WO1994002154A1 - Antiviral combinations - Google Patents

Antiviral combinations Download PDF

Info

Publication number
WO1994002154A1
WO1994002154A1 PCT/EP1993/001846 EP9301846W WO9402154A1 WO 1994002154 A1 WO1994002154 A1 WO 1994002154A1 EP 9301846 W EP9301846 W EP 9301846W WO 9402154 A1 WO9402154 A1 WO 9402154A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
ethyl
pyridinone
hiv
Prior art date
Application number
PCT/EP1993/001846
Other languages
French (fr)
Inventor
Janet Mary Cameron
Original Assignee
Glaxo Group Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxo Group Limited filed Critical Glaxo Group Limited
Priority to EP93915908A priority Critical patent/EP0650362A1/en
Priority to CA002140237A priority patent/CA2140237A1/en
Priority to JP6504123A priority patent/JPH07508997A/en
Priority to AU45688/93A priority patent/AU4568893A/en
Publication of WO1994002154A1 publication Critical patent/WO1994002154A1/en

Links

Classifications

    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants

Definitions

  • the present invention relates to combinations of antiviral agents. More specifically it is concerned with combinations of 1 ,3-oxathiolane nucleoside analogues with other antiviral agents, in particular agents effective against HIV.
  • HIV Human immunodeficiency virus
  • AIDS acquired immune deficiency syndrome
  • Nucleosides such as AZT, ddC and ddl inhibit HIV replication in vitro, and appear to exert their antiviral activity on the virus-encoded reverse transcriptase enzyme after metabolism by the cell to their 5'-triphosphate derivatives.
  • AZT reduces morbidity and mortality in patients with AIDS.
  • HIV infection of cells results in integration of the virus genome into the host chromosome, and so it has been necessary to continue AZT treatment for long periods of time.
  • the consequences of long-term AZT therapy are associated bone-marrow toxicity and the appearance of AZT-resistant variants of HIV-1.
  • some AIDS patients treated with ddC develop peripheral neuropathy and ddl has been shown to induce pancreatitis and peripheral neuropathy.
  • 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 alpha.
  • combinations of compounds can give rise to increased cytotoxicity.
  • AZT and recombinant interferon alpha have an increased cytotoxic effect on normal human bone marrow progenitor cells.
  • a combination comprising 3TC or a pharmaceutically acceptable derivitive thereof and a non- nucleoside inhibitor of HIV selected from a ⁇ [(benzoxazol-2-yl)methyl]amino ⁇ -5- alkyl-6-alkyl-2-(1 H)-pyridinone or a derivative thereof bearing an unsubstituted or substituted pyridyl or phenyl substituent.
  • the invention provides a combination of 3TC and a compound selected from 3- ⁇ [(4,7-dichlorobenzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)- pyridinone,
  • (+)-enantiomer that is to say no more than about 5% w/w of the (+)-enantiomer, preferably no more than about 2%, in particular less than about 1% w/w will be present.
  • a pharmaceutically acceptable derivative is meant any pharmaceutically acceptable salt, ester, or salt of such ester, of a parent compound or any other compound which, upon administration to the recipient, is capable of providing (directly or indirectly) the parent compound or an antivirally active metabolite or residue thereof.
  • 3TC may be modified to provide pharmaceutically acceptable derivatives thereof at functional groups in both the base moiety and at the hydroxymethyl group of the oxathioiane ring. Modification at all such functional groups are included within the scope of the invention. However of particular interest are pharmaceutically acceptable derivatives obtained by modification of the 2-hydroxymethyl group of the oxathioiane ring.
  • Preferred esters of 3TC include the compounds in which the hydrogen of the 2- hydroxymethyl group is replaced by an acyl function R-CO- in which the non- carbonyl moiety R of the ester is selected from hydrogen, straight or branched chain alkyl (e.g. methyl, ethyl, n-propyl, t-butyl, n-butyl), alkoxyalkyl (e.g. methoxymethyl), aralkyl (e.g. benzyl), aryloxyalkyl (e.g. phenoxymethyl), aryl (e.g.
