US20200316093A1 - Abiraterone acetate lipid formulations - Google Patents

Abiraterone acetate lipid formulations Download PDF

Info

Publication number
US20200316093A1
US20200316093A1 US16/906,603 US202016906603A US2020316093A1 US 20200316093 A1 US20200316093 A1 US 20200316093A1 US 202016906603 A US202016906603 A US 202016906603A US 2020316093 A1 US2020316093 A1 US 2020316093A1
Authority
US
United States
Prior art keywords
propylene glycol
glycerol
active
dosage form
fatty acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/906,603
Inventor
Hywel Williams
Prashant Agarwal
Eduardo Jule
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Capsugel Belgium NV
Original Assignee
Capsugel Belgium NV
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 Capsugel Belgium NV filed Critical Capsugel Belgium NV
Priority to US16/906,603 priority Critical patent/US20200316093A1/en
Assigned to CAPSUGEL BELGIUM NV reassignment CAPSUGEL BELGIUM NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JULE, Eduardo, AGARWAL, PRASHANT, WILLIAMS, HYWEL
Publication of US20200316093A1 publication Critical patent/US20200316093A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed

Definitions

  • lipid compositions comprising abiraterone acetate.
  • Lipid multiparticulates are known. See for example EP1030687, U.S. Pat. No. 6,572,892, EP1827382, U.S. Pat. Nos. 7,235,260, 7,887,844, EP1691787, U.S. Pat. No. 7,625,507.
  • Abiraterone acetate (Zytiga® tablets) was developed by Janssen, and approved in 2011 for use in combination with prednisone for the treatment of patients with metastatic castration-resistant prostate cancer.
  • Abiraterone acetate has the following structure:
  • Abiraterone acetate is a prodrug of abiraterone, which inhibits 17 ⁇ -hydroxylase/C17,20-lyase (CYP17) expressed in testicular, adrenal, and prostatic tumor tissues.
  • Abiraterone acetate is highly lipophilic (Log P 5.12) and as a result suffers from low aqueous solubility in the gastrointestinal tract.
  • Zytiga® tablets containing abiraterone acetate show a clinically significant positive food effect when administered with low-fat (7- and 5-fold increase in Cmax and AUC 0- ⁇ , respectively) or high-fat (17- and 10-fold increase in C max and AUC 0- ⁇ , respectively).
  • compositions comprising abiraterone acetate and a lipid matrix.
  • the lipid matrix comprises a) at least one fatty acid ester, b) at least one surfactant, and c) optionally an antioxidant.
  • the abiraterone acetate comprises at least 0.2 wt % of the composition
  • at least one fatty acid ester comprises at least 10 wt % of the composition
  • at least one surfactant comprises at least 10 wt % of the composition.
  • the compositions are liquid at ambient temperature.
  • the compositions are semi-solid at ambient temperature.
  • abiraterone is molecularly dispersed in the lipid matrix.
  • the lipid matrix comprises a) at least one low flow point excipient, b) at least one high flow point excipient, c) at least one low-flow point surfactant, and c) optionally an antioxidant.
  • the abiraterone acetate comprises at least 0.2 wt % of the composition
  • the at least one low flow point excipient comprises at least 10 wt % of the composition
  • the at least one high flow point excipient comprises at least 5 wt % of the composition
  • the at least one low-flow point surfactant comprises at least 10 wt % of the composition.
  • the compositions comprise a plurality of particles that are solid at ambient temperature, have a generally spherical shape, and have a mean diameter ranging from 40 ⁇ m to 3000 ⁇ m.
  • the lipid matrix is a liquid or semi-solid at ambient temperature.
  • compositions of the invention are filled into capsules.
  • compositions are administered orally to a patient in need of therapy.
  • the lipid matrix is selected from the group consisting of fatty alcohols, fatty acids, fatty acid esters of glycerol, glycols and poly glycols, fatty acid esters of polyglycerol, polyglycolized glycerides, C8-C18 triglycerides, stearoyl polyoxylglycerides, lauroyl macrogol-32 glycerides, caprylocaproyl macrogol-8 glycerides, oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, myristyl alcohol, lauryl alcohol, capric alcohol, glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, stearyl alcohol, behenyl alcohol, palmitic acid, stearic acid, paraffin wax, beeswax, candelilla wax, carnauba wax, polyethoxyl
  • the present disclosure relates to lipid compositions for abiraterone acetate, such as lipid multiparticulate formulations comprising abiraterone acetate and a lipid matrix or liquid fill hard capsules comprising abiraterone acetate and a lipid matrix.
  • the singular forms “a,” “an,” and “the” include the plural forms unless the context clearly dictates otherwise. Additionally, the term “includes” means “comprises.” Further, the term “coupled” generally means electrically, electromagnetically, and/or physically (e.g., mechanically or chemically) coupled or linked and does not exclude the presence of intermediate elements between the coupled or associated items absent specific contrary language.
  • w/w % and “wt %” means by weight as a percentage of the total weight or relative to another component in the composition.
  • excipient is meant any pharmaceutically acceptable ingredient that is included in the formulation that is not the active agent.
  • glyceride is meant a fatty acid ester of glycerol.
  • fatty acid ester of glycerol is meant mono, di, and triesters of glycerides.
  • Glycerides also include glycerol esters that have both fatty acid and poly alkyl oxide esters.
  • the term “flow point” is the temperature at which any portion of the mixture becomes sufficiently fluid that the mixture, as a whole, may be atomized. Generally, a mixture is sufficiently fluid for atomization when the viscosity of the molten mixture is less than 20,000 cp, or less than 15,000 cp, or less than 10,000 cp, less than 5000 cp, or even less than 1000 cp.
  • the viscosity can be measured by controlled stress rheometer, which measures viscosity as a function of temperature, and may use either a shear-type or rotational rheometer.
  • melting point refers to the temperature that marks the midpoint of the transition from a solid crystalline or semi-crystalline state to a liquid state.
  • melting point is the temperature where upon heating the solid material, the maximum exothermic heat flow occurs.
  • melting point will be used in reference to relative pure single component materials such as some actives or essentially single component excipients (e.g. stearyl alcohol) and flow point will be used in reference to multi-component materials or mixtures.
  • ambient temperature refers to a temperature of 20° C.
  • solid is a solid at ambient temperature, but becomes a liquid at temperatures above 30° C. or 40° C., or at body temperature.
  • capsule means a container suitable for enclosing solids or liquids, and includes empty capsule shells and components thereof such as caps and bodies that may be assembled together to form the capsule.
  • dosage form refers to a solid composition comprising an active ingredient.
  • compositions include abiraterone acetate, or a pharmaceutically acceptable salt form.
  • the compositions may contain one or more active agents.
  • active or “active agent” is meant a drug, medicament, pharmaceutical, therapeutic agent, nutraceutical, or other compound that may be desired to be administered to the body.
  • the active agent is crystalline in the compositions. In other embodiments, the active agent is non-crystalline in the compositions. In still another embodiment, the active agent may comprise crystalline and non-crystalline regions in the compositions. In some embodiments, the active agent is at least 60 wt % crystalline. In other embodiments, the active agent is at least 75 wt % crystalline. In another embodiment, the active agent is at least 90 wt % crystalline. In other embodiments, the active may dissolve into the excipients prior to forming the compositions. During the multiparticulate formation process, a portion of the crystalline active agent may dissolve into the excipient mixture up to the active agent's solubility limit in the excipient mixture at the processing conditions. When the excipient mixture is cooled to form the multiparticulates, the multiparticulate will comprise particles of crystalline active agent encapsulated in a solid solution of excipient and the dissolved active agent.
  • the lipid multiparticulates include fatty alcohols, fatty acids, fatty acid esters of glycols and poly glycols, fatty acid esters of polyglycerol and fatty acid esters of glycerol (glycerides) with flow points of less than 50° C.
  • the melting point is also less than 50° C.
  • a preferred class of low flow point excipients are low flow point glycerides.
  • low flow point excipient such as a glyceride, is meant that the melting point of the excipient, such as a glyceride, is less than 50° C.
  • the low flow point glyceride has a melting point of less than 40° C.
  • the low-flow point excipients are liquids at ambient temperature (typically at temperatures of from about 20° C. to about 25° C., preferably about 25° C.).
  • Exemplary low flow point excipients include glycerol monooleate (such as PeceolTM predominantly mono and dioleate (C 18 esters of glycerol), C 8 to C 10 triglycerides (Miglyol 812®, caprylic, C 8 , and capric acid, C 10 triglycerides), and glycerol monocaprylate (Imwitor 308®, mono and dicaprylate, C 8 esters of glycerol; also some mono-dicaprate acid, C 10 glycerol esters).
  • glycerol monooleate such as PeceolTM predominantly mono and dioleate (C 18 esters of glycerol)
  • C 8 to C 10 triglycerides Miglyol 812®, caprylic, C 8 , and capric acid, C 10 triglycerides
  • glycerol monocaprylate Imwitor 308®, mono and dicaprylate, C 8
  • the low-flow point excipient such as glyceride
  • the low-flow point excipient is a mixture of compounds, having a flow point of 50° C. or less.
  • the low-flow point excipient, such as glyceride has a flow point of 40° C. or less.
  • the low-flow point glyceride has a low flow point of 30° C. or less.
  • Exemplary low-flow point glycerides include polyglycolized glycerides, such as some of the Gelucire products manufactured by Gattefosse, such as Gelucire® 43/01 having a nominal melting point of 43° C. Mixtures of low flow point glycerides are also effective, such as mixtures of Gelucire° C. 43/01 (C 10 -C 18 triglycerides), Gelucire® 50/13 (stearoyl polyoxylglycerides), Gelucire® 44/14 (lauroyl macrogol-32 glycerides), Gelucire® 48/16 (polyoxyl stearate), and mixtures thereof. Other glycerides may also be used, such as fatty acid esters of glycols and poly glycols, and fatty acid esters of polyglycerols.
  • a function of the low flow point excipient is to ensure that at least a significant portion of the formulation matrix softens when ingested orally by a patient in need of therapy, at the temperature of the GI tract (about 37° C. for humans). This allows the formulation to break down by digestion in the gastro-intestinal (GI) tract, and ultimately to disperse in the GI tract to promote dissolution and absorption of the active.
  • the low flow point excipient provides a significant portion of the formulation matrix to be present in a non-crystalline liquid or amorphous state when ingested and softened in the GI tract.
  • Exemplary low flow point fatty alcohols include myristyl alcohol (Tm 38° C.), lauryl alcohol (Tm 23° C.) and capric alcohol (Tm 7° C.).
  • Exemplary high flow point glycerides include glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, and mixtures thereof.
  • the high flow point glyceride is a mixture of compounds that are formulated into a product and sold under a variety of trade names.
  • Exemplary high flow point and high melt point fatty alcohols include stearyl alcohol (Tm 58° C.) and behenyl alcohol (Tm 71° C.).
  • Exemplary high flow point and high melt point fatty acids include palmitic acid (Tm 63° C.) and stearic acid (Tm >70° C.).
  • Exemplary waxes include paraffin wax, beeswax, candelilla wax, carnauba wax, and mixtures thereof.
  • a function of the high flow point excipient is to aid in the manufacturability of the LMPs by enabling the LMP to congeal at a lower temperature to obtain solid particles during a melt-spray-congeal process.
  • the high flow point excipient aids the physical stability of the LMP formulation.
  • the high flow point excipient is not appreciably digested in the GI tract.
  • the LMPs include other excipients to improve the performance and chemical stability of the formulations.
  • surfactants and co-surfactants may be included in the compositions.
  • Exemplary surfactants and co-surfactants include polyethoxylated 12-hydroxysteric acid, also known as PEG15 hydroxystearate (Kolliphor® HS-15), propylene glycol monocaprylate (C 8 ) esters (CaproylTM 90), esterified alpha-tocopheryl polyethylene glycol succinate (TPGS), mono, di, tricaprylic (C 8 ) and capric acid (C 10 ) esters of glycerol and mono and diesters of PEG400 (Labrasol®), Propylene glycol monolaurate (C 12 ) esters (Labrafil® M1944CS), Polyoxyl 40 hydrogenated castor oil (Kolliphor® RH40), lecithins, and mixtures thereof.
  • the LMPs optionally include an antioxidant to maintain chemical stability of the abiraterone acetate.
  • antioxidants include vitamin E, tocopheryl polyethylene glycol succinate (TPGS), rosemary extract, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), and mixtures and combinations thereof.
  • a flow aid is used to improve the flow properties of the LMPs.
  • exemplary flow aids also known as glidants include calcium silicate, cab-o-sil, silicon dioxide, calcium phosphate tribasic, colloidal silicone dioxide, magnesium silicate, magnesium trisilicate, starch, talc, and other flow aids.
  • the lipid excipient is selected from the group comprising fatty alcohols, fatty acids, fatty acid esters of glycols and poly glycols, fatty acid esters of glycerol, polyglycerol, polyglycolized glycerides, C 10 -C 18 triglyceridesstearoyl polyoxylglycerides, lauroyl macrogol-32 glycerides, caprylocaproyl macrogol-8 glycerides, oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, myristyl alcohol, lauryl alcohol, capric alcohol, glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, stearyl alcohol, behenyl alcohol, palmitic acid, stearic acid, paraffin wax, beeswax, candelilla wax, carnauba wax, polyethoxyl
  • C 10 -C 18 triglyderides include but are not limited to glycerol tricaprylate, glycerol tricaprate, glycerol trilaurate, glyceryl triolein, glyceryl tristearate, glyceryl tripalmitate, almond oil, canola oil, cod liver oil, corn oil, cottonseed oil, evening primrose oil, fish oil, grape seed oil, olive oil, palm kernel oil, palm oil, peanut oil, rapeseed oil, safflower oil, sesame oil, soybean oil, sunflower oil, hydrogenated coconut oil, hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenated soybean oil, partially hydrogenated soybean oil and hydrogenated vegetable oil.
  • fatty acid esters of glycerol include but are not limited to glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate, which are sold under various trade names such as PeceolTM, Maisine 35-1, Geleol®, Capmul® and Imwitor.
  • propylene glycol fatty acid esters include but are not limited to propylene glycol monocaprylate, dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, propylene glycol monomyristate, which are sold under various trade names such as Capryol®, Lauroglycol®, Labrafac® and Capmul®.
  • sorbitan fatty acid esters include but are not limited to sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate and sorbitan trioleate, which are sold under various trade names such as Span® and Montane®.
  • polyoxyethylene sorbitan fatty acid esters examples include but are not limited to polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and polysorbate 85, which are sold under various trade names such as Tween® and Montanox®.
  • polyoxylglycerides examples include oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, and lauroyl macrogol-6 glycerides These are sold under different trade names such as Labrasol®, Gelucire® and Labrafil®.
  • Preferred embodiments of the composition for liquid (and/or solid/semi-solid) fill hard capsules (and/or soft capsules) are described in the table below:
  • Range wt % of Component Excipient composition Fatty acid Glycerol monooleate (e.g., Peceol); 10-60% wt esters of glycerol monolinoleate (eg Maisine 35-1) or glycerol glycerol monostearate (eg Geleol) or mixtures of triglyceride with these monoglycerides, for example 1:0.5, 1:1, 1:2, 1:3 or 1:4 triglyceride/monoglyceride ratios
  • These long-chain triglycerides could be corn oil, soybean oil, peanut oil etc
  • Propylene Propylene glycol monocaprylate Capryol TM 20-60% wt glycol fatty 90
  • the fatty acid ester of glyercol is selected from the group consisting of glycerol tricaprylate, glycerol tricaprate, glycerol trilaurate, glyceryl triolein, glyceryl tristearate, glyceryl tripalmitate, almond oil, canola oil, cod liver oil, corn oil, cottonseed oil, evening primrose oil, fish oil, grape seed oil, olive oil, palm kernel oil, palm oil, peanut oil, rapeseed oil, safflower oil, sesame oil, soybean oil, sunflower oil, hydrogenated coconut oil, hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenated soybean oil, partially hydrogenated soybean oil and hydrogenated vegetable oil, glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate.
  • the lipid propylene glycol fatty acid ester is selected from the group consisting of propylene glycol monocaprylate, propylene glycol dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, and propylene glycol monomyristate; and the polyoxylglyceride is selected from the group consisting of oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides and lauroyl macrogol-6 glycerides.
  • the surfactant is selected from the group consisting of Polyoxyl 40 hydrogenated castor oil, polysorbates, polyoxyl 35 castor oil, polyethoxylated 12-hydroxysteric acid, vitamin E, tocopheryl polyethylene glycol succinate (TPGS), caprylocaproyl macrogol-8 glycerides, lauroyl macrogol-32 glycerides, and macrogol stearate, or mixtures of these surfactants.
  • the fatty acid ester of glycerol is selected from the group consisting of glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate.
  • the propylene glycol fatty acid ester is selected from the group consisting of propylene glycol monocaprylate, propylene glycol dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, and propylene glycol monomyristate.
  • the fatty acid ester of glycerol is glycerol monooleate and the propylene glycol fatty acid ester is propylene glycol monocaprylate.
  • compositions of the present invention are liquid at ambient temperature. Such formulations can be filled into hard capsules, or softgel capsules. In another embodiment, the compositions of the invention are semi-solids, and may be filled into hard capsules or softgel capsules.
  • compositions comprise a plurality of particles that are solid at ambient temperature and are generally spherical in shape, having a size ranging from a mean diameter of 40 ⁇ m to 3000 ⁇ m, or even 50 ⁇ m to 1000 ⁇ m, or even 100 ⁇ m to 300 ⁇ m.
  • the LMPs comprising abiraterone acetate and all excipients that make up the matrix have a flow point above 25° C. In some embodiments, the flow point is above 30° C. or 35° C. or 40° C. In some embodiments, the flow point of the LMP composition is less than 50° C. or less than 40° C.
  • spherical By generally spherical is meant that while most particles are essentially spherical, they do not necessarily form “perfect” spheres. Such particle variations in spherical shapes are known to those persons of ordinary skill in the art of melt-spray-congeal processing and similar particulate forming methods. To measure the diameters of the particulates, there are several methods that can be used, including laser diffraction, optical microscopy, and/or SEM.
  • Lipid formulations are advantageous active agent forms because they are amenable for use in scaling dosage forms according to the weight of an individual animal, including humans, in need of treatment by simply scaling the mass of particles in the dosage form to comport with the animal's weight. They are further advantageous since they allow the incorporation of a large quantity of active into a simple dosage form such as a capsule. Lipid formulations also have numerous therapeutic advantages over other dosage forms, especially when taken orally, including (1) improved dispersal in the gastrointestinal (GI) tract, (2) relatively rapid and reproducible passage from the stomach, (3) more uniform GI tract transit time, and (4) reduced inter- and intra-patient variability.
  • GI gastrointestinal
  • the compositions comprise abiraterone acetate, at least one fatty acid ester, at least one surfactant, and optionally an antioxidant.
  • the abiraterone acetate comprises at least 0.2 wt % of the composition
  • the fatty acid ester comprises at least 10 wt % of the composition
  • the surfactant comprises at least 10 wt % of the composition.
  • the abiraterone acetate comprises at least 3 wt % of the composition
  • the fatty acid ester comprises at least 25 wt % of the composition
  • the surfactant comprises at least 40 wt % of the composition.
  • the abiraterone acetate comprises at least 5 wt % of the composition, the fatty acid ester comprises at least 40 wt % of the composition, and surfactant comprises at least 30 wt % of the composition. In one embodiment, the abiraterone acetate comprises at least 10 wt % of the composition, the fatty acid ester comprises at least 30 wt % of the composition, the surfactant comprises at least 50 wt % of the composition. In some embodiments, a low dose of the active may lead to improved bioavailability when dosed orally to a patient in need of therapy.
  • the abiraterone acetate comprises at least 2 wt % of the compositions, at least 3 wt % of the compositions, at least 5 wt % of the compositions, at least 7 wt % of the compositions, or at least 10 wt % of the compositions.
  • the compositions comprise abiraterone acetate, at least one low-flow point excipient, at least one high flow point excipients, at least one low-flow point surfactant, and optionally an antioxidant.
  • the abiraterone acetate comprises at least 0.2 wt % of the composition
  • the low flow-point excipient comprises at least 10 wt % of the composition
  • the high flow-point excipient comprises at least 5 wt % of the composition
  • the low-flow point surfactant comprises at least 10 wt % of the composition.
  • the abiraterone acetate comprises at least 5 wt % of the composition
  • the low flow-point excipient comprises at least 20 wt % of the composition
  • the high flow-point excipient comprises at least 5 wt % of the composition
  • the low-flow point surfactant comprises at least 30 wt % of the composition.
  • the abiraterone acetate comprises at least 2 wt % of the compositions, at least 3 wt % of the compositions, at least 5 wt % of the compositions, at least 7 wt % of the compositions, or at least 10 wt % of the compositions.
  • the LMP formulations also comprise a low flow point excipient.
  • the LMP matrix is comprised of at least 10 wt % to 50 wt % of the low flow point excipient.
  • the LMP formulation is comprised of at least 50 wt % to 75 wt % of the low flow point excipient.
  • the LMP formulations also comprise a high flow point excipient.
  • the compositions comprise at least 2 wt % of the high flow point excipient.
  • the LMP matrix is comprised of 1 wt % to 30 wt % of the high flow point excipient.
