WO1998057176A1 - Protein elongation factor 2 as a target for antifungal and antiparasitic agents - Google Patents

Protein elongation factor 2 as a target for antifungal and antiparasitic agents Download PDF

Info

Publication number
WO1998057176A1
WO1998057176A1 PCT/US1998/011903 US9811903W WO9857176A1 WO 1998057176 A1 WO1998057176 A1 WO 1998057176A1 US 9811903 W US9811903 W US 9811903W WO 9857176 A1 WO9857176 A1 WO 9857176A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
fungal
function
parasitic
cells
Prior art date
Application number
PCT/US1998/011903
Other languages
French (fr)
Inventor
Jennifer Nielsen-Kahn
Michael C. Justice
Dennis M. Schmatz
Ming-Jo Hsu
Theresa W. Ku
Original Assignee
Merck & Co., 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
Priority claimed from GBGB9714331.7A external-priority patent/GB9714331D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CA002291653A priority Critical patent/CA2291653A1/en
Priority to JP50307999A priority patent/JP2002511933A/en
Priority to EP98928944A priority patent/EP0995120A1/en
Priority to AU80627/98A priority patent/AU741017B2/en
Publication of WO1998057176A1 publication Critical patent/WO1998057176A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56961Plant cells or fungi

Definitions

  • PROTEIN ELONGATION FACTOR 2 AS A TARGET
  • Elongation factor 2 is an essential protein catalysing ribosomal translocation during protein synthesis in eukaryotic cells. It is highly conserved in all eukaryotes, and has been found to be largely interchangeable in in vitro protein synthesis systems reconstituted from such divergent organisms as human, wheat germ, and fungi. Despite the ubiquitous nature of EF2 in eukaryotic systems and the high degree of amino acid sequence homology between EF2s from various eukaryotic systems, a class of compounds, the sordarins, have now been identified to be selective inhibitors of fungal protein synthesis via a selective interaction with fungal EF2.
  • the present invention relates to elongation factor 2 (hereinafter referred to as "EF2") as a target for antifungal and antiparasitic agents.
  • EF2 elongation factor 2
  • the invention relates to a method for identifying potential antifungal and antiparasitic agents by determining whether a test compound is capable of specifically inhibiting pathogenic protein synthesis via a selective interaction with pathogen EF2.
  • the present invention describes the use of mechanism based assays with or without the use of a transformed eukaryotic organism with a heterologous EF2 to facilitate drug discovery.
  • the invention relates to a method for treating fungal infections by administering to a host suffering from a fungal or parasitic infection a therapeutically effective amount of a compound that specifically inhibits the pathogen's protein synthesis via EF2.
  • This invention provides a method for identifying and evaluating compounds having antifungal and antiparasitic activity comprising: A differential two plate assay containing genetically engineered sordarin sensitive (sSl) and resistant (sRl) strains or naturally selected sordarin resistant strains of yeast.
  • the readout of the assay is antimicrobial activity indicated by zones of inhibition which is more apparent against the sordarin sensitive strain relative to the sordarin resistant strain.
  • sSl sordarin sensitive
  • sRl sordarin resistant
  • EFT1 and EFT2 and at least one of these genes is required for survival.
  • the co-isogenic strains sSl and sRl were constructed by a series of genetic crosses that result in strains that are disrupted for both the EFT1 and ERG6 genes. The resultant strains are made more permeable due to the ergo disruption (ergosterol deficient), and have either a wild-type or resistant copy of EFT2 as the only source of EF2.
  • a known number of these yeast cells are either plated in solid medium or suspended in liquid medium and test compounds or fermentation extracts are applied with the intent of identifying samples which inhibit the growth of these yeast. The cultures are incubated at a specific temperature for a set period of time to allow for the growth of the test organisms (i.e.
  • Test samples of interest are those which show a differential effect on the sordarin sensitive strain(s) vs. the resistant strain(s). Those samples which are more potent against the wildtype by definition should be preventing growth via the EF2 target.
  • the present invention provides a method for identifying compounds specifically inhibiting pathogenic EF2 function comprising: (a) constructing fungal or protozoan cells dependent upon heterologous EF2 from fungal and parasitic pathogens or from the host species; (b) contacting said cell with a known dilution of a test compound or a natural product extract; and (c) quantitating the minimal inhibitory concentration (MIC) of test compound to completely inhibit growth in liquid or the measurement of an inhibitory zone on a solid substrate.
  • Test compounds or fermentation extracts of interest are those which display a differential degree of inhibition (i.e. more inhibitory activity against the wildtype vs. resistant strains of the test organisms). For example a sample which is more effective at inhibiting the growth of a yeast EF2 dependent organism vs. one that is dependent on human EF2.
  • the methods of the invention provides a facile and specific assay to screen compounds as potential antifungal and antiparasitic agents. It also allows for the evaluation of test compounds against the EF2 target of obligate pathogens that cannot be cultured in the laboratory.
  • EF2 may be cloned from pathogenic organisms for use in growth inhibition assays or purified from these pathogens for use in in vitro binding or translation inhibition assays.
  • the EF2 may be from pathogenic fungi of humans, animals or plants such as Candida, Aspergillus spp., Cryptococcus spp., Erysiphe and Puccinia. It may also be from protozoan parasites such as Plasmodium sp., Eimeria sp., Crypto sporidium sp. and Toxoplasma gondii and human and other desired host eukaryotic cells.