  • any alkyl moiety present advantageously contains 1 to 16 carbon atoms, particularly 1 to 4 carbon atoms.
  • Any aryl moiety present in such esters advantageously comprises a phenyl group.
  • esters may be a C- ⁇ .-j ⁇ alkyl ester, an unsubstituted benzyl ester or a benzyl ester substituted by at least one halogen (bromine, chlorine, fluorine or iodine), C- ⁇ ⁇ alkyl, C-j. ⁇ alkoxy, nitro or trifluoromethyl groups.
  • halogen bromine, chlorine, fluorine or iodine
  • Pharmaceutically acceptable salts of 3TC include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulphuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succinic, toluene-p- sulphonic, tartaric, acetic, citric, methanesulphonic, formic, benzoic, malonic, naphthalene-2-sulphonic and benzenesulphonic acids.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g. magnesium), ammonium and NR4 + (where R is C-j _4alkyl) salts.
  • 3TC is either synergistic with the second component of the combination and/or reduces the cytotoxic effects of the second component.
  • 3TC and the non-nucleoside inhibitors of HIV are realised over a wide ratio for example 1 :250 to 250:1 preferably 1 :50 to 50:1 , particularly about 1 :10 to 10:1 by weight.
  • each compound will be employed in the combination in an amount at which it exhibits antiviral activity when used alone.
  • a method for the treatment of a viral infection in a mammal comprising co-administration of 3TC or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV replication as defined herein.
  • Therapeutic methods comprising administration of a combination of a 3TC and more than one of the non- nucleoside inhibitors of HIV, either together or in a plurality of paired combinations, is also within the scope of the invention.
  • 3TC and the second antiviral agent may be administered either simultaneously (either separately or in combination) or sequentially. If administration is sequential, the delay in administering the second of the active ingredients should not be such as to lose the benefit of any synergistic effect of the combination. Preferably administration will be simultaneous.
  • a suitable dose will be in the range of from about 1 to about 750mg/kg e.g. from about 10 to about 75 mg/kg of bodyweight per day, such as 3 to about 120mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90mg/kg/day, most preferably in the range of 15 to 60mg/kg/day of each of the active ingredients of the combination.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • the combination is conveniently administered in unit dosage form; for example containing 10 to 1500mg, conveniently 20 to 10OOmg, most conveniently 50 to 700mg of each active ingredient per unit dosage form.
  • the combinations should be administered to achieve peak plasma concentrations of each of the active compound of about 1 to about 75 ⁇ M, preferably about 2 to 50 ⁇ M, most preferably about 3 to about 30 ⁇ M.
  • This may be achieved, for example, by the intravenous injection of a 0.1 to 5% solution of the active ingredients, optionally in saline, or orally administered as a bolus containing about 1 to about 100mg of each active ingredient.
  • Desirable blood levels may be maintained by a continuous infusion to provide about 0.01 to about 5.0mg/kg/hour or by intermittent infusions containing about 0.4 to about 15mg/kg of each active ingredient.
  • the active ingredients of the combination may be administered as the raw chemical it is preferable to present combinations as a pharmaceutical formulation.
  • the invention thus further provides a pharmaceutical formulation comprising 3TC or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV replication as defined herein together with one or more pharmaceutically acceptable carriers therefor and, optionally, other therapeutic and/or prophylactic ingredients.
  • the carrier(s) must be 'acceptable' in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
