Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPLEXES OF 6.ALPHA. 9.ALPHA. - DIFLUORO - 17. ALPHA. - [ (2 - FURANYLCARBONYL) OXY] - 11.BETA. -HYDROXY - 16- .ALPHA. -METHYL - 3OXO- ANDROSTA- 1,4 -DIENE - 17 .BETA. -CARBOTHIOIC ACID S-FLUOROMETHYL ESTER
Document Type and Number:
WIPO Patent Application WO/2004/024745
Kind Code:
A2
Abstract:
There is provided a crystalline chemical complex comprising a compound of formula (I) in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in the crystalline complex is of space group P212121 having unit cell dimensions of about 7.6 ± 0.6 Å, 12.7 ± 0.7 Å, and 33 ± 3 Å when determined at 120K.

Inventors:
BIGGADIKE KEITH (GB)
COOTE STEVEN JOHN (GB)
CRAIG ANDREW SIMON (GB)
JACEWICZ VICTOR WITOLD (GB)
MILLAN MICHAEL JOHN (GB)
SEAGER JOHN FREDERICK (GB)
THEOPHILUS ANDREW LEWIS (GB)
Application Number:
PCT/EP2003/010259
Publication Date:
March 25, 2004
Filing Date:
September 09, 2003
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GLAXO GROUP LTD (GB)
BIGGADIKE KEITH (GB)
COOTE STEVEN JOHN (GB)
CRAIG ANDREW SIMON (GB)
JACEWICZ VICTOR WITOLD (GB)
MILLAN MICHAEL JOHN (GB)
SEAGER JOHN FREDERICK (GB)
THEOPHILUS ANDREW LEWIS (GB)
International Classes:
A61K31/58; A61K31/59; C07J17/00; C07J31/00; C07J75/00; (IPC1-7): C07J75/00
Domestic Patent References:
WO2002012266A12002-02-14
WO2002012265A12002-02-14
WO2003066656A12003-08-14
Foreign References:
US4335121A1982-06-15
Attorney, Agent or Firm:
Giddings, Peter John (Corporate Intellectual Property 980 Great West Roa, Brentford Middlesex TW8 9GS, GB)
Download PDF:
Claims:
Claims :
1. A crystalline chemical complex comprising a compound of formula (I) in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in that the crystalline complex is of space group P2i2i2i having unit cell dimensions of about 7.7 0.6 A, 13.7 0.7 A, and 37 3 A when determined at 120K.
2. A complex according to claim 1 wherein the unit cell dimensions are 7.7 0.4 A, 13.8 0.3 A, and 36.8 3 A when determined at 120K.
3. A complex according to claim 1 or claim 2 wherein the crystal 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).
4. A complex according to any one of claims 1 to 3 wherein the guest molecule preferably has a relative molecular weight in the range 16 to 150.
5. A complex according to claim 1 wherein the guest molecule is toluene.
6. A complex according to claim 1 wherein the guest molecule is mxylene.
7. A complex according to claim 1 wherein the guest molecule is oxylene.
8. A complex according to claim 1 wherein the guest molecule is fluorobenzene.
9. A complex according to claim 1 wherein the guest molecule is chlorobenzene.
10. A complex according to claim 1 wherein the guest molecule is ethylbenzene.
11. A complex according to claim 1 wherein the ratio of compound of formula (I) to guest molecule is 1: 50.25.
12. A pharmaceutical composition comprising a complex according to any one of the preceding claims together with a physiologically acceptable diluent or carrier.
13. 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 complex according to any one of claims 1 to 11.
14. A pharmaceutical composition comprising a composition according to claim 1 in combination with another therapeutical active agent.
15. A pharmaceutical composition according to claim 14 wherein the other therapeutically active ingredient is a long acting ß2adrenoreceptor agonist.
16. A process for preparing a complex according to any one of claims 1 to 11 which comprises (a) crystallising the complex from a solution containing a compound of formula (I) and the guest molecule ; or (b) contacting the compound of formula (I) or a complex according to any one of claims 1 to 11 in solid form with a liquid containing the guest molecule and obtaining the composition therefrom; or (c) contacting a compound of formula (I) or a composition according to any one of claims 1 to 11 in solid form with a vapour containing the guest molecule.
17. A complex according to any one of claims 1 to 11 for use in human or veterinary medicine, in the treatment of patients with inflammatory and/or allergic conditions.
18. The use of a complex according to any one of claims 1 to 11 for the manufacture of a medicament for the treatment of patents with an inflammatory and/or an allergic condition.
19. The use as claimed in claim 18 for the treatment onceperday.
Description:
Novel Crystalline Complexes The present invention relates to a novel complex 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 complex 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. For example, US Patent 4335121 discloses 6a, 9oc-Difluoro-1 7c-(1- oxopropoxy)-11 1 ß-hydroxy-1 6cc-methyl-3-oxo-androsta-1, 4-diene-1 7ß-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.

