WO1998054308A2 - Polyketides and their synthesis and use - Google Patents

Polyketides and their synthesis and use Download PDF

Info

Publication number
WO1998054308A2
WO1998054308A2 PCT/GB1998/001559 GB9801559W WO9854308A2 WO 1998054308 A2 WO1998054308 A2 WO 1998054308A2 GB 9801559 W GB9801559 W GB 9801559W WO 9854308 A2 WO9854308 A2 WO 9854308A2
Authority
WO
WIPO (PCT)
Prior art keywords
rapamycin
gene
gene cluster
precursor
cluster
Prior art date
Application number
PCT/GB1998/001559
Other languages
French (fr)
Other versions
WO1998054308A3 (en
Inventor
Peter Francis Leadlay
James Staunton
Lake Ee Khaw
Original Assignee
Biotica Technology 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 Biotica Technology Limited filed Critical Biotica Technology Limited
Priority to AT98924463T priority Critical patent/ATE307198T1/en
Priority to DE69831943T priority patent/DE69831943T2/en
Priority to EP98924463A priority patent/EP0983348B1/en
Priority to AU76661/98A priority patent/AU7666198A/en
Publication of WO1998054308A2 publication Critical patent/WO1998054308A2/en
Publication of WO1998054308A3 publication Critical patent/WO1998054308A3/en
Priority to US10/307,595 priority patent/US7018808B2/en
Priority to US11/328,642 priority patent/US7381546B2/en
Priority to US12/129,295 priority patent/US20080287483A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H17/00Compounds containing heterocyclic radicals directly attached to hetero atoms of saccharide radicals
    • C07H17/04Heterocyclic radicals containing only oxygen as ring hetero atoms
    • C07H17/08Hetero rings containing eight or more ring members, e.g. erythromycins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/88Lyases (4.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/18Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms containing at least two hetero rings condensed among themselves or condensed with a common carbocyclic ring system, e.g. rifamycin
    • C12P17/182Heterocyclic compounds containing nitrogen atoms as the only ring heteroatoms in the condensed system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/44Preparation of O-glycosides, e.g. glucosides
    • C12P19/60Preparation of O-glycosides, e.g. glucosides having an oxygen of the saccharide radical directly bound to a non-saccharide heterocyclic ring or a condensed ring system containing a non-saccharide heterocyclic ring, e.g. coumermycin, novobiocin
    • C12P19/62Preparation of O-glycosides, e.g. glucosides having an oxygen of the saccharide radical directly bound to a non-saccharide heterocyclic ring or a condensed ring system containing a non-saccharide heterocyclic ring, e.g. coumermycin, novobiocin the hetero ring having eight or more ring members and only oxygen as ring hetero atoms, e.g. erythromycin, spiramycin, nystatin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to polyketides and their synthesis and use. It is particularly, but not exclusively, concerned with variants of rapamycin.
  • Rapamycin (see Fig. 2) is a lipophilic macrolide, of molecular weight 914, with a 1, 2, 3-tricarbonyl moiety linked to a pipecolic acid lactone . Sequencing of the putative biosynthetic genes of rapamycin has revealed the presence of three exceptionally large open reading frames encoding the modular polyketide synthase (Schwecke et al., P.N.A.S. 92 (17) 7839-7843 (1995)). On either side of these very large genes are ranged open reading frames which appear to encode enzymes that would be required for rapamycin biosynthesis.
  • the cluster also contains a novel gene (rapL) whose product is proposed to catalyse the formation of the rapamycin precursor L-pipecolate (2) through the cyclodeamination of L-lysine (1) (Molnar et al., Gene 169, 1-7 (1996) ) :-
  • rapL novel gene
  • rapamycin requires a modular polyketide synthase, which uses a shikimate-derived starter unit and which carries out a total of fourteen successive cycles of polyketide chain elongation that resemble the steps in fatty acid biosynthesis.
  • L- pipecolic acid is then incorporated into the chain, followed by closure of the macrocyclic ring, and both these steps are believed to be catalyzed by a pipecolate- incorporating enzyme (PIE), the product of the rapP gene. Further site-specific oxidations and O-methylation steps are then required to produce rapamycin.
  • PIE pipecolate- incorporating enzyme
  • the organism cannot produce rapamycin under normal growth conditions but can do so if fed pipecolate. Furthermore feeding the mutant organism with different substrates leads to the production of variants of rapamycin.
  • the same general method can be applied to other systems which involve a precursor compound which is produced by a gene product, e.g. the very closely related FK506 and immunomycin systems which also " involve pipecolate.
  • a process of modifying a gene cluster involved in the biosynthesis of a polyketide said gene cluster including a gene ("the precursor gene") responsible for the production of an enzyme which is responsible for the production of a precursor compound which is incorporated into said polyketide; said process comprising the step of deleting or inactivating said precursor gene.
  • said process of deleting or inactivating said precursor gene employs phage-mediated gene replacement .
  • the gene cluster is the gene cluster for the production of rapamycin in S . hycrroscopicus and the precursor gene is the rapL gene whose product is responsible for the production of L-pipecolate .
  • the invention provides a process for producing a polyketide comprising modifying a gene cluster by the process according to the first aspect and expressing the modified gene cluster in the presence of a variant precursor compound which is incorporated so that a variant polyketide is produced.
  • a variant precursor compound for the rapamycin system, examples include L-proline, L-trans-4-hydroxyproline, L-cis-4- hydroxyproline, L-cis-3-hydroxyproline and trans-3-aza- bicyclo [3,1,0] hexane-2-carboxylic acid.
  • the invention provides polyketides as producible by the above method, pharmaceuticals comprising such polyketides, and the use of such polyketides in preparing pharmaceutical compositions, e.g. immunosuppressants containing rapamycin variants.
  • Fig. 1 shows a portion of the rapamycin gene cluster, wild type and mutated, and the phage vector used to perform mutation;
  • Fig. 2 shows structures of rapamycin and some variants
  • Figs. 3 and 4 illustrate the effects of rapamycin and variants on human lymphoblastoid cell lines.
  • a chromosomal mutant of S . hycrroscopicus was created by phage ⁇ C31-mediated gene replacement using the method of Lomovskaya et al . [Microbiology (UK) 1997, 143 , 815-883] .
  • a unique BamH I site was found 42bp into the rapL gene (1032bp long) . This BamH I site was removed by end-filling with E.coli DNA polymerase I thus creating a frameshift in the rapL gene.
  • Escherichia coli DH10B Bacterial strains, phages and growth conditions Escherichia coli DH10B (GibcoBRL) was grown in
  • the rapamycin producer Streptomyces hygroscopicus NRRL 5491 (from ATCC) and its derivatives were maintained on SY agar (Soluble starch 1.5%; yeast extract 0.1%; K 2 HP0 4 0.1%; MgS0 4 x 7 H 2 0 0.1%; NaCl 0.3%; N-tri [Hydroxymethyl] methyl-2- aminoethanesulfonic acid (Tes) buffer 30 mM, pH7.4 ; agar 1.5%), and cultivated in Tryptic Soy Broth with 1.0% glucose, lOOmM MES pH6.0 , supplemented with 10 ug/ml viomycin when required.