  • the formulations may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association the active compound with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • compositions suitable for oral administration may conveniently 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, a suspension or as an emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • Tablets and capsules for oral administration may contain conventional excipients such as binding agents, fillers, lubricants, disintegrates, or wetting agents.
  • the tablets may be coated according to methods well known in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservatives.
  • the compounds according to the invention may also be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre- filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilising and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active ingredient in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • 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, and the suppositories may be conveniently formed by admixture of the active compound with the softened or melted carrier(s) followed by chilling and shaping in moulds.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • the compounds of the invention may be used as a liquid spray or dispersible powder or in the form of drops.
  • Drops may be formulated with an aqueous or non-aqueous base also comprising one more dispersing agents, solubilising agents or suspending agents. Liquid sprays are conveniently delivered from pressurised packs.
  • the compounds according to the invention are conveniently delivered from an insufflator, nebuliser or a pressurised pack or other convenient means of delivering an aerosol spray.
  • Pressurised packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form in, for example, capsules or cartridges or e.g. gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • compositions according to the invention may also contain other active ingredients such as antimicrobial agents, or preservatives.
  • 3TC may be obtained as described in International Patent Application No. W091/17159.
  • the non-nucleoside inhibitors of HIV replication may be obtained as described in EP0484071.
  • Chequerboard titrations are prepared by mixing 25 ⁇ l aliquots from each compound dilution both alone or in combination (to a final volume of 50 ⁇ l in new 96-well microtitre plates). Aliquots of MT-4 cells (10 6 cells/ml) in RPM1 1640 growth medium are infected with HIV-1 strain RF at a moi of 2 x 10"3 infectious doses/cell. Virus is adsorbed at room temperature for 90 minutes, after which the cells are washed in RPM1 1640 growth medium to remove unadsorbed virus and resuspended at 10 6 cells/ml in RPM1 1640 growth medium.
  • 50 ⁇ l of infected cell suspension are inoculated into wells containing compound or growth medium only.
  • 50 ⁇ l of mock-infected cell suspension are inoculated into wells not containing compound. The plates are then incubated for 7 days at 37°C in 5% C ⁇ 2/air.
  • Dose-response curves are plotted for each compound alone (IC50% values) and for reciprocal titrations of each compound at a fixed concentration of the second compound. Isobolograms of all compound combinations giving IC50% values are plotted.
  • the two compounds act additively. If the combination IC50% lie to the left of the line, the compounds are acting synergistically.
  • Cytotoxicity is determined by examination of the cells employed in Example 1 following drug treatment and/or by comparing the cvtotoxicities of 3TC and the non-nucleoside HIV inhibitors alone and in combination (at ⁇ g/ml ratios of 1 :1 , 1 :5 and 5:1 ) in uninfected peripheral blood lymphocytes and an established T- lymphocyte cell line; cytotoxicity is measured using a [ 3 H]-thymidine uptake assay.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Combinations of (2R, c^_i_^_s^_)-4-amino-1-(2-hydroxymethyl-1,3-oxathiolan-5-yl)-(1H)-pyrimidin-2-one or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV selected from a {[(benzoxazol-2-yl)methyl]amino}-5-alkyl-6-alkyl-2-(1H)-pyridinone or a derivative thereof bearing an unsubstituted or substituted pyridyl or phenyl substituent, pharmaceutical formulations of such combinations and the use of such combinations and formulations to treat viral infections, in particular HIV infections, are described.

