US20040220156A1 - Novel anti-inflammatory androstane derivative compositions - Google Patents

Novel anti-inflammatory androstane derivative compositions Download PDF

Info

Publication number
US20040220156A1
US20040220156A1 US10/853,500 US85350004A US2004220156A1 US 20040220156 A1 US20040220156 A1 US 20040220156A1 US 85350004 A US85350004 A US 85350004A US 2004220156 A1 US2004220156 A1 US 2004220156A1
Authority
US
United States
Prior art keywords
composition
compound
formula
crystalline
guest molecule
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/853,500
Inventor
Keith Biggadike
Steven Coote
Andrew Craig
Victor Jacewicz
Michael Millan
John Seager
Andrew Theophilus
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0019172.6A external-priority patent/GB0019172D0/en
Priority claimed from US09/958,050 external-priority patent/US7101866B2/en
Priority claimed from US10/067,010 external-priority patent/US6777399B2/en
Priority claimed from US10/200,364 external-priority patent/US6858593B2/en
Application filed by Individual filed Critical Individual
Priority to US10/853,500 priority Critical patent/US20040220156A1/en
Publication of US20040220156A1 publication Critical patent/US20040220156A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J17/00Normal steroids containing carbon, hydrogen, halogen or oxygen, having an oxygen-containing hetero ring not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a novel composition containing an anti-inflammatory and anti-allergic compound of the androstane series and to processes for its preparation.
  • the present invention also relates to pharmaceutical formulations containing the composition and to therapeutic uses thereof, particularly for the treatment of inflammatory and allergic conditions.
  • Glucocorticoids which have anti-inflammatory properties are known and are widely used for the treatment of inflammatory disorders or diseases such as asthma and rhinitis.
  • U.S. Pat. No. 4,335,121 discloses 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -(1-oxopropoxy)-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (known by the generic name of fluticasone propionate) and derivatives thereof.
  • fluticasone propionate known by the generic name of fluticasone propionate
  • glucocorticoids include suppression of the Hypothalamic-Pituitary-Adrenal (HPA) axis, effects on bone growth in children and on bone density in the elderly, ocular complications (cataract formation and glaucoma) and skin atrophy.
  • HPA Hypothalamic-Pituitary-Adrenal
  • Certain glucocorticoid compounds also have complex paths of metabolism wherein the production of active metabolites may make the pharmacodynamics and pharmacokinetics of such compounds difficult to understand. Whilst the modern steroids are very much safer than those originally introduced, it remains an object of research to produce new molecules which have excellent anti-inflammatory properties, with predictable pharmacokinetic and pharmacodynamic properties, with an attractive side effect profile, and with a convenient treatment regime.
  • a crystalline chemical composition comprising a compound of formula (I)
  • composition of the invention in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P2 1 2 1 2 1 having unit cell dimensions of about 7.7 ⁇ 0.6 ⁇ , 13.7 ⁇ 0.7 ⁇ , and 37 ⁇ 3 ⁇ when determined at 120K (hereinafter “a composition of the invention”).
  • the crystalline lattice is stabilised by a hydrogen bonding interaction between the hydrogen atom of the C11 hydroxy on the compound of formula (I) with the oxygen of the C3 carbonyl on a second molecule of the compound of formula (I).
  • FIG. 1 shows the spacial arrangement of 4 molecules of steroid and 8 guests within a single unit cell for an example composition. Detail of the hydrogen bond interaction between the steroid molecules is shown in FIG. 3.
  • FIG. 2 shows the conformation of the steroid molecule with 2 guest molecules present.
  • compositions of the invention when crystallographically pure also preferably exhibit one or more of the following 6 features when determined at ambient temperature (eg around 295K):
  • the chemical name of the compound of formula (I) is 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester.
  • the compound of formula (I) and compositions thereof have potentially beneficial anti-inflammatory or anti-allergic effects, particularly upon topical administration, demonstrated by, for example, its ability to bind to the glucocorticoid receptor and to illicit a response via that receptor, with long acting effect.
  • the compound of formula (I) and compositions thereof is useful in the treatment of inflammatory and/or allergic disorders, especially in once-per-day therapy.
  • Space group P2 1 2 1 2 1 is characterised by angles of 90° being present in each of the 3 axes.
  • the compound of formula (I) can form a crystalline composition of characteristic space group, unit cell dimensions and crystalline structure as evidenced by X-ray diffraction with a number of guest molecules.
  • the guest molecule preferably has a relative molecular weight in the range 16 to 150, more preferably 60 to 130, especially 70 to 120.
  • the guest molecule is a liquid at ambient temperature and pressure (eg 295K, 1.013 ⁇ 10 5 Pa).
  • guest molecules which are a liquid under pressure may also be capable of acting as a guest molecule (especially under pressurised conditions).
  • Substances which are solids at ambient temperature and pressure are also included.
  • Suitable guest molecules include solvents e.g.:
  • Aromatic compounds eg benzene and substituted derivatives, especially benzene substituted with one or more (eg 1 or 2) halogen or C 1-4 alkyl groups, for example toluene, o-xylene, m-xylene, fluorobenzene, chlorobenzene, ethylbenzene.
  • Preferred guest molecules are pharmaceutically acceptable substances and, as described below, compositions of the invention containing them may be used in therapy. However even if the guest molecule is not pharmaceutically acceptable then such compositions may be useful in the preparation of other compositions containing compound of formula (I), for example, other compositions of the invention containing guest molecules that are pharmaceutically acceptable or compound of formula (I) in unsolvated form.
  • the stoichiometry of the composition will usually be such that the ratio of compound to formula (I) to guest molecule, in molar terms, is 1:5-0.25, preferably 1:3.0-0.3, more preferably 1:2.2-0.6, especially 1:2.2-0.8.
  • a composition of the invention has a crystal structure which is quite distinct from that of compound of formula (I) in the absence of a guest molecule, eg. the compound of formula (I) as unsolvated polymorph Form 1 which has a space group of P2 1 (i.e. two of the axis angles are 90°) and cell dimensions of 7.6, 14.1, 11.8 ⁇ when determined at 150K.
  • a threshold level which will differ from guest to guest
  • the crystal structure of the composition starts to break down and converts to that of the structure of an unsolvated compound of formula (I), typically unsolvated polymorph Form 2 or 3.
  • the unit cell dimentions are about 7.7 ⁇ 0.4 ⁇ , 13.8 ⁇ 0.3 ⁇ , and 36.8 ⁇ 3 ⁇ when determined at 120K.
  • the unit cell dimensions are about 7.7 ⁇ 0.2 ⁇ , 13.8 ⁇ 0.2 ⁇ , and 36.8 ⁇ 2.7 ⁇ when determined at 120K.
  • Table 1 shows the unit cell dimensions and peak positions for a number of example compositions: TABLE 1 Guest molecule Unit cell dimensions Peak positions Toluene 7.8 13.7 34.2 4.9 6.8 11.8 m-xylene 7.8 13.8 35.9 4.8 6.8 11.5 o-xylene ND ND ND 5.0 6.9 11.7 fluorobenzene 7.7 13.9 38.7 ND ND ND ethylbenzene 7.7 13.8 37.8 ND ND ND chlorobenzene 7.8 13.7 39.3 4.3 6.6 —
  • Inhaled steroids are also absorbed through the lung and this route of absorption makes a significant contribution to systemic exposure. Reduced lung absorption could therefore provide an improved safety profile.
  • Studies with compound (I) have shown significantly lower exposure to compound (I) than with fluticasone propionate after dry powder delivery to the lungs of anaesthetised pigs.
  • Examples of disease states in which the compound of formula (I) and compositions thereof have utility include skin diseases such as eczema, psoriasis, allergic dermatitis, neurodermatitis, pruritis and hypersensitivity reactions; inflammatory conditions of the nose, throat or lungs such as asthma (including allergen-induced asthmatic reactions), rhinitis (including hayfever), nasal polyps, chronic obstructive pulmonary disease, interstitial lung disease, and fibrosis; inflammatory bowel conditions such as ulcerative colitis and Crohn's disease; and auto-immune diseases such as rheumatoid arthritis.
  • skin diseases such as eczema, psoriasis, allergic dermatitis, neurodermatitis, pruritis and hypersensitivity reactions
  • inflammatory conditions of the nose, throat or lungs such as asthma (including allergen-induced asthmatic reactions), rhinitis (including hayfever), nasal polyps, chronic obstructive pulmonary disease, intersti
  • the compound of formula (I) may also have use in the treatment of conjunctiva and conjunctivitis.
  • composition of the invention is expected to be most useful in the treatment of inflammatory disorders of the respiratory tract e.g. asthma, COPD and rhinitis particularly asthma and rhinitis.
  • composition of the invention is useful in human or veterinary medicine, in particular as an anti-inflammatory and anti-allergic agent.
  • composition of the invention for use in human or veterinary medicine, particularly in the treatment of patients with inflammatory and/or allergic conditions, especially for treatment once-per-day.
  • composition of the invention for the manufacture of a medicament for the treatment of patients with inflammatory and/or allergic conditions, especially for treatment once-per-day.
  • a method for the treatment of a human or animal subject with an inflammatory and/or allergic condition comprises administering to said human or animal subject an effective amount of the composition of the invention, especially for administration once-per-day.
  • composition of the invention may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical compositions comprising the composition of the invention together, if desirable, in admixture with one or more physiologically acceptable diluents or carriers.
  • pharmaceutical compositions suitable for once-per-day administration are of particular interest.
  • composition of the invention may, for example, be formulated for oral, buccal, sublingual, parenteral, local or rectal administration, especially local administration.
  • Local administration includes administration by insufflation and inhalation.
  • preparation for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (e.g. eye or nose drops), solutions/suspensions for nebulisation, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (e.g. for the treatment of aphthous ulcers) or liposome or microencapsulation preparations.
  • compositions for topical administration to the lung include dry powder compositions and spray compositions.
  • Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges for use in an inhaler or insufflator of, for example, gelatine.
  • Formulations generally contain a powder mix for inhalation of the compound of the invention and a suitable powder base (carrier substance) such as lactose or starch. Use of lactose is preferred.
  • a suitable powder base such as lactose or starch.
  • lactose is preferred.
  • Each capsule or cartridge may generally contain between 20 ⁇ g-10 mg of the compound of formula (I) in a composition of the invention optionally in combination with another therapeutically active ingredient.
  • the composition of the invention may be presented without excipients.
  • Packaging of the formulation may be suitable for unit dose or multi-dose delivery.
  • the formulation can be pre-metered (e.g. as in Diskus, see GB 2242134 or Diskhaler, see GB 2178965, 2129691 and 2169265) or metered in use (e.g. as in Turbuhaler, see EP 69715).
  • An example of a unit-dose device is Rotahaler (see GB 2064336).
  • the Diskus inhalation device comprises an elongate strip formed from a base sheet having a plurality of recesses spaced along its length and a lid sheet hermetically but peelably sealed thereto to define a plurality of containers, each container having therein an inhalable formulation containing a composition of the invention preferably combined with lactose.
  • the strip is sufficiently flexible to be wound into a roll.
  • the lid sheet and base sheet will preferably have leading end portions which are not sealed to one another and at least one of the said leading end portions is constructed to be attached to a winding means. Also, preferably the hermetic seal between the base and lid sheets extends over their whole width.
  • the lid sheet may preferably be peeled from the base sheet in a longitudinal direction from a first end of the said base sheet.
  • compositions which are non-pressurised and adapted to be administered as a dry powder topically to the lung via the buccal cavity (especially those which are free of excipient or are formulated with a diluent or carrier such as lactose or starch, most especially lactose) are of particular interest.
  • Spray compositions for topical delivery to the lung by inhalation may for example be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain the composition of the invention optionally in combination with another therapeutically active ingredient and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • the aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid or lecithin and cosolvents e.g. ethanol.
  • additional formulation excipients well known in the art such as surfactants e.g. oleic acid or lecithin and cosolvents e.g. ethanol.
  • One example formulation is excipient free and consists essentially of (e.g. consists of) composition of the invention (optionally together with a further active ingredient) and a propellant selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixture thereof.
  • Another example formulation comprises particulate composition of the invention, a propellant selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixture thereof and a suspending agent which is soluble in the propellant e.g. an oligolactic acid or derivative thereof as described in WO94/21229.
  • the preferred propellant is 1,1,1,2-tetrafluoroethane.
  • Pressurised formulations will generally be retained in a canister (e.g. an aluminium canister) closed with a valve (e.g. a metering valve) and fitted into an actuator provided with a mouthpiece.
  • Medicaments for administration by inhalation desirably have a controlled particle size.
  • the optimum particle size for inhalation into the bronchial system is usually 1-10 ⁇ m, preferably 2-5 ⁇ m. Particles having a size above 20 ⁇ m are generally too large when inhaled to reach the small airways.
  • the particles of the composition of the invention as produced may be size reduced by conventional means e.g. by micronisation. The desired fraction may be separated out by air classification or sieving.
  • the particles will be crystalline, prepared for example by a process which comprises mixing in a continuous flow cell in the presence of ultrasonic radiation a flowing solution of compound of formula (I) as medicament in a liquid solvent with a flowing liquid antisolvent for said medicament (e.g. as described in International Patent Application PCT/GB99104368) or else by a process which comprises admitting a stream of solution of the substance in a liquid solvent and a stream of liquid antisolvent for said substance tangentially into a cylindrical mixing chamber having an axial outlet port such that said streams are thereby intimately mixed through formation of a vortex and precipitation of crystalline particles of the substance is thereby caused (e.g. as described in International Patent Application PCT/GB00/04327).
  • the particle size of the excipient will be much greater than the inhaled medicament within the present invention.
  • the excipient is lactose it will typically be present as milled lactose, wherein not more than 85% of lactose particles will have a MMD of 60-90 ⁇ m and not less than 15% will have a MMD of less than 15 ⁇ m.
  • Formulations for administration topically to the nose include pressurised aerosol formulations and aqueous formulations administered to the nose by pressurised pump.
  • Formulations which are non-pressurised and adapted to be administered topically to the nasal cavity are of particular interest.
  • the formulation preferably contains water as the diluent or carrier for this purpose.
  • Aqueous formulations for administration to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • Aqueous formulations may also be administered to the nose by nebulisation.
  • Ointments, creams and gels may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents.
  • bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol.
  • Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
  • Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilising agents, suspending agents or preservatives.
  • formulations of the invention may be buffered by the addition of suitable buffering agents.
  • the proportion of the active compound of formula (I) in the local compositions according to the invention depends on the precise type of formulation to be prepared but will generally be within the range of from 0.001 to 10% by weight. Generally, however for most types of preparations advantageously the proportion used will be within the range of from 0.005 to 1% and preferably 0.01 to 0.5%. However, in powders for inhalation or insufflation the proportion used will usually be within the range of from 0.1 to 5%.
  • Aerosol formulations are preferably arranged so that each metered dose or “puff” of aerosol contains 1 ⁇ g-2000 ⁇ p e.g. 20 ⁇ g-2000 ⁇ p, preferably about 20 ⁇ g-500 ⁇ g of compound of formula (I) optionally in combination with another therapeutically active ingredient.
  • Administration may be once daily or several times daily, for example 2, 3, 4 or 8 times, giving for example 1, 2 or 3 doses each time.
  • the composition of the invention is delivered once or twice daily.
  • the overall daily dose with an aerosol will typically be within the range 10 ⁇ g-10 mg e.g. 100 ⁇ g-10 mg preferably, 200 ⁇ g-2000 ⁇ g.
  • Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system.
  • the compound according to the invention may, for example, be formulated in conventional manner for oral, parenteral or rectal administration.
  • Formulations for oral administration include syrups, elixirs, powders, granules, tablets and capsules which typically contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, wetting agents, suspending agents, emulsifying agents, preservatives, buffer salts, flavouring, colouring and/or sweetening agents as appropriate.
  • Dosage unit forms are, however, preferred as described below.
  • Preferred forms of preparation for internal administration are dosage unit forms i.e. tablets and capsules. Such dosage unit forms contain from 0.1 mg to 20 mg preferably from 2.5 to 10 mg of the compound of formula (I).
  • the compound according to the invention may in general may be given by internal administration in cases where systemic adreno-cortical therapy is indicated.
  • preparations for internal administration may contain from 0.05 to 10% of the active ingredient dependent upon the type of preparation involved.
  • the daily dose may vary from 0.1 mg to 60 mg, e.g. 5-30 mg, dependent on the condition being treated, and the duration of treatment desired.
  • Slow release or enteric coated formulations may be advantageous, particularly for the treatment of inflammatory bowel disorders.
  • the composition of the invention will be delivered once-per-day and the dose will be selected so that the compound has a therapeutic effect in the treatment of respiratory disorders (e.g. asthma or COPD, particularly asthma) over 24 hours or more.
  • respiratory disorders e.g. asthma or COPD, particularly asthma
  • compositions according to the invention may also be used in combination with another therapeutically active agent, for example, a ⁇ 2 adrenoreceptor agonist, an anti-histamine or an anti-allergic.
  • another therapeutically active agent for example, a ⁇ 2 adrenoreceptor agonist, an anti-histamine or an anti-allergic.
  • ⁇ 2 -adrenoreceptor agonists examples include salmeterol (e.g. as racemate or a single enantiomer such as the R-enantiomer), salbutamol, formoterol, salmefamol, fenoterol or terbutaline and salts thereof, for example the xinafoate salt of salmeterol, the sulphate salt or free base of salbutamol or the fumarate salt of formoterol.
  • Pharmaceutical compositions employing combinations with long-acting ⁇ 2 -adrenoreceptor agonists are particularly preferred, especially those which have a therapeutic effect (e.g. in the treatment of asthma or COPD, particularly asthma) over 24 hours or more.
  • the composition comprising the compound of formula (I) and the long-acting ⁇ 2 -adrenoreceptor agonists will be delivered once-per-day and the dose of each will be selected so that the composition has a therapeutic effect in the treatment of respiratory disorders effect (e.g. in the treatment of asthma or COPD, particularly asthma) over 24 hours or more.
  • respiratory disorders effect e.g. in the treatment of asthma or COPD, particularly asthma
  • anti-histamines examples include methapyrilene or loratadine.
  • NSAIDs e.g. sodium cromoglycate, nedocromil sodium, PDE4 inhibitors, leukotriene antagonists, iNOS inhibitors, tryptase and elastase inhibitors, beta-2 integrin antagonists and adenosine 2a agonists
  • antiinfective agents e.g. antibiotics, antivirals
  • composition of the invention in combination with a phosphodiesterase 4 (PDE4) inhibitor e.g. cilomilast or a salt thereof.
  • PDE4 phosphodiesterase 4
  • the compound according to the invention in combination with another therapeutically active ingredient as described above may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical formulations comprising the composition of the invention in combination with another therapeutically active ingredient together, if desirable, in admixture with one or more physiologically acceptable diluents or carriers.
  • the preferred route of administration for inflammatory disorders of the respiratory tract will generally be administration by inhalation.