  • SY agar Soluble starch 1.5%; yeast extract 0.1%; K 2 HP0 4 0.1%; MgS0 4 x 7 H 2 0 0.1%; NaCl 0.3%; N-tri [Hydroxymethyl] methyl-2- aminoethanesulfonic acid (Tes) buffer 30
  • ⁇ rapL carrying a frameshi-ft in the rapL gene for homologous recombination in S .hygroscopicus pUC3EcoRI was constructed by cloning a 3034bp Eco RI fragment (nucleotides 93956 to 96990 of the rap cluster) (T. Schwecke et al . , P.N.A.S. 92. 7839-7843 (1995)) encompassing the entire rapL gene flanked by rapK and part of the rapM genes respectively into an Eco Rl-cut pUC18 modified vector where the Bam HI site in the polylinker region has been removed.
  • a unique Bam HI site (starting at nucleotide 95036 of the rap cluster) was found 42bp into the rapL gene (nucleotide 95078 to 94047 of the rap cluster; 1032bp long) .
  • Plasmid pUC3Eco RI was digested with Bam HI and the cohesive ends were filled in by treating it with E. coli DNA polymerase I (Klenow fragment) .
  • the ligated plasmid DNA was redigested with Bam HI and used to transform E. coli . Ampicillin resistant transformants were selected and their plasmid DNA checked for the removal of the Bam HI site by restriction enzyme analysis. This was confirmed by DNA sequencing.
  • the 3kb insert was excised from the plasmid with Eco RI and the cohesive ends blunt-ended by treatment with E. coli DNA polymerase I (Klenow fragment) .
  • the blunt -ended insert was cloned into Pvu II cut phage vector KC515, resulting in ⁇ rapL.
  • Protoplasts of S . lividans J11326 were transfected with the phage construct as described by Hopwood et al . (1985) . Recombinant phage was identified using PCR analysis. Infection of S .
  • hygroscopicus NRRL 5491 with ⁇ rapL was done according to Lomovskaya ete al (Microbiology, 143, 875-883 (1997)) on DNA plates supplemented with glucose, MgS0 4 and Ca(N0 3 ) . Lysogens were selected by overlaying the plates with 50 ⁇ g ml "1 (final concentration) viomycin 24 h post-infection. Strains that had undergone a second recombination event deleting the integrated phage were identified by selecting viomycin sensitive isolates after three rounds of non-selective growth and sporulation on SY plates. The insertion and subsequent loss of the phage were confirmed by genomic Southern hybridizations.
  • Precursor feeding and fermentation of S . hygroscopicus ⁇ RapL Precursor feeding of S .hygroscopicus ⁇ RapL was performed routinely in 500 ml flasks containing 100ml of Tryptic Soy Broth with 1.0% glucose, lOOmM MES pH6.0 , supplemented with the appropriate pipecolic acid analogue, at a final concentration of lmg/ml .
  • S . hygroscopicus ⁇ RapL was also cultivated in 2 1 flasks containing 400 ml of chemically-defined media as described by Cheng et al (Appl . Microbiol . Biotechnol . 43 , 1096-1098, (1995)).
  • Trans 4-hydroxyproline was added to the medium aseptically to a final concentration of lmg/ml.
  • the fermentation was carried out at 28°C for 4 days, with an agitation rate of 500 rpm.
  • the cells were harvested and extracted with twice their volume of methanol overnight . Purification and analysis of rapamycin and its derivatives
  • Rapamycin induces a specific cell cycle arrest in Gl in the cell line 536, which is a human B lymphocytic line immortalised by Epstein Barr virus infection.
  • the potency of each analogue was compared to that of rapamycin using the 536 cells as a bioassay.
  • the 536 cells obtained from the human genetic mutant cell repository, Camden, New Jersey, USA) were cultured in Iscoves medium supplemented with 10% fetal calf serum. For bioassay, 536 cells were seeded into 96 well microtitre plates at 10,000 per well in lOO ⁇ l of growth medium.
  • Drug stocks of ImM in DMSO were prepared and further dilutions were made to give a constant final concentration of 0.1% DMSO in growth medium.
  • Control cultures were treated with 0.1% DMSO in growth medium; experimental cultures received a final concentration of 10 "7 M, 10 "8 M, 10 "9 M or 10 "10 M rapamycin or rapamycin analogue.
  • Each culture was set up in triplicate and replicate plates were labelled with l ⁇ Ci tritiated thymidine (Amersham International, specific activity 70Ci/mM) per well for 3h at either Oh; 24h; or 48h incubation with drugs.
  • the cultures were harvested onto glass fibre paper to trap the DNA following water lysis; free nucleotides were washed away. Radioactivity incorporated into the filter discs / trapped DNA was counted in a Packard scintillation counter using biodegradable scintillation fluid.
  • ⁇ RapL was isolated as described in Materials and Methods.
  • This mutation was investigated by Southern blot hybridization using the 3kb EcoRI fragment (93956-96990) to probe Bgl II/Bam HI digested chromosomal DNA. Analysis of the wild-type S . hygroscopicus shows the expected 5.9kb (representing nucleotides 89118-95036) and 2.7kb Bam HI/Bgl II fragments (representing nucleotides 95036-97710) after hybridisation.
  • HPLC-ESIMS High pressure liquid chromatograph-electrospray ionization mass spectrometry
  • the methanol extracts were combined and concentrated under reduced pressure.
  • the aqueous residue was diluted with 500 mL of distilled water, and extracted three times with 500 mL of distilled ethyl acetate.
  • the combined ethyl acetate extracts were dried with anhydrous sodium sulphate, and evaporated to dryness .
  • the resulting yellow residue was purified by flash column chromatography on a 150 mm x 30 mm (diameter) silica gel column [Merck 60] eluted isocratically witeh a 1:1 (v/v) mixture of acetone/hexane .
  • the fractions were analysed by electrospray mass spectrometry. MS-MS and Ms n were used to determine the structure of the new rapamycin in the fractions from the flash silica column.
  • the fractions containing trans-3-aza- bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin were further purified by reversed-phase preparative HPLC on a 250 x 20 mm (diameter) Prodigy 0DS3 column (Phenomenex) using gradient elution starting at 70/30 (v/v) acetonitrile/water rising linearly to 100% acetonitrile over 25 minutes.
  • the 2 L fermentation yielded about 4 mg of pure trans-3-aza-bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin.
  • the dose response of human lymphoblastoid cell lines 536 was measured.
  • the mean cpm of radiolabelled thymidine incorporated into the untreated controls shows that 0-3 h drug exposure had no appreciable effect on DNA synthesis up to lOOnM of rapamycin, prolylrapamycin, or 4-hydroxy-prolyl-26- demethoxy-rapamycin. This implies that none of the compounds were toxic to the 536 cell line.
  • the 536 cells showed a concentration- dependent inhibition of DNA synthesis with an ID50% of InM for rapamycin; and 3nM for prolylrapamycin.
  • rapamycin 4- hydroxy-prolyl-26-demethoxy-rapamycin was also inhibitory but did not reach 50% at lOOnM.
  • Previous experiments have shown that rapamycin is a profound inhibitor of Gl progression in the 536 cell line (Metcalfe et al . , Oncogene 15, 1635-1642 (1997)). This is also suggested in these experiments for the rapamycin analogues, since no significant effect was found at 3h but inhibition was observed once the cell population had time to proceed through a complete cell cycle (24h) and reach the drug arrest point .