In our application PCT. GB. 01.03495 we have identified a novel glucocorticoid compound which substantially meets these objectives.

We have now identified a novel crystalline complex of the novel glucocorticoid compound.

Thus, according to one aspect of the invention, there is provided a crystalline chemical complex comprising a compound of formula (I)

in which the crystal lattice is stabilised by the presence of a guest molecule, characterised in that the crystalline complex is of space group P2i2i2i having unit cell dimensions of about 7.7 0.6 A, 13.7 0.7 A, and 37 3 A when determined at 120K (hereinafter"a complex of the invention").

In the complex 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).

The nature of the crystal lattice can be seen by reference to Figure 1 which shows the spacial arrangement of 4 molecules of steroid and 8 guests within a single unit cell for an example complex. Detail of the hydrogen bond interaction between the steroid molecules is shown in Figure 3. Figure 2 shows the conformation of the steroid molecule with 2 guest molecules present.

We have determined the X-ray powder diffraction (XRPD) profiles for a number of complexes according to the invention. These XRPD profiles are also apparently characteristic of the crystalline complexes according to the invention. In particular they exhibit one or more of the following 3 features when determined at ambient temperature (eg around 295K): (a) A peak in the range of around 4.3-5. 0.

(b) A peak in the range of around 6.6-6. 9.

(c) A peak in the range of around 11.5-11. 8.

Typically they exhibit 2 or more of the above 3 features, especially 3 of the above features.

The XRPD profiles of complexes 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): (a) Absence of a peak at around 7.3 (eg around 7.1-7. 5) which is associated with the profile of 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 complexes of compound of formula (I) ; (c) Absence of a peak at around 12.5 (eg around 12. 3-12. 7) which is associated with the profile of 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 complexes of compound of formula (I) ; (e) Absence of a peak at around 10.5-11. 1 which is associated with the profile of another class of complexes of compound of formula (I) ; (f) Absence of a peak at around 12.2-12. 6 which is associated with the profile of another class of complexes of compound of formula (I).

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.

The chemical name of the compound of formula (I) is 6a, 9a-Difluoro-17a-[(2- furanylcarbonyl)oxy]-11ß-hydroxy-16α-methyl-3-oxo-androsta -1,4-diene-17ß- carbothioic acid S-fluoromethyl ester.

The compound of formula (I) and complexes 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 complexes and compositions thereof are useful in the treatment of inflammatory and/or allergic disorders, especially in once-per-day therapy.

Space group P2i2i2 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.

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 x 105Pa). 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.: Aromatic compounds eg benzene and substituted derivatives, especially benzene substituted with one or more (eg 1 or 2) halogen or C 1-4alkyl groups, for example toluene, o-xylene, m-xylene, fluorobenzene, chlorobenzene, ethylbenzene.

Preferred guest molecules are pharmaceutical acceptable substances and, as described below, complexes of the invention containing them may be used in therapy. However even if the guest molecule is not pharmaceutically acceptable then such complexes may be useful in the preparation of other complexes containing a compound of formula (I), for example, other complexes of the invention containing guest molecules that are pharmaceutically acceptable or compound of formula (I) in unsolvated form.

The stoichiometry of the complex 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.