  • the LMP matrix is comprised of 2 wt % to 20 wt % of the high flow point excipient.
  • the LMP matrix is comprised of 3 wt % to 15 wt % of the high flow point excipient.
  • the LMP formulations may also comprise an antioxidant.
  • the LMP formulations comprise from 0 wt % to 20 wt %, such as from 0.01 wt % to 20 wt %, of an antioxidant.
  • the LMP formulations comprise from 1 wt % to 15 wt % of an antioxidant.
  • the LMP formulations may also comprise a flow aid.
  • the LMP formulations may comprise from 0 wt % to 5 wt %, such as from 0.01 wt % to 5 wt %, of a flow aid.
  • the LMP formulations may comprise from 0.5 wt % to 2 wt % of a flow aid.
  • the compositions comprise an active agent, a low flow point excipient, and a high flow point excipient, where in the mass ratio of active agent is from 0.2 wt % to 60 wt %, the mass ratio of low flow point excipient from 10 wt % to 50 wt %, and the high flow point excipient ranges from 1 wt % to 30 wt % of the composition.
  • the mass ratio of active agent is from 5 wt % to 20 wt % of the composition
  • the mass ratio of the low flow point excipient is 50 wt % to 75 wt % of the composition
  • the mass ratio of the high flow point excipient ranges from 2 wt % to 20 wt % of the composition.
  • the mass ratio of active agent is from 5 wt % to 15 wt % of the composition
  • the mass ratio of the low flow point excipient is 50 wt % to 75 wt % of the composition
  • the mass ratio of the high flow point excipient ranges from 3 wt % to 15 wt % of the composition.
  • the compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a dispersing agent.
  • the compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a dispersing agent.
  • compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a flow aid.
  • compositions herein comprise, preferably consist of, from 3 wt % to 10 wt %, preferably from 4 wt % to 7 wt % of abiraterone acetate; from 10 wt % to 60 wt %, preferably from 20 wt % to 50 wt %, more preferably from 25 wt % to 35 wt %, more preferably from 27 wt % to 29 wt %, of a fatty acid ester of glycerol (preferably glycerol monooleate, like commercially available PeceolTM) or mixtures thereof; from 20 wt % to 60 wt %, preferably from 30 wt % to 50 wt %, more preferably from 32 wt % to 33 wt %, of at least one of a propylene glycol fatty acid ester (preferably Propylene glycol monocaprylate, like commercially available Capryo
  • the following process is used to prepare the compositions.
  • the active-free formulation is prepared by weighing the appropriate excipients into a vessel, and melting if necessary. The excipients are then mixed thoroughly to generate an isotropic solution. The mass of the abiraterone acetate is then added, and the active-free formulation is mixed at 50° C. until the active is completely dissolved (usually confirmed by polarized light microscopy). The final formulation is then cooled to ambient temperature for storage until filled into a capsule.
  • compositions may be formed by a “melt-spray-congeal” process, as described below. See, for example, U.S. Pat. Nos. 7,235,260, 7,887,844, EP1691787, U.S. Pat. No. 7,625,507.
  • a molten mixture is formed by mixing and heating the compositions.
  • Such compositions are comprised of abiraterone acetate, and a lipid matrix.
  • “Molten mixture” means that the mixture of abiraterone acetate and lipid matrix materials are sufficiently mixed and heated to fluidize the mixture sufficiently to allow it to be atomized into droplets.
  • the mixture is molten in the sense that it will flow when subjected to one or more forces such as pressure, shear, and centrifugal force, such as that exerted by a centrifugal or spinning-disk atomizer.
  • the molten mixture is delivered to an atomizer that breaks the molten mixture into small droplets.
  • Virtually any method can be used to deliver the molten mixture to the atomizer.
  • the molten mixture is delivered to the atomizer by use of pumps and/or various types of pneumatic devices such as pressurized vessels or piston pots or extruder.
  • the molten mixture is maintained at an elevated temperature during delivery to the atomizer to prevent its solidification and to keep it in a flowable state.
  • a centrifugal atomizer also known as rotary atomizers or spinning-disk atomizer
  • the molten mixture is fed onto a rotating surface, where it spreads outward and flows by centrifugal force.
  • the rotating surface may take several forms, examples of which include a flat disk, a cup, a vanned disk, and a slotted disk.
  • the surface of the disk may also be heated to aid in atomization of the molten mixture or cooled to aid in the solidification of the LMPs.
  • the droplets are congealed, typically by contact with a gas at a temperature below the solidification temperature of the composition. Typically, it is desirable that the droplets are congealed in less than 60 seconds, less than 10 seconds, or even in less than 1 second. In certain embodiments congealing at ambient temperature using an ambient temperature cooling medium, results in sufficiently rapid solidification of the droplets.
  • a cooling medium that is at a temperature that is at least 10° C. below ambient temperature. For some embodiments, it is preferred to utilize a cooling medium that is at least 20° C. below ambient temperature.
  • the compositions of the invention are placed into a capsule for delivery by oral ingestion.
  • exemplary capsules include hard gelatin capsules, soft gelatin capsules, HPMC capsules as well as capsules made from other materials.
  • the compositions may also be filled as a liquid into the capsule.
  • the LMPs can also be suspended in a liquid such as an aqueous solution and then ingested.
  • the LMPs may also be sprinkled on food and then ingested.
  • the LMPs may also be mixed with solid excipients and then compressed into tablets.
  • Liquid-filled capsules can be hard or soft in form and may conveniently deliver the active within a liquid formulation.
  • the fill formulation can contain either solubilized or suspended active, dependent on the choice of formulation, active solubility in this formulation and the dose per capsule.
  • active solubility in this formulation and the dose per capsule.
  • Other advantages of liquid-filled capsules over other dosage forms such as tablets and caplets include better patient compliance, greater flexibility in dosage form design, and less expensive manufacturing process.
  • Liquid-filled two piece hard capsules may be produced from a range of materials, with gelatin and hydrophilic polymers often the preferred choice.
  • the capsule shell may also contain a pharmaceutically acceptable plasticizer, coloring agents and lubricants.
  • Hard capsule shells are generally manufactured using dip molding processes.
  • LFHC may be used for a number of different applications including bioavailability enhancement of poorly water-soluble molecules, high-potency molecules, molecules susceptible to oxidation, molecules exhibiting low melting points, and molecules requiring controlled/sustained release formulations.
  • the fill formulation can contain or mixtures of lipidic excipients such as those outlined above.
  • the fill formulation may be liquid at ambient temperature or solid/semi-solid.
  • compositions of the invention may be tested in vivo in animals, or in vitro using appropriate test solutions. Bioavailability of the compositions can be tested in vivo in animals or humans using conventional methods for making such a determination.
  • An in vivo test such as a crossover study, may be used to determine whether a composition provides an enhanced active agent concentration in the blood (serum or plasma) versus time area under the curve (AUC) for a test subject, dosed with the composition relative to the active agent concentration in the blood versus time AUC for a test subject dosed with a control composition. The bioavailability is measured as the area under the curve (AUC) determined for each group.
  • AUC In vivo determinations of AUC can be made by plotting the serum or plasma concentration of active agent along the ordinate (y-axis) against time along the abscissa (x-axis).
  • the values for AUC represent a number of values taken from all of the subjects in a patient test population and are, therefore, mean values averaged over the entire test population.
  • the AUC of the test composition divided by the amount dosed is at least 1.25-fold the AUC of the control composition divided by the amount dosed. In another embodiment, the AUC of the test composition divided by the amount dosed is at least 2-fold the AUC of the control composition divided by the amount dosed. In still another embodiment, the AUC of the test composition divided by the amount dosed is at least 2.5-fold the AUC of the control composition divided by the amount dosed. In still another embodiment, the AUC of the test composition divided by the amount dosed is at least 3-fold the AUC of the control composition divided by the amount dosed.
  • compositions may be tested in vitro using an appropriate test solution.
  • test solutions include aqueous solutions at 37° C. comprising 0.1 N HCl, simulating gastric fluid without enzymes, or intestinal buffer solutions, or water.
  • Other in vitro tests solutions may be used as know by those skilled in the art.
  • the active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 50° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a pale yellow liquid, with evidence of some excipient solids on prolonged storage at room temperature.
  • Example 1 On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 1 formed a homogenous cloudy dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 1 containing an additional 0.1% butylated hydroxytoluene (BHT) antioxidant, was placed on accelerated stability for 1 month.
  • BHT butylated hydroxytoluene
  • the active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear colorless liquid.
  • Example 2 On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 2 formed a homogenous cloudy dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 2 containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.0% total impurities) instabilities were seen.
  • the active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear, colorless liquid.
  • Example 3 On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 3 formed a homogenous milky dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 3 containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.1% total impurities) instabilities were seen.
  • the active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear colorless liquid.
  • Example 4 On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 4 formed a homogenous cloudy to transparent dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 4 containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.1% total impurities) instabilities were seen.
  • Example 1 Example 2
  • Example 3 Solubilized (lipid + aq. phase) active concentration ( ⁇ g/mL) ⁇ 5 min 2411 3018 2755 2937 0 min 2392 3094 2805 2841 5 min 1555 2195 2551 2647 15 min 1642 1756 2360 2547 30 min 1416 1478 2374 2593 45 min 1000 1329 2200 2573 60 min 657 1421 2250 2515 Colloidal distribution of the active @ 60 min: Lipid phase 9% — 68% 84% Aq. phase 30% 57% 22% 17% Pellet phase 59% 34% 5% 2%
  • Example 1 and 2 showed evidence of active precipitation on digestion, as evidenced by the progressive decrease in solubilized concentrations with respect to time and the analysis of the pellet phase at 60 min.
  • Example 3 and 4 were more robust, showing negligible active precipitation on digestion.
  • the majority of the active was solubilized in a lipid phase that phase-separated on centrifugation of the samples and most likely consisting of larger lipophilic colloids including multi-lamellar vesicles.
  • Active drug in the aqueous phase (consisting of BS/PL mixed-micelles, lipid-enriched BS/PL mixed micelles and small vesicles) is considered the most readily available form for absorption.
  • Example 1 Active-free formulations were prepared as described for Example 1.
  • the above examples formed isotropic solutions at 50° C. that were colorless to yellow in color depending on the composition. At lower temperatures, there was some evidence of excipient solidification, e.g., in the case of Examples 8 and 9.
  • Example 7 and 9 containing an additional 0.1% BHT antioxidant were placed on accelerated stability for 1 month.
  • the results are summarized in the table below:
  • Example 9 Under accelerated conditions, Example 9 exhibited greater levels of unknown impurities relative to Example 7. Both Examples were physically stable over 1 month.
  • Example formulations 6-9 were then subjected to in vitro dispersion tests wherein 1 g of formulation was dispersed in 250 mL degassed water at 37° C. over 6 hours. The following results were obtained:
  • Example 6 Example 7
  • Example 8 Example 9 Macroscopic Milky Translucent/ Cloudy Cloudy appearance Cloudy Microscopic Aggregates and No particles were Poorly dispersed Poorly dispersed appearance poorly dispersed evident oil droplets oil droplets oil droplets Evidence of No precipitation No precipitation No precipitation active over 6 h over 6 h over 6 h over 6 h precipitation
  • Example 7 was robust to digestion, with minimal precipitation of the active to the pellet phase. Due to the optimized dispersion properties, Example 7 also provided a high percentage of active solubilized in the aqueous phase—this phase consists of small dispersed colloids including micelles and small vesicles. Solubilized active concentrations in both the aqueous phase (in which the majority (80%) of active was present) and lipid phase were >1500 ⁇ g/mL during the entire digestion test. As a comparison, the crystalline solubility of the active in the same medium was measured at 27 ⁇ g/mL.
  • Example 7 Variations of Example 7 containing different loadings of the active were prepared in the same manner as described for Example 1:
  • Example 11 Abiraterone acetate 3.00 6.00 10.00 Glycerol monooleate 29.10 28.20 27.00 (Peceol TM) Propylene glycol 33.95 32.90 31.50 monocaprylate (Capryol TM 90) Polyoxyl 40 hydrogenated 33.95 32.90 31.50 castor oil (Kolliphor ® RH40)
  • Examples 10-12 formed isotropic clear solutions that were pale yellow in color.
  • Example 11 was placed on stability over 3 months. The following results were obtained:
  • Example 12 Solubilized (lipid + aq. phase) active concentration ( ⁇ g/mL) ⁇ 5 min 884 1592 2505 0 min 846 1691 2477 5 min 719 1613 2038 15 min 712 1244 1984 30 min 720 1197 1972 45 min 712 1226 1962 60 min 646 1164 1925 Colloidal distribution of the active @ 60 min: Lipid phase 12% 10% 18% Aq. phase 75% 80% 76% Pellet phase 13% 10% 6%
  • Examples 10, 11 and 12 provided 30 mg, 60 mg and 100 mg of active, respectively, in the digestion test.
  • the results indicate no significant change in performance with increasing active loading in the formulation as evidenced by negligible change in % active recovered in the pellet phase and high solubilized active concentrations in the aqueous and lipid phase.
  • the commercial abiraterone acetate tablet (Zytiga® 250 mg) and Example 10 were both dosed.
  • the target dose delivered by Example 10 was 25 mg, i.e., 1/10 th of the Zytiga® dose, which was achieved using approximately 0.83 mL formulation filled in hard capsules (Licaps®, size 00el) prior to administration.
  • the following results were obtained (note that results are expressed as measured plasma abiraterone due to complete conversion from abiraterone acetate in vivo):
  • Example 10 The mean total absorption of the active from the commercial tablet was 413 h ⁇ ng/mL.
  • Example 10 provided approximately 50% of this exposure (227 h ⁇ ng/mL) despite dosing the dogs only 1/10 th of the Zytiga® dose. This result therefore highlights the low absorption of the active from the fasted state using Zytiga® which contains crystalline drug and the fact that a higher proportion of the administered dose can be absorbed using lipid formulations that are effective in maintaining substantially higher solubilized abiraterone acetate concentrations (such as Example 10).
  • Example 10 provided a marked reduction in inter-dog absorption variability (captured here by CV, the coefficient of variation) and variability in absorption rate (captured here by the T max ) in comparison to Zytiga®.
  • CV inter-dog absorption variability
  • T max variability in absorption rate
  • Example 11 was assessed on in vitro digestion under more dilute experimental conditions:
  • Example 11 0.33 g of Example 11 0.83 g of Example 11 Digestion time tested in 40 mL tested in 40 mL point digestion medium digestion medium Solubilized (lipid + aq. phase) active concentration ( ⁇ g/mL) ⁇ 5 min 553 1409 0 min 559 1363 5 min 448 1127 15 min 423 1128 30 min 382 1102 45 min 379 1111 60 min 352 1078 Colloidal distribution of the active @ 60 min: Lipid phase 6% 4% Aq. phase 70% 85% Pellet phase 24% 11%
  • Example 11 tested at 0.33 g and 0.83 g equated to ⁇ 20 mg and ⁇ 50 mg active doses.
  • the testing of lower formulation masses in the same medium and same medium volume generates higher bile salt/phospholipid-to-lipid formulation ratios. This is known by those skilled in the art to in some cases to “stress” lipid formulations and trigger increased precipitation of the active.
  • formulations such as Example 11 can be obtained that show consistent solubilization properties irrespective of the extent of formulation dilution. Indeed, as shown above, Example 11 maintained at least 75% of the active in the solubilized state during digestion under more stressful (i.e., more dilute) conditions.
  • Such consistent solubilization properties are desirable to the development of robust formulations since differences in dilution may be reasonably encountered in vivo through e.g., inter-subject variability in GI fluid volumes or in instances where patients are dosed different number of formulation capsules.
  • WO2014009434A1 discloses several castor oil containing lipid formulations for the active abiraterone acetate.
  • SMES 1 consists of 20% wt castor oil, 20% wt Capmul® MCM, 30% wt Span 80 and 30% Kolliphor® EL.
  • This placebo formulation was also prepared here followed by the complete incorporation of the active at a 60 mg/g loading to enable direct in vitro performance comparison to Example 11. Indeed, SMES 1 was subjected to in vitro dispersion and in vitro digestion tests as described above.
  • SMES 1 On dispersion, SMES 1 formed a milky dispersion indicating the presence of larger oil droplets in comparison to Example 11 which forms a more translucent dispersion. SMES 1 did however protect against precipitation of the active over 6 hours of dispersion.
  • the active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a water-jacketed tank, with prior excipient melting at 75° C. Excipients were then mixed thoroughly to generate a single phase solution.
  • the active, (abiraterone acetate) was added and mixed with the molten mixture. Then the molten mixture was allowed to cool to 60° C. The molten mixture was then injected onto a melt-spray congeal apparatus equipped with a spinning disk atomizer spinning at 2050 rpm, maintained at 60° C. and at a s pray rate of 120 g/min. Droplets of the molten mixture were formed and congealed at ⁇ 5° C. The resulting LMP particles had a generally spherical shape, and a mean particle size of 270 ⁇ m.
  • thermodynamic solubility of the active was evaluated in digested placebo formulations for determining solubility in digested lipidic colloidal phases.
  • Example 5 Active concentration ( ⁇ g/mL) Digestion Test 2 time Test 1 (undigested) ⁇ 5 548 427 0 623 435 5 757 519 15 754 665 30 593 502 45 401 640 60 381 513 AA solubility in crystalline 27 digestion media AA solubility in digested 121 LMP formulations
  • Example 5 showed evidence of active precipitation on digestion, as evidenced by the decrease in solubilized concentrations once the formulation started to disperse around 15 min.
  • Test 2 it is seen that digestion does not greatly impact compound solubilization which is positive. Digestion itself may have promoted “release” of the dissolved/dissolving drug though this might have been masked if released drug rapidly recrystallized.
  • Equilibrium solubility of crystalline active in the digested LMP's was measured at 121 ⁇ g/mL, which shows overall improvement in solubilization provided by the digested BS/PL-lipid mixed micellar system; active drug in the aqueous phase containing BS/PL-Lipid-enriched mixed micelles is considered the available form for passing drug through the unstirred water layer to the enterocyte for absorption.
  • the antioxidant, BHT was added at a 0.1% wt concentration to Example 5, which were then placed on accelerated chemical stability at 25° C./60% RH and 40° C./75% RH for 1 month.
  • the results are summarized in the table below.
  • Example 12 Abiraterone acetate 4.00 20.00 C 10 -C 18 Triglycerides 30.32 24.90 (Gelucire ® 43/01) Stearoyl macrogol-32 glycerides 60.64 49.90 (Gelucire ® 50/13) Glyceryl behenate (Compritol ® 888) 5.00 5.00 Butylated hydroxytoluene (BHT) 0.04 0.20
  • Example 12 was subjected to in vitro digestion tests are described for previous formulations. The following results were obtained:
  • Digestion Example 12 time Active concentration ( ⁇ g/mL) ⁇ 5 811 0 649 5 694 15 658 30 592 45 564 60 811
  • Example 12 generally provided higher solubilized active concentrations in comparison to Example 5, despite the use of a lower active loading (4% wt vs. 10% wt) and due to the formulation containing a higher fraction or pre-dissolved active in the formulation and the higher formulation:active ratio.
  • Example 12 was placed on accelerated chemical stability at 25° C./60% RH and 40° C./75% RH for 1 month. The results are summarized in the table below.
  • Dogs were randomized over 6 weeks to receive either the commercial abiraterone acetate tablet (Zytiga® 250 mg), Example 11 and Example 5, in a fasted and fed state.
  • the target dose in the lipid formulation (Example 11) and LMP formulation (Example 5) was 50 mg, namely 1 ⁇ 5 th of the Zytiga® dose. This dose was delivered using approximately 0.83 mL of Example 11, while for Example 5, using approximately 500 mg formulation. Both were filled in hard gelatin capsules (Licaps®, size 00el) prior to administration.
  • Example 11 achieved a fasted-state exposure of 852 h ⁇ ng/mL, which was slightly higher than Zytiga® under the same conditions despite dosing only 1 ⁇ 5 th of the dose.
  • Example 11 also substantially reduced fasted-state variability in absorption (reduced from 111% to 28%) while absorption from Example 11 was near independent of food—only a small decrease in the extent of absorption in the presence of food (762 h ⁇ ng/mL) was observed.
  • Example 5 did not provide the same level of absorption when compared to Example 11 at a 50 mg dose (AUC 0-24 h values were 351 and 562 h ⁇ ng/mL in the fasted and fed state, respectively, compared to 852 and 762 h ⁇ ng/mL for Example 11), but Example 5 did achieve a total exposure that was ⁇ 50% relative to Zytiga®, while also producing a substantial reduction in variability (36%). In terms of the food-effect, Example 5 exhibited a modest ⁇ 1.2-fold increase in exposure in the fed state.
  • Example 5 and Example 11 are able to support higher amounts of absorption in the fasted state. This in turn affords the possibility of lower doses, reduced variability and eliminated food-effect as illustrated here by lipid formulation Examples 5 and 11.

Abstract

Abiraterone acetate lipid formulations.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This is a continuation of U.S. application Ser. No. 15/565,400, filed on Oct. 9, 2017, which is the U.S. National Stage of International Application No. PCT/EP2016/056689, filed Mar. 25, 2016, which was published in English under PCT Article 21(2), which in turn claims the benefit of U.S. Provisional Application No. 62/145,864, filed Apr. 10, 2015; each of these prior applications is incorporated herein by reference in its entirety.
  • FIELD
  • Disclosed are lipid compositions comprising abiraterone acetate.
  • BACKGROUND
  • Lipid multiparticulates (LMPs) are known. See for example EP1030687, U.S. Pat. No. 6,572,892, EP1827382, U.S. Pat. Nos. 7,235,260, 7,887,844, EP1691787, U.S. Pat. No. 7,625,507.
  • Abiraterone acetate (Zytiga® tablets) was developed by Janssen, and approved in 2011 for use in combination with prednisone for the treatment of patients with metastatic castration-resistant prostate cancer. Abiraterone acetate has the following structure:
  • Figure US20200316093A1-20201008-C00001
  • Abiraterone acetate is a prodrug of abiraterone, which inhibits 17 α-hydroxylase/C17,20-lyase (CYP17) expressed in testicular, adrenal, and prostatic tumor tissues. Abiraterone acetate is highly lipophilic (Log P 5.12) and as a result suffers from low aqueous solubility in the gastrointestinal tract. Zytiga® tablets containing abiraterone acetate show a clinically significant positive food effect when administered with low-fat (7- and 5-fold increase in Cmax and AUC0-∞, respectively) or high-fat (17- and 10-fold increase in Cmax and AUC0-∞, respectively). As a result, patients taking Zytiga® need to avoid food for at least 2 hours before administration and at least one hour after. There is a benefit in developing a new abiraterone acetate formulation that improves bioavailability in the fasted state to in turn reduce the food effect and overall variability in absorption.
  • SUMMARY
  • Disclosed are compositions comprising abiraterone acetate and a lipid matrix. In one embodiment the lipid matrix comprises a) at least one fatty acid ester, b) at least one surfactant, and c) optionally an antioxidant. In one embodiment the abiraterone acetate comprises at least 0.2 wt % of the composition, at least one fatty acid ester comprises at least 10 wt % of the composition, at least one surfactant comprises at least 10 wt % of the composition. In one embodiment, the compositions are liquid at ambient temperature. In one embodiment, the compositions are semi-solid at ambient temperature. In one embodiment, abiraterone is molecularly dispersed in the lipid matrix.
  • In one embodiment, the lipid matrix comprises a) at least one low flow point excipient, b) at least one high flow point excipient, c) at least one low-flow point surfactant, and c) optionally an antioxidant. In one embodiment, the abiraterone acetate comprises at least 0.2 wt % of the composition, the at least one low flow point excipient comprises at least 10 wt % of the composition, the at least one high flow point excipient comprises at least 5 wt % of the composition, and the at least one low-flow point surfactant comprises at least 10 wt % of the composition. In one embodiment, the compositions comprise a plurality of particles that are solid at ambient temperature, have a generally spherical shape, and have a mean diameter ranging from 40 μm to 3000 μm.
  • In another embodiment, the lipid matrix is a liquid or semi-solid at ambient temperature.
  • In another embodiment, the compositions of the invention are filled into capsules.
  • In one embodiment, the compositions are administered orally to a patient in need of therapy.
  • In one embodiment, the lipid matrix is selected from the group consisting of fatty alcohols, fatty acids, fatty acid esters of glycerol, glycols and poly glycols, fatty acid esters of polyglycerol, polyglycolized glycerides, C8-C18 triglycerides, stearoyl polyoxylglycerides, lauroyl macrogol-32 glycerides, caprylocaproyl macrogol-8 glycerides, oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, myristyl alcohol, lauryl alcohol, capric alcohol, glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, stearyl alcohol, behenyl alcohol, palmitic acid, stearic acid, paraffin wax, beeswax, candelilla wax, carnauba wax, polyethoxylated 12-hydroxysteric acid, propylene glycol monocaprylate esters, propylene glycol dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, propylene glycol monomyristate, esterified alpha-tocopheryl polyethylene glycol succinate, propylene glycol monolaurate esters, polyoxyl 35 castor oil, polyoxyl 40 hydrogenated castor oil, lecithins, vitamin E, tocopheryl polyethylene glycol succinate (TPGS), sugar fatty acid esters, sorbitan fatty acid esters, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene-polyoxypropylene copolymers, propylene glycol, triacetin, isorpropyl myristate, diethylene glycol monoethyl ether, polyethylene glycol, glycerol, rosemary extract, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), and mixtures and combinations thereof.
  • DETAILED DESCRIPTION
  • The present disclosure relates to lipid compositions for abiraterone acetate, such as lipid multiparticulate formulations comprising abiraterone acetate and a lipid matrix or liquid fill hard capsules comprising abiraterone acetate and a lipid matrix.
  • The following description is exemplary in nature and is not intended to limit the scope, applicability, or configuration of the invention in any way. Various changes to the described embodiments may be made in the function and arrangement of the elements described herein without departing from the scope of the invention.
  • As used in this application and in the claims, the singular forms “a,” “an,” and “the” include the plural forms unless the context clearly dictates otherwise. Additionally, the term “includes” means “comprises.” Further, the term “coupled” generally means electrically, electromagnetically, and/or physically (e.g., mechanically or chemically) coupled or linked and does not exclude the presence of intermediate elements between the coupled or associated items absent specific contrary language.
  • Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, percentages, and so forth, as used in the specification or claims are to be understood as being modified by the term “about.” Accordingly, unless otherwise indicated, implicitly or explicitly, the numerical parameters set forth are approximations that may depend on the desired properties sought and/or limits of detection under standard test conditions/methods. When directly and explicitly distinguishing embodiments from discussed prior art, the embodiment numbers are not approximates unless the word “about” is recited.
  • Definitions
  • As used in the present disclosure, the following words, phrases, and symbols are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise.
  • As used herein, “optional” or “optionally” means that the subsequently described material, event or circumstance may or may not be present or occur, and that the description includes instances where the material, event or circumstance is present or occurs and instances in which it does not.
  • As used herein, “w/w %” and “wt %” means by weight as a percentage of the total weight or relative to another component in the composition.
  • The term “about” is intended to mean approximately, in the region of, roughly, or around. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. Unless otherwise indicated, it should be understood that the numerical parameters set forth in the following specification and attached claims are approximations. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, numerical parameters should be read in light of the number of reported significant digits and the application of ordinary rounding techniques.
  • The term excipient is meant any pharmaceutically acceptable ingredient that is included in the formulation that is not the active agent. The term glyceride is meant a fatty acid ester of glycerol. By “fatty acid ester of glycerol” is meant mono, di, and triesters of glycerides. Glycerides also include glycerol esters that have both fatty acid and poly alkyl oxide esters.
  • As used herein, the term “flow point” is the temperature at which any portion of the mixture becomes sufficiently fluid that the mixture, as a whole, may be atomized. Generally, a mixture is sufficiently fluid for atomization when the viscosity of the molten mixture is less than 20,000 cp, or less than 15,000 cp, or less than 10,000 cp, less than 5000 cp, or even less than 1000 cp. The viscosity can be measured by controlled stress rheometer, which measures viscosity as a function of temperature, and may use either a shear-type or rotational rheometer. As used herein, melting point refers to the temperature that marks the midpoint of the transition from a solid crystalline or semi-crystalline state to a liquid state. As measured by DSC, the melting point is the temperature where upon heating the solid material, the maximum exothermic heat flow occurs. In general, melting point will be used in reference to relative pure single component materials such as some actives or essentially single component excipients (e.g. stearyl alcohol) and flow point will be used in reference to multi-component materials or mixtures.
  • The term “ambient temperature” refers to a temperature of 20° C.
  • As used herein, the term “semi-solid” is a solid at ambient temperature, but becomes a liquid at temperatures above 30° C. or 40° C., or at body temperature.
  • Unless otherwise indicated, “capsule” means a container suitable for enclosing solids or liquids, and includes empty capsule shells and components thereof such as caps and bodies that may be assembled together to form the capsule.
  • Unless otherwise indicated, “dosage form” refers to a solid composition comprising an active ingredient.
  • Active Agents
  • Embodiments of the disclosed compositions include abiraterone acetate, or a pharmaceutically acceptable salt form. The compositions may contain one or more active agents. As used herein, by “active” or “active agent” is meant a drug, medicament, pharmaceutical, therapeutic agent, nutraceutical, or other compound that may be desired to be administered to the body.
  • In some embodiments, the active agent is crystalline in the compositions. In other embodiments, the active agent is non-crystalline in the compositions. In still another embodiment, the active agent may comprise crystalline and non-crystalline regions in the compositions. In some embodiments, the active agent is at least 60 wt % crystalline. In other embodiments, the active agent is at least 75 wt % crystalline. In another embodiment, the active agent is at least 90 wt % crystalline. In other embodiments, the active may dissolve into the excipients prior to forming the compositions. During the multiparticulate formation process, a portion of the crystalline active agent may dissolve into the excipient mixture up to the active agent's solubility limit in the excipient mixture at the processing conditions. When the excipient mixture is cooled to form the multiparticulates, the multiparticulate will comprise particles of crystalline active agent encapsulated in a solid solution of excipient and the dissolved active agent.
  • Lipid Excipients
  • The lipid multiparticulates include fatty alcohols, fatty acids, fatty acid esters of glycols and poly glycols, fatty acid esters of polyglycerol and fatty acid esters of glycerol (glycerides) with flow points of less than 50° C. When the low flow point excipient is a relatively pure material, the melting point is also less than 50° C. A preferred class of low flow point excipients are low flow point glycerides. By “low flow point” excipient, such as a glyceride, is meant that the melting point of the excipient, such as a glyceride, is less than 50° C. In some embodiments, the low flow point glyceride has a melting point of less than 40° C. In some embodiments, the low-flow point excipients are liquids at ambient temperature (typically at temperatures of from about 20° C. to about 25° C., preferably about 25° C.). Exemplary low flow point excipients include glycerol monooleate (such as Peceol™ predominantly mono and dioleate (C18 esters of glycerol), C8 to C10 triglycerides (Miglyol 812®, caprylic, C8, and capric acid, C10 triglycerides), and glycerol monocaprylate (Imwitor 308®, mono and dicaprylate, C8 esters of glycerol; also some mono-dicaprate acid, C10 glycerol esters).
  • In some embodiments, the low-flow point excipient, such as glyceride, is a mixture of compounds, having a flow point of 50° C. or less. In some embodiments, the low-flow point excipient, such as glyceride, has a flow point of 40° C. or less. In some embodiments, the low-flow point glyceride has a low flow point of 30° C. or less.
  • Exemplary low-flow point glycerides include polyglycolized glycerides, such as some of the Gelucire products manufactured by Gattefosse, such as Gelucire® 43/01 having a nominal melting point of 43° C. Mixtures of low flow point glycerides are also effective, such as mixtures of Gelucire° C. 43/01 (C10-C18 triglycerides), Gelucire® 50/13 (stearoyl polyoxylglycerides), Gelucire® 44/14 (lauroyl macrogol-32 glycerides), Gelucire® 48/16 (polyoxyl stearate), and mixtures thereof. Other glycerides may also be used, such as fatty acid esters of glycols and poly glycols, and fatty acid esters of polyglycerols.
  • A function of the low flow point excipient is to ensure that at least a significant portion of the formulation matrix softens when ingested orally by a patient in need of therapy, at the temperature of the GI tract (about 37° C. for humans). This allows the formulation to break down by digestion in the gastro-intestinal (GI) tract, and ultimately to disperse in the GI tract to promote dissolution and absorption of the active. In certain embodiments the low flow point excipient provides a significant portion of the formulation matrix to be present in a non-crystalline liquid or amorphous state when ingested and softened in the GI tract.
  • Exemplary low flow point fatty alcohols include myristyl alcohol (Tm 38° C.), lauryl alcohol (Tm 23° C.) and capric alcohol (Tm 7° C.).
  • High Flow Point Excipients
  • Exemplary high flow point glycerides include glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, and mixtures thereof. Often, the high flow point glyceride is a mixture of compounds that are formulated into a product and sold under a variety of trade names.
  • Exemplary high flow point and high melt point fatty alcohols include stearyl alcohol (Tm 58° C.) and behenyl alcohol (Tm 71° C.).
  • Exemplary high flow point and high melt point fatty acids include palmitic acid (Tm 63° C.) and stearic acid (Tm >70° C.).
  • Exemplary waxes include paraffin wax, beeswax, candelilla wax, carnauba wax, and mixtures thereof.
  • A function of the high flow point excipient is to aid in the manufacturability of the LMPs by enabling the LMP to congeal at a lower temperature to obtain solid particles during a melt-spray-congeal process. In certain embodiments the high flow point excipient aids the physical stability of the LMP formulation. In most embodiments, the high flow point excipient is not appreciably digested in the GI tract.
  • Other Excipients Including Surfactants, Co-Surfactants, and Antioxidants
  • In some embodiments, the LMPs include other excipients to improve the performance and chemical stability of the formulations. In some embodiments, surfactants and co-surfactants may be included in the compositions. Exemplary surfactants and co-surfactants include polyethoxylated 12-hydroxysteric acid, also known as PEG15 hydroxystearate (Kolliphor® HS-15), propylene glycol monocaprylate (C8) esters (Caproyl™ 90), esterified alpha-tocopheryl polyethylene glycol succinate (TPGS), mono, di, tricaprylic (C8) and capric acid (C10) esters of glycerol and mono and diesters of PEG400 (Labrasol®), Propylene glycol monolaurate (C12) esters (Labrafil® M1944CS), Polyoxyl 40 hydrogenated castor oil (Kolliphor® RH40), lecithins, and mixtures thereof.
  • In one embodiment, the LMPs optionally include an antioxidant to maintain chemical stability of the abiraterone acetate. Exemplary antioxidants include vitamin E, tocopheryl polyethylene glycol succinate (TPGS), rosemary extract, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), and mixtures and combinations thereof.
  • In some embodiments, a flow aid is used to improve the flow properties of the LMPs. Exemplary flow aids also known as glidants include calcium silicate, cab-o-sil, silicon dioxide, calcium phosphate tribasic, colloidal silicone dioxide, magnesium silicate, magnesium trisilicate, starch, talc, and other flow aids.
  • Lipid Excipients