Description

ANTIVIRAL COMBINATIONS
The present invention relates to combinations of antiviral agents. More specifically it is concerned with combinations of 1 ,3-oxathiolane nucleoside analogues with other antiviral agents, in particular agents effective against HIV.
Human immunodeficiency virus (HIV) causes a variety of clinical conditions including the acquired immune deficiency syndrome (AIDS) and chronic neurological disorders. Nucleosides such as AZT, ddC and ddl inhibit HIV replication in vitro, and appear to exert their antiviral activity on the virus-encoded reverse transcriptase enzyme after metabolism by the cell to their 5'-triphosphate derivatives.
AZT reduces morbidity and mortality in patients with AIDS. However, HIV infection of cells results in integration of the virus genome into the host chromosome, and so it has been necessary to continue AZT treatment for long periods of time. The consequences of long-term AZT therapy are associated bone-marrow toxicity and the appearance of AZT-resistant variants of HIV-1. Similarly, some AIDS patients treated with ddC develop peripheral neuropathy and ddl has been shown to induce pancreatitis and peripheral neuropathy.
The use of combinations of compounds may give rise to an equivalent antiviral effect with reduced toxicity, or an increase in drug efficacy if synergy between compounds occurs. Lower overall drug doses will possibly also reduce the frequency of occurrence of drug-resistant variants of HIV. Many different methods have been used to examine the effects of combinations of compounds acting together in different assay systems. All of these methods have limitations and for example, some methods have been applied to systems other than those for which they were derived. 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 alpha. However, it must be noted that combinations of compounds can give rise to increased cytotoxicity. AZT and recombinant interferon alpha have an increased cytotoxic effect on normal human bone marrow progenitor cells.
The compound (2R. cis)-4-amino-1-(2-hvdroxvmethvl-1.3-oxathiolan-5-vl) -(1H)- pyridmidin-2-one, also known as 3TC, is currently undergoing clinical trials for the treatment of conditions associated with infection by HIV.
We have now found that 3TC exhibits unexpected advantages when used in combination with certain non-nucleoside inhibtors of HIV replication.
There is thus provided in a first aspect of the invention a combination comprising 3TC or a pharmaceutically acceptable derivitive thereof and a non- nucleoside inhibitor of HIV selected from a {[(benzoxazol-2-yl)methyl]amino}-5- alkyl-6-alkyl-2-(1 H)-pyridinone or a derivative thereof bearing an unsubstituted or substituted pyridyl or phenyl substituent.
In a preferred aspect the invention provides a combination of 3TC and a compound selected from 3-{[(4,7-dichlorobenzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)- pyridinone,
3-{[(4,7-dimethylbenzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)- pyridinone,
3-{[(7-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone, 3-{[(7-methylbenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(4-fluorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(7-fluorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)-pyridinone,
3-{[(benzoaxazol-2-yl)methyi]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(4-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone, 3-{[(4-fluoro-7-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)- pyridinone,
3-[2-benzoaxazol-2-yl)ethyl]-5-ethyl-6-methyl-2-(1H)-pyridinone ,
3-[N-(5-ethyl-2-methoxy-6-methyl-3-pyridylmethyl)-amino]-5-ethyl-6-methyl-2-
(I H)-pyridinone 3-[N-(5,6-dimethyl-2-methoxy-3-pyridylmethyl)amino]-5-ethyl-6-methyl-2-(1 H)- pyridinone, 3-[N-(5-ethyl-2-methoxybenzyl)amino)-5-ethyl-6-methyl-2-(1H)-pyridinone,
3-[N-(2-methoxy-4,5-dimethylbenzyl)amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone and
3-[N-(2,6-dimethoxybenzyl)amino]-5-ethyl-6-methyl-2(1 H)-pyridinone.
3TC will normally be provided substantially free of the corresponding
(+)-enantiomer, that is to say no more than about 5% w/w of the (+)-enantiomer, preferably no more than about 2%, in particular less than about 1% w/w will be present.
By "a pharmaceutically acceptable derivative" is meant any pharmaceutically acceptable salt, ester, or salt of such ester, of a parent compound or any other compound which, upon administration to the recipient, is capable of providing (directly or indirectly) the parent compound or an antivirally active metabolite or residue thereof.
It will be appreciated by those skilled in the art that 3TC may be modified to provide pharmaceutically acceptable derivatives thereof at functional groups in both the base moiety and at the hydroxymethyl group of the oxathioiane ring. Modification at all such functional groups are included within the scope of the invention. However of particular interest are pharmaceutically acceptable derivatives obtained by modification of the 2-hydroxymethyl group of the oxathioiane ring.
Preferred esters of 3TC include the compounds in which the hydrogen of the 2- hydroxymethyl group is replaced by an acyl function R-CO- in which the non- carbonyl moiety R of the ester is selected from hydrogen, straight or branched chain alkyl (e.g. methyl, ethyl, n-propyl, t-butyl, n-butyl), alkoxyalkyl (e.g. methoxymethyl), aralkyl (e.g. benzyl), aryloxyalkyl (e.g. phenoxymethyl), aryl (e.g. phenyl optionally substituted by halogen, C1.4 alkyl or C1.4 alkoxy); sulphonate esters such as alkyl- or aralkylsulphonyl (e.g. methanesulphonyl); amino acid esters (e.g. L-valyl or L-isoleucyl) and mono-, di- or tri-phosphate esters. With regard to the above described esters, unless otherwise specified, any alkyl moiety present advantageously contains 1 to 16 carbon atoms, particularly 1 to 4 carbon atoms. Any aryl moiety present in such esters advantageously comprises a phenyl group.
In particular the esters may be a C-\ .-j βalkyl ester, an unsubstituted benzyl ester or a benzyl ester substituted by at least one halogen (bromine, chlorine, fluorine or iodine), C-μ ρalkyl, C-j.βalkoxy, nitro or trifluoromethyl groups.
Pharmaceutically acceptable salts of 3TC include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulphuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succinic, toluene-p- sulphonic, tartaric, acetic, citric, methanesulphonic, formic, benzoic, malonic, naphthalene-2-sulphonic and benzenesulphonic acids. Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g. magnesium), ammonium and NR4+ (where R is C-j _4alkyl) salts.
3TC is either synergistic with the second component of the combination and/or reduces the cytotoxic effects of the second component.
The advantageous effects of 3TC and the non-nucleoside inhibitors of HIV are realised over a wide ratio for example 1 :250 to 250:1 preferably 1 :50 to 50:1 , particularly about 1 :10 to 10:1 by weight. Conveniently each compound will be employed in the combination in an amount at which it exhibits antiviral activity when used alone.
It is expected that the present combinations will be generally useful against viral infections or virus-associated tumours in humans, and the method of their use to inhibit viral infectivity or tumour growth in vitro or in vivo is also within the scope of the present invention.
Thus there is provided in a second aspect a method for the treatment of a viral infection in a mammal, including man, comprising co-administration of 3TC or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV replication as defined herein. Therapeutic methods comprising administration of a combination of a 3TC and more than one of the non- nucleoside inhibitors of HIV, either together or in a plurality of paired combinations, is also within the scope of the invention.
It will be appreciated that 3TC and the second antiviral agent may be administered either simultaneously (either separately or in combination) or sequentially. If administration is sequential, the delay in administering the second of the active ingredients should not be such as to lose the benefit of any synergistic effect of the combination. Preferably administration will be simultaneous.
It will be appreciated by those skilled in the art that reference herein to treatment extends to prophylaxis as well as the treatment of established infections or symptoms.
It will be further appreciated that the amount of a combination of the invention required for use in treatment will vary not only with the particular compounds selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or veterinarian. In general however a suitable dose will be in the range of from about 1 to about 750mg/kg e.g. from about 10 to about 75 mg/kg of bodyweight per day, such as 3 to about 120mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90mg/kg/day, most preferably in the range of 15 to 60mg/kg/day of each of the active ingredients of the combination. The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example as two, three, four or more sub-doses per day.
The combination is conveniently administered in unit dosage form; for example containing 10 to 1500mg, conveniently 20 to 10OOmg, most conveniently 50 to 700mg of each active ingredient per unit dosage form.
Ideally the combinations should be administered to achieve peak plasma concentrations of each of the active compound of about 1 to about 75μM, preferably about 2 to 50μM, most preferably about 3 to about 30μM. This may be achieved, for example, by the intravenous injection of a 0.1 to 5% solution of the active ingredients, optionally in saline, or orally administered as a bolus containing about 1 to about 100mg of each active ingredient. Desirable blood levels may be maintained by a continuous infusion to provide about 0.01 to about 5.0mg/kg/hour or by intermittent infusions containing about 0.4 to about 15mg/kg of each active ingredient.
While it is possible that, for use in therapy, the active ingredients of the combination may be administered as the raw chemical it is preferable to present combinations as a pharmaceutical formulation.
The invention thus further provides a pharmaceutical formulation comprising 3TC or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV replication as defined herein together with one or more pharmaceutically acceptable carriers therefor and, optionally, other therapeutic and/or prophylactic ingredients. The carrier(s) must be 'acceptable' in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
Pharmaceutical formulations include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation. The formulations may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association the active compound with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