Unusually a complex 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 P21 (i. e. two of the axis angles are 90°) and cell dimensions of 7.6, 14.1, 11.8 A 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 complex 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 dimensions are about 7.7 0.4 A, 13.8 0.3 A, and 36.8 3 A when determined at 120K. Usually the unit cell dimensions are about 7.7 0.2 A, 13.8 0.2 A, and 36. 8 2.7 A when determined at 120K.

Table 1 shows the unit cell dimensions and peak positions for a number of example complexes : 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-

ND= not determined Compound (I) undergoes highly efficient hepatic metabolism to yield the 1 7-p carboxylic acid (X) as the sole major metabolite in rat and human in vitro systems. This metabolite has been synthesised and demonstrated to be >1000 fold less active than the parent compound in in vitro functional glucocorticoid assays.

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.

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.

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.

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.

We have also surprisingly found that the compound of Formula (I) shows a significantly longer mean absorption time (MAT) than fluticasone propionate, in man.

Measurements have shown that the compound of Formula (I) has a mean absorption time of an average of 12 hours while fluticasone propionate has a MAT of an average of 2.1 hours. Mathematical deconvolution of the plasma concentration time data shows that the time taken for 90% of lung dose to be absorbed is on average, 21 hours for compounds of Formula (I) compared with 11 hours for fluticasone propionate. It has been shown that oral bioavailability of the compound of Formula (I) is very low, and therefore systemic exposure is predominantly the result of absorption from the lung. This would suggest that the compound of Formula (I) has a significantly longer residence time in the lung than fluticasone propionate.

A longer residence time in the lung may lead to a longer duration of action and the lower daily systemic exposure may lead to an improved safety profile which, it is believed, allows 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.

In vitro human lung tissue affinity experiments indicate significantly higher affinity of the compound of formula (I) for human lung tissue.

This is supported also by cell transport and accumulation studies using human bronchial epithelial cells. These studies show reduced flux across the cell layer for compound of formula (I) compared with fluticasone propionate. The studies also show greater accumulation of the compound of formula (I) within the cell layer compared with fluticasone propionate.

Examples of disease states in which the compound of formula (I) and complexes 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.

The compound of formula (I) may also have use in the treatment of conjunctiva and conjunctivitis.

The complex 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.

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.

As mentioned above, the complex of the invention is useful in human or veterinary medicine, in particular as an anti-inflammatory and anti-allergic agent.

There is thus provided as a further aspect of the invention the complex 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.

According to another aspect of the invention, there is provided the use of the complex 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.

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 complex of the invention, especially for administration once-per-day.

The complex 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 complex 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.

Further, there is provided a process for the preparation of such pharmaceutical compositions which comprises mixing the ingredients.

The complex of the invention may, for example, be formulated for oral, buccal, sublingual, parenteral, local or rectal administration, especially local administration.

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.

Advantageously 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. Each capsule or cartridge may generally contain between 20wg-10mg of the compound of formula (I)

in a complex of the invention optionally in combination with another therapeutical active ingredient. Alternatively, the complex 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 complex 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.

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.

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 cosolvens e. g. ethanol. One example formulation is excipient free and consists essentially of (e. g. consists of) complex 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 complex 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 W094/21229. 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, um, preferably 2-51lm. Particles having a size above 20, um are generally too large when inhaled to reach the small airways. To achieve these particle sizes the particles of the complex 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/GB99/04368) 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).

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, um and not less than 15% will have a MMD of less than 15gm.

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.

Other possible presentations include the following : 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.

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.

If appropriate, the 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 1lig-2000, ug e. g. 20p, g-2000p, g, preferably about 2011g-5001lg of compound of formula (I) optionally in combination with another therapeutical 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 range 1011mg e. g. 100, ug-10mg preferably, 200pg-2000pg.

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.

For internal administration the complex 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 20mg preferably from 2.5 to 1 Omg of the compound of formula (I).

The complex according to the invention may in general may be given by internal administration in cases where systemic adreno-cortical therapy is indicated.

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 60mg, e. g. 5-30mg, 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.

Since the compound of formula (I) is long-acting, preferably the pharmaceutical 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.

