CA2212803C - Redispersible nanoparticulate film matrices with protective overcoats - Google Patents

Redispersible nanoparticulate film matrices with protective overcoats Download PDF

Info

Publication number
CA2212803C
CA2212803C CA002212803A CA2212803A CA2212803C CA 2212803 C CA2212803 C CA 2212803C CA 002212803 A CA002212803 A CA 002212803A CA 2212803 A CA2212803 A CA 2212803A CA 2212803 C CA2212803 C CA 2212803C
Authority
CA
Canada
Prior art keywords
composition according
drug
agents
solid carrier
composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002212803A
Other languages
French (fr)
Other versions
CA2212803A1 (en
Inventor
Mark A. Desieno
Gregg Stetsko
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Elan Pharma International Ltd
Original Assignee
Nanosystems LLC
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 Nanosystems LLC filed Critical Nanosystems LLC
Publication of CA2212803A1 publication Critical patent/CA2212803A1/en
Application granted granted Critical
Publication of CA2212803C publication Critical patent/CA2212803C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

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
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface
    • A61K9/1676Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface having a drug-free core with discrete complete coating layer containing drug
    • 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
    • A61K9/146Intimate 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 organic macromolecular compounds

Landscapes

  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Paints Or Removers (AREA)

Abstract

A pharmaceutical film matrix comprising nanoparticles of a low solubility drug associated with a steric stabilizer and a film dispersing agent is coated on the surface of carrier particles and overcoated with a protective layer.
Pharmaceutical compositions comprising the film matrix-coated carrier exhibit excellent bioavailability, are useful in methods of treating mammals and are extremely stable in that they are capable of being redispersed without extensive agglomeration into larger particles after being stored.

Description

REDISPERSIBLE NANOPARTICULATE FILM MATRICES
WITH PROTECTIVE OVERCOATS
Field Of The Invention This invention.relates to film matrixes of nanoparticulate drug particles and pharmaceutical compositions containing them. The invention further relates to the use of and compositions for treating mamma 1 s .
Background of Invention Bioavailability is the degree to which a drug becomes available to the target tissue after administration. Many factors can affect bioavailability including the dosage form and various properties, e.g., dissolution rate of the drug. Poor -bioavailability is a significant problem encountered in the development of pharmaceutical compositions, particularly those containing an active ingredient that is poorly soluble in water. Poorly water soluble drugs, i.e., those having a solubility less than about 10 mg/ml, tend to be eliminated from the gastrointestinal tract before being absorbed into the circulation. Moreover, poorly water soluble drugs tend to be unsafe for intravenous administration techniques, which are used primarily in conjunction with fully soluble drug substances.
It is known that the rate of dissolution of a particulate drug can increase with increasing surface area, i.e., decreasing particle size. Consequently, methods of making finely divided drugs have been studied and efforts have been made to control the size and size range of drug particles in pharmaceutical compositions. For example, dry. milling techniques have been used to reduce particle size and hence influence drug absorption. However, in conventional dry milling, as discussed by Lachman, et al., The Theory and 28516-31(S) Practice of Industrial Pharmacy, Chapter 2, "Milling", p. 45 (1986), the limit of fineness is reached in the region of 100 microns (100,000 nm) when material cakes on the milling chamber. Lachman, et al. note that wet grinding is beneficial in further reducing particle size, but that flocculation restricts the lower particle size limit to approximately 10 microns (10,000 nm). However, there tends to be a bias in the pharmaceutical art against wet milling due to concerns associated with contamination. Commercial airjet milling techniques have provided particles ranging in average particle size from as low as about 1 to 50 dim (1,000-50,000 nm).
U.S. Patent No. 5,145,684 discloses particles of a drug substance having a surface modifier absorbed on the surface thereof and methods for the preparation thereof by wet grinding. These particles have demonstrated significant pharmaceutical utility.
Suitable surface modifiers described include various polymers The surface modifiers disclosed include -x Pluronic F68 and F108, which are block copolymers of ethylene oxide and propylene oxide, '~etronie 908, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to~ethylene diamine; sodium dodecylsulfate, dialkyl esters of sodium sulfosuccinic acid such as Aerosol OTT"", sodium lauryl sulfate, and TritonT"" X-200.
This patent notes the difficulty of forming nanoparticulate drug containing compositions which have good bioavailability and also are generally impervious to increase in size due to agglomeration of drug particles.
It is desirable to provide a pharmaceutical composition containing a nanoparticulate drug which can be stored for periods of time without undue loss of *Trade-mark bioavailability due to agglomeration into larger size particles.
Summarv of the Invention We have discovered stable film matrixes containing nanoparticulate drug particles which can be coated on to a carrier particle to be formulated into pharmaceutical compositions exhibiting high bioavailability.
More specifically, in accordance with this invention, there are provided pharmaceutical film matrices comprising nanoparticles of a low solubility drug associated with a steric stabilizer and a film dispersing agent wherein dissolution of the film results in substantial redispersion of the drug nanoparticles.
This invention also provides a stable pharmaceutical composition comprising a carrier particle having on its surface a film comprising nanoparticles of a low solubility drug associated with a steric stabilizer and a film dispersing agent, wherein dissolution of the film results in substantial redispersion of the drug nanoparticles.
In a particularly valuable and important embodiment of the invention there is provided a pharmaceutical composition comprising a carrier particle having on its surface a film comprising nanoparticles of a low solubility drug associated with a steric stabilizer and a film dispersing agent, wherein dissolution of the film results in substantial redispersion of the drug nanoparticles and an overcoat applied over said film comprising polyvinylpyrrolidone/polyethylene glycol (PVP/PEG).

