WO2011021036A1 - Polyketides analogues and methods for their production - Google Patents

Polyketides analogues and methods for their production Download PDF

Info

Publication number
WO2011021036A1
WO2011021036A1 PCT/GB2010/051364 GB2010051364W WO2011021036A1 WO 2011021036 A1 WO2011021036 A1 WO 2011021036A1 GB 2010051364 W GB2010051364 W GB 2010051364W WO 2011021036 A1 WO2011021036 A1 WO 2011021036A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
crc
compound according
acyl
strain
Prior art date
Application number
PCT/GB2010/051364
Other languages
French (fr)
Inventor
Barrie Wilkinson
Christine Martin
Steven Moss
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
Publication of WO2011021036A1 publication Critical patent/WO2011021036A1/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
    • 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
    • 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
    • C12P17/188Heterocyclic compound containing in the condensed system at least one hetero ring having nitrogen atoms and oxygen atoms as the only ring heteroatoms

Definitions

  • the present invention relates to the use of the genes for biosynthesis of the allyl extender unit of FK506, the generation of novel FK506 and FK520 analogues and strains generated by manipulating one or more of these extender unit genes and optionally feeding non-natural extender units, or crotonic acid precursors thereof, to these strains and to the use of such compounds in therapy.
  • FK506 tacrolimus/fujimycin/Prograf
  • FK520 ascomycin or immunomycin
  • Figure 1 are lipophilic macrolides produced by a variety of actinomycetes, including Streptomyces tsukubuaensis No. 9993 (Hatanaka et al., 1989), Streptomyces sp.
  • FK525 Hatanaka H, et al., 1989
  • FK523 Hatanaka, H., et al., 1988
  • antascomicins Fehr, T., et al., 1996
  • a number of semisynthetic derivatives of these molecules have also been shown to be of utility, including pimecrolimus (SDZ ASM 981 , Elidel), which is a derivative of FK520 (Meingassner et al., 1997).
  • PKS polyketide synthases
  • FK506, FK520 and close analogues suppress the immune system by inhibiting signal transduction pathways required for T-cell activation and growth.
  • they have been shown to inhibit Ca 2+ -dependent T-cell proliferation, via initial formation of a complex with an FK-binding protein (FKBP), which binds to and blocks calcineurin (CaN).
  • FKBP FK-binding protein
  • This FK506-FKBP-CaN complex inhibits the activation of nuclear factor of activated t-cells (NF-AT), preventing its entrance into the nucleus, and subsequent activation of the promoter of lnterleukin-2 (IL-2), which initiates IL-2 production.
  • NF-AT nuclear factor of activated t-cells
  • IL-2 lnterleukin-2
  • FK506 can interfere with the action of calcineurin on substrates other than NFAT, including IKB, Na-K- ATPase and nitric oxide synthase, which may lead to some of the side-effects (Kapturczak et al., 2004).
  • TGF- ⁇ transforming growth factor beta
  • USES: FK506 in particular, is an important immunosuppressant used to aid prevention of organ rejection after transplantation.
  • it is used intravenously and orally for the prevention of organ rejection after allogeneic liver or kidney transplantation and in bone marrow transplantation. It has been shown to have potential utility in a wide variety of autoimmune, inflammatory and respiratory disorders, including Crohn's disease, Behcet syndrome, uveitis, psoriasis, atopic dermatitis, rheumatoid arthritis, nephritic syndrome, aplastic anaemia, biliary cirrhosis, asthma, pulmonary fibrosis, chronic obstructive pulmonary disease (COPD) and celiac disease.
  • COPD chronic obstructive pulmonary disease
  • FK506 FK506, which include renal dysfunction, gastrointestinal effects, neurological effects, hyperthrichosis and gingival hyperplasia.
  • Pimecrolimus and FK506 are both used in topical formulations, such as ointments and creams, as treatments for a variety of skin conditions, in particular atopic dermatitis (Nghiem et al., 2002).
  • Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to FK506 metabolism while the P-glycoprotein pump (MDR-1 ) modulates its bioavailability (Roy et al., 2006). The complexity of FK506 dosing is therefore enhanced by significant drug-drug interactions (Kapturczak et al., 2004).
  • the mechanism of toxicity of FK506 and FK520 has been related to the mechanism of action of immunosuppression (F. Dumont ef a/., 1992). This strong link between the mechanism of action and the toxicity has presented significant challenges to improving the therapeutic index through chemical modification. Segregation of efficacy and toxicity of new analogues may be possible by altering distribution or metabolism (NH Signal et al., 1991 ). By limiting the exposure of the compound to organs that are sensitive to such inhibition, such as the kidney, systemic toxicity can be avoided. Additionally, topical administration of the calcineurin inhibitor at the site of administration (such as skin, lungs, gut, eye etc.) can be maximized. One way this can be achieved is by using a 'soft drug' approach, which involves designing compounds to have limited systemic exposure such as through increased metabolism, higher blood/plasma protein binding, poor absorption or bioavailability.
  • variable metabolism of FK506 leads to some of the toxicity, due to variable levels of systemic exposure, which led to the need for constant drug monitoring (Armstrong and Oellerich, 2001 ). Therefore, analogues of FK506 with reduced or less variable metabolism could be useful in reducing toxicity, and reducing the need for constant monitoring of drug levels.
  • FK506 is also poorly bioavailable (Tamura et al., 2003), which leads to variable systemic exposure when dosed orally, and the frequent need for intravenous dosing. Therefore, analogues with improved oral bioavailability would be very useful, to reduce systemic toxicity through incorrect dosing, and improve the ease of oral dosing.
  • FK506 analogues altered in the side-chain at C21 may demonstrate modulated calcineurin binding.
  • FK506 and FK520 analogues with modulated calcineurin binding may be useful for reduction of toxicity and may demonstrate improved activities in diseases dependent upon immunophillin ligands, including, but not limited to, FKBP12, FKBP51 , FKBP52 and FKBP13, this may include increased neuroprotection, neuroregeneration and reduced neurotoxicity.
  • novel FK506 and FK520 analogues which may have utility in the maintenance of immunosuppression, both for organ transplantation, and for the treatment of inflammatory conditions, and for the treatment of fungal infections.
  • the present invention discloses novel FK506 and FK520 analogues which have altered
  • novel FK506 and FK520 analogues may also be useful for therapies requiring local availability but with poor systemic availability, including, but not limited to topically administered therapies for inflammatory disorders such as atopic dermatitis, asthma and inflammatory bowel diseases, which in particular are expected to show improvements in respect of one or more of the following properties: decreased metabolic stability, decreased bioavailability, decreased oral
  • bioavailability increased efflux via membrane transporters and high plasma protein binding
  • the complete biosynthetic cluster of FK506 is described. This includes the genes dedicated to provision of the allyl extender unit at C21 of FK506 which comprises some or all of ORF1 to ORF9 in Table 2. Analysis of gene function indicates a pathway in which a 3-carbon unit derived from propionyl-CoA is condensed with a 2-cabron unit derived from malonyl-CoA to give a 5-carbon chain. Selection and covalent attachment of at least one of these units is performed by ORF9, a dual acyltransferase-acylcarrier protein. The enzyme responsible for condensation is the ketosynthase ORF8.
  • the 3-carbon unit is most likely modified to an acrylic acid derivative prior to condensation by the action of an acyl-CoA dehydrogenase, most probably ORF6 but possibly ORF2.
  • an oxygenase such as ORF3.
  • the resulting acylcarrier protein- or coenzyme-A linked 5-carbon acid is modified to a crotonic acid derivative by the action of 'house-keeping' enzymes from the fatty acid biosynthetic pathway, specifically a beta- ketoacylreductase and a dehydratase.
  • Other possible enzymes involved in this chemistry include the short chain dehydrogenase/reductase ORF2.
  • the final step of biosynthesis is a reductive carboxylation catalysed by the ccr homologue ORF7 to a give a 3-carbon (allyl) substituted malonic acid thioester derivative.
  • Provision of the allyl extender unit may be engineered out of the cell by, for example, disruption of, or deletion of, one or more of the genes responsible for the provision of the allyl extender unit. Exogenous compounds which are accepted into this extender position may then be fed.
  • the FK506 cluster ccr gene (ORF7) may be retained in order to carboxylate the fed acid or ester appropriately for incorporation into the PKS.
  • a heterologous ccr gene may be expressed in the engineered organism in order to allow incorporation of a broader set of extender units.
  • FK506 analogues at C21 may be combined with previous strategies for the production of FK506 and FK520 analogues, of engineering a strain that cannot provide the natural starter unit and feeding exogenous acids or esters and generating engineered strains where the natural loading module from the FK506 or FK520 polyketide synthase is replaced by the loading module from the avermectin or an avermectin-like polyketide synthase, and optionally non-natural starter units are fed to these strains.
  • strains optionally having been mutated by classical methods or targeted inactivation or deletion of one or more genes responsible for post-PKS modification, and/or mutated by classical methods or targeted inactivation or deletion of one or more precursor supply genes, including bkd genes (Ward et al., 1999) and homologues thereof.
  • X a represents CH 2 , S, O, fused cyclopropyl unit and X b represents a bond;
  • Z represents keto or CH 2 ;
  • Ri represents a moiety selected from:
  • V W X Y R 2 represents H, alkyl, halo, hydroxyl or thiol
  • R 3 represents H, Ci -4 alkyl, halo, hydroxyl or thiol
  • R 4 represents H, Ci -4 alkyl, halo, hydroxyl or thiol
  • R 5 represents OMe, Me or H
  • R 6 represents OMe, Me or H
  • R 8 represents OH
  • R 9 represents H, OH, halo, thiol, CrC 4 alkyl
  • Rio, Rn and Ri 2 independently represent F, Cl, CrC 4 alkyl, ORi 3 , SRi 3 or NHRi 3 and Ri 3 represents H, CrC 4 alkyl or CrC 4 acyl, wherein two or three of Rio-Ri 2 are CrC 4 alkyl;
  • Ri 3 and Ri 4 independently represent H, F, Cl, CrC 4 alkyl, ORi 5 , SRi 5 or NHRi 5 and Ri 5 represents H, CrC 4 alkyl or CrC 4 acyl;
  • Ri6 Ri 7 and Ri 8 independently represent H, F, Cl, CrC 4 alkyl, ORi 9 , SRi 9 or NHRi 9 and Ri 9 represents H, CrC 4 alkyl or CrC 4 acyl,
  • R 20 and R 2 i independently represent H, F, Cl, CrC 4 alkyl, OR 22 , SR 22 or NHR 22 and R 22 represents H, CrC 4 alkyl or CrC 4 acyl,
  • R 23 and R 24 independently represent H, F, Cl, CrC 4 alkyl, OR 25 , SR 25 or NHR 25 and R 25 represents H, CrC 4 alkyl or CrC 4 acyl;
  • R 26 represents H, F, Cl, CrC 4 alkyl, OR 27 , SR 27 or NHR 27 , and R 27 represents H, CrC 4 alkyl or
  • R 28 and R 29 independently represent H, F, Cl, CrC 4 alkyl, OR 30 , SR 30 or NHR 30 and R 30 represents H, CrC 4 alkyl or CrC 4 acyl ;
  • R 3 i R 32 and R 33 independently represent H, F, Cl, CrC 4 alkyl, OR 34 , SR 34 or NHR 34 and R 34 represents H, CrC 4 alkyl or CrC 4 acyl;
  • R35 R36 and R 37 independently represent H, F, Cl, CrC 4 alkyl, OR 38 , SR 38 or NHR 38 and R 38 represents H, CrC 4 alkyl or CrC 4 acyl, save that OR 41 shall not represent OH;
  • R 39 R 40, R 4 i and R 42 independently represent H, F, Cl, CrC 4 alkyl, OR 43 , SR 43 or NHR 43 and R 43 represents H, CrC 4 alkyl or CrC 4 acyl,
  • R 44 and R 45 independently represent H, F, Cl, CrC 4 alkyl, OR 46 , SR 46 or NHR 46 and R 46 represents H, CrC 4 alkyl or CrC 4 acyl;
  • R 47 , R 48 and R 49 are independently selected from H, F, Cl, OR 50 , SR 50 and NHR 50 and R 50 is selected from H, Ci-C 4 alkyl and Ci-C 4 acyl;
  • A is selected from O, S and NR 5 i
  • R 5 i represents H, Ci-C 4 alkyl or Ci-C 4 acyl
  • R 52 and R 53 are independently selected from H, Ci-C 4 alkyl
  • R 54 represents Ci-C 4 alkyl or CrC 4 acyl
  • X c represents a bond or CH 2
  • R 54 represents H and when X c is
  • R 54 represents H, F, Cl, OH, SH, Ci-C 4 alkyl, OR 55 , SR 55 or NHR 55 and R 55 represents Ci-C 4 alkyl or Ci-C 4 acyl;
  • B represents O, S, or NR 6 O
  • R 6 o represents H, Ci-C 4 alkyl, or Ci-C 4 acyl
  • R 56 represents d-
  • R 62 represents Ci-C 4 alkyl or C r C 4 acyl
  • X d and X e independently represent bond or CH 2 provided that X d and X e do not both represent CH 2 and when X d is bond then R 57 represents H and when X e is CH 2 then R 57 represents H, F, Cl, OH,
  • R 63 represents CrC 4 alkyl or CrC 4 acyl and when X e is bond R 59 represents H and wherein when X e represents CH 2 then R 59 represents H, F, Cl, OH,
  • R 65 and R 66 independently represent H, F, Cl, OH, SH, C r C 4 alkyl, OR 67 , SR 67 or NHR 67 , and
  • R 67 represents Ci-C 4 alkyl or CrC 4 acyl
  • R 68 represents H, F, Cl, OH, SH, C r C 4 alkyl, OR 69 , SR 69 or NHR 69 , and R 69 represents C r C 4 alkyl or CrC 4 acyl;
  • R 70 and R 7i independently represent H, F, Cl, OH, SH, C r C 4 alkyl, OR 72 , SR 72 or NHR 72 and
  • R 72 represents CrC 4 alkyl or CrC 4 acyl ;
  • R 73 , R 74 and R 75 independently represent H, F, Cl, OH, SH, C r C 4 alkyl, OR 76 , SR 76 or NHR 76 and R 76 represents CrC 4 alkyl or CrC 4 acyl;
  • R 77 , R 78 and R 79 independently represent H, F, Cl, OR 82 , SR 82 , CrC 4 alkyl or CN, R 82 represents H, CrC 4 alkyl or CrC 4 acyl, R 80 and R 8 i independently represent H, F, Cl, OH, SH or CrC 4 alkyl, provided that at least one of R 77 , R 78 and R 79 is not H or CrC 4 alkyl;
  • R 83 , R 84 and R 85 are independently selected from H, F, Cl, OR 88 , SR 88 , C1-C4 alkyl and CN, R 88 represents H, CrC 4 alkyl or CrC 4 acyl, R 86 and R 87 independently represent H, F, Cl, OH, SH or CrC 4 alkyl and provided that at least one of R 83 , R 84 and R 85 does not represent H or CrC 4 alkyl;
  • the invention embraces all stereoisomers of the compounds defined by structure (I) as shown above.
  • the present invention provides FK506 or FK520 analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use as a pharmaceutical.
  • the present invention provides strains containing hybrid PKS, with a loading module conferring avermectin-like chain initiation, and the rest of the PKS conferring FK506/FK520-like chain processing and termination.
  • the present invention provides processes for production of FK506 and FK520 analogues defined by structure (I) above.
  • analogue means one analogue or more than one analogue.
  • analogue(s) refers to chemical compounds that are structurally similar to another but which differ slightly in composition (as in the replacement of one atom by another or in the presence or absence of a particular functional group).
  • FK506 and FK520 analogues refer to compounds related to FK506, FK520 and similar compounds in structure. Such compounds are 22-membered rings with one lactone and one amide bond. The N of the amide bond forms a 2-carboxyl piperidine or a 2-carboxyl pyrrolidine. This carboxyl group forms the lactone group, with an oxygen that is allylic to a double bond that is exo to the main 22-membered ring.
  • Such compounds include, without limitation, FK520, FK506, antascomicin, FK523, FK525, pimecrolimus and tsukubamycin as well as compounds of formula (I).
  • FK506 or FK520 producing strain refers to a strain (natural or recombinant) which is capable of producing one or more FK506 or FK520 analogues when fed appropriately.
  • recombinant strain of a FK506 or FK520 producing host refers to a recombinant strain based on a natural FK506 or FK520 producing strain which is capable of producing one or more FK506 or FK520 analogues when fed appropriately.
  • FK506 or FK520 cluster means the PKS and associated enzymes responsible for production of FK506 or FK520 analogues.
  • modifying gene(s) includes the genes required for post- polyketide synthase modifications of the polyketide, for example but without limitation cytochrome P-450 monooxygenases, ferredoxins and SAM-dependent O-methyltransferases.
  • these modifying genes include fkbD and fkbM, but a person of skill in the art will appreciate that PKS systems related to FK520 (for example but without limitation:
  • FK506, antascomicin, FK523, FK525 and tsukubamycin will have homologues of at least a subset of these genes, some of which are discussed further below.
  • precursor supply gene(s) includes the genes required for the supply of the natural or non-natural precursors, the genes required for the synthesis of any naturally or non-naturally incorporated precursors and the genes required for the incorporation of any naturally or non-naturally incorporated precursors.
  • these genes include fkbL, fkbO and fkbP but a person of skill in the art will appreciate that PKS systems related to FK506 and FK520 (for example but without limitation: antascomicin, FK523, FK525 and tsukubamycin) will have homologues of these genes, some of which are discussed further below.
  • extender unit(s) includes references to carboxylic acids or esters fed to cells that are then further processed by the addition of CoA or an acylcarrier protein, and/or also by reductive carboxylation, halogenation or any other cellular process that converts the fed compound into a functional extender unit for the PKS.
  • auxiliary gene(s) includes references to modifying genes, precursor supply genes or both modifying genes and precursor supply genes.
  • auxiliary gene is an oxygenase which may hydroxylate the starter unit.
  • basic product refers to the initial product of the polyketide synthase enzyme before the action of any modifying genes.
  • the term "precursor” includes the natural starter units (i.e. 4,5- dihydroxycyclohex-1-ene carboxylic acid), non-natural starter units (e.g non-cyclic or heterocyclic starter units), and naturally incorporated amino acids (i.e. pipecolic acid) and non- naturally incorporated amino acids
  • non-natural starter unit refers to any compounds which can be incorporated as a starter unit in polyketide synthesis that are not the starter unit usually incorporated by that PKS.
  • avermectin -I ike PKS means the PKS of a bacterium producing a avermectin-like polyketide such as nemadectin or milbemycin which contains a loading domain consisting of AT and ACP domain and which naturally incorporates branched chain starter acids such as those of the present invention.
  • acyl means an alkyl group in which the first carbon atom is a carbonyl moiety.
  • C1-4 acyl groups examples include C2-4 acyl groups such as -COMe and -COEt, especially COMe.
  • C2-4 acyl groups such as -COMe and -COEt, especially COMe.
  • CHO i.e. C1 acyl is a further example which is less preferred.
  • C1-4 alkyl groups include Me, Et, n-Pr, i-Pr, n-Bu, especially Me.
  • Physiologically functional derivatives of compounds of formula (I) include physiologically acceptable salts, esters and solvates.
  • Pharmaceutically acceptable salts include the non-toxic acid addition salt forms of the compounds of formula (I).
  • the pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid.
  • Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulphuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e.
  • ethanedioic malonic
  • succinic i.e. butanedioic acid
  • maleic fumaric
  • malic i.e. hydroxyl- butanedioic acid
  • tartaric citric
  • methanesulfonic ethanesulfonic
  • benzenesulfonic ethanedioic
  • salt forms can be converted by treatment with an appropriate base into the free base form.
  • Example esters include labile esters which are cleaved in the body, for example carboxylic acid esters formed with hydroxyl groups.
  • Example solvates include hydrates.
  • X a represents a bond. In another embodiment X a represents CH 2 . Preferably X a represents CH 2 .
  • X b represents a bond
  • Z 0.
  • R 5 represents OMe
  • R 6 represents OMe
  • R 2 represents H. In one embodiment R 3 represents H.
  • R 4 represents H.
  • the alkyl or alkenyl group of R 7 may optionally be substituted by one to three e.g. one or two or three halogen atoms. In one embodiment the alkyl or alkenyl group of R 7 is not substituted by halogen atoms. In one embodiment the alkyl or alkenyl group of R 7 is substituted by one halogen atom. In one embodiment the alkyl or alkenyl group of R 7 is substituted by two halogen atoms. In one embodiment the alkyl or alkenyl group of R 7 is substituted by three halogen atoms.
  • R 7 is a linear group.
  • R 7 is a branched group.
  • R 7 may for example be selected from CH 3 , CH 2 CH 2 CI, CH 2 CH 2 F,
  • R 1 represents [A]. In another embodiment R 1 represents [B]. In another embodiment R 1 represents [C]. In another embodiment R 1 represents [D]. In another embodiment R 1 represents [E]. In another embodiment R 1 represents [F]. In another embodiment R 1 represents [G]. In another embodiment R 1 represents [H]. In another embodiment R 1 represents [J]. In another embodiment R 1 represents [K]. In another embodiment R 1 represents [L]. In another embodiment R 1 represents [M]. In another embodiment R 1 represents [N]. In another embodiment R 1 represents [O]. In another embodiment R 1 represents [P]. In another embodiment R 1 represents [Q]. In another embodiment R 1 represents [R]. In another embodiment R 1 represents [S]. In another embodiment R 1 represents [T]. In another embodiment R 1 represents [U]. In another embodiment R 1 represents [V]. In another embodiment R 1 represents [W]. In another embodiment R 1 represents [X]. In another embodiment R 1 represents [Y].
  • Compounds of formula (I) may be produced by preventing the in vivo production of the natural extender unit for example but not restricted to interrupting or deleting one or more essential genes such as ORF8 (the KS) or ORF 6 (the AT/ACP), and feeding exogenous carboxylic acids (or derivatives thereof) which can be incorporated; or preventing the in vivo production of the natural extender unit as above while retaining the function of the ccr gene either in its natural position or by providing it in trans for example at an attachment site, and feeding exogenous carboxylic acids (or derivatives thereof) which can be incorporated; or preventing the in vivo production of the natural extender unit as above while retaining the function of a ccr for example a heterologous ccr which may have a different substrate tolerance for example but not limited to the nove chloro-ccr gene of salinosporamide, salG (Liu et al 2009) and feeding exogenous acids which can be incorporated.
  • ORF8 the
  • fkbO is required for the provision of the natural starter unit, 4,5-di- hydroxycyclohex-1-ene carboxylic acid.
  • Disruption, deletion or otherwise removing the function of FkbO (for example but not limited to a frameshift deletion or the use of UV mutagenesis followed by screening for a non-producer of FK506 that is restored to production by feeding exogenous 4,5-di-hydroxycyclohex-1-ene carboxylic acid to the culture medium) leads to a strain that can be used to make FK506 or FK520 analogues by feeding exogenous analogues of the natural starter unit to the production medium.
  • the loading modules of avermectin and avermectin-like PKSs consist of an AT and an ACP domain.
  • the strain is mutated to inactivate or delete of one or more genes that contribute to the biosynthesis or regulation of precursor supply.
  • the gene(s) that contribute to the biosynthesis or regulation of precursor supply may contribute to the biosynthesis or regulation of branched chain keto acid, such as bkd (Ward et al., 1999).
  • the strain may be mutated to inactivate or delete of one or more genes responsible for biosynthesis of pipecolic acid, such as fkbL (Wu et al., 2000).
  • Pipecolic acid is naturally incorporated into the chain as a final step prior to ring closure. This particular modification increases the yield of prolyl derivatives of formula (I) when proline is fed to the strain.
  • the strain may be mutated to have targeted inactivation or deletion of one or more genes responsible for post-PKS modification, for example the gene responsible for oxidation at the C-9 position, fkbJ (Motamedi and Shafiee, 1998, Wu et al., 2000). This particular modification increases the yield of C-9 desketo derivatives of formula (I).
  • a deletion may be the removal of a small part of a gene or most of a gene or all of a gene and it may be done in- frame, or it may be a frameshift. This equally applies to engineering methods to remove starter unit supply and to the use of alternative junctions to join the avermectin loading module to the rapamycin PKS.
  • an avermectin-like loading module may be transferred to the FK506 or FK520 cluster by taking just the Acyltransferase (AT) and Acyl Carrier Protein (ACP) domains from the avermectin PKS and joining to the FK506/FK520 PKS before the first ketosynthase (KS) domain.
  • the junction may be made between the KS from the avermectin PKS and the first extender AT from the FK506/FK520 PKS. Analogous possibilities are discussed in WO 98/01546.
  • the splice junction in the hybrid PKS between the avermectin load and the FK506/FK520 PKS may be before, within or after the KS of the first extension module.
  • part or all of the KS of the first extension domain of the recombinant strain is from the avermectin PKS.
  • the loading module (optionally together with part or all of the KS of the first extension domain) of an avermectin-like PKS may be used in place of the avermectin loading module, such as those loading modules found in the
  • a precursor supply gene or genes such as the bkd genes or homologues thereof may be manipulated by targeted inactivation or deletion or modified by other means such as exposing cells to UV radiation and selection of the phenotype indicating that branched chain alpha keto acid biosynthesis has been disrupted.
  • the optional targeting of the post-PKS genes may occur via a variety of mechanisms, e.g. by integration, targeted deletion of a region of the FK506 or FK520 cluster including all or some of the post-PKS genes optionally followed by insertion of gene(s) or other methods of rendering the post-PKS genes or their encoded enzymes non-functional e.g. chemical inhibition, site- directed mutagenesis or mutagenesis of the cell for example by the use of UV radiation.
  • WO2004/007709 provides methods for the alteration of a gene system which comprises a core portion responsible for the production of a basic product, and a multiplicity of modifying genes responsible for effecting relatively small modifications to the basic product - e.g.
  • the basic product may be a modular polyketide and the modifying genes may be concerned with modifications of a polyketide chain (such as oxidation at the 9 position), and the precursor supply genes may be involved in the production and/or incorporation of natural or non-natural precursors (e.g. pipecolate and/or 4,5- dihydroxycyclohex-1-ene carboxylic acid).
  • natural or non-natural precursors e.g. pipecolate and/or 4,5- dihydroxycyclohex-1-ene carboxylic acid.
  • the core portion may not function properly or even at all in the absence of a precursor supply gene (unless a natural or unnatural precursor compound is supplied or is otherwise available). Therefore, the deletion or inactivation of a precursor supply gene provides a system where it is possible to incorporate non-natural starter units with no competition from the natural starter unit.
