with the softened or melted carrier(s) followed by chilling and shaping in moulds.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents; and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Exemplary unit dosage formulations are those containing a daily dose or daily subdose of the active ingredients, as hereinbefore recited, or an appropriate fraction thereof. It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include such further agents as sweeteners, thickeners and flavoring agents.
  • the compounds of the combination of the present invention may be obtained in a conventional manner, known to those skilled in the art.
  • Tenofovir disoproxil fumarate can be prepared, for example, as described in U.S. Pat. No. 5,977,089. Methods for the preparation of FTC are described in WO 92/14743, incorporated herein by reference.
  • compositions of the present invention are administered to a human or other mammal in a safe and effective amount as described herein. These safe and effective amounts will vary according to the type and size of mammal being treated and the desired results of the treatment. Any of the various methods known by persons skilled in the art for packaging tablets, caplets, or other solid dosage forms suitable for oral administration, that will not degrade the components of the present invention, are suitable for use in packaging. The combinations may be packaged in glass and plastic bottles. Tablets, caplets, or other solid dosage forms suitable for oral administration may be packaged and contained in various packaging materials optionally including a dessicant, e.g. silica gel. Packaging may be in the form of unit dose blister packaging.
  • a package may contain one blister tray of tenofovir DF and another blister tray of emtricitabine pills, tablets, caplets, or capsule.
  • a patient would take one dose, e.g. a pill, from one tray and one from the other.
  • the package may contain a blister tray of the co-formulated combination of tenofovir DF and emtricitabine in a single pill, tablet, caplet or capsule.
  • the combinations of the invention include physiological functional derivatives of tenofovir DF and FTC.
  • the packaging material may also have labeling and information related to the pharmaceutical composition printed thereon.
  • an article of manufacture may contain a brochure, report, notice, pamphlet, or leaflet containing product information. This form of pharmaceutical information is referred to in the pharmaceutical industry as a “package insert.”
  • a package insert may be attached to or included with a pharmaceutical article of manufacture.
  • the package insert and any article of manufacture labeling provides information relating to the pharmaceutical composition.
  • the information and labeling provides various forms of information utilized by health-care professionals and patients, describing the composition, its dosage and various other parameters required by regulatory agencies such as the United States Food and Drug Agency.
  • the combinations of the inventions may be tested for in vitro activity against HIV and sensitivity, and for cytotoxicity in laboratory adapted cell lines, e.g. MT2 and in peripheral blood mononuclear cells (PBMC) according to standard assays developed for testing anti-HIV compounds, such as WO 02/068058 and U.S. Pat. No. 6,475,491.
  • Combination assays may be performed at varying concentrations of the compounds of the combinations to determine EC 50 by serial dilutions.
  • exemplary formulations A, B, C, D, E, and F are prepared by wet granulation of the ingredients with an aqueous solution, addition of extragranular components and then followed by addition of magnesium stearate and compression.
  • Formulation A mg/tablet Tenofovir Disoproxil Fumarate 300 emtricitabine 200 Microcrystalline Cellulose 200 Lactose Monohydrate 175 Croscarmellose Sodium 60 Pregelatinized Starch 50 Colloidal silicon dioxide 5 Magnesium Stearate 10 total: 1000
  • Formulation B mg/tablet Tenofovir Disoproxil fumarate 300 emtricitabine 100 Microcrystalline Cellulose 200 Lactose Monohydrate 180 Sodium Starch Glycollate 60 Pregelatinized Starch 50 Magnesium Stearate 10 total: 900
  • Formulation C mg/tablet Tenofovir Disoproxil fumarate 200 emtricitabine 200 Microcrystalline Cellulose 200 Lactose Monohydrate 180 Sodium Starch Glycollate 60 Pregelatinized Starch 50 Magnesium Stearate 10 total: 900
  • Formulation D mg/tablet Tenofovir Disoproxil fumarate 300 emtricitabine 25 Microcrystalline Cellulose 200 Lactose Monohydrate 180 Sodium Starch Glycollate 60 Pregelatinized Starch 50 Magnesium Stearate 10 total: 825
  • Formulation E mg/tablet Tenofovir Disoproxil fumarate 200 emtricitabine 25 Microcrystalline Cellulose 200 Lactose Monohydrate 180 Sodium Starch Glycollate 60 Pregelatinized Starch 50 Magnesium Stearate 10 total: 725
  • Formulation F mg/tablet Tenofovir Disoproxil fumarate 100 emtricitabine 100 Microcrystalline Cellulose 200 Lactose Monohydrate 180 Sodium Starch Glycollate 60 Pregelatinized Starch 50 Magnesium Stearate 10 total: 700 Formulation G (Controlled Release Formulation):
  • This formulation is prepared by wet granulation of the ingredients with an aqueous solution, followed by the addition of magnesium stearate and compression.
  • a capsule formulation is prepared by admixing the ingredients and filling into a two-part hard gelatin or hydroxypropyl methylcellulose capsule.
  • mg/capsule Active Ingredient 500 Microcrystalline Cellulose 143 Sodium Starch Glycollate 25 Magnesium Stearate 2 total: 670 Formulation I (Controlled Release Capsule):
  • the following controlled release capsule formulation is prepared by extruding ingredients a, b, and c using an extruder, followed by spheronization of the extrudate and drying. The dried pellets are then coated with release-controlling membrane (d) and filled into a two-piece, hard gelatin or hydroxypropyl methylcellulose capsule.
  • mg/capsule (a) Active Ingredient 500 (b) Microcrystalline Cellulose 125 (c) Lactose B.P. 125 (d) Ethyl Cellulose 13 total: 763 Formulation J (Oral Suspension):
  • Active Ingredient 500 mg Confectioner's Sugar 2000 mg Simethicone 300 mg Methylparaben 30 mg Propylparaben 10 mg Flavor, Peach 500 mg Purified Water q.s. to 5.00 ml Formulation K (Suppository):
  • Witepsol H15 is melted in a steam-jacketed pan at 45° C. maximum.
  • the active ingredients are sifted through a 200 micron sieve and added to the molten base with mixing, using a Silverson fitted with a cutting head, until a smooth dispersion is achieved. Maintaining the mixture at 45° C., the remaining Witepsol H15 is added to the suspension and stirred to ensure a homogenous mix.
  • the entire suspension is passed through a 250 micron stainless steel screen and, with continuous stirring, is allowed to cool to 40° C. At a temperature of 38° C. to 40° C., 2.02 g of the mixture is filled into suitable, 2 ml plastic molds.
  • the suppositories are allowed to cool to room temperature.
  • TDF tenofovir disoproxil fumarate
  • Equipment included a high shear mixer equipped with a pressure tank and spray nozzle tip to add the granulating water, a fluid-bed dryer, a mill, a tumble blender, a rotary tablet press, and a tablet deduster.
  • the dried, milled powder was blended with the extragranular microcrystalline cellulose and croscarmellose sodium and then blended with magnesium stearate. Powder samples were removed after mixing with the magnesium stearate. The blend samples were evaluated for, bulk density, mesh analysis and compressibility. The powder blend mixed with the magnesium stearate was compressed into tablets on a press setup.
  • TDF/emtricitabine tablet formulation The following Table 1 lists the quantitative composition of the TDF/emtricitabine tablet formulation.
  • Moisture content was measured by loss on drying using a heat lamp/balance system.
  • the powder blend was sampled with a sampling thief fitted with chambers to determine powder blend uniformity. Duplicate samples were removed from each of several locations in the blender. Blend uniformity analysis was performed on one sample from each location.
  • Particle size analysis of the final powder blend was determined by sifting a multi-gram sample through a screen using a sonic sifter.
  • the quantity of final powder blend retained on each sieve and the fines collector was determined by calculating the difference in weight between the sieves and fines collector before and after the test.
  • the geometric mean diameter particle size was calculated by logarithmic weighting of the sieved distribution.
  • Bulk density was determined by filling a graduated cylinder with the final powder blend and measuring the weight differential between the empty and filled graduate cylinder per unit volume.
  • Tablets were characterized for friability using a friabilator, a hardness tester, a thickness micrometer equipped with a printer, and a weighing balance.
  • Compression characteristics were determined using a rotary tablet press equipped with a flat-faced, beveled edged punch to a target weight of 400 mg.
  • the powder blends were compressed using target upper punch pressures ranging from approximately 100 to 250 MPa.
  • the apparent normalized ejection force was determined and normalized for tablet thickness and diameter.
  • Tablet hardness was determined using a hardness tester. Tablet thickness was determined using a micrometer, and tablet weights were determined using a top loading balance.