28516-31(S) 3a According to one aspect of the present invention, there is provided a pharmaceutical composa.tion comprising:
(a) a solid carrier, and coated onto the solid carrier a film comprising: (b) a nanoparticulate drug matrix composition comprising: (1) a nanoparticulate crystalline drug which has a solubility of less than about l0mg/ml in water and is dispersible in at least one liquid medium; (2) a steric stabilizer associated with the surface of the nanoparticulate drug; and (3) a water-soluble film dispersing agent, wherein the agent is associated with the surface of the nanoparticulate drug; wherein dissolution of t:he solid carrier results :in substantial redispersion of particles of the nanoparticulate drug.
The pharmaceutical compositions of this invention are useful in a method of treating mammals with a variety of drugs.
It is a particularly advantageous feature of this invention that the pharmaceutical compositions described herein exhibit high bioavailability.
Still another advantageous feature of this invention is that the compositions are stable against agglomeration of the drug nanoparticles into larger sizes because the compositions are readily redispersible.
Other advantageous features will become readily apparent upon reference to the following Description of Preferred Embodiments.
Description of Preferred Embodiments This invention is based partly on the discovery that drug particles having an extremely small effective average particle size can be prepared by wet milling in the presence of grinding media in conjunction with a surface modifier, and that such particles are stable and do not appreciably flocculate or agglomerate due to interparticle attractive forces and can be formulated into pharmaceutical compositions exhibiting unexpectedly high bioavailability. While the invention is described herein primarily in connection with its preferred utility, i.e., with respect to nanoparticulate drug substances for use in pharmaceutical compositions, it is also believed to be useful in other applications such as the preparation of particulate dispersions for use in image and magnetic recording elements.
The particles of this invention comprise a drug substance. The drug substance exists as a discrete, crystalline phase_ The crystalline phase differs from a noncrystalline or amorphous phase which results from precipitation techniques, such as described in EPO 275,796 cited above.
The invention can be practiced with a wide variety of drug substances. The drug substance preferably in present in an essentially pure form. The drug substance must be poorly soluble and dispersible in at least one liquid medium. By "poorly soluble" it is meant that the drug substance has a solubility in water of less than about 10 mg/ml and preferably-less than about 1 part in greater than 1,000,000 parts of water. A preferred liquid dispersion medium is water.
However, the invention can be practiced with other liquid media in which a drug substance is poorly soluble and dispersible including, for example, aqueous salt solutions, safflower oil and solvents such as ethanol, t-butanol, hexane and glycol. The pH of the aqueous dispersion media can be adjusted by techniques known in the art.
Suitable drug substances can be selected from a variety ofknown classes of drugs including, for example, analgesics, anti-inflammatory agents, anthelmintics, anti-arrhythmic agents, antibiotics (including penicillins), anticoagulants, antidepressants, antidiabetic agents, antiepileptics, antihistamines, antihypertensive agents, antimuscarinic agents, antimycobacterial agents, antineoplastic agents, immunosuppressants, antithyroid agents, antiviral agents, anxiolytic sedatives (hypnotics and neuroleptics), astringents, beta-adrenoceptor blocking .agents, blood products and substitutes, cardiac inotropic agents, contrast media, corticosteroids, cough suppressants (expectorants and mucolytics), diagnostic agents, diagnostic imaging agents, diuretics, dopaminergics (antiparkinsonian agents), haemostatics immuriological agents lipid regulating 28516-31(S) agents, muscle relaxants, parasympathomimetics, parathyroid calcitonin and biphosphonates, prostaglandins, radio-pharmaceuticals, sex hormones (including steroids), anti-allergic agents, stimulants and anoretics, sympathomimetics, thyroid agents, vasodilators and xanthines. Preferred drug substances include those intended for oral administration and intravenous administration. A description of these classes of drugs and a listing of species within each class can be found in Martindale, The Extra Pharmacopoeia, Twenty-ninth Edition, The Pharmaceutical Press, London, 1989. The drug substances are commercially available and/or can be prepared by techniques known in the art.
Preferred and representative illustrative species of drug substances useful in the practice of this invention include danazol, naproxen and cylosporin.
The matrix of this invention comprises a steric stabilizer. The steric stabilizer is a hydrophilic or amphophilic compound having_steric stabilizing properties such as a physical or chemical anchored molecule with backbone and side chains which import a.physical barrier to interaction of the small particles. Examples of steric stabilizers can be found in Lieberman, Rieger, Banker Pharmaceutical Dosage Forms: Disperse Systems, Volume 1.
Preferred steric stabilizers are gelatin, casein, lecithin, gum acacia, cholesterol, tragacanth, sorbitan esters, polyoxyethylene alkyl esters, polyoxyethylene caster oil derivatives, polyoxyethylene, sorbitan fatty acid esters, polyethylene glycols, polyoxyethylene stearates, carboxymethylcellulose calcium, carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, phthalate, non crystalline cellulose, polyvinyl alcohol and polyvinylpyrrolidone.
d-Alpha tocopheryl polyethylene glycol 1000 succinate is described by the manufacturer (Eastman Chemical Products, Inc., a division of Eastman Kodak Company, Kingsport, Tennessee 37622) in a product brochure dated February 4, 1983 as "prepared from crystalline d-Alpha Tocopheryl Acid Succinate NF by esterification of the acid group with polyethylene glycol 1000", as also being named "Vitamin E TPGS", as being a "[p]ale yellow, waxy solid" having a specific gravity at 45°C. of approximately 1.06 and a melting point of approximately 40°C., and in the opinion of the manufacturer as being "recognized as safe ("GRAS") when used as an oral dietary supplement of vitamin E."
The preferred amount of each of the cellulose derivative, polyethylene glycol or derivative thereof and waxy solid in the first composition of matter aspect of the invention is from about 5% to about 300 by weight of the coating composition.
The preferred amount of each of the hydropropyl methylcellulose, polyethylene glycol and polyoxyethylene-polyoxypropylene-polyoxyethylene block copolymer in the preferred ffirst composition of matter aspect of the invention is from about 5o to about 150 by weight of the coating composition.
The most preferred steric stabilizer is polyva.nylpyrrolidone_ The film matrix also comprises a film dispersing agent. The film dispersing agent is a water soluble compound, associated with the surface of the nanoparticle_ Film dispersing agents are those materials which oppose the binding capability of the R'O 96/25150 PCTIUS96/01845 _8_ film when placed in an aqueous environment enhancing film solubility or by swelling and disrupting films.
Preferred film dispersing agents are mannitol, sodium lauryl sulfate, mono and disaccharides. Sodium lauryl sulfate is particularly preferred. Sodium lauryl sulfate is also named as "[s]ulfuric acid monododecyl ester sodium salt; sodium starch glycolate; croscarmellose sodium" and "[s)odium monododecyl sulfate" and is described as ...a mixture of sodium alkyl sulfates consisting chiefly of sodium lauryl sulfate [CH3(CH2)10CH20S03Na].
The combined content of sodium chloride and sodium sulfate is not more than 8.0 percent.
In the dissolution step of the process aspect of the invention the temperature of warming can be in the range from room temperature to about 100°C. and is preferably in the range from 50°C. to 60°C. About 80~
of the total amount of water needed is used for the dissolution and suspension steps and the remainder is used for rinsing the last amounts of solution and suspension from the equipment. Preferably the polyethylene glycol or derivative thereof and the waxy solid are dissolved first, and the cellulose derivative is then added and dissolved. .The low solubility drug is added to the resulting solution with agitation to form a suspension. The dissolution and suspension steps are carried out with conventional mixing equipment. The suspension is preferably passed through a colloid mill before carrying out the coating step and agitation is maintained during the coating step. The coating and drying steps are preferably carried out in a fluid bed processor with inlet air temperature in the range from 50°C. to 70°C. with preheating of the sugar or sugar/starch beads. After drying the coated beads _g_ are sifted to produce coated beads of the desired particle size, preferably 16-60 mesh.
Preferred film matrices comprise nanoparticles of a low solubility drug having about 0.1% to about 60% of a steric stabilizer and about 0.05 to about 50% of a film dispersing agent, based on the weight of the low solubility drug, associated with the nanoparticle surface.
In a particularly preferred embodiment of this invention, the film matrix is overcoated with polyvinylpyrrolidone (PVP)/polyethylene glycol (PEG).
This overcoat provides improved stability to the pharmaceutical composition.
The overcoating is prepared by casting PVP
such as Providone with different types and units of PEG
such as PEG 600, PEG 1000 and PEG 1450 and mixtures thereof. Polyethylene glycol is described as "an addition polymer of ethylene oxide and water, represented by the formula H(OCH2CH2)nOH in which n represents the average number of oxyethylene groups."
The variants are-designated by "nominal value" of "average molecular weight", which is explained as follows:
The average molecular weight is not less than 95.0 percent and not more than 105.0 percent of the labeled nominal value if the labeled nominal value is below 1000; it is not less than 90.0 percent and note more than 110.0 percent of the labeled nominal value if the labeled nominal value is between 1000 and 7000; it is not less than 87.5 percent and not more than 112.5 percent of the labeled nominal value if the labeled nominal value is about 7000_ Polyethylene glycols having nominal average molecular weights in the range from 300 to 8000 are described. Polyethylene glycol 3350 is the preferred polyethylene glycol of the invention_ Providone, USP is the USAN name for 2-Pyrrolodinone, 1-ethenyl-homopolymer of 1-vinyl-2-pyrrolidinone polymer. It is sold under the trademarks KOLIDON (BASF), PLASDONE (International Specialty Products) and VINISIL (Abbott).
The weight ratio of PEG/PVP is generally from 1 to about 5_ The pharmaceutical film matrix is formed by dissolving the PEG and PVP in a suitable solvent.
Suitable carriers comprise sugar spheres, maltodextrin, microcrystalline cellulose, microcrystal cellulose/sodium carboxylmethylcellulose, granular dextrose, dicalcium phosphate, tricalcium phosphate, mono and discaccarides_ The preferred carriers are sugar spheres. Sugar Spheres are described as containing "not less than 62.5 percent and not more than 91.5 percent of surose (C12H22011). calculated on the dried basis, the remainder consisting chiefly of starch" and as consisting of "approximately spherical particles of a labeled nominal size range" and correspond to-the sugar or sugar/starch beads of the invention. They can also be or be referred to as granules, particles, pellets or nonpareils and are from about 2 millimeters or about 10 mesh to about 0.2 millimeter or about 80 mesh, preferably from about 20 mesh to about 70 mesh, in diameter or longest dimension before coating. After coating the preferred diameter or longest dimension is from about 16 mesh to about 60 mesh.
The overcoat can be coated over the film by spraying the solution onto the substrate by using a vented pan or fluid bed coating unit with inlet air temperature of between 40-80°C.

The dispersion of surface modified nanoparticles can be spray coated onto sugar spheres or onto a pharmaceutical excipient in a fluidized bed.
' The spray coating is accomplished by techniques well known in the art. Sugar spheres are described as containing "not less than 62.5 percent and not more than 91 _ 5 percent of surose ~C12H22~11) , calculated on the dried basis, the remainder consisting chiefly of startch" and as consisting of "approximately spherical particles of a labeled nominal size range" and correspond to the sugar or sugar/starch beads of the invention. They can also be or be referred to as granules, particles, pellets or nonpareils and are from about 2 millimeters or about 10 mesh to about 0.2 millimeter or about 80 mesh, preferably from about_20 mesh to about 70 mesh, in diameter or longest dimension before coating. After coating the preferred diameter or longest dimension is from about 16 mesh to about 60 mesh.
The composition can be formed into a capsule shell. The capsule shell of this composition of matter can be any pharmaceutically accpetable capsule shell but is preferably a gelating capsule shell and can be a soft gelatin capsule shell or a hard gelatin capsule shell but is preferably a hard gelatin capsule shell and is of suitable size for containing from about 0.5 milligram to about 700 milligrams of the composition of-matter. Conventional machinery and technique are used in filling the capsule shells.
The particles of this invention can be prepared in a method comprising the steps of dispersing a drug substance in a liquid dispersion medium and applying mechanical means in the presence of grinding media to reduce the particle size of the drug substance to an effective average particle size of less than about 400 nm. The particles can be reduced in size in the presence of a surface modifer. Alternatively, the particles can be contacted with a surface modifier after attrition.
A general procedure for preparing the particles of this invention is set forth below. The drug substance selected is obtained commercially and/or prepared by techniques known in the art in a conventional coarse form. It is preferred, but not essential, that the particle size of the coarse drug substance selected be less than about 100 um as determined by sieve analysis. 2f the coarse particle size of the drug substance is greater than about 100 m, then it is preferred that the particles of the drug substance be reduced in size to less than 100 dun using a conventional milling method such as airjet or fragmentation milling.
The coarse drug substance selected can then be added to a 11qu1d medium in which it is essentially insoluble to form a premix. The concentration of the drug substance in the liquid medium can vary from about 0.1-60%, and preferably is from 5-30%(w/w). It is preferred, but not essential, that the surface modifier be present in the premix. The concentration of the surface modifier can vary from about 0.1to about 90%, and preferably is 1-75%, more preferably 20-60%, by weight based on the total combined weight of the drug substance and surface modifier. The apparent viscosity of the premix suspension is preferably less than about 1000 centipoise.
The premix can be used directly by subjecting it to~mechanical means to reduce the average particle size in the dispersion to less than 400 nm. It is preferred that the premix be used directly when a ball mill is used for attrition. Alternatively, the drug substance-and, optionally, the surface modifier, can be dispersed in the liquid medium using suitable agitation, e.g., a roller mill or a Cowles type mixer, until a homogeneous dispersion is observed in which ' there are no large agglomerates visible to the naked eye. 2t is preferred that the premix be subjected to ~ such a premilling dispersion step when a recirculating media mill is used for attrition.
The mechanical means applied to reduce the particle size of the drug substance conveniently can take the form of a dispersion mill. Suitable dispersion mills include a ball mill, an attritor mill, a vibratory mill, and media mills such as a sand mill and a bead mill. A media mill is preferred due to the relatively shorter milling time required to provide the intended result, i.e., the desired reduction in particle size. For media milling, the apparent viscosity of the premix preferably, is from about 100 to about 1000 centipoise. For ball milling, the apparent viscosity of the premix preferably is from about 1 up to about 100 centipoise. Such ranges tend to afford an optimal balance between efficient particle fragmentation and media erosion.
The grinding media for the particle size reduction step can be selected from rigid media preferably spherical or particulate in form having an average size less than about 3 mm and, more preferably, less than about 1 mm. Such media desirably can provide the particles of the invention with shorter processing times and impart less wear to the milling equipment.
The selection of material for the grinding media is not believed to be critical. 44e have found that zirconium oxide, such as 95o Zr0 stabilized with magnesia, zirconium silicate, and glass grinding media provide particles having levels of contamination which are believed to be acceptable for the preparation of pharmaceutical compositions. However, other media, such as stainless steel, titania, alumina, and 95~ Zr0 stabilized with yttrium, are expected to be useful.
Preferred media have a density greater than about 3 g/cm3.
The attrition time can vary widely and depends primarily upon the particular mechanical means and processing conditions selected. For ball mills, processing timesof up to five days or longer may be required. On the other hand, processing times of less than 1day (residence times of one minute up to several hours) have provided the desired results using a high shear media mill.
The particles must be reduced in size at a temperature which does not significantly degrade the drug substance. Processing temperatures of less than about 30°C-40°C_ are ordinarily preferred. If desired, the processing equipment can be cooled with conventional cooling equipment. The method is conveniently carried out under conditions of ambient temperature and at processing pressures which are safe and effective for the milling process. For example, ambient processing pressures are typical of ball mills, attritor mills and vibratory mills. Processing pressures up to about 20 psi (1.4 kg/cm2) are typical of media milling.
The surface modifier, if it was not present in the premix, must be added to the dispersion after attrition in an amount as described for the premix above. Thereafter, the dispersion can be mixed, e.g., by shaking vigorously. Optionally, the dispersion can be subjected to a sonication step, e.g., using an ultrasonic power supply. For example, the dispersion can be subjected to ultrasonic energy having a frequency of 20-80 kHz for a time of about 1 to 120 seconds.