  • In one embodiment, the lipid excipient is selected from the group comprising fatty alcohols, fatty acids, fatty acid esters of glycols and poly glycols, fatty acid esters of glycerol, polyglycerol, polyglycolized glycerides, C10-C18 triglyceridesstearoyl polyoxylglycerides, lauroyl macrogol-32 glycerides, caprylocaproyl macrogol-8 glycerides, oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, myristyl alcohol, lauryl alcohol, capric alcohol, glycerol behenate, glycerol dibehenate, glycerol palmitate, hydrogenated castor oil, stearyl alcohol, behenyl alcohol, palmitic acid, stearic acid, paraffin wax, beeswax, candelilla wax, carnauba wax, polyethoxylated 12-hydroxysteric acid, propylene glycol fatty acid esters, esterified alpha-tocopheryl polyethylene glycol succinate, mono, propylene glycol monolaurate (C12) esters, polyoxyl 35 castor oil, polyoxyl 40 hydrogenated castor oil, lecithins, vitamin E, tocopheryl polyethylene glycol succinate (TPGS), sugar fatty acid esters, sorbitan fatty acid esters, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene-polyoxypropylene copolymers, rosemary extract, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), propylene glycol, triacetin, isorpropyl myristate, diethylene glycol monoethyl ether, polyethylene glycol, glycerol, and mixtures and combinations thereof. Such lipid excipients may be suitable for liquid fill hard capsule compositions.
  • Examples of C10-C18 triglyderides include but are not limited to glycerol tricaprylate, glycerol tricaprate, glycerol trilaurate, glyceryl triolein, glyceryl tristearate, glyceryl tripalmitate, almond oil, canola oil, cod liver oil, corn oil, cottonseed oil, evening primrose oil, fish oil, grape seed oil, olive oil, palm kernel oil, palm oil, peanut oil, rapeseed oil, safflower oil, sesame oil, soybean oil, sunflower oil, hydrogenated coconut oil, hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenated soybean oil, partially hydrogenated soybean oil and hydrogenated vegetable oil.
  • Examples of fatty acid esters of glycerol include but are not limited to glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate, which are sold under various trade names such as Peceol™, Maisine 35-1, Geleol®, Capmul® and Imwitor.
  • Examples of propylene glycol fatty acid esters include but are not limited to propylene glycol monocaprylate, dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, propylene glycol monomyristate, which are sold under various trade names such as Capryol®, Lauroglycol®, Labrafac® and Capmul®.
  • Examples of sorbitan fatty acid esters include but are not limited to sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate and sorbitan trioleate, which are sold under various trade names such as Span® and Montane®.
  • Examples of polyoxyethylene sorbitan fatty acid esters include but are not limited to polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and polysorbate 85, which are sold under various trade names such as Tween® and Montanox®.
  • Examples of the polyoxylglycerides are already listed, for example, oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides, and lauroyl macrogol-6 glycerides These are sold under different trade names such as Labrasol®, Gelucire® and Labrafil®. Preferred embodiments of the composition for liquid (and/or solid/semi-solid) fill hard capsules (and/or soft capsules) are described in the table below:
  • Range wt %
    of
    Component Excipient composition
    Fatty acid Glycerol monooleate (e.g., Peceol); 10-60% wt
    esters of glycerol monolinoleate (eg Maisine 35-1) or
    glycerol glycerol monostearate (eg Geleol) or
    mixtures of triglyceride with these
    monoglycerides, for example 1:0.5, 1:1,
    1:2, 1:3 or 1:4 triglyceride/monoglyceride
    ratios
    These long-chain triglycerides could be
    corn oil, soybean oil, peanut oil etc
    Propylene Propylene glycol monocaprylate (Capryol ™ 20-60% wt
    glycol fatty 90); propylene glycol dicaprate/dicaprylate
    acid esters or esters, propylene glycol heptanoate,
    polyoxyl- propylene glycol monolaurate, propylene
    glycerides glycol monostearate, propoylene glycol
    monooleate, propylene glycol
    monopalmitate, propylene glycol
    monomyristate (Capryol ®, Lauroglycol ®,
    Labrafac ® and Capmul), also oleoyl
    macrogol-6 glycerides (Labrafil ®
    M1944CS), linoleoyl macrogol-6 glycerides
    (Labrafil ® M2125CS) and lauroyl
    macrogol-6 glycerides (Labrafil ®
    M2130CS)
    Surfactant Polyoxyl 40 hydrogenated castor oil 15-60% wt
    (Kolliphor ® RH40): polysorbates
    (polysorbate 20, polysorbate 40,
    polysorbate 60, polysorbate 80 and
    polysorbate 85), polyoxyl 35 castor oil
    (Kolliphor EL), polyethoxylated 12-
    hydroxysteric acid (Solutol HS-15), vitamin
    E, tocopheryl polyethylene glycol succinate
    (TPGS), caprylocaproyl macrogol-8
    glycerides (Labrasol), lauroyl macrogol-32
    glycerides (Gelucire 44/14) and macrogol
    stearate (Gelucire 48/16), or mixtures of
    these surfactants
  • In one embodiment the lipid matrix comprises:
      • (a) a fatty acid ester of glycerol or mixtures thereof in an amount of from 10 to 60 wt % of said composition;
      • (b) at least one of a propylene glycol fatty acid ester and/or a polyoxylglyceride in an amount of from 20 to 60 wt % of said composition;
      • (c) a surfactant present in an amount of from 15 to 60 wt %.
  • In one embodiment the fatty acid ester of glyercol is selected from the group consisting of glycerol tricaprylate, glycerol tricaprate, glycerol trilaurate, glyceryl triolein, glyceryl tristearate, glyceryl tripalmitate, almond oil, canola oil, cod liver oil, corn oil, cottonseed oil, evening primrose oil, fish oil, grape seed oil, olive oil, palm kernel oil, palm oil, peanut oil, rapeseed oil, safflower oil, sesame oil, soybean oil, sunflower oil, hydrogenated coconut oil, hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenated soybean oil, partially hydrogenated soybean oil and hydrogenated vegetable oil, glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate.
  • In one embodiment the lipid propylene glycol fatty acid ester is selected from the group consisting of propylene glycol monocaprylate, propylene glycol dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, and propylene glycol monomyristate; and the polyoxylglyceride is selected from the group consisting of oleoyl macrogol-6 glycerides, linoleoyl macrogol-6 glycerides and lauroyl macrogol-6 glycerides.
  • In one embodiment the surfactant is selected from the group consisting of Polyoxyl 40 hydrogenated castor oil, polysorbates, polyoxyl 35 castor oil, polyethoxylated 12-hydroxysteric acid, vitamin E, tocopheryl polyethylene glycol succinate (TPGS), caprylocaproyl macrogol-8 glycerides, lauroyl macrogol-32 glycerides, and macrogol stearate, or mixtures of these surfactants.
  • In one embodiment the composition the lipid matrix comprises
      • (a) a fatty acid ester of glycerol present in an amount of from 20 to 40 wt % of the composition;
      • (b) at least one of a propylene glycol fatty acid ester and a polyoxylglyceride present in an amount of from 20 to 40 wt % of the composition; and
      • (c) the surfactant is present in an amount of from 20 to 40 wt %.
  • In one embodiment the fatty acid ester of glycerol is selected from the group consisting of glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate and glycerol monocaprate.
  • In one embodiment the propylene glycol fatty acid ester is selected from the group consisting of propylene glycol monocaprylate, propylene glycol dicaprate/dicaprylate esters, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propoylene glycol monooleate, propylene glycol monopalmitate, and propylene glycol monomyristate.
  • In one embodiment the fatty acid ester of glycerol is glycerol monooleate and the propylene glycol fatty acid ester is propylene glycol monocaprylate.
  • Physical Properties of the Lipid Matrix
  • In one embodiment, the compositions of the present invention are liquid at ambient temperature. Such formulations can be filled into hard capsules, or softgel capsules. In another embodiment, the compositions of the invention are semi-solids, and may be filled into hard capsules or softgel capsules.
  • In another embodiment, compositions comprise a plurality of particles that are solid at ambient temperature and are generally spherical in shape, having a size ranging from a mean diameter of 40 μm to 3000 μm, or even 50 μm to 1000 μm, or even 100 μm to 300 μm. In some embodiments, the LMPs comprising abiraterone acetate and all excipients that make up the matrix have a flow point above 25° C. In some embodiments, the flow point is above 30° C. or 35° C. or 40° C. In some embodiments, the flow point of the LMP composition is less than 50° C. or less than 40° C. By generally spherical is meant that while most particles are essentially spherical, they do not necessarily form “perfect” spheres. Such particle variations in spherical shapes are known to those persons of ordinary skill in the art of melt-spray-congeal processing and similar particulate forming methods. To measure the diameters of the particulates, there are several methods that can be used, including laser diffraction, optical microscopy, and/or SEM.
  • Lipid formulations are advantageous active agent forms because they are amenable for use in scaling dosage forms according to the weight of an individual animal, including humans, in need of treatment by simply scaling the mass of particles in the dosage form to comport with the animal's weight. They are further advantageous since they allow the incorporation of a large quantity of active into a simple dosage form such as a capsule. Lipid formulations also have numerous therapeutic advantages over other dosage forms, especially when taken orally, including (1) improved dispersal in the gastrointestinal (GI) tract, (2) relatively rapid and reproducible passage from the stomach, (3) more uniform GI tract transit time, and (4) reduced inter- and intra-patient variability.
  • Compositions
  • In some embodiments, the compositions comprise abiraterone acetate, at least one fatty acid ester, at least one surfactant, and optionally an antioxidant. In one embodiment, the abiraterone acetate comprises at least 0.2 wt % of the composition, the fatty acid ester comprises at least 10 wt % of the composition, the surfactant comprises at least 10 wt % of the composition. In still another embodiment, the abiraterone acetate comprises at least 3 wt % of the composition, the fatty acid ester comprises at least 25 wt % of the composition, the surfactant comprises at least 40 wt % of the composition. In yet another embodiment, the abiraterone acetate comprises at least 5 wt % of the composition, the fatty acid ester comprises at least 40 wt % of the composition, and surfactant comprises at least 30 wt % of the composition. In one embodiment, the abiraterone acetate comprises at least 10 wt % of the composition, the fatty acid ester comprises at least 30 wt % of the composition, the surfactant comprises at least 50 wt % of the composition. In some embodiments, a low dose of the active may lead to improved bioavailability when dosed orally to a patient in need of therapy.
  • In some embodiments, the abiraterone acetate comprises at least 2 wt % of the compositions, at least 3 wt % of the compositions, at least 5 wt % of the compositions, at least 7 wt % of the compositions, or at least 10 wt % of the compositions.
  • In another embodiment, the compositions comprise abiraterone acetate, at least one low-flow point excipient, at least one high flow point excipients, at least one low-flow point surfactant, and optionally an antioxidant. In one embodiment, the abiraterone acetate comprises at least 0.2 wt % of the composition, the low flow-point excipient comprises at least 10 wt % of the composition, the high flow-point excipient comprises at least 5 wt % of the composition, and the low-flow point surfactant comprises at least 10 wt % of the composition. In still another embodiment, the abiraterone acetate comprises at least 5 wt % of the composition, the low flow-point excipient comprises at least 20 wt % of the composition, the high flow-point excipient comprises at least 5 wt % of the composition, and the low-flow point surfactant comprises at least 30 wt % of the composition.
  • In some embodiments, the abiraterone acetate comprises at least 2 wt % of the compositions, at least 3 wt % of the compositions, at least 5 wt % of the compositions, at least 7 wt % of the compositions, or at least 10 wt % of the compositions.
  • The LMP formulations also comprise a low flow point excipient. In one embodiment, the LMP matrix is comprised of at least 10 wt % to 50 wt % of the low flow point excipient. In another embodiment, the LMP formulation is comprised of at least 50 wt % to 75 wt % of the low flow point excipient.
  • The LMP formulations also comprise a high flow point excipient. In one embodiment, the compositions comprise at least 2 wt % of the high flow point excipient. In another embodiment, the LMP matrix is comprised of 1 wt % to 30 wt % of the high flow point excipient. In still another embodiment, the LMP matrix is comprised of 2 wt % to 20 wt % of the high flow point excipient. In still another embodiment, the LMP matrix is comprised of 3 wt % to 15 wt % of the high flow point excipient.
  • The LMP formulations may also comprise an antioxidant. In one embodiment, the LMP formulations comprise from 0 wt % to 20 wt %, such as from 0.01 wt % to 20 wt %, of an antioxidant. In one embodiment, the LMP formulations comprise from 1 wt % to 15 wt % of an antioxidant.
  • The LMP formulations may also comprise a flow aid. In one embodiment, the LMP formulations may comprise from 0 wt % to 5 wt %, such as from 0.01 wt % to 5 wt %, of a flow aid. In another embodiment, the LMP formulations may comprise from 0.5 wt % to 2 wt % of a flow aid.
  • In some embodiments, the compositions comprise an active agent, a low flow point excipient, and a high flow point excipient, where in the mass ratio of active agent is from 0.2 wt % to 60 wt %, the mass ratio of low flow point excipient from 10 wt % to 50 wt %, and the high flow point excipient ranges from 1 wt % to 30 wt % of the composition. In other embodiments, the mass ratio of active agent is from 5 wt % to 20 wt % of the composition, the mass ratio of the low flow point excipient is 50 wt % to 75 wt % of the composition, and the mass ratio of the high flow point excipient ranges from 2 wt % to 20 wt % of the composition. In still another embodiment, the mass ratio of active agent is from 5 wt % to 15 wt % of the composition, the mass ratio of the low flow point excipient is 50 wt % to 75 wt % of the composition, and the mass ratio of the high flow point excipient ranges from 3 wt % to 15 wt % of the composition.
  • In one embodiment, the compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a dispersing agent.
  • In some embodiments, the compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a dispersing agent.
  • In other embodiments, the compositions comprise from 5 wt % to 10 wt % of the active agent, from 60 wt % to 80 wt % of the low flow point excipient, from 2 wt % to 12 wt % of an antioxidant, from 2 wt % to 20 wt % of a high flow point excipient, and from 1 wt % to 10 wt % of a flow aid.
  • In a preferred embodiment, the compositions herein comprise, preferably consist of, from 3 wt % to 10 wt %, preferably from 4 wt % to 7 wt % of abiraterone acetate; from 10 wt % to 60 wt %, preferably from 20 wt % to 50 wt %, more preferably from 25 wt % to 35 wt %, more preferably from 27 wt % to 29 wt %, of a fatty acid ester of glycerol (preferably glycerol monooleate, like commercially available Peceol™) or mixtures thereof; from 20 wt % to 60 wt %, preferably from 30 wt % to 50 wt %, more preferably from 32 wt % to 33 wt %, of at least one of a propylene glycol fatty acid ester (preferably Propylene glycol monocaprylate, like commercially available Capryol™ 90) or a polyoxylglyceride (like commercially available Gelucire™, Labrasol™, Labrafil™); and from 20 wt % to 60 wt %, preferably from 30 wt % to 50 wt %, more preferably from 32 wt % to 33 wt %, of a surfactant typically selected from a hydrogenated castor oil (preferably Polyoxyl 40 hydrogenated castor oil, like commercially available Kolliphor® RH40). The compositions herein may be in the form of an isotropic clear solution.
  • Processes for Making the Compositions
  • In one embodiment, the following process is used to prepare the compositions. The active-free formulation is prepared by weighing the appropriate excipients into a vessel, and melting if necessary. The excipients are then mixed thoroughly to generate an isotropic solution. The mass of the abiraterone acetate is then added, and the active-free formulation is mixed at 50° C. until the active is completely dissolved (usually confirmed by polarized light microscopy). The final formulation is then cooled to ambient temperature for storage until filled into a capsule.
  • In another embodiment, the compositions may be formed by a “melt-spray-congeal” process, as described below. See, for example, U.S. Pat. Nos. 7,235,260, 7,887,844, EP1691787, U.S. Pat. No. 7,625,507.
  • A molten mixture is formed by mixing and heating the compositions. Such compositions are comprised of abiraterone acetate, and a lipid matrix. “Molten mixture” means that the mixture of abiraterone acetate and lipid matrix materials are sufficiently mixed and heated to fluidize the mixture sufficiently to allow it to be atomized into droplets. Generally, the mixture is molten in the sense that it will flow when subjected to one or more forces such as pressure, shear, and centrifugal force, such as that exerted by a centrifugal or spinning-disk atomizer.
  • Once the molten mixture has been formed, it is delivered to an atomizer that breaks the molten mixture into small droplets. Virtually any method can be used to deliver the molten mixture to the atomizer. In certain embodiments of the disclosed methods the molten mixture is delivered to the atomizer by use of pumps and/or various types of pneumatic devices such as pressurized vessels or piston pots or extruder. In certain embodiments the molten mixture is maintained at an elevated temperature during delivery to the atomizer to prevent its solidification and to keep it in a flowable state.
  • When a centrifugal atomizer (also known as rotary atomizers or spinning-disk atomizer) is used, the molten mixture is fed onto a rotating surface, where it spreads outward and flows by centrifugal force. The rotating surface may take several forms, examples of which include a flat disk, a cup, a vanned disk, and a slotted disk. The surface of the disk may also be heated to aid in atomization of the molten mixture or cooled to aid in the solidification of the LMPs. Several mechanisms of atomization are observed with flat-disk and cup centrifugal atomizers, depending on the flow of molten mixture to the disk, the rotation speed of the disk, the diameter of the disk, the viscosity of the feed, and the surface tension and density of the feed. At low flow rates, the molten mixture spreads out across the surface of the disk and when it reaches the edge of the disk, forms a discrete droplet, which is then flung from the disk.
  • Once the molten mixture has been atomized, the droplets are congealed, typically by contact with a gas at a temperature below the solidification temperature of the composition. Typically, it is desirable that the droplets are congealed in less than 60 seconds, less than 10 seconds, or even in less than 1 second. In certain embodiments congealing at ambient temperature using an ambient temperature cooling medium, results in sufficiently rapid solidification of the droplets. However, as certain embodiments of the disclosed compositions are comprised of at least 50 wt % of a low flow point excipient, it is often advantageous to utilize a cooling medium that is at a temperature that is at least 10° C. below ambient temperature. For some embodiments, it is preferred to utilize a cooling medium that is at least 20° C. below ambient temperature.
  • Dosage Forms
  • In one embodiment, the compositions of the invention are placed into a capsule for delivery by oral ingestion. Exemplary capsules include hard gelatin capsules, soft gelatin capsules, HPMC capsules as well as capsules made from other materials. The compositions may also be filled as a liquid into the capsule. The LMPs can also be suspended in a liquid such as an aqueous solution and then ingested. The LMPs may also be sprinkled on food and then ingested. The LMPs may also be mixed with solid excipients and then compressed into tablets.
  • Liquid-filled capsules can be hard or soft in form and may conveniently deliver the active within a liquid formulation. The fill formulation can contain either solubilized or suspended active, dependent on the choice of formulation, active solubility in this formulation and the dose per capsule. There is a performance advantage to the development of liquid formulations in which the active is pre-solubilized within the fill formulation since this active does not require further dissolution in the GI tract. This is important since the dissolution of the active in the GI tract can be rate-limiting to the absorption of poorly water-soluble drugs. Other advantages of liquid-filled capsules over other dosage forms such as tablets and caplets include better patient compliance, greater flexibility in dosage form design, and less expensive manufacturing process.
  • Liquid-filled two piece hard capsules (LFHC) may be produced from a range of materials, with gelatin and hydrophilic polymers often the preferred choice. In addition to these polymers, the capsule shell may also contain a pharmaceutically acceptable plasticizer, coloring agents and lubricants. Hard capsule shells are generally manufactured using dip molding processes.
  • LFHC may be used for a number of different applications including bioavailability enhancement of poorly water-soluble molecules, high-potency molecules, molecules susceptible to oxidation, molecules exhibiting low melting points, and molecules requiring controlled/sustained release formulations. The fill formulation can contain or mixtures of lipidic excipients such as those outlined above. Dependent on the composition, the fill formulation may be liquid at ambient temperature or solid/semi-solid.
  • Evaluation of Compositions
  • The compositions of the invention may be tested in vivo in animals, or in vitro using appropriate test solutions. Bioavailability of the compositions can be tested in vivo in animals or humans using conventional methods for making such a determination. An in vivo test, such as a crossover study, may be used to determine whether a composition provides an enhanced active agent concentration in the blood (serum or plasma) versus time area under the curve (AUC) for a test subject, dosed with the composition relative to the active agent concentration in the blood versus time AUC for a test subject dosed with a control composition. The bioavailability is measured as the area under the curve (AUC) determined for each group. In vivo determinations of AUC can be made by plotting the serum or plasma concentration of active agent along the ordinate (y-axis) against time along the abscissa (x-axis). Generally, the values for AUC represent a number of values taken from all of the subjects in a patient test population and are, therefore, mean values averaged over the entire test population. By measuring the AUC for a population to which the test composition has been administered and comparing it with the AUC for the same population to which the control composition has been administered, the test composition can be evaluated. The determination of AUCs is a well-known procedure and is described, for example, in Welling, “Pharmacokinetics Processes and Mathematics,” ACS Monograph 185 (1986).
  • In one embodiment, the AUC of the test composition divided by the amount dosed is at least 1.25-fold the AUC of the control composition divided by the amount dosed. In another embodiment, the AUC of the test composition divided by the amount dosed is at least 2-fold the AUC of the control composition divided by the amount dosed. In still another embodiment, the AUC of the test composition divided by the amount dosed is at least 2.5-fold the AUC of the control composition divided by the amount dosed. In still another embodiment, the AUC of the test composition divided by the amount dosed is at least 3-fold the AUC of the control composition divided by the amount dosed.
  • In one embodiment, the compositions may be tested in vitro using an appropriate test solution. Exemplary test solutions include aqueous solutions at 37° C. comprising 0.1 N HCl, simulating gastric fluid without enzymes, or intestinal buffer solutions, or water. Other in vitro tests solutions may be used as know by those skilled in the art.
  • EXAMPLES Example 1
  • The following components were formulated:
  • Function Component Amount (wt %)
    Active Abiraterone acetate 10.0
    Lipid phase Glycerol monooleate (Peceol ™) 31.5
    Cosurfactant Propylene glycol monolaurate 31.5
    (Lauroglycol ™ 90)
    Surfactant PEG15 hydroxystearate 27.0
    (Kolliphor ® HS-15)
  • The active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 50° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a pale yellow liquid, with evidence of some excipient solids on prolonged storage at room temperature.
  • On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 1 formed a homogenous cloudy dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 1, containing an additional 0.1% butylated hydroxytoluene (BHT) antioxidant, was placed on accelerated stability for 1 month. The active recrystallized at 25° C./60% RH while samples stored at 40° C./75% RH were physically stable (no crystals) and chemically stable (0.3% total impurities).
  • Example 2
  • The following components were formulated:
  • Function Component Amount (wt %)
    Active Abiraterone acetate 10.00
    Lipid phase Glycerol monocaprylate (Imwitor 308 ®) 33.75
    Cosurfactant Propylene glycol monocaprylate 33.75
    (Capryol ™ 90)
    Surfactant TPGS (Kolliphor ® TPGS) 22.50
  • The active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear colorless liquid.
  • On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 2 formed a homogenous cloudy dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 2, containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.0% total impurities) instabilities were seen.
  • Example 3
  • The following components were formulated:
  • Function Component Amount (wt %)
    Active Abiraterone acetate 10.0
    Lipid phase Propylene glycol monocaprylate 54.0
    (Capryol ™ 90)
    Cosurfactant PEG-8 caprylic/capric acid glycerides 18.0
    (Labrasol ®)
    Surfactant PEG15 hydroxystearate 18.0
    (Kolliphor ® HS-15)
  • The active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear, colorless liquid.
  • On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 3 formed a homogenous milky dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 3, containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.1% total impurities) instabilities were seen.
  • Example 4
  • The following components were formulated:
  • Function Component Amount (wt %)
    Active Abiraterone acetate 10.0
    Lipid phase C8-C10 triglycerides (Miglyol 812 ®) 13.5
    Cosurfactant Propylene glycol monocaprylate 54.0
    (Capryol ™ 90)
    Surfactant PEG15 hydroxystearate 22.5
    (Kolliphor ® HS-15)
  • The active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a glass vial, with prior excipient melting, if needed. Excipients were then mixed thoroughly to generate an isotropic solution. The target mass of active (abiraterone acetate) was then added to another glass vial followed by the active-free formulation and mixing at 30° C. until the active was completely dissolved (confirmed by polarized light microscopy). The final formulation was a clear colorless liquid.
  • On in vitro dispersion in 250 mL degassed water (37° C.), 1 g of Example 4 formed a homogenous cloudy to transparent dispersion, with no evidence of active crystals over a 6 h time-period.
  • Example 4, containing an additional 0.1% BHT antioxidant, was placed on accelerated stability for 1 month. No physical or chemically (0.1% total impurities) instabilities were seen.
  • In Vitro Digestion Tests to Identify Best Performing Abiraterone Acetate Formulation at 10.0% wt Loading: Example 1, 2, 3 and 4
  • In vitro digestion tests were performed as follows: 1 g of each active-containing formulation was dispersed in 40 mL fasted intestinal medium (2 mM Tris-maleate, 1.4 mM CaCl2.2H2O, and 150 mM NaCl, 3 mM sodium taurodeoxycholate and 0.75 mM phosphatidylcholine, 37° C.) using a pH-stat titrator as described by the LFCS Consortium (Williams, H D. et al., J. Pharm. Sci. 101 (2012), p. 3360-3380). Digestion was initiated on addition of 4 mL porcine pancreatic extract, prepared as described previously (Williams, H D. et al., J. Pharm. Sci. 101 (2012), p. 3360-3380), and continuously monitored and maintained at pH 6.5 with 0.2 or 0.6 M NaOH for 60 min during which period samples were removed, centrifuged to produce a multiphase sample consisting of; a pellet phase, containing any precipitated active; an aqueous phase, containing the active in free solution and active solubilized in small micelles and vesicles; and sometimes a lipid phase, containing the active solubilized in undigested oil droplets and/or larger colloids such as multi-lamellar vesicles. Active concentrations in these phases were measured by HPLC. The performance of Examples 1, 2, 3 and 4 on digestion are summarized below:
  • Digestion time
    point Example 1 Example 2 Example 3 Example 4
    Solubilized (lipid + aq. phase)
    active concentration (μg/mL)
    −5 min 2411 3018 2755 2937
    0 min 2392 3094 2805 2841
    5 min 1555 2195 2551 2647
    15 min 1642 1756 2360 2547
    30 min 1416 1478 2374 2593
    45 min 1000 1329 2200 2573
    60 min  657 1421 2250 2515
    Colloidal distribution of the active @ 60 min:
    Lipid phase  9% 68% 84%
    Aq. phase 30% 57% 22% 17%
    Pellet phase 59% 34%  5%  2%
  • The results show that all lipid formulations example were able to solubilize the active at concentrations much higher than the equilibrium solubility of the crystalline active in the same medium (without the digested lipid formulation), which was measured at 27 μg/mL. Example 1 and 2 showed evidence of active precipitation on digestion, as evidenced by the progressive decrease in solubilized concentrations with respect to time and the analysis of the pellet phase at 60 min. Example 3 and 4 were more robust, showing negligible active precipitation on digestion. In both cases, the majority of the active was solubilized in a lipid phase that phase-separated on centrifugation of the samples and most likely consisting of larger lipophilic colloids including multi-lamellar vesicles. Active drug in the aqueous phase (consisting of BS/PL mixed-micelles, lipid-enriched BS/PL mixed micelles and small vesicles) is considered the most readily available form for absorption.
  • Examples 6-9 at Lower 7% w/w Active Loading
  • The following components were formulated:
  • Amount (wt %)
    Component Example 6 Example 7 Example 8 Example 9
    Abiraterone acetate  7.00  7.00  7.00  7.00
    Glycerol monooleate 27.90 27.90 27.90 46.50
    (Peceol ™)
    Propylene glycol 32.55
    monolaurate
    (Lauroglycol ™ 90)
    Propylene glycol 32.55
    monocaprylate
    (Capryol ™ 90)
    PEG-6 oleyl glycerides 32.55 23.25
    (Labrafil ® M1944CS)
    PEG15 hydroxystearate 32.55 32.55 23.25
    (Kolliphor ® HS-15)
    Polyoxyl 40 32.55
    hydrogenated
    castor oil
    (Kolliphor ® RH40)
  • Active-free formulations were prepared as described for Example 1. The above examples formed isotropic solutions at 50° C. that were colorless to yellow in color depending on the composition. At lower temperatures, there was some evidence of excipient solidification, e.g., in the case of Examples 8 and 9.
  • Example 7 and 9 containing an additional 0.1% BHT antioxidant, were placed on accelerated stability for 1 month. The results are summarized in the table below:
  • Impurities (%)
    25° C./60% RH 40° C./75% RH
    Physical Chemical Physical Chemical
    Stability Stability Stability Stability
    Example 7 - 1 month Stable 0.1% Stable 0.1%
    Example 9 - 1 month Stable <0.1% Stable 0.32%
  • Under accelerated conditions, Example 9 exhibited greater levels of unknown impurities relative to Example 7. Both Examples were physically stable over 1 month.
  • Example formulations 6-9 were then subjected to in vitro dispersion tests wherein 1 g of formulation was dispersed in 250 mL degassed water at 37° C. over 6 hours. The following results were obtained:
  • Dispersion
    observations Example 6 Example 7 Example 8 Example 9
    Macroscopic Milky Translucent/ Cloudy Cloudy
    appearance Cloudy
    Microscopic Aggregates and No particles were Poorly dispersed Poorly dispersed
    appearance poorly dispersed evident oil droplets oil droplets
    oil droplets
    Evidence of No precipitation No precipitation No precipitation No precipitation
    active over 6 h over 6 h over 6 h over 6 h
    precipitation
  • All examples provided the desirable property of precipitation of the active over 6 hours. Since the solubilization of poorly water-soluble actives is critical to absorption, Examples 6-9 may support the intestinal absorption of the active, abiraterone acetate. Example 7 exhibited superior dispersion properties and was progressed into in vitro digestion tests. These tests were performed as described above for Examples 1-4. The following results were obtained:
  • Digestion time
    point Example 7
    Solubilized (lipid + aq. phase)
    active concentration (μg/mL)
    −5 min 1858
    0 min 1855
    5 min 1961
    15 min 1611
    30 min 1592
    45 min 1544
    60 min 1504
    Colloidal distribution of the
    active @ 60 min:
    Lipid phase  7%
    Aq. phase 80%
    Pellet phase 13%
  • Example 7 was robust to digestion, with minimal precipitation of the active to the pellet phase. Due to the optimized dispersion properties, Example 7 also provided a high percentage of active solubilized in the aqueous phase—this phase consists of small dispersed colloids including micelles and small vesicles. Solubilized active concentrations in both the aqueous phase (in which the majority (80%) of active was present) and lipid phase were >1500 μg/mL during the entire digestion test. As a comparison, the crystalline solubility of the active in the same medium was measured at 27 μg/mL.
  • Variations of Example 7 containing different loadings of the active were prepared in the same manner as described for Example 1:
  • Amount (wt %)
    Component Example 10 Example 11 Example 12
    Abiraterone acetate 3.00 6.00 10.00
    Glycerol monooleate 29.10 28.20 27.00
    (Peceol ™)
    Propylene glycol 33.95 32.90 31.50
    monocaprylate
    (Capryol ™ 90)
    Polyoxyl 40 hydrogenated 33.95 32.90 31.50
    castor oil
    (Kolliphor ® RH40)
  • Examples 10-12 formed isotropic clear solutions that were pale yellow in color. Example 11 was placed on stability over 3 months. The following results were obtained:
  • 5° C. 25° C./60% RH 40° C./75% RH
    Example Physical Chemical Physical Chemical Physical Chemical
    11 Stability Stability Stability Stability Stability Stability
    1 month Stable <0.1% Stable <0.1% Stable <0.1%
  • Examples 10-12 were tested by in vitro digestion following the same method as previously described. The following results were obtained:
  • Digestion time
    point Example 10 Example 11 Example 12
    Solubilized (lipid + aq. phase)
    active concentration (μg/mL)
    −5 min 884 1592 2505
    0 min 846 1691 2477
    5 min 719 1613 2038
    15 min 712 1244 1984
    30 min 720 1197 1972
    45 min 712 1226 1962
    60 min 646 1164 1925
    Colloidal distribution of the active @ 60 min:
    Lipid phase 12% 10% 18%
    Aq. phase 75% 80% 76%
    Pellet phase 13% 10%  6%
  • Since 1 gram of formulation was evaluated, Examples 10, 11 and 12 provided 30 mg, 60 mg and 100 mg of active, respectively, in the digestion test. The results indicate no significant change in performance with increasing active loading in the formulation as evidenced by negligible change in % active recovered in the pellet phase and high solubilized active concentrations in the aqueous and lipid phase. Indeed, comparing Examples 7, 10, 11 and 12 which utilize the same excipients in the same ratios and only differ in active loading, a linear correlation (R2=0.9835) can be obtained between solubilized active concentrations following 60 min digestion and active loading in the formulation. This capacity of a formulation to maintain the active in solution across a range of active loadings is desirable for consistent in vivo performance at different doses. In addition, since good performance is maintained at a 10% w/w active loading, there is the gained knowledge that lower active loadings are not near a performance threshold. Solubilized active concentrations at the lowest loading (i.e., Example 10) were still significantly in excess of the crystalline solubility (27 μg/mL).
  • In Vivo Performance Assessment of Example 10 in Beagle Dogs Vs. Zytiga® Tablets
  • The in vivo absorption of the active was assessed in a cross-over study using fasted beagle dogs (n=6). In this study, the commercial abiraterone acetate tablet (Zytiga® 250 mg) and Example 10 were both dosed. The target dose delivered by Example 10 was 25 mg, i.e., 1/10th of the Zytiga® dose, which was achieved using approximately 0.83 mL formulation filled in hard capsules (Licaps®, size 00el) prior to administration. The following results were obtained (note that results are expressed as measured plasma abiraterone due to complete conversion from abiraterone acetate in vivo):
  • Dose AUC0-24 h Relative
    Treatment (mg) (h · ng/mL) ± SD CV Tmax (h) BA
    Zytiga ®
    250 413 ± 378 94% 2.4 ± 2.1 100.0%
    tablet fasted
    Example 10 25 227 ± 39  22% 0.8 ± 0.3 50.6%
    fasted
  • The mean total absorption of the active from the commercial tablet was 413 h·ng/mL. Example 10 provided approximately 50% of this exposure (227 h·ng/mL) despite dosing the dogs only 1/10th of the Zytiga® dose. This result therefore highlights the low absorption of the active from the fasted state using Zytiga® which contains crystalline drug and the fact that a higher proportion of the administered dose can be absorbed using lipid formulations that are effective in maintaining substantially higher solubilized abiraterone acetate concentrations (such as Example 10). In addition to supporting the total extent of absorption (captured here by AUC values), Example 10 provided a marked reduction in inter-dog absorption variability (captured here by CV, the coefficient of variation) and variability in absorption rate (captured here by the Tmax) in comparison to Zytiga®. The results of this study indicate that equivalent exposure to Zytiga® may be obtained at a substantially lower dose than 250 mg. Further analyses of the above results indicate that this equivalence might be achieved at a 50 mg active dose, which may be delivered in a capsule by Example 11 with no change in total formulation volume when compared to Example 10.
  • In Vitro Robustness Digestion Testing of Example 11
  • To further test formulation robustness as a digestion condition, Example 11 was assessed on in vitro digestion under more dilute experimental conditions:
  • 0.33 g of Example 11 0.83 g of Example 11
    Digestion time tested in 40 mL tested in 40 mL
    point digestion medium digestion medium
    Solubilized (lipid + aq. phase)
    active concentration (μg/mL)
    −5 min 553 1409
    0 min 559 1363
    5 min 448 1127
    15 min 423 1128
    30 min 382 1102
    45 min 379 1111
    60 min 352 1078
    Colloidal distribution of the
    active @ 60 min:
    Lipid phase  6%  4%
    Aq. phase 70% 85%
    Pellet phase 24% 11%
  • Example 11 tested at 0.33 g and 0.83 g equated to ˜20 mg and ˜50 mg active doses. The testing of lower formulation masses in the same medium and same medium volume generates higher bile salt/phospholipid-to-lipid formulation ratios. This is known by those skilled in the art to in some cases to “stress” lipid formulations and trigger increased precipitation of the active. Using rational design approaches for specific actives, however, formulations such as Example 11 can be obtained that show consistent solubilization properties irrespective of the extent of formulation dilution. Indeed, as shown above, Example 11 maintained at least 75% of the active in the solubilized state during digestion under more stressful (i.e., more dilute) conditions. Such consistent solubilization properties are desirable to the development of robust formulations since differences in dilution may be reasonably encountered in vivo through e.g., inter-subject variability in GI fluid volumes or in instances where patients are dosed different number of formulation capsules.
  • Example 11 In Vitro Performance Comparison to “SMES 1” in WO2014009434A1
  • WO2014009434A1 discloses several castor oil containing lipid formulations for the active abiraterone acetate. According to WO2014009434A1, SMES 1 consists of 20% wt castor oil, 20% wt Capmul® MCM, 30% wt Span 80 and 30% Kolliphor® EL. This placebo formulation was also prepared here followed by the complete incorporation of the active at a 60 mg/g loading to enable direct in vitro performance comparison to Example 11. Indeed, SMES 1 was subjected to in vitro dispersion and in vitro digestion tests as described above.
  • On dispersion, SMES 1 formed a milky dispersion indicating the presence of larger oil droplets in comparison to Example 11 which forms a more translucent dispersion. SMES 1 did however protect against precipitation of the active over 6 hours of dispersion.
  • On digestion of SMES 1, the following results were obtained:
  • Digestion time
    point “SMES 1”
    Solubilized (lipid + aq. phase)
    active concentration (μg/mL)
    −5 min 1582
    0 min 1678
    5 min 757
    15 min 614
    30 min 666
    45 min 774
    60 min 766
    Colloidal distribution of the
    active @ 60 min:
    Lipid phase 22%
    Aq. phase 47%
    Pellet phase 31%
  • The results show that SMES 1 exhibited a tendency toward precipitation of the active immediately on digestion of the formulation, with over a 50% decrease in solubilized active concentrations between t=0 and t−5 min, As a result, SMES 1 provided substantially lower solubilization after 1 hour digestion in comparison to Example 11 (766 ug/mL vs. 1164 ug/mL). In addition, due to poor dispersibility, the fraction of the active solubilized in the lipid phase was higher for SMES 1 in comparison to Example 11 (22% vs. 10%).
  • Example 5
  • The following components were formulated
  • Function Component Amount (wt %)
    Active Abiraterone Acetate 10.0
    Low flow point C10-C18 Triglycerides 28.3
    Lipid A (Gelucire ® 43/01)
    Low flow point Stearoyl macrogol-32 glycerides 56.6
    Surfactant B (Gelucire ® 50/13)
    High flow point Glyceryl behenate 5.0
    Lipid C (Compritol ® 888)
    Antioxidant Butylated hydroxytoluene (BHT) 0.1
  • The active-free formulation was prepared first by weighing the appropriate amount of each of the above excipients into a water-jacketed tank, with prior excipient melting at 75° C. Excipients were then mixed thoroughly to generate a single phase solution. The active, (abiraterone acetate) was added and mixed with the molten mixture. Then the molten mixture was allowed to cool to 60° C. The molten mixture was then injected onto a melt-spray congeal apparatus equipped with a spinning disk atomizer spinning at 2050 rpm, maintained at 60° C. and at a s pray rate of 120 g/min. Droplets of the molten mixture were formed and congealed at <5° C. The resulting LMP particles had a generally spherical shape, and a mean particle size of 270 μm.
  • In Vitro Performance Tests
  • In vitro digestion tests were performed as follows: 1 g of the active-containing formulation was dispersed in 40 mL fasted intestinal medium (2 mM Tris-maleate, 1.4 mM CaCl2.2H2O, and 150 mM NaCl, 3 mM sodium taurodeoxycholate and 0.75 mM phosphatidylcholine, 37° C.) using a pH-stat titrator as described by the LFCS Consortium (Williams, H D. et al., J. Pharm. Sci. 101 (2012), p. 3360-3380). Digestion was initiated on addition of 4 mL porcine pancreatic extract, prepared as described previously (Williams, H D. et al., J. Pharm. Sci. 101 (2012), p. 3360-3380), and continuously monitored and maintained at pH 6.5 with 0.2 or 0.6 M NaOH for 60 min during which period samples were removed, centrifuged to produce a multiphase sample consisting of; a pellet phase, containing any precipitated and suspended active; an aqueous phase, containing the active in free solution and active solubilized in small micelles and vesicles; and sometimes a lipid phase, containing the active solubilized/suspended in undigested and undispersed solid lipid phases and/or larger colloids such as multi-lamellar vesicles. Active concentration in the aqueous phase was measured by HPLC. The performance of Example 5 on digestion is summarized below. The same test was performed again without adding pancreatic enzyme to evaluate the impact of digestion on drug solubility in the lipidic colloidal species. Also thermodynamic solubility of the active was evaluated in digested placebo formulations for determining solubility in digested lipidic colloidal phases.
  • Example 5
    Active concentration (μg/mL)
    Digestion Test 2
    time Test 1 (undigested)
    −5 548 427
    0 623 435
    5 757 519
    15 754 665
    30 593 502
    45 401 640
    60 381 513
    AA solubility in crystalline 27
    digestion media
    AA solubility in digested 121
    LMP formulations
  • The results show that the formulation of example 5 was able to sustain the active at concentrations much higher than the equilibrium solubility of the crystalline active in the same medium, which was measured at 27 μg/mL. Example 5 showed evidence of active precipitation on digestion, as evidenced by the decrease in solubilized concentrations once the formulation started to disperse around 15 min. Through Test 2 it is seen that digestion does not greatly impact compound solubilization which is positive. Digestion itself may have promoted “release” of the dissolved/dissolving drug though this might have been masked if released drug rapidly recrystallized. Equilibrium solubility of crystalline active in the digested LMP's was measured at 121 μg/mL, which shows overall improvement in solubilization provided by the digested BS/PL-lipid mixed micellar system; active drug in the aqueous phase containing BS/PL-Lipid-enriched mixed micelles is considered the available form for passing drug through the unstirred water layer to the enterocyte for absorption.
  • The antioxidant, BHT, was added at a 0.1% wt concentration to Example 5, which were then placed on accelerated chemical stability at 25° C./60% RH and 40° C./75% RH for 1 month. The results are summarized in the table below.
  • Active loading
    Example 5 (% wt) 25° C./60% RH 40° C./75% RH
    (1 month) 10 <LOQ <LOQ
  • LMP Examples 12 and 13 were also successfully prepared in the same fashion as described above for Example 5:
  • Amount (wt %)
    Component Example 12 Example 13
    Abiraterone acetate 4.00 20.00
    C10-C18 Triglycerides 30.32 24.90
    (Gelucire ® 43/01)
    Stearoyl macrogol-32 glycerides 60.64 49.90
    (Gelucire ® 50/13)
    Glyceryl behenate (Compritol ® 888) 5.00 5.00
    Butylated hydroxytoluene (BHT) 0.04 0.20
  • Example 12 was subjected to in vitro digestion tests are described for previous formulations. The following results were obtained:
  • Digestion Example 12
    time Active concentration (μg/mL)
    −5 811
    0 649
    5 694
    15 658
    30 592
    45 564
    60 811
  • The results show that Example 12 generally provided higher solubilized active concentrations in comparison to Example 5, despite the use of a lower active loading (4% wt vs. 10% wt) and due to the formulation containing a higher fraction or pre-dissolved active in the formulation and the higher formulation:active ratio.
  • Example 12 was placed on accelerated chemical stability at 25° C./60% RH and 40° C./75% RH for 1 month. The results are summarized in the table below.
  • Active loading
    Example 12 (% wt) 25° C./60% RH 40° C./75% RH
    (1 month) 4 <LOQ <LOQ
  • In Vivo Performance Assessment of Example 11 in Beagle Dogs Vs. Zytiga® Tablets in Fed/Fasted Study
  • The in vivo absorption of the active was assessed in a further cross-over study in fasted and fed beagle dogs (n=6). In the fed legs of the study, dogs received ˜100 g of FDA high-fat diet daily via oral gavage for 3 days before treatment. This meal consisted of 1100-1200 calories, a total mass of 600-700 g and with ˜50% of the calories from fat. The meal was homogenized prior to administration. On the day of dosing, dogs received this meal 30 min prior to dosing.
  • Dogs were randomized over 6 weeks to receive either the commercial abiraterone acetate tablet (Zytiga® 250 mg), Example 11 and Example 5, in a fasted and fed state. The target dose in the lipid formulation (Example 11) and LMP formulation (Example 5) was 50 mg, namely ⅕th of the Zytiga® dose. This dose was delivered using approximately 0.83 mL of Example 11, while for Example 5, using approximately 500 mg formulation. Both were filled in hard gelatin capsules (Licaps®, size 00el) prior to administration.
  • The following results were obtained (results are expressed as measured abiraterone in the plasma due to complete conversion from abiraterone acetate). Results are also presented in FIG. 1.
  • Dose Cmax ± SD AUC0-24 h
    Treatment (mg) (ng/mL) (h · ng/mL) CV Tmax (h) Relative BA
    Zytiga ® tablet fasted 250  303.7 ± 313.4 713 111%  1.2 ± 0.6 100%
    Zytiga ® tablet fed 250 2401.7 ± 840.0 7098 39% 2.1 ± 1.0 ~1000% 
    Example 11 fasted 50 401.2 ± 99.1 852 28% 0.9 ± 0.2 119%
    Example 11 fed 50  373.3 ± 129.8 762 28% 1.4 ± 0.5 107%
    Example 5 fasted 50 161.4 ± 71.8 351 36% 1.0 ± 0.3  49%
    Example 5 fed 50 185.7 ± 39.1 562 25% 2.7 ± 0.5  79%
  • Consistent with the first dog study performed (described above), the fasted state absorption of the active from the Zytiga® tablet was relatively low and variable. Exposure in the fed state was ˜10-fold higher, increasing from 713 h·ng/mL to 7098 h·ng/mL, while the inter-dog variability decreased substantially from 111% in the fasted to state to 39% when co-administered with food. In the same cross-over study, Example 11 achieved a fasted-state exposure of 852 h·ng/mL, which was slightly higher than Zytiga® under the same conditions despite dosing only ⅕th of the dose. Example 11 also substantially reduced fasted-state variability in absorption (reduced from 111% to 28%) while absorption from Example 11 was near independent of food—only a small decrease in the extent of absorption in the presence of food (762 h·ng/mL) was observed.
  • Example 5 did not provide the same level of absorption when compared to Example 11 at a 50 mg dose (AUC0-24 h values were 351 and 562 h·ng/mL in the fasted and fed state, respectively, compared to 852 and 762 h·ng/mL for Example 11), but Example 5 did achieve a total exposure that was ˜50% relative to Zytiga®, while also producing a substantial reduction in variability (36%). In terms of the food-effect, Example 5 exhibited a modest ˜1.2-fold increase in exposure in the fed state.
  • This substantial increase in abiraterone acetate exposure in the presence of food in dogs on dosing Zytiga® tablets is consistent with evidence of a very large food effect (˜10-fold) in human subjects. By providing great solubilization of the active in the GI tract, both Example 5 and Example 11 are able to support higher amounts of absorption in the fasted state. This in turn affords the possibility of lower doses, reduced variability and eliminated food-effect as illustrated here by lipid formulation Examples 5 and 11.
  • See FIG. 1.
  • Abiraterone plasma concentration versus time data after oral administration of Zytiga® tablet (250 mg abiraterone acetate) or Example 11 capsule (50 mg abiraterone acetate) or Example 5 capsule (50 mg abiraterone acetate) to beagle dogs in either fasted or fed states. Mean±SD

Claims (15)

1. A dosage form, comprising:
a capsule; and
a lipid fill composition, comprising
abiraterone acetate;
a lipid matrix comprising (i) a fatty acid ester of glycerol, or a mixture thereof in an amount ranging from 10 wt % to 60 wt % of the lipid fill composition; and (ii) a propylene glycol fatty acid ester, or a polyoxylglyceride, or a combination thereof, wherein the propylene glycol fatty acid ester and/or the polyoxylglyceride is present in an amount ranging from 20 wt % to 60 wt % of the lipid fill composition; and
an optional antioxidant; wherein the abiraterone acetate is solubilized in the lipid matrix.
2. The dosage form of claim 1, wherein the abiraterone acetate is present in an amount ranging from 5 wt % to 20 wt %.
3. The dosage form of claim 1, wherein at least 60 wt % of the abiraterone acetate in the lipid fill composition is crystalline.
4. The dosage form of claim 1, wherein the fatty acid ester of glycerol is glycerol monolinoleate, glycerol monooleate, glycerol monostearate, glycerol monocaprylate, glycerol monocaprate, or mixtures thereof.
5. The dosage form of claim 1, wherein the propylene glycol fatty acid ester is selected from propylene glycol monocaprylate, a propylene glycol dicaprate/dicaprylate ester, propylene glycol heptanoate, propylene glycol monolaurate, propylene glycol monostearate, propylene glycol monooleate, propylene glycol monopalmitate, propylene glycol monomyristate, or a mixture thereof.
6. The dosage form of claim 1, wherein the polyoxylglyceride is selected from an oleoyl macrogol-6 glyceride, a linoleoyl macrogol-6 glyceride, a lauroyl macrogol-6 glyceride or a mixture thereof.
7. The dosage form of claim 1, wherein the lipid fill composition further comprises a surfactant.
8. The dosage form of claim 7, wherein the surfactant is present in an amount ranging from 15 wt % to 60 wt %.
9. The dosage form of claim 7, wherein the surfactant is polyoxyl 40 hydrogenated castor oil, polyoxyl 35 castor oil, polyethoxylated 12-hydroxteric acid, vitamin E, tocopheryl polyethylene glycol succinate, a caprylocaproyl macrogol-8 glyceride, a lauroyl macrogol-32 glyceride, a macrogol stearate, a polysorbate, or a mixture thereof.
10. The dosage form of claim 1, wherein the antioxidant is present and is selected from vitamin E, tocopheryl polyethylene glycol succinate, rosemary extract, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, or a mixture thereof.
11. The dosage form of claim 1, wherein the lipid fill composition comprises the abiraterone acetate; the fatty acid ester of glycerol in an amount ranging from 25 wt % to 35 wt %; and the propylene glycol fatty acid ester or the polyoxylglyceride in an amount ranging from 30 wt % to 50 wt %.
12. The dosage form of claim 11, wherein the lipid fill composition further comprises a surfactant, an antioxidant, or a combination thereof.
13. The dosage form of claim 12, wherein the surfactant is polyoxyl 40 hydrogenated castor oil.
14. The dosage form of claim 1, wherein the fatty acid ester of glycerol is glycerol monooleate; and the propylene glycol fatty acid is propylene glycol monolaurate, propylene glycol monocaprylate, or a mixture thereof.
15. The dosage form of claim 1, wherein the capsule is a hard capsule or a soft capsule.
US16/906,603 2015-04-10 2020-06-19 Abiraterone acetate lipid formulations Abandoned US20200316093A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/906,603 US20200316093A1 (en) 2015-04-10 2020-06-19 Abiraterone acetate lipid formulations

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562145864P 2015-04-10 2015-04-10
PCT/EP2016/056689 WO2016162229A1 (en) 2015-04-10 2016-03-25 Abiraterone acetate lipid formulations
US201715565400A 2017-10-09 2017-10-09
US16/906,603 US20200316093A1 (en) 2015-04-10 2020-06-19 Abiraterone acetate lipid formulations

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2016/056689 Continuation WO2016162229A1 (en) 2015-04-10 2016-03-25 Abiraterone acetate lipid formulations
US15/565,400 Continuation US10722527B2 (en) 2015-04-10 2016-03-25 Abiraterone acetate lipid formulations

Publications (1)

Publication Number Publication Date
US20200316093A1 true US20200316093A1 (en) 2020-10-08

Family

ID=55629030

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/565,400 Active US10722527B2 (en) 2015-04-10 2016-03-25 Abiraterone acetate lipid formulations
US16/906,603 Abandoned US20200316093A1 (en) 2015-04-10 2020-06-19 Abiraterone acetate lipid formulations

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/565,400 Active US10722527B2 (en) 2015-04-10 2016-03-25 Abiraterone acetate lipid formulations

Country Status (4)

Country Link
US (2) US10722527B2 (en)
EP (2) EP3280448B1 (en)
ES (1) ES2863500T3 (en)
WO (1) WO2016162229A1 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020118415A1 (en) * 2018-12-10 2020-06-18 CannTrust Inc. Cannabis-based self-emulsifying product
CN111481523B (en) * 2019-01-25 2023-03-24 鲁南制药集团股份有限公司 Soft capsule for treating prostatic cancer and preparation method thereof
CN110354094B (en) * 2019-01-25 2020-07-31 鲁南制药集团股份有限公司 CYP17 inhibitor soft capsule and preparation method thereof
CN109846826A (en) * 2019-01-25 2019-06-07 湖南华腾制药有限公司 Abiraterone acetate flexible lipidosome and preparation method thereof
WO2021076920A1 (en) 2019-10-16 2021-04-22 Capsugel Belgium Nv Method and composition for increasing muscle protein synthesis
AU2020384948A1 (en) 2019-11-14 2022-06-02 Suven Life Sciences Limited Amorphous pharmaceutical compositions of abiraterone acetate
EP4054348A1 (en) 2019-12-10 2022-09-14 Capsugel Belgium NV Particles containing a lipid matrix core and active ingredient
CN113133984A (en) * 2020-01-20 2021-07-20 鲁南制药集团股份有限公司 CYP17 inhibitor preparation
CN113133983B (en) * 2020-01-20 2023-12-12 鲁南制药集团股份有限公司 Pharmaceutical composition for treating prostate cancer
EP3854384A1 (en) 2020-01-27 2021-07-28 Adamed Pharma S.A. Pharmaceutical formulation comprising abiraterone acetate
CA3184236A1 (en) 2020-07-01 2022-01-06 Tyler White Dietary composition and method
EP3944860A1 (en) * 2020-07-30 2022-02-02 Galenicum Health S.L.U. Abiraterone for use in a method of treating cancer
WO2023150107A1 (en) 2022-02-02 2023-08-10 Lonza Greenwood Llc Fast acting joint health composition and use thereof
WO2023150113A1 (en) 2022-02-02 2023-08-10 Lonza Greenwood Llc Fast acting joint health composition and use thereof
WO2023150108A1 (en) 2022-02-02 2023-08-10 Lonza Greenwood Llc Extended release boswellic acid and manufacturing thereof
WO2023230216A1 (en) 2022-05-26 2023-11-30 Lonza Greenwood Llc Lipid microcapsules for viable and stable probiotics

Family Cites Families (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5604213A (en) 1992-03-31 1997-02-18 British Technology Group Limited 17-substituted steroids useful in cancer treatment
ATE186913T1 (en) 1992-03-31 1999-12-15 Btg Int Ltd 17-SUBSTITUTED STEROIDS USABLE IN TREATING CANCER
DE69811233T2 (en) 1997-10-27 2003-11-20 Merck Patent Gmbh SOLID SOLUTIONS AND DISPERSIONS OF A POOR WATER-SOLUBLE ACTIVE SUBSTANCE
FR2779962B1 (en) 1998-06-17 2002-12-06 Karim Ioualalen COSMETIC OR DERMOPHARMACEUTICAL COMPOSITION IN THE FORM OF PEARLS AND METHODS FOR THE PREPARATION
US20010053791A1 (en) 2000-03-16 2001-12-20 Babcock Walter C. Glycogen phosphorylase inhibitor
EP1351678A2 (en) 2001-01-02 2003-10-15 Elizabeth Shanahan-Prendergast Treatment for inhibiting neoplastic lesions using incensole and/or furanogermacrens
EP1336602A1 (en) 2002-02-13 2003-08-20 Giovanni Scaramuzzino Nitrate prodrugs able to release nitric oxide in a controlled and selective way and their use for prevention and treatment of inflammatory, ischemic and proliferative diseases
US7101576B2 (en) 2002-04-12 2006-09-05 Elan Pharma International Limited Nanoparticulate megestrol formulations
US8216609B2 (en) 2002-08-05 2012-07-10 Torrent Pharmaceuticals Limited Modified release composition of highly soluble drugs
US7985422B2 (en) 2002-08-05 2011-07-26 Torrent Pharmaceuticals Limited Dosage form
AU2003274823A1 (en) 2002-10-23 2004-05-13 Pantarhei Bioscience B.V. Pharmaceutical compositions comprising estetrol derivatives for use in cancer therapy
US6913708B2 (en) 2003-02-24 2005-07-05 3M Innovative Properties Company Cholesteric liquid crystal drying process and solvent
EP1636182A4 (en) 2003-06-12 2008-09-03 Merck & Co Inc Prodrugs of mitotic kinesin inhibitors
DE602004014805D1 (en) 2003-12-04 2008-08-14 Pfizer Prod Inc METHOD FOR THE PRODUCTION OF PHARMACEUTICAL MULTILAYFUL PRODUCTS
ES2600577T3 (en) 2003-12-04 2017-02-09 Bend Research, Inc. Spray-solidification process using an extruder to prepare multiparticulate compositions of crystalline drugs
WO2005053652A1 (en) 2003-12-04 2005-06-16 Pfizer Products Inc. Multiparticulate crystalline drug compositions containing a poloxamer and a glyceride
US9016221B2 (en) 2004-02-17 2015-04-28 University Of Florida Research Foundation, Inc. Surface topographies for non-toxic bioadhesion control
GB0418900D0 (en) 2004-08-24 2004-09-29 Btg Int Ltd Novel salt forms
JP2008521807A (en) * 2004-11-24 2008-06-26 メルク エンド カムパニー インコーポレーテッド Liquid and semisolid pharmaceutical formulations for oral administration of substituted amides
FR2879930B1 (en) 2004-12-23 2007-08-31 Oralance Pharma Sa NEW GALENIC SYSTEM FOR TRANSPORTING ASSETS, PROCESS FOR PREPARATION AND USE
WO2006116716A2 (en) 2005-04-27 2006-11-02 University Of Florida Materials and methods for enhanced degradation of mutant proteins associated with human disease
US20100004156A1 (en) 2005-07-27 2010-01-07 Shalesh Kaushal Small Compounds That Correct Protein Misfolding and Uses Thereof
KR101592659B1 (en) 2005-10-07 2016-02-11 더 유니버시티 오브 알라바마 Multi-functional ionic liquid composition for overcoming polymorphism and imparting improved properties for active pharmaceutical, biological, nutritional, and energetic ingredients
EP1787985A1 (en) 2005-11-09 2007-05-23 Unibioscreen S.A. Azonafide derivatives, methods for their production and pharmaceutical compositions therefrom
US8158152B2 (en) 2005-11-18 2012-04-17 Scidose Llc Lyophilization process and products obtained thereby
AU2007300734A1 (en) 2006-06-02 2008-04-03 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services RNA nanoparticles and nanotubes
CN101511361A (en) 2006-06-30 2009-08-19 先灵公司 Method of using substituted piperidines that increase p53 activity
JP2009542666A (en) 2006-06-30 2009-12-03 シェーリング コーポレイション Substituted piperidines that increase P53 activity and uses thereof
US20080051380A1 (en) 2006-08-25 2008-02-28 Auerbach Alan H Methods and compositions for treating cancer
US20080051375A1 (en) 2006-08-25 2008-02-28 Auerbach Alan H Methods for treating cancer comprising the administration of a vitamin d compound and an additional therapeutic agent, and compositions containing the same
ES2428634T3 (en) 2006-08-25 2013-11-08 Janssen Oncology, Inc. Compositions for cancer treatment
EP2081964A4 (en) 2006-10-12 2012-07-11 Univ Johns Hopkins Alginate and alginate lyase compositions and methods of use
WO2008062466A2 (en) 2006-10-13 2008-05-29 Reliance Life Sciences Pvt. Ltd. Cinnamic acid, vanillic acid and benzofuran derivatives for use in the treatment of inflammation and cancer
EP2128123A4 (en) 2007-01-29 2011-04-20 Medrx Co Ltd Salt of nonsteroidal anti-inflammatory drug and organic amine compound and use thereof
WO2008154382A1 (en) 2007-06-06 2008-12-18 University Of Maryland, Baltimore Hdac inhibitors and hormone targeted drugs for the treatment of cancer
WO2009009132A1 (en) 2007-07-12 2009-01-15 Cougar Biotechnology, Inc. Use of 17alpha-hydroxylase/c17, 20-lyase inhibitors for the treatment of cancer
EP2229147A2 (en) 2007-12-03 2010-09-22 The Johns Hopkins University Methods of synthesis and use of chemospheres
US20110033461A1 (en) 2008-03-12 2011-02-10 Vladimir Ratushny Combination Therapy for the Treatment of Cancer
RS54428B1 (en) 2008-05-13 2016-04-28 Yale University Chimeric small molecules for the recruitment of antibodies to cancer cells
US8835506B2 (en) 2008-06-05 2014-09-16 Stc.Unm Methods and related compositions for the treatment of cancer
SI2285350T1 (en) 2008-06-16 2018-03-30 Pfizer Inc. Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic nanoparticles
EP2340041A2 (en) 2008-09-08 2011-07-06 Hoffman/Barrett, L.L.C. Porphyrazine optical and dual optical/mr contrast and therapeutic agents
US20100233112A1 (en) 2009-01-16 2010-09-16 Jinlian Hu Shape memory polymer network using heterocyclic groups
US8791095B2 (en) 2009-02-05 2014-07-29 Tokai Pharmaceuticals, Inc. Steroidal CYP17 inhibitors/antiandrogens
KR20110113755A (en) 2009-02-10 2011-10-18 아스트라제네카 아베 Triazolo [4,3-b] pyridazine derivatives and their uses for prostate cancer
US9427418B2 (en) 2009-02-23 2016-08-30 Gtx, Inc. Estrogen receptor ligands and methods of use thereof
US9005608B2 (en) 2009-03-24 2015-04-14 Adds Pharmaceuticals Llc Stabilized solubility-enhanced formulations for oral delivery
CA2759098A1 (en) 2009-04-14 2010-10-21 Smith Holdings, Llc Methods and compositions for the treatment of medical conditions involving cellular programming
US20120121692A1 (en) 2009-05-08 2012-05-17 Shandong University Compounds and compositions comprising cdk inhibitors and methods for treating cancer
WO2010143199A1 (en) 2009-06-11 2010-12-16 Suven Nishtaa Pharma Private Limited Solid lipid dispersion for aqueous solubility enhancement of poorly water soluble drugs
RU2519123C2 (en) 2009-07-06 2014-06-10 Аэрпио Терапьютикс Инк. Compounds, compositions and methods for prevention of metastatic cancer cells
WO2011032100A1 (en) 2009-09-11 2011-03-17 Government Of The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Inhibitors of kshv vil6 and human il6
WO2011057064A1 (en) 2009-11-05 2011-05-12 Brian Long Igf1r inhibitor based treatment of prostate cancer
US8710035B2 (en) 2010-03-24 2014-04-29 The University Of Chicago Methods and compositions related to glucocorticoid receptor antagonists and breast cancer
BR112012027745A2 (en) 2010-04-27 2017-01-10 Univ Johns Hopkins method and immunogenic composition for treatment of neoplasia
US8476238B2 (en) 2010-06-29 2013-07-02 Mycomagic Biotechnology Co., Ltd. Uses of an immunomodulatory protein (GMI) from Ganoderma microsporum
WO2012012772A2 (en) 2010-07-22 2012-01-26 The Johns Hopkins University Drug eluting hydrogels for catheter delivery
ES2385240B1 (en) 2010-07-26 2013-09-23 Gp-Pharm, S.A. CAPSULES OF ACTIVE PHARMACEUTICAL PRINCIPLES AND POLYINSATURATED FATTY ACIDS FOR THE TREATMENT OF PROSTATE DISEASES.
MX345788B (en) 2010-08-04 2017-02-15 Pellficure Pharmaceuticals Inc * Combination therapy for the treatment of prostate carcinoma.
WO2012027462A2 (en) 2010-08-24 2012-03-01 Steward St. Elizabeth's Medical Center Of Boston, Inc. Compositions and methods for treating neoplasia
SG189095A1 (en) 2010-09-27 2013-05-31 Zestagen S A Compositions and methods for treating neoplasia
US9090633B2 (en) 2010-11-18 2015-07-28 Kasina Laila Innova Pharmaceuticals Private Limited Substituted 4-(arylamino) selenophenopyrimidine compounds and methods of use thereof
WO2012066578A2 (en) 2010-11-18 2012-05-24 Kasina Laila Innova Pharmaceuticals Private Limited Substituted 4-(selenophen-2(or 3)-ylamino)pyrimidine compounds and methods of use thereof
WO2012074842A2 (en) 2010-11-23 2012-06-07 The United States Of America, As Represented By The Secretary, National Institute Of Health Compositions and methods for treating neoplasia
CN103313599B (en) 2010-12-09 2016-03-23 卡斯纳莱拉创新药物私人有限公司 Replace 4-(arylamino) selenophen and pyrimidine compound and using method thereof
US9314473B2 (en) 2011-02-03 2016-04-19 Pop Test Oncology Limited Liability Company System and method for diagnosis and treatment
JP5804395B2 (en) 2011-02-03 2015-11-04 ポップ テスト オンコロジー エルエルシー Systems and methods for diagnosis and treatment
WO2012116277A1 (en) 2011-02-25 2012-08-30 Arena Pharmaceuticals, Inc. Cannabinoid receptor modulators
ES2657750T3 (en) 2011-04-01 2018-03-06 Genentech, Inc. Combination of AKT inhibitor compound and vemurafenib for use in therapeutic treatments
AU2012236493A1 (en) 2011-04-01 2013-10-31 University Of Florida Research Foundation, Inc. Thermo-sensitive, mucoadhesive or dermoadhesive, and penetration-enhancing formulations for topical delivery of therapeutics
EP2508525A1 (en) 2011-04-05 2012-10-10 Bayer Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline salts
US20140248263A1 (en) 2011-04-08 2014-09-04 The University Of British Columbia Bisphenol compounds and methods for their use
WO2012142208A1 (en) 2011-04-13 2012-10-18 The Trustees Of The University Of Pennsylvania Bifunctional akr1c3 inhibitors/androgen receptor modulators and methods of use thereof
WO2012145328A1 (en) 2011-04-18 2012-10-26 The University Of British Columbia Dibenzylphenyl compounds and methods for their use
WO2012145330A1 (en) 2011-04-18 2012-10-26 The University Of British Columbia Fluorene-9-bisphenol compounds and methods for their use
JP2014513730A (en) 2011-05-17 2014-06-05 武田薬品工業株式会社 Pharmaceutical compositions and methods for treating cancer
US20120295878A1 (en) 2011-05-17 2012-11-22 Cristiano Ferlini Method for the treatment of cancer
US20140127316A1 (en) 2011-05-20 2014-05-08 New York University Propolis and caffeic acid phenethyl ester and uses thereof
CA2839301C (en) 2011-06-15 2021-02-16 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
EP2734207A4 (en) 2011-07-18 2015-06-17 Tokai Pharmaceuticals Inc Novel compositions and methods for treating prostate cancer
WO2013028572A1 (en) 2011-08-19 2013-02-28 British Columbia Cancer Agency Branch Fluorinated bisphenol ether compounds and methods for their use
FR2980972B1 (en) 2011-10-05 2014-02-28 Commissariat Energie Atomique FORMULATIONS FOR THE DIAGNOSIS AND TREATMENT OF HORMONO - PENDANT CANCERS AND CANCERS OF STEROIDIAN HORMONE SYNTHESIS.
WO2013053691A1 (en) 2011-10-10 2013-04-18 Zach System Process for preparing 17-substituted steroids
WO2013067131A1 (en) 2011-11-02 2013-05-10 Medivation Technologies, Inc. Treatment methods
WO2013071177A1 (en) 2011-11-10 2013-05-16 Tokai Pharmaceuticals, Inc. Methods and compositions for inhibition of androgen receptor activity
LT2785349T (en) 2011-11-30 2019-12-10 Astrazeneca Ab Combination treatment of cancer
EP2793896A4 (en) 2011-12-22 2015-06-24 Tokai Pharmaceuticals Inc Methods and compositions for combination therapy using p13k/mtor inhibitors
CN103254265B (en) 2012-02-21 2016-07-13 上海希迈医药科技有限公司 Abiraterone acetate trifluoroacetate and its preparation method and application
EP2817019A4 (en) 2012-02-24 2016-04-06 Univ Chicago Methods and compositions related to glucocorticoid receptor antagonism and prostate cancer
US20130251804A1 (en) 2012-03-21 2013-09-26 Emphascience, Inc. Method to improve the safety of handling of high potency drugs in solid dosage forms without changing their efficacy
US9464326B2 (en) 2012-03-22 2016-10-11 University Of Maryland, Baltimore Total and phosphorylated IL-1 receptor-associated kinase-1 and IL-1 receptor-associated kinase-4 as a biomarker for cancer progression and chemotherapy resistance
CN111420032A (en) 2012-03-29 2020-07-17 阿尔托生物科学有限公司 Method for treating tumors
WO2013152342A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Anti-cancer mtor inhibitor and anti-androgen combination
US20140079636A1 (en) 2012-04-16 2014-03-20 Dinesh U. Chimmanamada Targeted therapeutics
US9365510B2 (en) 2012-04-16 2016-06-14 British Columbia Cancer Agency Branch Aziridine bisphenol ethers and related compounds and methods for their use
WO2013159225A1 (en) 2012-04-23 2013-10-31 Alphora Research Inc. Process for preparation of 17-substituted steroids
WO2013166110A1 (en) 2012-05-02 2013-11-07 Yale University Tlr-agonist-conjugated antibody recruiting molecules (tlr_arms)
GB201207886D0 (en) 2012-05-04 2012-06-20 Jagotec Ag Improvements in or relating to organic compounds
JP2015518888A (en) 2012-06-06 2015-07-06 ノバルティス アーゲー Combination of a 17-alpha-hydroxylase (C17,20-lyase) inhibitor and a specific PI-3K inhibitor for the treatment of tumor diseases
EP2866795A4 (en) 2012-06-29 2016-01-27 Univ New York State Res Found Polyenolic zinc-binding agents (pezbins) actively promote inactivation of cancer stem cells and potentiate cytotoxic anti-tumor drug substances
WO2014009437A1 (en) 2012-07-11 2014-01-16 Sandoz Ag Oxidation stability of abiraterone acetate
WO2014009434A1 (en) 2012-07-11 2014-01-16 Sandoz Ag Self-microemulsifying drug delivery system of abiraterone or abiraterone acetate
WO2014016830A1 (en) 2012-07-25 2014-01-30 Mapi Pharma Ltd. Process and intermediates for the preparation of abiraterone acetate
WO2014047199A1 (en) 2012-09-19 2014-03-27 The Research Foundation For The State University Of New York Novel prodrugs for selective anticancer therapy
SG11201501850VA (en) 2012-09-21 2015-04-29 Intensity Therapeutics Inc Method of treating cancer
AU2013326463B2 (en) 2012-10-04 2018-01-18 Ab Science Use of masitinib for treatment of cancer in patient subpopulations identified using predictor factors
WO2014058785A1 (en) 2012-10-10 2014-04-17 Novartis Ag Combination therapy
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
US20160199298A1 (en) 2012-10-15 2016-07-14 Chamaeleo Pharma Bvba Fosfestrol for use in curative or palliative treatment of cancer in female mammals
US20150283155A1 (en) 2012-10-15 2015-10-08 Chamaeleo Pharma Bvba Fosfestrol for use in curative or palliative treatment of prostate cancer
EP2917225A1 (en) 2012-11-09 2015-09-16 Synthon BV Process for making abiraterone-3-acetate
EP2934545A1 (en) 2012-12-19 2015-10-28 Bayer Pharma Aktiengesellschaft Combination comprising radium-223 for the treatment of cancer
KR101586789B1 (en) 2012-12-28 2016-01-19 주식회사 종근당 Sustained-release lipid pre-concentrate of cationic pharmacologically active substance and pharmaceutical composition comprising the same
JOP20200097A1 (en) 2013-01-15 2017-06-16 Aragon Pharmaceuticals Inc Androgen receptor modulator and uses thereof
US20140255413A1 (en) 2013-03-07 2014-09-11 Boehringer Ingelheim International Gmbh Combination therapy for neoplasia treatment
AU2014232508C1 (en) 2013-03-15 2023-01-19 Sun Pharmaceutical Industries Limited Abiraterone acetate formulation
US20150246060A1 (en) 2013-03-15 2015-09-03 Iceutica Inc. Abiraterone Acetate Formulation and Methods of Use
EP2792682B1 (en) 2013-04-19 2016-11-09 Zach System Process for producing a solid form of abiraterone acetate
US9511078B2 (en) 2013-06-19 2016-12-06 Kashiv Pharma, Llc Self-nanoemulsion of poorly soluble drugs
CZ306623B6 (en) 2013-07-04 2017-04-05 Zentiva, K.S. Salts of abiraterone acetate
CA2914841C (en) 2013-08-01 2021-11-02 Mw Encap Limited Compositions and preparation methods of low melting ionic salts of poorly-water soluble drugs
EP3034076B1 (en) 2013-08-13 2020-09-30 JC (Wuxi) COMPANY, Inc. Combined application of isothiocyanate compound and anti-cancer medicine
WO2015032873A1 (en) 2013-09-06 2015-03-12 Synthon B.V. High-load pharmaceutical compositions comprising abiraterone acetate
JP6548641B2 (en) 2013-10-28 2019-07-24 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Treatment of metastatic prostate cancer
WO2015114314A1 (en) 2014-01-28 2015-08-06 Cipla Limited Pharmaceutical composition comprising abiraterone
MX2016016173A (en) 2014-06-19 2017-03-28 Solural Pharma ApS Solid oral dosage form of lipophilic compounds.
WO2015200837A1 (en) 2014-06-27 2015-12-30 Fl Therapeutics Llc Abiraterone derivatives and non-covalent complexes with albumin
EP3160450A1 (en) 2014-06-30 2017-05-03 Galenicum Health S.L. Stable pharmaceutical compositions in the form of immediate release tablets
MD20170048A2 (en) 2014-09-18 2017-08-31 Iceutica Inc. Abiraterone acetate formulation and methods of use
HUP1500055A1 (en) 2015-02-09 2016-08-29 Druggability Technologies Ip Holdco Ltd Complexes of abiraterone acetate, process for the preparation thereof and pharmaceutical compositions containing them
CN104710499A (en) 2015-02-15 2015-06-17 重庆医药工业研究院有限责任公司 Crystal form of Abiraterone propionate and preparation method thereof
GB201514760D0 (en) 2015-08-19 2015-09-30 Karus Therapeutics Ltd Compounds and method of use
WO2017037647A1 (en) 2015-09-02 2017-03-09 Leiutis Pharmaceuticals Pvt Ltd Stable pharmaceutical compositions of abiraterone
PL3357513T3 (en) 2015-09-29 2024-04-02 Kangpu Biopharmaceuticals, Ltd. Pharmaceutical composition and application thereof
WO2017120218A1 (en) 2016-01-04 2017-07-13 The Regents Of The University Of Colorado, A Body Corporate Treatment of prostate cancer cells with fat oxidation inhibitors and enzalutamide
TWI726969B (en) 2016-01-11 2021-05-11 比利時商健生藥品公司 Substituted thiohydantoin derivatives as androgen receptor antagonists

Also Published As

Publication number Publication date
EP3280448A1 (en) 2018-02-14
US10722527B2 (en) 2020-07-28
WO2016162229A1 (en) 2016-10-13
EP3824908A1 (en) 2021-05-26
ES2863500T3 (en) 2021-10-11
US20180125863A1 (en) 2018-05-10
EP3280448B1 (en) 2020-12-30

Similar Documents

Publication Publication Date Title
US20200316093A1 (en) Abiraterone acetate lipid formulations
CA2618705C (en) Improved delivery of tetrahydrocannabinol
US9265724B2 (en) Oral dosage form of tetrahydrocannabinol and a method of avoiding and/or suppressing hepatic first pass metabolism via targeted chylomicron/lipoprotein delivery
US9241940B2 (en) Pharmaceutical composition comprising nanocrystals
US20160184258A1 (en) Oral gastrointestinal dosage form delivery system of cannabinoids and/or standardized marijuana extracts
US20060115527A1 (en) Controlled release preparation
US20070190080A1 (en) Non-aqueous compositions for oral delivery of insoluble bioactive agents
US20140357708A1 (en) Oral dosage form of tetrahydrocannabinol and a method of avoiding and/or suppressing hepatic first pass metabolism via targeted chylomicron/lipoprotein delivery
WO2016022936A1 (en) An improved oral gastrointestinal dosage form delivery system of cannabinoids and/or standardized marijuana extracts
Bansal et al. Solid self-nanoemulsifying delivery systems as a platform technology for formulation of poorly soluble drugs
CN102307576A (en) Methods for enhancing the release and absorption of water insoluble active agents
Goyal et al. Self microemulsifying drug delivery system: A method for enhancement of bioavailability
TW201444586A (en) Emulsion formulations
AU2016203127B2 (en) An improved oral gastrointestinal dosage form delivery system of cannabinoids and/or standardized marijuana extracts
EP1427392B1 (en) New self emulsifying drug delivery system
Parmar et al. SMEDDS: A dominant dosage form which improve bioavailability
AU2013213706B2 (en) An improved oral dosage form of tetrahydrocannabinol and a method of avoiding and/or suppressing hepatic first pass metabolism via targeted chylomicron/lipoprotein delivery
CN111093637A (en) Pharmaceutical composition
Kovačević Lipid nanocarriers for delivery of poorly soluble and poorly permeable drugs
Cannon et al. Emulsions, microemulsions, and lipid-based drug delivery systems for drug solubilization and delivery—Part II: Oral applications
Kazi Lipid‐based nano‐delivery for oral administration of poorly water soluble drugs (PWSDs): design, optimization and in-vitro assessment
Aldosari Development and evaluation of self-nanoemulsifying drug delivery systems for oral delivery of indomethacin
Sri et al. A review on SEDDS and Solid SEDDS: an emerging dosage forms for poorly soluble drugs
Cannon et al. 11 Emulsions, Microemulsions, and Lipid-Based Drug Delivery Systems for Drug Solubilization and Delivery—Part II: Oral Applications
Singh et al. Journal of Drug Discovery and Therapeutics 2 (17) 2014, 01-12

Legal Events

Date Code Title Description
AS Assignment

Owner name: CAPSUGEL BELGIUM NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WILLIAMS, HYWEL;AGARWAL, PRASHANT;JULE, EDUARDO;SIGNING DATES FROM 20171121 TO 20180118;REEL/FRAME:053128/0194

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE