WO2003024424A1 - Stabilization of active agents by formulation into nanoparticulate form - Google Patents

Stabilization of active agents by formulation into nanoparticulate form Download PDF

Info

Publication number
WO2003024424A1
WO2003024424A1 PCT/US2002/025979 US0225979W WO03024424A1 WO 2003024424 A1 WO2003024424 A1 WO 2003024424A1 US 0225979 W US0225979 W US 0225979W WO 03024424 A1 WO03024424 A1 WO 03024424A1
Authority
WO
WIPO (PCT)
Prior art keywords
active agent
ofthe
nanoparticulate
exposure
particle size
Prior art date
Application number
PCT/US2002/025979
Other languages
French (fr)
Inventor
Elaine Merisko-Liversidge
Linden Wei
Original Assignee
Elan Pharma International Ltd.
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25492513&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2003024424(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Elan Pharma International Ltd. filed Critical Elan Pharma International Ltd.
Priority to JP2003528521A priority Critical patent/JP2005505568A/en
Priority to DE60225571T priority patent/DE60225571T2/en
Priority to CA2460436A priority patent/CA2460436C/en
Priority to EP02775709A priority patent/EP1427395B1/en
Priority to DK02775709T priority patent/DK1427395T3/en
Publication of WO2003024424A1 publication Critical patent/WO2003024424A1/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
    • A61K9/146Intimate 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2991Coated