  • a compound that inhibits EF2 may be one that interferes with the translation of mRNA in target organisms.
  • Examples of compounds that inhibit EF2 include diptheria toxin and fusidic acid, however neither of these show any specificity for pathogen over host. Fusidic acid inhibits translation in many organisms by disrupting normal ribosome-EF2 interactions.
  • the compound that inhibits EF2 is preferably labeled to allow easy quantitation of the level of interaction between the compound and the enzyme.
  • a prefered radiolabel is tritium.
  • the test compound may be a synthetic compound, a mixture of synthetic compounds, a crude preparation, a purified preparation or an initial extract of a natural product obtained from plant, microorganism or animal sources.
  • sordarin is an antifungal antibiotic isolated from the mould Sordaria araneosa (see GB 1,162,027 and Helvetica Chimica Acta, 1971, 51:119-20).
  • Other compounds having the sordarin skeleton have also been reported as antifungal agents.
  • Japanese Kokai J62040292 discloses the compound zofimarin isolated from Zofiela marina sp.
  • Japanese Kokai J06157582 discloses the compound BE-31405 isolated from Penicillium sp.
  • SCH57404 is reported in J. Antibiotics.
  • sordaricin the aglycone
  • Sordaricin may be obtained from sordarin by acid hydrolysis (Hauser and Sigg, Helvetica Chimica Acta, 1971, 51: 119-20); similarly sordaricin methyl ester is obtained from sordarin methyl ester.
  • the total synthesis of sordaricin methyl ester is reported in Kato et al, J. Chem. Soc. Chem. Commun.. 1993, 1002-1004, which also discloses O-methoxymethyl sordaricin methyl ester.
  • Rl2 i s CHO R is not (g); Rl is (a) C1-C14 alkyl,
  • R5 is (a) R 1 or
  • R9 and RIO are independently
  • Rl l is (a) OH or
  • Rl3 is (a) H
  • Rl4 is (a) H
  • EF2 inhibitors are useful as antifungal and antiparasitic agents. As such, they may be used in the treatment and prevention of fungal and parasitic diseases in human, animals and plants.
  • fungal diseases against which EF2 inhibitors may be used, and their respective causative pathogens include: 1) Erysiphe, Puccinia, Septoria, Botrytis, Phytophthora, Plasmopora and other fungi which cause infections in plants and crops 2) Candida, Aspergillus, Cryptococcus, Fusarium, Penicillium and other fungi which cause fungal infections in man and animals 3) Plasmodium, Eimeria, Toxoplasma, Neospora, Cryptosporidium and other protozoa which infect man and animals.
  • the present invention provides a method for the treatment of fungal or parasitic infections comprising administering to a host suffering from a fungal or parasitic infection a therapeutically effective amount of a compound which inhibits EF2 function.
  • a therapeutically effective amount may be one that is sufficient to inhibit protein synthesis of the causative fungi or parasite.
  • EF2 inhibitors may be administered to a host in need of treatment in a manner similar to that used for other antifungal and antiparasitic agents; for example, they may be administered parenter- ally, orally, topically, or rectally.
  • the dosage to be administered will vary according to the particular compound used, the infectious organism involved, the particular host, the severity of the disease, physical condition of the host, and the selected route of administration; the appropriate dosage can be readily determined by a person skilled in the art.
  • the dosage may range from 0.01 mg/kg to 500 mg/kg.
  • the dosage may range from 0.01 mg/kg to 100 mg/kg.
  • the compositions of the present invention comprises an
  • compositions for human and veterninary usage, or in the form of feed composition.
  • composition is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions of one or more of the ingredients.
  • the composition of the present invention thus includes a composition when made by admixing an EF2 inhibitor and inert carrier.
  • compositions of the present invention comprise an EF2 inhibitor as an active ingredient, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients.
  • the compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administrations, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy.
  • an EF2 inhibitor can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed.
  • oral liquid preparations such as suspensions, elixirs and solutions
  • water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like may be used; or in the case of oral solid preparations such as powders, capsules and tablets, carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be included.
  • carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be included.
  • tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed.
  • tablets may be coated by standard aqueous or nonaqueous techniques.
  • compositions 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 or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion or a water-in-oil liquid emulsion.
  • Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients.
  • compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet may be prepared by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • each tablet contains from about 1 mg to about 500 mg of the active ingredient and each cachet or capsule contains from about 1 to about 500 mg of the active ingredient.
  • compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of these active compounds in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Suitable topical formulations include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like. These formulations may be prepared via conventional methods containing the active ingredient. To illustrate, a cream or ointment is prepared by mixing sufficient quantities of hydrophilic material and water, containing from about 5-10% by weight of the compound, in sufficient quantities to produce a cream or ointment having the desired consistency.
  • 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 combination with the softened or melted carrier(s) followed by chilling and shaping moulds.
