WO2022036267A1 - Compositions and methods of treatment - Google Patents

Compositions and methods of treatment Download PDF

Info

Publication number
WO2022036267A1
WO2022036267A1 PCT/US2021/046017 US2021046017W WO2022036267A1 WO 2022036267 A1 WO2022036267 A1 WO 2022036267A1 US 2021046017 W US2021046017 W US 2021046017W WO 2022036267 A1 WO2022036267 A1 WO 2022036267A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutical composition
oral
release pharmaceutical
pharmaceutically acceptable
Prior art date
Application number
PCT/US2021/046017
Other languages
French (fr)
Inventor
Steven August SMITH
Barrett LEVESQUE
Derek Moe
Colin Edward Rowlings
Nathaniel Santos MARTINEZ
Original Assignee
Gb004, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gb004, Inc. filed Critical Gb004, Inc.
Publication of WO2022036267A1 publication Critical patent/WO2022036267A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5084Mixtures of one or more drugs in different galenical forms, at least one of which being granules, microcapsules or (coated) microparticles according to A61K9/16 or A61K9/50, e.g. for obtaining a specific release pattern or for combining different drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5026Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • the present invention relates to the combination of certain pharmaceutical compositions containing tert-butyI-4- ⁇ [l-(4-chlorobenzyl)-3-hydroxy-2-oxo-l,2-dihydropyridin- 4-yl]methyl] piperazine- 1 -carboxylate (hereinafter also referred to as “Compound 1”) or a pharmaceutically acceptable salt thereof for use in the treatment of inflammatory bowel diseases; certain oral modified release pharmaceutical compositions comprising an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof; and to certain dosage regimens for treating inflammatory bowel diseases.
  • Compound 1 tert-butyI-4- ⁇ [l-(4-chlorobenzyl)-3-hydroxy-2-oxo-l,2-dihydropyridin- 4-yl]methyl] piperazine- 1 -carboxylate
  • Compound 1 a pharmaceutically acceptable salt thereof for use in the treatment of inflammatory bowel diseases
  • IBD Inflammatory bowel disease
  • mucosal inflammation changes in metabolic activity and vascular tissue damage lead to a reduction in tissue-oxygen tension (hypoxia), in which healing processes such as angiogenesis, cell migration, and re-epithelialization occur.
  • hypoxia tissue-oxygen tension
  • HIF-la hypoxia inducible factor-1 alpha
  • HIF-la prolyl hydroxylase PBD
  • HIF-la prolyl hydroxylase PBD
  • Inhibition of HIF-la prolyl hydroxylase thus leads to increased stabilization of HIF-la resulting in a up regulation of HIF-la which in turn leads to a corresponding increased response to inflammation.
  • treatment with effective HIF-la prolyl hydroxylase inhibitors can increase the level of the body's cellular inflammatory response.
  • HIF-la prolyl hydroxylase inhibitors can increase the amount of epithelial cell healing over that which the body would normally provide.
  • Compound 1 is a potent inhibitor of PHD and is selective for PHD-2 over PHD-1, resulting in specific stabilization of HIF-la and reduced risk of off-target effects.
  • the potential benefit of Compound 1 in the treatment of IBD is based on inhibition of the degradation of hypoxia inducible factor- 1 alpha (HIF-la) and sustained hypoxia inducible factor- 1 (HIF-1)- driven gene expression.
  • Compound 1 is disclosed in International Patent Application WO 2011/057121 and has the following structure:
  • Compound 1 has a low solubility/dissolution rate. Furthermore, the solubility of Compound 1 is highly dependent upon pH. For example, Compound 1 is soluble at pH 1 but is practically insoluble above pH 3, whilst some solubility is regained at very high pH values (i.e. above 10). Compounds which have pH-dependent solubility, particularly basic compounds, may exhibit undesirable pharmacokinetic properties such as problems in their absorption, possibly producing low or variable bioavailability between patients and between doses.
  • a factor which can affect the absorption of an orally administered drug is the changing pH experienced by the drug as it passes through the GI tract.
  • a drug may be absorbed in a number of different sites along the GI tract following oral administration; for example, the cheek lining, stomach, duodenumjejunum, ileum and colon.
  • the pH may be different at each site of absorption with the pH significantly different from the stomach (pH 1- 3.5) to the small intestine (pH 4-8).
  • pH 1- 3.5 the solubility of a drug
  • the drug may precipitate from solution as it passes through the GI tract. This can result in variability in the extent and/or rate of absorption between doses and between patients, because the drug needs to be in solution to be absorbed.
  • the Applicants have identified a particular hydrogen chloride salt form of Compound 1 with improved solubility properties, however, the salt is liable to dissociation into it's free-base form during formulation processing and/or storage. Such conversion can impact solubility and dissolution rate but also has consequences for the chemical stability of the compound. Unusually, the agent contains a tert-butyl oxy carbonyl (Boc) group and this group is liable to chemical degradation in the presence of an acidic environment.
  • Boc tert-butyl oxy carbonyl
  • compositions surprisingly provide systemic levels and colonic tissue concentrations comparable to HP-PCD formulations containing the compound in solubilized form.
  • pharmaceutically acceptable salt of Compound 1 is efficiently released from the granule based immediate release tablet dosage form and absorbed across the gastro-intestinal tract to provide a rapid increase in plasma concentration.
  • the geometric mean maximum plasma concentration (Cmax) achieved is approximately 8.85 ng/ml and the time at which the peak plasma concentration is observed (Tmax) is in the range of approximately 0.5-1.0 hour.
  • Tmax the time at which the peak plasma concentration is observed
  • the plasma concentrations of Compound 1 falls to less than approximately 5% of the Cmax within 24 hours.
  • the area under the plasma concentration-time curve from time zero to the last quantifiable concentration time point (AUCiast) is approximately 16.9 ng hr/mL.
  • the immediate release composition provides colonic tissue exposure, simply increasing dose levels to further improve tissue concentration levels may not be desirable. Any dose increase requires careful consideration to ensure any unintended safety or tolerability related issues do not occur. For example, previous studies have demonstrated that high systemic levels of PHD inhibitors can potentially influence EPO and/or VEGF regulation.
  • the Applicants have also surprisingly found that certain delayed release compositions comprising Compound 1 or a pharmaceutically acceptable salt thereof provide targeted release and biopharmaceutical performance without requiring high systemic levels.
  • certain delayed release compositions achieve similar colonic tissue concentration profiles to the above-mentioned immediate release granule composition and to a HP-PCD solution formulation.
  • tissue concentration levels at the higher dose of 240mg were found to be broadly equivalent to those achieved at 120mg. Hence, increasing dose did not result in increased tissue concentrations.
  • glucuronide conjugate upon oral administration of Compound 1, for example from an immediate release composition, the compound is rapidly absorbed and metabolized in the liver yielding a glucuronide conjugate.
  • This conjugate is subsequently subjected to biliary excretion into the intestine and it is expected that certain levels of the glucuronide conjugate pass through the gut lumen where P-glucuronidase enzymes hydrolyze the glucuronide back into the parent compound.
  • the parent drug can be distributed to the intestinal tract tissues allowing absorption into the colonic tissue. It is believed that other conjugates such as glycoside conjugates are also formed and could contribute to the increase in colonic tissue levels.
  • the unique contribution of this enterohepatic recirculation from an immediate release pharmaceutical composition, along with the luminal drug levels provided by a delayed release composition allows particularly high colonic tissue exposure to be achieved with acceptable safety and tolerability.
  • the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • the present invention provides an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof.
  • a composition may be typically a matrix dosage form, or a multiparticulate dosage form. Suitable multiparticulate dosage forms include subunits such as mini-tablets, beads, pellets, and granules. Such multiparticulates may be packaged, for example, in a capsule; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet or pill by compression or other processes known in the art.
  • mini-tablets have been found to be particularly suitable for the oral modified release pharmaceutical compositions of the present invention.
  • the Applicants have found that surprisingly it has been possible to achieve much higher drug loadings (greater than 50% w/w) of Compound 1 in both the immediate release component and the delayed release component of mini-tablet formulations.
  • the present invention provides methods of treating diseases or conditions mediated alone, or in part, by PHD (such as inflammatory bowel disease), comprising the administration of the combination according to the first aspect, or the modified release composition according to the second aspect.
  • PHD such as inflammatory bowel disease
  • the patent or application file contains at least one drawing executed in color.
  • FIG. l is a graphical plot of Compound 1 solubility (logio ng/mL) vs pH at 22°C, as described in Example 1.
  • FIG. 2 is a Flow diagram for a representative Compound 1 • HC1 High Shear Granulation Manufacturing Procedure.
  • FIG. 3 is a Flow diagram for the manufacturing process for Compound 1 • HC1 IR
  • FIG. 4 is a Flow diagram for the manufacturing process for Compound 1 • HC1
  • FIG. 5 is a Comparison of dissolution rates for Compound 1 from 60mg IR and
  • FIG. 6 is a Flow diagram for the Compound 1 • HC1 High Shear Granulation
  • FIG. 7 is a graphical plot of Compound 1 mean plasma concentration, over time 0-16hr for the clinical trial described in example 11 (all subjects).
  • FIG. 8 is a graphical plot of Compound 1 mean plasma concentration, over time 0-16hr for the clinical trial described in example 11 (without vomiting subjects).
  • FIG. 9 is a graphical representation of the Study Scheme for the clinical trial described in example 12.
  • FIG. 10 is a graphical plot of Compound 1 mean plasma concentration, over time
  • FIG. 11 is a graphical plot of Compound 1 mean plasma concentration, over time
  • FIG. 12 shows the box plot of the proportions of HIF-la positive cells evaluated at baseline and Day 8.
  • FIG. 13 is a graphical representation of the Study Scheme for the clinical trial described in example 13.
  • FIG. 14 is a graphical representation of the Study Scheme for the clinical trial described in Example 14.
  • FIG. 15 is a graphical plot showing Compound 1 mean plasma concentrations following single (day 1) and multiple (day 7) dosing of: (top) 120mg solution, 120mg immediate release tablet and 120mg & 240 mg delayed release tablets in fasted subjects; and (bottom) 120 mg immediate release tablet in fasted subjects and 240 mg immediate release tablet in fed subjects; over time 0-24hr for the clinical trial described in Example 14.
  • FIG. 16 is a graphical plot showing Compound 1 glucuronide metabolite mean plasma concentrations following [left] single (day 1) and [right] multiple (day 7) dosing of: 120 mg solution (white circles), 120 mg IR tablet fasted (white squares), 120 mg DR tablet fasted (black circles), 240 mg IR tablet fed (white triangles) and 240 mg DR tablet fasted (black squares); over time 0-24hr for the clinical trial described in Example 14.
  • FIG. 17 is a graphical plot of median colon tissue concentrations (rectum - left; sigmoid - right) for the dosing cohorts in the clinical trial described in Example 14.
  • FIG. 18 is a plot of Compound 1 total colon concentrations for the dosing cohorts in the clinical trial described in Example 14.
  • FIG. 19 shows USP2 Dissolution rates of Compound 1 from Example 23 pellets: (A) seal-coated pellets in pH 7.4 phosphate buffer with 2% CTAB; and (B) 20% Eudragit® FS 30 D coated pellets in pH 1.2 aqueous media for 2 hr followed by pH 7.4 phosphate buffer with 2% CTAB.
  • FIG. 20 shows USP2 Dissolution rates of Compound 1 from Example 29 capsules containing 1 :1 IR/DR mini -tablets.
  • Immediate Release or ‘IR’ as used in the present application in its conventional sense refers to a dosage form that provides for release of a compound immediately after administration.
  • an immediate release formulation means a formulation in which the dissolution rate of the drug from the formulation is 85 % or more after 10 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of an appropriate test fluid (such as 0.01N hydrochloric acid) is used and the paddle rotation speed is 75 rpm.
  • an appropriate test fluid such as 0.01N hydrochloric acid
  • ‘Immediate Release’ or ‘IR’ as used in the present application refers to a formulation or composition which rapidly disintegrates and disperses to release a drug compound after oral administration to a subject. More conveniently, it refers to a formulation or composition for which the dissolution rate of the drug from the formulation is 85 % or more after 10 minutes (such as after 5 minutes) from the beginning a dissolution test, which is carried out in accordance with a USP 2 dissolution test (paddle method) under the conditions that 900 mL of 0.01N hydrochloric acid is used, the temperature is 37°C and the paddle rotation speed is 75 rpm.
  • immediate release refers to a formulation or composition which typically provides systemic (plasma) levels of active pharmaceutical agent shortly after oral dosing. More conveniently, it refers to a formulation or composition which provides a geometric mean maximum plasma concentration (Cmax) of the compound after oral dosing of at least 5 ng/ml, such as at least 10 ng/ml within at least 120 minutes, conveniently within at least 60 minutes or 30 minutes.
  • Cmax geometric mean maximum plasma concentration
  • ‘Delayed Release’ or ‘DR’ as used in the present application refers to a dosage form that provides for release of a compound after administration at a slower rate than that from an immediate release formulation, or release of the active compound starts at a later point in time compared with an immediate release composition (such as at 30 min or more later such as, e.g., 1 hour or more later or 2 hours or more later or 3 hours or more later than an immediate release composition).
  • an immediate release composition such as at 30 min or more later such as, e.g., 1 hour or more later or 2 hours or more later or 3 hours or more later than an immediate release composition.
  • ‘Delayed Release’ or ‘DR as used in the present application refers to a formulation or composition which, after oral administration to a subject, does not undergo disintegration and dissolution in the acidic environment of the stomach.
  • delayed release refers to a formulation or composition which typically undergoes less than 5% dissolution in acidic aqueous media. More conveniently, it refers to a formulation or composition in which the dissolution rate of Compound 1 or a pharmaceutically acceptable salt thereof from the formulation is 5 % or less (such as 2% or less) after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a USP 2 dissolution test (paddle method) under the conditions that 900 mL of 0.01N hydrochloric acid is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm.
  • compositions being administered concurrently (for example, simultaneously, essentially simultaneously or within the same treatment protocol), sequentially or separately. However, the compositions need not be administered simultaneously or essentially simultaneously.
  • the immediate release composition may be administered first, followed by the delayed release composition, or the delayed release composition may be administered first, followed by the immediate release composition.
  • the combination is dosed once daily, the compositions of the invention are dosed within the same 24 hour period, preferably within the same 12 hour, 6 hour, 3 hour, 2 hour or 1 hour period.
  • the combination is dosed twice daily, the compositions of the invention are dosed within the same 12 hour period, preferably within the same 6 hour, 3 hour, 2 hour or 1 hour period.
  • compositions of the invention are dosed within the same 1 hour period, preferably within the same 30 minute period, after the patient has a meal.
  • the term ‘combination’ as used herein means the compositions are administered simultaneously or essentially simultaneously, more conveniently, simultaneously.
  • Granulate' as used in the present application refers to a granular material, which is the result of the agglomeration of fine particles into larger granules.
  • the granules may be packaged, for example, in a capsule for administration; dosed as a suspension or slurry in a liquid; dosed in a sachet; formed into pellets or beads by processes known in the art; or they may be formed into a tablet (e.g. a caplet), mini-tablet, micro-tablet or pill by compression or other processes known in the art.
  • Granules may typically be sized in the range 0.1 to 2.0 mm. Conveniently, the granules may have an average particle size in the range 0.2 to 0.5 mm.
  • the granules are formed into a mini-tablet or micro-tablet, most conveniently mini-tablets.
  • Mini-tablets may typically be sized in the range 1 to 4 mm.
  • minitablets are less than 3 mm in diameter, such as between about 2 and 3 mm in diameter, between about 2.2 and 2.8 mm in diameter, between about 2.3 and 2.7 mm in diameter, between about 2.4 and 2.6 mm in diameter, or about 2.5 mm in diameter.
  • excipient is used herein to refer to an essentially pharmacologically inert, non-toxic substance, e.g. that has been approved for inclusion in pharmaceutical products.
  • excipients classes includes fillers and diluents, binders, disintegrants, surfactants, wetting agents, lubricants, preservatives, colorants, flavouring agents, sweeteners and coatings.
  • the term 'intimate association' refers to at least two components which are intimately mixed together.
  • Compound 1 has a propensity to self-agglomerate in aqueous media such as pH 6.8 buffer. It has been found that co-processing of Compound 1 or a pharmaceutically acceptable salt thereof and one or more excipients (such as a wetting agent, in some cases even at very low levels) results in these components being intimately associated with one another leading to improved dispersion, and it is postulated that surface-to-surface interactions between particles of the Compound 1 or a pharmaceutically acceptable salt thereof are reduced in the intimate mixture such that dispersal is improved on exposure to aqueous media.
  • excipients such as a wetting agent
  • Compound 1 is tert-butyl 4-((l-(4- chlorobenzyl)-3-hydroxy -2-oxo- 1 ,2-dihydropyridin-4-yl)methyl)piperazine- 1 -carboxylate, which has the following structure:
  • Compound 1 is shown as a "free base”.
  • a pharmaceutically acceptable salt of Compound 1 is used.
  • a HC1 salt of Compound 1 (in a 1 : 1 molar ratio) is used.
  • the HC1 salt of Compound 1 is depicted below:
  • the molecular weight of Compound 1 is 433.93.
  • the molecular weight of the HC1 salt of Compound 1 is 470.39.
  • 1.0g of Compound 1 • HC1 salt contains 0.92g Compound 1 free base.
  • 1.08g Compound 1 • HC1 salt contains 1.0g Compound 1 free base:
  • references to the amount of Compound 1 will be understood to refer to the amount of the parent compound (free base equivalent), even if the compound is present as a salt of Compound 1.
  • reference to 120 mg of Compound 1 or a salt thereof will be understood to refer to 120 mg of the free base, or a salt of Compound 1 with 120 mg of free base equivalent; in the context of the anhydrous mono-hydrochloride salt of Compound 1, 130 mg of the salt provides 120 mg of Compound 1 (free base equivalent); in the context of the monohydrate mono-hydrochloride salt of Compound 1, 135 mg of the salt provides 120 mg of Compound 1 (anhydrous free base equivalent).
  • the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • a pharmaceutically acceptable salt of Compound 1 is used in the oral immediate release pharmaceutical composition.
  • the oral immediate release pharmaceutical composition is an immediate release solid pharmaceutical composition for oral administration comprising a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 : and one or more pharmaceutically acceptable excipients.
  • compositions display improved disintegration and dissolution properties leading to good absorption and/or bioavailability upon dosing.
  • Certain immediate release solid compositions have been found to give comparable systemic exposure of Compound 1 after 7 days of oral dosing as observed with an equivalent dose formulated as a HP-PCD solution (see Example 15).
  • certain immediate release compositions have been shown to deliver higher colonic tissue levels of Compound 1 than were achieved via oral solution dosing (see Example 16).
  • Certain compositions also possess improved chemical stability, powder flow and compression properties.
  • the compound used in the granulate is a pharmaceutically acceptable salt of Compound 1.
  • Suitable pharmaceutically acceptable salts include acid-addition salts of the basic piperazine nitrogen in Compound 1 and also metal salts of the weakly acidic hydroxyl group in Compound 1.
  • Acid-addition salts include salts with inorganic or organic acids.
  • Inorganic acid salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid salts.
  • Organic acid salts include trifluoroacetic, acetic, formic, citric, maleic, succinic, lactic, glycolic, tartaric, methanesulfonic and -toluenesulfonic acid salts.
  • the pharmaceutically acceptable salt is an inorganic acid salt. More conveniently, the pharmaceutically acceptable salt is a hydrochloric acid salt.
  • Metal salts include alkali metal salts and alkali earth metal salts, such as sodium, potassium, calcium or magnesium salts.
  • the pharmaceutically acceptable salt is a calcium salt.
  • the pharmaceutically acceptable salt is a hydrochloride salt. In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt and the ratio of Compound 1 to HC1 is about 1 : 1.
  • the hydrochloride salt is a hydrate.
  • the hydrochloride salt is a monohydrate.
  • the hydrochloride salt is a crystalline monohydrate, which is characterized by X-Ray (Cu K radiation in transmission mode using 40 kV / 40 mA generator settings) diffraction peaks at 15.1, 17.4, 19.8, 20.0 and 20.6 ⁇ 0.2 degrees 29 (Form A).
  • the hydrochloride salt is a crystalline monohydrate, which is characterized by a melting point of 191-194 °C.
  • the hydrochloride salt is anhydrous.
  • the hydrochloride salt is a crystalline anhydrous compound, which is characterized by X-Ray (Cu K radiation in transmission mode using 40 kV / 40 mA generator settings) diffraction peaks at 9.0, 15.2, 16.8, 18.6 and 20.3 ⁇ 0.2 degrees 29 (Form B).
  • the hydrochloride salt is a crystalline anhydrous compound, which is characterized by a melting point of 195-198 °C.
  • the granulate comprises a mixture of the hydrochloride salt as a crystalline monohydrate and the hydrochloride salt as an amorphous compound.
  • Analysis of the physical form of Compound 1 in the granulate may be carried out by techniques known to the person of skill in the art, e.g. XRPD, FTIR, Raman, or solid-state NMR.
  • the granulate comprises a mixture of the Compound 1 hydrochloride salt as a crystalline monohydrate and the Compound 1 hydrochloride salt as an amorphous compound, wherein the amount of amorphous compound present is less than 10% by weight of the total amount of Compound 1 hydrochloride salt present in the granulate.
  • the amount of amorphous compound present is less than 9% (such as less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, or less than 1%) by weight of the total amount of Compound 1 hydrochloride salt present in the granulate.
  • the granulate comprises a mixture of the hydrochloride salt as a crystalline monohydrate (Form A) and the hydrochloride salt as a crystalline anhydrous (Form B) compound.
  • the one or more pharmaceutically acceptable excipients comprise a wetting agent. It is postulated that when a wetting agent is intimately mixed with the pharmaceutically acceptable salt of Compound 1 within a granulate composition, the disintegration of the composition and dispersal of the pharmaceutically acceptable salt of Compound 1 in aqueous media is promoted.
  • the pharmaceutically acceptable salt of Compound 1 is in intimate association with the wetting agent in the granulate.
  • the pharmaceutically acceptable salt of Compound 1 is in intimate association with the wetting agent and one or more additional pharmaceutically acceptable excipients (optionally selected from disintegrant, binder and diluent).
  • the wetting agent is typically a surfactant or an emulsifier.
  • the wetting agent is a non-ionic surfactant.
  • Non-ionic surfactants can be characterized according to their hydrophilic-lipophilic balance (HLB). HLB values are commonly used to define emulsifiers and/or surfactants and refer to the hydrophilic-lipophilic balance of the given compound. HLB values can be calculated according to the methods of Griffin [Griffin, J. Soc. Cosmetic Chem. (1949), 311-326; Griffin, J. Soc. Cosmetic Chem. (1954), 249-256] as follows:
  • HLB 20 x (MW-H/MW-T) wherein MW-H is the molecular weight of the hydrophilic portion of the compound and MW-T is the molecular weight of the total compound.
  • PEG-80 sorbitan monooleate sold as Tween® 80 or Polysorbate 80
  • HLB values for a selection of emulsifiers and commercially available excipients are listed in the table below.
  • Ionic surfactants generally have higher HLB values than non-ionic surfactants.
  • Sodium lauryl sulfate (SLS) for example, has a HLB value of 40.
  • the wetting agent is generally regarded as safe for oral administration to humans.
  • the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25.
  • the wetting agent has a hydrophilic-lipophilic balance (HLB) between 8 and 20.
  • the wetting agent has a hydrophilic-lipophilic balance (HLB) between 12 and 18, such as between 13 and 18, between 13 and 17, or about 14 to 16.
  • the wetting agent is water soluble.
  • the wetting agent has an aqueous solubility greater than 10 g/litre, such as greater than 10 g/litre, greater than 20 g/litre, greater than 50 g/litre, or greater than 75 g/litre.
  • the wetting agent is water soluble and has a hydrophilic- lipophilic balance (HLB) between 12 and 18 In an embodiment, the wetting agent has an aqueous solubility greater than 10 g/litre and a hydrophilic-lipophilic balance (HLB) between 12 and 18. In an embodiment, the wetting agent has an aqueous solubility greater than 10 g/litre and a hydrophilic-lipophilic balance (HLB) between 13 and 18, such as between 13 and 17, or between 14 and 16. In an embodiment, the wetting agent has an aqueous solubility greater than 50 g/litre and a hydrophilic-lipophilic balance (HLB) between 13 and 18, such as between 13 and 17, or between 14 and 16.
  • HLB hydrophilic- lipophilic balance
  • the wetting agent is a non-ionic wetting agent selected from polyol esters, polyoxyethylene esters and poloxamers.
  • the polyol esters are selected from one or more of glycol esters, glycerol esters and sorbitan derivatives.
  • sorbitan derivates comprise polysorbate esters (such as polysorbate 20, polysorbate 40, polysorbate 60 and polysorbate 80).
  • the wetting agent is PEG-80 sorbitan monooleate (polysorbate 80).
  • the wetting agent is present in the composition at about 0.1 to 5.0% w/w of the total weight of the composition. It has been found that the advantageous effects associated with the presence of the wetting agent (improved wet granulation and superior disintegration of the composition and dispersal of the pharmaceutically acceptable salt of Compound 1 in aqueous media) can be realised even at low levels of wetting agent. Furthermore, even when the drug loading is high (for example in mini-tablets), low levels of wetting agent have been found to be effective. In an embodiment, the wetting agent is present in the composition at about 0.5 to 2.5% w/w.
  • the wetting agent is present in the composition at about 0.5 to 1.5% w/w, such as about 0.75 to 1.25% w/w, or about 1.0% w/w. In a convenient embodiment, the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w.
  • the wetting agent is a polysorbate ester and the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w.
  • the wetting agent is polysorbate 80 and the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w.
  • acidic environments are capable of cleaving the Boc group of Compound 1. It may therefore be advantageous to avoid incorporating overly acidic excipients in the compositions of the present invention, or carefully selecting grades of excipients such that their acidity is within acceptable ranges. It may be desirable to avoid excipients, such as buffering agents, capable of generating acidic micro-environments (e.g. pH less than 3) within the granulate.
  • excipients such as buffering agents, capable of generating acidic micro-environments (e.g. pH less than 3) within the granulate.
  • the one or more pharmaceutically acceptable excipients are not acidic.
  • the one or more pharmaceutically acceptable excipients have a pH of greater than 3.5 when dissolved or slurried in water.
  • the one or more pharmaceutically acceptable excipients have a pH of greater than 6 when dissolved or slurried in water. In a convenient embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 6.5 when dissolved or slurried in water. In an embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
  • the excipient microcrystalline cellulose is a purified, partially depolymerised cellulose prepared by treating cellulose pulp with mineral acids.
  • the pH of different batches of MCC may vary between approximately 5.0 and 7.5 (when a 5 g sample of MCC is slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the supernatant is measured).
  • the one or more pharmaceutically acceptable excipients do not comprise microcrystalline cellulose.
  • the composition has a moisture level of less than 6% w/w, such as less than 5% w/w or less than 4% w/w. Conveniently, the composition has a moisture level of about 3.5% w/w.
  • the one or more pharmaceutically acceptable excipients comprise a binder.
  • a binder helps to hold the granulate mixture together.
  • the binder is a cellulose ether-based binder (such as hydroxypropyl cellulose or methyl cellulose).
  • the binder is selected from hydroxy propyl cellulose, hypromellose, povidone, starch, methylcellulose, gelatin, pregelatinized starch, and xanthan gum.
  • the binder is hydroxy propyl cellulose.
  • the one or more pharmaceutically acceptable excipients comprise a disintegrant.
  • a disintegrant increases water wicking into the granulate core and therefore facilitates disintegration of the solid composition.
  • the disintegrant is selected from crospovidone, croscarmellose, sodium starch glycolate and low substituted hydroxypropyl cellulose. In a more convenient embodiment, the disintegrant is crospovidone.
  • the one or more pharmaceutically acceptable excipients comprise a diluent.
  • the diluent is selected from lactose, pregelatinized starch, microcrystalline cellulose and silicified microcrystalline cellulose.
  • the one or more pharmaceutically acceptable excipients comprise microcrystalline cellulose and lactose within the granulate.
  • the w/w ratio of microcrystalline celluloselactose within the granulate is between 1 : 1 and 3 : 1 (such as about 2: 1).
  • the diluent is selected from lactose and pregelatinized starch, or mixtures of the two. In an embodiment, the diluent is selected from lactose monohydrate and pregelatinized starch, or mixtures of the two.
  • the one or more pharmaceutically acceptable excipients comprise lactose monohydrate and pregelatinized starch within the granulate. Conveniently, the w/w ratio of pregelatinized starch lactose within the granulate is between 1 :2 and 2: 1 (such as about 1: 1).
  • the immediate release composition according to the present invention comprises about 10-40% w/w of the pharmaceutically acceptable salt of Compound 1.
  • the composition comprises 15-30% w/w of the pharmaceutically acceptable salt of Compound 1. In one embodiment, the composition comprises 15-20% (such as 16-18%) w/w of the pharmaceutically acceptable salt of Compound 1. In another embodiment, the composition comprises 25-30% (such as 26-28%) w/w of the pharmaceutically acceptable salt of Compound 1.
  • the immediate release composition according to the present invention comprises a high drug loading.
  • the immediate release composition according to the present invention comprises about 40-70% w/w of the pharmaceutically acceptable salt of Compound 1. More conveniently, the composition comprises 50-70% w/w of the pharmaceutically acceptable salt of Compound 1. In one embodiment, the composition comprises 60-70% (such as 62-66%) w/w of the pharmaceutically acceptable salt of Compound 1.
  • the immediate release composition according to this embodiment is a mini-tablet composition.
  • the immediate release composition according to the present invention comprises 20 to 150 mg (such as 50 to 70 mg, about 60 mg, 110 to 130 mg, or about 120 mg) of Compound 1 or a pharmaceutically acceptable salt thereof.
  • the immediate release composition according to the present invention comprises 120 to 360 mg (such as 180 to 300 mg, 200 to 280 mg, 220 to 260 mg, or about 240 mg) of Compound 1 or a pharmaceutically acceptable salt thereof.
  • the immediate release composition according to the present invention comprises 20 to 360 mg (such as about 60 mg, about 120 mg, or about 240 mg) of Compound 1 or the hydrochloride salt thereof.
  • the immediate release composition comprises about 120 mg of Compound 1.
  • the immediate release composition comprises about 135 mg of the monohydrate hydrochloride salt of Compound 1.
  • the one or more pharmaceutically acceptable excipients do not comprise a buffering agent.
  • Buffering agents are weak acids or bases which are capable of buffering the pH micro-environment the composition is exposed to in vivo after oral dosing. Examples of buffering agents include citric acid, lactic acid, tartaric acid.
  • the granulate is composed of granules, which may typically be sized in the range 0.1 to 2.0 mm. Conveniently, the granules within a granulate have an average particle size in the range 0.2 to 0.5 mm. In an embodiment, the granulate comprises granules having an average particle size between 200 and 500 pm, such as between 250 and 400 pm.
  • the granulate comprises granules having a particle size distribution of 0-10% less than between 74 pm; 10-20% between 74 and 125 pm; 10-20% between 125 and 177 pm; 30-50% between 177 and 420 pm; 5-30% between 420 and 595 pm; and 0-25% between 595 and 841 pm.
  • the granulate comprises granules having a particle size distribution of 30-50% of granules having particle sizes between 177 and 420 pm; and/or less than 20% of granules having particle sizes between 125 and 177 pm. In an embodiment, the granulate comprises granules having a particle size distribution of 30-40% of granules having particle sizes between 177 and 420 pm; and/or 10-20% of granules having particle sizes between 125 and 177 pm.
  • the granulate compositions according to the present invention typically have higher bulk densities, and lower Carr’s Index and Hausner ratio values. These properties are indicative that the granulates have superior powder flow and compression properties.
  • the composition has one or more of the following properties:
  • the composition has one or more of the following properties:
  • the composition has one or more of the following properties:
  • the composition has one or more of the following properties: a bulk density of > 0.5 g/ml; a Carr’s Index of ⁇ 25%; and a Hausner Ratio of ⁇ 1.3.
  • the composition has one or more of the following properties:
  • compositions according to the present invention undergo rapid disintegration and dispersal in aqueous media (such as 0.01N hydrochloric acid or pH 6.8 buffer).
  • aqueous media such as 0.01N hydrochloric acid or pH 6.8 buffer.
  • the dissolution rate of the drug from the immediate release composition is 75 % or more after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • the dissolution rate of the drug from the immediate release composition is 80 % or more after 15 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • the immediate release composition undergoes substantially complete disintegration and dispersal in 0.01N hydrochloric acid in less than 5 minutes at 37 °C using USP disintegration apparatus. In an embodiment, the immediate release composition undergoes substantially complete disintegration and dispersal in pH 6.8 aqueous media in less than 5 minutes at 37 °C using USP disintegration apparatus. In an embodiment, the immediate release composition undergoes substantially complete disintegration and dispersal in pH 1-7 aqueous media in less than 5 minutes at 37 °C using USP disintegration apparatus.
  • the immediate release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 40 °C and 75% RH for 3 months. In an embodiment, the immediate release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 6 months. In an embodiment, the immediate release composition provides less than 2% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 3 months.
  • the immediate release solid pharmaceutical composition as described herein may be formulated as a tablet.
  • tablet comprises tablets of any size suitable for oral administration, including micro-tablets and mini-tablets (which would typically be in the size range 1 to 4 mm).
  • the immediate release solid pharmaceutical composition is a mini-tablet.
  • the composition may be encapsulated within a capsule, wherein the capsule is non-functional (i.e. it is readily soluble in aqueous media, so that the immediate release properties of the composition are not significantly affected).
  • the immediate release composition is formulated as a tablet and the tablet has an average hardness of greater than 7 kp, such as greater than 7.5 kp, greater than 8 kp, greater than 8.5 kp or greater than 9 kp.
  • the immediate release composition is formulated as a tablet and the tablet has an average hardness of 7-12 kp, such as 7.5-11 kp, 8-11 kp, or 8.5-10 kp.
  • the immediate release composition is formulated as mini-tablets and the minitablets have an average hardness of 1-3 kp, such as 1-2 kp, 1-1.5 kp, or about 1.3 kp.
  • the tablet comprises a granulate component and an extra-granulate component.
  • the granulate component comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients selected from a filler, a diluent, a binder and a disintegrant.
  • the extra-granulate component comprises one or more pharmaceutically acceptable excipients selected from a filler, a diluent, a disintegrant, a lubricant and a glidant.
  • Suitable excipients for the extra-granulate component will be readily chosen by one of skill in the art and suitable fillers/diluents and disintegrants are described above.
  • Lubricants may be added to solid compositions to reduce friction and sticking during tablet processing. Suitable lubricants comprise magnesium stearate, calcium stearate, hydrogenated vegetable oil, stearic acid, sodium stearyl fumarate, mineral oil, hydrogenated vegetable oil and polyethylene glycol.
  • the extra-granulate component comprises magnesium stearate.
  • the composition comprises 0.25-1.5 % w/w of lubricant (such as magnesium stearate).
  • the extra-granulate component comprises magnesium stearate and hydrogenated vegetable oil .
  • the extra-granulate component comprises magnesium stearate and sodium stearyl fumarate.
  • the composition comprises 4-6 % w/w of one or more (e.g. two) lubricants (such as magnesium stearate and hydrogenated vegetable oil, or magnesium stearate and sodium stearyl fumarate).
  • the immediate release composition is formulated as mini-tablets and the mini-tablets comprise greater than 2.5 % w/w (such as greater than 3.5 % w/w, or greater than 4.5 % w/w) of one or more lubricants.
  • the immediate release composition is formulated as mini-tablets and the minitablets comprise between about 2.5 % and 7.5% w/w (such as between about 3.5 % and 6.5% w/w, or between about 4.5 % and 5.5% w/w) of one or more lubricants.
  • the Applicants have found that when the composition is formulated as mini-tablets, a combination of two lubricants at a total level of about 3 % to 7 % w/w provide significant beneficial effects, i.e. with respect to preventing the composition from sticking to the compression punches during tabletting and improving processability at scale.
  • the mini-tablets comprise a combination of magnesium stearate and a second lubricant at a total level of about 3 % to 7 % w/w.
  • the mini -tablets comprise a combination of magnesium stearate and hydrogenated vegetable oil at a total level of about 3 % to 7 % w/w.
  • the immediate release composition is formulated as mini-tablets and the minitablets comprise between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.0% w/w, or about 2.5 %) of magnesium stearate and between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.5% w/w, or about 2.75 %) of hydrogenated vegetable oil.
  • the mini-tablets comprise a combination of magnesium stearate and sodium stearyl fumarate at a total level of about 3 % to 7 % w/w.
  • the immediate release composition is formulated as mini-tablets and the mini-tablets comprise between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.0% w/w, or about 2.5 %) of magnesium stearate and between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.5% w/w, or about 2.75 %) of sodium stearyl fumarate.
  • Glidants may be added to solid compositions to improve flow properties during processing. Suitable glidants comprise colloidal silicon dioxide, ascorbyl palmitate, calcium palmitate, starch and talc.
  • the extra-granulate component comprises colloidal silicon dioxide.
  • the composition comprises 0.1-0.5 % w/w of glidant (such as colloidal silicon dioxide).
  • the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
  • the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
  • the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
  • the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
  • the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
  • the tablet comprises in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
  • the tablet comprises in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
  • the tablet comprises in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
  • the tablet comprises in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component: • 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
  • the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • composition does not comprise microcrystalline cellulose.
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component: • 60-70 % w/w of a pharmaceutically acceptable salt of Compound 1;
  • mini-tablets comprise in the extra-granulate component:
  • the immediate release composition is substantially encapsulated in a water-soluble coating.
  • the coating may be a capsule encapsulating the granulate, or it may be a coating substantially encapsulating the composition formulated as a tablet.
  • the composition is formulated as a tablet and the tablet is substantially encapsulated in a water- soluble (pH-independent - e.g. soluble in aqueous media at pH 1-8) coating.
  • the water-soluble coating is also a UV- resistant coating.
  • the coating is PVA-based.
  • the composition is formulated as mini-tablets and each mini-tablet is substantially encapsulated in a water-soluble (pH-independent - e.g. soluble in aqueous media at pH 1-8) coating.
  • the immediate release composition is prepared by a wet granulation method.
  • the immediate release composition is obtainable by wet granulation.
  • Wet granulation can be carried out by any known wet granulation process, including high-shear wet granulation and fluid bed granulation (see for example, Remington: The Science and Practice of Pharmacy, Edition, 22nd Edition, 2012). Conveniently, the wet granulation is carried out with high shear mixing.
  • the wet granulation is carried out by fluid bed granulation.
  • the immediate release solid pharmaceutical composition is formulated as mini-tablets and the wet granulation is carried out by fluid bed granulation.
  • the disintegrant, filler and pharmaceutically acceptable salt of Compound 1 are blended and then wet granulated in a fluid bed granulator in the presence of an aqueous solution of binder and wetting agent. After drying and optional milling, the granulate is then further blended with the extra-granular components (such as disintegrant, lubricant(s), optional filler and optional glidant). It has been found that fluid bed granulation offers advantages over other forms of granulation in terms of improved scalability of processing for compositions of the invention.
  • the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • the oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof is a delayed-release solid pharmaceutical composition for oral administration comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
  • compositions comprising Compound 1 or a pharmaceutically acceptable salt thereof provides benefits in respect to targeted release of Compound 1 and biopharmaceutical performance.
  • certain delayed release solid compositions of the present invention have been found to achieve colonic tissue concentration levels of Compound 1, that are at least comparable to those achieved via oral dosing of a pharmaceutically acceptable salt of Compound 1 as either an immediate release solution, or an immediate release solid composition, without the need for any appreciable systemic exposure.
  • Such compositions offer a number of advantages in respect to treatment of IBD, such as potentially minimising systemic side effects, e.g. those linked to elevated erythropoietin (EPO) and vascular endothelial growth factor (VEGF) levels.
  • EPO erythropoietin
  • VEGF vascular endothelial growth factor
  • the delayed-release solid pharmaceutical composition according to the present invention can be any composition suitable for oral administration, such as a tablet, a capsule, or granules or pellets, for example delivered in a sachet or capsule.
  • a delayed-release solid pharmaceutical composition for oral administration comprising Compound 1 (i.e. the free base) and one or more pharmaceutically acceptable excipients.
  • a delayed- release solid pharmaceutical composition for oral administration comprising a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients.
  • Suitable pharmaceutically acceptable salts include acid-addition salts of the basic piperazine nitrogen in Compound 1 and also metal salts of the weakly acidic hydroxyl group in Compound 1.
  • Acid-addition salts include salts with inorganic or organic acids.
  • Inorganic acid salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid salts.
  • Organic acid salts include trifluoroacetic, acetic, formic, citric, maleic, succinic, lactic, glycolic, tartaric, methanesulfonic and -toluenesulfonic acid salts.
  • the pharmaceutically acceptable salt is an inorganic acid salt. More conveniently, the pharmaceutically acceptable salt is a hydrochloric acid salt.
  • Metal salts include alkali metal salts and alkali earth metal salts, such as sodium, potassium, calcium or magnesium salts.
  • the pharmaceutically acceptable salt is a calcium salt.
  • the pharmaceutically acceptable salt is a hydrochloride salt. In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt and the ratio of Compound 1 to HC1 is about 1 : 1.
  • the hydrochloride salt is a hydrate.
  • the hydrochloride salt is a monohydrate.
  • the hydrochloride salt is a crystalline monohydrate, which is characterized by X-Ray diffraction peaks at 15.1, 17.4, 19.8 and 20.0 ⁇ 0.2 degrees 29 (Form A).
  • the hydrochloride salt is a crystalline monohydrate, which is characterized by a melting point of 191- 194 °C.
  • the hydrochloride salt is anhydrous.
  • the hydrochloride salt is a crystalline anhydrous compound, which is characterized by X-Ray diffraction peaks at 9.0, 16.8 and 18.6 ⁇ 0.2 degrees 29 (Form B).
  • the hydrochloride salt is a crystalline anhydrous compound, which is characterized by a melting point of 195-198 °C.
  • the delayed-release solid pharmaceutical composition is an erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix.
  • erodible matrix is meant aqueous-erodible or water-swellable or aqueous soluble, in the sense of being either erodible or swellable or dissolvable in pure water or requiring the presence of an acid or base to ionize the polymeric matrix sufficiently to cause erosion or dissolution.
  • the erodible matrix When contacted with an aqueous environment, the erodible matrix imbibes water and forms an aqueous-swollen gel or “ matrix ” that Compound 1 or a pharmaceutically acceptable salt thereof can pass or diffuse through depending on its physicochemical properties.
  • Suitable polymers for the erodible matrix are hydrogels such as synthetic polymers derived from vinyl, acrylate, methacrylate, urethane, ester and oxide monomers; or derivatives of naturally occurring polymers such as polysaccharides or proteins (including polysaccharides, gums, starches, alginates, collagen and cellulosics).
  • the delayed-release solid pharmaceutical composition is a capsule comprising granules or pellets encapsulated within a delayed release capsule.
  • the delayed-release solid pharmaceutical composition is a readily-soluble capsule comprising granules or pellets, wherein the individual granules or pellets are coated with a delayed release coating.
  • the delayed-release solid pharmaceutical composition comprises a core and a delayed release coating substantially encapsulating the core.
  • the delayed-release solid pharmaceutical composition is a tablet comprising a solid core and a delayed release coating substantially encapsulating the solid core.
  • tablet comprises tablets of any size suitable for oral administration, including micro-tablets and mini-tablets (which would typically be in the size range 1 to 4 mm).
  • the delayed- release solid pharmaceutical composition comprises mini-tablets and each mini-tablet comprises a solid core and a delayed release coating substantially encapsulating the solid core.
  • ‘substantially’ means that the coating covers the majority of the surface of the core, such as greater than 75%, greater than 85%, or preferably greater than 95% of the surface of the core.
  • the delayed release coating fully encapsulates the core.
  • the delayed release coating dissolves at pH values greater than about 5.5. In an embodiment, the delayed release coating dissolves at pH values greater than about 6.0. In an embodiment, the delayed release coating dissolves at pH values greater than about 7.0. Conveniently, the delayed release coating dissolves at about pH 5.5.
  • the delayed release coating comprises methyl acrylatemethacrylic acid copolymer, ethyl acrylate-methacrylic acid copolymer, hydroxy propyl methyl cellulose acetate succinate or cellulose acetate phthalate.
  • the delayed release coating comprises methyl acrylate-methacrylic acid copolymer or ethyl acrylatemethacrylic acid copolymer.
  • the delayed release coating is selected from Eudragit® L 100-55, Eudragit® FS30D, Eudragit® L100, Eudragit® L 12,5, Eudragit® L30 D- 55, Eudragit® S100 and Eudragit® S12,5, such as Eudragit® L 100-55.
  • the delayed-release composition comprises a greater than 6% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a greater than 8% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a greater than 10% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 12% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 14% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 10% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a 6 to 14% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a 7 to 13% (such as a 8 to 12%, or 9 to 11%) weight gain delayed release coating.
  • the delayed-release composition further comprises an additional sub-coating beneath the delayed release coating.
  • the sub-coating is soluble in aqueous media independent of the pH of the media.
  • the sub-coating is water-soluble at between pH 1 and 8.
  • the sub-coating is PVA-based.
  • the delayed-release composition comprises 2-6%, such as 3-5%, or about 4% weight gain of sub-coating.
  • the delayed-release composition comprises about 5% weight gain of sub-coating.
  • the delayed-release composition comprises a core and the core comprises a granulate.
  • the granulate may conveniently be a granulate as described above according to the first aspect of the invention. Therefore, all the embodiments described above for the first aspect (in relation to Compound 1 or a pharmaceutically acceptable salt thereof, the one or more pharmaceutically acceptable excipients, levels of the excipients, granule particle size distribution, bulk density, Carr’s index and Hausner ratio of the composition, the extra-granulate component and tablet hardness) all apply equally to a delayed-release composition core comprising a granulate according to the third aspect of the invention.
  • the delayed release composition according to the present invention comprises 20 to 150 mg (such as 50 to 70 mg, about 60 mg, 110 to 130 mg, or about 120 mg) of Compound 1 or a pharmaceutically acceptable salt thereof.
  • the delayed release composition according to the present invention comprises 120 to 360 mg (such as 180 to 300 mg, 200 to 280 mg, 220 to 260 mg, or about 240 mg) of Compound 1 or a pharmaceutically acceptable salt thereof.
  • the delayed release composition according to the present invention comprises 20 to 360 mg (such as about 60 mg, about 120 mg, or about 240 mg) of Compound 1 or the hydrochloride salt thereof.
  • the delayed-release composition is a multiparticulate composition comprising delayed release pellets, mini-tablets or beads.
  • the pellets or mini-tablets may be formed from a granulate as described hereinabove, wherein the individual pellets or mini-tablets are coated with a delayed release coating as described herein, or alternatively the pellets are encapsulated within a delayed release capsule.
  • the delayed release capsule may conveniently have the same properties as the delayed release coatings described herein.
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • composition does not comprise microcrystalline cellulose.
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
  • mini-tablets comprise in the extra-granulate component:
  • the delayed release coating dissolves at pH values greater than about 5.5, such as greater than about 6.0.
  • each mini-tablet comprises a greater than 6% weight gain (such as a greater than 8% weight gain) delayed release coating.
  • each pellet, or subunit, of the delayed release multiparticulate is an inert core coated with a drug layer comprising Compound 1 or a pharmaceutically acceptable salt thereof and the drug layer-coated core is then coated with a delayed release coating as described herein.
  • multiparticulates in the form of beads or pellets may be prepared by building Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g.
  • a binder dispersion for example HPMC
  • the seed core can be comprised of a sugar (for example a non-pareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose.
  • the inert core comprises sugar spheres mesh 45/60 (250-355 microns).
  • An example of a suitable composition and method is to spray a dispersion of Compound 1 / binder (e.g. HPMC) composition in water on to the seed core.
  • a delayed release coating is then employed to fabricate the membrane, which is applied over Compound 1 or a pharmaceutically acceptable salt thereof layered seed cores.
  • the DR pellets formed by this layering process on a seed core comprise the free base of Compound 1.
  • the delayed-release compositions do not undergo any significant disintegration or dispersal in the acidic environment of the stomach after oral administration. Therefore, in an embodiment, the delayed-release composition undergoes less than 5% dissolution in 0.01N HC1 after 30 mins at 37 °C using USP2 apparatus.
  • the dissolution rate of the drug from the delayed release composition is 10 % or less after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • the dissolution rate of the drug from the immediate release composition is 5% or less after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
  • the delayed release composition according to the present invention takes up only relatively small amounts of acid.
  • the delayed release composition increases in weight by less than 5% (such as less than 4% or less than 3%) after stirring for 2 hr in 0. IN hydrochloric acid in USP disintegration apparatus.
  • the delayed release coating may start to dissolve and the composition will undergo disintegration and dispersal.
  • the delayed release composition undergoes substantially complete disintegration in pH 6.8 aqueous media in less than 30 minutes (such as less than 15 minutes) at 37 °C using USP disintegration apparatus. In an embodiment, the delayed release composition undergoes substantially complete disintegration and dispersal in pH 6.8 aqueous media in less than 30 minutes (such as less than 15 minutes) at 37 °C using USP disintegration apparatus.
  • the delayed release composition undergoes greater than 75% dissolution within 240 minutes from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method) under the conditions that 900 mL of pH 6.8 with 2% CTAB buffer is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm.
  • the delayed release composition undergoes greater than 85% dissolution within 2400 minutes from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method) under the conditions that 900 mL of pH 6.8 with 2% CTAB buffer is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm.
  • the delayed release composition undergoes greater than 75% dissolution within 240 minutes (such as within 150 minutes, or within 90 minutes) from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method; temperature is 37 °C; paddle rotation speed is 75 rpm) under the conditions that 300 mL of pH 6.8 buffer is used for 30 minutes, followed by 900 mL of pH 6.8 buffer with 2% CTAB.
  • the delayed release composition undergoes greater than 75% dissolution within 240 minutes (such as within 150 minutes) from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method; temperature is 37 °C; paddle rotation speed is 75 rpm) under the conditions that 900 mL of 0.01N hydrochloric acid is used for 30 minutes, followed by 300 mL of pH 6.8 buffer for 30 minutes, followed by 900 mL of pH 6.8 buffer with 2% CTAB.
  • the chemical stability of Compound 1 or a pharmaceutical salt thereof in the solid compositions of the present invention has been found to be very good even after several months under accelerated storage conditions.
  • the delayed release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 40 °C and 75% RH for 3 months. In an embodiment, the delayed release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 6 months. In an embodiment, the delayed release composition provides less than 2% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 3 months. In an embodiment, the delayed release composition provides less than 0.5% (such as less than 0.25%) of a related impurity having a relative retention time (RRT) of 0.43-0.44 by HPLC (compared to the retention time of Compound 1) when stored at 40 °C and 75% RH for 6 months.
  • RRT relative retention time
  • the delayed-release composition comprises a granulate and the granulate is prepared by a wet granulation method.
  • the delayed-release composition comprises a granulate which is obtainable by wet granulation.
  • Wet granulation can be carried out by any known wet granulation process, including high-shear wet granulation and fluid bed granulation (see for example, Remington: The Science and Practice of Pharmacy, Edition, 22nd Edition, 2012). Conveniently, the wet granulation is carried out with high shear mixing.
  • the wet granulation is carried out by fluid bed granulation.
  • the delayed release solid pharmaceutical composition is formulated as mini-tablets and the wet granulation is carried out by fluid bed granulation.
  • the disintegrant, filler and pharmaceutically acceptable salt of Compound 1 are blended and then wet granulated in a fluid bed granulator in the presence of an aqueous solution of binder and wetting agent. After drying and optional milling, the granulate is then further blended with the extra-granular components (such as disintegrant, lubricant(s), optional filler and optional glidant).
  • Fluid bed granulation may offer advantages over other forms of granulation in terms of improved scalability of the processing.
  • the present invention provides an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a pharmaceutically acceptable salt thereof.
  • the oral modified release pharmaceutical compositions according to the present invention comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a pharmaceutically acceptable salt thereof.
  • the modified release composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a hydrochloride salt of Compound 1.
  • the immediate release component comprises a pharmaceutically acceptable salt of Compound 1.
  • the immediate release component comprises a hydrochloride salt of Compound 1.
  • the delayed release component comprises the hydrochloride salt of Compound 1. In an embodiment, the delayed release component comprises the free base of Compound 1.
  • the modified release composition comprises an immediate release component comprising the hydrochloride salt of Compound 1 and a delayed release component comprising the free base of Compound 1.
  • the modified release composition comprises an immediate release component comprising the hydrochloride salt of Compound 1 and a delayed release component comprising the hydrochloride salt of Compound 1.
  • the hydrochloride salt of Compound 1 may be the monohydrate or the anhydrous form, conveniently the monohydrate.
  • the modified release composition comprises the immediate release component and the delayed release component in a w/w ratio of between about 1 :4 and 4: 1.
  • the modified release composition comprises the immediate release component and the delayed release component in a w/w ratio of between about 1 :3 and 3: 1, such as between about 1 :2 and 2: 1, or conveniently about 1 :1.
  • the modified release composition comprises between 100 mg and 1000 mg of Compound 1. In an embodiment, the modified release composition comprises between 100 mg and 800 mg of Compound 1, such as between 100 mg and 800 mg, between 100 mg and 500 mg, or between 150 and 500 mg of Compound 1. In an embodiment, the modified release composition comprises about 120 mg or about 240 mg or about 480 mg of Compound 1. In a convenient embodiment, the modified release composition comprises about 240 mg of Compound 1. In this context, reference to the amount of Compound 1 will be understood to refer to the amount of the parent compound (free base), even if the compound is present as a salt of Compound 1.
  • the modified release composition comprises between 50 mg and 500 mg of Compound 1 in the immediate release component and between 50 mg and 500 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises between 50 mg and 250 mg of Compound 1 in the immediate release component and between 50 mg and 250 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises between 50 mg and 150 mg of Compound 1 in the immediate release component and between 50 mg and 150 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises about 60 mg of Compound 1 in the immediate release component and about 60 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises about 120 mg of Compound 1 in the immediate release component and about 120 mg of Compound 1 in the delayed release component.
  • the benefits of the present invention are not limited to a particular type of dosage form having a particular mechanism of drug release.
  • Modified release of Compound 1 or a pharmaceutically acceptable salt thereof may be accomplished by any means known in the pharmaceutical art, including but not limited to the use of osmotic dosage forms, matrix dosage forms, multiparticulate dosage forms, gastric retentive dosage forms, and pulsatile dosage forms.
  • Compound 1 or a pharmaceutically acceptable salt thereof is incorporated into an erodible or non-erodible matrix dosage form.
  • the drug is homogenously dispersed in a matrix material.
  • the modified release solid pharmaceutical composition is an erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix.
  • erodible matrix is meant aqueous-erodible or water-swellable or aqueous soluble, in the sense of being either erodible or swellable or dissolvable in pure water or requiring the presence of an acid or base to ionize the polymeric matrix sufficiently to cause erosion or dissolution.
  • the erodible matrix When contacted with an aqueous environment, the erodible matrix imbibes water and forms an aqueous-swollen gel or “matrix” that Compound 1 or a pharmaceutically acceptable salt thereof can pass or diffuse through depending on its physicochemical properties.
  • Suitable polymers for the erodible matrix are hydrogels such as synthetic polymers derived from vinyl, acrylate, methacrylate, urethane, ester and oxide monomers; or derivatives of naturally occurring polymers such as polysaccharides or proteins (including polysaccharides, gums, starches, alginates, collagen and cellulosics (e.g. HPC or HPMC)).
  • the modified release solid pharmaceutical composition is a non-erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix.
  • Non-erodible matrix materials are inert and drug release is predominantly via diffusion through the matrix.
  • Suitable materials for the inert matrix include water-insoluble plastics (such as PVC or PE), fatty materials (such as carnauba wax and triglycerides) and cross-linked PVP (crospovidone).
  • the matrix material may additionally contain a wide variety of pharmaceutically acceptable excipients known in the pharmaceutical arts, including excipients that ease the manufacturing process and/or improve the performance of the dosage form. Excipients include diluents or bulking agents, lubricants and binders.
  • the matrix polymers may be used alone, or as an appropriate combination of two or more thereof.
  • the matrix dosage form comprises an immediate release component and a delayed release component dispersed within a single polymer matrix.
  • the matrix dosage form comprises an immediate release component dispersed within a polymer matrix and a delayed release component dispersed within a different polymer matrix.
  • the matrix dosage form comprises Compound 1 or a pharmaceutically acceptable salt thereof dispersed within an erodible polymer matrix and Compound 1 or a pharmaceutically acceptable salt thereof dispersed within a non-erodible polymer matrix.
  • Multiparticulate systems include subunits such as mini-tablets, beads, pellets, and granules.
  • Multiparticulates generally comprise a plurality of mini-tablets, beads, pellets or granules that may range in size from about 10 pm to about 2 mm, more typically about 100 pm to 1 mm in diameter.
  • Such multiparticulates may be packaged, for example, in a capsule such as a gelatin capsule or a capsule formed from a polymer such as HPMCAS, HPMC or starch; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet (e.g. a caplet) or pill by compression or other processes known in the art .
  • Such multiparticulates may be made by any known process, such as wet- and drygranulation processes, extrusion/spheronization, roller-compaction, melt-congealing, or by spray-coating seed cores.
  • the multiparticulates are made by spraycoating seed cores.
  • the multiparticulates are made by wet-granulation.
  • the multiparticulates are made by extrusion/spheronization.
  • compositions comprising Compound 1 or a pharmaceutically acceptable salt thereof and optional excipients may be granulated to form multiparticulates of the desired size.
  • Excipients such as a binder and/or a wetting agent, may be blended with the composition to aid in processing and forming the multiparticulates.
  • Binders useful in fabrication of multiparticulates include microcrystalline cellulose, hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), and related materials or combinations thereof.
  • HPC hydroxypropyl cellulose
  • HPMC hydroxypropyl methyl cellulose
  • binders which are useful in granulation and tabletting such as starch, pregelatinized starch, and poly(N-vinyl-2-pyrrolidinone) (PVP) may also be used to form multiparticulates.
  • PVP poly(N-vinyl-2-pyrrolidinone)
  • wetting agents are useful in the fabrication of multiparticulates, particularly when multiparticulates are formed by wet granulation or spray-coating seed cores and Compound 1 is dispersed in an aqueous medium during processing. Wetting agents may also be beneficial in the disintegration and dispersal of multiparticulates comprising Compound 1 after oral dosing.
  • the wetting agent is typically a surfactant or an emulsifier. Conveniently the wetting agent is a nonionic surfactant. In an embodiment the wetting agent is a non-ionic wetting agent selected from polyol esters, polyoxyethylene esters and poloxamers.
  • the polyol esters are selected from one or more of glycol esters, glycerol esters and sorbitan derivatives (such as polysorbate esters).
  • the wetting agent is a poloxamer, (such as poloxamer 188), or a polysorbate ester (such as polysorbate 80).
  • the resulting particles may themselves constitute the multiparticulate or they may be coated by various film-forming materials such as water-swellable or water soluble polymers, and/or they may be combined with other excipients or vehicles to aid in dosing to patients.
  • the modified release composition of the present invention is a multiparticulate system comprising immediate release particulates and delayed release particulates.
  • the immediate release and delayed release particulates may be formed by the same process (e.g. spray-coating seed cores), or by different processes (e.g. IR particulates formed by wet-granulation and DR particulates formed by extrusion/spheronization).
  • the immediate release component of the modified release composition is a multiparticulate comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients selected from binder, wetting agent, diluent and disintegrant.
  • Each subunit of the multiparticulate can be individually coated with a membrane, such as a porous or a water-soluble coating.
  • the immediate release multiparticulate is an immediate release granulate as described hereinabove.
  • the immediate release granulate is obtained by a high shear wet granulation process as described hereinabove.
  • the immediate release granulate is obtained by an extrusion/spheronization process, wherein spherical pellets are obtained.
  • each subunit of the immediate release multiparticulate is an inert core coated with a drug layer.
  • multiparticulates in the form of beads or pellets may be prepared by building the Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying the Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g. a binder dispersion, for example HPMC) onto seed cores in a fluidized bed such as a Wurster coater or a rotary processor.
  • an appropriate solution / dispersion vehicle e.g. a binder dispersion, for example HPMC
  • the inert seed core can be comprised of a sugar (for example a nonpareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose.
  • the inert core comprises sugar spheres mesh 45/60 (250-355 microns).
  • An example of a suitable composition and method is to spray a dispersion of Compound 1 or a pharmaceutically acceptable salt thereof / binder (e.g. HPMC) composition in water on to the seed core.
  • the immediate release (IR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 10% to about 90% (w/w) of the weight of the IR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 5% to about 50% (w/w) of the total weight of the IR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 5: 1 to 15: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 5 : 1 ; and c) optionally a coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging from
  • the delayed release component of the modified release composition is a multiparticulate comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients selected from binder, wetting agent, diluent and disintegrant.
  • Each subunit of the multiparticulate can be individually coated with a membrane, such as a porous or a water-soluble coating, prior to coating with a delayed release coating.
  • the delayed release multiparticulate is a delayed release granulate as described hereinabove.
  • the delayed release granulate is obtained by a high shear wet granulation process as described hereinabove.
  • the delayed release granulate is obtained by an extrusion/spheronization process, wherein spherical pellets are obtained, which are subsequently coated with a delayed release coating as described herein.
  • each subunit of the delayed release multiparticulate is an inert core coated with a drug layer and the drug layer-coated core is coated with a polymeric material of the type useful for providing delayed release of Compound 1.
  • multiparticulates in the form of beads or pellets may be prepared by building the Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying the Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g.
  • a binder dispersion for example HPMC
  • the seed core can be comprised of a sugar (for example a non-pareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose.
  • the inert core comprises sugar spheres mesh 45/60 (250-355 microns).
  • An example of a suitable composition and method is to spray a dispersion of Compound 1 or a pharmaceutically acceptable salt thereof / binder (e.g. HPMC) composition in water on to the seed core.
  • a delayed release coating is then employed to fabricate the membrane, which is applied over Compound 1 or a pharmaceutically acceptable salt thereof layered seed cores.
  • the rate release from the coated multiparticulates can be controlled by factors such as the composition and binder content of the drug-coated core, the thickness and permeability of the delayed release coating, and the surface-to-volume ratio and size of the multiparticulates. It will be appreciated by those skilled in the art that increasing the thickness of the coating will decrease the release rate, whereas increasing the permeability of the coating or the size or surface-to- volume ratio of the multiparticulates will increase the release rate. If desired, the permeability of the coating may be adjusted by blending of two or more materials.
  • the delayed release (DR) pellet coating dissolves at pH values greater than about 5.5. In an embodiment, the delayed release coating dissolves at pH values greater than about 6.0. In an embodiment, the delayed release coating dissolves at pH values greater than about 7.0. Conveniently, the delayed release coating dissolves at about pH 5.5. In an embodiment, the delayed release pellet coating comprises methyl acrylate-methacrylic acid copolymer, ethyl acrylate-methacrylic acid copolymer, hydroxy propyl methyl cellulose acetate succinate or cellulose acetate phthalate.
  • the delayed release pellet coating comprises methyl acrylate-methacrylic acid copolymer or ethyl acrylatemethacrylic acid copolymer.
  • the delayed release pellet coating is selected from Eudragit® L 100-55, Eudragit® FS30D, Eudragit® L100, Eudragit® L 12,5, Eudragit® L30 D- 55, Eudragit® S100 and Eudragit® S12,5, such as Eudragit® L 100-55. More conveniently, the delayed release pellet coating is selected from Eudragit® FS30D and Eudragit® L30 D-55.
  • the delayed release (DR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 10% to about 80% (w/w) of the weight of the DR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 5% to about 50% (w/w) of the total weight of the DR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 5: 1 to 30: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 5: 1; c) optionally a sub-coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging
  • a polymer such
  • the delayed release (DR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 40% to about 60% (w/w) of the weight of the DR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 15% to about 35% (w/w) of the total weight of the DR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 15: 1 to 25: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 3: 1; c) optionally a sub-coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging from
  • a sub-coat can optionally be applied between the drug layer and the modified release layer if separation is needed.
  • the coat can be comprised of HPMC or magnesium stearate.
  • the IR or DR multiparticulates or pellets range in size from about 150 um to about 1000 pm, more conveniently about 350 pm.
  • the multiparticulate modified release composition will comprise a plurality of IR pellets and a plurality of DR pellets that may be packaged, for example, in a capsule such as a gelatin capsule or a capsule formed from a polymer such as HPMC AS, HPMC or starch; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet (e.g. a caplet) or pill by compression or other processes known in the art.
  • the multiparticulate modified release composition comprising the plurality of pellets is packaged in a capsule, such as a gelatin capsule or a capsule formed from a polymer such as HPMCAS, HPMC or starch.
  • the multiparticulate system comprises a plurality of immediate release mini -tablets and a plurality of delayed release mini-tablets encapsulated in a capsule.
  • mini-tablets have benefits compared to other multiparticulate systems in that they enable much higher drug loadings (up to approximately 64% w/w as described herein for Example 27 and 28) of Compound 1 to be achieved without detrimental impact of disintegration and/or dissolution properties.
  • an active compound such as Compound 1, which is particularly prone to agglomeration and poor disintegration/dissolution properties, even at relatively low drug loadings from compositions such as direct compression tablets.
  • High drug loadings are advantageous in terms of dosage form size and patient convenience and compliance. This was not previously possible with other compositions and formulations; for example immediate release pellets were prepared with approximately 20% w/w drug loadings (Examples 19 and 20).
  • the modified release composition comprises an immediate release component comprising mini-tablets and a delayed release component comprising mini -tablets.
  • the mini -tablets having an average diameter of 1.5 to 3.0 mm, such as 2.0 to 2.5 mm.
  • the mini -tablets comprise a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients.
  • the mini-tablets comprise a granulate, wherein the granulate comprises the hydrochloride salt of Compound 1 (conveniently as the mono-hydrate) and one or more pharmaceutically acceptable excipients.
  • the amounts refer to the Compound 1 free base amount, as the presence and nature of the salt and the presence of any solvates or hydrates will affect the total amount required to provide the free base amount; for example, 135 mg of the mono-hydrate hydrochloride salt of Compound 1 is equivalent to 120 mg of the parent free base.
  • the mini-tablets comprise a pharmaceutically acceptable salt of Compound 1 in a loading of greater than 40 % w/w (such as greater than 45 % w/w, greater than 50 % w/w, greater than 55 % w/w, or greater than 60 % w/w).
  • the mini-tablets comprise a pharmaceutically acceptable salt of Compound 1 in a loading of 40 to 75% w/w (such as 50 to 70 % w/w, 60 to 70 % w/w, 62 to 68 % w/w, or about 64 to 65 % w/w).
  • each mini-tablet comprises about 6 to 10 mg (such as about 8 to 9 mg) of a pharmaceutically acceptable salt of Compound 1.
  • the immediate release component comprises 10 to 20 mini-tablets, such as 14 to 16 mini-tablets.
  • the delayed release component comprises 10 to 20 mini-tablets, such as 14 to 16 mini-tablets.
  • the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1. In an embodiment, the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1.
  • the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1 and the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1.
  • the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1 and the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1.
  • the mini -tablets comprise a granulate, wherein the granulate comprises the hydrochloride salt of Compound 1 (conveniently as the mono-hydrate) and a wetting agent.
  • the wetting agent is a non-ionic wetting agent, optionally selected from polyol esters, polyoxyethylene esters and poloxamers.
  • Polyol esters include glycol esters, glycerol esters and sorbitan derivatives (such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80).
  • the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25, such as between 12 and 18. Most conveniently, the wetting agent is polysorbate 80.
  • HLB hydrophilic-lipophilic balance
  • the wetting agent is present in the composition at 0.5 to 3.0% w/w, such as 1.5 to 2.5% w/w.
  • the granulate comprises one or more lubricants, conveniently wherein the one or more lubricants are selected from magnesium stearate, calcium stearate, hydrogenated vegetable oil, stearic acid, sodium stearyl fumarate, mineral oil, hydrogenated vegetable oil, and polyethylene glycol.
  • the one or more lubricants are magnesium stearate and hydrogenated vegetable oil.
  • the one or more lubricants are magnesium stearate and sodium stearyl fumarate. In an embodiment, the one or more lubricants are present in the composition at a total level of between 3 and 7% w/w, such as between 4 and 6% w/w. In an embodiment, magnesium stearate is present in the composition at between 2 and 4% w/w, and hydrogenated vegetable oil is present in the composition at between 2 and 4% w/w. In an embodiment, magnesium stearate is present in the composition at between 2 and 4% w/w, and sodium stearyl fumarate is present in the composition at between 2 and 4% w/w.
  • the one or more pharmaceutically acceptable excipients does not comprise microcrystalline cellulose.
  • the one or more pharmaceutically acceptable excipients are not acidic, or have a pH of greater than 6 when dissolved or slurried in water. By avoiding acidic excipients, the stability of Compound 1 in the composition is improved.
  • the granulate used to provide the mini-tablets is prepared by fluid bed granulation.
  • the delayed release component comprises mini-tablets
  • the delayed release mini-tablets are individually substantially coated with a delayed release coating.
  • the delayed release coating dissolves at pH values greater than about 4.5, such as greater than about pH 5.5.
  • each delayed release mini -tablet comprises a greater than 6% (such as between 8 and 12%, or about 10%) weight gain delayed release coating.
  • an oral modified release pharmaceutical composition wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets, the mini-tablets comprising a hydrochloride salt of Compound 1.
  • an oral modified release pharmaceutical composition wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets, the mini-tablets comprising 40-75 % w/w of a hydrochloride salt of Compound 1.
  • an oral modified release pharmaceutical composition wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets, the mini -tablets comprising 40-75 % w/w of a hydrochloride salt of Compound 1; wherein each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core.
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
  • an oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
  • the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
  • the delayed release coating dissolves at pH values greater than about 5.5, such as greater than about 6.0.
  • each mini-tablet comprises a greater than 6% weight gain (such as a greater than 8% weight gain) delayed release coating.
  • the combination according to the first aspect or the modified release composition according to the second aspect of the invention can provide synergy in the form of high gastrointestinal exposure with acceptable safety and/or tolerability.
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 50 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 20 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 15 ng/ml, such as less than 10 ng/ml.
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 2 ng/ml and 100 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is between 2 ng/ml and 50 ng/ml or 2 ng/ml and 30 ng/ml or 5 ng/ml and 20 ng/ml.
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject of 100 mg to 1000 mg of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 50 ng/ml, such as less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject of 200 mg to 1000 mg (such as 240 mg to 960 mg, or about 480 mg, or about 960 mg) of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml or 50 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • a combination or modified release composition provides, after oral administration in the fed state to a subject of 200mg to 1000 mg of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, wherein about 50% of Compound 1 is derived from the immediate release component and about 50% of Compound 1 is derived from the delayed release component.
  • the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml or 50 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • a combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • the combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • the combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 15 and 35 ng/ml, such as between 20 and 30 ng/ml, or between 22 and 28 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • the combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 40 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • the combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 20 and 40 ng/ml, such as between 25 and 35 ng/ml, or between 27 and 33 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 15 and 35 ng/ml, such as between 20 and 30 ng/ml, or between 22 and 28 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 20 and 40 ng/ml, such as between 25 and 35 ng/ml, or between 27 and 33 ng/ml.
  • Cmax geometric mean maximum plasma concentration
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 .
  • the ratio of Cmax/AUCo-24 is between 0.04 and 0.3 h' 1 , such as between 0.04 and 0.2 h' 1 or between 0.06 and 0.16 h' 1 .
  • a combination according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 , such as between 0.04 and 0.3 h' 1 , between 0.04 and 0.2 h" between 0.06 and 0.16 h' 1 , between 0.2 and 0.35 h' 1 , or between 0.25 and 0.30 h' 1 .
  • a modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 , such as between 0.04 and 0.3 h' 1 , between 0.04 and 0.2 h' 1 , between 0.06 and 0.16 h' 1 , between 0.2 and 0.35 h' 1 , or between 0.25 and 0.30 h' 1 .
  • a combination according to the present invention produces, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 , such as between 0.04 and 0.3 h' 1 , between 0.04 and 0.2 h" between 0.06 and 0.16 h' 1 , between 0.1 and 0.2 h' 1 , or between 0.12 and 0.19 h' 1 .
  • a modified release composition according to the present invention produces, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 , such as between 0.04 and 0.3 h' 1 , between 0.04 and 0.2 h' 1 , between 0.06 and 0.16 h' 1 , between 0.1 and 0.2 h' 1 , or between 0.12 and 0.19 h' 1 .
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml (conveniently less than 75 ng/ml, 50 ng/ml, 30 ng/ml, 20 ng/ml, 15ng/ml or 10 ng/ml) and a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 .
  • Cmax geometric mean maximum plasma concentration
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 50 ng/ml (conveniently less than 30 ng/ml, 20 ng/ml, 15ng/ml or 10 ng/ml) and a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h' 1 .
  • Cmax geometric mean maximum plasma concentration
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.3 h' 1 .
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 15 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.2 h' 1 .
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 10 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.16 h' 1 .
  • Cmax geometric mean maximum plasma concentration
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml and a geometric mean maximum plasma concentration (Cmax) of the glucuronide metabolite of Compound 1 of greater than 500 ng/ml.
  • the geometric mean maximum plasma concentration (Cmax) of the glucuronide metabolite of Compound 1 is greater than 1000 ng/ml, 1500 ng/ml, 2000 ng/ml, 2500 ng/ml, 3000 ng/ml, 5000 ng/ml, 6000 ng/ml or 8000 ng/ml.
  • a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, an area under the plasma concentration-time curve from time zero up to 24 hours post-dose (AUC0-24) of the glucuronide metabolite of Compound 1 of greater than 10,000 ng*h/mL, conveniently greater than 15,000, 25,000, 35,000, 50,000, 70,000, or 100,000 ng*h/mL.
  • the combinations according to the first aspect or the modified release composition according to the second aspect of the invention advantageously provide high colon tissue exposure of Compound 1 after oral dosing. Colon tissue concentrations can be measured by biopsy as described in the Examples section.
  • the combination or modified release composition as described herein provides colonic tissue exposure greater than or equal to the systemic exposure of Compound 1 following oral solution dosing of the composition to a subject.
  • the combination or modified release composition as described herein delivers higher median levels of Compound 1 to the colon after oral dosing, than achieved with a corresponding dose formulated as an oral solution.
  • the combination or modified release composition as described herein provides median sigmoid colon tissue concentrations of greater than 100 ng/g (such as greater than 140 ng/g) after oral dosing to a fasted subject.
  • the combination as described herein provides median sigmoid colon tissue concentrations of greater than 100 ng/g (such as greater than 140 ng/g) after oral dosing to a fed subject.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations of Compound 1 greater than 100 ng/g.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations of Compound 1 greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg or 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • a combination according to the present invention produces, after oral administration in the fed state of 480 mg daily Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • a combination according to the present invention produces, after oral administration in the fed state of 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 480mg contained within an immediate release composition and 480 mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • a modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg or 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently wherein about 50% of Compound 1 is derived from the immediate release component and about 50% of Compound 1 from the delayed release component of the composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • a modified release composition according to the present invention produces, after oral administration in the fed state of 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 480mg contained within an immediate release composition and 480mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
  • Colonic biopsies may also allow for measurement of the proportion of hypoxia- inducible factor (HIF)-la positive cells in the biopsy samples both before (baseline) and after (e.g.
  • HIF hypoxia- inducible factor
  • a delayed release composition as described herein provides a greater than 10% (such as greater than 20%) increase from baseline in the proportion of HIF-la positive cells in the colon after oral dosing of the composition to a subject. In an embodiment, a delayed release composition as described herein provides a greater than 10% (such as greater than 20%) increase from baseline in the proportion of HIF-la positive cells in the colon after oral dosing of the composition comprising at least 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject.
  • a delayed release composition as described herein provides a greater than 10% (such as greater than 20%, greater than 30%, greater than 40%, or greater than 50%) increase from baseline in the proportion of HIF-la positive cells in the sigmoid colon tissue after oral dosing of the composition comprising at least 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject.
  • a combination or modified release composition according to the present invention produces, after oral administration, an increase in the proportion of HIF-la positive cells from baseline in greater than 10% of subjects, such as greater than 20% of subjects or greater than 30% of subjects.
  • the combinations or modified release compositions according to the present invention are capable of delivering high colonic tissue exposure of Compound 1 after oral dosing, without producing substantial side effects, which might be expected were the same dose of Compound 1 administered solely via an immediate release composition.
  • High systemic exposure of Compound 1 has the potential to trigger elevation of systemic levels of erythropoietin (EPO) and vascular endothelial growth factor (VEGF), which may lead to undesired side effects including those associated with increased hemoglobin and hematocrit (such as polycythemia following chronic exposure).
  • EPO erythropoietin
  • VEGF vascular endothelial growth factor
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g; plasma VEGF levels less than 500 pg/ml and plasma EPO levels less than 100 mIU/ml.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 200 ng/g; plasma VEGF levels less than 250 pg/ml and plasma EPO levels less than 50 mIU/ml.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 300 ng/g; plasma VEGF levels less than 150 pg/ml and plasma EPO levels less than 30 mIU/ml.
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g; and after 12 weeks daily dosing of Compound 1 fewer than 50% of subjects experience a treatment related adverse event.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 200 ng/g; and after 12 weeks daily dosing of Compound 1 or a pharmaceutically acceptable salt thereof fewer than 40% of subjects experience a treatment related adverse event.
  • a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 300 ng/g; and after 12 weeks daily dosing of Compound 1 or a pharmaceutically acceptable salt thereof fewer than 30% of subjects experience a treatment related adverse event.
  • colonic tissue concentrations such as colonic sigmoid tissue concentrations
  • the present invention also provides the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, for use in the treatment of diseases or conditions mediated alone, or in part, by PHD.
  • an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1
  • an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammatory bowel disease.
  • the treatment is for ulcerative colitis or Crohn’s disease.
  • a modified release composition as described herein for use in the treatment of inflammatory bowel disease is provided.
  • the treatment is for ulcerative colitis or Crohn’s disease.
  • the present invention provides a method of treating diseases or conditions mediated alone, or in part, by PHD, the method comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating an inflammatory bowel disease the method comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating ulcerative colitis or Crohn’s disease comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of treating diseases or conditions mediated alone, or in part, by PHD, the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating an inflammatory bowel disease the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • a method of treating ulcerative colitis or Crohn’s disease the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof.
  • the present invention provides for the use of a combination according to the first aspect of the invention, or a modified release composition according to the second aspect of the invention, in the manufacture of a medicament for treating diseases or conditions mediated alone, or in part, by PHD.
  • the disease or condition is inflammatory bowel disease.
  • the disease or condition is ulcerative colitis or Crohn’s disease.
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention is administered once or twice daily.
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention is administered orally once daily.
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention is administered orally once daily in the morning.
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention is administered orally twice daily.
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention is administered with food, or up to 30 minutes after a meal.
  • the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal.
  • the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal, once a day. In an embodiment, the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal, twice a day.
  • the combination or the modified release composition is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960 mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg.
  • the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg; and wherein about 50% of Compound 1 is derived from the immediate release composition and about 50% of Compound 1 is derived from the delayed release composition.
  • the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 480 mg; and wherein the immediate release composition comprises about 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof and about the delayed release composition comprises about 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof.
  • the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 960 mg; and wherein the immediate release composition comprises about 480 mg of Compound 1 and about the delayed release composition comprises about 480 mg of Compound 1.
  • the modified release composition is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg; and wherein about 50% of Compound 1 or a pharmaceutically acceptable salt thereof is derived from the immediate release component and about 50% of Compound 1 or a pharmaceutically acceptable salt thereof is derived from the delayed release component.
  • the combination or the modified release composition may be administered in further combination with one or more additional treatments.
  • the combination or the modified release composition may be administered with one or more additional treatments for inflammatory bowel disease (ulcerative colitis or Crohn’s disease).
  • the one or more additional treatments are selected from 5- aminosalicylates (5-ASAs) and steroids.
  • the 5-ASAs are selected from sulphasalazine, mesalazine and olsalazine.
  • the steroids are selected from prednisone and budesonide. Ulcerative Colitis
  • the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, are particularly useful in the treatment of ulcerative colitis.
  • the ulcerative colitis being treated is mild-to- moderate ulcerative colitis.
  • Suitable ulcerative colitis patients to be treated with the combinations or the modified release compositions described herein include those having a Mayo score of 5- 10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1 (the Examples contain details of these diagnostic assessments).
  • a method for treating ulcerative colitis comprising administering to the subject a combination, or a modified release composition, as described herein and wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency sub score of> 1.
  • the ulcerative colitis patient being treated is already receiving a different treatment for the condition.
  • the patient is already receiving an oral 5-aminosalicylate (5-ASA) drug for at least two weeks prior to the treatment commencing. Therefore, in an embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein and wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1, despite being on a stable dose of an oral 5-ASA for at least two weeks prior to the treatment.
  • an oral 5-aminosalicylate 5-ASA
  • 5 -ASA drugs may be given at different doses depending on a number of factors including the severity of the ulcerative colitis in a given patient. Patients with infrequent relapses may be treated with a low dose 5-ASA maintenance treatment, whereas higher doses (such as greater than or equal to 3 g per day) of 5-ASA may be needed in patients with mild or moderately active disease.
  • a ‘stable’ dose refers to the same dose of 5-ASA being administered to the patient with ulcerative colitis for at least two weeks prior to the treatment commencing and that same dose continuing during the treatment described in the present invention. In an embodiment that dose is greater than or equal to 3 g (such as 4 g) per day of 5-ASA.
  • the efficacy of the claimed treatment for ulcerative colitis can be assessed by various measures, which include the treated patient having a Modified Mayo score of ⁇ 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1. Therefore, in an embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of ⁇ 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1.
  • a method for treating ulcerative colitis comprising administering to the subject a combination, or a modified release composition, as described herein; wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1; and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of ⁇ 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1.
  • a method for treating ulcerative colitis comprising administering to the subject a combination, or a modified release composition, as described herein; wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1, despite being on a stable dose of an oral 5-ASA for at least two weeks prior to the treatment; and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of ⁇ 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1.
  • the slurry was adjusted to 10 °C and agitated for 1 hour.
  • the product slurry was filtered and the wet cake was washed with pre-cooled (10°C) purified water (3 x 1 vol).
  • the filtered cake was dried under vacuum at less than 35 °C.
  • Example 2.1 [0289] HPLC analysis of Example 2.1 showed a purity of 99.6% by area. LC-MS demonstrated an observed a mass of 434.2 m/z +ve ionization, consistent with the expected mass of 433.93 g/mol. High Performance Liquid Chromatography-Charged Aerosol Detection (HPLC- CAD) analysis confirmed Example 2.1 to be a mono-HCl salt.
  • Example 2.1 Thermogravimetric analysis of Example 2.1 showed an initial mass loss (from onset to ca. 80°C) of 3.4% related to loss of water (ca. 0.9 equiv). A loss of ca. 1 equivalent of water indicated Example 2.1 is a monohydrate. A second mass loss of 40.9% was observed (with an onset at ca. 160°C) related to the material melt and subsequent decomposition. Differential thermal analysis showed a shallow, broad endothermic event (onset at ca. 53°C) related to the loss of water. An initially sharp then broadening melt was recorded from an onset of ca. 190°C and a peak at ca. 194°C. DSC analysis showed a broad endothermic event with onset at ca.
  • Example 2.1 HPLC analysis of Example 2.1 showed a purity of 99.6% by area. LC-MS demonstrated an observed a mass of 434.2 m/z +ve ionization, consistent with the expected mass of 433.93 g/mol. High Performance Liquid Chromatography-Charged Aerosol Detection (HPLC- CAD) analysis confirmed Example 2.1 to be a mono-HCl salt.
  • Example 2.1 XRPD characterisation of Example 2.1 gave the peak list presented in Table 2 (referred to as Form A).
  • Polarized Light Microscopy (PLM) showed small birefringent crystals (ca. 20- 100pm) with plate-like morphology.
  • HPLC showed the material is 99.9% pure.
  • PLM analysis showed the material to have a form of fine (ca. 5-20 pm), birefringent crystals with irregular morphology and agglomeration.
  • TG analysis showed no solvent related mass loss, which correlates with Example
  • DT analysis showed a sharp melting endotherm with an onset at ca. 193°C and peak at ca. 195°C, followed by thermal decomposition.
  • DSC analysis showed no significant endothermic events related to solvent loss. An intense, sharp melting endotherm was observed with an onset at ca. 196°C and peak at ca. 198°C.
  • a wetting agent may advantageously be used to improve the disintegration and dispersal of Compound 1 hydrochloride salt from the resultant composition.
  • the wetting agent is dissolved in water or an aqueous medium and added to the shear-blended formulation to assist in granulation. It is postulated that when a wetting agent is intimately mixed with Compound 1 hydrochloride salt within a granulate composition, then the two-stage disintegration of the composition and dispersal of the API in aqueous media is promoted.
  • the wetting agent may be a surfactant or emulsifier.
  • the wetting agent is a non-ionic surfactant.
  • the wetting agent has a hydrophilic-lipophilic balance (HLB) between 10 and 25 (such as between 12 and 18).
  • HLB hydrophilic-lipophilic balance
  • Suitable examples of non-ionic wetting agents include polyol esters, polyoxyethylene esters and poloxamers. Examples of polyol esters include glycol esters, glycerol esters and sorbitan derivatives. Sorbitan derivates comprise polysorbates (such as polysorbate 20, polysorbate 40, polysorbate 60 and polysorbate 80). Most conveniently the wetting agent is polysorbate 80.
  • Hardness values and disintegration times, in water and pH 6.8 buffer, of formulations F2-F5 are provided in Table 5. Disintegration times increased, as compared to the direct compression formulations, and were similar in water and pH 6.8 buffer.
  • Formulations F6, F7 and F8 were prepared as detailed in Table 6 and the process described in Figure 2. These formulations incorporated the following modifications: using intra- granular microcrystalline cellulose (at approximately twice the level of intra-granular lactose monohydrate); removal of Starch 1500 from the formulation; and using HPC in place of PVP K29/32 as the primary binder.
  • An Opadry® Clear 03K19229 sub coat was applied to F7, to produce F7-SC1, which was then enterically coated with Acryl-Eze® White 93018359, to produce F7-SC1-EC2.
  • An immediate release Opadry® II White 85F 18422 coat was applied to F8 to a weight gain of 4%, to produce F8-IRC2.
  • F7 and F8 were compressed at similar hardness levels to produce plain, white to off-white, round, 11 mm Diam., shallow biconvex, bevel edge tablets.
  • F6 tablets were compressed at three hardness levels to produce plain, white to off white, round, 11mm diameter, shallow biconvex, bevel edge tablets.
  • the various components used in the formulation may be acquired from standard formulation excipient suppliers, such as: Lactose Monohydrate (Foremost #310);
  • Microcrystalline Cellulose (Avicel PH-102); Hydroxypropyl Cellulose (Klucel EXF Pharm); Crospovidone (Kollidon CL); Silicified Microcrystalline Cellulose (Prosolv SMCC HD 90); Crospovidone (Kollidon CL); Magnesium Stearate (Ligamed MF-2-V).
  • Step 3 Premix the screened materials from Step 2 in a suitably sized high-shear granulator.
  • Step 8 Transfer the milled granules from Step 8 to a suitably sized blender.
  • Step 17 Coat the tablets with Step 16 in suitably-sized pan.
  • the in-process controls applied at the compression stage (step 14) include individual tablet weight (370-430mg); individual tablet hardness (7-11 kilopond) and average weight of 10 tablets (3.800-4.200g).
  • 60mg DR tablets present as white to off-white, round film-coated tablets.
  • the strength of the active dosage form is 60mg (calculated as the anhydrous free base of Compound 1).
  • the composition of the Tablets and a representative batch for Compound 1 DR Tablets is provided in Table 10.
  • the typical batch blend size is 5.5 kg.
  • the theoretical batch size is 13,750 tablets.
  • Step 8 Transfer the milled granules from Step 8 into a suitably sized blender.
  • Step 17) Coat the tablets with Step 16 in suitably sized pan.
  • the in-process controls applied at the compression stage (step 14) include individual tablet weight (370-430mg); individual tablet hardness (7-11 kilopond) and average weight of 10 tablets (3.800-4.200g).
  • the F9/F10 core composition had the physical characteristics as set out in Table
  • F9/F10 blended formulations were compressed to produce plain, white to off- white, round, 11 mm Diam., shallow biconvex, bevel edge tablets.
  • EXAMPLE 9 ALTERNATIVE HIGH SHEAR WET GRANULATION TABLET FORMULATIONS - 120 MG TABLETS
  • the core blends of formulations Fl 1 and F12 were prepared according to a high shear wet granulation process as set out in Figure 6.
  • the amount of granulating solution containing dissolved HPC (wet portion) and Polysorbate 80 was applied in its entirety during mixing and incremental amounts of water were added to complete the granulation.
  • a granulating solution rate of addition of 55 g/min was used for Fl 1 and a rate of approximately 65 g/min was used for F 12.
  • Fl 1 and F 12 blended formulations were compressed to produce plain, white to off-white, caplet-shaped, biconvex, plain, 17.4 mm x 6.7 mm tablets. Physical Characteristics of Fl 1 & F12 tablets are presented in Table 19. Both formulations compressed very well. Weight uniformity was excellent, tablet hardness values remained consistent throughout the runs and friability was very low. The core tablets had disintegration times ranging from under 1 minute to approximately 1 minute and 45 seconds.
  • F12 DR tablets were prepared by sub-coating core tablets using Colorcon’s
  • Sub-coated tablets as prepared above were enteric-coated using Colorcon’s Acryl- Eze® 93018359 White to a weight gain of 14% in an O’Hara Labcoat II fully-perforated coating pan (12” insert) using the following processing parameters:
  • the F 12 DR tablets showed an average acid uptake of 3.1% and were intact with unchanged appearance after the acid treatment. Upon exposure to pH 6.8 buffer the coating dissolved after approximately 10.5 minutes and all the tablets had fully disintegrated after 11.5 minutes with no agglomerates remaining.
  • the Comparator SC1-EC2 tablets had been prepared by direct compression, without water addition to the processing. Despite the F7-SC1-EC2 tablets having had water added during the wet granulation processing, this did not result in increased degradation being seen in the six-month stability study; in fact slightly more degradation was observed for the direct compression tablets than was seen with the wet granulation tablets. As can be seen from Table 21, after 6 months at 25 °C / 60% RH and at 40 °C / 75% RH, the levels of total related impurities seen with the direct compression tablets were 0.91% and 2.70% respectively. None of the IR or DR tablets prepared by high shear wet granulation had such high levels of related impurities after 6 months under comparable conditions. [0341] It would therefore appear that, surprisingly, the chemical stability of Compound 1 on storage is slightly better when formulated in wet granulation compositions according to the present invention, as opposed to comparable compositions prepared by direct compression.
  • EXAMPLE 11 A PHASE 1 A RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, SINGLE ASCENDING DOSE CLINICAL STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF COMPOUND 1 (SOLUTION) IN HEALTHY MALE VOLUNTEERS.
  • Placebo dosing solutions were identical to active, except without Compound 1. Subjects received a single oral solution of 50mL or lOOmL; all subjects received a total volume of 150mL of liquid. Since HPpCD concentration remained constant, subjects in Cohort 5 received twice the amount of HPpCD relative to subjects in Cohorts 1 to 4. Dosing, including consumption of rinse water, was completed within 3 minutes, administered in the morning after an overnight fast, and maintained for up to 4 hours after dosing.
  • the primary objective of the study was to assess the safety and tolerability of ascending dose levels of Compound 1 after single oral dose administration.
  • the safety endpoints included AE incidence, clinically significant changes in vital signs, ECG parameters, clinical laboratory tests and physical examination.
  • the secondary objective was to characterize the single-dose pharmacokinetic (PK) parameters of Compound 1 after ascending doses.
  • PK pharmacokinetic
  • Subjects within each cohort were assigned to randomized treatment; 6 assigned to Compound 1 and 2 assigned to placebo. Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use. Each cohort consisted of white, black or African American, and Asian male subjects although the majority were white (47.5%). Subject demographics are summarized in Table 23.
  • each dosing container was rinsed with 25mL of water.
  • Clinical Laboratory Assessments included hematology (complete blood count (CBC), hemoglobin, hematocrit, red blood cell (RBC), mean corpuscular cell volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, red cell distribution width, mean platelet volume, white blood cell count with differential (neutrophils, immature granulocytes, lymphocytes, monocytes, eosinophils, basophils), platelets, and automated reticulocyte count); chemistry (sodium, potassium, bicarbonate, chloride, calcium, phosphorus, fasting glucose, creatinine, blood urea nitrogen, creatinine phosphokinase, uric acid, albumin, total protein, total bilirubin, alkaline phosphatase, ALT, AST, lactate dehydrogenase, total cholesterol and triglycerides); and urinalysis (bilirubin, blood glucose, ketones, pH, protein, specific gravity and microscopic examination).
  • CBC complete blood count
  • PK profiles (Compound 1 mean plasma concentration, over time 0-16 hours), are presented in Table 25, and Figures 7 and 8 for all subjects and subjects who did not experience vomiting (ie the 4 subjects in Cohort 4).
  • the profiles show Compound 1 was rapidly absorbed, with Tmax of less than 1 hour.
  • the peak concentrations, Cmax and AUC parameters increased with increasing dose and were higher for drug administered in a larger volume (i.e., Cohort 5, 240mg in lOOmL vehicle) compared with the other cohorts.
  • Elimination parameters (b/ 2 , CL/F) suggested rapid clearance from plasma and relatively large volume of distribution.
  • Hummel Power analysis revealed linear dose proportionality for Cmax, both for all subjects and for the subset excluding the four cohort 4 emesis subjects.
  • the slope value was close to 1 and the 90% confidence interval (CI) for the slope was contained within the critical interval (ie, 0.72-1.28).
  • AUCiast values were slightly above linearity with a slope of 1.167 for all subjects and 1.231 for subjects without emesis.
  • the upper bound of the 90% CI for the slope in both cases was above the critical range.
  • Primary Endpoint Conclusion - Safety and Tolerability All doses evaluated (20- 240mg) in 50mL or lOOmL vehicle were determined to be safe and well tolerated. No deaths or SAEs occurred and all of the TEAEs reported were mild in severity. No clinically significant laboratory abnormalities occurred.
  • EXAMPLE 12 A PHASE 1 A RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTIPLE ASCENDING DOSE CLINICAL STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF COMPOUND 1 (SOLUTION) IN HEALTHY MALE AND FEMALE VOLUNTEERS.
  • Compound 1 was administered as the HCl-salt (prepared analogously to Example 2.1). Compound 1 was administered orally in 100mL of a 10% hydroxypropyl-beta-cyclodextrin (HPpCD) solution, once daily for 8 days. Placebo consisted of a HPpCD solution administered at a volume and dose schedule corresponding with that of Compound 1. See Table 26.
  • HPpCD hydroxypropyl-beta-cyclodextrin
  • the primary objective of the study was to assess the safety and tolerability of ascending doses of Compound 1 after multiple oral doses administration.
  • the safety endpoints included TEAE incidence and severity, clinically significant changes in vital signs, ECG parameters, clinical laboratory tests and physical examination.
  • the secondary objective was to characterize the multiple dose pharmacokinetic (PK) parameters of Compound 1, after ascending doses.
  • Secondary endpoints included determination of PK parameters in plasma and urine, fecal excretion and assessment of colonic biopsy samples.
  • the first 3 dose cohorts were in an ascending-dose design. Prior to each dose escalation, a safety monitoring team determined if dose escalation was appropriate.
  • the planned dose levels are given in table 26.
  • the planned study scheme is presented as a flow chart in figure 10.
  • Subjects were healthy males or females, age 18 to 55, with a Body Mass Index (BMI) between 18 to 33 kg/m 2 .
  • Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use, and did not display any significant colorectal symptoms or findings.
  • BMI Body Mass Index
  • Subjects fasted for a minimum of 8 hours prior to obtaining blood samples Subjects fasted for a minimum of 8 hours prior to obtaining blood samples. During the treatment period, subjects fasted from at least 8 hours prior to dosing in the morning. After dosing, subjects are served a standard breakfast after the 2-hour blood sample has been collected (on Day 1 and 7), a standard lunch approximately 4 hours post-dose, a dinner approximately 10 hours post-dose, and a light snack 12-14 hours post-dose.
  • AUCiast area under concentration-time curve from time zero to last quantifiable concentration
  • Urine samples were collected at the following time points relative to dose administration on Day 1 and Day 7: pre-dose; 0-4, 4-8, and 8-24 hours post dose.
  • NA not available as 5 of 6 subjects had concentrations of Compound 1 below limit of quantitation.
  • N 2, 1, 1, 1, and 2 subjects in Cohort 2, 3, 4, 5 and 6, respectively, for whom Compound 1 concentrations were below the limit of quantitation
  • HIF hypoxia inducible factor
  • Table 34 presents the proportions of HIF-la positive cells evaluated at baseline and Day 8, and the change from baseline to Day 8 of the proportion of HIF-la positive cells.
  • the mean proportion of HIF-la positive cells ranged from 1% to 80% across all experimental groups.
  • the proportion of HIF-la positive cells increased from baseline to Day 8 across all groups and a numerically higher increase was seen for the combined group (“Pooled") (35.2 ⁇ 28.8 [mean ⁇ SD]) compared to the placebo group (13.7 ⁇ 20.9 [mean ⁇ SD]).
  • Figure 12 shows the box plot of the proportions of HIF-la positive cells evaluated at baseline and Day 8. TABLE 34. DESCRIPTIVE STATISTIC FOR MANUALLY ASSESSED PROPORTIONS OF HIF- la POSITIVE
  • EXAMPLE 13 A PHASE IB, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, CLINICAL STUDY TO EVALUATE THE SAFETY, TOLERABILITY, PHARMACOKINETICS AND PHARMACODYNAMICS OF COMPOUND 1 (SOLUTION)
  • Compound 1 is administered as the HCl-salt, thus:
  • 60mg dose refers to 65mg Compound 1 • HC1 salt - prepared analogously to Example
  • Exploratory Objective Explore target engagement and pharmacodynamic response; clinical and histologic activity; and evaluate the relationship between safety, disease activity, exposure parameters and pharmacogenetics
  • TEAEs treatment-emergent adverse events
  • Exploratory Endpoint Changes in biomarkers of target engagement, pharmacodynamics, clinical activity scores, UC-100 Mayo Clinic Score and Robarts Histopathology Index; safety, disease activity and exposure parameters; and proportion of patients with histologic remission and/or mucosal healing at Day 28.
  • HPpCD hydroxypropyl-beta-cyclodextrin
  • the total duration of the study per patients is 13 weeks, including a 5-week screening period (7-35 days), the 4-week treatment period and a 4-week follow-up period.
  • Dose are administered orally in the morning at approximately the same time each day, with patients fasting for at least 6 hours before and 2 hours after each dose.
  • the first dose is administered on Day 1.
  • a blood sample is taken 8 hours post-dose.
  • Patients are assessed on Days 7, 14, 21 and 28, followed by a Follow-up (FU) visit four weeks later.
  • FU Follow-up
  • Patients are age 18-75, male or female with a BMI 18-35kg/m 2 . Patients do not have Crohn’s disease or indeterminate colitis, pouchitis, evidence of Clostridium difficile infection, a current malignancy or previous history of cancer or a history of alcohol or substance abuse. Patients exhibit the following disease criteria:
  • Mayo Endoscopy sub score of 0 or 1, or 0 if baseline Mayo Endoscopy subscore 1.
  • UC-100 Score The composite index (1 + 16 x Mayo Clinic Stool Frequency Subscore [0 to 3] + 6 x Mayo Clinic Endoscopic Subscore [0 to 3] + 1 x Robarts Histopathology Index Score [0 to 33]).
  • RHI Robarts Histopathology Index
  • Blood samples ( ⁇ 3mL) are collected on the Day 1 and 28 visits, 0.5, 1, 2, 3, 4, 6, and 8, 12, and 24 hours post-dose. Stool samples, when available, are collected on Study Days 14 and 28.
  • Clinical Laboratory Assessments include hematology (white blood cell count, red blood cell, hemoglobin, hematocrit, platelet count); clinical chemistries (alanine aminotransferase, albumin, aspartate aminotransferase, alkaline phosphatase, bicarbonate, bilirubin, blood urea nitrogen, calcium, creatinine, chloride , gamma-glutamyl transferase, glucose, L-lactate dehydrogenase, potassium, total protein, sodium; and urinalysis (dipstick, including macroscopic appearance, bilirubin, blood, color, glucose, ketones, leukocyte esterase, nitrite, pH, protein, specific gravity, urobilinogen).
  • clinical chemistries alanine aminotransferase, albumin, aspartate aminotransferase, alkaline phosphatase, bicarbonate, bilirubin, blood urea nitrogen, calcium, creatinine, chloride ,
  • EXAMPLE 14 A PHASE 1 A, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTIPLE DOSE STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF TABLET FORMULATIONS OF COMPOUND 1 IN HEALTHY MALE AND FEMALE
  • the primary objective of the study is to assess the safety and tolerability of tablet formulations of Compound 1 • HC1 after multiple oral doses.
  • the safety endpoints include incidence of treatment-emergent adverse events (TEAEs) and changes in laboratory, vital sign and ECG parameters.
  • the secondary objective is to evaluate the pharmacodynamic response of tablet formulations after multiple oral doses, with an endpoint of change in of target engagement biomarkers (e.g., CAIX).
  • Target engagement is assessed by measuring HIF-1 related gene expression and protein abundance in colonic tissue, stool and blood.
  • An exploratory objective of the study is to evaluate the effect of pharmacogenetics (PGx) on pharmacokinetic (PK) parameters of tablet formulations as compared to oral solution.
  • Pharmacokinetics assessments include evaluation of blood, stool and colonic biopsy samples.
  • FIG. 15 A schematic of the study design is presented in Figure 15. 40 subjects are randomized in the study, stratified by gender with each stratum comprising half of the study population. Thirty subjects are randomized to Compound 1 and 10 to placebo. Up to 8 replacement subjects may be added (up to 2 in the solution groups and up to 6 in the tablet groups; each replacement is the same gender and assigned to the same treatment group as the subject being replaced). Subjects are randomized to 8 groups, according to Table 35.
  • the study duration is up to 65 days, and includes the following periods:
  • Subjects are healthy males or females, age 18-65 with a Body Mass Index (BMI) 18-33 kg/m 2 . Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use, and did not display any significant colorectal symptoms or findings.
  • BMI Body Mass Index
  • Subject are dosed orally in the morning, at the same time each day, once daily for 7 days. Doses are administered as follows:
  • Solution Compound 1 is administered in lOOmL of a water 10% hydroxypropyl -beta- cyclodextrin (HPpCD) solution. Each solution dose is mixed with 100 mL purified water, followed by a 100 mL water “rinse” taken orally from the same bottle.
  • HPpCD hydroxypropyl -beta- cyclodextrin
  • Tablet Compound 1 - IR tablets prepared according to Example 5; DR tablets prepared according to Example 6. 60mg tablets IR, DR or matching placebo formulations are similar and indistinguishable in terms of appearance, odor and taste. Subjects randomized to receive 120mg, are administered two 60mg tablets; subjects randomized to receive 240mg, are administered four 60mg tablets.
  • Fed subjects are dosed on a fed stomach 30 minutes after the start of a standardized meal. Subjects are required to consume the meal in 30 minutes or less.
  • Physical Examinations include vital signs (pulse rate, respiratory rate, temperature and blood pressure) and electrocardiogram (ECG).
  • vital signs pulse rate, respiratory rate, temperature and blood pressure
  • ECG electrocardiogram
  • a flexible sigmoidoscopy is performed prior to dosing and on Day 7 to evaluate the endoscopic appearance of the colonic mucosa and obtain biopsies.
  • 6 biopsies are collected in each of the two segments (e.g., Rectum and Sigmoid) for approximately 12 biopsies.
  • Biopsies assess PK, immunohistochemical and gene expression signals (e.g., CAIX) of biological activity
  • Blood samples are collected on Days 1 and 7, at pre-dose and 0.25, 0.5, 1, 2, 3, 4, 6, and 8, 12, and 24 hours post-dose.
  • EXAMPLE 15 PHARMACOKINETICS OF FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
  • IR and DR tablet dosing 120 mg or 240 mg dose was compared to solution dosing in a 10% hydroxypropyl-beta-cyclodextrin (HPpCD) solution (120mg dose) according to the protocol described in Example 14 above.
  • HPpCD 10% hydroxypropyl-beta-cyclodextrin
  • the composition and method of manufacture of the IR and DR tablet compositions used in this study are disclosed in Examples 5 and 6 above.
  • Tmax values of the DR tablet were relatively longer than the other tablet treatment groups on Day 7.
  • all Compound 1 treated subjects displayed a quantifiable concentration by 0.5 hours post-dose.
  • AUCinf area under the concentration-time curve from zero to infinity
  • AUClast area under the concentration-time curve from zero to the last quantifiable concentration time point
  • Cmax maximum plasma drug concentration
  • CV coefficient of variation
  • NC coefficient of variation
  • b Median (range) c PK parameter could only be determined in one subject and thus, not included in summary statistics.
  • b Median (range) c could be determined in less than 3 subjects and thus, not included in summary statistics.
  • the glucuronide and glucoside metabolites were rapidly formed and appeared in plasma after oral administration of the solution and tablet formulations of Compound 1 (median T max of ⁇ 0.5 h for tablet formulations) and their exposure is approximately 235-fold and 38-fold higher than the parent Compound 1, for the glucuronide and glucoside metabolites respectively, on Day 7.
  • the highest C max observed in the Example 14 patients for the these metabolites was in the 240 mg IR tablet (fed) cohort and was 4.64 pM (about 2800 ng/ml) and 0.68 pM (about 400 ng/ml), for the glucuronide and glucoside metabolites respectively on Day 7.
  • the apparent half-life of both metabolites is approximately 8 to 12 h, which is longer than that of the parent ( ⁇ 2 h).
  • EXAMPLE 16 COLONIC TISSUE CONCENTRATIONS OF TABLET FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
  • IR and DR tablet dosing 120 mg or 240 mg dose was compared to solution dosing in terms of colon tissue (sigmoid and rectum) concentrations of Compound 1 according to the protocol described in Example 14 above.
  • Figure 17 shows the total drug colonic concentrations across both sections (sigmoid + rectum) for the different study subject cohorts.
  • EXAMPLE 17 SAFETY OF FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
  • IR and DR tablet dosing (120 mg or 240 mg dose) was compared to solution dosing in terms of adverse events reported according to the study protocol described in Example 14 above.
  • EXAMPLE 18 A PHASE 2, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTI-CENTER STUDY TO EVALUATE COMPOUND 1 IN ADULT SUBJECTS WITH MILD-TO-MODERATE ACTIVE ULCERATIVE COLITIS
  • PHDi Small molecule prolyl hydroxylase inhibitors
  • TNBS trinitrobenzene sulfonic acid
  • DSS dextran sulfate sodium
  • TNFa tumor necrosis factor alpha
  • UC ulcerative colitis
  • Compound 1 a HIF-la stabilizer, is a potential oral therapeutic, gut-targeted treatment for UC with higher intestinal than systemic exposure.
  • This Phase 2 study assesses the efficacy and safety of Compound 1 when added to background UC therapy of 5 -aminosalicylate (5 -ASA) with or without steroids.
  • 5 -aminosalicylate 5 -aminosalicylate
  • the primary objective is to evaluate the effect of Compound 1 compared to placebo on clinical remission.
  • the primary endpoints are defined as the proportion of subjects with clinical remission at Week 12, defined as a Modified Mayo score ⁇ 2, with a rectal bleeding subscore of 0, stool frequency subscore of 0 or 1 (with a > 1 point decrease from baseline), and endoscopy subscore of 0 or 1. 18,3 Secondary Objective
  • the secondary objective is to evaluate the effect of Compound 1 on clinical response and endoscopic improvement.
  • the secondary endpoints include:
  • the study also has a safety objective related to monitoring the incidence of treatment-emergent adverse events (TEAEs).
  • TEAEs treatment-emergent adverse events
  • Total duration for study participation per subject is expected to be up to 20 weeks: 2-4 week screening period; 12 week treatment period; and 4 week follow-up period.
  • Blood plasma sample (PK) - treatment (weeks 0, 4 & 12). Blood samples will be collected pre-dose, and 0.5, 2 and 4 hours post-dose. The timepoints are in reference to the morning dose;
  • Blood sample (PD) - treatment (weeks 0, 2, 4, 8 & 12). Blood samples will be collected twice (pre-dose vs post-dose) as EDTA plasma and twice (pre-dose vs post-dose), as PaxGene. The timepoints are in reference to the morning dose;
  • Each dose of Investigational Product will consist of a combination of Compound 1 immediate-release tablets and Compound 1 delayed-release tablets or of matching placebo and will be taken orally BID on a fed stomach at approximately the same time each day, 8 to 12 hours apart.
  • the Compound 1 immediate-release tablets, Compound 1 delayed-release tablets, and placebo tablets are similar and indistinguishable in terms of appearance, odor, and taste, such that the blind is maintained.
  • the dose formulation is round film-coated tablets containing 60 mg of Compound 1 per tablet.
  • Placebo the dose formulation is round film-coated tablets matching the active formulation.
  • the tablets are taken orally on a fed stomach, 30 minutes after a meal.
  • the IP is taken BID (approximately 8 to 12 hours apart) at approximately the same time each day.
  • the diagnosis should be confirmed by clinical and endoscopic evidence and corroborated by a histopathology report (note: the screening endoscopy and histopathology results may be used to confirm the diagnosis if no prior report is readily available).
  • Contraception use by men or women should be consistent with local regulations regarding the methods of contraception for those participating in clinical studies.
  • Anti-diarrheal medications e.g., loperamide, diphenoxylate/atropine
  • NSAID non-steroidal anti-inflammatory agents
  • CYP3 A inducers e.g. oceprevir, cobicistat, conivaptan, danoprevir, ritonavir, elvitegravir, grapefruit juice, indinavir, itraconazole, ketoconazole, lopinavir, paritaprevir, ombitasvir, dasabuvir, posaconazole, saquinavir, telaprevir, tipranavir, troleandomycin, voriconazole, clarithromycin, diltiazem, idelalisib, nefazodone, nelfinavir, carbamazepine, enzalutamide, mitotane, phenytoin, rifampin, St. John’s wort) within 21 days or within 5 half-lives, whichever is longer, of the respective medication prior to Day 1.
  • oceprevir cobicistat, conivaptan, dan
  • Known pre-existing, unstable liver disease as defined by the presence of ascites, encephalopathy, coagulopathy, hypoalbuminemia, esophageal or gastric varices or persistent jaundice), cirrhosis, known biliary abnormalities, known pre-existing non-alcoholic steatohepatitis (NASH), or active infection with Hepatitis B or Hepatitis C.
  • NASH non-alcoholic steatohepatitis
  • Immunodeficiency including that due to human immunodeficiency virus (HIV), other than that explained by systemic corticosteroid use.
  • HAV human immunodeficiency virus
  • QTcF > 450 msec for males or QTcF > 470 msec for females at screening by central overread. If QTcF is above specified limit and there are no other clinically significant abnormalities, repeat assessment is allowed with permission by the Sponsor’s Medical Monitor (or designee).
  • ALT Alanine aminotransferase
  • AST aspartate aminotransferase
  • Subjects should remain on their stable UC background therapy, consisting of oral 5-ASA with or without prednisone ⁇ 20 mg/day (or equivalent) or budesonide or budesonide MMX of ⁇ 9 mg/day, from 2 weeks prior to screening flexible sigmoidoscopy/colonoscopy used to assess the baseline Mayo score and for the duration of their time on study.
  • Additional UC therapies may not be started between 2 weeks prior to the screening flexible sigmoidoscopy/colonoscopy to the Week 16 Follow-up visit. Subjects requiring introduction of new therapies to treat UC will be discontinued from IP.
  • Screening and Week 12 endoscopy subscores will be calculated by both a blinded central reader and a blinded local endoscopist. Differences between the 2 readers will be adjudicated by a second blinded central reader as described in the Imaging Charter. Centrally read endoscopic subscores will be used for both eligibility determination and efficacy analyses. Histologic scores (eg, RHI) will be evaluated by a blinded central reviewer.
  • Histologic scores eg, RHI
  • Flexible sigmoidoscopy/colonoscopy and histology assessments will consist of assessing the Mayo endoscopic subscores of the sigmoid colon at approximately 15 to 25 cm from the anal verge. Six biopsies will be obtained during the endoscopic procedures from the area that appears to be most affected by UC in this sigmoid colon segment.
  • Biopsies will be evaluated for PK, gene expression, and immunohistochemistry signals of biological activity. Mayo Score and Modified Mayo Score
  • Mayo score is used for inclusion into the study rather than a Modified Mayo score (used to evaluate efficacy) given the limited information available at the study inception regarding Modified Mayo score cut points that define a mild-to-moderate patient population.
  • the Mayo score is a conventional assessment of UC disease activity and is commonly used in clinical studies of UC.
  • the score is composed of four subscores: Stool frequency, Rectal bleeding, Physician Global Assessment (PGA), and Endoscopy (where the value of 1 does not include friability), each rated from 0-3, that are summed to give a total score ranging from 0 to 12 points (higher scores indicating greater severity).
  • the Modified Mayo score is an endpoint measure composed of three subscores: Stool frequency, Rectal bleeding, and Endoscopy (where the value of 1 does not include friability), each rated from 0 to 3, that are summed to give a total score ranging from 0 to 9 points (higher scores indicating greater severity).
  • the Partial Mayo score is an endpoint measure composed of three subscores: Stool frequency, Rectal bleeding, and PGA each rated from 0 to 3, that are summed to give a total score ranging from 0 to 9 points (higher scores indicating greater severity).
  • the 6-point Mayo score is an endpoint measure composed of two sub scores: Stool frequency and Rectal bleeding each rated from 0 to 3, that are summed to give a total score ranging from 0 to 6 points (higher scores indicating greater severity).
  • the RHI is a validated instrument that measures histological disease activity in UC.
  • the RHI consists of 4 items: chronic inflammatory infiltrate, lamina intestinal neutrophils, neutrophils in epithelium, and erosion or ulceration.
  • the RHI score ranges from 0-33 and is derived from Geboes score (Mosli et al., Gut (2017), 50-58).
  • the Geboes score is divided into 6 grades: architectural changes [grade 0], chronic inflammatory infiltrate [grade 1], lamina limba neutrophils and eosinophils [grade 2], neutrophils in epithelium [grade 3], crypt destruction [grade 4] and erosions or ulcerations [grade 5],
  • the Geboes score is calculated as the highest grade with a corresponding subgrade score > 0, excluding Geboes Grade 2 A, and ranges from 0 to 5.
  • the Ulcerative Colitis Index- 100 (UC-100) score is the composite index (1 + 16 x Stool frequency sub score [0 to 3] + 6 x Endoscopy sub score [0 to 3] + 1 x RHI score [0 to 33]) (Jairath et al., Lancet Gastroenterol Hepatol. (2019), 63-70).
  • the IR pellets comprise an inert core coated with a drug layer.
  • Inert core Sugar spheres mesh 45/60 (250-355 microns);
  • Drug layer Compound 1 (free base) - active agent
  • HPMC E5 Hydroxypropyl methyl cellulose
  • HPMC E5 42.68 g
  • poloxamer 188 3.05 g
  • PEG 6000 1.22 g
  • HPMC E5 42.68 g
  • poloxamer 188 3.05 g
  • PEG 6000 1.22 g
  • Compound 1 Free Base 75.0 g was added and the mixture stirred for 30 min to give a uniform dispersion (pH of dispersion in the range 6-8).
  • the coated beads were dried at 40°C inlet temperature at reduced fluidization.
  • Example 19 Coating the beads with Compound 1 free base gave improved uniformity of drug coating compared to coating with the HC1 salt (Example 20).
  • Example 19 pellets have the composition according to Table 42:
  • Example 20 IR pellets are prepared analogously to the Example 19 IR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base.
  • Example 20 pellets have the composition according to Table 43:
  • the DR pellets comprise an inert core coated with a drug layer.
  • the drug layer coated core is then covered with a seal coating and a delayed release coating.
  • Inert core Sugar spheres mesh 45/60 (250-355 microns)
  • Drug layer Compound 1 (free base) - active agent
  • HPMC E5 Hydroxypropyl methyl cellulose
  • Delayed release coat Eudragit® L 30 D 55 - pH 5.5 coat
  • the drug-layered core is prepared according to the fluidized bed spray coating process outlined above for Example 19, using the amounts listed in Table 40.
  • a seal coat is then applied to the drug-layered core, wherein HPMC E5 (18 g) dissolved in purified water (207 g) is spray coated on to drug-layered beads (600 g) using a GPCG 2 (Glatt) fluidized bed Labsystem under the following conditions in Table 44: TABLE 44: FLUIDIZED BED DRUG LAYERING CONDITIONS
  • the coated beads are dried at 40°C inlet temperature at reduced fluidization.
  • the drug layered pellets are thereby seal-coated to a 3% weight gain.
  • a delayed release coat is then applied as follows: Eudragit® L 30 D 55 (105 g) and PlasAcryl® T20 (10.5 g) are dissolved in purified water (461 ml) and then spray coated on to the seal-coated beads (350 g) using a GPCG 2 (Glatt) fluidized bed Labsystem under the following conditions in Table 45:
  • the coated beads are dried at 40°C inlet temperature at reduced fluidization and atomization air pressure.
  • pellets are thereby coated with a pH 5.5 delayed release coating to a 30% weight gain to give a pellet composition according to Table 46:
  • Example 22 DR pellets are prepared analogously to the Example 21 DR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base.
  • Example 23 seal-coated pellets were subjected to dissolution testing to determine the rate of Compound 1 release from the composition.
  • Fig. 19A shows the dissolution rate of Compound 1 from seal-coated pellets (60 mg compound) encapsulated in HPMC capsules according to USP2 dissolution testing for 10 hours in pH 7.4 phosphate buffer with 2% CTAB. Almost 90% drug release had occurred within 2 hours.
  • Example 23 pellets have the composition according to Table 47.
  • Example 23 DR pellets were subjected to dissolution testing to determine the rate of Compound 1 release from the composition.
  • Fig. 19B shows the dissolution rate of Compound 1 from Example 23 pellets (60 mg compound) coated with 20% Eudragit®® FS 30 D and encapsulated in HPMC capsules according to USP2 dissolution testing for 2 hours in pH 1.2 aqueous media followed by 10 hours in pH 7.4 phosphate buffer with 2% CTAB. Less than 5% drug release has occurred after 2 hr in acid, however, after switching to pH 7.4 almost 90% drug release occurred within 1 hour.
  • Example 24 DR pellets are prepared analogously to the Example 23 DR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base, to yield a multiparticulate composition according to Table 48.

Abstract

The present invention relates to methods of treating inflammatory bowel diseases comprising the administration of a combination of certain pharmaceutical compositions containing tert-ButyI-4-{[1-(4-chlorobenzyl)-3-hydroxy-2-oxo-1,2-dihydropyridin-4-yl]methyl}piperazine-1-carboxylate (hereinafter also referred to as "Compound 1") or a pharmaceutically acceptable salt thereof. Also provided are certain oral modified release pharmaceutical compositions comprising an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof, as well as methods and uses related to said modified release pharmaceutical composition and to certain dosage regimens for treating inflammatory bowel diseases.

Description

COMPOSITIONS AND METHODS OF TREATMENT
[0001] The present invention relates to the combination of certain pharmaceutical compositions containing tert-butyI-4-{[l-(4-chlorobenzyl)-3-hydroxy-2-oxo-l,2-dihydropyridin- 4-yl]methyl] piperazine- 1 -carboxylate (hereinafter also referred to as “Compound 1”) or a pharmaceutically acceptable salt thereof for use in the treatment of inflammatory bowel diseases; certain oral modified release pharmaceutical compositions comprising an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof; and to certain dosage regimens for treating inflammatory bowel diseases.
BACKGROUND
[0002] Inflammatory bowel disease (IBD) is characterized by repeated wounding and inflammation of the intestinal mucosa and typically involves a breach in intestinal barrier integrity, which allows the influx of luminal antigens and sets up a vicious cycle of inflammation and epithelial injury. During mucosal inflammation, changes in metabolic activity and vascular tissue damage lead to a reduction in tissue-oxygen tension (hypoxia), in which healing processes such as angiogenesis, cell migration, and re-epithelialization occur. Typically it is the epithelial cells on the surface of mucosal tissue that enter an induced state of hypoxia due to the presence of inflammation and the body's response to this hypoxic condition is to increase the presence of hypoxia inducible factor-1 alpha (HIF-la), which subsequently drives the expression of downstream HIF-1 target genes, inter alia, erythropoietin. As such, HIF-la is an important mediator in the body's response to inflammation.
[0003] The cellular concentration of HIF-la is regulated by prolyl hydroxylase (PHD) enzymes that serve to destabilize HIF-la during periods of normoxia resulting in the destruction of this protein. Inhibition of HIF-la prolyl hydroxylase thus leads to increased stabilization of HIF-la resulting in a up regulation of HIF-la which in turn leads to a corresponding increased response to inflammation. In subjects suffering from one or more inflammatory epithelial diseases, treatment with effective HIF-la prolyl hydroxylase inhibitors can increase the level of the body's cellular inflammatory response. In addition, during periods of low level inflammation in the case of chronic diseases, HIF-la prolyl hydroxylase inhibitors can increase the amount of epithelial cell healing over that which the body would normally provide. [0004] Compound 1 is a potent inhibitor of PHD and is selective for PHD-2 over PHD-1, resulting in specific stabilization of HIF-la and reduced risk of off-target effects. The potential benefit of Compound 1 in the treatment of IBD is based on inhibition of the degradation of hypoxia inducible factor- 1 alpha (HIF-la) and sustained hypoxia inducible factor- 1 (HIF-1)- driven gene expression.
[0005] Compound 1 is disclosed in International Patent Application WO 2011/057121 and has the following structure:
Figure imgf000004_0001
Compound 1
[0006] In order for a compound such as Compound 1 to provide optimal treatment for inflammatory bowel diseases, it is important that sufficiently high concentration levels of the compound are achieved at the site of action, i.e. the colonic tissue. Based on non-clinical animal models of colitis, colonic tissue concentrations of at least 140ng/g may be expected to provide optimal levels for efficacy.
[0007] Whilst Compound 1 has been found to be highly potent and effective in pre- clinical models of IBD, the unique physicochemical and biopharmaceutical properties of the molecule present particular challenges when trying to optimize colonic tissue levels through oral administration.
[0008] Compound 1 has a low solubility/dissolution rate. Furthermore, the solubility of Compound 1 is highly dependent upon pH. For example, Compound 1 is soluble at pH 1 but is practically insoluble above pH 3, whilst some solubility is regained at very high pH values (i.e. above 10). Compounds which have pH-dependent solubility, particularly basic compounds, may exhibit undesirable pharmacokinetic properties such as problems in their absorption, possibly producing low or variable bioavailability between patients and between doses.
[0009] A factor which can affect the absorption of an orally administered drug is the changing pH experienced by the drug as it passes through the GI tract. Typically a drug may be absorbed in a number of different sites along the GI tract following oral administration; for example, the cheek lining, stomach, duodenumjejunum, ileum and colon. The pH may be different at each site of absorption with the pH significantly different from the stomach (pH 1- 3.5) to the small intestine (pH 4-8). When the solubility of a drug varies with pH the drug may precipitate from solution as it passes through the GI tract. This can result in variability in the extent and/or rate of absorption between doses and between patients, because the drug needs to be in solution to be absorbed.
[0010] The Applicants have identified a particular hydrogen chloride salt form of Compound 1 with improved solubility properties, however, the salt is liable to dissociation into it's free-base form during formulation processing and/or storage. Such conversion can impact solubility and dissolution rate but also has consequences for the chemical stability of the compound. Unusually, the agent contains a tert-butyl oxy carbonyl (Boc) group and this group is liable to chemical degradation in the presence of an acidic environment.
[0011] The Applicants have found that stability of the hydrochloride salt can be maintained in certain immediate release granule-based compositions. When administered at a daily dose of 120 mg and 240 mg, the compositions surprisingly provide systemic levels and colonic tissue concentrations comparable to HP-PCD formulations containing the compound in solubilized form. Upon administration, the pharmaceutically acceptable salt of Compound 1 is efficiently released from the granule based immediate release tablet dosage form and absorbed across the gastro-intestinal tract to provide a rapid increase in plasma concentration. For example, after oral administration of the immediate release formulation described in Example 5 at a dose of 120 mg to healthy human subjects in the fasted state, the geometric mean maximum plasma concentration (Cmax) achieved is approximately 8.85 ng/ml and the time at which the peak plasma concentration is observed (Tmax) is in the range of approximately 0.5-1.0 hour. Following the Cmax, the plasma concentrations of Compound 1 falls to less than approximately 5% of the Cmax within 24 hours. The area under the plasma concentration-time curve from time zero to the last quantifiable concentration time point (AUCiast) is approximately 16.9 ng hr/mL.
[0012] Whilst the immediate release composition provides colonic tissue exposure, simply increasing dose levels to further improve tissue concentration levels may not be desirable. Any dose increase requires careful consideration to ensure any unintended safety or tolerability related issues do not occur. For example, previous studies have demonstrated that high systemic levels of PHD inhibitors can potentially influence EPO and/or VEGF regulation. [0013] As a potential alternative, the Applicants have also surprisingly found that certain delayed release compositions comprising Compound 1 or a pharmaceutically acceptable salt thereof provide targeted release and biopharmaceutical performance without requiring high systemic levels. Despite having a distinct systemic PK profile, certain delayed release compositions achieve similar colonic tissue concentration profiles to the above-mentioned immediate release granule composition and to a HP-PCD solution formulation. However, tissue concentration levels at the higher dose of 240mg were found to be broadly equivalent to those achieved at 120mg. Hence, increasing dose did not result in increased tissue concentrations.
[0014] There remains a need for an improved approach to achieve higher colonic tissue levels in order to maximize efficacy of Compound 1 but in a safe and tolerable way.
SUMMARY OF INVENTION
[0015] Surprisingly, the Applicants have found that administration of a combination of a solid oral immediate release pharmaceutical composition and a solid oral delayed release pharmaceutical composition offers the potential to provide high gastrointestinal exposure with acceptable safety and/or tolerability.
[0016] The Applicants have found that upon oral administration of Compound 1, for example from an immediate release composition, the compound is rapidly absorbed and metabolized in the liver yielding a glucuronide conjugate. This conjugate is subsequently subjected to biliary excretion into the intestine and it is expected that certain levels of the glucuronide conjugate pass through the gut lumen where P-glucuronidase enzymes hydrolyze the glucuronide back into the parent compound. The parent drug can be distributed to the intestinal tract tissues allowing absorption into the colonic tissue. It is believed that other conjugates such as glycoside conjugates are also formed and could contribute to the increase in colonic tissue levels. The unique contribution of this enterohepatic recirculation from an immediate release pharmaceutical composition, along with the luminal drug levels provided by a delayed release composition, allows particularly high colonic tissue exposure to be achieved with acceptable safety and tolerability.
[0017] Furthermore, the Applicant has found that there are particular benefits when a single oral modified release pharmaceutical composition comprising an immediate release component and a delayed release component (wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof) is used to achieve particularly high colonic tissue exposure combined with acceptable safety and tolerability.
[0018] In a first aspect, the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof. In a related aspect, there is provided a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof for use in treating an inflammatory bowel disease.
[0019] In a second aspect, the present invention provides an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof. Such a composition may be typically a matrix dosage form, or a multiparticulate dosage form. Suitable multiparticulate dosage forms include subunits such as mini-tablets, beads, pellets, and granules. Such multiparticulates may be packaged, for example, in a capsule; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet or pill by compression or other processes known in the art. In a convenient embodiment, mini-tablets have been found to be particularly suitable for the oral modified release pharmaceutical compositions of the present invention. The Applicants have found that surprisingly it has been possible to achieve much higher drug loadings (greater than 50% w/w) of Compound 1 in both the immediate release component and the delayed release component of mini-tablet formulations.
[0020] In a third aspect, the present invention provides methods of treating diseases or conditions mediated alone, or in part, by PHD (such as inflammatory bowel disease), comprising the administration of the combination according to the first aspect, or the modified release composition according to the second aspect. BRIEF DESCRIPTION OF THE DRAWINGS
[0021] The patent or application file contains at least one drawing executed in color.
Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
[0022] FIG. l is a graphical plot of Compound 1 solubility (logio ng/mL) vs pH at 22°C, as described in Example 1.
[0023] FIG. 2 is a Flow diagram for a representative Compound 1 • HC1 High Shear Granulation Manufacturing Procedure.
[0024] FIG. 3 is a Flow diagram for the manufacturing process for Compound 1 • HC1 IR
Tablets as described in Example 5.
[0025] FIG. 4 is a Flow diagram for the manufacturing process for Compound 1 • HC1
DR Tablets as described in Example 6.
[0026] FIG. 5 is a Comparison of dissolution rates for Compound 1 from 60mg IR and
DR Tablets, as described in Example 7.
[0027] FIG. 6 is a Flow diagram for the Compound 1 • HC1 High Shear Granulation
Manufacturing Procedure used for Formulations F9-F12.
[0028] FIG. 7 is a graphical plot of Compound 1 mean plasma concentration, over time 0-16hr for the clinical trial described in example 11 (all subjects).
[0029] FIG. 8 is a graphical plot of Compound 1 mean plasma concentration, over time 0-16hr for the clinical trial described in example 11 (without vomiting subjects).
[0030] FIG. 9 is a graphical representation of the Study Scheme for the clinical trial described in example 12.
[0031] FIG. 10 is a graphical plot of Compound 1 mean plasma concentration, over time
0-25hr for the clinical trial described in example 12, on day 1.
[0032] FIG. 11 is a graphical plot of Compound 1 mean plasma concentration, over time
0-25hr for the clinical trial described in example 12, on day 7.
[0033] FIG. 12 shows the box plot of the proportions of HIF-la positive cells evaluated at baseline and Day 8.
[0034] FIG. 13 is a graphical representation of the Study Scheme for the clinical trial described in example 13. [0035] FIG. 14 is a graphical representation of the Study Scheme for the clinical trial described in Example 14.
[0036] FIG. 15 is a graphical plot showing Compound 1 mean plasma concentrations following single (day 1) and multiple (day 7) dosing of: (top) 120mg solution, 120mg immediate release tablet and 120mg & 240 mg delayed release tablets in fasted subjects; and (bottom) 120 mg immediate release tablet in fasted subjects and 240 mg immediate release tablet in fed subjects; over time 0-24hr for the clinical trial described in Example 14.
[0037] FIG. 16 is a graphical plot showing Compound 1 glucuronide metabolite mean plasma concentrations following [left] single (day 1) and [right] multiple (day 7) dosing of: 120 mg solution (white circles), 120 mg IR tablet fasted (white squares), 120 mg DR tablet fasted (black circles), 240 mg IR tablet fed (white triangles) and 240 mg DR tablet fasted (black squares); over time 0-24hr for the clinical trial described in Example 14.
[0038] FIG. 17 is a graphical plot of median colon tissue concentrations (rectum - left; sigmoid - right) for the dosing cohorts in the clinical trial described in Example 14.
[0039] FIG. 18 is a plot of Compound 1 total colon concentrations for the dosing cohorts in the clinical trial described in Example 14.
[0040] FIG. 19 shows USP2 Dissolution rates of Compound 1 from Example 23 pellets: (A) seal-coated pellets in pH 7.4 phosphate buffer with 2% CTAB; and (B) 20% Eudragit® FS 30 D coated pellets in pH 1.2 aqueous media for 2 hr followed by pH 7.4 phosphate buffer with 2% CTAB.
[0041] FIG. 20 shows USP2 Dissolution rates of Compound 1 from Example 29 capsules containing 1 :1 IR/DR mini -tablets.
DETAILED DESCRIPTION
Definitions
[0042] Immediate Release’ or ‘IR’ as used in the present application in its conventional sense refers to a dosage form that provides for release of a compound immediately after administration. For example, an immediate release formulation means a formulation in which the dissolution rate of the drug from the formulation is 85 % or more after 10 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of an appropriate test fluid (such as 0.01N hydrochloric acid) is used and the paddle rotation speed is 75 rpm. Conveniently, ‘Immediate Release’ or ‘IR’ as used in the present application refers to a formulation or composition which rapidly disintegrates and disperses to release a drug compound after oral administration to a subject. More conveniently, it refers to a formulation or composition for which the dissolution rate of the drug from the formulation is 85 % or more after 10 minutes (such as after 5 minutes) from the beginning a dissolution test, which is carried out in accordance with a USP 2 dissolution test (paddle method) under the conditions that 900 mL of 0.01N hydrochloric acid is used, the temperature is 37°C and the paddle rotation speed is 75 rpm. In terms of in vivo pharmacokinetics, immediate release refers to a formulation or composition which typically provides systemic (plasma) levels of active pharmaceutical agent shortly after oral dosing. More conveniently, it refers to a formulation or composition which provides a geometric mean maximum plasma concentration (Cmax) of the compound after oral dosing of at least 5 ng/ml, such as at least 10 ng/ml within at least 120 minutes, conveniently within at least 60 minutes or 30 minutes.
[0043] ‘Delayed Release’ or ‘DR’ as used in the present application refers to a dosage form that provides for release of a compound after administration at a slower rate than that from an immediate release formulation, or release of the active compound starts at a later point in time compared with an immediate release composition (such as at 30 min or more later such as, e.g., 1 hour or more later or 2 hours or more later or 3 hours or more later than an immediate release composition). Conveniently, ‘Delayed Release’ or ‘DR as used in the present application refers to a formulation or composition which, after oral administration to a subject, does not undergo disintegration and dissolution in the acidic environment of the stomach. In terms of in vitro dissolution testing, delayed release refers to a formulation or composition which typically undergoes less than 5% dissolution in acidic aqueous media. More conveniently, it refers to a formulation or composition in which the dissolution rate of Compound 1 or a pharmaceutically acceptable salt thereof from the formulation is 5 % or less (such as 2% or less) after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a USP 2 dissolution test (paddle method) under the conditions that 900 mL of 0.01N hydrochloric acid is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm. [0044] The term ‘combination’ as used herein refers to compositions being administered concurrently (for example, simultaneously, essentially simultaneously or within the same treatment protocol), sequentially or separately. However, the compositions need not be administered simultaneously or essentially simultaneously. Thus the immediate release composition may be administered first, followed by the delayed release composition, or the delayed release composition may be administered first, followed by the immediate release composition. When the combination is dosed once daily, the compositions of the invention are dosed within the same 24 hour period, preferably within the same 12 hour, 6 hour, 3 hour, 2 hour or 1 hour period. When the combination is dosed twice daily, the compositions of the invention are dosed within the same 12 hour period, preferably within the same 6 hour, 3 hour, 2 hour or 1 hour period. When the combination is administered with food, the compositions of the invention are dosed within the same 1 hour period, preferably within the same 30 minute period, after the patient has a meal. Conveniently, the term ‘combination’ as used herein means the compositions are administered simultaneously or essentially simultaneously, more conveniently, simultaneously.
[0045] Granulate' as used in the present application refers to a granular material, which is the result of the agglomeration of fine particles into larger granules. The granules may be packaged, for example, in a capsule for administration; dosed as a suspension or slurry in a liquid; dosed in a sachet; formed into pellets or beads by processes known in the art; or they may be formed into a tablet (e.g. a caplet), mini-tablet, micro-tablet or pill by compression or other processes known in the art. Granules may typically be sized in the range 0.1 to 2.0 mm. Conveniently, the granules may have an average particle size in the range 0.2 to 0.5 mm. Conveniently, the granules are formed into a mini-tablet or micro-tablet, most conveniently mini-tablets. Mini-tablets may typically be sized in the range 1 to 4 mm. Conveniently, minitablets are less than 3 mm in diameter, such as between about 2 and 3 mm in diameter, between about 2.2 and 2.8 mm in diameter, between about 2.3 and 2.7 mm in diameter, between about 2.4 and 2.6 mm in diameter, or about 2.5 mm in diameter.
[0046] The term ‘pharmaceutically acceptable excipient’ is used herein to refer to an essentially pharmacologically inert, non-toxic substance, e.g. that has been approved for inclusion in pharmaceutical products. Examples of excipients classes includes fillers and diluents, binders, disintegrants, surfactants, wetting agents, lubricants, preservatives, colorants, flavouring agents, sweeteners and coatings.
[0047] As used herein, the term 'intimate association' refers to at least two components which are intimately mixed together. Compound 1 has a propensity to self-agglomerate in aqueous media such as pH 6.8 buffer. It has been found that co-processing of Compound 1 or a pharmaceutically acceptable salt thereof and one or more excipients (such as a wetting agent, in some cases even at very low levels) results in these components being intimately associated with one another leading to improved dispersion, and it is postulated that surface-to-surface interactions between particles of the Compound 1 or a pharmaceutically acceptable salt thereof are reduced in the intimate mixture such that dispersal is improved on exposure to aqueous media.
Compound 1
[0048] Compound 1, as referred to in the present invention, is tert-butyl 4-((l-(4- chlorobenzyl)-3-hydroxy -2-oxo- 1 ,2-dihydropyridin-4-yl)methyl)piperazine- 1 -carboxylate, which has the following structure:
Figure imgf000012_0001
[0049] As depicted above, Compound 1 is shown as a "free base". In some embodiments, a pharmaceutically acceptable salt of Compound 1 is used. Conveniently, a HC1 salt of Compound 1 (in a 1 : 1 molar ratio) is used. The HC1 salt of Compound 1 is depicted below:
Figure imgf000012_0002
[0050] The molecular weight of Compound 1 is 433.93. The molecular weight of the HC1 salt of Compound 1 is 470.39. 1.0g of Compound 1 • HC1 salt contains 0.92g Compound 1 free base. 1.08g Compound 1 • HC1 salt contains 1.0g Compound 1 free base:
Compound 1 • HC1 MW = 470.39 1.0g 1.08g Compound 1 MW = 433.93 0.92g 1.0g
HC1 MW = 36.46 0.08g 0.08g
[0051] Throughout the specification, unless specified otherwise, references to the amount of Compound 1 will be understood to refer to the amount of the parent compound (free base equivalent), even if the compound is present as a salt of Compound 1. Purely by way of example, reference to 120 mg of Compound 1 or a salt thereof, will be understood to refer to 120 mg of the free base, or a salt of Compound 1 with 120 mg of free base equivalent; in the context of the anhydrous mono-hydrochloride salt of Compound 1, 130 mg of the salt provides 120 mg of Compound 1 (free base equivalent); in the context of the monohydrate mono-hydrochloride salt of Compound 1, 135 mg of the salt provides 120 mg of Compound 1 (anhydrous free base equivalent).
Immediate Release (IR) Compositions
[0052] In a first aspect the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof. In a related aspect, there is provided a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof for use in treating an inflammatory bowel disease.
[0053] A pharmaceutically acceptable salt of Compound 1 is used in the oral immediate release pharmaceutical composition.
[0054] In a particular embodiment, the oral immediate release pharmaceutical composition is an immediate release solid pharmaceutical composition for oral administration comprising a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 : and one or more pharmaceutically acceptable excipients.
[0055] The Applicants have surprisingly found that stability of a salt form of Compound 1 can be maintained in certain granule-based compositions and dissociation into the free-base form and/or chemical degradation of the compound can be avoided or reduced. Certain granule-based compositions display improved disintegration and dissolution properties leading to good absorption and/or bioavailability upon dosing. Certain immediate release solid compositions have been found to give comparable systemic exposure of Compound 1 after 7 days of oral dosing as observed with an equivalent dose formulated as a HP-PCD solution (see Example 15). Furthermore, certain immediate release compositions have been shown to deliver higher colonic tissue levels of Compound 1 than were achieved via oral solution dosing (see Example 16). Certain compositions also possess improved chemical stability, powder flow and compression properties.
[0056] The compound used in the granulate is a pharmaceutically acceptable salt of Compound 1. Suitable pharmaceutically acceptable salts include acid-addition salts of the basic piperazine nitrogen in Compound 1 and also metal salts of the weakly acidic hydroxyl group in Compound 1.
[0057] Acid-addition salts include salts with inorganic or organic acids. Inorganic acid salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid salts. Organic acid salts include trifluoroacetic, acetic, formic, citric, maleic, succinic, lactic, glycolic, tartaric, methanesulfonic and -toluenesulfonic acid salts. Conveniently, the pharmaceutically acceptable salt is an inorganic acid salt. More conveniently, the pharmaceutically acceptable salt is a hydrochloric acid salt.
[0058] Metal salts include alkali metal salts and alkali earth metal salts, such as sodium, potassium, calcium or magnesium salts. Conveniently, the pharmaceutically acceptable salt is a calcium salt.
[0059] In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt. In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt and the ratio of Compound 1 to HC1 is about 1 : 1.
[0060] In an embodiment, the hydrochloride salt is a hydrate. In a convenient embodiment, the hydrochloride salt is a monohydrate. In a convenient embodiment, the hydrochloride salt is a crystalline monohydrate, which is characterized by X-Ray (Cu K radiation in transmission mode using 40 kV / 40 mA generator settings) diffraction peaks at 15.1, 17.4, 19.8, 20.0 and 20.6 ± 0.2 degrees 29 (Form A). In a convenient embodiment, the hydrochloride salt is a crystalline monohydrate, which is characterized by a melting point of 191-194 °C. [0061] In an embodiment, the hydrochloride salt is anhydrous. In a convenient embodiment, the hydrochloride salt is a crystalline anhydrous compound, which is characterized by X-Ray (Cu K radiation in transmission mode using 40 kV / 40 mA generator settings) diffraction peaks at 9.0, 15.2, 16.8, 18.6 and 20.3 ± 0.2 degrees 29 (Form B). In a convenient embodiment, the hydrochloride salt is a crystalline anhydrous compound, which is characterized by a melting point of 195-198 °C.
[0062] In an embodiment, the granulate comprises a mixture of the hydrochloride salt as a crystalline monohydrate and the hydrochloride salt as an amorphous compound. Analysis of the physical form of Compound 1 in the granulate may be carried out by techniques known to the person of skill in the art, e.g. XRPD, FTIR, Raman, or solid-state NMR. In a particular embodiment, the granulate comprises a mixture of the Compound 1 hydrochloride salt as a crystalline monohydrate and the Compound 1 hydrochloride salt as an amorphous compound, wherein the amount of amorphous compound present is less than 10% by weight of the total amount of Compound 1 hydrochloride salt present in the granulate. Conveniently, the amount of amorphous compound present is less than 9% (such as less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, or less than 1%) by weight of the total amount of Compound 1 hydrochloride salt present in the granulate.
[0063] In an alternative embodiment, the granulate comprises a mixture of the hydrochloride salt as a crystalline monohydrate (Form A) and the hydrochloride salt as a crystalline anhydrous (Form B) compound.
[0064] In an embodiment, the one or more pharmaceutically acceptable excipients comprise a wetting agent. It is postulated that when a wetting agent is intimately mixed with the pharmaceutically acceptable salt of Compound 1 within a granulate composition, the disintegration of the composition and dispersal of the pharmaceutically acceptable salt of Compound 1 in aqueous media is promoted. In an embodiment, the pharmaceutically acceptable salt of Compound 1 is in intimate association with the wetting agent in the granulate. In an embodiment, the pharmaceutically acceptable salt of Compound 1 is in intimate association with the wetting agent and one or more additional pharmaceutically acceptable excipients (optionally selected from disintegrant, binder and diluent).
[0065] The wetting agent is typically a surfactant or an emulsifier. In a convenient embodiment, the wetting agent is a non-ionic surfactant. Non-ionic surfactants can be characterized according to their hydrophilic-lipophilic balance (HLB). HLB values are commonly used to define emulsifiers and/or surfactants and refer to the hydrophilic-lipophilic balance of the given compound. HLB values can be calculated according to the methods of Griffin [Griffin, J. Soc. Cosmetic Chem. (1949), 311-326; Griffin, J. Soc. Cosmetic Chem. (1954), 249-256] as follows:
HLB = 20 x (MW-H/MW-T) wherein MW-H is the molecular weight of the hydrophilic portion of the compound and MW-T is the molecular weight of the total compound. For example, for the emulsifier PEG100 stearate, MW-H is the molecular weight of the ethylene glycol portions of the molecule which is 100 x 44 (MW ethylene oxide monomer = 44 g/mol) = 4400. Stearic acid has a molecular weight of 284.5 g/mol, so MW-T = 4684.5. Therefore, the HLB value for PEG100 stearate is calculated at 18.8. PEG-80 sorbitan monooleate (sold as Tween® 80 or Polysorbate 80) has a HLB value of 15. HLB values for a selection of emulsifiers and commercially available excipients are listed in the table below.
Figure imgf000016_0001
[0066] Ionic surfactants generally have higher HLB values than non-ionic surfactants. Sodium lauryl sulfate (SLS), for example, has a HLB value of 40.
[0067] In an embodiment, the wetting agent is generally regarded as safe for oral administration to humans.
[0068] In an embodiment, the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25. Conveniently, the wetting agent has a hydrophilic-lipophilic balance (HLB) between 8 and 20. More conveniently, the wetting agent has a hydrophilic-lipophilic balance (HLB) between 12 and 18, such as between 13 and 18, between 13 and 17, or about 14 to 16.
[0069] In an embodiment the wetting agent is water soluble. In an embodiment, the wetting agent has an aqueous solubility greater than 10 g/litre, such as greater than 10 g/litre, greater than 20 g/litre, greater than 50 g/litre, or greater than 75 g/litre.
[0070] In an embodiment, the wetting agent is water soluble and has a hydrophilic- lipophilic balance (HLB) between 12 and 18 In an embodiment, the wetting agent has an aqueous solubility greater than 10 g/litre and a hydrophilic-lipophilic balance (HLB) between 12 and 18. In an embodiment, the wetting agent has an aqueous solubility greater than 10 g/litre and a hydrophilic-lipophilic balance (HLB) between 13 and 18, such as between 13 and 17, or between 14 and 16. In an embodiment, the wetting agent has an aqueous solubility greater than 50 g/litre and a hydrophilic-lipophilic balance (HLB) between 13 and 18, such as between 13 and 17, or between 14 and 16.
[0071] In an embodiment the wetting agent is a non-ionic wetting agent selected from polyol esters, polyoxyethylene esters and poloxamers. In an embodiment, the polyol esters are selected from one or more of glycol esters, glycerol esters and sorbitan derivatives. In a convenient embodiment, sorbitan derivates comprise polysorbate esters (such as polysorbate 20, polysorbate 40, polysorbate 60 and polysorbate 80). Most conveniently the wetting agent is PEG-80 sorbitan monooleate (polysorbate 80).
[0072] In an embodiment, the wetting agent is present in the composition at about 0.1 to 5.0% w/w of the total weight of the composition. It has been found that the advantageous effects associated with the presence of the wetting agent (improved wet granulation and superior disintegration of the composition and dispersal of the pharmaceutically acceptable salt of Compound 1 in aqueous media) can be realised even at low levels of wetting agent. Furthermore, even when the drug loading is high (for example in mini-tablets), low levels of wetting agent have been found to be effective. In an embodiment, the wetting agent is present in the composition at about 0.5 to 2.5% w/w. In a convenient embodiment, the wetting agent is present in the composition at about 0.5 to 1.5% w/w, such as about 0.75 to 1.25% w/w, or about 1.0% w/w. In a convenient embodiment, the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w.
[0073] In an embodiment, the wetting agent is a polysorbate ester and the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w. In a convenient embodiment, the wetting agent is polysorbate 80 and the wetting agent is present in the composition at about 1.0 to 3.0% w/w, such as about 1.5 to 2.0% w/w, about 1.7 to 2.3% w/w, or about 1.9% w/w, or about 2.0% w/w.
[0074] It has been found that acidic environments are capable of cleaving the Boc group of Compound 1. It may therefore be advantageous to avoid incorporating overly acidic excipients in the compositions of the present invention, or carefully selecting grades of excipients such that their acidity is within acceptable ranges. It may be desirable to avoid excipients, such as buffering agents, capable of generating acidic micro-environments (e.g. pH less than 3) within the granulate. In a convenient embodiment, the one or more pharmaceutically acceptable excipients are not acidic. In a convenient embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 3.5 when dissolved or slurried in water. In a more convenient embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 6 when dissolved or slurried in water. In a convenient embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 6.5 when dissolved or slurried in water. In an embodiment, the one or more pharmaceutically acceptable excipients have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
[0075] For example, the excipient microcrystalline cellulose (MCC) is a purified, partially depolymerised cellulose prepared by treating cellulose pulp with mineral acids. As such, the pH of different batches of MCC may vary between approximately 5.0 and 7.5 (when a 5 g sample of MCC is slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the supernatant is measured). In an embodiment, the one or more pharmaceutically acceptable excipients do not comprise microcrystalline cellulose.
[0076] Typically, after formation of the granulate compositions they are dried and milled. It may be desirable to have a low water content. In an embodiment, the composition has a moisture level of less than 6% w/w, such as less than 5% w/w or less than 4% w/w. Conveniently, the composition has a moisture level of about 3.5% w/w.
[0077] In an embodiment, the one or more pharmaceutically acceptable excipients comprise a binder. A binder helps to hold the granulate mixture together. In an embodiment, the binder is a cellulose ether-based binder (such as hydroxypropyl cellulose or methyl cellulose). In a convenient embodiment, the binder is selected from hydroxy propyl cellulose, hypromellose, povidone, starch, methylcellulose, gelatin, pregelatinized starch, and xanthan gum. In a more convenient embodiment, the binder is hydroxy propyl cellulose.
[0078] In an embodiment, the one or more pharmaceutically acceptable excipients comprise a disintegrant. A disintegrant increases water wicking into the granulate core and therefore facilitates disintegration of the solid composition. In an embodiment, the disintegrant is selected from crospovidone, croscarmellose, sodium starch glycolate and low substituted hydroxypropyl cellulose. In a more convenient embodiment, the disintegrant is crospovidone.
[0079] In an embodiment, the one or more pharmaceutically acceptable excipients comprise a diluent. In an embodiment, the diluent is selected from lactose, pregelatinized starch, microcrystalline cellulose and silicified microcrystalline cellulose.
[0080] In an embodiment, the one or more pharmaceutically acceptable excipients comprise microcrystalline cellulose and lactose within the granulate. Conveniently, the w/w ratio of microcrystalline celluloselactose within the granulate is between 1 : 1 and 3 : 1 (such as about 2: 1).
[0081] In an embodiment, the diluent is selected from lactose and pregelatinized starch, or mixtures of the two. In an embodiment, the diluent is selected from lactose monohydrate and pregelatinized starch, or mixtures of the two. Conveniently, the one or more pharmaceutically acceptable excipients comprise lactose monohydrate and pregelatinized starch within the granulate. Conveniently, the w/w ratio of pregelatinized starch lactose within the granulate is between 1 :2 and 2: 1 (such as about 1: 1). [0082] In an embodiment, the immediate release composition according to the present invention comprises about 10-40% w/w of the pharmaceutically acceptable salt of Compound 1. Conveniently, the composition comprises 15-30% w/w of the pharmaceutically acceptable salt of Compound 1. In one embodiment, the composition comprises 15-20% (such as 16-18%) w/w of the pharmaceutically acceptable salt of Compound 1. In another embodiment, the composition comprises 25-30% (such as 26-28%) w/w of the pharmaceutically acceptable salt of Compound 1.
[0083] In an alternative embodiment, the immediate release composition according to the present invention comprises a high drug loading. Conveniently, the immediate release composition according to the present invention comprises about 40-70% w/w of the pharmaceutically acceptable salt of Compound 1. More conveniently, the composition comprises 50-70% w/w of the pharmaceutically acceptable salt of Compound 1. In one embodiment, the composition comprises 60-70% (such as 62-66%) w/w of the pharmaceutically acceptable salt of Compound 1. Conveniently, the immediate release composition according to this embodiment is a mini-tablet composition.
[0084] In an embodiment, the immediate release composition according to the present invention comprises 20 to 150 mg (such as 50 to 70 mg, about 60 mg, 110 to 130 mg, or about 120 mg) of Compound 1 or a pharmaceutically acceptable salt thereof. In an embodiment, the immediate release composition according to the present invention comprises 120 to 360 mg (such as 180 to 300 mg, 200 to 280 mg, 220 to 260 mg, or about 240 mg) of Compound 1 or a pharmaceutically acceptable salt thereof. In an embodiment, the immediate release composition according to the present invention comprises 20 to 360 mg (such as about 60 mg, about 120 mg, or about 240 mg) of Compound 1 or the hydrochloride salt thereof. In an embodiment the immediate release composition comprises about 120 mg of Compound 1. In an embodiment the immediate release composition comprises about 135 mg of the monohydrate hydrochloride salt of Compound 1.
[0085] In an embodiment, the one or more pharmaceutically acceptable excipients do not comprise a buffering agent. Buffering agents are weak acids or bases which are capable of buffering the pH micro-environment the composition is exposed to in vivo after oral dosing. Examples of buffering agents include citric acid, lactic acid, tartaric acid. [0086] The granulate is composed of granules, which may typically be sized in the range 0.1 to 2.0 mm. Conveniently, the granules within a granulate have an average particle size in the range 0.2 to 0.5 mm. In an embodiment, the granulate comprises granules having an average particle size between 200 and 500 pm, such as between 250 and 400 pm.
[0087] In an embodiment, the granulate comprises granules having a particle size distribution of 0-10% less than between 74 pm; 10-20% between 74 and 125 pm; 10-20% between 125 and 177 pm; 30-50% between 177 and 420 pm; 5-30% between 420 and 595 pm; and 0-25% between 595 and 841 pm.
[0088] In an embodiment, the granulate comprises granules having a particle size distribution of 30-50% of granules having particle sizes between 177 and 420 pm; and/or less than 20% of granules having particle sizes between 125 and 177 pm. In an embodiment, the granulate comprises granules having a particle size distribution of 30-40% of granules having particle sizes between 177 and 420 pm; and/or 10-20% of granules having particle sizes between 125 and 177 pm.
[0089] In contrast to fine powder formulations used in direct compression tabletting, the granulate compositions according to the present invention typically have higher bulk densities, and lower Carr’s Index and Hausner ratio values. These properties are indicative that the granulates have superior powder flow and compression properties.
[0090] In an embodiment, the composition has one or more of the following properties:
• a bulk density of > 0.4 g/ml;
• a Carr’s Index of < 40%; and
• a Hausner Ratio of < 1.7.
[0091] In an embodiment, the composition has one or more of the following properties:
• a bulk density of > 0.45 g/ml;
• a Carr’s Index of < 35%; and
• a Hausner Ratio of < 1.5.
[0092] In an embodiment, the composition has one or more of the following properties:
• a bulk density of > 0.5 g/ml;
• a Carr’s Index of < 30%; and
• a Hausner Ratio of < 1.35.
[0093] In an embodiment, the composition has one or more of the following properties: a bulk density of > 0.5 g/ml; a Carr’s Index of < 25%; and a Hausner Ratio of < 1.3.
[0094] In an embodiment, the composition has one or more of the following properties:
• a bulk density of > 0.55 g/ml;
• a Carr’s Index of < 20%; and
• a Hausner Ratio of < 1.25.
[0095] The immediate release compositions according to the present invention undergo rapid disintegration and dispersal in aqueous media (such as 0.01N hydrochloric acid or pH 6.8 buffer).
[0096] In an embodiment the dissolution rate of the drug from the immediate release composition is 75 % or more after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm. Conveniently, the dissolution rate of the drug from the immediate release composition is 80 % or more after 15 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
[0097] In an embodiment, the immediate release composition undergoes substantially complete disintegration and dispersal in 0.01N hydrochloric acid in less than 5 minutes at 37 °C using USP disintegration apparatus. In an embodiment, the immediate release composition undergoes substantially complete disintegration and dispersal in pH 6.8 aqueous media in less than 5 minutes at 37 °C using USP disintegration apparatus. In an embodiment, the immediate release composition undergoes substantially complete disintegration and dispersal in pH 1-7 aqueous media in less than 5 minutes at 37 °C using USP disintegration apparatus.
[0098] The chemical stability of Compound 1 in certain solid compositions of the present invention has been found to be very good even after several months under accelerated storage conditions. In an embodiment, the immediate release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 40 °C and 75% RH for 3 months. In an embodiment, the immediate release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 6 months. In an embodiment, the immediate release composition provides less than 2% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 3 months.
[0099] Conveniently, the immediate release solid pharmaceutical composition as described herein may be formulated as a tablet. The term ‘tablet’ comprises tablets of any size suitable for oral administration, including micro-tablets and mini-tablets (which would typically be in the size range 1 to 4 mm). Conveniently, the immediate release solid pharmaceutical composition is a mini-tablet. The composition may be encapsulated within a capsule, wherein the capsule is non-functional (i.e. it is readily soluble in aqueous media, so that the immediate release properties of the composition are not significantly affected). In an embodiment, the immediate release composition is formulated as a tablet and the tablet has an average hardness of greater than 7 kp, such as greater than 7.5 kp, greater than 8 kp, greater than 8.5 kp or greater than 9 kp. In an embodiment, the immediate release composition is formulated as a tablet and the tablet has an average hardness of 7-12 kp, such as 7.5-11 kp, 8-11 kp, or 8.5-10 kp. In another embodiment, the immediate release composition is formulated as mini-tablets and the minitablets have an average hardness of 1-3 kp, such as 1-2 kp, 1-1.5 kp, or about 1.3 kp.
[0100] When the composition is formulated as a tablet, prior to compression additional extra-granular excipients are typically mixed with the granules to improve the tablet processability and properties. Therefore, in an embodiment, the tablet comprises a granulate component and an extra-granulate component. In an embodiment, the granulate component comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients selected from a filler, a diluent, a binder and a disintegrant. In an embodiment, the extra-granulate component comprises one or more pharmaceutically acceptable excipients selected from a filler, a diluent, a disintegrant, a lubricant and a glidant. Suitable excipients for the extra-granulate component will be readily chosen by one of skill in the art and suitable fillers/diluents and disintegrants are described above. Lubricants may be added to solid compositions to reduce friction and sticking during tablet processing. Suitable lubricants comprise magnesium stearate, calcium stearate, hydrogenated vegetable oil, stearic acid, sodium stearyl fumarate, mineral oil, hydrogenated vegetable oil and polyethylene glycol. In an embodiment, the extra-granulate component comprises magnesium stearate. In an embodiment the composition comprises 0.25-1.5 % w/w of lubricant (such as magnesium stearate). In an alternative, convenient, embodiment, the extra-granulate component comprises magnesium stearate and hydrogenated vegetable oil . In an embodiment the extra-granulate component comprises magnesium stearate and sodium stearyl fumarate. In an embodiment the composition comprises 4-6 % w/w of one or more (e.g. two) lubricants (such as magnesium stearate and hydrogenated vegetable oil, or magnesium stearate and sodium stearyl fumarate).
[0101] Conveniently, when the composition is formulated as mini-tablets, it has been found that higher levels of lubricant are needed in order to prevent the composition from sticking to the compression punches during tabletting. Therefore, in an embodiment, the immediate release composition is formulated as mini-tablets and the mini-tablets comprise greater than 2.5 % w/w (such as greater than 3.5 % w/w, or greater than 4.5 % w/w) of one or more lubricants. In an embodiment, the immediate release composition is formulated as mini-tablets and the minitablets comprise between about 2.5 % and 7.5% w/w (such as between about 3.5 % and 6.5% w/w, or between about 4.5 % and 5.5% w/w) of one or more lubricants.
[0102] Surprisingly, the Applicants have found that when the composition is formulated as mini-tablets, a combination of two lubricants at a total level of about 3 % to 7 % w/w provide significant beneficial effects, i.e. with respect to preventing the composition from sticking to the compression punches during tabletting and improving processability at scale. More conveniently, the mini-tablets comprise a combination of magnesium stearate and a second lubricant at a total level of about 3 % to 7 % w/w. Most conveniently, the mini -tablets comprise a combination of magnesium stearate and hydrogenated vegetable oil at a total level of about 3 % to 7 % w/w. In an embodiment, the immediate release composition is formulated as mini-tablets and the minitablets comprise between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.0% w/w, or about 2.5 %) of magnesium stearate and between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.5% w/w, or about 2.75 %) of hydrogenated vegetable oil. Conveniently, the mini-tablets comprise a combination of magnesium stearate and sodium stearyl fumarate at a total level of about 3 % to 7 % w/w. In an embodiment, the immediate release composition is formulated as mini-tablets and the mini-tablets comprise between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.0% w/w, or about 2.5 %) of magnesium stearate and between about 1.5 % and 3.5% w/w (such as between about 2.0 % and 3.5% w/w, or about 2.75 %) of sodium stearyl fumarate. [0103] Glidants may be added to solid compositions to improve flow properties during processing. Suitable glidants comprise colloidal silicon dioxide, ascorbyl palmitate, calcium palmitate, starch and talc. In an embodiment, the extra-granulate component comprises colloidal silicon dioxide. In an embodiment the composition comprises 0.1-0.5 % w/w of glidant (such as colloidal silicon dioxide).
[0104] In an embodiment, the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
• 20-40 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 40-70 % w/w of one or more diluents;
• 1 - 10 % w/w of di si ntegrant;
• 1 - 10 % w/w of binder; and
• 0.5 -2.0 % w/w of wetting agent.
[0105] In an embodiment, the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
• 20-40 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 45-65 % w/w of one or more diluents;
• 3-8 % w/w of disintegrant;
• 3-8 % w/w of binder; and
• 1-2 % w/w of wetting agent.
[0106] In an embodiment, the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
• 20-30 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 55-65 % w/w of one or more diluents;
• 3-8 % w/w of disintegrant;
• 3-8 % w/w of binder; and
• 1-2 % w/w of wetting agent.
[0107] In an embodiment, the immediate release solid pharmaceutical composition comprises a granulate, wherein the granulate comprises:
• 30-40 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 45-55 % w/w of one or more diluents;
• 3-8 % w/w of disintegrant; • 3-8 % w/w of binder; and
• 1-2 % w/w of wetting agent.
[0108] In an embodiment, the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
• 10-30 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 30-60 % w/w of one or more diluents;
• 1-7 % w/w of disintegrant;
• 1-7 % w/w of binder; and
• 0.5 -2.0 % w/w of wetting agent; and the tablet comprises in the extra-granulate component:
• 20-30 % w/w of one or more diluents;
• 1-5 % w/w of disintegrant; and
• 0.25-1.5 % w/w of lubricant.
[0109] In an embodiment, the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
• 15-30 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 30-50 % w/w of one or more diluents;
• 3-5 % w/w of disintegrant;
• 3-5 % w/w of binder; and
• 0.5 - 1.5 % w/w of wetting agent; and the tablet comprises in the extra-granulate component:
• 20-30 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant; and
• 0.5-1.5 % w/w of lubricant.
[0110] In an embodiment, the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
• 15-20 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 40-50 % w/w of one or more diluents; • 3-6 % w/w of disintegrant;
• 3-6 % w/w of binder; and
• 0.5 - 1.5 % w/w of wetting agent; and the tablet comprises in the extra-granulate component:
• 22-27 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant; and
• 0.5-1.5 % w/w of lubricant.
[OHl] In an embodiment, the immediate release solid pharmaceutical composition is formulated as a tablet and the tablet comprises a granulate component and an extra-granulate component, wherein the tablet comprises in the granulate component:
• 25-30 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 30-40 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 4-6 % w/w of binder; and
• 0.5 - 1.5 % w/w of wetting agent; and the tablet comprises in the extra-granulate component:
• 22-27 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant; and
• 0.5-1.5 % w/w of lubricant.
[0112] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1.
[0113] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent.
[0114] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component: • 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
[0115] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the composition does not comprise microcrystalline cellulose.
[0116] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants.
[0117] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80.
[0118] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants. [0119] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of hydrogenated vegetable oil.
[0120] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of sodium stearyl fumarate.
[0121] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 10-30 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant;
• 1-4 % w/w of disintegrant; and
• 3-7 % w/w of one or more lubricants.
[0122] In an embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the mini-tablets comprise a granulate component and an extragranulate component, wherein the mini-tablets comprise in the granulate component: • 60-70 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 15-25 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant;
• 1-3 % w/w of disintegrant; and
• 4-6 % w/w of one or more lubricants.
[0123] Conveniently, the immediate release composition is substantially encapsulated in a water-soluble coating. The coating may be a capsule encapsulating the granulate, or it may be a coating substantially encapsulating the composition formulated as a tablet. In an embodiment, the composition is formulated as a tablet and the tablet is substantially encapsulated in a water- soluble (pH-independent - e.g. soluble in aqueous media at pH 1-8) coating. To prevent light degradation of the tablets on storage, in an embodiment, the water-soluble coating is also a UV- resistant coating. In an embodiment the coating is PVA-based. In an embodiment, the composition is formulated as mini-tablets and each mini-tablet is substantially encapsulated in a water-soluble (pH-independent - e.g. soluble in aqueous media at pH 1-8) coating.
[0124] In an embodiment, the immediate release composition is prepared by a wet granulation method. In an embodiment, the immediate release composition is obtainable by wet granulation. Wet granulation can be carried out by any known wet granulation process, including high-shear wet granulation and fluid bed granulation (see for example, Remington: The Science and Practice of Pharmacy, Edition, 22nd Edition, 2012). Conveniently, the wet granulation is carried out with high shear mixing.
[0125] In a convenient embodiment, the wet granulation is carried out by fluid bed granulation. In a convenient embodiment, the immediate release solid pharmaceutical composition is formulated as mini-tablets and the wet granulation is carried out by fluid bed granulation. In a typical fluid bed granulation process, the disintegrant, filler and pharmaceutically acceptable salt of Compound 1 are blended and then wet granulated in a fluid bed granulator in the presence of an aqueous solution of binder and wetting agent. After drying and optional milling, the granulate is then further blended with the extra-granular components (such as disintegrant, lubricant(s), optional filler and optional glidant). It has been found that fluid bed granulation offers advantages over other forms of granulation in terms of improved scalability of processing for compositions of the invention.
Delayed Release Solid Compositions
[0126] In a first aspect the present invention provides a method of treating an inflammatory bowel disease, comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof. In an embodiment, the oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof is a delayed-release solid pharmaceutical composition for oral administration comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
[0127] The oral delayed-release compositions comprising Compound 1 or a pharmaceutically acceptable salt thereof provides benefits in respect to targeted release of Compound 1 and biopharmaceutical performance. Surprisingly, certain delayed release solid compositions of the present invention have been found to achieve colonic tissue concentration levels of Compound 1, that are at least comparable to those achieved via oral dosing of a pharmaceutically acceptable salt of Compound 1 as either an immediate release solution, or an immediate release solid composition, without the need for any appreciable systemic exposure. Such compositions offer a number of advantages in respect to treatment of IBD, such as potentially minimising systemic side effects, e.g. those linked to elevated erythropoietin (EPO) and vascular endothelial growth factor (VEGF) levels.
[0128] The delayed-release solid pharmaceutical composition according to the present invention can be any composition suitable for oral administration, such as a tablet, a capsule, or granules or pellets, for example delivered in a sachet or capsule.
[0129] In an embodiment, there is provided a delayed-release solid pharmaceutical composition for oral administration comprising Compound 1 (i.e. the free base) and one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a delayed- release solid pharmaceutical composition for oral administration comprising a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients.
[0130] Suitable pharmaceutically acceptable salts include acid-addition salts of the basic piperazine nitrogen in Compound 1 and also metal salts of the weakly acidic hydroxyl group in Compound 1.
[0131] Acid-addition salts include salts with inorganic or organic acids. Inorganic acid salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid salts. Organic acid salts include trifluoroacetic, acetic, formic, citric, maleic, succinic, lactic, glycolic, tartaric, methanesulfonic and -toluenesulfonic acid salts. Conveniently, the pharmaceutically acceptable salt is an inorganic acid salt. More conveniently, the pharmaceutically acceptable salt is a hydrochloric acid salt.
[0132] Metal salts include alkali metal salts and alkali earth metal salts, such as sodium, potassium, calcium or magnesium salts. Conveniently, the pharmaceutically acceptable salt is a calcium salt.
[0133] In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt. In an embodiment, the pharmaceutically acceptable salt is a hydrochloride salt and the ratio of Compound 1 to HC1 is about 1 : 1.
[0134] In an embodiment, the hydrochloride salt is a hydrate. In a convenient embodiment, the hydrochloride salt is a monohydrate. In a convenient embodiment, the hydrochloride salt is a crystalline monohydrate, which is characterized by X-Ray diffraction peaks at 15.1, 17.4, 19.8 and 20.0 ± 0.2 degrees 29 (Form A). In a convenient embodiment, the hydrochloride salt is a crystalline monohydrate, which is characterized by a melting point of 191- 194 °C.
[0135] In an embodiment, the hydrochloride salt is anhydrous. In a convenient embodiment, the hydrochloride salt is a crystalline anhydrous compound, which is characterized by X-Ray diffraction peaks at 9.0, 16.8 and 18.6 ± 0.2 degrees 29 (Form B). In a convenient embodiment, the hydrochloride salt is a crystalline anhydrous compound, which is characterized by a melting point of 195-198 °C.
[0136] In one embodiment, the delayed-release solid pharmaceutical composition is an erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix. By erodible matrix is meant aqueous-erodible or water-swellable or aqueous soluble, in the sense of being either erodible or swellable or dissolvable in pure water or requiring the presence of an acid or base to ionize the polymeric matrix sufficiently to cause erosion or dissolution. When contacted with an aqueous environment, the erodible matrix imbibes water and forms an aqueous-swollen gel or “ matrix ” that Compound 1 or a pharmaceutically acceptable salt thereof can pass or diffuse through depending on its physicochemical properties. The aqueous-swollen matrix gradually erodes, swells, disintegrates or dissolves, thereby delaying the release of Compound 1 or a pharmaceutically acceptable salt thereof after oral administration. Suitable polymers for the erodible matrix are hydrogels such as synthetic polymers derived from vinyl, acrylate, methacrylate, urethane, ester and oxide monomers; or derivatives of naturally occurring polymers such as polysaccharides or proteins (including polysaccharides, gums, starches, alginates, collagen and cellulosics).
[0137] In another embodiment, the delayed-release solid pharmaceutical composition is a capsule comprising granules or pellets encapsulated within a delayed release capsule.
[0138] In an alternative embodiment, the delayed-release solid pharmaceutical composition is a readily-soluble capsule comprising granules or pellets, wherein the individual granules or pellets are coated with a delayed release coating.
[0139] In an alternative embodiment, the delayed-release solid pharmaceutical composition comprises a core and a delayed release coating substantially encapsulating the core. Conveniently, the delayed-release solid pharmaceutical composition is a tablet comprising a solid core and a delayed release coating substantially encapsulating the solid core. The term ‘tablet’ comprises tablets of any size suitable for oral administration, including micro-tablets and mini-tablets (which would typically be in the size range 1 to 4 mm). Conveniently, the delayed- release solid pharmaceutical composition comprises mini-tablets and each mini-tablet comprises a solid core and a delayed release coating substantially encapsulating the solid core.
[0140] In terms of the coating, ‘substantially’ means that the coating covers the majority of the surface of the core, such as greater than 75%, greater than 85%, or preferably greater than 95% of the surface of the core. In a convenient embodiment, the delayed release coating fully encapsulates the core.
[0141] In an embodiment, the delayed release coating dissolves at pH values greater than about 5.5. In an embodiment, the delayed release coating dissolves at pH values greater than about 6.0. In an embodiment, the delayed release coating dissolves at pH values greater than about 7.0. Conveniently, the delayed release coating dissolves at about pH 5.5.
[0142] In an embodiment, the delayed release coating comprises methyl acrylatemethacrylic acid copolymer, ethyl acrylate-methacrylic acid copolymer, hydroxy propyl methyl cellulose acetate succinate or cellulose acetate phthalate. In a convenient embodiment, the delayed release coating comprises methyl acrylate-methacrylic acid copolymer or ethyl acrylatemethacrylic acid copolymer. Conveniently, the delayed release coating is selected from Eudragit® L 100-55, Eudragit® FS30D, Eudragit® L100, Eudragit® L 12,5, Eudragit® L30 D- 55, Eudragit® S100 and Eudragit® S12,5, such as Eudragit® L 100-55.
[0143] The thickness of the coating is important to ensure that the composition does not take up acid and disintegrate in the acidic environment of the stomach. In an embodiment, the delayed-release composition comprises a greater than 6% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a greater than 8% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a greater than 10% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 12% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 14% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises about 10% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a 6 to 14% weight gain delayed release coating. In an embodiment, the delayed-release composition comprises a 7 to 13% (such as a 8 to 12%, or 9 to 11%) weight gain delayed release coating.
[0144] In an embodiment, the delayed-release composition further comprises an additional sub-coating beneath the delayed release coating. In an embodiment, the sub-coating is soluble in aqueous media independent of the pH of the media. In an embodiment, the sub-coating is water-soluble at between pH 1 and 8. In an embodiment the sub-coating is PVA-based. In an embodiment, the delayed-release composition comprises 2-6%, such as 3-5%, or about 4% weight gain of sub-coating. In an embodiment, the delayed-release composition comprises about 5% weight gain of sub-coating.
[0145] Conveniently, the delayed-release composition comprises a core and the core comprises a granulate. The granulate may conveniently be a granulate as described above according to the first aspect of the invention. Therefore, all the embodiments described above for the first aspect (in relation to Compound 1 or a pharmaceutically acceptable salt thereof, the one or more pharmaceutically acceptable excipients, levels of the excipients, granule particle size distribution, bulk density, Carr’s index and Hausner ratio of the composition, the extra-granulate component and tablet hardness) all apply equally to a delayed-release composition core comprising a granulate according to the third aspect of the invention.
[0146] In an embodiment, the delayed release composition according to the present invention comprises 20 to 150 mg (such as 50 to 70 mg, about 60 mg, 110 to 130 mg, or about 120 mg) of Compound 1 or a pharmaceutically acceptable salt thereof. In an embodiment, the delayed release composition according to the present invention comprises 120 to 360 mg (such as 180 to 300 mg, 200 to 280 mg, 220 to 260 mg, or about 240 mg) of Compound 1 or a pharmaceutically acceptable salt thereof. In an embodiment, the delayed release composition according to the present invention comprises 20 to 360 mg (such as about 60 mg, about 120 mg, or about 240 mg) of Compound 1 or the hydrochloride salt thereof.
[0147] In an alternative embodiment, the delayed-release composition is a multiparticulate composition comprising delayed release pellets, mini-tablets or beads. The pellets or mini-tablets may be formed from a granulate as described hereinabove, wherein the individual pellets or mini-tablets are coated with a delayed release coating as described herein, or alternatively the pellets are encapsulated within a delayed release capsule. The delayed release capsule may conveniently have the same properties as the delayed release coatings described herein.
[0148] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1.
[0149] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent. [0150] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
[0151] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the composition does not comprise microcrystalline cellulose.
[0152] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants.
[0153] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80. [0154] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants.
[0155] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of hydrogenated vegetable oil.
[0156] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of sodium stearyl fumarate.
[0157] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 50-70 % w/w of a pharmaceutically acceptable salt of Compound 1; • 10-30 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant;
• 1-4 % w/w of disintegrant; and
• 3-7 % w/w of one or more lubricants.
[0158] In an embodiment, the delayed release composition is formulated as mini-tablets and each mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component, wherein the mini-tablets comprise in the granulate component:
• 60-70 % w/w of a pharmaceutically acceptable salt of Compound 1;
• 15-25 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant;
• 1-3 % w/w of disintegrant; and
• 4-6 % w/w of one or more lubricants.
[0159] Conveniently, in any of the above delayed release mini-tablet embodiments, the delayed release coating dissolves at pH values greater than about 5.5, such as greater than about 6.0. Conveniently, in any of the above delayed release mini-tablet embodiments, each mini-tablet comprises a greater than 6% weight gain (such as a greater than 8% weight gain) delayed release coating.
[0160] Alternatively, each pellet, or subunit, of the delayed release multiparticulate is an inert core coated with a drug layer comprising Compound 1 or a pharmaceutically acceptable salt thereof and the drug layer-coated core is then coated with a delayed release coating as described herein. In this embodiment, multiparticulates in the form of beads or pellets may be prepared by building Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g. a binder dispersion, for example HPMC) onto seed cores in a fluidized bed such as a Wurster coater or a rotary processor. The seed core can be comprised of a sugar (for example a non-pareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose. In an embodiment, the inert core comprises sugar spheres mesh 45/60 (250-355 microns). An example of a suitable composition and method is to spray a dispersion of Compound 1 / binder (e.g. HPMC) composition in water on to the seed core. A delayed release coating is then employed to fabricate the membrane, which is applied over Compound 1 or a pharmaceutically acceptable salt thereof layered seed cores. In an embodiment, the DR pellets formed by this layering process on a seed core comprise the free base of Compound 1.
[0161] Due to the delayed release coating the delayed-release compositions do not undergo any significant disintegration or dispersal in the acidic environment of the stomach after oral administration. Therefore, in an embodiment, the delayed-release composition undergoes less than 5% dissolution in 0.01N HC1 after 30 mins at 37 °C using USP2 apparatus. In an embodiment the dissolution rate of the drug from the delayed release composition is 10 % or less after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm. In an embodiment the dissolution rate of the drug from the immediate release composition is 5% or less after 30 minutes from the beginning a dissolution test, which is carried out in accordance with a dissolution test (paddle method) described in the United States Pharmacopoeia (USP) under the conditions that 900 mL of 0.01N hydrochloric acid is used at 37 °C and the paddle rotation speed is 75 rpm.
[0162] When stirred in acidic aqueous media the delayed release composition according to the present invention takes up only relatively small amounts of acid. In an embodiment, the delayed release composition increases in weight by less than 5% (such as less than 4% or less than 3%) after stirring for 2 hr in 0. IN hydrochloric acid in USP disintegration apparatus. [0163] After oral administration, once the delayed-release composition enters the small intestines and the pH increases above approximately 5.5 the delayed release coating may start to dissolve and the composition will undergo disintegration and dispersal.
[0164] In an embodiment, the delayed release composition undergoes substantially complete disintegration in pH 6.8 aqueous media in less than 30 minutes (such as less than 15 minutes) at 37 °C using USP disintegration apparatus. In an embodiment, the delayed release composition undergoes substantially complete disintegration and dispersal in pH 6.8 aqueous media in less than 30 minutes (such as less than 15 minutes) at 37 °C using USP disintegration apparatus.
[0165] In an embodiment the delayed release composition undergoes greater than 75% dissolution within 240 minutes from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method) under the conditions that 900 mL of pH 6.8 with 2% CTAB buffer is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm. In an embodiment the delayed release composition undergoes greater than 85% dissolution within 2400 minutes from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method) under the conditions that 900 mL of pH 6.8 with 2% CTAB buffer is used, the temperature is 37 °C and the paddle rotation speed is 75 rpm.
[0166] In an embodiment the delayed release composition undergoes greater than 75% dissolution within 240 minutes (such as within 150 minutes, or within 90 minutes) from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method; temperature is 37 °C; paddle rotation speed is 75 rpm) under the conditions that 300 mL of pH 6.8 buffer is used for 30 minutes, followed by 900 mL of pH 6.8 buffer with 2% CTAB.
[0167] In an embodiment the delayed release composition undergoes greater than 75% dissolution within 240 minutes (such as within 150 minutes) from the beginning of a dissolution test, which is carried out in accordance with the USP 2 dissolution test (paddle method; temperature is 37 °C; paddle rotation speed is 75 rpm) under the conditions that 900 mL of 0.01N hydrochloric acid is used for 30 minutes, followed by 300 mL of pH 6.8 buffer for 30 minutes, followed by 900 mL of pH 6.8 buffer with 2% CTAB. [0168] The chemical stability of Compound 1 or a pharmaceutical salt thereof in the solid compositions of the present invention has been found to be very good even after several months under accelerated storage conditions. In an embodiment, the delayed release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 40 °C and 75% RH for 3 months. In an embodiment, the delayed release composition provides less than 5% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 6 months. In an embodiment, the delayed release composition provides less than 2% chemical degradation of Compound 1 by HPLC when stored at 25 °C and 60% RH for 3 months. In an embodiment, the delayed release composition provides less than 0.5% (such as less than 0.25%) of a related impurity having a relative retention time (RRT) of 0.43-0.44 by HPLC (compared to the retention time of Compound 1) when stored at 40 °C and 75% RH for 6 months.
[0169] In an embodiment, the delayed-release composition comprises a granulate and the granulate is prepared by a wet granulation method. In an embodiment, the delayed-release composition comprises a granulate which is obtainable by wet granulation. Wet granulation can be carried out by any known wet granulation process, including high-shear wet granulation and fluid bed granulation (see for example, Remington: The Science and Practice of Pharmacy, Edition, 22nd Edition, 2012). Conveniently, the wet granulation is carried out with high shear mixing.
[0170] In a convenient embodiment, the wet granulation is carried out by fluid bed granulation. In a convenient embodiment, the delayed release solid pharmaceutical composition is formulated as mini-tablets and the wet granulation is carried out by fluid bed granulation. In a typical fluid bed granulation process, the disintegrant, filler and pharmaceutically acceptable salt of Compound 1 are blended and then wet granulated in a fluid bed granulator in the presence of an aqueous solution of binder and wetting agent. After drying and optional milling, the granulate is then further blended with the extra-granular components (such as disintegrant, lubricant(s), optional filler and optional glidant). Fluid bed granulation may offer advantages over other forms of granulation in terms of improved scalability of the processing.
Modified Release Solid Compositions
[0171] In a second aspect, the present invention provides an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a pharmaceutically acceptable salt thereof.
[0172] The oral modified release pharmaceutical compositions according to the present invention comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a pharmaceutically acceptable salt thereof.
[0173] In an embodiment, the modified release composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1, or a hydrochloride salt of Compound 1.
[0174] In an embodiment, the immediate release component comprises a pharmaceutically acceptable salt of Compound 1. In an embodiment, the immediate release component comprises a hydrochloride salt of Compound 1.
[0175] In an embodiment, the delayed release component comprises the hydrochloride salt of Compound 1. In an embodiment, the delayed release component comprises the free base of Compound 1.
[0176] In an embodiment, the modified release composition comprises an immediate release component comprising the hydrochloride salt of Compound 1 and a delayed release component comprising the free base of Compound 1.
[0177] In a more convenient embodiment, the modified release composition comprises an immediate release component comprising the hydrochloride salt of Compound 1 and a delayed release component comprising the hydrochloride salt of Compound 1.
[0178] The hydrochloride salt of Compound 1 may be the monohydrate or the anhydrous form, conveniently the monohydrate.
[0179] In an embodiment, the modified release composition comprises the immediate release component and the delayed release component in a w/w ratio of between about 1 :4 and 4: 1. In a convenient embodiment, the modified release composition comprises the immediate release component and the delayed release component in a w/w ratio of between about 1 :3 and 3: 1, such as between about 1 :2 and 2: 1, or conveniently about 1 :1.
[0180] In an embodiment, the modified release composition comprises between 100 mg and 1000 mg of Compound 1. In an embodiment, the modified release composition comprises between 100 mg and 800 mg of Compound 1, such as between 100 mg and 800 mg, between 100 mg and 500 mg, or between 150 and 500 mg of Compound 1. In an embodiment, the modified release composition comprises about 120 mg or about 240 mg or about 480 mg of Compound 1. In a convenient embodiment, the modified release composition comprises about 240 mg of Compound 1. In this context, reference to the amount of Compound 1 will be understood to refer to the amount of the parent compound (free base), even if the compound is present as a salt of Compound 1.
[0181] In an embodiment, the modified release composition comprises between 50 mg and 500 mg of Compound 1 in the immediate release component and between 50 mg and 500 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises between 50 mg and 250 mg of Compound 1 in the immediate release component and between 50 mg and 250 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises between 50 mg and 150 mg of Compound 1 in the immediate release component and between 50 mg and 150 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises about 60 mg of Compound 1 in the immediate release component and about 60 mg of Compound 1 in the delayed release component. In an embodiment, the modified release composition comprises about 120 mg of Compound 1 in the immediate release component and about 120 mg of Compound 1 in the delayed release component.
[0182] The benefits of the present invention are not limited to a particular type of dosage form having a particular mechanism of drug release. Modified release of Compound 1 or a pharmaceutically acceptable salt thereof may be accomplished by any means known in the pharmaceutical art, including but not limited to the use of osmotic dosage forms, matrix dosage forms, multiparticulate dosage forms, gastric retentive dosage forms, and pulsatile dosage forms.
[0183] Two of these examples, namely matrix dosage forms and multiparticulate dosage forms, are described in greater detail below.
[0184] Matrix Systems (single unit dosage forms)
[0185] In one embodiment, Compound 1 or a pharmaceutically acceptable salt thereof is incorporated into an erodible or non-erodible matrix dosage form. Typically, in a matrix dosage form the drug is homogenously dispersed in a matrix material.
[0186] In one embodiment, the modified release solid pharmaceutical composition is an erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix. By erodible matrix is meant aqueous-erodible or water-swellable or aqueous soluble, in the sense of being either erodible or swellable or dissolvable in pure water or requiring the presence of an acid or base to ionize the polymeric matrix sufficiently to cause erosion or dissolution. When contacted with an aqueous environment, the erodible matrix imbibes water and forms an aqueous-swollen gel or “matrix” that Compound 1 or a pharmaceutically acceptable salt thereof can pass or diffuse through depending on its physicochemical properties. The aqueous-swollen matrix gradually erodes, swells, disintegrates or dissolves, thereby modifying the release of Compound 1 or a pharmaceutically acceptable salt thereof after oral administration. Suitable polymers for the erodible matrix are hydrogels such as synthetic polymers derived from vinyl, acrylate, methacrylate, urethane, ester and oxide monomers; or derivatives of naturally occurring polymers such as polysaccharides or proteins (including polysaccharides, gums, starches, alginates, collagen and cellulosics (e.g. HPC or HPMC)).
[0187] In another embodiment, the modified release solid pharmaceutical composition is a non-erodible matrix comprising Compound 1 or a pharmaceutically acceptable salt thereof dispersed in the matrix. Non-erodible matrix materials are inert and drug release is predominantly via diffusion through the matrix. Suitable materials for the inert matrix include water-insoluble plastics (such as PVC or PE), fatty materials (such as carnauba wax and triglycerides) and cross-linked PVP (crospovidone).
[0188] The matrix material may additionally contain a wide variety of pharmaceutically acceptable excipients known in the pharmaceutical arts, including excipients that ease the manufacturing process and/or improve the performance of the dosage form. Excipients include diluents or bulking agents, lubricants and binders.
[0189] The matrix polymers may be used alone, or as an appropriate combination of two or more thereof. In an embodiment, the matrix dosage form comprises an immediate release component and a delayed release component dispersed within a single polymer matrix. In an embodiment, the matrix dosage form comprises an immediate release component dispersed within a polymer matrix and a delayed release component dispersed within a different polymer matrix. In an embodiment, the matrix dosage form comprises Compound 1 or a pharmaceutically acceptable salt thereof dispersed within an erodible polymer matrix and Compound 1 or a pharmaceutically acceptable salt thereof dispersed within a non-erodible polymer matrix. Multiparticulate Systems
[0190] Multiparticulate systems include subunits such as mini-tablets, beads, pellets, and granules. Multiparticulates generally comprise a plurality of mini-tablets, beads, pellets or granules that may range in size from about 10 pm to about 2 mm, more typically about 100 pm to 1 mm in diameter. Such multiparticulates may be packaged, for example, in a capsule such as a gelatin capsule or a capsule formed from a polymer such as HPMCAS, HPMC or starch; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet (e.g. a caplet) or pill by compression or other processes known in the art .
[0191] Such multiparticulates may be made by any known process, such as wet- and drygranulation processes, extrusion/spheronization, roller-compaction, melt-congealing, or by spray-coating seed cores. In a convenient embodiment, the multiparticulates are made by spraycoating seed cores. In another embodiment, the multiparticulates are made by wet-granulation. In another embodiment, the multiparticulates are made by extrusion/spheronization.
[0192] For example, in wet- and dry-granulation processes the composition comprising Compound 1 or a pharmaceutically acceptable salt thereof and optional excipients may be granulated to form multiparticulates of the desired size. Excipients, such as a binder and/or a wetting agent, may be blended with the composition to aid in processing and forming the multiparticulates.
[0193] Binders useful in fabrication of multiparticulates include microcrystalline cellulose, hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), and related materials or combinations thereof. In general, binders which are useful in granulation and tabletting, such as starch, pregelatinized starch, and poly(N-vinyl-2-pyrrolidinone) (PVP) may also be used to form multiparticulates.
[0194] Wetting agents are useful in the fabrication of multiparticulates, particularly when multiparticulates are formed by wet granulation or spray-coating seed cores and Compound 1 is dispersed in an aqueous medium during processing. Wetting agents may also be beneficial in the disintegration and dispersal of multiparticulates comprising Compound 1 after oral dosing. The wetting agent is typically a surfactant or an emulsifier. Conveniently the wetting agent is a nonionic surfactant. In an embodiment the wetting agent is a non-ionic wetting agent selected from polyol esters, polyoxyethylene esters and poloxamers. In an embodiment, the polyol esters are selected from one or more of glycol esters, glycerol esters and sorbitan derivatives (such as polysorbate esters). Most conveniently the wetting agent is a poloxamer, (such as poloxamer 188), or a polysorbate ester (such as polysorbate 80).
[0195] In any case, the resulting particles may themselves constitute the multiparticulate or they may be coated by various film-forming materials such as water-swellable or water soluble polymers, and/or they may be combined with other excipients or vehicles to aid in dosing to patients.
[0196] In an embodiment, the modified release composition of the present invention is a multiparticulate system comprising immediate release particulates and delayed release particulates. The immediate release and delayed release particulates may be formed by the same process (e.g. spray-coating seed cores), or by different processes (e.g. IR particulates formed by wet-granulation and DR particulates formed by extrusion/spheronization).
[0197] In one embodiment, the immediate release component of the modified release composition is a multiparticulate comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients selected from binder, wetting agent, diluent and disintegrant. Each subunit of the multiparticulate can be individually coated with a membrane, such as a porous or a water-soluble coating.
[0198] In one embodiment, the immediate release multiparticulate is an immediate release granulate as described hereinabove. Conveniently, the immediate release granulate is obtained by a high shear wet granulation process as described hereinabove. Conveniently, the immediate release granulate is obtained by an extrusion/spheronization process, wherein spherical pellets are obtained.
[0199] In another embodiment, each subunit of the immediate release multiparticulate is an inert core coated with a drug layer. In this embodiment, multiparticulates in the form of beads or pellets may be prepared by building the Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying the Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g. a binder dispersion, for example HPMC) onto seed cores in a fluidized bed such as a Wurster coater or a rotary processor. The inert seed core can be comprised of a sugar (for example a nonpareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose. In an embodiment, the inert core comprises sugar spheres mesh 45/60 (250-355 microns). An example of a suitable composition and method is to spray a dispersion of Compound 1 or a pharmaceutically acceptable salt thereof / binder (e.g. HPMC) composition in water on to the seed core.
[0200] In one embodiment, the immediate release (IR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 10% to about 90% (w/w) of the weight of the IR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 5% to about 50% (w/w) of the total weight of the IR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 5: 1 to 15: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 5 : 1 ; and c) optionally a coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging from about 1 % to about 5 % (w/w) of the total weight of the IR pellet.
[0201] In one embodiment, the delayed release component of the modified release composition is a multiparticulate comprising Compound 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients selected from binder, wetting agent, diluent and disintegrant. Each subunit of the multiparticulate can be individually coated with a membrane, such as a porous or a water-soluble coating, prior to coating with a delayed release coating.
[0202] In one embodiment, the delayed release multiparticulate is a delayed release granulate as described hereinabove. Conveniently, the delayed release granulate is obtained by a high shear wet granulation process as described hereinabove. Conveniently, the delayed release granulate is obtained by an extrusion/spheronization process, wherein spherical pellets are obtained, which are subsequently coated with a delayed release coating as described herein.
[0203] In another embodiment, each subunit of the delayed release multiparticulate is an inert core coated with a drug layer and the drug layer-coated core is coated with a polymeric material of the type useful for providing delayed release of Compound 1. In this embodiment, multiparticulates in the form of beads or pellets may be prepared by building the Compound 1 or a pharmaceutically acceptable salt thereof composition (drug plus optionally any excipients) up on a seed core by a drug-layering technique such as powder coating or by applying the Compound 1 or a pharmaceutically acceptable salt thereof composition by spraying a solution or dispersion of Compound 1 or a pharmaceutically acceptable salt thereof in an appropriate solution / dispersion vehicle (e.g. a binder dispersion, for example HPMC) onto seed cores in a fluidized bed such as a Wurster coater or a rotary processor. The seed core can be comprised of a sugar (for example a non-pareil seed), starch or microcrystalline cellulose, conveniently microcrystalline cellulose. In an embodiment, the inert core comprises sugar spheres mesh 45/60 (250-355 microns). An example of a suitable composition and method is to spray a dispersion of Compound 1 or a pharmaceutically acceptable salt thereof / binder (e.g. HPMC) composition in water on to the seed core. A delayed release coating is then employed to fabricate the membrane, which is applied over Compound 1 or a pharmaceutically acceptable salt thereof layered seed cores. The rate release from the coated multiparticulates can be controlled by factors such as the composition and binder content of the drug-coated core, the thickness and permeability of the delayed release coating, and the surface-to-volume ratio and size of the multiparticulates. It will be appreciated by those skilled in the art that increasing the thickness of the coating will decrease the release rate, whereas increasing the permeability of the coating or the size or surface-to- volume ratio of the multiparticulates will increase the release rate. If desired, the permeability of the coating may be adjusted by blending of two or more materials.
[0204] In one embodiment, the delayed release (DR) pellet coating dissolves at pH values greater than about 5.5. In an embodiment, the delayed release coating dissolves at pH values greater than about 6.0. In an embodiment, the delayed release coating dissolves at pH values greater than about 7.0. Conveniently, the delayed release coating dissolves at about pH 5.5. In an embodiment, the delayed release pellet coating comprises methyl acrylate-methacrylic acid copolymer, ethyl acrylate-methacrylic acid copolymer, hydroxy propyl methyl cellulose acetate succinate or cellulose acetate phthalate. In a convenient embodiment, the delayed release pellet coating comprises methyl acrylate-methacrylic acid copolymer or ethyl acrylatemethacrylic acid copolymer. Conveniently, the delayed release pellet coating is selected from Eudragit® L 100-55, Eudragit® FS30D, Eudragit® L100, Eudragit® L 12,5, Eudragit® L30 D- 55, Eudragit® S100 and Eudragit® S12,5, such as Eudragit® L 100-55. More conveniently, the delayed release pellet coating is selected from Eudragit® FS30D and Eudragit® L30 D-55.
[0205] In one embodiment, the delayed release (DR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 10% to about 80% (w/w) of the weight of the DR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 5% to about 50% (w/w) of the total weight of the DR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 5: 1 to 30: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 5: 1; c) optionally a sub-coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging from about 1 % to about 5 % (w/w) of the total weight of the DR pellet; and d) a delayed release layer that encapsulates the optionally sub-coated drug layered core comprising methyl acrylate-methacrylic acid copolymer and/or ethyl acrylate-methacrylic acid copolymer and optionally an anti-tacking agent (such as PlasAcryl® T20) in an amount ranging from about 10 % to about 40 % (w/w) of the total weight of the DR pellet.
[0206] In one embodiment, the delayed release (DR) multiparticulates/pellets comprise: a) an inert core in an amount ranging from about 40% to about 60% (w/w) of the weight of the DR pellet; b) a drug layer that encapsulates the inert core comprising a mixture of Compound 1 or a pharmaceutically acceptable salt thereof, a wetting agent (such as for example pol oxamer 188) and optionally a binder (such as for example hydroxypropyl methylcellulose) in an amount ranging from about 15% to about 35% (w/w) of the total weight of the DR pellet, wherein the weight ratio of Compound 1 to the wetting agent ranges from about 15: 1 to 25: 1 and the weight ratio of Compound 1 to the binder (when present) ranges from about 1 : 1 to 3: 1; c) optionally a sub-coating layer that encapsulates the drug layered core comprising a polymer (such as HPMC or PVA) in an amount ranging from about 2 % to about 4 % (w/w) of the total weight of the DR pellet; and d) a delayed release layer that encapsulates the optionally sub-coated drug layered core comprising methyl acrylate-methacrylic acid copolymer and/or ethyl acrylate-methacrylic acid copolymer and optionally an anti-tacking agent (such as PlasAcryl® T20) in an amount ranging from about 15 % to about 30 % (w/w) of the total weight of the DR pellet.
[0207] In one aspect of this embodiment, a sub-coat can optionally be applied between the drug layer and the modified release layer if separation is needed. In one aspect of this embodiment, the coat can be comprised of HPMC or magnesium stearate.
[0208] Conveniently, the IR or DR multiparticulates or pellets range in size from about 150 um to about 1000 pm, more conveniently about 350 pm.
[0209] The multiparticulate modified release composition will comprise a plurality of IR pellets and a plurality of DR pellets that may be packaged, for example, in a capsule such as a gelatin capsule or a capsule formed from a polymer such as HPMC AS, HPMC or starch; dosed as a suspension or slurry in a liquid; dosed in a sachet; or they may be formed into a tablet (e.g. a caplet) or pill by compression or other processes known in the art. Conveniently, the multiparticulate modified release composition comprising the plurality of pellets is packaged in a capsule, such as a gelatin capsule or a capsule formed from a polymer such as HPMCAS, HPMC or starch.
[0210] As used herein and unless stated otherwise, it is to be understood that when using the term “ bead or " beads ” in relation to a multiparticulate formulation, the terms are used synonymously with the terms “ pellet ” or “ pellets ”, respectively.
Mini-tablets
[0211] In a convenient embodiment of the modified release composition of the present invention, the multiparticulate system comprises a plurality of immediate release mini -tablets and a plurality of delayed release mini-tablets encapsulated in a capsule. Surprisingly, it has been found that mini-tablets have benefits compared to other multiparticulate systems in that they enable much higher drug loadings (up to approximately 64% w/w as described herein for Example 27 and 28) of Compound 1 to be achieved without detrimental impact of disintegration and/or dissolution properties. This is particularly surprising for an active compound such as Compound 1, which is particularly prone to agglomeration and poor disintegration/dissolution properties, even at relatively low drug loadings from compositions such as direct compression tablets. High drug loadings are advantageous in terms of dosage form size and patient convenience and compliance. This was not previously possible with other compositions and formulations; for example immediate release pellets were prepared with approximately 20% w/w drug loadings (Examples 19 and 20).
[0212] In a convenient embodiment, the modified release composition comprises an immediate release component comprising mini-tablets and a delayed release component comprising mini -tablets. Conveniently, the mini -tablets having an average diameter of 1.5 to 3.0 mm, such as 2.0 to 2.5 mm.
[0213] In an embodiment, the mini -tablets comprise a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients. In an embodiment, the mini-tablets comprise a granulate, wherein the granulate comprises the hydrochloride salt of Compound 1 (conveniently as the mono-hydrate) and one or more pharmaceutically acceptable excipients.
[0214] For the avoidance of doubt, in the embodiments described herein, the amounts refer to the Compound 1 free base amount, as the presence and nature of the salt and the presence of any solvates or hydrates will affect the total amount required to provide the free base amount; for example, 135 mg of the mono-hydrate hydrochloride salt of Compound 1 is equivalent to 120 mg of the parent free base. In an embodiment, the mini-tablets comprise a pharmaceutically acceptable salt of Compound 1 in a loading of greater than 40 % w/w (such as greater than 45 % w/w, greater than 50 % w/w, greater than 55 % w/w, or greater than 60 % w/w). In an embodiment, the mini-tablets comprise a pharmaceutically acceptable salt of Compound 1 in a loading of 40 to 75% w/w (such as 50 to 70 % w/w, 60 to 70 % w/w, 62 to 68 % w/w, or about 64 to 65 % w/w). In an embodiment, each mini-tablet comprises about 6 to 10 mg (such as about 8 to 9 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the immediate release component comprises 10 to 20 mini-tablets, such as 14 to 16 mini-tablets. In an embodiment, the delayed release component comprises 10 to 20 mini-tablets, such as 14 to 16 mini-tablets. In an embodiment, the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1. In an embodiment, the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1. In an embodiment, the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1 and the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a pharmaceutically acceptable salt of Compound 1. In an embodiment, the immediate release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1 and the delayed release component comprises 80 to 160 mg (such as 100 to 140 mg, or about 120 mg) of a hydrochloride salt of Compound 1.
[0215] In an embodiment, the mini -tablets comprise a granulate, wherein the granulate comprises the hydrochloride salt of Compound 1 (conveniently as the mono-hydrate) and a wetting agent. Conveniently, the wetting agent is a non-ionic wetting agent, optionally selected from polyol esters, polyoxyethylene esters and poloxamers. Polyol esters include glycol esters, glycerol esters and sorbitan derivatives (such as polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80). Conveniently, the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25, such as between 12 and 18. Most conveniently, the wetting agent is polysorbate 80. Even when high loadings of Compound 1 have been achieved in mini-tablets, it has been discovered that the levels of the wetting agent can still be low and the mini-tablets demonstrate good dissolution and disintegration in aqueous media. In an embodiment, the wetting agent is present in the composition at 0.5 to 3.0% w/w, such as 1.5 to 2.5% w/w.
[0216] To provide mini -tablets with high drug loadings by wet granulation methods, it has been discovered that the addition of extra-granular lubricants is important to provide a granulate that has the desired flow properties and compressibility and does not stick during tableting. In an embodiment, the granulate comprises one or more lubricants, conveniently wherein the one or more lubricants are selected from magnesium stearate, calcium stearate, hydrogenated vegetable oil, stearic acid, sodium stearyl fumarate, mineral oil, hydrogenated vegetable oil, and polyethylene glycol. In an embodiment, the one or more lubricants are magnesium stearate and hydrogenated vegetable oil. In an embodiment, the one or more lubricants are magnesium stearate and sodium stearyl fumarate. In an embodiment, the one or more lubricants are present in the composition at a total level of between 3 and 7% w/w, such as between 4 and 6% w/w. In an embodiment, magnesium stearate is present in the composition at between 2 and 4% w/w, and hydrogenated vegetable oil is present in the composition at between 2 and 4% w/w. In an embodiment, magnesium stearate is present in the composition at between 2 and 4% w/w, and sodium stearyl fumarate is present in the composition at between 2 and 4% w/w.
[0217] In an embodiment, the one or more pharmaceutically acceptable excipients does not comprise microcrystalline cellulose. In an embodiment, the one or more pharmaceutically acceptable excipients are not acidic, or have a pH of greater than 6 when dissolved or slurried in water. By avoiding acidic excipients, the stability of Compound 1 in the composition is improved.
[0218] In an embodiment, the granulate used to provide the mini-tablets is prepared by fluid bed granulation.
[0219] When the delayed release component comprises mini-tablets, conveniently the delayed release mini-tablets are individually substantially coated with a delayed release coating. In an embodiment, the delayed release coating dissolves at pH values greater than about 4.5, such as greater than about pH 5.5. In an embodiment, each delayed release mini -tablet comprises a greater than 6% (such as between 8 and 12%, or about 10%) weight gain delayed release coating.
[0220] Therefore, according to an embodiment of the present invention there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets, the mini-tablets comprising a hydrochloride salt of Compound 1.
[0221] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets, the mini-tablets comprising 40-75 % w/w of a hydrochloride salt of Compound 1.
[0222] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets, the mini -tablets comprising 40-75 % w/w of a hydrochloride salt of Compound 1; wherein each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core.
[0223] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1.
[0224] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of wetting agent. [0225] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the one or more pharmaceutically acceptable excipients in the granulate or extragranulate component have a pH of greater than 6 (such as greater than 6.5), when a 5 g sample of the excipient is dissolved or slurried with 40 ml of water for 20 minutes, centrifuged and the pH of the solution or supernatant is measured.
[0226] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of wetting agent; wherein the composition does not comprise microcrystalline cellulose. [0227] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of wetting agent; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants.
[0228] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80.
[0229] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 3-7 % w/w of one or more lubricants.
[0230] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of hydrogenated vegetable oil.
[0231] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1; and
• 1-3 % w/w of polysorbate 80; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 2-4 % w/w of magnesium stearate; and
• 2-4% w/w of sodium stearyl fumarate.
[0232] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini -tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 50-70 % w/w of a hydrochloride salt of Compound 1;
• 10-30 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant; 1-4 % w/w of disintegrant; and
3-7 % w/w of one or more lubricants.
[0233] In an embodiment, there is provided an oral modified release pharmaceutical composition, wherein the composition comprises an immediate release component and a delayed release component, wherein each component is formulated as mini-tablets; wherein each immediate release mini-tablet comprises a granulate component and an extragranulate component; and each delayed release mini-tablet comprises a core and a delayed release coating substantially encapsulating the core, wherein the core comprises a granulate component and an extra-granulate component; wherein the immediate release mini-tablets and the delayed release mini-tablets comprise in the granulate component:
• 60-70 % w/w of a hydrochloride salt of Compound 1;
• 15-25 % w/w of one or more diluents;
• 2-4 % w/w of disintegrant;
• 2-4 % w/w of binder; and
• 1-3 % w/w of wetting agent; and the immediate release mini-tablets and the delayed release mini-tablets comprise in the extra-granulate component:
• 0.1 -0.5 % w/w of glidant;
• 1-3 % w/w of disintegrant; and
• 4-6 % w/w of one or more lubricants.
[0234] Conveniently, in any of the above delayed release mini-tablet embodiments, the delayed release coating dissolves at pH values greater than about 5.5, such as greater than about 6.0. Conveniently, in any of the above delayed release mini-tablet embodiments, each mini-tablet comprises a greater than 6% weight gain (such as a greater than 8% weight gain) delayed release coating. Pharmacokinetics/Pharmacodynamics
[0235] When administered orally, the combination according to the first aspect or the modified release composition according to the second aspect of the invention can provide synergy in the form of high gastrointestinal exposure with acceptable safety and/or tolerability.
[0236] The Applicants have discovered that Compound 1 appears to undergo enterohepatic recirculation. On oral dosing of an immediate release composition comprising the Compound 1 or a pharmaceutically acceptable salt thereof, the compound is rapidly absorbed and metabolized in the liver yielding a glucuronide conjugate, which is subsequently subjected to biliary excretion into the intestine. It is expected that certain levels of the glucuronide conjugate pass through the gut lumen where P -glucuronidase enzymes hydrolyze the glucuronide back into the parent compound. The parent drug can be distributed to the intestinal tract tissues allowing absorption into the colonic tissue. This glucuronide conjugate, also referred to as the ‘glucuronide metabolite’ of Compound 1 has the following chemical structure:
Figure imgf000060_0001
Compound 1 glucuronide metabolite
[0237] It is believed that other conjugates such as glycoside conjugates are also formed and could also contribute to the increase in colonic tissue levels. The structure of the glucoside metabolite of Compound 1 is shown below:
Figure imgf000061_0001
. mula: C28H38CIN3O9
OH Mass: 595.2 g/mol
Compound 1 glucoside metabolite
[0238] The unique contribution of this enterohepatic recirculation from an immediate release component, along with the luminal drug levels provided by a delayed release component, allows particularly high colonic tissue exposure to be achieved with acceptable safety and tolerability.
[0239] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 50 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 20 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 15 ng/ml, such as less than 10 ng/ml.
[0240] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 2 ng/ml and 100 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is between 2 ng/ml and 50 ng/ml or 2 ng/ml and 30 ng/ml or 5 ng/ml and 20 ng/ml.
[0241] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject of 100 mg to 1000 mg of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 50 ng/ml, such as less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
[0242] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject of 200 mg to 1000 mg (such as 240 mg to 960 mg, or about 480 mg, or about 960 mg) of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml or 50 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
[0243] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject of 200mg to 1000 mg of Compound 1 or a pharmaceutically acceptable salt thereof, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, wherein about 50% of Compound 1 is derived from the immediate release component and about 50% of Compound 1 is derived from the delayed release component. Conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 75 ng/ml or 50 ng/ml. More conveniently, the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml.
[0244] In an embodiment, a combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml. Conveniently, the combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml. Conveniently, the combination according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 15 and 35 ng/ml, such as between 20 and 30 ng/ml, or between 22 and 28 ng/ml.
[0245] In an embodiment, a combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml. Conveniently, the combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 40 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml. Conveniently, the combination according to the present invention provides, after oral daily administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 20 and 40 ng/ml, such as between 25 and 35 ng/ml, or between 27 and 33 ng/ml.
[0246] In an embodiment, a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml. Conveniently, a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml. Conveniently, a modified release composition according to the present invention provides, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 15 and 35 ng/ml, such as between 20 and 30 ng/ml, or between 22 and 28 ng/ml.
[0247] In an embodiment, a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml, such as less than 75 ng/ml or less than 50 ng/ml. Conveniently, a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml, 20 ng/ml, 15 ng/ml or 10 ng/ml. Conveniently, a modified release composition according to the present invention provides, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject in need of such treatment, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of between 20 and 40 ng/ml, such as between 25 and 35 ng/ml, or between 27 and 33 ng/ml.
[0248] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1. Conveniently, the ratio of Cmax/AUCo-24 is between 0.04 and 0.3 h'1, such as between 0.04 and 0.2 h'1 or between 0.06 and 0.16 h'1.
[0249] In an embodiment, a combination according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1, such as between 0.04 and 0.3 h'1, between 0.04 and 0.2 h" between 0.06 and 0.16 h'1, between 0.2 and 0.35 h'1, or between 0.25 and 0.30 h'1.
[0250] In an embodiment, a modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1, such as between 0.04 and 0.3 h'1, between 0.04 and 0.2 h'1 , between 0.06 and 0.16 h'1, between 0.2 and 0.35 h'1, or between 0.25 and 0.30 h'1.
[0251] In an embodiment, a combination according to the present invention produces, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as 480 mg contained within an immediate release composition and 480 mg contained within a delayed release composition) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1, such as between 0.04 and 0.3 h'1, between 0.04 and 0.2 h" between 0.06 and 0.16 h'1, between 0.1 and 0.2 h'1, or between 0.12 and 0.19 h'1.
[0252] In an embodiment, a modified release composition according to the present invention produces, after oral administration in the fed state of 960 mg of Compound 1 or a pharmaceutically acceptable salt thereof (conveniently as a composition comprising the immediate release component and the delayed release component in a w/w ratio of about 1 : 1) to a subject, a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1, such as between 0.04 and 0.3 h'1, between 0.04 and 0.2 h'1 , between 0.06 and 0.16 h'1, between 0.1 and 0.2 h'1, or between 0.12 and 0.19 h'1.
[0253] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml (conveniently less than 75 ng/ml, 50 ng/ml, 30 ng/ml, 20 ng/ml, 15ng/ml or 10 ng/ml) and a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 50 ng/ml (conveniently less than 30 ng/ml, 20 ng/ml, 15ng/ml or 10 ng/ml) and a ratio of Cmax/AUCo-24 between 0.04 and 0.4 h'1. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 30 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.3 h'1. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 15 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.2 h'1. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 10 ng/ml and a ratio of Cmax/AUCo-24 between 0.04 and 0.16 h'1.
[0254] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml and a geometric mean maximum plasma concentration (Cmax) of the glucuronide metabolite of Compound 1 of greater than 500 ng/ml. Conveniently, the geometric mean maximum plasma concentration (Cmax) of the glucuronide metabolite of Compound 1 is greater than 1000 ng/ml, 1500 ng/ml, 2000 ng/ml, 2500 ng/ml, 3000 ng/ml, 5000 ng/ml, 6000 ng/ml or 8000 ng/ml.
[0255] In an embodiment, a combination or modified release composition according to the present invention provides, after oral administration in the fed state to a subject, an area under the plasma concentration-time curve from time zero up to 24 hours post-dose (AUC0-24) of the glucuronide metabolite of Compound 1 of greater than 10,000 ng*h/mL, conveniently greater than 15,000, 25,000, 35,000, 50,000, 70,000, or 100,000 ng*h/mL. [0256] In an embodiment, the combinations according to the first aspect or the modified release composition according to the second aspect of the invention advantageously provide high colon tissue exposure of Compound 1 after oral dosing. Colon tissue concentrations can be measured by biopsy as described in the Examples section. In an embodiment, the combination or modified release composition as described herein provides colonic tissue exposure greater than or equal to the systemic exposure of Compound 1 following oral solution dosing of the composition to a subject. Advantageously, the combination or modified release composition as described herein delivers higher median levels of Compound 1 to the colon after oral dosing, than achieved with a corresponding dose formulated as an oral solution. In an embodiment, the combination or modified release composition as described herein provides median sigmoid colon tissue concentrations of greater than 100 ng/g (such as greater than 140 ng/g) after oral dosing to a fasted subject. In an embodiment, the combination as described herein provides median sigmoid colon tissue concentrations of greater than 100 ng/g (such as greater than 140 ng/g) after oral dosing to a fed subject.
[0257] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations of Compound 1 greater than 100 ng/g. Conveniently, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations of Compound 1 greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g.
[0258] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
[0259] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg or 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
[0260] In an embodiment, a combination according to the present invention produces, after oral administration in the fed state of 480 mg daily Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 240 mg contained within an immediate release composition and 240 mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
[0261] In an embodiment, a combination according to the present invention produces, after oral administration in the fed state of 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 480mg contained within an immediate release composition and 480 mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
[0262] In an embodiment, a modified release composition according to the present invention produces, after oral administration in the fed state of 480 mg or 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently wherein about 50% of Compound 1 is derived from the immediate release component and about 50% of Compound 1 from the delayed release component of the composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g).
[0263] In an embodiment, a modified release composition according to the present invention produces, after oral administration in the fed state of 960 mg daily of Compound 1 or a pharmaceutically acceptable salt thereof to a subject (conveniently as 480mg contained within an immediate release composition and 480mg contained within a delayed release composition), colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g (such as greater than 150 ng/g, greater than 200 ng/g, greater than 300 ng/g or greater than 400 ng/g). [0264] Colonic biopsies may also allow for measurement of the proportion of hypoxia- inducible factor (HIF)-la positive cells in the biopsy samples both before (baseline) and after (e.g. at day 7) treatment with a delayed release solid composition as described herein. In an embodiment, a delayed release composition as described herein provides a greater than 10% (such as greater than 20%) increase from baseline in the proportion of HIF-la positive cells in the colon after oral dosing of the composition to a subject. In an embodiment, a delayed release composition as described herein provides a greater than 10% (such as greater than 20%) increase from baseline in the proportion of HIF-la positive cells in the colon after oral dosing of the composition comprising at least 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject. In an embodiment, a delayed release composition as described herein provides a greater than 10% (such as greater than 20%, greater than 30%, greater than 40%, or greater than 50%) increase from baseline in the proportion of HIF-la positive cells in the sigmoid colon tissue after oral dosing of the composition comprising at least 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof to a subject.
[0265] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration, an increase in the proportion of HIF-la positive cells from baseline in greater than 10% of subjects, such as greater than 20% of subjects or greater than 30% of subjects.
[0266] The combinations or modified release compositions according to the present invention are capable of delivering high colonic tissue exposure of Compound 1 after oral dosing, without producing substantial side effects, which might be expected were the same dose of Compound 1 administered solely via an immediate release composition. High systemic exposure of Compound 1 has the potential to trigger elevation of systemic levels of erythropoietin (EPO) and vascular endothelial growth factor (VEGF), which may lead to undesired side effects including those associated with increased hemoglobin and hematocrit (such as polycythemia following chronic exposure).
[0267] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g; plasma VEGF levels less than 500 pg/ml and plasma EPO levels less than 100 mIU/ml. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 200 ng/g; plasma VEGF levels less than 250 pg/ml and plasma EPO levels less than 50 mIU/ml. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 300 ng/g; plasma VEGF levels less than 150 pg/ml and plasma EPO levels less than 30 mIU/ml.
[0268] In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 100 ng/g; and after 12 weeks daily dosing of Compound 1 fewer than 50% of subjects experience a treatment related adverse event. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 200 ng/g; and after 12 weeks daily dosing of Compound 1 or a pharmaceutically acceptable salt thereof fewer than 40% of subjects experience a treatment related adverse event. In an embodiment, a combination or modified release composition according to the present invention produces, after oral administration in the fed state to a subject, colonic tissue concentrations (such as colonic sigmoid tissue concentrations) of Compound 1 greater than 300 ng/g; and after 12 weeks daily dosing of Compound 1 or a pharmaceutically acceptable salt thereof fewer than 30% of subjects experience a treatment related adverse event.
Therapeutic Uses
[0269] In a further aspect, the present invention also provides the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, for use in the treatment of diseases or conditions mediated alone, or in part, by PHD.
[0270] Conveniently, there is provided a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammatory bowel disease. Conveniently, the treatment is for ulcerative colitis or Crohn’s disease.
[0271] Conveniently, there is provided a modified release composition as described herein for use in the treatment of inflammatory bowel disease. Conveniently, the treatment is for ulcerative colitis or Crohn’s disease.
[0272] The present invention provides a method of treating diseases or conditions mediated alone, or in part, by PHD, the method comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof. Conveniently, there is provided a method of treating an inflammatory bowel disease, the method comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof. Conveniently, there is provided a method of treating ulcerative colitis or Crohn’s disease, the method comprising administering to a subject a therapeutically effective combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
[0273] The present invention provides a method of treating diseases or conditions mediated alone, or in part, by PHD, the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof. Conveniently, there is provided a method of treating an inflammatory bowel disease, the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof. Conveniently, there is provided a method of treating ulcerative colitis or Crohn’s disease, the method comprising administering to a subject a therapeutically effective amount of a modified release composition as described herein comprising Compound 1 or a pharmaceutically acceptable salt thereof. [0274] The present invention provides for the use of a combination according to the first aspect of the invention, or a modified release composition according to the second aspect of the invention, in the manufacture of a medicament for treating diseases or conditions mediated alone, or in part, by PHD. Conveniently, the disease or condition is inflammatory bowel disease. Conveniently, the disease or condition is ulcerative colitis or Crohn’s disease.
[0275] In a convenient embodiment of any of the therapeutic uses described herein, the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, is administered once or twice daily. In an embodiment, the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, is administered orally once daily. In an embodiment, the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, is administered orally once daily in the morning. In an embodiment, the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, is administered orally twice daily.
[0276] In a convenient embodiment of any of the therapeutic uses described herein, the combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, is administered with food, or up to 30 minutes after a meal. In an embodiment, the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal.
[0277] In a convenient embodiment of any of the therapeutic uses described herein, the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal, once a day. In an embodiment, the combination or the modified release composition is administered orally with food, or up to 30 minutes after a meal, twice a day.
[0278] In a convenient embodiment of any of the therapeutic uses described herein, the combination or the modified release composition is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960 mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg.
[0279] In a convenient embodiment of any of the therapeutic uses described herein, the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg; and wherein about 50% of Compound 1 is derived from the immediate release composition and about 50% of Compound 1 is derived from the delayed release composition. In a convenient embodiment of any of the therapeutic uses described herein, the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 480 mg; and wherein the immediate release composition comprises about 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof and about the delayed release composition comprises about 240 mg of Compound 1 or a pharmaceutically acceptable salt thereof. In a convenient embodiment of any of the therapeutic uses described herein, the combination is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 960 mg; and wherein the immediate release composition comprises about 480 mg of Compound 1 and about the delayed release composition comprises about 480 mg of Compound 1.
[0280] In a convenient embodiment of any of the therapeutic uses described herein, the modified release composition is administered orally wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg, such as between 120 mg and 960mg, between 480 mg and 960 mg, about 480 mg, or about 960 mg; and wherein about 50% of Compound 1 or a pharmaceutically acceptable salt thereof is derived from the immediate release component and about 50% of Compound 1 or a pharmaceutically acceptable salt thereof is derived from the delayed release component.
[0281] In a further embodiment, the combination or the modified release composition may be administered in further combination with one or more additional treatments. Conveniently, the combination or the modified release composition may be administered with one or more additional treatments for inflammatory bowel disease (ulcerative colitis or Crohn’s disease). In an embodiment, the one or more additional treatments are selected from 5- aminosalicylates (5-ASAs) and steroids. In a more convenient embodiment, the 5-ASAs are selected from sulphasalazine, mesalazine and olsalazine. In a more convenient embodiment, the steroids are selected from prednisone and budesonide. Ulcerative Colitis
[0282] The combination according to the first aspect of the invention, or the modified release composition according to the second aspect of the invention, are particularly useful in the treatment of ulcerative colitis. In an embodiment, the ulcerative colitis being treated is mild-to- moderate ulcerative colitis. Suitable ulcerative colitis patients to be treated with the combinations or the modified release compositions described herein, include those having a Mayo score of 5- 10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1 (the Examples contain details of these diagnostic assessments).
[0283] Therefore, in an embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein and wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency sub score of> 1.
[0284] Conveniently, the ulcerative colitis patient being treated is already receiving a different treatment for the condition. Most conveniently, the patient is already receiving an oral 5-aminosalicylate (5-ASA) drug for at least two weeks prior to the treatment commencing. Therefore, in an embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein and wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1, despite being on a stable dose of an oral 5-ASA for at least two weeks prior to the treatment.
[0285] 5 -ASA drugs may be given at different doses depending on a number of factors including the severity of the ulcerative colitis in a given patient. Patients with infrequent relapses may be treated with a low dose 5-ASA maintenance treatment, whereas higher doses (such as greater than or equal to 3 g per day) of 5-ASA may be needed in patients with mild or moderately active disease. As used herein, a ‘stable’ dose refers to the same dose of 5-ASA being administered to the patient with ulcerative colitis for at least two weeks prior to the treatment commencing and that same dose continuing during the treatment described in the present invention. In an embodiment that dose is greater than or equal to 3 g (such as 4 g) per day of 5-ASA. [0286] The efficacy of the claimed treatment for ulcerative colitis can be assessed by various measures, which include the treated patient having a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1. Therefore, in an embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1. In a convenient embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein; wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1; and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1. In a convenient embodiment, there is provided a method for treating ulcerative colitis, the method comprising administering to the subject a combination, or a modified release composition, as described herein; wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1 and a stool frequency subscore of > 1, despite being on a stable dose of an oral 5-ASA for at least two weeks prior to the treatment; and wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0 and a stool frequency subscore of 0 or 1.
EXAMPLES
EXAMPLE 1 : COMPOUND 1 SOLUBILITY
[0287] The solubility of Compound 1 at various pH's was investigated. The results are presented in Table 1 and Figure 1, demonstrating that solubility is pH dependent. TABLE 1 : COMPOUND 1 PH-SOLUBILITY AT 22°C
Figure imgf000076_0001
EXAMPLE 2: FORMATION OF COMPOUND 1 HCL SALT
2, 1 Monohydrate (Form A)
[0288] Compound 1 free base (1.0 equiv.) and THF (4 vol.) were charged to a reactor and the contents were agitated for 20 minutes and then cooled to 3 °C. 1 M aq. HC1 solution (1.5 equiv) was slowly charged for 30 minutes while maintaining the internal temperature of 0 - 6 °C. The mixture was agitated for 20 minutes at 0-6 °C. The contents were filtered and washed with aq. THF solution (0.5 vol. of THF / 1 vol. of purified water) and purified water (6 vol.). The mixture was distilled under vacuum until target volume was achieved of approximately 10.0 L/kg (with respect to the free base) while maintaining the internal temperature of less than 25 °C. The slurry was adjusted to 10 °C and agitated for 1 hour. The product slurry was filtered and the wet cake was washed with pre-cooled (10°C) purified water (3 x 1 vol). The filtered cake was dried under vacuum at less than 35 °C.
[0289] HPLC analysis of Example 2.1 showed a purity of 99.6% by area. LC-MS demonstrated an observed a mass of 434.2 m/z +ve ionization, consistent with the expected mass of 433.93 g/mol. High Performance Liquid Chromatography-Charged Aerosol Detection (HPLC- CAD) analysis confirmed Example 2.1 to be a mono-HCl salt.
[0290] Thermogravimetric analysis of Example 2.1 showed an initial mass loss (from onset to ca. 80°C) of 3.4% related to loss of water (ca. 0.9 equiv). A loss of ca. 1 equivalent of water indicated Example 2.1 is a monohydrate. A second mass loss of 40.9% was observed (with an onset at ca. 160°C) related to the material melt and subsequent decomposition. Differential thermal analysis showed a shallow, broad endothermic event (onset at ca. 53°C) related to the loss of water. An initially sharp then broadening melt was recorded from an onset of ca. 190°C and a peak at ca. 194°C. DSC analysis showed a broad endothermic event with onset at ca. 70°C related to the loss of water during the first heating cycle (up to 150°C), as well as a sharp melt endothermic event with an onset at 191°C and peak at 194°C during the second heating cycle (up to 300°C). The melting point was consistent with TG/DTA.
[0291] HPLC analysis of Example 2.1 showed a purity of 99.6% by area. LC-MS demonstrated an observed a mass of 434.2 m/z +ve ionization, consistent with the expected mass of 433.93 g/mol. High Performance Liquid Chromatography-Charged Aerosol Detection (HPLC- CAD) analysis confirmed Example 2.1 to be a mono-HCl salt.
[0292] XRPD analysis was carried out on a PANalytical X’pert pro fitted with PIXcel detector, scanning the samples between 3 and 35° 29. The material was gently ground to release any agglomerates and loaded onto a multi-well plate with Kapton or Mylar polymer film to support the sample. The multi-well plate was then placed into the diffractometer and analyzed using Cu K radiation (al = 1.54060 A; a2 = 1.54443 A; P = 1.39225 A; al : a2 ratio = 0.5) running in transmission mode (step size 0.0130° 29) using 40 kV / 40 mA generator settings.
[0293] XRPD characterisation of Example 2.1 gave the peak list presented in Table 2 (referred to as Form A). Polarized Light Microscopy (PLM) showed small birefringent crystals (ca. 20- 100pm) with plate-like morphology.
TABLE 2: XRPD PEAK LIST OF EXAMPLE 2.1 (FORM A)
Figure imgf000077_0001
Figure imgf000078_0001
2.2 Anhydrous (Form B)
[0294] Compound 1 HC1 salt (1 eq; Example 2.1) and MeOH (5.5 vol.) were charged to a reactor and agitated at 20-25 °C for 10 minutes. The resulting beige slurry was then heated to 40 °C resulting in a clear, orange solution after 5-10 minutes. MTBE (8.5 vol.) was then added over 2 hours at a flow rate of 14.6 mL/minute. Following complete addition, the solution was slowly cooled to 2-7 °C at a rate of 0.1°C/minute. A pale beige slurry was observed and allowed to stir at this temperature for 16 hours. The resulting slurry was filtered, washed with MTBE (2 x 1 vol.), and dried at 40 °C under vacuum to afford a crystalline solid.
[0295] HPLC showed the material is 99.9% pure. PLM analysis showed the material to have a form of fine (ca. 5-20 pm), birefringent crystals with irregular morphology and agglomeration. TG analysis showed no solvent related mass loss, which correlates with Example
2.2 being an anhydrous form. DT analysis showed a sharp melting endotherm with an onset at ca. 193°C and peak at ca. 195°C, followed by thermal decomposition. DSC analysis showed no significant endothermic events related to solvent loss. An intense, sharp melting endotherm was observed with an onset at ca. 196°C and peak at ca. 198°C.
[0296] XRPD characterisation of Example 2.2 gave the peak list presented in Table 3 (referred to as Form B).
TABLE 3 : XRPD PEAK LIST OF EXAMPLE 2.2 (FORM B)
Figure imgf000078_0002
Figure imgf000079_0001
TABLET FORMULATION TRIALS
High Shear Wet Granulation Preparations
[0297] It was surprisingly found that high shear wet granulation processes intimately mix Compound 1 hydrochloride salt with solubility-enhancing excipients, such as surfactants, disintegrants and binders, to form dense homogenous granules suitable for tableting when combined with additional extra-granular components. When a tablet produced by this process disintegrates, the granules containing Compound 1 • HC1 and other excipients are dispersed within the media and subsequently disintegrate themselves to liberate Compound 1 into the media. This two-stage disintegration provides superior dispersion throughout the media and has been found to surprisingly avoid the formation of API agglomerates.
[0298] During early direct compression trials wetting agents or surfactants were found to have negative or minimal effects on the disintegration and/or dispersal of Compound 1 hydrochloride salt: the omission of SLS from the formulation did not have a negative impact on these properties; addition of 1% Polysorbate 80 to the pH 6.8 buffer media did not improve the dispersion characteristics, so was not considered for inclusion in the direct compression formulations; and whilst pol oxamer 188 was found to improve the aqueous dispersion of Compound 1 • HC1, nevertheless, when it was incorporated in the direct compression formulations it was found to be hindering tablet disintegration. It was therefore surprising that when the Applicants attempted to use certain wetting agents in granule formulations, they unexpectedly found that certain benefits were observed.
[0299] During the wet granulation process a wetting agent may advantageously be used to improve the disintegration and dispersal of Compound 1 hydrochloride salt from the resultant composition. The wetting agent is dissolved in water or an aqueous medium and added to the shear-blended formulation to assist in granulation. It is postulated that when a wetting agent is intimately mixed with Compound 1 hydrochloride salt within a granulate composition, then the two-stage disintegration of the composition and dispersal of the API in aqueous media is promoted.
[0300] The wetting agent may be a surfactant or emulsifier. Conveniently the wetting agent is a non-ionic surfactant. Conveniently, the wetting agent has a hydrophilic-lipophilic balance (HLB) between 10 and 25 (such as between 12 and 18). Suitable examples of non-ionic wetting agents include polyol esters, polyoxyethylene esters and poloxamers. Examples of polyol esters include glycol esters, glycerol esters and sorbitan derivatives. Sorbitan derivates comprise polysorbates (such as polysorbate 20, polysorbate 40, polysorbate 60 and polysorbate 80). Most conveniently the wetting agent is polysorbate 80.
EXAMPLE 3: CORE TABLETS 1-5
[0301] Five small-scale (batch size 250g) high shear granulation formulations, F1-F5, were prepared according to the core tablet formulations provided in Table 4 and the process described in Figure 2. Despite no benefit of Polysorbate 80 in direct compression tablets, Polysorbate 80 was introduced as a surfactant intended to assist in "wetting" Compound 1 • HC1 in aqueous media in the granule formulations. All formulations were successfully granulated and blended to produce free-flowing granulations suitable for tablet compression. The granulation process improved the flow of the final blend. Formulations F2, F3, F4 & F5 compressed well, while Fl could not be compressed due to insufficient hardness, to produce plain, white to off white, round, 11mm diameter, shallow biconvex, bevel edge tablet. The weight uniformity of the tablets was good.
[0302] Hardness values and disintegration times, in water and pH 6.8 buffer, of formulations F2-F5 are provided in Table 5. Disintegration times increased, as compared to the direct compression formulations, and were similar in water and pH 6.8 buffer.
TABLE 4: HIGH SHEAR GRANULATION FORMULATIONS Fl -5
Figure imgf000081_0001
TABLE 5: F2-F5 TABLET CHARACTERISTICS
Figure imgf000081_0002
EXAMPLE 4: CORE TABLETS F6, F7 AND F8
[0303] Formulations F6, F7 and F8 were prepared as detailed in Table 6 and the process described in Figure 2. These formulations incorporated the following modifications: using intra- granular microcrystalline cellulose (at approximately twice the level of intra-granular lactose monohydrate); removal of Starch 1500 from the formulation; and using HPC in place of PVP K29/32 as the primary binder. An Opadry® Clear 03K19229 sub coat was applied to F7, to produce F7-SC1, which was then enterically coated with Acryl-Eze® White 93018359, to produce F7-SC1-EC2. An immediate release Opadry® II White 85F 18422 coat was applied to F8 to a weight gain of 4%, to produce F8-IRC2.
TABLE 6: FORMULATIONS F6, F7 AND F8
Figure imgf000082_0001
[0304] F7 and F8 were compressed at similar hardness levels to produce plain, white to off-white, round, 11 mm Diam., shallow biconvex, bevel edge tablets. F6 tablets were compressed at three hardness levels to produce plain, white to off white, round, 11mm diameter, shallow biconvex, bevel edge tablets.
[0305] A summary of the physical blend characteristics of the high shear wet granulation formulations F6, F7 and F8 is provided in Table 7.
[0306] The bulk densities of the wet granulated formulations are high. As a result, the calculated Carr’s Index values and Hausner ratios are significantly than lower than formulations prepared by direct compression - indicating a free-flowing material. The high shear granulation produced spherical granules with a relatively coarser distribution in comparison to the direct compression blends. TABLE 7: PHYSICAL BLEND CHARACTERISTICS OF FORMULATIONS F6, F7 AND F8 PREPARED BY
HIGH SHEAR WET GRANULATION
Figure imgf000083_0001
[0307] Physical Characteristics of F6, F7, F8 Tablets are presented in Table 8. Tablet friability was similar at all hardness values. Disintegration times varied significantly, increasing as hardness increased.
TABLE 8: PHYSICAL CHARACTERISTICS OF F6, F7, F8 TABLETS
Figure imgf000083_0002
[0308] Disintegration studies were performed on F8-IRC2 tablets in pH 6.8 buffer and all tablets were found to disintegrate and disperse in under 4 minutes.
[0309] Disintegration studies were performed on F7-SC1-EC2 tablets in 0. IN HC1 and pH 6.8 buffer. After 2 hrs in 0. IN HC1 all the tablets were intact and gave an average acid uptake value of 2.9%. After returning the acid treated tablets to the USP disintegration apparatus and placing in pH 6.8 phosphate buffer media, all tablets disintegrated within approximately 10.5 minutes with no agglomerates remaining. [0310] In a parallel disintegration study on F7-SC1-EC2 tablets wherein the pH 6.8 buffer also contained 2% CTAB it was observed that it took over 1 hour and 5 minutes before all tablets fully disintegrated. No agglomerates were recovered and the tablet contents appeared to be well dispersed. Therefore, CTAB significantly slowed the disintegration of the enteric coated tablets.
Conclusion from wet granulation vs direct compression preparations
[0311] Although a direct compression manufacturing process for Compound 1 hydrochloride salt is physically viable, and an uncoated direct compression tablet can be developed that disintegrates very rapidly, the generally unaltered API liberated from coated tablets produced with this process does not adequately disperse in non-acidic media where API solubility is extremely low, even when a non-functional film coat is used. An optimized high shear wet granulation manufacturing process is viable for Compound 1 hydrochloride salt and effectively alters the physical characteristics of the API when combined with other components within the granulation, allowing the API from uncoated or coated tablets produced with this process to adequately disperse in all media. A coating is required to protect the API from exposure to light.
EXAMPLE 5: IMMEDIATE RELEASE (IR) TABLETS
[0312] The 60 mg IR tablets prepared according to Example 5 were used in the Phase la clinical study described in Example 14.
[0313] 60mg IR tablets present as white to off-white, round film-coated tablets. The strength of the active dosage form is 60mg (calculated as the anhydrous free base form of Compound 1). The composition of the Tablets and a representative batch for Compound 1 IR Tablets is provided in Table 9. The typical batch blend size is 5.5 kg. The theoretical batch size is 13,750 tablets. TABLE 9: COMPOSITION AND BATCH FORMULA FOR COMPOUND 1 IR TABLETS
Figure imgf000085_0001
NOTE: Water is employed as a granulating agent and to prepare the coating suspension. Water evaporates during drying and the coating process.
[0314] The various components used in the formulation may be acquired from standard formulation excipient suppliers, such as: Lactose Monohydrate (Foremost #310);
Microcrystalline Cellulose (Avicel PH-102); Hydroxypropyl Cellulose (Klucel EXF Pharm); Crospovidone (Kollidon CL); Silicified Microcrystalline Cellulose (Prosolv SMCC HD 90); Crospovidone (Kollidon CL); Magnesium Stearate (Ligamed MF-2-V).
[0315] 60mg IR Tablets were prepared via a high-shear wet granulation process. A summary of the process is provided below; a flow diagram for the manufacturing process is provided in Figure 3.
1) Weigh and dispense materials.
2) Screen or pass the following materials through Comil: a) Lactose monohydrate b) Compound 1 • HC1 c) Crospovidone (intra-granular) d) Hydroxypropyl cellulose e) Microcrystalline cellulose
3) Premix the screened materials from Step 2 in a suitably sized high-shear granulator.
4) Dissolve Polysorbate 80 in purified water.
5) Granulate the blend from Step 3 using granulating liquid from Step 4 in suitably sized high-shear granulator.
6) Screen or pass the wet granules through Comil.
7) Dry the wet granules in suitably sized fluid bed dryer.
8) Screen or pass the dry granules through Comil.
9) Transfer the milled granules from Step 8 to a suitably sized blender.
10) Screen or pass the following materials through Comil: a) Silicified microcrystalline cellulose b) Crospovidone (extra-granular)
11) Blend the material from Step 9 with screened materials from Step 10.
12) Screen magnesium stearate through a suitably sized mesh.
13) Lubricate blend from Step 11 with screened magnesium stearate from Step 12 in a suitably sized blender.
14) Compress the blend from Step 13.
15) Dedust and metal check the tablets.
16) Prepare coating solution with Opadry® II (85F 18422) White.
17) Coat the tablets with Step 16 in suitably-sized pan.
[0316] The in-process controls applied at the compression stage (step 14) include individual tablet weight (370-430mg); individual tablet hardness (7-11 kilopond) and average weight of 10 tablets (3.800-4.200g).
EXAMPLE 6: DELAYED RELEASE (DR) TABLETS
[0317] The 60 mg DR tablets prepared according to Example 6 were used in the Phase la clinical study described in Example 14.
[0318] 60mg DR tablets present as white to off-white, round film-coated tablets. The strength of the active dosage form is 60mg (calculated as the anhydrous free base of Compound 1). The composition of the Tablets and a representative batch for Compound 1 DR Tablets is provided in Table 10. The typical batch blend size is 5.5 kg. The theoretical batch size is 13,750 tablets.
TABLE 10: COMPOSITION AND BATCH FORMULA FOR COMPOUND 1 DR TABLETS
Figure imgf000087_0001
NOTE: Water is employed as a granulating agent and to prepare the coating suspension. Water evaporates during drying and the coating process.
[0319] 60 mg DR Tablets were prepared via a high-shear wet granulation process. A summary of the process is provided below; a flow diagram for the manufacturing process is provided in Figure 4.
1) Weigh and dispense materials.
2) Screen or pass the following materials through Comil: a) Lactose monohydrate b) Compound 1 • HC1 c) Crospovidone (Intra-granular) d) Hydroxypropyl cellulose e) Microcrystalline cellulose
3) Premix the screened materials from Step 2 in a suitably sized high-shear granulator. 4) Dissolve Polysorbate 80 in purified water.
5) Granulate the blend from Step 3 using granulating liquid from Step 4 in suitably sized high-shear granulator.
6) Screen or pass the wet granules through Comil.
7) Dry the wet granules in suitably sized Fluid Bed Dryer.
8) Screen or pass the dry granules through Comil.
9) Transfer the milled granules from Step 8 into a suitably sized blender.
10) Screen or pass the following materials through Comil: a) Silicified microcrystalline cellulose b) Crospovidone (Extra-granular)
11) Blend the material from Step 9 with screened materials from Step 10.
12) Screen magnesium stearate through a suitably sized mesh.
13) Lubricate blend from Step 11 with screened magnesium stearate from Step 12 in a suitably sized blender.
14) Compress the blend from Step 13.
15) Dedust and metal check the tablets.
16) Prepare coating solution with Opadry® (03K19229) Clear.
17) Coat the tablets with Step 16 in suitably sized pan.
18)Prepare coating solution with Acryl-Eze® (93018359) White.
19) Coat the sub-coated tablets with solution from Step 18 in a suitably sized pan.
[0320] The in-process controls applied at the compression stage (step 14) include individual tablet weight (370-430mg); individual tablet hardness (7-11 kilopond) and average weight of 10 tablets (3.800-4.200g).
EXAMPLE 7: DISSOLUTION RATES OF IR AND DR TABLETS
[0321] The dissolution rates of the IR and DR Tablets comprising Compound 1 • HC1, prepared as described in Examples 5 and 6 herein, were determined as detailed in Tables 11 and 12, respectively. The results are presented in Table 13 and shown graphically in Figure 5.
TABLE 11 : DISSOLUTION PARAMETERS FOR 60 MG IR TABLETS
Figure imgf000088_0001
Figure imgf000089_0001
TABLE 12: DISSOLUTION PARAMETERS FOR 60MG DR TABLETS
Figure imgf000089_0002
(CTAB: cetyltrimethylammonium bromide)
TABLE 13 : DISSOLUTION OF IR AND DR TABLETS
Figure imgf000089_0003
EXAMPLE 8: FURTHER HIGH SHEAR WET GRANULATION TABLET FORMULATIONS - 60 MG TABLETS
[0322] Investigations were carried out to explore alternative high shear wet granulation compositions. Pregelatinized starch (Starch 1500) and spray dried lactose monohydrate (Flowlac 100) were used as diluents. Optimization studies to deliver IR and DR tablets containing 60 mg of parent compound per tablet led to the formulations detailed in Table 14 (formulations F9 and F10).
TABLE 14: 60MG IR (F9) AND DR (F10) FORMULATIONS
Figure imgf000090_0001
[0323] The cores of formulations F9 and F 10 were prepared according to a high shear wet granulation process as set out in Figure 6.
[0324] The F9/F10 core composition had the physical characteristics as set out in Table
15. TABLE 15: F9/F 10 BLENDS PHYSICAL CHARACTERISTICS
Figure imgf000091_0001
[0325] F9/F10 blended formulations were compressed to produce plain, white to off- white, round, 11 mm Diam., shallow biconvex, bevel edge tablets.
[0326] Physical Characteristics of F9 tablets are presented in Table 16. Core tablets were coated using Colorcon’s Opadry® II 85F 18422 White to a weight gain of 4% in an O’Hara Labcoat II fully-perforated coating pan (12” insert) using the following processing parameters:
• Quantity of tablet cores: 800g/pan load
• Conditions: o Spray nozzle: 1.0 mm o Gun Angle: 8 o’clock o Gun Distance: Approx. 4” o Atomization Air: 15 psi o Pattern Air: 20 psi o Air Volume: 130 CFM o Pan Speed: 20 RPM o Spray Rate: Approx. 8.4-9.0 g/min o Inlet Air Temp: 58.5-60.8 °C o Exhaust Air Temp: 44.6-48.8 °C o Coating Dispersion: 20% solids, 4% tablet weight gain applied.
TABLE 16: PHYSICAL CHARACTERISTICS OF F9 TABLETS
Figure imgf000092_0001
[0327] The physical characteristics of the resulting reformulated tablet cores were very good and similar to those of the original high shear formulation containing microcrystalline cellulose (F6, F7, F8). However, the maximum achievable hardness values were slightly lower.
EXAMPLE 9: ALTERNATIVE HIGH SHEAR WET GRANULATION TABLET FORMULATIONS - 120 MG TABLETS
[0328] A higher strength 120mg tablet was developed based on the 60 mg formulations from Example 8, using the same high shear granulation process while keeping the size of the 120 mg strength tablet as small as possible. Table 17 lists the excipient levels used for the IR (Fl 1) and DR (F12) formulations.
TABLE 17: 120MG IR (F 11 ) AND DR (F 12) FORMULATIONS
Figure imgf000093_0001
[0329] The core blends of formulations Fl 1 and F12 were prepared according to a high shear wet granulation process as set out in Figure 6. The amount of granulating solution containing dissolved HPC (wet portion) and Polysorbate 80 was applied in its entirety during mixing and incremental amounts of water were added to complete the granulation. A granulating solution rate of addition of 55 g/min was used for Fl 1 and a rate of approximately 65 g/min was used for F 12.
[0330] The Fl 1 and F 12 core compositions had the physical characteristics as set out in Table 18. TABLE 18: Fl 1 & F12 BLENDS PHYSICAL CHARACTERISTICS
Figure imgf000094_0001
[0331] Fl 1 and F 12 blended formulations were compressed to produce plain, white to off-white, caplet-shaped, biconvex, plain, 17.4 mm x 6.7 mm tablets. Physical Characteristics of Fl 1 & F12 tablets are presented in Table 19. Both formulations compressed very well. Weight uniformity was excellent, tablet hardness values remained consistent throughout the runs and friability was very low. The core tablets had disintegration times ranging from under 1 minute to approximately 1 minute and 45 seconds.
TABLE 19: PHYSICAL CHARACTERISTICS OF Fl 1 & F12 TABLETS
Figure imgf000094_0002
[0332] Fl 1 IR tablets were coated using Colorcon’s Opadry® II 85F 18422 White to a weight gain of 4% in an O’Hara Labcoat II fully-perforated coating pan (12” insert) using the following processing parameters:
• Quantity of tablet cores: 800g/pan load
• Conditions: o Spray nozzle: 1.0 mm o Gun Angle: 8 o’clock o Gun Distance: Approx. 4” o Atomization Air: 15 psi o Pattern Air: 20 psi o Air Volume: 130 CFM o Pan Speed: 20 RPM o Spray Rate: Approx. 8.8-10.0 g/min o Inlet Air Temp: 59.0-60.0 °C o Exhaust Air Temp: 44.6-47.6 °C o Coating Dispersion: 20% solids, 4% tablet weight gain applied.
[0333] Disintegration testing was performed on these Fl 1 IR tablets in pH 6.8 buffer and all tablets were found to disintegrate and disperse from 1 min to 1 min and 13 secs.
[0334] F12 DR tablets were prepared by sub-coating core tablets using Colorcon’s
Opadry® Clear 03K19229 to a weight gain of 4% in an O’Hara Labcoat II fully-perforated coating pan (12” insert) using the following processing parameters:
• Quantity of tablet cores: 800g/pan load
• Conditions: o Spray nozzle: 1.0 mm o Gun Angle: 8 o’clock o Gun Distance: Approx. 4” o Atomization Air: 15 psi o Pattern Air: 20 psi o Air Volume: 130 CFM o Pan Speed: 20 RPM o Spray Rate: Approx. 8.4-9.1 g/min o Inlet Air Temp: 57.0-62.0 °C o Exhaust Air Temp: 44.6-45.3 °C o Coating Dispersion: 10% solids, 4% tablet weight gain applied. [0335] Sub-coated tablets as prepared above were enteric-coated using Colorcon’s Acryl- Eze® 93018359 White to a weight gain of 14% in an O’Hara Labcoat II fully-perforated coating pan (12” insert) using the following processing parameters:
• Quantity of tablet cores: 800g/pan load
• Conditions: o Spray nozzle: 1.0 mm o Gun Angle: 8 o’clock o Gun Distance: Approx. 4” o Atomization Air: 15 psi o Pattern Air: 20 psi o Air Volume: 130 CFM o Pan Speed: 20 RPM o Spray Rate: Approx. 7.5-9.8g/min o Inlet Air Temp: 42.5-50.0 °C o Exhaust Air Temp: 35.4-37.0 °C o Coating Dispersion: 20% solids, 14.0% tablet weight gain applied.
[0336] Following two hours immersion in 0. IN HC1 in the USP disintegration apparatus and calculation of the acid uptake value, the acid treated tablets were returned to the USP disintegration apparatus and placed in pH 6.8 Phosphate Buffer media and observations were made related to their disintegration performance.
[0337] The F 12 DR tablets showed an average acid uptake of 3.1% and were intact with unchanged appearance after the acid treatment. Upon exposure to pH 6.8 buffer the coating dissolved after approximately 10.5 minutes and all the tablets had fully disintegrated after 11.5 minutes with no agglomerates remaining.
IR/DR TABLET STABILITY
EXAMPLE 10: STABILITY DATA ON TABLET FORMULATIONS
[0338] IR Tablet formulations prepared by high shear wet granulation were subjected to stability testing under various storage conditions and the purity of Compound 1 by HPLC was assessed initially and then after 1, 3 and 6 months as outlined in Table 20. TABLE 20: COMPOUND 1 HPLC STABILITY ASSAYS FOR IR TABLET FORMULATIONS
Figure imgf000097_0001
*assessed at different test sites
[0339] DR Tablet formulations prepared by high shear wet granulation (F7-SC1-EC2) were also subjected to stability testing under various storage conditions and the purity of Compound 1 by HPLC was assessed initially and then after 1, 3 and 6 months as outlined in Table 21.
[0340] The Comparator SC1-EC2 tablets had been prepared by direct compression, without water addition to the processing. Despite the F7-SC1-EC2 tablets having had water added during the wet granulation processing, this did not result in increased degradation being seen in the six-month stability study; in fact slightly more degradation was observed for the direct compression tablets than was seen with the wet granulation tablets. As can be seen from Table 21, after 6 months at 25 °C / 60% RH and at 40 °C / 75% RH, the levels of total related impurities seen with the direct compression tablets were 0.91% and 2.70% respectively. None of the IR or DR tablets prepared by high shear wet granulation had such high levels of related impurities after 6 months under comparable conditions. [0341] It would therefore appear that, surprisingly, the chemical stability of Compound 1 on storage is slightly better when formulated in wet granulation compositions according to the present invention, as opposed to comparable compositions prepared by direct compression.
TABLE 21 : COMPOUND 1 HPLC STABILITY ASSAYS FOR DR TABLET
FORMULATIONS
Figure imgf000098_0001
* assessed at different test sites a When tested at same test site, F7-SC1-EC2 and Comparator SC1-EC2 had 0.16% and 0.6% respectively of a related substance with RRT of 0.43-0.44 by HPLC c.f. Compound 1 b When tested at same test site, F7-SC1-EC2 and Comparator SC1-EC2 had 0.20% and 0.72% respectively of a related substance with RRT of 0.43-0.44 by HPLC c.f. Compound 1 CLINICAL TRIALS
EXAMPLE 11 : A PHASE 1 A RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, SINGLE ASCENDING DOSE CLINICAL STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF COMPOUND 1 (SOLUTION) IN HEALTHY MALE VOLUNTEERS.
[0342] Compound 1 was administered as the HCl-salt (prepared analogously to Example 2.1). Compound 1 was administered orally in a pH adjusted water solution (pH = 2.5), also containing hydroxypropyl-P-cyclodextrin (HPpCD) as a solubilizing agent, as detailed in Table 22:
TABLE 22: COMPOUND 1 ORAL SOLUTION
Figure imgf000099_0001
[0343] Placebo dosing solutions were identical to active, except without Compound 1. Subjects received a single oral solution of 50mL or lOOmL; all subjects received a total volume of 150mL of liquid. Since HPpCD concentration remained constant, subjects in Cohort 5 received twice the amount of HPpCD relative to subjects in Cohorts 1 to 4. Dosing, including consumption of rinse water, was completed within 3 minutes, administered in the morning after an overnight fast, and maintained for up to 4 hours after dosing.
[0344] The primary objective of the study was to assess the safety and tolerability of ascending dose levels of Compound 1 after single oral dose administration. The safety endpoints included AE incidence, clinically significant changes in vital signs, ECG parameters, clinical laboratory tests and physical examination.
[0345] The secondary objective was to characterize the single-dose pharmacokinetic (PK) parameters of Compound 1 after ascending doses. The following PK parameters were determined from the plasma concentrations of Compound 1 using standard methods of noncompartmental analysis:
Cmax maximum plasma drug concentration
Tmax time to maximum plasma drug concentration t’/2 half-life
AUCiast area under the concentration-time curve from time 0 to last quantifiable concentration
AUC12 area under the concentration-time curve from time 0 to 12 hours
AUCo-inf area under the concentration-time curve extrapolated to infinity
Xz elimination rate constant
CL/F apparent clearance
V/F apparent volume of distribution
[0346] 40 healthy male subjects were randomized into 5 cohorts of 8 subjects each.
Subjects within each cohort were assigned to randomized treatment; 6 assigned to Compound 1 and 2 assigned to placebo. Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use. Each cohort consisted of white, black or African American, and Asian male subjects although the majority were white (47.5%). Subject demographics are summarized in Table 23.
TABLE 23 : STUDY SUBJECT DEMOGRAPHICS
Figure imgf000100_0001
[0347] Cohorts were studied sequentially starting with the lowest dose. The decision to enrol the cohort at the next dose level was reviewed by a safety monitoring team and based on the safety and PK data from previous dose cohorts. Progression to the next higher dose only occurred if the previous dose level was deemed safe and well tolerated. The treatments administered are summarized in Table 24. TABLE 24: COMPOUND 1 AND PLACEBO ADMINISTRATION
Figure imgf000101_0001
For Cohorts 1 to 4, one dosing container with 50mL of dosing solution was administered.
For Cohort 5, two dosing containers, each with 50mL of dosing solution, were administered. b For Cohorts 1 to 4, the dosing container was rinsed with a total of lOOmL of water.
For Cohort 5, each dosing container was rinsed with 25mL of water.
[0348] All subjects underwent a complete physical examination and their medical history and demographic information collected. The body systems evaluated included general appearance, skin, lymphatic, head and neck, EENT (eyes, ears, nose, and throat), chest and lungs, cardiovascular, abdomen, extremities, musculoskeletal and neuromuscular. Vital signs were measured. Blood pressure, pulse, respiratory rate and oral temperature; were taken. Blood and urine samples were collected at screening, pre- baseline, baseline, during the treatment period, and Day 8 follow-up visit; the following clinical laboratory evaluations were conducted.
[0349] Clinical Laboratory Assessments included hematology (complete blood count (CBC), hemoglobin, hematocrit, red blood cell (RBC), mean corpuscular cell volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, red cell distribution width, mean platelet volume, white blood cell count with differential (neutrophils, immature granulocytes, lymphocytes, monocytes, eosinophils, basophils), platelets, and automated reticulocyte count); chemistry (sodium, potassium, bicarbonate, chloride, calcium, phosphorus, fasting glucose, creatinine, blood urea nitrogen, creatinine phosphokinase, uric acid, albumin, total protein, total bilirubin, alkaline phosphatase, ALT, AST, lactate dehydrogenase, total cholesterol and triglycerides); and urinalysis (bilirubin, blood glucose, ketones, pH, protein, specific gravity and microscopic examination).
[0350] Blood samples were collected at baseline, day 1 (0.5, 1, 2, 3, 4, 6, 8, 12, 16, 24 hours post dose), day 2 (30, 36 and 48 hours post dose) and day 8 follow-up visit. Analysis of plasma samples was performed using a validated Liquid Chromatography- Mass Spectrometry and Liquid Chromatography - Tandem Mass Spectrometry (LC-MS/MS) method. [0351] 40 subjects received at least 1 dose. Ten subjects who received placebo across all cohorts were excluded from the PK analyses. Thus, only 30 subjects (75%) comprised the PK Population.
[0352] Four subjects from Cohort 4 experienced emesis within 1.5 hours post-dose, which likely affected PK results. Therefore, PK summaries were created with and without the subjects who experienced emesis. The 240mg dose level was repeated in Cohort 5 at a modified concentration - twice thevolume (lOOmL instead of 50mL) with half the concentration used in Cohort 4 (2.4mg/mL instead of 4.8mg/mL). The dose for Cohort 5 contained twice the amount of HPpCD relative to Cohort 4.
[0353] PK profiles (Compound 1 mean plasma concentration, over time 0-16 hours), are presented in Table 25, and Figures 7 and 8 for all subjects and subjects who did not experience vomiting (ie the 4 subjects in Cohort 4). The profiles show Compound 1 was rapidly absorbed, with Tmax of less than 1 hour. The peak concentrations, Cmax and AUC parameters increased with increasing dose and were higher for drug administered in a larger volume (i.e., Cohort 5, 240mg in lOOmL vehicle) compared with the other cohorts. Elimination parameters (b/2, CL/F) suggested rapid clearance from plasma and relatively large volume of distribution.
TABLE 25 : SUMMARY OF KEY MEAN PHARMACOKINETIC PARAMETERS (SD)
Figure imgf000102_0001
[0354] Hummel Power analysis revealed linear dose proportionality for Cmax, both for all subjects and for the subset excluding the four cohort 4 emesis subjects. The slope value was close to 1 and the 90% confidence interval (CI) for the slope was contained within the critical interval (ie, 0.72-1.28). AUCiast values were slightly above linearity with a slope of 1.167 for all subjects and 1.231 for subjects without emesis. The upper bound of the 90% CI for the slope in both cases was above the critical range. [0355] Primary Endpoint Conclusion - Safety and Tolerability: All doses evaluated (20- 240mg) in 50mL or lOOmL vehicle were determined to be safe and well tolerated. No deaths or SAEs occurred and all of the TEAEs reported were mild in severity. No clinically significant laboratory abnormalities occurred.
[0356] Secondary Endpoint Conclusion - Pharmacokinetics: Following a single oral solution formulation dose of Compound 1, the plasma concentration of Compound 1 increased rapidly (Tmax of 30 minutes), i.e. Compound 1 was rapid absorbed. Cmax and AUC increased in close to dose proportional manner over the entire dose range of Compound 1.
EXAMPLE 12: A PHASE 1 A RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTIPLE ASCENDING DOSE CLINICAL STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF COMPOUND 1 (SOLUTION) IN HEALTHY MALE AND FEMALE VOLUNTEERS.
[0357] Compound 1 was administered as the HCl-salt (prepared analogously to Example 2.1). Compound 1 was administered orally in 100mL of a 10% hydroxypropyl-beta-cyclodextrin (HPpCD) solution, once daily for 8 days. Placebo consisted of a HPpCD solution administered at a volume and dose schedule corresponding with that of Compound 1. See Table 26.
TABLE 26: PLANNED DOSES AND DOSE LEVELS
Figure imgf000103_0001
*Vol of 10% HPPCD solution
[0358] The primary objective of the study was to assess the safety and tolerability of ascending doses of Compound 1 after multiple oral doses administration. The safety endpoints included TEAE incidence and severity, clinically significant changes in vital signs, ECG parameters, clinical laboratory tests and physical examination.
[0359] The secondary objective was to characterize the multiple dose pharmacokinetic (PK) parameters of Compound 1, after ascending doses. Secondary endpoints included determination of PK parameters in plasma and urine, fecal excretion and assessment of colonic biopsy samples.
[0360] 40 healthy subjects were randomized into 5 dose cohorts of 8 subjects each.
Subjects received Compound 1 (n=6) or placebo (n=2), administered once daily for 8 days. The first 3 dose cohorts were in an ascending-dose design. Prior to each dose escalation, a safety monitoring team determined if dose escalation was appropriate. The planned dose levels are given in table 26. The planned study scheme is presented as a flow chart in figure 10.
[0361] Thus, the study planned to enroll up to 40 subjects (up to 30 assigned to Compound 1 and up to 10 assigned to placebo), with allowance for replacement subjects. The study actually enrolled 42 subjects, consisting of 32 subjects treated with Compound 1 (including 2 replacement subjects) and 10 subjects treated with placebo. Overall, 15 male and 17 female subjects were treated with Compound 1, and 5 male and 5 female subjects were treated with placebo.
[0362] Cohorts 1 and 2 evaluated male subjects exclusively; Cohort 3 (240mg) evaluated both male and female subjects. Upon completion of Cohort 3, Cohorts 4 and 5 were added to evaluate the 120mg and 60mg doses in females. As with Cohorts 1-3, Cohorts 4 and 5 were randomized to receive Compound 1 (n=6) or placebo (n=2).
[0363] Subjects were healthy males or females, age 18 to 55, with a Body Mass Index (BMI) between 18 to 33 kg/m2. Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use, and did not display any significant colorectal symptoms or findings. The actual subject demographics and baseline characteristics are summarized in Table 27.
TABLE 27: SUBJECT DEMOGRAPHICS
Figure imgf000104_0001
Figure imgf000105_0001
W = white; B = Black or African American; A = Asian; O = Other
[0364] The study included the following periods:
Screening (up to 30 days prior to dosing)
Treatment Period (Days 1-8)
Follow-Up telephone call 1 week after last dose administration (Day 15).
[0365] Subjects fasted for a minimum of 8 hours prior to obtaining blood samples. During the treatment period, subjects fasted from at least 8 hours prior to dosing in the morning. After dosing, subjects are served a standard breakfast after the 2-hour blood sample has been collected (on Day 1 and 7), a standard lunch approximately 4 hours post-dose, a dinner approximately 10 hours post-dose, and a light snack 12-14 hours post-dose.
[0366] Safety and tolerability were assessed by review of vital signs, laboratory results (serum chemistry, hematology and urinalysis), ECG, physical examination findings, AEs, and PK assessment following the Day 1 and Day 7 doses. All cohorts indicated Compound 1 was generally well tolerated at doses up to 120mg. The 240mg dose was less well tolerated, primarily due to episodes of dizziness and nausea. One event of vomiting led to early trial discontinuation in the 240mg dose group.
[0367] Blood samples were collected at the following time points relative to dose administration on Day 1 and Day 7: pre-dose; 0.5, 1, 2, 3, 4, 6, 8, 12, 16 and 24 hours post dose. Analysis and concentration determinations were performed using a liquid chromatography -mass spectrometry and liquid chromatography-tandem mass spectrometry (LC/MS/MS) method.
[0368] Mean (+/-SD) plasma concentrations of Compound 1, at the noted timepoints, on day 1 and day 7 are presented in Tables 28 and 29. Figures 10 and 11 show the mean plasma concentration-time profiles of Compound 1 on day 1 (single dose) and day 7 (multiple dose) respectively. TABLE 28: COMPOUND 1 CONCENTRATION (NG/ML) ON DAY 1
Figure imgf000106_0001
[0369] Mean PK parameters for Day 1 are presented in Table 30; mean PK parameters for Day 7 are presented in Table 31.
TABLE 30: MEAN (SD) PK PARAMETERS DAY 1
Figure imgf000106_0002
Figure imgf000107_0001
TABLE 31 : MEAN (SD) PK PARAMETERS DAY 7
Figure imgf000107_0002
a Tmax is presented as median (range). b AUCiast is presented as AU O-T) could not be estimated for several subjects. c Two additional subjects enrolled in this cohort as replacement for subjects who prematurely withdrew SD : standard deviation
Cmax : maximum plasma drug concentration
Tmax : time to reach Cmax
AUCiast : area under concentration-time curve from time zero to last quantifiable concentration
ARAUC : accumulation ratios of AUCO
ARc ax : accumulation ratio of Cmax
[0370] Fecal samples were collected, when possible, over 24 hours (0-24 hours) post administration on Day 1 and Day 7; the absolute amount of Compound 1 measured (mg) is shown Table 32.
TABLE 32: MEAN OBSERVED AMOUNT OF COMPOUND 1 (MG) IN FECAL SAMPLES ON DAYS 1 AND 7
Figure imgf000107_0003
[0371] Following ascending doses, mean fecal concentrations of Compound 1 increased in a dose-dependent manner and ranged across cohorts from approximately 40-225 pg/g on Day
1, and 95-506pg/g on Day 7. Mean recovery of Compound 1 in feces across cohorts ranged from approximately 5-22% on Day 1 and 19-52 % on Day 7.
[0372] Urine samples were collected at the following time points relative to dose administration on Day 1 and Day 7: pre-dose; 0-4, 4-8, and 8-24 hours post dose.
[0373] Colonic mucosal biopsy samples were acquired. Compound 1 tissue levels are presented in Table 33 and shown to increase in a dose-dependent manner. TABLE 33 : MEDIAN (RANGE) CONCENTRATIONS OF COMPOUND 1 IN COLON TISSUE
Figure imgf000108_0001
NA: not available as 5 of 6 subjects had concentrations of Compound 1 below limit of quantitation. N=2, 1, 1, 1, and 2 subjects in Cohort 2, 3, 4, 5 and 6, respectively, for whom Compound 1 concentrations were below the limit of quantitation
[0374] Colonic mucosal biopsy samples were also assessed for hypoxia inducible factor (HIF)-inducible gene expression. For Tier 1 association tests, two significant gene were detected (HM0X1, CA9). For Tier 2 analysis, 8 significant genes were detected (NDRG1, LCN2, BNIP3, TFF3, AKR1C3, BIRC3, TMEM45A and HILPDA). These genes are associated with HIF-la and/or hepcidin antimicrobial peptide (HAMP) suggesting the prolyl hydroxylase domain (PHD) target is engaged by Compound 1.
[0375] Manual assessment of the proportion of HIF-la positive cells was conducted in 10 regions of interest, selected in whole tissue sections. (Note, the study was amended to add this immunohistochemistry analysis; thus data is only available for cohorts 4 and 5 (120mg and 60mg), cohorts 1-3 lacked this type of biopsy).
[0376] Table 34 presents the proportions of HIF-la positive cells evaluated at baseline and Day 8, and the change from baseline to Day 8 of the proportion of HIF-la positive cells. The mean proportion of HIF-la positive cells ranged from 1% to 80% across all experimental groups. The proportion of HIF-la positive cells increased from baseline to Day 8 across all groups and a numerically higher increase was seen for the combined group ("Pooled") (35.2 ± 28.8 [mean ± SD]) compared to the placebo group (13.7 ± 20.9 [mean ± SD]).
[0377] Figure 12 shows the box plot of the proportions of HIF-la positive cells evaluated at baseline and Day 8. TABLE 34. DESCRIPTIVE STATISTIC FOR MANUALLY ASSESSED PROPORTIONS OF HIF- la POSITIVE
CELLS IN HIF- la STAINED SECTIONS
Figure imgf000109_0001
[0378] Serum samples were collected for PD analysis and revealed no Compound 1- related effects on circulating EPO and VEGF levels.
EXAMPLE 13: A PHASE IB, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, CLINICAL STUDY TO EVALUATE THE SAFETY, TOLERABILITY, PHARMACOKINETICS AND PHARMACODYNAMICS OF COMPOUND 1 (SOLUTION)
IN ADULT PATIENTS WITH ACTIVE ULCERATIVE COLITIS
[0379] Compound 1 is administered as the HCl-salt, thus:
"120mg dose" refers to 130mg Compound 1 • HC1 salt - prepared analogously to
Example 2.1 (equivalent to 120mg free Compound 1.)
"60mg dose" refers to 65mg Compound 1 • HC1 salt - prepared analogously to Example
2.1 (equivalent to 60mg free Compound 1.)
[0380] The objectives and endpoints of the study are as follows:
Primary Objective: Evaluate the safety and tolerability of Compound 1 daily dosing for 28 days Secondary Objective: Evaluate the pharmacokinetics (PK) of Compound 1
Exploratory Objective: Explore target engagement and pharmacodynamic response; clinical and histologic activity; and evaluate the relationship between safety, disease activity, exposure parameters and pharmacogenetics
Primary Endpoints: Incidence of treatment-emergent adverse events (TEAEs) and changes from baseline in laboratory, vital sign and electrocardiogram parameters
Secondary Endpoint: PK parameters
Exploratory Endpoint: Changes in biomarkers of target engagement, pharmacodynamics, clinical activity scores, UC-100 Mayo Clinic Score and Robarts Histopathology Index; safety, disease activity and exposure parameters; and proportion of patients with histologic remission and/or mucosal healing at Day 28.
[0381] In clinical trials, disease activity has traditionally been assessed by evaluating signs and symptoms of the disease, rather than the inflammatory process itself. Endoscopic appearance and histology have recently been validated as clinical trial outcome measures that provide for more direct assessment of disease activity and mucosal healing. Indeed, histology is an important prognostic factor and treatment target, providing insight into underlying histologic disease activity. The composite UC-100 score calculated as: (1 + 16 * Mayo Clinic stool frequency subscore [0 to 3] + 6 * Mayo Clinic endoscopic subscore [0 to 3] + 1 x Robarts histopathology index score [0 to 33]), ranges from 1 (no disease activity) to 100 (severe disease activity). Validation of the composite UC-100 Index allows clinical trials the ability to reliably measure the combination of symptoms, endoscopic appearance and histologic activity.
[0382] The study employs a two-cohort design, shown in schematic form in Figure 13. Cohort 1 evaluates Compound 1 120mg, relative to placebo.
Cohort 2 studies additional patients at the 120mg or 60mg dose levels. A safety monitoring team determines whether and when to initiate Cohort 2 following review of Cohort 1 data, and if so, the 120mg or 60mg dose to be evaluated. The total number of patients is approximately 30. Each cohort (Cohort 1 and 2) randomizes approximately 15 patients, with approximately 10 patients randomized to Compound 1 and 5 patients to placebo. [0383] Compound 1, or placebo, is administered one daily (QD), every day for 28 days, as an oral solution. Each dose is reconstituted with approximately lOOmL water, followed by an approximately lOOmL water "rinse" from the bottle contain Compound 1. Each dose also contains 10g hydroxypropyl-beta-cyclodextrin (HPpCD). Placebo dosing solutions are identical to active, except without Compound 1.
[0384] The total duration of the study per patients is 13 weeks, including a 5-week screening period (7-35 days), the 4-week treatment period and a 4-week follow-up period.
[0385] Dose are administered orally in the morning at approximately the same time each day, with patients fasting for at least 6 hours before and 2 hours after each dose. The first dose is administered on Day 1. A blood sample is taken 8 hours post-dose. Patients are assessed on Days 7, 14, 21 and 28, followed by a Follow-up (FU) visit four weeks later.
[0386] Patients are age 18-75, male or female with a BMI 18-35kg/m2. Patients do not have Crohn’s disease or indeterminate colitis, pouchitis, evidence of Clostridium difficile infection, a current malignancy or previous history of cancer or a history of alcohol or substance abuse. Patients exhibit the following disease criteria:
1) Positive ulcerative colitis diagnosis.
2) Complete Mayo clinic score of 3-12, with an endoscopic subscore > 1.
3) The presence of blood in the stool.
4) Colonic biopsies with evidence of histologic activity defined by a Robarts Histopathology Index > 4 with neutrophils in the epithelium (subscore > 1).
5) Currently receiving treatment with oral 5 -aminosalicylates (5-ASA) for ulcerative colitis or prednisone or equivalent, budesonide mmx or azathioprine or 6-mercaptopurine.
6) Evidence of UC extending > 15cm from the anal verge.
[0387] The efficacy endpoints are:
Complete Mayo Clinic Score: The sum of the Stool Frequency subscore, Rectal Bleeding subscore, Physician Global Assessment, and the Endoscopy subscore. Each subscore ranges from 0 to 3 points and the Complete Mayo score ranges from 0 to 12 points.
Improvement in Endoscopic Appearance: Mayo Endoscopy sub score of 0 or 1, or 0 if baseline Mayo Endoscopy subscore = 1. Mucosal Healing: Mayo Endoscopy subscore of 0 or 1, or 0 if baseline Mayo Endoscopy subscore =1, and Robarts Histopathology Index (RHI) Score <3 with lamina propria neutrophils sub score = 0 and neutrophils in epithelium sub score = 0.
Histologic Remission: RHI < 3 with lamina propria neutrophils subscore = 0 and neutrophils in epithelium sub score = 0.
UC-100 Score: The composite index (1 + 16 x Mayo Clinic Stool Frequency Subscore [0 to 3] + 6 x Mayo Clinic Endoscopic Subscore [0 to 3] + 1 x Robarts Histopathology Index Score [0 to 33]).
Robarts Histopathology Index [RHI] Score: Consists of four items: chronic inflammatory infiltrate, lamina propria neutrophils, neutrophils in epithelium, and erosion or ulceration. The RHI ranges from 0-33.
[0388] Safety is assessed by physical examination, vital signs (pulse, respiratory rate, temperature and blood pressure), an electrocardiogram (ECG) and laboratory assessments.
[0389] Blood samples (~3mL) are collected on the Day 1 and 28 visits, 0.5, 1, 2, 3, 4, 6, and 8, 12, and 24 hours post-dose. Stool samples, when available, are collected on Study Days 14 and 28.
[0390] Blood, stool and colonic biopsy samples evaluate the effect of Compound 1 on a range of potential target engagement and PD biomarkers.
[0391] Throughout the study, patients record stool frequency and rectal bleeding symptoms. The Rectal Bleeding and Stool Frequency components of the Mayo Score are calculated based on the most recent 3 days with data prior to the Visit, excluding the day of and the day after a flexible sigmoidoscopy/colonoscopy, and the day(s) of bowel preparation. These 3 days must occur within the 7 days prior to the Visit.
[0392] Flexible sigmoidoscopy evaluates the endoscopic appearance of the colonic mucosa. At Baseline and Day 28 (and Day 14 if applicable), 6 biopsies are collected from the rectum and sigmoid and evaluated for PK and histologic, immunohistochemical, and gene expression signals of biological activity.
[0393] Clinical Laboratory Assessments include hematology (white blood cell count, red blood cell, hemoglobin, hematocrit, platelet count); clinical chemistries (alanine aminotransferase, albumin, aspartate aminotransferase, alkaline phosphatase, bicarbonate, bilirubin, blood urea nitrogen, calcium, creatinine, chloride , gamma-glutamyl transferase, glucose, L-lactate dehydrogenase, potassium, total protein, sodium; and urinalysis (dipstick, including macroscopic appearance, bilirubin, blood, color, glucose, ketones, leukocyte esterase, nitrite, pH, protein, specific gravity, urobilinogen).
EXAMPLE 14: A PHASE 1 A, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTIPLE DOSE STUDY TO ASSESS THE SAFETY, TOLERABILITY, PHARMACOKINETICS, AND PHARMACODYNAMIC EFFECTS OF TABLET FORMULATIONS OF COMPOUND 1 IN HEALTHY MALE AND FEMALE
VOLUNTEERS.
[0394] The primary objective of the study is to assess the safety and tolerability of tablet formulations of Compound 1 • HC1 after multiple oral doses. The safety endpoints include incidence of treatment-emergent adverse events (TEAEs) and changes in laboratory, vital sign and ECG parameters.
[0395] The secondary objective is to evaluate the pharmacodynamic response of tablet formulations after multiple oral doses, with an endpoint of change in of target engagement biomarkers (e.g., CAIX). Target engagement is assessed by measuring HIF-1 related gene expression and protein abundance in colonic tissue, stool and blood.
[0396] An exploratory objective of the study is to evaluate the effect of pharmacogenetics (PGx) on pharmacokinetic (PK) parameters of tablet formulations as compared to oral solution. Pharmacokinetics assessments include evaluation of blood, stool and colonic biopsy samples.
[0397] A schematic of the study design is presented in Figure 15. 40 subjects are randomized in the study, stratified by gender with each stratum comprising half of the study population. Thirty subjects are randomized to Compound 1 and 10 to placebo. Up to 8 replacement subjects may be added (up to 2 in the solution groups and up to 6 in the tablet groups; each replacement is the same gender and assigned to the same treatment group as the subject being replaced). Subjects are randomized to 8 groups, according to Table 35.
TABLE 35 : STUDY INTERVENTIONS
Figure imgf000113_0001
Figure imgf000114_0001
[0398] The study duration is up to 65 days, and includes the following periods:
• Screening (2 - 28 days)
• Treatment period of 8 days (treatments administered once daily for 7 days)
• End of study visit, 10 days post-last dose [Day 17]
• Follow-up phone call, 28 days post-last dose [Day 35]
[0399] Subjects are healthy males or females, age 18-65 with a Body Mass Index (BMI) 18-33 kg/m2. Subjects were infection free (including HIV, hepatitis B or hepatitis C), no history of chronic disease or cancer, alcohol dependence, drug addiction or nicotine use, and did not display any significant colorectal symptoms or findings.
[0400] Subject are dosed orally in the morning, at the same time each day, once daily for 7 days. Doses are administered as follows:
Solution: Compound 1 is administered in lOOmL of a water 10% hydroxypropyl -beta- cyclodextrin (HPpCD) solution. Each solution dose is mixed with 100 mL purified water, followed by a 100 mL water "rinse" taken orally from the same bottle.
Tablet: Compound 1 - IR tablets prepared according to Example 5; DR tablets prepared according to Example 6. 60mg tablets IR, DR or matching placebo formulations are similar and indistinguishable in terms of appearance, odor and taste. Subjects randomized to receive 120mg, are administered two 60mg tablets; subjects randomized to receive 240mg, are administered four 60mg tablets.
Fasting subjects, fast for at least 10 hours before and 2 hours after each dose.
Fed subjects are dosed on a fed stomach 30 minutes after the start of a standardized meal. Subjects are required to consume the meal in 30 minutes or less.
[0401] Physical Examinations include vital signs (pulse rate, respiratory rate, temperature and blood pressure) and electrocardiogram (ECG).
[0402] A flexible sigmoidoscopy is performed prior to dosing and on Day 7 to evaluate the endoscopic appearance of the colonic mucosa and obtain biopsies. During the sigmoidoscopy, 6 biopsies are collected in each of the two segments (e.g., Rectum and Sigmoid) for approximately 12 biopsies. Biopsies assess PK, immunohistochemical and gene expression signals (e.g., CAIX) of biological activity
[0403] Blood samples are collected on Days 1 and 7, at pre-dose and 0.25, 0.5, 1, 2, 3, 4, 6, and 8, 12, and 24 hours post-dose.
[0404] Stool samples, when available, are collected on Days 5 and 7.
RESULTS FROM EXAMPLE 14 CLINICAL TRIAL
EXAMPLE 15: PHARMACOKINETICS OF FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
[0405] IR and DR tablet dosing (120 mg or 240 mg dose) was compared to solution dosing in a 10% hydroxypropyl-beta-cyclodextrin (HPpCD) solution (120mg dose) according to the protocol described in Example 14 above. This was a Phase la, randomized, double-blind, placebo-controlled, multiple dose study that evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics of tablet formulations of Compound 1 in male and female healthy adult subjects, and compared PK parameters of Compound 1 tablet formulations to Compound 1 oral solution and fasted to fed subjects. The composition and method of manufacture of the IR and DR tablet compositions used in this study are disclosed in Examples 5 and 6 above.
[0406] Mean plasma concentration-time profiles of Compound 1 on Day 1 and Day 7 are shown in Figure 15.
[0407] On Day 1, Compound 1 was rapidly absorbed from all fasting treatments with maximum plasma concentrations occurring at a median Tmax of 0.500 to 1.00 hours post-dose, but delayed to a median Tmax of 2.00 hours following administration of the IR tablet at a dose of 240 mg, fed.
[0408] Tmax values of the DR tablet were relatively longer than the other tablet treatment groups on Day 7. On Day 1, except for the 120 and 240 mg delayed release tablet formulations, all Compound 1 treated subjects displayed a quantifiable concentration by 0.5 hours post-dose. Five of 6 and 3 of 6 subjects receiving the 120 and 240 mg delayed release tablet formulations, respectively, displayed quantifiable concentrations below the lower limit of quantitation at all time points. There was considerable inter-subject variability in plasma concentrations up to 200% in all treatment groups.
[0409] On Day 7, all subjects had quantifiable concentrations throughout most of the sampling period except for two subjects in each of the delayed release tablet formulation groups.
[0410] Overall, mean Compound 1 concentrations on Day 7 were slightly higher than on Day 1 for all treatment groups, except for the 120 mg solution.
[0411] Summary statistics for Compound 1 PK parameters in plasma for Day 1 and Day 7 are presented in Tables 36 and 37, respectively.
TABLE 36 ARITHMETIC MEAN AND %CV PHARMACOKINETIC PARAMETERS IN PLASMA FOR COMPOUND 1 ON DAY 1
Figure imgf000117_0001
Abbreviations: AUCinf, area under the concentration-time curve from zero to infinity; AUClast, area under the concentration-time curve from zero to the last quantifiable concentration time point; Cmax, maximum plasma drug concentration; CV, coefficient of variation; NC, not calculated; Tmax, time to reach Cmax- a Includes subjects who had Compound 1 concentrations below the limit of quantitation at all time points (5 subjects in the 120 mg Delayed Release Tablet and 3 in the 240 mg Delayed Release Tablet treatment groups); for these subjects, Cmax and AUClast were set = 0.00 and Tmax was not calculated. b Median (range) c PK parameter could only be determined in one subject and thus, not included in summary statistics.
TABLE 37: ARITHMETIC MEAN AND %CV PHARMACOKINETIC PARAMETERS IN PLASMA FOR COMPOUND 1 ON DAY 7
Figure imgf000118_0001
Abbreviations:
ARAUCO-24 = accumulation ratio of AUC from time 0 to 24 hours; ARcmax = accumulation ratio of Cmax; AUCIast = area under the concentration-time curve from zero to the last quantifiable concentration time point; Cmax = maximum plasma drug concentration; CV = coefficient of variation; NC = not calculated; Tmax = time to reach Cmax- a Includes subjects who had Compound 1 concentrations below the limit of quantitation at all time points (2 in the 120 mg Delayed Release Tablet and 2 in the 240mg Delayed Release Tablet treatment groups; for these subjects, Cmax and AUCIast were set = 0.00 and Tmax was not calculated. b Median (range) c Could be determined in less than 3 subjects and thus, not included in summary statistics.
[0412] On Day 1, half-life (t’/2) and exposure (AUCinf) could not be estimated accurately due to the lack of evaluable data points available from most subjects except for those receiving the 120 mg solution fasting and 240 mg tablet fed treatments. The derived values for these parameters should be interpreted with caution.
[0413] Mean Cmax and AUClaSf for the fasted 120 mg solution were slightly higher than the fasted 120 mg IR tablet on Day 1 (34.5 ng/mL and 39.3 ng*h/mL vs. 8.85 ng/mL and 16.9 ng*h/mL, respectively), but comparable on Day 7 (14.4 ng/mL and 42.4 ng*h/mL and 10.6 ng/mL and 31.5 ng*h/mL). On Day 1, as previously stated, 5 of 6 and 3 of 6 subjects receiving the 120 and 240 mg delayed release tablet formulations had no detectable levels of Compound 1 in plasma and most PK parameters could not be accurately determined. The same was true on Day 7, where two of six subjects receiving the 120 and 240 delayed release tablet formulations had no detectable levels of Compound 1. Except for a slight delay in Tmax following administration of food with the 240 mg tablet on Day 1, the Cmax and AUClast, following dose normalization, were similar to that following administration of 120 mg tablet fasting.
[0414] Exposure of the solution was slightly higher than the tablet following a single dose but were comparable on Day 7. Following the first dose on Day 1, Compound 1 was rapidly absorbed from all fasting treatments with median Tmax of 0.500-1.00 h but delayed to 2.00 h after a meal. However, upon once daily dosing for 7 days, Compound 1 was rapidly absorbed with a median Tmax of 0.500 h for all treatments except for the 120 mg and 240 mg delayed release tablet formulations where the median Tmax values were 3.00 and 3.50 h, respectively. The majority of subjects receiving the delayed release tablets had no detectable levels of Compound 1 at any time point on Day 1; however, upon multiple dosing, more subjects had plasma concentrations above the lower limit of quantitation on Day 7.
Metabolite PK
[0415] Mean plasma concentration-time profiles of the glucuronide metabolite of Compound 1 on Day 1 and Day 7 following the dosing of Compound 1 to the various cohorts described in Example 14 are shown in Figure 16. The PK levels of the glucoside metabolite of Compound 1 were also similarly monitored in the Example 14 patients.
[0416] The glucuronide and glucoside metabolites were rapidly formed and appeared in plasma after oral administration of the solution and tablet formulations of Compound 1 (median Tmax of ~0.5 h for tablet formulations) and their exposure is approximately 235-fold and 38-fold higher than the parent Compound 1, for the glucuronide and glucoside metabolites respectively, on Day 7. The highest Cmax observed in the Example 14 patients for the these metabolites was in the 240 mg IR tablet (fed) cohort and was 4.64 pM (about 2800 ng/ml) and 0.68 pM (about 400 ng/ml), for the glucuronide and glucoside metabolites respectively on Day 7. The apparent half-life of both metabolites is approximately 8 to 12 h, which is longer than that of the parent (~ 2 h).
[0417] The highest AUCo-24 observed in the Example 14 cohorts for the glucuronide metabolite was 42600 ng*h/mL in the 240 mg IR tablet (fed) cohort.
EXAMPLE 16: COLONIC TISSUE CONCENTRATIONS OF TABLET FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
[0418] IR and DR tablet dosing (120 mg or 240 mg dose) was compared to solution dosing in terms of colon tissue (sigmoid and rectum) concentrations of Compound 1 according to the protocol described in Example 14 above.
[0419] A summary of mean and median tissue Compound 1 concentrations in the colon on Day 7 is provided in Table 38 and Figure 17.
[0420] Figure 17 shows the total drug colonic concentrations across both sections (sigmoid + rectum) for the different study subject cohorts.
TABLE 38 : TISSUE CONCENTRATIONS OF COMPOUND 1 IN THE COLON (NG/G), DAY 7
Figure imgf000120_0001
Abbreviation: CV = coefficient of variation.
[0421] The median Compound 1 concentrations in the rectum were higher for the delayed release tablets as compared to the immediate release tablet and solution. However, while the median concentration of Compound 1 in the sigmoid was also higher for the 120 mg delayed release tablet than for the immediate release tablet and solution, it was less for the 240 mg delayed release tablet than for the immediate release tablet; however, inter- subject variability was notably high across all formulations (CV% ranged from 65.4% to 227%).
[0422] Despite distinct systemic PK profiles, the immediate release and delayed release tablets delivered similar median levels of Compound 1 to the colon and more than the solution dose.
EXAMPLE 17: SAFETY OF FORMULATIONS OF COMPOUND 1 IN HEALTHY SUBJECTS
[0423] IR and DR tablet dosing (120 mg or 240 mg dose) was compared to solution dosing in terms of adverse events reported according to the study protocol described in Example 14 above.
[0424] Preliminary safety results in this study suggest that Compound 1 formulations up to 120 mg solution, 240 mg tablet, and 240 mg DR tablet are generally well tolerated in healthy subjects. Furthermore, no consistent or dose-related effects of Compound 1 on EPO or VEGF from baseline to Day 7 were observed when compared to placebo.
TABLE 39: ADVERSE EVENTS FOR EXAMPLE 14 STUDY COHORTS
Figure imgf000121_0001
Figure imgf000122_0001
EXAMPLE 18: A PHASE 2, RANDOMIZED, DOUBLE-BLIND, PLACEBO- CONTROLLED, MULTI-CENTER STUDY TO EVALUATE COMPOUND 1 IN ADULT SUBJECTS WITH MILD-TO-MODERATE ACTIVE ULCERATIVE COLITIS
18.1 Background
[0425] Small molecule prolyl hydroxylase inhibitors (PHDi’s) stabilize HIF-la protein and have shown efficacy in multiple mouse models of IBD that include trinitrobenzene sulfonic acid (TNBS) and dextran sulfate sodium (DSS)-induced colitis models and the tumor necrosis factor alpha (TNFa)-driven terminal ileitis model (Ablin, 1999; Minaiyan, 2012). Prolyl hydroxylases (PHDs) are key regulators of the HIF pathway (Eltzschig, 2014). These findings support PHD inhibition as a target for treating ulcerative colitis (UC). Compound 1, a HIF-la stabilizer, is a potential oral therapeutic, gut-targeted treatment for UC with higher intestinal than systemic exposure. The relatively low oral bioavailability (< 20%) and preferential distribution to the gastrointestinal (GI) tissues observed in nonclinical studies, combined with its mechanism of action in gut lumen and positive effects on gut healing and inflammation in animal models of colitis, provide rationale for further development in human subjects. This Phase 2 study assesses the efficacy and safety of Compound 1 when added to background UC therapy of 5 -aminosalicylate (5 -ASA) with or without steroids.
18.2 Primary Objective
[0426] The primary objective is to evaluate the effect of Compound 1 compared to placebo on clinical remission. The primary endpoints are defined as the proportion of subjects with clinical remission at Week 12, defined as a Modified Mayo score < 2, with a rectal bleeding subscore of 0, stool frequency subscore of 0 or 1 (with a > 1 point decrease from baseline), and endoscopy subscore of 0 or 1. 18,3 Secondary Objective
[0427] The secondary objective is to evaluate the effect of Compound 1 on clinical response and endoscopic improvement. The secondary endpoints include:
• Proportion of subjects with clinical response at Week 12, defined as reduction in the Modified Mayo score of > 2 points and > 35 percent reduction from baseline, including a decrease in Rectal bleeding subscore of >1 or absolute Rectal bleeding subscore of < 1;
• Proportion of subjects with histologic remission at Week 12, defined as Robarts Histopathology Index (RHI) < 3 with lamina propria neutrophils subscore = 0 and neutrophils in epithelium subscore = 0;
• Proportion of subjects with endoscopic improvement at Week 12, defined as endoscopic subscore of 0 or 1; and
• Proportion of subjects with mucosal healing at Week 12, defined as endoscopic improvement and histologic remission.
18.4 Safety Objective
[0428] The study also has a safety objective related to monitoring the incidence of treatment-emergent adverse events (TEAEs).
18.5 Further Exploratory Objectives
[0429] The study has the further exploratory objectives and associated endpoints:
Other safety outcomes
• Changes from Baseline in laboratory, vital signs, and electrocardiogram (ECG) parameters.
Other measures of the effect of Compound 1 on disease activity
• Proportion of subjects with resolution of Rectal bleeding at Week 12.
• Proportion of subjects with histologic response at Week 12.
• Proportion of subjects with Geboes score <2 at Week 12.
• Proportion of subjects with clinical remission by Mayo score and resolution of rectal bleeding at Week 12. • Proportion of subjects with clinical remission by Mayo score at Week 12.
• Proportion of subjects with clinical response by Mayo score at Week 12.
• Proportion of subjects with Modified Mayo score of < 2, with no individual subscore > 1 at Week 12.
• Proportion of subjects with symptomatic remission at Week 12.
• Proportion of subjects with modified symptomatic remission at Week 12.
• Proportion of subjects with partial Mayo score of <2 with no individual subscore >1 over time.
• Proportion of subjects with reduction of >2 points from baseline in partial Mayo score over time.
• Proportion of subjects with a 6-point Mayo score clinical remission at Week 12.
• Change from Baseline in Modified Mayo score at Week 12.
• Change from Baseline in Mayo score at Week 12.
• Change from Baseline in Partial Mayo score over time.
• Change from Baseline in Mayo subscores over time.
• Change from Baseline in Robarts Histopathology Index (RHI) score at Week 12.
• Change from Baseline in the UC-100 Index at Week 12.
Pharmacokinetics of Compound 1
• Plasma and colon tissue concentrations of Compound 1.
Target engagement (TE) and pharmacodynamic (PD) responses
• Change from baseline in tissue and blood markers of target engagement, e.g. HIF-la, EGLN1-2-3, etc.
• Change from baseline in markers of pharmacodynamics, e.g. fecal calprotectin, fecal lactoferrin, and neutrophil activity (myeloperoxidase).
• Change from baseline in tissue, blood, and stool in downstream genes and proteins, e.g. CAIX, MPO, c-reactive protein (CRP), TJP, CLDN1, etc. as a function of exposure (Cmax, Tmax, AUC), efficacy endpoints (e.g. clinical remission, clinical response), and/or safety endpoints. Response of baseline markers to treatment with Compound 1
• Relationship between tissue, blood, and stool baseline markers (e.g. HIF- la, EGLN1-2-3, calprotectin, etc.) and response to treatment, as measured by efficacy endpoints (e.g. clinical remission, clinical response), and/or safety endpoints.
Pharmacogenetics (PGx)
• Effect of naturally occurring genetic variation on the efficacy (e.g. clinical remission, clinical response), safety, and/or PK profile (Cmax, Tmax, AUC) and tissue concentrations associated with treatment with Compound 1.
18,6 Study Design
[0430] Randomized, double-blind, placebo-controlled, multi-center study to evaluate the efficacy, safety, tolerability, and PK of 2 dose regimens of Compound 1 in adult subjects with mild-to-moderate active UC and have disease activity despite treatment with 5-ASA.
[0431] After signing an informed consent form (ICF), subjects are screened for study eligibility over a 2 to 4 weeks Screening period. During the Screening period, subjects capture stool frequency and rectal bleeding symptoms in a provided electronic diary (eDiary) on a daily basis. Flexible sigmoidoscopy or colonoscopy is performed at screening, and biopsies are assessed for inclusion in the study. Subjects not meeting the eligibility criteria are deemed screen failures and do not continue participation in the study. All subjects participating in the study are required to maintain stable dose of background UC therapies throughout the study (for details refer to Section 18.12).
[0432] On Day 1, approximately 195 eligible subjects are randomized 1 : 1 : 1 to receive the following double-blind treatments for 12 weeks (approximately 65 subjects per treatment group):
• • Compound 1 - 480 mg twice daily (BID)
• • Compound 1 - 480 mg once daily (QD)
• • Placebo
[0433] Randomization is stratified by systemic corticosteroid use at baseline (yes/no). [0434] All subjects dose investigational product (IP) BID to maintain the blind - see section 1.7
[0435] Subsequently, subjects return to the clinic and are evaluated. A flexible sigmoidoscopy with biopsies is performed at the Week 12 visit (or at the Early Withdrawal from Study visit). All subjects also attend a Follow-up visit at the clinic 4 weeks after last dose of IP.
[0436] Total duration for study participation per subject is expected to be up to 20 weeks: 2-4 week screening period; 12 week treatment period; and 4 week follow-up period.
[0437] The study includes the following activities:
• Physical exam - screening, treatment (week 12) and follow-up;
• Vital signs (pulse rate, respiratory rate, temperature, blood pressure and weight) - screening, treatment (weeks 0, 2, 4, 8 & 12) and follow-up;
• 12-lead electrocardiogram - screening, treatment (week 12) and follow-up;
• Flexible sigmoidoscopy with biopsy - screening and treatment (week 12);
• Physician’s Global Assessment (PGA) - screening and treatment (weeks 0, 2, 4, 8 & 12);
• Blood plasma sample (PK) - treatment (weeks 0, 4 & 12). Blood samples will be collected pre-dose, and 0.5, 2 and 4 hours post-dose. The timepoints are in reference to the morning dose;
• Blood sample (PD) - treatment (weeks 0, 2, 4, 8 & 12). Blood samples will be collected twice (pre-dose vs post-dose) as EDTA plasma and twice (pre-dose vs post-dose), as PaxGene. The timepoints are in reference to the morning dose;
• Stool sample (PK & PD) - screening and treatment (week 12);
• Whole blood sample (PGx) - treatment (week 0). Collected pre-dose (morning).
18,7 Investigational Product! s) Administered
[0438] Each dose of Investigational Product (IP) will consist of a combination of Compound 1 immediate-release tablets and Compound 1 delayed-release tablets or of matching placebo and will be taken orally BID on a fed stomach at approximately the same time each day, 8 to 12 hours apart. [0439] The Compound 1 immediate-release tablets, Compound 1 delayed-release tablets, and placebo tablets are similar and indistinguishable in terms of appearance, odor, and taste, such that the blind is maintained.
[0440] For Compound 1 (Active), the dose formulation is round film-coated tablets containing 60 mg of Compound 1 per tablet. For Placebo (Control), the dose formulation is round film-coated tablets matching the active formulation.
[0441] The dose regimens for the three patient cohorts are as follows:
Compound 1 - 480 mg BID
[0442] Four immediate-release tablets each containing 60 mg Compound 1 and four delayed-release tablets each containing 60 mg Compound 1, administered twice daily.
Compound 1 - 480 mg QD
[0443] Four immediate-release tablets each containing 60 mg Compound 1 and four delayed-release tablets each containing 60 mg Compound 1, administered daily in the morning and eight placebo tablets administered daily in the evening.
Placebo
[0444] Eight placebo tablets administered twice daily.
18.8 IP Administration
[0445] The tablets are taken orally on a fed stomach, 30 minutes after a meal. The IP is taken BID (approximately 8 to 12 hours apart) at approximately the same time each day.
18.9 Study Population
[0446] Adult subjects with mild-to-moderate active UC and have disease activity despite taking 5-ASA and evidence of active UC in the setting of ongoing 5-ASA background therapy with or without prednisone, budesonide, or budesonide-MMX background therapy. Active UC is defined for this study by a baseline Mayo score of 5-10, inclusive, with an Endoscopy subscore of > 2, a Stool frequency subscore > 1, and a Rectal bleeding subscore > 1. 18, 10 Inclusion Criteria
[0447] Subjects are eligible to be included in the study only if all the following criteria are met:
Age and Sex
1. Adult male and female subjects aged > 18 years at the time of signing the informed consent form prior to initiation of any study specific activities/procedures.
Type of Subject and Disease Characteristics
2. UC diagnosed at least 3 months prior to first dose of IP on Day 1. The diagnosis should be confirmed by clinical and endoscopic evidence and corroborated by a histopathology report (note: the screening endoscopy and histopathology results may be used to confirm the diagnosis if no prior report is readily available).
3. Mayo score of 5-10, inclusive, with a centrally read Endoscopic subscore
> 2 and a Rectal bleeding subscore > 1 and Stool frequency score > 1.
4. Evidence of UC extending > 15 cm from the anal verge as determined by screening flexible sigmoidoscopy or colonoscopy.
5. Colonoscopy within the past 2 years (or during the Screening period) to screen for dysplasia (unless otherwise recommended by local and national guidelines) if the subject has had UC for 8 or more years duration.
Allowed UC Therapy
6) Currently receiving treatment for UC, on a stable dose for at least 2 weeks prior to flexible sigmoidoscopy or colonoscopy, with oral 5 -ASA (eg, mesalamine, sulfasalazine) alone, or with one of the following treatments: a) prednisone < 20 mg/day or equivalent; or b) budesonide or budesonide multi-matrix (MMX) of < 9 mg/day.
Body Mass Index (BMI)
7) BMI 18 to 35 kg/m2. Pregnancy and Contraception
[0448] Contraception use by men or women should be consistent with local regulations regarding the methods of contraception for those participating in clinical studies.
8) Women meeting the definition of non-childbearing potential or women of childbearing potential must agree to contraceptive guidance.
9) Men must agree to contraceptive guidance.
Informed Consent
10) Capable of giving signed informed consent.
18, 11 Exclusion Criteria
[0449] Subjects are excluded from the study if any of the following criteria apply:
Excluded Prior Therapy
1) Prior treatment with any biologic therapy used for the treatment of UC.
2) Systemic corticosteroids > 20 mg/day or equivalent, or budesonide or budesonide MMX of > 9 mg/day within 1 day of Day 1.
3) Enemas or suppository formulations of steroids or 5 -aminosalicylates within 2 weeks of screening flexible sigmoidoscopy/colonoscopy.
4) Tofacitinib, oral cyclosporine, sirolimus or myocophenolate mofetil within
8 weeks of Day 1.
5) Azathioprine, or 6-mercaptopurine within 1 day of Day 1.
6) Experimental agents within 5 half-lives of Day 1.
7) Anti-diarrheal medications (e.g., loperamide, diphenoxylate/atropine) during screening.
8) Epoetin alfa within 8 weeks of Day 1.
9) Cholestyramine or other drugs interfering with enterohepatic circulation within 14 days of Day 1.
10) Chronic non-steroidal anti-inflammatory agents (NSAID) use (occasional use of NSAID, acetaminophen, or aspirin up to 325 mg/day is allowed).
11) Treatment with strong cytochrome P450 (CYP) 3 A inhibitors and/or
CYP3 A inducers (e.g. oceprevir, cobicistat, conivaptan, danoprevir, ritonavir, elvitegravir, grapefruit juice, indinavir, itraconazole, ketoconazole, lopinavir, paritaprevir, ombitasvir, dasabuvir, posaconazole, saquinavir, telaprevir, tipranavir, troleandomycin, voriconazole, clarithromycin, diltiazem, idelalisib, nefazodone, nelfinavir, carbamazepine, enzalutamide, mitotane, phenytoin, rifampin, St. John’s wort) within 21 days or within 5 half-lives, whichever is longer, of the respective medication prior to Day 1.
Medical Conditions
12) Diagnosis of Crohn’s disease or indeterminate colitis, pouchitis.
13) Fulminant colitis, toxic megacolon, or in Investigator’s judgement likely to require colectomy or ileostomy within 16 weeks of Day 1.
14) Stool sample positive for Clostridium difficile (C difficile) toxin, stool pathogens, or ova and parasites infection on screening laboratory tests. If positive, subjects may be treated and retested.
15) A current malignancy, current colonic dysplasia or previous history of cancer in remission for less than 5 years prior to screening. Subjects are not excluded if they had localized carcinoma of the skin that was resected for cure, cervical dysplasia that has been adequately treated, or colonic dysplasia that has been completely removed.
16) Known pre-existing, unstable liver disease (as defined by the presence of ascites, encephalopathy, coagulopathy, hypoalbuminemia, esophageal or gastric varices or persistent jaundice), cirrhosis, known biliary abnormalities, known pre-existing non-alcoholic steatohepatitis (NASH), or active infection with Hepatitis B or Hepatitis C.
17) Immunodeficiency, including that due to human immunodeficiency virus (HIV), other than that explained by systemic corticosteroid use.
18) Other concurrent medical conditions: known, pre-existing clinically significant endocrine, autoimmune, metabolic, neurological, renal, gastrointestinal, hepatic, cardiovascular, respiratory (e.g. including active tuberculosis), hematological, or any other system abnormalities that are uncontrolled with standard treatment. 19) Any other medical condition or laboratory abnormality that, in the opinion of the Investigator, prohibits the subject from participating in the study.
Diagnostic assessments
20) QTcF > 450 msec for males or QTcF > 470 msec for females at screening by central overread. If QTcF is above specified limit and there are no other clinically significant abnormalities, repeat assessment is allowed with permission by the Sponsor’s Medical Monitor (or designee).
21) Hemoglobin <7.5 g/dL, platelet count <100,000/pL, absolute neutrophils count < 1200 at screening.
22) Alanine aminotransferase (ALT) or aspartate aminotransferase (AST) > 1.5 times upper limit of normal (ULN) at screening.
23) Total bilirubin > ULN at screening, unless likely related to diagnosis of Gilbert’s syndrome, in the opinion of the Investigator.
24) Estimated glomerular filtration rate (eGFR) <60 mL/min calculated by chronic kidney disease epidemiology collaboration (CKD-EPI).
Prior Clinical Study Experience
25) Treatment with an investigational medication or treatments (e.g. hyperbaric oxygen treatment) within the past 30 days or within 5 half-lives of the medication, whichever is longer, prior to screening.
26) Have previously received treatment with Compound 1.
Other Exclusions
27) A history or suspected history of alcohol misuse or substance abuse, per the discretion of the principal investigator, within 12 months prior to screening.
18, 12 Background UC Therapy
[0450] Subjects should remain on their stable UC background therapy, consisting of oral 5-ASA with or without prednisone < 20 mg/day (or equivalent) or budesonide or budesonide MMX of < 9 mg/day, from 2 weeks prior to screening flexible sigmoidoscopy/colonoscopy used to assess the baseline Mayo score and for the duration of their time on study.
[0451] Additional UC therapies may not be started between 2 weeks prior to the screening flexible sigmoidoscopy/colonoscopy to the Week 16 Follow-up visit. Subjects requiring introduction of new therapies to treat UC will be discontinued from IP.
18, 13 Efficacy Assessments
Clinical, Endoscopic and Histologic Assessment
[0452] Throughout the study, subjects will capture stool frequency and rectal bleeding symptoms on a daily basis in an eDiary. For evaluation of eligibility, the rectal bleeding and stool frequency subscores of the Mayo score will each be calculated based on the most recent 3 days with data within 14 days prior to Day 1, excluding the day of flexible sigmoidoscopy/colonoscopy and on the day before and day after bowel preparation, if performed. Subjects bowel preparation for flexible sigmoidoscopy/colonoscopy should follow standard clinical practice. Subjects will be instructed that a stool is defined as a trip to the toilet when the subject has either a bowel movement, or passes blood alone, blood and mucus, or mucus only.
[0453] Screening and Week 12 endoscopy subscores will be calculated by both a blinded central reader and a blinded local endoscopist. Differences between the 2 readers will be adjudicated by a second blinded central reader as described in the Imaging Charter. Centrally read endoscopic subscores will be used for both eligibility determination and efficacy analyses. Histologic scores (eg, RHI) will be evaluated by a blinded central reviewer.
[0454] Flexible sigmoidoscopy/colonoscopy and histology assessments will consist of assessing the Mayo endoscopic subscores of the sigmoid colon at approximately 15 to 25 cm from the anal verge. Six biopsies will be obtained during the endoscopic procedures from the area that appears to be most affected by UC in this sigmoid colon segment.
[0455] Biopsies will be evaluated for PK, gene expression, and immunohistochemistry signals of biological activity. Mayo Score and Modified Mayo Score
[0456] Mayo score is used for inclusion into the study rather than a Modified Mayo score (used to evaluate efficacy) given the limited information available at the study inception regarding Modified Mayo score cut points that define a mild-to-moderate patient population.
[0457] The Mayo score is a conventional assessment of UC disease activity and is commonly used in clinical studies of UC. The score is composed of four subscores: Stool frequency, Rectal bleeding, Physician Global Assessment (PGA), and Endoscopy (where the value of 1 does not include friability), each rated from 0-3, that are summed to give a total score ranging from 0 to 12 points (higher scores indicating greater severity).
[0458] The Modified Mayo score is an endpoint measure composed of three subscores: Stool frequency, Rectal bleeding, and Endoscopy (where the value of 1 does not include friability), each rated from 0 to 3, that are summed to give a total score ranging from 0 to 9 points (higher scores indicating greater severity).
[0459] The Partial Mayo score is an endpoint measure composed of three subscores: Stool frequency, Rectal bleeding, and PGA each rated from 0 to 3, that are summed to give a total score ranging from 0 to 9 points (higher scores indicating greater severity).
[0460] The 6-point Mayo score is an endpoint measure composed of two sub scores: Stool frequency and Rectal bleeding each rated from 0 to 3, that are summed to give a total score ranging from 0 to 6 points (higher scores indicating greater severity).
Robarts Histopathology Index (RHI) Score, Geboes Score and Ulcerative Colitis Index- 100
[0461] The RHI is a validated instrument that measures histological disease activity in UC. The RHI consists of 4 items: chronic inflammatory infiltrate, lamina propria neutrophils, neutrophils in epithelium, and erosion or ulceration. The RHI score ranges from 0-33 and is derived from Geboes score (Mosli et al., Gut (2017), 50-58).
[0462] The Geboes score is divided into 6 grades: architectural changes [grade 0], chronic inflammatory infiltrate [grade 1], lamina propria neutrophils and eosinophils [grade 2], neutrophils in epithelium [grade 3], crypt destruction [grade 4] and erosions or ulcerations [grade 5], The Geboes score is calculated as the highest grade with a corresponding subgrade score > 0, excluding Geboes Grade 2 A, and ranges from 0 to 5. [0463] The Ulcerative Colitis Index- 100 (UC-100) score is the composite index (1 + 16 x Stool frequency sub score [0 to 3] + 6 x Endoscopy sub score [0 to 3] + 1 x RHI score [0 to 33]) (Jairath et al., Lancet Gastroenterol Hepatol. (2019), 63-70).
[0464] Example 19 - Preparation of Immediate Release Pellets - Compound 1 Free Base
[0465] The IR pellets comprise an inert core coated with a drug layer.
Inert core = Sugar spheres mesh 45/60 (250-355 microns);
Drug layer = Compound 1 (free base) - active agent
Hydroxypropyl methyl cellulose (HPMC E5) - binder
Pol oxamer 188 - wetting agent
Coat = Hydroxypropyl methyl cellulose (HPMC E5) - seal coat (optional) TABLE 40: EXAMPLE 20 DRUG LAYERED CORE FORMULATION
Figure imgf000134_0001
[0466] HPMC E5 (42.68 g), poloxamer 188 (3.05 g) and PEG 6000 (1.22 g) were added to purified water (1097.6 g) and the mixture stirred to give a solution. Compound 1 Free Base (75.0 g) was added and the mixture stirred for 30 min to give a uniform dispersion (pH of dispersion in the range 6-8).
[0467] Sugar spheres mesh 45/60 beads (500 g) were charged to a GPCG 2 (Glatt) fluidized bed Labsystem and pre-heated to a product temperature of 37°C ± 2°C. The aqueous Compound 1 spray dispersion prepared above was sprayed onto the beads under the following conditions:
TABLE 41 : FLUIDIZED BED DRUG LAYERING CONDITIONS
Figure imgf000134_0002
Figure imgf000135_0001
[0468] After completion of spray coating, the coated beads were dried at 40°C inlet temperature at reduced fluidization.
[0469] This gave a core coated with a ca. 10% w/w Compound 1 layer.
[0470] The 10% coated beads were recharged to the GPCG 2 fluidized bed
Labsystem and further spray coated with an aqueous Compound 1 spray dispersion prepared according to the amounts in Table 40 under the conditions in Table 41, to give ca. 20% coated beads.
[0471] Coating the beads with Compound 1 free base gave improved uniformity of drug coating compared to coating with the HC1 salt (Example 20). Example 19 pellets have the composition according to Table 42:
TABLE 42: EXAMPLE 19 PELLET FORMULATION
Figure imgf000135_0002
EXAMPLE 20 - PREPARATION OF IMMEDIATE RELEASE PELLETS - COMPOUND 1 HCL SALT
[0472] Example 20 IR pellets are prepared analogously to the Example 19 IR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base. Example 20 pellets have the composition according to Table 43:
TABLE 43 : EXAMPLE 20 PELLET FORMULATION
Figure imgf000136_0001
EXAMPLE 21 - PREPARATION OF DELAYED RELEASE (PH 5.5) PELLETS - COMPOUND 1 FREE BASE
[0473] The DR pellets comprise an inert core coated with a drug layer. The drug layer coated core is then covered with a seal coating and a delayed release coating.
Inert core = Sugar spheres mesh 45/60 (250-355 microns)
Drug layer = Compound 1 (free base) - active agent
Hydroxypropyl methyl cellulose (HPMC E5) - binder
Pol oxamer 188 - wetting agent pH 7 PBS - buffering solution
Seal coat = Hydroxypropyl methyl cellulose (HPMC E5)
Delayed release coat = Eudragit® L 30 D 55 - pH 5.5 coat
PlasAcryl® T20 - anti-tacking agent
[0474] The drug-layered core is prepared according to the fluidized bed spray coating process outlined above for Example 19, using the amounts listed in Table 40.
[0475] A seal coat is then applied to the drug-layered core, wherein HPMC E5 (18 g) dissolved in purified water (207 g) is spray coated on to drug-layered beads (600 g) using a GPCG 2 (Glatt) fluidized bed Labsystem under the following conditions in Table 44: TABLE 44: FLUIDIZED BED DRUG LAYERING CONDITIONS
Figure imgf000137_0001
[0476] After completion of spray coating, the coated beads are dried at 40°C inlet temperature at reduced fluidization.
[0477] The drug layered pellets are thereby seal-coated to a 3% weight gain.
[0478] A delayed release coat is then applied as follows: Eudragit® L 30 D 55 (105 g) and PlasAcryl® T20 (10.5 g) are dissolved in purified water (461 ml) and then spray coated on to the seal-coated beads (350 g) using a GPCG 2 (Glatt) fluidized bed Labsystem under the following conditions in Table 45:
TABLE 45 : FLUIDIZED BED DRUG LAYERING CONDITIONS
Figure imgf000137_0002
Figure imgf000138_0001
[0479] After completion of spray coating, the coated beads are dried at 40°C inlet temperature at reduced fluidization and atomization air pressure.
[0480] The pellets are thereby coated with a pH 5.5 delayed release coating to a 30% weight gain to give a pellet composition according to Table 46:
TABLE 46: EXAMPLE 21 PELLET FORMULATION
Figure imgf000138_0002
EXAMPLE 22 - PREPARATION OF DELAYED RELEASE (PH 5.5) PELLETS - COMPOUND 1 HCL SALT
[0481] Example 22 DR pellets are prepared analogously to the Example 21 DR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base.
EXAMPLE 23 - PREPARATION OF DELAYED RELEASE (PH 7.0) PELLETS -
COMPOUND 1 FREE BASE
[0482] Drug-layered pellets covered in a 3% weight gain seal coating are prepared according to the procedures described in Example 21. [0483] Example 23 seal-coated pellets were subjected to dissolution testing to determine the rate of Compound 1 release from the composition. Fig. 19A shows the dissolution rate of Compound 1 from seal-coated pellets (60 mg compound) encapsulated in HPMC capsules according to USP2 dissolution testing for 10 hours in pH 7.4 phosphate buffer with 2% CTAB. Almost 90% drug release had occurred within 2 hours.
[0484] A delayed release coat is then applied according to the procedure described in Example 21, substituting Eudragit® FS 30 D for Eudragit® L 30 D 55, to give a ca. 30% weight gain DR coat. Example 23 pellets have the composition according to Table 47.
TABLE 47: EXAMPLE 23 PELLET FORMULATION
Figure imgf000139_0001
[0485] Example 23 DR pellets were subjected to dissolution testing to determine the rate of Compound 1 release from the composition. Fig. 19B shows the dissolution rate of Compound 1 from Example 23 pellets (60 mg compound) coated with 20% Eudragit®® FS 30 D and encapsulated in HPMC capsules according to USP2 dissolution testing for 2 hours in pH 1.2 aqueous media followed by 10 hours in pH 7.4 phosphate buffer with 2% CTAB. Less than 5% drug release has occurred after 2 hr in acid, however, after switching to pH 7.4 almost 90% drug release occurred within 1 hour.
[0486] Example 24 - Preparation of Delayed Release (pH 7.0) Pellets - Compound 1 HC1 salt
[0487] Example 24 DR pellets are prepared analogously to the Example 23 DR pellets substituting Compound 1 (HC1 salt) for Compound 1 free base, to yield a multiparticulate composition according to Table 48.
TABLE 48: EXAMPLE 24 PELLET FORMULATION
Figure imgf000139_0002
Figure imgf000140_0001
EXAMPLE 25 - PREPARATION OF 1 : 1 IR/DR MODIFIED RELEASE CAPSULES
[0488] HPMC capsules are filled with a mixture of IR and DR pellets according to the amounts specified in Table 49.
TABLE 49: IR/DR CAPSULE COMPOSITIONS
Figure imgf000140_0002
EXAMPLE 26 - PREPARATION OF IMMEDIATE RELEASE MINI-TABLETS - TRIAL EXPERIMENTS
[0489] Trials were conducted to investigate whether it was possible to produce minitablets (2.5 mm average diameter) with a high drug load (> 50% w/w Compound 1) to target 240 mg strength capsules comprising IR/DR minitablets.
[0490] The initial formulation used were Examples 26A to 26C prepared according to the compositions shown in Table 50. TABLE 50: FORMULA COMPOSITION OF EXAMPLE 26A-C TRIAL MINITABLETS
Figure imgf000141_0001
[0491] Examples 26A to 26C were formulated by fluid bed wet granulation followed by drying. The granulates had poor flow character and relatively low average particle size - particle size distribution (PSD) featured the greatest percentage of particles retained by the 75 pm or 90 pm sieves. When the granulate was subjected to compression to form mini -tablets, issues were encountered with material sticking to the compression punches. The average mini-tablet thickness was also less than 2.3 mm, below the target of 2.5 mm.
[0492] To attempt to overcome some of these issues, the level of the magnesium stearate lubricant was increased from 1% w/w to 1.5%, 2.0% and 2.5% w/w. 2.5% w/w of an alternative lubricant (sodium stearyl fumarate) was also investigated. In all cases, although the stickiness of the material to the punches was not eliminated, it decreased with increasing lubricant level. Sodium stearyl fumarate showed better compressibility and less sticking than the equivalent amount of magnesium stearate.
[0493] As tablet sticking was still an issue, different levels and combinations of lubricants were investigated, as shown in Table 51.
TABLE 51 : FORMULA COMPOSITION OF EXAMPLE 26D-F TRIAL MINITABLETS
Figure imgf000142_0001
[0494] Example 26D mini-tablets did not stick when compressed, but did suffer from poor powder flow. Example 26E exhibited negligible sticking to the compression punches and once it had been passed through a 40 mesh screen prior to compression, it exhibited improved powder flow and cohesion. Example 26F gave good flow and cohesion after having been passed through a 40 mesh screen prior to compression; this formulation was less optimal than Example 26E, however, as sticking on the lower punches at the end of compression was observed.
[0495] Example 26E provided increased average particle size (PSD) featured the greatest percentage of particles retained by the 150 pm to 250 pm sieves) and could be compressed to give mini-tablets having a mean thickness of 2.34 mm. [0496] Therefore, the trial experiments surprisingly identified compositions which were suitable for producing mini-tablets of the desired dimensions with a high drug load (> 60% w/w of Compound 1) by fluid bed granulation, despite low levels of wetting agent (polysorbate 80) being used. Furthermore, it was found that a combination of two lubricants (e.g. magnesium stearate and hydrogenated vegetable oil), at approximately 2.5% w/w loading each, was optimal in delivering a free flowing granulate that was compressible into mini-tablets without encountering sticking issues.
EXAMPLE 27 - PREPARATION OF IMMEDIATE RELEASE MINI-TABLETS -
COMPOUND 1 HCL SALT
[0497] The composition of the IR mini-tablets is provided in Table 52.
TABLE 52: FORMULA COMPOSITION OF IR MINITABLETS
Figure imgf000143_0001
Figure imgf000144_0001
1 This ingredient is a manufacturing aid that is removed during manufacturing, 10% Opadry® w/w.
[0498] The IR minitablets were prepared via a fluidized bed wet granulation process, as described below:
[0499] Compound 1 HC1 salt, lactose monohydrate, pregelatinized starch, hydroxypropyl cellulose, crospovidone and polysorbate 80, colloidal silicon dioxide, hydrogenated vegetable oil and magnesium stearate were weighed and dispensed into separate suitable sized containers.
[0500] Lactose monohydrate, pregelatinized starch and crospovidone (intragranular) were passed through a 20 mesh screen and premixed with Compound 1 HC1 salt in a 16 quarts V-blender. The loss on drying (LOD) of the pre-blend was measured. This blend was transferred to a fluidized bed granulator (Huttlin - large bowl).
[0501] Hydroxypropyl cellulose and polysorbate 80 were mixed in purified water slowly to minimize foam formation. This granulating liquid was then added to the blend in the fluidized bed granulator and granulation was carried out until all the liquid had been added. The LOD of the wet granulation was measured. The wet granules were dried in the same fluidized bed granulator and drying was continued until the target LOD result (less than 3% w/w) had been achieved. The dry granules were then passed through a Comil 197 and transferred to a 16 quarts V-blender and the weight recorded.
[0502] The following extragranular materials (crospovidone, colloidal silicon dioxide and hydrogenated vegetable oil) were passed through a 20 mesh screen and added to the granules in the V-blender and blended. To the blend was then added magnesium stearate (screened previously through a 30 or 40 mesh screen) and further blending was carried out.
[0503] The resultant blend was compressed using a rotary tablet machine (XL- 100 Korsch press) at an average core weight of 14 mg. During compression, the following in- process checks were monitored: weight, thickness, hardness, friability and disintegration time
[0504] The mini-tablets were then dedusted and metal checked and transferred to a suitably sized pan (LDCS XP - 1 IL) and sprayed with a solution of Opadry® 03F180012 White in purified water until the calculated target weight gain had been achieved during in- process monitoring.
EXAMPLE 28 - PREPARATION OF DELAYED RELEASE MINI-TABLETS -
COMPOUND 1 HCL SALT
[0505] The composition of the DR mini -tablets is provided in Table 53.
TABLE 53 : FORMULA COMPOSITION OF DR MINITABLETS
Figure imgf000145_0001
1 This ingredient is a manufacturing aid that is removed during manufacturing, 10% Opadry® w/w.
2 This ingredient is a manufacturing aid that is removed during manufacturing, 10% Acryl-Eze® w/w.
[0506] The DR minitablets were prepared via a fluidized bed wet granulation process, as described above for the IR minitablets; the IR minitablets were then charged to a suitably sized pan (LDCS XP - 1 IL) and sprayed with a solution of Acryl-Eze® (93018359) White in purified water until the calculated target weight gain had been achieved during in-process monitoring. EXAMPLE 29 - PREPARATION OF 240 MG IR/DR (1 : 1) CAPSULES CONTAINING MINI-TABLETS
[0507] The composition of the Example 29 IR/DR mini-tablet capsules is provided in
Table 54.
TABLE 54: FORMULA COMPOSITION OF 240 MG CAPSULE
Figure imgf000146_0001
theoretical unit weight for capsule size 00
[0508] Each capsule contains 15 IR minitablets (thereby providing 135.065 mg of Compound 1 HC1 salt, equivalent to 120 mg of parent compound) and 15 DR minitablets ((thereby providing 135.065 mg of Compound 1 HC1 salt, equivalent to 120 mg of parent compound).
[0509] The capsules (size 00 white opaque gelatin capsules) were filled in a capsule filler (Ultra 8) with equal amounts of the Example 27 IR minitablets and Example 28 DR minitablets prepared as described above. During encapsulation, the following in-process checks were monitored: weight and disintegration time. After encapsulation the capsules were dedusted and metal checked.
EXAMPLE 30 - DISSOLUTION RATES OF 240 MG IR/DR (1 : 1) CAPSULES CONTAINING MINI-TABLETS
[0510] The dissolution rates of the Example 29 IR/DR capsules comprising Compound 1 • HC1, were determined utilizing USP Apparatus 2 (Paddles) with quantitation by HPLC with UV detection. Table 55 describes the parameters for the dissolution testing.
TABLE 55: DISSOLUTION PARAMETERS
Figure imgf000146_0002
Figure imgf000147_0001
SLS = sodium lauryl sulfate
[0511] Procedure for the acid stage: place 750 mL 0.01N HC1 in the vessel and equilibrate the medium to 37.0 ± 0.5 °C. Drop the capsule in the vessel and start the dissolution apparatus at 75 rpm. At the specified time points, withdraw 1.5 mL of solution from mid-zone of the vessel and filter into a vial for analysis.
[0512] Procedure for the buffer stage: while the paddle is still rotating at 75 rpm, add 250 mL of 200 mM NasPOi buffer + 8% SLS, pH 7.0 (already equilibrated to 37.0 ± 0.5 °C) to the vessel to give a final buffer composition of 50 mM NasPOi buffer + 2% SLS, pH 6.8. Adjust to pH 6.8 if necessary. Continue the dissolution and withdraw 1.5 mL of solution from mid-zone of the vessel at specified time points. Filter into a vial for analysis.
[0513] The dissolution samples were then analyzed by HPLC using the parameters in Table 56 and the gradient in Table 57.
TABLE 56: HPLC PARAMETERS
Figure imgf000147_0002
TABLE 57: GRADIENT ELUTION
Figure imgf000148_0001
[0514] The results of the dissolution tests of Example 29 capsules are shown in Figure 20. The IR minitablets (-50%) dissolved rapidly in the acid stage media in about 10 mins. The remaining DR minitablets (-50%) did not begin to dissolve until the switch to pH 6.8 buffer conditions at 120 mins. After 300 mins approximately 95% dissolution had occurred.
EXAMPLE 31 - STABILITY STUDY OF 240 MG IR/DR (1 : 1) CAPSULES CONTAINING MINI-TABLETS
[0515] Capsules prepared according to Example 29 were stored and tested under the Table 58 conditions to analyze their stability in terms of drug loading/purity compared to time T = 0.
TABLE 58: CAPSULE STABILITY SCHEDULE
Figure imgf000148_0002
EXAMPLE 32 - PREPARATION OF CORE MINI-TABLETS
[0516] The composition of the blend used in the preparation of 30 core mini-tablets is provided in Table 59.
TABLE 59: COMPOSITION OF BLENDS FOR CORE MINI-TABLETS
Figure imgf000148_0003
Figure imgf000149_0001
1 Amount of Lactose Monohydrate is adjusted based on the Use-As-Value (UAV) of the API, in order to maintain the percent composition to 100%.
2 Amount of API is adjusted based on its UAV
3 Water aids granulation but is evaporated during drying
[0517] The blend was prepared via a fluidized bed wet granulation process, as described below:
[0518] Compound 1 HC1 salt, lactose monohydrate, pregelatinized starch, hydroxypropyl cellulose, crospovidone and polysorbate 80, colloidal silicon dioxide, sodium stearyl fumarate and magnesium stearate were weighed and dispensed into separate suitable sized containers.
[0519] Lactose monohydrate, pregelatinized starch and crospovidone (intragranular) were passed through a 20 mesh screen and premixed with Compound 1 HC1 salt in a 16 quarts V-blender. The loss on drying (LOD) of the pre-blend was measured. This blend was transferred to a fluidized bed granulator (Huttlin - large bowl).
[0520] Hydroxypropyl cellulose and polysorbate 80 were mixed in purified water slowly to minimize foam formation. This granulating liquid was then added to the blend in the fluidized bed granulator and granulation was carried out until all the liquid had been added. The LOD of the wet granulation was measured. The wet granules were dried in the same fluidized bed granulator and drying was continued until the target LOD result (less than or equal to 3% w/w) had been achieved. The dry granules were then passed through a Comil 197 and transferred to a 16 quarts V-blender and the weight recorded.
[0521] The following extra-granular materials (crospovidone and colloidal silicon dioxide) were passed through a 20 mesh screen and added to the granules in the V-blender and blended. To the blend was then added magnesium stearate (screened previously through a 30 or 40 mesh screen) and sodium stearyl fumarate (screened previously through a 40 mesh screen) and further blending was carried out.
[0522] The 420 mg granulate blend was then compressed into 30 core minitablets of average weight 14 mg per minitablet (8 mg free base API per minitablet).
[0523] Compression was carried out using a rotary tablet machine (XL- 100 Korsch press) with a 2.5 mm diameter tip, 9 multi -tip punch to provide the core minitablets having the properties shown in Table 60.
TABLE 60: PROPERTIES OF CORE MINI-TABLETS
Figure imgf000150_0001
EXAMPLE 33 - PREPARATION OF CAPSULES CONTAINING 15 IR MINI-TABLETS AND 15 DR MINI-TABLETS
[0524] After de-dusting and metal checking, the batch of 30 core mini-tablets from Example 32 (420 mg) was spray-coated with a solution of Opadry® 03F180012 White in purified water to give IR mini -tablets with a 5% weight gain coating (441 mg total weight).
[0525] A batch of 30 IR mini -tablets (441 mg) prepared as described above was spray-coated with a solution of Acryl-Eze® (93018359) White in purified water to give DR mini -tablets with a 10% weight gain coating (485.1 mg total weight).
[0526] Size 00 white opaque gelatin capsules were filled with the IR mini-tablets and DR mini -tablets, so that each capsule contained 15 IR mini -tablets and 15 DR mini -tablets. Therefore each capsule provides a total Compound 1 (free base) dose of 240 mg, split 120mg: 120mg between IR mini-tablets and DR mini-tablets.

Claims

1. A method of treating an inflammatory bowel disease comprising the administration of a combination of an oral immediate release pharmaceutical composition comprising a pharmaceutically acceptable salt of Compound 1 and an oral delayed release pharmaceutical composition comprising Compound 1 or a pharmaceutically acceptable salt thereof.
2. The method according to claim 1, wherein the combination is dosed once daily or twice daily.
3. The method according to claim 1 or 2, wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 100 mg and 1000 mg.
4. The method according to any one of claims 1-3, wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is between 400 mg and 1000 mg.
5. The method according to any one of claims 1-4, wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 480 mg.
6. The method according to any one of claims 1-4, wherein the total daily dose of Compound 1 or a pharmaceutically acceptable salt thereof is about 960 mg.
7. The method according to any one of claims 1-6, wherein the ratio of the amount of Compound 1 in the oral immediate release pharmaceutical composition relative to the amount of Compound 1 in the oral delayed release pharmaceutical composition is between 1 :4 and 4: 1 by weight.
8. The method according to any one of claims 1-7, wherein the ratio of the dose of Compound 1 in the oral immediate release pharmaceutical composition relative to the dose of Compound 1 in the oral delayed release pharmaceutical composition is between 1:2 and 2: 1 by weight.
9. The method according to any one of claims 1-8, wherein the ratio of the dose of Compound 1 in the oral immediate release pharmaceutical composition relative to the dose of Compound 1 in the oral delayed release pharmaceutical composition is about 1 : 1 by weight.
10. The method according to any one of claims 1-9, wherein the oral immediate release pharmaceutical composition comprises 50 to 500 mg of a pharmaceutically acceptable salt of Compound 1.
11. The method according to any one of claims 1-10, wherein the oral immediate release pharmaceutical composition comprises 100 to 250 mg of a pharmaceutically acceptable salt of Compound 1.
12. The method according to any one of claims 1-11, wherein the oral delayed release pharmaceutical composition comprises 50 to 500 mg of Compound 1 or a pharmaceutically acceptable salt thereof.
13. The method according to any one of claims 1-12, wherein the oral delayed release pharmaceutical composition comprises 100 to 250 mg of Compound 1 or a pharmaceutically acceptable salt thereof.
14. The method according to any one of claims 1-13, wherein the amount of a pharmaceutically acceptable salt of Compound 1 contained within the oral immediate release pharmaceutical composition is about 240 mg and the amount of Compound 1 or a pharmaceutically acceptable salt thereof contained within the oral delayed release pharmaceutical composition is also about 240 mg.
15. The method according to claim 14, wherein the combination is dosed twice daily.
16. The method according to any one of claims 1-15, wherein the combination is dosed with food, or up to 30 minutes after a meal.
17. The method according to claims 1-16, wherein the compound in either or both of the oral immediate release pharmaceutical composition and the oral delayed release pharmaceutical composition is a hydrochloride salt of Compound 1.
18. The method according to any one of claims 1-17, wherein the compound in both of the oral immediate release pharmaceutical composition and the oral delayed release pharmaceutical composition is a hydrochloride salt of Compound 1.
19. The method according to any one of claims 1-17, wherein the compound in the oral immediate release pharmaceutical composition is a hydrochloride salt of Compound 1 and the compound in the oral delayed release pharmaceutical composition is the free base of Compound 1.
20. The method according to any one of claims 17-19, wherein the ratio of Compound 1 to HC1 is about 1 : 1.
21. The method according to any one of claims 17-20, wherein the hydrochloride salt is a monohydrate.
22. The method according to any one of claims 1-21, wherein the oral immediate release pharmaceutical composition comprises a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients.
23. The method according to any one of claims 1-22, wherein the oral delayed release pharmaceutical composition is a solid composition comprising Compound 1 or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
151
24. The method according to claim 23, wherein the oral delayed release pharmaceutical composition comprises a core and a delayed release coating substantially encapsulating the core.
25. The method according to claim 24, wherein the core comprises a granulate.
26. The method according to claim 22 or 25, wherein the granulate comprises: 30-50% of granules having particle sizes between 177 and 420 pm; or less than 20% of granules having particle sizes between 125 and 177 pm.
27. The method according to any one of claims 22, 25, or 26, wherein the granulate comprises:
30-50% of granules having particle sizes between 177 and 420 pm; and less than 20% of granules having particle sizes between 125 and 177 pm.
28. The method according to claims 22-27, wherein the one or more pharmaceutically acceptable excipients comprises a wetting agent.
29. The method according to claim 28, wherein Compound 1 or a pharmaceutically acceptable salt thereof is in intimate association with the wetting agent.
30. The method according to claim 28 or 29, wherein the wetting agent is a nonionic wetting agent.
31. The method according to any one of claims 28-30, wherein the wetting agent is a polyol ester, a polyoxyethylene ester, or a poloxamer.
32. The method according to any one of claims 28-31, wherein the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25.
33. The method according to any one of claims 28-32, wherein the wetting agent has a hydrophilic-lipophilic balance (HLB) between 12 and 18.
152
34. The method according to claim 31, wherein the polyol ester comprises one or more glycol ester, glycerol ester, or sorbitan derivative.
35. The method according to claim 34, wherein the sorbitan derivative is polysorbate 20, polysorbate 40, polysorbate 60, or polysorbate 80.
36. The method according to any one of claims 28-35, wherein the wetting agent is present in the composition at 0.5 to 2.0% w/w.
37. The method according to any one of claims 28-36, wherein the wetting agent is present in the composition at 0.75 to 1.25% w/w.
38. The method according to any one of claims 22-37, wherein the one or more pharmaceutically acceptable excipients are not acidic, or have a pH of greater than 6 when dissolved or slurried in water.
39. The method according to any one of claims 22-38, wherein the composition has a moisture level of less than 6% w/w.
40. The method according to any one of claims 22-39, wherein the one or more pharmaceutically acceptable excipients comprises a binder.
41. The method according to claim 40, wherein the binder is a cellulose ether- based binder.
42. The method according to any one of claims 22-41, wherein the one or more pharmaceutically acceptable excipients comprises a disintegrant.
43. The method according to any one of claims 22-42, wherein the one or more pharmaceutically acceptable excipients does not comprise a buffering agent.
44. The method according to any one of claims 1-43, wherein the oral immediate release pharmaceutical composition is formulated as tablets, mini-tablets, or pellets.
45. The method according to any one of claims 1-44, wherein the oral delayed release pharmaceutical composition is formulated as tablets, mini-tablets, or pellets.
46. The method according to claim 44 or 45, wherein both the oral immediate release pharmaceutical composition and the oral delayed release pharmaceutical composition are formulated as mini-tablets.
47. The method according to any one of claims 1-46, wherein the oral immediate release pharmaceutical composition comprises the pharmaceutically acceptable salt of Compound 1 in an about 40-70% w/w loading.
48. The method according to claim 1-47, wherein the oral delayed release pharmaceutical composition comprises Compound 1, or a pharmaceutically acceptable salt thereof, in an about 40-70% w/w loading.
49. The method according to any one of claims 24-28, wherein the delayed release coating dissolves at pH values greater than about 5.5.
50. The method according to any one of claims 24-49, wherein the delayed release coating comprises a methacrylic acid copolymer.
51. The method according to claim 50, wherein the methacrylic acid copolymer is Eudragit® L 100-55, Eudragit® FS30D, Eudragit® L100, Eudragit® L 12,5, Eudragit® L30 D-55, Eudragit® S100, or Eudragit® S12,5.
52. The method according to any one of claims 24-51, wherein the oral delayed release pharmaceutical composition comprises a greater than 8% weight gain delayed release coating.
53. The method according to any one of claims 24-52, wherein the oral delayed release pharmaceutical composition comprises a greater than 10% weight gain.
54. The method according to any one of claims 24-53, wherein the oral delayed release pharmaceutical composition comprises about 12% weight gain delayed release coating.
55. The method according to any one of claims 24-53, wherein the oral delayed release pharmaceutical composition comprises about 14% weight gain delayed release coating.
56. The method according to any one of claims 1-55, wherein the oral immediate release pharmaceutical composition undergoes greater than 85% (such as greater than 95%) disintegration and dispersal in 0.01N hydrochloric acid in less than 4 minutes at 37 °C using USP2 disintegration apparatus.
57. The method according to any one of claims 1-56, wherein the oral immediate release pharmaceutical composition produces a geometric mean maximum plasma concentration of Compound 1 after oral administration of at least 5 ng/ml.
Figure imgf000157_0001
58. The method according to any one of claims 1-57, wherein the oral delayed release pharmaceutical composition undergoes less than 5% dissolution in 0.01N hydrochloric acid after 30 mins at 37 °C using USP2 apparatus.
59. The method according to any one of claims 1-58, wherein either or both of the oral immediate release pharmaceutical composition and the oral delayed release pharmaceutical composition provide colonic tissue exposure after oral dosing of greater than or equal to 100 ng/g.
60. The method according to any one of claims 1-59, wherein the combination, after oral administration in the fed state to a subject suffering from an inflammatory bowel disease produces at least one of the following:
155 a) produces in the subject a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml; or b) produces a ratio of Cmax /AUCo-24 between 0.04 and 0.4 h'1.
61. The method according to any one of claims 1-59, wherein the combination, after oral administration in the fed state to a subject suffering from an inflammatory bowel disease produces at least one of the following: a) produces in the subject a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 50 ng/ml; or b) produces a ratio of Cmax /AUCo-24 between 0.04 and 0.4 h'1.
62. The method according to claims 60 or 61, wherein the ratio of Cmax /AUCo-24 is between 0.04 and 0.3 h'1.
63. The method according to claims 60 or 61, wherein the ratio of Cmax /AUCo-24 is between 0.04 and 0.2 h'1.
64. The method according to claims 60 or 61, wherein the ratio of Cmax /AUCo-24 is between 0.06 and 0.16 h'1.
65. The method according to any one of claims 60-64, wherein the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml.
66. The method according to any one of claims 60-64, wherein the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 20 ng/ml.
67. The method according to any one of claims 1-66, wherein the daily dose of Compound 1 from immediate release compositions is about 240 mg and the daily dose of Compound 1 from delayed release compositions is about 240 mg.
156
68. The method according to any one of claims 1-66, wherein the daily dose of Compound 1 from immediate release compositions is about 480 mg and the daily dose of
Compound 1 from delayed release compositions is about 480 mg.
69. The method according to any one of claims 1-68, wherein the combination, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 100 ng/g.
70. The method according to any one of claims 1-69, wherein the combination, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 200 ng/g.
71. The method according to any one of claims 1-70, wherein the combination, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 300 ng/g.
72. The method according to any one of claims 1-71, wherein the inflammatory bowel disease is ulcerative colitis or Crohn’s disease.
73. The method according to claim 72, wherein the subject being treated with the combination has ulcerative colitis, and wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1, and a stool frequency sub score of > 1.
74. The method according to claim 73, wherein for at least two weeks prior to the treatment the subject has been on a stable dose of an oral 5-ASA.
75. The method according to claim 73 or 74, wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0, and a stool frequency subscore of 0 or 1.
157
76. An oral modified release pharmaceutical composition wherein the composition comprises an immediate release component and a delayed release component, wherein each component comprises Compound 1 or a pharmaceutically acceptable salt thereof.
77. The oral modified release pharmaceutical composition according to claim 76, wherein the immediate release component comprises the hydrochloride salt of Compound 1.
78. The oral modified release pharmaceutical composition according to claim 76 or 77, wherein the delayed release component comprises the free base form of Compound 1.
79. The oral modified release pharmaceutical composition according to any one of claims 76-78, wherein the ratio of immediate release component to delayed release component is between about 1 :4 and 4: 1.
80. The oral modified release pharmaceutical composition according to any one of claims 76-79, wherein the ratio of immediate release component to delayed release component is between about 1 :2 and 2: 1.
81. The oral modified release pharmaceutical composition according to any one of claims 76-80, wherein the ratio of immediate release component to delayed release component is about 1 : 1.
82. The oral modified release pharmaceutical composition according to any one of claims 76-81, wherein the immediate release component comprises 50 to 500 mg of a pharmaceutically acceptable salt of Compound 1.
83. The oral modified release pharmaceutical composition according to any one of claims 76-82, wherein the immediate release component comprises 100 to 250 mg of a pharmaceutically acceptable salt of Compound 1.
158
84. The oral modified release pharmaceutical composition according to any one of claims 76-83, wherein the delayed release component comprises 50 to 500 mg of Compound 1 or a pharmaceutically acceptable salt thereof.
85. The oral modified release pharmaceutical composition according to any one of claims 76-83, wherein the delayed release component comprises 100 to 250 mg of Compound 1 or a pharmaceutically acceptable salt thereof.
86. The oral modified release pharmaceutical composition according to any one of claims 76-85, wherein the composition is a matrix dosage form or a multi-particulate system.
87. The oral modified release pharmaceutical composition according to claim 86, wherein the composition is a matrix dosage form in the form of a tablet comprising a water erodible matrix.
88. The oral modified release pharmaceutical composition according to claim 86, wherein the composition is a multi-particulate composition comprising a population of pellets, mini-tablets, or beads.
89. The oral modified release pharmaceutical composition according to claim 88, wherein the multi-particulate composition comprises a population of pellets, mini-tablets, and/or beads encapsulated within a capsule.
90. The oral modified release pharmaceutical composition according to claim 88 or 89, wherein the immediate release component and the delayed release component both comprise mini-tablets.
91. The oral modified release pharmaceutical composition according to claim 90, wherein the mini -tablets have an average diameter of 1.5 to 3.0 mm.
92. The oral modified release pharmaceutical composition according to claim 90 or 91, wherein the mini -tablets have an average diameter of 2.0 to 2.5 mm.
159
93. The oral modified release pharmaceutical composition according to any one of claims 90-92, wherein the mini-tablets comprise a granulate, wherein the granulate comprises a pharmaceutically acceptable salt of Compound 1 and one or more pharmaceutically acceptable excipients.
94. The oral modified release pharmaceutical composition according to claim 93, wherein the granulate comprises the pharmaceutically acceptable salt of Compound 1 in a loading of greater than 40 % w/w.
95. The oral modified release pharmaceutical composition according to claim 93 or 94, wherein the granulate comprises the pharmaceutically acceptable salt of Compound 1 in a loading between about 60 to 70 % w/w.
96. The oral modified release pharmaceutical composition according to any one of claims 93-95, wherein the immediate release component comprises 80 to 160 mg of a pharmaceutically acceptable salt of Compound 1 and the delayed release component comprises 80 to 160 mg of a pharmaceutically acceptable salt of Compound 1.
97. The oral modified release pharmaceutical composition according to any one of claims 93-96, wherein the granulate comprises a wetting agent.
98. The oral modified release pharmaceutical composition according to claim 97, wherein the wetting agent is a non-ionic wetting agent.
99. The oral modified release pharmaceutical composition according to claim 98, wherein the non-ionic wetting agent is a polyol ester, a polyoxyethylene ester, or a poloxamer.
100. The oral modified release pharmaceutical composition according to any one of claims 97-99, wherein the wetting agent has a hydrophilic-lipophilic balance (HLB) between 5 and 25.
160
101. The oral modified release pharmaceutical composition according to any one of claims 97-100, wherein the wetting agent has a hydrophilic-lipophilic balance (HLB) between 12 and 18.
102. The oral modified release pharmaceutical composition according to claim 99, wherein the polyol ester is a glycol ester, a glycerol ester, or a sorbitan derivative.
103. The oral modified release pharmaceutical composition according to claim 102, wherein the sorbitan derivative is polysorbate 20, polysorbate 40, polysorbate 60, or polysorbate 80.
104. The oral modified release pharmaceutical composition according to any one of claims 97-103, wherein the wetting agent is present in the composition from 0.5 to 3.0% w/w.
105. The oral modified release pharmaceutical composition according to any one of claims 97-104, wherein the wetting agent is present in the composition from 1.5 to 2.5% w/w.
106. The oral modified release pharmaceutical composition according to any one of claims 93-105, wherein the granulate comprises one or more lubricants.
107. The oral modified release pharmaceutical composition according to claim 106, wherein the one or more lubricants are selected from magnesium stearate, calcium stearate, hydrogenated vegetable oil, stearic acid, sodium stearyl fumarate, mineral oil, hydrogenated vegetable oil, and polyethylene glycol.
108. The oral modified release pharmaceutical composition according to claim 107, wherein the one or more lubricants comprise magnesium stearate and either hydrogenated vegetable oil or sodium stearyl fumarate.
161
109. The oral modified release pharmaceutical composition according to any one of claims 106-108, wherein the one or more lubricants are present in the composition at a total level of between 3 and 7% w/w.
110. The oral modified release pharmaceutical composition according to any one of claims 106-109, wherein the one or more lubricants are present in the composition at a total level of between 4 and 6% w/w.
111. The oral modified release pharmaceutical composition according to claim 110, wherein magnesium stearate is present in the granulate at between 2 and 4% w/w, and hydrogenated vegetable oil or sodium stearyl fumarate is present in the composition at between 2 and 4% w/w.
112. The oral modified release pharmaceutical composition according to any one of claims 97-111, wherein the one or more pharmaceutically acceptable excipients are not acidic, or have a pH of greater than 6 when dissolved or slurried in water.
113. The oral modified release pharmaceutical composition according to any one of claims 90-112, wherein the delayed release mini-tablets are individually substantially coated with a delayed release coating.
114. The oral modified release pharmaceutical composition according to claim 113, wherein the delayed release coating dissolves at pH values greater than about 4.5.
115. The oral modified release pharmaceutical composition according to claim 113 or 114, wherein the delayed release coating dissolves at pH values greater than about pH 5.5.
116. The oral modified release pharmaceutical composition according to any one of claims 113-115, wherein each delayed release mini -tablet comprises a greater than 6% weight gain delayed release coating.
162
117. The oral modified release pharmaceutical composition according to any one of claims 113-116, wherein each delayed release mini-tablet comprises between 8 and 12% weight gain delayed release coating.
118. The oral modified release pharmaceutical composition according to any one of claims 76-117, wherein said composition, after oral administration in the fed state to a subject in need of treatment thereof exhibits at least one of the following: a) produces in the subject a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 100 ng/ml; or b) produces a ratio of Cmax /AUCo-24 between 0.04 and 0.4 h'1.
119. The oral modified release pharmaceutical composition according to any one of claims 76-117, wherein said composition, after oral administration in the fed state to a subject in need of treatment thereof exhibits both of the following: a) produces in the subject a geometric mean maximum plasma concentration (Cmax) of Compound 1 of less than 50 ng/ml; and b) produces a ratio of Cmax /AUCo-24 between 0.04 and 0.4 h'1.
120. The oral modified release pharmaceutical composition according to claim 118 or 119, wherein the ratio of Cmax /AUCo-24 is between 0.04 and 0.3 h'1.
121. The oral modified release pharmaceutical composition according to any one of claims 118-120, wherein the ratio of Cmax /AUCo-24 is between 0.04 and 0.2 h'1.
122. The oral modified release pharmaceutical composition according to any one of claims 118-121, wherein the ratio of Cmax /AUCo-24 is between 0.06 and 0.16 h'1.
123. The oral modified release pharmaceutical composition according to any one of claims 118-122, wherein the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 30 ng/ml.
163
124. The oral modified release pharmaceutical composition according to any one of claims 118-123, wherein the geometric mean maximum plasma concentration (Cmax) of Compound 1 is less than 20 ng/ml.
125. The oral modified release pharmaceutical composition according to any one of claims 118-124, wherein the composition when administered orally to the subject provides a dose of Compound 1 selected from a range of 120 to 600 mg.
126. The oral modified release pharmaceutical composition according to any one of claims 76-125, wherein the composition, when administered orally to a subject in a fed state, provides a dose of Compound 1 selected from about 240 mg and about 480 mg and a ratio of Cmax / AUCO-24 between 0.04 and 0.2 h'1.
127. The oral modified release pharmaceutical composition according to any one of claims 76-126, wherein the composition, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 100 ng/g.
128. The oral modified release pharmaceutical composition according to any one of claims 76-127, wherein the composition, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 200 ng/g.
129. The oral modified release pharmaceutical composition according to any one of claims 76-128, wherein the composition, after oral administration, provides colonic tissue concentrations of Compound 1 greater than 300 ng/g.
130. A method for treating a warm blooded subject, preferably a human, suffering from an inflammatory bowel disease, the method comprising administering to the subject an oral modified release pharmaceutical composition according to any one of claims 76-129.
131. The method according to claim 130, wherein the inflammatory bowel disease is ulcerative colitis or Crohn’s disease.
132. The method according to claim 130 or 131, wherein the composition is administered once daily or twice daily.
133. The method according to any one of claims 130-132, wherein the composition is administered with food, or up to 30 minutes after a meal.
134. The method for treating ulcerative colitis according to any one of claims 131— 133, wherein prior to the treatment the subject has a Mayo score of 5-10, an endoscopic subscore of > 2, a rectal bleeding subscore of > 1, and a stool frequency subscore of > 1.
135. The method according to claim 134, wherein for at least two weeks prior to the treatment the subject has been on a stable dose of an oral 5-ASA.
136. The method according to claim 134 or 135, wherein after at least 12 weeks of the treatment, the subject has a Modified Mayo score of < 2, an endoscopic subscore of 0 or 1, a rectal bleeding subscore of 0, and a stool frequency subscore of 0 or 1.
PCT/US2021/046017 2020-08-13 2021-08-13 Compositions and methods of treatment WO2022036267A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063065390P 2020-08-13 2020-08-13
US63/065,390 2020-08-13
US202163197269P 2021-06-04 2021-06-04
US63/197,269 2021-06-04

Publications (1)

Publication Number Publication Date
WO2022036267A1 true WO2022036267A1 (en) 2022-02-17

Family

ID=77655682

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/046017 WO2022036267A1 (en) 2020-08-13 2021-08-13 Compositions and methods of treatment

Country Status (1)

Country Link
WO (1) WO2022036267A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110112055A1 (en) * 2009-11-06 2011-05-12 Gardner Joseph H Compositions and methods for treating colitis
WO2021119033A1 (en) * 2019-12-09 2021-06-17 Gb004, Inc. Compositions and methods of treatment

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110112055A1 (en) * 2009-11-06 2011-05-12 Gardner Joseph H Compositions and methods for treating colitis
WO2011057121A1 (en) 2009-11-06 2011-05-12 Akebia Therapeutics Inc. Compositions and methods for treating colitis
WO2021119033A1 (en) * 2019-12-09 2021-06-17 Gb004, Inc. Compositions and methods of treatment

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2012
GRIFFIN, J. SOC. COSMETIC CHEM., 1949, pages 311 - 326
GRIFFIN, J., SOC. COSMETIC CHEM., 1954, pages 249 - 256
JAIRATH ET AL., LANCET GASTROENTEROL HEPATOL., 2019, pages 63 - 70
MOSLI ET AL., GUT, 2017, pages 50 - 58

Similar Documents

Publication Publication Date Title
US11648206B2 (en) Sustained-release formulations of colchicine and methods of using same
EP1878444B1 (en) Composition containing anti-dementia drug
KR101699912B1 (en) Pharmaceutical formulation comprising one or more fumaric acid esters in an erosion matrix
EP1443917B1 (en) Tamsulosin tablets
US20060246003A1 (en) Composition containing anti-dementia drug
JP5479909B2 (en) New formulation
JP5205053B2 (en) Tranexamic acid preparation
KR20090114330A (en) Pharmaceutical formulation
JP4457003B2 (en) Controlled release pharmaceutical composition
US20220175766A1 (en) Compositions and methods of treatment
JP2010536798A (en) Method and composition for controlling bioavailability of poorly soluble drugs
US20070292505A1 (en) Controlled release alfuzosin hydrochloride formulation
WO2022036267A1 (en) Compositions and methods of treatment
TW202207926A (en) Pharmaceutical formulations of abiraterone acetate and niraparib
JP6328138B2 (en) Of N- [5- [2- (3,5-dimethoxyphenyl) ethyl] -2H-pyrazol-3-yl] -4-[(3R, 5S) -3,5-dimethylpiperazin-1-yl] benzamide Pharmaceutical formulation
US10335376B2 (en) Raloxifene sprinkle composition
US20210353546A1 (en) Dual release pharmaceutical compositions comprising the combination of a beta-3 adrenoreceptor agonist and a muscarinic receptor antagonist
JP6697539B2 (en) Colchicine sustained-release preparation and method of using the same
EA046114B1 (en) ATR KINASE INHIBITOR BAY 1895344 FOR USE IN THE TREATMENT OF HYPERPROLIFERATIVE DISEASE

Legal Events

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

Ref document number: 21766323

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21766323

Country of ref document: EP

Kind code of ref document: A1