Definitions

  • the present invention is directed to methods for stabilizing active agents, particularly pharmaceutical agents, comprising formulating a at least one active agent, which is unstable under one or more environmental conditions, into a nanoparticulate composition.
  • the nanoparticulate active agent is stable under the same environmental conditions under which the active agent is unstable prior to formulating the agent into a nanoparticulate composition.
  • the nanoparticulate composition comprises an active agent and one or more surface stabilizers adhered to the surface ofthe active agent.
  • Active agents are subject to degradation. Such decomposition or degradation may be due to hydrolysis, oxidation, isomerization, epimerization, or photolysis.
  • the rate of degradation or decomposition varies considerably depending on the structural, physical, and chemical nature ofthe active agent. The rate of decomposition is also often significantly affected by numerous environmental factors, including temperature, light, radiation, enzyme or other catalysts, pH and ionic strength ofthe solution, solvent type, and buffer species.
  • Chemical instability due to degradation or decomposition is highly undesirable for several reasons.
  • a chemical compound is a pharmaceutical agent
  • degradation decreases its efficiency and shortens its effective shelf life.
  • the decrease in the content ofthe active ingredient in a pharmaceutical preparation renders the calculation of an effective dosage unpredictable and difficult.
  • degraded chemical agent may have highly undesirable or even severely toxic side effects.
  • Other methods include converting the drug into a more stable prodrug which, under physiological conditions, is processed to become a biologically active form ofthe compound.
  • Dosage form designs that improve the chemical stability of a drug include loading drugs into liposomes or polymers, e.g., during emulsion polymerization.
  • a lipid soluble drug is often required to prepare a suitable liposome.
  • unacceptably large amounts ofthe liposome or polymer may be required to prepare unit drug doses.
  • techniques for preparing such pharmaceutical compositions tend to be complex.
  • removal of contaminants at the end ofthe emulsion polymerization manufacturing process, such as potentially toxic unreacted monomer or initiator can be difficult and expensive.
  • a dosage fonn that can be used to increase the stability of an administered agent is a monolithic device, which is a rate-controlling polymer matrix throughout which a drug is dissolved or dispersed.
  • a dosage form is a reservoir device, which is a shell-like dosage form having a drug contained within a rate-controlling membrane.
  • U.S. Patent No. 4,725,442 refers to water insoluble drug materials solubilized in an organic liquid and incorporated in microcapsules of phospholipids.
  • One disadvantage of this dosage form is the toxic effects of the solubilizing organic liquids.
  • Other methods of forming reservoir dosage forms of pharmaceutical drug microcapsules include micronizing a slightly-soluble drug by high-speed stirring or impact comminution of a mixture ofthe drug and a sugar or sugar alcohol together with suitable excipients or diluents. See e.g. EP 411,629A.
  • One disadvantage of this method is that the resultant drug particles are larger than those obtained with milling.
  • a reservoir dosage form can also be formed by co-dispersion of a drug or a pharmaceutical agent in water with droplets of a carbohydrate polymer (see e.g. U.S. Patent No. 4,713,249 and WO 84/00294).
  • the major disadvantage of this procedure is that in many cases, a solubilizing organic co-solvent is required for the encapsulation procedure. Removal of traces of such harmful co-solvents can result in an expensive manufacturing process.
  • Nanoparticulate compositions are particles consisting of a poorly soluble active agent having adsorbed onto the surface thereof a non-crosslinked surface stabilizer.
  • the '684 patent also describes methods of making such nanoparticulate compositions.
  • the '684 patent teaches that nanoparticulate compositions are desirable because with a decrease in particle size, and a consequent increase in surface area, a composition can exhibit superior bioavailability.
  • Nanoparticulate compositions are also described, for example, in U.S. Patent Nos. 5,298,262 for "Use of Ionic Cloud Point Modifiers to Prevent Particle Aggregation During Sterilization;" 5,302,401 for “Method to Reduce Particle Size Growth During Lyophilization;” 5,318,767 for “X-Ray Contrast Compositions Useful in Medical Imaging;” 5,326,552 for “Novel Formulation For Nanoparticulate X-Ray Blood Pool Contrast Agents Using High Molecular Weight Non-ionic Surfactants;” 5,328,404 for “Method of X-Ray Imaging Using Iodinated Aromatic Propanedioates;” 5,336,507 for “Use of Charged Phospholipids to Reduce Nanoparticle Aggregation;” 5,340,564 for “Formulations Comprising Olin 10-G to Prevent Particle Aggregation and Increase Stability;” 5,346,702 for "Use of Non-Ionic Cloud Point Mod
  • Amorphous small particle compositions are described, for example, in U.S. Patent Nos. 4,783,484 for "Particulate Composition and Use Thereof as Antimicrobial Agent;” 4,826,689 for “Method for Making Uniformly Sized Particles from Water-Insoluble Organic Compounds;” 4,997,454 for “Method for Making Uniformly-Sized Particles From Insoluble Compounds;" 5,741,522 for "Ultrasmall, Non-aggregated Porous Particles of Uniform Size for Entrapping Gas Bubbles Within and Methods;" and 5,776,496, for "Ultrasmall Porous Particles for Enhancing Ultrasound Back Scatter.”
  • U.S. Patent Nos. 5,399,363 and 5,494,683, both for "Surface Modified Anticancer Nanoparticles,” refer to nanoparticulate compositions of anticancer agents, including taxol, also known as paclitaxel.
  • these patents do not address a means to overcome the chemical instability of an active agent, such as paclitaxel at a basic pH.
  • the two patents teach methods of making nanoparticulate paclitaxel compositions which are stable at a pH of 7.4.
  • this does not teach or suggest that a nanoparticulate paclitaxel composition is stable at an elevated pH, such as a basic pH of 9.
  • GIT gastrointestinal tract
  • the present invention is directed to the discovery that active agents which are unstable under one or more enviromnental conditions, when formulated into nanoparticulate compositions, are stable under those same environmental conditions.
  • Such environmental conditions can be, for example, exposure to a condition such as prolonged storage periods, elevated temperature, non-physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water, solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles.
  • One aspect ofthe invention is directed to a process for stabilizing active agents, particularly pharmaceutical agents, comprising formulating an active agent which is unstable under one or more environmental conditions into a nanoparticulate composition, wherein the active agent is then stable under those same environmental conditions.
  • the nanoparticulate composition comprises a poorly soluble active agent, such as a drug particle, and one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe active agent.
  • the nanoparticulate compositions have an effective average particle size of less than about two microns.
  • the present invention is further directed to a process for stabilizing rapamycin, which is unstable when exposed to water, comprising forming a nanoparticulate formulation of rapamycin having one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe drug.
  • the resultant nanoparticulate rapamycin composition exhibits dramatically superior stability when exposed to water.
  • the pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier, as well as any desired excipients.
  • Yet another aspect ofthe invention encompasses a process for stabilizing paclitaxel, which is unstable when exposed to a basic pH, comprising forming a nanoparticulate composition of paclitaxel having one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe drug.
  • the resultant nanoparticulate paclitaxel composition exhibits dramatically superior stability when exposed to a basic pH.
  • the pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier, as well as any desired excipients.
  • Figure 1 Shows the effect of 0.005 N NaOH (a basic pH level) on the rate of degradation of paclitaxel as compared to the rate of degradation of a nanoparticulate formulation of paclitaxel.
  • the present invention is directed to a method for stabilizing active agents, particularly pharmaceutical agents, which are unstable under one or more environmental conditions, comprising formulating an active agent into a nanoparticulate composition.
  • the nanoparticulate active agent is stable under the same one or more environmental conditions under which the active agent is unstable prior to formulation into a nanoparticulate composition.
  • Such one or more environmental conditions which can cause the active agent to degrade include, but are not limited to, prolonged storage periods, elevated temperature, non- physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water, solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles.
  • the method ofthe invention comprises first identifying and selecting an active agent which is unstable under one or more environmental conditions, followed by formulating particles ofthe active agent into a stable nanoparticulate composition.
  • the formulation step comprises combining the active agent particles and at least one non-crosslinked surface stabilizer, wherein after combining the at least one non-crosslinked surface stabilizer adsorbs to the surface ofthe active agent particles to produce a nanoparticulate composition in which the active agent particles have an effective average particle size of less than about 2 microns.
  • the claimed method is distinct from prior art methods of making nanoparticulate compositions, as prior methods did not teach that active agents which are unstable under one or more environmental conditions can be made stable under those same conditions by formulating the active agent into a nanoparticulate composition. Rather, prior methods of making nanoparticulate compositions teach that the nanoparticulate compositions are desirable as they can increase the bioavailability ofthe component active agent. See e.g., U.S. Patent No. 5,145,684.
  • active agents which are stable only under certain pH conditions such as paclitaxel
  • active agents which are unstable when exposed to elevated temperatures can be heat sterilized without degradation when formulated into a nanoparticulate composition. This can be useful in administering active agents to very young or immunocompromised patients.
  • active agents which are unstable when exposed to water i.e., hydrolysis
  • rapamycin can be formulated for administration in a liquid form. This can be beneficial as tablet dosage forms have a longer onset of activity as compared to liquid or IV dosage forms.
  • many infant and elderly patients are unable to swallow tablet dosage forms.
  • Chemical instability due to degradation is usually a result of hydrolysis, oxidation, isomerization, epimerization, or photolysis.
  • the rate of degradation is often determined by numerous enviromnental factors, including temperature, light, radiation, enzyme or other catalysts, pH and ionic strength ofthe solution, solvent type, or buffer species.
  • the molecules ofthe surface stabilizer shield the active agent, thereby protecting potentially labile chemical groups ofthe active agent from the potentially hostile environment.
  • Another possibility is that for a crystalline active agent particle, the crystalline structure in a nanoparticulate sized formulation results in greater active agent stability.
  • rapamycin is rapidly degraded when exposed to an aqueous environment.
  • the main degradation scheme of rapamycin is the cleavage ofthe macrocyclic lactone ring by the hydrolysis of an ester bond to form a secoacid (SECO).
  • SECO secoacid
  • the secoacid undergoes further dehydration and isomerization to form diketomorpholine analogs.
  • rapamycin when rapamycin is formulated into a nanoparticulate composition, minimal or no rapamycin degradation is observed, even following prolonged exposure to an aqueous medium.
  • paclitaxel Another example of an active agent that is unstable under certain environmental conditions, but which is stable in a nanoparticulate formulation under those same environmental conditions, is paclitaxel.
  • a basic pH i.e., a pH of about 9
  • paclitaxel rapidly degrades. Ringel et al, J Pharmac. Exp. Ther., 242:692-698, (1987).
  • paclitaxel is formulated into a nanoparticulate composition, minimal or no paclitaxel degradation is observed, even when the composition is exposed to a basic pH.
  • the method of stabilizing an active agent according to the present invention comprises formulating the active agent into a nanoparticulate formulation.
  • the nanoparticulate active agent formulation comprises at least one active agent and one or more surface stabilizers adsorbed to the surface ofthe active agent(s).
  • the nanoparticles ofthe invention comprise an active agent, such as a therapeutic or diagnostic agent, having one or more labile groups or exhibiting chemical instability when exposed to certain environmental conditions, such as prolonged storage periods, elevated temperature, non-physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water (which can cause hydrolysis), solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles.
  • active agents having, for example, cosmetic, diagnostic, or bioengineering uses are presumed suitable for the invention.
  • the active agent may be present either substantially in the form of one optically pure enantiomer or as a mixture, racemic or otherwise, of enantiomers.
  • a therapeutic agent can be a pharmaceutical, including biologies such as proteins and peptides, and a diagnostic agent is typically a contrast agent, such as an x-ray contrast agent, or any other type of diagnostic material.
  • the active agent particle exists as a discrete, crystalline phase, amorphous phase, semi-crystalline phase, semi-amorphous phase, or as a combination thereof.
  • the crystalline phase differs from a non-crystalline or amorphous phase which results from precipitation techniques, such as those described in EP Patent No. 275,796.
  • the active agent is preferably present in an essentially pure form, is poorly soluble, and is dispersible in at least one liquid medium.
  • “poorly soluble” it is meant that the active agent has a solubility in a liquid dispersion medium of less than about 30 mg/mL, less than about 10 mg/mL, or less than about 1 mg/mL.
  • the dispersion medium can be, for example, water, safflower oil, ethanol, t-butanol, glycerin, polyethylene glycol (PEG), hexane, or glycol.
  • the active agent can be selected from a variety of known classes of drugs, including, for example, COX-2 inhibitors, retinoids, NSAIDS, proteins, peptides, nucleotides, anti- obesity drugs, nutraceuticals, dietary supplements, carotenoids, corticosteroids, elastase inhibitors, anti-fungals, oncology therapies, anti-emetics, analgesics, cardiovascular agents, 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, anxiolytics, sedatives (hypnotics and neuroleptics), astringents, beta-adrenoceptor blocking agents, blood products and substitutes, cardiac ino
  • nutraceuticals and dietary supplements are disclosed, for example, in Roberts et al., Nutraceuticals: The Complete Encyclopedia of Supplements, Herbs, Vitamins, and Healing Foods (American Nutraceutical Association, 2001), which is specifically incorporated by reference.
  • a nutraceutical or dietary supplement also known as phytochemicals or functional foods, is generally any one of a class of dietary supplements, vitamins, minerals, herbs, or healing foods that have medical or pharmaceutical effects on the body.
  • nutraceuticals or dietary supplements include, but are not limited to, lutein, folic acid, fatty acids (e.g., DHA and ARA), fruit and vegetable extracts, vitamin and mineral supplements, phosphatidylserine, lipoic acid, melatonin, glucosamine/chondroitin, Aloe Vera, Guggul, glutamine, amino acids (e.g., iso-leucine, leucine, lysine, methionine, phenylanine, threonine, tryptophan, and valine), green tea, lycopene, whole foods, food additives, herbs, phytonutrients, antioxidants, flavonoid constituents of fruits, evening primrose oil, flax seeds, fish and marine animal oils, and probiotics. Nutraceuticals and dietary supplements also include bio-engineered foods genetically engineered to have a desired property, also known as "pharmafoods.”
  • Exemplary useful surface stabilizers include, but are not limited to, known organic and inorganic pharmaceutical excipients. Such excipients include various polymers, low molecular weight oligomers, natural products, and surfactants. Preferred surface stabilizers include nonionic, cationic, and ionic surfactants. Combinations of more than one surface stabilizer can be used in the invention. The surface stabilizers do not chemically interact with the active agent particles, and individually adsorbed molecules ofthe surface stabilizer are essentially free of intermolecular crosslinkages.
  • surface stabilizers include hydroxypropyl methylcellulose, hydroxypropylcellulose, polyvinylpyrrolidone, random copolymers of vinyl pyrrolidone and vinyl acetate, sodium lauryl sulfate, dioctylsulfosuccinate, gelatin, casein, lecithin (phosphatides), dextran, gum acacia, cholesterol, tragacanth, stearic acid, benzalkonium chloride, calcium stearate, glycerol monostearate, cetostearyl alcohol, cetomacrogol emulsifying wax, sorbitan esters, polyoxyethylene alkyl ethers (e.g., macrogol ethers such as cetomacrogol 1000), polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters (e.g., the commercially available Tweens ® such as e.g., Tween 20 ® and Tween 80 ® (
  • cationic surface stabilizers include, but are not limited to, polymers, biopolymers, polysaccharides, cellulosics, alginates, phospholipids, and nonpolymeric compounds, such as zwitterionic stabilizers, poly-n-methylpyridimum, anthryul pyridinium chloride, cationic phospholipids, chitosan, polylysine, polyvinylimidazole, polybrene, polymethylmethacrylate trimethylammoniumbromide bromide (PMMTMABr), hexyldesyltrimethylammonium bromide (HDMAB), and polyvinylpyrrolidone-2-dimethylaminoethyl methacrylate dimethyl sulfate.
  • zwitterionic stabilizers poly-n-methylpyridimum, anthryul pyridinium chloride, cationic phospholipids, chitosan, polylysine, polyvinylimi
  • cationic stabilizers include, but are not limited to, cationic lipids, sulfonium, phosphonium, and quarternary ammonium compounds, such as stearyltrimethylammonium chloride, benzyl-di(2-chloroethyl)ethylammonium bromide, coconut trimethyl ammonium chloride or bromide, coconut methyl dihydroxyethyl ammonium chloride or bromide, decyl triethyl ammonium chloride, decyl dimethyl hydroxyethyl ammonium chloride or bromide, C 12 .
  • cationic lipids such as stearyltrimethylammonium chloride, benzyl-di(2-chloroethyl)ethylammonium bromide, coconut trimethyl ammonium chloride or bromide, coconut methyl dihydroxyethyl ammonium chloride or bromide, decyl triethyl ammonium chloride,
  • Such exemplary cationic surface stabilizers and other useful cationic surface stabilizers are described in J. Cross and E. Singer, Cationic Surfactants: Analytical and Biological Evaluation (Marcel Dekker, 1994); P. andD. Rubingh (Editor), Cationic Surfactants: Physical Chemistry (Marcel Dekker, 1991); and J. Richmond, Cationic Surfactants: Organic Chemistry, (Marcel Dekker, 1990).
  • nonpolymeric primary stabilizers are any nonpolymeric compound, such benzalkonium chloride, a carbonium compound, a phosphonium compound, an oxonium compound, a halonium compound, a cationic organometallic compound, a quarternary phosphorous compound, a pyridinium compound, an anilinium compound, an ammonium compound, a hydroxylammonium compound, a primary ammonium compound, a secondary ammonium compound, a tertiary ammonium compound, and quarternary ammonium compounds ofthe formula NR 1 R 2 R 3 R 4 (+) .
  • benzalkonium chloride a carbonium compound, a phosphonium compound, an oxonium compound, a halonium compound, a cationic organometallic compound, a quarternary phosphorous compound, a pyridinium compound, an anilinium compound, an ammonium compound, a hydroxylammonium compound, a primary am
  • R r R 4 are CH 3 ;
  • one of R ⁇ -R 4 is CH 3 ;
  • three of R r R 4 are CH 3 ;
  • all of R r R 4 are CH 3 ;
  • two of R r R 4 are CH 3 , one of R ⁇ -R- 4 is C 6 H 5 CH 2 , and one of Rj-R 4 is an alkyl chain of seven carbon atoms or less;
  • two of R R 4 are CH 3 , one of RrR 4 is C 6 H 5 CH 2 , and one of R r R 4 is an alkyl chain of nineteen carbon atoms or more;
  • two of R r R 4 are CH 3 and one of R R 4 is the group C 6 H 5 (CH 2 ) n , where n>l;
  • two of Rj- - 4 are CH 3 , one of R r R 4 is C 6 H 5 CH 2 , and one of R r R 4
  • Such compounds include, but are not limited to, " behenalkonium chloride, benzethonium chloride, cetylpyridinium chloride, behentrimonium chloride, lauralkonium chloride, cetalkonium chloride, cetrimonium bromide, cetrimonium chloride, cethylamine hydrofluoride, chlorallyhnethenamine chloride (Quaternium-15), distearyldimonium chloride (Quaternium-5), dodecyl dimethyl ethylbenzyl ammonium chloride(Quaternium-14), Quaternium-22, Quaternium-26, Quaternium-18 hectorite, dimethylaminoethylchloride hydrochloride, cysteine hydrochloride, diethanolammonium POE (10) oletyl ether phosphate, diethanolammonium POE (3)oleyl ether phosphate, tallow alkonium chloride, dimethyl dioctadecylammoniumben
  • compositions ofthe invention contain nanoparticulate active agent particles which have an effective average particle size of less than about 2 microns, less than about 1 micron, less than about 600 nm, less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 200 nm, less than about 100 nm, or less than about 50 nm, as measured by light-scattering methods, microscopy, or other appropriate methods.
  • an effective average particle size of "less than about 2 microns” it is meant that at least 50% ofthe active agent particles have a weight average particle size of less than about 2 microns when measured by light scattering techniques, microscopy, or other appropriate methods.
  • at least 70% ofthe active agent particles have an average particle size of less than about 2 microns, more preferably at least 90% ofthe active agent particles have an average particle size of less than about 2 microns, and even more preferably at least about 95% ofthe particles have a weight average particle size of less than about 2 microns.
  • the relative amount of active agent and one or more surface stabilizers can vary widely.
  • the optimal amount ofthe one or more surface stabilizers can depend, for example, upon the particular active agent selected, the hydrophilic lipophilic balance (HLB), melting point, and water solubility ofthe surface stabilizer, and the surface tension of water solutions ofthe surface stabilizer, etc.
  • HLB hydrophilic lipophilic balance
  • the concentration ofthe at least one active agent can vary from about 99.5% to about 0.001%, from about 95% to about 0.1%, or from about 90% to about 0.5%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
  • the concentration ofthe at least one surface stabilizer can vary from about 0.001 to about 99.5%, from about 0.1% to about 95%, and from about 0.5% to about 90%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
  • Nanoparticulate active agent compositions can be made using methods known in the art such as, for example, milling, homogenization, and precipitation techniques. Exemplary methods of making nanoparticulate compositions are described in U.S. Patent No. 5,145,684. Methods of making nanoparticulate compositions are also described in U.S. Patent Nos. 5,518,187; 5,718,388; 5,862,999; 5,665,331; 5,662,883; 5,560,932; 5,543,133; 5,534,270; 5,510,118; and 5,470,583, all referenced in the background ofthe invention.
  • Milling an active agent to obtain a nanoparticulate composition comprises dispersing active agent particles in a liquid dispersion medium in which the active agent is poorly soluble, followed by applying mechanical means in the presence of grinding media to reduce the particle size ofthe active agent particles to the desired effective average particle size.
  • the dispersion medium can be, for example, water, safflower oil, ethanol, t-butanol, glycerin, polyethylene glycol (PEG), hexane, or glycol.
  • the active agent particles can be reduced in size in the presence of at least one surface stabilizer.
  • the active agent particles can be contacted with one or more surface stabilizers after attrition.
  • Other compounds, such as a diluent, can be added to the active agent/surface stabilizer composition during the size reduction process.
  • Dispersions can be manufactured continuously or in a batch mode.
  • the resultant nanoparticulate active agent dispersion can be utilized in solid or liquid dosage formulations, such as controlled release formulations, solid dose fast melt formulations, aerosol formulations, lyophilized formulations, tablets, capsules, solid lozenge, powders, etc.
  • Another method of forming the desired nanoparticulate active agent composition is by microprecipitation.
  • This is a method of preparing stable dispersions of poorly soluble active agents in the presence of one or more surface stabilizers and one or more colloid stability enhancing surface active agents free of any trace toxic solvents or solubilized heavy metal impurities.
  • Such a method comprises, for example: (1) dissolving an active agent in a suitable solvent; (2) adding the formulation from step (1) to a solution comprising at least one surface stabilizer; and (3) precipitating the formulation from step (2) using an appropriate non-solvent.
  • the method can be followed by removal of any formed salt, if present, by dialysis or diafiltration and concentration ofthe dispersion by conventional means.
  • the resultant nanoparticulate active agent dispersion can be utilized in solid or liquid dosage formulations, such as controlled release formulations, solid dose fast melt formulations, aerosol formulations, lyophilized formulations, tablets, solid lozenge, powders, capsules, etc.
  • nanoparticulate compositions ofthe present invention can be administered to humans and animals either orally, rectally, parenterally (intravenous, intramuscular, or subcutaneous), mtracisternally, intravaginally, intraperitoneally, locally (powders, ointments or drops), or as a buccal or nasal spray.
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance ofthe required particle size in the case of dispersions, and by the use of surfactants.
  • the nanoparticulate compositions may also contain adjuvants, such as preserving, wetting, emulsifying, and dispensing agents. Prevention ofthe growth of microorganisms can be ensured by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like. Prolonged absorption ofthe injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one ofthe following: (a) one or more inert excipients (or carrier), such as sodium citrate or dicalcium phosphate; (b) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; (c) binders, such as carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants, such as glycerol; (e) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (f) solution retarders, such as paraffin; (g) absorption accelerators, such as quaternary ammonium compounds; (h) wetting agents, such as
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may comprise inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers.
  • Exemplary emulsifiers are ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, such as cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil, glycerol, tetrahydro ⁇ urfuryl alcohol, polyethyleneglycols, fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • oils such as cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil
  • glycerol tetrahydro ⁇ urfuryl alcohol
  • polyethyleneglycols fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Actual dosage levels of active agent in the nanoparticulate compositions ofthe invention may be varied to obtain an amount of active agent that is effective to obtain a desired therapeutic response for a particular composition and method of administration.
  • the selected dosage level therefore depends upon the desired therapeutic effect, on the route of administration, on the desired duration of treatment, and other factors.
  • the total daily dose ofthe compounds of tins invention administered to a host in single or divided dose may be in amounts of, for example, from about 1 nanomole to about 5 micromoles per kilogram of body weight. Dosage unit compositions may contain such amounts of such submultiples thereof as may be used to make up the daily dose.
  • the purpose of this example was to determine the effect on the stability of paclitaxel at a basic pH when the drug is formulated into a nanoparticulate composition.
  • Paclitaxel is a naturally occurring diterpenoid which has demonstrated great potential as an anti-cancer drug. Paclitaxel can be isolated from the bark ofthe western yew, Taxus brevifolia, and is also found in several other yew species such as T baccata and T. cuspidata. Upon exposure to a basic pH (i.e., a pH of about 9), the drug rapidly degrades. Ringel et al., J Pharmac. Exp. Ther., 242:692-69% (1987).
  • paclitaxel Two formulations of paclitaxel were prepared: a solubilized formulation of paclitaxel and a nanoparticulate formulation of paclitaxel. The degradation of paclitaxel for both formulations was then compared.
  • Formulation I paclitaxel (Biolyse; Quebec, Canada) was solubilized in 1% methanol and 99% H 2 O to make a 2% paclitaxel solution.
  • Formulation II was prepared by milling the 2% paclitaxel solution with 1% Plurionic F108TM (BASF) in a 0.5 oz amber bottle containing 7.5 ml 0.5 mm Yttria-doped Zirconia media on a U.S. Stoneware Roller Mill for 72 hours.
  • BASF Plurionic F108TM
  • the resultant milled paclitaxel composition had an effective average particle size of about 220 nm, as measured by a Coulter Counter (Coulter Electronics Inc.). Both solubilized paclitaxel (Formulation I) and nanoparticulate paclitaxel (Formulation II) were incubated with 0.005 N NaOH solution (a basic solution). At the end ofthe incubation period, base degradation of paclitaxel was stopped by adding to the incubation solution 1/100 its volume of IN HCl. The recovery of paclitaxel was then measured at various time periods by HPLC.
  • solubilized paclitaxel rapidly degraded when exposed to basic pH conditions, as only about 20% ofthe paclitaxel was recoverable after a 20 minute incubation period.
  • nanoparticulate paclitaxel was essentially stable under basic conditions, as more than 90% ofthe drug was recoverable after the same incubation period.
  • the purpose of this example was to determine the stability of rapamycin when the drug is formulated into a nanoparticulate composition.
  • Rapamycin is useful as an immunosuppressant and as an antifungal antibiotic, and its use is described in, for example, U.S. Patent Nos. 3,929,992, 3,993,749, and 4,316,885, and in Belgian Pat. No. 877,700.
  • the compound which is only slightly soluble in water, i.e., 20 micrograms per mL, rapidly hydrolyzes when exposed to water.
  • rapamycin is highly unstable when exposed to an aqueous medium, special injectable formulations have been developed for administration to patients, such as those described in European Patent No. EP 041,795. Such formulations are often undesirable, as frequently the non-aqueous solubilizing agent exhibits toxic side effects.
  • Formulations I and II had effective average particle sizes of 194 nm and 199 nm, respectively.
  • Formulation I Frozen/thawed 2 days 95% ⁇ detection limit
  • Formulation II Frozen thawed 2 days 98% ⁇ detection limit
  • Formulation III Frozen thawed 2 days 97% ⁇ detection limit
  • the purpose of this example was to determine the effect of rapamycin concentration on the chemical stability of rapamycin in a nanoparticulate formulation following autoclaving.
  • rapamycin formulations were prepared by milling the following three slurries in a 250 ml PyrexTM bottle containing 125 ml 0.4 mm Yttria-doped Zirconia media for 72 hours on a U.S. Stoneware roller mill:
  • Each ofthe three dispersions was then diluted with water to prepare formulations having rapamycin concentrations of 4.4%, 2.2%, 1.1% and 0.5% as follows: (1) Formulation 1 : a mixture of 4.4% rapamycin and, prior to dilution, 1.25% Plurionic F68TM in an aqueous medium; (2) Formulation 2: a mixture of 4.4% rapamycin and, prior to dilution, 2.5% Plurionic F68TM in an aqueous medium;
  • Formulation 3 a mixture of 4.4% rapamycin and, prior to dilution, 5% Plurionic F68TM in an aqueous medium;
  • Fonnulation 4 a mixture of 2.2% rapamycin and, prior to dilution, 1.25% Plurionic F68TM in an aqueous medium;
  • Formulation 5 a mixture of 2.2% rapamycin and, prior to dilution, 2.5% Plurionic F68TM in an aqueous medium;
  • Fonnulation 6 a mixture of 2.2% rapamycin and, prior to dilution, 5% Plurionic F68TM in an aqueous medium;
  • Formulation 7 a mixture of 1.1% rapamycin and, prior to dilution, 1.25% Plurionic F68TM in an aqueous medium;
  • Formulation 8 a mixture of 1.1% rapamycin and, prior to dilution, 2.5% Plurionic F68TM in an aqueous medium;
  • Formulation 9 a mixture of 1.1 % rapamycin and, prior to dilution, 5% Plurionic F68TM in an aqueous medium;
  • Formulation 11 a mixture of 0.55% rapamycin and, prior to dilution, 2.5% Plurionic F68TM in an aqueous medium;
  • Formulation 12 a mixture of 0.55% rapamycin and, prior to dilution, 5% Plurionic F68TM in an aqueous medium;
  • the purpose of this example was to determine the chemical stability of a nanoparticulate rapamycin formulation following a prolonged storage period at room temperature.
  • a mixture of 20% rapamycin and 10% Plurionic F68TM in an aqueous medium was milled with 0.4 mm YTZ media (Performance Ceramic Co.) on a U.S. Stoneware mill for 72 hours at room temperature.
  • the final nanoparticulate composition had a mean particle size of between 180 to 230 nm, as measured by Coulter sizing.
  • the purpose of this example was to detennine the effect of long term storage on the chemical stability of rapamycin in a nanoparticulate composition.
  • rapamycin fonnulations were prepared as follows: Formulation 1, having a rapamycin concentration of 182.8 mg/mL; Formulation 2, having a rapamycin concentration of 191.4 mg/mL; and Formulation 3, having a rapamycin concentration of 192.7 mg/mL.
  • the formulations were prepared by milling the following three slurries in a 0.5 oz amber bottle containing 7.5 ml 0.8 mm Yttria-doped Zirconia media for 72 hours on a U.S. Stoneware roller mill:

Abstract

Methods for stabilizing active agents, particularly pharmaceutical agents, are described. The method comprises forming active agents into a nanoparticulate composition comprising the active agent and at least one surface stabilizer. The component active agent exhibits chemical stability, even following exposure to, for example, a prolonged storage period, an elevated temperature, light, radiation, radiation causing photolysis, non-physiological pH, enzymes or other catalysts, water or other solvent molecules, oxidizing agents or other free radicals, or freezing.

Description

STABILIZATION OF ACTIVE AGENTS BY FORMULATION INTO NANOPARTICULATE FORM
FIELD OF THE INVENTION
The present invention is directed to methods for stabilizing active agents, particularly pharmaceutical agents, comprising formulating a at least one active agent, which is unstable under one or more environmental conditions, into a nanoparticulate composition. The nanoparticulate active agent is stable under the same environmental conditions under which the active agent is unstable prior to formulating the agent into a nanoparticulate composition. The nanoparticulate composition comprises an active agent and one or more surface stabilizers adhered to the surface ofthe active agent.
BACKGROUND OF THE INVENTION A. Summary of Instability and/or Degradation of Active Agents
Active agents are subject to degradation. Such decomposition or degradation may be due to hydrolysis, oxidation, isomerization, epimerization, or photolysis. The rate of degradation or decomposition varies considerably depending on the structural, physical, and chemical nature ofthe active agent. The rate of decomposition is also often significantly affected by numerous environmental factors, including temperature, light, radiation, enzyme or other catalysts, pH and ionic strength ofthe solution, solvent type, and buffer species.
Chemical instability due to degradation or decomposition is highly undesirable for several reasons. For example, when a chemical compound is a pharmaceutical agent, degradation decreases its efficiency and shortens its effective shelf life. Moreover, the decrease in the content ofthe active ingredient in a pharmaceutical preparation renders the calculation of an effective dosage unpredictable and difficult. Furthermore, degraded chemical agent may have highly undesirable or even severely toxic side effects.
Because chemical stability is a critical aspect in the design and manufacture, as well as regulatory review and approval, of pharmaceutical compositions and dosage forms, in recent years extensive and systematic studies have been conducted on the mechanisms and kinetics of decomposition of pharmaceutical agents. For a brief review, see Alfred Martin, Physical Pharmacy: Physical Chemical Principles in the Pharmaceutical Sciences, 4th Edition, pp. 305-312 (Lee & Febiger, Philadelphia, 1993).
B. Prior Methods for Increasing the Stability of a Chemical Compound in Pharmaceutically Acceptable Formulations
Various methods have been devised to achieve improved chemical stability of a compound, including alteration of environmental parameters, such as buffer type, pH, storage temperature, and elimination of catalytic ions or ions necessary for enzyme activity using chelating agents.
Other methods include converting the drug into a more stable prodrug which, under physiological conditions, is processed to become a biologically active form ofthe compound.
Another method for improving the chemical stability of pharmaceutical agents employs novel dosage form designs. Dosage form designs that improve the chemical stability of a drug include loading drugs into liposomes or polymers, e.g., during emulsion polymerization. However, such techniques have problems and limitations. For example, a lipid soluble drug is often required to prepare a suitable liposome. Further, unacceptably large amounts ofthe liposome or polymer may be required to prepare unit drug doses. Further still, techniques for preparing such pharmaceutical compositions tend to be complex. Finally, removal of contaminants at the end ofthe emulsion polymerization manufacturing process, such as potentially toxic unreacted monomer or initiator, can be difficult and expensive.
Another example of a dosage fonn that can be used to increase the stability of an administered agent is a monolithic device, which is a rate-controlling polymer matrix throughout which a drug is dissolved or dispersed. Yet another example of such a dosage form is a reservoir device, which is a shell-like dosage form having a drug contained within a rate-controlling membrane.
An exemplary reservoir dosage form is described in U.S. Patent No. 4,725,442, which refers to water insoluble drug materials solubilized in an organic liquid and incorporated in microcapsules of phospholipids. One disadvantage of this dosage form is the toxic effects of the solubilizing organic liquids. Other methods of forming reservoir dosage forms of pharmaceutical drug microcapsules include micronizing a slightly-soluble drug by high-speed stirring or impact comminution of a mixture ofthe drug and a sugar or sugar alcohol together with suitable excipients or diluents. See e.g. EP 411,629A. One disadvantage of this method is that the resultant drug particles are larger than those obtained with milling. Yet another method of forming a reservoir dosage form is directed to polymerization of a monomer in the presence of an active drug material and a surfactant to produce small-particle microencapsulation (InternationalJournal of Pharmaceutics, 52:101-108 (1989)). This process, however, produces compositions containing contaminants, such as toxic monomers, which are difficult to remove. Complete removal of such monomers can be expensive, particularly when conducted on a manufacturing scale. A reservoir dosage form can also be formed by co-dispersion of a drug or a pharmaceutical agent in water with droplets of a carbohydrate polymer (see e.g. U.S. Patent No. 4,713,249 and WO 84/00294). The major disadvantage of this procedure is that in many cases, a solubilizing organic co-solvent is required for the encapsulation procedure. Removal of traces of such harmful co-solvents can result in an expensive manufacturing process.
C. Background Regarding Nanoparticulate Compositions
Nanoparticulate compositions, first described in U.S. Patent No. 5,145,684 ("the '684 patent"), are particles consisting of a poorly soluble active agent having adsorbed onto the surface thereof a non-crosslinked surface stabilizer. The '684 patent also describes methods of making such nanoparticulate compositions. The '684 patent teaches that nanoparticulate compositions are desirable because with a decrease in particle size, and a consequent increase in surface area, a composition can exhibit superior bioavailability.
Methods of making nanoparticulate compositions are described, for example, in U.S. Patent Nos. 5,518,187 and 5,862,999, both for "Method of Grinding Pharmaceutical Substances;" U.S. Patent No. 5,718,388, for "Continuous Method of Grinding Pharmaceutical Substances;" and U.S. Patent No. 5,510,118 for "Process of Preparing Therapeutic Compositions Containing Nanoparticles."
Nanoparticulate compositions are also described, for example, in U.S. Patent Nos. 5,298,262 for "Use of Ionic Cloud Point Modifiers to Prevent Particle Aggregation During Sterilization;" 5,302,401 for "Method to Reduce Particle Size Growth During Lyophilization;" 5,318,767 for "X-Ray Contrast Compositions Useful in Medical Imaging;" 5,326,552 for "Novel Formulation For Nanoparticulate X-Ray Blood Pool Contrast Agents Using High Molecular Weight Non-ionic Surfactants;" 5,328,404 for "Method of X-Ray Imaging Using Iodinated Aromatic Propanedioates;" 5,336,507 for "Use of Charged Phospholipids to Reduce Nanoparticle Aggregation;" 5,340,564 for "Formulations Comprising Olin 10-G to Prevent Particle Aggregation and Increase Stability;" 5,346,702 for "Use of Non-Ionic Cloud Point Modifiers to Minimize Nanoparticulate Aggregation During Sterilization;" 5,349,957 for "Preparation and Magnetic Properties of Very Small Magnetic- Dextran Particles;" 5,352,459 for "Use of Purified Surface Modifiers to Prevent Particle Aggregation During Sterilization;" 5,399,363 and 5,494,683, both for "Surface Modified Anticancer Nanoparticles;" 5,401,492 for "Water Insoluble Non-Magnetic Manganese Particles as Magnetic Resonance Enhancement Agents;" 5,429,824 for "Use of Tyloxapol as a Nanoparticulate Stabilizer;" 5,447,710 for "Method for Making Nanoparticulate X-Ray Blood Pool Contrast Agents Using High Molecular Weight Non-ionic Surfactants;" 5,451,393 for "X-Ray Contrast Compositions Useful in Medical Imaging;" 5,466,440 for "Formulations of Oral Gastrointestinal Diagnostic X-Ray Contrast Agents in Combination with Pharmaceutically Acceptable Clays;" 5,470,583 for "Method of Preparing Nanoparticle Compositions Containing Charged Phospholipids to Reduce Aggregation;" 5,472,683 for "Nanoparticulate Diagnostic Mixed Carbamic Anhydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging;" 5,500,204 for "Nanoparticulate Diagnostic Dimers as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging;" 5,518,738 for "Nanoparticulate NSAID Formulations;" 5,521,218 for "Nanoparticulate Iododipamide Derivatives for Use as X-Ray Contrast Agents;" 5,525,328 for "Nanoparticulate Diagnostic Diatrizoxy Ester X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging;" 5,543,133 for "Process of Preparing X-Ray Contrast Compositions Containing Nanoparticles;" 5,552,160 for "Surface Modified NSAID Nanoparticles;" 5,560,931 for "Formulations of Compounds as Nanoparticulate Dispersions in Digestible Oils or Fatty Acids;" 5,565,188 for "Polyalkylene Block Copolymers as Surface Modifiers for Nanoparticles;" 5,569,448 for "Sulfated Non-ionic Block Copolymer Surfactant as Stabilizer Coatings for Nanoparticle Compositions;" 5,571,536 for "Formulations of Compounds as Nanoparticulate Dispersions in Digestible Oils or Fatty Acids;" 5,573,749 for "Nanoparticulate Diagnostic Mixed Carboxylic Anydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging;" 5,573,750 for "Diagnostic Imaging X-Ray Contrast Agents;" 5,573,783 for "Redispersible Nanoparticulate Film Matrices With Protective Overcoats;" 5,580,579 for "Site-specific Adhesion Within the GI Tract Using Nanoparticles Stabilized by High Molecular Weight, Linear Poly(ethylene Oxide) Polymers;" 5,585,108 for "Formulations of Oral Gastrointestinal Therapeutic Agents in Combination with Pharmaceutically Acceptable Clays;" 5,587,143 for "Butylene Oxide-Ethylene Oxide Block Copolymers Surfactants as Stabilizer Coatings for Nanoparticulate Compositions;" 5,591,456 for "Milled Naproxen with Hydroxypropyl Cellulose as Dispersion Stabilizer;" 5,593,657 for "Novel Barium Salt Formulations Stabilized by Non-ionic and Anionic Stabilizers;" 5,622,938 for "Sugar Based Surfactant for Nanocrystals;" 5,628,981 for "Improved Formulations of Oral Gastrointestinal Diagnostic X-Ray Contrast Agents and Oral Gastrointestinal Therapeutic Agents;" 5,643,552 for "Nanoparticulate Diagnostic Mixed Carbonic Anhydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging;" 5,718,388 for "Continuous Method of Grinding Pharmaceutical Substances;" 5,718,919 for "Nanoparticles Containing the R(-)Enantiomer of Ibuprofen;" 5,747,001 for "Aerosols Containing Beclomethasone Nanoparticle Dispersions;" 5,834,025 for "Reduction of Intravenously Administered Nanoparticulate Formulation Induced Adverse Physiological Reactions;" 6,045,829 "Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors Using Cellulosic Surface Stabilizers;" 6,068,858 for "Methods of Making Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors Using Cellulosic Surface Stabilizers;" 6,153,225 for "Injectable Formulations of Nanoparticulate Naproxen;" 6,165,506 for "New Solid Dose Form of Nanoparticulate Naproxen;" 6,221,400 for "Methods of Treating Mammals Using Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors;" 6,264,922 for "Nebulized Aerosols Containing Nanoparticle Dispersions;" 6,267,989 for "Methods for Preventing Crystal Growth and Particle Aggregation in Nanoparticle Compositions;" 6,270,806 for "Use of PEG-Derivatized Lipids as Surface Stabilizers for Nanoparticulate Compositions;" 6,316,029 for "Rapidly Disintegrating Solid Oral Dosage Form," 6,375,986 for "Solid Dose Nanoparticulate Compositions Comprising a Synergistic Combination of a Polymeric Surface Stabilizer and Dioctyl Sodium Sulfosuccinate;" 6,428,814 for "Bioadhesive nanoparticulate compositions having cationic surface stabilizers;" and 6,432,381 for "Methods for targeting drug delivery to the upper and/or lower gastrointestinal tract," all of which are specifically incorporated by reference. In addition, U.S. Patent Application No. 20020012675 Al, published on January 31, 2002, for "Controlled Release Nanoparticulate Compositions," describes nanoparticulate compositions, and is specifically incorporated by reference.
Amorphous small particle compositions are described, for example, in U.S. Patent Nos. 4,783,484 for "Particulate Composition and Use Thereof as Antimicrobial Agent;" 4,826,689 for "Method for Making Uniformly Sized Particles from Water-Insoluble Organic Compounds;" 4,997,454 for "Method for Making Uniformly-Sized Particles From Insoluble Compounds;" 5,741,522 for "Ultrasmall, Non-aggregated Porous Particles of Uniform Size for Entrapping Gas Bubbles Within and Methods;" and 5,776,496, for "Ultrasmall Porous Particles for Enhancing Ultrasound Back Scatter."
In particular, U.S. Patent Nos. 5,399,363 and 5,494,683, both for "Surface Modified Anticancer Nanoparticles," refer to nanoparticulate compositions of anticancer agents, including taxol, also known as paclitaxel. However, these patents do not address a means to overcome the chemical instability of an active agent, such as paclitaxel at a basic pH. Specifically, the two patents teach methods of making nanoparticulate paclitaxel compositions which are stable at a pH of 7.4. However, this does not teach or suggest that a nanoparticulate paclitaxel composition is stable at an elevated pH, such as a basic pH of 9.
These references describe methods of increasing the bioavailability of poorly soluble active agents, in which the class of "active agents" encompassed is limited only by the solubility ofthe active agent. In contrast, the present invention is directed to a class of poorly soluble active agents which are unstable under one or more environmental conditions. The two classes of active agents are not the same, as all poorly soluble active agents are not unstable under one or more environmental conditions.
This is significant as the pH varies along the gastrointestinal tract (GIT). If a cancer is localized in a region ofthe GIT which has a basic pH, the conventional form of paclitaxel will be ineffective, or poorly effective, as the drug degrades at a high pH. This problem can be addressed by nanoparticulate forms of paclitaxel, which are stable at a high pH.
U.S. Patent No. 5,989,591 to Nagi, for "Rapamycin Formulations for Oral Administration," refers to solid dose compositions of rapamycin in which the rapamycin can have a nanoparticulate particle size. See Nagi at col. 6, lines 50-55. However, Nagi does not teach that the nanoparticulate rapamycin composition is stable when exposed to an aqueous environment. In fact, Nagi teaches that the nanoparticulate rapamycin composition is formulated into a tablet dosage form, in which rapamycin is present in a sugar coating over a solid core. See the Abstract and col. 3, lines 45-52.
This does not teach or suggest that an unstable form of rapamycin, i.e., rapamycin exposed to an aqueous environment, could be selected and made stable under that same environment by formulating the drug into a nanoparticulate composition. This is significant as tablet dosage forms have a longer onset of activity as compared to liquid or IV dosage forms, h addition, many infant and elderly patients are unable to swallow tablet dosage forms.
* * * *
There is a need in the art for a method of stabilizing active agents, which is efficient, cost-effective, and does not require the addition of potentially toxic solvents. The present invention satisfies this need.
SUMMARY OF THE INVENTION
The present invention is directed to the discovery that active agents which are unstable under one or more enviromnental conditions, when formulated into nanoparticulate compositions, are stable under those same environmental conditions. Such environmental conditions can be, for example, exposure to a condition such as prolonged storage periods, elevated temperature, non-physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water, solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles. One aspect ofthe invention is directed to a process for stabilizing active agents, particularly pharmaceutical agents, comprising formulating an active agent which is unstable under one or more environmental conditions into a nanoparticulate composition, wherein the active agent is then stable under those same environmental conditions. The nanoparticulate composition comprises a poorly soluble active agent, such as a drug particle, and one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe active agent. The nanoparticulate compositions have an effective average particle size of less than about two microns.
The present invention is further directed to a process for stabilizing rapamycin, which is unstable when exposed to water, comprising forming a nanoparticulate formulation of rapamycin having one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe drug. The resultant nanoparticulate rapamycin composition exhibits dramatically superior stability when exposed to water. The pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier, as well as any desired excipients.
Yet another aspect ofthe invention encompasses a process for stabilizing paclitaxel, which is unstable when exposed to a basic pH, comprising forming a nanoparticulate composition of paclitaxel having one or more non-crosslinked surface stabilizers adsorbed on to the surface ofthe drug. The resultant nanoparticulate paclitaxel composition exhibits dramatically superior stability when exposed to a basic pH. The pharmaceutical composition preferably comprises a pharmaceutically acceptable carrier, as well as any desired excipients.
Both the foregoing general description and the following detailed description are exemplary and explanatory and are intended to provide further explanation ofthe invention as claimed. Other objects, advantages, and novel features will be readily apparent to those skilled in the art from the following detailed description ofthe invention.
BRIEF DESCRIPTION OF THE FIGURE