  • the present invention provides a method for the incorporation of non-natural extender units into FK506 and FK520 analogues said method comprising removing the ability of the producing strain to provide the extender unit and feeding alternative compounds which may be incorporated, possibly following modification in vivo to become a suitable substrate for the PKS.
  • the present invention provides a method for additionally incorporating non-natural starter acids into FK506 and FK520 analogues, said method comprising either deleting or inactivating the ability of the cell to produce the natural starter unit, for example by targeted deletion or inactivation of fkbO, or by replacing the natural
  • FK506/FK520 loading module with the loading module of avermectin or an avermectin-like PKS and feeding starter units to this strain which optionally contains chromosomal DNA in which the precursor supply gene has been deleted or inactivated.
  • Suitable gene systems which express FK506/FK520 homologues include, but are not limited to, antascomicin, FK520 (Wu et al., 2000; U.S. 6,150,513; AF235504), FK506 (Motamedi et al., 1996; Motamedi et al., 1997;
  • the precursor supply gene which is deleted or inactivated is preferably a gene whose product is involved in branched chain keto acid formation, such as a bkd gene or homologue.
  • the gene system is preferably the FK506 or FK520 cluster.
  • the precursor supply gene deleted or inactivated is more preferably one or more of the bkd genes.
  • fkbM and/or fkbl are deleted or inactivated in addition to one or more of the bkd genes.
  • fkbL, or an analogue of rapL is also deleted to allow more efficient incorporation of pipecolate analogues (Motamedi and Shafiee, 1998, Khaw et al., 1998)
  • the present invention provides a method of producing compounds of formula (I) comprising:
  • strains may be further engineered so as not to produce the natural starter unit.
  • strains may be further engineered so that the gene fkbO is deleted or inactivated.
  • strains may be further engineered so that the natural loading module is replaced by the loading module from the avermectin or an avermectin like PKS.
  • one or more starter genes have been deleted or inactivated which produce a starter unit (e.g. starter acid). Additionally or instead the precursor for which one or more starter genes have been deleted or inactivated may be pipecolic acid. Optionally one or more genes responsible for post-PKS modification are also deleted or inactivated.
  • the recombinant strain is generated using the methods described in WO2004/007709 and in the examples below.
  • the host strain is a selected from the group consisting of
  • Streptomyces tsukubaensis No. 9993 (Ferm BP-927), Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822), Streptomyces sp. AA6554, Streptomyces hygroscopicus var. ascomyceticus MA 6475 ATCC 14891 , Streptomyces hygroscopicus var. ascomyceticus MA 6678 ATCC 55087, Streptomyces hygroscopicus var. ascomyceticus MA 6674, Streptomyces hygroscopicus var. ascomyceticus ATCC 55276, Streptomyces hygroscopicus subsp.
  • the host strain is selected from the group consisting of: S. hygroscopicus var. ascomyceticus ATCC 14891 , Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) or Streptomyces tsukubaensis No. 9993 (Ferm BP-927).
  • auxiliary genes may be deleted or inactivated in the host strain. If desired or necessary one or more of the deleted or inactivated genes of the host strain may be reintroduced by complementation (e.g. at an attachment site, on a self-replicating plasmid or by insertion into a homologous region of the chromosome).
  • polyketide gene clusters may be expressed in heterologous hosts (Pfeifer et al., 2001 ). Accordingly, the present invention includes the transfer of the FK506 or FK520 biosynthetic gene cluster with or without resistance and regulatory genes, either complete, engineered, containing mutations, or containing deletions, for complementation in heterologous hosts. Methods and vectors for the transfer as defined above of such large pieces of DNA are well known in the art (Rawlings, 2001 ; Staunton and Weissman, 2001 ) or are provided herein in the methods disclosed.
  • the present invention provides a method of producing compounds of formula (I) comprising:
  • a preferred heterologous host cell strain is a prokaryote, more preferably an actinomycete or Escherichia coli, still more preferably include, but are not limited to S.
  • PKS polyketide synthase
  • FK506 or FK520 analogue or other polyketide analogue a modified FK506 or FK520 analogue or other polyketide analogue.
  • PKS in a homologous or heterologous could be a hybrid PKS in which one or more domains have been removed, replaced or inserted, such replacements or insertions coming from other heterologous (or homologous) PKS clusters.
  • actinomycetes contain multiple biosynthetic gene clusters for different secondary metabolites, including polyketides and non-ribosomally synthesised peptides. Specifically, it has been demonstrated that strains of S. hygroscopicus produce a variety of polyketides and non-ribosomally synthesised peptides in addition to FK506, FK520, FK523, meridamycin, FK525, antascomicin or tsukubamycin.
  • biosynthetic gene clusters represent a competing requirement for biosynthetic precursors and an additional metabolic demand on the host strain.
  • elaiophylin elaiophylin, bialaphos, hygromycin, augustmycin, endomycin (A, B), glebomycin, hygroscopin, ossamycin and nigericin.
  • additional biosynthetic gene clusters represent a competing requirement for biosynthetic precursors and an additional metabolic demand on the host strain.
  • C21 extender units and starter units are suitably provided as the free carboxylic acid, but derivatives that may be employed include salts and esters.
  • C21 extender and starter unit substances are either known or may be prepared by a skilled person using conventional methods.
  • Standard methods known to those of skill in the art may be used to culture the host or recombinant strain in order to produce compounds of formula (I). Such methods include, without limitation, those described in the examples below; additional methods may also be found in Reynolds and Demain, 1997 and references therein.
  • Compounds of formula (I) are useful as pharmaceuticals for example, but without limitation, having potential utility as immunosuppressants, antifungal agents, anticancer agents, neuroregenerative agents, or agents for the treatment of psoriasis, rheumatoid arthritis, fibrosis and other hyperproliferative diseases.
  • the invention provides for the use of a compound of formula (I) as disclosed herein, in the preparation of a medicament for the prophylaxis and/or treatment of organ rejection after transplantation, autoimmune diseases, inflammatory disorders, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthrisis, fibrosis and/or other hyperproliferative disorders.
  • the invention provides for a method of treatment or prophylaxis of organ rejection after transplantation, autoimmune diseases, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthritis, fibrosis and/or other hyperproliferative disorders comprising administering a compound of formula (I) to a subject in need thereof.
  • the compounds of formula (I) disclosed herein may be used in the preparation of a medicament for the prevention of organ allograft rejection.
  • the compounds of formula (I) are used in the preparation of a medicament for the treatment of autoimmune diseases or inflammatory disorders.
  • autoimmune, inflammatory, proliferative and hyperproliferative diseases examples include but are not inclusively limited to autoimmune diseases, diabetes type I, acute or chronic rejection of an organ or tissue transplant, asthma, tumours or hyperprolific disorders, psoriasis, eczema, rheumatoid arthritis, fibrosis, allergies and food related allergies).
  • assays are well known to those of skill in the art, for example but without limitation: Immunosuppressant activity - Warner, IM., et al., 1992, Kahan et al.
  • the ability of the compounds of this invention to induce immunosuppression may be demonstrated in standard tests used for this purpose.
  • the compounds of this invention are useful in relation to antifibrotic, neuroregenerative and anti-angiogenic mechanisms, one skilled in the art would be able by routine experimentation to determine the ability of these compounds to prevent angiogenesis (e.g. Guba, M.,et al., 2002, ).
  • One of skill in the art would be able by routine experimentation to determine the utility of these compounds in stents (e.g. Morice, M. C, et al., 2002).
  • one of skill in the art would be able by routine experimentation to determine the neuroregenerative ability of these compounds (e.g. Myckatyn, T. M., et al., 2002, Steiner ef a/. 1997)
  • the compounds of formula (I) are also, or in particular, expected to be useful as a therapeutic or prophylactic agents for one or more of the following conditions: rejection reactions after transplantation of organs or tissues (for example heart, kidney, liver, bone marrow, skin, cornea, lung, pancreas, small intestine, limb, muscle, nerve, intervertebral disc, trachea, myoblast and cartilage); graft-versus-host reactions following bone marrow
  • autoimmune diseases for example rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes
  • infections caused by pathogenic microorganisms in particular fungal infections
  • inflammatory or hyperproliferative skin diseases or cutaneous manifestations of immunologically-mediated diseases for example psoriasis, atopic dermatitis, contact dermatitis, eczematoid dermatitis, pyoderma gangrenosum, seborrhoeic dermatitis, lichen planus, pemphigus, bullous
  • autoimmune or allergic diseases of the eye for example keratoconjunctivitis, vernal
  • conjunctivitis allergic conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical keratitis, corneal epithelial dystrophy, keratoleukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, iridocyclitis, sarcoidosis affecting the eye, endocrine ophthalmopathy); reversible obstructive airway diseases or asthma, in particular chronic or inveterate asthma (for example late asthma, airway hyperresponsiveness, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, and dust asthma), mucosal or vascular inflammations (for example gastric ulcers, ischaemic or thrombo
  • abnormality-associated diseases for example Down's syndrome
  • Addison's disease Human Immunodeficiency Virus (HIV) infection or AIDS
  • hypertrophic cicatrix and keloid due to trauma, burn, or surgery.
  • HIV Human Immunodeficiency Virus
  • the aforementioned compounds of the invention or a formulation thereof may be administered by any conventional method including topically (for example by inhalation, vaginally, intranasally, or by eye or ear drop), enterally (for example orally or rectally) or parenterally (for example by intravenous, intracavernosal, subcutaneous, intramuscular, intracardiac or
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • a compound of the invention Whilst it is possible for a compound of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more physiologically acceptable diluents or carriers.
  • the diluents or carrier(s) must be "physiologically acceptable” in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof. In some cases, the diluent or carrier will be water or saline which will be sterile and pyrogen free.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient (compound of the invention) with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformLy and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
  • Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
  • lactose lactose
  • starch a cellulose
  • milk sugar or high molecular weight polyethylene glycols.
  • compounds of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • a tablet may be made by compression or moulding, 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 a powder or granules, optionally mixed with a binder (eg povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (eg sodium starch glycolate, cross-linked povidone, cross-linked sodium
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example,
  • hydroxypropylmethylcellulose in varying proportions to provide desired release profile.
  • Formulations in accordance with the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • Aerosol formulations suitable for administering via inhalation can also be made using methods known in the art. Examples of this include administration of the compounds of the invention by inhalation in the form of a powder (e.g. micronized) or in the form of atomized solutions or suspensions.
  • the aerosol formulation may be placed in a suitable pressurized propellant, and may be used with additional equipment such as nebulizer or inhaler.
  • compositions are preferably applied as a topical ointment or cream.
  • the active agent may be employed with either a paraffinic or a water-miscible ointment base.
  • the active agent may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • a pharmaceutical composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. 5,399,163; U.S. 5,383,851 ; U.S. 5,312,335; U.S. 5,064,413; U.S. 4,941 ,880; U.S. 4,790,824; or U.S. 4,596,556.
  • a needleless hypodermic injection device such as the devices disclosed in U.S. 5,399,163; U.S. 5,383,851 ; U.S. 5,312,335; U.S. 5,064,413; U.S. 4,941 ,880; U.S. 4,790,824; or U.S. 4,596,556.
  • Examples of well-known implants and modules useful in the present invention include : US 4,487,603, which discloses an implantable micro- infusion pump for dispensing medication at a controlled rate; US 4,486,194, which discloses a therapeutic device for administering medicaments through the skin; US 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; US 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; US 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and US 4,475,196, which discloses an osmotic drug delivery system. Many other such implants, delivery systems, and modules are known to those skilled in the art.
  • the compounds can be administered as the sole active agent, or in combination with other pharmaceutical agents, such as other agents that stimulate or inhibit cell proliferation of immune responses.
  • agents include e.g. cyclosporine, rapamycin, FK506, leflunomide, butenamides, corticosteroids, Doxorubicin, and the like.
  • each active ingredient can be administered either in accordance with its usual dosage range, or at a lower dose level.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • compositions of the invention may optionally contain further active ingredients.
  • Such an engineered strain which strain is further engineered so that the natural loading module is replaced by the loading module from the avermectin or an avermectin like PKS.
  • extender unit genes have been deleted or inactivated and optionally one or more starter unit
  • -A process for producing a polyketide which comprises culturing such an engineered strain in the presence of a non-natural extender unit (and if appropriate a non-natural starter unit) and optionally isolating said polyketide.
  • Escherichia coli DH10B GibcoBRL
  • E. coli JM1 10 New England Biolabs
  • 2xTY medium as described by Sambrook et al. (2001 ).
  • E. coli ET12567(pUZ8002) was grown as described by Paget et al. (1999) in 2xTY medium with kanamycin (25 mg/L) and chloramphenicol (12.5 mg/L).
  • E. coli VCS257 was used for transfection of in vitro packaged cosmids. According to the instructions of Stratagene's Gigapack® III XL Packaging Extract the strain was kept on LB medium and grown on LB plus 0.2% maltose and 1OmM MgSO 4 for transfection. E. coli transformants were selected for with ampicillin (100 mg/L), kanamycin (50 mg/L), apramycin (50 mg/L).
  • the avermectin producer Streptomyces avermitilis (DSM41443J was grown on TSB at 28 0 C for genomic DNA isolation.
  • the FK506 producer Streptomyces tsukubaensis no. 9993 (FERM BP-927) (International Patent Organism Depositary, Tsukuba, Japan) and its derivatives were maintained on medium 1 agar plates or ISP4, ISP3, or ISP2 (see below) at 28 0 C.
  • BIOT-4168 Single spore isolates of both strains, termed BIOT-4168 (containing the genes for FK520 biosynthesis) and BIOT-4206 (containing the genes for FK506 biosynthesis), were used for strain construction.
  • BIOT-4081 , BIOT-4168, BIOT-3119, BIOT-4206 or strains which are described below were prepared after growth on MAM, ISP4, ISP3 or ISP2 agar medium, and preserved in 20% (w/v) glycerol and stored at -80 0 C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 medium and incubated for 5-21 days at 28 0 C.
  • Vegetative cultures were prepared by removing 5-10 agar plugs (6 mm in diameter) from the MAM, ISP4, ISP3 or ISP2 plate and inoculation of 200 - 350 ml. medium NGY in 2 L Erlenmeyer flasks with foam plug. Cultivation was carried out for 48 h at 28 0 C, 250 rpm (2.5 cm throw). The entire seed culture in one flask was transferred into 5 L PYDG containing 0.01-0.05% antifoam SAG 471 , in 7 L Applikon Fermentor. The fermentation medium was pre-adjusted at pH 6.0-7.0 post-sterilization.
  • the fermentation was carried out for 6 days at 28 0 C, with starting agitation set at 300-450 rpm, aeration rate at 0.5-0.8 v/v/m and dissolved oxygen (DO) level controlled with the agitation cascade at 20 - 40% air saturation. If required the pH may be maintained using acid or base addition on demand.
  • the selected feed (providing the starter unit for biosynthesis of target compound) was fed to the production medium 12 - 24 h post inoculation.
  • the feed compound was dissolved in 3 - 5 ml. methanol and added to the culture to give final concentration of 2 mM of the feed compound, the amount of methanol not exceeding 1 % of the total volume. Fermentation was continued for further five days post-feeding.
  • Water used for preparing media was prepared using Millipore ENx Analytical Grade Water Purification System.
  • Oatmeal is cooked/steamed in the water for 20 min, strained through a muslin and more water added to replace lost volume.
  • ISP Trace Elements Solution is added and pH adjusted to 7.2 with NaOH.
  • Agar is added before autoclaving at 121 0 C, 15 min.
  • the medium is adjusted to pH 7.0, with NaOH and then sterilised by autoclaving 121 0 C, 15 min.
  • the medium is adjusted to pH 7.0 with NaOH, and then sterilised by autoclaving 121 0 C, 15 min.
  • MES MES
  • PYDG + MES PYDG + MES
  • a paste is made using a little cold water and the starch. This is brought up to a volume of 500 ml_. All other ingredients are then added, and the pH of the media is adjusted to pH 7.0 - 7.4. Sterilise by autoclaving 121 0 C, 15 min.
  • Genomic DNA preparation was carried out as described in Sambrook et al. (2001 ). PCR was performed according to the instructions of the KOD Polymerase kit (Novagen). DNA sequencing was performed as described previously (Gaisser et al., 2000). Genome sequencing was carried out using 454 technology (Margulies et al., 2005) at Cogenics and the University of Cambridge. Genomic DNA preparation
  • Extractions were repeated until no more protein was visible at the interface, followed by a final chloroform/isoamylalcohol (49:1 ) extraction.
  • the upper phase was precipitated with 1/10 vol. 5 M NaCI and 1 vol. cold isopropanol. After a few min, the DNA was spooled out with a glass rod and washed in ice cold 70% EtOH. After brief drying, the recovered DNA was dissolved in 0.5 - 1 mL TE 10:1.
  • the proteinase K method Karl et al., 2000 was also applied successfully to recover genomic DNA from S. tsukubaensis.
  • a cosmid library of genomic DNA of S. tsukubaensis was constructed. High molecular weight DNA from several genomic DNA preps was partially digested with BfuC ⁇ , an isoschizomer of Sau3A, to a mean size of 30 - 60 kb, ligated to Supercos-1 , packaged into A phage using Gigapack® III XL Packaging Extract (Stratagene) and transfected into Escherichia coli VCS257. The titre was 6.7 x 10 5 cfu / ⁇ g vector. DNA of 10 cosmids was isolated and digested with EcoRI to check the insert size which was 40 kb on average.
  • 2000 clones were grown in 96-well microtitre plates (150 ⁇ L LB Ampl OO Kan50 per well) at 37 0 C and frozen at -80 0 C after mixing wells with 50 ⁇ L LB/glycerol 1 :1.
  • Escherichia coli ET12567 (pUZ8002) (Macneil et al., 1992, Paget et al., 1999) was transformed with pKC1 139B01 -derived plasmids by electroporation to generate the E. coli donor strains for spore conjugation (Kieser et al., 2000). Fresh spores were harvested in water from plates of Streptomyces tsukubaensis (BIOT-4206 or Biot-31 19). Spore suspensions were heat-shocked at 50 0 C for 10 min. They were then mixed with the E. coli donor strain, which had been washed twice with 2xTY, in a ratio of 3:1 Streptomycete to E.
  • Culture broth (0.9 ml.) were extracted with ethyl acetate (0.9 ml.) in a 2 ml. Eppendorf tube.
  • the broth was mixed with the solvent for 15 min on a shaking platform (vibrax) at 400 rpm.
  • the phases are then separated by centrifugation (2 min, 13,200 rpm).
  • An aliquot of the organic layer (0.1 ml.) is then transferred to either a clean glass LC-vial or a vial containing 5 ⁇ g of pimecrolimus (as an internal standard for quantification).
  • the solvent is removed in vacuo (3 min) and then re-dissolved in methanol (1 ml.) by gentle agitation on a shaking platform (5 min).
  • the HPLC system comprised an Agilent HP1100 equipped with a Hyperclone ODS2, C18, 3 micron 4.6 x 150 mm column (Phenomonex). Injection volume 10 ⁇ l_, oven 50 0 C, A: 0.1 % formic acid, B: 0.1% formic acid in MeCN. 1 mL/min; 0-1 min 65% B; 6.5 min 100% B; 10 min 100% B; 10.05 min 65% B, 12 min 65% B.
  • the HPLC system described above was coupled to a Bruker Daltonics Esquire3000 electrospray mass spectrometer. Positive-negative switching was used over a scan range of 500 to 1000 Dalton.
  • LC samples that have been spiked with 0.005 mg/mL pimecrolimus were analysed on the same instrument and with the same chromatographic conditions.
  • FK520 and FK506 analogues can be quantified in this manner, with the parent ion isolated as [M-H] " and the transition to 548.2 (for Fk520 analogues) or 560.2 (for FK506 analogues) used.
  • the amount of analyte present is then calculated by dividing the integral for the analyte transition (as detailed above) with that for the internal standard, pimecrolmius. This ratio is then compared with a standard calibration curve for FK520 or FK506 up to 100 ng on column with 50 ng on column pimecrolimus.
  • NMR spectra 1 H, 13 C, DQF-COSY, TOCSY, HMQC, HMBC, NOESY
  • 500 MHz for proton derived spectra, pro rata for other nuclei
  • Chemical shifts are described in parts per million (ppm) and are referenced to solvent signal e.g. CHCI 3 at ⁇ H 7.26 ( 1 H) and CHCI 3 at ⁇ c 77.0 ( 13 C). J values are given in Hertz (Hz).
  • Genomic DNA was isolated from S. hygroscopicus subsp. hygroscopicus (DSM 40822 assigned BIOT-4081 ) and Streptomyces tsukubaensis (FermBP927 assigned BIOT-3119) using standard protocols described in Kieser et al., (2000).
  • DNA sequencing of cosmids was carried out by the sequencing facility of the Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge CB2 1 QW using standard procedures.
  • a draft genome sequence was obtained using 454 technology by the sequencing facility of the Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge CB2 1 QW.
  • the initial draft genome was generated using a whole 454 chip and this data was subsequently improved by a further Vi chip and assembly of contigs carried out using PHredPHrap.
  • a cosmid library was prepared using isolated genomic DNA of the FK506-producing strain Streptomyces tsukubaensis (BIOT-31 19) using standard methods.
  • the genomic DNA was partially digested with BfuC ⁇ , dephosphorylated and ligated to supercos 1 cut with Xba ⁇ , dephosporylated with Shrimp Alkaline Phosphatase (Roche) and subsequently digested with BamYW. This was packaged into ⁇ phage used to transfect E. coli VCS257 and a library of 2000 clones generated.
  • Probes were made as decribed below for screening Streptomyces tsukubaensis (BIOT- 3119) to identify the FK506 cluster. Plasmid pAES2#2 was used as PCR template. It was obtained by cloning a 1.9kb PCR amplified fkbO fkbP fragment from Streptomyces
  • BIOT-3119 tsukubaensis into pUC19. This fragment had been recruited from BIOT-3119 genomic DNA using primers derived from FK520 fkbO and fkbP sequences.
  • cosmid 4D7 was selected for complete sequencing and covers the regions from 63048 to 109611 bp in the full sequence (SEQ ID No: 7) see table 1.
  • Cosmid 4D7 has an insert of ⁇ 46kb and overlaps with the G9 sequence by ca. 2kb.
  • Cosmid 4D7 contains the genes for biosynthesis of the allyl malonate extension unit and following a region of low homology to data there is a siderophore cluster.
  • Table 1 Regions of cluster sequence covered by contigs and cosmids
  • Table 2 Open reading frame predictions and gene assignments for FK506 cluster
  • fkbC and all genes downstream of fkbG were assigned based on glimmer predictions of ORFs with BLASTX / BLASTP predictions of functions.
  • Streptomyces hygroscopicus analysis of the enzymatic domains in the modular polyketide synthase. Gene 169: 9-16.
  • LL-F28249 antibiotic complex a new family of antiparasitic macrocyclic lactones. Isolation, characterization and structures of LL-F28249 alpha, beta, gamma, lambda. J. Antibiot. 41(4): 519-529.
  • Antascomicinc A, B, C, D and E Novel FKBP12 binding compounds from a
  • Cytokine-stimulated T cells induce macrophage IL-10 production dependent on phosphatidylinositol 3-kinase and p70S6K: implications for rheumatoid arthritis. Arthritis Res. 4(1):64-70. Epub 2001
  • Rapamycin inhibits arterial intimal thickening caused by both alloimmune and mechanical injury. Its effect on cellular, growth factor and cytokine response in injured vessels.
  • Immunosuppressant FK506 promotes neurite outgrowth in cultures of PC12 cells and sensory ganglia. Proceedings of the National Academy of Sciences of the United States of America 91 :3191-3195.
  • MacNeil DJ. , Gewain, K.M., Ruby, C. L., Dezeny, G., Gibbons, P. H., and MacNeil, T. (1992)
  • Streptomyces hygroscopicus analysis of genes flanking the polyketide synthase.
  • A mycophenolate mofetil, and sirolimus (rapamycin) inhibit allergen-induced proliferation and IL-5 production by PBMCs from atopic asthmatic patients.
  • Rabinovitch A., Suarez-Pinzon, W.L., Shapiro, A.M., Rajotte, R.V., Power, R. (2002).
  • the biosynthetic gene cluster for the polyketide immunosuppressant rapamycin is the biosynthetic gene cluster for the polyketide immunosuppressant rapamycin.
  • rapamycin the discovery of SDZ RAD. Transplantation Proceedings 30: 2192-2194. Sehgal, S. N., Baker, H., and Vezina, C. (1975) Rapamycin (AY-22,989), a new antifungal antibiotic II. Fermentation, isolation and characterization. The Journal of Antibiotics 28:
  • actinorhodin production by rpsL encoding ribosomal protein S12 mutations that confer streptomycin resistance in Streptomyces lividans and Streptomyces coelicolor A3(2).
  • Streptomyces hygroscopicus var. ascomyceticus contains genes for biosynthesis of unusual polyketide extender units. Gene 251 : 81-90.
  • Hsp56 component of steroid receptor complexes binds to immobilized FK506 and shows homology to FKBP-12 and FKBP-13. Journal of
  • Rapamycin inhibits hepatic stellate cell proliferation in vitro and limits fibrogenesis in an in vivo model of liver fibrosis. Gastroenterology. 117(5): 1198-204.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to compounds of formula (I) wherein the variables are as defined in the description and their use in therapy.