Abstract

The rapamycin gene cluster is an example of a gene cluster which includes a gene (rapL) leading to the formation of a precursor compound (pipecolic acid, in this case) which is required for inclusion in the larger product (rapamycin) produced by the enzymes encoded by the cluster. We have produced a mutant strain containing a rapamycin gene cluster in which the rapL gene is disabled. The strain does not naturally produce rapamycin but does so if fed with pipecolic acid. By feeding with alternative carboxylic acids we have produced variants of rapamycins. Tests have shown biological activity.

Description

POLYKETIDES AND THEIR SYNTHESIS AND USE
The present invention relates to polyketides and their synthesis and use. It is particularly, but not exclusively, concerned with variants of rapamycin.
Rapamycin (see Fig. 2) is a lipophilic macrolide, of molecular weight 914, with a 1, 2, 3-tricarbonyl moiety linked to a pipecolic acid lactone . Sequencing of the putative biosynthetic genes of rapamycin has revealed the presence of three exceptionally large open reading frames encoding the modular polyketide synthase (Schwecke et al., P.N.A.S. 92 (17) 7839-7843 (1995)). On either side of these very large genes are ranged open reading frames which appear to encode enzymes that would be required for rapamycin biosynthesis.
The cluster also contains a novel gene (rapL) whose product is proposed to catalyse the formation of the rapamycin precursor L-pipecolate (2) through the cyclodeamination of L-lysine (1) (Molnar et al., Gene 169, 1-7 (1996) ) :-
Figure imgf000003_0001
The biosynthesis of rapamycin requires a modular polyketide synthase, which uses a shikimate-derived starter unit and which carries out a total of fourteen successive cycles of polyketide chain elongation that resemble the steps in fatty acid biosynthesis. L- pipecolic acid is then incorporated into the chain, followed by closure of the macrocyclic ring, and both these steps are believed to be catalyzed by a pipecolate- incorporating enzyme (PIE), the product of the rapP gene. Further site-specific oxidations and O-methylation steps are then required to produce rapamycin. We have now found that we can genetically engineer an S . hycrroscopicus organism in which the (rapL) gene is inactivated. The organism cannot produce rapamycin under normal growth conditions but can do so if fed pipecolate. Furthermore feeding the mutant organism with different substrates leads to the production of variants of rapamycin. The same general method can be applied to other systems which involve a precursor compound which is produced by a gene product, e.g. the very closely related FK506 and immunomycin systems which also "involve pipecolate.
Thus according to the present invention in a first aspect there is provided a process of modifying a gene cluster involved in the biosynthesis of a polyketide, said gene cluster including a gene ("the precursor gene") responsible for the production of an enzyme which is responsible for the production of a precursor compound which is incorporated into said polyketide; said process comprising the step of deleting or inactivating said precursor gene. Suitably said process of deleting or inactivating said precursor gene employs phage-mediated gene replacement . In preferred embodiments of the invention the gene cluster is the gene cluster for the production of rapamycin in S . hycrroscopicus and the precursor gene is the rapL gene whose product is responsible for the production of L-pipecolate .
In a second aspect the invention provides a process for producing a polyketide comprising modifying a gene cluster by the process according to the first aspect and expressing the modified gene cluster in the presence of a variant precursor compound which is incorporated so that a variant polyketide is produced. For the rapamycin system, examples of the variant precursor compound include L-proline, L-trans-4-hydroxyproline, L-cis-4- hydroxyproline, L-cis-3-hydroxyproline and trans-3-aza- bicyclo [3,1,0] hexane-2-carboxylic acid.
In further aspects the invention provides polyketides as producible by the above method, pharmaceuticals comprising such polyketides, and the use of such polyketides in preparing pharmaceutical compositions, e.g. immunosuppressants containing rapamycin variants.
Some embodiments of the invention will now be described in greater detail with reference to the accompanying drawings in which;
Fig. 1 shows a portion of the rapamycin gene cluster, wild type and mutated, and the phage vector used to perform mutation;
Fig. 2 shows structures of rapamycin and some variants; and
Figs. 3 and 4 illustrate the effects of rapamycin and variants on human lymphoblastoid cell lines.
In order to facilitate the production of variant rapamycins, a chromosomal mutant of S . hycrroscopicus was created by phage φC31-mediated gene replacement using the method of Lomovskaya et al . [Microbiology (UK) 1997, 143 , 815-883] . A unique BamH I site was found 42bp into the rapL gene (1032bp long) . This BamH I site was removed by end-filling with E.coli DNA polymerase I thus creating a frameshift in the rapL gene. A 3 kb EcoR I fragment encompassing the entire rapL gene flanked by rapK and part of the rapM genes respectively was cloned into the phase vector, KC515. The recombinant phage was used to transfect S .hygroscopicus . A double recombination event resulted in the creation of a chromosomal mutant of S. hygroscopicus with a frameshift in rapL'.. This is summarised in Fig. 1.
Materials and Methods
Note: the reader is also referred to L.E. Khaw et. al. , J. Bacteriol. , 180 (4) 809-814 (1998) which is incorporated herein by reference, for both experimental details and discussion of the work and the background thereto .
Materials. All molecular biology enzymes and reagents were from commercial sources. Viomycin was a gift from Pfizer, L-pipecolic acid, L-proline, 3,4- dehydroproline, picolinic acid, pyrrole-2-carboxylic acid, trans 4-hydroxyproline, cis 4-hydroxyproline, cis 3-hydroxyproline and (+) -trans-3-aza-bicyclo
[3 , 1, 0] hexane-2-carboxylic acid were obtained from Aldrich Chemical Company.
Bacterial strains, phages and growth conditions Escherichia coli DH10B (GibcoBRL) was grown in