  • the powders were blended in a granulator and then granulated using water.
  • the impeller and chopper speeds were kept constant in the blender at a low setting during the granulation and wet massing operations.
  • the impeller and chopper were stopped and the granulator bowl was opened to observe the granulation consistency and texture.
  • the lid was closed and the wet massing phase was performed.
  • Acceptable granules had 40% w/w and 60% w/w water, respectively.
  • each wet granulation was deagglomerated with a mill fitted with a screen and an impeller.
  • the milled wet granules were charged into a fluid-bed dryer immediately following wet milling.
  • Milled wet granules were dried using an inlet air setpoint temperature of about 70° C. and airflow of approximately 100 cfm.
  • the target LOD was about 1.0% with a range of not more than (NMT) 1.5%.
  • the total fluid-bed drying time ranged from 53 to 75 minutes.
  • Final LOD ranged from 0.4% to 0.7% for all of the batches dried.
  • the final exhaust temperatures for all the batches ranged from 47° C. to 50° C.
  • final powder blend yield available for compression after blending ranged from 10.5 kg (87.5%) to 11.1 kg (92.5%).
  • the final powder blend bulk density ranged from 0.48 to 0.58 g/cc and the geometric mean diameter particle size ranged from 112 to 221 ⁇ m. Percent water and wet massing time affect final powder blend particle size and bulk density.
  • the final powder blend moisture level ranged from 0.8% to 1.1% LOD.
  • the final blends were compressed using a rotary tablet press and the tablets were film-coated.
  • Table 2 Three 300 gm formulations (Table 2) were granulated in a granulator equipped with a 1-L bowl. The quantities of intragranular components were based on a 300 g total batch size. The formulations in lots 1 and 2 differed in the amount of microcrystalline cellulose 30% vs. 20% w/w, respectively. Lots 2 and 3 were identical except for the type of binder. Lot 2 contained 5% w/w of pregelatinized starch and lot 3 contained 5% w/w povidone as binder.
  • the final blend batch size was adjusted to 100 g.
  • a portion, 81 g of the resulting blend from Lot 1 was blended with 15 g microcrystalline cellulose, 3 g croscarmellose sodium and 1 g magnesium stearate.
  • 86 g of the resulting granulation from Lot 2 and Lot 3 were each blended with 10 g microcrystalline cellulose, 3 g croscarmellose sodium and 1 g magnesium stearate.
  • Impurities related to tenofovir disoproxil fumarate and emtricitabine were characterized and measured in the bulk API (active pharmaceutical ingredient) before formulation in the three lots of Table 2, and again after formulation in the resulting tablets.
  • the impurities include by-products from hydrolysis of the exocyclic amino groups of tenofovir disoproxil fumarate and emtricitabine, and the hydrolysis of the disoproxil (POC) esters of tenofovir disoproxil fumarate. In each lot, the sum total of impurities related to tenofovir disoproxil fumarate and emtricitabine was less than 1% after formulation and tablet manufacture.
  • tenofovir disoproxil fumarate and emtricitabine tablets were evaluated by visual appearance, water content, label strength, impurity and degradation product contents, and tablet dissolution. Stability studies were conducted on drug product packaged in container-closure systems that are identical to the intended clinical and commercial container-closure system. There was no sign of discoloration or tablet cracking during the course of the stability study. Film-coated tenofovir disoproxil fumarate and emtricitabine tablets exhibited satisfactory stability at 40° C./75% RH (relative humidity) for up to six months when packaged and stored with silica gel desiccant.
  • RH relative humidity
  • a pharmaceutical composition comprising an effective amount of a compound of the formula:
  • R 1 and R 2 are independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 6 -C 20 arylalkyl, C 6 -C 20 substituted arylalkyl, acyloxymethyl esters —CH 2 C( ⁇ O)R 9 and acyloxymethyl carbonates —CH 2 C( ⁇ O)OR 9 where R 9 is C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl and C 6 -C 20 substituted aryl;
  • R 3 is selected from H, C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, or CH 2 OR 8 where R 8 is C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl and C 1 -C 6 haloalkyl;
  • R 4 and R 5 are independently selected from H, NH 2 , NHR and NR 2 where R is C 1 -C 6 alkyl;
  • R 6 and R 7 are independently selected from H and C 1 -C 6 alkyl
  • B is selected from adenine, guanine, cytosine, uracil, thymine, 7-deazaadenine, 7-deazaguanine, 7-deaza-8-azaguanine, 7-deaza-8-azaadenine, inosine, nebularine, nitropyrrole, nitroindole, 2-aminopurine, 2-amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, pseudouridine, 5-fluorocytosine, 5-chlorocytosine, 5-bromocytosine, 5-iodocytosine, pseudocytosine, pseudoisocytosine, 5-propynylcytosine, isocytosine, isoguanine, 7-deazaguanine, 2-thiopyrimidine, 6-thioguanine, 4-thiothymine, 4-thiouracil, O 6 -methylguanine, N 6 -methyladenine, O
  • R is selected from H, C 1 -C 18 alkyl, C 1 -C 18 is substituted alkyl, C 2 -C 18 alkenyl, C 2 -C 18 substituted alkenyl, C 2 -C 18 alkynyl, C 2 -C 18 substituted alkynyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 2 -C 20 heterocycle, C 2 -C 20 substituted heterocycle, phosphonate, phosphophosphonate, diphosphophosphonate, phosphate, diphosphate, triphosphate, polyethyleneoxy or a physiologically functional derivative thereof; and
  • R 1 and R 2 are independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 6 -C 20 arylalkyl, C 6 -C 20 substituted arylalkyl, acyloxymethyl esters —CH 2 C( ⁇ O)R 9 and acyloxymethyl carbonates —CH 2 C( ⁇ O)OR 9 where R 9 is C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl and C 6 -C 20 substituted aryl; and R 3 , R 4 , R 5 , R 6 and R 7 are independently H or C 1 -C 6 alkyl.
  • R 1 and R 2 are independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl, C 6 -C 20 substituted aryl, C 6 -C 20 arylalkyl, C 6 -C 20 substituted arylalkyl, acyloxymethyl esters —CH 2 C( ⁇ O)R 9 and acyloxymethyl carbonates —CH 2 C( ⁇ O)OR 9 where R 9 is C 1 -C 6 alkyl, C 1 -C 6 substituted alkyl, C 6 -C 20 aryl and C 6 -C 20 substituted aryl; and R 3 , R 4 , R 5 , R 6 and R 7 are independently H or C 1 -C 6 alkyl; and, in formula 2, B is cytosine or a 5-halocytosine.
  • R 1 and R 2 are independently selected from H, acyloxymethyl esters —CH 2 OC(—O)R 9 and acyloxymethyl carbonates —CH 2 C( ⁇ O)OR 9 where R 9 is C 1 -C 6 alkyl; and R 3 , R 4 , R 5 , R 6 and R 7 are independently H or C 1 -C 6 alkyl; and, in formula 2, B is cytosine or a 5-halocytosine and R is H.
  • R 1 and R 2 are independently selected from H and —CH 2 C( ⁇ O)OCH(CH 3 ) 2 ; R 3 is —CH 3 ; and R 4 , R 5 , R 6 and R 7 are H; and, in formula 2, B is 5-fluorocytosine and R is H.
  • a pharmaceutical composition comprising a pharmaceutically effective amount of [2-(6-amino-purin-9-yl)-1-methyl-ethoxymethyl]-phosphonic acid diisopropoxycarbonyloxymethyl ester fumarate (tenofovir disoproxil fumarate) or a physiologically functional derivative thereof and a pharmaceutically effective amount of (2R, 5S,)-4-amino-5-fluoro-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one (emtricitabine) or a physiologically functional derivative thereof; and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation of embodiment A1 to G7 further comprising a third active ingredient selected from the group consisting of a protease inhibitor, a nucleoside or nucleotide reverse transcriptase inhibitor, a non-nucleoside reverse transcriptase inhibitor, and an integrase inhibitor.
  • a method for the treatment or prevention of the symptoms or effects of an HIV infection in an infected animal which comprises administering to said animal a pharmaceutical composition of embodiments claims A1 to I9.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US10/540,794 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy Abandoned US20060246130A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US10/540,794 US20060246130A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US12/195,161 US8592397B2 (en) 2003-01-14 2008-08-20 Compositions and methods for combination antiviral therapy
US12/204,174 US8716264B2 (en) 2003-01-14 2008-09-04 Compositions and methods for combination antiviral therapy
US14/227,653 US20140213556A1 (en) 2003-01-14 2014-03-27 Compositions and methods for combination antiviral therapy
US14/523,783 US9744181B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US14/523,758 US9457036B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US15/586,500 US20170232019A1 (en) 2003-01-14 2017-05-04 Compositions and Methods for Combination Antiviral Therapy
US15/622,484 US20170273994A1 (en) 2003-01-14 2017-06-14 Compositions and Methods for Combination Antiviral Therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US44030803P 2003-01-14 2003-01-14
US44024603P 2003-01-14 2003-01-14
PCT/US2004/000832 WO2004064845A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US10/540,794 US20060246130A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/000832 A-371-Of-International WO2004064845A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/195,161 Continuation US8592397B2 (en) 2003-01-14 2008-08-20 Compositions and methods for combination antiviral therapy
US12/204,174 Continuation US8716264B2 (en) 2003-01-14 2008-09-04 Compositions and methods for combination antiviral therapy