  • Therapeutic agent combinations may be in any form, for example combinations may comprise a single dose containing separate particles of individual therapeutics, and optionally excipient material(s), alternatively, multiple therapeutics may be formed into individual multicomponent particles, formed for example by coprecipitation, and optionally containing excipient material(s).
  • composition of the invention may be prepared by the methodology described hereinafter, constituting a further aspect of this invention.
  • a first process for preparing a composition of the invention comprises crystallising the composition from a solution containing a compound of formula (I) and the guest molecule.
  • the solution containing the guest molecule could be the guest itself when this a liquid, or could be the guest dissolved in another liquid substance which substance does not act as a guest molecule.
  • the crystallisation process may be assisted by seeding with crystals of the composition of the invention.
  • the seed crystals of the composition of the invention need not contain the same guest molecule.
  • a second process for preparing a composition of the invention comprises contacting the compound of formula (I) or a composition according to the invention thereof in solid form with a liquid containing the guest molecule (for example by slurrying) and obtaining the composition therefrom.
  • the liquid containing the guest molecule could be the guest itself when this a liquid, or could be the guest dissolved in another liquid substance which substance does not act as a guest molecule.
  • a third process for preparing a composition of the invention comprises contacting a compound of formula (I) or a composition according to the invention thereof in solid form with a vapour containing the guest molecule. This process is suitable when the guest has acceptable volatility e.g. when the guest is a solvent.
  • the compound of formula (I) may be employed in the form of a composition with a guest molecule or in a form without a guest molecule (eg as unsolvated polymorph Form 1, 2 or 3).
  • the compound of formula (I) or a composition according to the invention may be dissolved in the solution or prepared in situ.
  • the input compound of formula (I) in the first, second and third processes is in the form of a substantially amorphous solid.
  • the compound of formula (I) in the form of a substantially amorphous solid is preferably in the form of substantially amorphous particles.
  • the compound of formula (I) in the form of substantially amorphous particles may be obtained by spray drying a solution containing the compound of formula (I). Any solvent that will dissolve the compound of formula (I) that can be evaporated safely in a spray drying process may be used.
  • Suitable solvents for forming the solution include, but are not limited to, methyl acetate, ethyl acetate, isopropyl acetate, acetone, 2-butanone, 3-pentanone, 4-methyl-2-pentanone, ethanol, methanol, 1-propanol, propan-2-ol, acetonitrile, chloroform, dichloromethane especially methylethylketone (2-butanone).
  • Solution concentration will typically be 0.5-50% specifically 10-40% eg 20-30%. Lower concentrations may be more suitable for preparing smaller particle sizes especially 24% e.g. 3.54%. The concentration that may be employed will be limited by the dissolution power of the solvent.
  • Methylethylketone is preferred since it dissolves compound of formula (I) at a relatively high concentration which results in production advantages.
  • the compound of formula (I) may be employed in non-solvated form or in the form of a composition of the invention (e.g. with acetone). Preferably it is employed as the non-solvated Form 1 polymorph.
  • Spray drying maybe performed, for example, using apparatus supplied by Buchi or Niro.
  • a pneumatic spray nozzle orifice of e.g. 0.04 inches is suitable, although alternate atomization methods such as rotary and pressure nozzles can be used.
  • Solution flow rate may typically be in the range 1-100 ml/min, especially 15-30 ml/min.
  • the inlet temperature and flow rate combination should be suitable to evaporate the solvent completely to minimize the risk of solvent trapped in the particle expediting an amorphous to crystalline transition.
  • Inlet temperatures can range from 50-250° C., typically 100-200° C.
  • Form 3 Peaks at around 18.6 and 19.2 degrees 2Theta.
  • Forms 1 appears likely to be the thermodynamically most stable form since Forms 2 and 3 are converted into Form 1 on heating.
  • a process for preparing a compound of formula (I) as crystalline unsolvated Form 1 polymorph comprises dissolving compound of formula (I) in methylisobutylketone or ethyl acetate and producing compound of formula (I) as unsolvated Form 1 by addition of an anti-solvent such as iso-octane or toluene.
  • the compound of formula (I) may be dissolved in ethyl acetate and compound of formula (I) as unsolvated Form 1 polymorph may be obtained by addition of toluene as anti-solvent.
  • the ethyl acetate solution is hot and once the toluene has been added the mixture is distilled to reduce the content of ethyl acetate.
  • the compound of formula (I) may be dissolved in methylisobutylketone and compound of formula (I) as crystalline unsolvated Form 1 polymorph may be obtained by addition of isooctane as anti-solvent.
  • a process for preparing a compound of formula (I) as unsolvated Form 2 polymorph comprises dissolving compound of formula (I) in unsolvated form in methanol or dry dichloromethane and recrystallising the compound of formula (I) as unsolvated Form 2 polymorph.
  • the compound of formula (I) will be dissolved in hot methanol or dry dichloromethane and allowed to cool.
  • a process for preparing a compound of formula (I) as unsolvated Form 3 polymorph comprises dissolving compound of formula (I) in particular as the composition with acetone in dichloromethane in the presence of water (typically 1-3% water by volume) and recrystallising the compound of formula (I) as unsolvated Form 3 polymorph.
  • compositions of the invention may also find use as manufacturing intermediates in the preparation of compound of formula (I) in unsolvated form, or in the preparation of other compositions of the invention, or in pharmaceutical compositions thereof.
  • a process for preparation of compound of formula (I) in unsolvated form comprises removing the guest molecule from a composition of the invention.
  • a process for preparing a compound of formula (I) comprises alkylation of a thioacid of formula (II)
  • the compound of formula (II) may be reacted with a compound of formula FCH 2 L wherein L represents a leaving group (e.g. a halogen atom, a mesyl or tosyl group or the like) for example, an appropriate fluoromethyl halide under standard conditions.
  • L represents a leaving group (e.g. a halogen atom, a mesyl or tosyl group or the like) for example, an appropriate fluoromethyl halide under standard conditions.
  • the fluoromethyl halide reagent is bromofluoromethane.
  • the compound of formula (II) is employed as a salt, particularly the salt with diisopropylethylamine.
  • the compound of formula (II) or a salt thereof is treated with bromofluoromethane optionally in the presence of a phase transfer catalyst.
  • a preferred solvent is methylacetate, or more preferably ethylacetate, optionally in the presence of water. The presence of water improves solubility of both starting material and product and the use of a phase transfer catalyst results in an increased rate of reaction.
  • phase transfer catalysts examples include (but are not restricted to) tetrabutylammonium bromide, tetrabutylammonium chloride, benzyltributylammonium bromide, benzyltributylammonium chloride, benzyltriethylammonium bromide, methyltributylammonium chloride and methyltrioctylammonium chloride.
  • THF has also successfully been employed as solvent for the reaction wherein the presence of a phase transfer catalyst again provides a significantly faster reaction rate.
  • the product present in an organic phase is washed firstly with aqueous acid e.g.
  • the step typically comprises the addition of a reagent suitable for performing the esterification e.g. an activated derivative of 2-furoic acid such as an activated ester or preferably a 2-furoyl halide e.g. 2-furoyl chloride (employed in at least 2 times molar quantity relative to the compound of formula (III)) in the presence of an organic base e.g. triethylamine.
  • a reagent suitable for performing the esterification e.g. an activated derivative of 2-furoic acid such as an activated ester or preferably a 2-furoyl halide e.g. 2-furoyl chloride (employed in at least 2 times molar quantity relative to the compound of formula (III)) in the presence of an organic base e.g. triethylamine.
  • the second mole of 2-furoyl chloride reacts with the thioacid moiety in the compound of formula (III) and needs to be removed e.g. by reaction with an
  • step (b) removal of the sulphur-linked 2-furoyl moiety from compound of formula (IIA) by reaction of the product of step (a) with an organic primary or secondary amine base capable of forming a water soluble 2-furoyl amide.
  • step (c1) when the product of step (b) is dissolved in a substantially water immiscible organic solvent, purifying the compound of formula (II) by washing out the amide by-product from step (b) with an aqueous wash, or
  • step (c2) when the product of step (b) is dissolved in a water miscible solvent, purifying the compound of formula (II) by treating the product of step (b) with an aqueous medium so as to precipitate out pure compound of formula (II) or a salt thereof.
  • the activated derivative of 2-furoic acid may be an activated ester of 2-furoic acid, but is more preferably a 2-furoyl halide, especially 2-furoyl chloride.
  • a suitable solvent for this reaction is ethylacetate or methylacetate (preferably methylacetate) (when step (c1) may be followed) or acetone (when step (c2) may be followed).
  • an organic base e.g. triethylamine will be present.
  • the organic base is diethanolamine.
  • the base may suitably be dissolved in a solvent e.g. methanol.
  • steps (a) and (b) will be performed at reduced temperature e.g. between 0 and 5° C.
  • the aqueous wash may be water, however the use of brine results in higher yields and is therefore preferred.
  • the aqueous medium is for example a dilute aqueous acid such as dilute HCl.
  • the activated derivative of 2-furoic acid may be an activated ester of 2-furoic acid, but is more preferably a 2-furoyl halide, especially 2-furoyl chloride.
  • a suitable solvent for his step is acetone. Normally an organic base e.g. triethylamine will be present.
  • a suitable solvent is DMF or dimethylacetamide. Normally an organic base e.g. triethylamine will be present.
  • steps (a) and (b) will be performed at reduced temperature e.g. between 0 and 5° C.
  • the product may be isolated by treatment with acid and washing with water.
  • the compound of formula (II) may advantageously be isolated in the form of a solid crystalline salt.
  • the preferred salt is a salt formed with a base such as triethylamine, 2,4,6-trimethylpyridine, diisopropylethylamine or N-ethylpiperidine.
  • Such salt forms of compound of formula (II) are more stable, more readily filtered and dried and can be isolated in higher purity than the free thioacid.
  • the most preferred salt is the salt formed with diisopropylethylamine.
  • the triethylamine salt is also of interest.
  • Step (a) comprises oxidation of a solution containing the compound of formula (V).
  • step (a) will be performed in the presence of a solvent comprising methanol, water, tetrahydrofuran, dioxan or diethylene glygol dimethylether.
  • a solvent comprising methanol, water, tetrahydrofuran, dioxan or diethylene glygol dimethylether.
  • preferred solvents are methanol, water or tetrahydrofuran, and more preferably are water or tetrahydrofuran, especially water and tetrahydrofuran as solvent.
  • Dioxan and diethylene glygol dimethylether are also preferred solvents which may optionally (and preferably) be employed together with water.
  • the solvent will be present in an amount of between 3 and 10 vol relative to the amount of the starting material (1 wt.), more preferably between 4 and 6 vol., especially 5 vol.
  • the oxidising agent is present in an amount of 1-9 molar equivalents relative to the amount of the starting material.
  • the oxidising agent may be present in an amount of between 1.1 and 10 wt. relative to the amount of the starting material (1 wt.), more preferably between 1.1 and 3 wt., especially 1.3 wt.
  • the oxidation step will comprise the use of a chemical oxidising agent. More preferably, the oxidising agent will be periodic acid or iodic acid or a salt thereof.
  • the oxidising agent will be periodic acid or sodium periodate, especially periodic acid.
  • the oxidation step may comprise any suitable oxidation reaction, e.g. one which utilises air and/or oxygen.
  • the solvent used in said reaction will preferably be methanol.
  • step (a) will involve incubating the reagents at room temperature or a little warmer, say around 25° C. e.g. for 2 hours.
  • the compound of formula (IV) may be isolated by recrystallisation from the reaction mixture by addition of an anti-solvent.
  • a suitable anti-solvent for compound of formula (IV) is water.
  • Step (b) will typically comprise the addition of a reagent suitable for converting a carboxylic acid to a carbothioic acid e.g. using hydrogen sulphide gas together with a suitable coupling agent e.g. carbonyldiimidazole (CDI) in the presence of a suitable solvent e.g. dimethylformamide.
  • a suitable coupling agent e.g. carbonyldiimidazole (CDI)
  • CDI carbonyldiimidazole
  • compositions comprising a compound of formula (I) together with a guest compound according to the invention may include the fact that the substance appears to demonstrate excellent anti-inflammatory properties, with predictable pharmacokinetic and pharmacodynamic behaviour, with an attractive side-effect profile, long duration of action, and is compatible with a convenient regime of treatment in human patients, in particular being amenable to once-per day dosing. Further advantages may include the fact that the substance has desirable physical and chemical properties which allow for ready manufacture and storage. Alternatively it may serve as a useful intermediate in the preparation of other forms of the compound of formula (I) or compositions thereof.
  • FIG. 1 Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a composition of the invention with toluene (guest molecule darkened).
  • FIG. 2 Figure showing hydrogen bond interactions between steroid and guest for the composition of the invention with toluene
  • FIG. 3 Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a composition of the invention with toluene, and hydrogen bond interactions between two steroid molecules
  • FIG. 4 Enlarged XRPD profile of composition of the invention with toluene
  • FIG. 5 Enlarged XRPD profile of composition of the invention with m-xylene
  • FIG. 6 Enlarged XRPD profile of composition of the invention with o-xylene
  • FIG. 7 Enlarged XRPD profile of composition of the invention with chlorobenzene
  • FIG. 8 XRPD profiles of unsolvated polymorphs 1, 2 and 3
  • LCMS was conducted on a Supelcosil LCABZ+PLUS column (3.3 cm ⁇ 4.6 mm ID) eluting with 0.1% HCO 2 H and 0.01 M ammonium acetate in water (solvent A), and 0.05% HCO 2 H 5% water in acetonitrile (solvent B), using the following elution gradient 0-0.7 min 0% B, 0.7-4.2 min 100% B, 4.2-5.3 min 0% B, 5.3-5.5 min 0% B at a flow rate of 3 ml/min.
  • the mass spectra were recorded on a Fisons VG Platform spectrometer using electrospray positive and negative mode (ES+ve and ES-ve).
  • the diffractometer used in each case can be determined by the end angle in the figure.
  • Dilute hydrochloric acid (approx 1M, 550 ml) is added maintaining a reaction temperature below 15° C. and the mixture stirred at 15° C.
  • the organic phase is separated and the aqueous phase is back extracted with methyl acetate (2 ⁇ 250 ml). All of the organic phases are combined, washed sequentially with brine (5 ⁇ 250 ml) and treated with di-isopropylethylamine (30 ml).
  • the reaction mixture is concentrated by distillation at atmospheric pressure to an approximate volume of 250 ml and cooled to 25-30° C. (crystallisation of the desired product normally occurs during distillation/subsequent cooling).
  • TBME Tertiary butyl methyl ether
  • a mobile suspension of Intermediate 1 (12.61 g, 19.8 mmol) in ethyl acetate (230 ml) and water (50 ml) is treated with a phase transfer catalyst (benzyltributylammonium chloride, 10 mol %), cooled to 3° C. and treated with bromofluoromethane (1.10 ml, 19.5 mmol, 0.98 equivalents), washing in with prechilled (0° C.) ethyl acetate (EtOAc) (20 ml). The suspension is stirred overnight, allowing to warm to 17° C.
  • a phase transfer catalyst benzyltributylammonium chloride, 10 mol %
  • the aqueous layer is separated and the organic phase is sequentially washed with 1M HCl (50 ml), 1% w/v NaHCO 3 solution (3 ⁇ 50 ml) and water (2 ⁇ 50 ml).
  • the ethylacetate solution is distilled at atmospheric pressure until the distillate reaches a temperature of approximately 73° C. at which point toluene (150 ml) is added. Distillation is continued at atmospheric pressure until all remaining EtOAc has been removed (approximate distillate temperature 103° C.).
  • the resultant suspension is cooled and aged at ⁇ 10° C. and filtered off. The bed is washed with toluene (2 ⁇ 30 ml) and the product oven dried under vacuum at 60° C.
  • the functional assay was based on that described by K. P. Ray et al., Biochem J. (1997), 328, 707-715.
  • A549 cells stably transfected with a reporter gene containing the NF- ⁇ B responsive elements from the ELAM gene promoter coupled to sPAP (secreted alkaline phosphatase) were treated with test compounds at appropriate doses for 1 hour at 37° C.
  • the cells were then stimulated with tumour necrosis factor (TNF, 10 ng/ml) for 16 hours, at which time the amount of alkaline phosphatase produced is measured by a standard colourimetric assay.
  • TNF tumour necrosis factor
  • Dose response curves were constructed from which EC 50 values were estimated.
  • the glucocorticoid receptor can function in at least two distinct mechanisms, by upregulating gene expression through the direct binding of GR to specific sequences in gene promotors, and by downregulating gene expression that is being driven by other transcription factors (such as NF ⁇ B or AP-1) through their direct interaction with GR.
  • the first cell line contains the firefly luciferase reporter gene under the control of a synthetic promoter that specifically responds to activation of the transcription factor NF ⁇ B when stimulated with TNF ⁇ .
  • the second cell line contains the renilla luciferase reporter gene under the control of a synthetic promotor that comprises 3 copies of the consensus glucocorticoid response element, and which responds to direct stimulation by glucocorticoids.
  • Anaesthetised pigs (2) were dosed intra-tracheally with a homogenous mixture of compound (I) (1 mg) and fluticasone propionate (1 mg) as a dry powder blend in lactose (10% w/w). Serial blood samples were taken for up to 8 h following dosing.
  • Plasma levels of compound (I) and fluticasone propionate were determined following extraction and analysis using LC-MS/MS methodology, the lower limits of quantitation of the methods were 10 and 20 ⁇ g/mL for compound (I) and fluticasone propionate respectively. Using these methods compound (I) was quantifiable up to 2 hours after dosing and fluticasone propionate was quantifiable up to 8 hours after dosing. Maximum plasma concentrations were observed for both compounds within 15 min after dosing. Plasma half-life data obtained from IV dosing (0.1 mg/kg) was used to calculate AUC (0-inf) values for compound (I). This compensates for the plasma profile of Compound (I) only being defined up to 2 hours after an IT dose and removes any bias due to limited data between compound (I) and fluticasone propionate.
  • C max and AUC (0-inf) values show markedly reduced systemic exposure to compound (I) compared to fluticasone propionate:— Cmax AUC (0-inf) (pg/mL) (hr ⁇ pg/mL) Pig 1 Pig 2 Pig 1 Pig 2 Compound of Formula (I) 117 81 254 221 Fluticasone propionate 277 218 455 495
  • Solution flow rate 30 ml/min using Isco 260D syringe pump (Isco Inc, Lincoln, Nebr., USA)
  • Particle collection was achieved in the conventional manner using a Fisher Kleinman XQ120-1.375 high efficiency cyclone (Fisher-Klosterman Inc, Louisville, Ky., USA). A white powder was recovered. The spray drying process was successful at producing smooth, spherical particles of amorphous 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester. System yield was 61%
  • compositions of the invention are provided in FIGS. 4, 5, 6 and 7 .
  • compositions of the invention substantially by reference to their XRPD profiles as shown in the Figures and Tables.
  • Dry powder composition containing 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester composition with toluene
  • a dry powder formulation may be prepared as follows:
  • a peelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared.
  • Dry powder composition containing 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester composition with toluene and a long acting ⁇ 2 -adrenoreceptor agonist
  • a dry powder formulation may be prepared as follows:
  • a peelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared.
  • Aerosol formulation containing 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester composition with toluene
  • An aluminium canister may be filled with a formulation as follows:
  • the canister may be fitted with a metering valve adapted to dispense 50 ⁇ l per actuation.
  • Aerosol formulation containing 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17′-carbothioic acid S-fluoromethyl ester composition with toluene and a long acting ⁇ 2 -adrenoreceptor agonist
  • An aluminium canister may be filled with a formulation as follows: 6 ⁇ , 9 ⁇ -Difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3 ⁇ m: 250 ⁇ g Long-acting ⁇ 2 -adrenoreceptor agonist 25 ⁇ g (micronised to a MMD of 3 ⁇ m): 1,1,1,2-tetrafluoroethane: to 50 ⁇ l
  • the canister may be fitted with a metering valve adapted to dispense 50 ⁇ l per actuation.
  • a formulation for intranasal delivery may be prepared as follows:
  • the formulation may be fitted into a spraypump capable of delivering a plurality of metered doses (Valois).