The pharmaceutical compositions according to the invention may also be used in combination with another therapeutical active agent, for example, a ß2 adrenoreceptor agonist, an anti-histamine or an anti-allergic. The invention thus provides, in a further aspect, a combination comprising the complex of the invention together with another therapeutically active agent, for example, a (32-adrenoreceptor agonist, an anti-histamine or an anti-allergic.

Examples of ß2-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.

Long-acting ß2-adrenoreceptor agonist are preferred, especially those having a therapeutic effect over a 24 hour period.

Preferred long acting ß2-adrenoreceptor agonists include those described in WO 02066422, W002070490 and W002076933.

Especially preferred long-acting ß2-adrenoreceptor agonists include compounds of formula (X): or a salt or solvate thereof, wherein: m is an integer of from 2 to 8; n is an integer of from 3 to 11,

with the proviso that m + n is 5 to 19, R is-XSO2NR16R17 wherein X is-(CH2) p-or C26 alkenylene ; R'6 and R17 are independently selected from hydrogen, C1-6alkyl, C3-7cycloalkyl, C (O) NR18R19, phenyl, and phenyl (C-4alkyl)-, or R16 and R17, together with the nitrogen to which they are bonded, form a 5-, 6-, or 7-membered nitrogen containing ring, and R16 and R17 are each optionally substituted by one or two groups selected from halo, C1-6alkyl, C106haloalkyl, C1-6alkoxy, hydroxy-substituted C1-6alkoxy, -CO2R18, -SO2NR18R19, -CONR18R19, - NR18C (O) R'9, or a 5-, 6-or 7-membered heterocylic ring; R18 and R19 are independently selected from hydrogen, C1-6alkyl, C3-6cycloalkyl, phenhyl, and phenyl (C1-4alkyl)-: and p is an integer of from 0 to 6, preferably from 0 to 4; R12 and R13 are independently selected from hydrogen, Chalky), C10-6alkoxy, halo. phenyl, and C16haloalkyl ; and R14 and R15 are independently selected from hydrogen and Cl-4alkyl with the proviso that the total number of carbon atoms in R14 and R"5 is not more than 4.

Since the compound of formula (I) is long-acting, preferably the pharmaceutical composition comprising the compound of formula (I) and the long-acting ß2- 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.

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).

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.

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.

The complex 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 complex 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.

Further, there is provided a process for the preparation of such pharmaceutical compositions which comprises mixing the ingredients.

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).

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.

The complex of the invention may be prepared by the methodology described hereinafter, constituting a further aspect of this invention.

A first process for preparing a complex of the invention comprises crystallising the complex 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 in the process.

Optionally, for better control and reproduceability, the crystallisation process may be assisted by seeding with crystals of the complex 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 complex of the invention comprises contacting the compound of formula (I) or a complex according to the invention thereof in solid form with a liquid containing the guest molecule (for example by slurrying) and obtaining the complex 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 in the process.

A third process for preparing a complex of the invention comprises contacting a compound of formula (I) or a complex thereof according to the invention 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.

In the second and third processes, the compound of formula (I) may be employed in the form of a complex with a guest molecule or in a form without a guest molecule (eg as unsolvated polymorph Form 1,2 or 3). In the first process the compound of formula (I) or a complex 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 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 2-4% e. g. 3. 5-4%. 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 15-30ml/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, 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 Figure 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 Form 2: Peaks at around 18.4 and 21.5 degrees 2Theta 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.

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 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 methyl isobutyl ketone 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. 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 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, complexes 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 complexes of the invention, or in pharmaceutical compositions thereof.

For example, a process for the preparation of compound of formula (I) in unsolvated form (typically unsolvated polymorph Form 1) comprises removing the guest molecule from a complex of the invention.

A process for preparing a compound of formula (I) comprises alkylation of a thioacid of formula (II)

or a salt thereof.

In this process the compound of formula (II) may be reacted with a compound of formula FCH2L 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 HCI 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).

Compounds of formula (II) may be prepared from the corresponding 17a-hydroxyl derivative of formula (III) :

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.

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.

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.

Thus we provide a process for preparing a compound of formula (II) which comprises: (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)

; and (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.

In two particularly convenient embodiments of this process we also provide methods for the efficient purification of the end product which comprise either (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 (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.

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. In step (c2) the aqueous medium is for example a dilute aqueous acid such as dilute HCI.

We also provide an alternative process for preparing a compound of formula (II) which comprises:

(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 (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).

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.

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).

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.

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.

Compounds of formula (III) may be prepared in accordance with procedures described in GB 2088877B. Compounds of formula (III) may also be prepared by a process comprising the following steps: O SH CL, :"OH 11 OH "OH"OH CH,,, CH, Step (a) CH"I cH, Step (b) CH3"'CH3 lu - F o/ () o o _ (up F F'' F F F

Step (a) comprises oxidation of a solution containing the compound of formula (V).

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 10vol relative to the amount of the starting material (1wt.), 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 10wt. relative to the amount of the starting material (1wt.), more preferably between 1.1 and 3wt., especially 1.3wt.

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. 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 solvate 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 solvate solvent within the crystal lattice.

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.

The advantages of the complex 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 complexes thereof.

Brief Description of the Figures: Figure 1: Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a complex of the invention with toluene (guest molecule darkened).

Figure 2: Figure showing hydrogen bond interactions between steroid and guest for the complex of the invention with toluene Figure 3: Figure showing the spacial arrangement of 4 steroid and 8 guest molecules in the unit cell of a complex of the invention with toluene, and hydrogen bond interactions between two steroid molecules Figure 4: Enlarged XRPD profile of complex of the invention with toluene

Figure 5: Enlarged XRPD profile of complex of the invention with m-xylene Figure 6: Enlarged XRPD profile of complex of the invention with o-xylene Figure 7: Enlarged XRPD profile of complex of the invention with chlorobenzene Figure 8: XRPD profiles of unsolvated polymorphs 1,2 and 3 The following non-limiting Examples illustrate the invention: EXAMPLES General 1 H-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 x 4.6 mm ID) eluting with 0. 1% HCO2H and 0.01 M ammonium acetate in water (solvent A), and 0.05% HCOzH 5% water in acetonitrile (solvent B), using the following elution gradient 0-0.7 min 0% B, 0.7-4. 2 min 1 00% 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 XRPD analyses shown in the figures were performed on a) a Philips X'pert MPD powder diffractometer, serial number DY667. The pattern was recorded using the following acquisition conditions: Tube anode: Cu, Start angle : 2.0 °20, End angle : 45.0 °20, Step size: 0.02 °29, Time per step: 1 second.

XRPD profiles were collected at ambient temperature (295K) (Figure 8); b) a Philips PW1710 powder diffractometer. The pattern was recorded using the following acquisition conditions: Tube anode: Cu, Start angle : 3.5 °20, End angle : 35.0 °20, Step size: 0.02 °29, Time per step: 2.3 seconds. XRPD profiles were collected at ambient temperature (295K) (Figures 4-7); X-ray diffraction pattern collections referred to in Table 1 were performed in the following manners: The crystal and molecular structures and corresponding unit cell dimensions were determined from three-dimensional X-ray diffraction data collected at 120 +/-2 K. All measurements were made using a Bruker SMART CCD diffractometer with graphite

monochromated Mo-Ka radiation (R = 0.71073 A) 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 (Fo2-Fc2) 2. The Bruker SHELX software was used throughout.