The relative amount of drug substance and surface modifier can vary widely and the optimal amount of the surface modifier can depend, for example, upon the particular drug substance and surface modifier selected, the critical micelle concentration of the surface modifier if it forms micelles, etc. The surface modifier preferably is present in an amount of about 0.1-10 mg per square meter surface area of the drug substance. The surface modifier can be present in an amount of 0.1-900, preferably 20-60o by weight based on the, total weight of the dry particle.
As indicated by the following examples, not every combination of surface modifier and drug substance provides the desired results. Consequently, the applicants have developed a simple screening process whereby compatible surface modifiers and drug substances can be selected which provide stable dispersions of the desired particles. First, coarse particles of a selected drug substance of interest are dispersed in a liquid in which the drug is essentially insoluble, e.g., water at 5~(w/w) and milled for 60 minutes in a DYNO-MILL under the standard milling conditions which are set forth in Example 1 which follows. The milled material is then divided into aliquots and surface modifiers are added at concentrations of 2, 10 and 50o by weight based on the total combined weight of the drug substance and surface modifier. The dispersions are then sonicated (1 minutes, 20 kHz) to disperse agglomerates and subjected to particle size analysis by examination under an optical microscope (1000 x magnification). If a stable dispersion is-observed, then the process for preparing the particular drug substance surface modifier combination can be optimized in accordance with the teachings above. By stable it is meant that the dispersion exhibits no flocculation or particle WO 96/25150 PCT/i1S96/01845 agglomeration visible to the naked eye at least 15 minutes, and preferably, at least two days or longer after preparation.
The resulting dispersion of this invention is stable and consists of the liquid dispersion medium and the above-described particles. The dispersion of surface modified drug nanoparticles can be spray coated onto sugar spheres or onto a pharmaceutically excipient in a fluidbed spray coater by techniques well known in the art.
Pharmaceutical compositions according to this invention include the particles described above and a pharmaceutically acceptable carrier thereof. Suitable pharmaceutically acceptable carriers are well known to those spilled in the art. These include non-toxic physiologically acceptable carriers, adjuvants or vehicles for parenteral injection, for oral administration in solid or liquid form, for rectal administration, and the like. A method of treating a mammal in accordance with this invention comprises the step of administering to the mammal in need of treatment an effective amount of the above-described pharmaceutical composition. The se-lected dosage level of the drug substance for treatment is effective to obtain a desired therapeutic response for a particular composition and method of administration. The selected dosage level therefore, depends upon the particular drug substance, the desired therapeutic effect, on the route of administration, on the desired duration of treatment and other factors. As noted, it is a particularly advantageous feature that the pharmaceutical compositions of this invention exhibit unexpectedly high bioavailability as illustrated in the examples which follow. Furthermore, it is contemplated that the drug particles of this invention provide more rapid onset of drug action and decreased gastrointestinal irritancy.
It is contemplated that the pharmaceutical compositions of this invention will be particularly useful in oral and parenteral, including intravenous, administration applications. It is expected that poorly water soluble drug substances, which prior to this invention, could not have been administered intravenously, may be administered safely in accordance with this invention. Additionally, drug substances which could not have been administered orally due to poor bioavailability may be effectively administered in accordance with this invention.
The following composition in accordance with the first composition of matter aspect of the invention was prepared using the process aspect of the invention.
Examt~le 1 Ingredient Amount (kg.) (5a,17a)-1'-(Methylsulfonyl)-1'H-pregn-20- 0_72 0 yno(3,2-c]pyrazol-17-0l Poloxamer 188, NF 0.0900 Polyethylene Glycol 3350, NF 0.144 Hydroxypropyl Methylcellulose, USP ~ 0.100 Sugar Spheres ( 3 0 , 3 5 mesh ) , NF' 0 . 4 5 0 Purified Water, USP (removed during processing) 2.4 6 Total amount of dry ingredients 1.500 A portion of this composition sufficient to provide 200 mg. of the steroid drug when filled into a hard gelatin capsule has the following composition:

Ingredient Ma_/Capsule (5a,17a.)-1'-(Methylsulfonyl)-1'H-pregn-20- 2 0 0.
yno[3,2-c)pyrazol-17-0l Poloxamer 188, NF 25.0 ~
Polyethylene Glycol 3350, NF 4 0.0 Hydroxypropyl Methylcellulose, USP 2 7.8 Sugar Spheres (30-,35 mesh), NF 12 5.
Total Capsule Fill Weight 4 18.
The amount of drug in each capsule can be varied by varying the capsule fill weight, the amount of drug in the coating composition or the amount of coating composition coated onto the sugar or sugar/starch beads.
The composition of Example 1 was shown to have improved bioavailability over a conventional tablet composition of the same drug when compared in the dog. The following conventional tablet composition was prepared using a conventional table preparation method.

28516-31(S) romparat~ve Example Inaredient Ma./Tablet (5a,17a)-1'-(Methylsulfonyl)-1'H-pregn-20- 50.0 yno[3,2-c]pyrazol-17-0l Microcrystalline Cellulose, NF (Avicel~pH 101) 60.0 Poloxamer~188, NF 6.0 Lactose, NF (Spray Dry) 161.5 Croscarmellose Sodium, FN (Ac-Di-Sol) 15.0 Magnesium, Stearate, NF 1.5 Providone* USP (PVP, K29-32) 6.0 Total 300.0 Example 2 - Preparation of a Pharmaceutical Composition Two danazol dispersions were prepared by dissolving the PVP in water, dispersin the danazol substance, and milling by use of an agitating bead mill. The dispersion was milled to a final mean particle size of between 150-250 nm.
The pharmaceutical compositions were prepared by spray coating onto 5 different solid carrier (sugar beads, granular sugar, maltodextrin, Avicel*p~-I 200 and Avicel*CL 611) at varying levels of film dispersing agent (additive) and danazol content as follows.
*Trade-mark 28516-31(S) Run # Additives Ratio crams Substrate Of Dana2ol/MZ
Addltlv substrate a to Danazol mg/gm 1 Blg-1186-074 low low Sugar Beads 2 Blg-1186-063 high high Maltodextrin 3 Blg-1186-075 low low Granular sugar 4 Blg-1186-060 high high Granualar sugar Blg-1186-069 low high Sugar Beads 6 Blg-1186-072 low high Avicel~'PH200 7 Blg-1186-076 low low Avicel'~PH200 8 Blg-1186-073 low high Avice h'CL611 9 Blg-1186-061 high high Aviceh'PH200 Blg-1186-071 low high Maltodextrin 11 Blg-1186-065 high low Granular sugar 12 Blg-1186-077 low low Avicel'~CL611 13 Blg-1186-066 high low Avicel~PH200 14 Blg-1186-068 high low Maltodextrin Blg-1186-078 low low Maltodextrin 16 Blg-1186-059 high high Sugar Beads 17 Blg-1186-067 high low Avicel*CL611 18 Blg-1186-070 low high Granular sugar 19 Blg-1186-062 high high Avice h'CL611 !.