  • the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like, and. substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.
  • compositions containing a compound of formula I may also be prepared in powder or liquid concentrate form .
  • conventional water soluble excipients such as lactose or sucrose, may be incorporated in the powders to improve their physical properties.
  • suitable powders of this invention comprise 50 to 100% w/w, and preferably 60 to 80% w/w of the combination and 0 to 50% w/w and preferably 20 to 40% w/w of conventional veterinary excipients.
  • These powders may either be added to animal feedstuffs, for example by way of an intermediate premix, or diluted in animal drinking water.
  • Liquid concentrates of this invention suitably contain a water-soluble compound combination and may optionally include a veterinarily acceptable water miscible solvent, for example polyethylene glycol, propylene glycol, glycerol, glycerol formal or such a solvent mixed with up to 30% v/v of ethanol.
  • the liquid concentrates may be administered to the drinking water of animals, particularly poultry.
  • EXAMPLES The following non-limiting examples are provided to illustrate the invention.
  • the assays may be run in 96 well or other appropriate sized plates or in the appropriate liquid medium.
  • yeast strain YEFD 12h/pURA3-EFT 1 that is deleted for both chromosomal copies of genes which encode EF2, has been obtained from the laboratory of James Bodley (Phan et al., Journal of Biological Chemistry (1993). 268:8665-8668). This strain also contains an episomal copy of a gene encoding EF2, and is essential for
  • Yeast strains expressing EF2 genes from pathogens of interest may be constructed by (1) transforming YEFD12h/pURA3- EFT1 with yeast expression plasmids that contain heterologous EF2 encoding genes, and (2) evicting the plasmid containing the native EF2 gene from the cell. This may be used in the growth inhibition assay
  • a competitive assay can be performed involving the displacement of a radiolabeled compound with specificity for pathogen EF2 such as H-Compound I binding to Saccharomyces cerevisiae EF2 in crude S30 extracts.
  • pathogen EF2 such as H-Compound I binding to Saccharomyces cerevisiae EF2 in crude S30 extracts.
  • binding competition can also be performed with purified EF2 in the presence of washed ribosomes
  • H-Compound I Specific binding of H-Compound I is found with Saccharomyces S30 extracts and requires the presence of ribosomes as well as EF2. The binding is displaceable by unlabelled L-793,422, sordarin and analogs. No binding is seen with mammalian cell or wheat germ S30 extracts.
  • the specificity of H-L793,422 for yeast resides in the EF2 molecule since substitution of yeast ribosomes with either rat or wheat germ has no effect on binding, while substitution of yeast EF2 with rat or wheat germ EF2 abolishes binding.
  • Buffer A 50 mM Tris-HCl PH 7.5,150 mM NaCl,10 mM MgCl 2 ,1 mM EDTA
  • Buffer B 50 mMTris-HCl PH 7.5,10 mM MgCl 2, 1 mM EDTA GTP- ⁇ -s (Sigma)
  • Yeast S-30 prepared as below.
  • 3H-Compound I (20 mCi/mg, 8000mCi/mmol; 0.004mg/ml)
  • Saccharomyces S30 extract A Saccharomyces cerevisiae strain containing wild-type EF2 is inoculated into medium containing in g/1: lOg Bacto Yeast Extract, 20g Bacto Peptone, 20 g dextrose and 60mg adenine and incubated with shaking at 30°C until mid to late logarithmic phase (A600-2). The cells are harvested and washed twice with water. Pellets may be stored at -70°C indefinitely prior to disruption. For breakage, cells are resuspended in 2 vol of buffer containing 50 mM Tris-Cl pH 7.4, 10% (wt/vol) glycerol, 2mM MgCl 2 ,
  • This assay involves the same procedure as disclosed above substituting 0.4 A260 units salt-washed S. cerevisiae ribosomes and 1 pmol purified EF2 for the 10 ⁇ g S30 in the above assay.
  • the ribosomes and EF2 are both prepared by published methods (L. Skogerson,
  • this assay may be used to either identify the component binding drug, or to examine competition by unknown agents. Results
  • Saccharomyces EF2 0.65 pmol purified Saccharomyces EF2 plus 4 pmol purified Saccharomyces ribosomes binds approximately 0.6 pmol labelled compound, with similar displacement (IC50 3ng/ml) by active analogs.
  • Replacement of Saccharomyces ribosomes by those of either rat liver or wheat germ does not reduce binding.
  • Ribosomes, EF1, EF2 and EF3 are purified from Saccharomyces cerevisiae, and the assay is performed, as described in (L. Skogerson, Methods in Enzymology, Vol LX,p676-685). Sordarin, its analogs and any unknowns may be titrated in this assay and an IC 50 value determined for inhibition.
  • Ribosomes and EF2 from other eukaryotic systems may be purified as described in the literature. When EF2 from rat liver (prepared as described by J. F. Collins, F. Raeburn and E.S. Maxwell. J. Biol.
  • An assay has been developed to identify antifungal compounds with sordarin like activities using S. cerevisiae as as a surrogate organism. It consists of a two plate differential zone assay using sordarin sensitive (sSl) and resistant strains (sRl) that contain an ergo deletion, which increases membrane permeability and facilitates the uptake of various substances. In this screen, active compounds show a clear zone on the sensitive strain plate and no zone for the resistant strain plate.
  • Methods Approximately 1 x 10 ⁇ 6° cells per ml are added to growth medium containing 2% agar. Medium and cells are mixed, poured into plates, and allowed to solidify. Test compounds or fermentation extracts are applied with the intent of identifying samples which inhibit the growth of these yeast. The cultures are incubated at 30 C for 16-24 hours. A similar assay can also be run in a high-throughput microtiter format by inoculating cells into liquid growth medium containing test compounds or fermentation extracts. Active compounds can be identified by assaying for growth inhibition, which can be determined by measuring the optical density of the individual cultures.
  • sordarin gives a clear zone of 20mm with the sensitive strain and no zone with the resistant strain.