Intermediates Intermediate 1: 6a, 9a-Difluoro-17a-f (2-furanylcarbonvl) oxyl-11ß-hvdroxy-16a- methyl-3-oxoandrosta-1, 4-diene-17ß-carbothioic acid diisopropylethylamine salt A stirred suspension of 6a, 9a-difluoro-11ß, 17a-dihydroxy-16a-methyl-3-oxo- androsta-1, 4-diene-17p-carbothioic acid (prepared in accordance with the procedure described in GB 2088877B) (49.5g) in methylacetate (500moi) is treated with triethylamine (35ml) maintaining a reaction temperature in the range 0-5°C. 2-Furoyl chloride (25ml) is added and the mixture stirred at 0-5°C for 1 hour. A solution of diethanolamine (52.8g) in methanol (50ml) is added and the mixture stirred at 0-5°C for at least 2 hours. Dilute hydrochloric acid (approx 1 M, 550mi) 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 (2x250ml). All of the organic phases are combined, washed sequentially with brine (5 x 250ml) and treated with di-isopropylethylamine (30ml). The reaction mixture is concentrated by distillation at atmospheric pressure to an approximate volume of 250moi and cooled to 25-30°C (crystallisation of the desired product normally occurs during distillation/subsequent cooling). Tertiary butyl methyl ether (TBME) (500ml) 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 (2x200ml) and dried under vacuum at approximately 40-50°C (75.3g, 98.7%). NMR (CDC) s) 5 : 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 50Hz), 4.45-4. 33 (1 H, 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]-11ß-hydroxy-16α - methvl-3-oxo-androsta-1s4-diene-17ß-carbothioic acid S-fluoromethyl ester Unsolvated Form 1 A mobile suspension of Intermediate 1 (12. 61g, 19. 8mmol) in ethyl acetate (230ml) and water (50ml) is treated with a phase transfer catalyst (benzyltributylammonium chloride, 10mol%) cooled to 3°C and treated with bromofluoromethane (1. 10ml,

19. 5mmol, 0.98 equivalents), washing in with prechilled (0°C) ethyl acetate (EtOAc) (20ml). The suspension is stirred overnight, allowing to warm to 17°C. The aqueous layer is separated and the organic phase is sequentially washed with 1 M HCI (50ml), 1 % w/v NaHCO3 solution (3x50ml) and water (2x50ml). The ethylacetate solution is distilled at atmospheric pressure until the distillate reaches a temperature of approximately 73°C at which point toluene (150mi) 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 (2x30ml) and the product oven dried under vacuum at 60°C to constant weight to yield the title compound (8.77g, 82%) LCMS retention time 3.66min, m/z 539 MH+, NMR 8 (CDCI3) includes 7.60 (1 H, m), 7.18-7. 11 (2H, m), 6.52 (1 H, dd, J 4. 2Hz), 6.46 (1H, s), 6.41 (1H, dd, J 10, 2Hz), 5.95 and 5.82 (2H dd, J 51,9Hz), 5.48 and 5.35 (1 H, 2m), 4.48 (1 H, m), 3.48 (1 H, m), 1.55 (3H, s), 1.16 (3H, s), 1.06 (3H, d, J 7Hz).

Intermediate 3: 6a. 9a-Difluoro-17a-r (2-furanylcarbonyl) oxyl-11B-hydroxy-16a- methyl-3-oxo-androsta-1, 4-diene-17 (3-carbothioic acid S-fluoromethyl ester. amorphous particles Intermediate 2 (30.04g) was dissolved in methylethylketone (850mi) 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, IL, USA). The other spray drying parameters were as follows : Temperature: 150 °C, outlet temperature 98 °C Solution flow rate: 30ml/min using Isco 260D syringe pump (Isco Inc, Lincoln, NE, USA) Atomisation Pressure: 2 Bar 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 6a, 9a-Difluoro-17a-[(2- furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo-androsta-1 4-diene-17ß carbothioic acid S-fluoromethyl ester. System yield was 61%

Intermediate 4 6cc, 9a-Difluoro-17a-f (2-furanyicarbonyl) oxy1-11 ß-hydroxy-16a- methyl-3-oxo-androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester Unsolvated Form 2 Pharmacological Activity In Vitro Pharmacological Activity 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.

For the experiments in this section, compound of formula (I) was used as unsolvated Form 1 (Intermediate 2) 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-KB 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, 10ng/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 EC50 values were estimated.

In this test the compound of formula (1) showed an EC5ovalue of <1nM.

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 NFKB or AP-1) through their direct interaction with GR.

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 NFKB when stimulated with TNFa. 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 1h 0. 5ng/ml TNFa (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 ! 50 values were determined.

Transactivation (GR) Transrepression (NFKB) 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 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.

