WO2011045593A1 - Novel macrocyles and methods for their production - Google Patents

Novel macrocyles and methods for their production Download PDF

Info

Publication number
WO2011045593A1
WO2011045593A1 PCT/GB2010/051713 GB2010051713W WO2011045593A1 WO 2011045593 A1 WO2011045593 A1 WO 2011045593A1 GB 2010051713 W GB2010051713 W GB 2010051713W WO 2011045593 A1 WO2011045593 A1 WO 2011045593A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compounds
formula
pharmaceutically acceptable
diseases
Prior art date
Application number
PCT/GB2010/051713
Other languages
French (fr)
Inventor
Steven James Moss
Matthew Alan Gregory
Ming-Qiang Zhang
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 WO2011045593A1 publication Critical patent/WO2011045593A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Definitions

  • the present invention relates to novel 31 -desmethoxy FK506 and FK520 ester analogues with potent local anti-inflammatory activity, but substantially reduced systemic activity.
  • These analogues are effective for therapeutic indications requiring potent local, but reduced systemic activity, such as pulmonary diseases (e.g. Asthma), gastrointestinal diseases (e.g. Inflammatory Bowel Disease), ocular diseases (e.g. Uveitis) and skin diseases (e.g.
  • 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), Sirepfomyces sp. MA6858, 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 ei a/., 1997).
  • PPS polyketide synthases
  • FK520 has a C-21 ethyl substituent
  • FK506 has a C-21 allyl substituent.
  • the biosynthetic pathway common to all known naturally produced FK506 and FK520 analogues leads to the necessity for an oxygen bound to the carbon at the C31 position.
  • 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 ⁇ , Na-K- ATPase and nitric oxide synthase, which may lead to some of the side-effects (Kapturczak et a/., 2004).
  • Treatment with FK506 may also be associated with up-regulation of transforming growth factor beta (TGF- ⁇ ).
  • TGF- ⁇ transforming growth factor beta
  • 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.
  • 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 (Elidel® cream) and FK506 (Protopic® ointment) 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).
  • topical formulations such as ointments and creams
  • ointments and creams treatments for a variety of skin conditions, in particular atopic dermatitis
  • atopic dermatitis Nghiem et al., 2002.
  • the FDA required a black box warning on both of these topical treatments, due to concerns over a risk of cancer (Rustin, 2007).
  • 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).
  • MDR-1 P-glycoprotein pump
  • the complexity of FK506 dosing is therefore enhanced by significant drug-drug interactions, variable metabolism and permeability (Tamura et al., 2003; Kapturczak et al., 2004; Izuishi et al., 1997).
  • the mechanism of toxicity of FK506 and FK520 has been related to the mechanism of action of immunosuppression (Dumont et al., 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 (Sigal 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.
  • Inhaled FK506 has been tested in humans and animal models for pulmonary obstructive diseases such as asthma by Astellas/Fujisawa (WO2005/063242). In human trials, pulmonary function was claimed to be better than placebo, but with a slightly higher number of adverse events (42% vs 37%) (Shimizu, 2005).
  • Drug transporters such as Pgp are expressed at the epithelium of the lung, and are thought to be a functionally important element in reducing tissue access of inhaled xenobiotics.
  • inhaled products which are substrates for Pgp would be anticipated to have reduced or nonlinear adsorption pharmacokinetics (Lechapt-Zalcman et al., 1997; Campbell et al., 2003). This is a concern with FK506, which is widely known to be a substrate for Pgp (Saeki et al., 1993).
  • calcineurin inhibitors such as FK506 could have potential benefits for the treatment of patients with steroid-dependent bronchial asthma, enabling either reduction or withdrawal of steroids (Sano et al., 1995). Indeed, in in vitro studies, both FK506 and cyclosporine have been shown to inhibit stimulated T-lymphocytes from both glucocorticoid resistant and sensitive patients (Corrigan et al., 1996).
  • Ester analogues of FK520 have been generated as part of a wide series of semisynthetic FK520 analogues for use as systemic immunosuppressants, with the aim to reduce direct toxicity and present structural variation at the FKBP/calcineurin interface (Wagner et al., 1998; US5.731 .320; US5.643.918; US5.563.172). These documents purport to disclose compounds containing two hydrogen atoms bound to the C-31 carbon, however such compounds cannot be considered enabled. In fact biosynthetic methods available at the time of the disclosures only permitted access to compounds having a hydroxyl at the C-31 position (or a derivative thereof).
  • novel FK506 and FK520 analogues in particular those with reduced systemic toxicity, which may have utility for the treatment of inflammatory conditions.
  • the present invention discloses novel FK506 and FK520 analogues which have reduced systemic activity compared with the currently available FK506 and FK520 analogues.
  • novel FK506 and FK520 analogues may 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, uveitis, psoriasis 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 systemic bioavailability, decreased oral bioavailability, high plasma protein binding, reduced efflux by Pgp and similar transporters, leading to improved permeability, improved lung pharmacokinetics and exposure (for example as shown by an improved lung:blood ratio after aerosol dosing), reduced CyP450 inhibition and metabolism, leading to fewer drug-drug interactions, improved formulation ability, improved potency, increased FKBP binding, improved toxicological profile, reduced nephrotoxicity and neurotoxicity, improved crystallinity or improved lipophilicity.
  • therapies for inflammatory disorders such as atopic dermatitis, asthma, uveitis, psori
  • the present invention provides a strategy for generating anti-inflammatory drugs with potent local activity, but reduced systemic toxicity (via rapid systemic inactivation).
  • the previously undescribed beneficial properties of 31 -desmethoxy FK506 and FK520 analogues, as compared with compounds having a 31 -methoxy group (which includes their improved ADME properties, such as a reduction in efflux by drug transporters, leading to greater permeability, and a reduction in P450 metabolism, leading to reduced drug-drug interactions) which the inventors have discovered are combined with an ester linked chain in the C-32 position which is rapidly cleaved by esterases to give the acid metabolite, which has reduced activity, leading to a reduction in systemic toxicity.
  • the present invention provides analogues of FK506 and FK520 which are lacking the methoxy group at C-31 via a biosynthetic change, and then have an ester linked group attached to the C32 hydroxyl group.
  • the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues according to the formula (I) below, or a pharmaceutically acceptable salt thereof:
  • Ri represents aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl
  • R 3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl;
  • Ri and R 3 are linked to form a lactone ring having 5 to 7 ring members which may optionally contain one or more additional heteroatoms selected from O, N and S.
  • the invention embraces all stereoisomers of the compounds defined by structure (I) as shown above.
  • the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use as a pharmaceutical.
  • the present invention provides processes for production of 31 - desmethoxy FK506 and FK520 ester analogues defined by formula (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).
  • 31 -desmethoxy FK506 and FK520 ester analogues refer to esters of compounds related to FK506, FK520 and similar compounds in structure, but with the 31 methoxy group replaced by a hydrogen atom.
  • 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 atom that is allylic to a double bond that is exo to the main 22-membered ring.
  • Such compounds include, without limitation, 31 - desmethoxy FK520 esters and 31 -desmethoxy FK506 esters 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.
  • 31 -desmethoxy FK506 or FK520 producing strain refers to a recombinant strain based on a natural FK506 or FK520 producing strain which is capable of producing one or more 31 -desmethoxy 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.
  • 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.
  • 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.
  • Alkyl groups may be straight chain or branched. Unless indicated otherwise, alkyl groups may be C1 -8alkyl, e.g. C1 -6alkyl, e.g. C1 -4 alkyl. Examples of C1 -4 alkyl groups include Me, Et, n-Pr, i-Pr, n-Bu, -CH 2 CHMe 2 (isobutyl) and CHMeCH 2 Me.
  • Alkoxy means -Oalkyl
  • Aryl groups include carbocyclic rings which may be mono or bicyclic and contain at least one aromatic ring. Apart from the aromatic ring any other ring may be unsaturated or partially or fully saturated. Typically aryl groups have 6 to 10 ring members. Examples of aryl groups include fully aromatic ring systems such as phenyl, naphthyl and partially aromatic ring systems such as indane, indene and tetralin. The preferred aryl group is phenyl.
  • Heteroaryl groups are aryl groups containing one or more (e.g. 1 to 4 such as 1 to 3 e.g. 1 or 2) hetero atoms selected from O, N and S. Typically heteroaryl groups have 5 to 10 ring members. Examples of monocyclic heteroaryl groups include 5 membered rings such as pyrazole, pyrrole, furan, thiophene, thiazole and 6 membered rings such as pyridine and pyrimidine. Examples of bicyclic heteroaryl groups include fully aromatic ring systems such as quinoline, isoquinoline, indole, isoindole and benzofuran and partially aromatic ring systems such chromane and tetrahydroquinoline.
  • monocyclic heteroaryl groups include 5 membered rings such as pyrazole, pyrrole, furan, thiophene, thiazole and 6 membered rings such as pyridine and pyrimidine.
  • Examples of bicyclic heteroaryl groups include fully aromatic
  • Aryl and heteroaryl groups may optionally be substituted by one or more (e.g. one to three) groups e.g. selected from halogen, hydroxyl, oxo, C1 -4alkyl, -COC1 -3alkyl, -N0 2 , -CN, - NH 2 , -NHC1 -4alkyl, N(C1 -4alkyl) 2 , -NHCOC1 -3alkyl and C1 -4alkoxy.
  • groups e.g. selected from halogen, hydroxyl, oxo, C1 -4alkyl, -COC1 -3alkyl, -N0 2 , -CN, - NH 2 , -NHC1 -4alkyl, N(C1 -4alkyl) 2 , -NHCOC1 -3alkyl and C1 -4alkoxy.
  • the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues, as set out above, methods for the preparation of these compounds, methods for the use of these compounds in medicine and the use of these compounds as intermediates or templates for further derivatisation.
  • R-i represents C1 -10 alkyl e.g. C1 -6 alkyl without any optional replacement of a carbon atom by a heteroatom.
  • R-i and/or R 3 represents C1 -10 alkyl (e.g. C1 -6 alkyl) wherein one or more carbon atoms are optionally replaced by a heteroatom selected from O, N and S
  • resultant groups include ethers and thioethers in which a CH 2 within an alkylene chain is replaced by O or S respectively and alcohols in which a terminal CH 3 is replaced by OH.
  • Amines may also be formed following replacement of -CHR- with-NR- or -CHRR' with -NRR'. Formation of unstable groups (e.g. -0-CH 2 -0-) should be avoided.
  • substitution with halogen atoms include substitution with F atoms e.g. replacement of CH 3 with CF 3 .
  • R-i represents C1 -4alkylaryl or C1 -4alkylheteroaryl, suitably it represents C1 - 2alkylaryl or C1 -2alkylheteroaryl e.g. -CH 2 aryl or -CH 2 heteroaryl.
  • R 3 represents C1 -4alkylaryl or C1 -4alkylheteroaryl, suitably it represents C1 - 2alkylaryl or C1 -2alkylheteroaryl e.g. -CH 2 aryl or -CH 2 heteroaryl.
  • Ri and R 3 may represent -(CH 2 ) 2-3 - thereby to form a 5-6 membered lactone ring.
  • Ri represents C1 to C4 alkyl.
  • R 3 represents H
  • R-i represents methyl
  • R 3 represents H as shown in the following structure:
  • R-i represents ethyl
  • R 3 represents H as shown in the following structure:
  • R-i represents isopropyl
  • R 3 represents H as shown in the following structure:
  • R-i represents n-propyl
  • R 3 represents H as shown in the following structure:
  • R-i represents isobutyl
  • R 3 represents H as shown in the following structure:
  • R-i represents n-butyl
  • R 3 represents H as shown in the following structure:
  • the invention embraces all stereoisomers of the compounds defined by formula (I) as shown above.
  • the present invention provides processes for production of 31 - desmethoxy FK506 or FK520 ester analogues defined by formula (I) above by culturing a 31 - desmethoxy FK506 or FK520 analogue producing strain, optionally with feeding and optionally isolating the compounds produced, then semisynthetically adding an ester group to the C32 hydroxyl.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable diluents or carriers.
  • the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants.
  • the present invention provides use of 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants.
  • the present invention provides a method for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases.
  • the present invention provides use of 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases.
  • the present invention provides a method for the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • 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.
  • salt forms can be converted by treatment with an appropriate base into the free base form.
  • the invention embraces compounds of formula (I) or a pharmaceutically acceptable salt thereof in the form of a pharmaceutically acceptable solvate, such as a hydrate.
  • Compounds of formula (I) may be produced either by total synthesis or by semi- synthesis from a compound of formula (II). Suitable analogues for semi-synthesis include 31 - desmethoxy FK506, 31 -desmethoxy FK520 or 31 -desmethoxy FK523.
  • Suitable analogues for semi-synthesis include 31 - desmethoxy FK506, 31 -desmethoxy FK520 or 31 -desmethoxy FK523.
  • (II) can be synthesised by total synthesis or by fermentation.
  • the producing organism can be supplemented with a suitable acid, or acid derivative, that will be incorporated by the biosynthetic machinery for FK506 or FK520 production in competition with the natural starter unit.
  • a suitable acid or acid derivative is 4- hydroxycyclohexanecarboxylic.
  • An alternative method is by genetic manipulation of the FK506 or FK520 producing organisms so that the the biosynthesis of the natural starter unit is disrupted.
  • the resultant strain can then be supplemented with a suitable acid, or acid derivative, including 4-hydroxylcyclohexanecarboxylic acid. A method for doing so is described in WO2004/007709.
  • Compound of formula (II) produced by fermentation 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 ai, 1998).
  • 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.
  • the compounds of formula (II) can then be derivatised by further chemical synthetic steps, by methods known to one skilled in the art (for example see March, Wiley Interscience) to compounds of formula I.
  • the 32-hydroxyl group of compounds of formula (II) is modified by reaction with a suitable compound of formula (III).
  • the etherification can be achieved by many methods known to one skilled in the art but preferably by reaction with a compound of formula
  • a deprotection step may be required, either in the formation of the compound of formula (III) or once it has been coupled to a compound of formula (II) in order to generate a compound of formula I).
  • R 3 is not H then the prerequisite bromoacyl bromide or alkyl 2- bromoalkanoate may be commercially available or may be prepared by methods known to the skilled person.
  • compounds of formula (I) may be prepared by reacting a corresponding compound of formula (I) in which R-i represents H and reacting it with an alcohol of formula R OH under ester forming conditions (e.g. acid or base catalysis).
  • Compounds of formula (I) are useful as pharmaceuticals for example, but without limitation, having potential as agents for the treatment of inflammatory diseases, in particular, those where local action is more relevant than systemic activity, such as the lung (eg asthma, COPD), gastrointestinal tract (eg Inflammatory Bowel Disease), eye (eg Uveitis) or skin (eg psoriasis, atopic dermatitis).
  • inflammatory diseases in particular, those where local action is more relevant than systemic activity, such as the lung (eg asthma, COPD), gastrointestinal tract (eg Inflammatory Bowel Disease), eye (eg Uveitis) or skin (eg psoriasis, atopic dermatitis).
  • 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 topical treatment of autoimmune diseases or inflammatory disorders.
  • 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 - eg Kahan et al., 1991 ; Allografts - eg Kirchner et al. 2000; Autoimmune / Inflammatory / Asthma - eg Carlson, R.P. et al., 1993; Diabetes I - Rabinovitch, A. et al., 2002; Psoriasis - Reitamo, S. et al., 2001 ; Rheumatoid arthritis - Foey, A., et al., 2002; Fibrosis - eg Gregory et al. 1993.
  • 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, py
  • 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
  • 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.
  • 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. 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
  • 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 powder) delivered as a dry powder or in an aerosol formulation or in the form of atomized solutions or suspensions.
  • a powder e.g. micronized powder
  • Exemplary dry powder inhalers include DISKUS, TURBUHALER, DISKHALER and
  • the aerosol formulation may comprise the drug suspended or dissolved in a suitable pressurized propellant (e.g. HFA134a and/or HFA227).
  • a suitable pressurized propellant e.g. HFA134a and/or HFA227).
  • Other equipment such as nebulizer or inhaler may be used (eg see Chan et al., 2003; Smyth et al., 2003; Dalby et al., 2003; Corcoran et al., 2006; Cryan et al., 2007; WO04/1 10335).
  • 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.
  • analogues eg see US 6,352,998; US6,598,153;
  • 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 or 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.
  • R 3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl.
  • Suitable and preferred x, R 2 and R 3 groups are as described above for compounds of formula (I).
  • Compounds of formula (IA) may be used as intermediates in the preparation of compounds of formula (I).
  • Compounds of formula (IA) are also produced by hydrolysis in vivo following administration of compounds of formula (I) therefore it may be useful to monitor the presence of compounds of formula (IA) during therapy.
  • Isolated compound of formula (IA) in various concentrations may therefore be used a reference standard for comparison of levels in the plasma (or urine or other body fluid) of treated subjects.
  • Figure 6 Plot of data from LCMS analysis of blood samples from pharmacokinetic study in male CD1 mice after (A) p.o. dosing at 10mg/kg and (B) i.v. dosing at 1 mg/kg with compounds 7, 8, 9 and 10 and FK506, 1.
  • Figure 7 Plot of data from LCMS analysis of samples ofter incubation of compounds 7, 9 and
  • Figure 8 Plot of data from analysis of (A) lung samples and (B) blood samples from pharmacokinetic study in male CD1 mice after aerosol dosing (20mg to groups of 9 mice over 10 minutes) with compounds 7, 9 and FK506, 1.
  • Vector pUC19 was obtained from New England Biolabs.
  • Cosmid Supercos-1 was obtained from Stratagene.
  • Vector pKC1 139B01 was obtained by inserting a linker into pKC1 139 (Bierman et al., 1992). The 674bp BglW PvuW fragment of pKC1 139 was replaced by the annealing product of oligos B01 and B02 to give the plasmid pKC1 139B01 (5789bp) containing the polylinker below:
  • Escherichia coli DH10B (GibcoBRL) and £ coli JM1 10 (New England Biolabs) were grown in 2xTY medium as described by Sambrook et al. (2001 ).
  • £. 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).
  • £ 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 10mM MgS0 4 for transfection. £ coli transformants were selected for with ampicillin (100 mg/L), kanamycin (50 mg/L), apramycin (50 mg/L).
  • 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 °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-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were
  • Vegetative cultures 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 °C,
  • BIOT-4081 , BIOT-4168, BIOT-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were 5 recovered on plates of MAM, ISP4, ISP3 or ISP2 and incubated for 5 - 21 days at 28 °C.
  • Vegetative cultures 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 °C, 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
  • BIOT-4081 BIOT-4168, BIOT-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were prepared after growth on MAM, ISP4, ISP3 or ISP2 agar medium, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 medium and incubated for 5-21 days at 28 °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 °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 °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 Elix 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 °C, 15 min.
  • the medium is adjusted to pH 7.0 with NaOH, and then sterilised by autoclaving 121 °C, 15 min.
  • MES is added to PYDG (PYDG + MES) it is added 21 .2 g/L prior to pH adjustment.
  • 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 °C, 15 min.
  • the medium is adjusted to pH 7.0 with NaOH, and then sterilised by autoclaving 121°C, 15 minutes.
  • the medium is adjusted to pH 6.4 with NaOH, and then sterilised by autoclaving 121°C, 15 minutes.
  • 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 pyrosequencing technology (Margulies et al., 2005) at Cogenics and the University of Cambridge.
  • 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., 1999) 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 ⁇ LB Amp100 Kan50 per well) at 37 °C and frozen at -80 °C after mixing wells with 50 ⁇ LB/glycerol 1 :1 .