Abstract

Inhibition of protein elongation factor (2) provides a target for identifying potential antifungal and antiparasitic compounds. EF2 inhibitors are useful as therapeutic agents against fungal and parasitic infections.

Description

TITLE OF THE INVENTION
PROTEIN ELONGATION FACTOR 2 AS A TARGET
FOR ANTIFUNGAL AND ANTIPARASITIC AGENTS
BACKGROUND OF THE INVENTION
Elongation factor 2 (EF2) is an essential protein catalysing ribosomal translocation during protein synthesis in eukaryotic cells. It is highly conserved in all eukaryotes, and has been found to be largely interchangeable in in vitro protein synthesis systems reconstituted from such divergent organisms as human, wheat germ, and fungi. Despite the ubiquitous nature of EF2 in eukaryotic systems and the high degree of amino acid sequence homology between EF2s from various eukaryotic systems, a class of compounds, the sordarins, have now been identified to be selective inhibitors of fungal protein synthesis via a selective interaction with fungal EF2. This finding demonstrates the potential for developing pathogen selective EF2 inhibitors which can kill invading organisms while sparing the host of any detrimental effects. Prior to this invention, EF2 has not been considered as a differential target for antifungal or antiparasitic agents.
SUMMARY OF THE INVENTION
The present invention relates to elongation factor 2 (hereinafter referred to as "EF2") as a target for antifungal and antiparasitic agents. In particular, the invention relates to a method for identifying potential antifungal and antiparasitic agents by determining whether a test compound is capable of specifically inhibiting pathogenic protein synthesis via a selective interaction with pathogen EF2. The present invention describes the use of mechanism based assays with or without the use of a transformed eukaryotic organism with a heterologous EF2 to facilitate drug discovery. Additionally, the invention relates to a method for treating fungal infections by administering to a host suffering from a fungal or parasitic infection a therapeutically effective amount of a compound that specifically inhibits the pathogen's protein synthesis via EF2. DETAILED DESCRIPTION OF THE INVENTION
This invention provides a method for identifying and evaluating compounds having antifungal and antiparasitic activity comprising: A differential two plate assay containing genetically engineered sordarin sensitive (sSl) and resistant (sRl) strains or naturally selected sordarin resistant strains of yeast. The readout of the assay is antimicrobial activity indicated by zones of inhibition which is more apparent against the sordarin sensitive strain relative to the sordarin resistant strain. There are two EF2 genes in Saccharomyces cerevisiae,
EFT1 and EFT2, and at least one of these genes is required for survival. The co-isogenic strains sSl and sRl were constructed by a series of genetic crosses that result in strains that are disrupted for both the EFT1 and ERG6 genes. The resultant strains are made more permeable due to the ergo disruption (ergosterol deficient), and have either a wild-type or resistant copy of EFT2 as the only source of EF2. A known number of these yeast cells are either plated in solid medium or suspended in liquid medium and test compounds or fermentation extracts are applied with the intent of identifying samples which inhibit the growth of these yeast. The cultures are incubated at a specific temperature for a set period of time to allow for the growth of the test organisms (i.e. 30°C for 16-24 hours). Test samples of interest are those which show a differential effect on the sordarin sensitive strain(s) vs. the resistant strain(s). Those samples which are more potent against the wildtype by definition should be preventing growth via the EF2 target.
In another aspect the present invention provides a method for identifying compounds specifically inhibiting pathogenic EF2 function comprising: (a) constructing fungal or protozoan cells dependent upon heterologous EF2 from fungal and parasitic pathogens or from the host species; (b) contacting said cell with a known dilution of a test compound or a natural product extract; and (c) quantitating the minimal inhibitory concentration (MIC) of test compound to completely inhibit growth in liquid or the measurement of an inhibitory zone on a solid substrate. Test compounds or fermentation extracts of interest are those which display a differential degree of inhibition (i.e. more inhibitory activity against the wildtype vs. resistant strains of the test organisms). For example a sample which is more effective at inhibiting the growth of a yeast EF2 dependent organism vs. one that is dependent on human EF2.
The methods of the invention provides a facile and specific assay to screen compounds as potential antifungal and antiparasitic agents. It also allows for the evaluation of test compounds against the EF2 target of obligate pathogens that cannot be cultured in the laboratory.
In the present invention, EF2 may be cloned from pathogenic organisms for use in growth inhibition assays or purified from these pathogens for use in in vitro binding or translation inhibition assays. The EF2 may be from pathogenic fungi of humans, animals or plants such as Candida, Aspergillus spp., Cryptococcus spp., Erysiphe and Puccinia. It may also be from protozoan parasites such as Plasmodium sp., Eimeria sp., Crypto sporidium sp. and Toxoplasma gondii and human and other desired host eukaryotic cells.
A compound that inhibits EF2 may be one that interferes with the translation of mRNA in target organisms. Examples of compounds that inhibit EF2 include diptheria toxin and fusidic acid, however neither of these show any specificity for pathogen over host. Fusidic acid inhibits translation in many organisms by disrupting normal ribosome-EF2 interactions. The compound that inhibits EF2 is preferably labeled to allow easy quantitation of the level of interaction between the compound and the enzyme. A prefered radiolabel is tritium.
The test compound may be a synthetic compound, a mixture of synthetic compounds, a crude preparation, a purified preparation or an initial extract of a natural product obtained from plant, microorganism or animal sources.
It has been found that the antifungal agent sordarin and analogs thereof inhibit protein synthesis in certain pathogenic fungi by inhibiting the fungal EF2 function. Sordarin is an antifungal antibiotic isolated from the mould Sordaria araneosa (see GB 1,162,027 and Helvetica Chimica Acta, 1971, 51:119-20). Other compounds having the sordarin skeleton have also been reported as antifungal agents. Japanese Kokai J62040292 discloses the compound zofimarin isolated from Zofiela marina sp.; Japanese Kokai J06157582 discloses the compound BE-31405 isolated from Penicillium sp.; and SCH57404 is reported in J. Antibiotics. 1995, 48:1171-1172. Semi-synthetic sordarin derivatives are reported in PCT Applications W096/14326 and W096/14327. Sordaricin, the aglycone, may be obtained from sordarin by acid hydrolysis (Hauser and Sigg, Helvetica Chimica Acta, 1971, 51: 119-20); similarly sordaricin methyl ester is obtained from sordarin methyl ester. The total synthesis of sordaricin methyl ester is reported in Kato et al, J. Chem. Soc. Chem. Commun.. 1993, 1002-1004, which also discloses O-methoxymethyl sordaricin methyl ester. The diacetate of 4-desformyl-4-hydroxymethyl sordaricin is disclosed in Mander and Robinson. J. Org. Chem.. 1991, 56(11):3395-3601. Neither sordaricin nor the reported derivatives thereof has been shown to have biological activity. Sordarin analogs of the formula
Figure imgf000006_0001
wherein
R is (a) C(=0)ORl,
(b) C(=0)NR2R3,
(c) C(=0)R4
(d) CH(R2)OR5,
(e) C(R6)(R7)(R8),
Figure imgf000007_0001
with the proviso that when Rl2 is CHO, R is not (g); Rl is (a) C1-C14 alkyl,
(b) C2-C14 alkenyl,
(c) C2-C14 alkynyl,
(d) C3-C20 cycloalkyl, (e) aryl or
(f) aryl Cl-6 alkyl; R2 and R3 are independently
(a) H or (b) Rl; R4 is (a) H,
Figure imgf000007_0002
R5 is (a) R1 or
(b) -(CH2)χO(CH2)yH; R6 is (a) H,
(b) C1-C14 alkyl,
(c) aryl,
(d) aryl Cl-6 alkyl, (e) -(CH2)yCHRn(CH2)zH, (f) -(CH2)yC≡C(CH2)zH,
(g) -(CH2)yC(R7)=CH(CH2)zH, (h) -(CH2)yC≡C(CH2)mR1 1,
(i) -(CH2)yC(R7)=CH(CH2)mR1 1, R7 and R8 are independently
(a) H, or
(b) C1-C14 alkyl;
R9 and RIO are independently
(a) H,
(a) C1-C14 alkyl,
(a) C2-C14 alkenyl,
(a) aryl Cl-6 alkyl;
Rl l is (a) OH or
(b) NR2R3;
Rl2 is (a) -C(=0)Rl4,
(b) -CH=NOH, or
(c) -CH2OCH3;
Rl3 is (a) H,
(b) -CH2C6H5,
(c) -CH2CH=CH2,
Figure imgf000008_0001
Rl4 is (a) H,
(b) C1-C4 alkyl,
(c) -CC13,
( ) -CBr3, (e) -CF3, or
( ) OH; n is 0 or 1 ; m is 1-6; x is 2-6; y is 0-6; z is 0-6 or a pharmaceutically or agriculturally acceptable salt thereof are disclosed in pending U.S. application serial number 60/026,993 filed October 7, 1996. Additional analogs of sordarin are disclosed in U.S. patent application 60/026,580 filed September 18, 1996.
EF2 inhibitors are useful as antifungal and antiparasitic agents. As such, they may be used in the treatment and prevention of fungal and parasitic diseases in human, animals and plants. Examples of fungal diseases against which EF2 inhibitors may be used, and their respective causative pathogens, include: 1) Erysiphe, Puccinia, Septoria, Botrytis, Phytophthora, Plasmopora and other fungi which cause infections in plants and crops 2) Candida, Aspergillus, Cryptococcus, Fusarium, Penicillium and other fungi which cause fungal infections in man and animals 3) Plasmodium, Eimeria, Toxoplasma, Neospora, Cryptosporidium and other protozoa which infect man and animals. In another aspect the present invention provides a method for the treatment of fungal or parasitic infections comprising administering to a host suffering from a fungal or parasitic infection a therapeutically effective amount of a compound which inhibits EF2 function. A therapeutically effective amount may be one that is sufficient to inhibit protein synthesis of the causative fungi or parasite.
EF2 inhibitors may be administered to a host in need of treatment in a manner similar to that used for other antifungal and antiparasitic agents; for example, they may be administered parenter- ally, orally, topically, or rectally. The dosage to be administered will vary according to the particular compound used, the infectious organism involved, the particular host, the severity of the disease, physical condition of the host, and the selected route of administration; the appropriate dosage can be readily determined by a person skilled in the art. For the treatment of fungal and parasitic diseases in human and animals, the dosage may range from 0.01 mg/kg to 500 mg/kg. For prophylactic use in human and animals, the dosage may range from 0.01 mg/kg to 100 mg/kg. The compositions of the present invention comprises an
EF2 inhibitor and an inert carrier. The compositons may be in the form of pharmaceutical compositions for human and veterninary usage, or in the form of feed composition. The term "composition" is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions of one or more of the ingredients. The composition of the present invention thus includes a composition when made by admixing an EF2 inhibitor and inert carrier.
The pharmaceutical compositions of the present invention comprise an EF2 inhibitor as an active ingredient, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients. The compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administrations, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy. In practical use, an EF2 inhibitor can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed. For example, in the case of oral liquid preparations such as suspensions, elixirs and solutions, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like may be used; or in the case of oral solid preparations such as powders, capsules and tablets, carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be included. Because of their ease of. administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. In addition to the common dosage forms set out above, EF2 inhibitors may also be administered by controlled release means and/or delivery devices. Pharmaceutical compositions 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 or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion or a water-in-oil liquid emulsion. Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation. For example, a tablet may be prepared by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Desirably, each tablet contains from about 1 mg to about 500 mg of the active ingredient and each cachet or capsule contains from about 1 to about 500 mg of the active ingredient.
Pharmaceutical compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of these active compounds in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
Suitable topical formulations include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like. These formulations may be prepared via conventional methods containing the active ingredient. To illustrate, a cream or ointment is prepared by mixing sufficient quantities of hydrophilic material and water, containing from about 5-10% by weight of the compound, in sufficient quantities to produce a cream or ointment having the desired consistency.
Pharmaceutical 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 combination with the softened or melted carrier(s) followed by chilling and shaping moulds. It should be understood that in addition to the aforementioned carrier ingredients the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like, and. substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.
Compositions containing a compound of formula I may also be prepared in powder or liquid concentrate form . In accordance with standard veterinary formulation practice, conventional water soluble excipients, such as lactose or sucrose, may be incorporated in the powders to improve their physical properties. Thus particularly suitable powders of this invention comprise 50 to 100% w/w, and preferably 60 to 80% w/w of the combination and 0 to 50% w/w and preferably 20 to 40% w/w of conventional veterinary excipients. These powders may either be added to animal feedstuffs, for example by way of an intermediate premix, or diluted in animal drinking water.
Liquid concentrates of this invention suitably contain a water-soluble compound combination and may optionally include a veterinarily acceptable water miscible solvent, for example polyethylene glycol, propylene glycol, glycerol, glycerol formal or such a solvent mixed with up to 30% v/v of ethanol. The liquid concentrates may be administered to the drinking water of animals, particularly poultry. EXAMPLES The following non-limiting examples are provided to illustrate the invention. The assays may be run in 96 well or other appropriate sized plates or in the appropriate liquid medium.
5
PREPARATION OF MATERIALS USED IN THE EXAMPLES
Compound I of the formula
Figure imgf000014_0001
I Q Compound I
is a sordarin analog. The preparation of Compound I is described in U.S. application serial number 60/026,993 filed October 7, 1996.
Construction of Yeast Strain
15 The yeast strain YEFD 12h/pURA3-EFT 1 , that is deleted for both chromosomal copies of genes which encode EF2, has been obtained from the laboratory of James Bodley (Phan et al., Journal of Biological Chemistry (1993). 268:8665-8668). This strain also contains an episomal copy of a gene encoding EF2, and is essential for
20 cell viability. Yeast strains expressing EF2 genes from pathogens of interest may be constructed by (1) transforming YEFD12h/pURA3- EFT1 with yeast expression plasmids that contain heterologous EF2 encoding genes, and (2) evicting the plasmid containing the native EF2 gene from the cell. This may be used in the growth inhibition assay
25 described below.
EXAMPLE 1 COMPETITIVE BINDING ASSAY
A competitive assay can be performed involving the displacement of a radiolabeled compound with specificity for pathogen EF2 such as H-Compound I binding to Saccharomyces cerevisiae EF2 in crude S30 extracts. As proof of the specificity of inhibitor found with the S30 binding assay, binding competition can also be performed with purified EF2 in the presence of washed ribosomes
Specific binding of H-Compound I is found with Saccharomyces S30 extracts and requires the presence of ribosomes as well as EF2. The binding is displaceable by unlabelled L-793,422, sordarin and analogs. No binding is seen with mammalian cell or wheat germ S30 extracts. The specificity of H-L793,422 for yeast resides in the EF2 molecule since substitution of yeast ribosomes with either rat or wheat germ has no effect on binding, while substitution of yeast EF2 with rat or wheat germ EF2 abolishes binding.
Materials :
Sephadex G-75 (G-75-120) Mini column : GS-QS quick-sep micro column ( Isolab )
Mini vials (4 ml ) Scintiverse
Buffer A : 50 mM Tris-HCl PH 7.5,150 mM NaCl,10 mM MgCl 2 ,1 mM EDTA Buffer B : 50 mMTris-HCl PH 7.5,10 mM MgCl 2, 1 mM EDTA GTP-γ-s (Sigma) Yeast S-30,prepared as below. 3H-Compound I (20 mCi/mg, 8000mCi/mmol; 0.004mg/ml)
In a microfuge tube 100 μl assay mixture contains :
10 μg yeast S-30, 25 μM GTP-γ-s ( 0.5 μl of 5mM stock ), dilutions of agent to be examined for ability to compete for binding and Buffer B to bring volume to 98μl. Vortex and incubate at room temperature 5 min. Add 2 μl H-Compound I (1 : 20 dilution in water). Vortex and incubate for 20-30 min.
To a Sephadex G-75 column was pre-soaked in Buffer A ( 20 gm 400ml) several hours. Mini columns were packed with G-75 precisely to the mark line (-1.6 ml ) and allowed to settle by washing with 2 ml Buffer A.
The lOOμl incubated mixtures are loaded onto G-75 columns (no collection) As soon as the sample has entered the gel bed (approximately 20 sec), 0.7 ml Buffer A is added and eluate is collected in mini vials 3 ml Scintiverse is added and vials counted.
Saccharomyces S30 extract: A Saccharomyces cerevisiae strain containing wild-type EF2 is inoculated into medium containing in g/1: lOg Bacto Yeast Extract, 20g Bacto Peptone, 20 g dextrose and 60mg adenine and incubated with shaking at 30°C until mid to late logarithmic phase (A600-2). The cells are harvested and washed twice with water. Pellets may be stored at -70°C indefinitely prior to disruption. For breakage, cells are resuspended in 2 vol of buffer containing 50 mM Tris-Cl pH 7.4, 10% (wt/vol) glycerol, 2mM MgCl2,
2 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride and broken by prolonged agitation with acid-washed 0.5mm glass beads. The supernatant is centrifuged at 30g for 15 min to sediment cell debris.
RESULTS:
10 μg S30 extract from wild-type Saccharomyces binds approximately 0.5 pmol labelled sordarin analog H-Compound I. This is displaced with an IC50 of 3ng/ml by sordarin or its more active analogs.
BINDING ASSAY WITH PURIFIED COMPONENTS:
This assay involves the same procedure as disclosed above substituting 0.4 A260 units salt-washed S. cerevisiae ribosomes and 1 pmol purified EF2 for the 10 μg S30 in the above assay. The ribosomes and EF2 are both prepared by published methods (L. Skogerson,
Methods in Enzymology, Vol LX,p676-685).
As with the S30 binding assay, this assay may be used to either identify the component binding drug, or to examine competition by unknown agents. Results
0.65 pmol purified Saccharomyces EF2 plus 4 pmol purified Saccharomyces ribosomes binds approximately 0.6 pmol labelled compound, with similar displacement (IC50 3ng/ml) by active analogs. Replacement of Saccharomyces ribosomes by those of either rat liver or wheat germ does not reduce binding. However, replacement of Saccharomyces EF2 by EF2 from either of these sources, abolishes binding to background levels.
EXAMPLE 3
MEASUREMENT OF PROTEIN SYNTHESIS IN PATHOGEN/HOST Another method of identifying specific inhibitors of the pathogen protein synthesis is to measure the incorporation of radio- labeled amino acids into TCA precipitable proteins in the pathogen and the host. Test samples with activity that indicates pathogen selectivity can then be further screened in the more specific EF2 assays.
Reconstituted protein synthesis
This assay involves inhibition of polyphenylalanine synthesis in a reconstituted in vitro translation system Ribosomes, EF1, EF2 and EF3 are purified from Saccharomyces cerevisiae, and the assay is performed, as described in (L. Skogerson, Methods in Enzymology, Vol LX,p676-685). Sordarin, its analogs and any unknowns may be titrated in this assay and an IC50 value determined for inhibition. Ribosomes and EF2 from other eukaryotic systems may be purified as described in the literature. When EF2 from rat liver (prepared as described by J. F. Collins, F. Raeburn and E.S. Maxwell. J. Biol. Chem :246 pp 1049- 1064 [1971]) or wheat germ (prepared as described by S.J. Lauer, E. Burks, J.D. Irvin and J.M. Ravel. J. Biol. Chem. 259: ρpl644-1648 [1984]) is substituted for yeast EF2 in the S. cerevisiae system, no inhibition is found for the sordarin class of compounds up to levels limited by drug solubility. On the other hand, substitution of yeast ribosomes by rat liver or wheat germ ribosomes does not affect the ability of the sordarin class to inhibit the reconstituted translation system.
Results
IC O for inhibition
Figure imgf000018_0001
EXAMPLE 4
Growth Inhibition Assay I.
An assay has been developed to identify antifungal compounds with sordarin like activities using S. cerevisiae as as a surrogate organism. It consists of a two plate differential zone assay using sordarin sensitive (sSl) and resistant strains (sRl) that contain an ergo deletion, which increases membrane permeability and facilitates the uptake of various substances. In this screen, active compounds show a clear zone on the sensitive strain plate and no zone for the resistant strain plate.
Methods Approximately 1 x 10 Λ6° cells per ml are added to growth medium containing 2% agar. Medium and cells are mixed, poured into plates, and allowed to solidify. Test compounds or fermentation extracts are applied with the intent of identifying samples which inhibit the growth of these yeast. The cultures are incubated at 30 C for 16-24 hours. A similar assay can also be run in a high-throughput microtiter format by inoculating cells into liquid growth medium containing test compounds or fermentation extracts. Active compounds can be identified by assaying for growth inhibition, which can be determined by measuring the optical density of the individual cultures.
Results
0.5μg of sordarin gives a clear zone of 20mm with the sensitive strain and no zone with the resistant strain.