2x (tryptone-yeast extract) medium as described by Sambrook et al , ("Molecular Cloning", Cold Spring Harbor (1989)) . Vector pUC18 was obtained from New England Biolabs. or Sigma Chemical Co. E. coli transformants were selected with 100 mg/ml ampicillin. The rapamycin producer Streptomyces hygroscopicus NRRL 5491 (from ATCC) and its derivatives were maintained on SY agar (Soluble starch 1.5%; yeast extract 0.1%; K2HP04 0.1%; MgS04 x 7 H20 0.1%; NaCl 0.3%; N-tri [Hydroxymethyl] methyl-2- aminoethanesulfonic acid (Tes) buffer 30 mM, pH7.4 ; agar 1.5%), and cultivated in Tryptic Soy Broth with 1.0% glucose, lOOmM MES pH6.0 , supplemented with 10 ug/ml viomycin when required. S.lividans J11326 (D A Hopwood et al : "Genetic Manipulation of Streptomyces : a laboratory manual". The John Innes Foundation, Norwich, England (1985)) was cultivated in YEME (Hopwood et al . , 1985) or Tap Water Medium (0.5% glucose; 1% sucrose; 0.5% tryptone; 0.25% yeast extract; 36mg EDTA; pH 7.1). Liquid cultures were grown at 30°C in Erlenmeyer flasks with shaking at 200-250 rpm. Infection with the atr actinophage KC515 (Hopwood (1985) op. cit. and K. F. Chater in: "The Bacteria" IX (119-158) , New York 1986) and its derivative ΦΔrapL (present work) were done on solid DNA medium supplemented with lOmM MgS04, 8mM Ca(N03) and 0.5% glucose (Hopwood et al . , 1985).
Isolation and in vitro manipulation of DNA DNA manipulations, PCR and electroporation procedures were carried out as described in Sambrook et al (1989) . Total S . hygroscopicus DNA was isolated using the Gibco genomic DNA isolation kit . Southern hybridizations were carried out with probes labelled with digoxigenin using the DIG DNA labelling kit (Boehringer Mannheim) . DNA fragments for labelling and subcloning were isolated with the Qiaex (Qiagen) gel extraction kit.
Construction of ΦΔrapL carrying a frameshi-ft in the rapL gene for homologous recombination in S .hygroscopicus pUC3EcoRI was constructed by cloning a 3034bp Eco RI fragment (nucleotides 93956 to 96990 of the rap cluster) (T. Schwecke et al . , P.N.A.S. 92. 7839-7843 (1995)) encompassing the entire rapL gene flanked by rapK and part of the rapM genes respectively into an Eco Rl-cut pUC18 modified vector where the Bam HI site in the polylinker region has been removed. A unique Bam HI site (starting at nucleotide 95036 of the rap cluster) was found 42bp into the rapL gene (nucleotide 95078 to 94047 of the rap cluster; 1032bp long) . Plasmid pUC3Eco RI was digested with Bam HI and the cohesive ends were filled in by treating it with E. coli DNA polymerase I (Klenow fragment) . The ligated plasmid DNA was redigested with Bam HI and used to transform E. coli . Ampicillin resistant transformants were selected and their plasmid DNA checked for the removal of the Bam HI site by restriction enzyme analysis. This was confirmed by DNA sequencing. The 3kb insert was excised from the plasmid with Eco RI and the cohesive ends blunt-ended by treatment with E. coli DNA polymerase I (Klenow fragment) . The blunt -ended insert was cloned into Pvu II cut phage vector KC515, resulting in ΦΔrapL. Protoplasts of S . lividans J11326 were transfected with the phage construct as described by Hopwood et al . (1985) . Recombinant phage was identified using PCR analysis. Infection of S . hygroscopicus NRRL 5491 with ΦΔrapL was done according to Lomovskaya ete al (Microbiology, 143, 875-883 (1997)) on DNA plates supplemented with glucose, MgS04 and Ca(N03) . Lysogens were selected by overlaying the plates with 50 μg ml"1 (final concentration) viomycin 24 h post-infection. Strains that had undergone a second recombination event deleting the integrated phage were identified by selecting viomycin sensitive isolates after three rounds of non-selective growth and sporulation on SY plates. The insertion and subsequent loss of the phage were confirmed by genomic Southern hybridizations.
Precursor feeding and fermentation of S . hygroscopicus ΔRapL Precursor feeding of S .hygroscopicus ΔRapL was performed routinely in 500 ml flasks containing 100ml of Tryptic Soy Broth with 1.0% glucose, lOOmM MES pH6.0 , supplemented with the appropriate pipecolic acid analogue, at a final concentration of lmg/ml . S . hygroscopicus ΔRapL was also cultivated in 2 1 flasks containing 400 ml of chemically-defined media as described by Cheng et al (Appl . Microbiol . Biotechnol . 43 , 1096-1098, (1995)). For large scale fermentation, 10 μl of spores of S . hygroscopicus ΔRapL was used to inoculate a 100 ml flask containing 30 ml of Tryptic Soy Broth medium. The flask was incubated on a rotary shaker (300 rpm) at 28°C for 4 days. 4 ml of the first seed culture was transferred to a 2 1 flask (second seed culture) containing 400 ml of the medium nd incubated on a rotary shaker (300 rpm) at 28°C for 4 days. The second seed culture was transferred to a 201 fermenter containing 15 1 of the medium. Trans 4-hydroxyproline was added to the medium aseptically to a final concentration of lmg/ml. The fermentation was carried out at 28°C for 4 days, with an agitation rate of 500 rpm. The cells were harvested and extracted with twice their volume of methanol overnight . Purification and analysis of rapamycin and its derivatives
After 3-4 days fermentation mycelia were collected by filtration and extracted with two volumes of methanol at room temperature for 1 h. The crude extracts were analysed by lc-ms using a Finnigan MAT (San Jose, CA) LCQ with a Hewlett-Packard 1100 HPLC. The large scale fermentation was worked up similarly. The crude extract was evaporated to dryness and then purified by flash chromatography (Merck silica gel 60, no. 9385) with acetone/hexane l/l. The fractions containing rapamycins were further purified by preparative HPLC on a 250 x 20 mm RP18 column (HPLC Technology, Macclesfield, UK) using standard conditions. The 15 1 fermentation yielded about 15 mg of pure prolyl-rapamycin and 3 mg of 4-hydroxy- prolyl-26-demethoxy-rapamycin. NMR spectra were determined on a Bruker DRX 500 spectrometer.
Biological activity of rapamycin analogues Rapamycin induces a specific cell cycle arrest in Gl in the cell line 536, which is a human B lymphocytic line immortalised by Epstein Barr virus infection. The potency of each analogue was compared to that of rapamycin using the 536 cells as a bioassay. The 536 cells (obtained from the human genetic mutant cell repository, Camden, New Jersey, USA) were cultured in Iscoves medium supplemented with 10% fetal calf serum. For bioassay, 536 cells were seeded into 96 well microtitre plates at 10,000 per well in lOOμl of growth medium. Drug stocks of ImM in DMSO were prepared and further dilutions were made to give a constant final concentration of 0.1% DMSO in growth medium. Control cultures were treated with 0.1% DMSO in growth medium; experimental cultures received a final concentration of 10"7M, 10"8M, 10"9M or 10"10M rapamycin or rapamycin analogue. Each culture was set up in triplicate and replicate plates were labelled with lμCi tritiated thymidine (Amersham International, specific activity 70Ci/mM) per well for 3h at either Oh; 24h; or 48h incubation with drugs. At the respective time points the cultures were harvested onto glass fibre paper to trap the DNA following water lysis; free nucleotides were washed away. Radioactivity incorporated into the filter discs / trapped DNA was counted in a Packard scintillation counter using biodegradable scintillation fluid.
Results
Characterisation of a frameshift chromosomal mutation in the rapL gene
To confirm that the rapL gene product is indeed involved in the biosynthesis of rapamycin as a precursor feeder, the frameshift chromosomal mutant S . hygroscopicus
ΔRapL was isolated as described in Materials and Methods.
This mutation was investigated by Southern blot hybridization using the 3kb EcoRI fragment (93956-96990) to probe Bgl II/Bam HI digested chromosomal DNA. Analysis of the wild-type S . hygroscopicus shows the expected 5.9kb (representing nucleotides 89118-95036) and 2.7kb Bam HI/Bgl II fragments (representing nucleotides 95036-97710) after hybridisation. When chromosomal DNA of S .hygroscopicus ΔRapL was treated similarly, only a 8.6kb Bam HI/Bgl II fragment (representing nucleotides 89118-97710) was detected, indicating that the Bam HI site at position 95036 has been removed. This was confirmed by PCR analysis. Chromosomal DNA was subjected to PCR using oligonucleotide primers identical to, respectively, the sequences from nucleotide 93950 to 93968; and from 96990 to 97010. The expected 3kb DNA fragment was amplified from wild type DNA and, following Bamlll digest, two bands roughly 2kb and lkb in size were detected. In samples containing S .hygroscopicus ΔRapL chromosomal DNA the 3kb PCR product amplified was found to be resistant to BamHI digestion.
Precursor feeding of the chromosomal mutant S . hygroscopicus ΔRapL
Growing cultures of the mutant S . hygroscopicus ΔRapL were fed with different amino acid precursors (table 1) . Only the three proline derivatives were found to be incorporated as judged by LC-MS. The main rapamycin derivative in the fermentations apart from prolyl rapamycin is a compound with m/z 908 which could correspond to a hydroxy-rapamycin lacking a methoxy group. Smaller amounts of a compound with m/z 938 were also detected which would correspond to hydroxy-prolyl- rapamycin. MS-fragmentation experiments as well as the characteristic UV spectra clearly indicated that these compounds are rapamycin derivatives with a hydroxyproline incorporated. In order to get enough material for NMR characterisation we fed hydroxyproline on a large scale to the mutant (15L broth) and isolated 3 mg of the compound with m/z 908 as described in material and methods. The NMR data (table 2) showed the chemical shifts and couplings expected for the hydroxy-proline spin system. The changed chemical shifts for the positions 26 and 27 and the unchanged shifts for positions 38-40 as compared to rapamycin proved that the methoxy group is missing at position 26. MS- fragmentation data (table 3) confirmed these findings. This can be inferred from the loss of the C15-C26- fragment leading to a fragment with m/z 644 for both of the new rapamycin derivatives. Furthermore, the loss of the C28 -C42-fragment (322 amu) can be seen for both compounds as well as for rapamycin, indicating that there is no modification in this part of the molecules. The ions at m/z 807 and 777 respectively which are equivalent to the loss of the amino acid (131 amu) confirm the presence of OH-proline. This means that the compound with m/z 938 is 4 -hydroxyprolyl -rapamycin.
Figure imgf000015_0001
Table 1
Figure imgf000016_0001
Table 2
Figure imgf000016_0002
Table 3 Preparation of trans-3-aza-bicyclo [3.1.01 hexane-2- carboxylic acid rapamycin
A 2 L fermentation of S . hygroscopicus ΔRapL fed with (+/- ) -trans-3-aza-bicyclo [3.1.0] hexane-2 -carboxylic acid (0.5 mg ml"1) was grown for 5 days in TSBGM medium (Khaw et al., (1998) J. Bacteriol . 180, 809-814.). The cells were collected by filtration and extracted with 1 L of methanol at 4°C overnight. High pressure liquid chromatograph-electrospray ionization mass spectrometry (HPLC-ESIMS) analysis of the crude methanol extract was performed at this stage using a Hewlett-Packard 1100 LC attached to a Finnigan-Mat LCQ mass spectrometer. Trans- 3-aza-bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin was detected in the fermentation broth.
The methanol extracts were combined and concentrated under reduced pressure. The aqueous residue was diluted with 500 mL of distilled water, and extracted three times with 500 mL of distilled ethyl acetate. The combined ethyl acetate extracts were dried with anhydrous sodium sulphate, and evaporated to dryness . The resulting yellow residue was purified by flash column chromatography on a 150 mm x 30 mm (diameter) silica gel column [Merck 60] eluted isocratically witeh a 1:1 (v/v) mixture of acetone/hexane .
The fractions were analysed by electrospray mass spectrometry. MS-MS and Msn were used to determine the structure of the new rapamycin in the fractions from the flash silica column. The fractions containing trans-3-aza- bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin were further purified by reversed-phase preparative HPLC on a 250 x 20 mm (diameter) Prodigy 0DS3 column (Phenomenex) using gradient elution starting at 70/30 (v/v) acetonitrile/water rising linearly to 100% acetonitrile over 25 minutes. The 2 L fermentation yielded about 4 mg of pure trans-3-aza-bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin.
High resolution MS of trans-3-aza- bicyclo [3.1.0] hexane-2 -carboxylic acid rapamycin on a Bruker BioApex FTICR mass spectrometer using electrospray ionisation gave a sodiated molecular ion at m/z 934.52776, which confirmed the molecular formula to be
Figure imgf000018_0001
Biological activity
The dose response of human lymphoblastoid cell lines 536 was measured. In the experiment shown in Figure 3 the mean cpm of radiolabelled thymidine incorporated into the untreated controls shows that 0-3 h drug exposure had no appreciable effect on DNA synthesis up to lOOnM of rapamycin, prolylrapamycin, or 4-hydroxy-prolyl-26- demethoxy-rapamycin. This implies that none of the compounds were toxic to the 536 cell line. After 24 and 48 hours (Fig. 4) the 536 cells showed a concentration- dependent inhibition of DNA synthesis with an ID50% of InM for rapamycin; and 3nM for prolylrapamycin. 4- hydroxy-prolyl-26-demethoxy-rapamycin was also inhibitory but did not reach 50% at lOOnM. Previous experiments have shown that rapamycin is a profound inhibitor of Gl progression in the 536 cell line (Metcalfe et al . , Oncogene 15, 1635-1642 (1997)). This is also suggested in these experiments for the rapamycin analogues, since no significant effect was found at 3h but inhibition was observed once the cell population had time to proceed through a complete cell cycle (24h) and reach the drug arrest point .

Claims

CLAIMS :
1. A process of modifying a gene cluster involved in the biosynthesis of a polyketide, said gene cluster including a gene ("the precursor gene") responsible for the production of an enzyme which is responsible for the production of a precursor compound which is incorporated into said polyketide; said process comprising the step of deleting or inactivating said precursor gene.
2. A process according to claim 1 wherein said process of deleting or inactivating said precursor gene employs phage-mediated gene replacement.
3. A process according to claim 1 or claim 2 wherein the gene cluster is the gene cluster for the production of rapamycin in S . hygroscopicus and the precursor gene is the rapL gene whose product is responsible" for the production of L-pipecolate .
4. A process for producing a polyketide comprising modifying a gene cluster by the process of claim 1, 2 or 3 and expressing the modified gene cluster to produce polyketide synthase enzymes which act in the presence of a variant precursor compound which is incorporated so that a variant polyketide is produced.
5. A process according to claim 4 as appendant on claim 3 wherein the variant precursor compound is selected from L-proline, L-trans-4-hydroxyproline, L-cis-4- hydroxyproline, L-cis-3-hydroxyproline and trans-3-aza- bicyclo [3, 1, 0] hexane-2-carboxylic acid.
6. A compound selected from prolyl-rapamycin, 4-hydroxy-propyl rapamycin, 4-hydroxyprolyl-2, 6- de ethoxy-rapamycin, 3-hydroxy-prolyl-rapamycin 3-hydroxy-prolyl-2, 6-demethoxy-rapamycin, and trans-3- aza-bicyclo [3, 1, 0] hexane-2-carboxylic acid rapamycin.
7. A pharmaceutical composition comprising a compound of claim 6.
8. Use of a compound of claim 6 in the manufacture of an immunosuppressant composition.
9. A modified gene cluster as produced by the process of any of claims 1-3.
10. A vector containing the gene cluster of claim 9.
11. A microorganism containing the gene cluster of claim 9 and capable of expressing it.
PCT/GB1998/001559 1997-05-28 1998-05-28 Polyketides and their synthesis and use WO1998054308A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AT98924463T ATE307198T1 (en) 1997-05-28 1998-05-28 POLYKETIDES AND THEIR PRODUCTION AND USE
DE69831943T DE69831943T2 (en) 1997-05-28 1998-05-28 POLYETHYDES AND THEIR PREPARATION AND APPLICATION
EP98924463A EP0983348B1 (en) 1997-05-28 1998-05-28 Polyketides and their synthesis and use
AU76661/98A AU7666198A (en) 1997-05-28 1998-05-28 Polyketides and their synthesis and use
US10/307,595 US7018808B2 (en) 1997-05-28 2002-12-02 Polyketides and their synthesis and use
US11/328,642 US7381546B2 (en) 1997-05-28 2006-01-10 Polyketides and their synthesis and use
US12/129,295 US20080287483A1 (en) 1997-05-28 2008-05-29 Polyketides and Their Synthesis and Use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB9710962.3A GB9710962D0 (en) 1997-05-28 1997-05-28 Polyketides and their synthesis
GB9710962.3 1997-05-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09424751 A-371-Of-International 1998-05-28
US10/307,595 Continuation US7018808B2 (en) 1997-05-28 2002-12-02 Polyketides and their synthesis and use

Publications (2)

Publication Number Publication Date
WO1998054308A2 true WO1998054308A2 (en) 1998-12-03
WO1998054308A3 WO1998054308A3 (en) 1999-04-08

Family

ID=10813155

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1998/001559 WO1998054308A2 (en) 1997-05-28 1998-05-28 Polyketides and their synthesis and use

Country Status (8)

Country Link
US (3) US7018808B2 (en)
EP (1) EP0983348B1 (en)
AT (1) ATE307198T1 (en)
AU (1) AU7666198A (en)
DE (1) DE69831943T2 (en)
ES (1) ES2251084T3 (en)
GB (1) GB9710962D0 (en)
WO (1) WO1998054308A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000047724A2 (en) * 1999-02-09 2000-08-17 Board Of Trustees Of The Leland Stanford Junior University Methods to mediate polyketide synthase module effectiveness
WO2001034816A1 (en) * 1999-10-29 2001-05-17 Kosan Biosciences, Inc. Rapamycin analogs
US6495348B1 (en) 1993-10-07 2002-12-17 Regents Of The University Of Minnesota Mitomycin biosynthetic gene cluster
WO2002101003A2 (en) * 2001-06-08 2002-12-19 Rhodia Chimie Stereoselective preparation of cyclic l-amino acids
WO2005100366A1 (en) * 2004-04-14 2005-10-27 Wyeth Proline cci-779 (proline-rapamycin 42-ester with 2,2-bis (hydroxymethyl) propionic acid) and two-step enzymatic synthesis of proline cci-779 and cci-779 using microbial lipase
WO2006016167A2 (en) * 2004-08-11 2006-02-16 Biotica Technology Limited 17-desmethylrapamycin and analogues thereof, methods for their roduction and their use as immunosupressants, anticancer agents, antifungal agents, etc.
US7001748B2 (en) 1999-02-09 2006-02-21 The Board Of Trustees Of The Leland Stanford Junior University Methods of making polyketides using hybrid polyketide synthases
US7300942B2 (en) 2002-07-16 2007-11-27 Biotica Technology Limited Production of polyketides and other natural products

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102144961A (en) * 2003-09-18 2011-08-10 参天制药株式会社 Transscleral delivery
US8663639B2 (en) * 2005-02-09 2014-03-04 Santen Pharmaceutical Co., Ltd. Formulations for treating ocular diseases and conditions
WO2006086750A1 (en) * 2005-02-09 2006-08-17 Macusight, Inc. Liquid formulations for treatment of diseases or conditions
JP5528708B2 (en) 2006-02-09 2014-06-25 参天製薬株式会社 Stable formulations and methods for preparing and using them
US8222271B2 (en) 2006-03-23 2012-07-17 Santen Pharmaceutical Co., Ltd. Formulations and methods for vascular permeability-related diseases or conditions
US20080265343A1 (en) * 2007-04-26 2008-10-30 International Business Machines Corporation Field effect transistor with inverted t shaped gate electrode and methods for fabrication thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0589703A1 (en) * 1992-09-24 1994-03-30 American Home Products Corporation Proline derivative of rapamycin, production and application thereof
WO1994010843A1 (en) * 1992-11-10 1994-05-26 Smithkline Beecham Corporation Rapamycin derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0589703A1 (en) * 1992-09-24 1994-03-30 American Home Products Corporation Proline derivative of rapamycin, production and application thereof
WO1994010843A1 (en) * 1992-11-10 1994-05-26 Smithkline Beecham Corporation Rapamycin derivatives

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DENOYA CD ET AL: "A second branched-chain alpha-keto acid dehydrogenase gene cluster (bkdFGH) from Streptomyces avermitilis: its relationship to avermectin biosynthesis and the construction of a bkdF mutant suitable for the production of novel antiparasitic avermectins." J BACTERIOL, JUN 1995, 177 (12) P3504-11, XP002079427 UNITED STATES *
DUTTON CJ ET AL: "Novel avermectins produced by mutational biosynthesis." J ANTIBIOT (TOKYO), MAR 1991, 44 (3) P357-65, XP002079430 JAPAN *
HAFNER EW ET AL: "Branched-chain fatty acid requirement for avermectin production by a mutant of Streptomyces avermitilis lacking branched-chain 2-oxo acid dehydrogenase activity." J ANTIBIOT (TOKYO), MAR 1991, 44 (3) P349-56, XP002079428 JAPAN *
KHAW LE ET AL: "Mutational biosynthesis of novel rapamycins by a strain of Streptomyces hygroscopicus NRRL 5491 disrupted in rapL, encoding a putative lysine cyclodeaminase." J BACTERIOL, FEB 1998, 180 (4) P809-14, XP002079433 UNITED STATES *
LOMOVSKAYA, N. ET AL: "Gene disruption and replacement in the rapamycin-producing Streptomyces hygroscopicus strain ATCC 29253" MICROBIOLOGY,, vol. 143, March 1997, pages 875-883, XP002079431 *
MOLNAR I ET AL: "Organisation of the biosynthetic gene cluster for rapamycin in Streptomyces hygroscopicus: analysis of genes flanking the polyketide synthase" GENE, vol. 169, no. 1, 22 February 1996, page 1-7 XP004042979 *
SCHWECKE T ET AL: "The biosynthetic gene cluster for the polyketide immunosuppressant rapamycin." PROC NATL ACAD SCI U S A, AUG 15 1995, 92 (17) P7839-43, XP002079432 UNITED STATES *
TANG L ET AL: "Amino acid catabolism and antibiotic synthesis: valine is a source of precursors for macrolide biosynthesis in Streptomyces ambofaciens and Streptomyces fradiae." J BACTERIOL, OCT 1994, 176 (19) P6107-19, XP002079429 UNITED STATES *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6495348B1 (en) 1993-10-07 2002-12-17 Regents Of The University Of Minnesota Mitomycin biosynthetic gene cluster
US7001748B2 (en) 1999-02-09 2006-02-21 The Board Of Trustees Of The Leland Stanford Junior University Methods of making polyketides using hybrid polyketide synthases
WO2000047724A3 (en) * 1999-02-09 2000-12-07 Univ Leland Stanford Junior Methods to mediate polyketide synthase module effectiveness
US6753173B1 (en) 1999-02-09 2004-06-22 Board Of Trustees Of The Leland Stanford Junior University Methods to mediate polyketide synthase module effectiveness
WO2000047724A2 (en) * 1999-02-09 2000-08-17 Board Of Trustees Of The Leland Stanford Junior University Methods to mediate polyketide synthase module effectiveness
WO2001034816A1 (en) * 1999-10-29 2001-05-17 Kosan Biosciences, Inc. Rapamycin analogs
US6670168B1 (en) 1999-10-29 2003-12-30 Kosan Bioscience, Inc. Recombinant Streptomyces hygroscopicus host cells that produce 17-desmethylrapamycin
WO2002101003A2 (en) * 2001-06-08 2002-12-19 Rhodia Chimie Stereoselective preparation of cyclic l-amino acids
WO2002101003A3 (en) * 2001-06-08 2004-02-26 Rhodia Chimie Sa Stereoselective preparation of cyclic l-amino acids
US7425530B2 (en) 2001-06-08 2008-09-16 Rhodia Chimie Stereoselective preparation of cyclic L-amino acids
US7645768B2 (en) 2002-07-16 2010-01-12 Wyeth Production of polyketides and other natural products
EP2277898A2 (en) 2002-07-16 2011-01-26 Biotica Technology Limited Rapamycin analogues
US7300942B2 (en) 2002-07-16 2007-11-27 Biotica Technology Limited Production of polyketides and other natural products
US9605001B2 (en) 2002-07-16 2017-03-28 Buck Institute For Research On Aging Production of polyketides and other natural products
US7390895B2 (en) 2002-07-16 2008-06-24 Biotica Technology Limited Production of polyketides and other natural products
EP1967520A2 (en) 2002-07-16 2008-09-10 Biotica Technology Limited Production of polyketides and other natural products
US9040259B2 (en) 2002-07-16 2015-05-26 Buck Institute For Research On Aging Production of polyketides and other natural products
EP2277898A3 (en) * 2002-07-16 2011-06-01 Biotica Technology Limited Rapamycin analogues
US7202256B2 (en) 2004-04-14 2007-04-10 Wyeth Proline CCI-779, production of and uses therefor, and two-step enzymatic synthesis of proline CCI-779 and CCI-779
WO2005100366A1 (en) * 2004-04-14 2005-10-27 Wyeth Proline cci-779 (proline-rapamycin 42-ester with 2,2-bis (hydroxymethyl) propionic acid) and two-step enzymatic synthesis of proline cci-779 and cci-779 using microbial lipase
JP2008509895A (en) * 2004-08-11 2008-04-03 バイオティカ テクノロジー リミテッド Manufacture of polyketides and other natural products
US7803808B2 (en) 2004-08-11 2010-09-28 Wyeth Llc Production of polyketides and other natural products
WO2006016167A2 (en) * 2004-08-11 2006-02-16 Biotica Technology Limited 17-desmethylrapamycin and analogues thereof, methods for their roduction and their use as immunosupressants, anticancer agents, antifungal agents, etc.
WO2006016167A3 (en) * 2004-08-11 2006-04-13 Biotica Tech Ltd 17-desmethylrapamycin and analogues thereof, methods for their roduction and their use as immunosupressants, anticancer agents, antifungal agents, etc.

Also Published As

Publication number Publication date
DE69831943D1 (en) 2006-03-02
US20060105436A1 (en) 2006-05-18
AU7666198A (en) 1998-12-30
US20030104585A1 (en) 2003-06-05
US7381546B2 (en) 2008-06-03
DE69831943T2 (en) 2006-07-06
US20080287483A1 (en) 2008-11-20
WO1998054308A3 (en) 1999-04-08
EP0983348A2 (en) 2000-03-08
ATE307198T1 (en) 2005-11-15
EP0983348B1 (en) 2005-10-19
US7018808B2 (en) 2006-03-28
GB9710962D0 (en) 1997-07-23
ES2251084T3 (en) 2006-04-16

Similar Documents

Publication Publication Date Title
US7381546B2 (en) Polyketides and their synthesis and use
EP2342335B1 (en) Novel gene cluster
Bibb et al. Analysis of the nucleotide sequence of the Streptomyces glaucescens tcmI genes provides key information about the enzymology of polyketide antibiotic biosynthesis.
Ward et al. Chalcomycin biosynthesis gene cluster from Streptomyces bikiniensis: novel features of an unusual ketolide produced through expression of the chm polyketide synthase in Streptomyces fradiae
JP2000515390A (en) Novel polyketide derivative and recombinant method for producing the same
JP2000511063A (en) Polyketides and their synthesis
EP0929681B1 (en) Rifamycin biosynthesis gene cluster
WO2005121327A2 (en) Biosynthetic gene cluster for the production of a complex polyketide
Arisawa et al. Direct fermentative production of acyltylosins by genetically-engineered strains of Streptomyces fradiae
JP2008278895A (en) Biosynthetic gene for producing butenyl-spinosyn insecticide
WILSON et al. Molecular analysis of tlrB, an antibiotic-resistance gene from tylosin-producing Streptomyces fradiae, and discovery of a novel resistance mechanism
CN110305881B (en) Biosynthetic gene cluster of polyketide neoenterocins and application thereof
EP1632569B1 (en) Strain belonging to the genus streptomyces and being capable of producing nemadictin and process for producing nemadictin using the strain
Tornus et al. Identification of four genes from the granaticin biosynthetic gene cluster of Streptomyces violaceoruber Tü22 involved in the biosynthesis of L-rhodinose
Novakova et al. Cloning and characterization of a new polyketide synthase gene cluster in Streptomyces aureofaciens CCM 3239
Paulus et al. New Alpiniamides From sp. IB2014/011-12 Assembled by an Unusual Hybrid Non-ribosomal Peptide Synthetase-AT Polyketide Synthase Enzyme.
SOCKALINGAM Genetic studies on a soil streptomyces sp. that produces an antifungal compoud
JP2009502187A (en) Genes involved in thiocoralin biosynthesis and their heterologous production
Praseuth et al. APPLIED CELLULAR PHYSIOLOGY AND METABOLIC ENGINEERING
Virolle et al. Constanze Paulus1, 2, Yuriy Rebets 2, Josef Zapp2, Christian Rückert 3, Jörn Kalinowski 3 and Andriy Luzhetskyy
SI20274A (en) Novel polyketides, anthrone derivatives
KR20140021083A (en) Two-component system genes derived from streptomyces acidiscabies atcc 49003 and the production method of antibiotics using the same genes
AU2002305118A1 (en) Biosynthetic genes for butenyl-spinosyn insecticide production

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A2

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)
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A3

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 09424751

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1998924463

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 1999500383

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1998924463

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: CA

WWG Wipo information: grant in national office

Ref document number: 1998924463

Country of ref document: EP