Publications (1)

Publication Number Publication Date
US20060246130A1 true US20060246130A1 (en) 2006-11-02

Family

ID=32776014

Family Applications (11)

Application Number Title Priority Date Filing Date
US10/757,122 Abandoned US20040224916A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US10/540,782 Abandoned US20060234982A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US10/540,794 Abandoned US20060246130A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US10/757,141 Abandoned US20040224917A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US12/195,161 Expired - Lifetime US8592397B2 (en) 2003-01-14 2008-08-20 Compositions and methods for combination antiviral therapy
US12/204,174 Expired - Lifetime US8716264B2 (en) 2003-01-14 2008-09-04 Compositions and methods for combination antiviral therapy
US14/227,653 Abandoned US20140213556A1 (en) 2003-01-14 2014-03-27 Compositions and methods for combination antiviral therapy
US14/523,758 Expired - Lifetime US9457036B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US14/523,783 Expired - Lifetime US9744181B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US15/586,500 Abandoned US20170232019A1 (en) 2003-01-14 2017-05-04 Compositions and Methods for Combination Antiviral Therapy
US15/622,484 Abandoned US20170273994A1 (en) 2003-01-14 2017-06-14 Compositions and Methods for Combination Antiviral Therapy

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/757,122 Abandoned US20040224916A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US10/540,782 Abandoned US20060234982A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy

Family Applications After (8)

Application Number Title Priority Date Filing Date
US10/757,141 Abandoned US20040224917A1 (en) 2003-01-14 2004-01-13 Compositions and methods for combination antiviral therapy
US12/195,161 Expired - Lifetime US8592397B2 (en) 2003-01-14 2008-08-20 Compositions and methods for combination antiviral therapy
US12/204,174 Expired - Lifetime US8716264B2 (en) 2003-01-14 2008-09-04 Compositions and methods for combination antiviral therapy
US14/227,653 Abandoned US20140213556A1 (en) 2003-01-14 2014-03-27 Compositions and methods for combination antiviral therapy
US14/523,758 Expired - Lifetime US9457036B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US14/523,783 Expired - Lifetime US9744181B2 (en) 2003-01-14 2014-10-24 Compositions and methods for combination antiviral therapy
US15/586,500 Abandoned US20170232019A1 (en) 2003-01-14 2017-05-04 Compositions and Methods for Combination Antiviral Therapy
US15/622,484 Abandoned US20170273994A1 (en) 2003-01-14 2017-06-14 Compositions and Methods for Combination Antiviral Therapy

Country Status (26)

Country Link
US (11) US20040224916A1 (hr)
EP (4) EP1583542B9 (hr)
JP (8) JP2006515624A (hr)
KR (3) KR100860136B1 (hr)
CN (2) CN105596356A (hr)
AP (1) AP2089A (hr)
AT (1) ATE398455T1 (hr)
AU (3) AU2004206827A1 (hr)
BR (1) BRPI0406760A (hr)
CA (2) CA2512319A1 (hr)
CY (1) CY1108355T1 (hr)
DE (1) DE602004014470D1 (hr)
DK (1) DK1583542T3 (hr)
EA (2) EA015145B1 (hr)
ES (1) ES2308136T3 (hr)
HK (1) HK1079122A1 (hr)
HR (2) HRP20050619A2 (hr)
IL (1) IL169243A (hr)
IS (1) IS7977A (hr)
MX (1) MXPA05007016A (hr)
NO (3) NO337917B1 (hr)
NZ (1) NZ540728A (hr)
PL (2) PL408254A1 (hr)
PT (1) PT1583542E (hr)
SI (1) SI1583542T1 (hr)
WO (2) WO2004064846A1 (hr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060234982A1 (en) * 2003-01-14 2006-10-19 Dahl Terrence C Compositions and methods for combination antiviral therapy
US20100055180A1 (en) * 2007-10-10 2010-03-04 Mallinckrodt Baker, Inc. Directly Compressible Granular Microcrystalline Cellulose Based Excipient, Manufacturing Process and Use Thereof
US20110092598A1 (en) * 2007-10-10 2011-04-21 Nandu Deorkar Driectly Compressible High Functionality Granular Microcrystalline Cellulose Based Excipient, Manufacturing Process and Use Thereof
WO2012154698A2 (en) * 2011-05-06 2012-11-15 Mckenna Charles E Method to improve antiviral activity of nucleotide analogue drugs
US8598185B2 (en) 2005-06-13 2013-12-03 Bristol-Myers Squibb & Gilead Sciences, Inc. Unitary pharmaceutical dosage form
US8871271B2 (en) 2005-06-13 2014-10-28 Gilead Sciences, Inc. Method and composition for pharmaceutical product
US20160199379A1 (en) * 2010-01-27 2016-07-14 Viiv Healthcare Company Antiviral therapy