  • a DIG labeled fkbO probe was used to detect cosmids containing this region of the FK506 biosynthetic cluster.
  • the probe was prepared by PCR using DIG labeled dNTP mix (Roche). It comprises 410 bp of 3'-terminal fkbO sequence. Sequence information for primer design had been obtained by 454 sequencing of BIOT-31 19 - see Figure 1 and SEQ ID NO: 1 . Primer sequences were:
  • UES2for (SEQ ID NO: 2) 5'-CACTCCTTCGATCTCCACGAGCAGGTCGCCACGGGC-3' and UES2rev (SEQ ID NO: 3) 5'-ACCCTGCCGTCCTCACGGCACACCACTACCCCACGG-3'.
  • Annealing temperatures between 66 and 71 °C and extension for 20 sec at 68 °C proved to be successful.
  • Escherichia coli ET12567 (pUZ8002) (Macneil et al., 1992, Paget et al., 1999) was transformed with pKC1 139B01 -derived plasmids by electroporation to generate the £ coli donor strains for spore conjugation (Kieser et al., 2000). Fresh spores were harvested in water from plates of Streptomyces hygroscopicus (BIOT-4168) or Streptomyces tsukubaensis (BIOT- 4206). Spore suspensions were heat-shocked at 50 °C for 10 min.
  • 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 ⁇ 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 HP1 100 equipped with a Hyperclone ODS2,
  • LC samples that have been spiked with 0.005 mg/mL pimecrolimus were analysed on the same instrument and with the same chromatographic conditions.
  • All FK520 and FK506 analogues can be quantified in this manner, for example, 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, pimecrolimus. 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, HMQC and HMBC
  • 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.
  • Method B Xbridge, C18, 3.5 micron 4.6 ⁇ 150 mm column (Waters). Oven 40°C, A: 0.1 % formic acid, B: 0.1 % formic acid in MeCN. 1 ml_/min; 0-2 min 10% B; 15 min 100% B; 17 min 100% B; 17.1 min 10% B, 20 min 10% B.
  • Method C Xbridge, C18, 3.5 micron 4.6 ⁇ 50 mm column (Waters). Oven 40°C, A: 0.01 % ammonium hydroxide, B: MeCN. 1 .7 mL/min; 0 min 5% B; 1 .5 min 95% B
  • Method D Ultimate AQ-C18 (2.1 x 50mm, 3 micron).
  • Method E Ultimate AQ-C18 (2.1 x 50mm, 3 micron).
  • Method F Ultimate AQ-C 18 (2.1 x 50mm, 3 micron).
  • NFAT-bla Jurkat cells were thawed and resuspended in Assay Media (OPTI-MEM, 0.5% dialyzed FBS, 0.1 mM NEAA, 1 mM Sodium Pyruvate, 100 U/mL/100 g/mL Pen/Strep) to a concentration of 781 ,250 cells/mL.
  • 4 ⁇ _ of a 10X serial dilution of the test compounds were added to appropriate wells of a TC-Treated assay plate.
  • 32 ⁇ _ of cell suspension was added to the wells and pre- incubated at 37°C/5% C0 2 in a humidified incubator with test compounds for 30 minutes.
  • Anti CD4:CD8 activator at the pre-determined EC80 concentration was added to wells containing the test compounds. The plate was incubated for 5 hours at 37°C/5% C0 2 in a humidified incubator. 8 ⁇ _ of 1 ⁇ Substrate Loading Solution was added to each well and the plate was incubated for 2 hours at room temperature. The plate was read on a fluorescence plate reader.
  • Water solubility may be tested as follows: A 10 mM stock solution of the FK506 or FK520 analogue is prepared in 100% DMSO at room temperature. Triplicate 0.01 mL aliquots are made up to 0.5 mL with either 0.1 M PBS, pH 7.3 solution or 100% DMSO in amber vials. The resulting 0.2 mM solutions are shaken, at room temperature on an IKA® vibrax VXR shaker for 6 h, followed by transfer of the resulting solutions or suspensions into 2 mL Eppendorf tubes and centrifugation for 30 min at 13200 rpm. Aliquots of the supernatant fluid are then analysed by the LCMS method as described above. Assessment of mouse plasma and human blood stability
  • 90 ⁇ _ of prewarmed blood or plasma was then added to each of the wells of plate B.
  • Triplicate 40 ⁇ _ aliquots were then removed immediately for the 0 h timepoint, then sequentially at 0.25, 0.5, 2, 8 and 24 hours after adding the blood or plasma, and transferred to the wells in plate A and mixed.
  • 80 ⁇ _ of 50% MeOH in dH 2 0 was added to each of the wells in plate A, mixed, then 380 ⁇ _ of pimecrolimus in acetonitrile (100ng/ml_) was added and mixed. Plate A was then kept at -70°C until analysis.
  • plate A was thawed, vortexed for 10 minutes at 300rpm in a shaker, then centrifuged at 4000rpm for 15 minutes. The supernatant was transferred to a new 96-well plate for LC/MS analysis (using method F).
  • mice Female BALB/c mice aged between 5 and 8 weeks were obtained from Harlan Olac (Shaw's Farm, Blackthorn, Bicester, Oxon, 0X25 1 TP, England). Animals were sensitised intra-peritoneally (i.p.) with 10C ⁇ g OVA in alum on day 0 to skew their immune profiles towards Th2. On day 7 animals were terminated by cervical dislocation and spleens were removed to ice-cold HBSS containing 10% HEPES.
  • Spleens were processed under aseptic conditions to obtain a single cell suspension. Cells were then pooled, washed, the red blood cells lysed and a cell count performed to determine cell numbers and viability. Cells were resuspended at a density of 2x10 7 /ml in RPMI with supplements. Two million cells were removed prior to culture for flow cytometry to assess levels of T and B lymphocytes present. This was determined using fluorescently conjugated antibodies to Thy1 .2 & B220 (CD45R). The remaining cells were cultured in triplicate in 96-well tissue culture plates with inhibitors at 3 concentrations (as determined by client) for 1 hour prior to the addition of OVA to the cultures (final concentration 200 ⁇ g ml).
  • Test compounds were reconstituted from the powdered form by addition of 100% DMSO to achieve concentrated stock solutions of equivalent molarity, based on the molecular weights provided for each compound. Compounds were then diluted 1 ,000 fold in culture media, resulting in a background level of 0.1 % DMSO in all wells. Subsequent 10-fold dilution of compounds was performed in RPMI/0.1 % DMSO. The final concentrations of test compounds in the wells were 2nM, 0.2nM & 0.02nM. Cells were incubated at a density of 106 per well. Positive control cultures were stimulated with a polyclonal activator of T cells (con A, 5 ⁇ g ml), in order to confirm the health of the starting population. All cultures were incubated for 72 hours at 37°C, 5% C0 2 . Supernatants were then removed and frozen down for subsequent measurement of key cytokines, IL-5 and IL-13, by Luminex.
  • microsphere was quantified using a Luminex machine (dual laser flow analyzer) and xPonent software (Luminex corporation).
  • a standard curve of quantified recombinant cytokines ranging from 3.2 - 10,000pg/ml, was used to convert detected PE fluorescence values to cytokine concentrations (pg/ml). All standards were run in duplicate and at least 50 microspheres are analysed per cytokine.
  • the cytokines present in the supernatants taken from spleen cultures were determined in each of the triplicates by comparison to the values observed with the recombinant cytokine standards. The cytokine concentration in each sample and the mean of replicates was noted. Cytokine production from splenocytes after OVA stimulation in vitro was compared in the presence and absence of inhibitors.
  • Blood was collected into standard lithium-heparin vacutainers from one healthy donor and subsequently diluted 1 :5 with RPMI media. The donor was not atopic and was not receiving any medication.
  • PBMCs were separated from whole blood using the AccuSpin System utilising
  • PBMCs Histopaque 1077 (Sigma). Following the appropriate washing steps, PBMCs were counted and then divided into relevant fractions for CD3/CD28 stimulation. Compounds were dissolved in DMSO, and diluted further in RPMI media to the working concentration. Final DMSO in the assay was 0.1 % in all samples.
  • PBMCs were diluted in RPMI containing 10% dialysed FBS to yield a concentration of 100,000 cells per 180 ⁇ .
  • anti-CD28 antibody (Calbiochem) was added to yield a final concentration of 2 ⁇ g ml in the assay and 180 ⁇ of this cell suspension was immediately added to the plates.
  • the plates were then placed in a humidified 37°C incubator for 24 hours. At this time, the plates were centrifuged at 400 x g for 5 minutes and 100 ⁇ supernatant transferred to clean 96 well plates. Supernatants were frozen rapidly at -80oC and stored until required for cytokine analysis by Bio-Plex assay.
  • cytokines were analysed by means of a Human Th1/Th2 multiplex assay kit (Bio-Rad) utilising a Bioplex platform. Cell supernatants were thawed on ice before being added directly to the assay plate. The cytokine analysis was carried out as per manufacture's instructions. Standard curves and quantification of samples were generated using Bioplex Manager software (BioRad). Assessment of Pgp efflux There are many methods which may be employed to investigate the rates of efflux by drug transporters such as Pgp. Many of these are reviewed in Szakacs et al., 2008 and
  • CyP450 metabolism and inhibition may be analysed by incubating compounds of interest with isolated hepatocytes, microsomes or isolated CyP450 enzymes, then measuring rate of metabolism of the test compound over a short period of time, such as 0-60 minutes, or using a fluorometric inhibition assay or microsomal inhibition assay to measure P450 inhibition (see Yan and Caldwell, 2001 ).
  • In vivo assays may also be used to measure the bioavailability of a compound.
  • a compound is administered to a test animal (e.g. mouse) both intravenously (i.v.) and orally (p.o.) and blood samples are taken at regular intervals to examine how the plasma (or whole blood) concentration of the drug varies over time.
  • the compounds may be dosed by aerosol using a nebulizer. The time course of plasma (or whole blood)
  • concentration over time can be used to calculate the PK parameters (using software such as Kinetica, Thermo Scientific) such as absolute bioavailability of the compound as a percentage using standard models.
  • the concentration of the compound of the invention or parent compound in the sample was determined via LCMS as follows:20 ⁇ _ of blood:H 2 0 (1 :1 , v/v)/PK sample was added with 20 ⁇ _ Internal standard (pimecrolimus) at 100 ng/mL, 20 ⁇ _ working solution/MeOH and 150 ⁇ _ of ACN, vortexed for 1 minute at 1500 rpm, and centrifuged at 12000 rpm for 5 min. The supernatant was then injected into LC-MS/MS. The time-course of blood concentrations was plotted and used to derive area under the whole blood concentration-time curve (AUC - which is directly proportional to the total amount of unchanged drug that reaches the systemic circulation). These values were used to generate the oral bioavailability (F%) and other PK parameters where possible. Where possible, the levels of the presumed primary metabolite, 11 , were analysed.
  • mice were dosed 20mg over 10 minutes to groups of 9 animals in a chamber using a nebulizer. At 0.25, 0.5, 2, 8, and 24 hours, three mice per timepoint were euthanized under anaesthesia with isofulrane. The animals were placed on ice and the chest cavity opened immediately to expose heart and lung. 100 ⁇ _ of blood sample was collected from each animal by cardial puncture into K 2 EDTA tubes. This blood sample was diluted with the same volume of diH 2 0 and put on dry ice immediately. After collection of the blood sample, lung tissue was rapidly collected, washed with cold saline, dried, weighed and snap frozen with dry ice. The whole process was completed in 2 minutes. Blood and lung samples were then stored at -70°C until analysis.
  • lung tissue was homogenized for 2 min with 3 volumes (v/w) of
  • homogenate/unknown PK sample was added to 30 ⁇ _ internal standard (pimecrolimus,100 ng/mL), 40 ⁇ _ of working solution/MeOH and 160 ⁇ _ of ACN, vortexed for 1 min at 1500 rpm, and centrifuged at 12000 rpm for 5 min. The supernatant was diluted with MeOH (1 :1 , v/v), vortexed and then injected into the LC-MS/MS system. Blood samples were prepared for analysis as described for intravenous and oral pharmacokinetics. The concentration of the relevant compound/s in the lung and blood samples was then determined via LCMS (see materials and methods, using LCMS methods D or E).
  • AUC - area under the whole blood concentration-time curve
  • Example 1 Generation of a Streptomyces hygroscopicus subsp. hygroscopicus strain in which the fkbO gene has been inactivated by introducing a small deletion inducing a frameshift.
  • Oligos SG165 SEQ ID NO: 1
  • SG166 SEQ ID NO: 2
  • Oligos SG165 SEQ ID NO: 1
  • SG166 SEQ ID NO: 2
  • aesFK520A SEQ ID NO: 10
  • DSM 40822 standard PCR reaction
  • genomic DNA Genomic DNA
  • a 5' extension was designed in each oligo to include the restriction sites present in the genomic sequence to aid cloning of the amplified fragment ( Figure 1 ).
  • This 1727 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAESA5 and the insert verified by sequencing.
  • Oligos SG167 SEQ ID NO: 3 and SG168 (SEQ ID NO: 4) were used to amplify a 2034 bp region of DNA, termed aesFK520B (SEQ ID NO: 1 1 ) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase.
  • a 5' extension was designed in oligo SG167 to introduce the restriction site to aid cloning of the amplified fragment ( Figure 1 ) and SG168 included the restriction site present in the genomic sequence.
  • This 2034 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAESBI and the insert verified by sequencing.
  • pAESABI The -1 .7 kb Bgl ⁇ IXho ⁇ fragment from pAESA5 and -2.0 kb Xho ⁇ IEcoR ⁇ fragment from pAESBI were cloned into the -5.9 kb Bgl ⁇ /EcoR ⁇ fragment of pKC1 139 (Bierman et al., 1992) to make pAESABI .
  • pAESABI therefore contained the upstream and downstream regions such that the double crossover event would result in the desired small deletion, introducing a frameshift in the fkbO gene.
  • Escherichia coli ET12567 harbouring the plasmid pUZ8002, was transformed with pAESABI by electroporation to generate the E. coli donor strain for conjugation.
  • This strain was used to transform Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) by spore conjugation (as described in Materials and Methods). Exconjugants were plated on R6 agar and incubated at 28°C.
  • pAESABI is able to self-replicate in Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) at 28°C.
  • Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 28°C to ensure the pAESABI plasmid with resistance marker was present. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured again on to MAM plates with apramycin at 37°C to induce the plasmid to integrate, as the plasmid cannot self-replicate at 37°C. The transformants were then subcultured for four subsequent rounds at 37°C on MAM plates with no antibiotic. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP3 medium at 28°C. Serial dilutions were made from the filtered collected spores and were plated on MAM plates to achieve single colonies.
  • Example 2 Generation of a Streptomyces hygroscopicus subsp. hygroscopicus strain in which the fkbO gene has been inactivated by introducing a large in-frame deletion.
  • 2.1 Cloning of DNA homologous to the upstream flanking region of fkbO disruption.
  • Oligos SG165 SEQ ID NO: 1
  • SG166 SEQ ID NO: 2
  • aesFK520A SEQ ID NO: 10
  • DSM 40822 standard PCR reaction
  • genomic DNA Genomic DNA
  • a 5' extension was designed in each oligo to include the restriction sites present in the genomic sequence to aid cloning of the amplified fragment ( Figure 2).
  • This 1727 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAESA5 and the insert verified by sequencing.
  • Oligos SG169 SEQ ID NO: 5 and SG168 (SEQ ID NO: 6) were used to amplify a 1460 bp region of DNA, termed aesFK520C (SEQ ID NO: 12) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase.
  • a 5' extension was designed in oligo SG169 to introduce the restriction site to aid cloning of the amplified fragment (Figure 2) and SG168 included the restriction site present in the genomic sequence.
  • This 1460 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAESC8 and the insert verified by sequencing.
  • Escherichia coli ET12567 harbouring the plasmid pUZ8002, was transformed with pAESAC4 by electroporation to generate the E. coli donor strain for conjugation.
  • This strain was used to transform Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) by spore conjugation (as described in Materials and methods).
  • Exconjugants were plated on R6 agar and incubated at 28°C.
  • pAESAC4 is able to self-replicate in Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) at 28°C.
  • Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 28°C to ensure the pAESAC4 plasmid with resistance marker was present. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured on to MAM plates with apramycin at 37°C to induce the plasmid to integrate, as the plasmid cannot self-replicate at 37°C. The transformants were then subcultured for four subsequent rounds at 37°C on MAM plates with no antibiotic. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP3 medium at 28°C. Serial dilutions were made from the filtered collected spores and were plated on MAM plates to achieve single colonies.
  • Oligos AES43 (SEQ ID NO: 6) and AES40 (SEQ ID NO: 7) were used to amplify a 2057 bp region of DNA, termed aesFK506D1 1 (SEQ ID NO: 13) from Streptomyces tsukubaensis no. 9993 (FERM BP-927) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase.
  • a 5' extension was designed in each oligo to introduce the restriction sites to aid cloning of the amplified fragment (Figure 3). This 2057 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAES8 and the insert verified by sequencing.
  • Oligos AES41 SEQ ID NO: 8 and AES42 (SEQ ID NO: 9) were used to amplify a 1985 bp region of DNA, termed aesFK506E3 (SEQ ID NO: 14) from Streptomyces tsukubaensis no. 9993 (FERM BP-927) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase.
  • a 5' extension was designed in each oligo to introduce the restriction sites to aid cloning of the amplified fragment ( Figure 3).
  • This 1985 bp fragment was cloned into pUC19 that had been linearised with Sma ⁇ , resulting in plasmid pAES9 and the insert verified by sequencing.
  • Escherichia coli ET12567 harbouring the plasmid pUZ8002, was transformed with pAESI O by electroporation to generate the E. coli donor strain for conjugation.
  • This strain was used to transform Streptomyces tsukubaensis no. 9993 (FERM BP-927) by spore conjugation (as described in Materials and methods).
  • Exconjugants were plated on R6 agar and incubated at 37°C.
  • pAESI O is not able to self-replicate in Streptomyces tsukubaensis no. 9993 (FERM BP-927) at 37°C and must integrate into the genome.
  • Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 37°C two to three times, to ensure that the pAESI O plasmid with resistance marker had integrated. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured for two subsequent rounds at 37°C on MAM plates with no antibiotic and then a final time at 28°C. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP4 medium at 28°C. Serial dilutions were made from the filtered collected spores and plated on ISP4 plates at 28°C to achieve single colonies.
  • BIOT-4254 Spore stocks of BIOT-4254 were recovered onto plates of ISP4 medium and incubated for 2 weeks at 28°C.
  • Vegetative cultures seed culture
  • the fermentation was carried out at 28 °C, with starting agitation at 200 rpm, aeration rate at 0.5 VA /M (7.5 SL/min) and dissolved oxygen (DO) level controlled with the agitation cascade at 30% air saturation.
  • aeration rate at 24 hours post- inoculation 5ml of 0.2 M irans-4-hydroxycyclohexanecarboxylic acid in methanol was added to the vessel.
  • the aeration rate was increased to 0.7 V/V/M (10.5 litres/min) and control ph 6.4 ⁇ 0.8 with 1 .0M NaOH and 0.5 M H 2 S0 4 .
  • the culture was harvested after 93 hours post-inoculation.
  • the fermentation was carried out at 28 °C, with starting agitation at 350 RPM, aeration rate at 0.5 V/V/M (2.5 SL/min) and dissolved oxygen (DO) level controlled with the agitation cascade at 30% air saturation.
  • aeration rate at 0.5 V/V/M (2.5 SL/min)
  • DO dissolved oxygen
  • a second fermentation batch was run as above except: NGY seed medium, PYDG production medium (pre-sterilisation pH adjusted at pH 7.0) and harvested after 1 17 hours.
  • the combined broths were extracted with ethyl acetate (2 x 1 volume equivalent) and the resultant organics reduced in vacuo to a crude extract (16 g).
  • This extract was dissolved in acetonitrile (1000 ml) and silica added (30 g). The solvent was removed in vacuo and the adsorbed silica added to the top of a packed silica column (170 mm x 55 mm diameter).
  • the column was eluted with ethyl actate / hexanes (1 :2, 0.9 L; 1 :1 , 1 L; 3:2, 1 L; 2:1 , 1.8 L) and fractions containing the target compound combined and taken to dryness (190 mg). This enriched extract was then purified by preparative HPLC over C18 to yield the target compound (37 mg).
  • IL-5 and IL-13 a mouse spleen cytokine release assay was used, as described in the materials and methods.
  • mice To confirm that the action of plasma enzymes (presumed to be esterases) was an important part of the rapid metabolism in mice, and to assess the relative rates of metabolism, mouse plasma stability studies were carried out, as described in the materials and methods, by spiking mouse plasma with test compounds at 2 ⁇ , to generate half lives for a series of compounds.
  • rapamycin in Streptomyces hygroscopicus analysis of the enzymatic domains in the modular polyketide synthase.” Gene 169(1 ): 9-16.
  • pristinaespiralis encoding enzymes involved in the biosynthesis of the 4-dimethylamino- L-phenylalanine precursor of pristinamycin I.” Molecular Microbiology 23(2): 191 -202.
  • Streptomyces pristinaespiralis encoding enzymes involved in the conversion of pristinamycin ll B to pristinamycin ll A (PIU): PI synthase and NADH:riboflavin 5'- phosphate oxidoreductase.” Journal of Bacteriology 177(18): 5206-5214.
  • Beta-lactamase an ideal reporter system for monitoring gene

Abstract

There are provided compounds of formula (I) or a pharmaceutically acceptable salt thereof: wherein the variables R1-R3 are as described in the description, said compounds being useful as anti-inflammatory agents with reduced systemic effects.