Blg-1186-064 high low Sugar Beads *Trade-mark 28516-31(S) i The beads were tested for aqueous dissolution rate using a distek 6 vessel dissolution bath at 37°C, stirring at 300 rpm using paddles. Test media was 1 liter distilled water, equilibrated to 37°C. A
*
stainless steel sampling probe was connected to Tygon tubing, and an Ismatec' SA peristaltic pump was used to control flow at -0.18 ml/min. An inline 0.020 ~zn filter was used to remove particulate danazol and carrier debris and to ensure that danazol detected was actually in solution. A fresh filter was used for each test vessel. An amount of formulation equivalent to 20 mg of danazol was added to the test vessel and absorbance was monitored continuously for 20 minutes at 285 nm using a Waters 990 Photodiode Array system. The Waters 990 software was then used to form a rate plot by taking the differential of the absorbance vs. time curve. The maximum point on the rate plot was noted as the maximum rate of aqueous dissolution. The two Avicel carriers used in this study were both insoluble.
The Avicel PH 200 was sized at approximately 60 Nm (uncoated) and retained its form as an insoluble particle during the dissolution process. Because of its size, a 10 ~.~m solvent filter was placed on the end of the stainless steel sampling probe preventing clogging~of the probe and the 0.020 dim in-line filter.
The Avicel CL 611 carrier was slightly smaller in size than the Avicel PH 200 in powder form, but immediately- swelled in the presence of water to a size of approximately 60 ~tm. During the dissolution assay, the constant agitation and 37°C temperature caused the Avicel CL 611 to break up into gelatinous particles in the 200 nm size range. All attempts to filter these particles failed. The gelatinous form of *
the Avicel CL 611 quickly and consistently clogged 0.2 N.m and 0.1 ~m filters placed inline before the 0.02 Nm *Trade-mark 28516-31(S) filter. For this reason, no aqueous dissolution data was obtained for the Avicel*CL 611 formulations.
Simple preparation for Acxueous Redisnersion Particlg Size Analysis -High Danazol Content Formulations: An amount of formulation equivalent to 50 mg of danazol was weighed into a 4 ml glass vial to which 1 ml of distilled water (room temperature) was added. The vial was capped and contents vortexed vigorously for 10 seconds. The prepared sample were then shaken at 300 rpm, 37°C for 10 minutes using a LabLine Shaker Incubator, after which particle sizing was performed. All samples were prepared in duplicate.
-Low Danazol Content Formulations: An amount of formulation equivalent to 50 mg of danazol was weighed into a 20 ml glass scintillation vial to which 5 ml of room temperature distilled water was added. The vial was capped and contents vortexed vigorously for 10 seconds. The prepared samples were then shaken at 300 rpm, 37°C for 10 minutes using a LabLine Shaker Incubator, after which particle sizing was performed.
All samples were prepared in duplicate.
".
-Special note for Avicel PH 200 samples: The Avicel PH
200 carrier was removed by filtering through a 5 ~.im filter just prior to sizing.
-Special note for Avicel CL 611 samples: Since the Avicel CL 611 carrier itself was breaking into the nanometer size range of danazol, no usable redispersion sizing data were obtained for these formulations.
-Sizing using the Zetasizer III: Samples were diluted in filtered (0,.45 elm) distilled water, and degassed before analysis. The AZ4 cell was used for all sizing procedures. Run time for size determinations was typically 120 seconds. Output from the Zetasizer III
*Trade-mark consisted of a mean particle size in nm and a 90%<value.
-Sizing using the Coulter N4MD: Samples were briefly vortexed then diluted 10 x..1.1 to -15 ml in filtered (0.22 Nm) deionized water. Diluted samples were shaken to obtain a homogeneous suspension. A 4.5 ml cuvette was cleaned and filtered (0.22 E.tm) deionized water, filled half way with filtered water, and sonicated for -5 seconds to remove air bubbles. A minimal amount of the diluted sample was then added to impart a slight opaqueness to the contents of the cuvette. The cuvette was capped and inverted several times to allow proper mixing. The outside of the cuvette was siped completely dry and clean and placed into the sizing chamber which was maintained at 37°C. The viscosity setting was 0.693 cp and the refractive index was set to 1.331. The cuvette contents were adjusted as necessary to achieve a sample intensity in the range of 1.5 x 105 to 2.5 x 105 counts/second. Run time for particle sizing was typically 200 seconds. Output consisted of a mean particle size in nm, standard deviation, and a o Dust value.
Sugar Bead Formulations Maximum aqueous dissolution rate values and redispersion particle sizing data-are found in Table 2.
The data clearly shows that the formulations containing the "high" additive ratio (Run #20 and #16) exhibited higher maximum aqueous dissolution rates and .smaller particle sizes upon redispersion in water than their "low" additive ratio counterparts.
Granular Dextrose Formulations Maximum aqueous dissolution rate values and redispersion particle sizing data are found in Table 3.
On the granular matrix, there appears to be little difference in maximum aqueous dissolution rate among 28516-31(S) the 4 formulations. It can be seen, however, that the "high" additive ratio formulations (Run #11 and #4) resulted in smaller particle sizes upon redispersion compared to the two "low" additive ratio formulations.
Maltodextrin Formu~tions Maximum aqueous dissolution rate values and redispersion particle sizing data are found in Table 4.
The same pattern is observed here as in the granular dextrose formulations. The maximum aqueous dissolution rates are all high and similar. The "high" additive ratio formulations (Run #14 and #2) exhibit smaller redispersion particle sizes than the "low" additive formulations.
Avicel PH 200 Formu:~iations Maximum aqueous dissolution rate values and redispersion particle sizing data are found in Table 5.
Results from the Avicel PH 200 matrix formulations closely mimic results of those formulations on the sugar bead matrix. The "high" additive ratio formulations (Run #13 and #9) exhibited higher maximum aqueous dissolution rates compared to the formulations containing the "low" additive ratio, and their redisper~ion particle sizes were much smaller as well, keeping with the trend observed in the previous three matrices.
*Trade-mark Table 2 Bead Formulations Run #1 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 5.3 3.6 299 670 371 180 17 3.4 3.4 308 680 347 150 8 4.6 3.8 Mean 4.4 3.6 304 359 SD 1.0 0.2 6 17 Run #5 Low/LowMaximum Time Size 90% CoulterSD % Dust to (nm) <Value Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10'5 AU/min) 3.1 4.4 271 580 274 45 18 3.6 3.8 283 620 >3~

3.1 4.2 Mean 3.3 4.1 277 >3~

S D 0.3 0.3 8 Run #20 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 6.4 2.0 223 430 268 100 0 8.4 1.8 224 45~ 279 50 0 8.7 2.4 Mean 7.8 ~, 2.1 224 274 SD 1.3 0.3 1 g WO 96!25150 PCT/US°._ Run #16 Low/LowMaximum Time Size 90%<ValueCoulterSD
to (nm) % Dust Aqueous Maximum (nm) N4MD
Size DissolutionRate (~) Rate (min) (x10-5 AU/tnin) 8.0 2.6 213 410 240 37 8.8 2.6 215 420 224 63 7.4 2.8 Mean 8.1 2.7 214 232 SD 0.7 0.1 1 11 Table 3 Granular Dextrose Formulations Run #3 Low/LowMaximum Time Size 90%<ValueCoulterSD
to (tun) % Dust Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 7-4 2.8 241 500 262 48 9 6.6 2.8 239 520 255 66 7 6.0 2.6 Mean 6.7 2.7 240 259 SD 0.7 0.1 1 5 Run #18 Low/L,owMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Ayueoue Maximum (nm) N4MD
Size DissolutionRate (~) Rate (min) (x10-5 AU/min) 8.4 2.8 306 700 289 36 10 5.8 2.8 315 710 272 50 16 6.5 3.0 Mean 6.9 2.9 311 281 SD 1.3 0.1 6 12 WO 96/25150 PCTlUS96/01845 Run #11 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm)-Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 6.0 2.8 210 380 229 26 0 7.0 3.0 205 400 224 62 g 6.7 2.8 Mean 6.6 2.9 208 227 SD 0.5 0.1 4 4 Run #4 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 9.0 2.8 214 390 211 29 g 7.2 2.6 216 420 240 42 1 9.0 2.4 Mean 8.4 2.6 215 226 SD 1.0 0.2 1 21 Table 4 Maltodextrin Formulations Run #15 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate ( Rate (min) (x10-~
AU/min) 7.0 3.0 972 2200 >3~
7.5 2.6 1282 3200 >3~
7.~ 2.7 -Mean 7.3 2.8 1127 SD 0.3 0.2 219 WO 96!25150 PCT/US96/01845 Run #10 Low/LowMaximum Time Size 90%cValueCoulterSD % Dust to (tun) Aqueous Maximum (tun) N4MD
Size DissolutionRue (nm) Rate (min) (x10'5 AU/min) 8.0 2.3 653 1150 >3Etm 7.8 2.5 530 1300 >3~

7.0 2.6 Mean 7.6 2.5 592 >3Etm SD 0.5 0.2 87 Run #14 Low/LowMaximum Titne Size 90%<ValueCoulterSD % Dust to (tun) Aqueous Maximum (tun) N4MD
Size DissolutionRate (~) Rate (min) (x10-5 AU/min) 6.9 3.0 307 680 331 120 7 6.5 2.8 326 720 286 35 5 7.4 2.7 Mean 6.9 2.8 317 309 SD 0.5 0 ~ 13 32 Run #2 Low/L,owMaximum Time Size 90%<ValueCoulterSD % Dust to (tun) Aqueous Maximum (nm) N4MD
Size DissolutionKate (~) Rate (min) (x10-5 AU/min) 83 2.6 343 730 267 46 7 9.0 2.4 247 520 281 39 0 Mean 8.7 2.5 295 274 SD 0.5 0.1 68 10 WO 96/25150 PCTlUS9610I845 _29_ Table 5 Avicel PH200 Formulations Run #7 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10-5 AU/min) 5.0 3.1 250 490 278 41 0 3.0 3.0 255 420 251 43 6 ean 4.0 3.1 253 265 SD 1.4 0.1 4 1y Run #6 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (nm) Aqueous Maximum (nm) N4MD
Size DissolutionRate Rate (min) (x10'5 AU/min) 3.2 3.2 254 525 254 44 5 2.9 3.3 256 500 258 45 4 ean 3.1 3.3 255 256 SD 0.2 0.1 1 3 Run #13 Low/LowMaximum Time Stze 90%<ValueCouiterSI>
to (nm) % [~u~t Aqueous Maximum (nm) N4MD
Stze DissoluuonRate Inm1 Rate (min) (x10-~
ALt/min) 6.'_' 3.0 21-l 400 2 I y ? 1 4 5.0 3.6 209 370 ~ 37 4 Mean 5.6 3.3 212 217 SD 0.8 0.4 4 q.

Run #9 Low/LowMaximum Time Size 90%<ValueCoulterSD % Dust to (tun) Aqueoue Maximum (tun) N4MD
Size DissolutionRats (~) Rate (min) (x10-5 AU/min) 6.5 3.0 229 370 212 29 5 5.7 3.3 212 420 207 32 5 Mean 6.1 3.2 221 210 SD 0.6 0.2 12 4 Example 3 --Preparation of Pharmaceutical Compositions and Test for Reconstitution A material having the following composition was formulated:
Drug Coat. % w.w 1. Danazol, USP 15.1 2. Povidone (1115/17), USP 4.52 3. Sodium Lauryl Sulfate, USP 1_13 4. Purified Water, USP ---Overcoat 5 _ Povi.done (1115 . 17 ) , USP 0 . 847 6. Polyethylene Glycol 1000, NF 0_148 7. Purified Water, USP ---Carrier 8. Sugar Spheres (20-25-mesh), NF 78.3-Range (Drug Coat) Danazol/Povidone Ratio 10 to 1 to 1 to 1 Range Overcoat to Povidone 0.5 to 3.6 (w/w) 28516-31(S) The pharmaceutical composition of Example 3 was used in vitro tests in fasted male beagle dogs and compared to a commercial Danocrine formulation as to relative oral bioavailability (mean AVC under the curve (AVC) at 20mg/kg ~ Standard Error of the Mean (SEM).
The oral AVC showed that the nanoparticulate Example 3 composition had a mean AVC. of 9860 ~ 2600 and 11988 ~
3448 in 2 separate runs and the commercial Danocrine composition had an AVC ~SEM of 1076 ~ 391 (20mg/kg).
This indicated that the formulation of Example 3 achieved a lOX increase in absorption over that of the standard Danocrine formulation.
Example 4 - Naproxen And Danazol Composi ions Naproxen 70.4 63.7 Povidone 4.22 3.82 Sucrose 21.1 19.1 Sodium Lauryl 4.22 3.82 Sulfate Mannitol --- 9.55 C D_ Danazol 40.0 ~ 20.0 Povidone 12.0 6.0 Sodium Lauryl 3.0 1.5 Sulfate Dextrose --- 72.5 Maltodextran 45.0 ---The reconstituted mean particle size was found to be less than 300 nm. The procedure used to *Trade-mark determine the ease of.reconstituting particles is as follows:
1. Weigh out appropriate amount of formulation, (eg, insoluble drug equivalent to 50 mg).
2. Add 1 to 10 ml of dispersion media (distilled water, O.lNHcl, simulated gastric or intestinal fluid, etc.) 3. VarteX the mixture for 10 seconds.
4. Shake the prepared sample at 300 RPM, 37°C for 10 minutes using a shako incubator such as a LabLine Shaker.
5. With a pipette draw off an appropriate amount of material from the preparation for particle sizing.
6. Size the resultant dispersion by using an appropriate technique such as a laser-light scattering, Franhaufa or disc centrifugation method.
It has been found that the PVP/PEG overcoat for compositions containing danazol, PVP and sodium lauryl sulfate coated on a bead, in particular, provides physical protection for the drug layer coated on the bead without inhibiting redispersion of the-drug in aqueous media.

Claims (32)

CLAIMS:
1. ~A pharmaceutical composition comprising:
(a) ~a solid carrier, and coated onto the solid carrier a film comprising:
(b) ~a nanoparticulate drug matrix composition comprising:
(1) ~a nanoparticulate crystalline drug which has a solubility of less than about 10mg/ml in water and is dispersible in at least one liquid medium;
(2) ~a steric stabilizer associated with the surface of the nanoparticulate drug; and (3) ~a water-soluble film dispersing agent, wherein the agent is associated with the surface of the nanoparticulate drug;

wherein dissolution of the solid carrier results in substantial redispersion of particles of the nanoparticulate drug.
2. ~The composition according to claim 1, wherein said liquid medium is selected from the group consisting of water, aqueous salt solutions, safflower oil, ethanol, t-butanol, hexane, and glycol.
3. ~The composition according to claim 1 or 2 wherein the drug has a solubility of less than about 1 part in greater than 1,000,000 parts of water.
4. ~The composition according to any one of claims 1 to 3, wherein the drug has an effective average particle size of less than about 400 nm.
5. The composition according to any one of claims 1 to 4, wherein the drug is selected from the group consisting of analgesics, anti-inflammatory agents, anthelmintics, anti-arrhythmic agents, antibiotics, anticoagulants, antidepressants, antidiabetic agents, antiepileptics, antihistamines, antihypertensive agents, antimuscarinic agents, antimycobacterial agents, antineoplastic agents, immunosuppressants, antithyroid agents, antiviral agents, anxiolytic sedatives, astringents, beta-adrenoceptor blocking agents, blood products, blood substitutes, cardiac inotropic agents, contrast media, corticosteroids, cough suppressants, diagnostic agents, diagnostic imaging agents, diuretics, dopaminergics, haemostatics, immunological agents, lipid regulating agents, muscle relaxants, parasympathomimetics, parathyroid calcitonin and biphosphonates, prostaglandins, radio-pharmaceuticals, sex hormones, anti-allergic agents, stimulants, anorectics, sympathomimetics, thyroid agents, vasodilators, and xanthines.
6. The composition of claim 5, wherein the drug is a steroid, danazol, naproxen, or cyclosporin.
7. The composition according to any one of claims 1 to 6, wherein the steric stabilizer is present in an amount of about 0.1% to about 90%, based on the weight of the dry particle.
8. The composition according to any one of claims 1 to 7, wherein the steric stabilizer is present in an amount of about 0.1% to about 60%, based on the weight of the drug.
9. The composition according to any one of claims 1 to 8, wherein the steric stabilizer is present in an amount of about 20% to about 60%, based on the weight of the dry particle.
10. The composition according to any one of claims 1 to 9, wherein the steric stabilizer is selected from the group consisting of gelatin, casein, lecithin, gum acacia, cholesterol, tragacanth, sorbitan esters, polyoxyethylene alkyl esters, polyoxyethylene caster oil derivatives, polyoxyethylene, sorbitan fatty acid esters, polyethylene glycols, polyoxyethylene stearates, carboxymethylcellulose calcium, carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, phthalate, non crystalline cellulose, polyvinyl alcohol, and polyvinylpyrrolidone.
11. The composition of claim 10, wherein the steric stabilizer is polyvinylpyrrolidone.
12. The composition according to any one of claims 1 to 11, wherein the film dispersing agent is present in an amount of about 0.5% to about 50%, based on the weight of the drug, or from about 0.5 to about 60%, based on the weight of the drug.
13. The composition according to any one of claims 1 to 12, wherein the nanoparticulate drug matrix comprises about 1 to about 1200% by weight of the composition.
14. The composition according to any one of claims 1 to 13, wherein the film dispersing agent is selected from the group consisting of mannitol, sodium lauryl sulfate, mono saccharides, and disaccharides.
15. The composition according to any one of claims 1 to 14, wherein the film dispersing agent is sodium lauryl sulfate.
16. The composition according to any one of claims 1 to 15, wherein the solid carrier is selected from the group consisting of sugar spheres, maltodextrin, microcrystalline cellulose, microcrystal cellulose/sodium carboxylmethylcellulose, granular dextrose, granular sugar, dicalcium phosphate, tricalcium phosphate, mono saccharides, and disaccharides.
17. The composition according to any one of claims 1 to 16, wherein the solid carrier is Avicel*, lactose, granular sugar, sugar beads, mannitol, dextrose, dicalcium phosphate, or maltodextrin.
18. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size of less than about 2 millimeters.
19. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size from about 10 mesh to about 0.2 millimeters.
20. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size from about 10 mesh to about 80 mesh.
21. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size from about 20 mesh to about 70 mesh
22. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size from about 16 mesh to about 60 mesh.

*Trade-mark
23. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size of 40-45 mesh.
24. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size of 30-35 mesh.
25. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size of 20-25 mesh.
26. The composition according to any one of claims 1 to 17, wherein the solid carrier has a size of 18-20 mesh.
27. The composition according to any one of claims 1 to 17, wherein the solid carrier has a sire of 16-18 mesh.
28. The composition of claim 25, wherein the solid carrier comprises sugar spheres of from 20 to 25 mesh.
29. The composition according to any one of claims 1 to 28, further comprising a protective coating over the nanoparticulate drug and film dispersing agent coated onto the solid carrier.
30. The composition according to claim 29, wherein said protective coating comprises polyvinylpyrrolidone and polyethylene glycol.
31. The composition of claim 30, wherein the polyethylene glycol has a molecular weight of from about 300 to about 8000.

32. The composition of claim 29 or 30, wherein the weight ratio of PEG/PVP is from 1 to 5.

33. The composition according to any one of claims 1 to 32, formulated into a pharmaceutically acceptable capsule shell.

34. The composition of claim 33, wherein the capsule shell is a gelatin capsule shell.

35. The composition of claim 34, wherein the capsule shell is a soft gelatin capsule shell.

36. The composition of claim 34, wherein the capsule shell is a hard gelatin capsule shell.

37. The composition according to any one of claims 1 to 36, further comprising a pharmaceutically acceptable carrier.

38. The composition according to any one of claims 1 to 37, formulated for oral administration.

39. The composition according to any one of claims 1 to 37, formulated for parenteral injection.

40. The composition according to any one of claims 1 to 37, formulated for rectal administration.

41. The composition according to any one of claims 1 to 37, formulated for intravenous injection.

42. Use of the composition according to any one of claims 1 to 41 for treating mammals.

to about 8000.
32. The compo
CA002212803A 1995-02-13 1996-02-09 Redispersible nanoparticulate film matrices with protective overcoats Expired - Fee Related CA2212803C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/387,651 1995-02-13
US08/387,651 US5573783A (en) 1995-02-13 1995-02-13 Redispersible nanoparticulate film matrices with protective overcoats
PCT/US1996/001845 WO1996025150A1 (en) 1995-02-13 1996-02-09 Redispersible nanoparticulate film matrices with protective overcoats

Publications (2)

Publication Number Publication Date
CA2212803A1 CA2212803A1 (en) 1996-08-22
CA2212803C true CA2212803C (en) 2003-10-07

Family

ID=23530812

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002212803A Expired - Fee Related CA2212803C (en) 1995-02-13 1996-02-09 Redispersible nanoparticulate film matrices with protective overcoats

Country Status (8)

Country Link
US (1) US5573783A (en)
EP (1) EP0812187B1 (en)
JP (3) JPH11500127A (en)
AT (1) ATE238770T1 (en)
AU (1) AU4866996A (en)
CA (1) CA2212803C (en)
DE (1) DE69627835T2 (en)
WO (1) WO1996025150A1 (en)

Families Citing this family (179)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH686761A5 (en) 1993-05-27 1996-06-28 Sandoz Ag Pharmaceutical formulations.
WO1996025918A1 (en) * 1995-02-24 1996-08-29 Nanosystems L.L.C. Aerosols containing nanoparticle dispersions
BE1009856A5 (en) 1995-07-14 1997-10-07 Sandoz Sa Pharmaceutical composition in the form of a solid release including macrolide and a vehicle.
US5766629A (en) 1995-08-25 1998-06-16 Sangstat Medical Corporation Oral cyclosporin formulations
US20050267302A1 (en) * 1995-12-11 2005-12-01 G.D. Searle & Co. Eplerenone crystalline form exhibiting enhanced dissolution rate
US5891548A (en) * 1996-10-03 1999-04-06 Dow Corning Corporation Encapsulated silica nanoparticles
US5993856A (en) * 1997-01-24 1999-11-30 Femmepharma Pharmaceutical preparations and methods for their administration
US20050004049A1 (en) * 1997-03-11 2005-01-06 Elan Pharma International Limited Novel griseofulvin compositions
DE19721947C2 (en) * 1997-05-21 1999-04-22 Diagnostikforschung Inst Use of pharmaceutical preparations containing particles, vesicles or polymers as well as non-steroidal anti-inflammatory drugs and / or platelet aggregation inhibitors for the visualization of the vessels, lymph nodes and bone marrow
NZ328751A (en) * 1997-09-16 1999-01-28 Bernard Charles Sherman Solid medicament containing an anionic surfactant and cyclosporin
JP2007246539A (en) * 1997-10-20 2007-09-27 Dainippon Sumitomo Pharma Co Ltd Rapidly soluble drug composition
UA72189C2 (en) 1997-11-17 2005-02-15 Янссен Фармацевтика Н.В. Aqueous suspensions of 9-hydroxy-risperidone fatty acid esters provided in submicron form
US5997905A (en) * 1998-09-04 1999-12-07 Mcneil-Ppc Preparation of pharmaceutically active particles
US8293277B2 (en) * 1998-10-01 2012-10-23 Alkermes Pharma Ireland Limited Controlled-release nanoparticulate compositions
US8236352B2 (en) 1998-10-01 2012-08-07 Alkermes Pharma Ireland Limited Glipizide compositions
WO2000018374A1 (en) * 1998-10-01 2000-04-06 Elan Pharma International, Ltd. Controlled release nanoparticulate compositions
US20070160675A1 (en) * 1998-11-02 2007-07-12 Elan Corporation, Plc Nanoparticulate and controlled release compositions comprising a cephalosporin
DK1126826T6 (en) * 1998-11-02 2019-06-24 Alkermes Pharma Ireland Ltd Multiparticulate modified release of methylphenidate
JP2002534370A (en) 1998-12-30 2002-10-15 デクセル リミテッド Dispersible concentrate for delivering cyclosporin
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20060034937A1 (en) * 1999-11-23 2006-02-16 Mahesh Patel Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20030180352A1 (en) * 1999-11-23 2003-09-25 Patel Mahesh V. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
SK12672001A3 (en) 1999-12-08 2002-04-04 Pharmacia Corporation Compositions of cyclooxygenase-2 inhibitor having rapid onset of therapeutic effect
EA008449B1 (en) * 1999-12-08 2007-06-29 Фармация Корпорейшн Eplerenone crystalline form
US20030083493A1 (en) * 1999-12-08 2003-05-01 Barton Kathleen P. Eplerenone drug substance having high phase purity
UA74539C2 (en) 1999-12-08 2006-01-16 Pharmacia Corp Crystalline polymorphous forms of celecoxib (variants), a method for the preparation thereof (variants), a pharmaceutical composition (variants)
US7179849B2 (en) * 1999-12-15 2007-02-20 C. R. Bard, Inc. Antimicrobial compositions containing colloids of oligodynamic metals
WO2001045674A1 (en) * 1999-12-20 2001-06-28 Cocensys, Inc. Process for producing nanometer particles by fluid bed spray-drying
US8771740B2 (en) * 1999-12-20 2014-07-08 Nicholas J. Kerkhof Process for producing nanoparticles by spray drying
JP2003518485A (en) 1999-12-23 2003-06-10 ファイザー・プロダクツ・インク Pharmaceutical composition giving improved drug concentration
US7732404B2 (en) 1999-12-30 2010-06-08 Dexcel Ltd Pro-nanodispersion for the delivery of cyclosporin
US6841211B1 (en) 2000-05-12 2005-01-11 Pechiney Emballage Flexible Europe Containers having improved barrier and mechanical properties
US6403231B1 (en) 2000-05-12 2002-06-11 Pechiney Emballage Flexible Europe Thermoplastic film structures having improved barrier and mechanical properties
US6447860B1 (en) 2000-05-12 2002-09-10 Pechiney Emballage Flexible Europe Squeezable containers for flowable products having improved barrier and mechanical properties
MY120279A (en) * 2000-05-26 2005-09-30 Pharmacia Corp Use of a celecoxib composition for fast pain relief
JP2004503601A (en) * 2000-07-13 2004-02-05 ファルマシア・コーポレーション Use of COX-2 inhibitors in the treatment and prevention of ocular COX-2-mediated diseases
US7276249B2 (en) * 2002-05-24 2007-10-02 Elan Pharma International, Ltd. Nanoparticulate fibrate formulations
US20030224058A1 (en) * 2002-05-24 2003-12-04 Elan Pharma International, Ltd. Nanoparticulate fibrate formulations
US7998507B2 (en) * 2000-09-21 2011-08-16 Elan Pharma International Ltd. Nanoparticulate compositions of mitogen-activated protein (MAP) kinase inhibitors
US7198795B2 (en) 2000-09-21 2007-04-03 Elan Pharma International Ltd. In vitro methods for evaluating the in vivo effectiveness of dosage forms of microparticulate of nanoparticulate active agent compositions
WO2002045691A2 (en) * 2000-12-06 2002-06-13 Pharmacia Corporation Laboratory scale milling process
ATE290324T1 (en) * 2000-12-15 2005-03-15 Quest Int A MOISTURE AND OXYGEN STABLE COMPOSITION AND METHOD FOR PRODUCING
US7193084B2 (en) 2000-12-22 2007-03-20 Baxter International Inc. Polymorphic form of itraconazole
US8067032B2 (en) * 2000-12-22 2011-11-29 Baxter International Inc. Method for preparing submicron particles of antineoplastic agents
US20040022862A1 (en) * 2000-12-22 2004-02-05 Kipp James E. Method for preparing small particles
US6977085B2 (en) * 2000-12-22 2005-12-20 Baxter International Inc. Method for preparing submicron suspensions with polymorph control
US9700866B2 (en) 2000-12-22 2017-07-11 Baxter International Inc. Surfactant systems for delivery of organic compounds
US6951656B2 (en) * 2000-12-22 2005-10-04 Baxter International Inc. Microprecipitation method for preparing submicron suspensions
US7037528B2 (en) * 2000-12-22 2006-05-02 Baxter International Inc. Microprecipitation method for preparing submicron suspensions
US20050048126A1 (en) 2000-12-22 2005-03-03 Barrett Rabinow Formulation to render an antimicrobial drug potent against organisms normally considered to be resistant to the drug
US20030072807A1 (en) * 2000-12-22 2003-04-17 Wong Joseph Chung-Tak Solid particulate antifungal compositions for pharmaceutical use
US6623761B2 (en) 2000-12-22 2003-09-23 Hassan Emadeldin M. Method of making nanoparticles of substantially water insoluble materials
US6884436B2 (en) * 2000-12-22 2005-04-26 Baxter International Inc. Method for preparing submicron particle suspensions
US6976647B2 (en) * 2001-06-05 2005-12-20 Elan Pharma International, Limited System and method for milling materials
JP2005504266A (en) * 2001-06-22 2005-02-10 エラン ファーマ インターナショナル,リミティド High-throughput screening methods using small-scale mills or microfluidics
US7758890B2 (en) 2001-06-23 2010-07-20 Lyotropic Therapeutics, Inc. Treatment using dantrolene
DE60217367T2 (en) * 2001-09-19 2007-10-18 Elan Pharma International Ltd. NANOPARTICLE COMPOSITIONS CONTAINING INSULIN
JP2005529060A (en) * 2001-09-25 2005-09-29 ファルマシア コーポレイション Solid form of N- (2-hydroxyacetyl) -5- (4-piperidyl) -4- (4-pyrimidinyl) -3- (4-chlorophenyl) pyrazole
US20060003012A9 (en) * 2001-09-26 2006-01-05 Sean Brynjelsen Preparation of submicron solid particle suspensions by sonication of multiphase systems
CA2461349C (en) 2001-09-26 2011-11-29 Baxter International Inc. Preparation of submicron sized nanoparticles via dispersion and solvent or liquid phase removal
US8603514B2 (en) 2002-04-11 2013-12-10 Monosol Rx, Llc Uniform films for rapid dissolve dosage form incorporating taste-masking compositions
US7357891B2 (en) 2001-10-12 2008-04-15 Monosol Rx, Llc Process for making an ingestible film
US20070281003A1 (en) 2001-10-12 2007-12-06 Fuisz Richard C Polymer-Based Films and Drug Delivery Systems Made Therefrom
US20110033542A1 (en) 2009-08-07 2011-02-10 Monosol Rx, Llc Sublingual and buccal film compositions
US8663687B2 (en) * 2001-10-12 2014-03-04 Monosol Rx, Llc Film compositions for delivery of actives
JP2005508939A (en) * 2001-10-12 2005-04-07 エラン ファーマ インターナショナル,リミティド Composition having combined immediate release and sustained release characteristics
US11207805B2 (en) 2001-10-12 2021-12-28 Aquestive Therapeutics, Inc. Process for manufacturing a resulting pharmaceutical film
US8765167B2 (en) 2001-10-12 2014-07-01 Monosol Rx, Llc Uniform films for rapid-dissolve dosage form incorporating anti-tacking compositions
US8900498B2 (en) 2001-10-12 2014-12-02 Monosol Rx, Llc Process for manufacturing a resulting multi-layer pharmaceutical film
US8900497B2 (en) 2001-10-12 2014-12-02 Monosol Rx, Llc Process for making a film having a substantially uniform distribution of components
US10285910B2 (en) 2001-10-12 2019-05-14 Aquestive Therapeutics, Inc. Sublingual and buccal film compositions
US20190328679A1 (en) 2001-10-12 2019-10-31 Aquestive Therapeutics, Inc. Uniform films for rapid-dissolve dosage form incorporating anti-tacking compositions
JPWO2003034302A1 (en) * 2001-10-15 2005-02-03 株式会社シュタルク CONTENT DISTRIBUTION SERVER AND CONTENT DISTRIBUTION SYSTEM HAVING THE SAME
US7112340B2 (en) * 2001-10-19 2006-09-26 Baxter International Inc. Compositions of and method for preparing stable particles in a frozen aqueous matrix
WO2003040223A2 (en) * 2001-11-03 2003-05-15 Nanophase Technology Corporation Nanostructured compositions
WO2003043586A2 (en) * 2001-11-20 2003-05-30 Advanced Inhalation Research, Inc. Compositions for sustained action product delivery
CA2468780A1 (en) * 2001-12-03 2003-06-12 C.R. Bard, Inc. Microbe-resistant medical device, microbe-resistant polymeric coating and methods for producing same
UA76810C2 (en) * 2001-12-10 2006-09-15 Мерк Енд Ко., Інк. Pharmaceutical compositions of tachikinine receptor antagonist in form of nanoparticles
MXPA04006017A (en) 2001-12-20 2005-06-08 Femmepharma Inc Vaginal delivery of drugs.
US20030129242A1 (en) * 2002-01-04 2003-07-10 Bosch H. William Sterile filtered nanoparticulate formulations of budesonide and beclomethasone having tyloxapol as a surface stabilizer
CA2475092C (en) 2002-02-04 2012-05-01 Christian F. Wertz Nanoparticulate compositions having lysozyme as a surface stabilizer
US20030215502A1 (en) * 2002-03-20 2003-11-20 Elan Pharma International Limited Fast dissolving dosage forms having reduced friability
US9101540B2 (en) * 2002-04-12 2015-08-11 Alkermes Pharma Ireland Limited Nanoparticulate megestrol formulations
ES2380318T3 (en) 2002-04-12 2012-05-10 Alkermes Pharma Ireland Limited Megestrol nanoparticular formulations
US7101576B2 (en) * 2002-04-12 2006-09-05 Elan Pharma International Limited Nanoparticulate megestrol formulations
US20040105889A1 (en) * 2002-12-03 2004-06-03 Elan Pharma International Limited Low viscosity liquid dosage forms
US20100226989A1 (en) * 2002-04-12 2010-09-09 Elan Pharma International, Limited Nanoparticulate megestrol formulations
AU2003234452A1 (en) * 2002-05-06 2003-11-11 Elan Pharma International Ltd. Nanoparticulate nystatin formulations
AU2003241477A1 (en) 2002-06-10 2003-12-22 Elan Pharma International, Ltd. Nanoparticulate polycosanol formulations and novel polycosanol combinations
DE60329188D1 (en) 2002-08-12 2009-10-22 Bend Res Inc DRUG PREPARATIONS CONSISTING OF MEDICINAL PRODUCTS IN HALF-ORIENTED FORM AND POLYMERS
UY27939A1 (en) 2002-08-21 2004-03-31 Glaxo Group Ltd COMPOUNDS
ATE487470T1 (en) * 2002-09-11 2010-11-15 Elan Pharma Int Ltd GEL-STABILIZED ACTIVE COMPOSITIONS IN NANOPARTICLE SIZE
WO2004032980A1 (en) * 2002-10-04 2004-04-22 Elan Pharma International Limited Gamma irradiation of solid nanoparticulate active agents
US20040173696A1 (en) * 2002-12-17 2004-09-09 Elan Pharma International Ltd. Milling microgram quantities of nanoparticulate candidate compounds
US9173836B2 (en) 2003-01-02 2015-11-03 FemmeParma Holding Company, Inc. Pharmaceutical preparations for treatments of diseases and disorders of the breast
JP2006515026A (en) 2003-01-02 2006-05-18 フェムファーマ ホールディング カンパニー, インコーポレイテッド Pharmaceutical preparations for the treatment of breast diseases and disorders
EP1593091B1 (en) * 2003-01-25 2013-07-17 Seno Medical Instruments, Inc. High contrast optoacoustical imaging using non-spherical nanoparticles
CA2513064C (en) * 2003-01-31 2009-11-10 Elan Pharma International, Ltd. Nanoparticulate topiramate formulations
US20040208833A1 (en) * 2003-02-04 2004-10-21 Elan Pharma International Ltd. Novel fluticasone formulations
SI21402A (en) * 2003-02-12 2004-08-31 LEK farmacevtska dru�ba d.d. Lined particles and pharmaceutical forms
US20100297252A1 (en) 2003-03-03 2010-11-25 Elan Pharma International Ltd. Nanoparticulate meloxicam formulations
US8512727B2 (en) * 2003-03-03 2013-08-20 Alkermes Pharma Ireland Limited Nanoparticulate meloxicam formulations
JP2007520999A (en) * 2003-04-08 2007-08-02 クエスト・インターナショナル・ビー・ブイ Oxygen stabilizing activator-containing composition
JP2007501683A (en) * 2003-05-22 2007-02-01 エラン ファーマ インターナショナル リミテッド Sterilization of nanoparticle active substance dispersions by gamma irradiation
ES2318330T3 (en) * 2003-08-08 2009-05-01 Elan Pharma International Limited NEW COMPOSITIONS OF METAXALONA.
ES2366646T3 (en) * 2003-11-05 2011-10-24 Elan Pharma International Limited COMPOSITIONS IN THE FORM OF NANOPARTICLES THAT HAVE A PEPTIDE AS A SURFACE STABILIZER.
US20050208145A1 (en) * 2004-03-19 2005-09-22 Thava Vasanthan Grain fiber compositions and methods of use
WO2006083326A2 (en) * 2004-08-07 2006-08-10 Cabot Corporation Gas dispersion manufacture of nanoparticulates and nanoparticulate-containing products and processing thereof
US20060083694A1 (en) 2004-08-07 2006-04-20 Cabot Corporation Multi-component particles comprising inorganic nanoparticles distributed in an organic matrix and processes for making and using same
US20080009568A1 (en) * 2004-10-18 2008-01-10 Nitin Kumar Methods to disperse and exfoliate nanoparticles
EA200701065A1 (en) * 2004-11-16 2007-12-28 Элан Фарма Интернэшнл Лтд. INJECTABLE COMPOSITIONS CONTAINING NANODISPERS Olanzapine
US20090155331A1 (en) * 2005-11-16 2009-06-18 Elan Pharma International Limited Injectable nanoparticulate olanzapine formulations
UA89513C2 (en) * 2004-12-03 2010-02-10 Элан Фарма Интернешнл Лтд. Nanoparticulate raloxifene hydrochloride composition
US20060159767A1 (en) * 2004-12-22 2006-07-20 Elan Pharma International Limited Nanoparticulate bicalutamide formulations
EP1835890A2 (en) * 2005-01-06 2007-09-26 Elan Pharma International Limited Nanoparticulate candesartan formulations
AU2006214443C1 (en) 2005-02-15 2011-11-24 Alkermes Pharma Ireland Limited Aerosol and injectable formulations of nanoparticulate benzodiazepine
WO2006099121A2 (en) * 2005-03-10 2006-09-21 Elan Pharma International Limited Formulations of a nanoparticulate finasteride, dutasteride and tamsulosin hydrochloride, and mixtures thereof
CA2601179A1 (en) * 2005-03-16 2006-09-21 Elan Pharma International Limited Nanoparticulate leukotriene receptor antagonist/corticosteroid formulations
JP2008533173A (en) * 2005-03-17 2008-08-21 エラン ファーマ インターナショナル リミテッド Nanoparticulate bisphosphonate composition
KR20070121786A (en) * 2005-03-23 2007-12-27 엘란 파마 인터내셔널 리미티드 Nanoparticulate corticosteroid and antihistamine formulations
BRPI0607537A2 (en) * 2005-04-12 2009-09-15 Elan Pharma Int Ltd nanoparticulate quinazoline derivative formulations
US20060246141A1 (en) * 2005-04-12 2006-11-02 Elan Pharma International, Limited Nanoparticulate lipase inhibitor formulations
CN101171000A (en) * 2005-04-12 2008-04-30 依兰药物国际有限公司 Nanoparticulate and controlled release compositions comprising cyclosporine
JP2009517485A (en) 2005-06-08 2009-04-30 エラン・ファルマ・インターナショナル・リミテッド Nanoparticulate and controlled release compositions containing cefditoren
CA2612384A1 (en) * 2005-06-15 2006-12-28 Elan Pharma International, Limited Nanoparticulate azelnidipine formulations
GB0516549D0 (en) * 2005-08-12 2005-09-21 Sulaiman Brian Milling system
EA017290B1 (en) * 2005-11-28 2012-11-30 Домейн Раша Инвестментс Лимитед Ganaxolone-based formulations
US8367112B2 (en) * 2006-02-28 2013-02-05 Alkermes Pharma Ireland Limited Nanoparticulate carverdilol formulations
US20080096819A1 (en) * 2006-05-02 2008-04-24 Allozyne, Inc. Amino acid substituted molecules
EP2395099A3 (en) * 2006-05-02 2012-05-16 Allozyne, Inc. Amino acid substituted molecules
AU2007265452A1 (en) * 2006-06-26 2008-01-03 Mutual Pharmaceutical Company, Inc. Active agent formulations, methods of making, and methods of use
US20090074872A1 (en) * 2006-06-26 2009-03-19 Mutual Pharmaceutical Company, Inc. Active Agent Formulations, Methods of Making, and Methods of Use
CL2007002689A1 (en) 2006-09-18 2008-04-18 Vitae Pharmaceuticals Inc COMPOUNDS DERIVED FROM PIPERIDIN-1-CARBOXAMIDA, INHIBITORS OF THE RENINE; INTERMEDIARY COMPOUNDS; PHARMACEUTICAL COMPOSITION; AND USE IN THE TREATMENT OF DISEASES SUCH AS HYPERTENSION, CARDIAC INSUFFICIENCY, CARDIAC FIBROSIS, AMONG OTHERS.
JP2010510988A (en) * 2006-11-28 2010-04-08 マリナス ファーマシューティカルズ Nanoparticle formulation, method for producing the same and use thereof
WO2008135828A2 (en) 2007-05-03 2008-11-13 Pfizer Products Inc. Nanoparticles comprising a drug, ethylcellulose, and a bile salt
WO2008135855A2 (en) 2007-05-03 2008-11-13 Pfizer Products Inc. Nanoparticles comprising a cholesteryl ester transfer protein inhibitor and a nonionizable polymer
US8722736B2 (en) 2007-05-22 2014-05-13 Baxter International Inc. Multi-dose concentrate esmolol with benzyl alcohol
US8426467B2 (en) 2007-05-22 2013-04-23 Baxter International Inc. Colored esmolol concentrate
US9545384B2 (en) 2007-06-04 2017-01-17 Bend Research, Inc. Nanoparticles comprising drug, a non-ionizable cellulosic polymer and tocopheryl polyethylene glocol succinate
US8974827B2 (en) 2007-06-04 2015-03-10 Bend Research, Inc. Nanoparticles comprising a non-ionizable cellulosic polymer and an amphiphilic non-ionizable block copolymer
CA2707840A1 (en) * 2007-08-20 2009-02-26 Allozyne, Inc. Amino acid substituted molecules
US8642062B2 (en) 2007-10-31 2014-02-04 Abbott Cardiovascular Systems Inc. Implantable device having a slow dissolving polymer
US9233078B2 (en) 2007-12-06 2016-01-12 Bend Research, Inc. Nanoparticles comprising a non-ionizable polymer and an Amine-functionalized methacrylate copolymer
WO2009073215A1 (en) 2007-12-06 2009-06-11 Bend Research, Inc. Pharmaceutical compositions comprising nanoparticles and a resuspending material
US20090238867A1 (en) * 2007-12-13 2009-09-24 Scott Jenkins Nanoparticulate Anidulafungin Compositions and Methods for Making the Same
US20090311335A1 (en) * 2008-06-12 2009-12-17 Scott Jenkins Combination of a triptan and an nsaid
CN102105140B (en) * 2008-07-02 2013-10-23 巴斯夫欧洲公司 Method for coating tablets
US20100159010A1 (en) * 2008-12-24 2010-06-24 Mutual Pharmaceutical Company, Inc. Active Agent Formulations, Methods of Making, and Methods of Use
US11304960B2 (en) 2009-01-08 2022-04-19 Chandrashekar Giliyar Steroidal compositions
US7828996B1 (en) * 2009-03-27 2010-11-09 Abbott Cardiovascular Systems Inc. Method for the manufacture of stable, nano-sized particles
FR2945950A1 (en) 2009-05-27 2010-12-03 Elan Pharma Int Ltd ANTICANCER NANOPARTICLE COMPOSITIONS AND METHODS FOR PREPARING THE SAME
AU2010254180B2 (en) 2009-05-27 2015-08-27 Alkermes Pharma Ireland Limited Reduction of flake-like aggregation in nanoparticulate active agent compositions
JP5947717B2 (en) 2009-06-19 2016-07-06 ナノフォーム ハンガリー リミテッド Nanostructured sildenafil base, pharmaceutically acceptable salts and co-crystals thereof, compositions thereof, methods of preparation thereof, and pharmaceutical compositions containing them
AR077692A1 (en) 2009-08-06 2011-09-14 Vitae Pharmaceuticals Inc SALTS OF 2 - ((R) - (3-CHLOROPHENYL) ((R) -1 - ((S) -2- (METHYLAMINE) -3 - ((R) -TETRAHYDRO-2H-PIRAN-3-IL) PROPILCARBAMOIL ) PIPERIDIN -3-IL) METOXI) METHYL ETILCARBAMATE
WO2011146583A2 (en) 2010-05-19 2011-11-24 Elan Pharma International Limited Nanoparticulate cinacalcet formulations
US9149959B2 (en) 2010-10-22 2015-10-06 Monosol Rx, Llc Manufacturing of small film strips
US20180153904A1 (en) 2010-11-30 2018-06-07 Lipocine Inc. High-strength testosterone undecanoate compositions
US9358241B2 (en) 2010-11-30 2016-06-07 Lipocine Inc. High-strength testosterone undecanoate compositions
US9034858B2 (en) 2010-11-30 2015-05-19 Lipocine Inc. High-strength testosterone undecanoate compositions
US20120148675A1 (en) 2010-12-10 2012-06-14 Basawaraj Chickmath Testosterone undecanoate compositions
JP6122003B2 (en) 2011-07-22 2017-04-26 ケモセントリックス, インコーポレイテッド Crystalline form of sodium salt of 4-tert-butyl-N- [4-chloro-2- (1-oxy-pyridine-4-carbonyl) -phenyl] -benzenesulfonamide
RU2607191C2 (en) 2011-07-22 2017-01-10 Хемоцентрикс, Инк. Polymorphic forms of sodium salt of 4-tert-butyl-n-[4-chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-benzenesulphonamide
RS57644B1 (en) 2011-09-01 2018-11-30 Glaxo Group Ltd Novel crystal form
IN2014DN09416A (en) 2012-04-25 2015-07-17 Spi Pharma Inc
EA032017B1 (en) 2014-06-25 2019-03-29 Глэксосмитклайн Интеллекчуал Проперти Дивелопмент Лимитед Crystalline salts of (s)-6-((1-acetylpiperidin-4-yl)amino)-n-(3-(3,4-dihydroisoquinolin-2(1h)-yl)-2-hydroxypropyl)pyrimidine-4-carboxamide
WO2016033549A2 (en) 2014-08-28 2016-03-03 Lipocine Inc. (17-ß)-3-OXOANDROST-4-EN-17-YL TRIDECANOATE COMPOSITIONS AND METHODS OF THEIR PREPARATION AND USE
US9498485B2 (en) 2014-08-28 2016-11-22 Lipocine Inc. Bioavailable solid state (17-β)-hydroxy-4-androsten-3-one esters
WO2016038519A1 (en) 2014-09-08 2016-03-17 Glaxosmithkline Intellectual Property Development Limited Crystalline forms of 2-(4-(4-ethoxy-6-oxo-1,6-dihydropyridin-3-yl)-2-fluorophenyl)-n-(5-(1,1,1-trifluoro-2-methylpropan-2-yl)isoxazol-3-yl)acetamide
US10011629B2 (en) 2015-05-01 2018-07-03 Cocrystal Pharma, Inc. Nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
KR20180082457A (en) 2015-10-16 2018-07-18 마리누스 파마슈티컬스 인코포레이티드 Injectable neuro-steroid agents containing nanoparticles
US11273131B2 (en) 2016-05-05 2022-03-15 Aquestive Therapeutics, Inc. Pharmaceutical compositions with enhanced permeation
EP3452023A1 (en) 2016-05-05 2019-03-13 Aquestive Therapeutics, Inc. Enhanced delivery epinephrine compositions
JP2019524816A (en) 2016-08-11 2019-09-05 オービッド・セラピューティクス・インコーポレイテッドOvid Therapeutics, Inc. Methods and compositions for the treatment of epileptic disorders
US10391105B2 (en) 2016-09-09 2019-08-27 Marinus Pharmaceuticals Inc. Methods of treating certain depressive disorders and delirium tremens
JP2020503269A (en) 2016-11-28 2020-01-30 リポカイン インコーポレーテッド Oral testosterone undecanoate therapy
WO2020118142A1 (en) 2018-12-07 2020-06-11 Marinus Pharmaceuticals, Inc. Ganaxolone for use in prophylaxis and treatment of pospartum depression
MX2021007335A (en) * 2018-12-19 2021-07-15 Galecto Biotech Ab Amorphous form of 5-bromopyridin-3-yl 3-deoxy-3-[4-(3,4,5-trifluo rophenyl)-1h-1,2,3-triazol-1-yl]-1-thio-alpha-d-galactopyranosid e.
WO2021026124A1 (en) 2019-08-05 2021-02-11 Marinus Pharmaceuticals, Inc. Ganaxolone for use in treatment of status epilepticus
KR20220134529A (en) 2019-12-06 2022-10-05 마리누스 파마슈티컬스 인코포레이티드 Ganaxolone for use in the treatment of complex tuberous sclerosis

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8414221D0 (en) * 1984-06-04 1984-07-11 Sterwin Ag Unit dosage form
FR2608988B1 (en) * 1986-12-31 1991-01-11 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF COLLOIDAL DISPERSIBLE SYSTEMS OF A SUBSTANCE, IN THE FORM OF NANOPARTICLES
US4946686A (en) * 1987-09-24 1990-08-07 Merck & Co., Inc. Solubility modulated drug delivery system
US5188836A (en) * 1990-07-27 1993-02-23 Warner-Lambert Company Sustained release formulations
ATE147976T1 (en) * 1991-01-15 1997-02-15 Hemosphere Inc PROTEIN NANOMATRICES AND PROCESS FOR PRODUCTION
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5399363A (en) * 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5223268A (en) * 1991-05-16 1993-06-29 Sterling Drug, Inc. Low solubility drug-coated bead compositions

Also Published As

Publication number Publication date
ATE238770T1 (en) 2003-05-15
JPH11500127A (en) 1999-01-06
JP2010241820A (en) 2010-10-28
AU4866996A (en) 1996-09-04
WO1996025150A1 (en) 1996-08-22
JP2007112816A (en) 2007-05-10
EP0812187A1 (en) 1997-12-17
DE69627835D1 (en) 2003-06-05
CA2212803A1 (en) 1996-08-22
EP0812187B1 (en) 2003-05-02
DE69627835T2 (en) 2004-02-19
US5573783A (en) 1996-11-12

Similar Documents

Publication Publication Date Title
CA2212803C (en) Redispersible nanoparticulate film matrices with protective overcoats
CA2059432C (en) Surface modified drug nanoparticles
US5622938A (en) Sugar base surfactant for nanocrystals
RU2240827C2 (en) Pharmaceutical composition, methods for its preparing, stable water-insoluble complexes, method for conversion of therapeutically active compound and therapeutically active compound
US8206746B2 (en) Microparticles of water-insoluble substances
EP1318788B1 (en) Solid dose nanoparticulate compositions
IE83410B1 (en) Surface modified drug nanoparticles
JP2001509518A (en) Nanocrystalline preparation of human immunodeficiency virus (HIV) protease inhibitor using cellulosic surface stabilizer and method for producing such preparation
KR20000035808A (en) Compositions comprising microparticles of water-insoluble substances and method for preparing same
WO2014074808A1 (en) Pharmaceutical core-shell composite powder and processes for making the same
WO1997004756A2 (en) Methacrylate backbone surfactants in nanoparticulate formulations
CA2470533A1 (en) Drug nanoparticles from template emulsions
US20040028747A1 (en) Crystalline drug particles prepared using a controlled precipitation process
AU743917B2 (en) Compositions comprising microparticles of water-insoluble substances

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20130211