For the experiments in this section, compound of formula (I) was used as 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 saline 30 min prior to ovalbumin challenge. Significant inhibition is achieved after a single dose of 30pg 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 thymus involution 3 daily IT doses of 100ug of compound (1) 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.

In vitro metabolism in rat and human hepatocytes 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-P 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, 10#M 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:- 17-p acid metabolite production Subject Cell density (amol/h) number (million cells/mL) Compound (I) Fluticasone 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.

Pharmacokinetics after intravenous (IV) and oral dosing in rats Compound (I) was dosed orally (0. 1mg/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).

Pharmacokinetics after intra-trachea ! dry powder dosing in the pig.

Anaesthetised pigs (2) were dosed intra-tracheally with a homogenous mixture of compound (I) (1 mg) and fluticasone propionate (1mg) as a dry powder blend in lactose (10% w/w). Serial blood samples were taken for up to 8h 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 20pg/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 15min 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.

Cmax and AUC (0-inf) values show markedly reduced systemic exposure to compound (I) compared to fluticasone propionate:- Cmax (pg/mL) AUC (0-inf) (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.

Examples Example 1: 6a, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11ß-hydroxy-16α -methyl-3- oxo-androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester Complex 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 complex as a white solid.

Stoichiometry of compound of formula (I) : guest = 1 : 3.1 from'H nmr (CDCl3).

Example 2: 6al 9a-Difluoro-17a-f (2-furanlcarbonyl) oxyl-1 3-hydroxy-16a-methyl-3- oxo-androsta-1. 4-diene-17ß-carbothioic acid S-fluoromethyl ester Complex 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 complex as a white solid.

Example 3 : 6cc, 9a-Difluoro-17a-r (2-furanylcarbonyl) oxyl-11 a-hydroxy-16a-methyl-3- oxo-androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester Complex with o-xvlene (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 complex as a white solid.

Stoichiometry of compound of formula (I) : guest = 1: 1.5 from'H nmr (CDCl3).

Example 4: 6a, 9a-Difluoro-17a-f (2-furanvlcarbonvl) oxyl-11 a-hydroxv-16a-methyl-3- oxo-androsta-1. 4-diene-17ß-carbothioic acid S-fluoromethyl ester Complex with o-xvlene (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 complex as a white solid.

Stoichiometry of compound of formula (I) : guest = 1 : 4 from'H nmr (CDCl3).

Example 5: 6cc, 9a-Difluoro-17a-f (2-furanvlcarbonyl) oxyl-11 a-hydroxy-16a-methyl-3- oxo-androsta-1. 4-diene-17i-carbothioic acid S-fluoromethyl ester Complex 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 complex as a white solid.

Further characterising data on complexes of the invention: Detailed XRPD profile peak information for various compositions of the invention is provided in Tables 2,3, 4 and 5.

The XRPD profiles of complexes of the invention are provided in Figures 4,5, 6 and 7.

We also claim complexes of the invention substantially by reference to their XRPD profiles as shown in the Figures and Tables.

Example A: Dry powder composition containing 6a, 9a-Difluoro-17a-f (2- furanvlcarbonyl) oxy]-11ß-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17ß- carbothioic acid S-fluoromethyl ester complex with toluene A dry powder formulation may be prepared as follows : 6a, 9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11ß-hydroxy-16α -methyl-3-oxo- androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester, composition with toluene prepared according to Example 1, MMD of 3p. m : 0.20mg milled lactose (wherein not greater than 85% of particles have a MMD of 60-90#m, and not less than 15% of particles have a MMD of less than 15m) : 12mg A pelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared.

Example B : Dry powder composition containing 6a, 9α-Difluoro-17α-[(2- furanylcarbonyl)oxyl]-11ß-hydroxy-16α-metyl-3-oxo-androsta -,4-diene-17ß- carbothioic acid S-fluoromethyl ester complex with toluene and a long acting ß= adrenoreceptor agonist A dry powder formulation may be prepared as follows : 6a, 9a-Difluoro-17a-[(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo- androsta-1, 4-diene-17p-carbothioic acid S-fluoromethyl ester composition with toluene prepared according to Example 1, MMD of 3, um : 0.20mg Long-acting p2-adrenoreceptor agonist (micronised to a MMD of 3, um) : 0.02mg milled lactose (wherein not greater than 85% of particles have a MMD of 60-90#m, and not less than 15% of particles have a MMD of less than 15Rm) : 12mg A pelable blister strip containing 60 blisters each filled with a formulation as just described may be prepared.

Example C: Aerosol formulation containing 6a, 9α-Difluoro-17α-[(2- furanylcarbonyl) oxyl-11 f3-ydroxy-16a-methyl-3-oxo-androsta-1, 4-diene-17a- carbothioic acid S-fluoromethyl ester complex with toluene prepared according to Example 1, MMD of zum : An aluminium canister may be filled with a formulation as follows :

6a, 9a-Difluoro-17a-[(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo- androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester complex with toluene prepared according to Example 1, MMD of 3#m : 250 pg 1,1, 1, 2-tetrafluoroethane : to 50p1 (amounts per actuation) in a total amount suitable for 120 actuations and the canister may be fitted with a metering valve adapted to dispense 50 pl per actuation.

Example D: Aerosol formulation containing 6a, 9α-Difluoro-17α-[(2- furanylcarbonyl)oxyl]-11ß-hydroxy-16α-methyl-3-oxo-androst a-1,4-diene-17ß- carbothioic acid S-fluoromethyl ester complex with toluene and a long acting ß2- adrenoreceptor agonist An aluminium canister may be filled with a formulation as follows : 6a, 9a-Difluoro-17a-[(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo- androsta-1, 4-diene-17ß-carbothioic acid S-fluoromethyl ester complex with toluene prepared according to Example 1, MMD of elm : 250 pg Long-acting ß2-adrenoreceptor agonist (micronised to a MMD of 3#m) : 25 pg 1,1, 1, 2-tetrafluoroethane : to 50p1 (amounts per actuation) in a total amount suitable for 120 actuations and the canister may be fitted with a metering valve adapted to dispense 50 pl per actuation.

Example E: Nasal formulation containing 6a, 9a-Difluoro-17a-f (2- furanvlcarbonyl) oxyl-11 3-hydroxy-16a-methyl-3-oxo-androsta-1, 4-diene-173- carbothioic acid S-fluoromethyl ester complex with toluene A formulation for intranasal delivery may be prepared as follows : 6a, 9a-Difluoro-17a-[(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo- androsta-1, 3-diene-17ß-carbothioic acid S-fluoromethyl ester complex with toluene prepared according to Example 1, MMD of cm : 10mg Polysorbate 20 0.8mg Sorbitan monolaurate 0.09mg Sodium dihydrogen phosphate dihydrate 94mg Dibasic sodium phosphate anhydrous 17.5mg Sodium chloride 48mg Demineralised water to 10ml

The formulation may be fitted into a spraypump capable of delivering a plurality of metered doses (alois).

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.

The patents and patent applications described in this application are herein incorporated by reference. Table 2: XRPD characteristic angles and relative intensities for 6a, 9a-Difluoro-17a- [(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo-androsta-1, 4-diene-17ß- carbothioic acid S-fluoromethyl ester, complex 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 Table 3: XRPD characteristic angles and relative intensities for 6a, 9a-Difluoro-17a- [(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo-androsta-1, 4-diene-17ß- carbothioic acid S-fluoromethyl ester, complex 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 Table 4: XRPD characteristic angles and relative intensities for 6a, 9a-Difluoro-17a- [(2-furanylcarbonyl) oxy]-11 ß-hydroxy-16a-methyl-3-oxo-androsta-1, 4-diene-17ß- carbothioic acid S-fluoromethyl ester, complex 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 Table 5: XRPD characteristic angles and relative intensities for 6a, 9a-Difluoro-17a- [(2-furanylcarbonyl)oxy]-11ß-hydroxy-16α-methyl-3-oxo-andr osta-,4-diene-17ß- carbothioic acid S-fluoromethyl ester, complex 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