Claims

WHAT IS CLAIMED IS:
1. A method of identifying compounds which selectively inhibit fungal or parasitic protein synthesis by impairing the function of the fungal or parasitic elongation factor 2 (EF2).
2. EF2 as a specific target for antifungal and antiparasitic agents.
3. A method for identifying a compound which inhibits
EF2 comprising
(a) adding a compound to a genetically engineered eukaryotic organism capable of detecting pathogen selective effects on EF2 function, and (b) determining whether said compound inhibits protein synthesis by assaying growth inhibition.
4. A method for identifying a compound which selectively inhibits EF2 which comprises (a) constructing fungal or protozoan cells that express a heterologous EF2 from fungal and parasitic pathogens;
(b) inoculating cells into liquid or onto solid growth medium containing test compounds or fermentation extracts; and
(c) quantitating the minimal inhibitory concentration (MIC) of the test compound to completely inhibit growth in liquid or the measurement of an inhibitory zone on a solid substrate.
5. The method of Claim 4 wherein the cells are inoculated onto a solid growth medium and the MIC of the test compound is the measurement of an inhibitory zone on a solid substrate.
6. The method of Claim 4 wherein the cells are inoculated into liquid growth medium and the MIC of the test compound is the measurement of complete inhibition of growth in the liquid.
7. A method for identifying a compound which inhibits EF2 comprising
(a) plating a known dilution of cells on the appropriate medium;
(b) contacting said cells with a known dilution of a test compound or natural product extract;
(c) incubating said cells at about 30┬░C for about 16 to 24 hours; and (d) quantitating the percent of growth inhibition specific for loss of EF2 function due to the test compound or natural product extract.
8. The method of Claim 3 wherein said eukaryotic organism is Saccharomyces cerevisiae deleted or mutated for EF2.
9. A modified Saccharomyces cerevisiae cell wherein the cell expresses a heterologous EF2.
10. A method for identifying compounds having antifungal activity comprising:
(a) contacting EF2 or an extract containing EF2 with a known amount of a labeled compound that interacts with EF2; £b) contacting said EF2 or said extract with a known dilution of a test compound or a natural product extract; and
(c) quantitating the percent inhibition of interaction of said labeled compound induced by said test compound.
11. The method of Claim 10 which further comprises quantitating the percent inhibition of interaction with a host EF2 of said labeled compound induced by said test compound.
12. A method for the treatment or prevention of fungal diseases comprising administering to a host a therapeutically or prophylactically effective amount of a compound which inhibits EF2 function of the fungi.
13. A method for the treatment or prevention of parasitic diseases comprising administering to a host a therapeutically or prophylactically effective amount of a compound which inhibits EF2 function of the parasite.
14. The method of Claim 12 wherein said fungal disease is caused by Candida, Aspergillus, Cryptococcus, Fusarium, Penicillium or Dermatophytes.
15. The method of Claim 13 wherein said parasitic disease is caused by Plasmodium, Eimeria, Isospora, Neospora,
Toxoplasma, Cryptosporidium, Trypanosoma, Leishmania or Theileria.
16. The method of Claim 12 wherein said compound impairs fungal EF2 function to a greater extent than host EF2 function.
17. The method of Claim 13 wherein said compound impairs parasitic EF2 function to a greater extent than host EF2 function.
18. A composition useful for the prevention or treatment of fungal diseases which comprises an inert carrier and an effective amount of a compound that selectively impairs fungal or parasitic EF2 function.
19. The composition of Claim 18 wherein said compound which selectively impairs fungal or parasitic EF2 function is sordarin.
PCT/US1998/011903 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic agents WO1998057176A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002291653A CA2291653A1 (en) 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic agents
JP50307999A JP2002511933A (en) 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic drugs
EP98928944A EP0995120A1 (en) 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic agents
AU80627/98A AU741017B2 (en) 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US4927197P 1997-06-10 1997-06-10
US60/049,271 1997-06-10
GB9714331.7 1997-07-07
GBGB9714331.7A GB9714331D0 (en) 1997-07-07 1997-07-07 Protein elongation factor 2 as a target for antifungal and antiparasitic agents

Publications (1)

Publication Number Publication Date
WO1998057176A1 true WO1998057176A1 (en) 1998-12-17

Family

ID=26311848

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/011903 WO1998057176A1 (en) 1997-06-10 1998-06-09 Protein elongation factor 2 as a target for antifungal and antiparasitic agents

Country Status (5)

Country Link
EP (1) EP0995120A1 (en)
JP (1) JP2002511933A (en)
AU (1) AU741017B2 (en)
CA (1) CA2291653A1 (en)
WO (1) WO1998057176A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7885665B2 (en) 2003-09-26 2011-02-08 Siemens Enterprise Communications, Inc. System and method for failsafe presence monitoring

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992003455A1 (en) * 1990-08-16 1992-03-05 Isis Pharmaceutics, Inc. INHIBITION OF $i(CANDIDA)
US5403713A (en) * 1988-11-14 1995-04-04 Brigham & Women's Hospital Antibodies specific for ELAM-1 and the use thereof
US5641627A (en) * 1993-10-25 1997-06-24 Ribogene, Inc. Methods for screening for antimycotics

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5403713A (en) * 1988-11-14 1995-04-04 Brigham & Women's Hospital Antibodies specific for ELAM-1 and the use thereof
WO1992003455A1 (en) * 1990-08-16 1992-03-05 Isis Pharmaceutics, Inc. INHIBITION OF $i(CANDIDA)
US5641627A (en) * 1993-10-25 1997-06-24 Ribogene, Inc. Methods for screening for antimycotics

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
JONES D. E., ET AL.: "MOLECULAR CLONING AND CHARACTERIZATION OF A CRYPTOSPORIDIUM PARVUM ELONGATION FACTOR-2 GENE.", MOLECULAR AND BIOCHEMICAL PARASITOLOGY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 71., 1 February 1995 (1995-02-01), NL, pages 143 - 147., XP002914520, ISSN: 0166-6851, DOI: 10.1016/0166-6851(95)00051-2 *

Also Published As

Publication number Publication date
JP2002511933A (en) 2002-04-16
AU8062798A (en) 1998-12-30
AU741017B2 (en) 2001-11-22
EP0995120A1 (en) 2000-04-26
CA2291653A1 (en) 1998-12-17

Similar Documents

Publication Publication Date Title
Van den Bossche et al. In vitro and in vivo effects of the antimycotic drug ketoconazole on sterol synthesis
Hall et al. The quorum-sensing molecules farnesol/homoserine lactone and dodecanol operate via distinct modes of action in Candida albicans
Urbina et al. Inhibitors of the fungal cell wall. Synthesis of 4-aryl-4-N-arylamine-1-butenes and related compounds with inhibitory activities on β (1–3) glucan and chitin synthases
JP4376297B2 (en) Method for screening for compounds that inhibit GPI biosynthesis in Plasmodium
Seyedmousavi et al. Emerging Aspergillus species almost exclusively associated with primary immunodeficiencies
Kozák et al. Tremorgenic and neurotoxic paspaline-derived indole-diterpenes: biosynthetic diversity, threats and applications
US6303302B1 (en) Regulation of fungal gene expression
Bertrand et al. N*-methyl coprogen B, a potential marker of the airway colonization by Scedosporium apiospermum in patients with cystic fibrosis
Monod et al. Secreted proteinases and other virulence mechanisms of Candida albicans
US6096511A (en) Protein elongation factor 2 as a target for antifungal and antiparasitic agents
Uchida et al. In vitro activity of novel imidazole antifungal agent NND-502 against Malassezia species
Kouvelis et al. Assessing the cytotoxic and mutagenic effects of secondary metabolites produced by several fungal biological control agents with the Ames assay and the VITOTOX® test
Weichert et al. Functional coupling between the unfolded protein response and endoplasmic reticulum/Golgi Ca2+-ATPases promotes stress tolerance, cell wall biosynthesis, and virulence of Aspergillus fumigatus
Strijbis et al. Carnitine-dependent transport of acetyl coenzyme A in Candida albicans is essential for growth on nonfermentable carbon sources and contributes to biofilm formation
Breakspear et al. Npc1 is involved in sterol trafficking in the filamentous fungus Fusarium graminearum
François et al. Alternative identification test relying upon sexual reproductive abilities of Candida lusitaniae strains isolated from hospitalized patients
AU741017B2 (en) Protein elongation factor 2 as a target for antifungal and antiparasitic agents
US20080152641A1 (en) Inhibitors of Siderophore Biosynthesis in Fungi
US20020160441A1 (en) Protein elongation factor 2 as a target for antifungal and antiparasitic agents
JPH10501408A (en) DNA encoding GLS1
Ulaszewski et al. A new mutation for multiple drug resistance and modified plasma membrane ATPase activity in Schizosaccharomyces pombe
PT626454E (en) FUNGUS POLAR FODDER SEARCH FOR FUNGICIDES
Hollomon A laboratory assay to determine the sensitivity of Rhynchosporium secalis to the fungicide triadimenol
Luis et al. Development of sexual structures influences metabolomic and transcriptomic profiles in Aspergillus flavus
Zhang et al. The usnic acid derivative peziculone targets cell walls of Gram-positive bacteria revealed by high-throughput CRISPRi-seq analysis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE GW HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN YU

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI 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: 2291653

Country of ref document: CA

Ref country code: CA

Ref document number: 2291653

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998928944

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 80627/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998928944

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 80627/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998928944

Country of ref document: EP