Description

POLYKETIDES ANALOGUES AND METHODS FOR THEIR PRODUCTION
Field of the Invention
The present invention relates to the use of the genes for biosynthesis of the allyl extender unit of FK506, the generation of novel FK506 and FK520 analogues and strains generated by manipulating one or more of these extender unit genes and optionally feeding non-natural extender units, or crotonic acid precursors thereof, to these strains and to the use of such compounds in therapy.
Background of the invention
FK506 (tacrolimus/fujimycin/Prograf) (Schreiber and Crabtree, 1992) and FK520 (ascomycin or immunomycin) (Wu et al., 2000) (Figure 1 ) are lipophilic macrolides produced by a variety of actinomycetes, including Streptomyces tsukubuaensis No. 9993 (Hatanaka et al., 1989), Streptomyces sp. MA6858, Streptomyces sp MA6548 Streptomyces kanamycetscus KCC S-0433, Strepiomyces davαligems CKD11 19 (Kirn and Park, 2008) which have been shown to produce FK506 (Muramatsu et al., 2005), and Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822), which is equivalent to Streptomyces hyqroscopicus vor ascomyceticus (ATCC 14891 }, pioducmg FK520 (Garπty et al., 1993). Other closely related macrolides include FK525 (Hatanaka H, et al., 1989), FK523 (Hatanaka, H., et al., 1988) and antascomicins (Fehr, T., et al., 1996). A number of semisynthetic derivatives of these molecules have also been shown to be of utility, including pimecrolimus (SDZ ASM 981 , Elidel), which is a derivative of FK520 (Meingassner et al., 1997).
FK506, 1 : R1= -CH=CH2, R2 = trans-OH FK520, 2 : R1= -CH3, R2 = trans-OH FK523, 3 : R1= -H, R2 = trans-OH Pimecrolimus, 4 : R1= -CH3, R2 = c/s-CI
Figure imgf000002_0001
BIOSYNTHESIS: FK506 and FK520 are synthesised by type I polyketide synthases (PKS). This biosynthesis involves a shikimate derived starter unit, followed by 10 extensions utilising malonyl, or substituted malonyl derivatives, thioesters, and addition of a lysine derived pipecolate group. The structure is completed by O-methylation at C-31 and oxidation at C-9 (Motamedi et al., 1996). FK520 and FK506 differ at the C-21 position. FK520 has a C-21 ethyl substituent, whereas FK506 has a C-21 allyl substituent.
MECHANISM OF ACTION: FK506, FK520 and close analogues suppress the immune system by inhibiting signal transduction pathways required for T-cell activation and growth. In particular, they have been shown to inhibit Ca2+-dependent T-cell proliferation, via initial formation of a complex with an FK-binding protein (FKBP), which binds to and blocks calcineurin (CaN). This FK506-FKBP-CaN complex inhibits the activation of nuclear factor of activated t-cells (NF-AT), preventing its entrance into the nucleus, and subsequent activation of the promoter of lnterleukin-2 (IL-2), which initiates IL-2 production. Additionally, FK506 can interfere with the action of calcineurin on substrates other than NFAT, including IKB, Na-K- ATPase and nitric oxide synthase, which may lead to some of the side-effects (Kapturczak et al., 2004).
Treatment with FK506 also seems to be associated with up-regulation of transforming growth factor beta (TGF-β). This cytokine not only has immunosuppressive properties, but may be associated with the development of allograft fibrosis, which can lead to serious
complications after long term treatment with these agents (Kapurtzak et al., 2004).
USES: FK506, in particular, is an important immunosuppressant used to aid prevention of organ rejection after transplantation. For example, it is used intravenously and orally for the prevention of organ rejection after allogeneic liver or kidney transplantation and in bone marrow transplantation. It has been shown to have potential utility in a wide variety of autoimmune, inflammatory and respiratory disorders, including Crohn's disease, Behcet syndrome, uveitis, psoriasis, atopic dermatitis, rheumatoid arthritis, nephritic syndrome, aplastic anaemia, biliary cirrhosis, asthma, pulmonary fibrosis, chronic obstructive pulmonary disease (COPD) and celiac disease.
Treatment of many of these disorders is currently limited to patients with severe disease that are either refractory or hypersensitive to standard treatments. This limitation is due to the side effects of administration of FK506, which include renal dysfunction, gastrointestinal effects, neurological effects, hyperthrichosis and gingival hyperplasia.
Chronic treatment with FK506 requires strict therapeutic monitoring due to its narrow therapeutic index and great inter-individual variability which lead to dangers of over (or under) dosing (Roy et al., 2006).
Pimecrolimus and FK506 are both used in topical formulations, such as ointments and creams, as treatments for a variety of skin conditions, in particular atopic dermatitis (Nghiem et al., 2002).
Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to FK506 metabolism while the P-glycoprotein pump (MDR-1 ) modulates its bioavailability (Roy et al., 2006). The complexity of FK506 dosing is therefore enhanced by significant drug-drug interactions (Kapturczak et al., 2004).
The mechanism of toxicity of FK506 and FK520 has been related to the mechanism of action of immunosuppression (F. Dumont ef a/., 1992). This strong link between the mechanism of action and the toxicity has presented significant challenges to improving the therapeutic index through chemical modification. Segregation of efficacy and toxicity of new analogues may be possible by altering distribution or metabolism (NH Signal et al., 1991 ). By limiting the exposure of the compound to organs that are sensitive to such inhibition, such as the kidney, systemic toxicity can be avoided. Additionally, topical administration of the calcineurin inhibitor at the site of administration (such as skin, lungs, gut, eye etc.) can be maximized. One way this can be achieved is by using a 'soft drug' approach, which involves designing compounds to have limited systemic exposure such as through increased metabolism, higher blood/plasma protein binding, poor absorption or bioavailability.
It has also been suggested that the variable metabolism of FK506 leads to some of the toxicity, due to variable levels of systemic exposure, which led to the need for constant drug monitoring (Armstrong and Oellerich, 2001 ). Therefore, analogues of FK506 with reduced or less variable metabolism could be useful in reducing toxicity, and reducing the need for constant monitoring of drug levels.
FK506 is also poorly bioavailable (Tamura et al., 2003), which leads to variable systemic exposure when dosed orally, and the frequent need for intravenous dosing. Therefore, analogues with improved oral bioavailability would be very useful, to reduce systemic toxicity through incorrect dosing, and improve the ease of oral dosing.
FK506 analogues altered in the side-chain at C21 may demonstrate modulated calcineurin binding. FK506 and FK520 analogues with modulated calcineurin binding may be useful for reduction of toxicity and may demonstrate improved activities in diseases dependent upon immunophillin ligands, including, but not limited to, FKBP12, FKBP51 , FKBP52 and FKBP13, this may include increased neuroprotection, neuroregeneration and reduced neurotoxicity.
Therefore, there remains a need to identify novel FK506 and FK520 analogues, which may have utility in the maintenance of immunosuppression, both for organ transplantation, and for the treatment of inflammatory conditions, and for the treatment of fungal infections. The present invention discloses novel FK506 and FK520 analogues which have altered
pharmaceutical properties compared with the currently available FK506 and FK520 analogues; these properties may be useful for therapies requiring good systemic bioavailability, including, but not limited to oral therapies to maintain immunosuppression, which in particular are expected to show improvements in respect of one or more of the following properties:
increased metabolic stability, increased bioavailability, increased oral bioavailability, reduced efflux via membrane transporters and low plasma protein binding; the novel FK506 and FK520 analogues may also be useful for therapies requiring local availability but with poor systemic availability, including, but not limited to topically administered therapies for inflammatory disorders such as atopic dermatitis, asthma and inflammatory bowel diseases, which in particular are expected to show improvements in respect of one or more of the following properties: decreased metabolic stability, decreased bioavailability, decreased oral
bioavailability, increased efflux via membrane transporters and high plasma protein binding; Other properties the molecules might be expected to have that are of general use, include improved formulation ability, improved potency, increased FKBP binding, improved
toxicological profile, reduced nephrotoxicity and neurotoxicity, improved crystallinity, improved water solubility or improved lipophilicity.
Summary of the Invention
In the present invention, the complete biosynthetic cluster of FK506 is described. This includes the genes dedicated to provision of the allyl extender unit at C21 of FK506 which comprises some or all of ORF1 to ORF9 in Table 2. Analysis of gene function indicates a pathway in which a 3-carbon unit derived from propionyl-CoA is condensed with a 2-cabron unit derived from malonyl-CoA to give a 5-carbon chain. Selection and covalent attachment of at least one of these units is performed by ORF9, a dual acyltransferase-acylcarrier protein. The enzyme responsible for condensation is the ketosynthase ORF8. The 3-carbon unit is most likely modified to an acrylic acid derivative prior to condensation by the action of an acyl-CoA dehydrogenase, most probably ORF6 but possibly ORF2. Alternatively it is possible that unsaturation of the terminal 2-carbons is achieved after condensation by the action of an oxygenase such as ORF3. After ORF8 catalysed condensation the resulting acylcarrier protein- or coenzyme-A linked 5-carbon acid is modified to a crotonic acid derivative by the action of 'house-keeping' enzymes from the fatty acid biosynthetic pathway, specifically a beta- ketoacylreductase and a dehydratase. Other possible enzymes involved in this chemistry include the short chain dehydrogenase/reductase ORF2. The final step of biosynthesis is a reductive carboxylation catalysed by the ccr homologue ORF7 to a give a 3-carbon (allyl) substituted malonic acid thioester derivative.
Provision of the allyl extender unit may be engineered out of the cell by, for example, disruption of, or deletion of, one or more of the genes responsible for the provision of the allyl extender unit. Exogenous compounds which are accepted into this extender position may then be fed. The FK506 cluster ccr gene (ORF7) may be retained in order to carboxylate the fed acid or ester appropriately for incorporation into the PKS. Alternatively a heterologous ccr gene may be expressed in the engineered organism in order to allow incorporation of a broader set of extender units. The generation of FK506 analogues at C21 may be combined with previous strategies for the production of FK506 and FK520 analogues, of engineering a strain that cannot provide the natural starter unit and feeding exogenous acids or esters and generating engineered strains where the the natural loading module from the FK506 or FK520 polyketide synthase is replaced by the loading module from the avermectin or an avermectin-like polyketide synthase, and optionally non-natural starter units are fed to these strains. These strains optionally having been mutated by classical methods or targeted inactivation or deletion of one or more genes responsible for post-PKS modification, and/or mutated by classical methods or targeted inactivation or deletion of one or more precursor supply genes, including bkd genes (Ward et al., 1999) and homologues thereof.
Thus in one aspect of the invention there is provided a compound of formula (I)
Figure imgf000006_0001
wherein
(ι) Xa and Xb represent a bond; or
(ιι) Xa represents a bond and Xb represents CH2, S, O; or
(in) Xa represents CH2, S, O, fused cyclopropyl unit and Xb represents a bond;
Z represents keto or CH2;
Ri represents a moiety selected from:
Figure imgf000007_0001
A B
Figure imgf000007_0002
D E F
Figure imgf000007_0003
I
G H
Figure imgf000007_0004
V W X Y R2 represents H, alkyl, halo, hydroxyl or thiol;
R3 represents H, Ci-4alkyl, halo, hydroxyl or thiol;
R4 represents H, Ci-4alkyl, halo, hydroxyl or thiol;
R5 represents OMe, Me or H;
R6 represents OMe, Me or H;
R7 represents Ci-6alkyl or Ci-6alkenyl, linear or branched, in either case optionally substituted by one to three (e.g. one or two especially one) halogen atoms, save that R7 does not represent -CH2CH2 or -CH2CH=CH2 and when R1 = P, Q, R, S, T, U, V, W, X or Y, then R7 does not represent CH3;
R8 represents OH;
R9 represents H, OH, halo, thiol, CrC4 alkyl;
Rio, Rn and Ri2 independently represent F, Cl, CrC4 alkyl, ORi3, SRi3 or NHRi3 and Ri3 represents H, CrC4 alkyl or CrC4 acyl, wherein two or three of Rio-Ri2 are CrC4 alkyl;
Ri3 and Ri4 independently represent H, F, Cl, CrC4 alkyl, ORi5, SRi5 or NHRi5 and Ri5 represents H, CrC4 alkyl or CrC4 acyl;
Ri6 Ri7 and Ri8 independently represent H, F, Cl, CrC4 alkyl, ORi9, SRi9 or NHRi9 and Ri9 represents H, CrC4 alkyl or CrC4 acyl,
R20 and R2i independently represent H, F, Cl, CrC4 alkyl, OR22, SR22 or NHR22 and R22 represents H, CrC4 alkyl or CrC4 acyl,
R23 and R24 independently represent H, F, Cl, CrC4 alkyl, OR25, SR25 or NHR25 and R25 represents H, CrC4 alkyl or CrC4 acyl;
R26 represents H, F, Cl, CrC4 alkyl, OR27, SR27 or NHR27, and R27 represents H, CrC4 alkyl or
CrC4 acyl;
R28 and R29 independently represent H, F, Cl, CrC4 alkyl, OR30, SR30 or NHR30 and R30 represents H, CrC4 alkyl or CrC4 acyl ;
R3i R32 and R33 independently represent H, F, Cl, CrC4 alkyl, OR34, SR34 or NHR34 and R34 represents H, CrC4 alkyl or CrC4 acyl;
R35 R36 and R37 independently represent H, F, Cl, CrC4 alkyl, OR38, SR38 or NHR38 and R38 represents H, CrC4 alkyl or CrC4 acyl, save that OR41 shall not represent OH;
R39 R40, R4i and R42 independently represent H, F, Cl, CrC4 alkyl, OR43, SR43 or NHR43 and R43 represents H, CrC4 alkyl or CrC4 acyl,
R44 and R45 independently represent H, F, Cl, CrC4 alkyl, OR46, SR46 or NHR46 and R46 represents H, CrC4 alkyl or CrC4 acyl;
R47, R48 and R49 are independently selected from H, F, Cl, OR50, SR50 and NHR50 and R50 is selected from H, Ci-C4alkyl and Ci-C4acyl;
A is selected from O, S and NR5i, R5i represents H, Ci-C4alkyl or Ci-C4acyl, R52 and R53 are independently selected from H, Ci-C4alkyl, OR54, SR54 and NHR54, R54 represents Ci-C4alkyl or CrC4acyl, Xc represents a bond or CH2, when Xc is bond then R54 represents H and when Xc is
CH2 then R54 represents H, F, Cl, OH, SH, Ci-C4 alkyl, OR55, SR55 or NHR55 and R55 represents Ci-C4alkyl or Ci-C4 acyl;
B represents O, S, or NR6O, R6o represents H, Ci-C4alkyl, or Ci-C4acyl, R56 represents d-
C4alkyl, OR6i, SR6i or NHR6i, R6i represents Ci-C4alkyl or Ci-C4acyl, R58 represents H, F, Cl,
OH, SH, CrC4 alkyl, OR62, SR62 or NHR62, R62 represents Ci-C4 alkyl or CrC4acyl, Xd and Xe independently represent bond or CH2 provided that Xd and Xe do not both represent CH2 and when Xd is bond then R57 represents H and when Xe is CH2 then R57 represents H, F, Cl, OH,
SH, Ci-C4alkyl, OR63, SR63 or NHR63, R63 represents CrC4 alkyl or CrC4 acyl and when Xe is bond R59 represents H and wherein when Xe represents CH2 then R59 represents H, F, Cl, OH,
SH, CrC4 alkyl, OR64, SR64 or NHR64, and R64 represents Ci-C4alkyl or Ci-C4acyl;
R65 and R66 independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR67, SR67 or NHR67, and
R67 represents Ci-C4alkyl or CrC4 acyl;
R68 represents H, F, Cl, OH, SH, CrC4alkyl, OR69, SR69 or NHR69, and R69 represents CrC4 alkyl or CrC4 acyl;
R70 and R7i independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR72, SR72 or NHR72 and
R72 represents CrC4 alkyl or CrC4 acyl ;
R73, R74 and R75 independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR76, SR76 or NHR76 and R76 represents CrC4 alkyl or CrC4 acyl;
R77, R78 and R79 independently represent H, F, Cl, OR82, SR82, CrC4 alkyl or CN, R82 represents H, CrC4 alkyl or CrC4 acyl, R80 and R8i independently represent H, F, Cl, OH, SH or CrC4 alkyl, provided that at least one of R77, R78 and R79 is not H or CrC4 alkyl;
R83, R84 and R85 are independently selected from H, F, Cl, OR88, SR88, C1-C4 alkyl and CN, R88 represents H, CrC4 alkyl or CrC4 acyl, R86 and R87 independently represent H, F, Cl, OH, SH or CrC4 alkyl and provided that at least one of R83, R84 and R85 does not represent H or CrC4 alkyl;
and physiologically functional derivatives thereof.
The above structure shows a representative tautomer and the invention embraces all tautomers of the compounds of formula (I) for example keto compounds where enol compounds are illustrated and vice versa. All isotopic variants (including deuterated and tritiated varians) are also embraced.
The invention embraces all stereoisomers of the compounds defined by structure (I) as shown above.
In a further aspect, the present invention provides FK506 or FK520 analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use as a pharmaceutical. In a further aspect, the present invention provides strains containing hybrid PKS, with a loading module conferring avermectin-like chain initiation, and the rest of the PKS conferring FK506/FK520-like chain processing and termination.
In a further aspect, the present invention provides processes for production of FK506 and FK520 analogues defined by structure (I) above.
Definitions:
The articles "a" and "an" are used herein to refer to one or to more than one (i.e. at least one) of the grammatical objects of the article. By way of example "an analogue" means one analogue or more than one analogue.
As used herein the term "analogue(s)" refers to chemical compounds that are structurally similar to another but which differ slightly in composition (as in the replacement of one atom by another or in the presence or absence of a particular functional group).
As used herein the terms "FK506 and FK520 analogues" / "FK506 or FK520 analogues" refer to compounds related to FK506, FK520 and similar compounds in structure. Such compounds are 22-membered rings with one lactone and one amide bond. The N of the amide bond forms a 2-carboxyl piperidine or a 2-carboxyl pyrrolidine. This carboxyl group forms the lactone group, with an oxygen that is allylic to a double bond that is exo to the main 22-membered ring. Such compounds include, without limitation, FK520, FK506, antascomicin, FK523, FK525, pimecrolimus and tsukubamycin as well as compounds of formula (I).
As used herein the term "FK506 or FK520 producing strain" refers to a strain (natural or recombinant) which is capable of producing one or more FK506 or FK520 analogues when fed appropriately.
As used herein the term "recombinant strain of a FK506 or FK520 producing host" refers to a recombinant strain based on a natural FK506 or FK520 producing strain which is capable of producing one or more FK506 or FK520 analogues when fed appropriately. As used herein the term "FK506 or FK520 cluster" means the PKS and associated enzymes responsible for production of FK506 or FK520 analogues.
As used herein the term "modifying gene(s)" includes the genes required for post- polyketide synthase modifications of the polyketide, for example but without limitation cytochrome P-450 monooxygenases, ferredoxins and SAM-dependent O-methyltransferases. In the FK520 system these modifying genes include fkbD and fkbM, but a person of skill in the art will appreciate that PKS systems related to FK520 (for example but without limitation:
FK506, antascomicin, FK523, FK525 and tsukubamycin) will have homologues of at least a subset of these genes, some of which are discussed further below.
As used herein the term "precursor supply gene(s)" includes the genes required for the supply of the natural or non-natural precursors, the genes required for the synthesis of any naturally or non-naturally incorporated precursors and the genes required for the incorporation of any naturally or non-naturally incorporated precursors. For example but without limitation in the FK506 and FK520 system these genes include fkbL, fkbO and fkbP but a person of skill in the art will appreciate that PKS systems related to FK506 and FK520 (for example but without limitation: antascomicin, FK523, FK525 and tsukubamycin) will have homologues of these genes, some of which are discussed further below. Additionally it may be useful to inactivate homologues of the bkd genes, which are involved in the generation of branched chain keto- acids, such as might be expected to incorporate preferentially into avermectin type loading modules, such as those described in Ward et al. 1999 (e.g. Wei et al., 2006).
As used herein, the term "extender unit(s)" includes references to carboxylic acids or esters fed to cells that are then further processed by the addition of CoA or an acylcarrier protein, and/or also by reductive carboxylation, halogenation or any other cellular process that converts the fed compound into a functional extender unit for the PKS.
As used herein, the term "auxiliary gene(s)" includes references to modifying genes, precursor supply genes or both modifying genes and precursor supply genes. One example of an auxiliary gene is an oxygenase which may hydroxylate the starter unit.
As used herein the term "basic product" refers to the initial product of the polyketide synthase enzyme before the action of any modifying genes.
As used herein, the term "precursor" includes the natural starter units (i.e. 4,5- dihydroxycyclohex-1-ene carboxylic acid), non-natural starter units (e.g non-cyclic or heterocyclic starter units), and naturally incorporated amino acids (i.e. pipecolic acid) and non- naturally incorporated amino acids
As used herein the term "non-natural starter unit" refers to any compounds which can be incorporated as a starter unit in polyketide synthesis that are not the starter unit usually incorporated by that PKS.
The term "avermectin -I ike PKS" means the PKS of a bacterium producing a avermectin-like polyketide such as nemadectin or milbemycin which contains a loading domain consisting of AT and ACP domain and which naturally incorporates branched chain starter acids such as those of the present invention.
The term "acyl" means an alkyl group in which the first carbon atom is a carbonyl moiety.
Examples of C1-4 acyl groups include C2-4 acyl groups such as -COMe and -COEt, especially COMe. CHO (i.e. C1 acyl) is a further example which is less preferred.
Examples of C1-4 alkyl groups include Me, Et, n-Pr, i-Pr, n-Bu, especially Me. Detailed Description of the Invention
Physiologically functional derivatives of compounds of formula (I) include physiologically acceptable salts, esters and solvates. Pharmaceutically acceptable salts include the non-toxic acid addition salt forms of the compounds of formula (I). The pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid. Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulphuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e.
ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic (i.e. hydroxyl- butanedioic acid), tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic,
p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.
Example esters include labile esters which are cleaved in the body, for example carboxylic acid esters formed with hydroxyl groups. Example solvates include hydrates.
In one embodiment Xa represents a bond. In another embodiment Xa represents CH2. Preferably Xa represents CH2.
In one embodiment Xb represents a bond.
In one embodiment Z represents =0.
In one embodiment R5 represents OMe.
In one embodiment R6 represents OMe.
In one embodiment R2 represents H. In one embodiment R3 represents H.
In one embodiment R4 represents H.
The alkyl or alkenyl group of R7 may optionally be substituted by one to three e.g. one or two or three halogen atoms. In one embodiment the alkyl or alkenyl group of R7 is not substituted by halogen atoms. In one embodiment the alkyl or alkenyl group of R7 is substituted by one halogen atom. In one embodiment the alkyl or alkenyl group of R7 is substituted by two halogen atoms. In one embodiment the alkyl or alkenyl group of R7 is substituted by three halogen atoms. Suitably if the alkyl or alkenyl group of R7 is substituted by halogen atoms, the halogen atoms are located on the terminal carbon atom of the alkyl or alkenyl group. In one embodiment R7 is a linear group. In another embodiment R7 is a branched group. R7 may for example be selected from CH3, CH2CH2CI, CH2CH2F,
CH2CH2CH3, CH2(CH2^CH3, CH2CH2Br, CH2CH2D, CH2CH2CH2F, CH2CH2CH2CI,
CH2CH2CH2Br and CH=CHCH3
In one embodiment R1 represents [A]. In another embodiment R1 represents [B]. In another embodiment R1 represents [C]. In another embodiment R1 represents [D]. In another embodiment R1 represents [E]. In another embodiment R1 represents [F]. In another embodiment R1 represents [G]. In another embodiment R1 represents [H]. In another embodiment R1 represents [J]. In another embodiment R1 represents [K]. In another embodiment R1 represents [L]. In another embodiment R1 represents [M]. In another embodiment R1 represents [N]. In another embodiment R1 represents [O]. In another embodiment R1 represents [P]. In another embodiment R1 represents [Q]. In another embodiment R1 represents [R]. In another embodiment R1 represents [S]. In another embodiment R1 represents [T]. In another embodiment R1 represents [U]. In another embodiment R1 represents [V]. In another embodiment R1 represents [W]. In another embodiment R1 represents [X]. In another embodiment R1 represents [Y].