Abstract

There is provided a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00001
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P2 12121 having unit cell dimensions of about 7.6±0.6 Å, 12.7±0.7 Å, and 33±3 Å when determined at 120K, wherein at about 295K said composition exhibits one or more of the following XRPD profiles: (i) a peak in the range of around 4.3-5.0; (ii) a peak in the range of around 6.6-6.9; or (iii) a peak in the range of around 11.5-11.8.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation of U.S. patent Ser. No. 10/241,658, filed Sep. 11, 2002, which is a Continuation-in-part of U.S. patent application Ser. No. 10/200,364 filed on 22 Jul. 2002 which is a Continuation-in-part of U.S. patent application Ser. No. 10/067,010 filed on 4 Feb. 2002 which is a Continuation-in-part of U.S. patent application Ser. No. 09/958,050 filed on 2 Oct. 2001, which is based upon International Patent Application No. PCT.GB01.03495 filed 3 Aug. 2001, which claims priority to United Kingdom Patent Application No. GB 0019172.6 filed 5 Aug. 2000.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to a novel composition containing an anti-inflammatory and anti-allergic compound of the androstane series and to processes for its preparation. The present invention also relates to pharmaceutical formulations containing the composition and to therapeutic uses thereof, particularly for the treatment of inflammatory and allergic conditions. [0002]
  • BACKGROUND OF THE INVENTION
  • Glucocorticoids which have anti-inflammatory properties are known and are widely used for the treatment of inflammatory disorders or diseases such as asthma and rhinitis. For example, U.S. Pat. No. 4,335,121 discloses 6α, 9α-Difluoro-17α-(1-oxopropoxy)-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (known by the generic name of fluticasone propionate) and derivatives thereof. The use of glucocorticoids generally, and especially in children, has been limited in some quarters by concerns over potential side effects. The side effects that are feared with glucocorticoids include suppression of the Hypothalamic-Pituitary-Adrenal (HPA) axis, effects on bone growth in children and on bone density in the elderly, ocular complications (cataract formation and glaucoma) and skin atrophy. Certain glucocorticoid compounds also have complex paths of metabolism wherein the production of active metabolites may make the pharmacodynamics and pharmacokinetics of such compounds difficult to understand. Whilst the modern steroids are very much safer than those originally introduced, it remains an object of research to produce new molecules which have excellent anti-inflammatory properties, with predictable pharmacokinetic and pharmacodynamic properties, with an attractive side effect profile, and with a convenient treatment regime. [0003]
  • SUMMARY OF THE INVENTION
  • We have now identified a novel glucocorticoid compound and a crystalline composition thereof which substantially meets these objectives. [0004]
  • Thus, according to one aspect of the invention, there is provided a crystalline chemical composition comprising a compound of formula (I) [0005]
    Figure US20040220156A1-20041104-C00002
  • in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of [0006] space group P2 12121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K (hereinafter “a composition of the invention”).
  • In the composition of the invention the crystalline lattice is stabilised by a hydrogen bonding interaction between the hydrogen atom of the C11 hydroxy on the compound of formula (I) with the oxygen of the C3 carbonyl on a second molecule of the compound of formula (I). [0007]
  • The nature of the crystal lattice can be seen by reference to FIG. 1 which shows the spacial arrangement of 4 molecules of steroid and 8 guests within a single unit cell for an example composition. Detail of the hydrogen bond interaction between the steroid molecules is shown in FIG. 3. FIG. 2 shows the conformation of the steroid molecule with 2 guest molecules present. [0008]
  • We have determined the XRPD profiles for a number of compositions according to the invention. These XRPD profiles are also apparently characteristic of the crystalline composition according to the invention. In particular they exhibit one or more of the following 3 features when determined at ambient temperature (eg around 295K): [0009]
  • (a) A peak in the range of around 4.3-5.0. [0010]
  • (b) A peak in the range of around 6.6-6.9. [0011]
  • (c) A peak in the range of around 11.5-11.8. [0012]
  • Typically they exhibit 2 or more of the above 3 features, especially 3 of the above features. [0013]
  • The XRPD profiles of compositions of the invention when crystallographically pure also preferably exhibit one or more of the following 6 features when determined at ambient temperature (eg around 295K): [0014]
  • (a) Absence of a peak at around 7.3 (eg around 7.1-7.5) which is associated with the profile of [0015] unsolvated Form 1, 2 and 3 polymorphs;
  • (b) Absence of a peak at around 7.5 (eg around 7.2-7.7) which is associated with the profile of another class of compositions of compound of formula (I); [0016]
  • (c) Absence of a peak at around 12.5 (eg around 12.3-12.7) which is associated with the profile of [0017] unsolvated Form 1, 2 and 3 polymorphs;
  • (d) Absence of a peak at around 8.8-9.6 which is associated with the profile of another class of compositions of compound of formula (I); [0018]
  • (e) Absence of a peak at around 10.5-11.1 which is associated with the profile of another class of compositions of compound of formula (I); [0019]
  • (f) Absence of a peak at around 12.2-12.6 which is associated with the profile of another class of compositions of compound of formula (I). [0020]
  • Preferably one or more preferably both of features (a) and (b) at least are exhibited. Preferably 3 or more preferably 4, especially 5, most especially all 6 of the above 6 features are exhibited. [0021]
  • The chemical name of the compound of formula (I) is 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester. [0022]
  • The compound of formula (I) and compositions thereof have potentially beneficial anti-inflammatory or anti-allergic effects, particularly upon topical administration, demonstrated by, for example, its ability to bind to the glucocorticoid receptor and to illicit a response via that receptor, with long acting effect. Hence, the compound of formula (I) and compositions thereof is useful in the treatment of inflammatory and/or allergic disorders, especially in once-per-day therapy. [0023]
  • [0024] Space group P2 12121 is characterised by angles of 90° being present in each of the 3 axes.
  • We have discovered that the compound of formula (I) can form a crystalline composition of characteristic space group, unit cell dimensions and crystalline structure as evidenced by X-ray diffraction with a number of guest molecules. [0025]
  • The guest molecule preferably has a relative molecular weight in the range 16 to 150, more preferably 60 to 130, especially 70 to 120. Preferably the guest molecule is a liquid at ambient temperature and pressure (eg 295K, 1.013×10[0026] 5 Pa). However guest molecules which are a liquid under pressure may also be capable of acting as a guest molecule (especially under pressurised conditions). Substances which are solids at ambient temperature and pressure are also included.
  • Examples of suitable guest molecules include solvents e.g.: [0027]
  • Aromatic compounds eg benzene and substituted derivatives, especially benzene substituted with one or more ([0028] eg 1 or 2) halogen or C1-4alkyl groups, for example toluene, o-xylene, m-xylene, fluorobenzene, chlorobenzene, ethylbenzene.
  • Preferred guest molecules are pharmaceutically acceptable substances and, as described below, compositions of the invention containing them may be used in therapy. However even if the guest molecule is not pharmaceutically acceptable then such compositions may be useful in the preparation of other compositions containing compound of formula (I), for example, other compositions of the invention containing guest molecules that are pharmaceutically acceptable or compound of formula (I) in unsolvated form. [0029]
  • The stoichiometry of the composition will usually be such that the ratio of compound to formula (I) to guest molecule, in molar terms, is 1:5-0.25, preferably 1:3.0-0.3, more preferably 1:2.2-0.6, especially 1:2.2-0.8. [0030]
  • Unusually a composition of the invention has a crystal structure which is quite distinct from that of compound of formula (I) in the absence of a guest molecule, eg. the compound of formula (I) as [0031] unsolvated polymorph Form 1 which has a space group of P21 (i.e. two of the axis angles are 90°) and cell dimensions of 7.6, 14.1, 11.8 Å when determined at 150K. Thus if the guest molecule is removed below a threshold level (which will differ from guest to guest) for example by heating (optionally at reduced pressure eg under vacuum) then the crystal structure of the composition starts to break down and converts to that of the structure of an unsolvated compound of formula (I), typically unsolvated polymorph Form 2 or 3.
  • Preferably the unit cell dimentions are about 7.7±0.4 Å, 13.8±0.3 Å, and 36.8±3 Å when determined at 120K. Usually the unit cell dimensions are about 7.7±0.2 Å, 13.8±0.2 Å, and 36.8±2.7 Å when determined at 120K. [0032]
  • Table 1 shows the unit cell dimensions and peak positions for a number of example compositions: [0033]
    TABLE 1
    Guest molecule Unit cell dimensions Peak positions
    Toluene 7.8 13.7 34.2 4.9 6.8 11.8
    m-xylene 7.8 13.8 35.9 4.8 6.8 11.5
    o-xylene ND ND ND 5.0 6.9 11.7
    fluorobenzene 7.7 13.9 38.7 ND ND ND
    ethylbenzene 7.7 13.8 37.8 ND ND ND
    chlorobenzene 7.8 13.7 39.3 4.3 6.6
  • Compound (I) undergoes highly efficient hepatic metabolism to yield the 17-β carboxylic acid (X) as the sole major metabolite in rat and human in vitro systems. [0034]
  • This metabolite has been synthesised and demonstrated to be >1000 fold less active than the parent compound in in vitro functional glucocorticoid assays. [0035]
    Figure US20040220156A1-20041104-C00003
  • This efficient hepatic metabolism is reflected by in vivo data in the rat, which have demonstrated plasma clearance at a rate approaching hepatic blood flow and an oral bioavailability of <1%, consistent with extensive first-pass metabolism. [0036]
  • In vitro metabolism studies in human hepatocytes have demonstrated that compound (I) is metabolised in an identical manner to fluticasone propionate but that conversion of (I) to the inactive acid metabolite occurs approximately 5-fold more rapidly than with fluticasone propionate. This very efficient hepatic inactivation would be expected to minimise systemic exposure in man leading to an improved safety profile. [0037]
  • Inhaled steroids are also absorbed through the lung and this route of absorption makes a significant contribution to systemic exposure. Reduced lung absorption could therefore provide an improved safety profile. Studies with compound (I) have shown significantly lower exposure to compound (I) than with fluticasone propionate after dry powder delivery to the lungs of anaesthetised pigs. [0038]
  • An improved safety profile is believed to allow the compound of formula (I) to demonstrate the desired anti-inflammatory effects when administered once-per day. Once-per-day dosing is considered to be significantly more convenient to patients than the twice-per day dosing regime that is normally employed for fluticasone propionate. [0039]
  • Examples of disease states in which the compound of formula (I) and compositions thereof have utility include skin diseases such as eczema, psoriasis, allergic dermatitis, neurodermatitis, pruritis and hypersensitivity reactions; inflammatory conditions of the nose, throat or lungs such as asthma (including allergen-induced asthmatic reactions), rhinitis (including hayfever), nasal polyps, chronic obstructive pulmonary disease, interstitial lung disease, and fibrosis; inflammatory bowel conditions such as ulcerative colitis and Crohn's disease; and auto-immune diseases such as rheumatoid arthritis. [0040]
  • The compound of formula (I) may also have use in the treatment of conjunctiva and conjunctivitis. [0041]
  • The composition of the invention is expected to be most useful in the treatment of inflammatory disorders of the respiratory tract e.g. asthma, COPD and rhinitis particularly asthma and rhinitis. [0042]
  • It will be appreciated by those skilled in the art that reference herein to treatment extends to prophylaxis as well as the treatment of established conditions. [0043]
  • As mentioned above, the composition of the invention is useful in human or veterinary medicine, in particular as an anti-inflammatory and anti-allergic agent. [0044]
  • There is thus provided as a further aspect of the invention the composition of the invention for use in human or veterinary medicine, particularly in the treatment of patients with inflammatory and/or allergic conditions, especially for treatment once-per-day. [0045]
  • According to another aspect of the invention, there is provided the use of the composition of the invention for the manufacture of a medicament for the treatment of patients with inflammatory and/or allergic conditions, especially for treatment once-per-day. [0046]
  • In a further or alternative aspect, there is provided a method for the treatment of a human or animal subject with an inflammatory and/or allergic condition, which method comprises administering to said human or animal subject an effective amount of the composition of the invention, especially for administration once-per-day. [0047]
  • The composition of the invention may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical compositions comprising the composition of the invention together, if desirable, in admixture with one or more physiologically acceptable diluents or carriers. Pharmaceutical compositions suitable for once-per-day administration are of particular interest. [0048]
  • Further, there is provided a process for the preparation of such pharmaceutical compositions which comprises mixing the ingredients. [0049]
  • The composition of the invention may, for example, be formulated for oral, buccal, sublingual, parenteral, local or rectal administration, especially local administration. [0050]
  • Local administration as used herein, includes administration by insufflation and inhalation. Examples of various types of preparation for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (e.g. eye or nose drops), solutions/suspensions for nebulisation, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (e.g. for the treatment of aphthous ulcers) or liposome or microencapsulation preparations. [0051]
  • Advantageously compositions for topical administration to the lung include dry powder compositions and spray compositions. [0052]
  • Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges for use in an inhaler or insufflator of, for example, gelatine. Formulations generally contain a powder mix for inhalation of the compound of the invention and a suitable powder base (carrier substance) such as lactose or starch. Use of lactose is preferred. Each capsule or cartridge may generally contain between 20 μg-10 mg of the compound of formula (I) in a composition of the invention optionally in combination with another therapeutically active ingredient. Alternatively, the composition of the invention may be presented without excipients. Packaging of the formulation may be suitable for unit dose or multi-dose delivery. In the case of multi-dose delivery, the formulation can be pre-metered (e.g. as in Diskus, see GB 2242134 or Diskhaler, see GB 2178965, 2129691 and 2169265) or metered in use (e.g. as in Turbuhaler, see EP 69715). An example of a unit-dose device is Rotahaler (see GB 2064336). The Diskus inhalation device comprises an elongate strip formed from a base sheet having a plurality of recesses spaced along its length and a lid sheet hermetically but peelably sealed thereto to define a plurality of containers, each container having therein an inhalable formulation containing a composition of the invention preferably combined with lactose. Preferably, the strip is sufficiently flexible to be wound into a roll. The lid sheet and base sheet will preferably have leading end portions which are not sealed to one another and at least one of the said leading end portions is constructed to be attached to a winding means. Also, preferably the hermetic seal between the base and lid sheets extends over their whole width. The lid sheet may preferably be peeled from the base sheet in a longitudinal direction from a first end of the said base sheet. [0053]
  • Pharmaceutical formulations which are non-pressurised and adapted to be administered as a dry powder topically to the lung via the buccal cavity (especially those which are free of excipient or are formulated with a diluent or carrier such as lactose or starch, most especially lactose) are of particular interest. [0054]
  • Spray compositions for topical delivery to the lung by inhalation may for example be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant. Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain the composition of the invention optionally in combination with another therapeutically active ingredient and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof. The aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid or lecithin and cosolvents e.g. ethanol. One example formulation is excipient free and consists essentially of (e.g. consists of) composition of the invention (optionally together with a further active ingredient) and a propellant selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixture thereof. Another example formulation comprises particulate composition of the invention, a propellant selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixture thereof and a suspending agent which is soluble in the propellant e.g. an oligolactic acid or derivative thereof as described in WO94/21229. The preferred propellant is 1,1,1,2-tetrafluoroethane. Pressurised formulations will generally be retained in a canister (e.g. an aluminium canister) closed with a valve (e.g. a metering valve) and fitted into an actuator provided with a mouthpiece. [0055]
  • Medicaments for administration by inhalation desirably have a controlled particle size. The optimum particle size for inhalation into the bronchial system is usually 1-10 μm, preferably 2-5 μm. Particles having a size above 20 μm are generally too large when inhaled to reach the small airways. To achieve these particle sizes the particles of the composition of the invention as produced may be size reduced by conventional means e.g. by micronisation. The desired fraction may be separated out by air classification or sieving. Preferably, the particles will be crystalline, prepared for example by a process which comprises mixing in a continuous flow cell in the presence of ultrasonic radiation a flowing solution of compound of formula (I) as medicament in a liquid solvent with a flowing liquid antisolvent for said medicament (e.g. as described in International Patent Application PCT/GB99104368) or else by a process which comprises admitting a stream of solution of the substance in a liquid solvent and a stream of liquid antisolvent for said substance tangentially into a cylindrical mixing chamber having an axial outlet port such that said streams are thereby intimately mixed through formation of a vortex and precipitation of crystalline particles of the substance is thereby caused (e.g. as described in International Patent Application PCT/GB00/04327). [0056]
  • When an excipient such as lactose is employed, generally, the particle size of the excipient will be much greater than the inhaled medicament within the present invention. When the excipient is lactose it will typically be present as milled lactose, wherein not more than 85% of lactose particles will have a MMD of 60-90 μm and not less than 15% will have a MMD of less than 15 μm. [0057]
  • Formulations for administration topically to the nose (e.g. for the treatment of rhinitis) include pressurised aerosol formulations and aqueous formulations administered to the nose by pressurised pump. Formulations which are non-pressurised and adapted to be administered topically to the nasal cavity are of particular interest. The formulation preferably contains water as the diluent or carrier for this purpose. Aqueous formulations for administration to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like. Aqueous formulations may also be administered to the nose by nebulisation. [0058]
  • Other possible presentations include the following: [0059]
  • Ointments, creams and gels, may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents. Such bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol. Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents. [0060]
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents. [0061]
  • Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilising agents, suspending agents or preservatives. [0062]
  • If appropriate, the formulations of the invention may be buffered by the addition of suitable buffering agents. [0063]
  • The proportion of the active compound of formula (I) in the local compositions according to the invention depends on the precise type of formulation to be prepared but will generally be within the range of from 0.001 to 10% by weight. Generally, however for most types of preparations advantageously the proportion used will be within the range of from 0.005 to 1% and preferably 0.01 to 0.5%. However, in powders for inhalation or insufflation the proportion used will usually be within the range of from 0.1 to 5%. [0064]
  • Aerosol formulations are preferably arranged so that each metered dose or “puff” of aerosol contains 1 μg-2000 μp e.g. 20 μg-2000 μp, preferably about 20 μg-500 μg of compound of formula (I) optionally in combination with another therapeutically active ingredient. Administration may be once daily or several times daily, for example 2, 3, 4 or 8 times, giving for example 1, 2 or 3 doses each time. Preferably the composition of the invention is delivered once or twice daily. The overall daily dose with an aerosol will typically be within the [0065] range 10 μg-10 mg e.g. 100 μg-10 mg preferably, 200 μg-2000 μg.
  • Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system. [0066]
  • For internal administration the compound according to the invention may, for example, be formulated in conventional manner for oral, parenteral or rectal administration. Formulations for oral administration include syrups, elixirs, powders, granules, tablets and capsules which typically contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, wetting agents, suspending agents, emulsifying agents, preservatives, buffer salts, flavouring, colouring and/or sweetening agents as appropriate. Dosage unit forms are, however, preferred as described below. [0067]
  • Preferred forms of preparation for internal administration are dosage unit forms i.e. tablets and capsules. Such dosage unit forms contain from 0.1 mg to 20 mg preferably from 2.5 to 10 mg of the compound of formula (I). [0068]
  • The compound according to the invention may in general may be given by internal administration in cases where systemic adreno-cortical therapy is indicated. [0069]
