WO2014120922A1 - Porous silica gel as a carrier for liquid technologies - Google Patents

Porous silica gel as a carrier for liquid technologies Download PDF

Info

Publication number
WO2014120922A1
WO2014120922A1 PCT/US2014/013848 US2014013848W WO2014120922A1 WO 2014120922 A1 WO2014120922 A1 WO 2014120922A1 US 2014013848 W US2014013848 W US 2014013848W WO 2014120922 A1 WO2014120922 A1 WO 2014120922A1
Authority
WO
WIPO (PCT)
Prior art keywords
inorganic oxide
composition according
oxide particles
active ingredient
biologically active
Prior art date
Application number
PCT/US2014/013848
Other languages
French (fr)
Inventor
Frederik Hendrik MONSUUR
Original Assignee
W. R. Grace & Co.-Conn.
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 W. R. Grace & Co.-Conn. filed Critical W. R. Grace & Co.-Conn.
Priority to KR1020157023665A priority Critical patent/KR102229036B1/en
Priority to US14/764,363 priority patent/US10660856B2/en
Priority to CN201480019478.7A priority patent/CN105102117A/en
Priority to PL14746507T priority patent/PL2950923T3/en
Priority to ES14746507T priority patent/ES2908958T3/en
Priority to EP14746507.4A priority patent/EP2950923B1/en
Priority to JP2015556128A priority patent/JP6513031B2/en
Publication of WO2014120922A1 publication Critical patent/WO2014120922A1/en
Priority to HK16105942.0A priority patent/HK1217927A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate 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
    • A61K9/143Intimate 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 with inorganic compounds
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/02Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material
    • B01J20/10Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/02Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material
    • B01J20/10Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate
    • B01J20/103Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate comprising silica
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28002Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their physical properties
    • B01J20/28004Sorbent size or size distribution, e.g. particle size
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28054Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their surface properties or porosity
    • B01J20/28057Surface area, e.g. B.E.T specific surface area
    • B01J20/28059Surface area, e.g. B.E.T specific surface area being less than 100 m2/g
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28054Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their surface properties or porosity
    • B01J20/28069Pore volume, e.g. total pore volume, mesopore volume, micropore volume
    • B01J20/28073Pore volume, e.g. total pore volume, mesopore volume, micropore volume being in the range 0.5-1.0 ml/g
    • 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
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01BNON-METALLIC ELEMENTS; COMPOUNDS THEREOF; METALLOIDS OR COMPOUNDS THEREOF NOT COVERED BY SUBCLASS C01C
    • C01B33/00Silicon; Compounds thereof
    • C01B33/113Silicon oxides; Hydrates thereof
    • C01B33/12Silica; Hydrates thereof, e.g. lepidoic silicic acid
    • C01B33/18Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof
    • C01B33/187Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof by acidic treatment of silicates
    • C01B33/193Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof by acidic treatment of silicates of aqueous solutions of silicates

Abstract

Compositions containing a biologically active ingredient and an inorganic oxide material are disclosed. Methods of making and using compositions containing a biologically active ingredient and an inorganic oxide material are also disclosed. The present invention relates to compositions comprising inorganic oxide porous material containing a biologically active ingredient in liquid form, methods of making such compositions, and methods of using them.

Description

POROUS SILICA GEL AS A CARRIER FOR LIQUID TECHNOLOGIES
TECHNICAL. FIELD
[0001] The present invention relates to compositions comprising inorganic oxide porous material containing a biologically active ingredient in liquid form, methods of making such compositions; and methods of using them.
BACKGROUND
[0002] The oral route remains the preferred route of drug administration due to its convenience and good patient compliance. Major problems in oral drug formulations are the erratic and incomplete absorption throughout the gastrointestinal (GI) tract, resulting in low and variable bioavailabilit ' and Sack of dose proportionality. These problems mainly result from poor aqueous solubility of the active ingredient, it has been reported that an estimated 40% of existing pharmaceutical active ingredients and an even higher proportion of all newly developed drugs are poorly soiubie or insoluble in water. This poses a major challenge to drug development, as there is a high need for producing suitable formulations to improve the solubility and bioavailability of such drugs.
[0003] Much research has been conducted into methods to cope with these problems.
Methods that have been developed include the reduction of particle size of the drug by micronisation or nanonisaiion as to increase surface area, thereby increasing dissolution rate of the active ingredient. Further methods include solubilization in surfactant systems, water- soluble molecular complexes with cyelodextrins, converting the drug in amorphous form by lyophilization or formation of solid dispersions in hydrophiiie carriers, microencapsulation, and the release from porous carrier materials.
[0004] One technique for promoting dissolution properties and oral bioavailability of poorly water-soluble drugs is by using them in liquid phase by dissolution or emulsion in non-volatile oils/ lipids. Such systems have been referred to as Hpid based drug delivery system (LBDDS). In these forms, the active ingredient is already in solution so that the drug is present on molecular level, avoiding the dissolution step from the crystalline state. The drugs in liquid phase are typically fdled into soft gelatin capsulees. The latter give rise to drawbacks, such as complications in manufacturing, low manageability and portability, risks of leakage, limited shelf-life due to stability problems during storage caused b interactions between the components, oxidation of the lipid components, issues of compatibility of the liquid formulation with the capsulee shell, critieality of storage temperature because of irreversible drugs excipients precipitation at lower temperatures.
[0005] To overcome these problems, so-called iiquisoiid formulations have been developed, which are porous carrier materials wherein the drugs remain in liquid form. Liquisolid forms are obtained by conversion of drugs in liquid form into acceptably flowing non-adherent and compressible powder mixtures by blending with selected carriers and coating materials. These then are converted into solid dosage forms such as tablets, pellets, and capsulees.
[0006] Due to increased wetting and surface area for dissolution, Iiquisoiid dosage forms of water insoluble drugs show improved dissolution properties and bioavailability. This technique was successfully applied for low dose water-insoluble drugs. However, as loadability and release of the drugs front the carriers used is limited, formulation of insoluble drags at higber doses is one of the limitations of the Iiquisoiid technique. Another problem associated with iiquisoiid formulations is their decreased flowability when loaded with higher amounts of drugs in liquid form. This causes these materials difficult to process in pharmaceutical manufacturing. In order to have acceptable flowability and compactabihty, high levels of carrier and coating materials have to be added thereby increasing the weight and volume of the resulting dosage forms,
[0007] One type of lipid based drug delivery systems are the self-emulsifying drug delivery systems (SEDDS). This type of emulsion-based drug formulations can be used in soft gelatin capsulees or as Iiquisoiid formulations. SEDDS are isotropic and thermodynamically stable mixtures of drug, oil lipid, surfactant/cosurfactant, thai, in contact with aqueous fluids, spontaneously form oil-in-water emulsions of droplets, ranging in droplet size approximately between 100-300 nrn. Systems forming emulsions with droplets of less than 50 nm are referred to as self-micro-emulsifying drug delivery systems (SMEDDS), and even smaller droplet sizes as seif-nanoemulsifying drug delivery system (SNEDDS). Self-emulsifying formulations spread readily in the gastrointestinal (Gi) tract, where the digestive motility of the stomach and the intestine provide the agitation necessary for selfemulsification. These systems advantageously present the drug in dissolved form and the small droplet size provides a large interfacial area for the drug absorption. When compared with emulsions, which are sensitive and metastabie dispersed forms, SEDDSs are physically stable formulations that are easy to manufacture. In particular for lipophilic drugs thai exhibit, limited and distorted absorption, these systems offer an improvement in the rate and extent of absorption resulting in more reproducible bioavailability.
[0008] Given the advantages of solid dosage forms, SEDDS, SNEDDS and SMEDDS have also been converted into solid-SEDDS, solid-SNEDDS or soHd-SMEDDS (S-SEDDS, S-5MEDDS or S-SMEDDS) using liquisoiid solidification procedures similar as described above. The resulting solid formulations in turn can be worked into various solid or semisolid dosage forms (tablets, pellets, capsules, creams, transdermal systems, etc).
SUMMARY
[0009] It is an object of this invention to provide further carriers for use in the release of active ingredients of various nature. It is a further object of this invention to provide carriers that allow higher drug loading compared to known systems, showing desired dissolution release profiles and concomitant oral bioavaiiabiUy characteristics. It is also an object of this invention to provide compact liquisoiid dosage forms of high dose water- insoluble drugs, which are of acceptable size to patients. It is an object of this invention to provide liquisoiid materials with optimum properties such as flow and bulk density. Still a further object concerns the provision of liquisoiid formulations that have good ffowabiliry characteristics and can be easily processed in pharmaceutical manufacturing.
[0010] One or more of these objects and other advantages are attained by the various aspects and embodiments of the present invention.
[001 1 ] in one embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume, as measured by nitrogen porosirnetry, of about 0.5 cm3/g or greater; wherein the inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1, or 1.6: 1 , or 1.7: 1 , or 1.8: 1, or 1.9: 1, up to 2: 1.
[0012] in another embodiment, the present invention relates to a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnr' g or greater; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1.5 : i , or 1.6: 1 , or 1.7: 1 , or 1.8: 1 , or 1.9: 1 , up to 2: 1.
[0013] In an even further embodiment, the present invention relates to a method of making composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/ 100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 em3/g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 ,5: 1 , or 3 ,6: 1, or 1.7: 1, or 1 .8: 1. or 1 .9: 1 , up to 2: 1.
[0014] In another embodiment, the present invention concerns a method of making a pharmaceutical and/or cosmetic composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnv'/g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1, or 1 ,6: 1 , or 1 .7: 1 , or 1 ,8: 1 , or 1 ,9: 1 , or at least 2: 1 .
[0015] in a further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 m 1/100 g; (b) pores having a pore volume, as measured by nitrogen porcsmietry, of about 0.5 cm3/g or greater; and (c) a pore size distribution having a relative span of about 1.5 or less, or about 1.4 or less, or about 1 ,3 or less, or about 1.2 or less, or about 1.1 or less, or about 1 .0 or less,
[0016] In another embodiment, the present invention concerns a composition comprising porous Inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cm'Vg or greater; and (c) a median pore size of 5 nm to 30 tint, wherein said inorganic oxide particles remain free flowing at a ratio of Hquid material to inorganic oxide particles of at least 1 : 1, or 1.3, or 1 ,5: 1, or 1 ,6: 1 , or 1.7: 1, or 1.8: 1 , or 1.9: 1, or at least 2: 1.
[0017] In an even further embodiment the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm " g or greater; and (c) a median particle size of from 3 microns to 10 mm; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1 , or 1 ,3, or 1.5: 1 , or 1 ,6: 1 , or 1.7: 1 , or 1.8: 1. or 1.9: 1 , or at least 2: 1.
[0018] fn another embodiment the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnrVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, decreases in volume of at least about 15% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40% after resting.
[0019] In a further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cnrVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, increases in bulk density by at least about 35% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40% after resting.
[0020] in another embodiment, the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cm'Vg or greater; wherein, after mixing the inorganic oxide particles and liquid material and then resting for at least 2 hours, at least about 400 mg of said composition may be loaded into a zero size capsule. In another embodiment at least about 410 mg, or at least about 420 trig, or at least about 430 rug of said composition may be loaded into a zero s e capsule.
[0021 ] In an even further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 enr/g or greater; and (c) a ratio of liquid material to inorganic oxide particles of at least 1 : 1 ; wherein at least 65% of the liquid material is desorbed from the particles upon desorption.
[0022] In one embodiment the inorganic oxide material comprises porous particles comprising pores having a mean pore diameter in the range of about 5 nm to about 30 nm.
[0023] In another embodiment, the porous inorganic oxide material containing a biologically active ingredient in a liquid material is in the form of particles, i.e. particles of porous inorganic oxide material containing a biologically active ingredient in liquid form. The average diameter of the particles of porous inorganic oxide material of the invention may be in the range of from about 3μηι to about 5 mm.
[0024] The compositions of the invention may contain further liquid organic auxiliary materials such as oils, non-volatile solvents, and surfactants.
[0025] In one embodiment, the particles of porous inorganic oxide material containing a biologically active ingredient in a Hquid material of the invention form a powder that is free-flowing. In a further embodiment, the powder has a Carr index of equal or lower than 25, or the powder has a Hausner index of about 1 to about 1 ,4. In one embodiment, the angle of repose of said powder is from about 30i! to about 45¾, These properties are measured before and after loading the biologically active ingredient on the porous inorganic oxide material.
[0026] In further embodiment, the composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material has a bulk bulk density, after loading and resting during at least 2 hours, of at least 450 g/1.
[0027] Upon loading with a biologically active ingredient in a liquid material, the resulting loaded porous Inorganic oxide material may show a limited (e.g. up to about 10%) or no increase, but in particular shows a decrease in volume when compared to the unloaded inorganic oxide porous material In one embodiment the decrease is up to about 30%, or up to about 20%, or up to about 10% (each % in this paragraph being volume/volume or v/v). Each of the changes in volume and/or density mentioned herein are after loadsng and resting up to 2 hours depending upon the liquid, and even up to 24 or 48 hours. In one embodiment, upon loading the inorganic oxide porous material with biologically active ingredient in a liquid material, a decrease of the volume or increase in density, in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs Is observed, after which time period no further decrease occurs and the volume stays substantially constant.
[0028] The compositions of the present invention advantageously do not .show the substantial increase of volume that is typically observed when loading art-known silicas with a biologically active ingredient in a liquid material This allows the manufacture of more compact dosage forms, which in turn contributes to better processabllity during pharmaceutical manufacturing and to better acceptability of the dosage forms by patients,
[0029] In a further aspect, the present invention concerns a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic o ide material has the oil adsorption, the pore volume, and BET surface area, as specified herein, in one embodiment the said composition comprising inorganic oxide porous material containing a biologically active ingredient is in the form of particles.
[0030] The present invention is further directed to methods of making the disclosed compositions, in one embodiment, the method of making a composition of the present invention comprises incorporating at least one biologically active ingredient in a liquid material into the porous inorganic oxide material having the oil adsorption, the pore volume, and BE T surface area, as specified herein.
[0031 ] In a further aspect, the present invention concerns a method of making a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic oxide material has the oil adsorption, the pore volume, and BET surface area, as specified herein, said method comprising combining said pharmaceutical dosage formulating ingredient with said composition.
[0032] The present invention is also directed to methods of using the disclosed compositions, in one embodiment, the method of using a composition of the present invention comprises administering a composition of the invention to a patient so as to deliver a biologically active material to the patient, wherein the composition comprises a porous inorganic oxide material containing a biologically active ingredient n a liquid material, wherein the inorganic oxide material has the oil adsorption, the pore volume, and BET surface area, as specified herein. The composition that is administered in particular is a pharmaceutical dosage form.
[0033] In one embodiment the said inorganic oxide material is in the form of particles, in particular particles havin an average diameter as further specified herein.
[0034] These and other features and advantages of the present invention will become apparent after a review of the following detailed description of the disclosed embodiments and the appended claims.
BRIEF DESCRIPTION OF THE DRAWINGS
[0035] The present invention is further described with reference to the appended figures, wherein:
[0036] FIG. 1 graphically displays the kinetic behaviour or volume change over time of an esemplasy composition of the present invention,
[0037] FIG. 2 graphically displays capsule loading capacity change before and after resting of an exemplary composition of the present Invention according to Example 4.
[0038] FIG. 3 graphically displays a release profile of acetaminophen from an exemplary composition of the present invention according to Example 6.
[0039] FIG. 4 graphically displays a release profile of ascorbic acid from an exemplary composition of the present, invention according to Example 6.
[0040] FIG, 5 graphically displays the release profile Gyburide from a solid SEDDS system of the present invention according to Example 8. DETAILED DESCRIPTION
[0041 ] To promote an understanding of the principles of the present invention, descriptions of specific embodiments of the invention follow and specific language is used to describe the specific embodiments. It will nevertheless be understood that no limitation of the scope of the inventio is intended by the use of specific language. Alterations, further modifications, and such further applications of the principles of the present invention discussed are contemplated as would normally occur to one ordinarily skilled in the art to which the invention pertains.
[0042] It must be noted that as used herein and in the appended claims, the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an oxide" includes a plurality of such oxides and reference to "oxide" includes reference to one or more oxides and equivalents thereof known to those skilled in the art. and so forth,
[0043] "About" modifying, for example, the quantity of an ingredient in a composition, concentrations, volumes, process temperatures, process times, recoveries or yields, flow rates, and like values, and ranges thereof, employed in describing the embodiments of the disclosure, refers to variation in the numerical quantity that may occur, for example, through typical measuring and handling procedures; through inadvertent error in these procedures; through differences in the ingredients used to carry out the methods; and like proximate considerations. The term "about" also encompasses amounts that differ due to aging of a formulation with a particular initial concentration or mixture, and amounts that differ due to mixing or processing a formulation with a particular initial concentration or mixture. Whether modified by the te m "about" the claims appended hereto include equivalents to these quantities.
[0044] As used herein, the term "biologically active ingredient" means an active pharmaceutical ingredient (API), which provides a pharmacological activity or otherwise have direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease, or to have direct effect In restoring, correcting or modifying physiological functions in humans. Even though this includes poorly soluble material, it may also include materials that range in solubility, including those listed in the BCS (Biopharmaceutic Classification System), which is a classification approach where drags (APIs) are divided into four classes based on the extent (high or low) of their aqueous solubility and permeability through the GI tract wail in particular intestinal. In this regard, these four classes are: (Group I) High Solubility and High Permeability drags, (Group Π) Low Solubility and High Permeability drugs, (Group ΙΠ) High Solubility and Low Permeability drugs and, (Group IV) Low solubility and Low Permeability drugs.
[0045] As used herein, the term "bulk density" means the mass of particulate matter, such as a powder, divided by the total volume occupied by the matter, and includes mtraparticle pore volume and interpariicle void volume,
[0046] As used herein, the term "free flowing" means the ability of a group of particles or a powder to move when a force (e.g., gravity, or other external force) is applied to it. Commonly used tests for measuring powder flow include the Hausner ratio, compressibility index (Carr index) or angle of repose. The compressibility index (Can index) and the Hausner ratio are determined by measurin both the bulk volume and the tapped volume of a powder.
[0047] As used herein, "inorganic oxides" is defined as binary oxygen compounds where the inorganic component is the cation and the oxide is the anion. The inorganic material includes metals may also include metalloids. Metals include those elements on the left of the diagonal line drawn from boron to polonium on the periodic table. Metalloids or semi-metals include those elements that are on the right of this line. Examples of inorganic oxides include silica, alumina, titania, zirconia, etc., and mixtures thereof.
[0048] As used herein, the term "intraparticle pore volume" means pore volume attributable to the spaces in the pore of the particles, as compared to interparticle pore volume, which is the pore volume attributable to the spaces between the particles (i.e., the interstitial space).
[0049] As used herein, the term "lipid materia!" or "lipid component" means organic materials comprising fatty acids and their derivatives, and substances related biosyntheticaiiy or functionally to these compounds. They include, but are not limited to, molecules that originate entirely or in part by carban ion-based condensations of thioesters (fatty acids, po!yketides, etc) and/or by carocation-based condensations of isoprene units (preno!s, sterols, etc.).
[0050] As used herein, the term "ordered porous material''' refers to porous particles that have an interna! structural order such that they possess a low angle X-ray diffraction patterns according to Bragg's Law. Such materials include ordered mesoporous silica, for example, MCM-41 , SBA- 15, TUD-1 , HMM-33 and FSM- 56,
! O [0051 ] As used herein, the term "non-ordered porous material" refers to porous particles possessing an internal structure such that they do not have a low angle X-ray diffraction pattern according to Bragg' s Law, Such materials may be formed via any known process including, but not limited to, a solution polymerization process such as for forming colloidal particles, a continuous flame hydrolysis technique such as for forming fused particles, a gel technique such as for forming gelled particles, and a precipitation technique such as for forming precipitated particles. The particles may be subsequently modified by autoclaving, flash drying, super critical fluid extracting, etching, or like processes. The particles may be composed of organic and/or inorganic materials and combinations thereof, in one exemplar}' embodiment the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides. The particles may be a variety of different symmetrical, asymmetrical or irregular shapes, including chain, rod or lath shape. The particles may have different structures including amorphous or crystalline, etc. The particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures, or that may be the same except for different surface treatments, Porosiiy of the particles may be intraparticle or interparticle in cases where smaller particles are agglomerated to form larger particles. In one exemplary embodiment the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides. Porous materials include organic and inorganic materials, or hybrids thereof, and may be in the form of particles, monoliths, membranes, coatings, and the like.
[0052] As used herein, the term "pore size distribution" means the relative abundance of each pore size in a representative volume of porous inorganic particles. As used herein "median pore size" is the pore diameter below which 50% of the intraparticle pore volume resides.
[0053] As used herein, the term "relative span" is defined as meaning a measure of the breadth of pore size distribution. The "span" is measured by subtracting the d¾o pore size (i.e., the pore size/diameter below which 10% of the pore volume resides) from the d¾¾ pore size (i.e., the pore size/diameter below which 90% by pore volume resides) as measured by mercury porosimetry. The term "relative span" is defined as the ratio of (d¾rdto)/dso. he span and relative span are determined using nitrogen sorption (BJH method) of the cumulative pore volume. [0054] As used herein the term "rested'' or "after resting" is used to indicate a period of time wherein the porous inorganic oxide materia! is allowed to stand undisturbed after loading with a biologically active ingredient in a liquid material.
[0055] Whenever used herein in relation to a ratio or a percentage of components, w/w means weight/weight and w/v means weight/volume.
[0056] The present invention is directed to compositions comprising a biologically active material or ingredient and an inorganic oxide material, wherein the inorganic oxide material comprises porous particles. Efficient loading of biologically active materials in liquids or a liquid material onto vehicles for drug delivery is a concern for many biologically active materials and this invention relates to various embodiments that provide solutions to this problem, Applicants of the present invention have found that certain porous inorganic oxide materials having a specific sets of physical properties provide exceptional liquid loading and release of biologically active materials that are in various liquids or a liquid material
[0057] in one embodiment, the present invention reiates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particies possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (h) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm¾ or greater; wherein the inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 ,5: 1 , or 1 .6:1 , or 1 ,7: 1 , or 1.8: 1, or 1.9: 1 , up to 2: 1.
[0058] In an even further embodiment the present invention relates to a method of making composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume* as measured by nitrogen porosimetry, of about 0,5 enrVg or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 .5: 1 , or 1.6: 1 , or 1.7: 1 , or 1 ,8: 1, or 1 ,9: 1, up to 2: 1.
[0059] In another embodiment, the present" invention concerns a method of making a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particies containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi 100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cmJ g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1 , or 1.6: 1, or 1.7: 1, or 1.8: 3 , or 1.9: 1, or at least 2: 1.
[0060] In another embodiment, the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 300 to about 500 ml/1 GO g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm3/g or greater; and (e) a median pore size of 5 nm to 30 nm, wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 3 : 1 , or 13, or 1 ,5: 1, or 1.6: 1 , or 1.7: 1 , or 1 .8: 1 , or 1 ,9: 1 , or at least 2: 1.
[0061 ] In an even further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/ 100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm Vg or greater; and (c) a median particle size of from 3 microns to 10 mm; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1 , or 1 ,3. or 1 .5: 1 , or 1.6: 1 , or 1 ,7: 1, or 1.8: 1 , or 1.9: L or at least 2: 1 .
[0062] In a further embodiment the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 eirf/g or greater; and (c) a pore size distribution where at least 0.5 cm3/g of pore volume are from pores ranging from 10 nm to 30 nm, at least 0.5 enrVg, at least 0.6 cmJ/g , at least 0.8 cnrVg* at least 1.0 cm Vg, at least 1 ,2 cmVg, at least 1.4 cm'Vg, or at least 1.6 cm 7g of pore volume arc from pores ranging from 10 nm to 30 nm,
[0063] In a further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm3/g or greater ; and (c) a pore size distribution having a relative span of about 1.5 or less, or about 1 ,4 or less, or about 1.3 or less, or about 1.2 or less, or about 1.1 or less, or about 1.0 or less.
[0064] The porous inorganic oxide material containing a biologically active ingredient in a liquid material may be in the form of particles, which may have a variety of different symmetrical, asymmetrical or irregular shapes, including chain, rod or lath shape. The particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures,
[0065] The average diameter of the particles of the porous inorganic oxide material of the invention may be in the range of from about 3 pm to about 5 mm, preferably from about 50 μίτ< (or about 44 μηι) to about 500 pm; or from about 70 pm (or about 74 pm) to about 200 μχη; or from about 50 pm (or about 44 μτ ) to about 350 pm (or about 149 pm); or from about 50 pm (or about 44 pm) to about 150 pm (or about 149 μπή: or from about 100 μπ\ (or about 105 μΐϊί) to about 120 μιη (or about 125 μιπ), or from about 48 pm (or about 44 pm) to about 65 μπι (or about 63 pm), or from about 90 μη\ to about 130 μπι. The average diameters disclosed herein preferably are determined by Malvern instrumentation. The desired particle sizes can be obtained by milling and subsequent mesh sieving,
[0066] The porous inorganic oxide materials may be formed via any known process including a solution polymerization process such as for forming colloidal particles, a continuous flame hydrolysis technique such as for forming fused particles, a gel technique such as for forming gelled particles, and a precipitation technique such as for forming precipitated particles. The particles may be subsequently modified by autociaving, flash drying, super critical fluid extracting, etching, or like processes. In one embodiment the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides. The particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures, or that may be the same except for different surface treatments,
[0067] Porous materials include organic and inorganic materials, or hybrids thereof and may be in the form of particles, monoliths, membranes, coatings, and the like.
[0068] The porous particles may be in various forms, such as precipitates, gels, fumed, colloidal, etc, and combinations thereof, unmodified or modified by subsequent processes, such as autociaving, super critical fluid extraction, flash drying, and the like, in one embodiment, porous inorganic oxide material that is suitable for use in the present invention includes precipitated inorganic oxide particles and inorganic oxide gel particles. These inorganic oxides are referred to herein as "parent inorganic oxides", "parent particles", or "parent dispersions".
[0069] In another embodiment, the porous inorganic oxide materials are non-ordered, and may further possess random intraparticle porosity. Such materials have been found, when combined with the biologically active ingredient and liquid material, to provide beneficial adsorption and desorpiion (and dissolution) characteristics. Even though any inorganic oxide composition may be suitable for use in this invention (e.g., Si02, AI2O3, AIPO4, MgO, TiOz, ZrO? etc.), one embodiment of the present invention includes amorphous precipitated silica and siliea gel. The inorganic oxides may also include mixed inorganic oxides such as SiC A^Os, MgO.SiO2.AbO3 and the like. Mixed inorganic oxides are prepared by conventional blending or coge!ling procedures, in embodiments comprising gels, the dispersions are derived from porous inorganic oxide geis such as gels comprising Si02, A !;.(),, A I FO4, MgO, TiOj, and ZrQ2, The gels can be hydrogeis, aerogels, or xerogels.
[0070] In one embodiment, the inorganic oxide gels include a non-ordered porous silica gel that includes interpartiele pore volume. Such a silica gel may be prepared by mixing an aqueous solution of an alkali metal silicate (e.g., sodium silicate) with a strong acid such as nitric or sulfuric acid, the mixing being done under suitable conditions of agitation to form a clear silica sol which sets into a hydrogel, i.e., macroge!, in less than about one-half hour. The resulting gel is then washed. The concentration of inorganic oxide, i.e., SiC ., formed in the hydrogel is usually in the range of about 10 and about 50, or between about 20 and about 35, or between about 30 and about 35 weight percent, with the pH of that gel being from about 1 to about 9, or 1 to about 4. A wide range of mixing temperatures can be employed, this range being typically from about 20 to about 50ν C. The newly formed hydrogeis are washed simply by immersion in a continuously moving stream of water, which leaches out the undesirable salts, leaving about 99.5 weight percent or more pure inorganic oxide behind. The pH, temperature, and duration of the washing will influence the physical properties of the siliea, such as surface area (SA) and pore volume (PV). Silica gei washed at 65-90" C. at pH's of 8-9 for 15-36 hours will usually have SA's of 250-400 and form aerogels with PV's of 1 A to 1.7 em5 g. [0071 ] Methods for preparing inorganic oxide gels such as alumina and mixed inorganic oxide gels such as siiiea/alismma cogels are also well known in the art, such as by conventional blending, co-gelation, co-precipitation, and the like. Methods for preparing such geis have been described in US 4,226,743. in general, alumina gels are prepared by mixing alkali rnetai aluminaies and aluminum sulfate, Cogels are prepared by cogeliing two or more metal oxides so that the geis are composited together. For example, silica alumina cogels can be prepared by gelling an alkali metal silicate with an acid or acid salt, and then adding alkali metal aluminate, aging the mixture and subsequently adding aluminum sulfate. The gel is then washed using conventional techniques. Another embodiment of this invention is derived from dispersions of certain precipitated inorganic oxides. Reinforced precipitated silica such as that described in US 4,157,920 can also be used to prepare the dispersion of this invention. For example, reinforced precipitated silicas can be prepared by first acidulating an alkali inorganic silicate to create an initial precipitate. The resulting precipitate is then reinforced or "post conditioned" by additional silicate and acid. The precipitate resulting from the second addition of silicate and acid comprises 10 to 70% by weight of the precipitate initially prepared. It is believed that the reinforced structure of this precipitate is more rigid than conventional precipitates as a result of the second precipitation, Once an inorganic oxide is selected for the parent dispersion, a liquid phase of the selected inorganic oxide is prepared. In general, the parent dispersion should be in a state that can be wet milled. The medium for the liquid phase can be aqueous or non-aqueous, e.g., organic. The liquid phase can he residual water in inorganic oxide gels which have been drained, but not yet dried, and to which additional water is added to reslurry the gel.
[0072] In another embodiment, dried inorganic oxides, e.g., xerogels, are dispersed in liquid medium. In some embodiments, the parent dispersion is then milled. The milling is conducted "wet", i.e., in liquid media. The general milling conditions can vary depending on the feed material, residence time, impeller speeds, and milling media particle size. The techniques for selecting and modifying these conditions to obtain the desired dispersions are known to those skilled in the art. The milling equipment used to mill the parent inorganic oxide particles should be of the type capable of severely milling and reducing materials to particles having the desired size, e.g., through mechanical action. Such mills are commercially available, with fluid energy mills, hammer mills, and sand mills being particularly suitable for this purpose. Hammer mills impart the necessary mechanical action through high speed metai blades, and sand mills impart the action through rapidly churning media such as zirconia or sand beads. Impact mills can also be used. In other embodiments, milling is not needed, such as for air-set inorganic oxide gels. Such gels are formed by air- spraying an intimate mixture of an alkali metal solution (e.g., sodium silicate) with a suitable acid (e.g.. sulfuric acid) at such a concentration so that mixture gels during flight, before being collected in a suitable medium, generally water. Any resulting dispersion or powder may also be further processed. For example, further processing is desirable if there is a need to prepare a relatively stable dispersion without the aid of dispersing agents, or if there is a significant population of particles that are larger than desired. Further processing may also be needed to insure that essentially all of the distribution of particles is below a certain size. In such a case, the dispersion or powder is processed to separate the smaller particles from the larger particles. This separation can be created by centrifuging the inorganic oxide particles into a supernatant phase, which comprises the smaller particles of the final product and a settled phase which comprises the larger particles. The supernatant phase is then removed from the settled phase, e.g., by decanting. In some instances, it may be preferable to centrifuge the supernatant two, three or more times to further remove large particles remaining after the initial centrifuge. It is also contemplated that the larger particles of a milled dispersion can separate over time under normal gravity conditions, and the supernatant can be removed by decanting. Depending on the product particle size targets, the settled phase also can be regarded as particles of this invention. The dispersion of particles or powder also can be modified after milling to insure a stable dispersion. This can be accomplished through pH adjustment, e.g., adding alkaline material, or by the addition of conventional dispersants.
[0073] The inorganic oxide material in the compositions of the present invention may comprise two or more different and distinct types of porous particles. In one embodiment each type of porous particles provides a specific desorption and/or dissolution rate profile for the biologically active material in a liquid material so as to form a composite desorption and/or dissolution rate profile for the biologically active material.
[0074] In one embodiment, the surface of the inorganic oxide material, in particular the surface in the pores, has not been chemically modified. The pores in the inorganic oxide material are open so that the active ingredient in a liquid material can enter the pores and become adsorbed at the surface of the pores, or can leave the pores as to release the active ingredient. [0075] The porous inorganic oxide material of the present invention has an oil adsorption of about 100 to about 600 ml/100 g, or of about 100 to about 500 ml/100 g, or of about 100 to about 450 ml/100 g, or of about 100 to about 400 ml/100 g, or of about 150 to about 400 ml/100 g, or of about 200 to about 400 nil/100 g . The oil adsorption values can be measured with standard methodology, in particular by titrating a predetermined quantity of the inorganic oxide materia! with an oil under constant mixing of the oil inorganic oxide material, until the mass shows excess of oil, such as done in ASTM D28 ! ,
[0076] The inorganic oxide material in the compositions of the present invention is porous. In one embodiment the pores have a mean pore diameter of greater than 5 nm, or from about 5 nm to about 30 nm; or from about 10 nm to about 30 nm. In a further embodiment, the mean pore diameter is about 20 to about 25 nm.
[0077] Desirably, the porous inorganic oxide material has a pore volume of about 0,5 cnrVg or greater, or about 0,6 cm'Vg or greater, or about 0,7 em3/g or greater. In some embodiments, the upper limit of the pore volume is about 3.0 cm'Vg, or about 2.3 cm'Vg.
[0078] Desirably, the porous inorganic oxide material has a BET surface area., as measured by nitrogen adsorption, of about 10 ni'7g or greater, or about 100 m'Vg or greater, or of about 200 nr'-'g or greater, or of about 300 m'Vg or greater. In some embodiments, the upper limit of the BET surface area is about 1000 m2/g, or about B00 Vg, or of about 600 m2/g. In other embodiments, the BET surface area may range from about 10 to about 1000 m'Vg, or about 100 to about 800 rn2ig, or about 150 to about 600 m'Vg, or about 200 to about 500 nrVg, or about 250 to about 400 m'Vg.
[0079] In one embodiment, the porous inorganic oxide material has (i) a mean pore diameter of from about 5 nm to about 30 nm, (si) a pore volume of about 0.7 cm'Vg or greater, and (iii) a surface area of about 20 to about 500 m2/g, or greater, in another embodiment, this porous inorganic oxide material is in the form of particles, which may have a diameter from about 50 μη? (or about 44 pm) to about 150 μιτη (or about 149 pm). n a further embodiment this inorganic oxide porous material has an oil adsorption of about 100 to about 500 ml/100 g. Such materials are attractive due to their superior properties in terms of high loading, hulk density, flowability, desorption and dissolution ehareteristies.
[0080] In the present invention, the measurements of pore volume are generated by
N2 porosity analysis and surface area are generated by the BET' technique, which are art- known techniques. [0081] The porous inorganic oxide materia? containing a biologically active ingredient in a liquid material may be obtained from porous inorganic oxide material that do not contain a biologically active ingredient in a liquid material, which biologically active ingredient in a liquid material subsequently is added so that it is adsorped by the porous inorganic oxide material . Or, alternatively, the biologically active ingredient in a liquid material may be added during one or more of the steps of the preparation of the porous inorganic, oxide material.
[0082] in yet another embodiment, the inorganic oxide can be dispersed in a liquid compound, which is subsequently used as a reaetant or solvent or medium, which forms the biologically active composition of the present invention.
[0083] The particles of porous inorganic oxide material of the invention may be free- flowing. In one embodiment said particles may have a Carr index of equal or lower than about 25, e.g, a Carr index of about 10 to about 25, In some embodiments the Carr index may be equal or lower than about 15, e.g. a Carr index of about 10 to about 15. The particles of porous inorganic oxide material of the invention may have a Hausner index of about 1 to about 1.4, in particular of about 1 ,2 to about 1.4, The particles of porous inorganic oxide materia! of the invention may have an angle of repose of about 30* to about 45". In some embodiments, the Carr and Hausner index values and angles of repose mentioned herein apply to any of the particles of the invention having a w/w ratio of the biologically active ingredient in a liquid material to inorganic oxide inorganic oxide, as specified herein, and in particular where said w/w ratio is between about 0.5 ; 1 to about 2 : 3. In some embodiments, the Carr and Hausner index values and angles of repose mentioned herein apply to any of the particles of the invention that are unloaded with biologically active ingredient in a liquid material.
[0084] The Carr index (CI,) is an indication of the flowability and compressibility of a powdery material and is calculated by the formula C = 100 x (l -pb/pt), where p¾ is the freely settled bulk bulk density of a powder, and pt is the tapped bulk density of the powder. A Carr index greater than 25 is considered to be an indication of poor flowability. Materials having a Carr index equal to or lower than 25 show good flowability and can also be referred to as "free-flowing" materials, The Hausner index (H,I.) is calculated by the formula H ~ [0085] In another embodiment, the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a iiquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 300 to about 500 rnI/ΪΟΟ g;. and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnr/g or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material decreases in volume of at least about 15% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40% after resting. Each of the changes in volume and/or density mentioned herein are after loading and resting up to 2 hours depending upon the Iiquid, and even up to 24 or 48 hours. In one embodiment, upon loading the inorganic oxide porous material with biologically active ingredient in a iiquid material, a decrease of the volume or increase in density, in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs is observed, after which time period no further decrease occurs and the volume stays substantially constant,
[0086] In a further embodiment, the present invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cnvVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, increases in bulk bulk density by at least about 15% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%. or at least about 40% after resting. Each of the changes in volume and¾r density mentioned herein are after loading and resting up to 2 hours depending upon the Iiquid, and even up to 24 or 48 hours. In one embodiment, upon loading the inorganic oxide porous material with biologically active ingredient in a liquid material, a decrease of the volume or increase in density, in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs is observed, after which time period no further decrease occurs and the volume stays substantially constant.
[0087] In another embodiment, the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 ΐο about 500 rnl ϊϋϋ g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0,5 cm3/g or greater; wherein, after mixing the inorganic oxide particles and liquid material and then resting for at least 2 hours, at least about 400 mg of the composition may be loaded into a zero size capsule. In another embodiment, at least about 410 mg, or at least about 420 mg, or at least about 430 mg of said composition may be loaded into a zero size capsule.
[0088] In an even further embodiment, the present Invention relates to a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/i OO g; (b) pores having an pore volume, as measured by nitrogen porosimetry. of about 0.5 cmJ g or greater; and (c) a ratio of liquid material to inorganic oxide particles of at least 1 : 1 ; wherein at least 65% of the liquid material is desorbed from the particles upon desorption. In another embodiment, at least 70%, or at least 75%, or at least 80%, or at least 85% of the liquid material is desorbed from the particles upon desorption. The liquid material is desorbed under conditions that simulate desorption of liquid material in biological fluids. Such tests are performed by intense mixing in aqueous medium, as set forth in Example 7.
[0089] As used herein, the term "in liquid form" or "liquid material" in relation to biologically active ingredients refers to such ingredients that in themselves are liquids or to biologicaliy active ingredients brought into liquid form by various techniques including, for example, desorption and/or dissolution or conversion into a self-emulsifying drug deliver)' system, Such materials may also include solid biologically active ingredients that are suspended, dispersed or incorporated with liquids.
[0090] The term "in liquid form" or "liquid material" refers to biologically active ingredients as such or brought into liquid form that are liquid at room tremperaiure or at physiological temperature, or are liquid at temperatures ranging from about 0t!C to about 60°C, in partuclar about 10°C to about 5GaC, or about 20°C to about 5 :C, Such materials may be solid at certain conditions (e.g., temperature, concentration, etc.) and liquid under other conditions.
[0091 ] The porous inorganic oxide material in accordance with this invention contains a biologically active ingredient in liquid form. The term "contain" means that the porous inorganic oxide material is loaded with a biologically active ingredient, the term "loaded" meaning thai the active ingredient is adsorbed at the surface of the inorganic oxide material including the surface within the pores of the inorganic oxide material. A major part of the active ingredient may be incorporated in the pores of the inorganic oxide material. Such inorganic oxide materials with adsorbed biologically active ingredient are referred as "loaded inorganic oxide materials". The terms "loaded" and "incorporated" in this context arc meant to he equivalent,
[0092] In one embodiment, the w/w ratio of the biologically active ingredient in a liquid material (which comprises the biologically active ingredient itself as well as any added materials in the iiquid) to inorganic oxide is in the range of about 0,5: 1 to about 5: 1 , or of about 0.5: 1 to about 3: 1, or of about 0.5: 1 to about 2: 1, or of about 1 : 1 to about 2: 1 ,
[0093] The composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material may have a bulk density- of at least 450 g/1. In some embodiments said bulk density is in the range of from 450 g 1 to 750 g/1, in particular in the range of from 500 g/1 to 700 g/1, or in the range of from 550 g/i to 650 g/1,
[0094] In one embodiment, the biologically active ingredient in a liquid material is a liquid lipid drug. Examples include vitamin A, vitamin E (dl-a-Tocopherol), paracetamol ascorbic acid, sesame oil miglyol, or combinations thereof.
[0095] In another embodiment the biologically active ingredient in a liquid material takes the form of a solution or dispersion of the biologically active ingredient in a nonvolatile solvent, e.g. having a boiling point of above about 150 C. Examples include glycerin, propylene glycol, liquid polyethylene glycols such as polyethylene glycol 200 and 400, polysorbates such as polysorbate 80, or an oil, Oils that can be used include long-chain triglyceride and medium-chain triglyceride oils with different degrees of saturation; vegetable oils such as olive oil, sunflower oil castor oil, linseed oil and the like; modified or hydro! yzed vegetable oils; semisynthetic medium-chain triglyceride oils having surfactant properties, for example Cremophor. In ease of dispersions, the active ingredient is preferably in the form of micro- or of nanoparticles. In one embodiment the concentration of the biologically active ingredient in the solution or dispersion is in the range of from 1 % to 90 %, or from ... (w w).
[0096] In one embodiment, the biologically active ingredient in a liquid material is a self-emulsifying drag delivery system (SEDDS) comprising an oily/Hpid component, a surfactant cosolvent, and a biologically active ingredient, [0097] The oily/iipid component is generally a fatty acid ester or a medium/long chain saturated, partially unsaturated or unsaturated hydrocarbon, in liquid, semisolid or solid form at room temperature (e.g., solid lipid nanopartic!es, oily suspensions, submicron lipid emulsions, lipid implants, lipid microtubules, lipid mierobubhies, or lipid microspheres, etc). Examples include mineral oil, vegetable oil, modified or hydrolyzed vegetable oils, silicone oil, lanolin, liposomes, refined animal oil, fatty acids, fatty alcohols, and mono-/ t~ tri- glyeerides, including long-chain triglyceride and medium-chain triglyceride oils with different degrees of saturation and semisynthetic medium-chain triglyceride oils having surfactant properties. Further oily/iipid components include oils comprised of one or more medium chain fatty acids esters of propylene glycol such as propylene glycol rnonocaprate, propylene glycol dicaprate, propylene glycol dicaprylate/dicaprate, propylene glycol dipelargonate, and propylene glycol dilaurate, triacetin, fats and oils such as olive oil, sesame oil, soybean oil, corn oil, rape oil, castor oil, coconut oil, and eucalyptus oik. capryiic/eapric acid triglyceride ( iglyol ' 812); a triglyceride such as tricaprylin and trilaurin; and po!yglycerm fatty acid esters such as tetraglycerin polyricinoleate, hexaglycerin polyricinoieate, condensed poiyricinoleate, and tetraglycerin mixed fatty acid esters . The term"medium chain fatty acid" is meant to refer to fatty acyi chains of between 6 and 14 carbons in length, more preferably between S and 12 carbons in length; "long chain fatty acid" is meant to refer to fatty acyl chains greater than 14 carbons in length; "short chain fatty acid" is meant to refer to fatty acyl chains less than 6 carbons in length.,
[0098] The oil components may be used in the SEDDS formulations of the present invention in any effective concentration, including, for example, in a concentration range of 5% to 80% (w/v).
[0099] Preferred surfactants comprise non-ionic surfactants with a relatively high hydrophiiie-lipophilic balance (HLB) value usually in concentration ranges between 30% and 60% (w/w).
[0100] The hydrophilic surfactant (HLB (hydrophile-lipophiSe balance) of 9,0 or higher that can be used include polyoxyethylene lauryl ethers (Laureth 2 (BL-2), Laureth 4.2 (BL-4.2), and Laureth 9 (BL-9), polyoxyethylene (20) sorbitan monococonut oil fatty acid ester ("Polysorbate 20"), Polysorbate 40, Polysorbate 80, Labrasol, D-ot -tocopherol polyethylene glycol 1000 succinate (Vitamin E TPGS NF), lauroyl polyoxyethylene glycerin (Gelucire 44/34), polyoxyethylene hydrogenated castor oil 40 , polyoxyethylene hydrogenaied castor oil 60 (HCQ-6G), poiyoxyethylene sorbitan rnonolaurate, poiyoxyethylene sorbitan monopalmitate, and poiyoxyethylene sorbitan monooleate,
[0101 ] Any effective non-aqueous protic cosoivent or combinations thereof, may be used in the SEDDS for use in the invention. Acceptable non-aqueous proiic solvents include any pharmaceutically acceptable mono-, dk tri-, or poly-hydroxy linear aliphatic and aromatic solvent, or combinations thereof, Examples of non-aqueous protic solvents include ethanol, propanol, benzyl alcohol, propylene glycol, liquid polyethylene glycols such as polyethylene glycoi 200 and 400, and glycerol The protic solvents may be used in the formulations of the present invention i any effective concentration, including, for example, in a concentration range of about 5% to about 50% (w/v).
[0102] Optionally, a chelating agent and/or a soluble antioxidant may be included in the SEDDS for use in the invention. Chelating agents may be added to enhance the stability of a hydrophobic drag in the SEDDS composition. Suitable optional chelating agents include any pharmaceutically acceptable chelating agent, such as citric acid, nia!eie acid, succinic acid, tartaric acid, EGTA (ethylene giyeol-bis ( (3-aminoethyl ether) tetraaeetic acid, or egtazie acid) and EDTA (ethylene diamine tetraaeetic acid, or edetic acid). Such chelating agents are available in various forms, e. g. 4 as sodium or potassium salts or as the free acids. Such chelating agents may be used in the formulations of the present invention in any effective concentration, including, for example, in a concentration range of between 0,01% and 10% (w/v),
[0103] For preparing an SEDDS formulation, for example, an absorption^promoter such as sodium salicylate, sodium deoxycholate, sodium myristate, or sodium dodecyl sulfate.
[0104] The SEDDS formulations may contain an auxiliary solvent such as ethanol, propylene glycol, polyethylene glycoi, diethylenetriaminepentaacetic acid, diethanolamine, triethanolamine, ethylenediamine, monoethanoiamine, or N,N-dimethy!acetarnide.
[0105] The SEDDS formulation may be prepared by dissolving the drug in a mix of oil, surfactant and cosoivent.
[0106] The biologically active material used in the compositions of the present invention may comprise any known biologically active material. The term "biologically active ingredient" is meant to cover any pharmaceutical or other active ingredient for administration to humans or animals, in particular to warm-blooded animals. The biologically active material may be an active pharmaceutical ingredient (API), which comprises include naiiirai, semi-synthetic or synthetic moiecules. In some embodiments, the biologically active material comprises two or more active pharmaceutical ingredients (APIs) in combination with one another. Other biologically active ingredients include ingredients that have an effect on the general well-being or have an effect on the outer appearance (cosmetic) such as the skin, hair, lips, and eyes. Such ingredients include any agents for use in cleansing, beautifying, promoting attractiveness, or altering the appearance, for example moisturizers, oils, anti-wrinkle agents, fragrances, and the like. Also included are ingredients for nutritious applications (in particular the so-called "nutraceutieal" ingredients), Such ingredients include food supplements such as, for example, dietary food supplements, vitamins, minerals, fiber, fatty acids, and amino acids. Examples of such ingredients are Vitamin C, omega-3 fatty acids, carotenes, and flavonoids. The term "biologically active" in relation to compositions for cosmetic or nutricious applications also includes activity relating to the improvement of the outer part of the body, in particular of the dermis, as well as the general well-being of an individual.
[0107] in one embodiment, the active ingredient has a molecular weight below about
1 ,000 (daitons), or below about 800, for example a molecular weight in the range of about 150 to about 1 ,000, or in the range of about 200 to about 800.
[01 OS] The active ingredient for use in the invention may be soluble or insoluble in water or aqueous media, in particular physiological aqueous media. According to generally accepted standards, any solvent solubility is defined as the amount of a solvent (g) required to dissolve 1 g of a compound, whereby the following solubility qualifications are defined: 10- 30 g ("soluble"); 30-100 g ("sparingly soluble"); 100-1000 g ("slightly soluble"); 1000-10000 g ("very slightly soluble" or "poorly soluble") and more than 10000 g (practically insoluble).
[0109] In one embodiment, the active ingredient is soluble or insoluble in water or aqueous media, in particular physiological aqueous media. In one embodiment, the pharmaceutically active ingredient belongs to the so-called BCS classes I through IV, Classes I and III are the soluble drugs. The Biopharmaceutical Classification System (BCS) classifies drug substances based on their aqueous solubility and intestinal permeability into four classes: Class i— High Permeability, High Solubility: Class II— High Permeability, Low Solubility: Class Ill-Low Permeability, High Solubility; Class IV-Low Permeability, Low Solubility. [01 10] In one embodiment, the active ingredient has a partition coefficient (expressed as log P) that is in the range from 4 to 9, in the range from 3 and 8. in a further embodiment, the active ingredient has a pKA that allows the molecule to be in neutral (non-ionic form) at about pH 5-8,
[01 1 1 ] Exemplary APIs include, aiorvastatin, amiodarone, eandesartan~cilexetil,: earvedilol, elopidogrel bisulfate, dipyridamole, eprosartan mesylate, epierenone, ezetirnibe, felodipine, furosemide, isradipine, iovastatin, metolazone, nicardipine, nisoldipine, olmesartan medoxornil, propafenone HC1, qinapril, ramipril, simvastatin, telmisartan, trandoiapril, valsartan and other cardio-vascular active drugs; acyclovir, adefovir, dipivoxil, amphotericin, Amprenavir, cefrxirne, ceftazidime, clarithromycin, clotrimazole, efavirenz, ganciclovir, itraconazole, norfloxacin, nystatin, ritonavir, saquinavir and other anti-infective drugs including anti-hacterial, anti-vira!, anti-fungal and anti-parasitic drugs; cisplatin, earbopiatin, docetaxel, etoposide, exemestane, idaruhicin. irinotecan, melphalan. mer- captopurine, mitotane, paclitaxel, valrubicin, vincristine and other drugs used in oncology; azathioprine, tacrolimus, cyclosporin, psmecroHrnus, siroHnms and other smmonosupressive drugs; clozapine, eniacapone, fluphenazine, imipramine, nefazodone, olanzapine, paroxetine, pimozide, sertraline, triazolam, zaleplon, ziprasidone, risperidone, carbamazepine and other drugs for CNS indications; danazol, dutasteride, medroxyprogesterone, estradiol raloxifene, sildenafil, tadalafi!, testosterone, vardenafii and other drugs used for reproductive health; celeeoxib, dihydroergotamine mesylate, eletriptan, ergoloidmesylates, ergotaniine tartrate; nabumetone, Ibuprofen, ketoprofen, triamcinolone, triamcinolone acetonide and other antiinflammatory and analgesic drugs; bosentan, budesonide, desloratadine, fexofenadin. Fluticasone, ioratadine, rnometasone, saimeterol, xinafoate, triamcinolon acetonide, zafirlukast and other drugs for respiratory indications; and dronabinol, famotidine, glyburide, hyoscyamine, isotretinoin, megestrol, mesaiamine, modafsnil, mosapride, nimodipine, perphenazine, propofol, sucralfate, thalidomide, trizanidine hydrochloride and other drugs for various indications including in particular gastro-intestinal disorders, diabetes and dermatology indications. in further embodiments the APIs include ezetimimbe, giucoroniude, tadalafii, fenofibrate, danazol, Itraconazole, carbamazepine, gnseofu!vin, nifedipin.
[01 12] The active ingredients further include sugars, polysaccharides, vitamins, amino acids, peptides, prostaglandins, nucleic acids, nucleotides, nucleosides, as well as derivatives thereof. Also included are peptides, proteins, protein fragments, antibodies, small antibody fragments, and the like. The latter include Fv" fragments, single-chain Fv (scFv) antibodies, antibody Fab fragments, antibody Fab' fragments, antibody fragments of heavy or light chain CDRs, or nanobodies. Also encompassed are small oligonucleic acid or peptide molecules such as aptamers, for example DNA aptamers, RNA aptaraers or peptide aptamers.
[01 13] In one embodiment, the biologically active ingredient in a liquid material when loaded in the inorganic oxide material shows an increased release compared to the active ingredient as such, or to formulations containing the active ingredient and ingredients that do not influence release. Increased release may for example be an increase of 10%, or of 20%, or of 30%, or of 50%, of the weight percentage of active ingredient released under physiological conditions (pH, temperature).
[01 14] In a further embodiment, the biologically active Ingredient in a liquid material when loaded in the inorganic oxide material shows immediate release from the compositions of the invention, the term "immediate release" meaning, for example, a release of at least 60% of the drug under physiological conditions (pll, temperature), such as within 60 minutes or less, such as within 30 or less, or within 20 minutes or less, or within 15 minutes or lexs,
[01 15] In the methods of making a composition of the present invention, the step of incorporating the biologically active material into the inorganic oxide materia! typically comprises a variety of loading methods, including the solvent method and the incipient wetness method, which methods have been described in the prior art, or mere mixing without use of any solvent or other mixing aid.
[01 16] In the (slurry) solvent method the inorganic oxide material is loaded with an aciive ingredient by treatment with a solution of the active ingredient in a liquid material after which the solvent is removed. The active ingredient in a liquid material thereby becomes adsorbed to the surface of the inorganic oxide material, including the surface within the pores of the inorganic oxide material, Appropriate organic solvents for use in this method include dichloromethane, 1 ,4-dioxane, tetrahydrofuran, 2-propanoL diethyl ether, ethyl acetate, acetonitrile, dimethySformamide, N-methyl~pyrrolidinone, hexane. For example, a solution containing about 50 nig of active ingredient per ml can be used for loading active ingredients in inorganic oxide material.
[01 17] In the incipient wetness method, also referred to as capillary impregnation or dry impregnation the inorganic oxide material is wetted with the active ingredient in a liquid material or in a concentrated solution and is drawn into the pores by capillar}' action. The
?7 porous inorganic oxide materials of the invention are particularly suited for this methodology as they show strong capillary action. In many instances, no or very little solvent needs to be used thereby avoiding the removal of the solvent after the loading step, This offers an additional advantage over known liquisolid formulations, which require additional ingredients needed to help adsorption, in particular solvents, involving methods of premixing the carrier or the drug in a liquid material (lipid or SEDDS) with a solvent to improve the adsorption. The presence of solvents in medicines and other products for human or animal use are critically scrutinized while many solvents are banned. Solvents also have environmental implications as they are considered an important source of pollution.
[01 18] In another embodiment, the liquid material may be loaded onto the inorganic oxide material by spraying, or any other known method of liquid adsorption onto porous materials.
[03 19] The biologically active ingredient in a liquid material either with or without solvent may have a viscosity that is selected such that it can be adequately adsorbed by the inorganic oxide materials in particular in terms of speed of adsorption, sufficient loading, and the like, it may for example have a viscosity below about 250 mPa.s, or below about 100 mPa.s, or below about 10 mPa.s, or below about 5 mPa.s, or below about i mPa.s. The lower limit of the viscosity may be about 0.1 mPa.s, or about 0.5 mPa.s.
[0120] The inorganic oxide materials of the invention are very efficient adsorbants of biologically active ingredient in liquid form. Contrary to known porous materials, where adsorption takes place in a very short time after being brought in contact with the liquid material, the inorganic oxide materials of the invention adsorp biologically active ingredient in liquid form during longer periods of time, in particular during several hours, for example up to about 2, 3, or 4 hours, k is believed that the decrease in volume after resting is related to capillar}' farces that continue to draw free liquid between the particles up into the pores of the particles. This attributes to the high loading capacity of the inorganic oxide materials of the invention.
[0121 ] The content of the active ingredient in the inorganic oxide material materials may be in the range of about 1 % to about 50%, or about 10% to about 30%, or about 15% to about 25%, for example about 20%. relative to the total weight of the loaded silica material (all percentages herein being weight/weight). [0122] The compositions of the present invention may in one or more additional steps formulated into a final dosage form, which may vary depending upon the marsner in which it is administered to the patient, Preferred are solid or semisolid dosage forms for oral administration, in particular pills, tablets, and eapsulees. Such dosage forms may be suitable for providing immediate or fast in vivo release of said biologically active species, or may be suitable for controlled release. This may include one or more pharmaceutically acceptable excipients
[0123] Regardless of the production method used to prepare the compositions containing a biologically active material and an inorganic oxide material in accordance with this invention, whether it is solvent-based or solventless, when the final dosage form comprises one or more pharmaceutically acceptable excipients, they may be introduced at any time during the process, including the step designed to load the biologically active material into the pores of the inorganic oxide material, or afterwards in a separate step,
[0124] The pharmaceutical compositions may also contain optional excipients. These may comprise any of the ingredients customarily employed in the art such as diluents, binding agents, granulating agents, giidants (flow aids), lubricants: disintegrants, sweeteners, flavors, and pigments to make the tablets visually attractive. Examples of such excipients include hydroxypropylmethyl cellulose, crospovidone, magnesium s earate, lactose, and talc.
[0125] The pharmaceutical compositions of the present invention may further comprise one or more pharmaceutically acceptable fillers selected, for example, from hydrocoiioids (such as xantl an gum), binding agents, giidants, lubricants, surfactants and diluents.
[0126] These include for instance binding agents such as starch, gelatin, glucose, alginic acid, sodium and calcium alginates, water-soluble acrylic (co) polymers, polyvinylpyrrolidone, polyaminoacids, ethylene-vinyi acetate copolymers and the like; natural and synthetic mineral fillers or giidants such as silica, magnesium silicates such as tale, diatornaceous earth, aluminum silicate such as kaolinite, montmorillonitc or mica, magnesium aluminum silicate such as attapulgite and vermieulite, carbon such as charcoal, sulphur and highly dispersed silicic acid polymers; water-soluble diluents such as lactose, sorbitol and the like.
[0127] The compositions of the present invention may also be formulated into forms suitable for topical application such as an ointment, a cream, a gel, a liniment or balm, etc... [0128] The present invention is further directed to methods of using any of the herein disclosed compositions. In some embodiments, the compositions, in particular the pharmaceutical compositions, of the present invention may be used as medicaments, in particular may be used as medicaments via the oral route.
[0129] The present invention a method of administering a composition to a patient so as to deliver at least one biologically active material to the patient wherein the composition comprises at least one pharmaceutical dosage formulating ingredient of a porous inorganic oxide material containing a biologically active ingredient in liquid form, wherein the inorganic oxide material has the oil adsorption, the intraparticie pore volume, and BET surface area, as specified herein. The compositions in this method are preferably administered by various means, including by oral, buccal, sublingual, periodontal, vaginal intrauterine, rectal, pulmonary, nasal, inhalation, intraocular, ophthalmic, auricular, and topical means,
[0130] One of the reasons for improved release of the biologically active material from the compositions of the present invention is due to the improved desorption of the liquid material from the inorganic oxide material. The presence of pores within the inorganic oxide materials (i.e., intraparticie porosity) that have certain features allows for a substantial amount of the biologically active ingredient to be adsorbed and then released. For example, the pore size distribution of the present inorganic oxide material is narrow (i.e., a small relative span), which allows for a number of pores to readily adsorb and desorb the liquid material. This contrary to known porous materials where the biologically active ingredient is adsorbed into or released from interstitial voids between lumps or particles of the carrier material, the size and shape of which are more aleatory and offer less room for loading molecules. This causes not only less loading capacity, but also less regular release profiles. Other factors that influence release are viscosity of the active ingredient In liquid form and the isoelectric point (logP).
[013 1 ] The compositions in accordance with the invention provide attractive drug delivery properties. They provide desirable desorption and/or dissolution rate profiles for a variety of biologically active materials (e.g., APIs), in some embodiments, the biologically active material exhibits a percent release desorption and/or dissolution rate of about 20 or greater within about 15 minutes of an initial time of contact with a dissolution medium. In some embodiments, the biologically active material exhibits a percent release dissolution rate of about 25 or greater (or about 30 or greater; or about 35 or greater) within about 15 minutes of an initial time of contact with a dissolution medium.
[0132] Further, in some embodiments, the biologically active material exhibits a percent release dissolution rate of about 20 or greater about 30 minutes after an initial time of contact with a dissolution medium, in some embodiments, the biologically active material exhibits a percent release dissolution rate of about 30 or greater about 30 minutes after an initial time of contact with a dissolution medium.
[0133] in some embodiments, the biologically active material exhibits a percent release dissolution rate of about 10 or greater about 60 minutes after an initial time of contact with a dissolution medium. In some embodiments, the biologically active material exhibits a percent release dissolution rate of about I S or greater (or about 20 or greater) about 60 minutes after an initial time of contact with a dissolution medium,
[0134] The compositions of the present invention in many instances show immediate release of the active ingredient but may be turned into controlled release compositions for example by coating the compositions with a suitable polymer. When mixing compositions with sleeted polymer coatings mixed release patterns can be obtained such as a combination of immediate and sustained release.
[0135] The inorganic oxide material may comprise two or more different and distinct types of porous particles with each distinct type of porous particles providing a specific desorption and/or dissolution rate profile for a single biologically active material (or two or more different biologically active materials) so as to form a composite desorption and/or dissolution rate profile.
[0136] A further aspect of this invention concerns particles of a inorganic oxide material, wherein the inorganic oxide material wherein the inorganic oxide material has the oil adsorption, the intraparticle pore volume, and BET surface area, as specified herein.
[0137] The compositions of the present invention not only show high loadabiiity of drugs in liquid form, they moreover show higher bulk density compared to existing Siquisolid systems. Further favorable properties include the excellent adsorptive capacity of the inorganic oxide material of the invention and the increased stability of the active ingredient. These advantages are in particular offered by the compositions of the invention in the form of particles. [0138] The compositions of the present invention can also be used in dermatology and cosmetic applications because of their good skin-comparability and lack of unpleasant skin feel
[0139] The following examples are meant to illustrate the present invention and should not be construed as a limitation of its scope,
Example 1
[0140] Sodium water glass with a Na content of 24,5% w/v and sulfuric acid of 45% w/v were mixed with a molar ratio of 0,85 to 0,99. After completion of the poly condensation the raw silica gel was crushed into pieces of several cm sizes. Then the by-product sodium sulfate was removed by washing the silica gel/sodium sulfate mixture with clear water. The aging of the silica by Oswald ripening took place in a water bath for 3-1 1 hours at 70-8Q6'C and at pH between 8 and 9. After the liquid/solid separation the formed silica hydro gel was crushed down to a particle size of about 300 μηι, T he subsequent drying step controlled the formation of the pore volume. In order to achieve pore volumes of about 1.7 cnrVg rapid drying for less than 4 seconds at a process air temperature of 1 8G*C was needed and was conducted in a Sab flash dryer type LABSPI FLASH (APV/Denmark). Silicas with pore volumes of < 1 em3/g were made by slow drying {packed bed drying) in a lab drying chamber at 100eC for 4h.
[0141 ] The following silica particles shown in Table 1 below were used in the subsequent examples.
Table 1
Figure imgf000034_0001
[0142] The DI G, D50 and D90 values indicate the 10th, 50th and 90th percentiles of the weight of the particle diameter distribution. These values were obtained from a Malvern f M rV!astersizer 2000 instrument available from Malvern Instruments Ltd. PV: pore volume; SA: surface area; APD: average pore diameter; BET SA: surface area; and Relative Span are determined using BJH nitrogen adsorption at a pressure of 0.995 using an ASAP 2420HV accelerated surface area and porosimeiry system available from Micromeretics Instrument Corporation.
Example 2
[0143] Procedure: 2 to 5 g (based on bulk density) of the solid carrier material was placed in a 300 ml beaker and the oil or surfactant was added drop wise from a Burette, while mixing with spatula according to ASTM D281 . The addition of oil or surfactant was continued until a thick paste-like mass formation. The addition of oil or surfactant was stopped when the mass appeared to contain excess oil The Burette reading was recorded when the mass contained no excess oil or surfactant. The adsorption capacity was calculated using the below equation:
Volume of oil added (mi) SG of oil 100
Oil adsorption g 100 g) ~
Weight of sample (g) [0144] The following table lists oils and and their adsorption capacity on Sample S I material as well as the specific gravity of the oils.
Table 2
Figure imgf000035_0001
[0145] The following table lists surfactants and their adsorpiion capacity on Sample
S 1 material as well as the specific gravity of the surfactants.
Table 3
Figure imgf000036_0001
[0346] In comparison, oiher carriers were tested for oil adsorption and following results were obtained.
Table 4
Figure imgf000036_0002
Example 3
[0147] Following the procedure of Example 2, the Cremophor and Labratli loaded materials were allowed to rest for 3 hours and then tested for free flowing properties. The results are shown n Tables 5 and 6.
Figure imgf000037_0001
Figure imgf000037_0002
[0148] Bulk Density is measured by USP 616 Method 1 using a 250 mi graduated cylinder (USP30-NF25). Tapped density is measured by USP 616 Method 2 (250 taps per minute) using a 250 mi graduated cylinder, with an ETD~f G20 Tap Density Tester available from Electro!ab. USP rating is an observed measure of powder flow and is rated according to the following Table 7. Table 7
Figure imgf000038_0001
Hxampis
[0149] Following the procedure of Example 2. 10 grains of Sample S I is loaded wiih
16.25 ml of sesame oil which yield a I ; 1 ,5 ratio of inorganic oxide material to oil. The volume of the mixture decreases from 65 ml to 50 nil over a period of 10 hours, which is a decrease in over 23% as shown in FIG. 1.
[0150] The Cremophor and Labrafii loaded materials of Example 3 were loaded into zero size capsules manually using a casule filling tray, such as the Cap- -Quick available from Empty Caps Company. As can be seen from FIG, 2, there ts a significant difference between the filling amounts before and after resting of the material The amounts of material loaded into the capsules are set forth in Tables 8 and 9.
arameters (Oessiophor
SI (256A, Ι ΐΰμΐϊί)
Loaded 1.5 ί}_
Gil added (mL) .14 7, 14
Carrier Material (g) 5
Filling Amount (mg)
451 429
After restmg_
Filling Amount (mg)
357 35 j
Before resting
Table 9
Figure imgf000039_0001
Example 6
[0153] Following the procedure of Example 2, acetaminophen and ascorbic acid loaded material was obtained and labeled carriers S I and S2, respectively The drug release from carriers S I and S2 was determined as follows:
[O S 52] The acetaminophen loaded carrier (S I) was subjected to dissolution studies as per USP 30 for Acetaminophen Tablets for 30 min. Dissolution test conditions comprised of use of USP dissolution apparatus 2 (Paddle) operated at speed of 50 RPM for 30 minut.es> Dissolution medium was 900 ml of pH 5.8 Phosphate Buffer at 37±0,5¾. For each carrier, 100 mg of the weighed amount of drug loaded carrier was used to dissolution studies.. ASiquots (5 ml) were withdrawn at 10, 20, 30 minute time intervals, filtered and diluted with dissolution fluid. Absorhance of aliquots was determined spectrometrieally at max 243nm. The release profile is illustrated in FIG, 3 The drug release from the both carriers SI and S2 met the USP criteria (NLT 80% in 30 minutes).
[0153] The ascorbic acid loaded carrier (S i ) was subjected to dissolution studies as per USP 30 for Ascorbic acid Tablets for 45 min. Dissolution test conditions comprised of use of USP dissolution apparatus 2 (Paddle) operated at speed of 50 PM for 45 minutes. Dissolution medium was 900 ml of water at 37±Q,5 C. For each carrier, 100 rng of the weighed amount of drug loaded carrier was used to dissolution studies. Aliquots (5 ml) were withdrawn at 10, 20, 30 minute time intervals, filtered and diluted with water. Absorbance of aliquots was determined spectromeiricaliy at X ax 266nm. The release profile is illustrated in FIG, 4. The drug release from the both carriers S i and S2 met the USP criteria (NLT 75% in 45 minutes).
Example 7
[0154] Oil release or desorption; The carrier material used in the following trials was the material designated S I prepared in Example 1 , 2 grams of oil loaded carrier ( 1 : 1 , w/w) prepared pursuant to method of Example 2 was mixed with 6 ml water in a beaker, vorlexed during 1 hour, and centrifuged at 5000 RPM for 10 min in a Heraeus Muitifuge 1 S~R centrifuge available from Thermo Electron Corporation. The supernatant, i.e. oil + water, was transferred into a petry dish and dried in hot air oven up to constant weight.
[0155] The results, which are based upon the w/w % release obtained in the trials., are as follows. For sesame oil, 81 % of the oil is released from the carrier materia! or inorganic oxide, and tor Miglyol 812, 81.3% of the oil is released from the carrier material or inorganic oxide.
Example 8
[0156] Solid SEDDS System Loading and Release (or Desorption): The carrier materials used in the following trials was the materials designated as SI and S2 in Example 1. A liquid SEDDS system was made up containing 0,6g of Giyburide as the API component, I Sg of Capryol®90 as the oil/vehicle component. 54.4g of Trascutoi® HP as a co-surfactan and 30g of Tween® 20 as a surfactant. This liquid SEDDS system was loaded onto SI and S2 by accurately weighing the required quantities of carrier and liquid SEDDS in the ratio of 1 : 1. Carrier and Liquid SEDDS were pre-heated at 60 °C for S 5 min, prior to mixing. Liquid SEDDS was added slowly to the carrier under stirring with metallic spatula. The prepared mixture was kept aside for around 24 hr to get free flowing powder.
[0157] The liquid SEDDS ioaded carriers (S I) and (S2), as well as non~mieronized
G!yhuride were subjected to dissolution studies as per USP<71 1> for 120 minutes. Dissolution test conditions comprised of use of USP dissolution apparatus 2 (paddle) operated at a speed of 75RPM. Dissolution medium was 500ml of pH 9.5 Borate Buffer (0.05M) at 37±1°C. For each carrier, a weighed amount of ioaded carrier or APi(non- micronized Gyburide) with an weight equivalent basis of 5rng Gyb ride were used for the dissolution studies. 5ml AHquots were withdrawn at 20, 30, 45, 60, and 120 minutes, filtered through 0.45 μ syringe filter and diluted with dissolution fluid, Samples were analyzed using HPLC (Waters Acquity H-class) using Grace Vision HT high load C I S column, Rocket Format, (53x7mm, 3μηι) as stationary phase and Acetonitrile: o-phosphoric acid 0.4% in water (50:50) as mobile phase at flow rate of LSmL/min with injection volume of 50 μΐ , Samples were detected at max 226nni. The release profile is illustrated in FIG.5, The drug release from both carriers SI and S2 met the USP criteria (NLT 70% drug release within 45 minutes, and NMT 3% relative SD).
[0158] To define the maximum amount of preloaded carrier in a tablet while maintaining optimum tahiettmg properties, the following experiments as shown in example 9 and 10 were performed:
Example 9
[0159] The carrier materials used in the following trials was the material designated
S2 in Example 1. Cremophor® EL was the liquid lipid that was loaded on to the silica. Silica 52 was loaded pursuant to method of Example § and was used as the oil loaded carrier component in tablet formulations. To deliver oil in tablet dosage form, tablets were made with two different processes: direct compression (DC) and wet granulation (WG).
[0160] Direct compression tablets were obtained by accurately weighed the quantities of excipients for blend preparation. Diluent (MCC ) and Oil loaded carriers were sieved through #40 mesh and mixed well for approximately 5 min. Binders and disiniegrant were sieved through #40 mesh and added to the blend, then mixed well for approximately 5 min. GHdant was passed through #40 mesh and added to the blend and mixed well for 5 min. Lubricant was sieved through # 60 mesh and added to the blend and mixed well for approximately 2min. This final blend was used for the compression of the tablets.
[0161] Wet granulation tablets were obtained by accurately weighed the quantities of excipients for blend preparation. Diluents (MCC) and Oil loaded carriers were sieved through #40 mesh and mixed well for approximately 5 min. Disintegrant was sieved through #40 mesh and added to the blend, then mixed well for approximately 5 min. Binder (stareh/pregelatinized starch) was prepared in a water solution (5%), Granules were then prepared by mixing Prepared diluents/oil loaded earrier disintegrant blend with the binder solution. Granules were dried at 50 °C to achieve LOD of 5-7%. Dried granules were passed through #20 mesh. Ghdant was passed through #40 mesh and added to the blend and mixed well for 5 min. Lubricant (what material) was sieved through # 60 mesh and added to dried granules and mixed well for approximately 2mirt. This final blend was used for the compression of the tablets.
[0162 Tablets were prepared using a Parle Elizabeth tools Pvt Ltd, Eliza press 200 mu!ti tooling single rotary tablet press operated at a Speed of 5 rpm, compresssion force of 20kN, ejection force 7QR and with a 12mm round bioconcave, D-tooiing punch. The formulations tabletted are listed in Table 10 below.
Table 10
Figure imgf000042_0001
[0163] Tablet hardness for all formulations was measured with EH 01 tablet hardness tester (Eleetrolab, India). The hardness for tested formulations are listed in Table Y below.
Figure imgf000043_0001
[0164] in all cases the tablets met the IJSP specifications for Weight variation (NMT
5%), Firabi!ity(<l ), and disintegration tirne(<I 5niin),
Example 10
[0165] High Concentration of Liquid Lipids Loaded onto Carrier and Compressed into Tablets: The carrier materials used in the following trials was the material designated S2 in Example 1. Tocopherol was the liquid lipid that was loaded on to the silica. Silica S2 was loaded with liquid lipid pursuant to method of Example 8 and was used as the oil loaded carrier component in tablet formulations. PVP30 was dispersed in ethanol (100mL) and added upon oil loaded carrier. Prepared blend was mixed thoroughly and allowed to dry at 50°C. Additional excipients were added pursuant to the method in Example 9 to obtain a direct compression blend with the following composition : 70% Tocopherol loaded silica S2(l : 1 loaded), 12.5%MCCPH102, 34% PVP30, 2% AcDiSoi, 1 % SYLOSD® 244FP, 0.5% Magnesium Stearate. Tablets were prepared using a Parle Elizabeth tools Pvt Ltd, Eliza press 200 mu!ti tooling single rotary tablet press operated at a Speed of 5 rprn, compression force of 20kN, ejection force <70N, and with a 12mm round bioconeave, D-tao!ing punch.
[0166] Tablet hardness for all formulations was measured with EH 01 tablet hardness tester (Eieetrolab. India) at tablet weight of 5GiK-5rng. Tablet hardenss results for these tablets was 40N. Friability was 0% and disintegration time was <l min.
Example 1 1
[0167] Oil Release (Tocopherol) from Tablets: The carrier materials used in the following trials was the material designated S2 in Example L Tocopherol was the liquid lipid that was loaded on to the silica. Silica S2 was loaded with liquid lipid pursuant to method of Example 8 and was used as the oil loaded carrier component in tablet. Tablets were prepared pursuant to the method in example 10. Tochopherol concentration in the tablet was 1 OOmg. [0168] Tablets prepared were .subjected to dissolution studies pursuant to the method described in Example 8. An aliquot of 2 mL was withdrawn at predetermined time interval and filtered through 0.22 μ membrane filter. The dissolution samples were analyzed by using HPLC (Waters UPLC Wavelength: 294 rtm Column: Rocket Format, 53x7mm, 3μ) Mobile Phase: 85% ACN: 10% MeOH: 5% H20.)
[0169] The Tocopherol release at 45 in was ¾! 00%.
[0170] While the invention has been described with a limited number of embodiments, these specific embodiments are not intended to limit the scope of the invention as otherwise described and claimed herein, It may be evident to those of ordinary skill in the art upon review of the exemplary embodiments herein that further modifications, equivalents, and variations are possible. All parts and percentages in the examples, as well as in the remainder of the specification, are by weight unless otherwise specified, Further, any range of numbers recited in the specification or claims, such as that representing a particular set of properties, units of measure, conditions, physical states or percentages, is intended to literally incorporate expressly herein by reference or otherwise, any number failing within such range, including any subset of numbers within any range so recited. For example, whenever a numerical range with a lower limit, and an upper limit Ry, is disclosed, any number R falling within the range is specifically disclosed, in particular, the following numbers R within the range are specifically disclosed: R ;;; RL + k(Ru -RL)> where k is a variable ranging from ! % to 100% with a 1% increment e.g., k is 1 %, 2%, 3%, 4%, 5%. ... 50%, 51 , 52%. , , , 95%, 96%, 97%, 98%, 99%, or 100%. Moreover, any numerical range represented by any two values of , as calculated above is also specifically disclosed. Any modifications of the invention, in addition to those shown and described herein, will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims. All publications cited herein are incorporated by reference in their entirety.

Claims

Claims
1. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 ml/100 g; and
(b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cmVg or greater;
wherein said inorganic oxide. particles remain free flowing at a weight ratio of liquid material, to inorganic oxide particles of at least 1 ,5:1 ,
2. The composition according to claim 1 , wherein the inorganic oxide particles comprises pores having an intraparticle pore volume of about 0.5 cnrVg or greater.
3. The composition according to claim .1 , wherein the inorganic oxide particles comprises a. BET surface area, as measured by nitrogen adsorption of about 20 orV'g or greater.
4. The composition according to claim 1, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nra to about 30 nro.
5. The composition according to claim 1, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1.5 or less.
6. The composition according to claim 1 , wherein the particles comprise a mean particle size of from about 3 ηι to about 5 mm.
7. The composition according to claim 1, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants.
8. The composition according to claim 1 , wherein said particles form a powder having a Carr index of equal or lower than 25.
9. The composition according to claim 1, wherein said porous inorganic oxide particles are non-ordered.
10. The composition according, to claim wherein the composition, after mixing the inorganic oxide particles and liquid material decreases in bulk density of at least 15% after resting up to 24 hours.
1 1 . The composition according to claim 1, wherein the biologically active ingredient is an active pharmaceutical ingredient.
12. The composition according to claim 1, wherein the biologically active ingredient is liquid.
13. The composition according to claim 1 , wherein the biologically active ingredient is dissolved m a non-volatile solven or a lipid material.
14. The composition according to claim 1 , wherein the biologically active ingredient is formulated in a SEDDS.
.
15. A pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess;
(a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cm3/g or greater;
wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least J .5: 1.
16. The composition according to claim. .15, wherein the particles comprise a pore size distribution where at least 0.5 cc/g of pore volume are from pores ranging from 10 nm to 30 sim.
17. The composition according to claim 15, wherein the inorganic oxide particles comprises pores having an intrapartic le pore volume of abou t 0.5 cnrVg or greater,
18. The composition according to claim 15, wherein the inorganic oxide particles comprises pores having .a mean pore diameter in the range of about 5 nro to about 30 nm,
1 . The composition according to claim 15, wherein the inorganic oxide particles comprises a BET surface area, as measured by nitrogen adsorption of about 20 rrf /g or greater,
20. The composition according to claim 15, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nm to about 30 nm,
21 . The composition according to claim 15, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1.5 or less.
22. The composition according to claim 15, wherein the particles comprise a mean, particle siz of from about 3prn to about 5 mm.
23. The composition according to claim 15, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants.
24. The composition according to claim 15, wherein said particles form a powder having a Carr index of equal or lower than 25.
25. The composition according to claim 15, wherein said porous inorganic oxide particles are non-ordered.
26. The composition according to claim 15, wherein the composition, after mixing the inorganic oxide particles and liquid material, decreases in bulk density of at least 15% after resting up to 24 hours.
27. The composition according to claim 15, wherein the biologically active ingredient is an active pharmaceutical ingredient.
28. The composition according to claim 15, wherein the biologically active ingredient is liquid.
29. The composition according to claim 15, wherein the biologically active ingredient is dissolved in a non-volatile solvent or a lipid material.
30. The composition according to claim 15, wherein the biologically active ingredient is formulated in a SEDDS.
31. A method of making composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess: (a) an oil adsorption of about i 00 to about 500 mi/ 100 g; and
(b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 em'Vg or greater;
wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least .1.5: 1.
32. A method of making a pharmaceutical composition comprising a least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid materia!, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 mi/100 g; and
(b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cm g or greater;
wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid materia! to inorganic oxide particles of at least 1.5 : 1 .
33. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 rol/100 g;
(b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0,5 cm7g or greater; and
(c) a pore size distribution having a relative span of about 1 .5 or less.
34. The composition according to claim 33, wherein the inorganic oxide particles comprises pores having an iniraparticle pore volume of about 0.5 cm'Vg or greater.
35. The composition according to claim 33, wherein the inorganic oxide particles comprises a BET surface area, as measured by nitrogen adsorption of about 20 m2/g or greater.
36. The composition according to claim 33, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nm to about 30 nm,
37. The com position according to claim 33, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1.3 or less.
38. The composition according to claim 33, wherein the particles comprise a mean particle siz of from about 3μιη to about 5 mm.
39. The composition according to claim 33, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants,
40. The composition according to claim 33. wherein said particles form a powder having a Cart index of equal or lower than 25.
41 . The composition according to claim 33, wherein said porous inorganic oxide particles are non-ordered.
42. The composition according to claim 33, wherein the composition, after mixing the inorganic oxide particles and liquid material, decreases in bulk density of at least 15% after resting up to 24 hours.
43. The composition according to claim 33, wherein the biologically active ingredient is an active pharmaceut cal ingredient.
44. The composition according to claim 33, wherein the biologically active ingredient is liquid.
45. The composition according to claim 33, wherein the biologically active ingredient is dissolved in a non-volatile solvent or lipid material.
46. The composition according to claim 33, wherein the biologically active ingredient is formulated in a SEDDS.
47. A composition comprising porous inorganic oxide particles containing biologically active ingredient h a liquid material, wherein the inorganic oxide particles possess:
(a) a oil adsorption of about 100 to about 500 ml/100 g;
(b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cm g or greater; and
(c) a median pore size of 5 nm to 30 nra.
wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1.
48. The composition according to claim 47, wherein the inorganic oxide particles comprises pores having an mtrapartide pore volume of about 0.5 cm g or greater.
49. The composition according to claim 47, wherein the inorganic oxide particles coraprises a BET surface area, as measured by nitrogen adsorption of about 20 m2/g or greater.
50. The composition according to claim 47, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nm to about 30 nm.
51. The composition according to claim 47, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1 .5 or less.
52. The composition according to claim 47, wherein the particles comprise a mean particle siz of from about 3 ηι to about 5 mm,
53. The composition, according to claim. 47, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants,
54. The coniposition accordmg to claim 47, wherein said particles form a powder having a Carr index of equal or lower than 25.
55. The composition according to claim 47. wherein said, porous inorganic oxide particles are non-ordered.
56. The composition according to claim 47, wherein the composition, after mixing the inorganic oxide particles and liquid material increases in bulk density of at least 15% after resting up to 24 hours.
57. The composition according to claim 47, wherein the biologically active ingredient is an active pharmaceutical ingredient.
58. The composition according to claim 47, wherein the biologically active ingredient is liquid.
59. The composition according to claim 47, wherein the biologically active ingredient is dissolved in a non-volatile solvent or a lipid material.
60. The composition according to claim 47, wherein the biologically active ingredient is formulated in a SEDDS.
61. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 mi/100 g;
(b) pores having an. pore volume, as measured by nitrogen porosimetry, of about 0.5 cnrVg or greater; and
(c) a median particle size of from 3 microns to 300 microns;
wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1.
62. The composition according to claim 61, wherein the inorganic oxide particles comprises pores having an intrapartic!e pore volume of about 0.5 cnrVg or greater.
63. The composition according to claim 6.1, wherein die inorganic oxide particles comprises a BET surface area, as measured by nitrogen adsorption of about 20 rrfVg .or greater..
64. The composition according to claim 61, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nm to about 30 m.
65. The composition according to claim 6.1, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1.5 or less.
66. The composition according to claim 61, wherein the particles comprise a mean particle siz of from about 3 η to about 5 mm.
67. The compositio according to claim 61, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants.
68. The composiiion according to claim 61 , wherein said pasticies form a powder having a Car!' index of equal or lower than 25,
69. The composition according to claim 61, wherein said porous inorganic oxide particles are non-ordered.
70. The composition according to claim 61, wherein the composition, after mixing the inorganic oxide particles and liquid material, increases in bulk density of at least 15% after resting up to 24 hours,
71 . The composition according to claim 61. wherein the biologically active ingredient is an active pharmaceutical ingredient,
72. The composition according to claim 61 , wherein the biologically active ingredient is liquid.
73. The composition according to claim 61. wherein the biologically active ingredient is dissolved in a non-volatile solvent or a lipid material
74. The composiiion according to claim 61, wherein the biologically active ingredient is formulated in a SEDDS.
75. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 mi/100 g: and
(b) pores ha ving an pore volume, as measured by nitrogen porosimetry, of about 0.5 cm g or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, decreases in volume of at least 15% after resting.
76. The composition according to claim 75, wherein the inorganic oxide particles comprises pores having an intrapartic!e pore volume of about 0.5 cm3/g or greater.
77. The composition according to claim 75, wherein the inorganic oxide particles comprises a BET surface area, as measured by nitroger! adsorption of about 20 m2/g or greater.
78. The composition according to claim 75, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 sim to about 30 am.
79. The composition according to claim 75, wherein the porous inorganic oxide particles comprise a pore size distribution having a relati ve span of about 1.5 or less.
80. The composition according to claim 75, wherein the particles comprise a mea particle siz of from about 3μπ¾ to about 5 mm,
81 ,. The composition according to claim 75, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants.
82. The composition according to claim 75, wherein said particles form a powder having a Carr index of equal or lower than 25,
83. The composition according to claim 75, wherein said porous inorganic oxide particles are non-ordered.
84. The composition according to claim 75, wherein the composition, after mixing the inorganic oxide particles and liquid material, increases in bulk density of at least 15% after resting up to 24 hours.
85. The composition according to claim 75, wherein the biologically active ingredient is an active pharmaceutical ingredient.
86. The composition according to claim 75, wherein the biologically active ingredient is liquid.
87. The composition according to claim 75, wherein the biologicall active ingredient is dissolved in a non-volatile solvent or a lipid material.
88. The composition according to claim 75, wherein the biologically active ingredient is formulated in a SEDDS.
89. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 ml/100 g; and
(b) pores having an pore volume, as measured by nitrogen porosimeiry, of about 0.5 cnrVg or greater:
wherein said composition, after mixing the inorganic oxide particles and liquid material, increases hi bulk density by at least 1 % after resting.
90. The composition according to claim 89. wherein the inorganic oxide particles comprises pores having an intraparticle pore volume of a bout 0.5 cmVg or greater.
91. The composition according to claim 89, wherein the inorganic oxide particles comprises a BET surface area, as measured by nitrogen adsorption of about 20 rrf /g or greater.
92. The composition according to claim 89, wherein the inorganic oxide particles comprises pores having a mean pore diameter in the range of about 5 nm to about 30 nm.
93. The composition according to claim 89, wherein the porous inorganic oxide particles comprise a pore size distribution having a relative span of about 1 .5 or less.
94. The composition according to claim 89, wherein the particles comprise a mean particle siz of from about 3um to about 5 mm.
95. The composition according to claim 89, wherein the liquid material comprises lipid materials, non-volatile solvents, and surfactants.
96. The composition according to claim 89, wherein said particles form a powder having a Carr index of equal or lower than 25.
97. The com position according to claim 89, wherein said porous inorganic oxide particles are non-ordered.
98. The composition according to claim 89, wherei the composition, after mixing the inorganic oxide particles and liquid material, increases in bulk density of at least 15% after resting up to 24 hours.
99. The composition according to claim 89, wherein the biologically active ingredient is an active pharmaceutical ingredient.
00,. The composition according to claim 89, wherein the bioiogicaliy active ingredient is i squid.
101. The composition according to claim 89, wherein the bioiogicaliy active ingredient is dissolved in a non-volatile solvent or a lipid material.
102. The composition according to claim 89, wherein the biologically active ingredient is formulated in a SEDDS.
103. A composition comprising porous inorganic oxide particles containing a biologically active ingredient in liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 ml/100 g; and
(b) pores having an pore vokime, as measured by nitrogen porosimetry, of about 0,5 cm'Vg or greater;
wherein, after mixing the inorganic oxide particles and liquid material and then resting for at least 2 hours, at least about 400 nig of said composition may be loaded into a zero size capsule.
104. A composition comprising porous inorganic oxide particles containing a biologically acti ve ingredient i a liquid material, wherein the inorganic oxide particles possess:
(a) an oil adsorption of about 100 to about 500 ml/100 g;
(b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cm3/g or greater; and
(c) a ratio of liquid material to inorganic oxide particles of at least 1 : 1 ;
wherein at least 65% of the liquid, material is desorbed from the particles upon desorption.
PCT/US2014/013848 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies WO2014120922A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
KR1020157023665A KR102229036B1 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
US14/764,363 US10660856B2 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
CN201480019478.7A CN105102117A (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
PL14746507T PL2950923T3 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
ES14746507T ES2908958T3 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
EP14746507.4A EP2950923B1 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
JP2015556128A JP6513031B2 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technology
HK16105942.0A HK1217927A1 (en) 2013-02-01 2016-05-25 Porous silica gel as a carrier for liquid technologies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361759723P 2013-02-01 2013-02-01
US61/759,723 2013-02-01

Publications (1)

Publication Number Publication Date
WO2014120922A1 true WO2014120922A1 (en) 2014-08-07

Family

ID=51262940

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/013848 WO2014120922A1 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies

Country Status (9)

Country Link
US (1) US10660856B2 (en)
EP (1) EP2950923B1 (en)
JP (2) JP6513031B2 (en)
KR (1) KR102229036B1 (en)
CN (2) CN111389345A (en)
ES (1) ES2908958T3 (en)
HK (1) HK1217927A1 (en)
PL (1) PL2950923T3 (en)
WO (1) WO2014120922A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112020004197A2 (en) * 2017-09-01 2020-09-08 Jw Pharmaceutical Corporation solid preparation comprising dutasteride and method for preparing the same
JP7124107B2 (en) * 2018-10-05 2022-08-23 富士化学工業株式会社 Porous silica particle composition
CN111751480B (en) * 2020-07-16 2021-06-15 北京理工大学 Application of vitamin E polyethylene glycol succinate
CN116159031B (en) * 2023-02-22 2023-08-22 上海国创医药股份有限公司 Fluvoxamine maleate tablet and preparation method thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4157920A (en) 1975-03-12 1979-06-12 J. M. Huber Corporation Novel precipitated siliceous products and methods for their use and production
US4226743A (en) 1979-03-12 1980-10-07 W. R. Grace & Co. Silica-alumina hydrogel catalyst
US20030152771A1 (en) * 1999-04-22 2003-08-14 Preston Barry W. Very high structure, highly absorptive hybrid silica and methods for making same
WO2004073689A1 (en) 2003-02-19 2004-09-02 Lifecycle Pharma A/S Use of a silica or silica derivative as a sorption material
US20050112232A1 (en) * 2002-04-30 2005-05-26 Jean-Marie Dollat Animal feed supplement
US20090311159A1 (en) * 2006-08-22 2009-12-17 Evonik Degussa Gmbh Fumed silica for use as auxiliary in pharmaceutical and cosmetic compositions
WO2011144346A1 (en) * 2010-05-21 2011-11-24 Grace Gmbh & Co. Kg Porous inorganic oxide particles and methods of making and using the same
US20120269792A1 (en) * 2001-11-14 2012-10-25 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4629588A (en) * 1984-12-07 1986-12-16 W. R. Grace & Co. Method for refining glyceride oils using amorphous silica
US5256386A (en) * 1987-06-29 1993-10-26 Eka Nobel Ab Method for preparation of silica particles
KR100212946B1 (en) * 1991-03-16 1999-08-02 크리스토퍼 피아세키 Peat-derived bioactive products and pharmaceutical and cosmetic compositions containing them
DE4429644A1 (en) * 1994-08-20 1996-02-22 Sued Chemie Ag iodine adsorbent
US5800834A (en) 1996-06-10 1998-09-01 Spireas; Spiridon Liquisolid systems and methods of preparing same
US6096337A (en) 1996-06-10 2000-08-01 Spireas; Spiridon Liquisolid systems and methods of preparing same
JPH10130013A (en) * 1996-09-09 1998-05-19 Toyota Central Res & Dev Lab Inc Cluster inclusion material
FR2767071B1 (en) * 1997-08-06 1999-09-10 Rhodia Chimie Sa COMPOSITION COMPRISING A LIQUID ABSORBED ON A PRECIPITATED SILICA BASE
JPH11157827A (en) * 1997-11-21 1999-06-15 Shionogi & Co Ltd New silicon dioxide
SE9704400D0 (en) 1997-11-28 1997-11-28 Astra Ab Porous inorganic particles as carriers for drug substances
US6342249B1 (en) 1998-12-23 2002-01-29 Alza Corporation Controlled release liquid active agent formulation dosage forms
US20030236236A1 (en) * 1999-06-30 2003-12-25 Feng-Jing Chen Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
US6790814B1 (en) 1999-12-03 2004-09-14 Procter & Gamble Company Delivery system having encapsulated porous carrier loaded with additives, particularly detergent additives such as perfumes
GB2365769A (en) 2000-08-18 2002-02-27 Secr Defence Skin preparations containing silicon
DE10153078A1 (en) * 2001-10-30 2003-05-22 Degussa Use of granules based on pyrogenic silicon dioxide in pharmaceutical compositions
DE10244397A1 (en) 2002-09-24 2004-04-01 Basf Ag Choline formulations
FR2850576B1 (en) 2003-02-05 2007-03-23 Ethypharm Sa COMPOSITION COMPRISING A MIXTURE OF ACTIVE INGREDIENTS AND PROCESS FOR PREPARING THE SAME
FR2851918B1 (en) 2003-03-06 2006-06-16 IMPREGNATED POWDER ENHANCING BIOAVAILABILITY AND / OR SOLUBILITY AND METHOD OF MANUFACTURE
US20060167117A1 (en) 2003-03-27 2006-07-27 Leaym Tine M Controlled release compositions
GB0312703D0 (en) 2003-06-03 2003-07-09 Oxonica Ltd Agricultural compositions
JP2005053728A (en) * 2003-08-01 2005-03-03 Dsl Japan Co Ltd Amorphous silica particle having high oil absorption and high structural performance
JP2005053744A (en) * 2003-08-05 2005-03-03 Dsl Japan Co Ltd High oil absorptive amorphous silica particle
EP1702886A4 (en) * 2003-09-11 2011-02-16 Taiyo Kagaku Kk Porous silica having substance carried thereon
US20070042046A1 (en) 2003-10-21 2007-02-22 Roghieh Saffie Methods for the treatment of cancer
GB0406819D0 (en) 2004-03-26 2004-04-28 Dow Corning Controlled release compositions
JP2005307120A (en) * 2004-04-26 2005-11-04 Mitsubishi Chemicals Corp Sustained release agent and sustained release material using the same
GB0420016D0 (en) * 2004-09-09 2004-10-13 Leuven K U Res & Dev Controlled release oral delivery system
CN100352430C (en) * 2005-03-09 2007-12-05 山东大学 Self microemulsifying preparation of curcumin and its preparing process
WO2006119779A2 (en) 2005-05-10 2006-11-16 Lifecycle Pharma A/S A pharmaceutical composition comprising an aldosterone antagonist in form of solid solution
FR2894822B1 (en) 2005-12-20 2011-11-18 Pf Medicament PHARMACEUTICAL COMPOSITION CONTAINING OMEGA-3 FATTY ACIDS
US8920821B2 (en) 2006-04-14 2014-12-30 Perrigo Israel Pharmaceuticals Ltd. Pharmaceutical compositions comprising silica microspheres
AU2007247770B2 (en) 2006-05-04 2013-07-18 Reformpharm Pty Ltd Drug release from nanoparticle-coated capsules
US7691400B2 (en) 2006-05-05 2010-04-06 Medtronic Vascular, Inc. Medical device having coating with zeolite drug reservoirs
KR20080026754A (en) * 2006-09-21 2008-03-26 주식회사 삼양사 Release controlled particle comprising a biologically active substance, and preparing method thereof
WO2008128292A1 (en) 2007-04-20 2008-10-30 University Of South Australia Nanoparticle-coated capsule formulation for dermal drug delivery
JPWO2009057808A1 (en) 2007-11-02 2011-03-17 Aspion株式会社 Slightly soluble drug-surfactant complex product and production method thereof
EP2072042A1 (en) * 2007-12-21 2009-06-24 Lek Pharmaceuticals D.D. Active pharmaceutical ingredient on solid support, amorphous and with improved solubility
KR100950548B1 (en) * 2008-01-10 2010-03-30 연세대학교 산학협력단 A porous hollow silica nanoparticle, preparation method thereof, drug carrier and pharmacetical composition comprising the same
DE102008000290A1 (en) 2008-02-13 2009-08-20 Evonik Degussa Gmbh Storage stable product systems for premix formulations
CA2718255C (en) 2008-03-11 2016-08-23 Aska Pharmaceutical Co., Ltd. Solid dispersion and pharmaceutical composition of the same, and production processes thereof
US8188022B2 (en) 2008-04-11 2012-05-29 Amcol International Corporation Multilayer fragrance encapsulation comprising kappa carrageenan
US8778400B2 (en) 2008-04-21 2014-07-15 University Of South Australia Nanoparticle-stabilized capsule formulation for treatment of inflammation
CA2733297A1 (en) 2008-08-12 2010-02-18 Castle Beach, L.L.C. Active delivery system formulations
GB0821928D0 (en) 2008-12-01 2009-01-07 Isis Innovation Antibiotic nanomaterial
DE102010003204A1 (en) * 2010-03-24 2011-12-15 Evonik Degussa Gmbh Coarse particle carrier silicic acids
NZ608598A (en) 2010-09-07 2015-03-27 Orexo Ab A transdermal drug administration device
PL2833718T3 (en) 2012-04-02 2019-05-31 Basf Se Method of preparation of granules by milling pesticide and silica, addition of adjuvant, and fluidized bed granulation

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4157920A (en) 1975-03-12 1979-06-12 J. M. Huber Corporation Novel precipitated siliceous products and methods for their use and production
US4226743A (en) 1979-03-12 1980-10-07 W. R. Grace & Co. Silica-alumina hydrogel catalyst
US20030152771A1 (en) * 1999-04-22 2003-08-14 Preston Barry W. Very high structure, highly absorptive hybrid silica and methods for making same
US20120269792A1 (en) * 2001-11-14 2012-10-25 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system
US20050112232A1 (en) * 2002-04-30 2005-05-26 Jean-Marie Dollat Animal feed supplement
WO2004073689A1 (en) 2003-02-19 2004-09-02 Lifecycle Pharma A/S Use of a silica or silica derivative as a sorption material
US20090311159A1 (en) * 2006-08-22 2009-12-17 Evonik Degussa Gmbh Fumed silica for use as auxiliary in pharmaceutical and cosmetic compositions
WO2011144346A1 (en) * 2010-05-21 2011-11-24 Grace Gmbh & Co. Kg Porous inorganic oxide particles and methods of making and using the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2950923A4

Also Published As

Publication number Publication date
EP2950923A1 (en) 2015-12-09
JP2016508502A (en) 2016-03-22
JP2019048819A (en) 2019-03-28
HK1217927A1 (en) 2017-01-27
JP6861679B2 (en) 2021-04-21
CN105102117A (en) 2015-11-25
ES2908958T3 (en) 2022-05-04
EP2950923B1 (en) 2022-03-09
PL2950923T3 (en) 2022-04-04
JP6513031B2 (en) 2019-05-15
US10660856B2 (en) 2020-05-26
US20150366805A1 (en) 2015-12-24
KR102229036B1 (en) 2021-03-17
EP2950923A4 (en) 2016-10-19
CN111389345A (en) 2020-07-10
KR20150115860A (en) 2015-10-14

Similar Documents

Publication Publication Date Title
JP6861679B2 (en) Porous silica gel as a carrier for liquid technology
Tan et al. Silica-lipid hybrid (SLH) microcapsules: a novel oral delivery system for poorly soluble drugs
EP2919903B1 (en) Compositions containing a biologically active material and a non-ordered inorganic oxide
JP2015521175A (en) Cannabinoid-containing granule, process for producing the same, and oral dosage unit containing such granule
Choudhari et al. Comparative evaluation of porous silica based carriers for lipids and liquid drug formulations
Midha et al. Prospectives of solid self-microemulsifying systems in novel drug delivery
CN108079308A (en) Ubiquinone10With the nano structured lipid carrier and preparation method of n-octacosanol compounding
Shazly et al. Dissolution improvement of solid self-emulsifying drug delivery systems of fenofibrate using an inorganic high surface adsorption material
CN107921017A (en) The method treated using cadotril composition
US11484501B2 (en) Mechanochemical activated dry amorphisation by milling equilibrium between ap mesoporous silica
Singh et al. Polymeric delivery systems for poorly soluble drugs
US20220354791A1 (en) Mesoporous polymeric particulate material
Nandal et al. Novel Approaches of Self Emulsifying Drug Delivery Systems and Recent Patents: A Comprehensive Review
LETCHMANAN ENHANCING BIO-PHARMACEUTICAL PROPERTIES OF POORLY SOLUBLE ANTI-MALARIAL DRUGS BY FORMULATING AMORPHOUS FORMS WITH BIOCOMPATIBLE EXCIPIENTS
WO2016201119A1 (en) Excipient and oral solid dosage forms for oily drugs

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480019478.7

Country of ref document: CN

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

Ref document number: 14746507

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2014746507

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015556128

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20157023665

Country of ref document: KR

Kind code of ref document: A