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8771733B2 (en) 2004-12-03 2014-07-08 Merck Sharp & Dohme Corp. Pharmaceutical composition containing an anti-nucleating agent
EP1868628B1 (en) * 2005-04-08 2014-06-11 Chimerix, Inc. Compounds, compositions and methods for the treatment of poxvirus infections
EP1865967A4 (en) * 2005-04-08 2011-02-09 Chimerix Inc COMPOUNDS, COMPOSITIONS AND METHODS FOR TREATING VIRAL INFECTIONS AND OTHER DISEASES
US8000981B2 (en) 2005-11-30 2011-08-16 The Invention Science Fund I, Llc Methods and systems related to receiving nutraceutical associated information
US10296720B2 (en) 2005-11-30 2019-05-21 Gearbox Llc Computational systems and methods related to nutraceuticals
US7974856B2 (en) 2005-11-30 2011-07-05 The Invention Science Fund I, Llc Computational systems and methods related to nutraceuticals
US8340944B2 (en) 2005-11-30 2012-12-25 The Invention Science Fund I, Llc Computational and/or control systems and methods related to nutraceutical agent selection and dosing
US8068991B2 (en) 2005-11-30 2011-11-29 The Invention Science Fund I, Llc Systems and methods for transmitting pathogen related information and responding
US8297028B2 (en) 2006-06-14 2012-10-30 The Invention Science Fund I, Llc Individualized pharmaceutical selection and packaging
US7827042B2 (en) 2005-11-30 2010-11-02 The Invention Science Fund I, Inc Methods and systems related to transmission of nutraceutical associated information
US7927787B2 (en) 2006-06-28 2011-04-19 The Invention Science Fund I, Llc Methods and systems for analysis of nutraceutical associated components
ATE474560T1 (de) * 2005-12-14 2010-08-15 Cipla Ltd Pharmazeutische kombination aus nucleotid und nucleosid-reverse-transkriptase-hemmern (wie tenofovir und lamivudin) in verschiedenen teilen der dosiereinheit
US9044509B2 (en) 2006-02-03 2015-06-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Inhibition of HIV infection through chemoprophylaxis
EP2049506B2 (en) 2006-07-07 2024-05-08 Gilead Sciences, Inc. Modulators of pharmacokinetic properties of therapeutics
RU2470846C2 (ru) * 2007-01-20 2012-12-27 Мерк Патент Гмбх Повторно закрываемый контейнер для упаковки мягких капсул
WO2008096369A2 (en) * 2007-02-05 2008-08-14 Matrix Laboratories Limited Pharmaceutical formulation for use in hiv therapy
US20100196273A1 (en) * 2007-02-12 2010-08-05 Board Of Regents, University Of Texas System Novel agent for in vivo pet imaging of tumor proliferation
PT2487166T (pt) 2007-02-23 2016-10-17 Gilead Sciences Inc Moduladores de propriedades farmacocinéticas de agentes terapêuticos
WO2009091586A2 (en) 2008-01-18 2009-07-23 North Carolina State University Peptides and methods of use as therapeutics and screening agents
CA2713105C (en) 2008-01-25 2016-06-07 Chimerix, Inc. Methods of treating viral infections
US7935817B2 (en) * 2008-03-31 2011-05-03 Apotex Pharmachem Inc. Salt form and cocrystals of adefovir dipivoxil and processes for preparation thereof
SI2296633T1 (sl) 2008-05-02 2015-11-30 Gilead Sciences, Inc. Uporaba trdnih nosilnih delcev za izboljšanje predelovalnih sposobnosti farmacevtskega sredstva
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
EP2376514A2 (en) 2008-12-23 2011-10-19 Pharmasset, Inc. Nucleoside analogs
PA8855801A1 (es) 2008-12-23 2010-07-27 Sintesis de nucleosidos de purina
AR074897A1 (es) 2008-12-23 2011-02-23 Pharmasset Inc Fosforamidatos de nucleosidos
HUE025822T2 (en) 2009-02-06 2016-04-28 Gilead Sciences Inc Dual layer tablets containing Elvitegravir, Cobicistat, Emtricitabine and Tenofovir
TWI583692B (zh) 2009-05-20 2017-05-21 基利法瑪席特有限責任公司 核苷磷醯胺
WO2011011519A1 (en) 2009-07-21 2011-01-27 Chimerix, Inc. Compounds, compositions and methods for treating ocular conditions
AU2011216243B2 (en) 2010-02-12 2015-07-09 Chimerix, Inc. Nucleoside phosphonate salts
PL3290428T3 (pl) 2010-03-31 2022-02-07 Gilead Pharmasset Llc Tabletka zawierająca krystaliczny (S)-2-(((S)-(((2R,3R,4R,5R)-5-(2,4-diokso-3,4-dihydropirymidyn-1(2H)-ylo)-4-fluoro-3-hydroksy-4-metylotetrahydrofuran-2-ylo)metoksy)(fenoksy)fosforylo)amino)propanian izopropylu
US8859756B2 (en) 2010-03-31 2014-10-14 Gilead Pharmasset Llc Stereoselective synthesis of phosphorus containing actives
UY33312A (es) 2010-03-31 2011-10-31 Pharmasset Inc Fosforamidato de nucleosido de purina
EP2563367A4 (en) 2010-04-26 2013-12-04 Chimerix Inc METHODS OF TREATING RETROVIRAL INFECTIONS AND ASSOCIATED DOSAGE REGIMES
EP2389929A1 (en) * 2010-05-30 2011-11-30 Abdi Ibrahim Ilac Sanayi ve Ticaret Anonim Sirketi Pharmaceutical formulations of tenofovir
CN114010776A (zh) 2010-06-09 2022-02-08 疫苗技术股份有限公司 用于增强抗逆转录病毒治疗的hiv感染者的治疗性免疫
KR20130095735A (ko) * 2010-08-01 2013-08-28 쟝수 치아타이 티안큉 파마수티컬 주식회사 테노포비어 디소프록실 푸마레이트의 결정
CN103491948B (zh) 2010-11-19 2016-11-02 吉利德科学公司 包含利匹韦林HCl和富马酸替诺福韦酯的治疗组合物
ES2716158T3 (es) 2010-11-30 2019-06-10 Gilead Pharmasset Llc 2'-spiro-nucleótidos para el tratamiento de hepatitis C
CN104185420B (zh) 2011-11-30 2017-06-09 埃默里大学 用于治疗或预防逆转录病毒和其它病毒感染的抗病毒jak抑制剂
EP2794624B1 (en) 2011-12-22 2019-05-15 Geron Corporation Guanine analogs as telomerase substrates and telomere length affectors
WO2013115916A1 (en) * 2012-02-03 2013-08-08 Gilead Sciences, Inc. Combination therapy comprising gs-7340 and cobicistat for use in the treatment of viral infections
EA026138B1 (ru) 2012-02-03 2017-03-31 Джилид Сайэнс, Инк. Комбинированная терапия, включающая тенофовир алафенамида гемифумарат и кобицистат, для применения для лечения вирусных инфекций
CN107312039B (zh) 2012-08-30 2019-06-25 江苏豪森药业集团有限公司 一种替诺福韦前药的制备方法
WO2014068265A1 (en) * 2012-10-29 2014-05-08 Cipla Limited Antiviral phosphonate analogues and process for preparation thereof
CA3012242C (en) 2012-12-21 2021-11-02 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
CN103127028A (zh) * 2013-03-14 2013-06-05 南京恒道医药科技有限公司 一种含有富马酸替诺福韦二吡呋酯的胶囊剂
AU2014286993B2 (en) 2013-07-12 2018-10-25 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their use for the treatment of HIV infections
NO2865735T3 (hr) 2013-07-12 2018-07-21
WO2015028875A2 (en) * 2013-08-29 2015-03-05 Teva Pharmaceuticals Industries Ltd. Unit dosage form comprising emtricitabine, tenofovir, darunavir and ritonavir and a monolithic tablet comprising darunavir and ritonavir
CZ2013985A3 (cs) * 2013-12-09 2015-06-17 Zentiva, K.S. Stabilní farmaceutická kompozice obsahující tenofovir disoproxil fumarát
TWI660965B (zh) 2014-01-15 2019-06-01 美商基利科學股份有限公司 泰諾福韋之固體形式
AU2015245217A1 (en) 2014-04-08 2016-10-13 Teva Pharmaceutical Industries Ltd. Unit dosage form comprising Emtricitabine, Tenofovir, Darunavir and Ritonavir
TW201613936A (en) 2014-06-20 2016-04-16 Gilead Sciences Inc Crystalline forms of(2R,5S,13aR)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
NO2717902T3 (hr) 2014-06-20 2018-06-23
TWI744723B (zh) 2014-06-20 2021-11-01 美商基利科學股份有限公司 多環型胺甲醯基吡啶酮化合物之合成
CN105399771B (zh) * 2014-07-21 2020-11-24 江苏豪森药业集团有限公司 替诺福韦前药晶型及其制备方法和用途
TWI695003B (zh) 2014-12-23 2020-06-01 美商基利科學股份有限公司 多環胺甲醯基吡啶酮化合物及其醫藥用途
EP3236972B1 (en) 2014-12-26 2021-07-28 Emory University Anti-viral n4-hydroxycytidine derivatives
EP3273946A1 (en) * 2015-03-27 2018-01-31 F. Hoffmann-La Roche AG Pharmaceutical formulation comprising sembragiline
EP3277691B1 (en) 2015-04-02 2019-01-30 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
EP4233846A3 (en) * 2015-06-30 2023-10-04 Gilead Sciences, Inc. Pharmaceutical formulations
TWI620754B (zh) * 2015-08-26 2018-04-11 Method for preparing amino phosphate derivative and preparation method thereof
MX2018005729A (es) 2015-11-09 2018-08-09 Gilead Sciences Inc Composiciones terapeuticas para tratamiento del virus de inmunodeficiencia humana.
WO2017106069A1 (en) 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Antiviral oxime phosphoramide compounds
EP3454862A4 (en) 2016-05-10 2020-02-12 C4 Therapeutics, Inc. SPIROCYCLIC DEGRONIMERS FOR TARGET PROTEIN REDUCTION
CN109641874A (zh) 2016-05-10 2019-04-16 C4医药公司 用于靶蛋白降解的c3-碳连接的戊二酰亚胺降解决定子体
CN109562107A (zh) 2016-05-10 2019-04-02 C4医药公司 用于靶蛋白降解的杂环降解决定子体
US10449208B2 (en) 2016-08-25 2019-10-22 Merck Sharp & Dohme Corp. Antiviral prodrugs of tenofovir
US10736908B2 (en) 2016-10-26 2020-08-11 Merck Sharp & Dohme Corp. Antiviral aryl-amide phosphodiamide compounds
TR201617448A2 (tr) * 2016-11-29 2018-06-21 Arven Ilac Sanayi Ve Ticaret Anonim Sirketi Tenofovi̇r ve emtri̇si̇tabi̇n i̇çeren kati oral farmasöti̇k bi̇leşi̇mler
JP2020504734A (ja) 2016-12-22 2020-02-13 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. 抗ウイルス性ベンジル−アミンホスホジアミド化合物
CN106749254B (zh) * 2017-01-10 2018-05-25 青岛科技大学 一种6-氨基嘌呤乙基萘乙酸酯类化合物及其作为植物生长调节剂的用途
WO2018153977A1 (en) * 2017-02-24 2018-08-30 Hexal Ag Stable composition of tenofovir alafenamide
GB201705087D0 (en) * 2017-03-30 2017-05-17 Univ Liverpool Method for producing a liquid composition
BR112019021918A2 (pt) * 2017-04-18 2020-05-26 Cipla Limited Terapia de combinação para uso no tratamento de infecções retroviriais
RU2662160C9 (ru) 2017-07-03 2018-10-22 Александрович Иващенко Андрей Комбинированный лекарственный препарат для терапии вирусных инфекций
KR102077060B1 (ko) * 2017-07-14 2020-02-13 주식회사 종근당 테노포비어 디소프록실 아스파르트산 염을 포함하는 약제학적 조성물
RU2666727C1 (ru) * 2017-07-18 2018-09-12 Андрей Александрович Иващенко Ингибитор вируса гепатита В (ВГВ)
TR201713954A2 (tr) 2017-09-20 2019-04-22 Sanovel Ilac Sanayi Ve Ticaret Anonim Sirketi Tenofovir, emtrisitabin ve efavirenzin farmasötik kombinasyonlari
CA3082191C (en) 2017-12-07 2021-09-21 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11826375B2 (en) 2018-07-19 2023-11-28 Merck Sharp & Dohme Llc Phosphinic amide prodrugs of tenofovir
RU2726210C2 (ru) * 2018-12-27 2020-07-09 Общество С Ограниченной Ответственностью "Пролонгированные Лекарства" Комбинация противовирусных средств, набор и способ лечения на ее основе
CN110261631A (zh) * 2019-07-26 2019-09-20 重庆德方信息技术有限公司 用于健康检测装置的滴液机构
CN110251476B (zh) * 2019-08-01 2022-08-09 海思科制药(眉山)有限公司 一种恩曲他滨替诺福韦药物组合物
CN113880898B (zh) * 2020-10-30 2023-07-25 杭州拉林智能科技有限公司 黄酮苷-有机胺类抗微生物剂复盐化合物及其制备方法和应用

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4724233A (en) * 1985-04-25 1988-02-09 Stichting Rega Vzw Therapeutical application of phosphonylmethoxyalkyl adenines
US4808716A (en) * 1985-04-25 1989-02-28 Ceskoslovenska Akademic Ved 9-(phosponylmethoxyalkyl) adenines, the method of preparation and utilization thereof
US4816570A (en) * 1982-11-30 1989-03-28 The Board Of Regents Of The University Of Texas System Biologically reversible phosphate and phosphonate protective groups
US4968788A (en) * 1986-04-04 1990-11-06 Board Of Regents, The University Of Texas System Biologically reversible phosphate and phosphonate protective gruops
US5047407A (en) * 1989-02-08 1991-09-10 Iaf Biochem International, Inc. 2-substituted-5-substituted-1,3-oxathiolanes with antiviral properties
US5151426A (en) * 1989-02-08 1992-09-29 Biochem Pharma Inc. 2-substituted-5-substituted-1,3-oxathiolanes with antiviral properties
US5179104A (en) * 1990-12-05 1993-01-12 University Of Georgia Research Foundation, Inc. Process for the preparation of enantiomerically pure β-D-(-)-dioxolane-nucleosides
US5204466A (en) * 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US5486806A (en) * 1994-11-09 1996-01-23 Firari; Harold A. Anti-hijacking and theft prevention device for motor vehicles
US5486520A (en) * 1991-05-20 1996-01-23 Biochempharma, Inc. 1,3-oxathiolanes useful in the treatment of hepatitis
US5538975A (en) * 1991-08-01 1996-07-23 Biochem Pharma, Inc. 1,3-oxathiolane nucleoside compounds and compositions
US5587480A (en) * 1990-11-13 1996-12-24 Biochem Pharma, Inc. Substituted 1,3-oxathiolanes and substituted 1,3-dithiolanes with antiviral properties
US5627186A (en) * 1991-05-16 1997-05-06 Glaxo Group Limited Antiviral combinations
US5663159A (en) * 1990-09-14 1997-09-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Prodrugs of phosphonates
US5696254A (en) * 1991-05-21 1997-12-09 Biochem Pharma Inc. Processes for the diastereoselective synthesis of nucleoside analogues
US5733788A (en) * 1996-07-26 1998-03-31 Gilead Sciences, Inc. PMPA preparation
US5763606A (en) * 1993-06-07 1998-06-09 Biochem Pharma, Inc. Stereoselective synthesis of nucleoside analogues using bicyclic intermediate
US5905082A (en) * 1991-06-03 1999-05-18 Glaxo Group Limited Crystalline oxathiolane derivatives
US5914331A (en) * 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
US5922695A (en) * 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
US5935946A (en) * 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
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
US6113920A (en) * 1996-10-31 2000-09-05 Glaxo Wellcome Inc. Pharmaceutical compositions
US6312662B1 (en) * 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds

Family Cites Families (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US568164A (en) 1896-09-22 Motor attachment for bicycles
CH389608A (de) 1960-01-19 1965-03-31 Sandoz Ag Verfahren zur Herstellung von neuen Äthern
US3524846A (en) * 1967-06-02 1970-08-18 Syntex Corp Process for the didealkylation of phosphonate esters
US3622677A (en) 1969-07-07 1971-11-23 Staley Mfg Co A E Compressed tablets containing compacted starch as binder-disintegrant ingredient
CH531000A (de) * 1970-03-11 1972-11-30 Sandoz Ag Verfahren zur Herstellung neuer Benzocycloheptathiophene
US3994974A (en) * 1972-02-05 1976-11-30 Yamanouchi Pharmaceutical Co., Ltd. α-Aminomethylbenzyl alcohol derivatives
US4003878A (en) * 1972-12-07 1977-01-18 Avtex Fibers Inc. Method of preparing an alkali-metal salt of an alkoxysulfonated benzoic acid glycol ester
GB1523865A (en) 1974-09-02 1978-09-06 Wellcome Found Purine compunds and salts thereof
DE2645710C2 (de) * 1976-10-09 1985-06-27 Merck Patent Gmbh, 6100 Darmstadt Phenoxy-amino-propanole, Verfahren zu ihrer Herstellung und pharmazeutische Zubereitung
US4384005A (en) 1980-09-26 1983-05-17 General Foods Corporation Non-friable, readily-soluble, compressed tablets and process for preparing same
EP0054512A3 (de) 1980-12-12 1983-08-03 Ciba-Geigy Ag Cephalosporinester, Verfahren zu ihrer Herstellung und sie enthaltende pharmazeutische Präparate
US4355032B2 (en) * 1981-05-21 1990-10-30 9-(1,3-dihydroxy-2-propoxymethyl)guanine as antiviral agent
JPS5879983A (ja) * 1981-11-06 1983-05-13 Kanebo Ltd 新規なベンズイミダゾ−ル誘導体、その製造法およびその医薬組成物
US4476248A (en) * 1983-02-28 1984-10-09 The Upjohn Company Crystallization of ibuprofen
DE3485225D1 (de) * 1983-08-18 1991-12-05 Beecham Group Plc Antivirale guanin-derivate.
US5155268A (en) * 1984-05-04 1992-10-13 The Upjohn Company Antiarrhythmic N-aminoalkylene alkyl and aryl sulfonamides
EP0182024B1 (en) 1984-09-20 1991-04-03 Beecham Group Plc Purine derivatives and their pharmaceutical use
GB8607684D0 (en) * 1986-03-27 1986-04-30 Ici America Inc Thiazepine compounds
CS264222B1 (en) * 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
IL84477A (en) 1986-11-18 1995-12-08 Bristol Myers Squibb Co History of Phosphonomethoxyalkylene Purinopyrimidine and Pharmaceutical Preparations Containing Them
EP0309491A1 (en) 1987-01-20 1989-04-05 Sri International Antiviral agents
GB8719367D0 (en) * 1987-08-15 1987-09-23 Wellcome Found Therapeutic compounds
ZA885709B (en) 1987-08-19 1989-04-26 Fujisawa Pharmaceutical Co Novel crystalline 7-(2-(2-aminothiazol-4-yl)-2-hydroxyiminoacetamido)-3-vinyl-3-cephem-4-carboxylic acid(syn isomer)
US5684164A (en) 1988-04-11 1997-11-04 Biochem Pharma Inc. Processes for preparing substituted 1,3-oxathiolanes with antiviral properties
US5466806A (en) 1989-02-08 1995-11-14 Biochem Pharma Inc. Processes for preparing substituted 1,3-oxathiolanes with antiviral properties
US5453503A (en) 1988-10-11 1995-09-26 Eli Lilly And Company Azetidinone intermediates to carbacephalosporins and process
CA2001715C (en) 1988-11-14 1999-12-28 Muzammil M. Mansuri Carbocyclic nucleosides and nucleotides
US6703396B1 (en) 1990-02-01 2004-03-09 Emory University Method of resolution and antiviral activity of 1,3-oxathiolane nuclesoside enantiomers
US6346627B1 (en) 1990-02-01 2002-02-12 Emory University Intermediates in the synthesis of 1,3-oxathiolane nucleoside enantiomers
GB9009861D0 (en) 1990-05-02 1990-06-27 Glaxo Group Ltd Chemical compounds
AU651835B2 (en) 1990-06-13 1994-08-04 Arnold Glazier Phosphorous prodrugs
US5177064A (en) * 1990-07-13 1993-01-05 University Of Florida Targeted drug delivery via phosphonate derivatives
EP0539487A1 (en) 1990-07-19 1993-05-05 Beecham Group Plc Antiviral phosphono-alken derivatives of purines
WO1992002511A1 (en) * 1990-08-10 1992-02-20 Bristol-Myers Squibb Company Novel process for the preparation of nucleotides
CA2054126A1 (en) 1990-10-26 1992-04-27 Michiyuki Sendai Cephem compounds, their production and use
US5208221A (en) * 1990-11-29 1993-05-04 Bristol-Myers Squibb Company Antiviral (phosphonomethoxy) methoxy purine/pyrimidine derivatives
GB9026164D0 (en) 1990-12-01 1991-01-16 Beecham Group Plc Pharmaceuticals
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
NZ241625A (en) 1991-02-22 1996-03-26 Univ Emory 1,3-oxathiolane derivatives, anti-viral compositions containing such and method of resolving racemic mixture of enantiomers
US6812233B1 (en) 1991-03-06 2004-11-02 Emory University Therapeutic nucleosides
US6177435B1 (en) 1992-05-13 2001-01-23 Glaxo Wellcome Inc. Therapeutic combinations
US5480875A (en) 1992-06-23 1996-01-02 Yamanouchi Pharmaceutical Co., Ltd. Crystal of monohydrate of heterocyclic bis(phosphonic acid) derivative
US5532225A (en) 1992-07-31 1996-07-02 Sri International Acyclic purine phosphonate nucleotide analogs as antiviral agents, and related synthetic methods
US5432172A (en) 1992-08-03 1995-07-11 The Research Foundation Of State University Of New York Biological applications of alkaloids derived from the tunicate Eudistoma sp.
US5519021A (en) 1992-08-07 1996-05-21 Merck & Co., Inc. Benzoxazinones as inhibitors of HIV reverse transcriptase
NZ248852A (en) 1992-10-28 1997-06-24 Squibb & Sons Inc Alpha-phosphono and phosphinosulphonates and medicaments thereof
JPH08506343A (ja) * 1993-02-03 1996-07-09 ジェンシア・インコーポレイテッド リキソフラノシル誘導体を含むアデノシンキナーゼ阻害物質
US5514798A (en) * 1993-06-02 1996-05-07 Gilead Sciences, Inc. Method and cyclic carbonates for nucleotide analogues
DE69426904T2 (de) 1993-06-29 2001-10-11 Mitsubishi Chem Corp Phosphonat-Nukleotid Ester-Derivate
JPH09506333A (ja) 1993-09-17 1997-06-24 ギリアード サイエンシーズ,インコーポレイテッド 治療化合物の投薬方法
US5798340A (en) * 1993-09-17 1998-08-25 Gilead Sciences, Inc. Nucleotide analogs
AU691527B2 (en) 1993-09-17 1998-05-21 Gilead Sciences, Inc. Nucleotide analogs
WO1995032957A1 (en) 1994-05-27 1995-12-07 Astra Aktiebolag Novel ethoxycarbonyloxymethyl derivatives of substituted benzimidazoles
US5514557A (en) 1994-06-06 1996-05-07 Genetic Testing Institute Inc. Method and kit for detecting antibodies specific for HLA and/or platelet glycoproteins
PE32296A1 (es) 1994-07-28 1996-08-07 Hoffmann La Roche Ester de l-monovalina derivado de 2-(2-amino-1,6-dihidro-6-oxo-purin-9-il) metoxi-1,3-propandiol y sus sales farmaceuticamente aceptables
US5512596A (en) * 1994-09-02 1996-04-30 Gilead Sciences, Inc. Aromatic compounds
US5684018A (en) 1994-12-13 1997-11-04 Merck & Co., Inc. Acyloxyisopropyl carbamates as prodrugs for amine drugs
MY115461A (en) 1995-03-30 2003-06-30 Wellcome Found Synergistic combinations of zidovudine, 1592u89 and 3tc
US5618964A (en) * 1995-06-07 1997-04-08 Bristol-Myers Squibb Company Prodrug esters of phosphonosulfonate squalene synthetase inhibitors and method
KR100571215B1 (ko) 1995-06-07 2006-10-24 트라이머리스 인코퍼레이티드 복합적인치료요법을이용한hiv와다른바이러스감염의치료
CA2261619C (en) 1996-07-26 2006-05-23 Gilead Sciences, Inc. Nucleotide analogs
US6121315A (en) * 1996-09-20 2000-09-19 Warner-Lambert Company Oral compositions containing a zinc compound
GB9622681D0 (en) 1996-10-31 1997-01-08 Glaxo Group Ltd Pharmaceutical compositions
US5965729A (en) 1997-02-05 1999-10-12 Merck & Co., Inc. Process for the crystallization of a reverse transcriptase inhibitor using an anti-solvent
US6143877A (en) 1997-04-30 2000-11-07 Epoch Pharmaceuticals, Inc. Oligonucleotides including pyrazolo[3,4-D]pyrimidine bases, bound in double stranded nucleic acids
ZA986614B (en) 1997-07-25 1999-01-27 Gilead Sciences Nucleotide analog composition
CN1900089A (zh) * 1997-07-25 2007-01-24 吉尔利德科学股份有限公司 核苷酸类似物组合物
US6270957B1 (en) * 1997-08-26 2001-08-07 Wisconsin Alumni Research Foundation Non-Imuunosuppressive cyclosporins and their use in the prevention and treatment of HIV infection
CO4970782A1 (es) * 1997-11-13 2000-11-07 Merck & Co Inc Terapia combinada para el tratamiento del sida
US6087383A (en) * 1998-01-20 2000-07-11 Bristol-Myers Squibb Company Bisulfate salt of HIV protease inhibitor
US6127121A (en) * 1998-04-03 2000-10-03 Epoch Pharmaceuticals, Inc. Oligonucleotides containing pyrazolo[3,4-D]pyrimidines for hybridization and mismatch discrimination
UA72207C2 (uk) * 1998-04-07 2005-02-15 Брістол- Майєрс Сквібб Фарма Компані Фармацевтична композиція ефавіренцу з добавкою дезінтегруючих агентів у вигляді швидкорозчинних капсул або таблеток та спосіб її виготовлення
US20020072493A1 (en) * 1998-05-19 2002-06-13 Yeda Research And Development Co. Ltd. Activated T cells, nervous system-specific antigens and their uses
EP1332757B1 (en) 1998-05-27 2012-06-13 Merck Sharp & Dohme Corp. Efavirenz compressed tablet formulation
PE20000559A1 (es) 1998-05-27 2000-07-05 Merck & Co Inc Formulacion de tabletas comprimidas de efavirenz
US20010014352A1 (en) * 1998-05-27 2001-08-16 Udit Batra Compressed tablet formulation
US6194391B1 (en) 1998-06-24 2001-02-27 Emory University 3′-azido-2′,3′-dideoxyuridine administration to treat HIV and related test protocol
ES2537540T3 (es) 1998-08-12 2015-06-09 Gilead Sciences, Inc. Método de fabricación de nucleósidos de 1,3-dioxolano y 1,3-oxatiolano
GB9820420D0 (en) 1998-09-18 1998-11-11 Glaxo Group Ltd Antiviral combinations
IL142910A0 (en) * 1998-11-02 2002-04-21 Triangle Pharmaceuticals Inc Combination therapy to treat hepatitis b virus
GB9909154D0 (en) 1999-04-22 1999-06-16 Nippon Glaxo Limited Pharmaceutical formulation
US6660845B1 (en) 1999-11-23 2003-12-09 Epoch Biosciences, Inc. Non-aggregating, non-quenching oligomers comprising nucleotide analogues; methods of synthesis and use thereof
ES2250363T3 (es) 2000-02-02 2006-04-16 Dorian Bevec Guanilhidrazonas aromaticas farmaceuticamente activas.
RS51561B (sr) * 2000-02-29 2011-08-31 Bristol-Myers Squibb Co. Formulacija male doze entekavira i upotreba
CA2416757C (en) * 2000-07-21 2011-02-15 Gilead Sciences, Inc. Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
KR20040040402A (ko) 2000-12-15 2004-05-12 트라이앵글 파마슈티칼스 인코포레이티드 이노신 모노포스페이트 데하이드로게나제 억제제와dapd의 병행 요법
US6900315B2 (en) 2001-02-06 2005-05-31 Yale University 2-amino-9H-purin-9-yl compounds and methods for inhibiting/treating HIV infections and AIDS related symptoms
AU2002335489B2 (en) 2001-03-01 2008-06-05 Abbott Laboratories Polymorphic and other crystalline forms of cis-FTC
MY169670A (en) 2003-09-03 2019-05-08 Tibotec Pharm Ltd Combinations of a pyrimidine containing nnrti with rt inhibitors
AU2002359518A1 (en) 2001-11-27 2003-06-10 Bristol-Myers Squibb Company Efavirenz tablet formulation having unique biopharmaceutical characteristics
KR100851770B1 (ko) 2002-01-16 2008-08-13 베링거 잉겔하임 파르마 게엠베하 운트 코 카게 텔미사르탄과 이뇨제를 포함하는 이중층 약제학적 정제 및이의 제조방법
WO2003061602A2 (en) * 2002-01-24 2003-07-31 Sangstat Medical Corporation Combination therapy for treatment of hiv infection
US20070010489A1 (en) 2002-04-26 2007-01-11 Arimilli Murty N Cellular accumulation of phosphonate analogs of hiv protease inhibitor compounds
MXPA05003637A (es) 2002-12-09 2005-08-16 Univ Georgia Res Found Dioxolan timina y combinaciones para utilizarse contra cepas de vih resistentes.
US7332183B2 (en) * 2002-12-26 2008-02-19 Pozen Inc. Multilayer dosage forms containing NSAIDs and triptans
US20040224916A1 (en) * 2003-01-14 2004-11-11 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
BRPI0411039A (pt) * 2003-06-06 2006-07-11 Ethypharm Sa comprimido orodispersìvel multicamada
CN101142229A (zh) 2005-01-18 2008-03-12 阿德拉·N·沙克 通过与KAP相互作用而用于细胞周期调节的HTm4
TWI471145B (zh) * 2005-06-13 2015-02-01 Bristol Myers Squibb & Gilead Sciences Llc 單一式藥學劑量型
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
US8232278B2 (en) 2005-06-24 2012-07-31 Gilead Sciences, Inc. Pyrido(3,2-D)pyrimidines and pharmaceutical compositions useful for treating hepatitis C
ES2438019T3 (es) * 2005-07-22 2014-01-15 Cytodyn, Inc. Procedimientos para reducir la carga viral en pacientes infectados por el VIH-1
US7448494B2 (en) * 2005-08-10 2008-11-11 Certain Teed Corporation Loose fill insulation packaged with additive
ATE474560T1 (de) 2005-12-14 2010-08-15 Cipla Ltd Pharmazeutische kombination aus nucleotid und nucleosid-reverse-transkriptase-hemmern (wie tenofovir und lamivudin) in verschiedenen teilen der dosiereinheit
GB0525898D0 (en) 2005-12-20 2006-02-01 Pharmo Bioscience As Screening compounds for activity in modulating chloride ion transport
EP2120940A2 (en) 2007-01-16 2009-11-25 Protelogiecs, Ltd. Methods for enhancing the therapeutic efficacy of topoisomerase inhibitors
US20110275679A1 (en) 2008-08-06 2011-11-10 Bionevia Pharmaceuticals, Inc. Flupirtine hydrochloride maleic acid cocrystal
WO2012003413A1 (en) 2010-06-30 2012-01-05 The Broad Institute, Inc. Novel solid forms of tacedinaline
CN103491948B (zh) 2010-11-19 2016-11-02 吉利德科学公司 包含利匹韦林HCl和富马酸替诺福韦酯的治疗组合物

Patent Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816570A (en) * 1982-11-30 1989-03-28 The Board Of Regents Of The University Of Texas System Biologically reversible phosphate and phosphonate protective groups
US4724233A (en) * 1985-04-25 1988-02-09 Stichting Rega Vzw Therapeutical application of phosphonylmethoxyalkyl adenines
US4808716A (en) * 1985-04-25 1989-02-28 Ceskoslovenska Akademic Ved 9-(phosponylmethoxyalkyl) adenines, the method of preparation and utilization thereof
US4968788A (en) * 1986-04-04 1990-11-06 Board Of Regents, The University Of Texas System Biologically reversible phosphate and phosphonate protective gruops
US5047407A (en) * 1989-02-08 1991-09-10 Iaf Biochem International, Inc. 2-substituted-5-substituted-1,3-oxathiolanes with antiviral properties
US5151426A (en) * 1989-02-08 1992-09-29 Biochem Pharma Inc. 2-substituted-5-substituted-1,3-oxathiolanes with antiviral properties
US5210085A (en) * 1990-02-01 1993-05-11 Emory University Method for the synthesis, compositions and use of 2'-deoxy-5-fluoro-3'-thiacytidine and related compounds
US5204466A (en) * 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US6114343A (en) * 1990-02-01 2000-09-05 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-flurocytosin-1-yl)-1,3-oxathiolane
US5914331A (en) * 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
US5814639A (en) * 1990-02-01 1998-09-29 Emory University Method for the synthesis, compositions and use of 2'-deoxy-5-fluoro-3'-thiacytidine and related compounds
US5792756A (en) * 1990-09-14 1998-08-11 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Prodrugs of phosphonates
US5663159A (en) * 1990-09-14 1997-09-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Prodrugs of phosphonates
US5587480A (en) * 1990-11-13 1996-12-24 Biochem Pharma, Inc. Substituted 1,3-oxathiolanes and substituted 1,3-dithiolanes with antiviral properties
US5179104A (en) * 1990-12-05 1993-01-12 University Of Georgia Research Foundation, Inc. Process for the preparation of enantiomerically pure β-D-(-)-dioxolane-nucleosides
US5859021A (en) * 1991-05-16 1999-01-12 Glaxo Group Limited Antiviral combinations
US5627186A (en) * 1991-05-16 1997-05-06 Glaxo Group Limited Antiviral combinations
US5486520A (en) * 1991-05-20 1996-01-23 Biochempharma, Inc. 1,3-oxathiolanes useful in the treatment of hepatitis
US5696254A (en) * 1991-05-21 1997-12-09 Biochem Pharma Inc. Processes for the diastereoselective synthesis of nucleoside analogues
US5744596A (en) * 1991-05-21 1998-04-28 Biochem Pharma Inc. Nucleoside analogues and synthetic intermediates
US5756706A (en) * 1991-05-21 1998-05-26 Biochem Pharma Inc. Processes for the diastereoselective synthesis of nucleoside analogues
US5905082A (en) * 1991-06-03 1999-05-18 Glaxo Group Limited Crystalline oxathiolane derivatives
US5618820A (en) * 1991-08-01 1997-04-08 Biochem Pharma, Inc. 1,3-oxathiolane nucleoside analogues and methods for using same
US5538975A (en) * 1991-08-01 1996-07-23 Biochem Pharma, Inc. 1,3-oxathiolane nucleoside compounds and compositions
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
US5763606A (en) * 1993-06-07 1998-06-09 Biochem Pharma, Inc. Stereoselective synthesis of nucleoside analogues using bicyclic intermediate
US5486806A (en) * 1994-11-09 1996-01-23 Firari; Harold A. Anti-hijacking and theft prevention device for motor vehicles
US6069249A (en) * 1996-07-26 2000-05-30 Gilead Sciences, Inc. Antiviral phosphonomethoxy nucleotide analogs having increased oral bioavailability
US5977089A (en) * 1996-07-26 1999-11-02 Gilead Sciences, Inc. Antiviral phosphonomethoxy nucleotide analogs having increased oral bioavailability
US6043230A (en) * 1996-07-26 2000-03-28 Gilead Sciences, Inc. Antiviral phosphonomethoxy nucleotide analogs having increased oral bioavailability
US5922695A (en) * 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
US5733788A (en) * 1996-07-26 1998-03-31 Gilead Sciences, Inc. PMPA preparation
US6113920A (en) * 1996-10-31 2000-09-05 Glaxo Wellcome Inc. Pharmaceutical compositions
US5935946A (en) * 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
US6312662B1 (en) * 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9457036B2 (en) 2003-01-14 2016-10-04 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US20060234982A1 (en) * 2003-01-14 2006-10-19 Dahl Terrence C Compositions and methods for combination antiviral therapy
US8716264B2 (en) 2003-01-14 2014-05-06 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US9744181B2 (en) 2003-01-14 2017-08-29 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US8592397B2 (en) 2003-01-14 2013-11-26 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US20090036408A1 (en) * 2003-01-14 2009-02-05 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
US8871271B2 (en) 2005-06-13 2014-10-28 Gilead Sciences, Inc. Method and composition for pharmaceutical product
US9018192B2 (en) 2005-06-13 2015-04-28 Bristol-Myers Squibb & Gilead Sciences, Inc. Unitary pharmaceutical dosage form
US8598185B2 (en) 2005-06-13 2013-12-03 Bristol-Myers Squibb & Gilead Sciences, Inc. Unitary pharmaceutical dosage form
US9545414B2 (en) 2005-06-13 2017-01-17 Bristol-Myers Squibb & Gilead Sciences, Llc Unitary pharmaceutical dosage form
US20100055180A1 (en) * 2007-10-10 2010-03-04 Mallinckrodt Baker, Inc. Directly Compressible Granular Microcrystalline Cellulose Based Excipient, Manufacturing Process and Use Thereof
US20110092598A1 (en) * 2007-10-10 2011-04-21 Nandu Deorkar Driectly Compressible High Functionality Granular Microcrystalline Cellulose Based Excipient, Manufacturing Process and Use Thereof
US20160199379A1 (en) * 2010-01-27 2016-07-14 Viiv Healthcare Company Antiviral therapy
US11234985B2 (en) 2010-01-27 2022-02-01 Viiv Healthcare Company Antiviral therapy
US10426780B2 (en) 2010-01-27 2019-10-01 Viiv Healthcare Company Antiviral therapy
US9550803B2 (en) 2011-05-06 2017-01-24 University Of Southern California Method to improve antiviral activity of nucleotide analogue drugs
WO2012154698A3 (en) * 2011-05-06 2013-01-31 Mckenna Charles E Method to improve antiviral activity of nucleotide analogue drugs
WO2012154698A2 (en) * 2011-05-06 2012-11-15 Mckenna Charles E Method to improve antiviral activity of nucleotide analogue drugs