Compounds of formula (I) may be produced by preventing the in vivo production of the natural extender unit for example but not restricted to interrupting or deleting one or more essential genes such as ORF8 (the KS) or ORF 6 (the AT/ACP), and feeding exogenous carboxylic acids (or derivatives thereof) which can be incorporated; or preventing the in vivo production of the natural extender unit as above while retaining the function of the ccr gene either in its natural position or by providing it in trans for example at an attachment site, and feeding exogenous carboxylic acids (or derivatives thereof) which can be incorporated; or preventing the in vivo production of the natural extender unit as above while retaining the function of a ccr for example a heterologous ccr which may have a different substrate tolerance for example but not limited to the nove chloro-ccr gene of salinosporamide, salG (Liu et al 2009) and feeding exogenous acids which can be incorporated. In order to enhance the incorporation of halogenated substrates salL could also be expressed in the host cell.
This can be done in combination with interrupting the starter unit biosynthesis and feeding exogenous acids such as in WO2004/007709 (which document is incorporated in its entirety by reference) or replacing the natural loading module of the FK506 or FK520 PKS with that of the avermectin or an avermectin-like PKS (such as nemadectin or milbemycin), and feeding an appropriate non-natural starter unit to the resultant strain such as in
PCT/GB2009/050689 (which document is incorporated in its entirety by reference).
In each case this would be followed by culturing the strain and optionally isolating the compounds thereafter.
For example fkbO is required for the provision of the natural starter unit, 4,5-di- hydroxycyclohex-1-ene carboxylic acid. Disruption, deletion or otherwise removing the function of FkbO (for example but not limited to a frameshift deletion or the use of UV mutagenesis followed by screening for a non-producer of FK506 that is restored to production by feeding exogenous 4,5-di-hydroxycyclohex-1-ene carboxylic acid to the culture medium) leads to a strain that can be used to make FK506 or FK520 analogues by feeding exogenous analogues of the natural starter unit to the production medium.
The loading modules of avermectin and avermectin-like PKSs consist of an AT and an ACP domain. In an optional embodiment the strain is mutated to inactivate or delete of one or more genes that contribute to the biosynthesis or regulation of precursor supply. For example the gene(s) that contribute to the biosynthesis or regulation of precursor supply may contribute to the biosynthesis or regulation of branched chain keto acid, such as bkd (Ward et al., 1999). By removal of the means of generation of branched chain keto acids, incorporation of other non-natural starter acids is facilitated due to lack of competition with the preferential substrate of the avermectin loading module. In an alternative embodiment the strain may be mutated to inactivate or delete of one or more genes responsible for biosynthesis of pipecolic acid, such as fkbL (Wu et al., 2000). Pipecolic acid is naturally incorporated into the chain as a final step prior to ring closure. This particular modification increases the yield of prolyl derivatives of formula (I) when proline is fed to the strain.
Additionally or instead in an alternative embodiment the strain may be mutated to have targeted inactivation or deletion of one or more genes responsible for post-PKS modification, for example the gene responsible for oxidation at the C-9 position, fkbJ (Motamedi and Shafiee, 1998, Wu et al., 2000). This particular modification increases the yield of C-9 desketo derivatives of formula (I).
If appropriate or necessary compounds so produced may be subject, after their isolation, to synthetic alteration using processes known to a skilled person e.g. alkylation of hydroxyl and amino groups and the like.
It should be understood by one skilled in the art that it may be possible to make different engineered strains that display the same phenotype eg there are multiple genes that can be either disrupted or deleted to remove the provision of the extender unit. A deletion may be the removal of a small part of a gene or most of a gene or all of a gene and it may be done in- frame, or it may be a frameshift. This equally applies to engineering methods to remove starter unit supply and to the use of alternative junctions to join the avermectin loading module to the rapamycin PKS. For example the specificity of an avermectin-like loading module may be transferred to the FK506 or FK520 cluster by taking just the Acyltransferase (AT) and Acyl Carrier Protein (ACP) domains from the avermectin PKS and joining to the FK506/FK520 PKS before the first ketosynthase (KS) domain. Alternatively, the junction may be made between the KS from the avermectin PKS and the first extender AT from the FK506/FK520 PKS. Analogous possibilities are discussed in WO 98/01546. Hence the splice junction in the hybrid PKS between the avermectin load and the FK506/FK520 PKS may be before, within or after the KS of the first extension module. Thus part or all of the KS of the first extension domain of the recombinant strain is from the avermectin PKS. The loading module (optionally together with part or all of the KS of the first extension domain) of an avermectin-like PKS may be used in place of the avermectin loading module, such as those loading modules found in the
milbemycin PKS or nemadectin PKS.
In one method of generating a suitable strain, a precursor supply gene or genes such as the bkd genes or homologues thereof may be manipulated by targeted inactivation or deletion or modified by other means such as exposing cells to UV radiation and selection of the phenotype indicating that branched chain alpha keto acid biosynthesis has been disrupted. The optional targeting of the post-PKS genes may occur via a variety of mechanisms, e.g. by integration, targeted deletion of a region of the FK506 or FK520 cluster including all or some of the post-PKS genes optionally followed by insertion of gene(s) or other methods of rendering the post-PKS genes or their encoded enzymes non-functional e.g. chemical inhibition, site- directed mutagenesis or mutagenesis of the cell for example by the use of UV radiation.
WO2004/007709 provides methods for the alteration of a gene system which comprises a core portion responsible for the production of a basic product, and a multiplicity of modifying genes responsible for effecting relatively small modifications to the basic product - e.g.
effecting oxidation, reduction, alkylation, dealkylation, acylation or cyclisation of the basic product, and a multiplicity of precursor supply genes which are involved in the production of particular precursor compounds. Thus the basic product may be a modular polyketide and the modifying genes may be concerned with modifications of a polyketide chain (such as oxidation at the 9 position), and the precursor supply genes may be involved in the production and/or incorporation of natural or non-natural precursors (e.g. pipecolate and/or 4,5- dihydroxycyclohex-1-ene carboxylic acid).
The core portion may not function properly or even at all in the absence of a precursor supply gene (unless a natural or unnatural precursor compound is supplied or is otherwise available). Therefore, the deletion or inactivation of a precursor supply gene provides a system where it is possible to incorporate non-natural starter units with no competition from the natural starter unit.
Therefore, the present invention provides a method for the incorporation of non-natural extender units into FK506 and FK520 analogues said method comprising removing the ability of the producing strain to provide the extender unit and feeding alternative compounds which may be incorporated, possibly following modification in vivo to become a suitable substrate for the PKS. In combination, the the present invention provides a method for additionally incorporating non-natural starter acids into FK506 and FK520 analogues, said method comprising either deleting or inactivating the ability of the cell to produce the natural starter unit, for example by targeted deletion or inactivation of fkbO, or by replacing the natural
FK506/FK520 loading module with the loading module of avermectin or an avermectin-like PKS and feeding starter units to this strain which optionally contains chromosomal DNA in which the precursor supply gene has been deleted or inactivated. Suitable gene systems which express FK506/FK520 homologues include, but are not limited to, antascomicin, FK520 (Wu et al., 2000; U.S. 6,150,513; AF235504), FK506 (Motamedi et al., 1996; Motamedi et al., 1997;
Motamedi and Shafiee, 1998; AF082100, Y10438), FK523, FK525 and tsukubamycin biosynthetic clusters. The precursor supply gene which is deleted or inactivated is preferably a gene whose product is involved in branched chain keto acid formation, such as a bkd gene or homologue. The gene system is preferably the FK506 or FK520 cluster. The precursor supply gene deleted or inactivated is more preferably one or more of the bkd genes. Optionally fkbM and/or fkbl are deleted or inactivated in addition to one or more of the bkd genes. Optionally fkbL, or an analogue of rapL is also deleted to allow more efficient incorporation of pipecolate analogues (Motamedi and Shafiee, 1998, Khaw et al., 1998)
Therefore in one aspect the present invention provides a method of producing compounds of formula (I) comprising:
(a) generating a recombinant strain of a FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated; and
(b) feeding a non-natural C21 extender unit to said recombinant strain;
(c) culturing said strain; and
(d) optionally isolating compounds of formula (I).
Also there is provided a method of producing compounds of formula (I) comprising:
(a) feeding a non-natural C21 extender unit to a recombinant strain of a FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 and FK520 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated;
(b) culturing said strain; and
(c) optionally isolating compounds of formula (I).
Also there is provided a method of producing compounds of formula (I) comprising:
(a) generating a recombinant strain of a non-FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated; and
(b) feeding a non-natural C21 extender unit to said recombinant strain;
(c) culturing said strain; and
(d) optionally isolating compounds of formula (I).
Also there is provided a method of producing compounds of formula (I) comprising:
(a) feeding a non-natural C21 extender unit to a recombinant strain of a non-FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 and FK520 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated;
(b) culturing said strain; and (c) optionally isolating compounds of formula (I)
Moreover the strains may be further engineered so as not to produce the natural starter unit. For example, the strains may be further engineered so that the gene fkbO is deleted or inactivated.
Also the strains may be further engineered so that the natural loading module is replaced by the loading module from the avermectin or an avermectin like PKS.
Thus there is also provided a method of producing compounds of formula (I) comprising:
(a) feeding a non-natural starter unit and a non-natural C21 extender unit to a recombinant strain of a FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 analogue synthesis in which the natural loading module has been replaced by the loading module from the avermectin or an avermectin-like PKS and in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated;
(b) culturing said strain; and
(c) optionally isolating compounds of formula (I).
Optionally one or more starter genes have been deleted or inactivated which produce a starter unit (e.g. starter acid). Additionally or instead the precursor for which one or more starter genes have been deleted or inactivated may be pipecolic acid. Optionally one or more genes responsible for post-PKS modification are also deleted or inactivated.
In a preferred embodiment the recombinant strain is generated using the methods described in WO2004/007709 and in the examples below.
In one embodiment, the host strain is a selected from the group consisting of
Streptomyces tsukubaensis No. 9993 (Ferm BP-927), Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822), Streptomyces sp. AA6554, Streptomyces hygroscopicus var. ascomyceticus MA 6475 ATCC 14891 , Streptomyces hygroscopicus var. ascomyceticus MA 6678 ATCC 55087, Streptomyces hygroscopicus var. ascomyceticus MA 6674, Streptomyces hygroscopicus var. ascomyceticus ATCC 55276, Streptomyces hygroscopicus subsp.
ascomyceticus ATCC 14891 , Sireptαmyees kanamyceϋcus KCC S-0433, Streptomyces davuϋgerus CKD11 19 (Kim and Park, 2008). Streptomyces hygroscopicus subsp.
yakushimaensis, Streptomyces sp. DSM 7348, Micromonospora n.sp. A92-306401 DSM 8429 Streptomyces sp. MA 6548 and Streptomyces sp. MA 6858 ATCC 55098. In a preferred embodiment the host strain is selected from the group consisting of: S. hygroscopicus var. ascomyceticus ATCC 14891 , Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) or Streptomyces tsukubaensis No. 9993 (Ferm BP-927). If desired or necessary one or more auxiliary genes may be deleted or inactivated in the host strain. If desired or necessary one or more of the deleted or inactivated genes of the host strain may be reintroduced by complementation (e.g. at an attachment site, on a self-replicating plasmid or by insertion into a homologous region of the chromosome).
If desired or necessary, further chemical steps, known to one skilled in the art may be used to generate the final compound (for example see March, Wiley Interscience)
It is well known to those skilled in the art that polyketide gene clusters may be expressed in heterologous hosts (Pfeifer et al., 2001 ). Accordingly, the present invention includes the transfer of the FK506 or FK520 biosynthetic gene cluster with or without resistance and regulatory genes, either complete, engineered, containing mutations, or containing deletions, for complementation in heterologous hosts. Methods and vectors for the transfer as defined above of such large pieces of DNA are well known in the art (Rawlings, 2001 ; Staunton and Weissman, 2001 ) or are provided herein in the methods disclosed.
Therefore in another aspect the present invention provides a method of producing compounds of formula (I) comprising:
(a) generating a recombinant strain of a non-FK506 producing host (i.e. a heterologous host) that contains a biosynthetic cluster that encodes polypeptides involved in FK506 and FK520 analogue synthesis in which the strain has been engineered by any of the methods described above; and
(b) feeding a non-natural extender unit and a non-natural starter unit to said
recombinant strain;
(c) culturing said strain; and
(d) optionally isolating compounds of formula (I).
In this context a preferred heterologous host cell strain is a prokaryote, more preferably an actinomycete or Escherichia coli, still more preferably include, but are not limited to S.
hygroscopicus, S. hygroscopicus sp. , S. hygroscopicus var. ascomyceticus, Streptomyces tsukubaensis, Streptomyces coelicolor, Streptomyces lividans, Saccharopolyspora erythraea, Streptomyces fradiae, Streptomyces avermitilis, Streptomyces cinnamonensis, Streptomyces rimosus, Streptomyces albus, Streptomyces griseofuscus, Streptomyces longisporoflavus, Streptomyces venezuelae, Micromonospora griseorubida, Amycolatopsis mediterranei or Actinoplanes sp. N902-109.
Those skilled in the art will appreciate that the methods of the present invention could be applied to recombinant host strains in which the polyketide synthase (PKS) has been altered by genetic engineering to express a modified FK506 or FK520 analogue or other polyketide analogue. For example the PKS in a homologous or heterologous could be a hybrid PKS in which one or more domains have been removed, replaced or inserted, such replacements or insertions coming from other heterologous (or homologous) PKS clusters. The prior art describes several methods for the production of novel polyketides by the deletion or inactivation of individual domains (WO93/13663, WO97/92358), construction of hybrid polyketide synthases (WO98/01546, WO00/00618, WO00/01827) or alteration of domain specificity by site-directed mutagenesis (WO02/14482).
It is well known that many actinomycetes contain multiple biosynthetic gene clusters for different secondary metabolites, including polyketides and non-ribosomally synthesised peptides. Specifically, it has been demonstrated that strains of S. hygroscopicus produce a variety of polyketides and non-ribosomally synthesised peptides in addition to FK506, FK520, FK523, meridamycin, FK525, antascomicin or tsukubamycin. These include, but are not limited to, elaiophylin, bialaphos, hygromycin, augustmycin, endomycin (A, B), glebomycin, hygroscopin, ossamycin and nigericin. These additional biosynthetic gene clusters represent a competing requirement for biosynthetic precursors and an additional metabolic demand on the host strain. In order to enhance production of the desired rapamycin, or other polyketide, analogues, it may therefore be advantageous to delete or inactivate any other biosynthetic gene clusters present in the host strain. Methods for the deletion or inactivation of biosynthetic gene clusters are well known in the art.
C21 extender units and starter units are suitably provided as the free carboxylic acid, but derivatives that may be employed include salts and esters.
The aforementioned C21 extender and starter unit substances are either known or may be prepared by a skilled person using conventional methods.
For instance the C21 extender units may be the appropriate derivative of malonic acid (e.g. to incorporate CH3 then methyl malonate (CH3-CH-(COOH)2) could be used) or else the corresponding C3-substituted acrylate (having one more carbon atom than that of the R7 side chain) could be used (e.g. to incorporate CH3(CH2)5- then CH3(CH2)3CH=CH-COOH would be used).
Standard methods known to those of skill in the art may be used to culture the host or recombinant strain in order to produce compounds of formula (I). Such methods include, without limitation, those described in the examples below; additional methods may also be found in Reynolds and Demain, 1997 and references therein.
Compounds of the invention may be isolated using standard methods known to those of skill in the art, including, without limitation, those described in the methods of the examples below. Alternatives to these methods which may also be considered by a person of skill in the art include those as described in Natural Products Isolation (Cannell et al., 1998).
Compounds of formula (I) are useful as pharmaceuticals for example, but without limitation, having potential utility as immunosuppressants, antifungal agents, anticancer agents, neuroregenerative agents, or agents for the treatment of psoriasis, rheumatoid arthritis, fibrosis and other hyperproliferative diseases. In a further aspect, the invention provides for the use of a compound of formula (I) as disclosed herein, in the preparation of a medicament for the prophylaxis and/or treatment of organ rejection after transplantation, autoimmune diseases, inflammatory disorders, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthrisis, fibrosis and/or other hyperproliferative disorders. In a further aspect, the invention provides for a method of treatment or prophylaxis of organ rejection after transplantation, autoimmune diseases, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthritis, fibrosis and/or other hyperproliferative disorders comprising administering a compound of formula (I) to a subject in need thereof. Additionally, the compounds of formula (I) disclosed herein may be used in the preparation of a medicament for the prevention of organ allograft rejection. In a preferred embodiment the compounds of formula (I) are used in the preparation of a medicament for the treatment of autoimmune diseases or inflammatory disorders.
One skilled in the art would be able by routine experimentation to determine the ability of these compounds to inhibit fungal growth (e.g. Baker, H., et al., 1978; NCCLS Reference method for broth dilution antifungal susceptibility testing for yeasts: Approved standard M27-A, 17(9). 1997). Additionally, one skilled in the art would be able by routine experimentation to determine the ability of these compounds to inhibit tumour cell growth, (see Dudkin, L., et al., 2001 ; Yu et al. 2001 ). In a further aspect the compounds of this invention are useful for inducing immunosuppression and therefore relate to methods of therapeutically or
prophylactically inducing a suppression of a human's or an animal's immune system for the treatment or prevention of rejection of transplanted organs or tissue, the treatment of autoimmune, inflammatory, proliferative and hyperproliferative diseases (examples include but are not inclusively limited to autoimmune diseases, diabetes type I, acute or chronic rejection of an organ or tissue transplant, asthma, tumours or hyperprolific disorders, psoriasis, eczema, rheumatoid arthritis, fibrosis, allergies and food related allergies). Such assays are well known to those of skill in the art, for example but without limitation: Immunosuppressant activity - Warner, IM., et al., 1992, Kahan et al. (1991 ) & Kahan & Camardo, 2001 ); Allografts - Fishbein, T. M., et al., 2002, Kirchner et al. 2000; Autoimmune / Inflammatory / Asthma - Carlson, R.P. et al., 1993, Powell, N. et al., 2001 ; Diabetes I - Rabinovitch, A. et al., 2002; Psoriasis - Reitamo, S. et al., 2001 ; Rheumatoid arthritis - Foey, A., et al., 2002; Fibrosis - Zhu, J. et al., 1999, Jain, S., et al., 2001 , Gregory et al. 1993
The ability of the compounds of this invention to induce immunosuppression may be demonstrated in standard tests used for this purpose. In a further aspect the compounds of this invention are useful in relation to antifibrotic, neuroregenerative and anti-angiogenic mechanisms, one skilled in the art would be able by routine experimentation to determine the ability of these compounds to prevent angiogenesis (e.g. Guba, M.,et al., 2002, ). One of skill in the art would be able by routine experimentation to determine the utility of these compounds in stents (e.g. Morice, M. C, et al., 2002). Additionally, one of skill in the art would be able by routine experimentation to determine the neuroregenerative ability of these compounds (e.g. Myckatyn, T. M., et al., 2002, Steiner ef a/. 1997)
The compounds of formula (I) are also, or in particular, expected to be useful as a therapeutic or prophylactic agents for one or more of the following conditions: rejection reactions after transplantation of organs or tissues (for example heart, kidney, liver, bone marrow, skin, cornea, lung, pancreas, small intestine, limb, muscle, nerve, intervertebral disc, trachea, myoblast and cartilage); graft-versus-host reactions following bone marrow
transplantation; autoimmune diseases (for example rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes); infections caused by pathogenic microorganisms, in particular fungal infections; inflammatory or hyperproliferative skin diseases or cutaneous manifestations of immunologically-mediated diseases (for example psoriasis, atopic dermatitis, contact dermatitis, eczematoid dermatitis, pyoderma gangrenosum, seborrhoeic dermatitis, lichen planus, pemphigus, bullous
pemphigoid, epidermolysis bullosa, rosacea, urticaria, angioedema, vasculitides, erythema, dermal eosinophilia, lupus erythematosus, acne, Netherton syndrome, and alopecia areata); autoimmune or allergic diseases of the eye (for example keratoconjunctivitis, vernal
conjunctivitis, allergic conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical keratitis, corneal epithelial dystrophy, keratoleukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, iridocyclitis, sarcoidosis affecting the eye, endocrine ophthalmopathy); reversible obstructive airway diseases or asthma, in particular chronic or inveterate asthma (for example late asthma, airway hyperresponsiveness, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, and dust asthma), mucosal or vascular inflammations (for example gastric ulcers, ischaemic or thrombotic vascular injury, ischaemic bowel diseases, enteritis, necrotizing enterocolitis, intestinal damage associated with thermal burns, leukotriene B4-mediated diseases); intestinal inflammations or allergies (for example coeliac disease, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease and ulcerative colitis); food-related allergic diseases with symptomatic manifestation remote from the gastrointestinal tract (for example migraine, rhinitis and eczema); renal diseases (for example interstitial nephritis, Goodpasture's syndrome, haemolytic uraemic syndrome, nephrotic syndrome (for example glomerulonephritis) and diabetic nephropathy); nervous system diseases (for example multiple myositis, Guillain-Barre syndrome, Meniere's disease, multiple neuritis, solitary neuritis, cerebral infarction, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and radiculopathy); ischaemic diseases (for example head injury, brain haemorrhage, cerebral thrombosis, cerebral embolism, cardiac arrest, stroke, transient ischemic attack, hypertensive encephalopathy, cerebral infarction); endocrine diseases (for example hyperthyroidism and Basedow's disease); haematic diseases (for example pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anaemia, agranulocytosis, pernicious anaemia, megaloblastic anaemia, and anerythroplasia); bone diseases (for example osteoporosis); respiratory diseases (for example sarcoidosis affecting the respiratory tract, pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), Bronchiolitis Obliterans Syndrome (BOS) and idiopathic interstitial pneumonia); skin diseases (for example dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photosensitivity, and cutaneous T-cell lymphoma); circulatory diseases (for example arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, and myocardosis); collagen diseases (for example scleroderma, Wegener's granulomatosis, and Sjogren's syndrome); adiposis; eosinophilic fasciitis; periodontal diseases (for example damage to gingiva, periodontium, alveolar bone or substantia ossea dentis); male pattern alopecia, alopecia senile; muscular dystrophy; pyoderma and Sezary syndrome; chromosome
abnormality-associated diseases (for example Down's syndrome); Addison's disease; Human Immunodeficiency Virus (HIV) infection or AIDS; hypertrophic cicatrix and keloid due to trauma, burn, or surgery.
The aforementioned compounds of the invention or a formulation thereof may be administered by any conventional method including topically (for example by inhalation, vaginally, intranasally, or by eye or ear drop), enterally (for example orally or rectally) or parenterally (for example by intravenous, intracavernosal, subcutaneous, intramuscular, intracardiac or
intraperitoneal injection) or via a medical device (for example via a stent). The treatment may consist of a single dose or a plurality of doses over a period of time.
Whilst it is possible for a compound of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more physiologically acceptable diluents or carriers. The diluents or carrier(s) must be "physiologically acceptable" in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof. In some cases, the diluent or carrier will be water or saline which will be sterile and pyrogen free.
The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient (compound of the invention) with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformLy and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the
compounds of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
A tablet may be made by compression or moulding, 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 a powder or granules, optionally mixed with a binder (eg povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (eg sodium starch glycolate, cross-linked povidone, cross-linked sodium
carboxymethyl cellulose), surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example,
hydroxypropylmethylcellulose in varying proportions to provide desired release profile.
Formulations in accordance with the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a
predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
Aerosol formulations suitable for administering via inhalation can also be made using methods known in the art. Examples of this include administration of the compounds of the invention by inhalation in the form of a powder (e.g. micronized) or in the form of atomized solutions or suspensions. The aerosol formulation may be placed in a suitable pressurized propellant, and may be used with additional equipment such as nebulizer or inhaler.
For applications to external tissues, for example the mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active agent may be employed with either a paraffinic or a water-miscible ointment base.
Alternatively, the active agent may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
The compounds of the invention may also be administered using medical devices known in the art. For example, in one embodiment, a pharmaceutical composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. 5,399,163; U.S. 5,383,851 ; U.S. 5,312,335; U.S. 5,064,413; U.S. 4,941 ,880; U.S. 4,790,824; or U.S. 4,596,556. Examples of well-known implants and modules useful in the present invention include : US 4,487,603, which discloses an implantable micro- infusion pump for dispensing medication at a controlled rate; US 4,486,194, which discloses a therapeutic device for administering medicaments through the skin; US 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; US 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; US 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and US 4,475,196, which discloses an osmotic drug delivery system. Many other such implants, delivery systems, and modules are known to those skilled in the art.
The compounds can be administered as the sole active agent, or in combination with other pharmaceutical agents, such as other agents that stimulate or inhibit cell proliferation of immune responses. These agents include e.g. cyclosporine, rapamycin, FK506, leflunomide, butenamides, corticosteroids, Doxorubicin, and the like. In such combinations, each active ingredient can be administered either in accordance with its usual dosage range, or at a lower dose level.
It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
Pharmaceutical compositions of the invention may optionally contain further active ingredients.
Further aspects of the invention include:
-A hybrid PKS containing starter and extension modules for producing FK506 derivatives in which one or more of the gene products responsible for provision of the C21 extender unit (based on the structure of FK506) is absent or inactive;
-An engineered FK506 producing strain containing such a hybrid PKS ;
-An engineered non- FK506 producing strain containing such a hybrid PKS;
-Such a strain in which the ccr gene product responsible for carboxylation of the C21 extender unit is not absent or inactivated;
-An engineered non-FK506 producing strain containing a functional PKS which strain comprises one or more genes responsible for provision of the C21 extender unit (based on the structure of FK506) from the FK506 biosynthetic cluster;
-Such an engineered non-FK506 producing strain wherein the functional PKS is a hybrid PKS including the AT from module 7 of the FK506 biosynthetic cluster (this module which incorporates an allyl extender unit may, for example, be included as part of a module swap);
-Such an engineered strain which strain is further engineered so as not to produce the natural starter unit, for example which strain is further engineered so that the gene fkbO is
deleted or inactivated;
Such an engineered strain which strain is further engineered so that the natural loading module is replaced by the loading module from the avermectin or an avermectin like PKS.
-An engineered non- FK506 producing strain containing an FK506 biosynthetic cluster engineered according to the invention, for example such a strain in which one or more C21
extender unit genes have been deleted or inactivated and optionally one or more starter unit
genes have been deleted or inactivated and optionally pipecolic acid biosynthesis genes have been deleted or inactivated and/or a strain in which one or more post PKS modification genes have been deleted or inactivated.
-A process for producing a polyketide which comprises culturing such an engineered strain in the presence of a non-natural extender unit (and if appropriate a non-natural starter unit) and optionally isolating said polyketide.
Materials and Methods
Materials
Bacterial strains and growth conditions
Escherichia coli DH10B (GibcoBRL) and E. coli JM1 10 (New England Biolabs) were grown in 2xTY medium as described by Sambrook et al. (2001 ). E. coli ET12567(pUZ8002) was grown as described by Paget et al. (1999) in 2xTY medium with kanamycin (25 mg/L) and chloramphenicol (12.5 mg/L). E. coli VCS257 was used for transfection of in vitro packaged cosmids. According to the instructions of Stratagene's Gigapack® III XL Packaging Extract the strain was kept on LB medium and grown on LB plus 0.2% maltose and 1OmM MgSO4 for transfection. E. coli transformants were selected for with ampicillin (100 mg/L), kanamycin (50 mg/L), apramycin (50 mg/L).
The avermectin producer Streptomyces avermitilis (DSM41443J was grown on TSB at 28 0C for genomic DNA isolation.
The FK506 producer Streptomyces tsukubaensis no. 9993 (FERM BP-927) (International Patent Organism Depositary, Tsukuba, Japan) and its derivatives were maintained on medium 1 agar plates or ISP4, ISP3, or ISP2 (see below) at 28 0C.
The FK520 producer, Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822, purchased from DSMZ, Braunschweig, Germany) (known to be equivalent to Streptomyces hygroscopicus subsp. ascomyceticus ATCC 14891 ) and its derivatives were maintained on medium 1 agar plates, ISP2, ISP3 or ISP4 (see below) at 28 0C.
Production of FK520 was carried out by fermentation of Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822), also termed BIOT-4081. Streptomyces tsukubaensis no. 9993 (FERM BP-927), also termed BIOT-31 19 was used for producing FK506. Single spore isolates of both strains, termed BIOT-4168 (containing the genes for FK520 biosynthesis) and BIOT-4206 (containing the genes for FK506 biosynthesis), were used for strain construction.
Strains were grown on MAM, ISP4, ISP3 or ISP2 agar at 28 0C for 5 - 21 days and used to inoculate seed medium NGY. The inoculated seed medium was incubated with shaking between 200 and 300 rpm at 5.0 or 2.5 cm throw at 28 0C for 48 h. For production of FK520 or FK506 the fermentation medium PYDG or PYDG+MES buffer (PYDM) were inoculated with 2.5%-10% of the seed culture and incubated with shaking between 200 and 300 rpm with a 5 or 2.5 cm throw at 28 0C for six days. The culture was then harvested for extraction.
Production of FK520, FK506 or analogues in Tubes
Spore stocks of BIOT-4081 , BIOT-4168, BIOT-3119, BIOT-4206 or strains which are described below were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 0C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 and incubated for 5-21 days at 28 0C. Vegetative cultures (seed culture) were prepared by removing one agar plug (6 mm in diameter) from the MAM, ISP4, ISP3 or ISP2 plate and transferring into 7 ml. medium NGY in 50 ml. polypropylene centrifuge tubes (cat no. 227261 , purchased from Greiner Bio-One Ltd, Stonehouse, Gloucestershire, UK) with foam plugs, or in Erlenmeyer flasks as described below. The culture tubes were incubated at 28 0C, 300 rpm, 2.5 cm throw for 48 h. From the seed culture 0.5 ml. were transferred into 7 ml. production medium PYDG or PYDG+MES in 50 ml. centrifuge tubes with foam plugs. Cultivation was carried out for 6 days at 28 0C and 300 rpm (2.5 cm throw). When necessary a selected precursor was fed to the production medium 24 h post inoculation. The feed compound was dissolved in 0.05 - 0.1 ml. methanol and added to the culture to give a final concentration of 2.12 mM of the feed compound.
Production of FK520, FK506 or analogues in Flasks
Spore stocks of BIOT-4081 , BIOT-4168, BIOT-3119, BIOT-4206 or strains which are described below were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 0C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 and incubated for 5 - 21 days at 28 0C. Vegetative cultures (seed culture) were prepared by removing 4 - 10 agar plugs (6 mm in diameter) from the MAM, ISP4, ISP3 or ISP2 plate and inoculating into 50 - 250 ml. medium NGY in 250 ml. or 2000 ml. Erlenmeyer flasks with foam plugs. The seed flasks were incubated at 28 0C, 200 - 250 rpm (5 or 2.5 cm throw) for 48 h. From the seed culture 2 - 10% (v/v) was transferred into 50 or 250 ml. production medium PYDG (or PYDG + MES) in 250 ml. or 2000 ml. Erlenmeyer flasks respectively with foam plugs. Cultivation was carried out for 6 days at 28 0C and 200 - 250 rpm (5 or 2.5 cm throw). If necessary a selected precursor was fed to the production medium 24 h post inoculation. The feed compound was dissolved in 0.375 - 1 ml_ methanol and added to the culture to give final concentration of 2.12 mM of the feed compound.
Production of FK520, FK506 or analogues in stirred bioreactors
Spore stocks of BIOT-4081 , BIOT-4168, BIOT-3119, BIOT-4206 or strains which are described below were prepared after growth on MAM, ISP4, ISP3 or ISP2 agar medium, and preserved in 20% (w/v) glycerol and stored at -80 0C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 medium and incubated for 5-21 days at 28 0C.
Vegetative cultures (seed culture) were prepared by removing 5-10 agar plugs (6 mm in diameter) from the MAM, ISP4, ISP3 or ISP2 plate and inoculation of 200 - 350 ml. medium NGY in 2 L Erlenmeyer flasks with foam plug. Cultivation was carried out for 48 h at 28 0C, 250 rpm (2.5 cm throw). The entire seed culture in one flask was transferred into 5 L PYDG containing 0.01-0.05% antifoam SAG 471 , in 7 L Applikon Fermentor. The fermentation medium was pre-adjusted at pH 6.0-7.0 post-sterilization. The fermentation was carried out for 6 days at 28 0C, with starting agitation set at 300-450 rpm, aeration rate at 0.5-0.8 v/v/m and dissolved oxygen (DO) level controlled with the agitation cascade at 20 - 40% air saturation. If required the pH may be maintained using acid or base addition on demand. For production of analogues of FK520 or FK506, the selected feed (providing the starter unit for biosynthesis of target compound) was fed to the production medium 12 - 24 h post inoculation. The feed compound was dissolved in 3 - 5 ml. methanol and added to the culture to give final concentration of 2 mM of the feed compound, the amount of methanol not exceeding 1 % of the total volume. Fermentation was continued for further five days post-feeding.
Media Recipes
Water used for preparing media was prepared using Millipore ENx Analytical Grade Water Purification System.
Medium 1: Modified A-medium (MAM)
Component Source per L
Wheat starch Sigma 10 g
Corn steep powder Sigma 2.5 g
Yeast extract Difco 3 g
Calcium carbonate Sigma 3 g
Iron sulphate Sigma 0.3 g
BACTO agar Difco 2O g No pH adjustment is made. Sterilised by autoclaving 121 0C, 15 min.
Medium 9: R6 (Kieser et al., 2000)
Component Source per L
Sucrose Fisher 20O g
Dextrin Avedex 1 O g
Casamino acids Difco i g
MgSO4.7H2O Sigma 0.05 g
ISP Trace Element See below 1 ml_
Solution
K2SO4 Sigma 0.1 g
Water to 70O mL
Add 20 g BACTO agar and a stirrer bar to each 1 L Duran and
autoclave at 121 0C for 15 min.
After autoclaving add:
Component Source per L
(previously sterilised individually
by autoclaving 121 0C, 15 min)
0.65 M L-glutamic acid, mono Sigma 100 ml_ (10.99 g)
sodium salt
0.48 M CaCI2.2H2O Sigma 100 ml_ (7.06 g)
0.1 M MOPS pH 7.2 Fisher 100 ml_ (2.09 g)
ISP3
Component Source per L
Oatmeal Tesco 2O g
BACTO agar Difco 18 g
ISP Trace Element See 1 ml.
Solution below
Oatmeal is cooked/steamed in the water for 20 min, strained through a muslin and more water added to replace lost volume. ISP Trace Elements Solution is added and pH adjusted to 7.2 with NaOH. Agar is added before autoclaving at 121 0C, 15 min.
/SP Trace Elements Solution
Component Source per L
FeSO4JH2O Sigma 0.1 g
MnCI2.4H2O Sigma 0.1 g
ZnSO4JH2O Sigma 0.1 g Distilled water 100
mL
Stored in the dark at■ 40C. 2xTY
Component Source per L
Tryptone Difco 16g
Yeast extract Difco 1Og
NaCI Sigma 5g
BACTO agar Difco 15g
Sterilised by autoclaving 1210C, 15 min. LB
Component Source per L
Tryptone Difco 1Og
Yeast extract Difco 5g
NaCI Sigma 1Og
BACTO agar Difco 15g
Sterilised by autoclaving 1210C, 15 min.
TSB
Component Source per L
Bacto Tryptic Soy BD 3Og
Broth
Sterilised by autoclaving 1210C, 15 min. NGY
Component Source per L
Difco Nutrient Broth Difco 8g
Glucose Sigma 10g
Yeast Extract Difco 5g
The medium is adjusted to pH 7.0, with NaOH and then sterilised by autoclaving 1210C, 15 min.
PYDG
Component Source per L
Peptone from Milk Sigma 15g
Solids Yeast Extract Difco 1.5 g
Dextrin Avedex 45 g
Glucose Sigma 5 g
The medium is adjusted to pH 7.0 with NaOH, and then sterilised by autoclaving 121 0C, 15 min. When MES is added to PYDG (PYDG + MES) it is added 21.2 g/L prior to pH adjustment.
ISP4
Component Source per L
Soluble Starch BDH 1 O g
K2HPO4 Sigma 1.O g
MgSO4.7H2O Sigma 1.O g
NaCI RDH 1.O g
(NH4)2SO4 RDH 2.O g
CaCO3 Caltec 2.O g
BACTO agar Difco 2O g
ISP Trace Elements See 1 ml.
Solution above
A paste is made using a little cold water and the starch. This is brought up to a volume of 500 ml_. All other ingredients are then added, and the pH of the media is adjusted to pH 7.0 - 7.4. Sterilise by autoclaving 121 0C, 15 min.
ISP2
Component Source per L
Yeast extract Difco 4 g
Malt extract Difco 10 g
Glucose Sigma 4 g
Bacto agar Difco 2O g
Sterilise by autoclaving 121 0C, 15 min.
DNA manipulation and sequencing
DNA manipulations and electroporation procedures were carried out as described in Sambrook et al. (2001 ). PCR was performed according to the instructions of the KOD Polymerase kit (Novagen). DNA sequencing was performed as described previously (Gaisser et al., 2000). Genome sequencing was carried out using 454 technology (Margulies et al., 2005) at Cogenics and the University of Cambridge. Genomic DNA preparation
Strains were grown in shake flasks containing 25 mL TSB or ISP2 medium at 250 - 300 rpm and 28 0C and harvested after 2 to 3 days. Cell pellets were washed with 10.3% sucrose and frozen at -20 0C until used. The following method was most successful for genomic DNA isolation from S. hygroscopicus, S. tsukubaensis and S. avermitilis. A pellet originating from 12.5 mL of culture was resuspended in 1 mL STE buffer (100 mM NaCI, 10 mM Tris HCI pH8, 1 mM EDTA). 20 mL STE buffer supplemented with 2 mg/mL lysozyme were added and the resuspension incubated for 30 min at 37 0C. 20 μL of RNaseA (10 mg/mL) were added and the mixture incubated for another 30 min at 37 0C. 4.8 mL EDTA (0.1 M final concentration) were added to stop the reaction. 1.4 mL 20% SDS were added. After careful mixing the lysate was incubated on ice for 5 min, then extracted with one volume of phenol/chloroform/isoamylalcohol (25:24:1 ) and centrifuged at 2300 g and 4 0C for at least 15 min up to 1 h. Extractions were repeated until no more protein was visible at the interface, followed by a final chloroform/isoamylalcohol (49:1 ) extraction. The upper phase was precipitated with 1/10 vol. 5 M NaCI and 1 vol. cold isopropanol. After a few min, the DNA was spooled out with a glass rod and washed in ice cold 70% EtOH. After brief drying, the recovered DNA was dissolved in 0.5 - 1 mL TE 10:1. The proteinase K method (Kieser et al., 2000) was also applied successfully to recover genomic DNA from S. tsukubaensis.
Cosmid library preparation for S. tsukubaensis
A cosmid library of genomic DNA of S. tsukubaensis, was constructed. High molecular weight DNA from several genomic DNA preps was partially digested with BfuC\, an isoschizomer of Sau3A, to a mean size of 30 - 60 kb, ligated to Supercos-1 , packaged into A phage using Gigapack® III XL Packaging Extract (Stratagene) and transfected into Escherichia coli VCS257. The titre was 6.7 x 105 cfu / μg vector. DNA of 10 cosmids was isolated and digested with EcoRI to check the insert size which was 40 kb on average. 2000 clones were grown in 96-well microtitre plates (150 μL LB Ampl OO Kan50 per well) at 37 0C and frozen at -80 0C after mixing wells with 50μL LB/glycerol 1 :1.
Colony hybridization
Thawn microplate-cultures were stamped onto positively charged filter membranes (Roche) which had been placed on LB AmplOO Kan50 plates. After overnight growth at 37 0C membranes were taken off. Cells were lysed and cell debris removed according to the DIG Application Manual for Filter Hybridization (Roche). DNA was crosslinked by exposing membranes to UV (302nm) for 5 min. Membranes were kept between two sheets of filter paper soaked with 2xSSC at 4 0C or used immediately for hybridization. Hybridization was carried out using standard hybridization buffer and DIG labeled fkbO probe (see above) at a hybridization temperature of 68 0C. Stringent washes were performed at 68 0C. The nonradioactive DIG Nucleic Acid Detection kit from Roche was used to identify 5 positive clones on 4 library plates. The procedure followed the instructions of the DIG Application Manual for Filter Hybridization (Roche).
Transformation of Streptomyces tsukubaensis by conjugation
Escherichia coli ET12567 (pUZ8002) (Macneil et al., 1992, Paget et al., 1999) was transformed with pKC1 139B01 -derived plasmids by electroporation to generate the E. coli donor strains for spore conjugation (Kieser et al., 2000). Fresh spores were harvested in water from plates of Streptomyces tsukubaensis (BIOT-4206 or Biot-31 19). Spore suspensions were heat-shocked at 50 0C for 10 min. They were then mixed with the E. coli donor strain, which had been washed twice with 2xTY, in a ratio of 3:1 Streptomycete to E. coli, and the mixture shaken at 37 0C, 300 rpm, 2.5 cm throw for 1.5 - 2 h. The conjugation mixture was then plated on R6 medium and incubated at 37 0C. After -20 h, the plates were overlaid with 2xTY containing apramycin sulphate and nalidixic acid and incubation continued at 37 0C. Plasmids with pKC1139B01 backbone are not able to self-replicate in Streptomycetes at 37 0C and are forced to integrate into the genome. For more details on the screening procedure for conjugants see below.
Culture broth sample extraction and analysis
Culture broth (0.9 ml.) were extracted with ethyl acetate (0.9 ml.) in a 2 ml. Eppendorf tube. The broth was mixed with the solvent for 15 min on a shaking platform (vibrax) at 400 rpm. The phases are then separated by centrifugation (2 min, 13,200 rpm). An aliquot of the organic layer (0.1 ml.) is then transferred to either a clean glass LC-vial or a vial containing 5 μg of pimecrolimus (as an internal standard for quantification). The solvent is removed in vacuo (3 min) and then re-dissolved in methanol (1 ml.) by gentle agitation on a shaking platform (5 min).
Analysis by LCMS
The HPLC system comprised an Agilent HP1100 equipped with a Hyperclone ODS2, C18, 3 micron 4.6 x 150 mm column (Phenomonex). Injection volume 10 μl_, oven 500C, A: 0.1 % formic acid, B: 0.1% formic acid in MeCN. 1 mL/min; 0-1 min 65% B; 6.5 min 100% B; 10 min 100% B; 10.05 min 65% B, 12 min 65% B. The HPLC system described above was coupled to a Bruker Daltonics Esquire3000 electrospray mass spectrometer. Positive-negative switching was used over a scan range of 500 to 1000 Dalton. Alternatively, LC samples that have been spiked with 0.005 mg/mL pimecrolimus were analysed on the same instrument and with the same chromatographic conditions. However the MS was conducted in multiple reaction monitoring mode (MRM mode) in order to quantify the amount of FK analog in the sample. Details of the quantification are: negative scan mode, m/z = 450-850
MRM setup:
transitions [Da] fragmentation amplitude [V] pimecrolimus (IS): 808.4→ 548.3 1.15
FK520 790.5→ 548.3 1.15
FK506 802.4→ 560.3 1.15 all parent ions are isolated with a width of 3 amu.
All FK520 and FK506 analogues can be quantified in this manner, with the parent ion isolated as [M-H]" and the transition to 548.2 (for Fk520 analogues) or 560.2 (for FK506 analogues) used.
The amount of analyte present is then calculated by dividing the integral for the analyte transition (as detailed above) with that for the internal standard, pimecrolmius. This ratio is then compared with a standard calibration curve for FK520 or FK506 up to 100 ng on column with 50 ng on column pimecrolimus.
NMR spectra (1H, 13C, DQF-COSY, TOCSY, HMQC, HMBC, NOESY) of purified material were recorded on a Bruker Advance DRX500 spectrometer which operated at 500 MHz (for proton derived spectra, pro rata for other nuclei) at 27 0C,. Chemical shifts are described in parts per million (ppm) and are referenced to solvent signal e.g. CHCI3 at δH 7.26 (1H) and CHCI3 at δc 77.0 (13C). J values are given in Hertz (Hz).
Example 1 - Sequencing of the FK506 biosynthetic cluster
Genomic DNA was isolated from S. hygroscopicus subsp. hygroscopicus (DSM 40822 assigned BIOT-4081 ) and Streptomyces tsukubaensis (FermBP927 assigned BIOT-3119) using standard protocols described in Kieser et al., (2000). DNA sequencing of cosmids was carried out by the sequencing facility of the Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge CB2 1 QW using standard procedures. A draft genome sequence was obtained using 454 technology by the sequencing facility of the Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge CB2 1 QW. The initial draft genome was generated using a whole 454 chip and this data was subsequently improved by a further Vi chip and assembly of contigs carried out using PHredPHrap.
A cosmid library was prepared using isolated genomic DNA of the FK506-producing strain Streptomyces tsukubaensis (BIOT-31 19) using standard methods. The genomic DNA was partially digested with BfuC\, dephosphorylated and ligated to supercos 1 cut with Xba\, dephosporylated with Shrimp Alkaline Phosphatase (Roche) and subsequently digested with BamYW. This was packaged into λ phage used to transfect E. coli VCS257 and a library of 2000 clones generated.
Probes were made as decribed below for screening Streptomyces tsukubaensis (BIOT- 3119) to identify the FK506 cluster. Plasmid pAES2#2 was used as PCR template. It was obtained by cloning a 1.9kb PCR amplified fkbO fkbP fragment from Streptomyces
tsukubaensis (BIOT-3119) into pUC19. This fragment had been recruited from BIOT-3119 genomic DNA using primers derived from FK520 fkbO and fkbP sequences.
AES09 5'- CCATGCATCCGACGCCGTTTCGGCGCTCTATACGCGGA-S' (SEQ I D NO: 1 )
AES24 5'- CCAGATCTGAGGTCGAAGCGGGTGAACCAGCGGCCGGT-S' (SEQ I D NO: 2)
Primers for probe construction were the following: UES2For and UES2Rev were used to amplify a region within FK506 fkbO and UES1 For and UES1 Rev were used to amplify a region within FK506 fkbP to generate probes using DIG-labelled dNTPs.
UES2For S'-CACTCCTTCGATCTCCACGAGCAGGTCGCCACGGGC-S' (SEQ ID NO: 3) UES2Rev S'-ACCCTGCCGTCCTCACGGCACACCACTACCCCACGG-S' (SEQ ID NO: 4) UESI For 5'-ACAGGCGATGTGGTGCACGACCACCGCCAGCACGTG-S' (SEQ I D NO: 5) UESI Rev 5'-GGTTTCTGCAGGAACTGGATCCGGACAG-S' (SEQ I D NO: 6)
Southern blot experiments were carried out using the DIG Reagents and Kits for Non- Radioactive Nucleic Acid Labelling and Detection according to the manufacturers' instructions (Roche). The DIG-labelled FK506-fkbO DNA fragment was used as a homologous probe to screen 400 clones of the cosmid library of the FK506 producing Streptomyces tsukubaensis (BIOT-3119) using the DIG Reagents and Kits for Non-Radioactive Nucleic Acid Labelling and Detection according to the manufacturers' instructions for colony hybridisation (Roche). 5 positive cosmids were identified from 400 screened clones using the fkbO-probe and a hybridisation temperature of 68°C (a second screen using a hybridisation temperature of 77°C confirmed the same positive clones). The positive clones were then probed with the fkbP probe using a hybridisation temperature of 77°C and again were positive. The 5 cosmids were isolated, digested with EcoR\ and sent for end sequencing with T7 and T3 commercially available primers. Of these, cosmid G9 was selected and fully sequenced by the sequencing facility of the Biochemistry Department, University of Cambridge. The sequence of G9 covers the regions from 19180 to 64312 bp in the full sequence (SEQ ID No: 7) see table 1.
In order to identify a cosmid containing the flanking genes an fkbL probe was generated. A DIG-labelled probe of a region within fkbL was amplified using genomic DNA from the FK506 producing BIOT-31 19. Colony hybridisation of 800 clones lead to the identification of 4 potentially f/ob/.-containing cosmids and these were end-sequenced. Of these, cosmid 4D7 was selected for complete sequencing and covers the regions from 63048 to 109611 bp in the full sequence (SEQ ID No: 7) see table 1. Cosmid 4D7 has an insert of ~46kb and overlaps with the G9 sequence by ca. 2kb. Cosmid 4D7 contains the genes for biosynthesis of the allyl malonate extension unit and following a region of low homology to data there is a siderophore cluster.
Concurrently a draft genome sequence was being prepared as described above. It was apparent in the first draft that the PKS modules had collapsed. However one contig was identified covering the flanking genes fkbM, fkbD, fkbN and fkbQ (genes are names
analogously to the FK520 cluster when homologous genes are present) followed by an ABC transporter. This contig #2476 is 9612 bp in length and covers the region 1 to 9612 bp in the full sequence (SEQ ID No: 7) see table 1. There remained an -10 kbp gap within fkbA. This was closed by a combination of sequencing cosmid D5 which had been identified by probing with a region within the fkbO gene, and by incorporating the cosmid sequences into the draft genome in order to decollapse the PKS modules. Sequencing of cosmid D5 was achieved by subcloning and primer walking.
Incorporation of the cosmid sequences into the draft genome was successful but breaking them up into 5 kbp fragments and performing an assembly with these fragments. This lead to the complete FK506 cluster sequence as provided in the sequence listing SEQ ID NO: 7
Table 1 : Regions of cluster sequence covered by contigs and cosmids
Figure imgf000035_0001
Table 2: Open reading frame predictions and gene assignments for FK506 cluster
Figure imgf000035_0002
Figure imgf000036_0001
Note: fkbC and all genes downstream of fkbG were assigned based on glimmer predictions of ORFs with BLASTX / BLASTP predictions of functions.
Note 2: downstream of ORF9 are a further set of genes which may be involved in regulation and transport
References
Armstrong, V.W. and Oellerich, M. (2001 ) New developments in the immunosuppressive drug monitoring of cyclosporine, tacrolimus and azathioprine. Clinical Biochemistry 34(1 ): 9- 16
Aparicio, J. F., Molnar, I., Schwecke, T., Konig, A., Haydock, S. F., Khaw, L. E., Staunton, J., and
Leadlay, P. F. (1996) Organization of the biosynthetic gene cluster for rapamycin in
Streptomyces hygroscopicus: analysis of the enzymatic domains in the modular polyketide synthase. Gene 169: 9-16.
Baker, H., Sidorowicz, A., Sehgal, S. N., and Vezina, C. (1978) Rapamycin (AY-22,989), a new antifungal antibiotic. III. In vitro and in vivo evaluation. Journal of Antibiotics 31 : 539-
545.
Bierman, M., Logan, R., O'Brien, K., Seno, E. T., Nagaraja Rao, R., and Schoner, B. E. (1992)
Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to
Streptomyces spp. Gene 116: 43-49.
Blanc, V., Lagneaux, D., Didier, P., Gil, P., Lacroix, P., and Crouzet, J. (1995) Cloning and analysis of structural genes from Streptomyces pristinaespiralis encoding enzymes involved in the conversion of pristinamycin 116 to pristinamycin IIA (PI IA): PIU synthase and NAD H: riboflavin 5'-phosphate oxidoreductase. Journal of Bacteriology 177: 5206-
5214.
Blanc, V., Gil, P., Bamas-Jacques, N., Lorenzon, S., Zagorec, M., Schleuniger, J., Bisch, D.,
Blanche, F., Debussche, L., Crouzet, J., and Thibaut, D. (1997) Identification and analysis of genes from Streptomyces pristinaespiralis encoding enzymes involved in the biosynthesis of the 4-dimethylamino-L-phenylalanine precursor of pristinamycin I.
Molecular Microbiology 23 : 191 -202.
Cannell, R. J. P, Ed., (1998) "Natural Products Isolation", Methods in Biotechnology, 4, Humana Press.
Cao, W., Mohacsi, P., Shorthouse, R., Pratt, R. and Morris, R. E. (1995). Effects of rapamycin on growth factor-stimulated vascular smooth muscle cell DNA synthesis. Inhibition of basic fibroblast growth factor and platelet-derived growth factor action and antagonism of rapamycin by FK506. Transplantation 59(3): 390-395.
Carlson, R. P., Hartman, D.A., Tomchek, L.A., Walter, T.L., Lugay, J. R., Calhoun, W., Sehgal, S. N., Chang, J.Y. (1993). Rapamycin, a potential disease-modifying antiarthritic drug. J. Pharmacol. Exp. Ther. 266(2): 1 125-38.
Carter, G.T., Nietsche, J.A., Hertz, M. R., Williams, D. R., Siegel, M. M., Morton, G.O., James, J. C, Borders, D. B. (1988). LL-F28249 antibiotic complex: a new family of antiparasitic macrocyclic lactones. Isolation, characterization and structures of LL-F28249 alpha, beta, gamma, lambda. J. Antibiot. 41(4): 519-529.
Chambraud, B., Radanyi, C, Camonis, J. H., Shazand, K., Rajkowski, K., and Baulieu, E. E.
(1996) FAP48, a new protein that forms specific complexes both immunophilins FKBP59 and FKBP12. Prevention by the immunosuppressant drugs FK506 and rapamycin. Journal of Biological Chemistry 271 : 32923-32929.
Chang, J.Y., Sehgal, S. N., and Bansbach, CC. (1991 ) FK506 and rapamycin: novel
pharmacological probes of the immune response. Trends in Pharmacological Sciences
12: 218-223.
Chen, J., Zheng, X.F., Brown, EJ. , and Schreiber, S. L. (1995) Identification of an 11-kDa
FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue. Proceedings of the National
Academy of Sciences of the United States of America 92: 4947-4951.
Choi, S-S., Hur, Y-A., Sherman, D. H., Kim, E-S. (2007) Isolation of the biosynthetic gene
cluster for tautomycetin, a linear polyketide T-cell specific immunomodulator from
Streptomyces sp. CK4412 Microbiology 153:1095-1 102
DiLeIIa, A.G., and Craig, RJ. (1991 ) Exon organization of the human FKBP-12 gene:
correlation with structural and functional protein domains. Biochemistry 30: 8512-8517. Du, L. C, Sanchez, C, Chen, M., Edwards, DJ., and Shen, B. (2000) The biosynthetic gene cluster for the antitumor drug bleomycin from Streptomyces verticillus ATCC 15003 supporting functional interactions between nonribosomal peptide synthetases and a polyketide synthase. Chemistry & Biology 7: 623-642.
Dudkin, L., Dilling, M. B., Cheshire, PJ., Harwood, F. C, Hollingshead, M., Arbuck, S. G., Travis,
R., Sausville, E.A., Houghton, PJ. (2001 ). Biochemical correlates of mTOR inhibition by the rapamycin ester CCI-779 and tumor growth inhibition. Clin. Cancer Res.
7(6): 1758-64
Dumont, F., Staruch, MJ., Koprak, S. L., Siekierka, Lin, C. S., Harrison, R., Sewell, T., Kindt,
V. M., Beattie, T. R., Wyvratt, M. and Sigal, N. H. (1992)The immunosuppressive and toxic effects of FK506 are mechanistically related: Pharmacology of a novel antagonist of FK506 and Rapamycin. Journal of Experimental Medicine, 176:751-760 (1992) Dutton, C. J., S. P. Gibson, et al. (1991 ). "Novel avermectins produced by mutational
biosynthesis." J. Antibiot. 44(3): 357-365.
Fang, J., Zhang, Y., Huang, L., Jia, X., Zhang, Q., Zhang, X., Tang, G. and Liu, W. (2008).
Cloning and characterisation of the tetrocarcin A gene cluster from Micromonospora chalcae NRRL 11289 reveals a highly conserved strategy for tetronate biosynthesis in spirotetronate antibiotics. J. Bact. 190(17): 6014-6025
Fehr, T., Sanglier, J-J., Schuler, W., Gschwind, L., Ponelle, M., Schilling, W., Wioland, C.
(1996). Antascomicinc A, B, C, D and E: Novel FKBP12 binding compounds from a
Micromonospora strain. J. Antibiot. 49(3): 230-233.
Findlay J.A, and Radics, L. (1980) Canadian Journal of Chemistry 58:579. Fishbein, T. M., Florman, S., Gondolesi, G., Schiano, T., LeLeiko, N., Tschernia, A., Kaufman,
S. (2002). Intestinal transplantation before and after the introduction of sirolimus.
Transplantation. 73(10): 1538-42.
Foey, A., Green, P., Foxwell, B., Feldmann, M., Brennan, F. (2002). Cytokine-stimulated T cells induce macrophage IL-10 production dependent on phosphatidylinositol 3-kinase and p70S6K: implications for rheumatoid arthritis. Arthritis Res. 4(1):64-70. Epub 2001
Oct 10.
Gaisser, S., Reather, J., Wirtz, G., Kellenberger, L., Staunton, J., and Leadlay, P. F. (2000) A defined system for hybrid macrolide biosynthesis in Saccharopolyspora erythraea.
Molecular Microbiology 36: 391-401.
Galat, A. (2000) Sequence diversification of the FK506-binding proteins in several
different genomes. European Journal of Biochemistry 267: 4945-4959.
Garrity, G. M., Heimbuch, B. K., Motamedi, H., Shafiee, A. (1993) Genetic relationships among
Actinomycetes that produce the immunosuppressant macrolides FK506, FK520/FK523 and rapamycin. Journal of Industrial Microbiology 12(1): 42-47
Gregory, C. R., Huie, P., Billingham, M. E. and Morris, R. E. (1993). Rapamycin inhibits arterial intimal thickening caused by both alloimmune and mechanical injury. Its effect on cellular, growth factor and cytokine response in injured vessels. Transplantation
55(6):1409-1418.
Guba, M., von Breitenbuch, P., Steinbauer, M., Koehl, G., Flegel, S., Hornung, M., Bruns, C. J.,
Zuelke, C, Farkas, S., Anthuber, M., Jauch, K. W., and Geissler, E. K. (2002) Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nature Medicine 8: 128-135.
Hamilton, G. S., and Steiner, J. P. (1998) Immunophilins: Beyond immunosuppression. Journal of Medicinal Chemistry 41 : 51 19-5143.
Hatanaka, H., Kino, T., Miyata, S., Inamura, N., Kuroda, A., Goto, T., Tanaka, H., Okuhara, M.
(1988). FR-900520 and FR-900523, novel immunosuppressants isolated from a
Streptomyces. II. Fermentation, isolation and physico-chemical and biological characteristics. J. Antibiot. (Tokyo). 41(11):1592-601.
Hatanaka H, Kino T, Asano M, Goto T, Tanaka H, Okuhara M. (1989). FK-506 related
compounds produced by Streptomyces tsukubaensis No. 9993. J. Antibiot. (Tokyo).
42(4): 620-2.
Hendrickson, B.A., Zhang, W., Craig, RJ. , Jin, YJ. , Bierer, B. E., Burakoff, S., and DiLeIIa, A.G.
(1993) Structural organization of the genes encoding human and murine FK506-binding protein (FKBP)13 and comparison to FKBP1. Gene 134: 271-275.
Hopwood, D.A. (1997) Genetic contributions to understanding polyketide synthases. Chemical
Reviews 97: 2465-2497. Hosted, T. J., and Baltz, R. H. (1997) Use of rpsL for dominance selection and gene replacement in Streptomyces roseosporus. Journal of Bacteriology 179: 180-186.
Hung, DT. , and Schreiber, S. L. (1992) cDNA cloning of a human 25 kDa FK506 and rapamycin binding protein. Biochemical and Biophysical Research Communications 184: 733-738. Hung, DT. , Jamison, T. F., and Schreiber, S. L. (1996) Understanding and controlling the cell cycle with natural products. Chemistry & Biology 3: 623-639.
Jain, S., Bicknell, G. R., Whiting, P. H., Nicholson, M. L. (2001 ). Rapamycin reduces expression of fibrosis-associated genes in an experimental model of renal ischaemia reperfusion injury. Transplant Proc. 33(1 -2):556-8.
Jin, Y.J., Burakoff, S. J., and Bierer, B. E. (1992) Molecular cloning of a 25-kDa high affinity rapamycin binding protein, FKBP25. Journal of Biological Chemistry 267: 10942-10945. Kahan, B. D., Chang, J.Y., and Sehgal, S.N. (1991 ) Preclinical evaluation of a new potent
immunosuppressive agent, rapamycin. Transplantation 52: 185-191.
Kahan, B. D., and Camardo, J. S. (2001 ) Rapamycin: Clinical results and future
opportunities. Transplantation 72:1181-1193.
Kapturczak, H. U., Meier-Kriesche, and Kaplan, B. (2004) Pharmacology of Calcineurin
Antagonists. Transplantation Proceedings 36(supp 2S): 25S-32S
Khaw, L. E., Bohm, G.A., Metcalfe, S., Staunton, J., and Leadlay, P. F. (1998) Mutational
biosynthesis of novel rapamycins by a strain of Streptomyces hygroscopicus NRRL
5491 disrupted in rapL, encoding a putative lysine cyclodeaminase. Journal of
Bacteriology 180: 809-814.
Kieser, T., Bibb, M.J., Buttner, M.J., Chater, K.F., and Hopwood, D.A. (2000) Practical
Streptomyces Genetics, John lnnes Foundation, Norwich.
Kirby, B., and Griffiths, C. E. M. (2001 ) Psoriasis: the future. British Journal of Dermatology
144:37-43.
Kirchner, G. I., Winkler, M., Mueller L., Vidal, C, Jacobsen, W., Franzke, A., Wagner, S., Blick,
S., Manns M. P., and Sewing K.-F.(2000) Pharmacokinetics of SDZ RAD and cyclosporin including their metabolites in seven kidney graft patients after the first dose of SDZ RAD. British Journal of Clinical Pharmacology 50:449-454.
Konig, A., Schwecke, T., Molnar, I., Bohm, G., Lowden, P.A.S., Staunton, J., and
Leadlay, P. F. (1997) The pipecolate-incorporating enzyme for the biosynthesis of the immunosuppressant rapamycin. Nucleotide sequence analysis, disruption and heterologus expression of rapP from Streptomyces hygroscopicus. European Journal of
Biochemistry 247: 526-534.
Konz, D. and M. A. Marahiel (1999). "How do peptide synthetases generate structural
diversity?" Chemistry & Biology 6(2): R39-R48.
Liu, Y., Hazzard, C, Eustaquio, A.S., Reynolds, K. and Moore B. S. (2009) Biosynthesis of Salinosporamide from α,β-Unsaturated Fatty Acids: Implications for Extending
Polyketide Synthase Diversity. J. Am. Chem. Soc.
Lowden, P.A.S., Bohm, G., Staunton, J., and Leadlay, P. F. (1996) The nature of the starter unit for the rapamycin polyketide sythase. Angewandte Chemie 35: 2249-2251.
Lowden, P. A. S., (1997) Ph.D. Dissertation, University of Cambridge. "Studies on the
biosynthesis of rapamycin".
Lowden, P.A.S., Wilkinson, B., Bohm, G.A., Handa, S., Floss, H. G., Leadlay, P. F., and
Staunton, J. (2001 ) Origin and true nature of the starter unit for the rapamycin polyketide synthase. Angewandte Chemie-lnternational Edition 40: 777-779.
Lyons, W.E., George, E. B., Dawson, T.M., Steiner, J. P., and Snyder, S. H. (1994)
Immunosuppressant FK506 promotes neurite outgrowth in cultures of PC12 cells and sensory ganglia. Proceedings of the National Academy of Sciences of the United States of America 91 :3191-3195.
MacNeil, DJ. , Gewain, K.M., Ruby, C. L., Dezeny, G., Gibbons, P. H., and MacNeil, T. (1992)
Analysis of Streptomyces avermitilis genes required for avermectin biosynthesis utilizing a novel integration vector. Gene 111 : 61-68.
Maes, B. D. and Vanrenterghem, Y. F. Ch. (2004) Cyclosporine: Advantages Versus
Disadvantages Vis-a-vis Tacrolimus. Transplantation Proceedings 36 (Suppl 2S): 40S-
49S
Marahiel, M.A., Stachelhaus, T., and Mootz, H. D. (1997) Modular peptide synthetases involved in nonribosomal peptide synthesis. Chemical Reviews 97: 2651-2673.
Margulies et al., (2005) Genome sequencing in microfabricated high-density picolitre reactors.
Nature 437: 376-380
March, J. Advanced Organic Chemistry - Reactions, Mechanisms and Structure. 4th Edition.
Wiley Interscience.
Meingassner, J. G., Grassberger, M., Fahrngruber, H., Moore, H. D., Schuurman, H. and Stutz.,
A. (1997) A novel anti-inflammatory drug, SDZ ASM 981 , for the topical and oral treatment of skin diseases: in vivo pharmacology. British Journal of Dermatology
137:568-576
Molnar, I., Aparicio, J. F., Haydock, S. F., Khaw, L. E., Schwecke, T., Konig, A., Staunton, J., and
Leadlay, P. F. (1996) Organisation of the biosynthetic gene cluster for rapamycin in
Streptomyces hygroscopicus: analysis of genes flanking the polyketide synthase. Gene
169: 1-7.
Morice, M. C, Serruys, P. W., Sousa, J. E., Fajadet, J., Ban Hayashi, E., Perin, M., Colombo, A.,
Schuler, G., Barragan, P., Guagliumi, G., Molnar, F., Falotico, R. (2002). RAVEL Study
Group. Randomized Study with the Sirolimus-Coated Bx Velocity Balloon-Expandable
Stent in the Treatment of Patients with de Novo Native Coronary Artery Lesions. A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization. N. Eng.l J. Med. 346(23): 1773-80.
Motamedi, H., Shafiee, A., Cai, SJ. , Streicher, S. L., Arison, B. H., and Miller, R.R. (1996)
Characterization of methyltransferase and hydroxylase genes involved in the
biosynthesis of the immunosuppressants FK506 and FK520. Journal of Bacteriology
178: 5243-5248.
Motamedi, H., Cai, S. J., Shafiee, A., and Elliston, K. O. (1997) Structural organization of a
multifunctional polyketide synthase involved in the biosynthesis of the macrolide immunosuppressant FK506. European Journal of Biochemistry 244: 74-80.
Motamedi, H., and Shafiee, A. (1998) The biosynthetic gene cluster for the macrolactone ring of the immunosuppressant FK506. European Journal of Biochemistry 256: 528-534.
Muramatsu, H., Mokhtar, S. I., Katsuoka, M. and Ezaki, M. (2005) Phylogenetic Analysis of
Immunosuppressant FK506-Producing Streptomycete Strains. Actinomycetologica
19:33-39
Myckatyn, T. M., Ellis, R.A., Grand, A.G., Sen, S. K., Lowe, J. B. 3rd, Hunter, D.A., Mackinnon,
S. E. (2002). The effects of rapamycin in murine peripheral nerve isografts and allografts. Plast. Reconstr. Surg. 109(7):2405-17.
Navia, M.A. (1996) Protein-drug complexes important for immunoregulation and organ
transplantation. Current Opinion in Structural Biology 6: 838-847.
Nghiem, P., Pearson, G., and Langley, R. G. (2002) Tacrolimus and pimecrolimus: From clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis. Journal of the
American Academy of Dermatology 46: 228-241
NCCLS Reference Method for Broth Dilution Antifungal Susceptibility Testing for Yeasts:
Approved Standard M27-A, vol. 17 No. 9. (1997).
Okazaki, T., Ono, M., Aoki, A., Fukuda, R. (1983) Milbemycins, a new family of macrolide
antibiotics: producing organism and its mutants. J. Antibiot. (1983) 36(4): 438-441 Oliynyk, M., Stark, C.B.W., Bhatt, A., Jones, M.A., Hughes-Thomas, Z.A., Wilkinson, C,
Oliynyk, Z., Demydchuk, Y., Staunton, J., Leadlay, P. F. (2003) Analysis of the biosynthetic gene cluster for the polyether antibiotic monensin in Streptomyces cinnamonensis and evidence for the role of monB and monC genes in oxidative cyclization MoI. Micro. 49(5): 1179-1 190
Paget, M.S. B., Chamberlin, L., Atrih, A., Foster, SJ. , and Buttner, MJ. (1999) Evidence that the extracytoplasmic function sigma factor σE is required for normal cell wall structure in
Streptomyces coel lcolor A3(2). Journal of Bacteriology 181 : 204-211 )
Paiva, N. L., Demain, A.L., and Roberts, M. F. (1991 ) Incorporation of acetate, propionate, and methionine into rapamycin By Streptomyces hygroscopicus. Journal of Natural Products
54: 167-177. Paiva, N. L., Demain, A.L., and Roberts, M. F. (1993) The immediate precursor of the nitrogen- containing ring of rapamycin is free pipecolic acid. Enzyme and Microbial Technology
15: 581-585.
Patterson, C. E., Schaub, T., Coleman, E. J., and Davies E. C. (2000) Developmental regulation of FKBP65. An ER-localized extracellular matrix binding-protein. Molecular Biology of the Cell 11 :3925-3935.
Pfeifer, B.A., Admiraal, SJ. , Gramajo, H., Cane, D. E., and Khosla, C. (2001 ) Biosynthesis of complex polyketides in a metabolically engineered strain of E. coli. Science 291 : 1790-
1792.
Powell, N., Till, S., Bungre, J., Corrigan, C. (2001 ). The immunomodulatory drugs cyclosporin
A, mycophenolate mofetil, and sirolimus (rapamycin) inhibit allergen-induced proliferation and IL-5 production by PBMCs from atopic asthmatic patients.
J. Allergy Clin. Immunol. 108(6):915-7
Rabinovitch, A., Suarez-Pinzon, W.L., Shapiro, A.M., Rajotte, R.V., Power, R. (2002).
Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. Diabetes. 51(3):638-45.
Rawlings, BJ. (2001 ) Type I polyketide biosynthesis in bacteria (Part A). Natural Product
Reports 18: 190-227.
Reitamo, S., Spuls, P., Sassolas, B., Lahfa, M., Claudy, A., Griffiths, C. E.; Sirolimus European
Psoriasis Study Group. (2001 ). Efficacy of sirolimus (rapamycin) administered concomitantly with a subtherapeutic dose of cyclosporin in the treatment of severe psoriasis: a randomized controlled trial. Br. J. Dermatol. 145(3):438-45.
Reynolds, K.A., and Demain, A.L. (1997) "Rapamycin, FK506 and Ascomycin-related
Compounds" pp497-520, Biotechnology of Antibiotics: Second Edition, Revised and
Expanded. W.R.Strohl, Ed.
Rosen, M. K., and Schreiber, S. L. (1992) Natural products as probes of cellular function: studies of immunophilins. Angewandte Chemie-lnternational Edition in English 31 : 384-400. Roy, J. N., Barama, A., Poirier, C, Vinet, B. Roger, M. (2006) Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients
Pharmacogenetics 16(9): 659-665
Roymans, D., and Siegers, H. (2001 ) Phosphaditidylinositol 3-kinases in tumor progression.
European Journal of Biochemistry 268:487-498.
Schwarzer, D., and Marahiel, M.A. (2001 ) Multimodular biocatalysts for natural product
assembly. Naturwissenschaften 88: 93-101.
Sambrook, J., Fritsch, E. F., and Maniatis, T. (1989) Molecular cloning: a laboratory manual,
2nd ed. Cold Spring Harbor Laboratory Press, N.Y.
Sambrook, J., and Russell, D.W. (2001 ) Molecular cloning: a laboratory manual, 3rd ed. Cold Spring Harbor Laboratory Press, N. Y.
Schreiber, S. L., and Crabtree, G. R. (1992) The mechanism of action of cyclosporine A and
FK506. Immunology Today 13: 136-142.
Schwecke, T., Aparicio, J. F., Molnar, I., Konig, A., Khaw, L. E., Haydock, S. F., Oliynyk, M.,
Caffrey, P., Cortes, J., Lester, J. B., Bohm, G.A., Staunton, J., and Leadlay, P. F. (1995)
The biosynthetic gene cluster for the polyketide immunosuppressant rapamycin.
Proceedings of the National Academy of Sciences of the United States of America 92:
7839-7843.
Sedrani, R., Cottens, S., Kallen, J., and Schuler, W. (1998) Chemical modifications of
rapamycin: the discovery of SDZ RAD. Transplantation Proceedings 30: 2192-2194. Sehgal, S. N., Baker, H., and Vezina, C. (1975) Rapamycin (AY-22,989), a new antifungal antibiotic II. Fermentation, isolation and characterization. The Journal of Antibiotics 28:
727-733.
Shepherd, P. R , Withers, D. J., and Siddle K. (1998) Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling. Biochemical Journal 333: 471-490.
Shima, J., Hesketh, A., Okamoto, S., Kawamoto, S., and Ochi, K. (1996) Induction of
actinorhodin production by rpsL (encoding ribosomal protein S12) mutations that confer streptomycin resistance in Streptomyces lividans and Streptomyces coelicolor A3(2).
Journal of Bacteriology MZ: 7276-7284.
Sigal, N. H., and Dumont, FJ. (1992) Cyclosporine A, FK-506, and rapamycin: pharmacological probes of lymphocyte signal transduction. Annual Review of Immunology 10: 519-560. Sigal, N. H., Dumont, F., Durette, P., Siekierka, JJ. , Peterson, L., Rich, D., Dunlap, B. E.,
Staruch, MJ., Melino, M. R., Koprak, S. L., Williams, D., Witzel,B. and Pisano, J. M.
(1991 ) Is Cyclophilin Involved in the Immunosuppressive and Nephrotoxic Mechanism of Action of Cyclosporin A? Journal of Experimental Medicine 173:619-628
Staunton, J., and Weissman, KJ. (2001 ) Polyketide biosynthesis: a millennium review. Natural
Product Reports 18: 380-416.
Steiner, J. P., Hamilton, G. S., Ross, D. T., Valentine, H. L., Guo, H., Connolly, M.A., Liang, S.,
Ramsey, C, Li, J.-H J., Huang, W., Howorth, P., Soni, R., Fuller, M., Sauer, H.,
Nowotnik, A.C., and Suzdak, P. D. (1997) Neutrophic immunophilin ligands stimulate structural and functional recovery in neurodegenerative animal models. Proceedings of the National Academy of Sciences of the United States of America 94:2019-2024.
Strassler, C, Linden, A., and Heimgartner, H. (1997) Novel heterospirocyclic 3-amino-2/-/- azirines as synthons for heterocyclic α-amino acids. HeIv. Chim. Acta. 80: 1528-1554 Tamura, S., Tokunaga, Y., Ibuki, R., Amidon, G. L., Sezaki, H. and Yamashita, S. (2003) The site specific transport and metabolism of Tacrolimus in the rat small intestine. The
Journal of Pharmacology and Experimental Therapeutics 306: 310-316 Takiguchi, Y., Mishima, H., Okuda, M., Terao, M., Aoki, A., Fukuda, R. Milbemycins, a new family of macrolide antibiotics: fermentation, isolation and physico-chemical properties.
J. Antibiot. (1980) 33(10): 1120-7
Van Duyne, G. D., Standaert, R. F., Karplus, P.A., Schreiber, S. L., and Clardy, J. (1993) Atomic structures of the human immunophilin FKBP-12 complexes with FK506 and rapamycin.
Journal of Molecular Biology 229: 105-124.
Vilella-Bach, M., Nuzzi, P., Fang, Y.M., and Chen, J. (1999) The FKBP12-rapamycin-binding domain is required for FKBP12-rapamycin-associated protein kinase activity and Gi progression. Journal of Biological Chemistry 274: 4266-4272.
Waller, J. R., and Nicholson, M. L. (2001 ) Molecular mechanisms of renal allograft fibrosis.
British Journal of Surgery 88: 1429-1441.
Warner, L. M., Adams, L. M., Chang, J.Y., Sehgal, S.N. (1992). A modification of the in vivo mixed lymphocyte reaction and rapamycin's effect in this model. Clin. Immunol.
Immunopathol. 64(3):242-7.
Ward, D. E., Ross, R.P., Van der Weijden, C. C., Snoep, J. L., Claiborne, A.L. Catabolism of branched-chain . alpha. -keto acids in Enterococcus faecalis: the bkd gene cluster, enzymes, and metabolic route. J. Bact. (1999), 181(17): 5433-5442.
Wei, X., Liang, Y., Zheng, Y. Enhancement and Selective Production of Oligomycin through
Inactivation of Avermectin's Starter Unit in Streptomyces avermitilis. Biotechnology
Letters (2006), 28(12), 911-916
Wilkinson, B., Foster, G., Rudd, B.A.M., Taylor, N. L., Blackaby, A.P., Sidebottom, PJ. , Cooper,
D. J., Dawson, MJ. , Buss, A.D., Gaisser, S., Bohm, I. U., Rowe, CJ., Cortes, J.,
Leadlay, P. F. and Staunton, J. (2000). Novel octaketide macrolides related to 6- deoxoerythronolide B provide evidence for iterative operation of the erythromycin polyketide synthase. Chemistry & Biology 7: 1 11-1 17.
Wu, K., Chung, L., Revill, W.P., Katz, L., and Reeves, CD. (2000) The FK520 gene cluster of
Streptomyces hygroscopicus var. ascomyceticus (ATCC 14891 ) contains genes for biosynthesis of unusual polyketide extender units. Gene 251 : 81-90.
Yem, A.W., Tomasselli, A.G., Heinrikson, R. L., Zurcher-Neely, H., Ruff, V.A., Johnson, R.A., and Deibel, M. R. (1992) The Hsp56 component of steroid receptor complexes binds to immobilized FK506 and shows homology to FKBP-12 and FKBP-13. Journal of
Biological Chemistry 267: 2868-2871.
Yu, K., Toral-Barza, L., Discafani, C, Zhang, W.G., Skotnicki, J., Frost, P., Gibbons, JJ. (2001 ) mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocrine-Related Cancer 8:249-258.
Zhu, J., Wu J., Frizell, E., Liu, S. L., Bashey, R., Rubin, R., Norton, P., Zern, M.A. (1999).
Rapamycin inhibits hepatic stellate cell proliferation in vitro and limits fibrogenesis in an in vivo model of liver fibrosis. Gastroenterology. 117(5): 1198-204.
Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.
All documents referred to herein, including patents and patent applications, are incorporated by reference in their entirety.

Claims

Claims
1. A compound of formula (I)
Figure imgf000047_0001
Wherein:
(ι) Xa and Xb represent a bond; or
(ιι) Xa represents a bond and Xb represents CH2, S, O; or
(in) Xa represents CH2, S, O, fused cyclopropyl unit and Xb represents a bond;
Z represents keto or CH2;
Ri represents a moiety selected from:
Figure imgf000048_0001
A B
Figure imgf000048_0002
D E F
Figure imgf000048_0003
I
G H
Figure imgf000048_0004
V W X Y R2 represents H, alkyl, halo, hydroxyl or thiol;
R3 represents H, Ci-4alkyl, halo, hydroxyl or thiol;
R4 represents H, Ci-4alkyl, halo, hydroxyl or thiol;
R5 represents OMe, Me or H;
R6 represents OMe, Me or H;
R7 represents Ci-6alkyl or Ci-6alkenyl, linear or branched, in either case optionally substituted by one to three halogen atoms, save that R7 does not represent -CH2CH2 or -CH2CH=CH2 and when R1 = P, Q, R, S, T, U, V, W, X or Y, then R7 does not represent CH3;
R8 represents OH;
R9 represents H, OH, halo, thiol, CrC4 alkyl;
Rio, Rn and Ri2 independently represent F, Cl, CrC4 alkyl, ORi3, SRi3 or NHRi3 and Ri3 represents H, CrC4 alkyl or CrC4 acyl, wherein two or three of Rio-Ri2 are CrC4 alkyl;
Ri3 and Ri4 independently represent H, F, Cl, CrC4 alkyl, ORi5, SRi5 or NHRi5 and Ri5 represents H, CrC4 alkyl or CrC4 acyl;
Ri6 Ri7 and Ri8 independently represent H, F, Cl, CrC4 alkyl, ORi9, SRi9 or NHRi9 and Ri9 represents H, CrC4 alkyl or CrC4 acyl,
R20 and R2i independently represent H, F, Cl, CrC4 alkyl, OR22, SR22 or NHR22 and R22 represents H, CrC4 alkyl or CrC4 acyl,
R23 and R24 independently represent H, F, Cl, CrC4 alkyl, OR25, SR25 or NHR25 and R25 represents H, CrC4 alkyl or CrC4 acyl;
R26 represents H, F, Cl, CrC4 alkyl, OR27, SR27 or NHR27, and R27 represents H, CrC4 alkyl or
CrC4 acyl;
R28 and R29 independently represent H, F, Cl, CrC4 alkyl, OR30, SR30 or NHR30 and R30 represents H, CrC4 alkyl or CrC4 acyl ;
R3i R32 and R33 independently represent H, F, Cl, CrC4 alkyl, OR34, SR34 or NHR34 and R34 represents H, CrC4 alkyl or CrC4 acyl;
R35 R36 and R37 independently represent H, F, Cl, CrC4 alkyl, OR38, SR38 or NHR38 and R38 represents H, CrC4 alkyl or CrC4 acyl, save that OR41 shall not represent OH;
R39 R40, R4i and R42 independently represent H, F, Cl, CrC4 alkyl, OR43, SR43 or NHR43 and R43 represents H, CrC4 alkyl or CrC4 acyl,
R44 and R45 independently represent H, F, Cl, CrC4 alkyl, OR46, SR46 or NHR46 and R46 represents H, CrC4 alkyl or CrC4 acyl;
R47, R48 and R49 are independently selected from H, F, Cl, OR50, SR50 and NHR50 and R50 is selected from H, Ci-C4alkyl and Ci-C4acyl;
A is selected from O, S and NR5i, R5i represents H, Ci-C4alkyl or Ci-C4acyl, R52 and R53 are independently selected from H, Ci-C4alkyl, OR54, SR54 and NHR54, R54 represents Ci-C4alkyl or
Ci-C4acyl, Xc represents a bond or CH2, when Xc is bond then R54 represents H and when Xc is CH2 then R54 represents H, F, Cl, OH, SH, Ci-C4 alkyl, OR55, SR55 or NHR55 and R55 represents Ci-C4alkyl or Ci-C4 acyl;
B represents O, S, or NR6O, R6o represents H, Ci-C4alkyl, or Ci-C4acyl, R56 represents d-
C4alkyl, OR6i, SR6i or NHR6i, R6i represents Ci-C4alkyl or Ci-C4acyl, R58 represents H, F, Cl,
OH, SH, CrC4 alkyl, OR62, SR62 or NHR62, R62 represents Ci-C4 alkyl or CrC4acyl, Xd and Xe independently represent bond or CH2 provided that Xd and Xe do not both represent CH2 and when Xd is bond then R57 represents H and when Xe is CH2 then R57 represents H, F, Cl, OH,
SH, Ci-C4alkyl, OR63, SR63 or NHR63, R63 represents CrC4 alkyl or CrC4 acyl and when Xe is bond R59 represents H and wherein when Xe represents CH2 then R59 represents H, F, Cl, OH,
SH, CrC4 alkyl, OR64, SR64 or NHR64, and R64 represents Ci-C4alkyl or Ci-C4acyl;
R65 and R66 independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR67, SR67 or NHR67, and
R67 represents Ci-C4alkyl or CrC4 acyl;
R68 represents H, F, Cl, OH, SH, Ci-C4alkyl, OR69, SR69 or NHR69, and R69 represents CrC4 alkyl or CrC4 acyl;
R70 and R7i independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR72, SR72 or NHR72 and
R72 represents CrC4 alkyl or CrC4 acyl ;
R73, R74 and R75 independently represent H, F, Cl, OH, SH, CrC4 alkyl, OR76, SR76 or NHR76 and R76 represents CrC4 alkyl or CrC4 acyl;
R77, R78 and R79 independently represent H, F, Cl, OR82, SR82, CrC4 alkyl or CN, R82 represents H, CrC4 alkyl or CrC4 acyl, R80 and R8i independently represent H, F, Cl, OH, SH or CrC4 alkyl, provided that at least one of R77, R78 and R79 is not H or CrC4 alkyl;
R83, R84 and R85 are independently selected from H, F, Cl, OR88, SR88, C1-C4 alkyl and CN, R88 represents H, CrC4 alkyl or CrC4 acyl, R86 and R87 independently represent H, F, Cl, OH, SH or CrC4 alkyl and provided that at least one of R83, R84 and R85 does not represent H or CrC4 alkyl;
or a physiologically functional derivative thereof.
2. A compound according to claim 1 wherein Z represents =0
3. A compound according to claim 1 or claim 2 wherein Xa represents a bond.
4. A compound according to claim 1 or claim 2 wherein Xa represents CH2.
5. A compound according to claim 1 or claim 2 wherein Xb represents a bond.
6. A compound according to any one of claims 1 to 5 wherein R5 represents OMe.
7. A compound according to any one of claims 1 to 6 wherein R6 represents OMe.
8. A compound according to any one of claims 1 to 7 wherein R2 represents H.
9. A compound according to any one of claims 1 to 8 wherein R3 represents H.
10. A compound according to any one of claims 1 to 9 wherein R4 represents H.
1 1. A compound according to any one of claims 1 to 10 wherein R7 represents -CH3, - CH2CH2CI, -CH2CH2F, -CH2CH2CH3, -CH2(CH2)4CH3, -CH2CH2Br, -CH2CH2D, -CH2CH2CH2F, - CH2CH2CH2CI, -CH2CH2CH2Br or -CH=CHCH3.
12. A compound according to any one of claims 1 to 1 1 wherein R1 represents [A].
13. A compound according to any one of claims 1 to 1 1 wherein R1 represents [B].
14. A compound according to any one of claims 1 to 1 1 wherein R1 represents [C].
15. A compound according to any one of claims 1 to 1 1 wherein R1 represents [D].
16. A compound according to any one of claims 1 to 1 1 wherein R1 represents [E].
17. A compound according to any one of claims 1 to 1 1 wherein R1 represents [F].
18. A compound according to any one of claims 1 to 1 1 wherein R1 represents [G].
19. A compound according to any one of claims 1 to 1 1 wherein R1 represents [H].
20. A compound according to any one of claims 1 to 1 1 wherein R1 represents [I].
21. A compound according to any one of claims 1 to 1 1 wherein R1 represents [J].
22. A compound according to any one of claims 1 to 1 1 wherein R1 represents [K].
23. A compound according to any one of claims 1 to 1 1 wherein R1 represents [L].
24. A compound according to any one of claims 1 to 1 1 wherein R1 represents [M].
25. A compound according to any one of claims 1 to 1 1 wherein R1 represents [N].
26. A compound according to any one of claims 1 to 1 1 wherein R1 represents [O].
27. A compound according to any one of claims 1 to 1 1 wherein R1 represents [P].
28. A compound according to any one of claims 1 to 1 1 wherein R1 represents [Q].
29. A compound according to any one of claims 1 to 1 1 wherein R1 represents [R].
30. A compound according to any one of claims 1 to 1 1 wherein R1 represents [S].
31. A compound according to any one of claims 1 to 1 1 wherein R1 represents [T].
32. A compound according to any one of claims 1 to 1 1 wherein R1 represents [U].
33. A compound according to any one of claims 1 to 11 wherein R1 represents [V].
34. A compound according to any one of claims 1 to 1 1 wherein R1 represents [W].
35. A compound according to any one of claims 1 to 1 1 wherein R1 represents [X].
36. A compound according to any one of claims 1 to 1 1 wherein R1 represents [Y].
37. A compound according to any one of claims 1 to 36 for use as a pharmaceutical.
38. A pharmaceutical composition comprising a compound according to any one of claims 1 to 36 together with one or more physiologically acceptable diluents or carriers.
39. A compound according to any one of claims 1 to 36 for use in the treatment or prophylaxis of organ rejection after transplantation, autoimmune diseases, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthrisis, fibrosis and/or other
hyperproliferative disorders.
40. Use of a compound according to any one of claims 1 to 36 in the manufacture of a medicament for the treatment or prophylaxis of organ rejection after transplantation, autoimmune diseases, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthrisis, fibrosis and/or other hyperproliferative disorders.
41. A method of treatment or prophylaxis of organ rejection after transplantation, autoimmune diseases, fungal infections, cancer, neurodegeneration, psoriasis, rheumatoid arthrisis, fibrosis and/or other hyperproliferative disorders which comprises administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 36.
42. A method of producing compounds of formula (I) according to any one of claims 1 to 36 comprising:
(a) generating a recombinant strain of a FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated; and
(b) feeding a non-natural C21 extender unit to said recombinant strain;
(c) culturing said strain; and
(d) optionally isolating compounds of formula (I).
43. A method of producing compounds of formula (I) according to any one of claims 1 to 36 comprising:
(a) feeding a non-natural C21 extender unit to a recombinant strain of a FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 and FK520 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated;
(b) culturing said strain; and
(c) optionally isolating compounds of formula (I).
44. A method of producing compounds of formula (I) according to any one of claims 1 to 36 comprising:
(a) generating a recombinant strain of a non-FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated; and
(b) feeding a non-natural C21 extender unit to said recombinant strain;
(c) culturing said strain; and
(d) optionally isolating compounds of formula (I).
45. A method of producing compounds of formula (I) according to any one of claims 1 to 36 comprising:
(a) feeding a non-natural C21 extender unit to a recombinant strain of a non-FK506 producing host that contains a biosynthetic cluster that encodes polypeptides involved in FK506 and FK520 analogue synthesis in which one or more of the genes responsible for provision of the C21 extender unit (based on the structure of FK506) has been deleted or inactivated; (b) culturing said strain; and
(c) optionally isolating compounds of formula (I)
46. A method according to any one of claims 41 to 45 in which the ccr gene responsible for carboxylation of the C21 extender unit is not deleted or inactivated, or else is comprised elsewhere in functional form in the host.
47. A hybrid PKS containing starter and extension modules for producing FK506 derivatives in which one or more of the gene products responsible for provision of the C21 extender unit (based on the structure of FK506) is absent or inactive.
48. An engineered FK506 producing strain containing a hybrid PKS according to claim 47.
49. An engineered non- FK506 producing strain containing a hybrid PKS according to claim 47.
50. A strain according to claim 48 or 49 in which the ccr gene product responsible for carboxylation of the C21 extender unit is not absent or inactivated.
51. An engineered non-FK506 producing strain containing a functional PKS which strain comprises one or more genes responsible for provision of the C21 extender unit (based on the structure of FK506) from the FK506 biosynthetic cluster.
52. An engineered non-FK506 producing strain according to claim 51 wherein the functional PKS is a hybrid PKS including the AT from module 7 of the FK506 biosynthetic cluster.
53. An engineered strain according to any any of claims 48 to 52 which strain is further engineered so as not to produce the natural starter unit.
54. An engineered strain according to any one of claims 48 to 52 which strain is further engineered so that the gene fkbO is deleted or inactivated.
55. An engineered strain according to any one of claims 48 to 52 which strain is further engineered so that the natural loading module is replaced by the loading module from the avermectin or an avermectin like PKS.
56. A process for producing a polyketide which comprises culturing an engineered strain according to any one of claims 48 to 55 in the presence of a non-natural C21 extender unit and optionally isolating said polyketide.
57. A method according to any of the claims 41 to 46 where additional genes are incorporated into the host at an attachment site in order to generate the extender unit.
58. A method according to claim 57 where salL is incorporated as an additional gene.
PCT/GB2010/051364 2009-08-20 2010-08-18 Polyketides analogues and methods for their production WO2011021036A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0914589A GB0914589D0 (en) 2009-08-20 2009-08-20 Novel compounds and methods for their production
GB0914589.7 2009-08-20

Publications (1)

Publication Number Publication Date
WO2011021036A1 true WO2011021036A1 (en) 2011-02-24

Family

ID=41171688

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2010/051364 WO2011021036A1 (en) 2009-08-20 2010-08-18 Polyketides analogues and methods for their production

Country Status (2)

Country Link
GB (1) GB0914589D0 (en)
WO (1) WO2011021036A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011068341A2 (en) * 2009-12-03 2011-06-09 (주)제노텍 Gene and protein for biosynthesis of tricyclocompounds
WO2012089349A3 (en) * 2010-12-31 2012-12-27 Acies Bio D.O.O. Novel polyketide compounds and methods of making same
US20130230559A1 (en) * 2010-08-24 2013-09-05 Ewha University - Industry Collaboration Foundatio Novel tacrolimus analogues, a neuroprotective composition comprising the same, an immunosuppressive composition comprising the same, a method for preparing the same, and a mutant for producing the same
CN114591286A (en) * 2021-04-12 2022-06-07 南京大学 Novel macrolide compound acautalides A-C and preparation method and application thereof

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
EP0427680A1 (en) * 1989-11-09 1991-05-15 Sandoz Ltd. Heteroatoms-containing tricyclic compounds
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
WO1993013663A1 (en) 1992-01-17 1993-07-22 Abbott Laboratories Method of directing biosynthesis of specific polyketides
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
WO1997002358A1 (en) 1995-07-06 1997-01-23 The Leland Stanford Junior University Cell-free synthesis of polyketides
WO1998001546A2 (en) 1996-07-05 1998-01-15 Biotica Technology Limited Polyketides and their synthesis
WO2000000618A2 (en) 1998-06-29 2000-01-06 Biotica Technology Limited Polyketides and their synthesis
WO2000001827A2 (en) 1998-07-06 2000-01-13 Biotica Technology Limited Polyketides, their preparation, and materials for use therein
US6150513A (en) 1998-09-16 2000-11-21 Kosan Biosciences, Inc. Polyketide synthase enzymes and recombinant DNA constructs therefor
WO2002014482A2 (en) 2000-08-14 2002-02-21 Biotica Technology Limited Polyketides and their synthesis
WO2004007709A2 (en) 2002-07-16 2004-01-22 Biotica Technology Limited Production of polyketides

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
EP0427680A1 (en) * 1989-11-09 1991-05-15 Sandoz Ltd. Heteroatoms-containing tricyclic compounds
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
WO1993013663A1 (en) 1992-01-17 1993-07-22 Abbott Laboratories Method of directing biosynthesis of specific polyketides
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5399163A (en) 1992-07-24 1995-03-21 Bioject Inc. Needleless hypodermic injection methods and device
WO1997002358A1 (en) 1995-07-06 1997-01-23 The Leland Stanford Junior University Cell-free synthesis of polyketides
WO1998001546A2 (en) 1996-07-05 1998-01-15 Biotica Technology Limited Polyketides and their synthesis
WO2000000618A2 (en) 1998-06-29 2000-01-06 Biotica Technology Limited Polyketides and their synthesis
WO2000001827A2 (en) 1998-07-06 2000-01-13 Biotica Technology Limited Polyketides, their preparation, and materials for use therein
US6150513A (en) 1998-09-16 2000-11-21 Kosan Biosciences, Inc. Polyketide synthase enzymes and recombinant DNA constructs therefor
WO2002014482A2 (en) 2000-08-14 2002-02-21 Biotica Technology Limited Polyketides and their synthesis
WO2004007709A2 (en) 2002-07-16 2004-01-22 Biotica Technology Limited Production of polyketides

Non-Patent Citations (116)

* Cited by examiner, † Cited by third party
Title
"Methods in Biotechnology", vol. 4, 1998, HUMANA PRESS, article "Natural Products Isolation"
APARICIO, J.F.; MOLNAR, I.; SCHWECKE, T.; KONIG, A.; HAYDOCK, S.F.; KHAW, L.E.; STAUNTON, J.; LEADLAY, P.F.: "Organization of the biosynthetic gene cluster for rapamycin in Streptomyces hygroscopicus: analysis of the enzymatic domains in the modular polyketide synthase", GENE, vol. 169, 1996, pages 9 - 16
ARMSTRONG, V.W.; OELLERICH, M.: "New developments in the immunosuppressive drug monitoring of cyclosporine, tacrolimus and azathioprine", CLINICAL BIOCHEMISTRY, vol. 34, no. 1, 2001, pages 9 - 16
BAKER, H.; SIDOROWICZ, A.; SEHGAL, S.N.; VÉZINA, C.: "Rapamycin (AY-22,989), a new antifungal antibiotic", IN VITRO AND IN VIVO EVALUATION. JOURNAL OFANTIBIOTICS, vol. 31, 1978, pages 539 - 545
BIERMAN, M.; LOGAN, R.; O'BRIEN, K.; SENO, E.T.; NAGARAJA RAO, R.; SCHONER, B.E.: "Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to Streptomyces spp", GENE, vol. 116, 1992, pages 43 - 49
BLANC, V.; GIL, P.; BAMAS-JACQUES, N.; LORENZON, S.; ZAGOREC, M.; SCHLEUNIGER, J.; BISCH, D.; BLANCHE, F.; DEBUSSCHE, L.; CROUZET,: "Identification and analysis of genes from Streptomyces pristinaespiralis encoding enzymes involved in the biosynthesis of the 4-dimethylamino-L-phenylalanine precursor of pristinamycin I", MOLECULAR MICROBIOLOGY, vol. 23, 1997, pages 191 - 202
BLANC, V.; LAGNEAUX, D.; DIDIER, P.; GIL, P.; LACROIX, P.; CROUZET, J.: "Cloning and analysis of structural genes from Streptomyces pristinaespiralis encoding enzymes involved in the conversion of pristinamycin IIB to pristinamycin IIA (PIIA): PIIA synthase and NADH:riboflavin 5'-phosphate oxidoreductase", JOURNAL OF BACTERIOLOGY, vol. 177, 1995, pages 5206 - 5214
CANNELL ET AL., NATURAL PRODUCTS ISOLATION, 1998
CAO, W.; MOHACSI, P.; SHORTHOUSE, R.; PRATT, R.; MORRIS, R.E.: "Effects of rapamycin on growth factor-stimulated vascular smooth muscle cell DNA synthesis. Inhibition of basic fibroblast growth factor and platelet-derived growth factor action and antagonism of rapamycin by FK506", TRANSPLANTATION, vol. 59, no. 3, 1995, pages 390 - 395
CARLSON, R.P.; HARTMAN, D.A.; TOMCHEK, L.A.; WALTER, T.L.; LUGAY, J.R.; CALHOUN, W.; SEHGAL, S.N.; CHANG, J.Y.: "Rapamycin, a potential disease-modifying antiarthritic drug", J. PHARMACOL. EXP. THER., vol. 266, no. 2, 1993, pages 1125 - 38
CARTER, G.T.; NIETSCHE, J.A.; HERTZ, M.R.; WILLIAMS, D.R.; SIEGEL, M.M.; MORTON, G.O.; JAMES, J.C.; BORDERS, D.B.: "LL-F28249 antibiotic complex: a new family of antiparasitic macrocyclic lactones. Isolation, characterization and structures of LL-F28249 alpha, beta, gamma, lambda", J. ANTIBIOT., vol. 41, no. 4, 1988, pages 519 - 529
CHAMBRAUD, B.; RADANYI, C.; CAMONIS, J.H.; SHAZAND, K.; RAJKOWSKI, K.; BAULIEU, E.E.: "FAP48, a new protein that forms specific complexes both immunophilins FKBP59 and FKBP12. Prevention by the immunosuppressant drugs FK506 and rapamycin", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, 1996, pages 32923 - 32929
CHANG, J.Y.; SEHGAL, S.N.; BANSBACH, C.C.: "FK506 and rapamycin: novel pharmacological probes of the immune response", TRENDS IN PHARMACOLOGICAL SCIENCES, vol. 12, 1991, pages 218 - 223
CHEN, J.; ZHENG, X.F.; BROWN, E.J.; SCHREIBER, S.L.: "Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 92, 1995, pages 4947 - 4951
CHOI, S-S.; HUR, Y-A.; SHERMAN, D.H.; KIM, E-S.: "Isolation of the biosynthetic gene cluster for tautomycetin, a linear polyketide T-cell specific immunomodulator from Streptomyces sp", CK4412 MICROBIOLOGY, vol. 153, 2007, pages 1095 - 1102
CHUNG L ET AL: "DELETION OF RAPQONML FROM THE RAPAMYCIN GENE CLUSTER OF STREPTOMYCES HYGROSCOPICUS GIVES PRODUCTION OF THE 16-O-DESMETHYL-27-DESMETHOXY ANALOG", JOURNAL OF ANTIBIOTICS, JAPAN ANTIBIOTICS RESEARCH ASSOCIATION, TOKYO, JP, vol. 54, no. 3, 1 March 2001 (2001-03-01), pages 250 - 256, XP001194490, ISSN: 0021-8820 *
DILELLA, A.G.; CRAIG, R.J.: "Exon organization of the human FKBP-12 gene: correlation with structural and functional protein domains", BIOCHEMISTRY, vol. 30, 1991, pages 8512 - 8517
DU, L.C.; SÁNCHEZ, C.; CHEN, M.; EDWARDS, D.J.; SHEN, B.: "The biosynthetic gene cluster for the antitumor drug bleomycin from Streptomyces verticillus A TCC15003 supporting functional interactions between nonribosomal peptide synthetases and a polyketide synthase", CHEMISTRY & BIOLOGY, vol. 7, 2000, pages 623 - 642
DUDKIN, L.; DILLING, M.B.; CHESHIRE, P.J.; HARWOOD, F.C.; HOLLINGSHEAD, M.; ARBUCK, S.G.; TRAVIS, R.; SAUSVILLE, E.A.; HOUGHTON, P: "Biochemical correlates of mTOR inhibition by the rapamycin ester CCI-779 and tumor growth inhibition", CLIN. CANCER RES., vol. 7, no. 6, 2001, pages 1758 - 64
DUMONT, F.; STARUCH, M.J.; KOPRAK, S.L.; SIEKIERKA, LIN, C.S.; HARRISON, R.; SEWELL, T.; KINDT, V.M.; BEATTIE, T.R.; WYVRATT, M.;: "The immunosuppressive and toxic effects of FK506 are mechanistically related: Pharmacology of a novel antagonist of FK506 and Rapamycin", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 176, 1992, pages 751 - 760
DUTTON, C. J.; S. P. GIBSON ET AL.: "Novel avermectins produced by mutational biosynthesis", J. ANTIBIOT., vol. 44, no. 3, 1991, pages 357 - 365
FANG, J.; ZHANG, Y.; HUANG, L.; JIA, X.; ZHANG, Q.; ZHANG, X.; TANG, G.; LIU, W.: "Cloning and characterisation of the tetrocarcin A gene cluster from Micromonospora chalcae NRRL 11289 reveals a highly conserved strategy for tetronate biosynthesis in spirotetronate antibiotics", J. BACT., vol. 190, no. 17, 2008, pages 6014 - 6025
FEHR, T.; SANGLIER, J-J.; SCHULER, W.; GSCHWIND, L.; PONELLE, M.; SCHILLING, W.; WIOLAND, C.: "Antascomicinc A, B, C, D and E: Novel FKBP12 binding compounds from a Micromonospora strain", J. ANTIBIOT., vol. 49, no. 3, 1996, pages 230 - 233
FINDLAY J.A; RADICS, L., CANADIAN JOURNAL OF CHEMISTRY, vol. 58, 1980, pages 579
FISHBEIN, T.M.; FLORMAN, S.; GONDOLESI, G.; SCHIANO, T.; LELEIKO, N.; TSCHERNIA, A.; KAUFMAN, S.: "Intestinal transplantation before and after the introduction of sirolimus", TRANSPLANTATION, vol. 73, no. 10, 2002, pages 1538 - 42
FOEY, A.; GREEN, P.; FOXWELL, B.; FELDMANN, M.; BRENNAN, F.: "Cytokine-stimulated T cells induce macrophage IL-10 production dependent on phosphatidylinositol 3-kinase and p70S6K: implications for rheumatoid arthritis", ARTHRITIS RES., vol. 4, no. 1, 2002, pages 64 - 70
GAISSER, S.; REATHER, J.; WIRTZ, G.; KELLENBERGER, L.; STAUNTON, J.; LEADLAY, P.F.: "A defined system for hybrid macrolide biosynthesis in Saccharopolyspora erythraea", MOLECULAR MICROBIOLOGY, vol. 36, 2000, pages 391 - 401
GALAT, A.: "Sequence diversification of the FK506-binding proteins in several different genomes", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 267, 2000, pages 4945 - 4959
GARRITY, G.M.; HEIMBUCH, B.K.; MOTAMEDI, H.; SHAFIEE, A.: "Genetic relationships among Actinomycetes that produce the immunosuppressant macrolides FK506, FK520/FK523 and rapamycin", JOURNAL OF INDUSTRIAL MICROBIOLOGY, vol. 1 2, no. 1, 1993, pages 42 - 47
GREGORY, C.R.; HUIE, P.; BILLINGHAM, M.E.; MORRIS, R.E.: "Rapamycin inhibits arterial intimal thickening caused by both alloimmune and mechanical injury. Its effect on cellular, growth factor and cytokine response in injured vessels", TRANSPLANTATION, vol. 55, no. 6, 1993, pages 1409 - 1418
GUBA, M.; VON BREITENBUCH, P.; STEINBAUER, M.; KOEHL, G.; FLEGEL, S.; HORNUNG, M.; BRUNS, C.J.; ZUELKE, C.; FARKAS, S.; ANTHUBER,: "Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor", NATURE MEDICINE, vol. 8, 2002, pages 128 - 135
HAMILTON, G.S.; STEINER, J.P.: "Immunophilins: Beyond immunosuppression", JOURNAL OF MEDICINAL CHEMISTRY, vol. 41, 1998, pages 5119 - 5143
HATANAKA H; KINO T; ASANO M; GOTO T; TANAKA H; OKUHARA M: "FK-506 related compounds produced by Streptomyces tsukubaensis No. 9993", J. ANTIBIOT. (TOKYO), vol. 42, no. 4, 1989, pages 620 - 2
HATANAKA, H.; KINO, T.; MIYATA, S.; INAMURA, N.; KURODA, A.; GOTO, T.; TANAKA, H.; OKUHARA, M.: "FR-900520 and FR-900523, novel immunosuppressants isolated from a Streptomyces. II. Fermentation, isolation and physico-chemical and biological characteristics", J. ANTIBIOT. (TOKYO), vol. 41, no. 11, 1988, pages 1592 - 601
HENDRICKSON, B.A.; ZHANG, W.; CRAIG, R.J.; JIN, Y.J.; BIERER, B.E.; BURAKOFF, S.; DILELLA, A.G.: "Structural organization of the genes encoding human and murine FK506-binding protein (FKBP)13 and comparison to FKBP1", GENE, vol. 134, 1993, pages 271 - 275
HOPWOOD, D.A.: "Genetic contributions to understanding polyketide synthases", CHEMICAL REVIEWS, vol. 97, 1997, pages 2465 - 2497
HOSTED, T.J.; BALTZ, R.H.: "Use of rpsL for dominance selection and gene replacement in Streptomyces roseosporus", JOURNAL OF BACTERIOLOGY, vol. 179, 1997, pages 180 - 186
HUNG, D.T.; JAMISON, T.F.; SCHREIBER, S.L.: "Understanding and controlling the cell cycle with natural products", CHEMISTRY & BIOLOGY, vol. 3, 1996, pages 623 - 639
HUNG, D.T.; SCHREIBER, S.L.: "cDNA cloning of a human 25 kDa FK506 and rapamycin binding protein", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 184, 1992, pages 733 - 738
JAIN, S.; BICKNELL, G.R.; WHITING, P.H.; NICHOLSON, M.L.: "Rapamycin reduces expression of fibrosis-associated genes in an experimental model of renal ischaemia reperfusion injury", TRANSPLANT PROC., vol. 33, no. 1-2, 2001, pages 556 - 8
JIN, Y.J.; BURAKOFF, S.J.; BIERER, B.E.: "Molecular cloning of a 25-kDa high affinity rapamycin binding protein, FKBP25", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 267, 1992, pages 10942 - 10945
KAHAN, B.D.; CAMARDO, J.S.: "Rapamycin: Clinical results and future opportunities", TRANSPLANTATION, vol. 72, 2001, pages 1181 - 1193
KAHAN, B.D.; CHANG, J.Y.; SEHGAL, S.N.: "Preclinical evaluation of a new potent immunosuppressive agent, rapamycin", TRANSPLANTATION, vol. 52, 1991, pages 185 - 191
KAPTURCZAK, H.U.; MEIER-KRIESCHE; KAPLAN, B.: "Pharmacology of Calcineurin Antagonists", TRANSPLANTATION PROCEEDINGS, vol. 36, no. 2S, 2004, pages 25S - 32S
KHAW, L.E.; BOHM, G.A.; METCALFE, S.; STAUNTON, J.; LEADLAY, P.F.: "Mutational biosynthesis of novel rapamycins by a strain of Streptomyces hygroscopicus NRRL 5491 disrupted in rapL, encoding a putative lysine cyclodeaminase", JOURNAL OF BACTERIOLOGY, vol. 180, 1998, pages 809 - 814
KIESER, T.; BIBB, M.J.; BUTTNER, M.J.; CHATER, K.F.; HOPWOOD, D.A.: "Practical Streptomyces Genetics", 2000, JOHN INNES FOUNDATION
KIRBY, B.; GRIFFITHS,C.E.M.: "Psoriasis: the future", BRITISH JOURNAL OF DERMATOLOGY, vol. 144, 2001, pages 37 - 43
KIRCHNER, G.I.; WINKLER, M.; MUELLER L.; VIDAL, C.; JACOBSEN, W.; FRANZKE, A.; WAGNER, S.; BLICK, S.; MANNS M.P.; SEWING K.-F.: "Pharmacokinetics of SDZ RAD and cyclosporin including their metabolites in seven kidney graft patients after the first dose of SDZ RAD", BRITISH JOURNAL OF CLINICAL PHARMACOLOGY, vol. 50, 2000, pages 449 - 454
KONIG, A.; SCHWECKE, T.; MOTNAR, I.; BOHM, G.; LOWDEN, P.A.S.; STAUNTON, J.; LEADLAY, P.F.: "The pipecolate-incorporating enzyme for the biosynthesis of the immunosuppressant rapamycin. Nucleotide sequence analysis, disruption and heterologus expression of rapP from Streptomyces hygroscopicus", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 247, 1997, pages 526 - 534
KONZ, D.; M. A. MARAHIEL: "How do peptide synthetases generate structural diversity?", CHEMISTRY & BIOLOGY, vol. 6, no. 2, 1999, pages R39 - R48
LIU, Y.; HAZZARD, C.; EUSTAQUIO, A.S.; REYNOLDS, K.; MOORE B.S.: "Biosynthesis of Salinosporamide from a,D-Unsaturated Fatty Acids: Implications for Extending Polyketide Synthase Diversity", J. AM. CHEM. SOC., 2009
LOWDEN, P. A. S.: "Ph.D. Dissertation", 1997, UNIVERSITY OF CAMBRIDGE, article "Studies on the biosynthesis of rapamycin"
LOWDEN, P.A.S.; BOHM, G.; STAUNTON, J.; LEADLAY, P.F.: "The nature of the starter unit for the rapamycin polyketide sythase", ANGEWANDTE CHEMIE, vol. 35, 1996, pages 2249 - 2251
LOWDEN, P.A.S.; WILKINSON, B.; BOHM, G.A.; HANDA, S.; FLOSS, H.G.; LEADLAY, P.F.; STAUNTON, J.: "Origin and true nature of the starter unit for the rapamycin polyketide synthase", ANGEWANDTE CHEMIE-INTERNATIONAL EDITION, vol. 40, 2001, pages 777 - 779
LYONS, W.E.; GEORGE, E.B.; DAWSON, T.M.; STEINER, J.P.; SNYDER, S.H.: "Immunosuppressant FK506 promotes neurite outgrowth in cultures of PC12 cells and sensory ganglia", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 91, 1994, pages 3191 - 3195
MACNEIL, D.J.; GEWAIN, K.M.; RUBY, C.L.; DEZENY, G.; GIBBONS, P.H.; MACNEIL, T.: "Analysis of Streptomyces avermitilis genes required for avermectin biosynthesis utilizing a novel integration vector", GENE, vol. 111, 1992, pages 61 - 68
MAES, B.D.; VANRENTERGHEM, Y.F.CH.: "Cyclosporine: Advantages Versus Disadvantages Vis-a-vis Tacrolimus", TRANSPLANTATION PROCEEDINGS, vol. 36, no. 2S, 2004, pages 40S - 49S
MARAHIEL, M.A.; STACHELHAUS, T.; MOOTZ, H.D.: "Modular peptide synthetases involved in nonribosomal peptide synthesis", CHEMICAL REVIEWS, vol. 97, 1997, pages 2651 - 2673
MARCH, J: "Advanced Organic Chemistry - Reactions, Mechanisms and Structure", WILEY INTERSCIENCE
MARGULIES ET AL.: "Genome sequencing in microfabricated high-density picolitre reactors", NATURE, vol. 437, 2005, pages 376 - 380
MEINGASSNER, J.G.; GRASSBERGER, M.; FAHRNGRUBER, H.; MOORE, H.D.; SCHUURMAN, H.; STUTZ., A.: "A novel anti-inflammatory drug, SDZ ASM 981, for the topical and oral treatment of skin diseases: in vivo pharmacology", BRITISH JOURNAL OF DERMATOLOGY, vol. 137, 1997, pages 568 - 576
MOLNÁR, I.; APARICIO, J.F.; HAYDOCK, S.F.; KHAW, L.E.; SCHWECKE, T.; K6NIG, A.; STAUNTON, J.; LEADLAY, P.F.: "Organisation of the biosynthetic gene cluster for rapamycin in Streptomyces hygroscopicus: analysis of genes flanking the polyketide synthase", GENE, vol. 169, 1996, pages 1 - 7
MORICE, M.C.; SERRUYS, P.W.; SOUSA, J.E.; FAJADET, J.; BAN HAYASHI, E.; PERIN, M.; COLOMBO, A.; SCHULER, G.; BARRAGAN, P.; GUAGLIU: "RAVEL Study Group. Randomized Study with the Sirolimus-Coated Bx Velocity Balloon-Expandable Stent in the Treatment of Patients with de Novo Native Coronary Artery Lesions. A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization", N. ENG.1 J. MED., vol. 346, no. 23, 2002, pages 1773 - 80
MOTAMEDI H ET AL: "Characterization of methyltransferase and hydroxylase genes involved in the biosynthesis of the immunosupressants FK506 and FK520", JOURNAL OF BACTERIOLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, WASHINGTON, DC; US, vol. 178, no. 17, 1 July 1996 (1996-07-01), pages 5243 - 5248, XP002137077, ISSN: 0021-9193 *
MOTAMEDI H ET AL: "Integrative vectors for heterologous gene expression in Streptomyces spp", GENE, ELSEVIER, AMSTERDAM, NL, vol. 160, no. 1, 4 July 1995 (1995-07-04), pages 25 - 31, XP004042173, ISSN: 0378-1119, DOI: DOI:10.1016/0378-1119(95)00191-8 *
MOTAMEDI, H.; CAI, S.J.; SHAFIEE, A.; ELLISTON, K.O.: "Structural organization of a multifunctional polyketide synthase involved in the biosynthesis of the macrolide immunosuppressant FK506", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 244, 1997, pages 74 - 80
MOTAMEDI, H.; SHAFIEE, A.: "The biosynthetic gene cluster for the macrolactone ring of the immunosuppressant FK506", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 256, 1998, pages 528 - 534
MOTAMEDI, H.; SHAFIEE, A.; CAI, S.J.; STREICHER, S.L.; ARISON, B.H.; MILLER, R.R.: "Characterization of methyltransferase and hydroxylase genes involved in the biosynthesis of the immunosuppressants FK506 and FK520", JOURNAL OF BACTERIOLOGY, vol. 178, 1996, pages 5243 - 5248
MURAMATSU, H.; MOKHTAR, S.I.; KATSUOKA, M.; EZAKI, M.: "Phylogenetic Analysis of Immunosuppressant FK506-Producing Streptomycete Strains", ACTINOMYCETOLOGICA, vol. 19, 2005, pages 33 - 39
MYCKATYN, T.M.; ELLIS, R.A.; GRAND, A.G.; SEN, S.K.; LOWE, J.B. 3RD; HUNTER, D.A.; MACKINNON, S.E.: "The effects of rapamycin in murine peripheral nerve isografts and allografts", PLAST. RECONSTR. SURG., vol. 109, no. 7, 2002, pages 2405 - 17
NAVIA, M.A.: "Protein-drug complexes important for immunoregulation and organ transplantation", CURRENT OPINION IN STRUCTURAL BIOLOGY, vol. 6, 1996, pages 838 - 847
NCCLS REFERENCE METHOD FOR BROTH DILUTION ANTIFUNGAL SUSCEPTIBILITY TESTING FOR YEASTS: APPROVED STANDARD M27-A, vol. 17, no. 9, 1997
NGHIEM, P.; PEARSON, G.; LANGLEY, R.G.: "Tacrolimus and pimecrolimus: From clever prokaryotes to inhibiting calcineurin and treating atopic dermatitis", JOURNAL OF THE AMERICAN ACADEMY OF DERMATOLOGY, vol. 46, 2002, pages 228 - 241
OKAZAKI, T.; ONO, M.; AOKI, A.; FUKUDA, R.: "Milbemycins, a new family of macrolide antibiotics: producing organism and its mutants", J. ANTIBIOT. (1983), vol. 36, no. 4, 1983, pages 438 - 441
OLIYNYK, M.; STARK, C.B.W.; BHATT, A.; JONES, M.A.; HUGHES-THOMAS, Z.A.; WILKINSON, C.; OLIYNYK, Z.; DEMYDCHUK, Y.; STAUNTON, J.;: "Analysis of the biosynthetic gene cluster for the polyether antibiotic monensin in Streptomyces cinnamonensis and evidence for the role of monB and monC genes in oxidative cyclization", MOL. MICRO., vol. 49, no. 5, 2003, pages 1179 - 1190
PAGET, M.S.B.; CHAMBERLIN, L.; ATRIH, A.; FOSTER, S.J.; BUTTNER, M.J.: "Evidence that the extracytoplasmic function sigma factor CyE is required for normal cell wall structure in Streptomyces coelicolor A3(2)", JOURNAL OF BACTERIOLOGY, vol. 181, 1999, pages 204 - 211
PAIVA, N.L.; DEMAIN, A.L.; ROBERTS, M.F.: "Incorporation of acetate, propionate, and methionine into rapamycin By Streptomyces hygroscopicus", JOURNAL OF NATURAL PRODUCTS, vol. 54, 1991, pages 167 - 177
PAIVA, N.L.; DEMAIN, A.L.; ROBERTS, M.F.: "The immediate precursor of the nitrogen- containing ring of rapamycin is free pipecolic acid", ENZYME AND MICROBIAL TECHNOLOGY, vol. 15, 1993, pages 581 - 585
PATTERSON, C.E.; SCHAUB, T.; COLEMAN, E.J.; DAVIES E.C.: "Developmental regulation of FKBP65. An ER-localized extracellular matrix binding-protein", MOLECULAR BIOLOGY OF THE CELL, vol. 11, 2000, pages 3925 - 3935
PFEIFER, B.A.; ADMIRAAL, S.J.; GRAMAJO, H.; CANE, D.E.; KHOSLA, C.: "Biosynthesis of complex polyketides in a metabolically engineered strain of E. coli", SCIENCE, vol. 291, 2001, pages 1790 - 1792
POWELL, N.; TILL, S.; BUNGRE, J.; CORRIGAN, C.: "The immunomodulatory drugs cyclosporin A, mycophenolate mofetil, and sirolimus (rapamycin) inhibit allergen-induced proliferation and IL-5 production by PBMCs from atopic asthmatic patients", J. ALLERGY CLIN. IMMUNOL., vol. 108, no. 6, 2001, pages 915 - 7
RABINOVITCH, A.; SUAREZ-PINZON, W.L.; SHAPIRO, A.M.; RAJOTTE, R.V.; POWER, R.: "Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice", DIABETES, vol. 51, no. 3, 2002, pages 638 - 45
RAWLINGS, B.J.: "Type I polyketide biosynthesis in bacteria (Part A)", NATURAL PRODUCT REPORTS, vol. 18, 2001, pages 190 - 227
REITAMO, S.; SPULS, P.; SASSOLAS, B.; LAHFA, M.; CLAUDY, A.; GRIFFITHS, C.E.: "Br. J. Dermatol.", vol. 145, 2001, SIROLIMUS EUROPEAN PSORIASIS STUDY GROUP, article "Efficacy of sirolimus (rapamycin) administered concomitantly with a subtherapeutic dose of cyclosporin in the treatment of severe psoriasis: a randomized controlled trial", pages: 438 - 45
REYNOLDS, K.A.; DEMAIN, A.L.: "Biotechnology of Antibiotics", 1997, article "Rapamycin, FK506 and Ascomycin-related Compounds", pages: 497 - 520
ROSEN, M.K.; SCHREIBER, S.L.: "Natural products as probes of cellular function: studies of immunophilins", ANGEWANDTE CHEMIE-INTERNATIONAL EDITION IN ENGLISH, vol. 31, 1992, pages 384 - 400
ROY, J.N.; BARAMA, A.; POIRIER, C.; VINET, B.; ROGER, M.: "Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients", PHARMACOGENETICS, vol. 16, no. 9, 2006, pages 659 - 665
ROYMANS, D.; SLEGERS, H.: "Phosphaditidylinositol 3-kinases in tumor progression", EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 268, 2001, pages 487 - 498
SAMBROOK, J.; FRITSCH, E.F.; MANIATIS, T.: "Molecular cloning: a laboratory manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SAMBROOK, J.; RUSSELL, D.W.: "Molecular cloning: a laboratory manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
SCHREIBER, S.L.; CRABTREE, G.R.: "The mechanism of action of cyclosporine A and FK506", IMMUNOLOGY TODAY, vol. 13, 1992, pages 136 - 142
SCHWARZER, D.; MARAHIEL, M.A.: "Multimodular biocatalysts for natural product assembly", NATURWISSENSCHAFTEN, vol. 88, 2001, pages 93 - 101
SCHWECKE, T.; APARICIO, J.F.; MOLNÁR, I.; K6NIG, A.; KHAW, L.E.; HAYDOCK, S.F.; OLIYNYK, M.; CAFFREY, P.; CORTÉS, J.; LESTER, J.: "The biosynthetic gene cluster for the polyketide immunosuppressant rapamycin", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OFAMERICA, vol. 92, 1995, pages 7839 - 7843
SEDRANI, R.; COTTENS, S.; KALLEN, J.; SCHULER, W.: "Chemical modifications of rapamycin: the discovery of SDZ RAD", TRANSPLANTATION PROCEEDINGS, vol. 30, 1998, pages 2192 - 2194
SEHGAL, S.N.; BAKER, H.; VÉZINA, C.: "Rapamycin (AY-22,989), a new antifungal antibiotic . Fermentation, isolation and characterization", THE JOURNAL OF ANTIBIOTICS, vol. 28, 1975, pages 727 - 733
SHAFIEE A ET AL: "CHEMICAL AND BIOLOGICAL CHARACTERIZATION OF TWO FK506 ANALOGS PRODUCED BY TARGETED GENE DISRUPTION IN STREPTOMYCES SP. MA6548", JOURNAL OF ANTIBIOTICS, JAPAN ANTIBIOTICS RESEARCH ASSOCIATION, TOKYO, JP, vol. 50, no. 5, 1 January 1997 (1997-01-01), pages 418 - 423, XP000906741, ISSN: 0021-8820 *
SHEPHERD, P.R; WITHERS, D.J.; SIDDLE K.: "Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling", BIOCHEMICAL JOURNAL, vol. 333, 1998, pages 471 - 490
SHIMA, J.; HESKETH, A.; OKAMOTO, S.; KAWAMOTO, S.; OCHI, K.: "Induction of actinorhodin production by rpsL (encoding ribosomal protein S12) mutations that confer streptomycin resistance in Streptomyces lividans and Streptomyces coelicolor A3(2)", JOURNAL OF BACTERIOLOGY, vol. 178, 1996, pages 7276 - 7284
SIGAL, N.H.; DUMONT, F.; DURETTE, P.; SIEKIERKA, J.J.; PETERSON, L.; RICH, D.; DUNLAP, B.E.; STARUCH, M.J.; MELINO, M.R.; KOPRAK,: "Is Cyclophilin Involved in the Immunosuppressive and Nephrotoxic Mechanism of Action of Cyclosporin A?", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 173, 1991, pages 619 - 628
SIGAL, N.H.; DUMONT, F.J.: "Cyclosporine A, FK-506, and rapamycin: pharmacological probes of lymphocyte signal transduction", ANNUAL REVIEW OF IMMUNOLOGY, vol. 10, 1992, pages 519 - 560
STAUNTON, J.; WEISSMAN, K.J.: "Polyketide biosynthesis: a millennium review", NATURAL PRODUCT REPORTS, vol. 18, 2001, pages 380 - 416
STEINER, J.P.; HAMILTON, G.S.; ROSS, D. T.; VALENTINE, H.L.; GUO, H.; CONNOLLY, M.A.; LIANG, S.; RAMSEY, C.; LI, J.-H.J.; HUANG, W: "Neutrophic immunophilin ligands stimulate structural and functional recovery in neurodegenerative animal models", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 94, 1997, pages 2019 - 2024
STRÄSSLER, C.; LINDEN, A.; HEIMGARTNER, H.: "Novel heterospirocyclic 3-amino-2H-azirines as synthons for heterocyclic a-amino acids", HELV. CHIM. ACTA., vol. 80, 1997, pages 1528 - 1554
TAKIGUCHI, Y.; MISHIMA, H.; OKUDA, M.; TERAO, M.; AOKI, A.; FUKUDA, R.: "Milbemycins, a new family of macrolide antibiotics: fermentation, isolation and physico-chemical properties", J. ANTIBIOT., vol. 33, no. 10, 1980, pages 1120 - 7
TAMURA, S.; TOKUNAGA, Y.; IBUKI, R.; AMIDON, G.L.; SEZAKI, H.; YAMASHITA, S.: "The site specific transport and metabolism of Tacrolimus in the rat small intestine", THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 306, 2003, pages 310 - 316
VAN DUYNE, G.D.; STANDAERT, R.F.; KARPLUS, P.A.; SCHREIBER, S.L.; CLARDY, J.: "Atomic structures of the human immunophilin FKBP-12 complexes with FK506 and rapamycin", JOURNAL OF MOLECULAR BIOLOGY, vol. 229, 1993, pages 105 - 124
VILELLA-BACH, M.; NUZZI, P.; FANG, Y.M.; CHEN, J.: "The FKBP12-rapamycin-binding domain is required for FKBP12-rapamycin-associated protein kinase activity and G1 progression", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 274, 1999, pages 4266 - 4272
WALLER, J.R.; NICHOLSON, M.L.: "Molecular mechanisms of renal allograft fibrosis", BRITISH JOURNAL OF SURGERY, vol. 88, 2001, pages 1429 - 1441
WARD, D.E.; ROSS, RP.; VAN DER WEIJDEN, C.C.; SNOEP, J.L.; CLAIBORNE, A.L.: "Catabolism of branched-chain .alpha.-keto acids in Enterococcus faecalis: the bkd gene cluster, enzymes, and metabolic route", J. BACT., vol. 181, no. 17, 1999, pages 5433 - 5442
WARNER, L.M.; ADAMS, L.M.; CHANG, J.Y.; SEHGAL, S.N.: "A modification of the in vivo mixed lymphocyte reaction and rapamycin's effect in this modeI.Cin", IMMUNOL. IMMUNOPATHOL., vol. 64, no. 3, 1992, pages 242 - 7
WEI, X.; LIANG, Y.; ZHENG, Y.: "Enhancement and Selective Production of Oligomycin through Inactivation of Avermectin's Starter Unit in Streptomyces avermitilis", BIOTECHNOLOGY LETTERS, vol. 28, no. 12, 2006, pages 911 - 916
WILKINSON, B.; FOSTER, G.; RUDD, B.A.M.; TAYLOR, N.L.; BLACKABY, A.P.; SIDEBOTTOM, P.J.; COOPER, D.J.; DAWSON, M.J.; BUSS, A.D.; G: "Novel octaketide macrolides related to 6- deoxoerythronolide B provide evidence for iterative operation of the erythromycin polyketide synthase", CHEMISTRY & BIOLOGY, vol. 7, 2000, pages 111 - 117
WU, K.; CHUNG, L.; REVILL, W.P.; KATZ, L.; REEVES, C.D.: "The FK520 gene cluster of Streptomyces hygroscopicus var. ascomyceticus (ATCC 14891) contains genes for biosynthesis of unusual polyketide extender units", GENE, vol. 251, 2000, pages 81 - 90
YEM, A.W.; TOMASSELLI, A.G.; HEINRIKSON, R.L.; ZURCHER-NEELY, H.; RUFF, V.A.; JOHNSON, R.A.; DEIBEL, M.R.: "The Hsp56 component of steroid receptor complexes binds to immobilized FK506 and shows homology to FKBP-12 and FKBP-13", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 267, 1992, pages 2868 - 2871
YU, K.; TORAL-BARZA, L.; DISCAFANI, C.; ZHANG, W.G.; SKOTNICKI, J.; FROST, P.; GIBBONS, J.J.: "mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer", ENDOCRINE-RELATED CANCER, vol. 8, 2001, pages 249 - 258
ZHU, J.; WU J.; FRIZELL, E.; LIU, S.L.; BASHEY, R.; RUBIN, R.; NORTON, P.; ZERN, M.A.: "Rapamycin inhibits hepatic stellate cell proliferation in vitro and limits fibrogenesis in an in vivo model of liver fibrosis", GASTROENTEROLOGY, vol. 117, no. 5, 1999, pages 1198 - 204

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011068341A2 (en) * 2009-12-03 2011-06-09 (주)제노텍 Gene and protein for biosynthesis of tricyclocompounds
WO2011068341A3 (en) * 2009-12-03 2011-12-22 (주)제노텍 Gene and protein for biosynthesis of tricyclocompounds
US20130230559A1 (en) * 2010-08-24 2013-09-05 Ewha University - Industry Collaboration Foundatio Novel tacrolimus analogues, a neuroprotective composition comprising the same, an immunosuppressive composition comprising the same, a method for preparing the same, and a mutant for producing the same
US9505779B2 (en) * 2010-08-24 2016-11-29 Intron Biotechnology, Inc. Tacrolimus analogues, a neuroprotective composition comprising the same, an immunosuppressive composition comprising the same, a method for preparing the same, and a mutant for producing the same
WO2012089349A3 (en) * 2010-12-31 2012-12-27 Acies Bio D.O.O. Novel polyketide compounds and methods of making same
CN114591286A (en) * 2021-04-12 2022-06-07 南京大学 Novel macrolide compound acautalides A-C and preparation method and application thereof

Also Published As

Publication number Publication date
GB0914589D0 (en) 2009-09-30

Similar Documents

Publication Publication Date Title
AU2005270989B2 (en) 17-desmethylrapamycin and analogues thereof, methods for their production and their use as immunosupressants, anticancer agents, antifungal agents, etc.
JP5789190B2 (en) New gene cluster
EP1589031B1 (en) Production of polyketides and other natural products
WO2010004304A1 (en) Novel compounds and methods for their production
US8030325B2 (en) Biosynthetic gene cluster for the production of a complex polyketide
WO2011021036A1 (en) Polyketides analogues and methods for their production
EP2658855A2 (en) Novel polyketide compounds and methods of making same
WO2010106366A1 (en) Fk506 and fk520 analogues and their pharmaceutical uses
US20100022766A1 (en) Biosynthetic gene cluster for the production of a complex polyketide
WO2011045594A1 (en) Novel macrocycles and methods for their production
WO2011045593A1 (en) Novel macrocyles and methods for their production

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10747258

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10747258

Country of ref document: EP

Kind code of ref document: A1