Description

NOVEL MACROCYLES AND METHODS FOR THEIR PRODUCTION
Field of the Invention
The present invention relates to novel 31 -desmethoxy FK506 and FK520 ester analogues with potent local anti-inflammatory activity, but substantially reduced systemic activity. These analogues are effective for therapeutic indications requiring potent local, but reduced systemic activity, such as pulmonary diseases (e.g. Asthma), gastrointestinal diseases (e.g. Inflammatory Bowel Disease), ocular diseases (e.g. Uveitis) and skin diseases (e.g.
Psoriasis and Atopic Dermatitis).
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), Sirepfomyces sp. MA6858, Streptomyces sp. MA6548, Streptomyces kanamyceticus KCC S-0433, Streptomyces davuligerus CKD1 1 19 (Kim and Park, 2007) which have been shown to produce FK506 (Muramatsu et al., 2005), and Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822), which is equivalent to Streptomyces hygroscopicus var ascomyceticus (ATCC 14891 ), producing FK520 (Garrity 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 ei a/., 1997).
CH=CH2, R2 = trans-OH CH3, R2 = trans-OH H, R2 = trans-OH : R-, = -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 thioesters, or substituted malonyl thioesters, and addition of a lysine derived pipecolate group and cyclization by a non-ribosomal peptide synthetase (NRPS). The structure is then finally 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. The biosynthetic pathway common to all known naturally produced FK506 and FK520 analogues leads to the necessity for an oxygen bound to the carbon at the C31 position.
It has been shown previously, that if fkbO is inactivated in an FK506 or FK520 producing strain, unnatural starter acids, such as hydroxy cyclohexane carboxylic acid, can be fed to the fermentation broth and are incorporated, leading to novel compounds such as 31 - desmethoxy FK506 and 31 -desmethoxy FK520 (WO 2004/007709). Previous to the publication of this patent application, methods for generating 31 -desmethoxy-31 -Hydrogen derivatives of FK506 and FK520 had not been described.
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 ΙκΒ, Na-K- ATPase and nitric oxide synthase, which may lead to some of the side-effects (Kapturczak et a/., 2004).
Treatment with FK506 may also 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 leads to dangers of over (or under) dosing (Roy et al., 2006).
Pimecrolimus (Elidel® cream) and FK506 (Protopic® ointment) 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). On January 19th 2006, the FDA required a black box warning on both of these topical treatments, due to concerns over a risk of cancer (Rustin, 2007).
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, variable metabolism and permeability (Tamura et al., 2003; Kapturczak et al., 2004; Izuishi et al., 1997).
The mechanism of toxicity of FK506 and FK520 has been related to the mechanism of action of immunosuppression (Dumont et al., 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 (Sigal 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 (Bodor and Buchwald, 2001 ; Bodor and Buchwald, 2005).
Inhaled FK506 has been tested in humans and animal models for pulmonary obstructive diseases such as asthma by Astellas/Fujisawa (WO2005/063242). In human trials, pulmonary function was claimed to be better than placebo, but with a slightly higher number of adverse events (42% vs 37%) (Shimizu, 2005).
Drug transporters such as Pgp are expressed at the epithelium of the lung, and are thought to be a functionally important element in reducing tissue access of inhaled xenobiotics. In particular, inhaled products which are substrates for Pgp would be anticipated to have reduced or nonlinear adsorption pharmacokinetics (Lechapt-Zalcman et al., 1997; Campbell et al., 2003). This is a concern with FK506, which is widely known to be a substrate for Pgp (Saeki et al., 1993).
It has been suggested that, due to their different mechanism of action to steroids, calcineurin inhibitors such as FK506 could have potential benefits for the treatment of patients with steroid-dependent bronchial asthma, enabling either reduction or withdrawal of steroids (Sano et al., 1995). Indeed, in in vitro studies, both FK506 and cyclosporine have been shown to inhibit stimulated T-lymphocytes from both glucocorticoid resistant and sensitive patients (Corrigan et al., 1996).
Ester analogues of FK520 have been generated as part of a wide series of semisynthetic FK520 analogues for use as systemic immunosuppressants, with the aim to reduce direct toxicity and present structural variation at the FKBP/calcineurin interface (Wagner et al., 1998; US5.731 .320; US5.643.918; US5.563.172). These documents purport to disclose compounds containing two hydrogen atoms bound to the C-31 carbon, however such compounds cannot be considered enabled. In fact biosynthetic methods available at the time of the disclosures only permitted access to compounds having a hydroxyl at the C-31 position (or a derivative thereof). Specifically, there was no known method for achieving no substitution whatever at the C-31 position (i.e. to produce a methylene group), as the biosynthesis (as it is now understood) requires a carbon-oxygen bond at the C31 position and all of the exemplified compounds have a methoxy at this position, as do FK506 and FK520 (Wallace et al., 1994; Reynolds et al., 1997; Lowden et al., 2001 ). There was also no suggestion as to any benefit that removal of the C-31 methoxy may lend to the final product, for example by increasing tissue (in particular lung) permeability or reduction in drug-drug interactions, nor any suggestion that these compounds might be useful for topical use, with a reduction in systemic activity.
Therefore, there remains a need to identify novel FK506 and FK520 analogues, in particular those with reduced systemic toxicity, which may have utility for the treatment of inflammatory conditions. The present invention discloses novel FK506 and FK520 analogues which have reduced systemic activity compared with the currently available FK506 and FK520 analogues. The novel FK506 and FK520 analogues may 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, uveitis, psoriasis 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 systemic bioavailability, decreased oral bioavailability, high plasma protein binding, reduced efflux by Pgp and similar transporters, leading to improved permeability, improved lung pharmacokinetics and exposure (for example as shown by an improved lung:blood ratio after aerosol dosing), reduced CyP450 inhibition and metabolism, leading to fewer drug-drug interactions, improved formulation ability, improved potency, increased FKBP binding, improved toxicological profile, reduced nephrotoxicity and neurotoxicity, improved crystallinity or improved lipophilicity.
Summary of the Invention
The present invention provides a strategy for generating anti-inflammatory drugs with potent local activity, but reduced systemic toxicity (via rapid systemic inactivation). The previously undescribed beneficial properties of 31 -desmethoxy FK506 and FK520 analogues, as compared with compounds having a 31 -methoxy group (which includes their improved ADME properties, such as a reduction in efflux by drug transporters, leading to greater permeability, and a reduction in P450 metabolism, leading to reduced drug-drug interactions) which the inventors have discovered are combined with an ester linked chain in the C-32 position which is rapidly cleaved by esterases to give the acid metabolite, which has reduced activity, leading to a reduction in systemic toxicity. This gives a series of 31 -desmethoxy FK506 and FK520 esters with potent local activity when dosed directly to organs such as the lung, gastrointestinal tract, eye or skin, which are then rapidly inactivated upon systemic exposure.
Therefore, in a first aspect the present invention provides analogues of FK506 and FK520 which are lacking the methoxy group at C-31 via a biosynthetic change, and then have an ester linked group attached to the C32 hydroxyl group.
In a more specific aspect, the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues according to the formula (I) below, or a pharmaceutically acceptable salt thereof:
Figure imgf000006_0001
wherein R2 represents -CH3, -CH2CH3! or -CH2CH=CH2;
Ri represents C1 -10 alkyl (e.g. C1 -6 alkyl) wherein one or more carbon atoms are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups;
or Ri represents aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl; and
R3 represents a group -(CO)XC1 -10 alkyl wherein one or more carbon atoms of the alkyl chain are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups and x represents 0 or 1 ;
or R3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl;
or Ri and R3 are linked to form a lactone ring having 5 to 7 ring members which may optionally contain one or more additional heteroatoms selected from O, N and S.
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.
The invention embraces all stereoisomers of the compounds defined by structure (I) as shown above.
In a further aspect, the present invention provides 31 -desmethoxy FK506 or FK520 ester 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 processes for production of 31 - desmethoxy FK506 and FK520 ester analogues defined by formula (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 "31 -desmethoxy FK506 and FK520 ester analogues" / "31 - desmethoxy FK506 or FK520 ester analogues" refer to esters of compounds related to FK506, FK520 and similar compounds in structure, but with the 31 methoxy group replaced by a hydrogen atom. 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 atom that is allylic to a double bond that is exo to the main 22-membered ring. Such compounds include, without limitation, 31 - desmethoxy FK520 esters and 31 -desmethoxy FK506 esters 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 "31 -desmethoxy FK506 or FK520 producing strain" refers to a recombinant strain based on a natural FK506 or FK520 producing strain which is capable of producing one or more 31 -desmethoxy 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.
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.
Alkyl groups may be straight chain or branched. Unless indicated otherwise, alkyl groups may be C1 -8alkyl, e.g. C1 -6alkyl, e.g. C1 -4 alkyl. Examples of C1 -4 alkyl groups include Me, Et, n-Pr, i-Pr, n-Bu, -CH2CHMe2 (isobutyl) and CHMeCH2Me.
Alkoxy means -Oalkyl.
Aryl groups include carbocyclic rings which may be mono or bicyclic and contain at least one aromatic ring. Apart from the aromatic ring any other ring may be unsaturated or partially or fully saturated. Typically aryl groups have 6 to 10 ring members. Examples of aryl groups include fully aromatic ring systems such as phenyl, naphthyl and partially aromatic ring systems such as indane, indene and tetralin. The preferred aryl group is phenyl.
Heteroaryl groups are aryl groups containing one or more (e.g. 1 to 4 such as 1 to 3 e.g. 1 or 2) hetero atoms selected from O, N and S. Typically heteroaryl groups have 5 to 10 ring members. Examples of monocyclic heteroaryl groups include 5 membered rings such as pyrazole, pyrrole, furan, thiophene, thiazole and 6 membered rings such as pyridine and pyrimidine. Examples of bicyclic heteroaryl groups include fully aromatic ring systems such as quinoline, isoquinoline, indole, isoindole and benzofuran and partially aromatic ring systems such chromane and tetrahydroquinoline.
Aryl and heteroaryl groups may optionally be substituted by one or more (e.g. one to three) groups e.g. selected from halogen, hydroxyl, oxo, C1 -4alkyl, -COC1 -3alkyl, -N02, -CN, - NH2, -NHC1 -4alkyl, N(C1 -4alkyl)2, -NHCOC1 -3alkyl and C1 -4alkoxy.
Detailed Description of the Invention
The present invention provides 31 -desmethoxy FK506 or FK520 ester analogues, as set out above, methods for the preparation of these compounds, methods for the use of these compounds in medicine and the use of these compounds as intermediates or templates for further derivatisation.
Preferably R2 represents -CH2CH=CH2
Suitably Ri represents C1 -10 alkyl (e.g. C1 -6 alkyl) wherein one or more carbon atoms are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups.
In one embodiment, R-i represents C1 -10 alkyl e.g. C1 -6 alkyl without any optional replacement of a carbon atom by a heteroatom.
When R-i and/or R3 represents C1 -10 alkyl (e.g. C1 -6 alkyl) wherein one or more carbon atoms are optionally replaced by a heteroatom selected from O, N and S, examples of resultant groups include ethers and thioethers in which a CH2 within an alkylene chain is replaced by O or S respectively and alcohols in which a terminal CH3 is replaced by OH. Amines may also be formed following replacement of -CHR- with-NR- or -CHRR' with -NRR'. Formation of unstable groups (e.g. -0-CH2-0-) should be avoided.
Examples of substitution with halogen atoms include substitution with F atoms e.g. replacement of CH3with CF3.
Examples of substitution with =0 groups include replacement of -CH2- with -CO- which carbonyl group may, for example, be part of a ketone, ester or amide functionality depending on the nature of neighbouring atoms.
When R-i represents C1 -4alkylaryl or C1 -4alkylheteroaryl, suitably it represents C1 - 2alkylaryl or C1 -2alkylheteroaryl e.g. -CH2aryl or -CH2heteroaryl.
When R3 represents C1 -4alkylaryl or C1 -4alkylheteroaryl, suitably it represents C1 - 2alkylaryl or C1 -2alkylheteroaryl e.g. -CH2aryl or -CH2heteroaryl.
When R-i and R3 are joined for form a lactone ring, for example Ri and R3 together may represent -(CH2)2-3- thereby to form a 5-6 membered lactone ring.
Preferably Ri represents C1 to C4 alkyl.
Preferably R3 represents H
In one example embodiment, R-i represents methyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000010_0001
In another example embodiment, R-i represents ethyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000011_0001
In another example embodiment, R-i represents isopropyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000011_0002
In another example embodiment, R-i represents n-propyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000011_0003
In another example embodiment, R-i represents isobutyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000012_0001
In another example embodiment, R-i represents n-butyl, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000012_0002
The above structures show 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.
The invention embraces all stereoisomers of the compounds defined by formula (I) as shown above.
In a further aspect, the present invention provides processes for production of 31 - desmethoxy FK506 or FK520 ester analogues defined by formula (I) above by culturing a 31 - desmethoxy FK506 or FK520 analogue producing strain, optionally with feeding and optionally isolating the compounds produced, then semisynthetically adding an ester group to the C32 hydroxyl.
In a further aspect, the present invention provides a pharmaceutical composition comprising a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable diluents or carriers.
In a further aspect, the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants.
In a further aspect, the present invention provides use of 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants.
In a further aspect, the present invention provides a method for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases and/or rejection of transplants which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof.
In a further aspect, the present invention provides 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases.
In a further aspect, the present invention provides use of 31 -desmethoxy FK506 or FK520 ester analogues such as compounds of formula (I) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases.
In a further aspect, the present invention provides a method for the treatment or prevention of atopic dermatitis, asthma, uveitis, psoriasis and inflammatory bowel diseases which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a 31 -desmethoxy FK506 or FK520 ester analogue such as a compound of formula (I) or a pharmaceutically acceptable salt thereof.
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. hydroxylbutanedioic 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.
The invention embraces compounds of formula (I) or a pharmaceutically acceptable salt thereof in the form of a pharmaceutically acceptable solvate, such as a hydrate. Compounds of formula (I) may be produced either by total synthesis or by semi- synthesis from a compound of formula (II). Suitable analogues for semi-synthesis include 31 - desmethoxy FK506, 31 -desmethoxy FK520 or 31 -desmethoxy FK523. Compounds of formula
(II) can be synthesised by total synthesis or by fermentation. To generate compounds of formula (II) by fermentation the producing organism can be supplemented with a suitable acid, or acid derivative, that will be incorporated by the biosynthetic machinery for FK506 or FK520 production in competition with the natural starter unit. A suitable acid or acid derivative is 4- hydroxycyclohexanecarboxylic. An alternative method is by genetic manipulation of the FK506 or FK520 producing organisms so that the the biosynthesis of the natural starter unit is disrupted. The resultant strain can then be supplemented with a suitable acid, or acid derivative, including 4-hydroxylcyclohexanecarboxylic acid. A method for doing so is described in WO2004/007709.
Compound of formula (II) produced by fermentation 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 ai, 1998).
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 , Streptomyces kanamyceticus KCC S-0433, Streptomyces c!avu!igerus CKD1 1 19, 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).
The compounds of formula (II) can then be derivatised by further chemical synthetic steps, by methods known to one skilled in the art (for example see March, Wiley Interscience) to compounds of formula I. The 32-hydroxyl group of compounds of formula (II) is modified by reaction with a suitable compound of formula (III). The etherification can be achieved by many methods known to one skilled in the art but preferably by reaction with a compound of formula
(III) in the presence of a rhodium-acetate catalyst (Wagner et ai 1998). Optionally other reactive centres on compounds of formula (II) may be protected by methods known to one skilled in the art.
Figure imgf000015_0001
(III)
Compounds of formula (III) may be made by many methods known to those skilled in the art. When R3 = H they can be synthesised in two steps from 2-bromoacetyl bromide.
Reaction with the correct alcohol for R-i (e.g. reaction with n-butanol for R-i = n-butyl) yields an alkyl 2-bromoacetate. This can in turn be reacted with Ν,Ν'-ditosyl hydrazine to generate the alkyl diazoacetate. Suitable modification on the alcohol can lead to the diversity in R-i
(optionally a deprotection step may be required, either in the formation of the compound of formula (III) or once it has been coupled to a compound of formula (II) in order to generate a compound of formula I). When R3 is not H then the prerequisite bromoacyl bromide or alkyl 2- bromoalkanoate may be commercially available or may be prepared by methods known to the skilled person. Alternatively, compounds of formula (I) may be prepared by reacting a corresponding compound of formula (I) in which R-i represents H and reacting it with an alcohol of formula R OH under ester forming conditions (e.g. acid or base catalysis).
Compounds of formula (I) are useful as pharmaceuticals for example, but without limitation, having potential as agents for the treatment of inflammatory diseases, in particular, those where local action is more relevant than systemic activity, such as the lung (eg asthma, COPD), gastrointestinal tract (eg Inflammatory Bowel Disease), eye (eg Uveitis) or skin (eg psoriasis, atopic dermatitis). 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 topical 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). 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 - eg Kahan et al., 1991 ; Allografts - eg Kirchner et al. 2000; Autoimmune / Inflammatory / Asthma - eg Carlson, R.P. et al., 1993; Diabetes I - Rabinovitch, A. et al., 2002; Psoriasis - Reitamo, S. et al., 2001 ; Rheumatoid arthritis - Foey, A., et al., 2002; Fibrosis - eg 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. 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. Steiner ei 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 powder) delivered as a dry powder or in an aerosol formulation or in the form of atomized solutions or suspensions.
Exemplary dry powder inhalers include DISKUS, TURBUHALER, DISKHALER and
CLICKHALER. The aerosol formulation may comprise the drug suspended or dissolved in a suitable pressurized propellant (e.g. HFA134a and/or HFA227). Other equipment such as nebulizer or inhaler may be used (eg see Chan et al., 2003; Smyth et al., 2003; Dalby et al., 2003; Corcoran et al., 2006; Cryan et al., 2007; WO04/1 10335).
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. There are many examples in the literature of topical formulations for natural products such as FK506, FK520 and analogues (eg see US 6,352,998; US6,598,153;
US6,673,808; US6,515,016; US6,403,122).
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 or 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.
Also claimed as an aspect of the invention are compounds of formula (IA) or a salt thereof (such as a pharmaceutically acceptable salt):
Figure imgf000021_0001
wherein
R2 represents -CH3, -CH2CH3! or -CH2CH=CH2;
Ri represents H; and
R3 represents a group -(CO)XC1 -10 alkyi wherein one or more carbon atoms of the alkyi chain are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups and x represents 0 or 1 ;
or R3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl.
Suitable and preferred x, R2 and R3 groups are as described above for compounds of formula (I).
An example compound of formula (IA) is a compound in which R-i represents H, R2 represents -CH2CH=CH2 and R3 represents H as shown in the following structure:
Figure imgf000021_0002
above for compounds of formula (I). Suitably the alcohol function represented by Ri = H in a compound of formula (lll)is protected e.g. as an ester by a tnmethylsilylalkylene group or other hydroxyl protecting group which may be removed (e.g. by hydrolysis) following reaction with the compound of formula (II).
Compounds of formula (IA) may be used as intermediates in the preparation of compounds of formula (I). Compounds of formula (IA) are also produced by hydrolysis in vivo following administration of compounds of formula (I) therefore it may be useful to monitor the presence of compounds of formula (IA) during therapy. Isolated compound of formula (IA) in various concentrations may therefore be used a reference standard for comparison of levels in the plasma (or urine or other body fluid) of treated subjects. Brief description of the Figures
Figure 1 : 1H NMR of Compound 7
Figure 2: 1H NMR of Compound 8
Figure 3: 1H NMR of Compound 9
Figure 4: 1H NMR of Compound 10
Figure 5: 1H NMR of Compound 11
Figure 6: Plot of data from LCMS analysis of blood samples from pharmacokinetic study in male CD1 mice after (A) p.o. dosing at 10mg/kg and (B) i.v. dosing at 1 mg/kg with compounds 7, 8, 9 and 10 and FK506, 1.
Figure 7: Plot of data from LCMS analysis of samples ofter incubation of compounds 7, 9 and
10 and FK506, 1 with human whole blood.
Figure 8: Plot of data from analysis of (A) lung samples and (B) blood samples from pharmacokinetic study in male CD1 mice after aerosol dosing (20mg to groups of 9 mice over 10 minutes) with compounds 7, 9 and FK506, 1.
Materials and Methods
Materials
All molecular biology enzymes and reagents were from commercial sources. Vector pUC19 was obtained from New England Biolabs. Cosmid Supercos-1 was obtained from Stratagene. Vector pKC1 139B01was obtained by inserting a linker into pKC1 139 (Bierman et al., 1992). The 674bp BglW PvuW fragment of pKC1 139 was replaced by the annealing product of oligos B01 and B02 to give the plasmid pKC1 139B01 (5789bp) containing the polylinker below:
Primers 5' gatccactagttcggacgcatatgctggatatccaagtatctagaac B01 (SEQ ID NO: 21)
5' gttctagatacttggatatccagcatatgcgtccgaactagtg B02 (SEQ ID NO:
22)
Resulting polylinker
Spel Ndel EcoRV Xbal
5' gatccactagttcggacgcatatgctggatatccaagtatctagaac 3'
3 ' gtgatcaagcctgcgtatacgacctataggttcatagatcttg 5'
Starter materials
All of the feeds used for starting units were obtained from commercial sources. 4-trans- hydroxycyclohexanecarboxylic acid was purchased from TCI Europe.
Bacterial strains and growth conditions
Escherichia coli DH10B (GibcoBRL) and £ coli JM1 10 (New England Biolabs) were grown in 2xTY medium as described by Sambrook et al. (2001 ). £. 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). £ 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 10mM MgS04 for transfection. £ coli transformants were selected for with ampicillin (100 mg/L), kanamycin (50 mg/L), apramycin (50 mg/L).
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 °C.
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 °C.
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 °C 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 °C 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 °C for six days. The culture was then harvested for extraction.
Other actinomycete strains were generated from these available strains using methods described below.
5
Production of FK520, FK506 or analogues in Tubes
Spore stocks of BIOT-4081 , BIOT-4168, BIOT-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were
10 recovered on plates of MAM, ISP4, ISP3 or ISP2 and incubated for 5-21 days at 28 °C. 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 °C,
15 300 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 °C 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. 0
Production of FK520, FK506 or analogues in Flasks
Spore stocks of BIOT-4081 , BIOT-4168, BIOT-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were cultured on MAM, ISP4, ISP3 or ISP2 plates, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were 5 recovered on plates of MAM, ISP4, ISP3 or ISP2 and incubated for 5 - 21 days at 28 °C. 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 °C, 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
30 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 °C 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.
35
Production of FK520, FK506 or analogues in stirred bioreactors Spore stocks of BIOT-4081 , BIOT-4168, BIOT-31 19, BIOT-4206 or strains which are described below, such as BIOT-4131 , BIOT-4132 and BIOT-4254 were prepared after growth on MAM, ISP4, ISP3 or ISP2 agar medium, and preserved in 20% (w/v) glycerol and stored at -80 °C. Spores were recovered on plates of MAM, ISP4, ISP3 or ISP2 medium and incubated for 5-21 days at 28 °C.
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 °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 °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. 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 Elix 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 20 g
No pH adjustment is made. Sterilised by autoclaving 121 °C, 15 min. Medium 9: R6 (Kieser et al., 2000) Component Source per L
Sucrose Fisher 200 g
Dextrin Avedex 10 g
Casamino acids Difco g
MgS04.7H20 Sigma 0.05 g
ISP Trace Element See below 1 mL
Solution
K2S04 Sigma 0.1 g
Water to 700 mL
Add 20 g BACTO agar and a stirrer bar to each 1 L Duran and
autoclave at 121 °C for 15 min.
After autoclaving add:
Component Source per L
(previously sterilised individually
by autoclaving 121 °C, 15 min)
0.65 M L-glutamic acid, mono Sigma 100 mL (10.99 g)
sodium salt
0.48 M CaCI2.2H20 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 20 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 °C, 15 min.
ISP Trace Elements Solution
Component Source per L
FeS04.7H20 Sigma 0.1 g
MnCI2.4H20 Sigma 0.1 g
ZnS04.7H20 Sigma 0.1 g
Distilled water 100
mL Stored in the dark at 4 °C.
2xTY
Component Source per L
Tryptone Difco 16 g
Yeast extract Difco 10 g
NaCI Sigma 5 g
BACTO agar Difco 15 g
Sterilised by autoclaving 121 °C, 15 min.
LB
Component Source per L
Tryptone Difco 10 g
Yeast extract Difco 5 g
NaCI Sigma 10 g
BACTO agar Difco 15 g
Sterilised by autoclaving 121 °C, 15 min.
TSB
Component Source per L
Bacto Tryptic Soy BD 30 g
Broth
Sterilised by autoclaving 121 °C, 15 min.
NGY
Component Source per L
Difco Nutrient Broth Difco 8 g
Glucose Sigma 10 g
Yeast Extract Difco 5 g
The medium is adjusted to pH 7.0, with NaOH and then sterilised by autoclaving 121 °C, 15 min. PYDG
Component Source per L
Peptone from Milk Sigma 15 g
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 °C, 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 10 g
K2HP04 Sigma 1 .0 g
MgS04.7H20 Sigma 1 .0 g
NaCI RDH 1 .0 g
(NH4)2S04 RDH 2.0 g
CaC03 Caltec 2.0 g
BACTO agar Difco 20 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 °C, 15 min.
ISP2
Component Source per L
Yeast extract Difco 4 g
Malt extract Difco 10 g
Glucose Sigma 4 g
Bacto agar Difco 20 g
Sterilise by autoclaving 121 °C, 15 min.
FK Seed Medium
component Source per L
1 .0 % Soy Peptone 10.0 g
2.0 % Glucose 20.0 g
0.5 % Bakers Yeast 5.0 g
0.2 % NaCI 2.0 g
1 % Trace Elements 10 mL
Solution The medium is adjusted to pH 7.0 with NaOH, and then sterilised by autoclaving 121°C, 15 minutes.
FK Production Medium
component Source per L
2.0 % Nutrisoy Soy 20.0 g
Bean Flour
2.0 % Glucose 20.0 g
0.6 % Bakers Yeast 6.0 g
0.25 % K2HP04 2.5 g
0.25 % KH2P04 2.5 g
0.5 % NaCI 5.0 g
3.0 % Glycerol 30.0 g
2.0% Soybean Oil 20.0 g
1 .0 % Trace 10 mL
Elements Solution
The medium is adjusted to pH 6.4 with NaOH, and then sterilised by autoclaving 121°C, 15 minutes.
FK Trace Elements Solution
component Source per L of
distilled
H20
ZnS04.7H20 0.05 g
MgS04.7H20 0.125 g
MnS04.H20 0.01 g
FeS04.7H20 0.02 g
Store in the dark at 4 °C
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 pyrosequencing 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 °C and harvested after 2 to 3 days. Cell pellets were washed with 10.3% sucrose and frozen at -20 °C 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 °C. 20 μί of RNaseA (10 mg/mL) were added and the mixture incubated for another 30 min at 37 °C. 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 °C 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., 1999) 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 μί LB Amp100 Kan50 per well) at 37 °C and frozen at -80 °C after mixing wells with 50μί LB/glycerol 1 :1 .
FkbO probe preparation
A DIG labeled fkbO probe was used to detect cosmids containing this region of the FK506 biosynthetic cluster. The probe was prepared by PCR using DIG labeled dNTP mix (Roche). It comprises 410 bp of 3'-terminal fkbO sequence. Sequence information for primer design had been obtained by 454 sequencing of BIOT-31 19 - see Figure 1 and SEQ ID NO: 1 . Primer sequences were:
UES2for (SEQ ID NO: 2) 5'-CACTCCTTCGATCTCCACGAGCAGGTCGCCACGGGC-3' and UES2rev (SEQ ID NO: 3) 5'-ACCCTGCCGTCCTCACGGCACACCACTACCCCACGG-3'.
Annealing temperatures between 66 and 71 °C and extension for 20 sec at 68 °C proved to be successful.
Colony hybridization
Thawed microplate-cultures were stamped onto positively charged filter membranes (Roche) which had been placed on LB Amp100 Kan50 plates. After overnight growth at 37 °C 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 (300 mM NaCI, 15 mM Sodium citrate) at 4 °C 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 °C. Stringent washes were performed at 68 °C. 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).
Conjugation of Streptomyces hygroscopicus subsp. hygroscopicus and Streptomyces tsukubaensis
Escherichia coli ET12567 (pUZ8002) (Macneil et al., 1992, Paget et al., 1999) was transformed with pKC1 139B01 -derived plasmids by electroporation to generate the £ coli donor strains for spore conjugation (Kieser et al., 2000). Fresh spores were harvested in water from plates of Streptomyces hygroscopicus (BIOT-4168) or Streptomyces tsukubaensis (BIOT- 4206). Spore suspensions were heat-shocked at 50 °C for 10 min. They were then mixed with the £ coli donor strain, which had been washed twice with 2xTY, in a ratio of 3:1 Streptomycete to £. coli, and the mixture shaken at 37 °C, 300 rpm, 2.5 cm throw for 1 .5 - 2 h. The conjugation mixture was then plated on R6 medium and incubated at 37 °C. After -20 h, the plates were overlaid with 2xTY containing apramycin sulphate and nalidixic acid and incubation continued at 37 °C. Plasmids with pKC1 139B01 backbone are not able to self- replicate in Streptomycetes at 37 °C 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 μ 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).
Analysis by LCMS
The HPLC system comprised an Agilent HP1 100 equipped with a Hyperclone ODS2,
C18, 3 micron 4.6 χ 150 mm column (Phenomonex). Injection volume 10 μί, oven 50°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.
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 positive scan mode,
MRM setup:
transitions [Da]
pimecrolimus (IS): 827.7→ 774.5
FK506 821 .7→ 768.5 (or 774.6)
7 878.0→ 824.7 8 906.0→ 720.6
9 891 .9→ 720.4
10 906.0→ 720.6
11 850.0→ 796.7 all parent ions are isolated with a width of 3 amu.
All FK520 and FK506 analogues can be quantified in this manner, for example, 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, pimecrolimus. 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, HMQC and HMBC) of purified material were recorded on a Bruker Advance DRX500 spectrometer, fitted with a Bruker Avance 500 Cryo Ultrashield, which operated at 500 MHz (for proton derived spectra, pro rata for other nuclei) at 27 °C. Chemical shifts are described in parts per million (ppm) and are referenced to solvent signal e.g. CHCI3 at δΗ 7.26 (1H) and CHCI3 at δ0 77.0 (13C). J values are given in Hertz (Hz).
LC-MS analysis of compounds prepared by synthesis: Samples were run on an Agilent 1200 coupled to a single-quad mass spectrometer and separate runs recorded positive and negative MS data. Method A: Xbridge, C18, 3.5 micron 4.6 χ 150 mm column (Waters). Oven 40°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.
Method B: Xbridge, C18, 3.5 micron 4.6 χ 150 mm column (Waters). Oven 40°C, A: 0.1 % formic acid, B: 0.1 % formic acid in MeCN. 1 ml_/min; 0-2 min 10% B; 15 min 100% B; 17 min 100% B; 17.1 min 10% B, 20 min 10% B.
Method C: Xbridge, C18, 3.5 micron 4.6 χ 50 mm column (Waters). Oven 40°C, A: 0.01 % ammonium hydroxide, B: MeCN. 1 .7 mL/min; 0 min 5% B; 1 .5 min 95% B
LCMS analysis of FK506 and FK520 analogues in biological assays: Samples were run on an API 4000.
Method D: Ultimate AQ-C18 (2.1 x 50mm, 3 micron). A: 0.025%FA/1 mM NH4OAc/H20; B: 0.025%FA/1 mM NH4OAc/MeOH. 0.4mL/min; 0.2min 10% B; 0.7min 60% B; 1.1 min 60% B; 1.4min 95% B; 2.3min 95% B; 2.4min 10%B, 3.5min 10%B.
Method E: Ultimate AQ-C18 (2.1 x 50mm, 3 micron). A: 0.025%FA/1 mM NH4OAc/H20; B: 0.025%FA/1 mM NH4OAc/MeOH. 0.4mL/min; 0.2min 20% B; 0.7min 60% B; 1.1 min 60% B; 1.4min 98% B; 2.3min 98% B; 2.4min 20%B, 3.5min stop.
Method F: Ultimate AQ-C 18 (2.1 x 50mm, 3 micron). A: 0.025%FA/1 mM NH4OAc/H20; B: 0.025%FA/1 mM NH4OAc/MeOH. 0.4mL/min; 0.3min 5% B; 0.5min 95% B; 1.7min 95% B; 1 .8min 5% B; 3.0min stop. Assessment of effect on NFA T gene expression
To assess the effect of compounds on inhibition of the IL-2 pathway, the Invitrogen GENEblazer NFAT gene reporter assay was used (Hanson, 2006; Qureshi, 2007) .
The general process of carrying out the assay was as follows: NFAT-bla Jurkat cells were thawed and resuspended in Assay Media (OPTI-MEM, 0.5% dialyzed FBS, 0.1 mM NEAA, 1 mM Sodium Pyruvate, 100 U/mL/100 g/mL Pen/Strep) to a concentration of 781 ,250 cells/mL. 4 μΙ_ of a 10X serial dilution of the test compounds were added to appropriate wells of a TC-Treated assay plate. 32 μΙ_ of cell suspension was added to the wells and pre- incubated at 37°C/5% C02 in a humidified incubator with test compounds for 30 minutes. Anti CD4:CD8 activator at the pre-determined EC80 concentration was added to wells containing the test compounds. The plate was incubated for 5 hours at 37°C/5% C02 in a humidified incubator. 8 μΙ_ of 1 μΜ Substrate Loading Solution was added to each well and the plate was incubated for 2 hours at room temperature. The plate was read on a fluorescence plate reader.
Assessment of water solubility
Water solubility may be tested as follows: A 10 mM stock solution of the FK506 or FK520 analogue is prepared in 100% DMSO at room temperature. Triplicate 0.01 mL aliquots are made up to 0.5 mL with either 0.1 M PBS, pH 7.3 solution or 100% DMSO in amber vials. The resulting 0.2 mM solutions are shaken, at room temperature on an IKA® vibrax VXR shaker for 6 h, followed by transfer of the resulting solutions or suspensions into 2 mL Eppendorf tubes and centrifugation for 30 min at 13200 rpm. Aliquots of the supernatant fluid are then analysed by the LCMS method as described above. Assessment of mouse plasma and human blood stability
In general, compounds were dissolved in DMSO, then spiked to a final concentration of 0.1 or 2μΜ into either human whole blood, or murine plasma and incubated at 25°C. Samples were then taken at 0, 0.25, 0.5, 2, 8 and 24 hours after addition, and analysed for levels of compound using LCMS methods described above.
Specific methods were as follows: 40μΙ_ of ultrapure H20 were added into a 96-well deep plate (plate A), then kept at 4°C. 10μΙ_ of prewarmed spiking solution B, containing test compound at 10x the necessary concentration in DMSO:MeOH:H20 (1 :4.5:4.5, v/v) (eg 100μΙ_ of 0.01 mM test compound in DMSO was added to 900μΙ_ of MeOH:H20 (1 :1 ) to give a spiking solution to generate a final test compound concentration in the blood or plasma of 0.1 μΜ) was added to all wells designated for timepoints in another 96 well plate (plate B), this was prewarmed at 37°C for 10 minutes. 90μΙ_ of prewarmed blood or plasma was then added to each of the wells of plate B. Triplicate 40μΙ_ aliquots were then removed immediately for the 0 h timepoint, then sequentially at 0.25, 0.5, 2, 8 and 24 hours after adding the blood or plasma, and transferred to the wells in plate A and mixed. 80μΙ_ of 50% MeOH in dH20 was added to each of the wells in plate A, mixed, then 380μΙ_ of pimecrolimus in acetonitrile (100ng/ml_) was added and mixed. Plate A was then kept at -70°C until analysis.
For analysis, plate A was thawed, vortexed for 10 minutes at 300rpm in a shaker, then centrifuged at 4000rpm for 15 minutes. The supernatant was transferred to a new 96-well plate for LC/MS analysis (using method F).
Assessment of cell permeability and efflux
Cell permeability was tested using the BIOCOAT® HTS Caco-2 Assay System developed by BD Biosciences as follows: The test compound was dissolved in DMSO and then diluted further in buffer to produce a final 1 μΜ dosing concentration. The fluorescence marker lucifer yellow was also included to monitor membrane integrity. Test compound was then applied to the apical surface of Caco-2 cell monolayers and compound permeation into the basolateral compartment is measured. This is performed in the reverse direction (basolateral to apical) to investigate active transport. LC-MS/MS was used to quantify levels of both the test and standard control compounds (such as Propanolol and Acebutolol). Standard criteria for compounds to be classified as showing efflux-limited permeability were:
Papp (B→A) / Papp (A→B) > 3, and Papp (B→A) > 1 .0 x 10"6 cm/s
Assessment of effect on murine cytokine release To look at the effect of compounds on cytokine production from mouse spleen cells, an in vitro murine cytokine release T-cell assay was used. Female BALB/c mice aged between 5 and 8 weeks were obtained from Harlan Olac (Shaw's Farm, Blackthorn, Bicester, Oxon, 0X25 1 TP, England). Animals were sensitised intra-peritoneally (i.p.) with 10C^g OVA in alum on day 0 to skew their immune profiles towards Th2. On day 7 animals were terminated by cervical dislocation and spleens were removed to ice-cold HBSS containing 10% HEPES. Spleens were processed under aseptic conditions to obtain a single cell suspension. Cells were then pooled, washed, the red blood cells lysed and a cell count performed to determine cell numbers and viability. Cells were resuspended at a density of 2x107/ml in RPMI with supplements. Two million cells were removed prior to culture for flow cytometry to assess levels of T and B lymphocytes present. This was determined using fluorescently conjugated antibodies to Thy1 .2 & B220 (CD45R). The remaining cells were cultured in triplicate in 96-well tissue culture plates with inhibitors at 3 concentrations (as determined by client) for 1 hour prior to the addition of OVA to the cultures (final concentration 200μg ml). Test compounds were reconstituted from the powdered form by addition of 100% DMSO to achieve concentrated stock solutions of equivalent molarity, based on the molecular weights provided for each compound. Compounds were then diluted 1 ,000 fold in culture media, resulting in a background level of 0.1 % DMSO in all wells. Subsequent 10-fold dilution of compounds was performed in RPMI/0.1 % DMSO. The final concentrations of test compounds in the wells were 2nM, 0.2nM & 0.02nM. Cells were incubated at a density of 106 per well. Positive control cultures were stimulated with a polyclonal activator of T cells (con A, 5μg ml), in order to confirm the health of the starting population. All cultures were incubated for 72 hours at 37°C, 5% C02. Supernatants were then removed and frozen down for subsequent measurement of key cytokines, IL-5 and IL-13, by Luminex.
Culture supernatants were thawed and centrifuged at 1 ,000g for 5 minutes at room temperature. Luminex analysis performed to determine the concentration of IL-5 and IL-13 in each sample. Samples were analysed using multiplex xMAP bead technology. Cytokines were measured according to the manufacturer's instructions. Briefly, aliquots of samples, standards and quality controls were incubated overnight with cytokine antibody-coated capture beads at 4°C in 96 well filter plates. Washed beads were further incubated with biotin-labelled cytokine- specific antibodies for 1 hour at room temperature. Beads were washed to remove excess biotinylated antibody, followed by 30 minute incubation with streptavidin conjugated to PE. After final washing, the mean fluorescence intensity of PE bound to each individual
microsphere was quantified using a Luminex machine (dual laser flow analyzer) and xPonent software (Luminex corporation). A standard curve of quantified recombinant cytokines, ranging from 3.2 - 10,000pg/ml, was used to convert detected PE fluorescence values to cytokine concentrations (pg/ml). All standards were run in duplicate and at least 50 microspheres are analysed per cytokine. The cytokines present in the supernatants taken from spleen cultures were determined in each of the triplicates by comparison to the values observed with the recombinant cytokine standards. The cytokine concentration in each sample and the mean of replicates was noted. Cytokine production from splenocytes after OVA stimulation in vitro was compared in the presence and absence of inhibitors.
Assessment of effect on human PBMC cytokine release
Blood was collected into standard lithium-heparin vacutainers from one healthy donor and subsequently diluted 1 :5 with RPMI media. The donor was not atopic and was not receiving any medication.
PBMCs were separated from whole blood using the AccuSpin System utilising
Histopaque 1077 (Sigma). Following the appropriate washing steps, PBMCs were counted and then divided into relevant fractions for CD3/CD28 stimulation. Compounds were dissolved in DMSO, and diluted further in RPMI media to the working concentration. Final DMSO in the assay was 0.1 % in all samples.
Two 96 well plates were coated with 0^g per well anti-CD3 antibody (clone UCHT1 , Calbiochem) at 4°C overnight with the exception of wells for unstimulated controls. After washing off unbound anti-CD3 antibody, 20μΙ of compound or medium containing DMSO was added to the plates. PBMCs were diluted in RPMI containing 10% dialysed FBS to yield a concentration of 100,000 cells per 180μΙ. For unstimulated control wells, 180μΙ of cell suspension was added to relevant wells. To the remaining cell suspension, anti-CD28 antibody (Calbiochem) was added to yield a final concentration of 2μg ml in the assay and 180μΙ of this cell suspension was immediately added to the plates.
The plates were then placed in a humidified 37°C incubator for 24 hours. At this time, the plates were centrifuged at 400 x g for 5 minutes and 100μΙ supernatant transferred to clean 96 well plates. Supernatants were frozen rapidly at -80oC and stored until required for cytokine analysis by Bio-Plex assay.
Levels of cytokines were analysed by means of a Human Th1/Th2 multiplex assay kit (Bio-Rad) utilising a Bioplex platform. Cell supernatants were thawed on ice before being added directly to the assay plate. The cytokine analysis was carried out as per manufacture's instructions. Standard curves and quantification of samples were generated using Bioplex Manager software (BioRad). Assessment of Pgp efflux There are many methods which may be employed to investigate the rates of efflux by drug transporters such as Pgp. Many of these are reviewed in Szakacs et al., 2008 and
Hegedus et al., 2009, and include ATPase assays. Assessment of CyP450 metabolism and inhibition
CyP450 metabolism and inhibition may be analysed by incubating compounds of interest with isolated hepatocytes, microsomes or isolated CyP450 enzymes, then measuring rate of metabolism of the test compound over a short period of time, such as 0-60 minutes, or using a fluorometric inhibition assay or microsomal inhibition assay to measure P450 inhibition (see Yan and Caldwell, 2001 ).
In vivo assessment of pharmacokinetics
In vivo assays may also be used to measure the bioavailability of a compound.
Generally, a compound is administered to a test animal (e.g. mouse) both intravenously (i.v.) and orally (p.o.) and blood samples are taken at regular intervals to examine how the plasma (or whole blood) concentration of the drug varies over time. Alternatively, the compounds may be dosed by aerosol using a nebulizer. The time course of plasma (or whole blood)
concentration over time can be used to calculate the PK parameters (using software such as Kinetica, Thermo Scientific) such as absolute bioavailability of the compound as a percentage using standard models.
In vivo assessment of oral and intravenous pharmacokinetics
For FK506 and FK520 analogues, whole blood was analysed. Compounds were formulated in 5% ethanol / 5% cremophor EL / 90% saline for both p.o. and i.v. administration. Groups of 3 male CD1 mice were dosed with either 1 mg/kg i.v. or 10mg/kg p.o. Blood samples (40μΙ_) were taken via saphenous vein, pre-dose and at 0.25, 0.5, 2, 8, and 24 hours, and diluted with an equal amount of dH20 and put on dry ice immediately. Samples were stored at - 70°C until analysis. The concentration of the compound of the invention or parent compound in the sample was determined via LCMS as follows:20 μΙ_ of blood:H20 (1 :1 , v/v)/PK sample was added with 20 μΙ_ Internal standard (pimecrolimus) at 100 ng/mL, 20 μΙ_ working solution/MeOH and 150 μΙ_ of ACN, vortexed for 1 minute at 1500 rpm, and centrifuged at 12000 rpm for 5 min. The supernatant was then injected into LC-MS/MS. The time-course of blood concentrations was plotted and used to derive area under the whole blood concentration-time curve (AUC - which is directly proportional to the total amount of unchanged drug that reaches the systemic circulation). These values were used to generate the oral bioavailability (F%) and other PK parameters where possible. Where possible, the levels of the presumed primary metabolite, 11 , were analysed.
In vivo assessment of aerosol pharmacokinetics
For FK506 and FK520 analogues, whole blood was analysed. Compounds were formulated in 100% ethanol for aerosol administration. Mice were dosed 20mg over 10 minutes to groups of 9 animals in a chamber using a nebulizer. At 0.25, 0.5, 2, 8, and 24 hours, three mice per timepoint were euthanized under anaesthesia with isofulrane. The animals were placed on ice and the chest cavity opened immediately to expose heart and lung. 100μΙ_ of blood sample was collected from each animal by cardial puncture into K2EDTA tubes. This blood sample was diluted with the same volume of diH20 and put on dry ice immediately. After collection of the blood sample, lung tissue was rapidly collected, washed with cold saline, dried, weighed and snap frozen with dry ice. The whole process was completed in 2 minutes. Blood and lung samples were then stored at -70°C until analysis.
For analysis, lung tissue was homogenized for 2 min with 3 volumes (v/w) of
homogenizing solution (PBS: ACN=1 :9). 40 μΙ_ of blank blood:H20 (1 :1 , v/v)/lung
homogenate/unknown PK sample was added to 30 μΙ_ internal standard (pimecrolimus,100 ng/mL), 40 μΙ_ of working solution/MeOH and 160 μΙ_ of ACN, vortexed for 1 min at 1500 rpm, and centrifuged at 12000 rpm for 5 min. The supernatant was diluted with MeOH (1 :1 , v/v), vortexed and then injected into the LC-MS/MS system. Blood samples were prepared for analysis as described for intravenous and oral pharmacokinetics. The concentration of the relevant compound/s in the lung and blood samples was then determined via LCMS (see materials and methods, using LCMS methods D or E). The time-course of lung and blood concentrations was plotted and used to derive area under the whole blood concentration-time curve (AUC - which is directly proportional to the total amount of unchanged drug that reaches the systemic circulation). These values were used to generate the ratio of lung concentration to blood concentration. Where possible, the levels of the presumed primary metabolite, 11 , were also analysed.
Example 1 - Generation of a Streptomyces hygroscopicus subsp. hygroscopicus strain in which the fkbO gene has been inactivated by introducing a small deletion inducing a frameshift.
1 .1 Cloning of DNA homologous to the upstream flanking region of fkbO disruption.
Oligos SG165 (SEQ ID NO: 1 ) and SG166 (SEQ ID NO: 2) were used to amplify a 1727 bp region of DNA, aesFK520A (SEQ ID NO: 10) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in each oligo to include the restriction sites present in the genomic sequence to aid cloning of the amplified fragment (Figure 1 ). This 1727 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAESA5 and the insert verified by sequencing.
SG165 CCAGATCTCGTCGGGCACCTTGAAGTAGGCGAGCCG
(SEQ ID NO: 1 )
SG166 CCCTCGAGGTCCGGTGATCCGGTCTTCTCGAAGC
Xho\
(SEQ ID NO: 2)
1 .2 Cloning of DNA homologous to the downstream flanking region of fkbO disruption.
Oligos SG167 (SEQ ID NO: 3) and SG168 (SEQ ID NO: 4) were used to amplify a 2034 bp region of DNA, termed aesFK520B (SEQ ID NO: 1 1 ) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in oligo SG167 to introduce the restriction site to aid cloning of the amplified fragment (Figure 1 ) and SG168 included the restriction site present in the genomic sequence. This 2034 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAESBI and the insert verified by sequencing.
SG167 CCCTCGAGCGCACAGCGCCCTGTCGAGTCCGGCATG
Xho\
(SEQ ID NO: 3)
SG168 CCGAATTCGAGGTCGAAGCGGGTGAACCAGCGTC
EcoRI
(SEQ ID NO: 4)
The -1 .7 kb Bgl\\IXho\ fragment from pAESA5 and -2.0 kb Xho\IEcoR\ fragment from pAESBI were cloned into the -5.9 kb Bgl\\/EcoR\ fragment of pKC1 139 (Bierman et al., 1992) to make pAESABI . pAESABI therefore contained the upstream and downstream regions such that the double crossover event would result in the desired small deletion, introducing a frameshift in the fkbO gene.
1 .3 Transformation of Streptomyces hygroscopicus subsp. hygroscopicus
Escherichia coli ET12567, harbouring the plasmid pUZ8002, was transformed with pAESABI by electroporation to generate the E. coli donor strain for conjugation. This strain was used to transform Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) by spore conjugation (as described in Materials and Methods). Exconjugants were plated on R6 agar and incubated at 28°C. pAESABI is able to self-replicate in Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) at 28°C. Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 28°C to ensure the pAESABI plasmid with resistance marker was present. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured again on to MAM plates with apramycin at 37°C to induce the plasmid to integrate, as the plasmid cannot self-replicate at 37°C. The transformants were then subcultured for four subsequent rounds at 37°C on MAM plates with no antibiotic. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP3 medium at 28°C. Serial dilutions were made from the filtered collected spores and were plated on MAM plates to achieve single colonies.
1 .4 Screening for secondary crosses
Single colonies were patched in duplicate onto MAM supplemented with 0.050 mg/mL apramycin and MAM containing no antibiotics, and grown at 28 °C for 3-4 days. Patches that grew on the no antibiotic plate but did not grow on the apramycin plate were screened to test if the desired double recombination effect had occurred. A 6 mm circular plug from each patch that had lost the marker was used to inoculate individual 50 mL falcon tubes containing 7 mL FK seed medium (See Media Recipes) without antibiotics and grown for 2 days at 28°C, 300 rpm with a 1 inch throw. These were then used to inoculate (0.5 mL into 7 mL - 7% inoculum) FK production medium (See Media Recipes) in a 50 mL falcon tube at 28°C, 300 rpm with a 1 inch throw. After 24 hours, each falcon tube was fed with 0.050 mL 0.32 M A-trans- hydroxycyclohexane carboxylic acid to give a final concentration of 2.12 mM acid and shaking incubation was continued as before. The cultures were sampled after 6 days growth and analysed by LC-MS, using the methods described above.
14 out of 79 apramycin sensitive strains tested had undergone the desired recombination event to give the disruption of the fkbO gene. These strains produced a metabolite that was 30 amu less than FK520 itself. This compound was shown to be 31 - desmethoxyFK520 following isolation and characterisation of that compound. One strain was ultimately selected, designated "AB5-10". This strain was later renamed "BIOT-4131 ".
Example 2 - Generation of a Streptomyces hygroscopicus subsp. hygroscopicus strain in which the fkbO gene has been inactivated by introducing a large in-frame deletion. 2.1 Cloning of DNA homologous to the upstream flanking region of fkbO disruption.
Oligos SG165 (SEQ ID NO: 1 ) and SG166 (SEQ ID NO: 2) were used to amplify a 1727 bp region of DNA, termed aesFK520A (SEQ ID NO: 10) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al. 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in each oligo to include the restriction sites present in the genomic sequence to aid cloning of the amplified fragment (Figure 2). This 1727 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAESA5 and the insert verified by sequencing.
SG165 CCAGATCTCGTCGGGCACCTTGAAGTAGGCGAGCCG
fig/i I
(SEQ ID NO: 1 )
SG166 CCCTCGAGGTCCGGTGATCCGGTCTTCTCGAAGC
Xho\
(SEQ ID NO: 2)
2.2 Cloning of DNA homologous to the downstream flanking region of fkbO disruption.
Oligos SG169 (SEQ ID NO: 5) and SG168 (SEQ ID NO: 6) were used to amplify a 1460 bp region of DNA, termed aesFK520C (SEQ ID NO: 12) from Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in oligo SG169 to introduce the restriction site to aid cloning of the amplified fragment (Figure 2) and SG168 included the restriction site present in the genomic sequence. This 1460 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAESC8 and the insert verified by sequencing.
SG169 CCCTCGAGAACATGGCCCGGGTCATCGGCGCGG
Xho\
(SEQ ID NO: 5)
SG168 CCGAATTCGAGGTCGAAGCGGGTGAACCAGCGTC
EcoRI
(SEQ ID NO: 4)
The -1 .7 kb Bgl\\/Xho\ fragment from pAESA5 and -1 .5 kb Xho\/EcoR\ fragment from pAESBI were cloned into the -5.9 kb Bgl\\IEcoR\ fragment of pKC1 139 (Bierman et al., 1992) to make pAESAC4. pAESAC4 therefore contained the upstream and downstream regions such that the double crossover event would result in the desired large deletion of the fkbO gene. 2.3 Transformation of Streptomyces hygroscopicus subsp. hygroscopicus
Escherichia coli ET12567, harbouring the plasmid pUZ8002, was transformed with pAESAC4 by electroporation to generate the E. coli donor strain for conjugation. This strain was used to transform Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) by spore conjugation (as described in Materials and methods). Exconjugants were plated on R6 agar and incubated at 28°C. pAESAC4 is able to self-replicate in Streptomyces hygroscopicus subsp. hygroscopicus (DSM 40822) at 28°C. Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 28°C to ensure the pAESAC4 plasmid with resistance marker was present. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured on to MAM plates with apramycin at 37°C to induce the plasmid to integrate, as the plasmid cannot self-replicate at 37°C. The transformants were then subcultured for four subsequent rounds at 37°C on MAM plates with no antibiotic. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP3 medium at 28°C. Serial dilutions were made from the filtered collected spores and were plated on MAM plates to achieve single colonies.
2.4 Screening for secondary crosses
Single colonies were patched in duplicate onto MAM supplemented with 0.050 mg/mL apramycin and MAM containing no antibiotics, and grown at 28 °C for 3-4 days. Patches that grew on the no antibiotic plate but did not grow on the apramycin plate were screened to test if the desired double recombination effect had occurred. A 6 mm circular plug from each patch that had lost the marker was used to inoculate individual 50 mL falcon tubes containing 7 mL FK seed medium (See Media Recipes) without antibiotics and grown for 2 days at 28°C, 300 rpm with a 1 inch throw. These were then used to inoculate (0.5 mL into 7 mL - 7% inoculum) FK production medium (See Media Recipes) in a 50 mL falcon tube at 28°C, 300 rpm with a 1 inch throw. After 24 hours, each falcon tube was fed with 0.050 mL 0.32 M 4-irans-hydroxy cyclohexane carboxylic acid to give a final concentration of 2.12 mM acid and shaking incubation was continued as before. The cultures were sampled after 6 days growth and analysed by LC-MS using the methods described above.
15 out of 59 apramycin sensitive strains tested had undergone the desired recombination event to give the disruption of the fkbO gene. When fed as described above these strains produced a metabolite that was 30 amu less than FK520 itself. This compound was shown to be 31 -desmethoxyFK520 by comparison with isolated and charaterised material. One of these strains was designated as "AC1 -5" and later renamed to BIOT-4132. Example 3 - Generation of a Streptomyces tsukubaensis strain in which the fkbO gene has been inactivated by introducing a small deletion inducing a frameshift.
3.1 Cloning of DNA homologous to the upstream flanking region of fkbO disruption.
Oligos AES43 (SEQ ID NO: 6) and AES40 (SEQ ID NO: 7) were used to amplify a 2057 bp region of DNA, termed aesFK506D1 1 (SEQ ID NO: 13) from Streptomyces tsukubaensis no. 9993 (FERM BP-927) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in each oligo to introduce the restriction sites to aid cloning of the amplified fragment (Figure 3). This 2057 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAES8 and the insert verified by sequencing.
AES43 CCAAGCTTGAGCGCCTCGTCCCAGAGCGCGGCCTGGTC
Hind\\\
(SEQ ID NO: 6)
AES40 CCATGCATCGGGACACCGTCCGTGAGCGACACCTCGGCATGACC
Λ/s/'l
(SEQ ID NO: 7) 3.2 Cloning of DNA homologous to the downstream flanking region of fkbO disruption.
Oligos AES41 (SEQ ID NO: 8) and AES42 (SEQ ID NO: 9) were used to amplify a 1985 bp region of DNA, termed aesFK506E3 (SEQ ID NO: 14) from Streptomyces tsukubaensis no. 9993 (FERM BP-927) in a standard PCR reaction (Sambrook and Russell, 2001 ) using genomic DNA (Kieser et al., 2000) as the template and hot start KOD DNA polymerase. A 5' extension was designed in each oligo to introduce the restriction sites to aid cloning of the amplified fragment (Figure 3). This 1985 bp fragment was cloned into pUC19 that had been linearised with Sma\, resulting in plasmid pAES9 and the insert verified by sequencing.
AES41 CCATGCATCCGATGCCGTCGCGGCGCTCTACACGCGGG
Λ/s/'l
(SEQ ID NO: 8)
AES42 CCAGATCTGAAGGGCTCGGCGGTCACACCGGGCAGCGC
fig/i I
(SEQ ID NO: 9)
The -2.0 kb Hind\\\/Nsi\ fragment from pAES8 and -2.0 kb NsiUBglW I fragment from pAES9 were cloned into the -6.0 kb H/ndlll/Sg/ll fragment of pKC1 139 (Bierman et al., 1992) to make pAES10. pAES10 therefore contained the upstream and downstream regions such that the double crossover event would result in the desired small deletion, introducing a frameshift in the fkbO gene. 3.3 Transformation of Streptomyces tsukubaensis
Escherichia coli ET12567, harbouring the plasmid pUZ8002, was transformed with pAESI O by electroporation to generate the E. coli donor strain for conjugation. This strain was used to transform Streptomyces tsukubaensis no. 9993 (FERM BP-927) by spore conjugation (as described in Materials and methods). Exconjugants were plated on R6 agar and incubated at 37°C. pAESI O is not able to self-replicate in Streptomyces tsukubaensis no. 9993 (FERM BP-927) at 37°C and must integrate into the genome. Transformants were subcultured onto MAM plates with apramycin (0.050 mg/mL) at 37°C two to three times, to ensure that the pAESI O plasmid with resistance marker had integrated. Subculturing to allow secondary recombination was carried out as follows: the transformants were subcultured for two subsequent rounds at 37°C on MAM plates with no antibiotic and then a final time at 28°C. The transformants from the fourth subculture on antibiotic free plates were plated for spore harvest on ISP4 medium at 28°C. Serial dilutions were made from the filtered collected spores and plated on ISP4 plates at 28°C to achieve single colonies.
3.4 Screening for secondary crosses
Single colonies were patched in duplicate onto ISP4 supplemented with 0.050 mg/mL apramycin and ISP4 containing no antibiotics, and grown at 28 °C for 3-4 days. Patches that grew on the no antibiotic plate but did not grow on the apramycin plate were screened to test if the desired double recombination effect had occurred. A 6 mm circular plug from each patch that had lost the marker was used to inoculate individual 50 mL falcon tubes containing 7 mL NGY (See Media Recipes) without antibiotics and grown for 2 days at 28°C, 300 rpm with a 1 inch throw. These were then used to inoculate (0.5 mL into 7 mL - 7% inoculum) PYDG+MES (See Media Recipes) in a 50 mL falcon tube at 28°C, 300 rpm with a 1 inch throw. After 24 hours, each falcon tube was fed with 0.050 mL 0.32 M 4-irans-hydroxy cyclohexane carboxylic acid to give a final concentration of 2.12 mM acid and shaking incubation will be continued as before. The cultures were sampled after 6 days growth and analysed by LC-MS by the methods described above. Of the 93 patches that were screened, 49 had undergone the desired double recombination event. One of these strains was designated BIOT-4254.
Example 4 - Isolation of 5 (31 -desmethoxy FK506)
Spore stocks of BIOT-4254 were recovered onto plates of ISP4 medium and incubated for 2 weeks at 28°C. Vegetative cultures (seed culture) were prepared by adding 0.2 ml of BIOT-4254 working stock and inoculating into 400 ml NGY medium in 2 litre Erlenmeyer flasks with a foam bung. Cultivation was carried out for 48 hours at 28°C, 250 rpm (2.5 cm throw). The entire seed culture in one flask was transferred into 15 litres of medium PYDG pre- adjusted at pH 7.0 in 22 L Braun Biostat Fermentors. The fermentation was carried out at 28 °C, with starting agitation at 200 rpm, aeration rate at 0.5 VA /M (7.5 SL/min) and dissolved oxygen (DO) level controlled with the agitation cascade at 30% air saturation. At 24 hours post- inoculation 5ml of 0.2 M irans-4-hydroxycyclohexanecarboxylic acid in methanol was added to the vessel. After 24 hours post inoculation the aeration rate was increased to 0.7 V/V/M (10.5 litres/min) and control ph 6.4 ± 0.8 with 1 .0M NaOH and 0.5 M H2S04. The culture was harvested after 93 hours post-inoculation.
The broths were combined and the cells harvested by centrifugation. The cells were extracted with EtOAc/MeOH (50:50) and the organics were dried in vacuo to yield a crude extract (19.5 g). The extract was subjected to normal phase column chromatography (on Silica Gel 60 (40 x 2.5 cm)) eluted with CHCI3/MeOH (from 100:0 to 95:5 in incremental changes). Fractions containing the target compound were combined and reduced in vacuo. The enriched extract was then subjected to normal phase column chromatography (on Silica Gel 60 (25 x 2 cm)) eluted with 50:50 Hexane and EtOAc. Fractions containing the target compound were combined and taken to dryness, to yield the target compound as an off-white solid (667 mg)
Example 5 - Isolation of 6 (31 -desmethoxy FK520)
Spore stocks of BIOT-4131 were recovered onto plates of MAM medium and incubated for 5 days at 28°C. Vegetative cultures (seed culture) were prepared by removing 4 agar plugs (5 mm in diameter) from the MAM plate and inoculating into 250 ml FK seed medium in 2 litre Erlenmeyer flasks with foam bung. Cultivation was carried out for 50 hours at 28°C, 250 rpm (2.5 cm throw). The entire seed culture in one flask was transferred into 5 litres of FK production medium (pre-sterilisation pH adjusted at pH 6.4) in 7 L Applikon Fermentor. The fermentation was carried out at 28 °C, with starting agitation at 350 RPM, aeration rate at 0.5 V/V/M (2.5 SL/min) and dissolved oxygen (DO) level controlled with the agitation cascade at 30% air saturation. At 24 hours post-inoculation 5 ml from a 2M stock solution of trans-4- hydroxycyclohexane carboxylic acid in methanol was added to the vessel. The culture was harvested after 1 16 hours post-inoculation.
A second fermentation batch was run as above except: NGY seed medium, PYDG production medium (pre-sterilisation pH adjusted at pH 7.0) and harvested after 1 17 hours. The combined broths were extracted with ethyl acetate (2 x 1 volume equivalent) and the resultant organics reduced in vacuo to a crude extract (16 g). This extract was dissolved in acetonitrile (1000 ml) and silica added (30 g). The solvent was removed in vacuo and the adsorbed silica added to the top of a packed silica column (170 mm x 55 mm diameter). The column was eluted with ethyl actate / hexanes (1 :2, 0.9 L; 1 :1 , 1 L; 3:2, 1 L; 2:1 , 1.8 L) and fractions containing the target compound combined and taken to dryness (190 mg). This enriched extract was then purified by preparative HPLC over C18 to yield the target compound (37 mg).
Example 6 - Preparation of Λ/,Λ/'-ditosylhydrazine
To a mixture of p-toluenesulfonyl hydrazide (9.32 g, 50 mmol) and p-toluenesulfonyl chloride (14.3 g, 75 mmol) in CH2CI2 (50 ml) was added pyridine (6 ml, 75 mmol). The mixture was stirred for 1.5 h at room temperature. Et20 (200 ml) and H20 (100 ml) were added and stirred at 0 °C for 15 min. The mixture was filtered, and washed with Et20. The solid was recrystallized from CH3OH to give Λ/,Λ/'-ditosylhydrazine as a white solid (1 1.5 g, 68% yield).
Example 7 - Synthesis of 7
Figure imgf000047_0001
To a solution of 5 (150 mg, 0.19 mmol) and rhodium acetate dimer (2.2 mg, 0.0038 mmol) in CH2CI2 (3 ml) was added dropwise a solution of ethyl diazoacetate (21 μΙ, 0.19 mmol) in CH2CI2 (1 ml) at 30 °C under N2. After the addition was completed, the reaction was stirred for 30 min at 30 °C and then additional ethyl diazoacetate (42 μΙ, 0.38 mmol) in CH2CI2 (2 ml) was added dropwise. After the addition, stirring was continued at 30 °C for 30 min under N2. Then the solvent was removed in vacuo and the residue was purified by preparative TLC (petroleum ether / acetone =3:1 ) to give 7 as a white solid (85 mg, 51 % yield) (see figure 1 for NMR). LCMS data (method A): retention time = 7.8 minutes, [M+Na]+ = 882.5.
Example 8 - Synthesis of 8
Figure imgf000048_0001
8.1 The preparation of intermediate n-butyl diazoacetate
To a solution of butanol (2.8 ml, 30 mmol) in CH2CI2 (30 ml) was added Et3N (14 ml, 60 mmol). Bromoacetyl bromide (3.9 ml, 45 mmol) was added dropwise at 0 °C . The mixture was stirred for 30 min at room temperature, and then water was added, and extracted with CH2CI2. The combined organic extracts were washed with NaHC03 and brine, dried (Na2S04), filtered and concentrated to give butyl 2-bromoacetate as a brown oil (5.9 g) which was used without further purification.
Butyl 2-bromoacetate (0.98 g, 5 mmol) and Ν,Ν'-ditosyl hydrazine (3.4 g, 10 mmol, see example 6) were dissolved in THF (25 ml) and cooled to 0 °C . DBU (3.7 ml, 25 mmol) was added dropwise and the mixture was stirred for 30 min at room temperature, then quenched by the addition of saturated NaHC03 solution, and extracted with Et20. The combined organic extracts were washed with water and brine, dried (Na2S04), filtered and concentrated to give a residue which was purified by flash chromatography (petroleum ether / ethyl acetate =10:1 ) to give n-butyl diazoacetate as a yellow oil (0.4 g, 57% yield).
8.2 The preparation of 8
A stirred solution of 5 (100 mg, 0.13 mmol) in CH2CI2 (2 ml) containing rhodium acetate dimer (1 .5 mg, 0.0026 mmol) under N2 was heated to 30 °C , while n-butyl diazoacetate (18 mg, 0.13 mmol) in CH2CI2 (1 ml) was added dropwise. After the addition was completed, the reaction was stirred for 30 min at 30 °C and additional n-butyl diazoacetate (37 mg, 0.26 mmol) in CH2CI2 (2 ml) was added dropwise. Stirring was continued at 30 °C for 30 min under N2 after the addition was completed. The solvent was removed in vacuo and the residue was purified by preparative TLC (petroleum ether / acetone =3:1 ) to give 8 as a white solid (45 mg, 39% yield) (see figure 2 for NMR).
LCMS data (method B): retention time = 17.5 minutes, [M+Na]+ = 910.5. Example 9 - Synthesis of 9
Figure imgf000049_0001
9.1 The preparation of intermediate isopropyl diazoacetate
To a solution of 'PrOH (2.3 ml, 30 mmol) in CH2CI2 (30 ml) was added Et3N (14 ml, 60 mmol). Bromoacetyl bromide (3.9 ml, 45 mmol) was added dropwise at 0 °C . The mixture was stirred for 30 min at room temperature, and then water was added, and extracted with CH2CI2. The combined organic extracts were washed with NaHC03 and brine, dried (Na2S04), filtered and concentrated to give isopropyl 2-bromoacetate as a brown oil (5.9 g) which was used without further purification.
Isopropyl 2-bromoacetate (0.91 g, 5 mmol) and Ν,Ν'-ditosyl hydrazine (3.4 g, 10 mmol; see example 6) were dissolved in THF (25 ml) and cooled to 0 °C . DBU (3.7 ml, 25 mmol) was added dropwise and the mixture was stirred for 30 min at room temperature. The reaction was quenched by the addition of saturated NaHC03 solution, and extracted with Et20. The combined organic extracts were washed with water and brine, dried (Na2S04), filtered and concentrated to give a residue which was purified by flash chromatography (petroleum ether / ethyl acetate =10:1 ) to give isopropyl diazoacetate as a yellow oil (0.3 g, 47 % yield).
9.2 The preparation of 9
A stirred solution of 5 (100 mg, 0.13mmol) in CH2CI2 (2 ml) containing rhodium acetate dimer (1.5 mg, 0.0026 mmol) under N2 was heated to 30 °C, while isopropyl diazoacetate (20 mg, 0.13 mmol) in CH2CI2 (1 ml) was added dropwise. After the addition was completed, the reaction was stirred for 30 min at 30 °C and additional isopropyl diazoacetate (40 mg, 0.26 mmol) in CH2CI2 (2 ml) was added dropwise. Stirring was continued at 30 °C under N2for 30 min after addition was completed. The solvent was removed in vacuo and the residue was purified by preparative TLC (petroleum ether / acetone =3:1 ) to give 9 as a yellow oil (50 mg, 44 % yield) (see figure 3 for NMR)..
LCMS data (method A): retention time = 8.0 minutes, [M+Na]+ = 896.5 Example 10 - Synthesis
Figure imgf000050_0001
10
10.1 The preparation of isobutyl diazoacetate
To a solution of isobutyl alcohol (2.75 ml, 30 mmol) in CH2CI2 (30 ml) was added Et3N (14 ml, 60 mmol). The bromoacetyl bromide (3.9 ml, 45 mmol) was added dropwise at 0 °C. The mixture was stirred for 30 min at room temperature, water was added, and extracted with
CH2CI2. The combined organic extracts were washed with NaHC03 and brine, dried (Na2S04), filtered and concentrated to give isobutyl 2-bromoacetate as a brown oil (6 g) which was used without further purification.
Isobutyl 2-bromoacetate (0.195 g, 1 mmol) and Ν,Ν'-ditosyl hydrazine (0.68 g, 2 mmol, see example 6) were dissolved in THF (5 ml) and cooled to 0 °C. DBU (0.74 ml, 5 mmol) was added dropwise and the mixture was stirred for 30 min at room temperature. The reaction was quenched by the addition of saturated NaHC03 solution, and extracted with Et20. The combined extracts were washed with water and brine, dried (Na2S04), filtered and concentrated to give a residue which was purified by flash chromatography (petroleum ether / ethyl acetate =10:1 ) to give isobutyl diazoacetate as a yellow oil (78 mg, 55% yield).
10.2 Preparation of 10
A stirred solution of 5 (1 10 mg, 0.141 mmol) in CH2CI2 (2 ml) containing rhodium acetate dimer (1.5 mg, 0.0026 mmol) under N2 was heated to 30 °C, while isobutyl diazoacetate (20 mg, 0.141 mmol) in CH2CI2 (1 ml) was added dropwise. After the addition was completed, the reaction was stirred for 30 min at 30 °C and additional isobutyl diazoacetate (40 mg, 0.282 mmol) in CH2CI2 (2 ml) was added dropwise. Stirring was continued at 30 °C under N2for 30 min after the addition was completed. The solvent was removed in vacuo and the residue was purified by preparative TLC (petroleum ether / acetone =3:1 ) to give 10 as a yellow oil (28 mg, 22% yield) (see figure 4 for NMR)..
LCMS data (method B): retention time = 17.0 minutes, [M+Na]+ = 910.4
Example 11 - preparation of 11
Figure imgf000051_0001
TAS-F
DMF, rt
Figure imgf000051_0002
11.1 The preparation of 2-(trimethylsilyl)ethyl diazoacetate
To a solution of 2-(trimethylsilyl)ethanol (4.3 ml, 30 mmol) in CH2CI2 (30 ml) was added Et3N (14 ml, 60 mmol). Bromoacetyl bromide (3.9 ml, 45 mmol) was added dropwise at 0 °C. The mixture was stirred for 30 min at room temperature, and then water was added, and extracted with CH2CI2. The combined organic extracts were washed with NaHC03 and brine, dried (Na2S04), filtered and concentrated to give 2-(trimethylsilyl)ethyl 2-bromoacetate as a brown oil (7.2 g) which was used without further purification.
2-(trimethylsilyl)ethyl 2-bromoacetate (1 .2 g, 5 mmol) and Ν,Ν'-ditosyl hydrazine (3.4 g, 10 mmol, see example 6) were dissolved in THF (25 ml) and cooled to 0 °C . DBU (3.7 ml, 25 mmol) was added dropwise and the mixture was stirred for 30 min at room temperature. The reaction was quenched by the addition of saturated NaHC03 solution, and extracted with Et20. The combined extracts were washed with water and brine, dried (Na2S04), filtered and concentrated to give a residue which was purified by flash chromatography (petroleum ether / ethyl acetate =10:1 ) to give 2-(trimethylsilyl)ethyl diazoacetate as a yellow oil (0.55 g, 59% yield).
11.2 Synthesis of 11
To a stirred solution of 5 (100 mg, 0.13 mmol) and rhodium acetate dimer (1 .5 mg, 0.0026 mmol) in CH2CI2 (2 ml) was added dropwise 2-(trimethylsilyl)ethyl diazoacetate (24 mg, 0.13 mmol) in CH2CI2 (1 ml) at 30 °C under N2. After the addition was completed, the reaction was stirred for 30 min at 30 °C , then additional 2-(trimethylsilyl)ethyl diazoacetate (48 mg, 0.26 mmol) in CH2CI2 (2 ml) was added dropwise and stirring was continued for 30 min at 30 °C under N2. The solvent was removed in vacuo and the residue was purified by preparative TLC (petroleum ether / acetone =3:1 ) to give a yellow oil (60 mg). To a solution of this yellow oil (30 mg, 0.032 mmol) in DMF (1 ml) was added tris(dimethylamino)sulfonium
difluorotrimethylsilicate (35 mg, 0.129 mmol). The resulting solution was stirred overnight, then partitioned between NaH2P04 (1 M, 5 ml) and Et20 (10 ml). The organic phase was washed with Na2HP04 (1 .0 M, 2 x 3 ml), dried over sodium sulfate, filtered and concentrated to afford a light yellow oil. The crude acid was purified by preparative-TLC (DCM: i-PrOH=20 ml: 1 ml, 2 drops AcOH) to give pure 11 (17 mg) (see figure 5 for NMR)..
LCMS data (method C): retention time = 1 .27 minutes, [M+Na]+ = 849.3
Example 12 - Biological data - NFAT gene reporter
To assess direct effect of the compounds on inhibition of the IL-2 pathway, the Invitrogen GENEblazer NFAT gene reporter assay was used as described in the materials and methods. IC50 values are displayed in table 1 for all compounds tested, the limits of ranges are shown.
Table 1.
Figure imgf000053_0001
Example 13 - Biological data - inhibition of murine cytokine release
To assess effect of the compounds on inhibition of the asthma-relevant cytokines, such as
IL-5 and IL-13, a mouse spleen cytokine release assay was used, as described in the materials and methods.
Data for three concentrations of test article is shown in tables 2 and 3. The data displayed below was obtained from a mean of triplicate values and shows the percentage inhibition of IL5 or IL13 at three concentrations compared to OVA alone. This data confirms the action of FK506, 1 and 7 on cytokines that are thought to be important in asthma progression, and also the reduced activity of 11 , the presumed primary metabolite of the ester series.
Table 2.
Percentage inhibition of IL-5 compared to OVA control
Test article 2μΜ 0.2μΜ 0.02μΜ
FK506, 1 73.9 59.4 25.5
7 48.4 34.3 34.4
11 1 1.2 17.7 2.2 Table 3.
Figure imgf000054_0001
Example 14 - Biological data - mouse in vivo oral and iv PK
To assess the pharmacokinetics of the compounds in an in vivo setting, compounds were dosed po at 10mg/kg and iv at 1 mg/kg to groups of male CD1 mice. Pharmacokinetic analysis was carried out as described in the materials and methods. Plots of the data are shown in figure 6, and the PK parameters are shown in table 4. As can be seen from this data, the systemic bioavailability of the ester series 7, 8, 9 and 10 are substantially reduced from FK506. Levels of 11 , the presumed primary metabolite, were also analysed, and revealed that substantial amounts of this compound were present in samples from the 7, 8, 9 and 10 PK studies (data not shown).
Table 4.
Figure imgf000054_0002
Example 15 - Biological data - caco-2 permeability and efflux
To look at the permeability of the compounds in vitro, a caco-2 permeability assay was used with 1 μΜ of test compounds, as described in the materials and methods. Attempts were made to analyse the ester series, 7, 8, 9 and 10, but difficulties were encountered with nonspecific binding, leading the results to be uninterpretable. As such, and to analyse the effect of removal of the 31 -H on the compounds being a substrate for Pgp, FK506 (1 ) and 31 - desmethoxy FK506 (5) were compared in the caco-2 assay. Data is shown in table 5, and suggests that removal of the 31 -H is beneficial in reducing the efflux of compounds, possibly via a reduced interaction with transporters such as Pgp. Table 5.
Figure imgf000055_0001
Example 16 - Mouse plasma stability
To confirm that the action of plasma enzymes (presumed to be esterases) was an important part of the rapid metabolism in mice, and to assess the relative rates of metabolism, mouse plasma stability studies were carried out, as described in the materials and methods, by spiking mouse plasma with test compounds at 2μΜ, to generate half lives for a series of compounds.
As seen in table 6, all of the ester compounds tested showed rapid metabolism, whereas FK506, 1 was relatively stable in mouse plasma.
Table 6.
Figure imgf000056_0001
Example 17 - Human blood stability
To look at the stability of the ester series of compounds in human blood, human blood stability studies were carried out, as described in the materials and methods, by spiking whole human blood with test compounds at 0.1 μΜ. The data from analysis was then plotted to compare the stability in human blood.
Once again, as seen in figure 7, all of the ester compounds tested showed rapid metabolism, with no parent remaining after 4 hours, whereas FK506, 1 was relatively stable in human blood.
Example 18 - Biological data - mouse in vivo aerosol PK
To assess the pharmacokinetics of the compounds when dosed topically to the lungs in an in vivo setting, 20mg of the test articles was dosed in a 100% ethanol vehicle by aerosol using a nebuliser to groups of 9 male CD1 mice (see materials and methods) for 10 minutes at 3ml_/10min. At the designated time points, lung and blood samples were taken and pharmacokinetic analysis was carried out as described in the materials and methods. Plots of the data are shown in figure 8, and the PK parameters are shown in table 8, with data generated using AUC calculations based on 0-24 hour timepoints (AUCiast)- As can be seen from this data, the lung:blood ratio is substantially improved in the ester series as compared to FK506. Levels of 11 , the presumed primary metabolite, were also analysed, and revealed that substantial amounts of this compound were also present in lung samples from the 7 and 9 PK studies, with much less in the blood samples.
Table 8 11, 11,
Parent Parent Parent metabolite 11, metabolite metabolite Blood AUC Lung AUC ratio Blood AUC Lung AUC ratio
Compound (ng*hi7ml_) (ng*hr/ml_) Blood:Lung (ng*hr/ml_) (ng*hr/mL) Blood:Lung
FK506, 1 9.3 476 51 x N/A N/A N/A
9 1 .55 862 556x <1 228 >228x
7 <1 310 >310x 5.23 382 73x
References
Aparicio, J. F., I. Molnar, et al. (1996). "Organization of the biosynthetic gene cluster for
rapamycin in Streptomyces hygroscopicus: analysis of the enzymatic domains in the modular polyketide synthase." Gene 169(1 ): 9-16.
Armstrong, V. W. and M. Oellerich (2001 ). "New developments in the immunosuppressive drug monitoring of cyclosporine, tacrolimus, and azathioprine." Clin Biochem 34(1 ): 9-16. Baker, H., A. Sidorowicz, et al. (1978). "Rapamycin (AY-22,989), a new antifungal antibiotic.
III. In vitro and in vivo evaluation." Journal of Antibiotics 31 (6): 539-545.
Bierman, M., R. Logan, et al. (1992). "Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to Streptomyces spp." Gene 116(1 ): 43-49.
Blanc, V., P. Gil, et al. (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(2): 191 -202.
Blanc, V., D. Lagneaux, et al. (1995). "Cloning and analysis of structural genes from
Streptomyces pristinaespiralis encoding enzymes involved in the conversion of pristinamycin llB to pristinamycin llA (PIU): PI synthase and NADH:riboflavin 5'- phosphate oxidoreductase." Journal of Bacteriology 177(18): 5206-5214.
Bodor, N. and P. Buchwald (2001 ). "Drug targeting by retrometabolic design: soft drugs and chemical delivery systems." J Recept Signal Transduct Res 21 (2-3): 287-310.
Bodor, N. and P. Buchwald (2005). "Ophthalmic drug design based on the metabolic activity of the eye: soft drugs and chemical delivery systems." AAPS J 7(4): E820-33.
Brizuela, L., G. Chrebet, et al. (1991 ). "Antifungal properties of the immunosuppressant FK- 506: identification of an FK-506-responsive yeast gene distinct from FKB1." Mol Cell
Biol 11 (9): 4616-26. Campbell, L, A. N. Abulrob, et al. (2003). "Constitutive expression of p-glycoprotein in normal lung alveolar epithelium and functionality in primary alveolar epithelial cultures." J
Pharmacol Exp Ther 304(1 ): 441 -52.
Cannell, R., Ed. (1998). Natural Products Isolation, Humana Press.
Cao, W., P. Mohacsi, et al. (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): 390-5.
Carlson, R. P., W. L. Baeder, et al. (1993). "Effects of orally administered rapamycin in animal models of arthritis and other autoimmune diseases." Ann N Y Acad Sci 685: 86-1 13.
Carlson, R. P., D. A. Hartman, et al. (1993). "Rapamycin, a potential disease-modifying
antiarthritic drug." J Pharmacol Exp Ther 266(2): 1 125-38.
Chambraud, B., C. Radanyi, et al. (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 (51 ): 32923-32929.
Chan, H. K. and N. Y. Chew (2003). "Novel alternative methods for the delivery of drugs for the treatment of asthma." Adv Drug Deliv Rev 55(7): 793-805.
Chang, J. Y., S. N. Sehgal, et al. (1991 ). "FK506 and rapamycin: novel pharmacological probes of the immune response." Trends in Pharmacological Sciences 12(6): 218-223.
Chen, J., X. F. Zheng, et al. (1995). "Identification of an 1 1 -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(1 1 ): 4947-4951 .
Choi, S. S., Y. A. Hur, et al. (2007). "Isolation of the biosynthetic gene cluster for tautomycetin, a linear polyketide T cell-specific immunomodulator from Streptomyces sp. CK4412."
Microbiology 153(Pt 4): 1095-102.
Corcoran, T. E. (2006). "Inhaled delivery of aerosolized cyclosporine." Adv Drug Deliv Rev
58(9-10): 1 1 19-27.
Cryan, S. A., N. Sivadas, et al. (2007). "In vivo animal models for drug delivery across the lung mucosal barrier." Adv Drug Deliv Rev 59(1 1 ): 1 133-51 .
Dalby, R. and J. Suman (2003). "Inhalation therapy: technological milestones in asthma
treatment." Adv Drug Deliv Rev 55(7): 779-91 .
DiLella, A. G. and R. J. Craig (1991 ). "Exon organization of the human FKBP-12 gene:
correlation with structural and functional protein domains." Biochemistry 30(35): 8512-7. Du, L, C. Sanchez, et al. (2000). "The biosynthetic gene cluster for the antitumor drug
bleomycin from Streptomyces verticillus ATCC15003 supporting functional interactions between nonribosomal peptide synthetases and a polyketide synthase." Chem Biol 7(8): 623-42.
Dudkin, L, M. B. Dilling, et al. (2001 ). "Biochemical correlates of rmTOR inhibition by the
rapamycin ester CCI-779 and tumor growth inhibition." Clin Cancer Res 7(6): 1758-64. Dumont, F. J., M. J. Staruch, et al. (1992). "The immunosuppressive and toxic effects of FK-
506 are mechanistically related: pharmacology of a novel antagonist of FK-506 and rapamycin." J Exp Med 176(3): 751 -60.
Dutton, C. J., S. P. Gibson, et al. (1991 ). "Novel avermectins produced by mutational
biosynthesis." Journal of Antibiotics 44(3): 357-365.
Egorin, M. J., T. F. Lagattuta, et al. (2002). "Pharmacokinetics, tissue distribution, and
metabolism of 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (NSC
707545) in CD2F1 mice and Fischer 344 rats." Cancer Chemother Pharmacol 49(1 ): 7-
19.
Fang, J., Y. Zhang, et al. (2008). "Cloning and characterization of the tetrocarcin A gene cluster from Micromonospora chalcea NRRL 1 1289 reveals a highly conserved strategy for tetronate biosynthesis in spirotetronate antibiotics." J Bacteriol 190(17): 6014-25.
Fehr, T., J. J. Sanglier, et al. (1996). "Antascomicins A, B, C, D and E. Novel FKBP12 binding compounds from a Micromonospora strain." J Antibiot (Tokyo) 49(3): 230-3.
Foey, A., P. Green, et al. (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.
Gaisser, S., J. Reather, et al. (2000). "A defined system for hybrid macrolide biosynthesis in
Saccharopolyspora erythraea." Molecular Microbiology 36(2): 391 -401 .
Garrity, G. M., B. K. Heimbuch, et al. (1993). "Genetic relationships among actinomycetes that produce the immunosuppressant macrolides FK506, FK520/FK523 and rapamycin." J
Ind Microbiol 12(1 ): 42-7.
Gregory, C. R., P. Huie, et al. (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): 1409-18.
Hanson, B. J. (2006). "Multiplexing Fluo-4 NW and a GeneBLAzer transcriptional assay for high-throughput screening of G-protein-coupled receptors." J Biomol Screen 11 (6): 644- 51 .
Hatanaka, H., T. Kino, et al. (1989). "FK-506 related compounds produced by Streptomyces tsukubaensis No. 9993." J Antibiot (Tokyo) 42(4): 620-2. Hatanaka, H., T. Kino, et al. (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 (1 1 ): 1592-601 .
Hegedus, C, G. Szakacs, et al. (2009). "Ins and outs of the ABCG2 multidrug transporter: an update on in vitro functional assays." Adv Drug Deliv Rev 61 (1 ): 47-56.
Izuishi, K., H. Wakabayashi, et al. (1997). "Effects of an immunosuppressive agent, tacrolimus (FK-506), on the activities of cytochrome P-450-linked monooxygenase systems in rat liver microsomes." Int J Biochem Cell Biol 29(6): 921 -8.
Kahan, B. D., J. Y. Chang, et al. (1991 ). "Preclinical evaluation of a new potent
immunosuppressive agent, rapamycin." Transplantation 52(2): 185-191 .
Kapturczak, M. H., H. U. Meier-Kriesche, et al. (2004). "Pharmacology of calcineurin
antagonists." Transplant Proc 36(2 Suppl): 25S-32S.
Kieser, T., M. J. Bibb, et al., Eds. (1999). Practical Streptomyces Genetics, John Innes
Foundation.
Kim, H. S. and Y. I. Park (2007). "Lipase activity and tacrolimus production in Streptomyces clavuligerus CKD 1 1 19 mutant strains." J Microbiol Biotechnol 17(10): 1638-44.
Kirchner, G. I., M. Winkler, et al. (2000). "Pharmacokinetics of SDZ RAD and cyclosporin
including their metabolites in seven kidney graft patients after the first dose of SDZ RAD." Br J Clin Pharmacol 50(5): 449-54.
Lechapt-Zalcman, E., I. Hurbain, et al. (1997). "MDR1 -Pgp 170 expression in human
bronchus." Eur Respir J 10(8): 1837-43.
Lowden, P. A. S. (1997). Studies on the Biosynthesis of Rapamycin. Churchill College,
University of Cambridge.
Lowden, P. A. S., B. Wilkinson, et al. (2001 ). "Origin and true nature of the starter unit for the rapamycin polyketide synthase." Angewandte Chemie-lnternational Edition 40(4): 777-
779.
Macian, F. (2005). "NFAT proteins: key regulators of T-cell development and function." Nat Rev Immunol 5(6): 472-84.
Margulies, M., M. Egholm, et al. (2005). "Genome sequencing in microfabricated high-density picolitre reactors." Nature 437(7057): 376-80.
Meingassner, J. G., M. Grassberger, et al. (1997). "A novel anti-inflammatory drug, SDZ ASM 981 , for the topical and oral treatment of skin diseases: in vivo pharmacology." Br J Dermatol 137(4): 568-76.
Morice, M. C, P. W. Serruys, et al. (2002). "A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization." N Engl J Med 346(23): 1773-
80. Motamedi, H., A. Shafiee, et al. (1996). "Characterization of methyltransferase and hydroxylase genes involved in the biosynthesis of the immunosuppressants FK506 and FK520."
Journal of Bacteriology 178(17): 5243-5248.
Muramatsu, H., M. SI, et al. (2005). "Phylogenetic Analysis of Immunosuppressant FK506- Producing Streptomycete Strains." Actinomycetologica 19: 33-39.
Qureshi, S. A. (2007). "Beta-lactamase: an ideal reporter system for monitoring gene
expression in live eukaryotic cells." Biotechniques 42(1 ): 91 -6.
Rabinovitch, A., W. L. Suarez-Pinzon, et al. (2002). "Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice." Diabetes 51 (3): 638-45.
Reitamo, S., P. Spuls, et al. (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., K. K. Wallace, et al. (1997). "Biosynthesis of the shikimate-derived starter unit of the immunosuppressant ascomycin: stereochemistry of the 1 ,4-conjugate
elimination." Journal of Antibiotics 50(8): 701 -703.
Roy, J. N., A. Barama, et al. (2006). "Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients." Pharmacogenet Genomics
16(9): 659-65.
Rustin, M. H. (2007). "The safety of tacrolimus ointment for the treatment of atopic dermatitis: a review." Br J Dermatol 157(5): 861 -73.
Saeki, T., K. Ueda, et al. (1993). "Human P-glycoprotein transports cyclosporin A and FK506."
J Biol Chem 268(9): 6077-80.
Sambrook, J. and D. Russel, Eds. (2001 ). Molecular Cloning: A laboratory manual (third
edition), Cold Spring Harbor Laboratory Press.
Schreiber, S. L. and G. R. Crabtree (1992). "The mechanism of action of cyclosporine A and
FK506." Immunology Today 13(4): 136-142.
Shimizu, M. (2005). Astellas R&D Meeting - Clinical Development.
Sigal, N. H., F. Dumont, et al. (1991 ). "Is cyclophilin involved in the immunosuppressive and nephrotoxic mechanism of action of cyclosporin A?" J Exp Med 173(3): 619-28.
Smyth, H. D. (2003). "The influence of formulation variables on the performance of alternative propellant-driven metered dose inhalers." Adv Drug Deliv Rev 55(7): 807-28.
Steiner, J. P., G. S. Hamilton, et al. (1997). "Neurotrophic immunophilin ligands stimulate
structural and functional recovery in neurodegenerative animal models." Proc Natl Acad Sci U S A 94(5): 2019-24. Szakacs, G., A. Varadi, et al. (2008). "The role of ABC transporters in drug absorption, distribution, metabolism, excretion and toxicity (ADME-Tox)." Drug Discov Today 13(9- 10): 379-93.
Tamura, S., Y. Tokunaga, et al. (2003). "The site-specific transport and metabolism of
tacrolimus in rat small intestine." J Pharmacol Exp Ther 306(1 ): 310-6.
Wagner, R., T. A. Rhoades, et al. (1998). "32-Ascomycinyloxyacetic acid derived
immunosuppressants. Independence of immunophilin binding and immunosuppressive potency." J Med Chem 41 (1 1 ): 1764-76.
Wallace, K. K., K. A. Reynolds, et al. (1994). "Biosynthetic studies of ascomycin (FK520): formulation of the (1 R,3R4R)-3,4-dihydroxycyclohexanecarboxylic acid-derived moiety."
JACS 116(25): 1 1600-1 1601 .
Wu, K., L. Chung, et al. (2000). "The FK520 gene cluster of Streptomyces hygroscopicus var. ascomyceticus (ATCC 14891 ) contains genes for biosynthesis of unusual polyketide extender units." Gene 251 (1 ): 81 -90.
Yan, Z. and G. W. Caldwell (2001 ). "Metabolism profiling, and cytochrome P450 inhibition & induction in drug discovery." Curr Top Med Chem 1 (5): 403-25.
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) or a pharmaceutically acceptable salt thereof:
Figure imgf000063_0001
wherein
R2 represents -CH3, -CH2CH3! or -CH2CH=CH2;
Ri represents C1 -10 alkyi wherein one or more carbon atoms are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups;
or Ri represents aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl; and
R3 represents a group -(CO)XC1 -10 alkyi wherein one or more carbon atoms of the alkyi chain are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups and x represents 0 or 1 ;
or R3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl;
or Ri and R3 are linked to form a lactone ring having 5 to 7 ring members which may optionally contain one or more additional heteroatoms selected from O, N and S.
2. A compound according to claim 1 wherein R2 represents -CH2CH=CH2.
3. A compound according to claim 1 or claim 2 wherein R-i represents C1 -4alkyl.
4. A compound according to any one of claims 1 to 3 wherein R3 represents H.
5. A compound according to claim 1 selected from: WO 2011/045593
Figure imgf000064_0001
Figure imgf000065_0001
and pharmaceutically acceptable salts of any one thereof.
6. A compound according to any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof for use as a pharmaceutical.
7. A pharmaceutical composition comprising a compound of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable diluents or carriers.
8. A compound of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof for use in the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases, rejection of transplant, atopic dermatitis, asthma, uveitis, psoriasis and/or inflammatory bowel diseases.
9. Use of a compounds of formula (I) according to any one of claims 1 to 5 or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases, rejection of transplant, atopic dermatitis, asthma, uveitis, psoriasis and/or inflammatory bowel diseases.
10. A method for the treatment or prevention of immune disorders, inflammatory diseases, fungal diseases, rejection of transplant, atopic dermatitis, asthma, uveitis, psoriasis and/or inflammatory bowel diseases which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a compound of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof.
11. A process for the preparation of a compound of formula (I) or a pharmaceutically acceptable salt thereof which comprises reacting a compound of formula (II)
Figure imgf000066_0001
(II)
with a compound of formula (III)
Figure imgf000066_0002
wherein R2 represents -CH3, -CH2CH3! or -CH2CH=CH2;
Ri represents H; and
R3 represents a group -(CO)XC1 -10 alkyi wherein one or more carbon atoms of the alkyi chain are optionally replaced by a heteroatom selected from O, N and S and which is optionally substituted by one or more halogen atoms or =0 groups and x represents 0 or 1 ;
or R3 represents H, aryl, C1 -4alkylaryl, heteroaryl or C1 -4heteroaryl.
13. A compound according to claim 12 wherein R-i represents C1 -4alkyl.
14. A compound according to claim 12 which is:
Figure imgf000067_0001
or a salt thereof.
PCT/GB2010/051713 2009-10-12 2010-10-12 Novel macrocyles and methods for their production WO2011045593A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0917816.1 2009-10-12
GBGB0917816.1A GB0917816D0 (en) 2009-10-12 2009-10-12 Novel compounds and methods for their production

Publications (1)

Publication Number Publication Date
WO2011045593A1 true WO2011045593A1 (en) 2011-04-21

Family

ID=41402876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2010/051713 WO2011045593A1 (en) 2009-10-12 2010-10-12 Novel macrocyles and methods for their production

Country Status (2)

Country Link
GB (1) GB0917816D0 (en)
WO (1) WO2011045593A1 (en)

Citations (22)

* 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
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
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
US5563172A (en) 1991-09-05 1996-10-08 Abbott Laboratories Macrocyclic amide and urea immunomodulators
US6352998B2 (en) 1994-10-26 2002-03-05 Novartis Ag Pharmaceutical compositions
US6403122B1 (en) 1996-09-12 2002-06-11 Galena As Immunomodulatory pharmaceutical composition adopted to form lyotrophic liquid crystals in contact with an aqueous phase
US6515016B2 (en) 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
US6598153B1 (en) 1999-12-10 2003-07-22 International Business Machines Corporation Processor and method that accelerate evaluation of pairs of condition-setting and branch instructions
US6673808B1 (en) 1998-04-27 2004-01-06 Fujisawa Pharmaceutical Co., Ltd. Medicinal compositions
WO2004007709A2 (en) 2002-07-16 2004-01-22 Biotica Technology Limited Production of polyketides
WO2004110335A1 (en) 2003-06-10 2004-12-23 Astellas Pharma Inc. Aerosol preparation comprising sealed container and enclosed therein aerosol composition containing macrolide compound
WO2005063242A1 (en) 2003-12-30 2005-07-14 Astellas Pharma Inc. Use of macrolides for treating or preventing airflow obstruction
US20060160838A1 (en) * 2005-01-20 2006-07-20 Array Biopharma Inc. Macrocyclic analogs for the treatment of immunoregulatory disorders and respiratory diseases

Patent Citations (25)

* 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
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5731320A (en) 1991-09-05 1998-03-24 Abbott Laboratories Macrocyclic amide and urea immunomodulators
US5643918A (en) 1991-09-05 1997-07-01 Abbott Laboratories Marcrocyclic amide and urea immunomodulators
US5563172A (en) 1991-09-05 1996-10-08 Abbott Laboratories Macrocyclic amide and urea immunomodulators
US5399163A (en) 1992-07-24 1995-03-21 Bioject Inc. Needleless hypodermic injection methods and device
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US6352998B2 (en) 1994-10-26 2002-03-05 Novartis Ag Pharmaceutical compositions
US6403122B1 (en) 1996-09-12 2002-06-11 Galena As Immunomodulatory pharmaceutical composition adopted to form lyotrophic liquid crystals in contact with an aqueous phase
US6515016B2 (en) 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
US6673808B1 (en) 1998-04-27 2004-01-06 Fujisawa Pharmaceutical Co., Ltd. Medicinal compositions
US6598153B1 (en) 1999-12-10 2003-07-22 International Business Machines Corporation Processor and method that accelerate evaluation of pairs of condition-setting and branch instructions
WO2004007709A2 (en) 2002-07-16 2004-01-22 Biotica Technology Limited Production of polyketides
WO2004110335A1 (en) 2003-06-10 2004-12-23 Astellas Pharma Inc. Aerosol preparation comprising sealed container and enclosed therein aerosol composition containing macrolide compound
WO2005063242A1 (en) 2003-12-30 2005-07-14 Astellas Pharma Inc. Use of macrolides for treating or preventing airflow obstruction
US20060160838A1 (en) * 2005-01-20 2006-07-20 Array Biopharma Inc. Macrocyclic analogs for the treatment of immunoregulatory disorders and respiratory diseases

Non-Patent Citations (76)

* Cited by examiner, † Cited by third party
Title
"Molecular Cloning: A laboratory manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
"Natural Products Isolation", 1998, HUMANA PRESS
"Practical Streptomyces Genetics", 1999, JOHN INNES FOUNDATION
APARICIO, J. F.; MOLNÁR ET AL.: "Organization of the biosynthetic gene cluster for rapamycin in Streptomyces hygroscopicus: analysis of the enzymatic domains in the modular polyketide synthase.", GENE, vol. 169, no. 1, 1996, pages 9 - 16, XP004042980, DOI: doi:10.1016/0378-1119(95)00800-4
ARMSTRONG, V. W.; M. OELLERICH; 2001: "New developments in the immunosuppressive drug monitoring of cyclosporine, tacrolimus, and azathioprine.", CLIN BIOCHEM, vol. 34, no. 1, pages 9 - 16, XP027214182
BAKER, H.; A. SIDOROWICZ ET AL.: "Rapamycin (AY-22,989), a new antifungal antibiotic. III. In vitro and in vivo evaluation.", JOURNAL OF ANTIBIOTICS, vol. 31, no. 6, 1978, pages 539 - 545
BIERMAN, M.; R. LOGAN ET AL.: "Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to Streptomyces spp.", GENE, vol. 116, no. 1, 1992, pages 43 - 49, XP023542508, DOI: doi:10.1016/0378-1119(92)90627-2
BLANC, V.; D. LAGNEAUX ET AL.: "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, no. 18, 1995, pages 5206 - 5214
BLANC, V.; P. GIL ET AL.: "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, no. 2, 1997, pages 191 - 202, XP002934995, DOI: doi:10.1046/j.1365-2958.1997.2031574.x
BODOR, N.; P. BUCHWALD: "Drug targeting by retrometabolic design: soft drugs and chemical delivery systems.", J RECEPT SIGNAL TRANSDUCT RES, vol. 21, no. 2-3, 2001, pages 287 - 310
BODOR, N.; P. BUCHWALD: "Ophthalmic drug design based on the metabolic activity of the eye: soft drugs and chemical delivery systems.", AAPS J, vol. 7, no. 4, 2005, pages E820 - 33, XP035718775, DOI: doi:10.1208/aapsj070479
BRIZUELA, L.; G. CHREBET ET AL.: "Antifungal properties of the immunosuppressant FK-506: identification of an FK-506-responsive yeast gene distinct from FKB1.", MOL CELL BIOL, vol. 11, no. 9, 1991, pages 4616 - 26
CAMPBELL, L.; A. N. ABULROB ET AL.: "Constitutive expression of p-glycoprotein in normal lung alveolar epithelium and functionality in primary alveolar epithelial cultures.", J PHARMACOL EXP THER, vol. 304, no. 1, 2003, pages 441 - 52
CAO, W.; P. MOHACSI ET AL.: "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 - 5, XP000600683
CARLSON, R. P.; D. A. HARTMAN ET AL.: "Rapamycin, a potential disease-modifying antiarthritic drug.", J PHARMACOL EXP THER, vol. 266, no. 2, 1993, pages 1125 - 38
CARLSON, R. P.; W. L. BAEDER ET AL.: "Effects of orally administered rapamycin in animal models of arthritis and other autoimmune diseases.", ANN N Y ACAD SCI, vol. 685, 1993, pages 86 - 113
CHAMBRAUD, B.; C. RADANYI ET AL.: "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, no. 51, 1996, pages 32923 - 32929
CHAN, H. K.; N. Y. CHEW: "Novel alternative methods for the delivery of drugs for the treatment of asthma.", ADV DRUG DELIV REV, vol. 55, no. 7, 2003, pages 793 - 805, XP055075721, DOI: doi:10.1016/S0169-409X(03)00078-4
CHANG, J. Y.; S. N. SEHGAL ET AL.: "FK506 and rapamycin: novel pharmacological probes of the immune response.", TRENDS IN PHARMACOLOGICAL SCIENCES, vol. 12, no. 6, 1991, pages 218 - 223, XP023860665, DOI: doi:10.1016/0165-6147(91)90555-7
CHEN, J.; X. F. ZHENG ET AL.: "Identification of an 11-kDa FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-associated protein and characterization of a critical serine residue.", PROCEEDINQS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 92, no. 11, 1995, pages 4947 - 4951, XP002018161, DOI: doi:10.1073/pnas.92.11.4947
CHEN, Y.-W.; SMITH M.L.; SHEETS, M.; BALLARON, S.; TREVILLYAN, J.M.; BURKE, S.E.; ROSENBERG, T.; HENRY, C.; WAGNER, R.; BAUCH, J.: "Zotarolimus, a Novel Sirolimus Analogue With Potent Anti-proliferative Activity on Coronary Smooth Muscle Cells and Reduced Potential for Systemic Immunosuppression", JOURNAL OF CARDIOVASCULAR PHARMACOLOGY, vol. 49, no. 4, 2007, pages 228 - 235, XP002613587, ISSN: 1533-4023 *
CHOI, S. S.; Y. A. HUR ET AL.: "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 - 102
CORCORAN, T. E.: "Inhaled delivery of aerosolized cyclosporine.", ADV DRUG DELIV REV, vol. 58, no. 9-10, 2006, pages 1119 - 27, XP024892122, DOI: doi:10.1016/j.addr.2006.07.016
CRYAN, S. A.; N. SIVADAS ET AL.: "In vivo animal models for drug delivery across the lung mucosal barrier.", ADV DRUG DELIV REV, vol. 59, no. 11, 2007, pages 1133 - 51
DALBY, R.; J. SUMAN: "Inhalation therapy: technological milestones in asthma treatment.", ADV DRUG DELIV REV, vol. 55, no. 7, 2003, pages 779 - 91
DILELLA, A. G.; R. J. CRAIG: "Exon organization of the human FKBP-12 gene: correlation with structural and functional protein domains.", BIOCHEMISTRY, vol. 30, no. 35, 1991, pages 8512 - 7
DU, L.; C. SANCHEZ ET AL.: "The biosynthetic gene cluster for the antitumor drug bleomycin from Streptomyces verticillus ATCC15003 supporting functional interactions between nonribosomal peptide synthetases and a polyketide synthase.", CHEM BIOL, vol. 7, no. 8, 2000, pages 623 - 42
DUDKIN, L.; M. B. DILLING ET AL.: "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, XP001093805
DUMONT, F. J.; M. J. STARUCH ET AL.: "The immunosuppressive and toxic effects of FK-506 are mechanistically related: pharmacology of a novel antagonist of FK-506 and rapamycin.", J EXP MED, vol. 176, no. 3, 1992, pages 751 - 60, XP000606993, DOI: doi:10.1084/jem.176.3.751
DUTTON, C. J.; S. P. GIBSON ET AL.: "Novel avermectins produced by mutational biosynthesis.", JOURNAL OF ANTIBIOTICS, vol. 44, no. 3, 1991, pages 357 - 365, XP002079430
EGORIN, M. J.; T. F. LAGATTUTA ET AL.: "Pharmacokinetics, tissue distribution, and metabolism of 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (NSC 707545) in CD2F1 mice and Fischer 344 rats.", CANCER CHEMOTHER PHARMACOL, vol. 49, no. 1, 2002, pages 7 - 19
FANG, J.; Y. ZHANG ET AL.: "Cloning and characterization of the tetrocarcin A gene cluster from Micromonospora chalcea NRRL 11289 reveals a highly conserved strategy for tetronate biosynthesis in spirotetronate antibiotics.", J BACTERIOL, vol. 190, no. 17, 2008, pages 6014 - 25
FEHR, T.; J. J. SANGLIER ET AL.: "Antascomicins A, B, C, D and E. Novel FKBP12 binding compounds from a Micromonospora strain.", J ANTIBIOT (TOKYO), vol. 49, no. 3, 1996, pages 230 - 3
FOEY, A.; P. GREEN ET AL.: "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, XP021020652, DOI: doi:10.1186/ar385
GAISSER, S.; J. REATHER ET AL.: "A defined system for hybrid macrolide biosynthesis in Saccharopolyspora erythraea.", MOLECULAR MICROBIOLOGY, vol. 36, no. 2, 2000, pages 391 - 401, XP002210930, DOI: doi:10.1046/j.1365-2958.2000.01856.x
GARRITY, G. M.; B. K. HEIMBUCH ET AL.: "Genetic relationships among actinomycetes that produce the immunosuppressant macrolides FK506, FK520/FK523 and rapamycin.", J IND MICROBIOL, vol. 12, no. L, 1993, pages 42 - 7, XP000906748, DOI: doi:10.1007/BF01570127
GREGORY, C. R.; P. HUIE ET AL.: "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 - 18, XP000569310
HANSON, B. J.: "Multiplexing Fluo-4 NW and a GeneBLAzer transcriptional assay for high-throughput screening of G-protein-coupled receptors.", J BIOMOL SCREEN, vol. 11, no. 6, 2006, pages 644 - 51, XP008103072, DOI: doi:10.1177/1087057106289982
HATANAKA, H.; T. KINO ET AL.: "FK-506 related compounds produced by Streptomyces tsukubaensis No. 9993", J ANTIBIOT (TOKYO), vol. 42, no. 4, 1989, pages 620 - 2, XP009142387
HATANAKA, H.; T. KINO ET AL.: "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, XP009064959
HEGEDUS, C.; G. SZAKACS ET AL.: "Ins and outs of the ABCG2 multidrug transporter: an update on in vitro functional assays.", ADV DRUG DELIV REV, vol. 61, no. 1, 2009, pages 47 - 56, XP025908638, DOI: doi:10.1016/j.addr.2008.09.007
IUPAC: "Aryl groups", INTERNET CITATION, 1997, pages 1, XP002582725, Retrieved from the Internet <URL:http://www.iupac.org/goldbook/A00464.pdf> [retrieved on 20100519] *
IZUISHI, K.; H. WAKABAYASHI ET AL.: "Effects of an immunosuppressive agent, tacrolimus (FK-506), on the activities of cytochrome P-450-linked monooxygenase systems in rat liver microsomes.", INT J BIOCHEM CELL BIOL, vol. 29, no. 6, 1997, pages 921 - 8
KAHAN, B. D.; J. Y. CHANG ET AL.: "Preclinical evaluation of a new potent immunosuppressive agent, rapamycin.", TRANSPLANTATION, vol. 52, no. 2, 1991, pages 185 - 191
KAPTURCZAK, M. H.; H. U. MEIER-KRIESCHE ET AL.: "Pharmacology of calcineurin antagonists.", TRANSPLANT PROC, vol. 36, 2004, pages 25S - 32S
KIM, H. S.; Y. . PARK: "Lipase activity and tacrolimus production in Streptomyces clavuligerus CKD 1119 mutant strains.", J MICROBIOL BIOTECHNOL, vol. 17, no. 10, 2007, pages 1638 - 44
KIRCHNER, G. I.; M. WINKLER ET AL.: "Pharmacokinetics of SDZ RAD and cyclosporin including their metabolites in seven kidney graft patients after the first dose of SDZ RAD.", BR J CLIN PHARMACOL, vol. 50, no. 5, 2000, pages 449 - 54, XP001066397, DOI: doi:10.1046/j.1365-2125.2000.00293.x
LECHAPT-ZALCMAN, E.; HURBAIN ET AL.: "MDR1-Pgp 170 expression in human bronchus.", EUR RESPIR J, vol. 10, no. 8, 1997, pages 1837 - 43
LOWDEN, P. A. S.: "Studies on the Biosynthesis of Rapamycin", 1997, CHURCHILL COLLEGE, UNIVERSITY OF CAMBRIDGE
LOWDEN, P. A. S.; B. WILKINSON ET AL.: "Origin and true nature of the starter unit for the rapamycin polyketide synthase.", ANGEWANDTE CHEMIE-INTERNATIONAL EDITION, vol. 40, no. 4, 2001, pages 777 - 779
MACIAN, F.: "NFAT proteins: key regulators of T-cell development and function.", NAT REV IMMUNOL, vol. 5, no. 6, 2005, pages 472 - 84
MARGULIES, M.; M. EGHOLM ET AL.: "Genome sequencing in microfabricated high-density picolitre reactors.", NATURE, vol. 437, no. 7057, 2005, pages 376 - 80
MEINGASSNER, J. G.; M. GRASSBERGER ET AL.: "A novel anti-inflammatory drug, SDZ ASM 981, for the topical and oral treatment of skin diseases: in vivo pharmacology.", BR J DERMATOL, vol. 137, no. 4, 1997, pages 568 - 76, XP008008917, DOI: doi:10.1111/j.1365-2133.1997.tb03788.x
MORICE, M. C.; P. W. SERRUYS ET AL.: "A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization.", N ENQL J MED, vol. 346, no. 23, 2002, pages 1773 - 80, XP009073187, DOI: doi:10.1056/NEJMoa012843
MOTAMEDI, H.; A. SHAFIEE ET AL.: "Characterization of methyltransferase and hydroxylase genes involved in the biosynthesis of the immunosuppressants FK506 and FK520.", JOURNAL OF BACTERIOLOGY, vol. 178, no. 17, 1996, pages 5243 - 5248, XP002137077
MURAMATSU, H.; M. SI ET AL.: "Phylogenetic Analysis of Immunosuppressant FK506-Producing Streptomycete Strains.", ACTINOMYCETOLOQICA, vol. 19, 2005, pages 33 - 39
QURESHI, S. A.: "Beta-lactamase: an ideal reporter system for monitoring gene expression in live eukaryotic cells.", BIOTECHNIQUES, vol. 42, no. 1, 2007, pages 91 - 6
RABINOVITCH, A.; W. L. SUAREZ-PINZON ET AL.: "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
REITAMO, S.; P. SPULS ET AL.: "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, vol. 145, no. 3, 2001, pages 438 - 45
REYNOLDS, K. A.; K. K. WALLACE ET AL.: "Biosynthesis of the shikimate-derived starter unit of the immunosuppressant ascomycin: stereochemistry of the 1,4-conjugate elimination.", JOURNAL OF ANTIBIOTICS, vol. 50, no. 8, 1997, pages 701 - 703
ROY, J. N.; A. BARAMA ET AL.: "Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients.", PHARMACOGENET GENOMICS, vol. 16, no. 9, 2006, pages 659 - 65
RUSTIN, M. H.: "The safety of tacrolimus ointment for the treatment of atopic dermatitis: a review.", BR J DERMATOL, vol. 157, no. 5, 2007, pages 861 - 73
SAEKI, T.; K. UEDA ET AL.: "Human P-glycoprotein transports cyclosporin A and FK506.", J BIOL CHEM, vol. 268, no. 9, 1993, pages 6077 - 80
SCHREIBER, S. L.; G. R. CRABTREE: "The mechanism of action of cyclosporine A and FK506.", IMMUNOLOGY TODAY, vol. 13, no. 4, 1992, pages 136 - 142
SHIMIZU, M., ASTELLAS R&D MEETING - CLINICAL DEVELOPMENT, 2005
SIGAL, N. H.; F. DUMONT ET AL.: "Is cyclophilin involved in the immunosuppressive and nephrotoxic mechanism of action of cyclosporin A?", J EXP MED, vol. 173, no. 3, 1991, pages 619 - 28, XP002449993, DOI: doi:10.1084/jem.173.3.619
SMYTH, H. D.: "The influence of formulation variables on the performance of alternative propellant-driven metered dose inhalers.", ADV DRUG DELIV REV, vol. 55, no. 7, 2003, pages 807 - 28
STEINER, J. P.; G. S. HAMILTON ET AL.: "Neurotrophic immunophilin ligands stimulate structural and functional recovery in neurodegenerative animal models.", PROC NATL ACAD SCI US A, vol. 94, no. 5, 1997, pages 2019 - 24, XP002525434, DOI: doi:10.1073/pnas.94.5.2019
SZAKACS, G.; A. VARADI ET AL.: "The role of ABC transporters in drug absorption, distribution, metabolism, excretion and toxicity (ADME-Tox).", DRUG DISCOV TODAY, vol. 13, no. 9-10, 2008, pages 379 - 93, XP022649912, DOI: doi:10.1016/j.drudis.2007.12.010
TAMURA, S.; Y. TOKUNAGA ET AL.: "The site-specific transport and metabolism of tacrolimus in rat small intestine.", J PHARMACOL EXP THER, vol. 306, no. 1, 2003, pages 310 - 6
WAGNER, R.; RHOADES, T.A.; SUN OR, S.; LANES, B.C.; HSIEH, G.; MOLLISON, K.W.; LULY, J.R.: "32-Ascomycinyloxyacetic Acid Derived Immunosuppressants. Independence of immunophilin Binding and Immunosuppressive Potency", JOURNAL OF MEDICINAL CHEMISTRY, vol. 41, 1998, pages 1764 - 1776, XP002613586, ISSN: 0022-2623 *
WAGNER, R.; T. A. RHOADES ET AL.: "32-Ascomycinyloxyacetic acid derived immunosuppressants. Independence of immunophilin binding and immunosuppressive potency.", J MED CHEM, vol. 41, no. 11, 1998, pages 1764 - 76, XP002613586, DOI: doi:10.1021/jm960066y
WALLACE, K. K.; K. A. REYNOLDS ET AL.: "Biosynthetic studies of ascomycin (FK520): formulation of the (1 R,3R4R)-3,4-dihydroxycyclohexanecarboxylic acid-derived moiety.", JACS, vol. 116, no. 25, 1994, pages 11600 - 11601
WU, K.; L. CHUNG ET AL.: "The FK520 gene cluster of Streptomyces hygroscopicus var. ascomyceticus (ATCC 14891) contains genes for biosynthesis of unusual polyketide extender units.", GENE, vol. 251, no. 1, 2000, pages 81 - 90, XP004202089, DOI: doi:10.1016/S0378-1119(00)00171-2
YAN, Z.; G. W. CALDWELL: "Metabolism profiling, and cytochrome P450 inhibition & induction in drug discovery.", CURR TOP MED CHEM, vol. 1, no. 5, 2001, pages 403 - 25
YURA H ET AL: "Synthesis and pharmacokinetics of a novel macromolecular prodrug of Tacrolimus (FK506), FK506-dextran conjugate", JOURNAL OF CONTROLLED RELEASE, ELSEVIER, AMSTERDAM, NL, vol. 57, no. 1, 1 January 1999 (1999-01-01), pages 87 - 99, XP004155643, ISSN: 0168-3659, DOI: DOI:10.1016/S0168-3659(98)00150-3 *

Also Published As

Publication number Publication date
GB0917816D0 (en) 2009-11-25

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.
JP5422788B2 (en) 39-desmethoxy derivative of rapamycin
US8962329B2 (en) Gene cluster
WO2010004304A1 (en) Novel compounds and methods for their production
AU2005219822A1 (en) Macrolides and methods for producing same
EP3019504A2 (en) Novel compounds
EP2797929B1 (en) Novel rapamycin analogue
EP1720548A1 (en) Non-immunosuppressive immunophilin ligands as neuroprotective and/or neuroregenerative agents
US8030325B2 (en) Biosynthetic gene cluster for the production of a complex polyketide
US20090209572A1 (en) 36-Des(3-Methoxy-4-Hydroxycyclohexyl) 36-(3-Hydroxycycloheptyl) Derivatives of Rapamycin for the Treatment of Cancer and Other Disorders
WO2011021036A1 (en) Polyketides analogues and methods for their production
JP2009536181A (en) 18,21-didesoxymacbecin derivatives for cancer treatment
WO2012089349A2 (en) Novel polyketide compounds and methods of making same
WO2011045594A1 (en) Novel macrocycles and methods for their production
WO2011045593A1 (en) Novel macrocyles and methods for their production
US20100022766A1 (en) Biosynthetic gene cluster for the production of a complex polyketide
WO2010106366A1 (en) Fk506 and fk520 analogues and their pharmaceutical uses
CA2651411A1 (en) 39-desmethoxy-39-methyl derivatives of rapamycin
JP2010509307A (en) 18,21-didesoxymacbecin derivatives for the treatment of cancer
MXPA06009994A (en) Macrolides and methods for producing same

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: 10766101

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 18.07.2012)

122 Ep: pct application non-entry in european phase

Ref document number: 10766101

Country of ref document: EP

Kind code of ref document: A1