  • In general terms preparations, for internal administration may contain from 0.05 to 10% of the active ingredient dependent upon the type of preparation involved. The daily dose may vary from 0.1 mg to 60 mg, e.g. 5-30 mg, dependent on the condition being treated, and the duration of treatment desired. [0070]
  • Slow release or enteric coated formulations may be advantageous, particularly for the treatment of inflammatory bowel disorders. [0071]
  • Since the compound of formula (I) is long-acting, preferably the composition of the invention will be delivered once-per-day and the dose will be selected so that the compound has a therapeutic effect in the treatment of respiratory disorders (e.g. asthma or COPD, particularly asthma) over 24 hours or more. [0072]
  • The pharmaceutical compositions according to the invention may also be used in combination with another therapeutically active agent, for example, a β[0073] 2 adrenoreceptor agonist, an anti-histamine or an anti-allergic. The invention thus provides, in a further aspect, a combination comprising the composition of the invention together with another therapeutically active agent, for example, a β2-adrenoreceptor agonist, an anti-histamine or an anti-allergic.
  • Examples of β[0074] 2-adrenoreceptor agonists include salmeterol (e.g. as racemate or a single enantiomer such as the R-enantiomer), salbutamol, formoterol, salmefamol, fenoterol or terbutaline and salts thereof, for example the xinafoate salt of salmeterol, the sulphate salt or free base of salbutamol or the fumarate salt of formoterol. Pharmaceutical compositions employing combinations with long-acting β2-adrenoreceptor agonists (e.g. salmeterol and salts thereof) are particularly preferred, especially those which have a therapeutic effect (e.g. in the treatment of asthma or COPD, particularly asthma) over 24 hours or more.
  • Since the compound of formula (I) is long-acting, preferably the composition comprising the compound of formula (I) and the long-acting β[0075] 2-adrenoreceptor agonists will be delivered once-per-day and the dose of each will be selected so that the composition has a therapeutic effect in the treatment of respiratory disorders effect (e.g. in the treatment of asthma or COPD, particularly asthma) over 24 hours or more.
  • Examples of anti-histamines include methapyrilene or loratadine. [0076]
  • Other suitable combinations include, for example, other anti-inflammatory agents e.g. NSAIDs (e.g. sodium cromoglycate, nedocromil sodium, PDE4 inhibitors, leukotriene antagonists, iNOS inhibitors, tryptase and elastase inhibitors, beta-2 integrin antagonists and adenosine 2a agonists)) or antiinfective agents (e.g. antibiotics, antivirals). [0077]
  • Also of particular interest is use of the composition of the invention in combination with a phosphodiesterase 4 (PDE4) inhibitor e.g. cilomilast or a salt thereof. [0078]
  • The combination referred to above may conveniently be presented for use in the form of a pharmaceutical formulation and thus pharmaceutical formulations comprising a combination as defined above together with a physiologically acceptable diluent or carrier represent a further aspect of the invention. [0079]
  • The compound according to the invention in combination with another therapeutically active ingredient as described above may be formulated for administration in any convenient way, and the invention therefore also includes within its scope pharmaceutical formulations comprising the composition of the invention in combination with another therapeutically active ingredient together, if desirable, in admixture with one or more physiologically acceptable diluents or carriers. The preferred route of administration for inflammatory disorders of the respiratory tract will generally be administration by inhalation. [0080]
  • Further, there is provided a process for the preparation of such pharmaceutical compositions which comprises mixing the ingredients. [0081]
  • Therapeutic agent combinations may be in any form, for example combinations may comprise a single dose containing separate particles of individual therapeutics, and optionally excipient material(s), alternatively, multiple therapeutics may be formed into individual multicomponent particles, formed for example by coprecipitation, and optionally containing excipient material(s). [0082]
  • The individual compounds of such combinations may be administered either sequentially in separate pharmaceutical compositions as well as simultaneously in combined pharmaceutical formulations. Appropriate doses of known therapeutic agents will be readily appreciated by those skilled in the art. [0083]
  • The composition of the invention may be prepared by the methodology described hereinafter, constituting a further aspect of this invention. [0084]
  • A first process for preparing a composition of the invention comprises crystallising the composition from a solution containing a compound of formula (I) and the guest molecule. The solution containing the guest molecule could be the guest itself when this a liquid, or could be the guest dissolved in another liquid substance which substance does not act as a guest molecule. [0085]
  • Optionally, for better control and reproduceability, the crystallisation process may be assisted by seeding with crystals of the composition of the invention. The seed crystals of the composition of the invention need not contain the same guest molecule. [0086]
  • A second process for preparing a composition of the invention comprises contacting the compound of formula (I) or a composition according to the invention thereof in solid form with a liquid containing the guest molecule (for example by slurrying) and obtaining the composition therefrom. The liquid containing the guest molecule could be the guest itself when this a liquid, or could be the guest dissolved in another liquid substance which substance does not act as a guest molecule. [0087]
  • A third process for preparing a composition of the invention comprises contacting a compound of formula (I) or a composition according to the invention thereof in solid form with a vapour containing the guest molecule. This process is suitable when the guest has acceptable volatility e.g. when the guest is a solvent. [0088]
  • In the second and third processes, the compound of formula (I) may be employed in the form of a composition with a guest molecule or in a form without a guest molecule (eg as [0089] unsolvated polymorph Form 1, 2 or 3). In the first process the compound of formula (I) or a composition according to the invention may be dissolved in the solution or prepared in situ.
  • In one particular embodiment of this aspect of the invention the input compound of formula (I) in the first, second and third processes is in the form of a substantially amorphous solid. Preferably the compound of formula (I) in the form of a substantially amorphous solid is preferably in the form of substantially amorphous particles. For example the compound of formula (I) in the form of substantially amorphous particles may be obtained by spray drying a solution containing the compound of formula (I). Any solvent that will dissolve the compound of formula (I) that can be evaporated safely in a spray drying process may be used. Suitable solvents for forming the solution include, but are not limited to, methyl acetate, ethyl acetate, isopropyl acetate, acetone, 2-butanone, 3-pentanone, 4-methyl-2-pentanone, ethanol, methanol, 1-propanol, propan-2-ol, acetonitrile, chloroform, dichloromethane especially methylethylketone (2-butanone). Solution concentration will typically be 0.5-50% specifically 10-40% eg 20-30%. Lower concentrations may be more suitable for preparing smaller particle sizes especially 24% e.g. 3.54%. The concentration that may be employed will be limited by the dissolution power of the solvent. Methylethylketone is preferred since it dissolves compound of formula (I) at a relatively high concentration which results in production advantages. The compound of formula (I) may be employed in non-solvated form or in the form of a composition of the invention (e.g. with acetone). Preferably it is employed as the [0090] non-solvated Form 1 polymorph. Spray drying maybe performed, for example, using apparatus supplied by Buchi or Niro. A pneumatic spray nozzle orifice of e.g. 0.04 inches is suitable, although alternate atomization methods such as rotary and pressure nozzles can be used. Solution flow rate may typically be in the range 1-100 ml/min, especially 15-30 ml/min. The inlet temperature and flow rate combination should be suitable to evaporate the solvent completely to minimize the risk of solvent trapped in the particle expediting an amorphous to crystalline transition. Inlet temperatures can range from 50-250° C., typically 100-200° C.
  • Compound of formula (I) in unsolvated form which is itself a useful substance has been found to exist in 3 crystalline polymorphic forms, [0091] Forms 1, 2 and 3, although Form 3 may be an unstable variant of Form 2. The Forms are characterised by their XRPD patterns shown in FIG. 8. Broadly speaking the Forms are characterised in their XRPD profiles by the absence of guest molecules and by peaks as follows:
  • Form 1: Peak at around 18.9 degrees 2Theta [0092]
  • Form 2: Peaks at around 18.4 and 21.5 degrees 2Theta [0093]
  • Form 3: Peaks at around 18.6 and 19.2 degrees 2Theta. [0094]
  • Forms 1 appears likely to be the thermodynamically most stable form since [0095] Forms 2 and 3 are converted into Form 1 on heating.
  • A process for preparing a compound of formula (I) as crystalline [0096] unsolvated Form 1 polymorph comprises dissolving compound of formula (I) in methylisobutylketone or ethyl acetate and producing compound of formula (I) as unsolvated Form 1 by addition of an anti-solvent such as iso-octane or toluene.
  • According to a first preferred embodiment of this process the compound of formula (I) may be dissolved in ethyl acetate and compound of formula (I) as [0097] unsolvated Form 1 polymorph may be obtained by addition of toluene as anti-solvent. In order to improve the yield, preferably the ethyl acetate solution is hot and once the toluene has been added the mixture is distilled to reduce the content of ethyl acetate.
  • According to a second preferred embodiment of this process the compound of formula (I) may be dissolved in methylisobutylketone and compound of formula (I) as crystalline [0098] unsolvated Form 1 polymorph may be obtained by addition of isooctane as anti-solvent.
  • A process for preparing a compound of formula (I) as [0099] unsolvated Form 2 polymorph comprises dissolving compound of formula (I) in unsolvated form in methanol or dry dichloromethane and recrystallising the compound of formula (I) as unsolvated Form 2 polymorph. Typically the compound of formula (I) will be dissolved in hot methanol or dry dichloromethane and allowed to cool.
  • A process for preparing a compound of formula (I) as [0100] unsolvated Form 3 polymorph comprises dissolving compound of formula (I) in particular as the composition with acetone in dichloromethane in the presence of water (typically 1-3% water by volume) and recrystallising the compound of formula (I) as unsolvated Form 3 polymorph.
  • As mentioned above, compositions of the invention may also find use as manufacturing intermediates in the preparation of compound of formula (I) in unsolvated form, or in the preparation of other compositions of the invention, or in pharmaceutical compositions thereof. [0101]
  • For example, a process for preparation of compound of formula (I) in unsolvated form (typically unsolvated polymorph Form 1) comprises removing the guest molecule from a composition of the invention. [0102]
  • A process for preparing a compound of formula (I) comprises alkylation of a thioacid of formula (II) [0103]
    Figure US20040220156A1-20041104-C00004
  • or a salt thereof. [0104]
  • In this process the compound of formula (II) may be reacted with a compound of formula FCH[0105] 2L wherein L represents a leaving group (e.g. a halogen atom, a mesyl or tosyl group or the like) for example, an appropriate fluoromethyl halide under standard conditions. Preferably, the fluoromethyl halide reagent is bromofluoromethane. Preferably the compound of formula (II) is employed as a salt, particularly the salt with diisopropylethylamine.
  • In a preferred process for preparing the compound of formula (I), the compound of formula (II) or a salt thereof is treated with bromofluoromethane optionally in the presence of a phase transfer catalyst. A preferred solvent is methylacetate, or more preferably ethylacetate, optionally in the presence of water. The presence of water improves solubility of both starting material and product and the use of a phase transfer catalyst results in an increased rate of reaction. Examples of phase transfer catalysts that may be employed include (but are not restricted to) tetrabutylammonium bromide, tetrabutylammonium chloride, benzyltributylammonium bromide, benzyltributylammonium chloride, benzyltriethylammonium bromide, methyltributylammonium chloride and methyltrioctylammonium chloride. THF has also successfully been employed as solvent for the reaction wherein the presence of a phase transfer catalyst again provides a significantly faster reaction rate. Preferably the product present in an organic phase is washed firstly with aqueous acid e.g. dilute HCl in order to remove amine compounds such as triethylamine and diisopropylethylamine and then with aqueous base e.g. sodium bicarbonate in order to remove any unreacted precursor compound of formula (II). [0106]
  • Compounds of formula (II) may be prepared from the corresponding 17α-hydroxyl derivative of formula (III): [0107]
    Figure US20040220156A1-20041104-C00005
  • using for example, the methodology described by G. H. Phillipps et al., (1994) Journal of Medicinal Chemistry, 37, 3717-3729. For example the step typically comprises the addition of a reagent suitable for performing the esterification e.g. an activated derivative of 2-furoic acid such as an activated ester or preferably a 2-furoyl halide e.g. 2-furoyl chloride (employed in at least 2 times molar quantity relative to the compound of formula (III)) in the presence of an organic base e.g. triethylamine. The second mole of 2-furoyl chloride reacts with the thioacid moiety in the compound of formula (III) and needs to be removed e.g. by reaction with an amine such as diethylamine. [0108]
  • This method suffers disadvantages, however, in that the resultant compound of formula (II) is not readily purified of contamination with the by-product 2-furoyldiethylamide. We have therefore invented several improved processes for performing this conversion. [0109]
  • In a first such improved process we have discovered that by using a more polar amine such as diethanolamine, a more water soluble by-product is obtained (in this case 2-furoyldiethanolamide) which permits compound of formula (II) or a salt thereof to be produced in high purity since the by-product can efficiently be removed by water washing. [0110]
  • Thus we provide a process for preparing a compound of formula (II) which comprises: [0111]
  • (a) reacting a compound of formula (III) with an activated derivative of 2-furoic acid as in an amount of at least 2 moles of the activated derivative per mole of compound of formula (III) to yield a compound of formula (IIA) [0112]
    Figure US20040220156A1-20041104-C00006
  • ; and [0113]
  • (b) removal of the sulphur-linked 2-furoyl moiety from compound of formula (IIA) by reaction of the product of step (a) with an organic primary or secondary amine base capable of forming a water soluble 2-furoyl amide. [0114]
  • In two particularly convenient embodiments of this process we also provide methods for the efficient purification of the end product which comprise either [0115]
  • (c1) when the product of step (b) is dissolved in a substantially water immiscible organic solvent, purifying the compound of formula (II) by washing out the amide by-product from step (b) with an aqueous wash, or [0116]
  • (c2) when the product of step (b) is dissolved in a water miscible solvent, purifying the compound of formula (II) by treating the product of step (b) with an aqueous medium so as to precipitate out pure compound of formula (II) or a salt thereof. [0117]
  • In step (a) preferably the activated derivative of 2-furoic acid may be an activated ester of 2-furoic acid, but is more preferably a 2-furoyl halide, especially 2-furoyl chloride. A suitable solvent for this reaction is ethylacetate or methylacetate (preferably methylacetate) (when step (c1) may be followed) or acetone (when step (c2) may be followed). Normally an organic base e.g. triethylamine will be present. In step (b) preferably the organic base is diethanolamine. The base may suitably be dissolved in a solvent e.g. methanol. Generally steps (a) and (b) will be performed at reduced temperature e.g. between 0 and 5° C. In step (c1) the aqueous wash may be water, however the use of brine results in higher yields and is therefore preferred. [0118]
  • In step (c2) the aqueous medium is for example a dilute aqueous acid such as dilute HCl. [0119]
  • We also provide an alternative process for preparing a compound of formula (II) which comprises: [0120]
  • (a) reacting a compound of formula (III) with an activated derivative of 2-furoic acid in an amount of at least 2 moles of activated derivative per mole of compound of formula (III) to yield a compound of formula (IIA); and [0121]
  • (b) removal of the sulphur-linked 2-furoyl moiety from compound of formula (IIA) by reaction of the product of step (a) with a further mole of compound of formula (III) to give two moles of compound of formula (II). [0122]
  • In step (a) preferably the activated derivative of 2-furoic acid may be an activated ester of 2-furoic acid, but is more preferably a 2-furoyl halide, especially 2-furoyl chloride. A suitable solvent for his step is acetone. Normally an organic base e.g. triethylamine will be present. In step (b) a suitable solvent is DMF or dimethylacetamide. Normally an organic base e.g. triethylamine will be present. Generally steps (a) and (b) will be performed at reduced temperature e.g. between 0 and 5° C. The product may be isolated by treatment with acid and washing with water. [0123]
  • This aforementioned process is very efficient in that it does not produce any furoylamide by-product (thus affording inter alia environmental advantages) since the excess mole of furoyl moiety is taken up by reaction with a further mole of compound of formula (II) to form an additional mole of compound of formula (II). [0124]
  • Further general conditions for the conversion of compound of formula (III) to compound of formula (II) in the two processes just described will be well known to persons skilled in the art. [0125]
  • According to a preferred set of conditions, however, we have found that the compound of formula (II) may advantageously be isolated in the form of a solid crystalline salt. The preferred salt is a salt formed with a base such as triethylamine, 2,4,6-trimethylpyridine, diisopropylethylamine or N-ethylpiperidine. Such salt forms of compound of formula (II) are more stable, more readily filtered and dried and can be isolated in higher purity than the free thioacid. The most preferred salt is the salt formed with diisopropylethylamine. The triethylamine salt is also of interest. [0126]
  • Compounds of formula (III) may be prepared in accordance with procedures described in GB 2088877B. [0127]
  • Compounds of formula (III) may also be prepared by a process comprising the following steps: [0128]
    Figure US20040220156A1-20041104-C00007
  • Step (a) comprises oxidation of a solution containing the compound of formula (V). [0129]
  • Preferably, step (a) will be performed in the presence of a solvent comprising methanol, water, tetrahydrofuran, dioxan or diethylene glygol dimethylether. So as to enhance yield and throughput, preferred solvents are methanol, water or tetrahydrofuran, and more preferably are water or tetrahydrofuran, especially water and tetrahydrofuran as solvent. Dioxan and diethylene glygol dimethylether are also preferred solvents which may optionally (and preferably) be employed together with water. Preferably, the solvent will be present in an amount of between 3 and 10 vol relative to the amount of the starting material (1 wt.), more preferably between 4 and 6 vol., especially 5 vol. Preferably the oxidising agent is present in an amount of 1-9 molar equivalents relative to the amount of the starting material. For example, when a 50% w/w aqueous solution of periodic acid is employed, the oxidising agent may be present in an amount of between 1.1 and 10 wt. relative to the amount of the starting material (1 wt.), more preferably between 1.1 and 3 wt., especially 1.3 wt. Preferably, the oxidation step will comprise the use of a chemical oxidising agent. More preferably, the oxidising agent will be periodic acid or iodic acid or a salt thereof. [0130]
  • Most preferably, the oxidising agent will be periodic acid or sodium periodate, especially periodic acid. Alternatively (or in addition), it will also be appreciated that the oxidation step may comprise any suitable oxidation reaction, e.g. one which utilises air and/or oxygen. When the oxidation reaction utilises air and/or oxygen, the solvent used in said reaction will preferably be methanol. Preferably, step (a) will involve incubating the reagents at room temperature or a little warmer, say around 25° C. e.g. for 2 hours. The compound of formula (IV) may be isolated by recrystallisation from the reaction mixture by addition of an anti-solvent. A suitable anti-solvent for compound of formula (IV) is water. Surprisingly we have discovered that it is highly desirable to control the conditions under which the compound of formula (IV) is precipitated by addition of anti-solvent e.g. water. When the recrystallisation is performed using chilled water (e.g. water/ice mixture at a temperature of 0-5° C.) although better anti-solvent properties may be expected we have found that the crystalline product produced is very voluminous, resembles a soft gel and is very difficult to filter. Without being limited by theory we believe that this low density product contains a large amount of solvated solvent within the crystal lattice. By contrast when conditions of around 10° C. or higher are used (e.g. around ambient temperature) a granular product of a sand like consistency which is very easily filtered is produced. Under these conditions, crystallisation typically commences after around 1 hour and is typically completed within a few hours (e.g. 2 hours). Without being limited by theory we believe that this granular product contains little or no solvated solvent within the crystal lattice. [0131]
  • Step (b) will typically comprise the addition of a reagent suitable for converting a carboxylic acid to a carbothioic acid e.g. using hydrogen sulphide gas together with a suitable coupling agent e.g. carbonyldiimidazole (CDI) in the presence of a suitable solvent e.g. dimethylformamide. [0132]
  • The advantages of the composition comprising a compound of formula (I) together with a guest compound according to the invention may include the fact that the substance appears to demonstrate excellent anti-inflammatory properties, with predictable pharmacokinetic and pharmacodynamic behaviour, with an attractive side-effect profile, long duration of action, and is compatible with a convenient regime of treatment in human patients, in particular being amenable to once-per day dosing. Further advantages may include the fact that the substance has desirable physical and chemical properties which allow for ready manufacture and storage. Alternatively it may serve as a useful intermediate in the preparation of other forms of the compound of formula (I) or compositions thereof. [0133]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a composition of the invention with toluene (guest molecule darkened). [0134]