Figure 1 : Shows the effect of 0.005 N NaOH (a basic pH level) on the rate of degradation of paclitaxel as compared to the rate of degradation of a nanoparticulate formulation of paclitaxel. DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to a method for stabilizing active agents, particularly pharmaceutical agents, which are unstable under one or more environmental conditions, comprising formulating an active agent into a nanoparticulate composition. The nanoparticulate active agent is stable under the same one or more environmental conditions under which the active agent is unstable prior to formulation into a nanoparticulate composition. Such one or more environmental conditions which can cause the active agent to degrade include, but are not limited to, prolonged storage periods, elevated temperature, non- physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water, solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles.
The method ofthe invention comprises first identifying and selecting an active agent which is unstable under one or more environmental conditions, followed by formulating particles ofthe active agent into a stable nanoparticulate composition. The formulation step comprises combining the active agent particles and at least one non-crosslinked surface stabilizer, wherein after combining the at least one non-crosslinked surface stabilizer adsorbs to the surface ofthe active agent particles to produce a nanoparticulate composition in which the active agent particles have an effective average particle size of less than about 2 microns.
The claimed method is distinct from prior art methods of making nanoparticulate compositions, as prior methods did not teach that active agents which are unstable under one or more environmental conditions can be made stable under those same conditions by formulating the active agent into a nanoparticulate composition. Rather, prior methods of making nanoparticulate compositions teach that the nanoparticulate compositions are desirable as they can increase the bioavailability ofthe component active agent. See e.g., U.S. Patent No. 5,145,684.
This distinction is significant as the present invention can expand the use of many active agents which may have only been useable under limited conditions prior to the present invention. For example, active agents which are stable only under certain pH conditions, such as paclitaxel, can be formulated for administration at any desired pH level. This is desirable as the pH varies along the human gastrointestinal tract. Similarly, active agents which are unstable when exposed to elevated temperatures can be heat sterilized without degradation when formulated into a nanoparticulate composition. This can be useful in administering active agents to very young or immunocompromised patients. Moreover, active agents which are unstable when exposed to water (i.e., hydrolysis), such as rapamycin, can be formulated for administration in a liquid form. This can be beneficial as tablet dosage forms have a longer onset of activity as compared to liquid or IV dosage forms. In addition, many infant and elderly patients are unable to swallow tablet dosage forms.
A. Active Agents Formulated into Nanoparticulate Compositions Exhibit Increased Stability of the Component Active Agent
Chemical instability due to degradation is usually a result of hydrolysis, oxidation, isomerization, epimerization, or photolysis. Apart from the structural, physical, and chemical nature ofthe active agent, the rate of degradation is often determined by numerous enviromnental factors, including temperature, light, radiation, enzyme or other catalysts, pH and ionic strength ofthe solution, solvent type, or buffer species.
While not intending to be bound by theory, one possibility is that the molecules ofthe surface stabilizer shield the active agent, thereby protecting potentially labile chemical groups ofthe active agent from the potentially hostile environment. Another possibility is that for a crystalline active agent particle, the crystalline structure in a nanoparticulate sized formulation results in greater active agent stability.
For example, rapamycin is rapidly degraded when exposed to an aqueous environment. The main degradation scheme of rapamycin is the cleavage ofthe macrocyclic lactone ring by the hydrolysis of an ester bond to form a secoacid (SECO). The secoacid undergoes further dehydration and isomerization to form diketomorpholine analogs.
However, as described in the examples below, when rapamycin is formulated into a nanoparticulate composition, minimal or no rapamycin degradation is observed, even following prolonged exposure to an aqueous medium.
Another example of an active agent that is unstable under certain environmental conditions, but which is stable in a nanoparticulate formulation under those same environmental conditions, is paclitaxel. Upon exposure to a basic pH (i.e., a pH of about 9), paclitaxel rapidly degrades. Ringel et al, J Pharmac. Exp. Ther., 242:692-698, (1987). However, when paclitaxel is formulated into a nanoparticulate composition, minimal or no paclitaxel degradation is observed, even when the composition is exposed to a basic pH.
B. Methods of Preparing Nanoparticulate Compositions
1. Active Agent and Surface Stabilizer Components
The method of stabilizing an active agent according to the present invention comprises formulating the active agent into a nanoparticulate formulation. The nanoparticulate active agent formulation comprises at least one active agent and one or more surface stabilizers adsorbed to the surface ofthe active agent(s).
a. Active Agent Particles
The nanoparticles ofthe invention comprise an active agent, such as a therapeutic or diagnostic agent, having one or more labile groups or exhibiting chemical instability when exposed to certain environmental conditions, such as prolonged storage periods, elevated temperature, non-physiological pH, light, radiation, radiation causing photolysis, enzymes or other catalysts, water (which can cause hydrolysis), solvent molecules, oxidizing agents, free radicals, and freezing-thawing temperature cycles. In addition, active agents having, for example, cosmetic, diagnostic, or bioengineering uses are presumed suitable for the invention.
The active agent may be present either substantially in the form of one optically pure enantiomer or as a mixture, racemic or otherwise, of enantiomers.
A therapeutic agent can be a pharmaceutical, including biologies such as proteins and peptides, and a diagnostic agent is typically a contrast agent, such as an x-ray contrast agent, or any other type of diagnostic material. The active agent particle exists as a discrete, crystalline phase, amorphous phase, semi-crystalline phase, semi-amorphous phase, or as a combination thereof. The crystalline phase differs from a non-crystalline or amorphous phase which results from precipitation techniques, such as those described in EP Patent No. 275,796.
The active agent is preferably present in an essentially pure form, is poorly soluble, and is dispersible in at least one liquid medium. By "poorly soluble" it is meant that the active agent has a solubility in a liquid dispersion medium of less than about 30 mg/mL, less than about 10 mg/mL, or less than about 1 mg/mL. The dispersion medium can be, for example, water, safflower oil, ethanol, t-butanol, glycerin, polyethylene glycol (PEG), hexane, or glycol.
The active agent can be selected from a variety of known classes of drugs, including, for example, COX-2 inhibitors, retinoids, NSAIDS, proteins, peptides, nucleotides, anti- obesity drugs, nutraceuticals, dietary supplements, carotenoids, corticosteroids, elastase inhibitors, anti-fungals, oncology therapies, anti-emetics, analgesics, cardiovascular agents, 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, anxiolytics, 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, immunological agents, lipid regulating 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, xanthines, alpha-hydroxy formulations, cystic-fibrosis therapies, asthma therapies, emphysema therapies, respiratory distress syndrome therapies, chronic bronchitis therapies, chronic obstructive pulmonary disease therapies, organ-transplant rejection therapies, therapies for tuberculosis and other infections ofthe lung, and respiratory illness therapies associated with acquired immune deficiency syndrome.
Exemplary nutraceuticals and dietary supplements are disclosed, for example, in Roberts et al., Nutraceuticals: The Complete Encyclopedia of Supplements, Herbs, Vitamins, and Healing Foods (American Nutraceutical Association, 2001), which is specifically incorporated by reference. A nutraceutical or dietary supplement, also known as phytochemicals or functional foods, is generally any one of a class of dietary supplements, vitamins, minerals, herbs, or healing foods that have medical or pharmaceutical effects on the body. Exemplary nutraceuticals or dietary supplements include, but are not limited to, lutein, folic acid, fatty acids (e.g., DHA and ARA), fruit and vegetable extracts, vitamin and mineral supplements, phosphatidylserine, lipoic acid, melatonin, glucosamine/chondroitin, Aloe Vera, Guggul, glutamine, amino acids (e.g., iso-leucine, leucine, lysine, methionine, phenylanine, threonine, tryptophan, and valine), green tea, lycopene, whole foods, food additives, herbs, phytonutrients, antioxidants, flavonoid constituents of fruits, evening primrose oil, flax seeds, fish and marine animal oils, and probiotics. Nutraceuticals and dietary supplements also include bio-engineered foods genetically engineered to have a desired property, also known as "pharmafoods."
A description of these classes of active agents and a listing of species within each class can be found in Martindale, The Extra Pharmacopoeia, Twenty-ninth Edition (The Pharmaceutical Press, London, 1989), specifically incorporated by reference. The active agents are commercially available and/or can be prepared by techniques known in the art.
b. Surface Stabilizers
Exemplary useful surface stabilizers include, but are not limited to, known organic and inorganic pharmaceutical excipients. Such excipients include various polymers, low molecular weight oligomers, natural products, and surfactants. Preferred surface stabilizers include nonionic, cationic, and ionic surfactants. Combinations of more than one surface stabilizer can be used in the invention. The surface stabilizers do not chemically interact with the active agent particles, and individually adsorbed molecules ofthe surface stabilizer are essentially free of intermolecular crosslinkages.
Representative examples of surface stabilizers include hydroxypropyl methylcellulose, hydroxypropylcellulose, polyvinylpyrrolidone, random copolymers of vinyl pyrrolidone and vinyl acetate, sodium lauryl sulfate, dioctylsulfosuccinate, gelatin, casein, lecithin (phosphatides), dextran, gum acacia, cholesterol, tragacanth, stearic acid, benzalkonium chloride, calcium stearate, glycerol monostearate, cetostearyl alcohol, cetomacrogol emulsifying wax, sorbitan esters, polyoxyethylene alkyl ethers (e.g., macrogol ethers such as cetomacrogol 1000), polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters (e.g., the commercially available Tweens® such as e.g., Tween 20® and Tween 80® (ICI Speciality Chemicals)); polyethylene glycols (e.g., Carbowaxs 3550® and 934® (Union Carbide)), polyoxyethylene stearates, colloidal silicon dioxide, phosphates, carboxymethylcellulose calcium, carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylmethylcellulose phthalate, noncrystalline cellulose, magnesium aluminium silicate, triethanolamine, polyvinyl alcohol (PVA), 4-(l, 1,3,3- tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde (also known as tyloxapol, superione, and triton), poloxamers (e.g., Pluronics F68® and F108®, which are block copolymers of ethylene oxide and propylene oxide); poloxamines (e.g., Tetronic 908®, also known as Poloxamine 908®, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine (BASF Wyandotte Corporation, Parsippany, NJ.)); Tetronic 1508® (T- 1508) (BASF Wyandotte Corporation), Tritons X-200®, which is an alkyl aryl polyether sulfonate (Rohm and Haas); Crodestas F-l 10®, which is a mixture of sucrose stearate and sucrose distearate (Croda Inc.); p-isononylphenoxypoly-(glycidol), also known as Olin-IOG® or Surfactant 10-G®(Olin Chemicals, Stamford, CT); Crodestas SL-40® (Croda, Inc.); and SA9OHCO, which is Ci8H37CH2C(O)N(CH3)-CH2(CHOH)4(CH20H)2 (Eastman Kodak Co.); decanoyl-N- methylglucamide; n-decyl β-D-glucopyranoside; n-decyl β-D-maltopyranoside; n-dodecyl β- D-glucopyranoside; n-dodecyl β-D-maltoside; heptanoyl-N-methylglucamide; n-heptyl-β-D- glucopyranoside; n-heptyl β-D-thioglucoside; n-hexyl β-D-glucopyranoside; nonanoyl-N- methylglucamide; n-noyl β-D-glucopyranoside; octanoyl-N-methylglucamide; n-octyl-β-D- glucopyranoside; octyl β-D-thioglucopyranoside; PEG-phospholipid, PEG-cholesterol, PEG- cholesterol derivative, PEG- vitamin A, PEG-vitamin E, lysozyme, and the like.
Examples of useful cationic surface stabilizers include, but are not limited to, polymers, biopolymers, polysaccharides, cellulosics, alginates, phospholipids, and nonpolymeric compounds, such as zwitterionic stabilizers, poly-n-methylpyridimum, anthryul pyridinium chloride, cationic phospholipids, chitosan, polylysine, polyvinylimidazole, polybrene, polymethylmethacrylate trimethylammoniumbromide bromide (PMMTMABr), hexyldesyltrimethylammonium bromide (HDMAB), and polyvinylpyrrolidone-2-dimethylaminoethyl methacrylate dimethyl sulfate.
Other useful cationic stabilizers include, but are not limited to, cationic lipids, sulfonium, phosphonium, and quarternary ammonium compounds, such as stearyltrimethylammonium chloride, benzyl-di(2-chloroethyl)ethylammonium bromide, coconut trimethyl ammonium chloride or bromide, coconut methyl dihydroxyethyl ammonium chloride or bromide, decyl triethyl ammonium chloride, decyl dimethyl hydroxyethyl ammonium chloride or bromide, C12.15dimethyl hydroxyethyl ammonium chloride or bromide, coconut dimethyl hydroxyethyl ammonium chloride or bromide, myristyl trimethyl ammonium methyl sulphate, lauryl dimethyl benzyl ammonium chloride or bromide, lauryl dimethyl (ethenoxy)4 ammonium chloride or bromide, N-alkyl (C12, 18)dimethylbenzyl ammonium chloride, N-alkyl (C14.18)dimethyl-benzyl ammonium chloride, N-tetradecylidmethylbenzyl ammonium chloride monohydrate, dimethyl didecyl ammonium chloride, N-alkyl and (C12-14) dimethyl 1-napthyhnethyl ammonium chloride, trimethylammonium halide, alkyl-trimethylammonium salts and dialkyl-dimethylammonium salts, lauryl trimethyl ammonium chloride, ethoxylated alkyamidoalkyldialkylammonium salt and/or an ethoxylated trialkyl ammonium salt, dialkylbenzene dialkylammonium chloride, N- didecyldimethyl ammonium chloride, N-tetradecyldimethylbenzyl ammonium, chloride monohydrate, N-alkyl(C12.14) dimethyl 1-naphthylmethyl ammonium chloride and dodecyldimethylbenzyl ammonium chloride, dialkyl benzenealkyl ammonium chloride, lauryl trimethyl ammonium chloride, alkylbenzyl methyl ammonium chloride, alkyl benzyl dimethyl ammonium bromide, C12, C15, C trimethyl ammonium bromides, dodecylbenzyl triethyl ammonium chloride, poly-diallyldimethylammonium chloride (DADMAC), dimethyl ammonium chlorides, allcyldimethylammonium halogenides, tricetyl methyl ammonium chloride, decyltrimethylammonium bromide, dodecyltriethylammonium bromide, tetradecyltrimethylammonium bromide, methyl trioctylammonium chloride (ALIQUAT 336™), POLYQUAT 10™, tetrabutylammonium bromide, benzyl trimethylammonium bromide, choline esters (such as choline esters of fatty acids), benzalkonium chloride, stearalkonium chloride compounds (such as stearyltrimonium chloride and Di- stearyldimonium chloride), cetyl pyridinium bromide or chloride, halide salts of quaternized polyoxyethylalkylamines, MIRAPOL™ and ALKAQUAT™ (Alkaril Chemical Company), alkyl pyridinium salts; amines, such as alkylamines, dialkylamines, alkanolamines, polyethylenepolyamines, N,N-dialkylaminoalkyl acrylates, and vinyl pyridine, amine salts, such as lauryl amine acetate, stearyl amine acetate, alkylpyridinium salt, and alkylimidazohum salt, and amine oxides; imide azolinium salts; protonated quaternary acrylamides; methylated quaternary polymers, such as poly[diallyl dimethylammonium chloride] and poly-[N-methyl vinyl pyridinium chloride]; and cationic guar.
Such exemplary cationic surface stabilizers and other useful cationic surface stabilizers are described in J. Cross and E. Singer, Cationic Surfactants: Analytical and Biological Evaluation (Marcel Dekker, 1994); P. andD. Rubingh (Editor), Cationic Surfactants: Physical Chemistry (Marcel Dekker, 1991); and J. Richmond, Cationic Surfactants: Organic Chemistry, (Marcel Dekker, 1990).
Particularly preferred nonpolymeric primary stabilizers are any nonpolymeric compound, such benzalkonium chloride, a carbonium compound, a phosphonium compound, an oxonium compound, a halonium compound, a cationic organometallic compound, a quarternary phosphorous compound, a pyridinium compound, an anilinium compound, an ammonium compound, a hydroxylammonium compound, a primary ammonium compound, a secondary ammonium compound, a tertiary ammonium compound, and quarternary ammonium compounds ofthe formula NR1R2R3R4 (+). For compounds ofthe formula NR1R2R3R4 (+):
(i) none of RrR4 are CH3; (ii) one of R{ -R4 is CH3 ; (iii) three of RrR4 are CH3; (iv) all of RrR4 are CH3; (v) two of RrR4 are CH3, one of Rχ-R-4 is C6H5CH2, and one of Rj-R4 is an alkyl chain of seven carbon atoms or less; (vi) two of R R4 are CH3, one of RrR4 is C6H5CH2, and one of RrR4 is an alkyl chain of nineteen carbon atoms or more; (vii) two of RrR4 are CH3 and one of R R4 is the group C6H5(CH2)n, where n>l; (viii) two of Rj- -4 are CH3, one of RrR4 is C6H5CH2, and one of RrR4 comprises at least one heteroatom; (ix) two of RrR4 are CH3, one of RrR4 is C6HSCH2, and one of RrR4 comprises at least one halogen; (x) two of RrR4 are CH3, one of RrR4 is C6H5CH2, and one of RrR4 comprises at least one cyclic fragment; (xi) two of RrR4 are CH3 and one of RrR4 is a phenyl ring; or (xii) two of R!-R4 are CH3 and two of RrR4 are purely aliphatic fragments.
Such compounds include, but are not limited to,"behenalkonium chloride, benzethonium chloride, cetylpyridinium chloride, behentrimonium chloride, lauralkonium chloride, cetalkonium chloride, cetrimonium bromide, cetrimonium chloride, cethylamine hydrofluoride, chlorallyhnethenamine chloride (Quaternium-15), distearyldimonium chloride (Quaternium-5), dodecyl dimethyl ethylbenzyl ammonium chloride(Quaternium-14), Quaternium-22, Quaternium-26, Quaternium-18 hectorite, dimethylaminoethylchloride hydrochloride, cysteine hydrochloride, diethanolammonium POE (10) oletyl ether phosphate, diethanolammonium POE (3)oleyl ether phosphate, tallow alkonium chloride, dimethyl dioctadecylammoniumbentonite, stearalkonium chloride, domiphen bromide, denatonium benzoate, myristalkonium chloride, laurtrimonium chloride, ethylenediamine dihydrochloride, guanidine hydrochloride, pyridoxine HCl, iofetamine hydrochloride, meglumine hydrochloride, methylbenzethonium chloride, myrtrimonium bromide, oleyltrimonium chloride, polyquaternium-1, procainehydrochloride, cocobetaine, stearalkonium bentonite, stearalkoniumhectonite, stearyl trihydroxyethyl propylenediamine dihydrofluoride, tallowtrimonium chloride, and hexadecyltrimethyl ammonium bromide.
Most of these surface stabilizers are known pharmaceutical excipients and are described in detail in the Handbook of Pharmaceutical Excipients, published jointly by the American Pharmaceutical Association and The Pharmaceutical Society of Great Britain (The Pharmaceutical Press, 2000), specifically incorporated by reference. The surface stabilizers are commercially available and/or can be prepared by techniques known in the art.
c. Nanoparticulate Active Agent/Surface Stabilizer Particle Size
The compositions ofthe invention contain nanoparticulate active agent particles which have an effective average particle size of less than about 2 microns, less than about 1 micron, less than about 600 nm, less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 200 nm, less than about 100 nm, or less than about 50 nm, as measured by light-scattering methods, microscopy, or other appropriate methods.
By "an effective average particle size of "less than about 2 microns," it is meant that at least 50% ofthe active agent particles have a weight average particle size of less than about 2 microns when measured by light scattering techniques, microscopy, or other appropriate methods. Preferably, at least 70% ofthe active agent particles have an average particle size of less than about 2 microns, more preferably at least 90% ofthe active agent particles have an average particle size of less than about 2 microns, and even more preferably at least about 95% ofthe particles have a weight average particle size of less than about 2 microns.
d. Concentration of Nanoparticulate Active Agent and Surface Stabilizer
The relative amount of active agent and one or more surface stabilizers can vary widely. The optimal amount ofthe one or more surface stabilizers can depend, for example, upon the particular active agent selected, the hydrophilic lipophilic balance (HLB), melting point, and water solubility ofthe surface stabilizer, and the surface tension of water solutions ofthe surface stabilizer, etc.
The concentration ofthe at least one active agent can vary from about 99.5% to about 0.001%, from about 95% to about 0.1%, or from about 90% to about 0.5%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
The concentration ofthe at least one surface stabilizer can vary from about 0.001 to about 99.5%, from about 0.1% to about 95%, and from about 0.5% to about 90%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
2. Methods of Making Nanoparticulate Formulations
Nanoparticulate active agent compositions can be made using methods known in the art such as, for example, milling, homogenization, and precipitation techniques. Exemplary methods of making nanoparticulate compositions are described in U.S. Patent No. 5,145,684. Methods of making nanoparticulate compositions are also described in U.S. Patent Nos. 5,518,187; 5,718,388; 5,862,999; 5,665,331; 5,662,883; 5,560,932; 5,543,133; 5,534,270; 5,510,118; and 5,470,583, all referenced in the background ofthe invention.
1. Milling to Obtain Nanoparticulate Active Agent Dispersions
Milling an active agent to obtain a nanoparticulate composition comprises dispersing active agent particles in a liquid dispersion medium in which the active agent is poorly soluble, followed by applying mechanical means in the presence of grinding media to reduce the particle size ofthe active agent particles to the desired effective average particle size. The dispersion medium can be, for example, water, safflower oil, ethanol, t-butanol, glycerin, polyethylene glycol (PEG), hexane, or glycol.
The active agent particles can be reduced in size in the presence of at least one surface stabilizer. Alternatively, the active agent particles can be contacted with one or more surface stabilizers after attrition. Other compounds, such as a diluent, can be added to the active agent/surface stabilizer composition during the size reduction process. Dispersions can be manufactured continuously or in a batch mode. The resultant nanoparticulate active agent dispersion can be utilized in solid or liquid dosage formulations, such as controlled release formulations, solid dose fast melt formulations, aerosol formulations, lyophilized formulations, tablets, capsules, solid lozenge, powders, etc.
2. Precipitation to Obtain Nanoparticulate Active Agent Compositions
Another method of forming the desired nanoparticulate active agent composition is by microprecipitation. This is a method of preparing stable dispersions of poorly soluble active agents in the presence of one or more surface stabilizers and one or more colloid stability enhancing surface active agents free of any trace toxic solvents or solubilized heavy metal impurities. Such a method comprises, for example: (1) dissolving an active agent in a suitable solvent; (2) adding the formulation from step (1) to a solution comprising at least one surface stabilizer; and (3) precipitating the formulation from step (2) using an appropriate non-solvent. The method can be followed by removal of any formed salt, if present, by dialysis or diafiltration and concentration ofthe dispersion by conventional means. The resultant nanoparticulate active agent dispersion can be utilized in solid or liquid dosage formulations, such as controlled release formulations, solid dose fast melt formulations, aerosol formulations, lyophilized formulations, tablets, solid lozenge, powders, capsules, etc.
3. Homogenization to Obtain Active Agent Nanoparticulate Compositions
Exemplary homogenization methods of preparing active agent nanoparticulate compositions are described in U.S. Patent No. 5,510,118, for "Process of Preparing Therapeutic Compositions Containing Nanoparticles."
C. Methods of Using Nanoparticulate Active Agent Formulations
The nanoparticulate compositions ofthe present invention can be administered to humans and animals either orally, rectally, parenterally (intravenous, intramuscular, or subcutaneous), mtracisternally, intravaginally, intraperitoneally, locally (powders, ointments or drops), or as a buccal or nasal spray.
Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance ofthe required particle size in the case of dispersions, and by the use of surfactants. The nanoparticulate compositions may also contain adjuvants, such as preserving, wetting, emulsifying, and dispensing agents. Prevention ofthe growth of microorganisms can be ensured by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like. Prolonged absorption ofthe injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one ofthe following: (a) one or more inert excipients (or carrier), such as sodium citrate or dicalcium phosphate; (b) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; (c) binders, such as carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants, such as glycerol; (e) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (f) solution retarders, such as paraffin; (g) absorption accelerators, such as quaternary ammonium compounds; (h) wetting agents, such as cetyl alcohol and glycerol monostearate; (i) adsorbents, such as kaolin and bentonite; and (j) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. For capsules, tablets, and pills, the dosage forms may also comprise buffering agents.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage forms may comprise inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers. Exemplary emulsifiers are ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, such as cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil, glycerol, tetrahydro±urfuryl alcohol, polyethyleneglycols, fatty acid esters of sorbitan, or mixtures of these substances, and the like.
Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Actual dosage levels of active agent in the nanoparticulate compositions ofthe invention may be varied to obtain an amount of active agent that is effective to obtain a desired therapeutic response for a particular composition and method of administration. The selected dosage level therefore depends upon the desired therapeutic effect, on the route of administration, on the desired duration of treatment, and other factors. The total daily dose ofthe compounds of tins invention administered to a host in single or divided dose may be in amounts of, for example, from about 1 nanomole to about 5 micromoles per kilogram of body weight. Dosage unit compositions may contain such amounts of such submultiples thereof as may be used to make up the daily dose. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the body weight, general health, sex, diet, time and route of administration, rates of absorption and excretion, combination with other active agents, and the severity ofthe particular disease being treated.
The following examples are given to illustrate the present invention. It should be understood, however, that the invention is not to be limited to the specific conditions or details described in these examples. Throughout the specification, any an all references to publicly available documents are specifically incorporated by reference.
Example 1
The purpose of this example was to determine the effect on the stability of paclitaxel at a basic pH when the drug is formulated into a nanoparticulate composition.
Paclitaxel is a naturally occurring diterpenoid which has demonstrated great potential as an anti-cancer drug. Paclitaxel can be isolated from the bark ofthe western yew, Taxus brevifolia, and is also found in several other yew species such as T baccata and T. cuspidata. Upon exposure to a basic pH (i.e., a pH of about 9), the drug rapidly degrades. Ringel et al., J Pharmac. Exp. Ther., 242:692-69% (1987).
Two formulations of paclitaxel were prepared: a solubilized formulation of paclitaxel and a nanoparticulate formulation of paclitaxel. The degradation of paclitaxel for both formulations was then compared. For Formulation I, paclitaxel (Biolyse; Quebec, Canada) was solubilized in 1% methanol and 99% H2O to make a 2% paclitaxel solution. Formulation II was prepared by milling the 2% paclitaxel solution with 1% Plurionic F108™ (BASF) in a 0.5 oz amber bottle containing 7.5 ml 0.5 mm Yttria-doped Zirconia media on a U.S. Stoneware Roller Mill for 72 hours. The resultant milled paclitaxel composition had an effective average particle size of about 220 nm, as measured by a Coulter Counter (Coulter Electronics Inc.). Both solubilized paclitaxel (Formulation I) and nanoparticulate paclitaxel (Formulation II) were incubated with 0.005 N NaOH solution (a basic solution). At the end ofthe incubation period, base degradation of paclitaxel was stopped by adding to the incubation solution 1/100 its volume of IN HCl. The recovery of paclitaxel was then measured at various time periods by HPLC.
As shown in Figure 1, solubilized paclitaxel rapidly degraded when exposed to basic pH conditions, as only about 20% ofthe paclitaxel was recoverable after a 20 minute incubation period. In contrast, nanoparticulate paclitaxel was essentially stable under basic conditions, as more than 90% ofthe drug was recoverable after the same incubation period.
Example 2
The purpose of this example was to determine the stability of rapamycin when the drug is formulated into a nanoparticulate composition.
Rapamycin is useful as an immunosuppressant and as an antifungal antibiotic, and its use is described in, for example, U.S. Patent Nos. 3,929,992, 3,993,749, and 4,316,885, and in Belgian Pat. No. 877,700. The compound, which is only slightly soluble in water, i.e., 20 micrograms per mL, rapidly hydrolyzes when exposed to water. Because rapamycin is highly unstable when exposed to an aqueous medium, special injectable formulations have been developed for administration to patients, such as those described in European Patent No. EP 041,795. Such formulations are often undesirable, as frequently the non-aqueous solubilizing agent exhibits toxic side effects.
Two different formulations of rapamycin were prepared and then exposed to different environmental conditions. The degradation of rapamycin for each ofthe formulations was then compared. The two formulations were prepared as follows:
(1) Formulation I, a mixture of 5% rapamycin and 2.5% Plurionic F68™ (BASF) in an aqueous medium; and
(2) Formulation II, a mixture of 5% rapamycin and 1.25% Plurionic FI 08™ (BASF) in an aqueous medium. Each ofthe two formulations was milled for 72 hours in a 0.5 ounce bottle containing 0.4 mm Yttria beads (Performance Ceramics Media) on a U.S. Stoneware Mill. Particle sizes ofthe resultant nanoparticulate compositions were measured by a Coulter Counter (Model No. N4MD). Following milling, Formulations I and II had effective average particle sizes of 162 nm and 171 nm, respectively.
The samples were then diluted to about 2% rapamycin with Water For Injection (WFI), bottled, and then either stored at room temperature or frozen upon completion of milling and then thawed and stored at room temperature. After ten days of storage at room temperature, Formulations I and II had effective average particle sizes of 194 nm and 199 nm, respectively.
The strength ofthe rapamycin in the formulations was measured by HPLC, the results of which are shown below in Table I.
TABLE I
Stability of Nanoparticulate Rapamycin under Different Storage Conditions
Sample Description Storage Storage Ending Strength SECO %* Conditions Time Starting Strength
Formulation I RT 2 days 97% <detection limit
Formulation II RT 2 days 99% <detection limit
Formulation III RT 2 days 96% <detection limit
Formulation I Frozen/thawed 2 days 95% <detection limit
Formulation II Frozen thawed 2 days 98% <detection limit
Formulation III Frozen thawed 2 days 97% <detection limit
Formulation I RT 3 wks 95% <detection limit
Formulation II RT 3 wks 98% <detection limit
Formulation III RT 3 wks 98% <detection limit
*SECO, or secoacid, is the primary degradation product of rapamycin. The detection limit is 0.2%.
The results show that the nanoparticulate rapamycin formulation exhibited minimal degradation of rapamycin following prolonged storage periods or exposure to the environmental conditions of freezing and thawing.
Example 3
The purpose of this example was to determine the effect of rapamycin concentration on the chemical stability of rapamycin in a nanoparticulate formulation following autoclaving.
Three rapamycin formulations were prepared by milling the following three slurries in a 250 ml Pyrex™ bottle containing 125 ml 0.4 mm Yttria-doped Zirconia media for 72 hours on a U.S. Stoneware roller mill:
(a) 5% raρamycin/1.25% Plurionic F68™
(b) 5% raρamycin 2.5% Plurionic F68™
(c) 5 % rapamycin/5% Plurionic F68™
Each ofthe three dispersions was then diluted with water to prepare formulations having rapamycin concentrations of 4.4%, 2.2%, 1.1% and 0.5% as follows: (1) Formulation 1 : a mixture of 4.4% rapamycin and, prior to dilution, 1.25% Plurionic F68™ in an aqueous medium; (2) Formulation 2: a mixture of 4.4% rapamycin and, prior to dilution, 2.5% Plurionic F68™ in an aqueous medium;
(3) Formulation 3: a mixture of 4.4% rapamycin and, prior to dilution, 5% Plurionic F68™ in an aqueous medium;
(4) Fonnulation 4: a mixture of 2.2% rapamycin and, prior to dilution, 1.25% Plurionic F68™ in an aqueous medium;
(5) Formulation 5: a mixture of 2.2% rapamycin and, prior to dilution, 2.5% Plurionic F68™ in an aqueous medium;
(6) Fonnulation 6: a mixture of 2.2% rapamycin and, prior to dilution, 5% Plurionic F68™ in an aqueous medium;
(7) Formulation 7: a mixture of 1.1% rapamycin and, prior to dilution, 1.25% Plurionic F68™ in an aqueous medium;
(8) Formulation 8: a mixture of 1.1% rapamycin and, prior to dilution, 2.5% Plurionic F68™ in an aqueous medium;
(9) Formulation 9: a mixture of 1.1 % rapamycin and, prior to dilution, 5% Plurionic F68™ in an aqueous medium;
(10) Formulation 10: amixture of 0.55% rapamycin and, prior to dilution, 1.25% Plurionic F68™ in an aqueous medium;
(11) Formulation 11 : a mixture of 0.55% rapamycin and, prior to dilution, 2.5% Plurionic F68™ in an aqueous medium; and
(12) Formulation 12: a mixture of 0.55% rapamycin and, prior to dilution, 5% Plurionic F68™ in an aqueous medium;
All twelve ofthe nanoparticulate formulations were autoclaved for 25 minutes at 12 C. The formulations were then stored at 4°C for 61 days, followed by testing for rapamycin degradation. No degradation, as measured by the percent ofthe SECO degradation product, was detected for any ofthe formulations. Example 4
The purpose of this example was to determine the chemical stability of a nanoparticulate rapamycin formulation following a prolonged storage period at room temperature.
A mixture of 20% rapamycin and 10% Plurionic F68™ in an aqueous medium was milled with 0.4 mm YTZ media (Performance Ceramic Co.) on a U.S. Stoneware mill for 72 hours at room temperature. The final nanoparticulate composition had a mean particle size of between 180 to 230 nm, as measured by Coulter sizing.
After two weeks of storage at room temperature, no SECO degradation product was detected in any ofthe nanoparticulate preparations, indicating that there was minimal or no degradation of rapamycin in the stored nanoparticulate formulation samples.
Example 5
The purpose of this example was to detennine the effect of long term storage on the chemical stability of rapamycin in a nanoparticulate composition.
Three different nanoparticulate rapamycin fonnulations were prepared as follows: Formulation 1, having a rapamycin concentration of 182.8 mg/mL; Formulation 2, having a rapamycin concentration of 191.4 mg/mL; and Formulation 3, having a rapamycin concentration of 192.7 mg/mL.
The formulations were prepared by milling the following three slurries in a 0.5 oz amber bottle containing 7.5 ml 0.8 mm Yttria-doped Zirconia media for 72 hours on a U.S. Stoneware roller mill:
(1) 20% rapamycin/10% Plurionic F68
(2) 20% rapamycin/5%) Plurionic F68
(3) 20% rapamycin/2.5% Plurionic F68
Following storage for two and half months, no SECO degradation product was detected in any ofthe samples. These results show that various dosage strengths of rapamycin can be used in nanoparticulate formulations without any impact on the increased chemical stability ofthe drug.
* * * * It will be apparent to those skilled in the art that various modifications and variations can be made in the methods and compositions ofthe present invention without departing from the spirit or scope ofthe invention. Thus, it is intended that the present invention cover the modifications and variations of this invention, provided they come within the scope ofthe appended claims and their equivalents.

Claims

Claims:
1. A method for chemically stabilizing an active agent comprising:
(a) identifying and selecting at least one active agent which is unstable under one or more environmental conditions, wherein the active agent is poorly soluble in at least one liquid medium; and
(b) formulating particles ofthe active agent into a stable nanoparticulate composition by a method comprising:
(1) combining the active agent particles which are unstable under one or more environmental conditions; and
(2) at least one non-crosslinked surface stabilizer, wherein after said combining the at least one non-crosslinked surface stabilizer adsorbs to the surface ofthe active agent particles to produce a composition in which the active agent particles have an effective average particle size of less than about 2 microns; wherein the resultant nanoparticulate active agent composition is stable under the one or more environmental conditions under which the active agent is unstable prior to formulating the active agent into a nanoparticulate composition.
2. The method of claim 1, wherein the at least one active agent is present in an amount selected from the group consisting of from about 99.5% to about 0.001%, from about 95% to about 0.1%, and from about 90% to about 0.5%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
3. The method of claim 1 or 2, wherein the at least one surface stabilizer is present in an amount selected from the group consisting of from about 0.001 to about 99.5%, from about 0.1% to about 95%, and from about 0.5% to about 90%, by weight, based on the total combined weight ofthe at least one active agent and at least one surface stabilizer, not including other excipients.
4. The method of any of the previous claims, wherem after step (b) the active agent has an effective average particle size of less than about 1 micron.
5. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 600 nm.
6. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 500 nm.
7. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 400 nm.
8. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 300 nm.
9. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 200 nm.
10. The method of any ofthe previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 100 nm.
11. The method of any of the previous claims, wherein after step (b) the active agent has an effective average particle size of less than about 50 nm.
12. The method of any ofthe previous claims, wherein the resultant composition is an injectable formulation.
13. The method of any ofthe previous claims, wherein the one or more environmental conditions are selected from the group consisting of exposure to a prolonged storage period, exposure to an elevated temperature, exposure to light, exposure to radiation, exposure to radiation causing photolysis, exposure to non-physiological pH, exposure to enzymes or other catalysts, exposure to water or other solvent molecules, exposure to oxidizing agents or other free radicals, and exposure to freezing-thawing temperature cycles.
14. The method of any ofthe previous claims, wherein the active agent is rapamycin.
15. The method of claim 14, wherem said rapamycin is stable following exposure to hydrolysis conditions.
16. The method of any of claims 1-13, wherein the active agent is paclitaxel.
17. The method of claim 16, wherein said paclitaxel is stable following exposure to basic pH conditions.
18. The method of any of the previous claims, wherein the active agent particles are in a crystalline phase, amorphous phase, semi-crystalline phase, semi-amorphous phase, or a combination thereof.
19. The method of any ofthe previous claims, wherein the surface stabilizer is selected from the group consisting of anionic, ionic, and cationic compounds.
PCT/US2002/025979 2001-09-14 2002-09-13 Stabilization of active agents by formulation into nanoparticulate form WO2003024424A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2003528521A JP2005505568A (en) 2001-09-14 2002-09-13 Stabilization of active agents by formulation into nanoparticulate form
DE60225571T DE60225571T2 (en) 2001-09-14 2002-09-13 STABILIZATION OF ACTIVE AGENTS BY FORMULATION AS NANOPARTICULAR COMPOSITIONS
CA2460436A CA2460436C (en) 2001-09-14 2002-09-13 Stabilization of active agents by formulation into nanoparticulate form
EP02775709A EP1427395B1 (en) 2001-09-14 2002-09-13 Stabilization of active agents by formulation into nanoparticulate form
DK02775709T DK1427395T3 (en) 2001-09-14 2002-09-13 Stabilization of active ingredients by formulation in nanoparticulate form

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/952,032 US20030054042A1 (en) 2001-09-14 2001-09-14 Stabilization of chemical compounds using nanoparticulate formulations
US09/952,032 2001-09-14

Publications (1)

Publication Number Publication Date
WO2003024424A1 true WO2003024424A1 (en) 2003-03-27

Family

ID=25492513

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/025979 WO2003024424A1 (en) 2001-09-14 2002-09-13 Stabilization of active agents by formulation into nanoparticulate form

Country Status (11)

Country Link
US (4) US20030054042A1 (en)
EP (2) EP1829530A3 (en)
JP (2) JP2005505568A (en)
AT (1) ATE388690T1 (en)
CA (1) CA2460436C (en)
CY (1) CY1108130T1 (en)
DE (1) DE60225571T2 (en)
DK (1) DK1427395T3 (en)
ES (1) ES2303553T3 (en)
PT (1) PT1427395E (en)
WO (1) WO2003024424A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034916A1 (en) * 2003-10-03 2005-04-21 Novartis Ag Pharmaceutical multiparticulate composition comprising mycophenolic acid or mycophenolate sodium and combination with rapamycin
JP2006089386A (en) * 2004-09-21 2006-04-06 Nippon Tenganyaku Kenkyusho:Kk Suspension medicine composition containing steroid or steroid derivative
JP2006528176A (en) * 2003-07-23 2006-12-14 エラン ファーマ インターナショナル リミテッド Novel sildenafil free base composition
JP2008013562A (en) * 2006-06-30 2008-01-24 Elan Corp Plc Nanoparticle-like controlled release composition comprising cephalosporin
JP2008505982A (en) * 2004-05-18 2008-02-28 ブリストル−マイヤーズ スクイブ カンパニー Lactam compound-containing nanoparticle dispersion
JP2008536856A (en) * 2005-04-13 2008-09-11 エラン・ファルマ・インターナショナル・リミテッド Nanoparticulate and sustained release composition comprising a prostaglandin derivative
JP2008540691A (en) * 2005-05-16 2008-11-20 エラン・ファルマ・インターナショナル・リミテッド Nanoparticles and controlled release compositions comprising cephalosporin
EP1435909B1 (en) * 2001-10-19 2009-12-02 Baxter International Inc. Stable composition comprising particles in a frozen aqueous matrix
EP1810665B1 (en) 2005-12-29 2015-03-04 Cordis Corporation Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
EP3104844B1 (en) 2014-02-14 2020-02-12 Druggability Technologies IP Holdco Limited Complexes of sirolimus and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
EP3928772A1 (en) 2020-06-26 2021-12-29 Algiax Pharmaceuticals GmbH Nanoparticulate composition

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
US8853260B2 (en) * 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
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
US7758890B2 (en) 2001-06-23 2010-07-20 Lyotropic Therapeutics, Inc. Treatment using dantrolene
BR0212833A (en) * 2001-09-26 2004-10-13 Baxter Int Preparation of submicron sized nanoparticles by dispersion and solvent or liquid phase removal
US20080220075A1 (en) * 2002-03-20 2008-09-11 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
CA2479665C (en) * 2002-03-20 2011-08-30 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
WO2005065657A2 (en) * 2003-12-31 2005-07-21 Pfizer Products Inc. Solid compositions of low-solubility drugs and poloxamers
WO2005120487A2 (en) * 2004-06-11 2005-12-22 Egon Tech Preparation for the prevention and treatment of stress conditions as well as functional and organic disorders of the nervous system and metabolic disorders
US20060147515A1 (en) * 2004-12-02 2006-07-06 Zhongzhou Liu Bioactive dispersible formulation
US7727554B2 (en) * 2004-12-21 2010-06-01 Board Of Regents Of The University Of Nebraska By And Behalf Of The University Of Nebraska Medical Center Sustained-release nanoparticle compositions and methods for using the same
AU2006259594A1 (en) * 2005-06-14 2006-12-28 Baxter Healthcare S.A. Pharmaceutical formulations for minimizing drug-drug interactions
WO2007033239A2 (en) * 2005-09-13 2007-03-22 Elan Pharma International, Limited Nanoparticulate tadalafil formulations
EP1954245A2 (en) * 2005-11-15 2008-08-13 Baxter International Inc. Compositions of lipoxygenase inhibitors
US8022054B2 (en) 2005-11-28 2011-09-20 Marinus Pharmaceuticals Liquid ganaxolone formulations and methods for the making and use thereof
US8367112B2 (en) * 2006-02-28 2013-02-05 Alkermes Pharma Ireland Limited Nanoparticulate carverdilol formulations
US8173152B2 (en) 2006-03-24 2012-05-08 Auxilium Us Holdings, Llc Stabilized compositions containing alkaline labile drugs
US20080050450A1 (en) * 2006-06-26 2008-02-28 Mutual Pharmaceutical Company, Inc. Active Agent Formulations, Methods of Making, and Methods of Use
EP2101735A2 (en) * 2006-11-28 2009-09-23 Marinus Pharmaceuticals, Inc. Nanoparticulate formulations and methods for the making and use thereof
EP1938800A1 (en) * 2006-12-06 2008-07-02 Ranbaxy Laboratories Limited Sirolimus nanodispersion
US20090152176A1 (en) * 2006-12-23 2009-06-18 Baxter International Inc. Magnetic separation of fine particles from compositions
KR20150002886A (en) * 2007-03-07 2015-01-07 아브락시스 바이오사이언스, 엘엘씨 Nanoparticle comprising rapamycin and albumin as anticancer agent
US20100166869A1 (en) * 2007-05-03 2010-07-01 Desai Neil P Methods and compositions for treating pulmonary hypertension
US8722736B2 (en) * 2007-05-22 2014-05-13 Baxter International Inc. Multi-dose concentrate esmolol with benzyl alcohol
US20080293814A1 (en) * 2007-05-22 2008-11-27 Deepak Tiwari Concentrate esmolol
US8426467B2 (en) * 2007-05-22 2013-04-23 Baxter International Inc. Colored esmolol concentrate
PL2155188T3 (en) * 2007-06-01 2014-03-31 Abraxis Bioscience Llc Methods and compositions for treating recurrent cancer
CA2707010A1 (en) * 2007-12-14 2009-06-25 Kazuhisa Sugimoto .alpha.-lipoic acid nanoparticles and methods for preparing thereof
EP2249808A2 (en) * 2008-01-11 2010-11-17 Cipla Limited Solid pharmaceutical dosage form
WO2009113070A1 (en) * 2008-03-12 2009-09-17 Do-Coop Technologies Ltd. Freeze-free method for storage of polypeptides
US8183233B2 (en) * 2008-05-15 2012-05-22 Baxter International Inc. Stable pharmaceutical formulations
US20100098770A1 (en) * 2008-10-16 2010-04-22 Manikandan Ramalingam Sirolimus pharmaceutical formulations
US20100159010A1 (en) * 2008-12-24 2010-06-24 Mutual Pharmaceutical Company, Inc. Active Agent Formulations, Methods of Making, and Methods of Use
KR20120022984A (en) * 2009-04-21 2012-03-12 셀렉타 바이오사이언시즈, 인크. Immunonanotherapeutics providing a th1-biased response
JP6072539B2 (en) 2009-05-27 2017-02-01 アルカーメス ファーマ アイルランド リミテッド Reduction of flaky aggregation in nanoparticulate active agent compositions
WO2011135580A2 (en) * 2010-04-28 2011-11-03 Cadila Healthcare Limited Pharmaceutical compositions of sirolimus
CN101829061A (en) * 2010-05-14 2010-09-15 无锡纳生生物科技有限公司 Taxol nanoparticle composition and preparation method thereof
MX2012013713A (en) 2010-05-26 2013-01-28 Selecta Biosciences Inc Nanocarrier compositions with uncoupled adjuvant.
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US8912215B2 (en) * 2011-12-13 2014-12-16 Everon Biosciences, Inc. Rapamycin composition
US9095521B2 (en) 2012-02-02 2015-08-04 Washington University Methods for improving muscle strength
WO2016007194A1 (en) * 2014-07-10 2016-01-14 Gerald Lee Wolf Companion nanoparticles for theranosis of macrophage-dependent diseases
US20160346221A1 (en) 2015-06-01 2016-12-01 Autotelic Llc Phospholipid-coated therapeutic agent nanoparticles and related methods
CA3026452C (en) 2015-06-04 2023-03-21 Crititech, Inc. Nozzle assembly and methods for use
EA036155B1 (en) 2015-10-16 2020-10-06 Маринус Фармасьютикалс, Инк. Injectable neurosteroid formulations containing nanoparticles
US10792477B2 (en) 2016-02-08 2020-10-06 Orbusneich Medical Pte. Ltd. Drug eluting balloon
WO2017176628A1 (en) 2016-04-04 2017-10-12 Crititech, Inc. Methods for solid tumor treatment
BR112019002538A2 (en) 2016-08-11 2019-05-21 Ovid Therapeutics Inc. use of a pharmaceutical composition comprising an allosteric modulator, use of a pharmaceutical composition comprising garboxadol or a pharmaceutically acceptable salt thereof, and pharmaceutical composition for parenteral administration
US10391105B2 (en) 2016-09-09 2019-08-27 Marinus Pharmaceuticals Inc. Methods of treating certain depressive disorders and delirium tremens
JP2020523285A (en) 2017-06-09 2020-08-06 クリチテック,インコーポレイテッド Treatment of epithelial cysts by intracystic injection of antitumor particles
KR102303762B1 (en) 2017-06-14 2021-09-23 크리티테크, 인크. Methods of treating lung disorders
CN111278436A (en) 2017-10-03 2020-06-12 克里蒂泰克公司 Local delivery of anti-tumor particles in combination with systemic delivery of immunotherapeutic agents for the treatment of cancer
US11266662B2 (en) 2018-12-07 2022-03-08 Marinus Pharmaceuticals, Inc. Ganaxolone for use in prophylaxis and treatment of postpartum depression
CN109431997B (en) * 2018-12-20 2021-06-04 武汉科福新药有限责任公司 Local rapamycin injection and preparation method thereof
CA3145923A1 (en) 2019-08-05 2021-02-11 David Czekai Ganaxolone for use in treatment of status epilepticus
MX2022006014A (en) 2019-12-06 2022-06-22 Marinus Pharmaceuticals Inc Ganaxolone for use in treating tuberous sclerosis complex.

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399363A (en) * 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
WO1999049846A2 (en) * 1998-03-30 1999-10-07 Rtp Pharma Inc. Compositions containing microparticles of water-insoluble substances and method for their preparation
US5989591A (en) * 1997-03-14 1999-11-23 American Home Products Corporation Rapamycin formulations for oral administration
WO2001026635A2 (en) * 1999-10-08 2001-04-19 Elan Pharma International Ltd. Bioadhesive nanoparticulate compositions having cationic surface stabilizers
US6267989B1 (en) * 1999-03-08 2001-07-31 Klan Pharma International Ltd. Methods for preventing crystal growth and particle aggregation in nanoparticulate compositions

Family Cites Families (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2671750A (en) * 1950-09-19 1954-03-09 Merck & Co Inc Stable noncaking aqueous suspension of cortisone acetate and method of preparing the same
US3536074A (en) * 1968-03-29 1970-10-27 Alfred Aufhauser Oral administration of a pill,tablet or capsule
US3959457A (en) * 1970-06-05 1976-05-25 Temple University Microparticulate material and method of making such material
JPS4932056B1 (en) * 1970-12-22 1974-08-27
US4073943A (en) * 1974-09-11 1978-02-14 Apoteksvarucentralen Vitrum Ab Method of enhancing the administration of pharmalogically active agents
US4107288A (en) * 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
US4001200A (en) * 1975-02-27 1977-01-04 Alza Corporation Novel polymerized, cross-linked, stromal-free hemoglobin
US4001401A (en) * 1975-02-02 1977-01-04 Alza Corporation Blood substitute and blood plasma expander comprising polyhemoglobin
US4053590A (en) * 1975-02-27 1977-10-11 Alza Corporation Compositions of matter comprising macromolecular hemoglobin
DK143689C (en) * 1975-03-20 1982-03-15 J Kreuter PROCEDURE FOR THE PREPARATION OF AN ADVERTISED VACCINE
US4226248A (en) * 1978-10-26 1980-10-07 Manoli Samir H Phonocephalographic device
US4344934A (en) * 1978-11-20 1982-08-17 American Home Products Corporation Therapeutic compositions with enhanced bioavailability
DE3013839A1 (en) * 1979-04-13 1980-10-30 Freunt Ind Co Ltd METHOD FOR PRODUCING AN ACTIVATED PHARMACEUTICAL COMPOSITION
US4247406A (en) * 1979-04-23 1981-01-27 Widder Kenneth J Intravascularly-administrable, magnetically-localizable biodegradable carrier
US4316885A (en) * 1980-08-25 1982-02-23 Ayerst, Mckenna And Harrison, Inc. Acyl derivatives of rapamycin
US4534899A (en) * 1981-07-20 1985-08-13 Lipid Specialties, Inc. Synthetic phospholipid compounds
US4572203A (en) * 1983-01-27 1986-02-25 Feinstein Steven B Contact agents for ultrasonic imaging
US4718433A (en) * 1983-01-27 1988-01-12 Feinstein Steven B Contrast agents for ultrasonic imaging
US4725442A (en) * 1983-06-17 1988-02-16 Haynes Duncan H Microdroplets of water-insoluble drugs and injectable formulations containing same
US4671954A (en) * 1983-12-13 1987-06-09 University Of Florida Microspheres for incorporation of therapeutic substances and methods of preparation thereof
US4598064A (en) * 1984-06-27 1986-07-01 University Of Iowa Research Foundation Alpha-alpha cross-linked hemoglobins
US4600531A (en) * 1984-06-27 1986-07-15 University Of Iowa Research Foundation Production of alpha-alpha cross-linked hemoglobins in high yield
US4639364A (en) * 1984-11-14 1987-01-27 Mallinckrodt, Inc. Methods and compositions for enhancing magnetic resonance imaging
US4584130A (en) * 1985-03-29 1986-04-22 University Of Maryland Intramolecularly cross-linked hemoglobin and method of preparation
FR2608942B1 (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 NANOCAPSULES
US5006650A (en) * 1987-02-11 1991-04-09 The Upjohn Company Novel N-1 substituted beta-lactams as antibiotics
US5723147A (en) * 1987-02-23 1998-03-03 Depotech Corporation Multivesicular liposomes having a biologically active substance encapsulated therein in the presence of a hydrochloride
KR890700586A (en) * 1987-02-27 1989-04-25 로버어트 에이 아마테이지 Antibacterial beta-lactam containing pyridone carboxylic acid or acid derivative
US5015737A (en) * 1987-07-22 1991-05-14 The Upjohn Company Therapeutically useful beta-lactams
US4844882A (en) * 1987-12-29 1989-07-04 Molecular Biosystems, Inc. Concentrated stabilized microbubble-type ultrasonic imaging agent
US4929446A (en) * 1988-04-19 1990-05-29 American Cyanamid Company Unit dosage form
US4951673A (en) * 1988-08-19 1990-08-28 Alliance Pharmaceutical Corp. Magnetic resonance imaging with perfluorocarbon hydrides
US5041292A (en) * 1988-08-31 1991-08-20 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
US5114703A (en) * 1989-05-30 1992-05-19 Alliance Pharmaceutical Corp. Percutaneous lymphography using particulate fluorocarbon emulsions
GB8914060D0 (en) * 1989-06-19 1989-08-09 Wellcome Found Agents for potentiating the effects of antitumour agents and combating multiple drug resistance
US5116599A (en) * 1989-07-31 1992-05-26 Johns Hopkins Univ. Perfluoro-t-butyl-containing compounds for use in fluorine-19 nmr and/or mri
FR2651680B1 (en) * 1989-09-14 1991-12-27 Medgenix Group Sa NOVEL PROCESS FOR THE PREPARATION OF LIPID MICROPARTICLES.
JP2687245B2 (en) * 1989-09-29 1997-12-08 富士写真フイルム株式会社 Manufacturing method of magnetic recording medium
US5250283A (en) * 1990-03-28 1993-10-05 Molecular Biosystems, Inc. Organic contrast agent analog and method of making same
US5091188A (en) * 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
CA2019719A1 (en) * 1990-06-25 1991-12-25 William J. Thompson Mouthwash
US5059699A (en) * 1990-08-28 1991-10-22 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol
US5110606A (en) * 1990-11-13 1992-05-05 Affinity Biotech, Inc. Non-aqueous microemulsions for drug delivery
AU642066B2 (en) * 1991-01-25 1993-10-07 Nanosystems L.L.C. X-ray contrast compositions useful in medical imaging
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5143716A (en) * 1991-02-01 1992-09-01 Unger Evan C Phosphorylated sugar alcohols, Mono- and Di-Saccharides as contrast agents for use in magnetic resonance imaging of the gastrointestinal region
US5416071A (en) * 1991-03-12 1995-05-16 Takeda Chemical Industries, Ltd. Water-soluble composition for sustained-release containing epo and hyaluronic acid
US5434143A (en) * 1991-05-10 1995-07-18 Boron Biologicals, Inc. Pharmaceutical compositions comprising phosphite-borane compounds
GB9111580D0 (en) * 1991-05-30 1991-07-24 Wellcome Found Nucleoside derivative
US5442062A (en) * 1991-10-24 1995-08-15 The Upjohn Company Imidazole derivatives and pharmaceutical compositions containing the same
US5292650A (en) * 1991-10-29 1994-03-08 Eli Lilly And Company Preparation of hapalindole-related alkaloids from blue-green algae
AU3922193A (en) * 1992-03-23 1993-10-21 Georgetown University Liposome encapsulated taxol and a method of using the same
CA2086874E (en) * 1992-08-03 2000-01-04 Renzo Mauro Canetta Methods for administration of taxol
AU660852B2 (en) * 1992-11-25 1995-07-06 Elan Pharma International Limited Method of grinding pharmaceutical substances
US5298262A (en) * 1992-12-04 1994-03-29 Sterling Winthrop Inc. Use of ionic cloud point modifiers to prevent particle aggregation during sterilization
US5302401A (en) * 1992-12-09 1994-04-12 Sterling Winthrop Inc. Method to reduce particle size growth during lyophilization
US5429824A (en) * 1992-12-15 1995-07-04 Eastman Kodak Company Use of tyloxapole as a nanoparticle stabilizer and dispersant
US5326552A (en) * 1992-12-17 1994-07-05 Sterling Winthrop Inc. Formulations for nanoparticulate x-ray blood pool contrast agents using high molecular weight nonionic surfactants
US5401492A (en) * 1992-12-17 1995-03-28 Sterling Winthrop, Inc. Water insoluble non-magnetic manganese particles as magnetic resonance contract enhancement agents
US6537579B1 (en) * 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
US5916596A (en) * 1993-02-22 1999-06-29 Vivorx Pharmaceuticals, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6753006B1 (en) * 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
US5665382A (en) * 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of pharmaceutically active agents for in vivo delivery
DE69433723T3 (en) * 1993-02-22 2008-10-30 Abraxis Bioscience, Inc., Los Angeles PROCESS FOR IN VIVO ADMINISTRATION OF BIOLOGICAL SUBSTANCES AND COMPOSITIONS USED THEREFROM
US6096331A (en) * 1993-02-22 2000-08-01 Vivorx Pharmaceuticals, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US6749868B1 (en) * 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US5362478A (en) * 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
US5395619A (en) * 1993-03-03 1995-03-07 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
US5264610A (en) * 1993-03-29 1993-11-23 Sterling Winthrop Inc. Iodinated aromatic propanedioates
TW406020B (en) * 1993-09-29 2000-09-21 Bristol Myers Squibb Co Stabilized pharmaceutical composition and its method for preparation and stabilizing solvent
US5766627A (en) * 1993-11-16 1998-06-16 Depotech Multivescular liposomes with controlled release of encapsulated biologically active substances
US5731334A (en) * 1994-01-11 1998-03-24 The Scripps Research Institute Method for treating cancer using taxoid onium salt prodrugs
IL128775A (en) * 1994-03-07 2001-05-20 Dow Chemical Co Composition comprising a dendritic polymer in complex with at least one unit of biological response modifier and a process for the preparation thereof
US5565478A (en) * 1994-03-14 1996-10-15 The United States Of America As Represented By The Department Of Health & Human Services Combination therapy using signal transduction inhibitors with paclitaxel and other taxane analogs
GB9405593D0 (en) * 1994-03-22 1994-05-11 Zeneca Ltd Pharmaceutical compositions
US5731355A (en) * 1994-03-22 1998-03-24 Zeneca Limited Pharmaceutical compositions of propofol and edetate
TW384224B (en) * 1994-05-25 2000-03-11 Nano Sys Llc Method of preparing submicron particles of a therapeutic or diagnostic agent
US5718388A (en) * 1994-05-25 1998-02-17 Eastman Kodak Continuous method of grinding pharmaceutical substances
US5543152A (en) * 1994-06-20 1996-08-06 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
US5628981A (en) * 1994-12-30 1997-05-13 Nano Systems L.L.C. Formulations of oral gastrointestinal diagnostic x-ray contrast agents and oral gastrointestinal therapeutic agents
US5518738A (en) * 1995-02-09 1996-05-21 Nanosystem L.L.C. Nanoparticulate nsaid compositions
US5593657A (en) * 1995-02-09 1997-01-14 Nanosystems L.L.C. Barium salt formulations stabilized by non-ionic and anionic stabilizers
US5622938A (en) * 1995-02-09 1997-04-22 Nano Systems L.L.C. Sugar base surfactant for nanocrystals
US5500204A (en) * 1995-02-10 1996-03-19 Eastman Kodak Company Nanoparticulate diagnostic dimers as x-ray contrast agents for blood pool and lymphatic system imaging
US5591456A (en) * 1995-02-10 1997-01-07 Nanosystems L.L.C. Milled naproxen with hydroxypropyl cellulose as a dispersion stabilizer
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles
US5528328A (en) * 1995-02-21 1996-06-18 O'farrill; Dave Camera filter quick release adapter
EP0810853B1 (en) * 1995-02-24 2004-08-25 Elan Pharma International Limited Aerosols containing nanoparticle dispersions
US5718919A (en) * 1995-02-24 1998-02-17 Nanosystems L.L.C. Nanoparticles containing the R(-)enantiomer of ibuprofen
US5747001A (en) * 1995-02-24 1998-05-05 Nanosystems, L.L.C. Aerosols containing beclomethazone nanoparticle dispersions
US5643552A (en) * 1995-03-09 1997-07-01 Nanosystems L.L.C. Nanoparticulate diagnostic mixed carbonic anhydrides as x-ray contrast agents for blood pool and lymphatic system imaging
US5521218A (en) * 1995-05-15 1996-05-28 Nanosystems L.L.C. Nanoparticulate iodipamide derivatives for use as x-ray contrast agents
US5635406A (en) * 1995-06-07 1997-06-03 Abbott Laboratories Stabilized standards and calibrators containing rapamycin and tacrolimus bound to anti-rapamycin and anti-tacrolimus antibodies
DE69624921T2 (en) * 1995-06-09 2003-09-11 Novartis Ag rapamycin derivatives
GB9515214D0 (en) * 1995-07-25 1995-09-20 Univ Strathclyde Plant extracts
US5962019A (en) * 1995-08-25 1999-10-05 Sangstat Medical Corporation Oral cyclosporin formulations
US5834025A (en) * 1995-09-29 1998-11-10 Nanosystems L.L.C. Reduction of intravenously administered nanoparticulate-formulation-induced adverse physiological reactions
US5631741A (en) * 1995-12-29 1997-05-20 Intel Corporation Electronic carbon paper
EP0874809B1 (en) * 1996-01-03 2003-08-27 SmithKline Beecham plc Carbamoyloxy derivatives of mutiline and their use as antibacterials
US5744460A (en) * 1996-03-07 1998-04-28 Novartis Corporation Combination for treatment of proliferative diseases
US5637625A (en) * 1996-03-19 1997-06-10 Research Triangle Pharmaceuticals Ltd. Propofol microdroplet formulations
JP3942641B2 (en) * 1996-07-30 2007-07-11 ノバルティス アクチエンゲゼルシャフト Pharmaceutical composition for the treatment of transplant rejection, autoimmune disease or inflammatory condition comprising cyclosporin A and 40-O- (2-hydroxyethyl) -rapamycin
JP2000516244A (en) * 1996-08-22 2000-12-05 リサーチ・トライアングル・ファーマシューティカルズ Composition containing fine particles of water-insoluble substance and method for producing the same
US6458373B1 (en) * 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US6051563A (en) * 1997-02-12 2000-04-18 U.S. Bioscience, Inc. Methods for the administration of amifostine and related compounds
US6045829A (en) * 1997-02-13 2000-04-04 Elan Pharma International Limited Nanocrystalline formulations of human immunodeficiency virus (HIV) protease inhibitors using cellulosic surface stabilizers
WO1998035666A1 (en) * 1997-02-13 1998-08-20 Nanosystems Llc Formulations of nanoparticle naproxen tablets
SI20424A (en) * 1998-03-05 2001-06-30 Agouron Pharmaceuticals, Inc. Non-peptide GnRH agents
US6228985B1 (en) * 1998-05-21 2001-05-08 Schering Corporation Derivatives of aminobenzoic and aminobiphenylcarboxylic acids useful as anti-cancer agents
US5962536A (en) * 1998-07-31 1999-10-05 Komer; Gene Injectable propofol formulations
US6028108A (en) * 1998-10-22 2000-02-22 America Home Products Corporation Propofol composition comprising pentetate
US6140373A (en) * 1998-10-23 2000-10-31 Abbott Laboratories Propofol composition
US6071952A (en) * 1998-12-02 2000-06-06 Mylan Pharmaceuticals, Inc. Stabilized injectable pharmaceutical compositions containing taxoid anti-neoplastic agents
US6225311B1 (en) * 1999-01-27 2001-05-01 American Cyanamid Company Acetylenic α-amino acid-based sulfonamide hydroxamic acid tace inhibitors
US6200085B1 (en) * 1999-03-08 2001-03-13 Stuart Gee Transport system for farm combines and other large vehicles
US6177477B1 (en) * 1999-03-24 2001-01-23 American Home Products Corporation Propofol formulation containing TRIS
US6100302A (en) * 1999-04-05 2000-08-08 Baxter International Inc. Propofol formulation with enhanced microbial characteristics
US6362234B1 (en) * 2000-08-15 2002-03-26 Vyrex Corporation Water-soluble prodrugs of propofol for treatment of migrane
US6399087B1 (en) * 2000-12-20 2002-06-04 Amphastar Pharmaceuticals, Inc. Propofol formulation with enhanced microbial inhibition
CA2479665C (en) * 2002-03-20 2011-08-30 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
EP2623095A1 (en) * 2004-11-16 2013-08-07 Elan Pharma International Limited Injectable nanoparticulate olanzapine formulations
SE530813C2 (en) * 2007-01-31 2008-09-16 Tolerans Ab Method and apparatus of a rotary stapler

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399363A (en) * 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5494683A (en) * 1991-01-25 1996-02-27 Eastman Kodak Company Surface modified anticancer nanoparticles
US5989591A (en) * 1997-03-14 1999-11-23 American Home Products Corporation Rapamycin formulations for oral administration
WO1999049846A2 (en) * 1998-03-30 1999-10-07 Rtp Pharma Inc. Compositions containing microparticles of water-insoluble substances and method for their preparation
US6267989B1 (en) * 1999-03-08 2001-07-31 Klan Pharma International Ltd. Methods for preventing crystal growth and particle aggregation in nanoparticulate compositions
WO2001026635A2 (en) * 1999-10-08 2001-04-19 Elan Pharma International Ltd. Bioadhesive nanoparticulate compositions having cationic surface stabilizers

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1435909B1 (en) * 2001-10-19 2009-12-02 Baxter International Inc. Stable composition comprising particles in a frozen aqueous matrix
JP4860469B2 (en) * 2003-07-23 2012-01-25 エラン ファーマ インターナショナル,リミティド Novel sildenafil free base composition
JP2006528176A (en) * 2003-07-23 2006-12-14 エラン ファーマ インターナショナル リミテッド Novel sildenafil free base composition
WO2005034916A1 (en) * 2003-10-03 2005-04-21 Novartis Ag Pharmaceutical multiparticulate composition comprising mycophenolic acid or mycophenolate sodium and combination with rapamycin
JP2008505982A (en) * 2004-05-18 2008-02-28 ブリストル−マイヤーズ スクイブ カンパニー Lactam compound-containing nanoparticle dispersion
JP2006089386A (en) * 2004-09-21 2006-04-06 Nippon Tenganyaku Kenkyusho:Kk Suspension medicine composition containing steroid or steroid derivative
JP2008536856A (en) * 2005-04-13 2008-09-11 エラン・ファルマ・インターナショナル・リミテッド Nanoparticulate and sustained release composition comprising a prostaglandin derivative
JP2008540691A (en) * 2005-05-16 2008-11-20 エラン・ファルマ・インターナショナル・リミテッド Nanoparticles and controlled release compositions comprising cephalosporin
EP1810665B1 (en) 2005-12-29 2015-03-04 Cordis Corporation Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
JP2008013562A (en) * 2006-06-30 2008-01-24 Elan Corp Plc Nanoparticle-like controlled release composition comprising cephalosporin
EP3104844B1 (en) 2014-02-14 2020-02-12 Druggability Technologies IP Holdco Limited Complexes of sirolimus and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
EP3928772A1 (en) 2020-06-26 2021-12-29 Algiax Pharmaceuticals GmbH Nanoparticulate composition
WO2021259669A1 (en) 2020-06-26 2021-12-30 Algiax Pharmaceuticals Gmbh Nanoparticulate composition

Also Published As

Publication number Publication date
DE60225571T2 (en) 2009-04-23
JP2005505568A (en) 2005-02-24
CA2460436C (en) 2011-05-10
CA2460436A1 (en) 2003-03-27
EP1829530A2 (en) 2007-09-05
DE60225571D1 (en) 2008-04-24
EP1829530A3 (en) 2009-05-06
CY1108130T1 (en) 2014-02-12
PT1427395E (en) 2008-05-20
US20090175951A1 (en) 2009-07-09
US20120087984A1 (en) 2012-04-12
EP1427395A1 (en) 2004-06-16
US20070224279A1 (en) 2007-09-27
DK1427395T3 (en) 2008-06-30
US20030054042A1 (en) 2003-03-20
EP1427395B1 (en) 2008-03-12
ES2303553T3 (en) 2008-08-16
JP2010280687A (en) 2010-12-16
ATE388690T1 (en) 2008-03-15

Similar Documents

Publication Publication Date Title
CA2460436C (en) Stabilization of active agents by formulation into nanoparticulate form
EP1471887B1 (en) Nanoparticulate compositions having lysozyme as a surface stabilizer
US7695739B2 (en) In vitro methods for evaluating the in vivo effectiveness of dosage forms of microparticulate or nanoparticulate active agent compositions
CA2523035C (en) Sterilization of dispersions of nanoparticulate active agents with gamma radiation
EP1503737B1 (en) Nanoparticulate nystatin formulations
EP1585502B1 (en) Fast-disintegrating solid dosage forms being not friable and comprising pullulan
EP1443912B1 (en) Compositions having a combination of immediate release and controlled release characteristics
US20080152585A1 (en) Low viscosity liquid dosage forms
US20110165251A1 (en) Liquid dosage compositions of stable nanoparticulate active agents
US20040105778A1 (en) Gamma irradiation of solid nanoparticulate active agents
WO2008008733A2 (en) Nanoparticulate sorafenib formulations
US20110159054A1 (en) Nanoparticulate bicalutamide formulations
CA2914782C (en) Nanoparticles of indirubin, derivatives thereof and methods of making and using same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BY BZ CA CH CN CO CR CU CZ DE DM DZ EC EE ES FI GB GD GE GH HR HU ID IL IN IS JP KE KG KP KR LC LK LR LS LT LU LV MA MD MG MN MW MX MZ NO NZ OM PH PL PT RU SD SE SG SI SK SL TJ TM TN TR TZ UA UG US UZ VC VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZM ZW AM AZ BY KG KZ RU TJ TM AT BE BG CH CY CZ DK EE ES FI FR GB GR IE IT LU MC PT SE SK TR BF BJ CF CG CI GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003528521

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2460436

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002775709

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002775709

Country of ref document: EP