Also Published As

Publication number Publication date
ATE398455T1 (de) 2008-07-15
AU2004206821A1 (en) 2004-08-05
US20060234982A1 (en) 2006-10-19
US20150111855A1 (en) 2015-04-23
AP2005003348A0 (en) 2005-06-30
DK1583542T3 (da) 2008-09-22
CA2512475A1 (en) 2004-08-05
US20170273994A1 (en) 2017-09-28
EP1583542B9 (en) 2008-10-22
CN102670629A (zh) 2012-09-19
NZ540728A (en) 2008-08-29
PL378368A1 (pl) 2006-04-03
NO20053817D0 (no) 2005-08-12
US8592397B2 (en) 2013-11-26
WO2004064845A8 (en) 2004-09-23
EP1585527A1 (en) 2005-10-19
EP1583542A1 (en) 2005-10-12
JP2021004264A (ja) 2021-01-14
CY1108355T1 (el) 2014-02-12
EA201100293A1 (ru) 2011-08-30
EP1923063A3 (en) 2009-04-08
WO2004064845A1 (en) 2004-08-05
PT1583542E (pt) 2008-09-17
PL408254A1 (pl) 2014-07-21
IL169243A (en) 2008-12-29
US20090143314A1 (en) 2009-06-04
JP2010120957A (ja) 2010-06-03
NO20171193A1 (no) 2005-10-13
CN102670629B (zh) 2016-02-24
AU2004206821B2 (en) 2009-02-26
US9457036B2 (en) 2016-10-04
JP2006515624A (ja) 2006-06-01
HRP20050619A2 (en) 2005-10-31
NO337917B1 (no) 2016-07-11
JP2015098488A (ja) 2015-05-28
KR20050092755A (ko) 2005-09-22
JP2014037430A (ja) 2014-02-27
WO2004064846A1 (en) 2004-08-05
CA2512319A1 (en) 2004-08-05
EA200501134A1 (ru) 2005-12-29
US20040224917A1 (en) 2004-11-11
US20170232019A1 (en) 2017-08-17
US9744181B2 (en) 2017-08-29
IS7977A (is) 2005-08-12
US8716264B2 (en) 2014-05-06
NO20053817L (no) 2005-10-13
KR20090053867A (ko) 2009-05-27
US20150111856A1 (en) 2015-04-23
US20040224916A1 (en) 2004-11-11
HK1079122A1 (en) 2006-03-31
JP2017057232A (ja) 2017-03-23
US20090036408A1 (en) 2009-02-05
JP2019052174A (ja) 2019-04-04
JP2006516570A (ja) 2006-07-06
EP1583542B1 (en) 2008-06-18
ES2308136T3 (es) 2008-12-01
US20140213556A1 (en) 2014-07-31
CA2512475C (en) 2009-06-02
DE602004014470D1 (de) 2008-07-31
HRP20140379A2 (hr) 2014-07-18
NO20150656A1 (no) 2005-10-13
AU2004206821C1 (en) 2009-10-01
NO340951B1 (no) 2017-07-24
EP1923063A2 (en) 2008-05-21
JP4996241B2 (ja) 2012-08-08
BRPI0406760A (pt) 2005-12-20
CN105596356A (zh) 2016-05-25
SI1583542T1 (sl) 2008-12-31
EA015145B1 (ru) 2011-06-30
MXPA05007016A (es) 2005-09-12
AP2089A (en) 2010-01-11
AU2009200414B2 (en) 2011-12-08
KR100860136B1 (ko) 2008-09-25
KR20080032014A (ko) 2008-04-11
AU2004206827A1 (en) 2004-08-05
AU2009200414A1 (en) 2009-02-26
EP3025718A1 (en) 2016-06-01

Similar Documents

Publication Publication Date Title
US9744181B2 (en) Compositions and methods for combination antiviral therapy
AU2011253996B2 (en) Compositions and methods for combination antiviral therapy
DAHL et al. Patent 2512475 Summary
AU2014216017A1 (en) Compositions and methods for combination antiviral therapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: GILEAD SCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DAHL, TERRENCE C.;MENNING, MARK M.;OLIYAI, REZA;REEL/FRAME:017378/0645

Effective date: 20051206

STCB Information on status: application discontinuation

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