  • FIG. 2: Figure showing hydrogen bond interactions between steroid and guest for the composition of the invention with toluene [0135]
  • FIG. 3: Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a composition of the invention with toluene, and hydrogen bond interactions between two steroid molecules [0136]
  • FIG. 4: Enlarged XRPD profile of composition of the invention with toluene [0137]
  • FIG. 5: Enlarged XRPD profile of composition of the invention with m-xylene [0138]
  • FIG. 6: Enlarged XRPD profile of composition of the invention with o-xylene [0139]
  • FIG. 7: Enlarged XRPD profile of composition of the invention with chlorobenzene [0140]
  • FIG. 8: XRPD profiles of [0141] unsolvated polymorphs 1, 2 and 3
  • DETAILED DESCRIPTION
  • The following non-limiting Examples illustrate the invention: [0142]
  • EXAMPLES
  • General [0143]
  • [0144] 1H-nmr spectra were recorded at 400 MHz and the chemical shifts are expressed in ppm relative to tetramethylsilane. The following abbreviations are used to describe the multiplicities of the signals: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), dd (doublet of doublets), ddd (doublet of doublet of doublets), dt (doublet of triplets) and b (broad). Biotage refers to prepacked silica gel cartridges containing KP-Sil run on flash 12i chromatography module. LCMS was conducted on a Supelcosil LCABZ+PLUS column (3.3 cm×4.6 mm ID) eluting with 0.1% HCO2H and 0.01 M ammonium acetate in water (solvent A), and 0.05% HCO2H 5% water in acetonitrile (solvent B), using the following elution gradient 0-0.7 min 0% B, 0.7-4.2 min 100% B, 4.2-5.3 min 0% B, 5.3-5.5 min 0% B at a flow rate of 3 ml/min. The mass spectra were recorded on a Fisons VG Platform spectrometer using electrospray positive and negative mode (ES+ve and ES-ve).
  • The XRPb analyses shown in the figures were performed on [0145]
  • a) a Phillips X'pert MPD powder diffractometer, serial number DY667. The pattern was recorded using the following acquisition conditions: Tube anode: Cu, Start angle: 2.0 °2θ, End angle: 45.0 °2θ, Step size: 0.02 °2θ, Time per step: 1 second. [0146]
  • XRPD profiles were collected at ambient temperature (295K) (FIG. 8); [0147]
  • b) a Philips PW1710 powder diffractometer. The pattern was recorded using the following acquisition conditions: Tube anode: Cu, Start angle: 3.5 °2θ, End angle: 35.0 °2θ, Step size: 0.02 °2θ, Time per step: 2.3 seconds. XRPD profiles were collected at ambient temperature (295K) (FIGS. [0148] 4-7);
  • The diffractometer used in each case can be determined by the end angle in the figure. [0149]
  • X-ray diffraction pattern collections referred to in Table 1 were performed in the following manners: [0150]
  • The crystal and molecular structures and corresponding unit cell dimensions were determined from three-dimensional X-ray diffraction data collected at 120+/−2K. All measurements were made using a Bruker SMART CCD diffractometer with graphite monochromated Mo-Kα radiation (λ=0.71073 Å) from a fine focus sealed tube source. The structure was solved by direct methods and refined using full-matrix least-squares procedures which minimized the function Sw(Fo[0151] 2−Fc2)2. The Bruker SHELX software was used throughout.
  • Intermediates [0152]
  • Intermediate 1: 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid diisopropylethylamine salt [0153]
  • A stirred suspension of 6α, 9α-difluoro-11β, 17α-dihydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid (prepared in accordance with the procedure described in GB 2088877B) (49.5 g) in methylacetate (500 ml) is treated with triethylamine (35 ml) maintaining a reaction temperature in the range 0-5° C. 2-Furoyl chloride (25 ml) is added and the mixture stirred at 0-5° C. for 1 hour. A solution of diethanolamine (52.8 g) in methanol (50 ml) is added and the mixture stirred at 0-5° C. for at least 2 hours. Dilute hydrochloric acid (approx 1M, 550 ml) is added maintaining a reaction temperature below 15° C. and the mixture stirred at 15° C. The organic phase is separated and the aqueous phase is back extracted with methyl acetate (2×250 ml). All of the organic phases are combined, washed sequentially with brine (5×250 ml) and treated with di-isopropylethylamine (30 ml). The reaction mixture is concentrated by distillation at atmospheric pressure to an approximate volume of 250 ml and cooled to 25-30° C. (crystallisation of the desired product normally occurs during distillation/subsequent cooling). Tertiary butyl methyl ether (TBME) (500 ml) is added, the slurry further cooled and aged at 0-5° C. for at least 10 minutes. The product is filtered off, washed with chilled TBME (2×200 ml) and dried under vacuum at approximately 40-50° C. (75.3 g, 98.7%). NMR (CDCl[0154] 3) δ: 7.54-7.46 (1H, m), 7.20-7.12 (1H, dd), 7.07-6.99 (1H, dd), 6.48-6.41 (2H, m), 6.41-6.32 (1H, dd), 5.51-5.28 (1H, dddd 2JH-F 50 Hz), 4.45-4.33(1H, bd), 3.92-3.73 (3H, bm), 3.27-3.14 (2H, q), 2.64-2.12 (5H, m), 1.88-1.71 (2H, m), 1.58-1.15 (3H, s), 1.50-1.38 (15H, m), 1.32-1.23 (1H, m), 1.23-1.15 (3H s), 1.09-0.99 (3H, d)
  • Intermediate 2: 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl [0155] ester Unsolvated Form 1
  • A mobile suspension of Intermediate 1 (12.61 g, 19.8 mmol) in ethyl acetate (230 ml) and water (50 ml) is treated with a phase transfer catalyst (benzyltributylammonium chloride, 10 mol %), cooled to 3° C. and treated with bromofluoromethane (1.10 ml, 19.5 mmol, 0.98 equivalents), washing in with prechilled (0° C.) ethyl acetate (EtOAc) (20 ml). The suspension is stirred overnight, allowing to warm to 17° C. The aqueous layer is separated and the organic phase is sequentially washed with 1M HCl (50 ml), 1% w/v NaHCO[0156] 3 solution (3×50 ml) and water (2×50 ml). The ethylacetate solution is distilled at atmospheric pressure until the distillate reaches a temperature of approximately 73° C. at which point toluene (150 ml) is added. Distillation is continued at atmospheric pressure until all remaining EtOAc has been removed (approximate distillate temperature 103° C.). The resultant suspension is cooled and aged at <10° C. and filtered off. The bed is washed with toluene (2×30 ml) and the product oven dried under vacuum at 60° C. to constant weight to yield the title compound (8.77 g, 82%) LCMS retention time 3.66 min, m/z 539 MH+, NMR δ (CDCl3) includes 7.60 (1H, m), 7.18-7.11 (2H, m), 6.52 (1H, dd, J 4.2 Hz), 6.46 (1H, s), 6.41 (1H, dd, J 10, 2 Hz), 5.95 and 5.82 (2H dd, J 51, 9 Hz), 5.48 and 5.35 (1H, 2 m), 4.48 (1H, m), 3.48 (1H, m), 1.55 (3H, s), 1.16 (3H, s), 1.06 (3H, d, J 7 Hz).
  • Pharmacological Activity [0157]
  • In Vitro Pharmacological Activity [0158]
  • Pharmacological activity was assessed in a functional in vitro assay of glucocorticoid agonist activity which is generally predictive of anti-inflammatory or anti-allergic activity in vivo. [0159]
  • For the experiments in this section, compound of formula (I) was used as unsolvated Form 1 (Intermediate 2) [0160]
  • The functional assay was based on that described by K. P. Ray et al., Biochem J. (1997), 328, 707-715. A549 cells stably transfected with a reporter gene containing the NF-κB responsive elements from the ELAM gene promoter coupled to sPAP (secreted alkaline phosphatase) were treated with test compounds at appropriate doses for 1 hour at 37° C. The cells were then stimulated with tumour necrosis factor (TNF, 10 ng/ml) for 16 hours, at which time the amount of alkaline phosphatase produced is measured by a standard colourimetric assay. Dose response curves were constructed from which EC[0161] 50 values were estimated.
  • In this test the compound of formula (1) showed an EC[0162] 50 value of <1 nM.
  • The glucocorticoid receptor (GR) can function in at least two distinct mechanisms, by upregulating gene expression through the direct binding of GR to specific sequences in gene promotors, and by downregulating gene expression that is being driven by other transcription factors (such as NFκB or AP-1) through their direct interaction with GR. [0163]
  • In a variant of the above method, to monitor these functions, two reporter plasmids have been generated and introduced separately into A549 human lung epithelial cells by transfection. The first cell line contains the firefly luciferase reporter gene under the control of a synthetic promoter that specifically responds to activation of the transcription factor NFκB when stimulated with TNFα. The second cell line contains the renilla luciferase reporter gene under the control of a synthetic promotor that comprises 3 copies of the consensus glucocorticoid response element, and which responds to direct stimulation by glucocorticoids. Simultaneous measurement of transactivation and transrepression was conducted by mixing the two cell lines in a 1:1 ratio in 96 well plate (40,000 cells per well) and growing overnight at 37° C. Test compounds were dissolved in DMSO, and added to the cells at a final DMSO concentration of 0.7%. After incubation for 1 h 0.5 ng/ml TNFα (R&D Systems) was added and after a further 15 hours at 37° C., the levels of firefly and renilla luciferase were measured using the Packard Firelite kit following the manufacturers' directions. Dose response curves were constructed from which EC[0164] 50 values were determined.
    Transactivation Transrepression
    (GR) (NFκB)
    ED50 (nM) ED50 (nM)
    Compound of Formula (I) 0.06 0.20
    Metabolite (X) >250 >1000
    Fluticasone propionate 0.07 0.16
  • In Vivo Pharmacological Activity [0165]
  • Pharmacological activity in vivo was assessed in an ovalbumin sensitised Brown Norway rat eosinophilia model. This model is designed to mimic allergen induced lung eosinophilia, a major component of lung inflammation in asthma. [0166]
  • For the experiments in this section, compound of formula (I) was used as [0167] unsolvated Form 1.
  • Compound of formula (I) produced dose dependant inhibition of lung eosinophilia in this model after dosing as an intra-tracheal (IT) suspension in [0168] saline 30 min prior to ovalbumin challenge. Significant inhibition is achieved after a single dose of 30 μg of compound of formula (I) and the response was significantly (p=0.016) greater than that seen with an equivalent dose of fluticasone propionate in the same study (69% inhibition with compound of formula (I) vs 41% inhibition with fluticasone propionate).
  • In a rat model of [0169] thymus involution 3 daily IT doses of 100 μg of compound (I) induced significantly smaller reductions in thymus weight (p=0.004) than an equivalent dose of fluticasone propionate in the same study (67% reduction of thymus weight with compound (I) vs 78% reduction with fluticasone propionate).
  • Taken together these results indicate a superior therapeutic index for compound (I) compared to fluticasone propionate. [0170]
  • In Vitro Metabolism in Rat and Human Hepatocytes [0171]
  • Incubation of compound (I) with rat or human hepatocytes shows the compound to be metabolised in an identical manner to fluticasone propionate with the 17-β carboxylic acid (X) being the only significant metabolite produced. Investigation of the rate of appearance of this metabolite on incubation of compound (I) with human hepatocytes (37° C., 10M drug concentration, hepatocytes from 3 subjects, 0.2 and 0.7 million cells/mL) shows compound (i) to be metabolised ca. 5-fold more rapidly than fluticasone propionate:— [0172]
    17-β acid metabolite production
    (pmol/h)
    Subject Cell density Fluticasone
    number (million cells/mL) Compound (I) propionate
    1 0.2 48.9 18.8
    1 0.7 73.3 35.4
    2 0.2 118 9.7
    2 0.7 903 23.7
    3 0.2 102 6.6
    3 0.7 580 23.9
  • Median metabolite production 102-118 pmol/h for compound (I) and 18.8-23.0 pmol/h for fluticasone propionate. [0173]
  • Pharmacokinetics After Intravenous (IV) and Oral Dosing in Rats [0174]
  • Compound (I) was dosed orally (0.1 mg/kg) and IV (0.1 mg/kg) to male Wistar Han rats and pharmacokinetic parameters determined. Compound (I) showed negligible oral bioavailability (0.9%) and plasma clearance of 47.3 mL/min/kg, approaching liver blood flow (plasma clearance of fluticasone propionate=45.2 mL/min/kg). [0175]
  • Pharmacokinetics After Intra-Tracheal Dry Powder Dosing in the Pig. [0176]
  • Anaesthetised pigs (2) were dosed intra-tracheally with a homogenous mixture of compound (I) (1 mg) and fluticasone propionate (1 mg) as a dry powder blend in lactose (10% w/w). Serial blood samples were taken for up to 8 h following dosing. [0177]
  • Plasma levels of compound (I) and fluticasone propionate were determined following extraction and analysis using LC-MS/MS methodology, the lower limits of quantitation of the methods were 10 and 20 μg/mL for compound (I) and fluticasone propionate respectively. Using these methods compound (I) was quantifiable up to 2 hours after dosing and fluticasone propionate was quantifiable up to 8 hours after dosing. Maximum plasma concentrations were observed for both compounds within 15 min after dosing. Plasma half-life data obtained from IV dosing (0.1 mg/kg) was used to calculate AUC (0-inf) values for compound (I). This compensates for the plasma profile of Compound (I) only being defined up to 2 hours after an IT dose and removes any bias due to limited data between compound (I) and fluticasone propionate. [0178]
  • C[0179] max and AUC (0-inf) values show markedly reduced systemic exposure to compound (I) compared to fluticasone propionate:—
    Cmax AUC (0-inf)
    (pg/mL) (hr · pg/mL)
    Pig 1 Pig 2 Pig 1 Pig 2
    Compound of Formula (I) 117  81 254 221
    Fluticasone propionate 277 218 455 495
  • The pharmacokinetic parameters for both compound (I) and fluticasone propionate were the same in the anaesthetised pig following intravenous administration of a mixture of the two compounds at 0.1 mg/kg. The clearance of these two glucocorticoids is similar is this experimental pig model. [0180]
  • Intermediate 3: 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester, amorphous particles [0181]
  • Intermediate 2 (30.04 g) was dissolved in methylethylketone (850 ml) to give a 3.5% solution. The solution was spray dried using a Niro Mobile Minor spray drier (Niro Inc, Columbia, Md., USA). The spray orifice was a two fluid pneumatic nozzle with 0.04 inch orifice diameter (Spray Systems Co, Wheaton, Ill., USA). The other spray drying parameters were as follows: [0182]
  • Temperature: 150° C., outlet temperature 98° C. [0183]
  • Solution flow rate: 30 ml/min using Isco 260D syringe pump (Isco Inc, Lincoln, Nebr., USA) [0184]
  • Atomisation Pressure: 2 Bar [0185]
  • Particle collection was achieved in the conventional manner using a Fisher Klosterman XQ120-1.375 high efficiency cyclone (Fisher-Klosterman Inc, Louisville, Ky., USA). A white powder was recovered. The spray drying process was successful at producing smooth, spherical particles of amorphous 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester. System yield was 61% [0186]
  • Intermediate 4 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl [0187] ester Unsolvated Form 2
  • EXAMPLES Example 1 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester Composition with toluene
  • Intermediate 4 (200 mg) was slurried in toluene (5 mL) for 3 hours. The solid was then recovered by filtration to afford the title compound as a white solid. [0188]
  • Stoichiometry of compound of formula (I): guest=1:3.1 from [0189] 1H nmr (CDCl3).
  • Example 2 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester Composition with m-xylene
  • Intermediate 4 (200 mg) was slurried in m-xylene (6 mL) for 2 hours. The solid was then recovered by filtration to afford the title compound as a white solid. [0190]
  • Example 3 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester Composition with o-xylene (first alternative method)
  • Intermediate 3 (100 mg) was slurried in o-xylene (1 mL) at 21° C. for 16 hours. The solid was recovered by filtration to afford the title compound as a white solid. [0191]
  • Stoichiometry of compound of formula (I): guest=1:1.5 from [0192] 1H nmr (CDCl3).
  • Example 4 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester Composition with o-xylene (second alternative method)
  • Intermediate 2 (3.0 g) was dissolved in a mixture of o-xylene (60 mL) and acetonitrile (60 mL). The solvent was allowed to evaporate under ambient conditions until the crystallisation had proceeded sufficiently. The solid was then recovered by filtration to afford the title compound as a white solid. [0193]
  • Stoichiometry of compound of formula (I): guest=1:4 from [0194] 1H nmr (CDCl3).
  • Example 5 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester Composition with chlorobenzene
  • Intermediate 4 (200 mg) was slurried in m-xylene (5 mL) for 3 hours. The solid was then recovered by filtration to afford the title compound as a white solid. [0195]
  • Further Characterising Data on Compositions of the Invention: [0196]
  • Detailed XRPD profile peak information for various compositions of the invention is provided in Tables 2, 3, 4 and 5. [0197]
  • The XRPD profiles of compositions of the invention are provided in FIGS. 4, 5, [0198] 6 and 7.
  • We also claim compositions of the invention substantially by reference to their XRPD profiles as shown in the Figures and Tables. [0199]
  • Example A Dry powder composition containing 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene
  • A dry powder formulation may be prepared as follows: [0200]
  • 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester, composition with toluene prepared according to Example 1, MMD of 3 μm: 0.20 mg milled lactose (wherein not greater than 85% of particles have a MMD of 60-90 μm, and not less than 15% of particles have a MMD of less than 15 μm): 12 mg [0201]
  • A peelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared. [0202]
  • Example B Dry powder composition containing 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene and a long acting β2-adrenoreceptor agonist
  • A dry powder formulation may be prepared as follows: [0203]
  • 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3 μm: 0.20 mg Long-acting P[0204] 2-adrenoreceptor agonist (micronised to a MMD of 3 μm): 0.02 mg milled lactose (wherein not greater than 85% of particles have a MMD of 60-90 μm, and not less than 15% of particles have a MMD of less than 15 μm): 12 mg
  • A peelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared. [0205]
  • Example C Aerosol formulation containing 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene
  • prepared according to Example 1, MMD of 3 μm: [0206]
  • An aluminium canister may be filled with a formulation as follows: [0207]
  • 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3 μm: 250 [0208] μg
    1,1,1,2-tetrafluoroethane: to 50 μl
  • (amounts per actuation) [0209]
  • in a total amount suitable for 120 actuations and the canister may be fitted with a metering valve adapted to dispense 50 μl per actuation. [0210]
  • Example D Aerosol formulation containing 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17′-carbothioic acid S-fluoromethyl ester composition with toluene and a long acting β2-adrenoreceptor agonist
  • An aluminium canister may be filled with a formulation as follows: 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3 μm: 250 μg [0211]
    Long-acting β2-adrenoreceptor agonist   25 μg
    (micronised to a MMD of 3 μm):
    1,1,1,2-tetrafluoroethane: to 50 μl
  • (amounts per actuation) [0212]
  • in a total amount suitable for 120 actuations and the canister may be fitted with a metering valve adapted to dispense 50 μl per actuation. [0213]
  • Example E Nasal formulation containing 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene
  • A formulation for intranasal delivery may be prepared as follows: [0214]
  • 6α, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3 μm: 10 [0215] mg
    Polysorbate
    20 0.8 mg
    Sorbitan monolaurate 0.09 mg
    Sodium dihydrogen phosphate dihydrate 94 mg
    Dibasic sodium phosphate anhydrous 17.5 mg
    Sodium chloride 48 mg
    Demineralised water to 10 ml
  • The formulation may be fitted into a spraypump capable of delivering a plurality of metered doses (Valois). [0216]
  • 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 or step or group of integers but not to the exclusion of any other integer or step or group of integers or steps. [0217]
  • The patents and patent applications described in this application are herein incorporated by reference. [0218]
    TABLE 2
    XRPD characteristic angles and relative intensities for 6α,
    9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-
    16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid
    S-fluoromethyl ester, composition with toluene
    Angle Rel. Intensity
    2-Theta ° %
    4.9 25.6
    6.8 12.2
    8.0 43.6
    9.9 1.7
    11.8 15.5
    13.0 13.1
    14.0 12.9
    15.0 100.0
    16.3 53.2
    16.7 12.0
    17.9 14.2
    18.7 83.2
    19.4 13.3
    20.1 26.6
    21.1 19.7
    21.7 17.2
    22.8 14.5
    23.5 37.2
    23.9 34.6
    24.5 14.0
    25.3 10.5
    25.8 15.5
    26.1 20.5
    27.0 18.8
    28.8 8.2
    30.7 39.2
    31.1 21.4
    32.7 14.0
    34.0 14.0
    34.7 18.6
  • [0219]
    TABLE 3
    XRPD characteristic angles and relative intensities for 6α,
    9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-
    16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid
    S-fluoromethyl ester, composition with m-xylene
    Angle Rel. Intensity
    2-Theta ° %
    4.8 38.7
    6.8 12.7
    7.9 22.8
    9.6 6.2
    11.5 24.1
    13.0 11.8
    13.7 26.9
    14.5 100.0
    15.8 17.9
    16.1 25.7
    16.4 12.4
    17.0 7.7
    17.6 12.5
    18.3 52.1
    19.3 34.9
    20.4 6.4
    21.2 22.2
    22.3 16.3
    22.7 18.4
    23.3 21.1
    24.3 14.0
    25.1 12.8
    25.6 14.6
    26.3 8.1
    26.8 9.5
    27.6 6.9
    28.0 5.0
    28.6 5.8
    29.8 18.8
    31.4 8.8
    32.5 8.0
    33.1 8.7
    34.3 11.7
  • [0220]
    TABLE 4
    XRPD characteristic angles and relative intensities for 6α,
    9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-
    16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid
    S-fluoromethyl ester, composition with o-xylene
    Angle Rel. Intensity
    2-Theta ° %
    5.0 11.4
    6.9 13.6
    8.1 21.7
    9.9 1.6
    11.7 18.0
    13.1 15.4
    13.8 27.2
    14.8 100.0
    16.2 39.3
    16.6 16.8
    17.1 9.4
    17.7 12.0
    18.5 60.6
    19.3 10.7
    19.7 18.5
    20.6 5.1
    21.5 20.0
    22.8 21.3
    23.4 22.5
    24.4 17.6
    24.9 7.4
    25.6 21.5
    26.3 12.4
    26.8 12.0
    28.0 8.8
    28.7 7.0
    29.0 6.6
    30.2 16.6
    32.2 8.1
    32.5 9.4
    33.4 6.6
    34.2 12.3
  • [0221]
    TABLE 5
    XRPD characteristic angles and relative intensities for 6α,
    9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-
    16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid
    S-fluoromethyl ester, composition with chlorobenzene
    Angle Rel. Intensity
    2-Theta ° %
    4.3 34.4
    6.6 12.4
    10.7 10.2
    12.7 11.1
    13.3 10.5
    13.6 23.4
    14.6 22.9
    15.3 24.8
    16.9 43.2
    17.3 44.4
    17.6 100.0
    19.1 26.7
    19.4 12.4
    20.1 32.3
    20.9 24.3
    22.1 15.2
    23.0 47.0
    23.6 20.7
    24.5 10.5
    25.2 9.6
    27.4 12.7
    28.5 6.2
    29.6 28.1
    31.8 17.5
    33.3 6.2