Pharmaceutical formulations suitable for oral administration may conveniently 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, a suspension or as an emulsion. The active ingredient may also be presented as a bolus, electuary or paste. Tablets and capsules for oral administration may contain conventional excipients such as binding agents, fillers, lubricants, disintegrates, or wetting agents. The tablets may be coated according to methods well known in the art. Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservatives.
The compounds according to the invention may also be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre- filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilising and/or dispersing agents. Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
For topical administration to the epidermis the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch. Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
Formulations suitable for topical administration in the mouth include lozenges comprising active ingredient in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Pharmaceutical formulations 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, and the suppositories may be conveniently formed by admixture of the active compound with the softened or melted carrier(s) followed by chilling and shaping in moulds.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
For intra-nasal administration the compounds of the invention may be used as a liquid spray or dispersible powder or in the form of drops.
Drops may be formulated with an aqueous or non-aqueous base also comprising one more dispersing agents, solubilising agents or suspending agents. Liquid sprays are conveniently delivered from pressurised packs.
For administration by inhalation the compounds according to the invention are conveniently delivered from an insufflator, nebuliser or a pressurised pack or other convenient means of delivering an aerosol spray. Pressurised packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurised aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Alternatively, for administration by inhalation or insufflation, the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form in, for example, capsules or cartridges or e.g. gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
When desired the above described formulations adapted to give sustained release of the active ingredient may be employed.
The pharmaceutical compositions according to the invention may also contain other active ingredients such as antimicrobial agents, or preservatives.
3TC may be obtained as described in International Patent Application No. W091/17159.
The non-nucleoside inhibitors of HIV replication may be obtained as described in EP0484071.
The following examples illustrate the invention but are not intended as a limitation thereof.
EXAMPLE 1
Antiviral Activities Alone or in Combination
Compounds are first serially-diluted in 2-fold decrements in 96-well microtitre plates. Chequerboard titrations are prepared by mixing 25μl aliquots from each compound dilution both alone or in combination (to a final volume of 50μl in new 96-well microtitre plates). Aliquots of MT-4 cells (106 cells/ml) in RPM1 1640 growth medium are infected with HIV-1 strain RF at a moi of 2 x 10"3 infectious doses/cell. Virus is adsorbed at room temperature for 90 minutes, after which the cells are washed in RPM1 1640 growth medium to remove unadsorbed virus and resuspended at 106cells/ml in RPM1 1640 growth medium. 50μl of infected cell suspension are inoculated into wells containing compound or growth medium only. 50μl of mock-infected cell suspension are inoculated into wells not containing compound. The plates are then incubated for 7 days at 37°C in 5% Cθ2/air.
After incubation, 10μl of 3-[4,5-dimethyl thiazol-2-yl]-2,5- diphenyltetrazolium bromide (MTT) at 7.5mg/ml are added to all wells and the plates incubated for a further 90 minutes at 37°C. 150μl of 10% (v/v) Triton X-100 in isopropanol are then added and the cells resuspended. After 15 minutes at room temperature the plates are analysed in a Multiskan MC (Flow Laboratories, Irvine, UK) reader at 405nm. Conversion of yellow MTT to its formazan derivative is maximum in the uninfected untreated cells, and absent in untreated infected cells.
Dose-response curves are plotted for each compound alone (IC50% values) and for reciprocal titrations of each compound at a fixed concentration of the second compound. Isobolograms of all compound combinations giving IC50% values are plotted.
If the IC50% values of compound combination lies on a line joining the IC50% values of each compound on its own, then the two compounds act additively. If the combination IC50% lie to the left of the line, the compounds are acting synergistically.
EXAMPLE 2
Cvtotoxicities of Compounds Alone and in Combination.
Cytotoxicity is determined by examination of the cells employed in Example 1 following drug treatment and/or by comparing the cvtotoxicities of 3TC and the non-nucleoside HIV inhibitors alone and in combination (at μg/ml ratios of 1 :1 , 1 :5 and 5:1 ) in uninfected peripheral blood lymphocytes and an established T- lymphocyte cell line; cytotoxicity is measured using a [3H]-thymidine uptake assay.

Claims

Claims
1. A combination comprising (2R. cisV4-amino-1-(2-hydroxymethyl-1.3- oxathiolan-5-yl)-(1 H)-pyrimidin-2-one or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV selected from a {[(benzoxazol-2-yl)methyl]amino}-5-alkyl-6-alkyl-2-(1 H)-pyridinone or a derivative thereof bearing an unsubstituted or substituted pyridyl or phenyl substituent.
2. A combination according to claim 1 wherein the non-nucleoside inhibitor of HIV is selected from 3-{[(4,7-dichlorobenzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)- pyridinone,
3-{[(4,7-dimethylbenzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)- pyridinone,
3-{[(7-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)-pyridinone, 3-{[(7-methylbenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(4-fluorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)-pyridinone,
3-{[(7-fluorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(benzoaxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-{[(4-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)-pyridinone, 3-{[(4-fluoro-7-chlorobenzoxazol-2-yl)methyl]amino)-5-ethyl-6-methyl-2-(1H)- pyridinone,
3-[2-benzoaxazol-2-yl)ethyl]-5-ethyl-6-methyl-2-(1H)-pyridinone ,
3-[N-(5-ethyl-2-methoxy-6-methyl-3-pyridylmethyl)-amino]-5-ethyl-6-methyl-2-
(1 H)-pyridinone 3-[N-(5,6-dimethyl-2-methoxy-3-pyridylmethyl)amino]-5-ethyl-6-methyl-2-(1 H)- pyridinone,
3-[N-(5-ethyl-2-methoxybenzyl)amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone,
3-[N-(2-methoxy-4,5-dimethylbenzyl)amino)-5-ethyl-6-methyl-2-(1 H)-pyridinone and 3-[N-(2,6-dimethoxybenzyl)amino]-5-ethyl-6-methyl-2(1 H)-pyridinone. 3. A combination according to claim 1 or claim 2 wherein the ratio of (2R, cis)- 4-amino-1-(2-hydroxymethyl-1 ,3-oxathiolan-5-yl)-(1 H)-pyrimidin-2-one to the non-nucleoside inhibitor of HIV is from 1 :250 to 250:1 by weight.
4. A pharmaceutical formulation comprising a combination as defined in any one of claims 1 to 3 together with a pharmaceutically acceptable carrier therefor.
5. A method for the treatment of a mammal, including man, suffering from or susceptible to infection by HIV comprising co-administration of (2R, cisV4- amino-1-(2-hydroxymethyl-1 ,3-oxathiolan-5-yl)-(1 H)-pyrimidin-2-one or a pharmaceutically acceptable derivative thereof and a non-nucleoside inhibitor of HIV selected from a {[(benzoxazol-2-yl)methyl]amino}-5-alkyl-6-alkyl-2-(1 H)- pyridinone or a derivative thereof, bearing an unsubstituted or substituted pyridyl or phenyl substituent.
6. A method according to claim 5 wherein administration is sequential.
7. A method according to claim 5 wherein administration is simultaneous.
PCT/EP1993/001846 1992-07-16 1993-07-13 Antiviral combinations WO1994002154A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP93915908A EP0650362A1 (en) 1992-07-16 1993-07-13 Antiviral combinations
CA002140237A CA2140237A1 (en) 1992-07-16 1993-07-13 Antiviral combinations
JP6504123A JPH07508997A (en) 1992-07-16 1993-07-13 Antiviral combination
AU45688/93A AU4568893A (en) 1992-07-16 1993-07-13 Antiviral combinations

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB929215176A GB9215176D0 (en) 1992-07-16 1992-07-16 Antiviral combinations
GB9215176.0 1992-07-16

Publications (1)

Publication Number Publication Date
WO1994002154A1 true WO1994002154A1 (en) 1994-02-03

Family

ID=10718842

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1993/001846 WO1994002154A1 (en) 1992-07-16 1993-07-13 Antiviral combinations

Country Status (6)

Country Link
EP (1) EP0650362A1 (en)
JP (1) JPH07508997A (en)
AU (1) AU4568893A (en)
CA (1) CA2140237A1 (en)
GB (1) GB9215176D0 (en)
WO (1) WO1994002154A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5763042A (en) * 1994-06-28 1998-06-09 Reichhold Chemicals, Inc. Reinforcing structural rebar and method of making the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991017159A1 (en) * 1990-05-02 1991-11-14 Iaf Biochem International Inc. 1,3-oxathiolane nucleoside analogues
EP0484071A2 (en) * 1990-11-01 1992-05-06 Merck & Co. Inc. Synergism of HIV reverse transcriptase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991017159A1 (en) * 1990-05-02 1991-11-14 Iaf Biochem International Inc. 1,3-oxathiolane nucleoside analogues
EP0484071A2 (en) * 1990-11-01 1992-05-06 Merck & Co. Inc. Synergism of HIV reverse transcriptase inhibitors

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
AIDS RES. HUM. RETROVIRUS vol. 8, no. 2, February 1992, pages 119 - 134 E. DECLERCQ 'HIV inhibitors targeted at the reverse transcriptase' *
CURR. OPIN. INFECT. DIS. vol. 5/6, May 1992, pages 798 - 805 C. FLEXNER 'New antiretroviral agents in clinical development' *
PROC. NATL. ACAD. SCI. USA vol. 88, no. 15, August 1991, pages 6863 - 6867 M. GOLDMAN ET AL. 'Pyridinone derivatives: Specific HIV-1 reverse transcriptase inhibitors with antiviral activity' *
PROC. NATL. ACAD. SCI. USA vol. 90, June 1993, pages 5653 - 5656 M. TISDALE ET AL. 'Rapid in vitro selection of HIV-1 resistant to 3'-thiacytidine inhibitors due to a mutation in the YMDD region of reverse transcriptase' *

Also Published As

Publication number Publication date
GB9215176D0 (en) 1992-08-26
AU4568893A (en) 1994-02-14
EP0650362A1 (en) 1995-05-03
CA2140237A1 (en) 1994-02-03
JPH07508997A (en) 1995-10-05

Similar Documents

Publication Publication Date Title
US5627186A (en) Antiviral combinations
EP0844878A1 (en) 1,3-oxathiolane nucleoside analogues in the treatment of hepatitis c
EP1628682B1 (en) Composition comprising a pde4 inhibitor and a pde5 inhibitor for the treatment of copd
JPH0125A (en) Pharmaceutical composition for treating HIV infection comprising dsRNA and reverse transcriptase inhibitor
EA032868B1 (en) Combination for treating hiv infection
US20240058347A1 (en) Dosages for hdac treatment with reduced side effects
JP2818299B2 (en) Use of dideoxynucleoside analogs in the treatment of viral infections
AU2004275771A1 (en) Iloprost in combination therapies for the treatment of pulmonary arterial hypertension
RU2417083C2 (en) Application of soluble guanylate cyclase activators for treating acute and chronic diseases
US20030050329A1 (en) Synergistic combination comprising roflumilast and a PDE-3 inhibitor
US5612333A (en) Antiviral combinations
WO1994002154A1 (en) Antiviral combinations
US10953011B2 (en) Methods of treating virally associated cancers with histone deacetylase inhibitors
CA2213621A1 (en) Antiviral combinations of bch-189 and ritonavir
Wutzler et al. Neuraminidase inhibitors in the treatment of influenza A and B–overview and case reports
RU2139059C1 (en) Antiviral composition containing nucleoside analog, pharmaceutical composition, method of treatment
IT202100024668A1 (en) Compositions and methods for combined antiviral therapy
WO2023244968A1 (en) Methods of treating autoimmune disorders with histone deacetylase inhibitors
MXPA06003273A (en) Iloprost in combination therapies for the treatment of pulmonary arterial hypertension

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR BY CA CH CZ DE DK ES FI GB HU JP KP KR KZ LK LU MG MN MW NL NO NZ PL PT RO RU SD SE SK UA US VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 1994 356321

Country of ref document: US

Date of ref document: 19941229

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1993915908

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2140237

Country of ref document: CA

WWP Wipo information: published in national office

Ref document number: 1993915908

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1993915908

Country of ref document: EP