Claims (40)

What is claimed is:
1. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00008
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7+0.7 Å, and 37±3 Å when determined at 120K,
wherein at about 295K said composition exhibits one or more of the following XRPD profiles:
(i) a peak in the range of around 4.3-5.0;
(ii) a peak in the range of around 6.6-6.9; or
(iii) a peak in the range of around 11.5-11.8.
2. The composition of claim 1, which exhibits XRPD profiles (i)-(iii).
3. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00009
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
wherein at about 295K said composition exhibits one or more of the following XRPD profiles:
(a) absence of a peak at around 7.3;
(b) absence of a peak at around 7.5;
(c) absence of a peak at around 12.5;
(d) absence of a peak at around 8.8-9.6;
(e) absence of a peak at around 10.5-11.1; or
(f) absence of a peak at around 12.2-12.6.
4. The composition of claim 3, which exhibits 3 or more of XRPD profiles (a)-(f).
5. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00010
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
wherein the compound of formula (I) is a nonsolvated Form 1 polymorph.
6. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00011
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
which is non-pressurised and adapted to be administered as a dry powder topically to the lung via the buccal cavity.
7. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00012
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7+0.6 Å, 13.7±0.7 Å, and 37+3 Å when determined at 120K,
which is non-pressurised and adapted to be administered topically to the nasal cavity.
8. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00013
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7+0.7 Å, and 37+3 Å when determined at 120K,
which further comprises a PDE4 inhibitor.
9. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00014
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7+0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
wherein the composition is selected from the group consisting of an ointment, lotion, cream, gel, foam, preparation for delivery by transdermal patch, powder, spray, aerosol, capsule or cartridge for use in an inhaler or insufflator or drop, solution or suspension for nebulisation, suppositories, pessaries, retention enemas, chewable or suckable tablets or pellets, liposome preparation and microencapsulation preparation.
10. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00015
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37+3 Å when determined at 120K,
wherein the composition is a dry powder or spray.
11. The composition according to claim 10, wherein the composition is a dry powder.
12. The composition according to claim 10, wherein the composition is a spray.
13. A pharmaceutical composition comprising a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00016
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K in combination with another therapeutically active agent, wherein the other therapeutically active ingredient is at least one selected from the group consisting of salmeterol, salbutamol, formoterol, salmefamol, fenoterol and terbutaline and salts thereof.
14. A crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00017
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K, wherein said compound of formula (I) is present in the amount of 0.001 to 10% by weight of said composition.
15. A pharmaceutical composition comprising a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00018
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
in combination with another therapeutically active agent, wherein said another therapeutically active agent is an anti-histamine, anti-inflammatory agent or antiinfective agent.
16. The composition according to claim 15, wherein said anti-histamine is methapyrilene or loratadine, said anti-inflammatory agent is an NSAID and said antiinfective agent is an antibiotic or antiviral.
17. A method for the treatment of a human or animal subject with an anti-inflammatory and/or allergic condition comprising administering to said human or animal subject an effective amount of a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00019
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
wherein said effective amount of the composition is administered topically to said human or animal subject.
18. The method for the treatment of a human or animal subject with an anti-inflammatory and/or allergic condition according to claim 17, wherein said effective amount of the composition is administered topically as a dry powder to the lung via the buccal cavity.
19. The method for the treatment of a human or animal subject with an anti-inflammatory and/or allergic condition comprising administering to said human or animal subject an effective amount of a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00020
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P2121 2 1 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
wherein said effective amount of the composition is administered once per day.
20. A method for the treatment of at least one condition selected from the group consisting of skin disease, inflammatory condition of the nose, throat or lungs, inflammatory bowel condition, auto-immune disease, conjunctiva and conjunctivitis in a human or animal subject, which comprises administering an effective amount of a pharmaceutical composition comprising a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00021
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P2±212, having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
and delivering said pharmaceutical composition to said human or animal subject in need thereof for the treatment of said at least one condition.
21. The method of treatment as recited in claim 20, wherein the skin disease is at least one selected from the group consisting of eczema, psoriasis, allergic dermatitis neurodermatitis, pruritis and hypersensitivity reactions.
22. The method of treatment as recited in claim 20, wherein the inflammatory condition of the nose, throat or lungs is at least one selected from the group consisting of asthma, rhinitis, nasal polyps, chronic obstructive pulmonary disease, interstitial lung disease and fibrosis.
23. The method of treatment as recited in claim 22, wherein the inflammatory condition of the nose, throat or lungs is asthma.
24. The method of treatment as recited in claim 22, wherein the inflammatory condition of the nose, throat or lungs is rhinitis.
25. The method of treatment as recited in claim 22, wherein the inflammatory condition of the nose, throat or lungs is chronic obstructive pulmonary disease.
26. The method of treatment as recited in claim 22, wherein the inflammatory bowel condition is at least one selected from the group consisting of ulcerative colitis and Crohn's disease.
27. The method of treatment as recited in claim 20, wherein said composition is administered by inhalation or by nebulisation.
28. The method of treatment as recited in claim 20, wherein said composition is administered orally, buccally, sublingually, parenterally, locally or rectally.
29. The method for the treatment of a human or animal subject with an anti-inflammatory and/or allergic condition according to claim 20, wherein said effective amount of the composition is administered topically to said human or animal subject.
30. An inhaler comprising a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00022
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K.
31. The inhaler of claim 30, further comprising propellant.
32. The inhaler of claim 30, further comprising at least one excipient selected from the group consisting of surfactant and cosolvent.
33. The inhaler of claim 30, wherein the composition is free of excipient.
34. The inhaler of claim 30, wherein the composition is retained in a pressurized canister closed with a valve.
35. The inhaler of claim 30, wherein said composition has a particle size in the range of 1-10 μm.
36. A process for preparing a crystalline chemical composition comprising a compound of formula (I)
Figure US20040220156A1-20041104-C00023
in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline composition is of space group P212121 having unit cell dimensions of about 7.7±0.6 Å, 13.7±0.7 Å, and 37±3 Å when determined at 120K,
which process comprises
(a) crystallising the composition from a solution containing a compound of formula (I) and the guest molecule; or
(b) contacting the compound of formula (I) or another composition according claim 1 in solid form with a liquid containing the guest molecule and obtaining the composition therefrom; or
contacting a compound of formula (I) or another composition according to claim 1 in solid form with a vapour containing the guest molecule,
wherein the compound of formula (I), which is crystallized in (a), or contacted with a liquid containing the guest molecule in (b), or contacted with a vapor containing the guest molecule in (c), is in the form of a substantially amorphous solid.
37. The process of claim 36, wherein the compound of formula (I) is in the form of substantially amorphous paticles.
38. The process of claim 36, wherein the compound of formula (I) as crystalline unsolvated Form 1 polymorph is prepared by dissolving the compound of formula (I) in methylisobutylketone or ethyl acetate and producing the compound of formula (I) as unsolvated Form 1 by addition of an anti-solvent.
39. The process of claim 36, wherein the compound of formula (I) as crystalline unsolvated Form 1 polymorph is prepared by dissolving the compound of formula (I) in ethyl acetate and producing the compound of formula (I) as unsolvated Form 1 by addition of toluene as the anti-solvent.
40. The process of claim 36, wherein the compound of formula (I) as crystalline unsolvated Form 1 polymorph is prepared by dissolving the compound of formula (I) in methylisobutylketone and producing the compound of formula (I) as unsolvated Form 1 by addition of isooctane as the anti-solvent.
US10/853,500 2000-08-05 2004-05-25 Novel anti-inflammatory androstane derivative compositions Abandoned US20040220156A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/853,500 US20040220156A1 (en) 2000-08-05 2004-05-25 Novel anti-inflammatory androstane derivative compositions

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GBGB0019172.6A GB0019172D0 (en) 2000-08-05 2000-08-05 Novel compounds
GB0019172.6 2000-08-05
US09/958,050 US7101866B2 (en) 2000-08-05 2001-08-03 Anti-inflammatory androstane derivative
US10/067,010 US6777399B2 (en) 2000-08-05 2002-02-04 Anti-inflammatory androstane derivative compositions
US10/200,364 US6858593B2 (en) 2000-08-05 2002-07-22 Anti-inflammatory androstane derivative compositions
US10/241,658 US6777400B2 (en) 2000-08-05 2002-09-11 Anti-inflammatory androstane derivative compositions
US10/853,500 US20040220156A1 (en) 2000-08-05 2004-05-25 Novel anti-inflammatory androstane derivative compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/241,658 Continuation US6777400B2 (en) 2000-08-05 2002-09-11 Anti-inflammatory androstane derivative compositions

Publications (1)

Publication Number Publication Date
US20040220156A1 true US20040220156A1 (en) 2004-11-04

Family

ID=31991223

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/241,658 Expired - Fee Related US6777400B2 (en) 2000-08-05 2002-09-11 Anti-inflammatory androstane derivative compositions
US10/853,500 Abandoned US20040220156A1 (en) 2000-08-05 2004-05-25 Novel anti-inflammatory androstane derivative compositions

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/241,658 Expired - Fee Related US6777400B2 (en) 2000-08-05 2002-09-11 Anti-inflammatory androstane derivative compositions

Country Status (5)

Country Link
US (2) US6777400B2 (en)
EP (1) EP1569951A2 (en)
JP (1) JP2006502172A (en)
AU (1) AU2003266390A1 (en)
WO (1) WO2004024745A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050054622A1 (en) * 2001-10-20 2005-03-10 Keith Biggadike Novel anti-inflammatory androstane derivatives
US9290698B2 (en) 2010-07-15 2016-03-22 Battelle Memorial Institute Biobased polyols for potential use as flame retardants in polyurethane and polyester applications

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL129407A0 (en) 1996-10-18 2000-02-17 Vertex Pharma Inhibitors of serine proteases particularly hepatitis C virus NS3 protease pharmaceutical compositions containing the same and the use thereof
GB0017988D0 (en) * 2000-07-21 2000-09-13 Glaxo Group Ltd Novel process
US6759398B2 (en) * 2000-08-05 2004-07-06 Smithkline Beecham Corporation Anti-inflammatory androstane derivative
GB0019172D0 (en) 2000-08-05 2000-09-27 Glaxo Group Ltd Novel compounds
US6777399B2 (en) * 2000-08-05 2004-08-17 Smithkline Beecham Corporation Anti-inflammatory androstane derivative compositions
AU2001276497B2 (en) 2000-08-05 2005-04-07 Glaxo Group Limited 17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
US6858596B2 (en) * 2000-08-05 2005-02-22 Smithkline Beecham Corporation Formulation containing anti-inflammatory androstane derivative
SV2003000617A (en) 2000-08-31 2003-01-13 Lilly Co Eli INHIBITORS OF PROTEASA PEPTIDOMIMETICA REF. X-14912M
UA77656C2 (en) 2001-04-07 2007-01-15 Glaxo Group Ltd S-fluoromethyl ester of 6-alpha, 9-alpha-difluoro-17-alpha-[(2-furanylcarbonyl)oxy]-11-beta-hydroxy-16- alpha-methyl-3-oxoandrosta-1,4-dien-17-beta-carbothioacid as anti-inflammatory agent
KR100831534B1 (en) * 2001-04-30 2008-05-22 글락소 그룹 리미티드 Anti-inflammatory 17.beta.-carbothioate ester derivatives of androstane with a cyclic ester group in position 17.alpha
ATE399174T1 (en) * 2001-06-12 2008-07-15 Glaxo Group Ltd NEW ANTI-INFLAMMATORY 17.ALPHA.-HETEROCYCLIC ESTERS OF 17.BETA.-CARBOTHIOATE ANDROSTANE DERIVATIVES
US20050175545A1 (en) * 2002-02-04 2005-08-11 Keith Biggadike Formulation for inhalation comprising a glucocorticoid and a beta 2-adrenoreceptor agonist
GB0202635D0 (en) * 2002-02-05 2002-03-20 Glaxo Wellcome Mfg Pte Ltd Formulation containing novel anti-inflammatory androstane derivative
GB2389530B (en) * 2002-06-14 2007-01-10 Cipla Ltd Pharmaceutical compositions
PE20050374A1 (en) 2003-09-05 2005-05-30 Vertex Pharma SERINE PROTEASE INHIBITORS, IN PARTICULAR HCV PROTEASE NS3-NS4A
WO2005032521A2 (en) * 2003-10-02 2005-04-14 Collegium Pharmaceutical, Inc. Non-flammable topical anesthetic liquid aerosols
US20060188449A1 (en) * 2003-10-03 2006-08-24 Jane Hirsh Topical aerosol foams
ZA200700030B (en) * 2004-06-08 2009-06-24 Vertex Pharma Pharmaceutical compositions
GB0507165D0 (en) * 2005-04-08 2005-05-18 Glaxo Group Ltd Novel crystalline pharmaceutical product
US7964624B1 (en) 2005-08-26 2011-06-21 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases
AR055395A1 (en) 2005-08-26 2007-08-22 Vertex Pharma INHIBITING COMPOUNDS OF THE ACTIVITY OF SERINA PROTEASA NS3-NS4A OF HEPATITIS C VIRUS
AU2006332726B2 (en) 2005-12-28 2012-12-13 Vertex Pharmaceuticals Incorporated. Solid forms of N-[2,4-bis(1,1-dimethylethyl)-5-hydroxyphenyl]-1,4-dihydro-4-oxoquinoline-3-carboxamide
US7780671B2 (en) * 2006-01-23 2010-08-24 Zimmer Technology, Inc. Bone resection apparatus and method for knee surgery
US8039475B2 (en) 2006-02-27 2011-10-18 Vertex Pharmaceuticals Incorporated Co-crystals and pharmaceutical compositions comprising the same
KR20080112303A (en) 2006-03-16 2008-12-24 버텍스 파마슈티칼스 인코포레이티드 Deuterated hepatitis c protease inhibitors
EP2674428B1 (en) 2006-04-07 2016-04-06 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US10022352B2 (en) 2006-04-07 2018-07-17 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US7645789B2 (en) 2006-04-07 2010-01-12 Vertex Pharmaceuticals Incorporated Indole derivatives as CFTR modulators
GB0615108D0 (en) * 2006-07-28 2006-09-06 Glaxo Group Ltd Novel formulations
US8563573B2 (en) 2007-11-02 2013-10-22 Vertex Pharmaceuticals Incorporated Azaindole derivatives as CFTR modulators
US8575208B2 (en) 2007-02-27 2013-11-05 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases
ES2379905T3 (en) * 2007-02-27 2012-05-04 Vertex Pharmceuticals Incorporated Co-crystals and pharmaceutical compositions comprising them
CL2008002549A1 (en) * 2007-08-30 2010-09-03 Vertex Pharma Cocrystal comprising vx-950 and a cocrystal former selected from 3-methoxy-4-hydroxybenzoic acid, 2,4-dihydroxybenzoic acid and 2,5-dihydroxybenzoic acid; Preparation method; Pharmaceutical composition comprising cocrystal, useful as an antiviral agent in the treatment of hcv.
US8652443B2 (en) * 2008-02-14 2014-02-18 Precision Dermatology, Inc. Foamable microemulsion compositions for topical administration
WO2010016931A2 (en) * 2008-08-07 2010-02-11 Plus Chemicals Sa Polymorphs of fluticasone furoate and process for preparation thereof
EP2408798A1 (en) 2009-03-19 2012-01-25 Plus Chemicals S.A. Polymorphs of fluticasone furoate and processes for preparation thereof
HRP20230661T1 (en) 2010-03-25 2023-09-29 Vertex Pharmaceuticals Incorporated Synthesis and intermediates of (r)-1(2,2 -difluorobenzo[d][1,3]dioxol-5yl)-n-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2yl)-1h-indol-5yl)cyclopropanecarboxamide
US8802868B2 (en) 2010-03-25 2014-08-12 Vertex Pharmaceuticals Incorporated Solid forms of (R)-1(2,2-difluorobenzo[D][1,3]dioxo1-5-yl)-N-(1-(2,3-dihydroxypropyl-6-fluoro-2-(1-hydroxy-2-methylpropan2-yl)-1H-Indol-5-yl)-Cyclopropanecarboxamide
NZ603042A (en) 2010-04-22 2015-02-27 Vertex Pharma Pharmaceutical compositions comprising cftr modulators and administrations thereof
CA2796646A1 (en) 2010-04-22 2011-10-27 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions and administrations thereof
EP2560650A1 (en) 2010-04-22 2013-02-27 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions and administrations thereof
WO2011133751A2 (en) 2010-04-22 2011-10-27 Vertex Pharmaceuticals Incorporated Process of producing cycloalkylcarboxamido-indole compounds
NZ606805A (en) 2010-08-23 2015-03-27 Vertex Pharma Pharmaceutical composition of (r)-1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-n-(1-(2,3-dihydroxy propyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1h-indol-5-yl) cyclopropanecarboxamide and administration therof
CA2874851A1 (en) 2012-06-08 2013-12-12 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cftr-mediated disorders
AU2013290444B2 (en) 2012-07-16 2018-04-26 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions of (R)-1-(2,2-diflurorbenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1H-indol-5-yl) cyclopropanecarboxamide and administration thereof
UA117464C2 (en) 2012-11-02 2018-08-10 Вертекс Фармасьютікалз Інкорпорейтед Pharmaceutical compositions for the treatment of cftr mediated diseases
PL3424534T3 (en) 2014-04-15 2021-11-22 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
WO2019010092A1 (en) 2017-07-01 2019-01-10 Vertex Pharmaceuticals Incorporated Compositions and methods for treatment of cystic fibrosis
TWI719349B (en) 2017-10-19 2021-02-21 美商維泰克斯製藥公司 Crystalline forms and compositions of cftr modulators
WO2019113089A1 (en) 2017-12-04 2019-06-13 Vertex Pharmaceuticals Incorporated Compositions for treating cystic fibrosis
TWI810243B (en) 2018-02-05 2023-08-01 美商維泰克斯製藥公司 Pharmaceutical compositions for treating cystic fibrosis
CN111662353A (en) * 2019-03-05 2020-09-15 上海谷森医药有限公司 Preparation method of fluticasone furoate crystal form 1
UY38696A (en) 2019-05-14 2020-11-30 Vertex Pharma ALPHA-1 ANTITRYPSIN MODULATORS
WO2020242935A1 (en) 2019-05-29 2020-12-03 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
CN112823009A (en) 2019-08-28 2021-05-18 上海谷森医药有限公司 Fluticasone furoate liposome preparation and preparation method thereof
US20240002386A1 (en) 2020-11-17 2024-01-04 Vertex Pharmaceuticals Incorporated Solid forms of 4-(5-(4-fluorophenyl)-6-(tetrahydro-2h-pyran-4-yl)-1,5-dihydropyrrolo[2,3-f]indazol-7-yl)benzoic acid
EP4340809A1 (en) 2021-05-17 2024-03-27 Cybin IRL Limited Formulations of psilocybin
AU2022284952A1 (en) 2021-06-04 2023-12-14 Vertex Pharmaceuticals Incorporated Solid dosage forms and dosing regimens comprising (2r,3s,4s,5r)-4-[[3-(3,4-difluoro-2-methoxy-phenyl)-4,5-dimethyl-5-(trifluoromethyl) tetrahydrofuran-2-carbonyl]amino]pyridine-2-carboxamide
WO2023247665A1 (en) 2022-06-22 2023-12-28 Cybin Irl Limited Solid dispersions of psilocybin
WO2024031081A1 (en) 2022-08-04 2024-02-08 Vertex Pharmaceuticals Incorporated Compositions for the treatment of cftr-mediated diseases

Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3067197A (en) * 1961-04-26 1962-12-04 Pfizer & Co C 11-oxygenated 6alpha-fluoro-16-methylene-delta-pregnenes and derivatives
US3639434A (en) * 1967-02-02 1972-02-01 Boots Pure Drug Co Ltd 17-acyloxysteroids and their manufacture
US3828080A (en) * 1972-01-20 1974-08-06 Glaxo Lab Ltd Androstane-17beta-carboxylic acids and processes for the preparation thereof
US3856828A (en) * 1972-07-19 1974-12-24 Glaxo Lab Ltd Anti-inflammatory steroids of the androstane series having a halo-substituted c{11 {14 c{11 {11 alkoxy carbonyl group at the 17{62 {0 position
US3981894A (en) * 1974-08-30 1976-09-21 Glaxo Laboratories Limited Chemical compounds
US3989686A (en) * 1972-06-15 1976-11-02 Glaxo Laboratories Limited Anaesthetic steroids of the androstane series and process for preparing same
US4093721A (en) * 1974-08-30 1978-06-06 Glaxo Laboratories Limited Pharmaceutical compositions of 6α,9α-difluoro-androst-4-ene-17β-carboxylates and derivatives thereof
US4187301A (en) * 1978-04-05 1980-02-05 Syntex (U.S.A.) Inc. 17 Beta-thiocarboxylic acid esters of 6 alpha, 6 beta-difluoro-3-oxoandrost-4-enes
US4188385A (en) * 1978-04-05 1980-02-12 Syntex (U.S.A.) Inc. Thioetianic acid derivatives
US4198403A (en) * 1978-04-05 1980-04-15 Syntex (U.S.A.) Inc. 17 Beta-thiocarboxylic acid esters of 4-halo-3-oxoandrost-4-enes
US4261984A (en) * 1978-04-05 1981-04-14 Syntex (U.S.A.) Inc. 17β-thiocarboxylic acid esters of 3-oxo-4-halo-16β-methylandrost-4-enes
US4263289A (en) * 1978-04-05 1981-04-21 Syntex (U.S.A.) Inc. Thio etianic acid derivatives
US4267173A (en) * 1979-11-05 1981-05-12 Schering Corporation Use of 6β-fluoro-7α-halogenocorticoids as topical anti-inflammatories and pharmaceutical formulations useful therefor
US4285937A (en) * 1976-02-24 1981-08-25 Ciba-Geigy Corporation Novel androstadiene-17-carboxylic acid esters
US4310466A (en) * 1979-08-31 1982-01-12 Syntex (U.S.A.) Inc. Thio etianic acid derivatives
US4335121A (en) * 1980-02-15 1982-06-15 Glaxo Group Limited Androstane carbothioates
US4377575A (en) * 1978-04-25 1983-03-22 Hoechst Aktiengesellschaft Corticoid-17-(alkyl carbonates) and process for their manufacture
US4472393A (en) * 1981-02-02 1984-09-18 Schering Corporation 3,20-Dioxo-1,4-pregnadiene-17α-ol 17-aromatic heterocycle carboxylates
US4607028A (en) * 1983-08-18 1986-08-19 Ciba-Geigy Corporation Novel carboxylic acid esters
US4710495A (en) * 1980-07-10 1987-12-01 Otsuka Pharmaceutical Co., Ltd. Soft steroids having anti-inflammatory activity
US4861765A (en) * 1985-06-26 1989-08-29 Jouveinal 21-alkyl-, cycloalkyl- or aryl-substituted thio steroids and pharmaceutical compositions containing them
US4992474A (en) * 1983-04-18 1991-02-12 Glaxo Group Ltd. Phenethanolamine derivatives
US4994439A (en) * 1989-01-19 1991-02-19 California Biotechnology Inc. Transmembrane formulations for drug administration
US4996335A (en) * 1980-07-10 1991-02-26 Nicholas S. Bodor Soft steroids having anti-inflammatory activity
US5250293A (en) * 1991-04-22 1993-10-05 Gleich Gerald J Method for the treatment of hypersensitivity diseases by administration of anionic polymers
US5420120A (en) * 1993-12-17 1995-05-30 Alcon Laboratories, Inc. Anti-inflammatory glucocorticoid compounds for topical ophthalmic use
US5658549A (en) * 1991-12-12 1997-08-19 Glaxo Group Limited Aerosol formulations containing propellant 134a and fluticasone propionate
US5707984A (en) * 1995-12-08 1998-01-13 G. D. Searle & Co. Steroid nitrite/nitrate ester derivatives useful as anti-inflammatory drugs
US5837699A (en) * 1994-01-27 1998-11-17 Schering Corporation Use of mometasone furoate for treating upper airway passage diseases
US5849265A (en) * 1994-09-28 1998-12-15 Glaxo Wellcome Inc. Pharmaceutical aerosol formulation comprising a medicament, a propellant and a fluorinated surfactant
US5919776A (en) * 1996-12-20 1999-07-06 Merck & Co., Inc. Substituted aminoquinolines as modulators of chemokine receptor activity
US5972920A (en) * 1998-02-12 1999-10-26 Dermalogix Partners, Inc. Formulation containing a carrier, active ingredient, and surfactant for treating skin disorders
US5981517A (en) * 1996-05-09 1999-11-09 Soft Drugs, Inc. Androstene derivatives
US6127353A (en) * 1991-09-06 2000-10-03 Schering Corporation Mometasone furoate monohydrate, process for making same and pharmaceutical compositions
US6136294A (en) * 1998-09-22 2000-10-24 Aeropharm Technology Inc. Amino acid stabilized medical aerosol formulation
US6197761B1 (en) * 1995-12-29 2001-03-06 Glaxo Wellcome Inc. 17β-2-oxo-tetrahydrofuranyl)-carbothioic acid ester, -carboxylic acid ester and -carboxylic acid amide androstane derivatives
US6261539B1 (en) * 1998-12-10 2001-07-17 Akwete Adjei Medicinal aerosol formulation
US6395300B1 (en) * 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6537983B1 (en) * 2001-04-07 2003-03-25 Smithkline Beecham Corporation Anti-inflammatory androstane derivatives

Family Cites Families (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR580494A (en) 1923-07-10 1924-11-07 Const Metalliques Schiltigheim Method of establishing ventilation nozzles or other pipes
GB1384372A (en) 1971-01-20 1975-02-19 Glaxo Lab Ltd Dereivatives of 17alpha-hydroxyandrost-4-ene-17beta-carboxylic acids
DE2336693A1 (en) 1973-07-19 1975-02-06 Nassheuer Ind Ofenbau Jean Radiant heating pipe with igniter and flame monitor - has electrode eccentrically outside gas feed pipe
DE2538569A1 (en) 1975-08-29 1977-03-03 Siemens Ag Metallising thermosetting plastics contg. reinforcing fibres - after preliminary etching with chromosulphuric acid
SE449106B (en) 1980-07-10 1987-04-06 Otsuka Pharma Co Ltd STEROID WITH ANTI-INFLAMMATORY EFFECT AND COMPOSITION CONTAINING THIS
EP0179583A1 (en) 1984-10-04 1986-04-30 Merck & Co. Inc. A system for enhancing the water dissolution rate and solubility of poorly soluble drugs
ZA872389B (en) 1987-04-02 1987-11-25 Advanced Polymer Systems Inc Composition and method for delivering a steroid active ingredient
IL95590A (en) 1989-09-08 1996-06-18 Glaxo Group Ltd Pharmaceutical compositions comprising salmeterol and fluticasone propionate
DE3931041C2 (en) 1989-09-16 2000-04-06 Boehringer Ingelheim Kg Esters of thienyl carboxylic acids with amino alcohols, their quaternization products, processes for their preparation and medicaments containing them
JP3087763B2 (en) 1990-11-30 2000-09-11 三井化学株式会社 Novel heterocyclic compound and pharmaceutical composition containing the same
GB9103764D0 (en) 1991-02-22 1991-04-10 Glaxo Group Ltd Compositions
TW247878B (en) 1991-07-02 1995-05-21 Takeda Pharm Industry Co Ltd
DE69413955T2 (en) 1993-03-17 1999-04-01 Minnesota Mining & Mfg AEROSOL COMPOSITION CONTAINING A DERIVATIVE DERIVATIVE FROM ESTER, AMID OR MERCAPTOESTER
DE4328819A1 (en) 1993-08-27 1995-03-02 Hoechst Ag Corticosteroid-17-alkylcarbonate-21/0 / -carboxylic acid and carbonic acid esters, process for their preparation and medicaments containing them
DE4333920A1 (en) 1993-10-05 1995-04-13 Hoechst Ag Corticoid-17,21-dicarboxylic acid esters and corticosteroid-17-carboxylic acid ester-21-carbonic acid esters, processes for their preparation and medicaments containing them
IL109656A (en) 1994-05-15 1998-02-22 Chemagis Ltd Process for the manufacture of androstane - 17 - carbothioates and androstane - 17 - carbothioates prepared thereby
GB9410222D0 (en) 1994-05-21 1994-07-06 Glaxo Wellcome Australia Ltd Medicaments
AR002009A1 (en) 1994-12-22 1998-01-07 Astra Ab PHARMACEUTICAL COMPOSITION, PROCEDURE FOR THE MANUFACTURE OF A PROLIPOSOMA POWDER AS USED IN SUCH COMPOSITION, PROCEDURE FOR LAMANUFACTURE OF SUCH COMPOSITION, USE OF SUCH PHARMACEUTICAL COMPOSITION IN THE MANUFACTURE OF A DISPOSAL MEDICINAL PRODUCT.
DE19528145A1 (en) 1995-08-01 1997-02-06 Boehringer Ingelheim Kg New drugs and their use
GB9521696D0 (en) 1995-10-23 1996-01-03 Bayer Ag Combination of LTD4 receptor antagonists with glucocorticosteriods
BR9709650A (en) 1996-06-04 1999-08-10 Procter & Gamble Nasal aerosol containing an intransal steroid and an antihistamine
GB9622173D0 (en) 1996-10-24 1996-12-18 Glaxo Group Ltd Particulate Products
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
ATE530180T1 (en) 1997-04-02 2011-11-15 Brigham & Womens Hospital METHOD FOR DETERMINING THE INDIVIDUAL RISK PROFILES OF ATHEROSCLEROTIC DISEASES
AU8439998A (en) 1997-06-30 1999-01-25 Glaxo Group Limited Compounds
SE9704186D0 (en) 1997-11-14 1997-11-14 Astra Ab New composition of matter
SE9704833D0 (en) 1997-12-22 1997-12-22 Astra Ab New formulation
GB9828721D0 (en) 1998-12-24 1999-02-17 Glaxo Group Ltd Novel apparatus and process
AU3411000A (en) 1999-03-24 2000-10-09 Glenayre Electronics, Inc Computation and quantization of voiced excitation pulse shapes in linear predictive coding of speech
ES2165768B1 (en) 1999-07-14 2003-04-01 Almirall Prodesfarma Sa NEW DERIVATIVES OF QUINUCLIDINE AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
WO2001015744A1 (en) 1999-08-31 2001-03-08 The Brigham And Women's Hospital, Inc. Systemic inflammatory markers as diagnostic tools in the prevention of atherosclerotic diseases
US7413536B1 (en) 1999-09-14 2008-08-19 Xenoport, Inc. Substrates and screening methods for transport proteins
US6596261B1 (en) 2000-01-25 2003-07-22 Aeropharm Technology Incorporated Method of administering a medicinal aerosol formulation
US20020133032A1 (en) 2000-02-25 2002-09-19 Jufang Barkalow Method for the preparation of fluticasone and related 17beta-carbothioic esters using a novel carbothioic acid synthesis and novel purification methods
GB0009591D0 (en) 2000-04-18 2000-06-07 Glaxo Group Ltd Medical combinations
GB0009606D0 (en) 2000-04-18 2000-06-07 Glaxo Group Ltd Therapeutic combinations
GB0009583D0 (en) 2000-04-18 2000-06-07 Glaxo Group Ltd Respiratory formulations
GB0009592D0 (en) 2000-04-18 2000-06-07 Glaxo Group Ltd Respiratory combinations
TW519651B (en) 2000-06-27 2003-02-01 Intel Corp Embedded security device within a nonvolatile memory device
GB0015876D0 (en) 2000-06-28 2000-08-23 Novartis Ag Organic compounds
GB0016040D0 (en) 2000-06-29 2000-08-23 Glaxo Group Ltd Novel process for preparing crystalline particles
GB0017988D0 (en) 2000-07-21 2000-09-13 Glaxo Group Ltd Novel process
JP2004504357A (en) 2000-07-26 2004-02-12 アルコン,インコーポレイテッド Pharmaceutical suspension compositions containing no polymeric suspending agent
US6777399B2 (en) * 2000-08-05 2004-08-17 Smithkline Beecham Corporation Anti-inflammatory androstane derivative compositions
AU2001276497B2 (en) 2000-08-05 2005-04-07 Glaxo Group Limited 17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
WO2002026723A1 (en) 2000-09-29 2002-04-04 Glaxo Group Limited Compounds useful in the treatment of inflammatory diseases
US20020132803A1 (en) 2001-01-05 2002-09-19 Mahendra Dedhiya Fluticasone suspension formulation, spray pattern method, and nasal spray apparatus
GB0103630D0 (en) 2001-02-14 2001-03-28 Glaxo Group Ltd Chemical compounds
DE60220887T2 (en) 2001-03-08 2008-02-28 Glaxo Group Ltd., Greenford AGONISTS OF BETA ADRENORE RECEPTORS
JP4143413B2 (en) 2001-03-22 2008-09-03 グラクソ グループ リミテッド Formanilide derivatives as β2-adrenergic receptor agonists
AU2002303425A1 (en) 2001-04-24 2002-11-05 Epigenesis Pharmaceuticals, Inc. Composition, formulations and kit for treatment of respiratory and lung disease with non-glucocorticoid steroids and/or ubiquinone and a bronchodilating agent

Patent Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3067197A (en) * 1961-04-26 1962-12-04 Pfizer & Co C 11-oxygenated 6alpha-fluoro-16-methylene-delta-pregnenes and derivatives
US3639434A (en) * 1967-02-02 1972-02-01 Boots Pure Drug Co Ltd 17-acyloxysteroids and their manufacture
US3828080A (en) * 1972-01-20 1974-08-06 Glaxo Lab Ltd Androstane-17beta-carboxylic acids and processes for the preparation thereof
US3989686A (en) * 1972-06-15 1976-11-02 Glaxo Laboratories Limited Anaesthetic steroids of the androstane series and process for preparing same
US3856828A (en) * 1972-07-19 1974-12-24 Glaxo Lab Ltd Anti-inflammatory steroids of the androstane series having a halo-substituted c{11 {14 c{11 {11 alkoxy carbonyl group at the 17{62 {0 position
US3981894A (en) * 1974-08-30 1976-09-21 Glaxo Laboratories Limited Chemical compounds
US4093721A (en) * 1974-08-30 1978-06-06 Glaxo Laboratories Limited Pharmaceutical compositions of 6α,9α-difluoro-androst-4-ene-17β-carboxylates and derivatives thereof
US4285937A (en) * 1976-02-24 1981-08-25 Ciba-Geigy Corporation Novel androstadiene-17-carboxylic acid esters
US4261984A (en) * 1978-04-05 1981-04-14 Syntex (U.S.A.) Inc. 17β-thiocarboxylic acid esters of 3-oxo-4-halo-16β-methylandrost-4-enes
US4187301A (en) * 1978-04-05 1980-02-05 Syntex (U.S.A.) Inc. 17 Beta-thiocarboxylic acid esters of 6 alpha, 6 beta-difluoro-3-oxoandrost-4-enes
US4263289A (en) * 1978-04-05 1981-04-21 Syntex (U.S.A.) Inc. Thio etianic acid derivatives
US4188385A (en) * 1978-04-05 1980-02-12 Syntex (U.S.A.) Inc. Thioetianic acid derivatives
US4198403A (en) * 1978-04-05 1980-04-15 Syntex (U.S.A.) Inc. 17 Beta-thiocarboxylic acid esters of 4-halo-3-oxoandrost-4-enes
US4377575A (en) * 1978-04-25 1983-03-22 Hoechst Aktiengesellschaft Corticoid-17-(alkyl carbonates) and process for their manufacture
US4310466A (en) * 1979-08-31 1982-01-12 Syntex (U.S.A.) Inc. Thio etianic acid derivatives
US4267173A (en) * 1979-11-05 1981-05-12 Schering Corporation Use of 6β-fluoro-7α-halogenocorticoids as topical anti-inflammatories and pharmaceutical formulations useful therefor
US4335121A (en) * 1980-02-15 1982-06-15 Glaxo Group Limited Androstane carbothioates
US4710495A (en) * 1980-07-10 1987-12-01 Otsuka Pharmaceutical Co., Ltd. Soft steroids having anti-inflammatory activity
US4996335A (en) * 1980-07-10 1991-02-26 Nicholas S. Bodor Soft steroids having anti-inflammatory activity
US4472393A (en) * 1981-02-02 1984-09-18 Schering Corporation 3,20-Dioxo-1,4-pregnadiene-17α-ol 17-aromatic heterocycle carboxylates
US4992474A (en) * 1983-04-18 1991-02-12 Glaxo Group Ltd. Phenethanolamine derivatives
US4607028A (en) * 1983-08-18 1986-08-19 Ciba-Geigy Corporation Novel carboxylic acid esters
US4861765A (en) * 1985-06-26 1989-08-29 Jouveinal 21-alkyl-, cycloalkyl- or aryl-substituted thio steroids and pharmaceutical compositions containing them
US4994439A (en) * 1989-01-19 1991-02-19 California Biotechnology Inc. Transmembrane formulations for drug administration
US5250293A (en) * 1991-04-22 1993-10-05 Gleich Gerald J Method for the treatment of hypersensitivity diseases by administration of anionic polymers
US6127353A (en) * 1991-09-06 2000-10-03 Schering Corporation Mometasone furoate monohydrate, process for making same and pharmaceutical compositions
US5658549A (en) * 1991-12-12 1997-08-19 Glaxo Group Limited Aerosol formulations containing propellant 134a and fluticasone propionate
US5420120A (en) * 1993-12-17 1995-05-30 Alcon Laboratories, Inc. Anti-inflammatory glucocorticoid compounds for topical ophthalmic use
US6057307A (en) * 1994-01-27 2000-05-02 Schering Corporation Use of mometasone furoate for treating airway passage and lung diseases
US5837699A (en) * 1994-01-27 1998-11-17 Schering Corporation Use of mometasone furoate for treating upper airway passage diseases
US5889015A (en) * 1994-01-27 1999-03-30 Schering Corporation Use of mometasone furoate for treating lower airway passage and lung diseases
US5849265A (en) * 1994-09-28 1998-12-15 Glaxo Wellcome Inc. Pharmaceutical aerosol formulation comprising a medicament, a propellant and a fluorinated surfactant
US5707984A (en) * 1995-12-08 1998-01-13 G. D. Searle & Co. Steroid nitrite/nitrate ester derivatives useful as anti-inflammatory drugs
US6197761B1 (en) * 1995-12-29 2001-03-06 Glaxo Wellcome Inc. 17β-2-oxo-tetrahydrofuranyl)-carbothioic acid ester, -carboxylic acid ester and -carboxylic acid amide androstane derivatives
US5981517A (en) * 1996-05-09 1999-11-09 Soft Drugs, Inc. Androstene derivatives
US5919776A (en) * 1996-12-20 1999-07-06 Merck & Co., Inc. Substituted aminoquinolines as modulators of chemokine receptor activity
US5972920A (en) * 1998-02-12 1999-10-26 Dermalogix Partners, Inc. Formulation containing a carrier, active ingredient, and surfactant for treating skin disorders
US6136294A (en) * 1998-09-22 2000-10-24 Aeropharm Technology Inc. Amino acid stabilized medical aerosol formulation
US6136294C1 (en) * 1998-09-22 2002-09-24 Aeropharm Technology Inc Amino acid stabilized medical aerosol formulation
US6261539B1 (en) * 1998-12-10 2001-07-17 Akwete Adjei Medicinal aerosol formulation
US6395300B1 (en) * 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6537983B1 (en) * 2001-04-07 2003-03-25 Smithkline Beecham Corporation Anti-inflammatory androstane derivatives

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050054622A1 (en) * 2001-10-20 2005-03-10 Keith Biggadike Novel anti-inflammatory androstane derivatives
US7405206B2 (en) * 2001-10-20 2008-07-29 Glaxo Group Limited Anti-inflammatory androstane derivatives
US9290698B2 (en) 2010-07-15 2016-03-22 Battelle Memorial Institute Biobased polyols for potential use as flame retardants in polyurethane and polyester applications

Also Published As

Publication number Publication date
AU2003266390A1 (en) 2004-04-30
WO2004024745A2 (en) 2004-03-25
AU2003266390A8 (en) 2004-04-30
EP1569951A2 (en) 2005-09-07
JP2006502172A (en) 2006-01-19
US6777400B2 (en) 2004-08-17
WO2004024745A3 (en) 2005-07-14
US20030144257A1 (en) 2003-07-31

Similar Documents

Publication Publication Date Title
US6777400B2 (en) Anti-inflammatory androstane derivative compositions
AU2001276497B2 (en) 17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
US6878698B2 (en) Anti-inflammatory androstane derivatives
US6858593B2 (en) Anti-inflammatory androstane derivative compositions
US6759398B2 (en) Anti-inflammatory androstane derivative
US6777399B2 (en) Anti-inflammatory androstane derivative compositions
AU2001276497A1 (en) 17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
AU2001275760A1 (en) 6.alpha., 9.alpha.-difluoro-17.alpha.-&#39;(2-furanylcarboxyl) oxy!-11.beta.-hydroxy-16.alpha.-methyl-3-oxo-androst-1,4,-diene-17-carbothioic acid S-fluoromethyl ester as an anti-inflammatory agent
US7132532B2 (en) Compounds useful in the manufacture of an anti-inflammatory androstane derivative
US20050152845A1 (en) Amorphous fluticasone 2-furoate, pharmaceutical compositions thereof and its conversion to the crystalline unsolvated form

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION